WO2022067336A1 - Compositions and methods for vasodilator delivery - Google Patents

Compositions and methods for vasodilator delivery Download PDF

Info

Publication number
WO2022067336A1
WO2022067336A1 PCT/US2021/071589 US2021071589W WO2022067336A1 WO 2022067336 A1 WO2022067336 A1 WO 2022067336A1 US 2021071589 W US2021071589 W US 2021071589W WO 2022067336 A1 WO2022067336 A1 WO 2022067336A1
Authority
WO
WIPO (PCT)
Prior art keywords
nanostructured
formulation
nonlamellar
iloprost
phase
Prior art date
Application number
PCT/US2021/071589
Other languages
French (fr)
Inventor
Matthew DUFFY
Kamal MUBARAK
Original Assignee
Algorithm Sciences, 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 Algorithm Sciences, Inc. filed Critical Algorithm Sciences, Inc.
Priority to JP2023519289A priority Critical patent/JP2023543037A/en
Priority to MX2023003506A priority patent/MX2023003506A/en
Publication of WO2022067336A1 publication Critical patent/WO2022067336A1/en

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • A61K31/5578Eicosanoids, e.g. leukotrienes or prostaglandins having a pentalene ring system, e.g. carbacyclin, iloprost
    • 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
    • A61K9/127Liposomes
    • A61K9/1274Non-vesicle bilayer structures, e.g. liquid crystals, tubules, cubic phases, cochleates; Sponge phases

Definitions

  • the present invention relates to the field of pharmaceutical compositions for administering a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof).
  • a vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof.
  • Other embodiments are directed to methods of treatment and methods of manufacturing the compositions disclosed herein.
  • Constriction of blood vessels is a serious condition that prevents proper blood flow to surrounding tissues.
  • pulmonary hypertension is an elevated blood pressure that affects the arteries in the lungs and the right side of the heart.
  • PAH pulmonary arterial hypertension
  • blood vessels in the lungs are narrowed, blocked or damaged which slows blood flow through the lungs, and blood pressure in the lung arteries becomes elevated. This causes the heart to work harder and results in cardiac complications.
  • Pulmonary hypertension may become worse over time and can be life-threatening.
  • Other diseases associated with constricted blood vessels include scleroderma and Raynaud’s syndrome.
  • Iloprost is a vasodilator that can be used to treat diseases associated with constricted blood vessels.
  • the chemical name for iloprost is (E)-(3aS, 4R, 5R, 6aS)-hexahydro-5-hydroxy-4- [(E)(3S,4RS)-3-hydroxy-4-methyl-l-octen-6-ynyl]- ⁇ 2(1H) ⁇ -pentalenevaleric acid.
  • Racemic iloprost consists of a mixture of the 4R and 4S diastereoisomers at a ratio of approximately 50:50.
  • Iloprost is commercially available as a solution for inhalation in 1 mL single-use glass ampules containing either 10 mcg/mL or 20 mcg/mL and is administered by a nebulized delivery system.
  • the approved regimen includes dosing every 6-9 hours during waking hours. This is very inconvenient and difficult for compliance and may result in underdosing or overdosing by patients.
  • vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof
  • disease states such as arterial hypertension, pulmonary arterial hypertension, scleroderma and Reynaud’s disease.
  • the vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof
  • diseases associated with constriction of blood vessels arterial hypertension, pulmonary arterial hypertension, scleroderma and Reynaud’s disease
  • the pharmaceutical compositions e.g., by an implantable pump
  • a pharmaceutical composition comprising a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable excipient (e.g., a lipid material such as a lyotropic liquid crystal material).
  • a vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof
  • a pharmaceutically acceptable excipient e.g., a lipid material such as a lyotropic liquid crystal material.
  • the pharmaceutical composition is contained in an implantable pump.
  • the present invention is directed to the use of a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) in the preparation of a medicament for the treatment of a disease requiring vasodilatory therapy such as hypertension (e.g., pulmonary hypertension).
  • a vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof
  • the medicament is contained in an implantable pump.
  • vasodilator includes a single vasodilator as well as a mixture of two or more different vasodilators
  • excipient includes a single excipient as well as a mixture of two or more different excipients, and the like.
  • the term “about” in connection with a measured quantity or time refers to the normal variations in that measured quantity or time, as expected by one of ordinary skill in the art in making the measurement and exercising a level of care commensurate with the objective of measurement.
  • the term “about” includes the recited number ⁇ 10%, such that “about 10” would include from 9 to 11, or “about 1 hour” would include from 54 minutes to 66 minutes.
  • the terms "therapeutically effective” and an “effective amount” refer to that amount of an active agent or the rate at which it is administered needed to produce a desired therapeutic result.
  • treatment of and “treating” include the administration of an active agent(s) with the intent to lessen the severity of a condition.
  • prevention of and “preventing” include the avoidance of the onset of a condition by a prophylactic administration of the active agent.
  • parenteral means administration by injection, implantation or infiltration by a route such as intravenous, inhalation, intrarterial, intracardiac, intraspinal, intraosseous, intrarticular, intrasynovial, intracutaneous, subcutaneous or intramuscular.
  • injection means administration to a discrete site through the skin or into tissue of a human or animal.
  • implantation means embedding a device and/or pharmaceutical composition into the skin, tissue, muscles, tendons, joints, or other body parts of a human or animal.
  • the term refers to a surgical procedure where a device and/or pharmaceutical composition is put into the body and the skin is closed over it.
  • aqueous formulation refers to a water base formulation or compositionwithout a lipid material.
  • formulation and composition are used interchangeably.
