US20190134059A1 - Fulvestrant formulations and methods of their use - Google Patents

Fulvestrant formulations and methods of their use Download PDF

Info

Publication number
US20190134059A1
US20190134059A1 US16/099,265 US201716099265A US2019134059A1 US 20190134059 A1 US20190134059 A1 US 20190134059A1 US 201716099265 A US201716099265 A US 201716099265A US 2019134059 A1 US2019134059 A1 US 2019134059A1
Authority
US
United States
Prior art keywords
microns
fulvestrant
suspension
pharmaceutical composition
aqueous
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/099,265
Inventor
Feng-Jing Chen
Steven L. Krill
Rama Abu Shmeis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eagle Pharmaceuticals Inc
Original Assignee
Eagle Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eagle Pharmaceuticals Inc filed Critical Eagle Pharmaceuticals Inc
Priority to US16/099,265 priority Critical patent/US20190134059A1/en
Assigned to EAGLE PHARMACEUTICALS, INC. reassignment EAGLE PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, FENG-JING, KRILL, STEVEN L., SHMEIS, RAMA ABU
Publication of US20190134059A1 publication Critical patent/US20190134059A1/en
Assigned to JPMORGAN CHASE BANK, N.A., AS ADMINISTRATIVE AGENT reassignment JPMORGAN CHASE BANK, N.A., AS ADMINISTRATIVE AGENT SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EAGLE PHARMACEUTICALS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the disclosure is directed to fulvestrant-containing formulations and methods of their use in the treatment of disease.
  • Fulvestrant or 7-(9-(4,4,5,5,5-pentafluoropentylsulfinyl)nonyl)estra-1,3,5(10)-triene-3,17-diol, has the structure of formula (1):
  • Fulvestrant is a selective estrogen receptor degrader (SERD) indicated for the treatment of hormone receptor positive metastatic breast cancer in postmenopausal women with disease progression following anti-estrogen therapy.
  • SESD selective estrogen receptor degrader
  • Fulvestrant As with other steroidal-like compounds, fulvestrant has physical properties which make preparing fulvestrant pharmaceutical compositions difficult. Fulvestrant is a particularly lipophilic molecule, even when compared with other steroidal compounds, and its aqueous solubility is extremely low.
  • fulvestrant Due to the poor solubility and oral bioavailability of fulvestrant, the drug is currently administered via intramuscular injection of an oil-based fulvestrant formulation.
  • the current commercial formulation of fulvestrant, FASLODEXTM is dosed at 500 mg and requires that two 5 mL injections of a 50 mg/mL fulvestrant formulation be administered intramuscularly. Each 5 mL injection contains 10% w/v alcohol, 10% w/v benzyl alcohol, and 15% w/v benzyl benzoate as co-solvents and made up to 100% w/v with castor oil as a further co-solvent and release rate modifier.
  • the present disclosure provides formulations comprising fulvestrant particles.
  • the disclosure also provides fulvestrant suspensions, preferably those having a fulvestrant concentration of equal to or greater than about 50 mg/mL.
  • the disclosure also provides formulations comprising fulvestrant particles and a non-oil vehicle. Some aspects of the disclosure are directed to pharmaceutical compositions comprising fulvestrant particles having an LD Dv(90) greater than or equal to about 7 microns.
  • compositions comprising fulvestrant particles having a CE Dv(90) less than about 200 microns, for example, between about 10 microns and about 200 microns, a CE Dv(50) less than about 60 microns, for example, between about 5 microns and about 60 microns, and a CE Dv(10) less than 25 microns, for example, between about 1 microns and about 25 microns.
  • aspects of the disclosure are directed to pharmaceutical compositions comprising fulvestrant at a concentration of about 100 mg/mL, whereupon administration to a subject, the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of the pharmaceutical compositions of the disclosure is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of a reference listed fulvestrant product.
  • Other aspects of the disclosure are directed to fulvestrant formulations having a concentration of about 100 mg/mL and particular pharmacokinetic profiles.
  • the disclosure is directed to pharmaceutical compositions comprising fulvestrant particles, wherein the fulvestrant concentration is about 40 to 125 mg/mL.
  • FIG. 1A depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FASLODEXTM commercial fulvestrant formulations
  • FIG. 1A depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 1B depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FASLODEXTM commercial fulvestrant formulations
  • FIG. 1B depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 2A depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FASLODEXTM commercial fulvestrant formulations
  • FIG. 2A depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 2B depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FASLODEXTM commercial fulvestrant formulations
  • FIG. 2B depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 2C depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 3 depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 4 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 5 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 6 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 7 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 8 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 9 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure.
  • FIG. 10 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 11 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 12 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure
  • FIG. 13 depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 14 depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEXTM) and some exemplary fulvestrant formulations of the present disclosure to canines; and
  • FIG. 15 depicts schematic representations of aspects of methods of preparation of some exemplary fulvestrant formulations of the present disclosure.
  • pharmaceutically acceptable indicates that the designated entity such as, for example, e.g., a pharmaceutically acceptable excipient is generally chemically and/or physically compatible with other ingredients in a formulation, and/or is generally physiologically compatible with the recipient thereof.
  • pharmaceutical composition refers to a formulation as described herein that includes one or more pharmaceutically acceptable excipients, that is suitable for administration to a subject. It should be understood that the term “pharmaceutical composition” encompasses (a) suspensions and (b) suspensions which have been dried such that one or more solvents have been removed partially or completely, either by evaporation or sublimation, including, but not limited to, lyophilized cakes.
  • subject(s), “individual(s),” and “patient(s)”, refer to mammals, including humans.
  • human(s) refers to and includes, a human child, adolescent, or adult.
  • treats refer to and include ameliorative, palliative, and/or curative uses and results, or any combination thereof.
  • the methods described herein can be used prophylactically. It should be understood that “prophylaxis” or a prophylactic use or result do not refer to nor require absolute or total prevention (i.e., a 100% preventative or protective use or result).
  • prophylaxis or a prophylactic use or result refer to uses and results in which administration of a compound or formulation diminishes or reduces the severity of a particular condition, symptom, disorder, or disease described herein; diminishes or reduces the likelihood of experiencing a particular condition, symptom, disorder, or disease described herein; or delays the onset or relapse (reoccurrence) of a particular condition, symptom, disorder, or disease described herein; or any combination of the foregoing.
  • therapeutic and “therapeutically effective amount”, refer to an amount of a compound or formulation that (a) treats a particular condition, symptom, disorder, or disease described herein; (b) attenuates, ameliorates or eliminates one or more symptoms of a particular condition, disorder, or disease described herein; (c) delays the onset or relapse (reoccurrence) of a particular condition, symptom, disorder, or disease described herein. It should be understood that the terms “therapeutic” and “therapeutically effective” encompass any one of the aforementioned effects (a)-(c), either alone or in combination with any of the others (a)-(c).
  • therapeutic agent refers to any substance included in a formulation that is useful in the treatment of a disease, condition, or disorder or comorbidity (i.e., a disease, condition, or disorder that exists simultaneously with breast cancer) and is not fulvestrant.
  • suspension refers to solid particles dispersed in a liquid vehicle.
  • formulation refers to a mixture of components.
  • formulation encompasses pharmaceutical compositions, and suspensions, as well as suspensions that have been dried such that one or more solvents have been removed partially or completely (e.g., lyophilized cakes).
  • Dv(10)”, “Dv(50)” and “Dv(90)” are defined as the volume weighted particle diameters where a cumulative 10%, 50% or 90% v/v of the particles have an equal or smaller diameter, respectively, when measured. For example, if a particle population has a Dv(50) of about 25 microns, 50% of the particles in volume have a diameter of less than or equal to about 25 microns.
  • Dn(10)”, “Dn(50)” and “Dn(90)” are defined as the number weighted particle diameters where a cumulative 10%, 50% or 90% of the particles have an equal or smaller diameter, respectively, when measured. For example, if a particle population has a Dn(50) of about 25 microns, 50% of the particles in number have a diameter of less than or equal to about 25 microns.
  • Particle size and particle size distributions can be determined by measurement via laser diffraction.
  • Particle size analysis by laser diffraction methods is known in the art and is explained more fully by ISO 13320:2009(E), “Particle size analysis—Laser diffraction methods,” International Organization for Standardization which is incorporated by reference herein in its entirety for all purposes.
  • Particle sizes determined by laser diffraction are represented as the diameter of a sphere having equivalent volume to the particle volume as determined by Mie theory of light scattering. Tables 1-7 and 23-27 and FIGS.
  • PSD laser diffraction particle size and particle size distribution
  • Particle size and particle size distributions can also be determined by microscopy image capture and analysis.
  • Microscopy image capture and analysis captures a two dimensional (2D) image of a 3D particle and calculates various size and shape parameters from the 2D image.
  • Particle sizes determined by microscopy image capture and analysis are represented as the diameter of a circle with the equivalent area as the 2D image of the particle, referred to herein as a circle equivalent or “CE” diameter.
  • CEC circle equivalent or “CE” diameter.
  • Particle size analysis by microscopy image capture and analysis is known in the art and is explained more fully by ISO 13322-1:2014, “Particle size analysis—Image analysis methods—Part 1: Static image analysis methods,” International Organization for Standardization, which is incorporated by reference herein in its entirety for all purposes.
  • Tables 41-50 provide microscopy image capture and analysis particle size and particle size distribution data for some exemplary embodiments of the present invention, with measurement samples taken during methods of preparation, after an initial suspension is formed, or after lyophilization and reconstitution, as indicated.
  • the invention is directed to suspensions comprising fulvestrant particles and a vehicle.
  • the fulvestrant particles may have different particle size distributions as described more fully elsewhere herein.
  • a “vehicle” is a suspending medium, preferably a pharmaceutically acceptable suspending medium.
  • the vehicle is a non-oil vehicle.
  • “oils” are non-polar substances that have no or low miscibility with water. Castor oil is an example of an oil.
  • the vehicle comprises water, i.e., is aqueous.
  • an “aqueous” vehicle is a vehicle that comprises at least about 50% w/w water.
  • the aqueous vehicle comprises at least about 60% w/w, at least about 70% w/w, at least about 80% w/w, at least about 85% w/w, at least about 90% w/w, at least about 95% w/w, at least about 96% w/w, at least about 97% w/w, at least about 98% w/w, or at least about 99% w/w water.
  • the vehicle is water.
  • the vehicle is a non-aqueous medium.
  • a vehicle comprises a single suspending medium.
  • a vehicle comprises a mixture of two or more suspending mediums, which may be aqueous or non-aqueous.
  • the vehicle comprises both water and a non-aqueous solvent.
  • the suspension is substantially oil-free.
  • a “substantially oil-free” suspension is a suspension comprising a vehicle that comprises at most about 10% w/w oil.
  • a substantially oil-free suspension comprises a vehicle that comprises less than about 5% w/w oil, less than about 2% w/w oil, less than about 1% w/w oil, less than about 0.5% w/w oil, less than about 0.1% w/w oil, or comprises a vehicle that is free of oil.
  • Fulvestrant suspensions of the disclosure can have fulvestrant present at a concentration of about 40 mg/mL to about 125 mg/mL in a vehicle.
  • the fulvestrant present in the fulvestrant suspensions may have different particle size distributions as described more fully elsewhere herein.
  • fulvestrant is present at a concentration equal to or greater than about 40 mg/mL.
  • fulvestrant is present at a concentration of about 40 to about 75 mg/mL.
  • fulvestrant is present at a concentration of about 75 mg/mL to about 125 mg/mL.
  • fulvestrant is present at a concentration of about 40 mg/mL, about 45 mg/mL, about 50 mg/mL, about 55 mg/mL, about 60 mg/mL, about 65 mg/mL, about 70 mg/mL, or about 75 mg/mL.
  • fulvestrant is present in the suspension at a concentration equal to or greater than about 75 mg/mL. In further embodiments, fulvestrant is present in the suspension at a concentration of about 75 to about 125 mg/mL. In particular embodiments, fulvestrant is present in the suspension at a concentration of about 80 mg/mL, about 85 mg/mL, about 90 mg/mL, about 95 mg/mL, about 100 mg/mL, about 105 mg/mL, about 110 mg/mL, about 115 mg/mL, about 120 mg/mL, or about 125 mg/mL.
  • fulvestrant is present in the suspension at a concentration of about 75 mg/mL to about 95 mg/mL, about 80 mg/mL to about 100 mg/mL, about 90 mg/mL to about 110 mg/ml, about 95 mg/mL to about 105 mg/mL, about 95 mg/mL to about 115 mg/mL, about 100 mg/mL to about 110 mg/mL, about 110 mg/mL to about 125 mg/mL, including all ranges and subranges there between.
  • compositions Comprising Fulvestrant
  • compositions comprising fulvestrant.
  • These pharmaceutical compositions may be prepared by combining fulvestrant, as described herein, with one or more additional excipients, preferably pharmaceutically acceptable excipients.
  • the pharmaceutical compositions may further comprise a stabilizer, or one or more stabilizers, or two or more stabilizers.
  • the stabilizer is selected from the group consisting of surfactants, polymers, cross-linked polymers, buffering agents, electrolytes, and non-electrolytes.
  • the pharmaceutical composition comprises a combination of two or more stabilizers selected from the group consisting of surfactants, polymers, cross-linked polymers, buffering agents, electrolytes, and non-electrolytes.
  • the pharmaceutical compositions comprising fulvestrant comprise about 0.2 mg/mL to about 75 mg/mL of one or more stabilizers, and all ranges and subranges therebetween.
  • the pharmaceutical composition comprises about 0.2 to 0.7 mg/mL, 0.5 to 1 mg/mL, 1 to 5 mg/mL, 2 to 8 mg/mL, 5 to 6 mg/mL, 5 to 10 mg/mL, 8 to 12 mg/mL, 10 to 15 mg/mL, 15 to 20 mg/mL, 20 to 30 mg/mL, 30 to 40 mg/mL, 40 to 50 mg/mL, 45 to 55 mg/mL, 50 to 60 mg/mL, or 60 to 75 mg/mL of one or more stabilizers, and all ranges and subranges there between.
  • the pharmaceutical composition comprises about 0.2 mg/mL, 0.5 mg/mL, 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, 5 mg/mL, 5.5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, 10 mg/mL, 12 mg/mL, 15 mg/mL, 17 mg/mL, 20 mg/mL, 25 mg/mL, 30 mg/mL, 35 mg/mL, 40 mg/mL, 45 mg/mL, 50 mg/mL, 55 mg/mL, 60 mg/mL, 65 mg/mL, 70 mg/mL, or about 75 mg/mL of one or more stabilizers.
  • the stabilizer is a surfactant.
  • the stabilizer can be, but is not limited to, polyethylene oxide (PEO), a PEO derivative, polysorbate 80, polysorbate 20, poloxamer 188 (including, but not limited to, PLURONIC® F-68 poloxamer sold by BASF Corp. (Wyandotte, Mich., USA)), poloxamer 124 (including, but not limited to, PLURONIC® L44 poloxamer sold by BASF Corp. (Wyandotte, Mich., USA)), poloxamer 407 (including, but not limited to, PLURONIC® F127 poloxamer sold by BASF Corp.
  • polyethoxylated vegetable oils including but not limited to KOLLIPHOR® EL, formerly known as CREMOPHOR® EL sold by BASF Corp. (Wyandotte, Mich., USA)
  • sorbitan palmitate including, but not limited to, SPANTM 40 sold by Croda International Plc
  • lecithin poly(vinyl alcohol) (“PVA”)
  • human serum albumin and mixtures thereof.
  • the stabilizer is a polymer.
  • the stabilizer can be, but is not limited to, a polyvinylpyrrolidone (“PVP”) (such as, but not limited to povidone K12, povidone K17, PLASDONETM C-12 povidone, PLASDONETM C-17 povidone, PLASDONETM C-30 povidone, and mixtures thereof), polyethylene glycol 3350, and mixtures thereof.
  • PVP polyvinylpyrrolidone
  • the stabilizer is an electrolyte, i.e., a salt that dissociates into anions and cations in aqueous solution.
  • the stabilizer can be, but is not limited to, sodium chloride, calcium chloride, and mixtures thereof.
  • the stabilizer is a non-electrolyte, i.e., is non-ionic.
  • the stabilizer can be, but is not limited to, dextrose, glycerol (also referred to as glycerin), mannitol, or mixtures thereof.
  • the stabilizer is a cross-linked polymer.
  • the stabilizer can be, but is not limited to, carboxymethylcellulose sodium (CMC).
  • CMC carboxymethylcellulose sodium
  • the stabilizer is CMC 7LF, CMC 7MF, CMC 7HF, or mixtures thereof.
  • the stabilizer is a buffering agent, for example, NaH 2 PO 4 .H 2 O, NaH 2 PO 4 .2H 2 O, anhydrous NaH 2 PO 4 , sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or mixtures thereof.
  • a buffering agent for example, NaH 2 PO 4 .H 2 O, NaH 2 PO 4 .2H 2 O, anhydrous NaH 2 PO 4 , sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or mixtures thereof.
  • combinations of non-electrolyte stabilizers and electrolyte stabilizers may be used.
  • the combination of stabilizers may comprise two or more non-electrolyte stabilizers.
  • the combination of stabilizers may comprise two or more electrolyte stabilizers.
  • the combination of stabilizers may comprise one or more non-electrolyte stabilizers and one or more electrolyte stabilizers.
  • the combination of stabilizers may comprise two or more of mannitol, dextrose, and sodium chloride.
  • combinations of surfactant stabilizers and polymer stabilizers may be used.
  • the combination of stabilizers may comprise two or more surfactant stabilizers.
  • the combination of stabilizers may comprise two or more polymer stabilizers.
  • the combination of stabilizers may comprise one or more surfactant stabilizers and one or more polymer stabilizers.
  • the combination of stabilizers may comprise two or more of polysorbate 80, polysorbate 20, and poloxamer 188.
  • the combination of stabilizers may comprise one or more of polysorbate 80, polysorbate 20, and poloxamer 188 and one or more of povidone K12, povidone K17, PLASDONETM C-12 povidone, PLASDONETM C-17 povidone, PLASDONETM C-30 povidone, and polyethylene glycol 3350.
  • the combination of stabilizers may comprise polysorbate 80 and one or more of PLASDONETM C-12 povidone and povidone K12.
  • the pharmaceutical compositions comprising fulvestrant comprise CMC (carboxymethylcellulose sodium).
  • CMC carboxymethylcellulose sodium
  • the CMC is prepared and heat sterilized before being combined with the fulvestrant during methods of preparation (described more fully elsewhere herein).
  • the viscosity of a CMC solution can be modulated by the degree of heating applied, which can allow for the formation of a plurality of fulvestrant pharmaceutical compositions having identical constituents, but with different viscosity values. These different viscosity values can affect the physical stability of the fulvestrant pharmaceutical compositions and the pharmacokinetic characteristics upon administration to subjects.
  • fulvestrant pharmaceutical compositions comprising CMC may be prepared in two or more parts with each part comprising a different amount of CMC. In other embodiments one or more such parts may be a suspension free of any CMC. In further embodiments, the parts can be mixed in an appropriate ratio to obtain a desired pharmaceutical composition.
  • the pharmaceutical compositions in the form of liquid suspensions comprising fulvestrant and one or more stabilizers may exhibit different sedimentation behaviors to form either flocculated or caked suspension upon storage.
  • pharmaceutical compositions in the form of liquid suspensions comprising fulvestrant after being stored, can be redispersed back into a homogeneous suspension with an acceptable particle size distribution upon redispersion.
  • Exemplary liquid suspension formulations described herein were prepared and tested for sedimentation and redispersion. The tested formulations exhibited different sedimentation behaviors, but all were redispersible back to an acceptable, homogeneous suspension after a 3-month storage period at room temperature.
  • the pharmaceutical compositions comprising fulvestrant have a pH of from about 3-10, for example, about 3, 4, 5, 6, 7, 8, 9, or about 10.
  • the pharmaceutical composition has a pH of from about 5-8.
  • the pharmaceutical composition has a pH of from about 6-8.
  • the pharmaceutical composition has a pH of from about 3-7.
  • the pharmaceutical composition has a pH of about 6.0 to 8.0.
  • the pharmaceutical composition has a pH of about 6.0 to 7.0, 6.5 to 7.0, 6.5 to 7.5, 6.7 to 7.2, 7.0 to 7.2, 7.0 to 7.5, or 7.0 to 8.0.
  • the pharmaceutical composition has a pH of about 7.0.
  • the pharmaceutical composition further comprises one or more buffering agents, i.e., an agent that when added to a pharmaceutical composition, results in a pharmaceutical composition that resists pH changes or that results in a change in pH, such as, but not limited to, NaH 2 PO 4 .H 2 O, NaH 2 PO 4 .2H 2 O, anhydrous NaH 2 PO 4 , sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or mixtures thereof.
  • the pharmaceutical composition comprises about 1 mM to 20 mM, of one or more buffering agents, and all ranges and subranges therebetween.
  • the pharmaceutical composition comprises about 1 to 2 mM, 1 to 3 mM, 1 to 5 mM, 2 to 8 mM, 5 to 6 mM, 5 to 10 mM, 8 to 12 mM, 10 to 15 mM, or 15 to 20 mM of one or more buffering agents, and all ranges and subranges there between.
  • the pharmaceutical composition comprises about 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, 15 mM, 16 mM, 17 mM, 18 mM, 19 mM, or 20 mM of one or more buffering agents.
  • the pharmaceutical composition has an osmolarity from about 280 mOsm/L to about 310 mOsm/L, for example, about 280, 285, 290, 300, 305, or about 310 mOsm/L. In further embodiments of the invention, the pharmaceutical composition has an osmolarity from about 290 mOsm/L to about 300 mOsm/L. In yet further embodiments of the invention, the pharmaceutical composition has an osmolarity of about 290 mOsm/L.
  • the osmolarity may be selected through the use of appropriate amounts of the one or more stabilizers, e.g., stabilizers that also act as tonicifiers, such as, but not limited to, the non-electrolyte stabilizers and electrolyte stabilizers described herein.
  • the osmolarity may be selected through the use of appropriate amounts of one or more buffering agents that act as tonicifiers in a pharmaceutical composition, such as, but not limited to, the buffering agents described herein.
  • the pharmaceutical composition has an absolute viscosity measured at 25° C. from about 1.0 cP to about 1000 cP, and all ranges and subranges therebetween. In particular embodiments of the invention, the pharmaceutical composition has an absolute viscosity measured at 25° C.
  • the pharmaceutical composition has an absolute viscosity measured at 25° C. of about 1.0 cP, 1.1 cP, 1.2 cP, 1.3 cP, 1.4 cP, 1.5 cP, 1.6 cP, 1.7 cP, 1.8 cP, 1.9 cP, 2.0 cP, 2.1 cP, 2.2 cP, 2.3 cP, 2.4 cP, 2.5 cP, 2.6 cP, 2.7 cP, 2.8 cP, 2.9 cP, 3.0 cP, 3.5 cP, 4.0 cP, 4.5 cP, 5.0 cP, 10 cP, 15 cP, or 20 cP.
  • compositions having a fulvestrant concentration of 50 mg/mL or 100 mg/mL have an absolute viscosity measured at 25° C. that is from about 2-fold to about 500-fold lower than FASLODEXTM, and all ranges and subranges therebetween.
  • fulvestrant pharmaceutical compositions having a fulvestrant concentration of 50 mg/mL or 100 mg/mL have an absolute viscosity measured at 25° C.
  • fulvestrant pharmaceutical compositions having a fulvestrant concentration of 50 mg/mL or 100 mg/mL have an absolute viscosity measured at 25° C. that is substantially equivalent to FASLODEXTM.
  • Table 21 provides density measurements of some exemplary fulvestrant pharmaceutical compositions of the present disclosure.
  • Table 22 provides viscosity measurements of some exemplary fulvestrant pharmaceutical compositions of the present disclosure.
  • the pharmaceutical composition comprises one or more additional pharmaceutically acceptable excipients.
  • a pharmaceutically acceptable excipient is generally chemically and/or physically compatible with other ingredients in a pharmaceutical composition or pharmaceutical composition, and/or is generally physiologically compatible with the recipient thereof.
  • the one or more additional pharmaceutically acceptable excipients are selected from the group consisting of preservatives, antioxidants, or mixtures thereof.
  • the additional pharmaceutically acceptable excipient is a preservative such as, but not limited to, phenol, cresol, p-hydroxybenzoic ester, chlorobutanol, or mixtures thereof.
  • the additional pharmaceutically acceptable excipient is an antioxidant such as, but not limited to, ascorbic acid, sodium pyrosulfite, palmitic acid, butylated hydroxyanisole, butylated hydroxytoluene, tocopherols, or mixtures thereof.
  • an antioxidant such as, but not limited to, ascorbic acid, sodium pyrosulfite, palmitic acid, butylated hydroxyanisole, butylated hydroxytoluene, tocopherols, or mixtures thereof.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5.8 mg/mL of one or more stabilizers, and water for injection (WFI) q.s. to volume.
  • WFI water for injection
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 0.8 mg/mL of one or more polymers, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 0.8 mg/mL of povidone K12 (PVP 12K), and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5.8 mg/mL of one or more stabilizers, about 9 mg/mL of one or more electrolytes, about 10 mM of one or more buffering agents, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 0.8 mg/mL of one or more polymers, about 9 mg/mL of sodium chloride, about 10 mM of one or more of NaH 2 PO 4 .H 2 O, NaH 2 PO 4 .2H 2 O, and anhydrous NaH 2 PO 4 (preferably a mixture of about 0.61 mg/mL NaH 2 PO 4 .2H 2 O and about 0.85 mg/mL of anhydrous NaH 2 PO 4 ), and WFI q.s. to volume
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 55 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 50 mg/mL of dextrose, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more stabilizers, about 9 mg/mL of one or more electrolytes, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 9 mg/mL of sodium chloride, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 9 mg/mL of sodium chloride, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 55 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 56.6 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 1.6 mg/mL of one or more polymers, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.6 mg/mL of PLASDONETM C-12 povidone, povidone K12, or a mixture thereof, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 57.4 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 2.4 mg/mL of one or more polymers, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 2.4 mg/mL of PLASDONETM C-12 povidone, povidone K12, or a mixture thereof, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, between about 1 mg/mL and 2.4 mg/mL of PVP, sorbitan palmitate, poloxamer 188, poloxamer 124, poloxamer 427, polyethoxylated castor oil, PVA, or a mixture thereof, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 2.4 mg/mL of PVA, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.0 mg/mL of polyethoxylated castor oil, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 2.0 mg/mL of poloxamer 188, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of poloxamer 188, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of sorbitan palmitate, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of poloxamer 124, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of poloxamer 407, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • compositions comprising fulvestrant are shown in Tables 1-20 and 23-27.
  • exemplary formulations are identified as “F###” where each “#” is a numeral, e.g., F001, F002, and so on.
  • the exemplary formulations sharing an initial identification “F###” share identical concentrations of constituent components (mg/mL), but may vary in their properties due to different methods of preparation, particle size distributions of fulvestrant, or other differences in processing, storage, or handling.
  • Such exemplary formulation sharing an initial identification scheme “F###” are further identified by extra alphanumeric characters.
  • the exemplary formulations F003a, F003b, and F003k2 have the same concentrations of constituent components but may differ in, e.g., the underlying methods of preparation and resulting particle size distributions.
  • formulations are identified by only ending non-zero numerals # or ## and subsequent alphanumeric characters; for example, formulation F003a may be referred to as “Variant 3a”, formulation F005a2 may be referred to as “Variant 5a2”, and the like.
  • Lot Some exemplary formulations are identified and referred to as “Lot”s, with references to the same Lot number referring to exemplary formulations having the same concentrations of constituent components, but may vary in their properties due to different methods of preparation, particle size distributions of fulvestrant, or other differences in processing, storage, or handling.
  • Formulation L3F Formulation L6 Fulvestrant 50 100 Polysorbate 80 5 5 PVP 12K 0.8 0.8 NaCl 9 9 Phosphate buffer* 10 mM 10 mM WFI q.s. to volume q.s.
  • Particular embodiments of the disclosure comprise solid fulvestrant particles, for example a fulvestrant suspension comprising solid fulvestrant particles.
  • a fulvestrant suspension comprising solid fulvestrant particles.
  • at least about 90% of the total fulvestrant in the formulation is present as solid particles.
  • at least about 80% of the total fulvestrant in the formulation is present as solid particles.
  • solid fulvestrant particles are particles consisting of crystalline and/or amorphous fulvestrant.
  • fulvestrant particles comprise crystalline and/or amorphous fulvestrant as well as other excipients.
  • fulvestrant particles comprise crystalline and/or amorphous fulvestrant coated or surface modified by a surface modifier adsorbed on the surface of the particle.
  • the surface modifier can be a stabilizer such as, but not limited to surfactants, polymers, electrolytes, and non-electrolytes, and mixtures thereof.
  • fulvestrant in forms other than a solid particle, such as, but not limited to, solubilized fulvestrant as a free molecule or associated with a suspension such as micelles, microemulsions, emulsion, liposome, and combinations thereof, or complexed with other formulation constituents in a vehicle.
  • a suspension such as micelles, microemulsions, emulsion, liposome, and combinations thereof, or complexed with other formulation constituents in a vehicle.
  • such other forms of fulvestrant are in equilibrium with the fulvestrant solid particles.
  • the fulvestrant particles comprise about 90-99.9% by weight of fulvestrant and 0.1-10% by weight of a surface modifier adsorbed on the surface of said particle.
  • the surface modifier is a stabilizer such as, but not limited to surfactants, polymers, electrolytes, and non-electrolytes, and mixtures thereof.
  • fulvestrant particles comprise at least about 90% fulvestrant. In other embodiments of the invention fulvestrant particles comprise at least about 92%, 95%, 97%, 98%, 99%, 99.5%, or 99.9% fulvestrant.
  • one or more solvents, such as water, present in the pharmaceutical composition can be removed partially or completely by appropriate techniques known to the art, such as lyophilization or spray drying, to form a dried pharmaceutical composition for reconstitution.
  • the dried pharmaceutical composition can comprise up to about 1%, about 2%, about 5%, or about 10% of the one or more solvents.
  • the dried pharmaceutical composition can be reconstituted with appropriate diluent known to the art, such as, but not limited to water for injection (WFI), normal saline (NS), and 5% dextrose in water (D5W) prior to administration.
  • the diluent can further comprise an organic solvent or one or more of the excipients described herein. Dried pharmaceutical compositions formed by lyophilization may be in the form of a lyophilized cake.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In yet further embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In other embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 0.5 microns. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 0.5 microns. In other embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 microns. In still other embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1.5 microns.
  • the fulvestrant particles have a laser diffraction diameter less than about 1.5 microns. In yet other embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 0.5 microns. In other embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In still other embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 1.5 microns. In yet other embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns.
  • the fulvestrant particles have an LD Dv(90) between about 4 microns and about 120 microns, between about 4 microns and about 100 microns, between about 4 microns and about 75 microns, between about 4 microns and about 60 microns, between about 4 microns and about 50 microns, between about 4 microns and about 40 microns, between about 4 microns and about 30 microns, between about 4 microns and about 20 microns, between about 4 microns and about 15 microns, between about 4 microns and about 10 microns, between about 20 microns and about 60 microns, between about 20 microns and about 45 microns, between about 20 microns and about 30 microns, between about 30 microns and about 50 microns, or between about 4 microns and about 9 microns.
  • the fulvestrant particles have a LD Dv(90) equal to about 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, about 60 microns, about 65 microns, about 70 microns, about 75 microns, about 80 microns, about 85 microns, about 90 microns, about 95 microns, about 100 microns, about 105 microns, about 110 microns, about 115 microns, or about 120 microns.
  • the fulvestrant particles have an LD Dv(90) less than or equal to about 120 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 100 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 80 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 60 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 50 microns.
  • the fulvestrant particles have an LD Dv(90) less than or equal to about 40 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 30 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 25 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 18 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 16 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 14 microns.
  • the particles have an LD Dv(90) less than or equal to about 11 microns. In yet further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 9 microns. In yet further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 7 microns. In yet further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 5 microns. In particular embodiments of the invention, particles have an LD Dv(90) between about 9-14 microns. In other embodiments of the invention, the particles have an LD Dv(90) between about 12-14 microns.
  • the particles have an LD Dv(90) between about 9-11 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 7-9 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-8 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-7 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 3-6 microns.
  • the fulvestrant particles have an LD Dv(50) between about 2 microns and about 35 microns, between about 2 microns and about 25 microns, between about 2 microns and about 20 microns, between about 2 microns and about 15 microns, between about 2 microns and about 10 microns, between about 2 microns and about 8 microns, between about 2 microns and about 7 microns, between about 2 microns and about 6 microns, between about 2 microns and about 5 microns, between about 2 microns and about 4 microns, between about 5 microns and about 10 microns, between about 5 microns and about 15 microns, between about 7 microns and about 10 microns, between about 8 microns and about 10 microns, or between about 9 microns and about 16 microns.
  • the fulvestrant particles have a LD Dv(50) equal to about 2 microns, 3 microns, 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 25 microns, about 30 microns, or about 35 microns.
  • the fulvestrant particles have an LD Dv(50) less than or equal to about 9 microns. In other embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 7 microns. In other embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 6 microns. In yet other embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 5 microns. In particular embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 4 microns. In further embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 3 microns.
  • the particles have an LD Dv(50) between about 4-6 microns. In further embodiments of the invention, the particles have an LD Dv(50) between about 3-5 microns. In yet further embodiments of the invention, the particles have an LD Dv(50) between about 3-4 microns. In yet further embodiments of the invention, the particles have an LD Dv(50) between about 2-3 microns.
  • the fulvestrant particles have an LD Dv(10) no more than about 3 microns, about 2 microns, or about 1 microns. In further embodiments of the invention, the particles have an LD Dv(10) between about 1 micron and about 3 microns. In still further embodiments of the invention, the particles have an LD Dv(10) greater than or equal to about 2 microns. In yet further embodiments of the invention, the particles have an LD Dv(10) between about 1.5 microns to about 2.5 microns. In yet further embodiments of the invention, the particles have an LD Dv(10) between about 1 micron to about 2 microns.
  • the particles have an LD Dv(10) between about 1.0 micron to about 1.5 microns. In even further embodiments of the invention, the particles have an LD Dv(10) of about 2 microns. In even further embodiments of the invention, the particles have an LD Dv(10) of about 1.5 microns.
  • the fulvestrant particles have an LD Dv(90) less than or equal to about 25 microns and an LD Dv(50) less than or equal to about 9 microns.
  • the particles have an LD Dv(90) less than or equal to about 16 microns and an LD Dv(50) less than or equal to about 6 microns.
  • the particles have an LD Dv(90) less than or equal to about 11 microns and an LD Dv(50) less than or equal to about 5 microns.
  • the particles have an LD Dv(90) less than or equal to about 9 microns and an LD Dv(50) less than or equal to about 4 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 8 microns and an LD Dv(50) less than or equal to about 4 microns.
  • the fulvestrant particles have an LD Dv(90) between about 9-14 microns and an LD Dv(50) between about 4-6 microns. In still other embodiments of the invention, the particles have an LD Dv(90) between about 9-11 microns and an LD Dv(50) between about 4-6 microns. In particular embodiments of the invention, the particles have an LD Dv(90) between about 12-14 microns and an LD Dv(50) between about 4-6 microns. In further embodiments of the invention, the particles have an LD Dv(90) between about 6-8 microns and an LD Dv(50) between about 2-4 microns.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In yet further embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In other embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • the fulvestrant particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns. In other embodiments of the invention, the particles have an LD Dv(90) between about 9-11 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 12-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns.
  • the particles have an LD Dv(90) between about 6-9 microns, an LD Dv(50) between about 2-4 microns, and an LD Dv(10) between about 1-2 microns.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron.
  • at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns.
  • at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • the fulvestrant particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron.
  • the particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron.
  • the fulvestrant particles have an LD Dv(90) between about 30 microns and about 110 microns, an LD Dv(50) between about 5 microns and about 30 microns, and an LD Dv(10) between about 1.5 microns and about 3 microns. In other embodiments of the invention, the particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns.
  • the particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • the particles have an LD Dv(90) between about 6-9 microns, an LD Dv(50) between about 2-4 microns, an LD Dv(10) between about 1-2 microns, and the fulvestrant particles have a laser diffraction diameter greater than or equal to about 0.5 microns.
  • the particles have an LD Dv(90) between about 6-9 microns, an LD Dv(50) between about 2-4 microns, an LD Dv(10) between about 1-2 microns, and at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 0.5 microns.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron.
  • at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron.
  • the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns.
  • at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • the fulvestrant particles have a CE Dv(10) between about 1 microns and about 25 microns, between about 2 microns and about 25 microns, between about 3 microns and about 7 microns, between about 4 microns and about 15 microns, between about 4 microns and about 10 microns, between about 4 microns and about 8 microns, between about 6 microns and about 8 microns, between about 6 microns and about 7 microns, or between about 1 microns and about 10 microns.
  • the fulvestrant particles have a CE Dv(10) equal to about 1 micron, about 2 microns, about 3 microns, about 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 21 microns, about 22 microns, about 23 microns, about 24 microns, or about 25 microns.
  • the fulvestrant particles have a CE Dv(50) between about 5 microns and about 60 microns, between about 5 microns and about 50 microns, between about 9 microns and about 20 microns, between about 9 microns and about 15 microns, between about 10 microns and about 50 microns, between about 10 microns and about 40 microns, between about 10 microns and about 30 microns, between about 10 microns and about 20 microns, between about 15 microns and about 30 microns, between about 15 microns and about 25 microns, between about 15 microns and about 20 microns, or between about 10 microns and about 15 microns.
  • a CE Dv(50) between about 5 microns and about 60 microns, between about 5 microns and about 50 microns, between about 9 microns and about 20 microns, between about 9 microns and about 15 microns, between about 10 microns and about 50 microns, between about 10 microns and
  • the fulvestrant particles have a CE Dv(50) equal to about 5 micron, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 21 microns, about 22 microns, about 23 microns, about 24 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, or about 60 microns.
  • the fulvestrant particles have a CE Dv(90) between about 10 microns and about 200 microns, between about 25 microns and about 150 microns, between about 25 microns and about 125 microns, between about 25 microns and about 100 microns, between about 25 microns and about 75 microns, between about 25 microns and about 50 microns, between about 25 microns and about 40 microns, between about 25 microns and about 35 microns, between about 35 microns and about 90 microns, between about 35 microns and about 75 microns, between about 35 microns and about 50 microns, between about 35 microns and about 45 microns, between about 50 microns and about 100 microns, between about 50 microns and about 75 microns, or between about 20 microns and about 40 microns.
  • the fulvestrant particles have a CE Dv(90) equal to about 10 microns, about 15 microns, about 20 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, about 60 microns, about 65 microns, about 70 microns, about 75 microns, about 80 microns, about 85 microns, about 90 microns, about 95 microns, about 100 microns, about 105 microns, about 110 microns, about 115 microns, about 120 microns, about 125 microns, about 130 microns, about 135 microns, about 140 microns, about 145 microns, about 150 microns, about 155 microns, about 160 microns, about 165 microns, about 170 microns, about 175 microns, or about 200 microns.
  • CE Dv(90) equal to about 10 microns, about
  • the fulvestrant particles have a CE Dv(90) between about 35 microns and about 90 microns, a CE Dv(50) between about 10 microns and about 35 microns, and a CE Dv(10) between about 4 microns and about 10 microns. In other embodiments of the invention, the particles have a CE Dv(90) between about 25 microns and about 60 microns, a CE Dv(50) between about 10 microns and about 25 microns, and a CE Dv(10) between about 4 microns and about 8 microns.
  • the particles have a CE Dv(90) between about 20 microns and about 35 microns, a CE Dv(50) between about 10 microns and about 20 microns, and a CE Dv(10) between about 4 microns and about 8 microns. In still other embodiments of the invention, the particles have a CE Dv(90) between about 30 microns and about 100 microns, a CE Dv(50) between about 10 microns and about 50 microns, and a CE Dv(10) between about 4 microns and about 10 microns.
  • the particles have a CE Dv(90) between about 50 microns and about 100 microns, a CE Dv(50) between about 20 microns and about 50 microns, a CE Dv(10) between about 6 microns and about 8 microns. In yet other embodiments of the invention, the particles have a CE Dv(90) between about 50 microns and about 75 microns, a CE Dv(50) between about 30 microns and about 40 microns, a CE Dv(10) between about 8 microns and about 10 microns.
  • the particles have a CE Dv(90) between about 20 microns and about 60 microns, a CE Dv(50) between about 9 microns and about 20 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In still further embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 50 microns, a CE Dv(50) between about 9 microns and about 20 microns, and a CE Dv(10) between about 3 microns and about 7 microns.
  • the particles have a CE Dv(90) between about 20 microns and about 45 microns, a CE Dv(50) between about 9 microns and about 20 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In yet further embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 40 microns, a CE Dv(50) between about 9 microns and about 15 microns, and a CE Dv(10) between about 3 microns and about 7 microns.
  • the particles have a CE Dv(90) between about 20 microns and about 35 microns, a CE Dv(50) between about 9 microns and about 15 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In still other embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 45 microns, a CE Dv(50) between about 9 microns and about 15 microns, and a CE Dv(10) between about 3 microns and about 7 microns.
  • the fulvestrant particles have a CE Dn(90) between about 4 microns and about 20 microns, between about 6 microns and about 15 microns, between about 6 microns and about 12 microns, between about 8 microns and about 12 microns, between about 8 microns and about 11 microns, between about 4 microns and about 10 microns, between about 4 microns and about 8 microns, between about 4 microns and about 7 microns, or between about 4 microns and about 6 microns.
  • the fulvestrant particles have a CE Dn(90) equal to about 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, or about 20 microns.
  • the fulvestrant particles have a CE Dn(50) between about 2.0 microns and about 10.0 microns, between about 2.0 microns and about 8.0 microns, between about 2.0 microns and about 6.0 microns, between about 2.0 microns and about 5.0 microns, between about 3.0 microns and about 5.0 microns, between about 3.5 microns and about 4.5 microns, between about 2.0 microns and about 4.0 microns, between about 2.5 microns and about 4.5 microns, or between about 2.5 microns and about 3.5 microns.
  • the fulvestrant particles have a CE Dn(50) equal to about 2.0 microns, about 2.5 microns, about 3.0 microns, about 3.5 microns, about 4.0 microns, about 4.5 microns, about 5.0 microns, about 5.5 microns, about 6.0 microns, about 6.5 microns, about 7.0 microns, about 7.5 microns, about 8.0 microns, about 8.5 microns, about 9.0 microns, about 9.5 microns, or about 10.0 microns.
  • the fulvestrant particles have a CE Dn(10) between about 0.5 microns and about 2.0 microns, between about 0.5 microns and about 1.5 microns, between about 1.0 microns and about 1.5 microns, between about 0.8 microns and about 1.2 microns, between about 0.9 microns and about 1.1 microns, or between about 0.5 microns and about 1.0 microns.
  • the fulvestrant particles have a CE Dn(10) equal to about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1.0, about 1.1, about 1.2, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2.0 microns.
  • the fulvestrant particles have a CE Dn(90) between about 4 microns and about 20 microns, a CE Dn(50) between about 2.0 microns and about 10.0 microns, and a CE Dn(10) between about 0.5 microns and about 2.0 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dn(90) between about 6 microns and about 12 microns, a CE Dn(50) between about 2.0 microns and about 6.0 microns, and a CE Dn(10) between about 0.5 microns and about 1.5 microns.
  • the fulvestrant particles have a CE Dn(90) between about 8 microns and about 1 microns, a CE Dn(50) between about 3.0 microns and about 5.0 microns, and a CE Dn(10) between about 0.8 microns and about 1.2 microns.
  • formulations of the invention can be prepared from commercially available fulvestrant having different particle size distributions, such as, for example, recrystallized, micronized fulvestrant, or a combination thereof.
  • the formulations are prepared with sterilized, commercially available fulvestrant.
  • commercially available fulvestrant is used in the formulations of the present invention without further processing for size reduction.
  • fulvestrant particles suitable for use in formulations of the invention can be prepared from commercially available fulvestrant by any suitable methods known in the art. Suitable methods include, but are not limited to, size-reduction techniques such as milling, grinding, crushing, compression, attrition, low shear mixing, high shear mixing, high pressure homogenization, lyophilization, precipitation, or combinations thereof.
  • Desired particle size distributions for fulvestrant particles can be achieved by processing steps at one or more stages of formulation preparation.
  • the desired particle size distribution can be formed by processing fulvestrant material prior to suspension in media, by techniques described more fully elsewhere herein.
  • the desired particle size distribution can be formed by processing after suspension in media, by techniques described more fully elsewhere herein, including but not limited to high shear mixing and high pressure homogenization.
  • the desired particle size distribution can be formed by a combination of the processing prior to and after suspension in media.
  • Suitable milling techniques include, but are not limited to, dry milling, wet milling, and cryogenic milling.
  • Suitable milling machines include ball mills, pebble mills, rod mills, roller mills, colloid mills, impact mills, and jet mills.
  • the particles can be reduced in size in the presence of one or more excipients or stabilizers, such as but not limited to a surfactants, polymers, electrolytes, and non-electrolytes, and mixtures thereof.
  • the particles can be contacted with one or more excipients or stabilizers after they are reduced in size.
  • the formulations can be prepared from an un-milled commercially available fulvestrant by reducing the fulvestrant particle size with high shear mixing. In yet further embodiments of the invention, the formulations can be prepared from an un-milled, commercially available fulvestrant by reducing the fulvestrant particle size with high shear mixing followed by high pressure homogenization.
  • the formulations can be prepared from commercially available micronized fulvestrant by reducing the micronized fulvestrant particle size with high shear mixing (“HSM”). In yet further embodiments of the invention, the formulations can be prepared from an un-milled commercially available fulvestrant by reducing the fulvestrant particle size with high shear mixing followed by high pressure homogenization (“HPH”).
  • HSM high shear mixing
  • HPH high pressure homogenization
  • the formulations can be prepared using high pressure homogenization.
  • the high pressure homogenization process reduces particle size by subjecting the particle population to one or more of cavitation, shear, and impact within a homogenization chamber under operating pressures from about 5,000 psi to about 45,000 psi, for example, about 5,000, 10,000, 15,000, 20,000, 25,000, 30,000, 35,000, 40,000 or about 45,000 psi.
  • the high pressure homogenization process is performed at about 40,000 psi.
  • the high pressure homogenization process is performed at operating pressures from about 15,000 psi to about 20,000 psi.
  • the formulations can be prepared by passing the formulation suspension through a homogenization chamber under operating pressures for one or more passes, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, or 50 passes.
  • Some exemplary preparation processes are identified with alphanumeric reference identifiers, such as “Process A1,” “Process A2,” and so on.
  • Some exemplary formulations can share identical concentrations of constituent components (mg/mL), but may vary in their properties due to the different preparation processes, storage, or handling, which can result in different particle size distributions due to more or less size reduction, more or less aggregation or agglomeration, or both during processing, storage, or handling.
  • methods of forming an aqueous fulvestrant suspension comprise mixing an aqueous medium and at least one stabilizer to form a suspension vehicle, adding an amount of fulvestrant to the suspension vehicle, and dispersing the fulvestrant in the suspension vehicle to form the aqueous fulvestrant suspension.
  • these methods can further comprise homogenizing the aqueous fulvestrant suspension.
  • the methods with or without the homogenizing step can further comprise concentrating the fulvestrant suspension by phase separating the suspension and removing a portion of the supernatant.
  • the methods can further comprise adding one or more electrolytes, non-electrolytes, buffering agents, or cross-linked polymers to the homogenized aqueous fulvestrant suspension and mixing the one or more electrolytes, non-electrolytes, buffering agents, or cross-linked polymers into the suspension.
  • the methods comprise a dispersing step performed using high shear mixing, a homogenizing step performed using high pressure homogenization, or both a dispersing step performed using high shear mixing and a homogenizing step performed using high pressure homogenization.
  • the one or more stabilizers may be incorporated into the formulations at one or more stages of the methods of forming the formulations.
  • at least a portion or all of the stabilizers of a formulation are added to an aqueous medium along with an amount of fulvestrant prior to some or all of any mixing, homogenization, or supernatant-removal steps.
  • at least a portion or all of the one or more stabilizers of a formulation are added to the fulvestrant suspensions after some or all of any mixing, homogenization, or supernatant-removal steps have been completed.
  • At least a portion or all of the surfactant and polymer stabilizers of the formulations are combined with the aqueous medium and fulvestrant prior to some or all of any mixing, homogenization, or supernatant-removal steps and at least a portion or all of the electrolyte, non-electrolyte, buffering agents, and cross-linked polymers of the formulations are added to the suspension after some or all of any mixing, homogenization, or supernatant-removal steps.
  • the fulvestrant particles described herein can be prepared in a method comprising the steps of dispersing fulvestrant particles in a liquid suspension medium and applying mechanical means in the presence of grinding media to reduce the particle size of fulvestrant to the desired size.
  • a solvent, such as water, present in a formulation can be removed by appropriate techniques known to the art, such as lyophilization or spray drying, to form a dried formulation suitable for later reconstitution.
  • Lyophilization can be used to produce a lyophilized (lyo) cake.
  • the dried formulation can be reconstituted back into a liquid suspension using an appropriate diluent. Different volumes of diluent can be used to produce reconstituted suspensions with different fulvestrant concentrations as needed.
  • the diluent can be aqueous in general but can further comprise an organic solvent and/or any excipient as described elsewhere herein.
  • suspensions can be prepared with higher or lower concentrations of constituent components than desired in formulations for administration, formed into dried formulations and placed into vials in appropriate amounts of dried formulation to achieve target dose amounts of fulvestrant per vial for later reconstitution of diluent to form the desired formulation for administration.
  • FIG. 15 Some exemplary methods of preparation of dried pharmaceutical compositions are depicted schematically in FIG. 15 .
  • the pharmaceutical compositions and dried pharmaceutical compositions can be prepared using aseptic process or terminally sterilized by a compatible sterilization technique, such as, but not limited to, gamma irradiation.
  • a compatible sterilization technique such as, but not limited to, gamma irradiation.
  • said polymer such as microcrystalline cellulose (CMC) or its salts including sodium CMC, can be sterilized by autoclave in a solution then combined with rest of the pharmaceutical composition that is prepared aseptically or terminally sterilized.
  • the pharmaceutical compositions are bioequivalent to the commercial pharmaceutical composition, FASLODEXTM.
  • the single dose PK parameters in postmenopausal advanced breast cancer patients administered FASLODEXTM dosed intramuscularly with 500 mg with an additional dose at day 15 are reported as, in geometric mean and coefficient of variation (%), Cmax 25.1 (35.3) ng/mL, Cmin 16.3 (25.9) ng/mL, and AUC 11,400 (33.4) ng ⁇ hr/mL.
  • the 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) of the pharmaceutical composition of the invention is within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of FASLODEXTM.
  • the 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) of the pharmaceutical composition of the invention is within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of FASLODEXTM in the fasting state.
  • the 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) of the pharmaceutical composition of the invention is within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of FASLODEXTM in the fed state.
  • the 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) of the pharmaceutical composition of the invention having a fulvestrant concentration of 100 mg/mL is within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of FASLODEXTM.
  • the 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) of the pharmaceutical composition of the invention having a fulvestrant concentration of 100 mg/mL is within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of FASLODEXTM in the fasting state.
  • the 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) of the pharmaceutical composition of the invention having a fulvestrant concentration of 100 mg/mL is within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of FASLODEXTM in the fed state.
  • the pharmaceutical composition has the single dose and multiple dose pharmacokinetic parameters shown in Tables 28 and 29.
  • Table 28 shows pharmacokinetic parameters for 500 mg dosage of pharmaceutical compositions of the disclosure.
  • the fulvestrant blood plasma concentration data are shown for a 500 mg initial dose with an additional 500 mg dose given on day 15.
  • the fulvestrant blood plasma concentration data are shown for measurement at month 3, after a 500 mg dosage on days 1, 15, 20, and once monthly thereafter.
  • Table 29 shows pharmacokinetic parameters for a single 250 mg dosage of pharmaceutical compositions of the disclosure. In Table 29, data are expressed as geometric mean (CV %), except for T max , which is shown as a median value with a range indicated in parentheses.
  • a dose of about 500 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEXTM.
  • a dose of less than 500 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEXTM.
  • a dose of about 400 to 450 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEXTM.
  • a dose of about 350 to 400 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEXTM.
  • a dose of about 300 to 350 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEXTM.
  • a dose of about 250 to 300 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEXTM.
  • a 500 mg dose of a pharmaceutical composition of the invention provides 90% confidence intervals (CI) of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of a 500 mg dose of FASLODEXTM.
  • a dose of less than 500 mg of a pharmaceutical composition of the invention provides 90% confidence intervals (CI) of the relative mean C ma , AUC (0-t) and AUC (0- ⁇ ) within 80% to 125% of the relative mean C max , AUC (0-t) and AUC (0- ⁇ ) , respectively, of a 500 mg dose of FASLODEXTM.
  • fulvestrant pharmaceutical compositions of the invention can be administered as a single intramuscular injection, with the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0- ⁇ ) of fulvestrant is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0- ⁇ ), respectively, of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEXTM administered intramuscularly as two 5 mL injections.
  • such fulvestrant pharmaceutical compositions administered as a single intramuscular injection comprise a dose of about 500 mg of fulvestrant.
  • such fulvestrant pharmaceutical compositions administered as a single intramuscular injection comprise a dose of about 500 mg of fulvestrant in an injection volume of about 3.0 mL to about 5.0 mL, about 3.5 mL to about 4.5 mL, or about 4.0 mL.
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80% of the relative mean Cmax of FASLODEXTM.
  • Such embodiments may provide benefits by providing a therapeutically effect amount of fulvestrant exposure to a subject while reducing the degree of one or more Cmax-driven side-effects or toxicities in comparison to the degree of side-effects or toxicities experienced by a subject from receiving a therapeutically effective amount of fulvestrant exposure from one or more dosages of FASLODEXTM.
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, or less than 40% of the relative mean Cmax of FASLODEXTM.
  • such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM
  • the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, or less than 40% of the relative mean Cmax of FASLODEXTM in the fasting state.
  • such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, or less than 40% of the relative mean Cmax of FASLODEXTM in the fed state.
  • such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM
  • the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, within about 45% to about 55%, within about 55% to about 65%, within about 65% to about 75%, within about 50% to about 60%, within about 60% to about 70%, or within about 70% to about 80% of the relative mean Cmax of FASLODEXTM.
  • such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM
  • the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, within about 45% to about 55%, within about 55% to about 65%, within about 65% to about 75%, within about 50% to about 60%, within about 60% to about 70%, or within about 70% to about 80% of the relative mean Cmax of FASLODEXTM in the fasting state.
  • such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100
  • the 90% confidence intervals (CI) of the relative mean AUC (0-t) , relative mean AUC (0- ⁇ ) , or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC (0-t) and relative mean AUC (0- ⁇ ) , respectively, of FASLODEXTM
  • the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, within about 45% to about 55%, within about 55% to about 65%, within about 65% to about 75%, within about 50% to about 60%, within about 60% to about 70%, or within about 70% to about 80% of the relative mean Cmax of FASLODEXTM in the fed state.
  • such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100
  • the invention is directed to methods of treatment comprising administration of a pharmaceutically effective amount of any of the fulvestrant pharmaceutical compositions described herein to a patient in need thereof.
  • the invention is directed to a method of treating breast cancer, comprising administering a pharmaceutically acceptable amount of any of the fulvestrant pharmaceutical compositions described herein.
  • the breast cancer is metastatic breast cancer.
  • the breast cancer is hormone receptor (HR)-positive breast cancer.
  • the invention is directed to a method of treating hormone receptor (HR)-positive breast cancer in a post-menopausal woman comprising administration of a pharmaceutically effective amount of any of the fulvestrant pharmaceutical compositions described herein.
  • the invention is directed to a method of treating hormone receptor (HR)-positive breast cancer in a post-menopausal woman with disease progression following antiestrogen therapy comprising administration of a pharmaceutically effective amount of any of the fulvestrant pharmaceutical compositions described herein.
  • the invention is directed to a method of treating HR-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced or metastatic breast cancer in a woman with disease progression after endocrine therapy.
  • HR hormone receptor
  • HER2 human epidermal growth factor receptor 2
  • a fulvestrant pharmaceutical composition as described herein is administered on days 1, 15, 29, and once monthly thereafter.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered on days 1, 15, 29, and once monthly thereafter.
  • a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered on days 1, 15, 29, and once monthly thereafter.
  • a fulvestrant pharmaceutical composition as described herein is administered as a single injection.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single injection.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 5 mL injection.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 4 mL injection.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 3 mL injection.
  • a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single injection. In further embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 2.5 mL injection. In yet further embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 5 mL injection.
  • a fulvestrant pharmaceutical composition as described herein is administered as two injections.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two injections.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two 5 mL injections.
  • a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two 2 mL injections, two 2.5 mL injections, two 3 mL injections, two 3.5 mL injections, or two 4 mL injections.
  • a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two injections. In yet other embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two 2.5 mL injections.
  • fulvestrant pharmaceutical compositions described herein may be administered alone, or in combination with one or more additional therapeutic agents as defined herein.
  • An additional therapeutic agent may be used to treat one or more core symptoms and/or comorbidities associated with cancer in general or breast cancer in particular.
  • fulvestrant is formulated (and administered) with at least one therapeutic agent as a fixed dose.
  • fulvestrant is formulated (and administered) separately from the therapeutic agent(s).
  • a EGFR kinase inhibitor e.g., a EGFR kinase inhibitor, a PDGFR kinase inhibitor, a FGFR kinase inhibitor, or any of the other cytotoxic, chemotherapeutic, antihormonal, anti-angiogenic, antiproliferative, pro-apoptotic, anti-HER2, radiation or a radiopharmaceutical, signal transduction inhibitors, or other anti-cancer agents or treatments
  • agents that can be used in combination with the fulvestrant pharmaceutical compositions of the disclosure include palbociclib, letrozole, anastrozole, doxorubicin, paclitaxel, docetaxel, vinorelbine, and 5-fluorouracil.
  • therapeutic agents that may be used in combination with fulvestrant include, but are not limited to, agents or treatments for one or more of pain, nausea, emesis, hot flushes, constipation, and dizziness.
  • Some exemplary fulvestrant pharmaceutical compositions were prepared with 50 mg/mL and 100 mg/mL concentrations of fulvestrant in aqueous suspensions.
  • Tables 1-20 show aspects of the pharmaceutical compositions of the pharmaceutical compositions and the methods of preparation of some of the pharmaceutical compositions, also referred to as formulations, variants, or Lots in the Tables.
  • Tables 4-7 and 23-27 and FIGS. 4-12 and 15 show aspects of the methods of preparation used to prepare some of the pharmaceutical compositions.
  • the formulations B, E, I, J, K, L, L3F, L6, F003a, F003b, F003e, F004a, F003k2, F003k3, F005a2, F003l, F005b1, F015a1, F015a3, F005d1, F005c3, F005g5 tested in Studies 1-3 below were prepared via one or more of process steps of (1) low shear mixing, indicated in the Tables 23-27 and FIGS.
  • phase separation was performed by phase separating the pharmaceutical composition and withdrawing the desired amount of supernatant to concentrate the suspensions to the target concentrations of fulvestrant, either 50 mg/mL or 100 mg/mL, depending on the pharmaceutical composition.
  • phase separation was performed by overnight settling in a clear glass centrifuge tube. Application of centrifuge for phase separation could also be utilized.
  • Fulvestrant active pharmaceutical ingredient (which may also be referred to as “API” herein and in the Tables and Figures) was obtained in un-milled forms or in milled, micronized, or recrystallized forms from commercial suppliers.
  • As-obtained particle size distributions for un-milled API varied from an LD Dv(90) of about 240 microns to an LD Dv(90) of about 2130 microns.
  • As-obtained particle size distributions for milled, micronized, and recrystallized API varied from an LD Dv(90) of about 7 microns to an LD Dv(90) of about 18 microns.
  • Fulvestrant API may be obtained in various particle size distributions from commercial sources and processed as described elsewhere herein to achieve the desired particle size distributions. Particle size distributions can be monitored throughout the processing steps through analysis of samples as described elsewhere herein.
  • the formulations B, E, I, J, K, L, L3F, L6, F003a, F003b, F003e, F004a, F003k2, F003k3, F005a2, F003l, F005b1, F015a1, F015a3, F005d1, F005c3, F005g5 tested in Studies 1-3 below were prepared via high shear mixing (HSM) steps.
  • the preparation of formulations can be performed with an IKA T10 Basic Disperser with an IKA S10N-10G dispersing tool. At the speeds indicated ( ⁇ 20,000 to 30,000 rpm), the mixture of fulvestrant and suspension vehicle was processed in cycles until the total processing time indicated was reached.
  • a formulation was vortexed at ⁇ 3000 rpm for 30 seconds then sonicated for 1 minute to remove or reduce foam generated during the high shear mixing by the disperser.
  • Formulations were also rested as needed in between cycles at room temperature to allow the disperser to cool down and avoid overheating of the product and the equipment. Sonication was performed with a Branson 3800 Ultrasonic Bath (Branson Ultrasonics Corp., Danbury, Conn.) at a frequency of 40 kHz. Other mixing and ultrasonic apparatuses may also be used to achieve mixing and particle size distribution as desired.
  • high pressure homogenization was performed.
  • high pressure homogenization (HPH) steps were performed with a Nano DeBEE High Pressure Homogenizer (BEE International, South Easton, Mass.) in a Labconco XPert Filtered Balance System (Model 3950630) (Labconco, Kans. City, Mo.), installed the 100 ml sample holder and Z5 nozzle in parallel flow configuration on Nano DeBEE High Pressure Homogenizer.
  • the homogenizer was primed with filing water until the process pressure reached the processing pressure as indicated in the Tables and Figures. Water was removed from the system using the plunger to minimize the dilution of the batch by the residual priming water.
  • references to “Assay” refers to high-performance liquid chromatography (HPLC) measurement of the fulvestrant concentration of the pharmaceutical composition at intermediate processing steps or in final result as prepared.
  • HPLC high-performance liquid chromatography
  • the “Assay” results are given in absolute measured mg/mL or as a percentage (%) or (% LC), where percentages indicate the concentration of fulvestrant relative to the 50 mg/mL label claim of the commercially available FASLODEXTM product. Total impurities were also measured and are provided in the figures as a percentage by area (% a/a) where indicated.
  • HPLC was performed with Agilent Technologies Agilent 1260 Infinity Quaternary LC module G1311B (Agilent Technologies, Santa Clara, Calif.). Other HPLC apparatuses may also be used to analyze the fulvestrant concentrations.
  • particle size and particle size distributions were analyzed with Malvern Mastersizer 3000 (Malvern Instruments Ltd., Malvern, Worcestershire, UK), with an attached sample dispersion unit with an in-line sonication probe for agglomerate dispersion prior to analysis via laser diffraction.
  • Malvern Mastersizer 3000 Malvern Instruments Ltd., Malvern, Worcestershire, UK
  • particle size and particle size distributions were analyzed with Malvern Morphologi G3 (Malvern Instruments Ltd., Malvern, Worcestershire, UK), to determine circle equivalent (CE) diameters via microscopy image capture and analysis.
  • Malvern Morphologi G3 Malvern Instruments Ltd., Malvern, Worcestershire, UK
  • Fulvestrant pharmaceutical compositions B, E, I, J, K, and L were prepared as described elsewhere herein and in the Figures.
  • a preclinical study was performed to determine the pharmacokinetics of the pharmaceutical compositions following a single intramuscular administration of 15.4 mg/kg to female dogs.
  • the pharmacokinetics of 15.4 mg/kg IM FASLODEXTM fulvestrant injection, 250 mg/5 mL were also determined and used for comparison to the three prototype pharmaceutical compositions.
  • the 15.4 mg/kg dose used in this study is the canine equivalent, in mg/m 2 , of the maximum dose (500 mg) for human use and was scaled for use in canine by dividing the dose (based on a 60 kg human) by a canine species conversion factor of 0.54.
  • the FASLODEXTM test articles contained a small molecule that was used as received and no adjustment was made for purity, salt correction, etc.
  • the FASLODEXTM test articles were gently agitated prior to dispensing and dose delivery.
  • Pharmaceutical Composition B, Pharmaceutical Composition E, Pharmaceutical Composition I, Pharmaceutical Composition J, Pharmaceutical Composition K, and Pharmaceutical Composition L were stored at room temperature and protected from light prior to use, and gently agitated prior to dispensing and dose delivery.
  • IM doses were administered with a 20 G needle via bolus injection into the same large muscle mass (using the Z-track injection technique) in the left hind limb of each animal. Attempts were made for consistent injections between animals [selection of the dose site (muscle), depth, etc.]. The hair was clipped from the injection site prior to dosing. The injection site was marked following dosing and remarked as necessary throughout the study.
  • Blood samples were collected at various time intervals to measure the blood plasma concentration of fulvestrant. Blood samples for Groups 1-3 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 456 (on Day 20), 528 (on Day 23), 600 (on Day 26), and 672 (on Day 29) hours postdose.
  • Blood samples for Groups 4-8 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 72 (on Day 4), 96 (on Day 5), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 528 (on Day 23), and 672 (on Day 29) hours postdose.
  • Plasma samples were initially placed on dry ice prior to being stored in the appropriate freezer ( ⁇ 60 to ⁇ 90° C.). Samples were shipped on dry ice for bioanalytical analysis.
  • Table 31 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition).
  • An “Assay %” is shown for the fulvestrant pharmaceutical compositions of the present disclosure used in the study.
  • the “Assay %” represents the percentage equivalence of the particular pharmaceutical composition in comparison to the FASLODEXTM label claim fulvestrant concentration, with “Assay %” values determined via HPLC, measurement samples taken pre- and post-dose, with one value selected for normalization.
  • Tables 32-37 are normalized using the “Assay %” values to compare PK results based upon the actual mg/kg of fulvestrant administered, assuming linear scaling.
  • FIGS. 1B, 2A, 2C, and 3 depict graphs of the dose normalized fulvestrant mean plasma concentrations.
  • Fulvestrant pharmaceutical compositions L3F and L6 were prepared as described elsewhere herein and in the Figures.
  • a preclinical study was performed to determine the pharmacokinetics of the pharmaceutical compositions following a single intramuscular administration of 15.4 mg/kg to female dogs.
  • the pharmacokinetics of 15.4 mg/kg IM FASLODEXTM fulvestrant injection, 250 mg/5 mL were also determined and used for comparison to the three prototype pharmaceutical compositions.
  • the 15.4 mg/kg dose used in this study is the canine equivalent, in mg/m 2 , of the maximum dose (500 mg) for human use and was scaled for use in canine by dividing the dose (based on a 60 kg human) by a canine species conversion factor of 0.54.
  • the FASLODEXTM test articles contained a small molecule that was used as received and no adjustment was made for purity, salt correction, etc.
  • the FASLODEXTM test articles were gently agitated prior to dispensing and dose delivery.
  • Pharmaceutical Compositions L3F and L6 were stored at room temperature and protected from light prior to use, and gently agitated prior to dispensing and dose delivery.
  • Dose Volume Dose Group Test Article (mg/kg) (mg/mL) (mg/kg) 1 FASLODEX TM 15.4 50 0.308 2 Pharmaceutical 15.4 100 0.154 Composition L3F 3 Pharmaceutical 15.4 100 0.154 Composition L6
  • IM doses were administered with a 20 G needle via bolus injection into the same large muscle mass (using the Z-track injection technique) in the left hind limb of each animal. Attempts were made for consistent injections between animals [selection of the dose site (muscle), depth, etc.]. The hair was clipped from the injection site prior to dosing. The injection site was marked following dosing and remarked as necessary throughout the study.
  • Plasma (minimum of 0.8 mL volume) was separated from blood cells within 0.75 hours of blood collection and frozen. Plasma samples were initially placed on dry ice prior to being stored in the appropriate freezer ( ⁇ 60 to ⁇ 90° C.). Samples were shipped on dry ice for bioanalytical analysis.
  • Table 31 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition).
  • An “Assay %” is shown for the fulvestrant pharmaceutical compositions of the present disclosure used in the study.
  • the “Assay %” represents the percentage equivalence of the particular pharmaceutical composition in comparison to the FASLODEXTM label claim fulvestrant concentration, with “Assay %” values determined via HPLC, measurement samples taken pre- and post-dose, with one value selected for normalization.
  • Tables 32-37 are normalized using the “Assay %” values to compare PK results based upon the actual mg/kg of fulvestrant administered, assuming linear scaling.
  • FIGS. 2A and 2C depict graphs of the dose normalized fulvestrant mean plasma concentrations.
  • Fulvestrant formulations F003a, F003b, F004a, F003e, F003k2, F003k3, F005a2, F0031, F005b1, F015a1, F005d1, F005c3, F015a3, F005g5, Del-1S, Del-2S, F005H3, Lot 15, Lot 26, Lot 27, Lot 28, Lot 42, Lot 43, Lot 45, Lot 46, Lot 47, and Lot 48 were prepared as described elsewhere herein and in the Figures. In some instances, the formulations were prepared using different processes as indicated, referred to by an alphanumeric process identifier, such as “Process A1,” “Process A2,” and the like.
  • a fulvestrant pharmaceutical composition for intravenous injection referred to as batch FV-004/15M, was prepared as described below.
  • a preclinical study was performed to determine the pharmacokinetics of the pharmaceutical compositions following a single intramuscular (IM) administration of 15.4 mg/kg to female dogs.
  • the pharmacokinetics of 15.4 mg/kg IM FASLODEXTM (fulvestrant injection, 250 mg/5 mL) were also determined and used for comparison to the three prototype pharmaceutical compositions.
  • the 15.4 mg/kg dose used in this study is the canine equivalent, in mg/m 2 , of the maximum dose (500 mg) for human use and was scaled for use in canine by dividing the dose (based on a 60 kg human) by a canine species conversion factor of 0.54.
  • compositions F003a, F003b, F004a, F003e, F003k2, F003k3, F005a2, F0031, F005b1, F015a1, F005d1, F005c3, F015a3, FV-004/15M, Del-1S, and Del-2S were stored at room temperature and protected from light prior to use, and gently agitated via inversion prior to dispensing and dose delivery. If visible clumps of material were seen on the vial inside wall or inner seal of the vial cap after 3 minutes of inversion, the tightly capped vial was vortexed at moderate intensity and unlimited duration until clumps were not visible.
  • FASLODEXTM was supplied as two 5-mL clear neutral glass (Type 1) syringe barrels, each containing a 250 mg/5 mL (50 mg/mL) solution for intramuscular injection. Upon receipt, FASLODEXTM was stored refrigerated (2°-8° C.) and protected from light. The procedure to prepare and administer FASLODEXTM was performed as outlined in the manufacturer's prescribing information.
  • Lyophilized pharmaceutical compositions of formulations F005g5, F005H3, F015a3, Lot 15, Lot 26, Lot 27, Lot 28, Lot 42, Lot 43, Lot 45, Lot 46, Lot 47, and Lot 48 were reconstituted prior to dosing. Using an empty syringe and hypodermic needle, about 5 mL of air was withdrawn from the head space of the vial (above the lyophilized contents) via the septum and the syringe and needle were discarded.
  • a sterile syringe and hypodermic needle 5 mL of sterile water for injection, USP were added to the vial by piercing the septum and injecting a stream of water slowly around the inner wall of the neck of the vial to wet the lyophilized cake without touching any of the vial contents.
  • the needle was removed from the septum and the vial was gently swirled until a visually homogeneous particulate suspension formed, with no visual clumps or material attached to the inside wall of the vial. If a homogeneous suspension was not formed after 5 minutes of swirling, the vial was vortexed until a homogeneous suspension was formed. The vials were not shaken to avoid generating bubbles or excessive foam.
  • Group 18 was administered an intravenous batch of fulvestrant (batch FV-004/15M) prepared as follows by (% w/v): 2% fulvestrant, 10% EtOH, 79% propylene glycol, 1% Poloxamer 407, 8% Water for Injection, USP.
  • Fulvestrant API was stored at 2-8° C., protected from light. Care was taken to protect the API from humidity during weighing. Fulvestrant powder was dissolved in ethanol and swirled and vortexed as needed to dissolve completely. Propylene glycol was added and mixed to dissolve to a clear liquid state.
  • Poloxamer 407 was dissolved in water for injection, USP in a separate vessel and mixed, vortexed, and sonicated as needed to dissolve into to a clear liquid state.
  • the Poloxamer 407 in water for injection solution was added to the fulvestrant/ethanol solution.
  • Propylene glycol was added and the solution was mixed and vortexed to achieve a clear liquid.
  • the solution was filtered through a 0.2 ⁇ m or 0.22 ⁇ m syringe (to ensure that all liquid volume was usable) tip filter (PVDF) into a clear glass vessel.
  • the prepared formulation as stored at room temperature for up to four hours prior to dosing under protection from light exposure. Intravenous doses were administered via the cephalic (or other suitable) vein as a slow injection over approximately 1 minute. Batch FV-004/15M was administered intravenously at a dose of 2.5 mg/kg.
  • IM doses were administered with a 20 G needle via bolus injection into the same large muscle mass (using the Z-track injection technique) in the left hind limb of each animal. Attempts were made for consistent injections between animals [selection of the dose site (muscle), depth, etc.]. The hair was clipped from the injection site prior to dosing. The injection site was marked following dosing and remarked as necessary throughout the study.
  • Blood samples for Groups 1-7 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 72 (on Day 4), 96 (on Day 5), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 528 (on Day 23), and 672 (on Day 29) hours postdose.
  • Blood samples for Groups 8-14 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 72 (on Day 4), 96 (on Day 5), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 456 (on Day 20), 528 (on Day 23), 600 (on Day 26), 672 (on Day 29), 696 (Day 30), 768 (Day 33), 816 (Day 35), 864 (Day 37), 936 (Day 40) and 1008 (Day 43) hours post-dose.
  • Blood samples for Groups 15-17 and 19-21 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, 12 (Day 1), 24 (Day 2), 48 (Day 3), 72 (Day 4), 96 (Day 5), 120 (Day 6), 192 (Day 9), 264 (Day 12), 336 (Day 15), 384 (Day 17), 456 (Day 20), 528 (Day 23), 600 (Day 26), 672 (Day 29), 696 (Day 30), 768 (Day 33), 816 (Day 35), 864 (Day 37), 936 (Day 40), 1008 (Day 43), and 1176 (Day 50) hours postdose.
  • Blood samples for Group 18 were collected predose and at 0.033 (2 minutes), 0.1 (6 minutes), 0.13 (8 minutes), 0.27 (16 minutes), 0.52 (31 minutes), 0.77 (46 minutes), 1, 2, 3, 4, 6, 8, 10, and 12 hours postdose on Day 1 and at 24 (on Day 2), 30 (on Day 2), 48 (on Day 3) and 72 (on Day 4) hours postdose, with postdose measurements from the start of dose administration, which took about 1 minute to complete.
  • Blood samples for Groups 22-48 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, 12 (Day 1), 24 (Day 2), 48 (Day 3), 72 (Day 4), 96 (Day 5), 120 (Day 6), 144 (Day 7), 168 (Day 8), 192 (Day 9), 216 (Day 10), 240 (Day 11), 264 (Day 12), 336 (Day 15), 384 (Day 17), 456 (Day 20), 528 (Day 23), 600 (Day 26), 672 (Day 29), 696 (Day 30), 768 (Day 33), 816 (Day 35), 864 (Day 37), 936 (Day 40), 1008 (Day 43), and 1176 (Day 50) hours postdose.
  • Table 31 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition).
  • An “Assay %” is shown in Table 31 for the fulvestrant pharmaceutical compositions of the present disclosure used in the study.
  • the “Assay %” represents the percentage equivalence of the particular pharmaceutical composition in comparison to the FASLODEXTM label claim fulvestrant concentration, with “Assay %” values determined via HPLC, measurement samples taken pre- and post-dose, with one value selected for normalization.
  • the data in Tables 32-37 are normalized using the “Assay %” values to compare PK results based upon the actual mg/kg of fulvestrant administered, assuming linear scaling.
  • FIGS. 1A, 1B, 2A, 2B, and 2C depict graphs of the dose normalized fulvestrant mean plasma concentrations.
  • FIGS. 13 and 14 depict fulvestrant plasma measurements for administration of Faslodex Lot MB948 to three female dogs and administration of fulvestrant formulation Lot 27 processed by Process A2 to three female dogs (referred to in FIG. 14 as subjects 924, 925, and 926).
  • fulvestrant pharmaceutical compositions of the present disclosure were examined via optical and scanning electron microscopy.
  • Suspensions of fulvestrant pharmaceutical compositions Variants B, E, I, J, K, L, L3F, L6, F003a, F003b, F004a, F003e, F00k2, and F003k3 were examined via optical microscopy.
  • Optical microscopy was performed at 400 ⁇ magnification with a polarized light filter using fully dispersed homogeneous suspension samples.
  • Example 1 Sample 1
  • Sample 2 test samples
  • CE diameters were measured and number-weighted and volume-weighted particle size distribution parameters were determined as shown in Tables 41 and 42.
  • CE diameter ranges of measurement aliquots are also shown, with the lower range value of 0.54 microns representing the lower limit of detection for the apparatus setup.
  • Test samples were analyzed with a Malvern Mastersizer 3000 apparatus for laser diffraction particle size characterization of LD diameters.
  • Fulvestrant pharmaceutical compositions F005g5, Lot 27, Lot 28, and Lot 45, described elsewhere herein, were prepared by the methods of preparation shown schematically as process 101 of FIG. 15 .
  • Samples of each Lot were prepared using different processes as indicated in Tables 43 to 50 to achieve fulvestrant particle sizes and particle size distributions.
  • the preparation processes are referred to with alphanumeric identifiers, such as “Process A1,” “Process A2,” and the like, with each process representing a set of fulvestrant-particle-size-reduction steps, as more fully described elsewhere herein, to achieve final fulvestrant particle size and particle size distributions as shown in the Tables 43 to 50.
  • Test samples were evaluated for particle size both prior to drying via lyophilization, which are indicated as “(100 ⁇ Suspensions)”, and after lyophilization and reconstitution, which are indicated as “(100 ⁇ Reconstituted Suspensions)” in the Tables 43 to 50.
  • Samples were analyzed with a Malvern Morphologi G3 apparatus for microscopy image capture and analysis. CE diameters were measured and volume-weighted particle size distribution parameters were determined. The CE diameter ranges of measurement samples are also shown, with the lower range value of 0.54 microns representing the lower limit of detection for the apparatus setup. Samples were analyzed with a Malvern Mastersizer 3000 apparatus for laser diffraction particle size characterization of LD diameters.
  • ranges are used herein for chemical or physical properties, such as particle size or particle size distribution, formulation component concentrations, or pharmacokinetic properties, all combinations, and subcombinations of ranges for specific embodiments therein are intended to be included.