  • FIG. 1 is a graphic illustration of Mean (SD) Plasma Concentrations (pg/mL) of Iloprost versus Time Post Start of Infusion in Female Dogs Following a Single Continuous Intravenous Infusion of Aqueous Formulation or Aqueous lipid-based Formulation for up to 24
  • FIGS. 2 to 6 are a graphic illustration of Individual PK Profiles of Log-transformed
  • FIGS. 7 to 11 are a graphic illustration of Individual PK Profiles of Log-transformed Concentration Values vs Time and Linear Regression to Estimate the Elimination Rate in Dog Plasma after IV Infusion of Iloprost Aqueous lipid-based for up to 24 Hours at 5760 ng/kg of the Pharmokinetic (PK) study.
  • PK Pharmokinetic
  • FIG. 12 is an image of the optical microscopcy of the blank aqueous lipid mixture of the Preparation example.
  • FIG. 13 is an image of the optical microscopy of the Manufacturing Example.
  • FIG. 14 is a bargraph illustrating intracellular cAMP levels in isolated hPASMCs upon treatment with Iloprost in an aqueous lipid formulation and aqueous lipid dispersion.
  • FIG. 15 is a bargraph illustrating intracellular cAMP levels in isolated hPASMCs upon treatment with Iloprost Trometamol solution.
  • FIG. 16 is a line graph illustrating intracellular cAMP levels in isolated hPASMCs upon treatment with Iloprost in an aquous lipid formulation with Iloprost Trometamol formulation.
  • the invention is directed to a pharmaceutical composition
  • a vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition comprising a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) that can be administered (e.g., by implantable pump) to facilitate the treatment of relevant disease states.
  • the pharmaceutical composition comprises iloprost or a pharmaceutically acceptable salt thereof and a lipid material (e.g., a lyotropic liquid crystal material).
  • a lipid material e.g., a lyotropic liquid crystal material
  • the pharmaceutical composition is an aqueous lipid solution).
  • the lyotropic liquid crystal material comprises both polar and non-polar nano-domains.
  • the material comprises nanostructured nonlamellar liquid crystalline phases.
  • the material comprises reversed nanostructured liquid crystalline phases.
  • the material comprises distinct nanostructured nonlamellar liquid crystalline material and a liquid phase embedded within said distinct nanostructured nonlamellar liquid crystalline material, said liquid phase being selected from the group consisting of a hydrophobe-rich phase and a polar solvent-rich phase.
  • the pharmaceutical composition disclosed herein is contained in an implantable pump that can deliver the active agent for a time period of, e.g., at least 3 days, at least 7 days, at least 10 days, at least 14 days, at least 21 days, at least 28 days, at least 30 days, at least 45 days, at least 60 days, at least 90 days, at least 100 days, at least 120 days, at least 150 days, at least 180 days, at least 200 days, at least 210 days, at least 240 days, at least 270 days, at least 300 days, or at least one year.
  • a time period of e.g., at least 3 days, at least 7 days, at least 10 days, at least 14 days, at least 21 days, at least 28 days, at least 30 days, at least 45 days, at least 60 days, at least 90 days, at least 100 days, at least 120 days, at least 150 days, at least 180 days, at least 200 days, at least 210 days, at least 240 days, at least 270 days, at least 300 days, or at least one year.
  • the pharmaceutical composition disclosed herein is contained in an implantable pump that can deliver the active agent for a time period of, e.g., about 3 days, about 7 days, about 10 days, about 14 days, about 21 days, about 28 days, about 30 days, about 45 days, about 60 days, about 90 days, about 100 days, about 120 days, about 150 days, about 180 days, about 200 days, about 210 days, about 240 days, about 270 days, about 300 days, or about one year.
  • the pharmaceutical composition disclosed herein is contained in an implantable pump that can deliver the active agent for a time period of, e.g., from about 3 days to about 400 days, from about 7 day to about 200 days, or from about 30 days to about 180 days.
  • the formulation e.g., a lipid formulation such as a lyotropic liquid crystal formulation
  • the vasodilator e.g., iloprost or pharmaceutically acceptable salt thereof
  • the formulation disclosed herein increases the potency of the drug.
  • it may increase the potency of iloprost or pharmaceutically acceptable salt thereof on prostaglandin receptors. This may decrease the minimum effective dose and result in less drug being administered and/or decrease the frequency of dosing.
  • the formuation dislcosed herein may have a pH of about 6 to about 10, about 7 to about 9 or about 7.5 to 8.5.
  • the formulation disclosed herein may have a density of about 0.8 to about 1.2 g/mL, about 0.9 to about 1.1 g/mL or about 1 g/mL.
  • the formulation disclosed herein may have a particle size distribution (D90) of the drug of about 0.1 nm to about 10 nm, about 0.5 nm to about 5 nm about 1 nm to about 3 nm, about 1.25 nm to about 2.75 nm, about 1.5 nm to about 2.5 nm, or about 1.75 nm to about 2.25 nm.
  • D90 particle size distribution
  • the formulation disclosed herein provides iloprost or pharmaceutically acceptable salt thereof to the patient in a daily amount from about 1000 ng/kg to about 10,000 ng/kg, from about 2000 ng/kg to about 8000 ng/kg, from about 5000 ng/kg to about 6500 ng/kg, from about 5250 ng/kg to about 6250 ng/kg, from about 5500 ng/kg to about 6000 ng/kg, or from about 5700 ng/kg to about 5800 ng/kg.
  • the formulation disclosed herein is administered daily, twice daily, three times daily, 4 times daily, 6 times daily, once weekly, twice weekly, three times weekly, 4 times weekly or any oyjer suitable disong mitin.
  • the administration is continuous (e.g., by intravenous infusion) over a time period of about 30 minutes to about 7 days, about 1 hour to about 3 days, about 2 hours to about 2 days, or about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 12 hours or about 24 hours.
  • formulation disclosed herein provides a maximum plama concentration (C max ) of iloprost that is higher when compared to the adminsitartiion of a comparable aqueous formulation.