Abstract

The disclosure is directed to fulvestrant formulations including suspensions of fulvestrant particles suitable for injection. The formulations can comprise fulvestrant particles having an LD Dv(10) less than about 3 microns, for example, between about 1 micron to about 3 microns, an LD Dv(50) less than about 35 microns, for example, between about 2 microns and about 35 microns, and an LD Dv(90) less than about 120 microns, for example, between about 4 microns and about 120 microns. The formulations can comprise fulvestrant particles having a CE Dv(90) less than about 200 microns, for example, between about 10 microns and about 200 microns, a CE Dv(50) less than about 60 microns, for example, between about 5 microns and about 60 microns, and a CE Dv(10) less than about 25 microns, for example, between about 1 micron and about 25 microns.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 62/332,842, filed May 6, 2016, and U.S. Provisional Application No. 62/420,555, filed Nov. 10, 2016, the entireties of which are incorporated by reference herein.
  • FIELD
  • The disclosure is directed to fulvestrant-containing formulations and methods of their use in the treatment of disease.
  • BACKGROUND
  • Fulvestrant, or 7-(9-(4,4,5,5,5-pentafluoropentylsulfinyl)nonyl)estra-1,3,5(10)-triene-3,17-diol, has the structure of formula (1):
  • Figure US20190134059A1-20190509-C00001
  • Fulvestrant is a selective estrogen receptor degrader (SERD) indicated for the treatment of hormone receptor positive metastatic breast cancer in postmenopausal women with disease progression following anti-estrogen therapy.
  • As with other steroidal-like compounds, fulvestrant has physical properties which make preparing fulvestrant pharmaceutical compositions difficult. Fulvestrant is a particularly lipophilic molecule, even when compared with other steroidal compounds, and its aqueous solubility is extremely low.
  • Due to the poor solubility and oral bioavailability of fulvestrant, the drug is currently administered via intramuscular injection of an oil-based fulvestrant formulation. The current commercial formulation of fulvestrant, FASLODEX™, is dosed at 500 mg and requires that two 5 mL injections of a 50 mg/mL fulvestrant formulation be administered intramuscularly. Each 5 mL injection contains 10% w/v alcohol, 10% w/v benzyl alcohol, and 15% w/v benzyl benzoate as co-solvents and made up to 100% w/v with castor oil as a further co-solvent and release rate modifier. Administration of the formulation is slow (1-2 minutes per injection) and painful, due to the viscous oil-based vehicle used to solubilize fulvestrant. A warning has been added to the FASLODEX™ label concerning painful injections, sciatica, neuropathic pain, and peripheral neuropathy.
  • It has been previously reported (U.S. Pat. No. 6,774,122 to AstraZeneca) that intra-muscular injections of fulvestrant in the form of an aqueous suspension were not suitable for use. Those suspensions resulted in extensive local tissue irritation at the injection site as well as a poor release profile due to the presence of fulvestrant in the form of solid particles. Furthermore, the fulvestrant release rate was reported as not clinically significant.
  • There is a need for fulvestrant formulations with improved dosing properties. The disclosure is directed to these and other important needs.
  • SUMMARY
  • The present disclosure provides formulations comprising fulvestrant particles. The disclosure also provides fulvestrant suspensions, preferably those having a fulvestrant concentration of equal to or greater than about 50 mg/mL. The disclosure also provides formulations comprising fulvestrant particles and a non-oil vehicle. Some aspects of the disclosure are directed to pharmaceutical compositions comprising fulvestrant particles having an LD Dv(90) greater than or equal to about 7 microns. Further aspects of the disclosure are directed to pharmaceutical compositions comprising fulvestrant particles having a CE Dv(90) less than about 200 microns, for example, between about 10 microns and about 200 microns, a CE Dv(50) less than about 60 microns, for example, between about 5 microns and about 60 microns, and a CE Dv(10) less than 25 microns, for example, between about 1 microns and about 25 microns. Other aspects of the disclosure are directed to pharmaceutical compositions comprising fulvestrant at a concentration of about 100 mg/mL, whereupon administration to a subject, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of the pharmaceutical compositions of the disclosure is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of a reference listed fulvestrant product. Other aspects of the disclosure are directed to fulvestrant formulations having a concentration of about 100 mg/mL and particular pharmacokinetic profiles. In other aspects, the disclosure is directed to pharmaceutical compositions comprising fulvestrant particles, wherein the fulvestrant concentration is about 40 to 125 mg/mL.
  • Methods of making and using the products described herein are also described.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The summary, as well as the following detailed description, is further understood when read in conjunction with the appended drawings. For the purpose of illustrating the disclosure, there are shown in the drawings exemplary embodiments of the disclosure; however, the disclosure is not limited to the specific methods, compositions, and devices disclosed. In the drawings:
  • FIG. 1A depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 1B depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 2A depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 2B depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 2C depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 3 depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 4 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 5 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 6 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 7 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 8 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 9 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 10 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 11 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 12 depicts aspects of exemplary methods of preparation for fulvestrant suspensions of the present disclosure;
  • FIG. 13 depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines;
  • FIG. 14 depicts pharmacokinetic data for administration of commercial fulvestrant formulations (FASLODEX™) and some exemplary fulvestrant formulations of the present disclosure to canines; and
  • FIG. 15 depicts schematic representations of aspects of methods of preparation of some exemplary fulvestrant formulations of the present disclosure.
  • All callouts and annotations in the Figures are hereby incorporated into this description as if fully set forth herein.
  • DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
  • The present disclosure may be understood more readily by reference to the following detailed description taken in connection with the accompanying figures and examples, which form a part of this disclosure. It is to be understood that this disclosure is not limited to the specific devices, methods, applications, conditions or parameters described and/or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting of the claimed disclosure.
  • As used in the specification including the appended claims, the singular forms “a,” “an,” and “the” include the plural, and reference to a particular numerical value includes at least that particular value, unless the context clearly dictates otherwise.
  • When a range of values is expressed, another embodiment includes from the one particular value and/or to the other particular value. All ranges are inclusive and combinable. Further, reference to values stated in ranges include each and every value within that range. When values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. The term “about” as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass reasonable variations of the value, such as, for example, ±10% from the specified value. For example, the phrase “about 50%” can include ±10% of 50, or from 45% to 55%.
  • It is to be appreciated that certain features of the disclosure which are, for clarity, described herein in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the disclosure that are, for brevity, described in the context of a single embodiment, may also be provided separately or in any subcombination.
  • Terms
  • As used herein, whether by itself or in conjunction with another term or terms, it should be understood that the phrases “method of treating” and “method of treatment” may be used interchangeably with the phrase “for use in the treatment of” a particular disease.
  • As used herein, whether by itself or in conjunction with another term or terms, “pharmaceutically acceptable” indicates that the designated entity such as, for example, e.g., a pharmaceutically acceptable excipient is generally chemically and/or physically compatible with other ingredients in a formulation, and/or is generally physiologically compatible with the recipient thereof.
  • As used herein, “pharmaceutical composition” refers to a formulation as described herein that includes one or more pharmaceutically acceptable excipients, that is suitable for administration to a subject. It should be understood that the term “pharmaceutical composition” encompasses (a) suspensions and (b) suspensions which have been dried such that one or more solvents have been removed partially or completely, either by evaporation or sublimation, including, but not limited to, lyophilized cakes.
  • As used herein, whether by themselves or in conjunction with another term or terms, “subject(s),” “individual(s),” and “patient(s)”, refer to mammals, including humans. The term human(s) refers to and includes, a human child, adolescent, or adult.
  • As used herein, whether by themselves or in conjunction with another term or terms, “treats,” “treating,” “treated,” and “treatment,” refer to and include ameliorative, palliative, and/or curative uses and results, or any combination thereof. In other embodiments, the methods described herein can be used prophylactically. It should be understood that “prophylaxis” or a prophylactic use or result do not refer to nor require absolute or total prevention (i.e., a 100% preventative or protective use or result). As used herein, prophylaxis or a prophylactic use or result refer to uses and results in which administration of a compound or formulation diminishes or reduces the severity of a particular condition, symptom, disorder, or disease described herein; diminishes or reduces the likelihood of experiencing a particular condition, symptom, disorder, or disease described herein; or delays the onset or relapse (reoccurrence) of a particular condition, symptom, disorder, or disease described herein; or any combination of the foregoing.
  • As used herein, whether used alone or in conjunction with another term or terms, “therapeutic” and “therapeutically effective amount”, refer to an amount of a compound or formulation that (a) treats a particular condition, symptom, disorder, or disease described herein; (b) attenuates, ameliorates or eliminates one or more symptoms of a particular condition, disorder, or disease described herein; (c) delays the onset or relapse (reoccurrence) of a particular condition, symptom, disorder, or disease described herein. It should be understood that the terms “therapeutic” and “therapeutically effective” encompass any one of the aforementioned effects (a)-(c), either alone or in combination with any of the others (a)-(c).
  • As used herein, whether used alone or in conjunction with another term or terms, “therapeutic agent” refers to any substance included in a formulation that is useful in the treatment of a disease, condition, or disorder or comorbidity (i.e., a disease, condition, or disorder that exists simultaneously with breast cancer) and is not fulvestrant.
  • As used herein, whether used alone or in conjunction with another term or terms, “suspension” refers to solid particles dispersed in a liquid vehicle.
  • As used herein, whether used alone or in conjunction with another term or terms, “formulation” refers to a mixture of components. The term “formulation” encompasses pharmaceutical compositions, and suspensions, as well as suspensions that have been dried such that one or more solvents have been removed partially or completely (e.g., lyophilized cakes).
  • As used herein “Dv(10)”, “Dv(50)” and “Dv(90)” are defined as the volume weighted particle diameters where a cumulative 10%, 50% or 90% v/v of the particles have an equal or smaller diameter, respectively, when measured. For example, if a particle population has a Dv(50) of about 25 microns, 50% of the particles in volume have a diameter of less than or equal to about 25 microns.
  • As used herein, Dn(10)”, “Dn(50)” and “Dn(90)” are defined as the number weighted particle diameters where a cumulative 10%, 50% or 90% of the particles have an equal or smaller diameter, respectively, when measured. For example, if a particle population has a Dn(50) of about 25 microns, 50% of the particles in number have a diameter of less than or equal to about 25 microns.
  • Particle size and particle size distributions can be determined by measurement via laser diffraction. Particle size analysis by laser diffraction methods is known in the art and is explained more fully by ISO 13320:2009(E), “Particle size analysis—Laser diffraction methods,” International Organization for Standardization which is incorporated by reference herein in its entirety for all purposes. Particle sizes determined by laser diffraction are represented as the diameter of a sphere having equivalent volume to the particle volume as determined by Mie theory of light scattering. Tables 1-7 and 23-27 and FIGS. 4-12 provide laser diffraction particle size and particle size distribution (“PSD”) data for some exemplary embodiments of the present invention, with measurements taken during methods of preparation, on the day of formulation (“Day 0”), and at various later dates after formulation, as indicated. Measurements were taken “as is” and “sonicated.” Data for “sonicated” samples indicates that the measurement sample was subjected to sonication to disperse agglomerates and provide stable repeat measurements, as more fully described in ISO 13320:2009(E). Values measured via laser diffraction are indicated as such in the Figures and Tables, or are referred to herein by “laser diffraction Dv(##)”, “LD Dv(##)”, “laser diffraction diameter”, or “LD diameter.”
  • Particle size and particle size distributions can also be determined by microscopy image capture and analysis. Microscopy image capture and analysis captures a two dimensional (2D) image of a 3D particle and calculates various size and shape parameters from the 2D image. Particle sizes determined by microscopy image capture and analysis are represented as the diameter of a circle with the equivalent area as the 2D image of the particle, referred to herein as a circle equivalent or “CE” diameter. Particle size analysis by microscopy image capture and analysis is known in the art and is explained more fully by ISO 13322-1:2014, “Particle size analysis—Image analysis methods—Part 1: Static image analysis methods,” International Organization for Standardization, which is incorporated by reference herein in its entirety for all purposes. Values measured by microscopy image capture and analysis are referred to herein by “circle equivalent diameter,” “CE diameter,” “circle equivalent Dv(##),” “CE Dv(##)”, or “CE Dn(##)”. Tables 41-50 provide microscopy image capture and analysis particle size and particle size distribution data for some exemplary embodiments of the present invention, with measurement samples taken during methods of preparation, after an initial suspension is formed, or after lyophilization and reconstitution, as indicated.
  • A. Formulations. Suspensions Comprising Fulvestrant Particles and a Vehicle
  • In particular embodiments, the invention is directed to suspensions comprising fulvestrant particles and a vehicle. The fulvestrant particles may have different particle size distributions as described more fully elsewhere herein. As used herein, a “vehicle” is a suspending medium, preferably a pharmaceutically acceptable suspending medium. In certain embodiments, the vehicle is a non-oil vehicle. As used herein, “oils” are non-polar substances that have no or low miscibility with water. Castor oil is an example of an oil. In other embodiments of the invention, the vehicle comprises water, i.e., is aqueous. As used herein, an “aqueous” vehicle is a vehicle that comprises at least about 50% w/w water. In some embodiments, the aqueous vehicle comprises at least about 60% w/w, at least about 70% w/w, at least about 80% w/w, at least about 85% w/w, at least about 90% w/w, at least about 95% w/w, at least about 96% w/w, at least about 97% w/w, at least about 98% w/w, or at least about 99% w/w water. In certain embodiments of the invention, the vehicle is water. In yet other embodiments of the invention, the vehicle is a non-aqueous medium. In some embodiments, a vehicle comprises a single suspending medium. In other embodiments, a vehicle comprises a mixture of two or more suspending mediums, which may be aqueous or non-aqueous. In still other embodiments of the invention, the vehicle comprises both water and a non-aqueous solvent. In particular embodiments of the invention, the suspension is substantially oil-free. As used herein, a “substantially oil-free” suspension is a suspension comprising a vehicle that comprises at most about 10% w/w oil. In some preferred embodiments, a substantially oil-free suspension comprises a vehicle that comprises less than about 5% w/w oil, less than about 2% w/w oil, less than about 1% w/w oil, less than about 0.5% w/w oil, less than about 0.1% w/w oil, or comprises a vehicle that is free of oil.
  • Fulvestrant suspensions of the disclosure can have fulvestrant present at a concentration of about 40 mg/mL to about 125 mg/mL in a vehicle. The fulvestrant present in the fulvestrant suspensions may have different particle size distributions as described more fully elsewhere herein. In particular embodiments of the invention, fulvestrant is present at a concentration equal to or greater than about 40 mg/mL. In further embodiments, fulvestrant is present at a concentration of about 40 to about 75 mg/mL. In other embodiments, fulvestrant is present at a concentration of about 75 mg/mL to about 125 mg/mL. In still further embodiments, fulvestrant is present at a concentration of about 40 mg/mL, about 45 mg/mL, about 50 mg/mL, about 55 mg/mL, about 60 mg/mL, about 65 mg/mL, about 70 mg/mL, or about 75 mg/mL.
  • In certain embodiments, fulvestrant is present in the suspension at a concentration equal to or greater than about 75 mg/mL. In further embodiments, fulvestrant is present in the suspension at a concentration of about 75 to about 125 mg/mL. In particular embodiments, fulvestrant is present in the suspension at a concentration of about 80 mg/mL, about 85 mg/mL, about 90 mg/mL, about 95 mg/mL, about 100 mg/mL, about 105 mg/mL, about 110 mg/mL, about 115 mg/mL, about 120 mg/mL, or about 125 mg/mL. In other embodiments, fulvestrant is present in the suspension at a concentration of about 75 mg/mL to about 95 mg/mL, about 80 mg/mL to about 100 mg/mL, about 90 mg/mL to about 110 mg/ml, about 95 mg/mL to about 105 mg/mL, about 95 mg/mL to about 115 mg/mL, about 100 mg/mL to about 110 mg/mL, about 110 mg/mL to about 125 mg/mL, including all ranges and subranges there between.
  • Pharmaceutical Compositions Comprising Fulvestrant
  • Other embodiments of the disclosure include pharmaceutical compositions comprising fulvestrant. These pharmaceutical compositions may be prepared by combining fulvestrant, as described herein, with one or more additional excipients, preferably pharmaceutically acceptable excipients.
  • In certain embodiments, the pharmaceutical compositions may further comprise a stabilizer, or one or more stabilizers, or two or more stabilizers. In still further embodiments of the invention, the stabilizer is selected from the group consisting of surfactants, polymers, cross-linked polymers, buffering agents, electrolytes, and non-electrolytes. In yet further embodiments of the invention, the pharmaceutical composition comprises a combination of two or more stabilizers selected from the group consisting of surfactants, polymers, cross-linked polymers, buffering agents, electrolytes, and non-electrolytes.
  • In certain embodiments of the invention, the pharmaceutical compositions comprising fulvestrant comprise about 0.2 mg/mL to about 75 mg/mL of one or more stabilizers, and all ranges and subranges therebetween. In particular embodiments of the invention, the pharmaceutical composition comprises about 0.2 to 0.7 mg/mL, 0.5 to 1 mg/mL, 1 to 5 mg/mL, 2 to 8 mg/mL, 5 to 6 mg/mL, 5 to 10 mg/mL, 8 to 12 mg/mL, 10 to 15 mg/mL, 15 to 20 mg/mL, 20 to 30 mg/mL, 30 to 40 mg/mL, 40 to 50 mg/mL, 45 to 55 mg/mL, 50 to 60 mg/mL, or 60 to 75 mg/mL of one or more stabilizers, and all ranges and subranges there between. In further embodiments of the invention, the pharmaceutical composition comprises about 0.2 mg/mL, 0.5 mg/mL, 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, 5 mg/mL, 5.5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, 10 mg/mL, 12 mg/mL, 15 mg/mL, 17 mg/mL, 20 mg/mL, 25 mg/mL, 30 mg/mL, 35 mg/mL, 40 mg/mL, 45 mg/mL, 50 mg/mL, 55 mg/mL, 60 mg/mL, 65 mg/mL, 70 mg/mL, or about 75 mg/mL of one or more stabilizers.
  • In yet further embodiments of the invention, the stabilizer is a surfactant. For example, the stabilizer can be, but is not limited to, polyethylene oxide (PEO), a PEO derivative, polysorbate 80, polysorbate 20, poloxamer 188 (including, but not limited to, PLURONIC® F-68 poloxamer sold by BASF Corp. (Wyandotte, Mich., USA)), poloxamer 124 (including, but not limited to, PLURONIC® L44 poloxamer sold by BASF Corp. (Wyandotte, Mich., USA)), poloxamer 407 (including, but not limited to, PLURONIC® F127 poloxamer sold by BASF Corp. (Wyandotte, Mich., USA)), polyethoxylated vegetable oils, polyethoxylated castor oil (including but not limited to KOLLIPHOR® EL, formerly known as CREMOPHOR® EL sold by BASF Corp. (Wyandotte, Mich., USA)), sorbitan palmitate (including, but not limited to, SPAN™ 40 sold by Croda International Plc), lecithin, poly(vinyl alcohol) (“PVA”), human serum albumin, and mixtures thereof.
  • In particular embodiments of the invention, the stabilizer is a polymer. For example, the stabilizer can be, but is not limited to, a polyvinylpyrrolidone (“PVP”) (such as, but not limited to povidone K12, povidone K17, PLASDONE™ C-12 povidone, PLASDONE™ C-17 povidone, PLASDONE™ C-30 povidone, and mixtures thereof), polyethylene glycol 3350, and mixtures thereof.
  • In other embodiments of the invention, the stabilizer is an electrolyte, i.e., a salt that dissociates into anions and cations in aqueous solution. For example, the stabilizer can be, but is not limited to, sodium chloride, calcium chloride, and mixtures thereof.
  • In still other embodiments of the invention, the stabilizer is a non-electrolyte, i.e., is non-ionic. For example, the stabilizer can be, but is not limited to, dextrose, glycerol (also referred to as glycerin), mannitol, or mixtures thereof.
  • In other embodiments of the invention, the stabilizer is a cross-linked polymer. For example, the stabilizer can be, but is not limited to, carboxymethylcellulose sodium (CMC). In some embodiments of the invention, the stabilizer is CMC 7LF, CMC 7MF, CMC 7HF, or mixtures thereof.
  • In other embodiments of the invention, the stabilizer is a buffering agent, for example, NaH2PO4.H2O, NaH2PO4.2H2O, anhydrous NaH2PO4, sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or mixtures thereof.
  • In further embodiments of the invention, combinations of non-electrolyte stabilizers and electrolyte stabilizers may be used. In some embodiments, the combination of stabilizers may comprise two or more non-electrolyte stabilizers. In other embodiments, the combination of stabilizers may comprise two or more electrolyte stabilizers. In further embodiments, the combination of stabilizers may comprise one or more non-electrolyte stabilizers and one or more electrolyte stabilizers. In yet further embodiments, the combination of stabilizers may comprise two or more of mannitol, dextrose, and sodium chloride.
  • In certain embodiments of the invention, combinations of surfactant stabilizers and polymer stabilizers may be used. In some embodiments, the combination of stabilizers may comprise two or more surfactant stabilizers. In other embodiments, the combination of stabilizers may comprise two or more polymer stabilizers. In further embodiments, the combination of stabilizers may comprise one or more surfactant stabilizers and one or more polymer stabilizers. In yet further embodiments, the combination of stabilizers may comprise two or more of polysorbate 80, polysorbate 20, and poloxamer 188. In still further embodiments, the combination of stabilizers may comprise one or more of polysorbate 80, polysorbate 20, and poloxamer 188 and one or more of povidone K12, povidone K17, PLASDONE™ C-12 povidone, PLASDONE™ C-17 povidone, PLASDONE™ C-30 povidone, and polyethylene glycol 3350. In yet still further embodiments, the combination of stabilizers may comprise polysorbate 80 and one or more of PLASDONE™ C-12 povidone and povidone K12.
  • In certain embodiments, the pharmaceutical compositions comprising fulvestrant comprise CMC (carboxymethylcellulose sodium). In some embodiments, the CMC is prepared and heat sterilized before being combined with the fulvestrant during methods of preparation (described more fully elsewhere herein). In further embodiments of the invention, the viscosity of a CMC solution can be modulated by the degree of heating applied, which can allow for the formation of a plurality of fulvestrant pharmaceutical compositions having identical constituents, but with different viscosity values. These different viscosity values can affect the physical stability of the fulvestrant pharmaceutical compositions and the pharmacokinetic characteristics upon administration to subjects. In some embodiments, fulvestrant pharmaceutical compositions comprising CMC may be prepared in two or more parts with each part comprising a different amount of CMC. In other embodiments one or more such parts may be a suspension free of any CMC. In further embodiments, the parts can be mixed in an appropriate ratio to obtain a desired pharmaceutical composition.
  • In certain embodiments of the invention, the pharmaceutical compositions in the form of liquid suspensions comprising fulvestrant and one or more stabilizers may exhibit different sedimentation behaviors to form either flocculated or caked suspension upon storage. In some embodiments of the invention, after being stored, pharmaceutical compositions in the form of liquid suspensions comprising fulvestrant can be redispersed back into a homogeneous suspension with an acceptable particle size distribution upon redispersion. Exemplary liquid suspension formulations described herein were prepared and tested for sedimentation and redispersion. The tested formulations exhibited different sedimentation behaviors, but all were redispersible back to an acceptable, homogeneous suspension after a 3-month storage period at room temperature.
  • In certain embodiments of the invention, the pharmaceutical compositions comprising fulvestrant have a pH of from about 3-10, for example, about 3, 4, 5, 6, 7, 8, 9, or about 10. In further embodiments of the invention, the pharmaceutical composition has a pH of from about 5-8. In further embodiments of the invention, the pharmaceutical composition has a pH of from about 6-8. In further embodiments of the invention, the pharmaceutical composition has a pH of from about 3-7. In certain embodiments of the invention, the pharmaceutical composition has a pH of about 6.0 to 8.0. In particular embodiments of the invention, the pharmaceutical composition has a pH of about 6.0 to 7.0, 6.5 to 7.0, 6.5 to 7.5, 6.7 to 7.2, 7.0 to 7.2, 7.0 to 7.5, or 7.0 to 8.0. In further embodiments of the invention, the pharmaceutical composition has a pH of about 7.0.
  • In particular embodiments of the invention, the pharmaceutical composition further comprises one or more buffering agents, i.e., an agent that when added to a pharmaceutical composition, results in a pharmaceutical composition that resists pH changes or that results in a change in pH, such as, but not limited to, NaH2PO4.H2O, NaH2PO4.2H2O, anhydrous NaH2PO4, sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or mixtures thereof. In certain embodiments of the invention, the pharmaceutical composition comprises about 1 mM to 20 mM, of one or more buffering agents, and all ranges and subranges therebetween. In particular embodiments of the invention, the pharmaceutical composition comprises about 1 to 2 mM, 1 to 3 mM, 1 to 5 mM, 2 to 8 mM, 5 to 6 mM, 5 to 10 mM, 8 to 12 mM, 10 to 15 mM, or 15 to 20 mM of one or more buffering agents, and all ranges and subranges there between. In further embodiments of the invention, the pharmaceutical composition comprises about 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, 15 mM, 16 mM, 17 mM, 18 mM, 19 mM, or 20 mM of one or more buffering agents.
  • In certain embodiments of the invention, the pharmaceutical composition has an osmolarity from about 280 mOsm/L to about 310 mOsm/L, for example, about 280, 285, 290, 300, 305, or about 310 mOsm/L. In further embodiments of the invention, the pharmaceutical composition has an osmolarity from about 290 mOsm/L to about 300 mOsm/L. In yet further embodiments of the invention, the pharmaceutical composition has an osmolarity of about 290 mOsm/L. In some embodiments, the osmolarity may be selected through the use of appropriate amounts of the one or more stabilizers, e.g., stabilizers that also act as tonicifiers, such as, but not limited to, the non-electrolyte stabilizers and electrolyte stabilizers described herein. In some embodiments, the osmolarity may be selected through the use of appropriate amounts of one or more buffering agents that act as tonicifiers in a pharmaceutical composition, such as, but not limited to, the buffering agents described herein.
  • In certain embodiments of the invention, the pharmaceutical composition has an absolute viscosity measured at 25° C. from about 1.0 cP to about 1000 cP, and all ranges and subranges therebetween. In particular embodiments of the invention, the pharmaceutical composition has an absolute viscosity measured at 25° C. from about 750 cP to about 1000 cP, about 500 to about 750 cP, about 250 cP to about 500 cP, about 100 cP to about 250 cP, about 50 cP to about 100 cP, about 25 cP to about 50 cP, about 10 cP to about 25 cP, about 1 cP to about 10 cP, about 1 cP to about 5 cP, about 1.0 cP to about 4.0 cP, about 1.0 cP to about 3.0 cP, about 1.0 cP to about 2.5 cP, about 1.0 cP to about 2.0 cP, about 1.5 cP to about 2.0 cP. In further embodiments of the invention, the pharmaceutical composition has an absolute viscosity measured at 25° C. of about 1.0 cP, 1.1 cP, 1.2 cP, 1.3 cP, 1.4 cP, 1.5 cP, 1.6 cP, 1.7 cP, 1.8 cP, 1.9 cP, 2.0 cP, 2.1 cP, 2.2 cP, 2.3 cP, 2.4 cP, 2.5 cP, 2.6 cP, 2.7 cP, 2.8 cP, 2.9 cP, 3.0 cP, 3.5 cP, 4.0 cP, 4.5 cP, 5.0 cP, 10 cP, 15 cP, or 20 cP.
  • In yet further embodiments of the invention, pharmaceutical compositions having a fulvestrant concentration of 50 mg/mL or 100 mg/mL have an absolute viscosity measured at 25° C. that is from about 2-fold to about 500-fold lower than FASLODEX™, and all ranges and subranges therebetween. In further embodiments of the invention, fulvestrant pharmaceutical compositions having a fulvestrant concentration of 50 mg/mL or 100 mg/mL have an absolute viscosity measured at 25° C. that is 500-fold lower, about 400-fold lower, about 300-fold lower, about 250-fold lower, about 200-fold lower, about 150-fold lower, about 100-fold lower, about 50-fold lower, about 40-fold lower, about 30-fold lower, about 20-fold lower, about 10-fold lower, about 5-fold lower, about 4-fold lower, about 3-fold lower, about 2-fold lower, or about 1.5-fold lower than FASLODEX™. In further embodiments of the invention, for example, fulvestrant pharmaceutical compositions having a fulvestrant concentration of 50 mg/mL or 100 mg/mL, have an absolute viscosity measured at 25° C. that is substantially equivalent to FASLODEX™. Table 21 provides density measurements of some exemplary fulvestrant pharmaceutical compositions of the present disclosure. Table 22 provides viscosity measurements of some exemplary fulvestrant pharmaceutical compositions of the present disclosure.
  • In certain embodiments of the invention, the pharmaceutical composition comprises one or more additional pharmaceutically acceptable excipients. As used herein, a pharmaceutically acceptable excipient is generally chemically and/or physically compatible with other ingredients in a pharmaceutical composition or pharmaceutical composition, and/or is generally physiologically compatible with the recipient thereof. In some embodiments, the one or more additional pharmaceutically acceptable excipients are selected from the group consisting of preservatives, antioxidants, or mixtures thereof. In yet further embodiments of the invention, the additional pharmaceutically acceptable excipient is a preservative such as, but not limited to, phenol, cresol, p-hydroxybenzoic ester, chlorobutanol, or mixtures thereof. In yet further embodiments of the invention, the additional pharmaceutically acceptable excipient is an antioxidant such as, but not limited to, ascorbic acid, sodium pyrosulfite, palmitic acid, butylated hydroxyanisole, butylated hydroxytoluene, tocopherols, or mixtures thereof.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5.8 mg/mL of one or more stabilizers, and water for injection (WFI) q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 0.8 mg/mL of one or more polymers, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 0.8 mg/mL of povidone K12 (PVP 12K), and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5.8 mg/mL of one or more stabilizers, about 9 mg/mL of one or more electrolytes, about 10 mM of one or more buffering agents, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 0.8 mg/mL of one or more polymers, about 9 mg/mL of sodium chloride, about 10 mM of one or more of NaH2PO4.H2O, NaH2PO4.2H2O, and anhydrous NaH2PO4 (preferably a mixture of about 0.61 mg/mL NaH2PO4.2H2O and about 0.85 mg/mL of anhydrous NaH2PO4), and WFI q.s. to volume
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 55 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 50 mg/mL of dextrose, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more stabilizers, about 9 mg/mL of one or more electrolytes, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 9 mg/mL of sodium chloride, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 50 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 9 mg/mL of sodium chloride, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 55 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 56.6 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 1.6 mg/mL of one or more polymers, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.6 mg/mL of PLASDONE™ C-12 povidone, povidone K12, or a mixture thereof, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 57.4 mg/mL of one or more stabilizers, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of one or more surfactants, about 2.4 mg/mL of one or more polymers, about 50 mg/mL of one or more non-electrolytes, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 2.4 mg/mL of PLASDONE™ C-12 povidone, povidone K12, or a mixture thereof, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, between about 1 mg/mL and 2.4 mg/mL of PVP, sorbitan palmitate, poloxamer 188, poloxamer 124, poloxamer 427, polyethoxylated castor oil, PVA, or a mixture thereof, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 2.4 mg/mL of PVA, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In yet further embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.0 mg/mL of polyethoxylated castor oil, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 2.0 mg/mL of poloxamer 188, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of poloxamer 188, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of sorbitan palmitate, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of poloxamer 124, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • In certain embodiments of the invention, the pharmaceutical composition comprises about 100 mg/mL fulvestrant, about 5 mg/mL of polysorbate 80, about 1.5 mg/mL of poloxamer 407, about 50 mg/mL of mannitol, and WFI q.s. to volume.
  • Aspects of some exemplary embodiments of pharmaceutical compositions comprising fulvestrant are shown in Tables 1-20 and 23-27.
  • In the Figures and specification, references are made to exemplary formulations. Some exemplary formulations are identified as “F###” where each “#” is a numeral, e.g., F001, F002, and so on. The exemplary formulations sharing an initial identification “F###” share identical concentrations of constituent components (mg/mL), but may vary in their properties due to different methods of preparation, particle size distributions of fulvestrant, or other differences in processing, storage, or handling. Such exemplary formulation sharing an initial identification scheme “F###” are further identified by extra alphanumeric characters. For example, the exemplary formulations F003a, F003b, and F003k2 have the same concentrations of constituent components but may differ in, e.g., the underlying methods of preparation and resulting particle size distributions. In some instances in the Figures, formulations are identified by only ending non-zero numerals # or ## and subsequent alphanumeric characters; for example, formulation F003a may be referred to as “Variant 3a”, formulation F005a2 may be referred to as “Variant 5a2”, and the like. Some exemplary formulations are identified and referred to as “Lot”s, with references to the same Lot number referring to exemplary formulations having the same concentrations of constituent components, but may vary in their properties due to different methods of preparation, particle size distributions of fulvestrant, or other differences in processing, storage, or handling.
  • TABLE 1
    Target Formulation (mg/mL) Formulation B Formulation E Formulation I
    Fulvestrant 50 50 50
    Polysorbate 80 5 5 5
    PVP 12K 0.8 0.8 0.8
    NaCl 9
    WFI q.s. to volume q.s. to volume q.s. to volume
    Assay (% LC) 100.0 93.5 94.0
    Total Impurities (% a/a) 0.2 0.3 0.2
    PSD (via laser diffraction) as is sonicated as is sonicated as is sonicated
    Day 0 (μm) LD Dv(10) 7.4 6.0 2.4 1.9 NA NA
    LD Dv(50) 35.0 32.0 5.2 3.9
    LD Dv(90) 143 129 11.1 8.7
    (μm) day 5 day 1 day 4
    LD Dv(10) 7.6 2.3 2.3 1.2 2.5 0.04
    LD Dv(50) 31.9 5.7 5.0 1.8 5.6 0.8
    LD Dv(90) 107 19.2 10.9 2.5 12.3 3.2
    (μm) day 14 Day 10 Day 11
    LD Dv(10) 5.9 5.1 1.9 1.8 2.3 0.06
    LD Dv(50) 27.5 24.4 4.0 3.8 4.8 1.3
    LD Dv(90) 86.3 76.0 9.1 9.4 9.4 3.7
    (μm) Day 77 Day 71 Day 12
    LD Dv(10) 5.4 4.8 2.1 1.8 2.4 0.2
    LD Dv(50) 26.3 21.0 4.7 3.8 5.1 3.0
    LD Dv(90) 88.7 62.2 12.5 9.7 11.4 6.3
    pH 7.4 (day 11) 6.9 (day 7) 7.1 (day 4) 
    pH N/A N/A 6.9 (day 11)
  • TABLE 2
    Target Formulation (mg/mL) Formulation J Formulation K Formulation L
    Fulvestrant 50 50 100
    Polysorbate 80 5 5 5
    PVP 12K 0.8 0.8 0.8
    NaCl 9 9 9
    Phosphate buffer* 10 mM 10 mM 10 mM
    WFI q.s. to volume q.s. to volume q.s. to volume
    Assay (% LC) 80.9 82.4 94.1
    Total Impurities (% a/a) 0.2 0.2 0.2
    PSD (via laser diffraction) as is sonicated as is sonicated as is sonicated
    Day 0 (μm) LD Dv(10) 2.2 1.8 0.04 0.04 2.4 1.9
    LD Dv(50) 5.4 4.2 1.9 1.1 5.0 4.1
    LD Dv(90) 11.8 10.3 4.7 3.6 10.4 8.4
    (μm) day 5 day 4 Day 0
    LD Dv(10) 2.0 1.7 0.03 0.04 2.4 1.9
    LD Dv(50) 4.7 3.8 1.4 1.1 5.0 4.1
    LD Dv(90) 9.7 8.2 3.6 3.0 10.4 8.4
    (μm) Day 12 Day 11 Day 11
    LD Dv(10) 2.1 1.8 1.8 0.05 2.0 2.0
    LD Dv(50) 5.2 4.2 3.6 2.1 4.0 4.0
    LD Dv(90) 14.8 14.5 6.9 4.9 7.6 7.8
    (μm) Day 13 Day 12 Day 11
    LD Dv(10) 2.3 1.9 2.2 0.06 2.5 2.2
    LD Dv(50) 5.9 4.6 4.5 2.3 5.3 5.0
    LD Dv(90) 16.5 16.0 11.1 5.6 10.8 10.6
    pH 7.0 (day 4)  7.0 (day 1) 
    pH 7.0 (day 11) 7.1 (day 12)
  • TABLE 3
    Target Formulation (mg/mL)
    Formulation L3F Formulation L6
    Fulvestrant
    50 100
    Polysorbate 80 5 5
    PVP 12K 0.8 0.8
    NaCl 9 9
    Phosphate buffer* 10 mM 10 mM
    WFI q.s. to volume q.s. to volume
    Assay (mg/mL) 83.8 113.9
    Total Impurities (% a/a) 0.7 0.2
    PSD (via laser diffraction) as is sonicated as is sonicated
    Day 0 (μm) LD Dv(10) 2.1 2.0 2.2 2.1
    LD Dv(50) 5.9 5.5 6.9 6.7
    LD Dv(90) 14.7 14.2 17.6 17.7
    (μm) Day 14 Day 1
    LD Dv(10) 2.1 2.0 2.1 2.1
    LD Dv(50) 5.7 5.5 6.8 6.6
    LD Dv(90) 14.4 14.0 18.0 17.7
    pH 7.1 (day 1)
  • TABLE 4
    Target Formulation F003a F003b F004a
    Fulvestrant
    100 100 100
    Polysorbate 80 5 5 5
    Dextrose 50 50
    NaCl 9
    WFI q.s. to volume q.s. to volume q.s. to volume
    Manufacturing Process API size reduction by API size reduction by API size reduction by
    HSM only HSM followed by HPH HSM followed by HPH
    Assay (mg/mL) 95.0 96.3 99.8
    Total Impurities (% 0.42 0.28 0.27
    PSD (via laser diffraction) as is sonicated as is sonicated as is sonicated
    Day 0 LD 1.9 1.9 2.2 1.6 1.9 1.5
    (μm) LD 5.3 5.2 6.0 3.9 5.8 3.5
    LD 13.0 12.7 12.1 7.6 12.2 7.7
    (μm) Day 13 Day 13 Day 13
    LD Dv(10) 1.9 1.9 2.1 1.7 1.7 1.5
    LD Dv(50) 5.3 5.2 5.5 4.2 4.0 3.3
    LD Dv(90) 13.4 13.0 11.3 8.2 8.7 6.8
    (μm) Day 13 Day 13 Day 13
    LD Dv(10) 1.8 1.7 2.1 1.7 1.7 1.6
    LD Dv(50) 5.1 4.9 5.9 4.3 4.0 3.4
    LD Dv(90) 13.0 13.2 12.1 8.4 8.6 6.3
    pH 7.3 (Day 0)  7.5 (Day 0) 
    pH 7.1 (Day 13) 7.1 (Day 13)
  • TABLE 5
    Target Formulation (mg/mL) F003e F003k2 F003k3
    Fulvestrant 100 100 100
    Polysorbate 80 5 5 5
    Dextrose 50 50 50
    WFI q.s. to voltune q.s. to volume q.s. to volume
    Manufacturing Process Micronized API Micronized API Micronized API dispersed
    dispersed by vortex/ dispersed by HSM by HSM followed by HPH
    sonics or HSM for size reduction
    Assay (mg/mL) 100.5 97.8 99.6
    Total Impurities (% a/a) 0.34 0.43 0.42
    PSD (via laser diffraction) as is sonicated as is sonicated as is sonicated
    Day 0 (μm) LD Dv(10) 1.5 1.5 1.5 1.4 2.2 1.4
    LD Dv(50) 3.9 3.8 2.9 2.6 7.0 3.4
    LD Dv(90) 13.0 12.8 6.7 6.3 13.3 7.4
    (μm) Day 1 Day 5 Day 1
    LD Dv(10) 1.5 1.5 NA NA NA NA
    LD Dv(50) 4.0 4.0
    LD Dv(90) 12.4 12.5
    (μm) Day 13 Day 12 Day 8
    LD Dv(10) 1.5 1.5 1.6 1.5 1.5 1.3
    LD Dv(50) 4.1 4.1 3.2 2.9 2.9 2.6
    LD Dv(90) 14.0 14.3 7.0 6.5 5.7 5.2
    (μm) Day 22 Day 12 Day 8
    LD Dv(10) 1.5 1.5 1.5 1.4 1.4 1.1
    LD Dv(50) 3.9 3.9 3.0 2.8 2.8 1.9
    LD Dv(90) 12.6 12.8 6.7 6.1 6.2 3.5
    pH 7.2 (day 1)  7.2 (day 0)
    pH 4.5 (Day 12) 6.8 (Day 8)
  • TABLE 6
    Target Formulation (mg/mL) F003i F005a2 F005b1
    Fulvestrant
    100 100 100
    Polysorbate 80 5 5 5
    Dextrose 50
    Mannitol 50 50
    WFI q.s. to volume q.s. to volume q.s. to volume
    Manufacturing Process Reconstituted lyophilized Reconstituted lyophilized Micronized API
    suspension, micronized suspension, API sized dispersed by HSM
    API dispersed by HSM reduction by HSM
    Assay (mg/mL) 99.2 93.2 100.2
    Total Impurities (% a/a) 0.46 0.55 0.36
    PSD (via laser diffraction) as is sonicated as is sonicated as is sonicated
    Day 0 (μm) LD 4.6 1.7 2.1 2.0 1.5 1.4
    LD 54.2 5.6 6.5 5.6 3.2 2.7
    LD 112 16.2 18.4 13.5 7.9 6.6
    (μm) Day 9 Day 8 Day 13
    LD 2.5 1.5 2.1 2.0
    LD 39.5 4.3 6.2 5.4
    LD Dv(90) 95.1 13.5 16.7 13.2
    pH 6.5 (Day 3) 7.4 (Day 0)
    pH 7.2 (Day 9)
  • TABLE 7
    Target Formulation (mg/mL) F005c2 F0017a F0015a
    Fulvestrant
    100 100 100
    Polysorbate 80 5 15 25
    Mannitol 50 50 50
    WFI q.s. to volume q.s. to volume q.s. to volume
    Manufacturing Process Micronized API dispersed Micronized API Micronized API
    by HSM followed by HPH dispersed by HSM dispersed by HSM
    for size reduction
    Assay (mg/mL) 100.2 103.8
    Total Impurities (% a/a) 0.39
    PSD (via laser diffraction) as is sonicated as is sonicated as is sonicated
    Day 0 (μm) LD 1.3 1.1 1.5 1.4 1.5 1.4
    LD 2.3 1.9 3.0 2.6 2.9 2.6
    LD 4.9 3.7 7.2 6.4 6.9 6.4
    pH 7.1 (day 0) 7.1 (day 0)
  • TABLE 8
    Formulation/Variants
    mg/mL I J K L M N O, O2
    Fulvestrant
    50 50 50 100 100 100 100
    Polysorbate 80 5 5 5 5 5 5 5
    Povidone K12 0.8 0.8 0.8 0.8 0.8 0.8 0.8
    Sodium Chloride 9 9 9 9 9 4.5
    Dextrose 25 50
    NaH2PO4•2H2O 0.61 0.61 0.61
    Na2HPO4 0.85 0.85 0.85
    WFI QS QS QS QS QS QS QS
  • TABLE 9
    Formulation
    F001 F002 F003 F004 F005 F015 F016 F017
    variants (mg/mL)
    F003a, b, e, F005a, a2, b1,
    f, g, h, i, j, c, c2, c3, d, g, F015a, F017a,
    F001e B, E k2, k3, l F004a d1, g4, g5, h3 a1, a3 a1, a3
    Fulvestrant
    100 100 100 100 100 100 100 100
    Polysorbate 80 5 5 5 5 5 25 5 15
    Povidone K12 0.8 0.8
    Sodium Chloride 9
    Dextrose 50
    Mannitol 50 50 50 50
    WFI QS QS QS QS QS QS QS QS
  • TABLE 10
    Formulation
    mg/mL F003k+ F006 F007 F008 F009 F010 F011 F012 F013 F014
    Fulvestrant
    200 100 100 100 100 100 100 100 100 100
    Polysorbate 80 5 25 5 5 5 5 5 5 5 5
    Dextrose 50 50 50 50 50 50 50 50 50 50
    Polysorbate 20 15
    Poloxamer 188 2
    PVP K12 20
    PVP K17 5
    PEG 3350 60
    CMC7LF PH 30
    CMC7MF PH 20
    CMC7HF PH 5
    WFI QS QS QS QS QS QS QS QS QS QS
  • TABLE 11
    (mg/mL) F018 F019 F020 F021 F022 F023 F024 F025 F026 F027 F028
    Fulvestrant
    100 100 100 100 100 100 100 100 100 100 100
    Polysorbate 80 5 5 5 5 5 5 5 5 5 5 5
    Polysorbate 20 5 15 5 15 5
    Poloxamer 188 2 2 2 2
    Mannitol 50 50 50 50 50 50 50 50 50 50 50
    Dextrose
    NaCl
    Glycerol
    PVP K12
    5 10 20 10
    PVP K17 5
    PEG 3350 60
    CMC7LF PH
    CMC7MF PH
    CMC7HF PH
    WFI qs qs qs qs qs qs qs qs qs qs qs
  • TABLE 12
    (mg/mL) F029 F030 F031 F032 F033 F034 F035 F036
    Fulvestrant
    100 100 100 100 100 100 100 100
    Polysorbate 80 5 5 5 5 5 5 5 5
    Polysorbate 20 5
    Poloxamer 188 2
    Mannitol 50 25 12.5 37.5 50 50 50 50
    Dextrose 25 37.5 12.5
    NaCl
    Glycerol
    30
    PVP K12 5
    PVP K17
    PEG 3350
    CMC7LF PH 3 2 1 1
    CMC7MF PH
    CMC7HF PH
    WFI qs qs qs qs qs qs qs qs
  • TABLE 13
    (mg/mL) F037 F038 F039 F040 F041 F042 F043 F044 F045 F046 F047
    Fulvestrant
    100 100 100 100 100 100 100 100 100 100 100
    Polysorbate 80 5 5 5 5 5 5 5 5 5 5 5
    Polysorbate 20 5 5
    Poloxamer 188 2 2
    Mannitol 25 25 25 25 12.5 12.5 12.5 12.5 37.5 37.5 37.5
    Dextrose 25 25 25 25 37.5 37.5 37.5 37.5 12.5 12.5 12.5
    NaCl
    Glycerol
    PVP K12
    5 5
    PVP K17
    PEG 3350
    CMC7LF PH 3 2 1 1 3 2 1 1 3 2 1
    CMC7MF PH
    CMC7HF PH
    WFI qs qs qs qs qs qs qs qs qs qs qs
  • TABLE 14
    (mg/mL) F048 F049 F050 F051 F052 F053 F054 F055
    Fulvestrant* 100 100 100 100 100 100 100 100
    Polysorbate 80 5 5 5 5 5 5 5 5
    Polysorbate 20 5 5
    Poloxamer 188 2 2
    Mannitol 37.5 25 25 25 25 25 25 25
    Dextrose 12.5 25 25 25 25 25
    NaCl 4.9 9 4.9 4.9 4.9 4.9 4.9
    Glycerol
    PVP K12
    5 5
    PVP K17
    PEG 3350
    CMC7LF PH 1 3 2 1 1
    CMC7MF PH
    CMC7HF PH
    WFI qs qs qs qs qs qs qs qs
  • TABLE 15
    (mg/mL) F056 F057 F058 F059 F060 F061 F062
    Fulvestrant
    100 100 100 100 100 100 100
    Polysorbate 80
    Polysorbate 20 5 10 15 5 5 10 10
    Poloxamer 188
    Mannitol 50 50 50 50 50 50 50
    Dextrose
    NaCl
    Glycerol
    PVP K12
    PVP K17
    PEG 3350 10 30 10 30
    PEG 4000
    CMC7LF PH
    CMC7MF PH
    CMC7HF PH
    WFI qs qs qs qs qs qs qs
  • TABLE 16
    (mg/mL) F063 F064 F065 F066 F067 F068 F069 F070
    Fulvestrant
    100 100 100 100 100 100 100 100
    Polysorbate 80
    Polysorbate 20 15 15 5 5 10 10 15 15
    Poloxamer 188
    Mannitol 50 50 50 50 50 50 50 50
    Dextrose
    NaCl
    Glycerol
    PVP K12
    PVP K17
    PEG 3350 10 30
    PEG 4000 3 7.5 3 7.5 3 7.5
    CMC7LF PH
    CMC7MF PH
    CMC7HF PH
    WFI qs qs qs qs qs qs qs qs
  • TABLE 17
    Formulations/Variants
    Component Q R S T U V W X
    Fulvestrant (mg/mL) 100  100  100  100  100  100  100  100 
    Polysorbate 80 25 5 5  5  5
    (mg/mL)
    Polysorbate 20 5
    (mg/mL)
    Poloxamer 188 5
    (mg/mL)
    Lecithin (mg/mL) 5
    PVP K12 (mg/mL)   0.8 5   0.8   0.8   0.8   0.8
    PVP K17 (mg/mL)   0.8
    PEG 3350 (mg/mL) 50
    Dextrose (mg/mL) 50
    Sodium Chloride 9 13 
    (mg/mL)
    NaOH QS to QS to QS to QS to QS to QS to QS to pH QS to
    pH 7.0 pH 7.0 pH 7.0 pH 7.0 pH 7.0 pH 7.0 7.0 pH 7.0
    HCl QS to QS to QS to QS to QS to QS to QS to pH QS to
    pH 7.0 pH 7.0 pH 7.0 pH 7.0 pH 7.0 pH 7.0 7.0 pH 7.0
    WFI QS to QS to QS to QS to QS to QS to QS QS to
    volume volume volume volume volume volume to volume volume
  • TABLE 18
    Formulations/Variants
    Component
    1 2 3 4 5 6 7 8
    Fulvestrant 100  100  100  100  100  100  100  100 
    (mg/mL)
    Polysorbate 80 5 5 5 5
    (mg/mL)
    Polysorbate 20 5
    (mg/mL)
    Poloxamer 188 5
    (mg/mL)
    Human Serum 5
    Albumin (mg/mL)
    Lecithin (mg/mL) 5
    PVP K12   0.8 5   0.8   0.8   0.8   0.8
    (mg/mL)
    PVP K17   0.8
    (mg/mL)
    PEG 3350 50 
    (mg/mL)
    Dextrose 50 50 
    (mg/mL)
    Mannitol (mg/mL) 50 
    Citric buffer QS to QS to QS to QS to QS to QS to QS to pH QS to
    pH 7.0 pH 7.0 pH 7.0 pH 7.0 pH 7.0 pH 7.0 7.0 pH 7.0
    WFI QS to QS to QS to QS to QS to QS to QS QS to
    volume volume volume volume volume volume to volume volume
  • TABLE 19
    Formulation (mg/mL)
    F005g4 Lot Lot Lot Lot Lot Lot Lot
    F005g5 Lot
    15 26 27 28 42 43 X1 X2
    Fulvestrant
    100  100  100  100  100  100  100  100  100 
    Polysorbate 80 5 5  5 5 5  5  5  5  5
    PVPC12   1.6   1.6
    Span 40   1.5
    Pluronic F-68   1.5
    Pluronic L44   1.5
    Pluronic F127   1.5
    Mannitol (before 50  50  50  50 50 50 50
    homogenization)
    Mannitol (after 50 50 
    homogenization)
  • TABLE 20
    Formulation (mg/mL)
    Lot Lot Lot Lot
    45 46 47 48
    Fulvestrant 100 100 100 100
    Polysorbate 80 5 5 5 5
    PVPC12 2.4
    Span 40
    Pluronic F-68 2
    Cremophor EL 1
    PVA 2.4
    Mannitol 50 50 50 50
    (before
    homogenization)
  • TABLE 21
    Sample Density
    Name (g/ml)
    F003h 1.032
    F003f 1.032
    F003e 1.032
    F003k2 1.030
  • TABLE 22
    Sample Viscosity
    Name Formulation (cps)
    Placebo 5 mg/mL PS80 + 50 mg/mL 1.1
    Dextrose
    F003h [Described elsewhere] 1.8
    F003f [Described elsewhere] 1.9
    F003k [Described elsewhere] 2.0
    F003e [Described elsewhere] 1.5
  • B. Fulvestrant Particles
  • Particular embodiments of the disclosure comprise solid fulvestrant particles, for example a fulvestrant suspension comprising solid fulvestrant particles. In certain embodiments of the invention, at least about 90% of the total fulvestrant in the formulation is present as solid particles. In further embodiments of the invention, at least about 80% of the total fulvestrant in the formulation is present as solid particles.
  • In certain embodiments of the invention, solid fulvestrant particles are particles consisting of crystalline and/or amorphous fulvestrant. In other embodiments of the invention, fulvestrant particles comprise crystalline and/or amorphous fulvestrant as well as other excipients. In still other embodiments, fulvestrant particles comprise crystalline and/or amorphous fulvestrant coated or surface modified by a surface modifier adsorbed on the surface of the particle. The surface modifier can be a stabilizer such as, but not limited to surfactants, polymers, electrolytes, and non-electrolytes, and mixtures thereof.
  • Other embodiments of the present invention may further comprise fulvestrant in forms other than a solid particle, such as, but not limited to, solubilized fulvestrant as a free molecule or associated with a suspension such as micelles, microemulsions, emulsion, liposome, and combinations thereof, or complexed with other formulation constituents in a vehicle. In further embodiments of the invention, such other forms of fulvestrant are in equilibrium with the fulvestrant solid particles.
  • In particular embodiments of the invention, the fulvestrant particles comprise about 90-99.9% by weight of fulvestrant and 0.1-10% by weight of a surface modifier adsorbed on the surface of said particle. In particular embodiments of the invention, the surface modifier is a stabilizer such as, but not limited to surfactants, polymers, electrolytes, and non-electrolytes, and mixtures thereof. In certain embodiments of the invention, fulvestrant particles comprise at least about 90% fulvestrant. In other embodiments of the invention fulvestrant particles comprise at least about 92%, 95%, 97%, 98%, 99%, 99.5%, or 99.9% fulvestrant.
  • In further embodiments of the invention, one or more solvents, such as water, present in the pharmaceutical composition can be removed partially or completely by appropriate techniques known to the art, such as lyophilization or spray drying, to form a dried pharmaceutical composition for reconstitution. In certain embodiments of the invention, the dried pharmaceutical composition can comprise up to about 1%, about 2%, about 5%, or about 10% of the one or more solvents. The dried pharmaceutical composition can be reconstituted with appropriate diluent known to the art, such as, but not limited to water for injection (WFI), normal saline (NS), and 5% dextrose in water (D5W) prior to administration. In further embodiments of the invention, the diluent can further comprise an organic solvent or one or more of the excipients described herein. Dried pharmaceutical compositions formed by lyophilization may be in the form of a lyophilized cake.
  • Fulvestrant Particle Sizes
  • In certain embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In yet further embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In other embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 0.5 microns. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 0.5 microns. In other embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 microns. In still other embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1.5 microns. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1.5 microns. In yet other embodiments of the invention, the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • In further embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 0.5 microns. In other embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In still other embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 1.5 microns. In yet other embodiments of the invention, about 98% of fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) between about 4 microns and about 120 microns, between about 4 microns and about 100 microns, between about 4 microns and about 75 microns, between about 4 microns and about 60 microns, between about 4 microns and about 50 microns, between about 4 microns and about 40 microns, between about 4 microns and about 30 microns, between about 4 microns and about 20 microns, between about 4 microns and about 15 microns, between about 4 microns and about 10 microns, between about 20 microns and about 60 microns, between about 20 microns and about 45 microns, between about 20 microns and about 30 microns, between about 30 microns and about 50 microns, or between about 4 microns and about 9 microns. In other embodiments of the invention, the fulvestrant particles have a LD Dv(90) equal to about 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, about 60 microns, about 65 microns, about 70 microns, about 75 microns, about 80 microns, about 85 microns, about 90 microns, about 95 microns, about 100 microns, about 105 microns, about 110 microns, about 115 microns, or about 120 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 120 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 100 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 80 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 60 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 50 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 40 microns. In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 30 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 25 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 18 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 16 microns. In further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 14 microns. In still further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 11 microns. In yet further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 9 microns. In yet further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 7 microns. In yet further embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 5 microns. In particular embodiments of the invention, particles have an LD Dv(90) between about 9-14 microns. In other embodiments of the invention, the particles have an LD Dv(90) between about 12-14 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 9-11 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 7-9 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-8 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-7 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 3-6 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(50) between about 2 microns and about 35 microns, between about 2 microns and about 25 microns, between about 2 microns and about 20 microns, between about 2 microns and about 15 microns, between about 2 microns and about 10 microns, between about 2 microns and about 8 microns, between about 2 microns and about 7 microns, between about 2 microns and about 6 microns, between about 2 microns and about 5 microns, between about 2 microns and about 4 microns, between about 5 microns and about 10 microns, between about 5 microns and about 15 microns, between about 7 microns and about 10 microns, between about 8 microns and about 10 microns, or between about 9 microns and about 16 microns. In other embodiments of the invention, the fulvestrant particles have a LD Dv(50) equal to about 2 microns, 3 microns, 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 25 microns, about 30 microns, or about 35 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(50) less than or equal to about 9 microns. In other embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 7 microns. In other embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 6 microns. In yet other embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 5 microns. In particular embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 4 microns. In further embodiments of the invention, the particles have an LD Dv(50) less than or equal to about 3 microns. In further embodiments of the invention, the particles have an LD Dv(50) between about 4-6 microns. In further embodiments of the invention, the particles have an LD Dv(50) between about 3-5 microns. In yet further embodiments of the invention, the particles have an LD Dv(50) between about 3-4 microns. In yet further embodiments of the invention, the particles have an LD Dv(50) between about 2-3 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(10) no more than about 3 microns, about 2 microns, or about 1 microns. In further embodiments of the invention, the particles have an LD Dv(10) between about 1 micron and about 3 microns. In still further embodiments of the invention, the particles have an LD Dv(10) greater than or equal to about 2 microns. In yet further embodiments of the invention, the particles have an LD Dv(10) between about 1.5 microns to about 2.5 microns. In yet further embodiments of the invention, the particles have an LD Dv(10) between about 1 micron to about 2 microns. In yet further embodiments of the invention, the particles have an LD Dv(10) between about 1.0 micron to about 1.5 microns. In even further embodiments of the invention, the particles have an LD Dv(10) of about 2 microns. In even further embodiments of the invention, the particles have an LD Dv(10) of about 1.5 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) less than or equal to about 25 microns and an LD Dv(50) less than or equal to about 9 microns. In particular embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 16 microns and an LD Dv(50) less than or equal to about 6 microns. In other embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 11 microns and an LD Dv(50) less than or equal to about 5 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 9 microns and an LD Dv(50) less than or equal to about 4 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) less than or equal to about 8 microns and an LD Dv(50) less than or equal to about 4 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) between about 9-14 microns and an LD Dv(50) between about 4-6 microns. In still other embodiments of the invention, the particles have an LD Dv(90) between about 9-11 microns and an LD Dv(50) between about 4-6 microns. In particular embodiments of the invention, the particles have an LD Dv(90) between about 12-14 microns and an LD Dv(50) between about 4-6 microns. In further embodiments of the invention, the particles have an LD Dv(90) between about 6-8 microns and an LD Dv(50) between about 2-4 microns. In further embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In yet further embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In other embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns. In other embodiments of the invention, the particles have an LD Dv(90) between about 9-11 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 12-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-9 microns, an LD Dv(50) between about 2-4 microns, and an LD Dv(10) between about 1-2 microns. In further embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In yet further embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In other embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • In certain embodiments of the invention, the fulvestrant particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In other embodiments of the invention, the particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In yet other embodiments of the invention, the fulvestrant particles have an LD Dv(90) between about 30 microns and about 110 microns, an LD Dv(50) between about 5 microns and about 30 microns, and an LD Dv(10) between about 1.5 microns and about 3 microns. In other embodiments of the invention, the particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, the particles have an LD Dv(90) between about 9-14 microns, an LD Dv(50) between about 4-6 microns, and an LD Dv(10) between about 2-3 microns, and at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-9 microns, an LD Dv(50) between about 2-4 microns, an LD Dv(10) between about 1-2 microns, and the fulvestrant particles have a laser diffraction diameter greater than or equal to about 0.5 microns. In yet other embodiments of the invention, the particles have an LD Dv(90) between about 6-9 microns, an LD Dv(50) between about 2-4 microns, an LD Dv(10) between about 1-2 microns, and at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 0.5 microns. In further embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 1 micron. In yet further embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 1 micron. In other embodiments of the invention the fulvestrant particles have a laser diffraction diameter greater than or equal to about 2 microns. In still other embodiments of the invention, at least a portion of the fulvestrant particles have a laser diffraction diameter less than about 2 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dv(10) between about 1 microns and about 25 microns, between about 2 microns and about 25 microns, between about 3 microns and about 7 microns, between about 4 microns and about 15 microns, between about 4 microns and about 10 microns, between about 4 microns and about 8 microns, between about 6 microns and about 8 microns, between about 6 microns and about 7 microns, or between about 1 microns and about 10 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dv(10) equal to about 1 micron, about 2 microns, about 3 microns, about 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 21 microns, about 22 microns, about 23 microns, about 24 microns, or about 25 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dv(50) between about 5 microns and about 60 microns, between about 5 microns and about 50 microns, between about 9 microns and about 20 microns, between about 9 microns and about 15 microns, between about 10 microns and about 50 microns, between about 10 microns and about 40 microns, between about 10 microns and about 30 microns, between about 10 microns and about 20 microns, between about 15 microns and about 30 microns, between about 15 microns and about 25 microns, between about 15 microns and about 20 microns, or between about 10 microns and about 15 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dv(50) equal to about 5 micron, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, about 20 microns, about 21 microns, about 22 microns, about 23 microns, about 24 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, or about 60 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dv(90) between about 10 microns and about 200 microns, between about 25 microns and about 150 microns, between about 25 microns and about 125 microns, between about 25 microns and about 100 microns, between about 25 microns and about 75 microns, between about 25 microns and about 50 microns, between about 25 microns and about 40 microns, between about 25 microns and about 35 microns, between about 35 microns and about 90 microns, between about 35 microns and about 75 microns, between about 35 microns and about 50 microns, between about 35 microns and about 45 microns, between about 50 microns and about 100 microns, between about 50 microns and about 75 microns, or between about 20 microns and about 40 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dv(90) equal to about 10 microns, about 15 microns, about 20 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, about 60 microns, about 65 microns, about 70 microns, about 75 microns, about 80 microns, about 85 microns, about 90 microns, about 95 microns, about 100 microns, about 105 microns, about 110 microns, about 115 microns, about 120 microns, about 125 microns, about 130 microns, about 135 microns, about 140 microns, about 145 microns, about 150 microns, about 155 microns, about 160 microns, about 165 microns, about 170 microns, about 175 microns, or about 200 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dv(90) between about 35 microns and about 90 microns, a CE Dv(50) between about 10 microns and about 35 microns, and a CE Dv(10) between about 4 microns and about 10 microns. In other embodiments of the invention, the particles have a CE Dv(90) between about 25 microns and about 60 microns, a CE Dv(50) between about 10 microns and about 25 microns, and a CE Dv(10) between about 4 microns and about 8 microns. In other embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 35 microns, a CE Dv(50) between about 10 microns and about 20 microns, and a CE Dv(10) between about 4 microns and about 8 microns. In still other embodiments of the invention, the particles have a CE Dv(90) between about 30 microns and about 100 microns, a CE Dv(50) between about 10 microns and about 50 microns, and a CE Dv(10) between about 4 microns and about 10 microns. In yet other embodiments of the invention, the particles have a CE Dv(90) between about 50 microns and about 100 microns, a CE Dv(50) between about 20 microns and about 50 microns, a CE Dv(10) between about 6 microns and about 8 microns. In yet other embodiments of the invention, the particles have a CE Dv(90) between about 50 microns and about 75 microns, a CE Dv(50) between about 30 microns and about 40 microns, a CE Dv(10) between about 8 microns and about 10 microns. In yet other embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 60 microns, a CE Dv(50) between about 9 microns and about 20 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In still further embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 50 microns, a CE Dv(50) between about 9 microns and about 20 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In other embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 45 microns, a CE Dv(50) between about 9 microns and about 20 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In yet further embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 40 microns, a CE Dv(50) between about 9 microns and about 15 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In further embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 35 microns, a CE Dv(50) between about 9 microns and about 15 microns, and a CE Dv(10) between about 3 microns and about 7 microns. In still other embodiments of the invention, the particles have a CE Dv(90) between about 20 microns and about 45 microns, a CE Dv(50) between about 9 microns and about 15 microns, and a CE Dv(10) between about 3 microns and about 7 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dn(90) between about 4 microns and about 20 microns, between about 6 microns and about 15 microns, between about 6 microns and about 12 microns, between about 8 microns and about 12 microns, between about 8 microns and about 11 microns, between about 4 microns and about 10 microns, between about 4 microns and about 8 microns, between about 4 microns and about 7 microns, or between about 4 microns and about 6 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dn(90) equal to about 4 microns, about 5 microns, about 6 microns, about 7 microns, about 8 microns, about 9 microns, about 10 microns, about 11 microns, about 12 microns, about 13 microns, about 14 microns, about 15 microns, about 16 microns, about 17 microns, about 18 microns, about 19 microns, or about 20 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dn(50) between about 2.0 microns and about 10.0 microns, between about 2.0 microns and about 8.0 microns, between about 2.0 microns and about 6.0 microns, between about 2.0 microns and about 5.0 microns, between about 3.0 microns and about 5.0 microns, between about 3.5 microns and about 4.5 microns, between about 2.0 microns and about 4.0 microns, between about 2.5 microns and about 4.5 microns, or between about 2.5 microns and about 3.5 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dn(50) equal to about 2.0 microns, about 2.5 microns, about 3.0 microns, about 3.5 microns, about 4.0 microns, about 4.5 microns, about 5.0 microns, about 5.5 microns, about 6.0 microns, about 6.5 microns, about 7.0 microns, about 7.5 microns, about 8.0 microns, about 8.5 microns, about 9.0 microns, about 9.5 microns, or about 10.0 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dn(10) between about 0.5 microns and about 2.0 microns, between about 0.5 microns and about 1.5 microns, between about 1.0 microns and about 1.5 microns, between about 0.8 microns and about 1.2 microns, between about 0.9 microns and about 1.1 microns, or between about 0.5 microns and about 1.0 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dn(10) equal to about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1.0, about 1.1, about 1.2, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2.0 microns.
  • In certain embodiments of the invention, the fulvestrant particles have a CE Dn(90) between about 4 microns and about 20 microns, a CE Dn(50) between about 2.0 microns and about 10.0 microns, and a CE Dn(10) between about 0.5 microns and about 2.0 microns. In other embodiments of the invention, the fulvestrant particles have a CE Dn(90) between about 6 microns and about 12 microns, a CE Dn(50) between about 2.0 microns and about 6.0 microns, and a CE Dn(10) between about 0.5 microns and about 1.5 microns. In further embodiments of the invention, the fulvestrant particles have a CE Dn(90) between about 8 microns and about 1 microns, a CE Dn(50) between about 3.0 microns and about 5.0 microns, and a CE Dn(10) between about 0.8 microns and about 1.2 microns.
  • C. Preparation Methods
  • In certain embodiments of the invention, formulations of the invention can be prepared from commercially available fulvestrant having different particle size distributions, such as, for example, recrystallized, micronized fulvestrant, or a combination thereof. In further embodiments of the invention, the formulations are prepared with sterilized, commercially available fulvestrant. In particular embodiments, commercially available fulvestrant is used in the formulations of the present invention without further processing for size reduction.
  • In other embodiments of the invention, fulvestrant particles suitable for use in formulations of the invention can be prepared from commercially available fulvestrant by any suitable methods known in the art. Suitable methods include, but are not limited to, size-reduction techniques such as milling, grinding, crushing, compression, attrition, low shear mixing, high shear mixing, high pressure homogenization, lyophilization, precipitation, or combinations thereof.
  • Desired particle size distributions for fulvestrant particles can be achieved by processing steps at one or more stages of formulation preparation. In some embodiments, the desired particle size distribution can be formed by processing fulvestrant material prior to suspension in media, by techniques described more fully elsewhere herein. In other embodiments, the desired particle size distribution can be formed by processing after suspension in media, by techniques described more fully elsewhere herein, including but not limited to high shear mixing and high pressure homogenization. In still other embodiments, the desired particle size distribution can be formed by a combination of the processing prior to and after suspension in media.
  • Suitable milling techniques include, but are not limited to, dry milling, wet milling, and cryogenic milling. Suitable milling machines include ball mills, pebble mills, rod mills, roller mills, colloid mills, impact mills, and jet mills. In certain embodiments of the invention, the particles can be reduced in size in the presence of one or more excipients or stabilizers, such as but not limited to a surfactants, polymers, electrolytes, and non-electrolytes, and mixtures thereof. Alternatively, the particles can be contacted with one or more excipients or stabilizers after they are reduced in size.
  • In certain embodiments of the invention, the formulations can be prepared from an un-milled commercially available fulvestrant by reducing the fulvestrant particle size with high shear mixing. In yet further embodiments of the invention, the formulations can be prepared from an un-milled, commercially available fulvestrant by reducing the fulvestrant particle size with high shear mixing followed by high pressure homogenization.
  • In certain embodiments of the invention, the formulations can be prepared from commercially available micronized fulvestrant by reducing the micronized fulvestrant particle size with high shear mixing (“HSM”). In yet further embodiments of the invention, the formulations can be prepared from an un-milled commercially available fulvestrant by reducing the fulvestrant particle size with high shear mixing followed by high pressure homogenization (“HPH”).
  • In some embodiments of the invention, the formulations can be prepared using high pressure homogenization. In further embodiments of the invention, the high pressure homogenization process reduces particle size by subjecting the particle population to one or more of cavitation, shear, and impact within a homogenization chamber under operating pressures from about 5,000 psi to about 45,000 psi, for example, about 5,000, 10,000, 15,000, 20,000, 25,000, 30,000, 35,000, 40,000 or about 45,000 psi. In yet further embodiments of the invention, the high pressure homogenization process is performed at about 40,000 psi. In some embodiments of the invention, the high pressure homogenization process is performed at operating pressures from about 15,000 psi to about 20,000 psi. In further embodiments, the formulations can be prepared by passing the formulation suspension through a homogenization chamber under operating pressures for one or more passes, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, or 50 passes.
  • In the Figures and specification, references are made to exemplary formulations and processes for preparing exemplary formulations. Some exemplary preparation processes are identified with alphanumeric reference identifiers, such as “Process A1,” “Process A2,” and so on. Some exemplary formulations can share identical concentrations of constituent components (mg/mL), but may vary in their properties due to the different preparation processes, storage, or handling, which can result in different particle size distributions due to more or less size reduction, more or less aggregation or agglomeration, or both during processing, storage, or handling.
  • Methods of Forming Aqueous Fulvestrant Suspensions
  • In some embodiments of the invention, methods of forming an aqueous fulvestrant suspension comprise mixing an aqueous medium and at least one stabilizer to form a suspension vehicle, adding an amount of fulvestrant to the suspension vehicle, and dispersing the fulvestrant in the suspension vehicle to form the aqueous fulvestrant suspension. In further embodiments, these methods can further comprise homogenizing the aqueous fulvestrant suspension. In yet further embodiments, the methods with or without the homogenizing step can further comprise concentrating the fulvestrant suspension by phase separating the suspension and removing a portion of the supernatant. In particular further embodiments, after the concentrating step the methods can further comprise adding one or more electrolytes, non-electrolytes, buffering agents, or cross-linked polymers to the homogenized aqueous fulvestrant suspension and mixing the one or more electrolytes, non-electrolytes, buffering agents, or cross-linked polymers into the suspension. In some embodiments of the invention, the methods comprise a dispersing step performed using high shear mixing, a homogenizing step performed using high pressure homogenization, or both a dispersing step performed using high shear mixing and a homogenizing step performed using high pressure homogenization.