  • C max maximum plama concentration
  • the Cmax provided by the present formulation is up to about 100% higher, up to about about 90% higher, up to about 80% higher, up to about 75% higher, up to about 65% higher, up to about 50% higher, up to about 40% higher, up to about 25% higher, up to about 15% higher, up to about 10% higher or up to about 5% higher than a comparable aqueuous formulation.
  • the formulation disclosed herein provides a half-life of iloprost that decreases as compared to a comparable aqueous formulation.
  • the decrease is about 75% or less, about 50% or less, about 35% or less, about 25% or less, about 10% or less or about 5% or less as compared to an aqueous formulation.
  • the formulation disclosed herein provides a total clearance of iloprost that increases when compared to an aqueous formulation.
  • the total clearance provided by the present formulation is up to about 100% higher, up to about about 90% higher, up to about 80% higher, up to about 75% higher, up to about 65% higher, up to about 50% higher, up to about 40% higher, up to about 25% higher, up to about 15% higher, up to about 10% higher or up to about 5% higher than a comparable aqueuous formulation
  • the formulation disclosed herein has a relative mean plasma exposure (AUC 0-48 ) that increases as compared to a comparable aqueous formulation.
  • the (AUC 0-48 ) provided by the present formulation is up to about 100% higher, up to about about 90% higher, up to about 80% higher, up to about 75% higher, up to about 65% higher, up to about 50% higher, up to about 40% higher, up to about 25% higher, up to about 15% higher, up to about 10% higher or up to about 5% higher than a comparable aqueuous formulation.
  • the formulation may be used to treat any condition to provide vasodilator therapy or iloprost therapy such as pulmonary arterial hypertension, or hypertension.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 10 nM to about 40 nM, or about 20 nM to about 30 nM at a concentration of about 1950 nM iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 15 nM, or about 4 nM to about 12 nM at a concentration of about 650 nM iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 15 nM, or about 4 nM to about 12 nM at a concentration of about 195 nM iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 15 nM, or about 4 nM to about 12 nM at a concentration of about 65 nM iloprost.
  • the formulation disclosed herein provides an increase in cAMP in human PASMCs when compared to an aqueous formulation iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 5 nM to about 30 nM, or about 10 nM to about 20 nM at a concentration of about 2310nm iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 20 nM, or about 5 nM to about 15 nM at a concentration of about 770 nM iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 12 nM, or about 5 nM to about 10 nM at a concentration of about 231 nM iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 12 nM, or about 5 nM to about 10 nM at a concentration of about 77 nM iloprost.
  • the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of at least about 15 nM, at at least about 20 nM, from about 15 nM to about 50 nM, from about 20 nM to about 30 nM, or from about 22 nM to about 28 nM at a concentration of about 1000 nM to about 3000 nM, about 1500 nM to about 2500 nM, about 1900 nM to about 2400 nM or about 1950 nM or about 2310 nM iloprost.
  • the composition disclosed herein can be contained in an implantable pump.
  • the pump may provide a continuous release or a pulsed release of the drug.
  • the pulses may be, e.g., about every 2 hours, about every 4 hours, about every 6 hours, about every 8 hours, about every 12 hours, about every 24 hours, about every 48 hours, about every 72 hours or about every 168 hours.
  • the implantable pump may be implanted in any suitable body space such as the abdomen, neck area (e.g., under the collarbone), buttock, thigh or upper arm.
  • the composition is released from the pump into the surrounding tissue and the drug (e.g., iloprost or pharmaceutically acceptable salt thereof) is subsequently absorbed into the circulatory system.
  • a catheter delivers the composition from the device (e.g., implanted in the abdomen or under the collarbone) directly into the circulatory system (e.g., to the venous system such as subclavian vein or the subjugular vein).
  • the pump is in the form of an implantable infusion device that comprises a lightweight, inert housing that contains a pharmaceutical composition disclosed herein for administration to a patient, and an enclosed, gas-impermeable variable volume chamber which contains a pressure source, such as a propellant or mechanical spring.
  • the variable volume chamber may be in the form of a rigid-walled expandable bellows structure or a nonstretchable flexible bag and is attached to the housing so as not to obstruct the entry port.
  • a recharging fluid flow path may be provided adjacent to where the variable volume chamber is attached to the housing.
  • the composition is delivered from the device via a catheter in response to expansion of the variable volume chamber against the volume of infusate in the housing.
  • a separate bolus injection port is provided which allows infusate to be safely introduced directly into the catheter and overrides the controlled pressure-driven delivery.
  • the lightweight housing may include integrally formed needle stops and suture fastening loops.
  • the device is in the form of an implantable infusion device comprising a housing including an inner wall and having an interior region defined at least in part by the inner wall, at least a portion of the interior region defining an pharmaceutical composition reservoir adapted to store a pharmaceutical composition as disclosed herein for delivery to a patient; a closed variable volume chamber having an outer wall, the variable volume chamber disposed within the interior region of the housing with the outer wall of the variable volume chamber being spaced from the inner wall of the housing, the variable volume chamber being supported within the interior region from an internal portion of the inner wall of the housing; a pressure source contained within the variable volume chamber; a pharmaceutical composition recharging fluid flow path adapted to recharge the infusate reservoir by carrying infusate to the infusate reservoir through the internal portion of the housing from which the variable volume chamber is supported within the interior region; and an exit fluid flow path adapted to deliver infusate from the infusate reservoir out of the housing, e.g., directly into the circulatory system.
  • the implantable pump comprises a filter for microbial agents.
  • the filter can comprise a material such as, e.g., plastic, ceramic, glass or fiber.
  • the filter comprises plastic and does not comprise polyvinylidene fluoride. In other embodiments, the filter does not comprise plastic.