  • In further embodiments of the invention that include one or more stabilizers, the one or more stabilizers may be incorporated into the formulations at one or more stages of the methods of forming the formulations. In some embodiments, at least a portion or all of the stabilizers of a formulation are added to an aqueous medium along with an amount of fulvestrant prior to some or all of any mixing, homogenization, or supernatant-removal steps. In still other embodiments, at least a portion or all of the one or more stabilizers of a formulation are added to the fulvestrant suspensions after some or all of any mixing, homogenization, or supernatant-removal steps have been completed. In further embodiments, at least a portion or all of the surfactant and polymer stabilizers of the formulations are combined with the aqueous medium and fulvestrant prior to some or all of any mixing, homogenization, or supernatant-removal steps and at least a portion or all of the electrolyte, non-electrolyte, buffering agents, and cross-linked polymers of the formulations are added to the suspension after some or all of any mixing, homogenization, or supernatant-removal steps.
  • The fulvestrant particles described herein can be prepared in a method comprising the steps of dispersing fulvestrant particles in a liquid suspension medium and applying mechanical means in the presence of grinding media to reduce the particle size of fulvestrant to the desired size.
  • In further embodiments of the invention, a solvent, such as water, present in a formulation can be removed by appropriate techniques known to the art, such as lyophilization or spray drying, to form a dried formulation suitable for later reconstitution. Lyophilization can be used to produce a lyophilized (lyo) cake. The dried formulation can be reconstituted back into a liquid suspension using an appropriate diluent. Different volumes of diluent can be used to produce reconstituted suspensions with different fulvestrant concentrations as needed. The diluent can be aqueous in general but can further comprise an organic solvent and/or any excipient as described elsewhere herein.
  • In some embodiments of the invention, at least a portion of the formulation components other than fulvestrant can be omitted from the suspension and incorporated as part of the diluent and introduced into the suspension upon reconstitution by the diluent to arrive at the final formulation. In further embodiments, suspensions can be prepared with higher or lower concentrations of constituent components than desired in formulations for administration, formed into dried formulations and placed into vials in appropriate amounts of dried formulation to achieve target dose amounts of fulvestrant per vial for later reconstitution of diluent to form the desired formulation for administration.
  • Some exemplary methods of preparation of dried pharmaceutical compositions are depicted schematically in FIG. 15.
  • In some embodiments, the pharmaceutical compositions and dried pharmaceutical compositions can be prepared using aseptic process or terminally sterilized by a compatible sterilization technique, such as, but not limited to, gamma irradiation. When a polymer is used as an excipient in the pharmaceutical composition, said polymer such as microcrystalline cellulose (CMC) or its salts including sodium CMC, can be sterilized by autoclave in a solution then combined with rest of the pharmaceutical composition that is prepared aseptically or terminally sterilized.
  • Some aspects of exemplary embodiments of methods of preparation of the invention are shown in Tables 4-7 and 23-27 and FIGS. 4-12 and 15 which describe aspects of the preparation methods for exemplary fulvestrant formulations.
  • TABLE 23
    Target Formulation (mg/mL)
    B E I J K L
    Fulvestrant 50 50 50 50 50 100
    Polysorbate 80 5 5 5 5 5 5
    PVP 12K 0.8 0.8 0.8 0.8 0.8 0.8
    NaCl 9 9 9 9
    Phosphate 10 mM 10 mM 10 mM
    buffer
    WFI q.s. to q.s. to q.s. to q.s. to q.s. to volume q.s. to volume
    Starting API Un-milled Un-milled API Un-milled API Un-milled API Un-milled API Un-milled API
    (PSD via laser API L D Dv90: 780 L D Dv90: 780 L D Dv90: 1890 L D Dv90: 1890 L D Dv90: 1890
    diffraction) L D Dv90: microns microns microns microns microns
    780
    Manufacturing API size API size API size API size API size reduction API size reduction by
    process reduction reduction by reduction by reduction by by HSM followed HSM followed by
    by HSM HSM followed HSM followed HSM followed by HPH in the HPH in the presence
    by HPH by HPH, then by HPH in the presence of salts, of salts
    subsequent salt presence of then further HSM
    addition salts
    Process End L D Dv90: Total 15 min Total 42 min Total 45 min Total 45 min HSM Total 25 min HSM at
    Point Targets 143 HSM at HSM at HSM at at ~25,000 rpm
    (PSD via laser micron ~20,000 rpm ~20,000 rpm ~25,000 rpm ~25,000 rpm (L D Dv90:<~80
    diffraction) HSM: (L D Dv90:<~80 (L D Dv90:<~40 (L D Dv90:<~50 (L D Dv90:<~50 micron)
    Total 15 min micron) micron) micron) micron) L D Dv90: 13.8
    at HPH: 15 passes L D Dv90: 12.6 L D Dv90: 11.8 L D Dv90: 4.7 microns
    ~20,000 rpm in reverse flow micron micron micron HPH: 15 passes in
    through z5 HPH: 30 passes HPH: 3 passes HPH: 12 passes in parallel flow through
    nozzle at in parallel flow in parallel flow parallel flow z8 nozzle at ~30,000 psi
    ~30,000 psi through z5 through z5 through z5 nozzle at
    L D Dv90: 11.1 nozzle at nozzle at ~40,000 psi to
    micron ~40,000 psi ~40,000 psi L D Dv90: ~8
    micron then
    additional 5 min
    HSM at ~25,000 rpm
  • TABLE 24
    Target Formulation (mg/mL)
    L3F L6 F003a F003b F004a F003e
    Fulvestrant 100  100  100  100  100  100 
    Polysorbate 80 5 5 5 5 5 5
    PVP 12K   0.8   0.8
    NaCl 9 9
    Phosphate 10 mM 10 mM 9
    buffer
    Dextrose 50  50  50 
    WFI q.s. to q.s. to q.s. to q.s. to q.s. to q.s. to
    Starting API Un-milled Un-milled Un-milled API Un-milled API Un-milled API Jet-milled
    (PSD via laser API API L D Dv90: L D Dv90: 240 L D Dv90: 240 API
    diffraction) L D Dv90: 240 micron micron micron
    Manufacturing API size API size API size API size API size API size
    process reduction reduction by reduction by reduction by reduction by dispersed
    by HSM HSM HSM (F001e), HSM followed HSM followed by HSM
    then by HPH, then by HPH, then and
    subsequent subsequent subsequent sonication/
    dextrose dextrose NaCl addition vortex
    addition addition
    Process End L D Dv90: L D Dv90: L D Dv90: Total 15 min Total 15 min L D Dv90:
    Point Targets 14.7 17.6 micron 13.6 micron HSM at HSM at 12.2
    (PSD via laser micron and and L D Dv50: ~25,000 rpm to ~25,000 rpm to micron
    diffraction) and L D Dv50: 6.9 5.7 micron target target HSM:
    L D Dv50: micron HSM: Total L D Dv90:<~40 L D Dv90:<~40 Total 5 min
    5.9 HSM: Total 60-120 min micron micron at
    micron >120 min at at L D Dv90: 15.1 L D DV90: 15.1 ~25,000 rpm
    HSM: ~25,000-30,000 rpm ~25,000 rpm micron micron to
    Total before HPH: 15 passes HPH: 15 passes L D Dv90:
    >120 min dextrose in parallel flow in parallel flow 12.6
    at addition to through z5 through z5 micron by
    ~25,000-30,000 rpm L D Dv90: 13.0 nozzle at nozzle at sonication/
    micron ~40,000 psi ~40,000 psi vortex
    before dextrose before NaCl
    addition to addition to
    L D Dv90: 12.1 L D Dv90:
    micron 12.2 micron
    pH = 7.3 pH = 7.5
  • TABLE 25
    Target Formulation (mg/mL)
    F003k2 F003k3 F003l F005a2
    Fulvestrant
    100 100 100 100
    Polysorbate 80 5 5 5 5
    Dextrose 50 50 50
    Mannitol 50
    WFI q.s. to q.s. to q.s. to volume q.s. to volume
    Starting API Jet-mill API Jet-mill API Jet-mill API L D Dv90: Un-milled API L D Dv90:
    (PSD via laser L D Dv90: 7-8 L D Dv90: 7-8 7-13 micron 240 micron
    diffraction) micron micron
    Manufacturing API dispersed API dispersed by API dispersed by HSM API size reduction by
    process by HSM, then HSM then size (F003i, f, j). Suspension HSM (F001e), then
    subsequent reduction by HPH lyophilized, reconstituted mannitol addition.
    dextrose in the presence of then composited Suspension lyophilized,
    addition dextrose reconstituted then
    composited
    Process End L D Dv90: 6.7 Total 5 min HSM Total 5 min HSM at L D Dv90: 13.6 micron
    Point Targets micron after at ~25,000 rpm for and L D Dv50: 5.7 micron
    (PSD via laser dextrose ~25,000 rpm dispersing each HSM: Total 60-120 min.
    diffraction) addition (Dv90: 9.2 individual API lot at
    HSM: Total 5 min micron) for F003i, f, j before ~25,000 rpm before
    at dispersing API lyophilization, L D Dv90: mannitol addition to
    ~25,000 rpm L D Dv90: 13.3 7.0, 12.9, 7.3 micron L D Dv90: 13.2 micron
    micron HPH: 9 After reconstitution, (F005a)
    passes in parallel L D Dv90: 121, 113, 113 After lyophilization,
    flow through z5 micron. reconstitution, composite
    nozzle at Composite F003l, F005a2, Dv90: 18.4
    ~40,000 psi; L D Dv90: 112 micron micron
    dextrose co-
    processed
  • TABLE 26
    Target Formulation (mg/mL)
    F005b1 F005c2 F005c3 F005d1 F015a1 F015a3
    Fulvestrant 100 100 100 100 100 100
    Polysorbate 80 5 5 5 5 25 25
    Mannitol 50 50 50 50 50 50
    WFI q.s. to q.s. to q.s. to q.s to q.s. to q.s. to
    Starting API Jet-mill API Jet-mill Jet-mill API Jet-mill API Jet-mill API Jet-mill API
    (PSD via laser L D Dv90: 7-8 API L D Dv90: 7-8 L D Dv90: 7-8 L D Dv90: 7-8 L D Dv90: 7-8
    diffraction) micron L D Dv90: micron micron micron micron
    7-8 micron
    Manufacturing API API Lyophilized API size API Lyophilized
    process dispersed by dispersed F005c2 reduction by dispersed by F015a1
    HSM in the by HSM HSM followed HSM in the
    presence of then size by HPH, then presence of
    mannitol reduction subsequent mannitol
    by HPH in mannitol
    the addition
    presence of
    mannitol
    Process End L D Dv90: Total 5 min After To total 5 min L D Dv90: After
    Point Targets 7.9 HSM at reconstitution, HSM at 6.9 reconstitution,
    (PSD via laser micron ~25,000 rpm L D Dv90: ~25,000 rpm for micron L D Dv90:
    diffraction) HSM: Total for 112 micron dispersing API HSM: Total 22.7
    5 min at dispersing L D Dv90: ~7 5 min at micron
    ~25,000 rpm API micron ~25,000 rpm
    (L D Dv90: HPH with 15
    ~7 micron) passes in parallel
    To HPH flow through z5
    with 15 nozzle at
    passes in ~40,000 psi
    parallel resulted in
    flow Dv90:
    through z5 ~16 micron
    nozzle at (F001h4)
    ~40,000 psi L D Dv90: 4.0
    resulted in micron after
    L D Dv90: mannitol
    17.2 micron addition
    pH = 7.1 pH = 7.1
  • TABLE 27
    Target Formulation (mg/mL)
    F015a4 F017a1 F017a3 F005g4 F005g5
    Fulvestrant
    100 100 100 100 100
    Polysorbate 80 25 15 15 5 5
    Mannitol 50 50 50 50 50
    WFI q.s. to q.s. to q.s. to q.s. to q.s. to
    Starting API Jet-mill API Jet-mill API Jet-mill API Recrystallized API Recrystallized
    (PSD via laser L D Dv90: 7-8 L D Dv90: 7-8 L D Dv90: 7-8 L D Dv90: 18 API
    diffraction) micron micron micron micron L D Dv90: 18
    micron
    Manufacturing F015a3 gamma API dispersed by Lyophilized, API dispersed by Lyophilized
    process irradiated at 35 HSM in the gamma HSM in the F005g4
    kGy presence of irradiated (35 presence of
    mannitol KGy) F017a1 mannitol
    Process End After L D Dv90: 7.2 After Total 5 min HSM at
    Point Targets reconstitution, micron reconstitution, ~25,000 rpm for
    (PSD via laser L D Dv90: 22.7 HSM: Total 5 min L D Dv90: 31.9 dispersing API
    diffraction) micron at micron (L D Dv90: ~20
    ~25,000 rpm micron)
    L D Dv90: ~10
    micron after HPH
    with 9 passes in
    parallel flow
    through z5 nozzle at
    ~40,000 psi
    Final L D Dv90: 7.5
    micron after
    concentration
  • D. Pharmacokinetics
  • In certain embodiments of the invention, the pharmaceutical compositions are bioequivalent to the commercial pharmaceutical composition, FASLODEX™. The single dose PK parameters in postmenopausal advanced breast cancer patients administered FASLODEX™ dosed intramuscularly with 500 mg with an additional dose at day 15 are reported as, in geometric mean and coefficient of variation (%), Cmax 25.1 (35.3) ng/mL, Cmin 16.3 (25.9) ng/mL, and AUC 11,400 (33.4) ng·hr/mL.
  • In further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of the pharmaceutical composition of the invention is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of FASLODEX™. In yet further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of the pharmaceutical composition of the invention is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of FASLODEX™ in the fasting state. In still further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of the pharmaceutical composition of the invention is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of FASLODEX™ in the fed state.
  • In other embodiments of the invention, the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of the pharmaceutical composition of the invention having a fulvestrant concentration of 100 mg/mL is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of FASLODEX™. In still other embodiments of the invention, the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of the pharmaceutical composition of the invention having a fulvestrant concentration of 100 mg/mL is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of FASLODEX™ in the fasting state. In yet other embodiments of the invention, the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of the pharmaceutical composition of the invention having a fulvestrant concentration of 100 mg/mL is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of FASLODEX™ in the fed state.
  • In particular embodiments of the invention, the pharmaceutical composition has the single dose and multiple dose pharmacokinetic parameters shown in Tables 28 and 29. Table 28 shows pharmacokinetic parameters for 500 mg dosage of pharmaceutical compositions of the disclosure. For the data labeled “Single Dose” in Table 28, the fulvestrant blood plasma concentration data are shown for a 500 mg initial dose with an additional 500 mg dose given on day 15. For the data labeled “Multiple Dose Steady State” in Table 28, the fulvestrant blood plasma concentration data are shown for measurement at month 3, after a 500 mg dosage on days 1, 15, 20, and once monthly thereafter. Table 29 shows pharmacokinetic parameters for a single 250 mg dosage of pharmaceutical compositions of the disclosure. In Table 29, data are expressed as geometric mean (CV %), except for Tmax, which is shown as a median value with a range indicated in parentheses.
  • TABLE 28
    Cmax (ng/mL) Cmin (ng/mL) AUC (ng · hr/mL)
    Single Dose1  20.08-31.375 13.04-20.375  9,120-14,250
    Multiple Dose 22.4-35.0 9.76-15.25 10,480-16,375
    Steady State2
  • TABLE 29
    I II III IV V VI VII
    Cmax 8.20 4.76 8.2   4-8.5 11.8 8.3  8-12
    (μg/L) (63.8) (68.1) (6.6) (8.8)
    Cmin 2.62 2.38 2.6 2.0-3.0
    (μg/L) (33.4) (47.7)
    Tmax 6.97 8.8 7 6-9 4.2 4.6 4-5
    (days) (1.86-7.95) (6.97-12.0) (8.3) (11.2)
    AUC28 148 88.4 148  80-150 369 333 325-375
    (μg · day/L) (45.3) (47.3) (4.1) (3.0)
  • In particular embodiments, a dose of about 500 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEX™. In certain embodiments, a dose of less than 500 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEX™. In further embodiments, a dose of about 400 to 450 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEX™. In still further embodiments, a dose of about 350 to 400 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEX™. In yet further embodiments, a dose of about 300 to 350 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEX™. In even further embodiments, a dose of about 250 to 300 mg of a fulvestrant pharmaceutical composition of the invention is bioequivalent to 500 mg of the commercial pharmaceutical composition, FASLODEX™.
  • In other embodiments of the invention, a 500 mg dose of a pharmaceutical composition of the invention provides 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of a 500 mg dose of FASLODEX™.
  • In other embodiments of the invention, a dose of less than 500 mg of a pharmaceutical composition of the invention provides 90% confidence intervals (CI) of the relative mean Cma, AUC(0-t) and AUC(0-∞) within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of a 500 mg dose of FASLODEX™.
  • In some embodiments of the invention, fulvestrant pharmaceutical compositions of the invention can be administered as a single intramuscular injection, with the 90% confidence intervals (CI) of the relative mean Cmax, AUC(0-t) and AUC(0-∞) of fulvestrant is within 80% to 125% of the relative mean Cmax, AUC(0-t) and AUC(0-∞), respectively, of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections. In further embodiments, such fulvestrant pharmaceutical compositions administered as a single intramuscular injection comprise a dose of about 500 mg of fulvestrant. In yet further embodiments, such fulvestrant pharmaceutical compositions administered as a single intramuscular injection comprise a dose of about 500 mg of fulvestrant in an injection volume of about 3.0 mL to about 5.0 mL, about 3.5 mL to about 4.5 mL, or about 4.0 mL.
  • In certain embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80% of the relative mean Cmax of FASLODEX™. It is believed that such embodiments may provide benefits by providing a therapeutically effect amount of fulvestrant exposure to a subject while reducing the degree of one or more Cmax-driven side-effects or toxicities in comparison to the degree of side-effects or toxicities experienced by a subject from receiving a therapeutically effective amount of fulvestrant exposure from one or more dosages of FASLODEX™.
  • In some embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, or less than 40% of the relative mean Cmax of FASLODEX™. In further embodiments, such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • In yet further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, or less than 40% of the relative mean Cmax of FASLODEX™ in the fasting state. In further embodiments, such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • In still further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, or less than 40% of the relative mean Cmax of FASLODEX™ in the fed state. In further embodiments, such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • In some embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, within about 45% to about 55%, within about 55% to about 65%, within about 65% to about 75%, within about 50% to about 60%, within about 60% to about 70%, or within about 70% to about 80% of the relative mean Cmax of FASLODEX™. In further embodiments, such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • In yet further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, within about 45% to about 55%, within about 55% to about 65%, within about 65% to about 75%, within about 50% to about 60%, within about 60% to about 70%, or within about 70% to about 80% of the relative mean Cmax of FASLODEX™ in the fasting state. In further embodiments, such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • In still further embodiments of the invention, the 90% confidence intervals (CI) of the relative mean AUC(0-t), relative mean AUC(0-∞), or both of fulvestrant pharmaceutical compositions of the invention is within 80% to 125% of the relative mean AUC(0-t) and relative mean AUC(0-∞), respectively, of FASLODEX™, and the relative mean Cmax of fulvestrant pharmaceutical compositions of the invention is about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, within about 45% to about 55%, within about 55% to about 65%, within about 65% to about 75%, within about 50% to about 60%, within about 60% to about 70%, or within about 70% to about 80% of the relative mean Cmax of FASLODEX™ in the fed state. In further embodiments, such fulvestrant pharmaceutical compositions are administered as a single intramuscular injection and comprise a dose of about 500 mg of fulvestrant at a concentration of about 100 mg/mL.
  • E. Methods of Treatment
  • In further embodiments, the invention is directed to methods of treatment comprising administration of a pharmaceutically effective amount of any of the fulvestrant pharmaceutical compositions described herein to a patient in need thereof. In particular embodiments, the invention is directed to a method of treating breast cancer, comprising administering a pharmaceutically acceptable amount of any of the fulvestrant pharmaceutical compositions described herein. In certain embodiments, the breast cancer is metastatic breast cancer. In other embodiments of the invention, the breast cancer is hormone receptor (HR)-positive breast cancer. In still other embodiments of the invention, the invention is directed to a method of treating hormone receptor (HR)-positive breast cancer in a post-menopausal woman comprising administration of a pharmaceutically effective amount of any of the fulvestrant pharmaceutical compositions described herein. In yet other embodiments, the invention is directed to a method of treating hormone receptor (HR)-positive breast cancer in a post-menopausal woman with disease progression following antiestrogen therapy comprising administration of a pharmaceutically effective amount of any of the fulvestrant pharmaceutical compositions described herein. In yet further embodiments, the invention is directed to a method of treating HR-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced or metastatic breast cancer in a woman with disease progression after endocrine therapy.
  • In particular embodiments of the invention, a fulvestrant pharmaceutical composition as described herein is administered on days 1, 15, 29, and once monthly thereafter. In further embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered on days 1, 15, 29, and once monthly thereafter. In still further embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered on days 1, 15, 29, and once monthly thereafter.
  • In certain embodiments of the invention, a fulvestrant pharmaceutical composition as described herein is administered as a single injection. In other embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single injection. In yet other embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 5 mL injection. In further embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 4 mL injection. In yet further embodiments, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 3 mL injection. In still other embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single injection. In further embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 2.5 mL injection. In yet further embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as a single 5 mL injection.
  • In particular embodiments of the invention, a fulvestrant pharmaceutical composition as described herein is administered as two injections. In further embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two injections. In still further embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two 5 mL injections. In yet further embodiments of the invention, a 500 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two 2 mL injections, two 2.5 mL injections, two 3 mL injections, two 3.5 mL injections, or two 4 mL injections. In other embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two injections. In yet other embodiments of the invention, a 250 mg dose of any of the fulvestrant pharmaceutical compositions as described herein is administered as two 2.5 mL injections.
  • The fulvestrant pharmaceutical compositions described herein may be administered alone, or in combination with one or more additional therapeutic agents as defined herein. An additional therapeutic agent may be used to treat one or more core symptoms and/or comorbidities associated with cancer in general or breast cancer in particular. In one aspect, fulvestrant is formulated (and administered) with at least one therapeutic agent as a fixed dose. In another aspect, fulvestrant is formulated (and administered) separately from the therapeutic agent(s).
  • Some examples of therapeutic agents that may be used in combination with fulvestrant include, but are not limited to, e.g., a EGFR kinase inhibitor, a PDGFR kinase inhibitor, a FGFR kinase inhibitor, or any of the other cytotoxic, chemotherapeutic, antihormonal, anti-angiogenic, antiproliferative, pro-apoptotic, anti-HER2, radiation or a radiopharmaceutical, signal transduction inhibitors, or other anti-cancer agents or treatments. Examples of particular agents that can be used in combination with the fulvestrant pharmaceutical compositions of the disclosure include palbociclib, letrozole, anastrozole, doxorubicin, paclitaxel, docetaxel, vinorelbine, and 5-fluorouracil. In other embodiments, therapeutic agents that may be used in combination with fulvestrant include, but are not limited to, agents or treatments for one or more of pain, nausea, emesis, hot flushes, constipation, and dizziness.
  • Those skilled in the art will appreciate that numerous changes and modifications can be made to the preferred embodiments of the disclosure and that such changes and modifications can be made without departing from the spirit of the disclosure. It is, therefore, intended that the following examples and appended claims cover all such equivalent variations as fall within the true spirit and scope of the disclosure.
  • The disclosures of each patent, patent application, and publication cited or described in this document are hereby incorporated herein by reference, in its entirety.
  • EXAMPLES Example: Preparation of Fulvestrant Pharmaceutical Compositions/Variants
  • Some exemplary fulvestrant pharmaceutical compositions were prepared with 50 mg/mL and 100 mg/mL concentrations of fulvestrant in aqueous suspensions. Tables 1-20 show aspects of the pharmaceutical compositions of the pharmaceutical compositions and the methods of preparation of some of the pharmaceutical compositions, also referred to as formulations, variants, or Lots in the Tables. Tables 4-7 and 23-27 and FIGS. 4-12 and 15 show aspects of the methods of preparation used to prepare some of the pharmaceutical compositions.
  • Where indicated in the Tables and Figures, the formulations B, E, I, J, K, L, L3F, L6, F003a, F003b, F003e, F004a, F003k2, F003k3, F005a2, F003l, F005b1, F015a1, F015a3, F005d1, F005c3, F005g5 tested in Studies 1-3 below were prepared via one or more of process steps of (1) low shear mixing, indicated in the Tables 23-27 and FIGS. 4-12 as “Mix” or “Mix with Vortex Mixer” steps; (2) high shear mixing, indicated as “HSM” or “Homogenize” steps; (3) high pressure homogenization, indicated as “HPH” or “Process with Nano DeBee” steps; (4) concentration via supernatant removal; and (5) application of sonication. Where indicated, supernatant removal was performed by phase separating the pharmaceutical composition and withdrawing the desired amount of supernatant to concentrate the suspensions to the target concentrations of fulvestrant, either 50 mg/mL or 100 mg/mL, depending on the pharmaceutical composition. Where indicated, phase separation was performed by overnight settling in a clear glass centrifuge tube. Application of centrifuge for phase separation could also be utilized.
  • Fulvestrant active pharmaceutical ingredient (which may also be referred to as “API” herein and in the Tables and Figures) was obtained in un-milled forms or in milled, micronized, or recrystallized forms from commercial suppliers. As-obtained particle size distributions for un-milled API varied from an LD Dv(90) of about 240 microns to an LD Dv(90) of about 2130 microns. As-obtained particle size distributions for milled, micronized, and recrystallized API varied from an LD Dv(90) of about 7 microns to an LD Dv(90) of about 18 microns. Fulvestrant API may be obtained in various particle size distributions from commercial sources and processed as described elsewhere herein to achieve the desired particle size distributions. Particle size distributions can be monitored throughout the processing steps through analysis of samples as described elsewhere herein.
  • Where indicated in the Tables and Figures, the formulations B, E, I, J, K, L, L3F, L6, F003a, F003b, F003e, F004a, F003k2, F003k3, F005a2, F003l, F005b1, F015a1, F015a3, F005d1, F005c3, F005g5 tested in Studies 1-3 below were prepared via high shear mixing (HSM) steps. The preparation of formulations can be performed with an IKA T10 Basic Disperser with an IKA S10N-10G dispersing tool. At the speeds indicated (˜20,000 to 30,000 rpm), the mixture of fulvestrant and suspension vehicle was processed in cycles until the total processing time indicated was reached. Between each cycle, a formulation was vortexed at ˜3000 rpm for 30 seconds then sonicated for 1 minute to remove or reduce foam generated during the high shear mixing by the disperser. Formulations were also rested as needed in between cycles at room temperature to allow the disperser to cool down and avoid overheating of the product and the equipment. Sonication was performed with a Branson 3800 Ultrasonic Bath (Branson Ultrasonics Corp., Danbury, Conn.) at a frequency of 40 kHz. Other mixing and ultrasonic apparatuses may also be used to achieve mixing and particle size distribution as desired.
  • In some embodiments, high pressure homogenization was performed. In certain embodiments, high pressure homogenization (HPH) steps were performed with a Nano DeBEE High Pressure Homogenizer (BEE International, South Easton, Mass.) in a Labconco XPert Filtered Balance System (Model 3950630) (Labconco, Kans. City, Mo.), installed the 100 ml sample holder and Z5 nozzle in parallel flow configuration on Nano DeBEE High Pressure Homogenizer. The homogenizer was primed with filing water until the process pressure reached the processing pressure as indicated in the Tables and Figures. Water was removed from the system using the plunger to minimize the dilution of the batch by the residual priming water. Approximately ˜50 ml of the suspension for HPH processing was loaded from the 50 mL clear Pyrex glass bottle on Nano DeBEE High Pressure Homogenizer. The Nano DeBEE was run in continuous mode until the pressure reached the indicated target processing pressure. The suspension was then processed for the indicated number passes at the processing pressure. To avoid losing the prime of the system and consequently the processing pressure, only total ˜40 mL (8 strokes of ˜5 mL per stroke) of the suspension was processed and collected from each pass. The 40 mL suspension was then loaded back to the reservoir for the suspension to be processed in the next pass. After the processing was completed, 40 ml fine suspension was collected in a 100 mL clear Pyrex glass bottle by running Nano DeBEE High Pressure Homogenizer until no sample was pumped out. In certain embodiments, high pressure homogenization was performed with other apparatuses at processing pressures ranging from about 5,000 psi to about 45,000 psi. Other high pressure homogenization apparatuses may also be used to achieve the desired particle size distributions described herein.
  • Some formulations for Study 3 below were lyophilized and reconstituted with sterile water for injection, USP prior to administration, as indicated in the Example below.
  • References to “Assay” refers to high-performance liquid chromatography (HPLC) measurement of the fulvestrant concentration of the pharmaceutical composition at intermediate processing steps or in final result as prepared. The “Assay” results are given in absolute measured mg/mL or as a percentage (%) or (% LC), where percentages indicate the concentration of fulvestrant relative to the 50 mg/mL label claim of the commercially available FASLODEX™ product. Total impurities were also measured and are provided in the figures as a percentage by area (% a/a) where indicated. HPLC was performed with Agilent Technologies Agilent 1260 Infinity Quaternary LC module G1311B (Agilent Technologies, Santa Clara, Calif.). Other HPLC apparatuses may also be used to analyze the fulvestrant concentrations.
  • In some aspects, particle size and particle size distributions were analyzed with Malvern Mastersizer 3000 (Malvern Instruments Ltd., Malvern, Worcestershire, UK), with an attached sample dispersion unit with an in-line sonication probe for agglomerate dispersion prior to analysis via laser diffraction.
  • In some aspects, particle size and particle size distributions were analyzed with Malvern Morphologi G3 (Malvern Instruments Ltd., Malvern, Worcestershire, UK), to determine circle equivalent (CE) diameters via microscopy image capture and analysis.
  • Measurements of pH were obtained at ambient room temperature with a Thermo Scientific Orion Star A211 pH Meter (Thermo Fisher Scientific Inc., Waltham, Mass.).
  • Example: Pharmacokinetic Study 1 of Intramuscular Administration to Female Dogs
  • Fulvestrant pharmaceutical compositions B, E, I, J, K, and L were prepared as described elsewhere herein and in the Figures. A preclinical study was performed to determine the pharmacokinetics of the pharmaceutical compositions following a single intramuscular administration of 15.4 mg/kg to female dogs. The pharmacokinetics of 15.4 mg/kg IM FASLODEX™ (fulvestrant injection, 250 mg/5 mL) were also determined and used for comparison to the three prototype pharmaceutical compositions. The 15.4 mg/kg dose used in this study is the canine equivalent, in mg/m2, of the maximum dose (500 mg) for human use and was scaled for use in canine by dividing the dose (based on a 60 kg human) by a canine species conversion factor of 0.54.
  • Twenty-four non-naïve female beagle dogs were used in the study. The animals weighed between approximately 5-10 kg. Animal welfare for this study was in compliance with the U.S. Department of Agriculture's (USDA) Animal Welfare Act (9 Code of Federal Regulations (CFR) Parts 1, 2 and 3). The Guide for the Care and Use of Laboratory Animals, Institute of Laboratory Animal Resources, National Academy Press, Washington, D.C., was followed. The facility maintained an Animal Welfare Assurance statement with the National Institutes of Health, Office of Laboratory Animal Welfare.
  • The FASLODEX™ test articles contained a small molecule that was used as received and no adjustment was made for purity, salt correction, etc. The FASLODEX™ test articles were gently agitated prior to dispensing and dose delivery. Pharmaceutical Composition B, Pharmaceutical Composition E, Pharmaceutical Composition I, Pharmaceutical Composition J, Pharmaceutical Composition K, and Pharmaceutical Composition L were stored at room temperature and protected from light prior to use, and gently agitated prior to dispensing and dose delivery.
  • The animals were not fasted prior to dosing. Each animal received a single intramuscular (IM) dose of only one of the appropriate test article pharmaceutical compositions as outlined in the following study design table, Table 30. IM doses were administered with a 20 G needle via bolus injection into the same large muscle mass (using the Z-track injection technique) in the left hind limb of each animal. Attempts were made for consistent injections between animals [selection of the dose site (muscle), depth, etc.]. The hair was clipped from the injection site prior to dosing. The injection site was marked following dosing and remarked as necessary throughout the study. Specifications for all dose delivery were recorded and reported in the study report [including, but not limited to needle gauge/length, syringe size/barrel type with manufacturer and part number, estimated injection depth into the muscle, approximate duration required to administer the injection; any substantial resistance (either flow through the syringe/needle and/or into the muscle during administration)] was documented.
  • TABLE 30
    Pharmaceutical
    Composition
    Fulvestrant Dose Dose
    No. of Concentration Level Volume
    Group Test Article Females (mg/mL) (mg/kg) (mL/kg)
    1 FASLODEX ™ 3 50 15.4 0.308
    2 Pharmaceutical 3 50 15.4 0.308
    Composition B
    3 Pharmaceutical 3 50 15.4 0.308
    Composition E
    4 FASLODEX ™ 3 50 15.4 0.308
    5 Pharmaceutical 3 50 15.4 0.308
    Composition I
    6 Pharmaceutical 3 50 15.4 0.308
    Composition J
    7 Pharmaceutical 3 50 15.4 0.308
    Composition K
    8 Pharmaceutical 3 100 15.4 0.154
    Composition L
  • All animals were observed at least twice a day for morbidity, mortality, injury, and availability of food and water. Any animals in poor health were identified for further monitoring and possible euthanasia.
  • Blood samples were collected at various time intervals to measure the blood plasma concentration of fulvestrant. Blood samples for Groups 1-3 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 456 (on Day 20), 528 (on Day 23), 600 (on Day 26), and 672 (on Day 29) hours postdose. Blood samples for Groups 4-8 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 72 (on Day 4), 96 (on Day 5), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 528 (on Day 23), and 672 (on Day 29) hours postdose.
  • Whole venous blood samples of approximately 2 mL each were collected from a peripheral vein of all animals for determination of fulvestrant exposure. Blood was collected with sodium heparin anticoagulant (glass tube, no gel separator). All blood samples were placed on wet ice following collection until centrifuged. Blood was centrifuged at 3500 rpm for 7 minutes at 2 to 8° C. Plasma (minimum of 0.8 mL volume) was separated from blood cells within 0.75 hours of blood collection and frozen. Plasma samples were initially placed on dry ice prior to being stored in the appropriate freezer (−60 to −90° C.). Samples were shipped on dry ice for bioanalytical analysis.
  • A model independent method was used to determine Cmax and AUC values from fulvestrant plasma concentration-time data. Results are shown in Tables 31-37 and FIGS. 1B, 2A, 2C, and 3. Table 31 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition). An “Assay %” is shown for the fulvestrant pharmaceutical compositions of the present disclosure used in the study. The “Assay %” represents the percentage equivalence of the particular pharmaceutical composition in comparison to the FASLODEX™ label claim fulvestrant concentration, with “Assay %” values determined via HPLC, measurement samples taken pre- and post-dose, with one value selected for normalization. The data in Tables 32-37 are normalized using the “Assay %” values to compare PK results based upon the actual mg/kg of fulvestrant administered, assuming linear scaling. FIGS. 1B, 2A, 2C, and 3 depict graphs of the dose normalized fulvestrant mean plasma concentrations.
  • TABLE 31
    PK parameters based upon the nominal dose 15.4 mg/kg
    Assay (%) Assay (%)
    (measured (measured
    Variant pre-dose) post-dose)
    (Geometric Mean of AUC0-14 d AUC0-28 d Starred value used for
    n as indicated) Cmax (ng/mL) (hr * ng/mL) (hr * ng/mL) normalization
    Faslodex 35.0 7015 8917
    (LW466, n = 9)
    Faslodex 45.7 7666 9306
    (MB122, n = 6)
    Faslodex 32.0 7177 9018
    (MB948, n = 3)
    Faslodex 36.9 8149 9817
    (MC949, n = 4)
    Faslodex 37.5 7408 9195
    (All, n = 22)
    B (n = 3) 8.7 1930 3250 100.0* 138.9
    E (n = 3) 29.1 5750 8380 93.4* 91.8
    I (n = 3) 41.5 8840 12300 95.6* 99.6
    J (n = 3) 44.8 5750 8100 87.6* 93.6