  • Exemplary infusion pumps that can be adapted for use in the devices, systems (device/pharmaceutical composition combinations) and methods disclosed herein are described in US2003/0208184; US2014/0228765; US2014/0194851; US2013/0023857; US3, 840,009; US5,167,633; US5, 514103; US3,951,147; US5,045,064; US5,395,324; US5,769,823; US5,575,770; US10, 173,004; US10, 010670; US9,968,734; US9,700,669, US9, 180,282; US9, 125,982; US8,551,044; US8,545,477 and US 8,273,058, the disclosures of which are hereby incorporated by reference for all purposes.
  • the pharmaceutical formulation comprising iloprost or a pharmaceutically acceptable salt thereof; a carrier; and a particle or material comprising a distinct nanostructured nonlamellar liquid crystalline material; and one or more pockets or droplets of a liquid phase embedded within said distinct nanostructured nonlamellar liquid crystalline material, said liquid phase being selected from the group consisting of an oil-rich liquid phase and a polar solvent-rich liquid phase.
  • the distinct nanostructured nonlamellar liquid crystalline material is a reversed phase nonlamellar liquid crystalline material.
  • the distinct nanostructured nonlamellar liquid crystalline material comprises a reversed hexagonal phase material, a reversed bicontinuous cubic phase material, a reversed discrete cubic phase material, or a reversed intermediate phase material.
  • the distinct nanostructured nonlamellar liquid crystalline material is polymerized.
  • the pharmaceutical formulation further comprises a stabilizing layer exterior to said particle or material.
  • the stabilizing layer is selected from the group consisting of a charged moiety, a polymer, and a surfactant.
  • the particle or material further comprises a coating comprising the vasodilator (e.g., iloprost or pharmaceutically acceptable salt thereof).
  • the vasodilator e.g., iloprost or pharmaceutically acceptable salt thereof.
  • the liquid phase is an oil and said oil is selected from the group consisting of benzyl benzoate, estragole, eugenol, isoeugenol, linalool, and the essential oils of basil, bay, bois de rose (rosewood), carrot seed, clovebud, eucalyptus, ginger, grapefruit, hyssop, lemon, balsam of Peru, mugwort, myrrh gum, biter orange, oregano, palmarosa, patchouly, peppermint, petitgrain, rosemary, santalwood oil, spearmint, thuja (cedar leaf), thyme, vanilla, and ylang ylang (cananga).
  • said oil is selected from the group consisting of benzyl benzoate, estragole, eugenol, isoeugenol, linalool, and the essential oils of basil, bay, bois de rose (rosewood), carrot seed, clovebud, eucalyptus
  • the liquid phase is a polar solvent and said polar solvent is selected from the group consisting of water, glycerol, and N,N-dimethylacetamide.
  • the vasodilator e.g., iloprost or pharmaceutically acceptable salt thereof
  • the vasodilator is dissolved or dispersed in the liquid phase or in the distinct nanostructured nonlamellar liquid crystalline material.
  • the one or more pockets or droplets have a diameter of 50 nm or greater.
  • the liquid phase includes at least one of an oil and a polar solvent.
  • the liquid phase is a hydrophobe-rich phase or a polar solvent- rich phase.
  • the distinct nanostructured nonlamellar liquid crystalline material consists essentially of a reversed bicontinuous cubic phase material.
  • the distinct nanostructured nonlamellar liquid crystalline material comprises or consists essentially of a reversed discrete cubic phase material.
  • the distinct nanostructured nonlamellar liquid crystalline material comprises a water insoluble lipid or surfactant.
  • the pharmaceutical formulation comprises a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) and a coated particle comprising a) an interior core comprising a matrix comprising of i) at least one nanostructured liquid phase or a dehydrated variant thereof, ii) at least one nanostructured liquid crystalline phase or a dehydrated variant thereof or iii) a combination of (1) at least one nanostructured liquid phase or a dehydrated variant thereof and (2) at least one nanostructured liquid crystalline phase or a dehydrated variant thereof and b) an exterior coating comprising nonlamellar domains wherein the iloprost or pharmaceutically acceptable salt thereof is in a), b) or a combination thereof.
  • a vasodilator e.g., iloprost or a pharmaceutically acceptable salt thereof
  • a coated particle comprising a) an interior core comprising a matrix comprising of i) at least one nanostructured liquid phase or a dehydrated variant thereof, i
  • the nanostructured liquid phase material comprises a nanostructured LI phase material, a nanostructured L2 phase material, a microemulsion that is nanostructured, or a nanostructured L3 phase material.
  • the nanostructured liquid phase material comprises a nanostructured normal or reversed cubic phase material, a nanostructured normal or reversed hexagonal phase material, a nanostructured normal or reversed intermediate phase material, or nanostructured lamellar phase material.
  • the nanostructured liquid phase material comprises a polar solvent and a surfactant or a lipid.
  • the nanostructured liquid phase material comprises a polar solvent, a surfactant or a lipid and an amphiphile or hydrophobe.
  • the nanostructured liquid phase material comprises a block copolymer.
  • the nanostructured liquid phase material comprises a block copolymer and a solvent.
  • the nanostructured liquid phase material comprises a polar solvent and a surfactant.
  • the nanostructured liquid phase material comprises a polar solvent, a surfactant and an amphiphile or hydrophobe.
  • the interior core comprises the vasodilator (e.g., iloprost or pharmaceutically acceptable salt thereof) disposed within said matrix.
  • the vasodilator e.g., iloprost or pharmaceutically acceptable salt thereof
  • the interior core comprises a reversed cubic phase material.
  • the nonlamellar domain is amorphous.
  • the nonlamellar domain is a polymer such as polylactic glycolic acid.
  • the nonlamellar domain comprises a sugar such as trehalose.