    K (n = 3) 69.7 7540 9630 84.6* 91.2
    L (n = 3) 63.9 8430 11000 94.1* 97.3
    L3F (n = 3) 22.6 5140 7130 83.8* 79.0
    L6 (n = 3) 24.7 6050 9360 113.9* 113.9
    F003a (n = 3) 27.7 5860 8610 95.0* 97.1
    F003b (n = 3) 32.5 7210 9650 96.3* 97.4
    F003e (n = 3) 28.5 6400 9080 100.6* 100.5
    F004a (n = 3) 31.7 4310 6190 99.8* 100.4
    F003k2 (n = 4) 33 3910 5960 97.8* 100.1
    F003k3 (n = 4) 26.8 5430 7060 99.6* 100.7
    F005a2 (L, n = 3) 19.7 4370 6840 93.2*
    F003l (L, n = 3) 25.1 5510 8680 99.2*
    F005b1 (n = 3) 49.7 9420 12100 100.2*
    F015a1 (n = 3) 63.6 4750 7120 103.8*
    F015a3 (L, n = 3) 34.4 3850 6000 98.6*
    F005d1 (n = 3) 37.9 7180 9910 100.5* 99.2
    F005c3 (L, n = 3) 17.7 3680 5820 95.2*
  • TABLE 32
    PK parameters normalized against the actual dose
    Cmax
    Variant (Geometric (ng/mL AUC0-14 d AUC0-28 d
    Mean of n as per (hr * ng/mL per (hr * ng/mL per
    indicated) mg/kg) mg/kg) mg/kg)
    Faslodex (LW466, 2.3 456 579
    n = 9)
    Faslodex (MB122, 3.0 498 604
    n = 6)
    Faslodex (MB948, 2.1 466 586
    n = 3)
    Faslodex (MC949, 2.4 529 637
    n = 4)
    Faslodex 2.4 481 597
    (All, n = 22)
    B (n = 3) 0.6 125 211
    E (n = 3) 2.0 400 583
    I (n = 3) 2.8 600 835
    J (n = 3) 3.3 426 600
    K (n = 3) 5.3 579 739
    L (n = 3) 4.4 582 759
    L3F (n = 3) 1.8 398 552
    L6 (n = 3) 1.4 345 534
    F003a (n = 3) 1.9 401 589
    F003b (n = 3) 2.2 486 651
    F003e (n = 3) 1.8 413 586
    F004a (n = 3) 2.1 280 403
    F003k2 (n = 4) 2.2 260 396
    F003k3 (n = 4) 1.7 354 460
    F005a2 (L, n = 3) 1.4 304 477
    F003l (L, n = 3) 1.6 361 568
    F005b1 (n = 3) 3.2 610 784
    F015a1 (n = 3) 4.0 297 445
    F015a3 (L, n = 3) 2.3 254 395
    F005d1 (n = 3) 2.4 464 640
    F005c3 (L, n = 3) 1.2 251 397
  • TABLE 33
    Comparison of normalized PK parameters against all Faslodex lots
    Variant Cmax
    (Geometric Mean of ratio to AUC0-14 d ratio AUC0-28 d ratio
    n as indicated) Faslodex (%) to Faslodex (%) to Faslodex (%)
    Faslodex (LW466, 93 95 97
    n = 9)
    Faslodex (MB122, 122 103 101
    n = 6)
    Faslodex (MB948, 85 97 98
    n = 3)
    Faslodex (MC949, 98 110 107
    n = 4)
    Faslodex 100 100 100
    (All, n = 22)
    B (n = 3) 23 26 35
    E (n = 3) 83 83 98
    I (n = 3) 116 125 140
    J (n = 3) 136 89 101
    K (n = 3) 219 120 124
    L (n = 3) 181 121 127
    L3F (n = 3) 72 83 93
    L6 (n = 3) 58 72 89
    F003a (n = 3) 78 83 99
    F003b (n = 3) 90 101 109
    F003e (n = 3) 75 86 98
    F004a (n = 3) 85 58 67
    F003k2 (n = 4) 90 54 66
    F003k3 (n = 4) 72 74 77
    F005a2 (L, n = 3) 56 63 80
    F003l (L, n = 3) 67 75 95
    F005b1 (n = 3) 132 127 131
    F015a1 (n = 3) 163 62 75
    F015a3 (L, n = 3) 93 53 66
    F005d1 (n = 3) 100 96 107
    F005c3 (L, n = 3) 50 52 66
  • TABLE 34
    Comparison of normalized PK parameters against Faslodex lot
    LW466
    Cmax
    Variant ratio to
    (Geometric Mean of n Faslodex AUC0-14 d ratio AUC0-28 d ratio
    as indicated) (%) to Faslodex (%) to Faslodex (%)
    Faslodex (LW466, 100 100 100
    n = 9)
    Faslodex (MB122, 131 109 104
    n = 6)
    Faslodex (MB948, 92 102 101
    n = 3)
    Faslodex (MC949, 106 116 110
    n = 4)
    Faslodex 107 106 103
    (All, n = 22)
    B (n = 3) 25 28 36
    E (n = 3) 89 88 101
    I (n = 3) 124 132 144
    J (n = 3) 146 94 104
    K (n = 3) 236 127 128
    L (n = 3) 194 128 131
    L3F (n = 3) 77 87 95
    L6 (n = 3) 62 76 92
    F003a (n = 3) 83 88 102
    F003b (n = 3) 97 107 112
    F003e (n = 3) 81 91 101
    F004a (n = 3) 91 62 70
    F003k2 (n = 4) 97 57 68
    F003k3 (n = 4) 77 78 79
    F005a2 (L, n = 3) 60 67 82
    F003l (L, n = 3) 72 79 98
    F005b1 (n = 3) 142 134 135
    F015a1 (n = 3) 175 65 77
    F015a3 (L, n = 3) 100 56 68
    F005d1 (n = 3) 108 102 111
    F005c3 (L, n = 3) 53 55 69
  • TABLE 35
    Comparison of normalized PK parameters against Faslodex lot
    MB122
    Cmax
    Variant ratio to
    (Geometric Mean of n Faslodex AUC0-14 d ratio AUC0-28 d ratio
    as indicated) (%) to Faslodex (%) to Faslodex (%)
    Faslodex (LW466, 76 92 96
    n = 9)
    Faslodex (MB122, 100 100 100
    n = 6)
    Faslodex (MB948, 70 94 97
    n = 3)
    Faslodex (MC949, 81 106 105
    n = 4)
    Faslodex 82 97 99
    (All, n = 22)
    B (n = 3) 19 25 35
    E (n = 3) 68 80 96
    I (n = 3) 95 121 138
    J (n = 3) 112 86 99
    K (n = 3) 180 116 122
    L (n = 3) 148 117 126
    L3F (n = 3) 59 80 91
    L6 (n = 3) 47 69 88
    F003a (n = 3) 64 80 97
    F003b (n = 3) 74 98 108
    F003e (n = 3) 62 83 97
    F004a (n = 3) 69 56 67
    F003k2 (n = 4) 74 52 65
    F003k3 (n = 4) 59 71 76
    F005a2 (L, n = 3) 46 61 79
    F003l (L, n = 3) 55 72 94
    F005b1 (n = 3) 108 123 130
    F015a1 (n = 3) 134 60 74
    F015a3 (L, n = 3) 76 51 65
    F005d1 (n = 3) 82 93 106
    F005c3 (L, n = 3) 41 50 66
  • TABLE 36
    Comparison of normalized PK parameters against Faslodex lot
    MB948
    Cmax
    Variant ratio to
    (Geometric Mean of n Faslodex AUC0-14 d ratio AUC0-28 d ratio
    as indicated) (%) to Faslodex (%) to Faslodex (%)
    Faslodex (LW466, 109 98 99
    n = 9)
    Faslodex (MB122, 143 107 103
    n = 6)
    Faslodex (MB948, 100 100 100
    n = 3)
    Faslodex (MC949, 115 114 109
    n = 4)
    Faslodex 117 103 102
    (All, n = 22)
    B (n = 3) 27 27 36
    E (n = 3) 97 86 99
    I (n = 3) 136 129 143
    J (n = 3) 160 91 103
    K (n = 3) 257 124 126
    L (n = 3) 212 125 130
    L3F (n = 3) 84 85 94
    L6 (n = 3) 68 74 91
    F003a (n = 3) 91 86 100
    F003b (n = 3) 105 104 111
    F003e (n = 3) 89 89 100
    F004a (n = 3) 99 60 69
    F003k2 (n = 4) 105 56 68
    F003k3 (n = 4) 84 76 79
    F005a2 (L, n = 3) 66 65 81
    F003l (L, n = 3) 79 77 97
    F005b1 (n = 3) 155 131 134
    F015a1 (n = 3) 191 64 76
    F015a3 (L, n = 3) 109 54 67
    F005d1 (n = 3) 118 100 109
    F005c3 (L, n = 3) 58 54 68
  • TABLE 37
    Comparison of normalized PK parameters against Faslodex lot
    MB949
    Cmax
    Variant ratio to
    (Geometric Mean of n Faslodex AUC0-14 d ratio AUC0-28 d ratio
    as indicated) (%) to Faslodex (%) to Faslodex (%)
    Faslodex (LW466, 95 86 91
    n = 9)
    Faslodex (MB122, 124 94 95
    n = 6)
    Faslodex (MB948, 87 88 92
    n = 3)
    Faslodex (MC949, 100 100 100
    n = 4)
    Faslodex 102 91 94
    (All, n = 22)
    B (n = 3) 24 24 33
    E (n = 3) 84 76 91
    I (n = 3) 118 113 131
    J (n = 3) 139 81 94
    K (n = 3) 223 109 116
    L (n = 3) 184 110 119
    L3F (n = 3) 73 75 87
    L6 (n = 3) 59 65 84
    F003a (n = 3) 79 76 92
    F003b (n = 3) 91 92 102
    F003e (n = 3) 77 78 92
    F004a (n = 3) 86 53 63
    F003k2 (n = 4) 91 49 62
    F003k3 (n = 4) 73 67 72
    F005a2 (L, n = 3) 57 58 75
    F003l (L, n = 3) 69 68 89
    F005b1 (n = 3) 134 115 123
    F015a1 (n = 3) 166 56 70
    F015a3 (L, n = 3) 94 48 62
    F005d1 (n = 3) 102 88 100
    F005c3 (L, n = 3) 50 47 62
  • Example: Pharmacokinetic Study 2 of Intramuscular Administration to Female Dogs
  • Fulvestrant pharmaceutical compositions L3F and L6 were prepared as described elsewhere herein and in the Figures. A preclinical study was performed to determine the pharmacokinetics of the pharmaceutical compositions following a single intramuscular administration of 15.4 mg/kg to female dogs. The pharmacokinetics of 15.4 mg/kg IM FASLODEX™ (fulvestrant injection, 250 mg/5 mL) were also determined and used for comparison to the three prototype pharmaceutical compositions. The 15.4 mg/kg dose used in this study is the canine equivalent, in mg/m2, of the maximum dose (500 mg) for human use and was scaled for use in canine by dividing the dose (based on a 60 kg human) by a canine species conversion factor of 0.54.
  • Nine non-naïve female beagle dogs were used in the study. The animals weighed between approximately 5-13 kg. Animal welfare for this study was in compliance with the U.S. Department of Agriculture's (USDA) Animal Welfare Act (9 Code of Federal Regulations (CFR) Parts 1, 2 and 3). The Guide for the Care and Use of Laboratory Animals, Institute of Laboratory Animal Resources, National Academy Press, Washington, D.C., was followed. The facility maintained an Animal Welfare Assurance statement with the National Institutes of Health, Office of Laboratory Animal Welfare.
  • The FASLODEX™ test articles contained a small molecule that was used as received and no adjustment was made for purity, salt correction, etc. The FASLODEX™ test articles were gently agitated prior to dispensing and dose delivery. Pharmaceutical Compositions L3F and L6 were stored at room temperature and protected from light prior to use, and gently agitated prior to dispensing and dose delivery.
  • TABLE 38
    Fulvestrant
    Dosage Conc. Dose Volume
    Dose Group Test Article (mg/kg) (mg/mL) (mg/kg)
    1 FASLODEX ™ 15.4 50 0.308
    2 Pharmaceutical 15.4 100 0.154
    Composition
    L3F
    3 Pharmaceutical 15.4 100 0.154
    Composition
    L6
  • The animals were not fasted prior to dosing. Each animal received a single intramuscular (IM) dose of only one of the appropriate test article pharmaceutical compositions as outlined in the following study design table, Table 38. IM doses were administered with a 20 G needle via bolus injection into the same large muscle mass (using the Z-track injection technique) in the left hind limb of each animal. Attempts were made for consistent injections between animals [selection of the dose site (muscle), depth, etc.]. The hair was clipped from the injection site prior to dosing. The injection site was marked following dosing and remarked as necessary throughout the study. Specifications for all dose delivery were recorded and reported in the study report [including, but not limited to needle gauge/length, syringe size/barrel type with manufacturer and part number, estimated injection depth into the muscle, approximate duration required to administer the injection; any substantial resistance (either flow through the syringe/needle and/or into the muscle during administration)] was documented.
  • All animals were observed at least twice a day for morbidity, mortality, injury, and availability of food and water. Any animals in poor health were identified for further monitoring and possible euthanasia.
  • Whole venous blood samples of approximately 2 mL each were collected from a peripheral vein of all animals for determination of fulvestrant exposure. Samples were collected at the following target timepoints; predose, 0.25, 0.5, 1, 2, 4, 8, 12, 24 (Day 2), 48 (Day 3), 72 (Day 4), 96 (Day 5), 120 (Day 6), 192 (Day 9), 264 (Day 12), 336 (Day 15), 384 (Day 17), 528 (Day 23), and 672 (Day 29) hours after administration. Blood was collected with sodium heparin anticoagulant (glass tube, no gel separator). All blood samples were placed on wet ice following collection until centrifuged. Blood was centrifuged at 3500 rpm for 7 minutes at 2 to 8° C. Plasma (minimum of 0.8 mL volume) was separated from blood cells within 0.75 hours of blood collection and frozen. Plasma samples were initially placed on dry ice prior to being stored in the appropriate freezer (−60 to −90° C.). Samples were shipped on dry ice for bioanalytical analysis.
  • A model independent method was used to determine Cmax and AUC values from fulvestrant plasma concentration-time data. Results are shown in Tables 31-37 and FIGS. 2A and 2C. Table 31 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition). An “Assay %” is shown for the fulvestrant pharmaceutical compositions of the present disclosure used in the study. The “Assay %” represents the percentage equivalence of the particular pharmaceutical composition in comparison to the FASLODEX™ label claim fulvestrant concentration, with “Assay %” values determined via HPLC, measurement samples taken pre- and post-dose, with one value selected for normalization. The data in Tables 32-37 are normalized using the “Assay %” values to compare PK results based upon the actual mg/kg of fulvestrant administered, assuming linear scaling. FIGS. 2A and 2C depict graphs of the dose normalized fulvestrant mean plasma concentrations.
  • Example: Pharmacokinetic Study 3 of Intramuscular and Intravenous Administration to Female Dogs
  • Fulvestrant formulations F003a, F003b, F004a, F003e, F003k2, F003k3, F005a2, F0031, F005b1, F015a1, F005d1, F005c3, F015a3, F005g5, Del-1S, Del-2S, F005H3, Lot 15, Lot 26, Lot 27, Lot 28, Lot 42, Lot 43, Lot 45, Lot 46, Lot 47, and Lot 48 were prepared as described elsewhere herein and in the Figures. In some instances, the formulations were prepared using different processes as indicated, referred to by an alphanumeric process identifier, such as “Process A1,” “Process A2,” and the like. A fulvestrant pharmaceutical composition for intravenous injection, referred to as batch FV-004/15M, was prepared as described below. A preclinical study was performed to determine the pharmacokinetics of the pharmaceutical compositions following a single intramuscular (IM) administration of 15.4 mg/kg to female dogs. The pharmacokinetics of 15.4 mg/kg IM FASLODEX™ (fulvestrant injection, 250 mg/5 mL) were also determined and used for comparison to the three prototype pharmaceutical compositions. The 15.4 mg/kg dose used in this study is the canine equivalent, in mg/m2, of the maximum dose (500 mg) for human use and was scaled for use in canine by dividing the dose (based on a 60 kg human) by a canine species conversion factor of 0.54.
  • One hundred fifty-six non-naïve female beagle dogs of body weight range of 5.65 to 11.40 kilograms were used in the study and assigned to Groups 1-48, as shown in Table 39 below.
  • Animal welfare for this study was in compliance with the U.S. Department of Agriculture's (USDA) Animal Welfare Act (9 Code of Federal Regulations (CFR) Parts 1, 2 and 3). The Guide for the Care and Use of Laboratory Animals, Institute of Laboratory Animal Resources, National Academy Press, Washington, D.C., was followed. The facility maintained an Animal Welfare Assurance statement with the National Institutes of Health, Office of Laboratory Animal Welfare.
  • TABLE 39
    Fulvestrant Dose Dose
    Group Number of Dose Concentration Level Volume
    Number Test Article Females Route (mg/mL) (mg/kg) (mL/kg)
    1 Faslodex (lot 3 IM 50 15.4 0.308
    LW466)
    2 Faslodex (lot 3 IM 50 15.4 0.308
    MB122)
    3 Faslodex (lot 3 IM 50 15.4 0.308
    MB948)
    4 Formulation F003a 3 IM 100 15.4 0.154
    5 Formulation F003b 3 IM 100 15.4 0.154
    6 Formulation F004a 3 IM 100 15.4 0.154
    7 Formulation F003e 3 IM 100 15.4 0.154
    8 Formulation F003k2 4 IM 100 15.4 0.154
    9 Formulation F003k3 4 IM 100 15.4 0.154
    10 Faslodex (lot 4 IM 50 15.4 0.308
    MC949)
    11 Formulation F005a2 3 IM 100 15.4 0.154
    12 Formulation F003l 4 IM 100 15.4 0.154
    13 Formulation F005b1 3 IM 100 15.4 0.154
    14 Formulation F015a1 3 IM 100 15.4 0.154
    15 Formulation F005d1 3 IM 100 15.4 0.154
    16 Formulation F005c3 4 IM 100 15.4 0.154
    17 Formulation F015a3 3 IM 100 15.4 0.154
    18 Fulvestrant 4 IV 20 2.5 0.125
    (batch FV-004/15M)
    19 Formulation F005g5 4 IM 100 15.4 0.154
    20 Formulation Del-1S 4 IM 100 15.4 0.154
    21 Formulation Del-2S 4 IM 100 15.4 0.154
    22 Formulation F005H3 3 IM 100 15.4 0.154
    23 Lot 15, by Process 4 IM 100 15.4 0.154
    E1
    24 Lot 15, by Process 3 IM 100 15.4 0.154
    E2
    25 Lot 26, by Process F1 3 IM 100 15.4 0.154
    26 Lot 26, by Process F2 3 IM 100 15.4 0.154
    27 Lot 26, by Process F3 3 IM 100 15.4 0.154
    28 Lot 26, by Process F4 3 IM 100 15.4 0.154
    29 Lot 26, by Process J1 3 IM 100 15.4 0.154
    30 Lot 26, by Process J2 3 IM 100 15.4 0.154
    31 Lot 26, by Process J3 3 IM 100 15.4 0.154
    32 Lot 26, by Process J4 3 IM 100 15.4 0.154
    33 Lot 42, by Process G1 3 IM 100 15.4 0.154
    34 Lot 42, by Process G2 3 IM 100 15.4 0.154
    35 Lot 43, by Process H1 3 IM 100 15.4 0.154
    36 Lot 43, by Process H2 3 IM 100 15.4 0.154
    37 Lot 27, by Process A1 3 IM 100 15.4 0.154
    38 Lot 27, by Process A2 3 IM 100 15.4 0.154
    39 Lot 27, by Process A3 3 IM 100 15.4 0.154
    40 Lot 27, by Process A4 3 IM 100 15.4 0.154
    41 Lot 28, by Process B1 3 IM 100 15.4 0.154
    42 Lot 28, by Process B2 3 IM 100 15.4 0.154
    43 Lot 28, by Process B3 3 IM 100 15.4 0.154
    44 Lot 28, by Process B4 3 IM 100 15.4 0.154
    45 Lot 45, by Process C1 3 IM 100 15.4 0.154
    46 Lot 46 3 IM 100 15.4 0.154
    47 Lot 47 3 IM 100 15.4 0.154
    48 Lot 48 3 IM 100 15.4 0.154
  • Pharmaceutical Compositions F003a, F003b, F004a, F003e, F003k2, F003k3, F005a2, F0031, F005b1, F015a1, F005d1, F005c3, F015a3, FV-004/15M, Del-1S, and Del-2S were stored at room temperature and protected from light prior to use, and gently agitated via inversion prior to dispensing and dose delivery. If visible clumps of material were seen on the vial inside wall or inner seal of the vial cap after 3 minutes of inversion, the tightly capped vial was vortexed at moderate intensity and unlimited duration until clumps were not visible. FASLODEX™ was supplied as two 5-mL clear neutral glass (Type 1) syringe barrels, each containing a 250 mg/5 mL (50 mg/mL) solution for intramuscular injection. Upon receipt, FASLODEX™ was stored refrigerated (2°-8° C.) and protected from light. The procedure to prepare and administer FASLODEX™ was performed as outlined in the manufacturer's prescribing information.
  • Lyophilized pharmaceutical compositions of formulations F005g5, F005H3, F015a3, Lot 15, Lot 26, Lot 27, Lot 28, Lot 42, Lot 43, Lot 45, Lot 46, Lot 47, and Lot 48 were reconstituted prior to dosing. Using an empty syringe and hypodermic needle, about 5 mL of air was withdrawn from the head space of the vial (above the lyophilized contents) via the septum and the syringe and needle were discarded. Using a sterile syringe and hypodermic needle, 5 mL of sterile water for injection, USP were added to the vial by piercing the septum and injecting a stream of water slowly around the inner wall of the neck of the vial to wet the lyophilized cake without touching any of the vial contents. The needle was removed from the septum and the vial was gently swirled until a visually homogeneous particulate suspension formed, with no visual clumps or material attached to the inside wall of the vial. If a homogeneous suspension was not formed after 5 minutes of swirling, the vial was vortexed until a homogeneous suspension was formed. The vials were not shaken to avoid generating bubbles or excessive foam.
  • Group 18 was administered an intravenous batch of fulvestrant (batch FV-004/15M) prepared as follows by (% w/v): 2% fulvestrant, 10% EtOH, 79% propylene glycol, 1% Poloxamer 407, 8% Water for Injection, USP. Fulvestrant API was stored at 2-8° C., protected from light. Care was taken to protect the API from humidity during weighing. Fulvestrant powder was dissolved in ethanol and swirled and vortexed as needed to dissolve completely. Propylene glycol was added and mixed to dissolve to a clear liquid state. Poloxamer 407 was dissolved in water for injection, USP in a separate vessel and mixed, vortexed, and sonicated as needed to dissolve into to a clear liquid state. The Poloxamer 407 in water for injection solution was added to the fulvestrant/ethanol solution. Propylene glycol was added and the solution was mixed and vortexed to achieve a clear liquid. The solution was filtered through a 0.2 μm or 0.22 μm syringe (to ensure that all liquid volume was usable) tip filter (PVDF) into a clear glass vessel. The prepared formulation as stored at room temperature for up to four hours prior to dosing under protection from light exposure. Intravenous doses were administered via the cephalic (or other suitable) vein as a slow injection over approximately 1 minute. Batch FV-004/15M was administered intravenously at a dose of 2.5 mg/kg.
  • The animals were not fasted prior to dosing. Each animal in Groups 4-9, 11-17, and 19-48 received a single intramuscular (IM) dose of only one of the appropriate test article pharmaceutical compositions as outlined in Table 39. IM doses were administered with a 20 G needle via bolus injection into the same large muscle mass (using the Z-track injection technique) in the left hind limb of each animal. Attempts were made for consistent injections between animals [selection of the dose site (muscle), depth, etc.]. The hair was clipped from the injection site prior to dosing. The injection site was marked following dosing and remarked as necessary throughout the study. Specifications for all dose delivery were recorded and reported in the study report [including, but not limited to needle gauge/length, syringe size/barrel type with manufacturer and part number, estimated injection depth into the muscle, approximate duration required to administer the injection; any substantial resistance (either flow through the syringe/needle and/or into the muscle during administration)] was documented. Animals in Groups 13-17 and 19-48 were administered 1 tablet or capsule (25 mg) of (PO) diphenhydramine at approximately 1 hour prior to dosing.
  • All animals were observed at least twice a day for morbidity, mortality, injury, and availability of food and water. Any animals in poor health were identified for further monitoring and possible euthanasia.
  • Whole venous blood samples of approximately 2 mL each were collected from a peripheral vein of all animals for determination of fulvestrant exposure. Blood samples for Groups 1-7 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 72 (on Day 4), 96 (on Day 5), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 528 (on Day 23), and 672 (on Day 29) hours postdose. Blood samples for Groups 8-14 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, and 12 (on Day 1); and 24 (on Day 2), 48 (on Day 3), 72 (on Day 4), 96 (on Day 5), 120 (on Day 6), 192 (on Day 9), 264 (on Day 12), 336 (on Day 15), 384 (on Day 17), 456 (on Day 20), 528 (on Day 23), 600 (on Day 26), 672 (on Day 29), 696 (Day 30), 768 (Day 33), 816 (Day 35), 864 (Day 37), 936 (Day 40) and 1008 (Day 43) hours post-dose. Blood samples for Groups 15-17 and 19-21 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, 12 (Day 1), 24 (Day 2), 48 (Day 3), 72 (Day 4), 96 (Day 5), 120 (Day 6), 192 (Day 9), 264 (Day 12), 336 (Day 15), 384 (Day 17), 456 (Day 20), 528 (Day 23), 600 (Day 26), 672 (Day 29), 696 (Day 30), 768 (Day 33), 816 (Day 35), 864 (Day 37), 936 (Day 40), 1008 (Day 43), and 1176 (Day 50) hours postdose. Blood samples for Group 18 were collected predose and at 0.033 (2 minutes), 0.1 (6 minutes), 0.13 (8 minutes), 0.27 (16 minutes), 0.52 (31 minutes), 0.77 (46 minutes), 1, 2, 3, 4, 6, 8, 10, and 12 hours postdose on Day 1 and at 24 (on Day 2), 30 (on Day 2), 48 (on Day 3) and 72 (on Day 4) hours postdose, with postdose measurements from the start of dose administration, which took about 1 minute to complete. Blood samples for Groups 22-48 were collected predose and at 0.25, 0.5, 1, 2, 4, 8, 12 (Day 1), 24 (Day 2), 48 (Day 3), 72 (Day 4), 96 (Day 5), 120 (Day 6), 144 (Day 7), 168 (Day 8), 192 (Day 9), 216 (Day 10), 240 (Day 11), 264 (Day 12), 336 (Day 15), 384 (Day 17), 456 (Day 20), 528 (Day 23), 600 (Day 26), 672 (Day 29), 696 (Day 30), 768 (Day 33), 816 (Day 35), 864 (Day 37), 936 (Day 40), 1008 (Day 43), and 1176 (Day 50) hours postdose. Blood was collected with sodium heparin anticoagulant (glass tube, no gel separator). All blood samples were placed on wet ice following collection until centrifuged. Blood was centrifuged at 3500 rpm for 7 minutes at 2 to 8° C. Plasma (minimum of 0.8 mL volume) was separated from blood cells within 0.75 hours of blood collection and frozen. Plasma samples were initially placed on dry ice prior to being stored in the appropriate freezer (−60 to −90° C.). Samples were shipped on dry ice for bioanalytical analysis to determine absolute ng/mL fulvestrant in the plasma.
  • TABLE 40
    PK parameters compared to
    Faslodex lots based upon the
    nominal dose 15.4 mg/kg
    AUC0-14 d AUC0-28 d
    Formulation PK parameters based upon the Cmax ratio ratio
    (Geometric Mean, nominal dose 15.4 mg/kg ratio to to to
    n = 3 unless indicated Cmax AUC0-14 d AUC0-28 d Faslodex Faslodex Faslodex
    otherwise) (ng/mL) (hr * ng/mL) (hr * ng/mL) (%) (%) (%)
    Faslodex (All, n = 22) 37.5 7408 9195 100 100 100
    F005g5 32.1 6950 10400 86 94 113
    Lot 15, by Process E1 20.1 3890 5500 54 53 60
    Lot 15, by Process E2 20.3 4190 6230 54 57 68
    Lot 26, by Process F1 19.5 4150 6650 52 56 72
    Lot 26, by Process F2 30.5 5510 7850 81 74 85
    Lot 26, by Process F3 25.7 5350 8260 68 72 90
    Lot 26, by Process F4 30.8 7210 10200 82 97 111
    Lot 26, by Process J1 25.2 5490 7910 67 74 86
    Lot 26, by Process J2 20.8 4640 7190 55 63 78
    Lot 26, by Process J3 21.1 4720 7310 56 64 80
    Lot 26, by Process J4 21.3 4320 5900 57 58 64
    Lot 42, by Process G1 28.1 4640 7410 75 63 81
    Lot 42, by Process G2 33.8 6110 8800 90 82 96
    Lot 43, by Process H1 25.0 5190 7500 67 70 82
    Lot 43, by Process H2 22.2 4430 6250 59 60 68
    Lot 27, by Process A1 92.6 6150 8710 247 83 95
    Lot 27, by Process A2 27.9 6240 9170 74 84 100
    Lot 27, by Process A3 26.3 5100 7680 70 69 84
    Lot 27, by Process A4 29.1 6030 8620 78 81 94
    Lot 28, by Process B1 21.9 3950 6230 58 53 68
    Lot 28, by Process B2 36.3 7010 9870 97 95 107
    Lot 28, by Process B3 31.6 6880 9660 84 93 105
    Lot 28, by Process B4 31.0 6330 9000 83 85 98
    Lot 45, by Process C1 29.8 5320 8560 79 72 93
    Lot 46 30.2 5570 8240 80 75 90
    Lot 47 27.9 5230 8070 74 71 88
    Lot 48 26.3 4900 7410 70 66 81
  • A model independent method was used to determine Cmax and AUC values from fulvestrant plasma concentration-time data. Results are shown in Tables 31-37 and 40 and FIGS. 1A, 1B, 2A, 2B, 2C, 13, and 14. Table 31 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition). An “Assay %” is shown in Table 31 for the fulvestrant pharmaceutical compositions of the present disclosure used in the study. The “Assay %” represents the percentage equivalence of the particular pharmaceutical composition in comparison to the FASLODEX™ label claim fulvestrant concentration, with “Assay %” values determined via HPLC, measurement samples taken pre- and post-dose, with one value selected for normalization. The data in Tables 32-37 are normalized using the “Assay %” values to compare PK results based upon the actual mg/kg of fulvestrant administered, assuming linear scaling. FIGS. 1A, 1B, 2A, 2B, and 2C depict graphs of the dose normalized fulvestrant mean plasma concentrations. Table 40 shows the pharmacokinetic data from the 15.4 mg/kg dosages as nominally dosed (based on the target fulvestrant concentration for each pharmaceutical composition) in comparison to the geometric mean of all Faslodex lots tested (n=22). FIGS. 13 and 14 depict fulvestrant plasma measurements for administration of Faslodex Lot MB948 to three female dogs and administration of fulvestrant formulation Lot 27 processed by Process A2 to three female dogs (referred to in FIG. 14 as subjects 924, 925, and 926).
  • Example: Microscopic Imaging of Fulvestrant Particles in Suspensions
  • Some exemplary fulvestrant pharmaceutical compositions of the present disclosure were examined via optical and scanning electron microscopy. Suspensions of fulvestrant pharmaceutical compositions Variants B, E, I, J, K, L, L3F, L6, F003a, F003b, F004a, F003e, F00k2, and F003k3 were examined via optical microscopy. Optical microscopy was performed at 400× magnification with a polarized light filter using fully dispersed homogeneous suspension samples.
  • Example: Particle Size Distribution Characterization of Fulvestrant Pharmaceutical Compositions
  • Batches of fulvestrant pharmaceutical composition Lot 27, described elsewhere herein, were prepared by the methods of preparation 101 of FIG. 15. Samples were taken periodically during high shear mixing, prior to any high pressure homogenization steps. Some test samples, referred to as “Sample 1”, were taken after approximately five hours of high shear mixing and other test samples, referred to as “Sample 2”, were taken after approximately 13.7 hours to high shear mixing. Particle sizes of test samples were analyzed using optical microscopy with a Malvern Morphologi G3 apparatus for microscopy image capture and analysis. CE diameters were measured and number-weighted and volume-weighted particle size distribution parameters were determined as shown in Tables 41 and 42. The CE diameter ranges of measurement aliquots are also shown, with the lower range value of 0.54 microns representing the lower limit of detection for the apparatus setup. Test samples were analyzed with a Malvern Mastersizer 3000 apparatus for laser diffraction particle size characterization of LD diameters.
  • TABLE 41
    Volume-Weighted Distribution Parameters
    Circle
    CE CE Equivalent
    Dv Dv CE (CE)- LD LD LD
    (10) (50) Dv (90) diameter- Dv10 Dv50 Dv90
    Formulation (μm) (μm) (μm) range (μm) (μm) (μm) (μm)
    Lot 27 6.113 13.77 32.71 0.54-49.72 1.81 6.68 16.6
    (Sample 1)
    Lot 27 6.509 14.34 28.64 0.54-54.14 1.85 6.93 17.7
    (Sample 1)
    Lot 27 6.378 12.76 23.90 0.54-55.04
    (Sample 1)
    Lot 27 5.297 10.73 24.57
    (Sample 1)
    Lot 27 6.015 13.03 25.25
    (Sample 1)
    Lot 27 5.446 11.41 22.10
    (Sample 1)
    Lot 27 7.222 14.93 28.99
    (Sample 1)
    Lot 27 8.747 18.31 32.93
    (Sample 1)
    Lot 27 7.663 14.96 26.58
    (Sample 1)
    Lot 27 6.733 13.61 24.70 0.54-42.64 1.96 7.54 19.3
    (Sample 2)
    Lot 27 7.182 14.12 25.15 0.54-44.65
    (Sample 2)
    Lot 27 6.920 13.26 22.63 0.54-38.24
    (Sample 2)
    Lot 27 7.400 14.86 27.82
    (Sample 2)
    Lot 27 6.857 14.08 27.39
    (Sample 2)
    Lot 27 8.117 16.58 30.90
    (Sample 2)
  • TABLE 42
    Number-Weighted Distribution Parameters
    Circle
    Equivalent
    (CE)-
    CE Dn10 CE Dn50 CE Dn90 diameter-
    Formulation (μm) (μm) (μm) range (μm)
    Lot 27 (Sample 1) 1.10 3.92 8.91 0.54-49.72
    Lot 27 (Sample 1) 1.14 4.06 9.61 0.54-54.14
    Lot 27 (Sample 1) 0.95 4.16 9.85 0.54-55.04
    Lot 27 (Sample 2) 1.08 4.24 10.34 0.54-42.64
    Lot 27 (Sample 2) 0.85 3.71 10.52 0.54-44.65
    Lot 27 (Sample 2) 0.91 4.07 10.55 0.54-38.24
  • Example: Particle Size Distribution Characterization of Fulvestrant Pharmaceutical Compositions
  • Fulvestrant pharmaceutical compositions F005g5, Lot 27, Lot 28, and Lot 45, described elsewhere herein, were prepared by the methods of preparation shown schematically as process 101 of FIG. 15. Samples of each Lot were prepared using different processes as indicated in Tables 43 to 50 to achieve fulvestrant particle sizes and particle size distributions. The preparation processes are referred to with alphanumeric identifiers, such as “Process A1,” “Process A2,” and the like, with each process representing a set of fulvestrant-particle-size-reduction steps, as more fully described elsewhere herein, to achieve final fulvestrant particle size and particle size distributions as shown in the Tables 43 to 50. Test samples were evaluated for particle size both prior to drying via lyophilization, which are indicated as “(100× Suspensions)”, and after lyophilization and reconstitution, which are indicated as “(100× Reconstituted Suspensions)” in the Tables 43 to 50. Samples were analyzed with a Malvern Morphologi G3 apparatus for microscopy image capture and analysis. CE diameters were measured and volume-weighted particle size distribution parameters were determined. The CE diameter ranges of measurement samples are also shown, with the lower range value of 0.54 microns representing the lower limit of detection for the apparatus setup. Samples were analyzed with a Malvern Mastersizer 3000 apparatus for laser diffraction particle size characterization of LD diameters.
  • TABLE 43
    Volume-Weighted Distribution Parameters
    (100x Reconstituted Suspensions)
    Circle
    Equivalent
    CE CE CE (CE)-
    Dv10 Dv50 Dv90 diameter-
    Formulation Process (μm) (μm) (μm) range (μm)
    Lot 27 Process 6.606 16.14 36.36 0.54-59.46
    A1
    Process 7.031 31.23 84.50 0.54-107.10
    A2
    Process 6.725 24.83 53.11 0.54-84.16
    A3
    Process 6.790 43.74 98.32 0.54-105.94
    A4
    Lot 28 Process 5.333 17.64 41.18 0.54-53.33
    B1
    Process 4.863 12.71 42.29 0.54-75.19
    B2
    Process 5.376 15.85 49.64 0.54-75.62
    B3
    Process 4.144 25.86 82.19 0.54-107.08
    B4
    Lot 45 Process 8.904 35.32 66.41 0.54-85.05
    C1
    Process 8.135 34.79 56.77 0.54-81.50
    C2
  • TABLE 44
    Volume-Weighted Distribution Parameters
    (100x Reconstituted Suspensions)
    Circle
    Equivalent As-Is Sonicated
    CE CE CE (CE)- LD LD LD LD LD LD
    Dv10 Dv50 Dv90 diameter- Dv10 Dv50 Dv90 Dv10 Dv50 Dv90
    Formulation Process (μm) (μm) (μm) range (μm) (μm) (μm) (μm) (μm) (μm) (μm)
    Lot 45 Process 5.140 14.44 42.62 0.54-71.31 3.42 8.21 33.8 2.81 5.57 10.6
    C9
    Lot 45 Process 6.558 17.10 44.70 0.54-74.57 3.37 7.81 32.6 2.82 5.58 10.5
    C9
    Lot 45 Process 6.648 19.97 56.47 0.54-112.57 3.50 9.34 43.1 2.81 5.57 10.5
    C9
    Lot 45 Process 6.541 15.68 37.75 0.54-55.93 3.20 7.08 24.3 2.80 5.52 10.7
    C10
    Lot 45 Process 5.787 13.11 32.25 0.54-55.82 3.33 7.83 33.9 2.98 5.80 11.1
    C10
    Lot 45 Process 5.375 14.59 55.02 0.54-88.17 3.38 8.02 35.6 2.97 5.79 11.1
    C10
    F005g5 As 3.07 29.2 95.1 2.20 5.25 14.0
    shown
    in
    Table
    34
  • TABLE 45
    Volume-Weighted Distribution Parameters
    (100x Reconstituted Suspensions)
    Circle
    Equivalent
    CE (CE)-
    Dv10 CE Dv50 CE Dv90 diameter-
    Formulation Process (μm) (μm) (μm) range (μm)
    Lot 27 Process A2 4.547 11.10 33.19 0.54-55.75
    Lot 27 Process A2 4.290 10.15 23.66 0.54-49.72
    Lot 27 Process A2 5.908 12.98 25.55 0.54-42.63
    Lot 27 Process A2 6.075 12.81 23.11 0.54-47.71
    Lot 27 Process A2 7.022 15.59 30.94 0.54-69.41
    Lot 27 Process A2 5.399 12.22 34.36 0.54-51.56
    Lot 27 Process A13 4.89 11.84 32.47
    Lot 27 Process A13 6.46 15.33 32.47
    Lot 27 Process A13 4.93 11.90 31.23
    Lot 27 Process A13 5.54 14.17 50.26
    Lot 27 Process A13 4.19 10.16 32.22
    Lot 27 Process A13 5.91 13.71 33.81
    Lot 27 Process A13 5.10 13.26 45.32
    Lot 27 Process A13 4.61 11.47 39.79
    Lot 27 Process A13 4.79 12.50 46.19
    Lot 27 Process A13 4.71 10.33 22.64
    Lot 27 Process A13 5.66 12.66 29.72
    Lot 27 Process A13 4.96 11.62 37.81
    Lot 27 Process A13 4.22 12.46 42.59
    Lot 27 Process A13 5.80 19.69 57.80
    Lot 27 Process A13 5.12 11.53 28.37
    Lot 27 Process A13 3.84 9.28 32.42
    Lot 27 Process A13 4.64 10.32 25.28
    Lot 27 Process A13 4.86 11.69 32.44
  • TABLE 46
    Volume-Weighted Distribution Parameters
    (100x Reconstituted Suspensions)
    As-Is Sonicated
    LD LD LD LD LD
    Dv10 Dv50 Dv90 Dv10 LD Dv50 Dv90
    Formulation Process (μm) (μm) (μm) (μm) (μm) (μm)
    Lot 27 Process 2.03 8.41 36.0 1.20 3.93 7.97
    A13
    Lot 27 Process 1.96 7.83 35.0 1.14 3.69 7.57
    A13
    Lot 27 Process 2.08 9.14 41.3 1.20 3.92 7.94
    A13
    Lot 27 Process 2.10 9.65 40.3 1.15 3.66 7.53
    A13
    Lot 27 Process 1.67 6.10 26.5 1.08 3.42 7.19
    A13
    Lot 27 Process 1.70 5.71 21.6 1.17 3.80 7.78
    A13
    Lot 27 Process 2.53 15.8 48.4 1.27 4.07 8.72
    A13
    Lot 27 Process 2.01 12.6 57.6 1.01 3.04 6.85
    A13
    Lot 27 Process 1.62 6.34 29.5 0.96 2.76 5.89
    A13
  • TABLE 47
    Volume-Weighted Distribution Parameters
    (100x Suspensions)
    Circle
    Equivalent
    CE (CE)-
    Dv10 CE Dv50 CE Dv90 diameter-
    Formulation Process (μm) (μm) (μm) range (μm)
    Lot 27 Process A15 5.199 10.4 17.52 0.54-27.89
    Process A16 3.766 7.411 12.15 0.54-25.43
    Process A17 3.053 6.256 10.81 0.54-38.27
    Process A18 3.727 7.277 13.91 0.54-31.30
    Lot 28 Process B5 4.442 8.585 15.04 0.54-41.23
    Process B6 4.134 7.773 13.25 0.54-22.59
    Process B7 3.835 7.311 13.11 0.54-25.94
    Process B8 3.417 6.644 11.62 0.54-28.25
    Lot 45 Process C3 7.160 14.79 25.70 0.54-47.55
    Process C4 8.587 18.15 36.68 0.54-62.10
  • TABLE 48
    Volume-Weighted Distribution Parameters
    (100x Suspensions)
    As-Is Sonicated
    LD LD LD LD LD
    Dv10 Dv50 LD Dv90 Dv10 Dv50 Dv90
    Formulation Process (μm) (μm) (μm) (μm) (μm) (μm)
    Lot 27 Process 1.08 3.29 7.68 0.91 2.62 6.98
    A15
    Process 1.03 3.15 7.00 0.83 2.32 5.72
    A16
    Process 0.97 2.96 6.52 0.78 2.13 5.01
    A17
    Process 0.92 2.91 6.32 0.72 1.96 4.38
    A18
    Lot 45 Process 1.50 6.03 12.0 0.83 2.30 5.69
    C3
    Process 1.59 5.82 11.0 0.85 2.37 5.74
    C4
  • TABLE 49
    Volume-Weighted Distribution Parameters
    (100x Suspensions)
    Circle
    Equivalent As-Is Sonicated
    CE CE CE (CE)- LD LD LD LD LD LD
    Dv10 Dv50 Dv90 diameter- Dv10 Dv50 Dv90 Dv10 Dv50 Dv90
    Formulation Process (μm) (μm) (μm) range (μm) (μm) (μm) (μm) (μm) (μm) (μm)
    Lot 45 Process 12.320 29.88 57.24 0.54-90.58 1.51 5.40 10.7 0.80 2.11 4.64
    C3
    Lot 45 Process 7.454 17.37 40.29 0.54-76.61 1.71 6.36 12.5 0.82 2.26 5.20
    C4
    Lot 45 Process 12.130 27.59 50.08 0.54-87.69 1.73 6.38 12.7 0.79 2.15 4.77
    C5
    Lot 45 Process 8.193 17.89 35.05 0.54-54.50 1.63 5.85 11.7 0.79 2.13 4.68
    C5
    Lot 45 Process 8.690 19.36 35.53 0.54-64.77 1.70 6.12 12.0 0.77 2.05 4.45
    C5
    Lot 45 Process 8.218 17.00 46.80 0.54-80.21 1.46 4.92 9.59 1.36 4.71 9.55
    C6
    Lot 45 Process 6.455 14.94 38.47 0.54-65.55 1.27 4.21 8.61 0.77 2.11 4.66
    C7
    Lot 45 Process 7.795 17.20 31.60 0.54-45.03 1.55 5.37 10.7 0.77 2.14 4.81
    C8
    Lot 45 Process 9.257 20.16 39.47 0.54-84.26 1.64 5.64 11.2 0.78 2.19 4.99
    C8
    Lot 45 Process 10.050 21.35 36.43 0.54-59.43 1.59 5.55 11.1 0.76 2.10 4.66
    C8
  • TABLE 50
    Volume-Weighted Distribution Parameters
    (100x Suspensions)
    Circle
    Equivalent As-Is As-Is As-Is
    CE CE CE (CE)- LD LD LD
    Dv10 Dv50 Dv90 diameter- Dv10 Dv50 Dv90
    Formulation Process (μm) (μm) (μm) range (μm) (μm) (μm) (μm)
    Lot 27 Process A5 5.506 12.94 26.24 0.54-52.71 1.54 5.70 11.3
    Lot 27 Process A6 8.740 19.30 35.62 0.54-54.43 1.42 5.10 10.1
    Lot 27 Process A7 7.862 18.92 36.09 0.54-52.63 1.48 5.30 10.4
    Lot 27 Process A8 8.184 17.49 33.02 0.54-63.56 1.46 5.04 9.69
    Lot 27 Process A9 7.451 15.99 29.94 0.54-55.58 1.49 5.00 9.65
    Lot 27 Process A10 7.673 18.50 36.71 0.54-74.22 1.59 5.55 10.6
    Lot 27 Process A11 9.093 20.01 45.26 0.54-61.26 1.72 6.11 12.1
    Lot 27 Process A12 6.355 14.92 46.31 0.54-63.98 1.66 5.77 10.9
    Lot 27 Process A13 7.029 15.66 31.06 0.54-55.89 1.07 3.51 7.32
    Lot 27 Process A14 7.870 20.15 43.79 0.54-62.35 1.74 5.87 11.0
    Lot 27 Process A14 8.499 19.06 37.90 0.54-70.88 1.67 5.49 10.5
    Lot 27 Process A14 8.072 17.43 30.90 0.54-56.50 1.76 6.01 11.5
    Lot 27 Process A14 8.078 18.98 45.32 0.54-69.82 1.81 6.43 12.5
  • When ranges are used herein for chemical or physical properties, such as particle size or particle size distribution, formulation component concentrations, or pharmacokinetic properties, all combinations, and subcombinations of ranges for specific embodiments therein are intended to be included.
  • The disclosures of each patent, patent application, and publication cited or described in this document are hereby incorporated herein by reference, in its entirety.
  • Those skilled in the art will appreciate that numerous changes and modifications can be made to the preferred embodiments of the invention and that such changes and modifications can be made without departing from the spirit of the invention. It is, therefore, intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention.

Claims (28)

1. A suspension comprising fulvestrant particles and a vehicle;
wherein optionally the fulvestrant particles have
an LD Dv(10) between about 1 micron to about 3 microns, preferably an LD Dv(10) of about 1-2 microns,
an LD Dv(50) between about 2 microns and about 35 microns, preferably an LD Dv(50) of about 2-4 microns, and
an LD Dv(90) between about 4 microns and about 120 microns, preferably an LD Dv(90) of about 6-9 microns, and;
wherein optionally the vehicle is a non-oil vehicle, preferably water or non-aqueous vehicle;
wherein optionally fulvestrant is at a concentration of about 100 mg/mL; and
wherein optionally the suspension is substantially oil-free.
2. A suspension comprising fulvestrant and a vehicle,
wherein the fulvestrant is at a concentration equal to or greater than about 50 mg/mL, preferably about 50 mg/mL or about 100 mg/mL;
wherein optionally the vehicle is a non-oil vehicle, preferably water or non-aqueous vehicle;
wherein optionally the suspension is substantially oil-free.
3. A pharmaceutical composition comprising fulvestrant particles and a non-oil vehicle,
wherein optionally the pharmaceutical composition further comprises at least one stabilizer selected from the group consisting of surfactants, polymers, cross-linked polymers, buffering agents, electrolytes, and non-electrolytes, preferably wherein the at least one stabilizer is a cross-linked polymer, preferably carboxymethylcellulose sodium, or the at least one stabilizer is selected from:
the group of polyethylene oxide (PEO), a PEO derivative, polysorbate 80, polysorbate 20, poloxamer 188, poloxamer 124, poloxamer 407, polyethoxylated vegetable oils, polyethoxylated castor oil, sorbitan palmitate, lecithin, poly(vinyl alcohol), human serum albumin, and mixtures thereof,
the group consisting of polyvinylpyrrolidone, povidone K12, povidone K17, PLASDONE™ C-12 povidone, PLASDONE™ C-17 povidone, PLASDONE™ C-30 povidone, polyethylene glycol 3350, and mixtures thereof,
the group consisting of sodium chloride, calcium chloride, and mixtures thereof, or
the group consisting of dextrose, glycerol, mannitol, and mixtures thereof;
wherein optionally the pharmaceutical composition further comprises at least one buffering agent selected from the group consisting of NaH2PO4.H2O, NaH2PO4.2H2O, anhydrous NaH2PO4, sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or a mixture thereof; and
wherein optionally the pharmaceutical composition further comprises solubilized fulvestrant.
4. A pharmaceutical composition comprising fulvestrant particles,
wherein the fulvestrant particles have one or more of:
an LD Dv(10) between about 1 micron to about 3 microns, preferably between about 1 micron to about 2 microns or between about 2 microns and about 3 microns;
an LD Dv(50) between about 2 microns and about 35 microns, preferably between about 2 microns to about 6 microns, more preferably between about 2 microns and about 4 microns; and
an LD Dv(90) between about 4 microns and about 120 microns, preferably between about 7 microns to about 15 microns, more preferably between about 12 microns to about 14 microns or between about 9 microns to about 11 microns, or preferably between about 6 microns and about 9 microns, more preferably between about 6 microns and about 8 microns, most preferably between about 7 microns and about 8 microns;
wherein optionally the pharmaceutical composition is in the form of a suspension, preferably an aqueous suspension.
5. A pharmaceutical composition comprising fulvestrant,
wherein the fulvestrant is at a concentration of about 100 mg/mL and wherein upon administration to a subject, the 90% confidence intervals (CI) of the relative mean AUC(0-t) and AUC(0-∞) of fulvestrant of the pharmaceutical composition is within 80% to 125% of the relative mean AUC(0-t) and AUC(0-∞), respectively, of fulvestrant upon administration of a reference listed fulvestrant product, for example, FASLODEX™;
wherein optionally the reference listed fulvestrant product comprises fulvestrant at a concentration of about 50 mg/mL;
wherein optionally upon administration to a subject, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 40% to 80% of the relative mean Cmax of fulvestrant after administration of the reference listed fulvestrant product.
6. A pharmaceutical composition comprising fulvestrant wherein the fulvestrant is at a concentration of about 100 mg/mL and wherein after an initial administration to a subject of the pharmaceutical composition comprising 500 mg fulvestrant on day 1 and an additional administration of the pharmaceutical composition comprising 500 mg fulvestrant on day 15 to the subject provides:
an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
wherein optionally the pharmaceutical composition is administered as a single injection of about 5 mL or administered as two injections of about 2.5 mL.
7. A pharmaceutical composition comprising fulvestrant particles wherein the fulvestrant is at a concentration of about 40 to about 125 mg/mL.
8. The pharmaceutical composition of claim 7,
wherein at least about 70% of the fulvestrant is present as fulvestrant particles, preferably at least about 80% of the fulvestrant is present as fulvestrant particles, more preferably at least about 90% of the fulvestrant is present as fulvestrant particles.
9. The pharmaceutical composition of claim 7,
wherein no more than about 20% of the fulvestrant is solubilized, preferably no more than about 10% of the fulvestrant is solubilized, more preferably no more than about 5% of the fulvestrant is solubilized.
10. The pharmaceutical composition of claim 6,
wherein the fulvestrant particles:
have one or more of:
an LD Dv(10) between about 1 micron to about 3 microns;
an LD Dv(50) between about 2 microns and about 35 microns; and
an LD Dv(90) between about 4 microns and about 120 microns;
have one or more of:
an LD Dv(10) between about 1-2 microns;
an LD Dv(50) between about 2-4 microns; and
an LD Dv(90) between about 6-9 microns;
have an LD Dv(90) of about 12-14 microns;
have an LD Dv(90) of about 9-11 microns;
have an LD Dv(90) of about 6-9 microns;
have an LD Dv(90) of about 6-8 microns; or
have an LD Dv(90) of about 7-8 microns.
11. A method of forming an aqueous fulvestrant suspension comprising:
mixing an aqueous medium and at least one stabilizer to form a suspension vehicle, the at least one stabilizer preferably comprising one surfactant and one polymer or one surfactant and one non-electrolyte;
adding an amount of fulvestrant to the suspension vehicle; and
dispersing the fulvestrant in the suspension vehicle to form the aqueous fulvestrant suspension, preferably performed using high shear mixing;
optionally further comprising homogenizing the aqueous fulvestrant suspension, the homogenizing preferably performed using high pressure homogenization, preferably at a pressure of about 15,000 psi to about 45,000 psi, the method preferably further comprising adding an electrolyte to the homogenized aqueous fulvestrant suspension and mixing the electrolyte into the suspension or adding a non-electrolyte to the homogenized aqueous fulvestrant suspension and mixing the non-electrolyte into the suspension;
the method optionally further comprising concentrating the fulvestrant suspension by phase separating the suspension and removing a portion of the supernatant;
the method optionally further comprising drying the aqueous suspension to form a dried pharmaceutical composition, the method preferably further comprising sterilizing the dried pharmaceutical composition using gamma irradiation, the method more preferably further comprising reconstituting the dried pharmaceutical composition into a second aqueous suspension by adding at least one of water for injection (WFI), normal saline (NS), and 5% dextrose in water (D5W).
12. An aqueous fulvestrant suspension prepared according to the method of claim 11.
13. A pharmaceutical composition comprising the aqueous fulvestrant suspension of claim 12,
wherein optionally the pharmaceutical composition comprises fulvestrant particles:
having one or more of:
an LD Dv(10) between about 1 micron to about 3 microns;
an LD Dv(50) between about 2 microns and about 35 microns; and
an LD Dv(90) between about 4 microns and about 120 microns;
having one or more of:
an LD Dv(10) between about 1-2 microns;
an LD Dv(90) between about 6-9 microns; and
an LD Dv(50) between about 2-4 microns;
having one or more of:
a CE Dv(90) between about 10 microns and about 200 microns;
a CE Dv(50) between about 5 microns and about 60 microns; and
a CE Dv(10) between about 1 microns and about 25 microns;
having a CE Dv(90) between about 10 microns and about 200 microns;
having a CE Dv(50) between about 5 microns and about 60 microns; or
having a CE Dv(10) between about 1 microns and about 25 microns.
14. An aqueous fulvestrant suspension comprising:
an amount of fulvestrant, preferably about 500 mg, about 450 mg, about 400 mg, about 350 mg, about 300 mg, or about 250 mg;
about 0.2 mg/mL to about 75 mg/mL of one or more stabilizers; and
an amount of aqueous medium;
wherein upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean AUC(0-t) and AUC(0-∞) of fulvestrant is within 80% to 125% of the relative mean AUC(0-t) and AUC(0-∞), respectively, of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
wherein optionally the suspension has a volume of about 3.0 to about 5.0 mL, preferably about 3.5 to about 4.5 mL, more preferably about 4.0 mL.
15. The aqueous fulvestrant suspension of claim 14,
wherein the one or more stabilizers:
is selected from the group consisting of surfactants, polymers, cross-linked polymers, buffering agents, electrolytes, and non-electrolytes,
is selected from the group consisting of polyethylene oxide (PEO), a PEO derivative, polysorbate 80, polysorbate 20, poloxamer 188, poloxamer 124, poloxamer 407, polyethoxylated vegetable oils, polyethoxylated castor oil, sorbitan palmitate, lecithin, poly(vinyl alcohol), human serum albumin, and mixtures thereof,
is selected from the group consisting of polyvinylpyrrolidone, povidone K12, povidone K17, PLASDONE™ C-12 povidone, PLASDONE™ C-17 povidone, PLASDONE™ C-30 povidone, polyethylene glycol 3350, and mixtures thereof,
is selected from the group consisting of sodium chloride, calcium chloride, and mixtures thereof, or
is selected from the group consisting of dextrose, glycerol, mannitol, and mixtures thereof.
16. The aqueous fulvestrant suspension of claim 14, wherein the one or more stabilizers comprises a cross-linked polymer, preferably carboxymethylcellulose sodium.
17. The aqueous fulvestrant suspension of claim 14, further comprising at least one buffering agent selected from the group consisting of NaH2PO4.H2O, NaH2PO4.2H2O, anhydrous NaH2PO4, sodium citrate, citric acid, Tris, sodium hydroxide, HCl, or mixtures thereof.
18. The aqueous fulvestrant suspension of claim 14,
wherein the fulvestrant particles:
have one or more of:
an LD Dv(10) between about 1 micron to about 3 microns;
an LD Dv(50) between about 2 microns and about 35 microns; and
an LD Dv(90) between about 4 microns and about 120 microns; or
have one or more of:
an LD Dv(10) between about 1-2 microns;
an LD Dv(50) between about 2-4 microns; and
an LD Dv(90) between about 6-9 microns.
19. The aqueous fulvestrant suspension of claim 14,
wherein the fulvestrant particles:
have one or more of:
a CE Dv(10) between about 4-10 microns,
a CE Dv(50) between about 10-35 microns, and
a CE Dv(90) between about 35-110 microns;
have one or more of:
a CE Dv(10) between about 4-8 microns,
a CE Dv(50) between about 10-25 microns, and
a CE Dv(90) between about 25-60 microns; or
have one or more of:
a CE Dv(10) between about 4-8 microns,
a CE Dv(50) between about 10-20 microns, and
a CE Dv(90) between about 20-35 microns.
20. The aqueous fulvestrant suspension of claim 14,
wherein:
the suspension comprises about 56 mg/mL to about 59 mg/mL of one or more stabilizers; and
the one or more stabilizers comprises:
about 1.0 mg/mL to about 4.0 mg/mL of one or more polyvinylpyrrolidones;
about 5.0 mg/mL of polysorbate 80; and
about 50 mg/mL of mannitol; or
wherein:
the suspension comprises about 56.6 mg/mL to about 57.4 mg/mL of one or more stabilizers; and
the one or more stabilizers comprises:
about 1.6 mg/mL to about 2.4 mg/mL of one or more polyvinylpyrrolidones;
about 5.0 mg/mL of polysorbate 80; and
about 50 mg/mL of mannitol; or
wherein the suspension comprises:
about 1.6 mg/mL to about 2.4 mg/mL of PLASDONE™ C-12 povidone, povidone K12, or a combination thereof;
about 5.0 mg/mL of polysorbate 80; and
about 50 mg/mL of mannitol; or
wherein the suspension comprises:
about 100 mg/mL fulvestrant;
about 1.6 mg/mL of PLASDONE™ C-12 povidone, povidone K12, or a combination thereof;
about 5.0 mg/mL of polysorbate 80; and
about 50 mg/mL of mannitol; or
wherein the suspension comprises:
about 100 mg/mL fulvestrant;
about 2.4 mg/mL of PLASDONE™ C-12 povidone, povidone K12, or a combination thereof;
about 5.0 mg/mL of polysorbate 80; and
about 50 mg/mL of mannitol.
21. The pharmaceutical composition of claim 6,
wherein after the initial administration the additional administration provides:
both a Cmax in the range of 10.04-12.55 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 12.55-15.06 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 15.06-17.57 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 17.57-20.08 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 10.04-11.295 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 11.295-12.55 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 12.55-13.805 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 13.805-15.06 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 15.06-16.315 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 16.315-17.57 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 17.57-18.825 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 18.825-20.08 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 15.06-18.825 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant;
both a Cmax in the range of 12.55-18.825 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant; or
both a Cmax in the range of 10.04-15.06 ng/mL of fulvestrant and an AUC in the range of 9,120-14,250 ng·hr/mL of fulvestrant.
22. The pharmaceutical composition of claim 3, wherein the fulvestrant particles:
have one or more of:
a CE Dv(90) between about 35 microns and about 110 microns;
a CE Dv(50) between about 10 microns and about 35 microns; and
a CE Dv(10) between about 4 microns and about 10 microns;
have one or more of:
a CE Dv(90) between about 25 microns and about 60 microns;
a CE Dv(50) between about 10 microns and about 25 microns; and
a CE Dv(10) between about 4 microns and about 8 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 35 microns;
a CE Dv(50) between about 10 microns and about 20 microns; and
a CE Dv(10) between about 4 microns and about 8 microns;
have one or more of:
a CE Dv(90) between about 30 microns and about 100 microns;
a CE Dv(50) between about 10 microns and about 50 microns; and
a CE Dv(10) between about 4 microns and about 10 microns;
have one or more of:
a CE Dv(90) between about 50 microns and about 100 microns;
a CE Dv(50) between about 20 microns and about 50 microns; and
a CE Dv(10) between about 6 microns and about 8 microns;
have one or more of:
a CE Dv(90) between about 50 microns and about 75 microns;
a CE Dv(50) between about 30 microns and about 40 microns; and
a CE Dv(10) between about 8 microns and about 10 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 60 microns;
a CE Dv(50) between about 9 microns and about 20 microns; and
a CE Dv(10) between about 3 microns and about 7 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 50 microns;
a CE Dv(50) between about 9 microns and about 20 microns; and
a CE Dv(10) between about 3 microns and about 7 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 45 microns;
a CE Dv(50) between about 9 microns and about 20 microns; and
a CE Dv(10) between about 3 microns and about 7 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 40 microns;
a CE Dv(50) between about 9 microns and about 15 microns; and
a CE Dv(10) between about 3 microns and about 7 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 35 microns;
a CE Dv(50) between about 9 microns and about 15 microns; and
a CE Dv(10) between about 3 microns and about 7 microns;
have one or more of:
a CE Dv(90) between about 20 microns and about 45 microns;
a CE Dv(50) between about 9 microns and about 15 microns; and
a CE Dv(10) between about 3 microns and about 7 microns;
have a CE Dv(90) between about 10 microns and about 200 microns;
have a CE Dv(50) between about 5 microns and about 60 microns;
have a CE Dv(10) between about 1 microns and about 25 microns;
have one or more of:
a CE Dv(90) between about 10 microns and about 200 microns;
a CE Dv(50) between about 5 microns and about 60 microns; and
a CE Dv(10) between about 1 microns and about 25 microns;
have one or more of:
a CE Dv(90) between about 10 microns and about 200 microns; and
a CE Dv(50) between about 5 microns and about 60 microns;
have one or more of:
a CE Dv(50) between about 5 microns and about 60 microns; and
a CE Dv(10) between about 1 microns and about 25 microns; or
have one or more of:
a CE Dv(90) between about 10 microns and about 200 microns; and
a CE Dv(10) between about 1 microns and about 25 microns.
23. The aqueous fulvestrant suspension of claim 14,
wherein:
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 40% to 80% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 60% to 80% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 50% to 75% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 40% to 50% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 50% to 60% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 60% to 70% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections;
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 70% to 80% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections; or
upon administration of the aqueous fulvestrant suspension to a subject in a single intramuscular injection, the 90% confidence intervals (CI) of the relative mean Cmax of fulvestrant is within 70% to 80% of the relative mean Cmax of fulvestrant after administration of 500 mg of fulvestrant in the form of FASLODEX™ administered intramuscularly as two 5 mL injections.
24. A method of treating a subject having breast cancer, comprising administering to the subject
the suspension of claim 1;
wherein optionally the suspension, pharmaceutical composition, or aqueous fulvestrant suspension comprises about 50 mg/mL or about 100 mg/mL fulvestrant;
wherein optionally the suspension, pharmaceutical composition, or aqueous fulvestrant suspension is administered in combination with one or more additional therapeutic agents, preferably palbociclib.
25. The method of claim 24, wherein the breast cancer is hormone receptor (HR)-positive breast cancer.
26. The method of claim 24, wherein the subject is a post-menopausal human woman with disease progression following antiestrogen therapy.
27. The method of claim 24, wherein the breast cancer is HR-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced or metastatic breast cancer.
28. The method of claim 24, wherein the subject is a human woman with disease progression after endocrine therapy.
US16/099,265 2016-05-06 2017-05-05 Fulvestrant formulations and methods of their use Abandoned US20190134059A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/099,265 US20190134059A1 (en) 2016-05-06 2017-05-05 Fulvestrant formulations and methods of their use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662332842P 2016-05-06 2016-05-06
US201662420555P 2016-11-10 2016-11-10
US16/099,265 US20190134059A1 (en) 2016-05-06 2017-05-05 Fulvestrant formulations and methods of their use
PCT/US2017/031376 WO2017193048A1 (en) 2016-05-06 2017-05-05 Fulvestrant formulations and methods of their use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/031376 A-371-Of-International WO2017193048A1 (en) 2016-05-06 2017-05-05 Fulvestrant formulations and methods of their use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/379,534 Continuation US20210346396A1 (en) 2016-05-06 2021-07-19 Fulvestrant formulations and methods of their use