  • the exterior coating comprises a semi-crystalline nonlamellar material.
  • the exterior coating comprises at least 2% nonlamellar domains, at least 10% nonlamellar domains or at least 50% nonlamellar domains.
  • the exterior coating comprises the vasodilator (iloprost or pharmaceutically acceptable salt thereof).
  • compositions and implants disclosed herein include iloprost or a pharmaceutically acceptable salt thereof.
  • Pharmaceutically acceptable salts include, but are not limited to, inorganic acid salts such as hydrochloride, hydrobromide, sulfate, phosphate and the like; organic acid salts such as formate, acetate, trifluoroacetate, maleate, tartrate and the like; sulfonates such as methanesulfonate, benzenesulfonate, p-toluenesulfonate, and the like; amino acid salts such as arginate, asparginate, glutamate and the like, and metal salts such as sodium salt, potassium salt, cesium salt and the like; alkaline earth metals such as calcium salt, magnesium salt and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, tromethamine salt, triethanolamine salt, dicyclohexylamine salt
  • vasodilators that can be utilized in the present invention (alone or in combination with iloprost) include an alpha-adrenergic receptor antagonist (a-blocker), an angiotensin converting enzyme (ACE) inhibitor, an angiotensin receptor blocker (ARB), a beta 2 -adrenergic receptor agonist ( 2 -agonist), Calcium channel blocker (CCB), centrally acting sympathetic blocker, direct acting vasodilator, endothelin receptor antagonist, ganglion blocker, nitrodilator, phosphodiesterase inhibitor, potassium channel opener, renin inhibitor or combination thereof.
  • a-blocker alpha-adrenergic receptor antagonist
  • ACE angiotensin converting enzyme
  • ARB angiotensin receptor blocker
  • 2 -agonist beta 2 -adrenergic receptor agonist
  • CB Calcium channel blocker
  • centrally acting sympathetic blocker direct acting vasodilator, endothelin receptor antagonist, ganglion blocker,
  • Particular agents include prazosin, terazosin, doxazosin, trimazosin, phentolamine, phenoxybenzamine, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, quinapril, ramipril, candesartan, eprosartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, epinephrine, norepinephrine, dopamine, dobutamine, isoproterenol, amlodipine, felodipine, isradipine, nicardipine, nifedipine, nimodipine, nitrendipine, clonidine
  • compositions according to the present invention may comprise one or more pharmaceutically acceptable carriers and excipients appropriate for parenteral administration.
  • pharmaceutically acceptable carriers and excipients are described in the Handbook of Pharmaceutical Excipients, American Pharmaceutical Association (6 th Edition, 2009 Publication), which is incorporated by reference herein.
  • Carriers and excipients suitable for parenteral compositions include antioxidants, buffering agents, diluents, surfactants, solubilizers, stabilizers, hydrophilic polymers, additional absorption or permeability enhancers, preservatives, osmotic agents, isotonicity agents, pH adjusting agents, solvents, co-solvents, viscosity agents, gelling agents, suspending agents or combinations thereof.
  • Suitable surfactants for the formulations disclosed herein include, but are not limited to Polysorbate 80 NF, polyoxyethylene 20 sorbitan monolaurate, polyoxyethylene (4) sorbitan monolaurate, polyoxyethylene 20 sorbitan monopalmitate, polyoxyethylene 20 sorbitan monostearate, polyoxyethylene (4) sorbitan monostearate, polyoxyethylene 20 sorbitan tristearate, polyoxyethylene (5) sorbitan monooleate, polyoxyethylene 20 sorbitan trioleate, polyoxyethylene 20 sorbitan monoisostearate, sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monostearate, sorbitan trilaurate, sorbitan trioleate, sorbitan tristearate, and the like, and combinations thereof.
  • the pharmaceutical composition is used in an implantable pump, implantable depot or similar implantable device that can provide administration of the active agent over an extended period of time.
  • it may be necessary to have a concentrated formulation in order to have the a suitable amount of active agent in a practical size for implant.
  • the iloprost is in a concentration from about 5 mg/mL to about 50 mg/mL, about 6 mg/mL to about 40 mg/mL, about 7 mg/mL to about 40 mg/mL, about 8 mg/mL to about 30 mg/mL, about 10 mg/mL to about 20 mg/mL, about 5 mg/mL to about 15 mg/mL or about 15 mg/mL to about 25 mg/mL.
  • the concentration is about 5 mg/mL, about 6 mg/mL, about 7 mg/mL, about 8 mg/mL, about 9 mg/mL, about 10 mg/mL, about 11 mg/mL about 12 mg/mL.
  • the device for implant can contain from about 5 mL to about 30 mL, about 10 mL to about 20 mL or about 15 mL to about 25 mL. In other embodiments the device for implant can contain about 10 mL, about 15 mL, about 20 m, about 25 mL, about 30 mL, about 40 mL or about 50 mL.
  • solubilizing agent such as an alcohol (e.g., benzyl alcohol or ethyl alcohol), a polyethylene glycol or dimethylsulfoxide (DMSO).
  • alcohol e.g., benzyl alcohol or ethyl alcohol
  • DMSO dimethylsulfoxide
  • the lipid mixture of the present disclosure may be prepared according to the following method.
  • a cubic phase is first prepared.
  • the cubic phase is prepared by mixing one or more of the following ingredients: vitamin E, a phospholipid and/or a lecithin.
  • the vitamin E may be a tocopherol, such as D,L-a-tocopherol.
  • the phospholipid may be derived from soybean, rapeseed (canola), egg or sunflower.
  • the lecithin may be naturally occurring and/or hydrogenated lecitihin fractions.
  • the phospholipid and/or lecithin may be Phospolipon ®, such as Phospholipon 90G, or Phospholipon 85G, LIPOID SI 00 or LIPOID E80.