Publications (1)

Publication Number Publication Date
US20190134059A1 true US20190134059A1 (en) 2019-05-09

Family

ID=58709642

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/099,265 Abandoned US20190134059A1 (en) 2016-05-06 2017-05-05 Fulvestrant formulations and methods of their use
US17/379,534 Pending US20210346396A1 (en) 2016-05-06 2021-07-19 Fulvestrant formulations and methods of their use

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/379,534 Pending US20210346396A1 (en) 2016-05-06 2021-07-19 Fulvestrant formulations and methods of their use

Country Status (15)

Country Link
US (2) US20190134059A1 (en)
EP (1) EP3452011A1 (en)
JP (3) JP2019516789A (en)
KR (1) KR102438425B1 (en)
CN (1) CN109310621A (en)
AU (2) AU2017261321B2 (en)
BR (1) BR112018072768A2 (en)
CA (1) CA3022834A1 (en)
CO (1) CO2018013257A2 (en)
IL (2) IL262465A (en)
MA (1) MA44862A (en)
MX (2) MX2018013414A (en)
UA (1) UA125514C2 (en)
WO (1) WO2017193048A1 (en)
ZA (1) ZA201807031B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113694017A (en) * 2020-05-11 2021-11-26 鲁南制药集团股份有限公司 Fulvestrant injection preparation and preparation method thereof
CN114504551A (en) * 2020-11-16 2022-05-17 北京厚燊维康科技有限责任公司 Formulations useful for photodynamic therapy or diagnosis
EP4074304A4 (en) * 2019-12-11 2024-01-10 Shanghai Bocimed Pharmaceutical Co Ltd Fulvestrant pharmaceutical composition, preparation method therefor, and application thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11590077B2 (en) 2016-05-06 2023-02-28 Eagle Pharmaceuticals, Inc. Fulvestrant formulations and methods of their use
BR112020009141A2 (en) * 2017-11-08 2020-10-27 Eagle Pharmaceuticals, Inc. method to treat breast cancer in an individual
CN114617847A (en) * 2020-12-10 2022-06-14 上海博志研新药物技术有限公司 Fulvestrant pharmaceutical composition, preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030114430A1 (en) * 2000-05-15 2003-06-19 Macleod Steven K. Stabilized injectable suspension of a steroidal drug
US20080161276A1 (en) * 2004-08-04 2008-07-03 Markus Johnsson Compositions Forming Non-Lamellar Dispersions

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8853260B2 (en) * 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
GB0000313D0 (en) * 2000-01-10 2000-03-01 Astrazeneca Uk Ltd Formulation
WO2003006064A1 (en) * 2001-07-07 2003-01-23 Astrazeneca Ab Pharmaceutical formulation for the intramuscular administration of fulvestrant
GB0116619D0 (en) * 2001-07-07 2001-08-29 Astrazeneca Ab Formulation
CN101108168B (en) * 2007-08-03 2010-12-08 西安力邦医药科技有限责任公司 Method of manufacturing fulvestrant sustained-release microspheres
US9180088B2 (en) * 2008-03-07 2015-11-10 Scidose, Llc Fulvestrant formulations
TW201016220A (en) * 2008-10-31 2010-05-01 Scinopharm Taiwan Ltd Micronization form of 7α-[9-(4,4,5,5,5-pentafluoropentylsufinyl)nonyl]estra-1,3,5(10)-triene-3,17β-diol and process for the preparation thereof
MX2011013797A (en) * 2009-06-18 2012-01-30 Abbott Lab Stable nanoparticulate drug suspension.
US20130280330A9 (en) * 2009-07-31 2013-10-24 Xi'an Libang Medical Technology Co., Ltd. Microsphere drug carrier, preparation method, composition and use thereof
CN102264372B (en) * 2009-08-31 2012-07-11 西安力邦医药科技有限责任公司 Fulvestrant nanosphere/microsphere and preparative method and use thereof
PT2616078T (en) 2010-09-16 2020-03-31 Shimoda Biotech Pty Ltd Fulvestrant compositions and methods of use
BR112013029758A2 (en) * 2011-05-20 2018-10-09 Capital, Business Y Gestion De Finanzas S.L. pharmaceutical composition
US11179468B2 (en) * 2012-04-09 2021-11-23 Eagle Pharmaceuticals, Inc. Fulvestrant formulations
MX2014014831A (en) 2012-06-08 2015-02-12 Hoffmann La Roche Mutant selectivity and combinations of a phosphoinositide 3 kinase inhibitor compound and chemotherapeutic agents for the treatment of cancer.
CN104337761B (en) * 2013-08-07 2019-03-26 江苏豪森药业集团有限公司 Fulvestrant pharmaceutical composition
EP3041512A4 (en) * 2013-09-06 2017-05-10 Ahmed, Salah U. Fulvestrant compositions
EP3052493A4 (en) * 2013-10-03 2017-06-14 Millennium Pharmaceuticals, Inc. Enhanced treatment regimens using pi3k inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030114430A1 (en) * 2000-05-15 2003-06-19 Macleod Steven K. Stabilized injectable suspension of a steroidal drug
US20080161276A1 (en) * 2004-08-04 2008-07-03 Markus Johnsson Compositions Forming Non-Lamellar Dispersions

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4074304A4 (en) * 2019-12-11 2024-01-10 Shanghai Bocimed Pharmaceutical Co Ltd Fulvestrant pharmaceutical composition, preparation method therefor, and application thereof
CN113694017A (en) * 2020-05-11 2021-11-26 鲁南制药集团股份有限公司 Fulvestrant injection preparation and preparation method thereof
CN114504551A (en) * 2020-11-16 2022-05-17 北京厚燊维康科技有限责任公司 Formulations useful for photodynamic therapy or diagnosis

Also Published As

Publication number Publication date
MA44862A (en) 2019-03-13
US20210346396A1 (en) 2021-11-11
AU2017261321B2 (en) 2023-03-09
KR20190005183A (en) 2019-01-15
EP3452011A1 (en) 2019-03-13
BR112018072768A2 (en) 2019-02-19
ZA201807031B (en) 2023-10-25
MX2018013414A (en) 2019-06-06
JP2023109959A (en) 2023-08-08
RU2018142068A (en) 2020-06-08
CA3022834A1 (en) 2017-11-09
CO2018013257A2 (en) 2018-12-28
MX2022013199A (en) 2022-12-13
CN109310621A (en) 2019-02-05
IL285928A (en) 2021-10-31
RU2018142068A3 (en) 2020-09-17
WO2017193048A1 (en) 2017-11-09
UA125514C2 (en) 2022-04-13
JP2021169510A (en) 2021-10-28
AU2017261321A1 (en) 2018-11-15
JP2019516789A (en) 2019-06-20
JP7312523B2 (en) 2023-07-21
AU2023203392A1 (en) 2023-06-29
KR102438425B1 (en) 2022-09-01
IL262465A (en) 2018-12-31

Similar Documents

Publication Publication Date Title
US20210346396A1 (en) Fulvestrant formulations and methods of their use
JP5513713B2 (en) Compositions that produce non-layered dispersions
JP3977418B2 (en) Nanosuspension for drug administration with increased saturation solubility and dissolution rate
RU2642234C2 (en) Antagonists neurokinin-1 compositions for intravenous introduction
CN103002877B (en) It is used as the antirheumatic medicine for alleviating disease(DMARD)With the extended release preparation of the methotrexate (MTX) of anticancer agent
JP5627039B2 (en) Nanodispersions of drugs and methods for their preparation
CN103153282B (en) The pharmaceutical dosage form of &#39;, 9 &#39;-dihydro-3 &#39; H-spiral shell [cyclohexane extraction-1,1 &#39;-pyrans is [3,4, B] indole also]-4-amine containing 6 &#39;-fluoro-(N-methyl-or N, N-dimethyl-)-4-phenyl-4
JPH06510772A (en) Pharmaceutical carrier made of solid lipid microparticles (solid lipid nanospheres)
CN106794251B (en) Aripiprazole prodrug compositions
CN103263385B (en) A kind of celecoxib long-acing nano injection and preparation method thereof
JP7157830B2 (en) Aripiprazole prodrug composition
RU2775225C2 (en) Fulvestrant compositions and their application methods
JP2021502337A (en) Sustained release formulation for intra-articular application
WO2011047639A1 (en) Pharmaceutical solution of taxanes comprising chelating agent and preparation method thereof
RU2817019C2 (en) Aripiprazole prodrug compositions
CN113117092A (en) Non-aqueous sustained-release drug delivery system

Legal Events

Date Code Title Description
AS Assignment

Owner name: EAGLE PHARMACEUTICALS, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEN, FENG-JING;KRILL, STEVEN L.;SHMEIS, RAMA ABU;REEL/FRAME:047421/0458

Effective date: 20170515

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: JPMORGAN CHASE BANK, N.A., AS ADMINISTRATIVE AGENT, ILLINOIS

Free format text: SECURITY INTEREST;ASSIGNOR:EAGLE PHARMACEUTICALS, INC.;REEL/FRAME:061513/0804

Effective date: 20191108