  • the phospholipid may be from LIPOID ®, such as LIPOID SI 00 or LIPOID E80.
  • the phospholipid and/or lecithin may include one or more of the following ingredients: phosphatidylcholine (PC), lyso-phosphati-dycholine (LPC) and phosphatidylethanolamine (PE).
  • PC phosphatidylcholine
  • LPC lyso-phosphati-dycholine
  • PE phosphatidylethanolamine
  • the phospholipid and/or lecithin may include about 80 to 85% phosphatidylcholine (PC) + lyso-phosphati-dycholine (LPC), about 7 to about 9.5% phosphatidylethanolamine (PE), and about 2 to about 3 wt% sphingomyelin (SPM).
  • the phospholipid and/or lechithin may include greater than 95% phosphatidylcholine (PC).
  • the ratio of vitamin E 90 (e.g., D,L-a- tocopherol) to one or both of the phospholipid and lecithin may be from about 1:5 to about 5:1, from about 1:4 to about 4:1, from about 1:3 to about 3:1, from about 1:2 to about 2:1, or about 1: 1.
  • the mixture may be heated to fully dissolve one or both of the phospholipid and lecithinat a temperature of about 60°C to about 250°C, or any one of about about 70°C, about 80°C, about 90°C, about 100°C, or about 120°C to any one of about 130°C, about 140°C, about 150°C, about 160°C, about 170°C, about 180°C, about 190°C, or about 200°C.
  • the temperature should be set to facilitate lipid dissolution without burning.
  • a deoxycholic acid may be added to the above mixture before preparation, after preparation or during preparation.
  • the deoxycholic acid is dissolved in water and added to the mixture above.
  • the deoxycholic acid may be sodium deoxycholate.
  • the resultant composition may be mixed to homogenize the composition, e.g., from about 1 minute to about 30 minutes. After homogenization, the mixture may be hydrated, e.g., from about 15 minutes to about 24 hours, or any of about 1 hour, 2 hours, 4 hours, 6 hours, 8 hours or 10 hours to any of about 12 hours, about 14 hours, about 16 hours, about 18 hours, about 20 hours, about 22 hours, or about 24 hours, to form the cubic phase.
  • a separate mixture of a deoxycholic acid may be incorporated.
  • a deoxycholic acid is dissolved in water and incorporated with the cubic phase and mixed until no lamellar phase is observed (e.g. for about 30 minutes to about 8 hours). Once no lamellar phase is observed, glycine and/or water may be added. The final mixture may be microfluidized to form a nano-dispersion.
  • PK pharmacokinetic
  • Ten female beagle dogs were assigned to one of two treatment groups using a standard, by weight, randomization procedure.
  • Iloprost formulated as Aqueous formulation or Aqueuous lipid-based formulation was administered by a single IV infusion to two groups of female beagle dogs (5 animals per group), Group 1 administered with Aqueous formulation, and Group 2 administered with Aqueous lipid-based formulation.
  • Each formulation was administered to animals via a surgically implanted catheter into the femoral vein by a single continuous intravenous infusion for up to 24 hours at a dose level of 5760 ng/kg and a dose volume of 3.6 mL/kg, see Table 1.
  • Plasma samples were analyzed for Iloprost using the validated analytical method based on a liquid-liquid extraction, followed by LC-MS/MS analysis.
  • the bioanalytical assay provided a linear range from 20.0 to 20,000 pg/mL (LLOQ: 20.0 pg/mL) for plasma based on a 200 pL sample volume. Plasma concentrations were reported to three significant figures and exported from the Watson® LIMS.
  • Non-compartmental methods were used to calculate the plasma PK parameters of Iloprost using the PhoenixTM WinNonlin software (version 8.1, Pharsight Corp., Mountain View, CA).
  • PK analysis for individual animals in Groups 1 and 2 were performed using Iloprost plasma concentration data at nominal time points. Mean plasma concentration for each time point was calculated using Watson® LIMS (version 7.4.2). For data below the limit of quantitation (BLQ) before dosing at time zero, a value of zero was assigned for mean calculations. Of note, one sample (animal 2501 at 27-hour point) was not received by MBL, and one sample (animal 2504 at 25.5 hour) was not processed for analysis due to quantity not sufficient for testing. Therefore, these two time points were omitted from PK calculations.
  • AUC 0- ⁇ Reliability of AUC 0- ⁇ were evaluated by the coverage of AUC 0-48 in relation to AUC 0- ⁇ for individual animals and the number of samples used to fit a good regression line in the terminal log-linear phase of the individual concentration-time profiles by PhoenixTM WinNonlin software.
  • Iloprost Aqueous formulation 5760 ng/kg, IV Infusion for up to 24 hours
  • Iloprost Aqueous lipid-based formulation 5760 ng/kg, IV Infusion for up to 24 hours a. Time post start of infusion;
  • Iloprost Aqueous formulation 5760 ng/kg, IV Infusion for up to 24 hours a: Dose normalized C max is calculated as C max Dose b: Dose normalized AUC 0-48 is calculated as AUCo-rs Dose
  • Aqueous lipid-based formulation 5760 ng/kg, IV Infusion for up to 24 hours a: Dose normalized C max is calculated as C max Dose b: Dose normalized AUCo-rs is calculated as AUCo-rs Dose
  • Iloprost formulated as Aqueous formulation and Aqueous lipid-based formulation was administrated in dogs by IV infusion for up to 24 hours. Following an initial of infusion, the plasma C max was observed at 24.03 hours (median T max ) for Group 1 (Aqueous formulation) and at 24.08 hours for Group 2 (Aqueous lipid-based formulation).
  • Mean plasma exposure ( AUC 0-48 ) was 4286 hr*pg/mL for Group 1 and 5359 hr*pg/mL for Group 2.
  • Mean C max was 2602 pg/mL for Group 1 and 3969 pg/mL for Group 2.
  • Estimated elimination half-life (ti/2) in plasma ranged from 0.0303 to 0.797 hour for Group 1 and from 0.0289 to 0.437 hour for Group 2.
  • Total clearance (CL) ranged from 292 to 184111 mL/hr/kg for Group 1 and from 7708 to 292462 mL/hr/kg for Group 2.
  • Apparent volume of distribution (Vd) ranged from 335 to 8038 mL/kg for Group 1 and from 322 to 89971 mL/kg for Group 2.
  • the relative mean plasma exposure ( AUC 0-48 ) and mean C max of Group 2 to Group 1 were 125% and 153%, respectively (Table 5), indicating improved pharmacokinetics characterized by approximate 25% higher exposure (AUC 0-48 ) and 53% higher C max in Aqueous lipid-based formulation compared with Aqueous formulation. It should be noted that small numbers of animals or insufficient qunatificable data points for some animals caused large variations (%CVs of AUC 0-48 and C max ranging from 191.4% to 218.8% in the two groups) and impacted the results.
  • Iloprost peak or near peak concentrations were achieved at 24.03 hours (median T max ) for Group 1 (Aqueous formulation) and at 24.08 hours for Group 2 (Aqueous lipid-based formulation).
  • Mean plasma exposure ( AUC 0-48 ) was 4286 hr*pg/mL for Group 1 and 5359 hr*pg/mL for Group 2.
  • Mean C max was 2602 pg/mL for Group 1 and 3969 pg/mL for Group 2.
  • Estimated elimination half-life (ti/2) in plasma ranged from 0.0303 to 0.797 hour for Group 1 and from 0.0289 to 0.437 hour for Group 2.
  • Total clearance (CL) ranged from 292 to 184111 mL/hr/kg for Group 1 and from 7708 to 292462 mL/hr/kg for Group 2
  • apparent volume of distribution (Vd) ranged from 335 to 8038 mL/kg for Group 1 and from 322 to 89971 mL/kg for Group 2.
  • the relative mean plasma exposure (AUC 0-48 ) and mean C max of Group 2 to Group 1 were 125% and 153%, respectively.
  • a cubic phase was first prepared.
  • D To prepare the cubic phase, 47.24 g of D, L-a-tocopherol and 34.25 g of Phospholipon 90G were mixed to form a lipid mix.
  • the lipid mix was heated on a hot plate set to 90°C to fully dissolve the Phospholipon 90G.
  • 2.68 g of sodium deoxy cholate was dissolved in 27.42 g of sterile water for injection into the lipid mix.
  • the sodium deoxy cholate injection was added to the lipid mix, which was vigorously mixed to form a cubic gel.
  • the cubic gel was then observed under polarized light under a microscope and images were recorded. If lamellar phase is observed, then continue homogenization or kneading using mortar-pestle until most of the lamellar phase disappears. An image of the final cubic gel was recorded before hydration start time and after hydratation completion if necessary. The adjustments should be recorded. The beaker was then covered with parafilm and hydrated overnight for about 18 hours.
  • a blank aqueous lipid dispersion was prepared.
  • a 4L glass beaker 1432.0 g of WFI water and 4.0 g of sodium deoxy cholate was added.
  • the beaker was transferred under a Silverson homogenizer (L5M-A) using a square slot high shear screen, and the stirring speed was set to 5,000 rpm.
  • the solution was mixed until the sodium deoxy cholate was dissolved, about 4-5 minutes.
  • Amount of water Sum of the formulation component added to the beaker (amount of cubic gel + amount of DI water + amount of Sodium deoxycholate + amount of glycine) - amount of dispersion
  • aqueous lipid mixture is prepared accoding to the following process, where the aqueous lipid mixture has a concentration of about 2 mg/mL.
  • a 400 mL beaker 1.103 g of iloprost and 125 g of an aqueous lipid mixture was added.
  • the beaker was secured under a Silverson homogenizer (L5M-A).
  • the small-scale square hole screen homogenizing head was lowered into the solution in the beaker and the solution was mixed at 5,000 rpm. This was performed until all the iloprost was dissolved, i.e. until the iloprost was no longer visible.
  • a magnetic stir bar should be used instead of the homogenizer and the solution should be mixed for a longer interval, at least 2 days, but at a lower speed then was used with the homogenizer.
  • the solution was filter using a Meissner 0.2 pm polypropylene vanguard filter into a 100 mL beaker.
  • a 2mL sample was then prepared for assay to test the concentration of the solution.
  • the sample was found to have a concentration of 6.4 mg/mL.
  • 60.31 g of the 6.4 mg/mL of the solution was diluted to 193.0 g by adding 132.69 g of the aqueous lipid mixture to prepare a 2 mg/mL solution. This was then mixed for about 30 minutes.
  • hPASMCs were isolated from small-resistance arteries obtained from nonimplanted human Caucasian donor lungs. These cells were plated in 96-well plates the day before the experiment, and the cells were serum-starved overnight. The hPASMCs were treated with 3-isobutyl-l -methylxanthine (IBMX lOOpM; an inhibitor of both extracellular and intracellular phosphodiesterases) prior to the exposure with the different Iloprost formulations and respective vehicle controls.
  • IBMX lOOpM 3-isobutyl-l -methylxanthine
  • the Iloprost in an aqueous lipid formulation was used in the concentration range of 1.9nM - 1950nM, while the Iloprost Trometamol solution was used in the concentration range of 2.3nM - 2310 nM.
  • Forskolin (5pL) a permanent cAMP activator, was used as a positive control.
  • the cells were treated, and after 15 minutes, the hPASMCs were rinsed twice with ice-cold PBS (200 ⁇ l), and then IX lysis buffer was added (100 ⁇ l/well). The samples were then stored at -80°C until the cAMP levels were measured using the Cyclic AMP XP® Assay Kit.
  • An “upper solution”, is obtained by dissolving Pluronic F-68 (a polvpropyleneoxide-polvethyleneoxide block copolymer surfactant commercially available from BASF), and acetic acid together and adding to the test tube as a layer of solution above the previous solution that includes the iloprost. Immediately the test tube containing the liquid crystalline mixture and the upper solution is shaken and sonicated for in a small, table-top ultrasonicator (Model FS6, manufactured by Fisher Scientific) to form a dispersion.
  • Pluronic F-68 a polvpropyleneoxide-polvethyleneoxide block copolymer surfactant commercially available from BASF
  • acetic acid acetic acid

Abstract

Disclosed in certain embodiments is a parenteral formulation comprising a therapeutically effective amount of iloprost or a pharmaceutically acceptable salt thereof and a lyotropic liquid crystal material.

Description

COMPOSITIONS AND METHODS FOR VASODILATOR DELIVERY
CROSS REFERENCE TO RELATED APPLICATION(S)
[0001] The present application claims priority to U.S. provisional application No. 63/083,150 filed on September 25, 2020, the entire contents of which are incorporated by reference in their entirety.
FIELD OF THE INVENTION
[0002] In certain embodiments, the present invention relates to the field of pharmaceutical compositions for administering a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof). Other embodiments are directed to methods of treatment and methods of manufacturing the compositions disclosed herein.
BACKGROUND OF THE INVENTION
[0003] Constriction of blood vessels is a serious condition that prevents proper blood flow to surrounding tissues. One disease is pulmonary hypertension which is an elevated blood pressure that affects the arteries in the lungs and the right side of the heart. In pulmonary arterial hypertension (PAH), blood vessels in the lungs are narrowed, blocked or damaged which slows blood flow through the lungs, and blood pressure in the lung arteries becomes elevated. This causes the heart to work harder and results in cardiac complications. Pulmonary hypertension may become worse over time and can be life-threatening. Other diseases associated with constricted blood vessels include scleroderma and Raynaud’s syndrome.
[0004] Iloprost is a vasodilator that can be used to treat diseases associated with constricted blood vessels. The chemical name for iloprost is (E)-(3aS, 4R, 5R, 6aS)-hexahydro-5-hydroxy-4- [(E)(3S,4RS)-3-hydroxy-4-methyl-l-octen-6-ynyl]-Δ2(1H)Δ-pentalenevaleric acid. Racemic iloprost consists of a mixture of the 4R and 4S diastereoisomers at a ratio of approximately 50:50. [0005] Iloprost is commercially available as a solution for inhalation in 1 mL single-use glass ampules containing either 10 mcg/mL or 20 mcg/mL and is administered by a nebulized delivery system. The approved regimen includes dosing every 6-9 hours during waking hours. This is very inconvenient and difficult for compliance and may result in underdosing or overdosing by patients.
[0006] There exists a need in the art for a pharmaceutical composition for providing treatment of constricted blood vessels (e.g., pulmonary arterial hypertension) that avoids the problems of frequent nebulized administration.
OBJECTS AND SUMMARY OF THE INVENTION
[0007] It is an object of certain embodiments of the present invention to provide a pharmaceutical composition comprising a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) for treating disease states such as arterial hypertension, pulmonary arterial hypertension, scleroderma and Reynaud’s disease.
[0008] It is an object of certain embodiments of the present invention to provide an implantable pump that contains the pharmaceutical composition disclosed herein that can release the vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) over an extended period of time.
[0009] It is an object of certain embodiments of the present invention to provide a method of treating diseases associated with constriction of blood vessels (arterial hypertension, pulmonary arterial hypertension, scleroderma and Reynaud’s disease) comprising administration to a patient in need thereof the pharmaceutical compositions (e.g., by an implantable pump) disclosed herein. [0010] It is an object of certain embodiments of the present invention to provide a method of preparing the pharmaceutical compositions and the implantable pumps disclosed herein.
[0011] The above objects and others may be achieved by the present invention which in certain embodiments is directed to a pharmaceutical composition comprising a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable excipient (e.g., a lipid material such as a lyotropic liquid crystal material). In alternative embodiments, the pharmaceutical composition is contained in an implantable pump.
[0012] In certain embodiments, the present invention is directed to the use of a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) in the preparation of a medicament for the treatment of a disease requiring vasodilatory therapy such as hypertension (e.g., pulmonary hypertension). In certain embodiments, the medicament is contained in an implantable pump.
[0013] As used herein, the singular forms “a,” “an,” and “the” include plural references unless the context clearly indicates otherwise. Thus, for example, reference to “a vasodilator” includes a single vasodilator as well as a mixture of two or more different vasodilators; and reference to an “excipient” includes a single excipient as well as a mixture of two or more different excipients, and the like.
[0014] As used herein, the term “about” in connection with a measured quantity or time, refers to the normal variations in that measured quantity or time, as expected by one of ordinary skill in the art in making the measurement and exercising a level of care commensurate with the objective of measurement. In certain embodiments, the term “about” includes the recited number ±10%, such that “about 10” would include from 9 to 11, or “about 1 hour” would include from 54 minutes to 66 minutes.
[0015] As used herein, the terms "therapeutically effective" and an “effective amount” refer to that amount of an active agent or the rate at which it is administered needed to produce a desired therapeutic result.
[0016] The terms “treatment of’ and “treating” include the administration of an active agent(s) with the intent to lessen the severity of a condition.
[0017] The terms “prevention of’ and “preventing” include the avoidance of the onset of a condition by a prophylactic administration of the active agent. [0018] The term “parenteral” means administration by injection, implantation or infiltration by a route such as intravenous, inhalation, intrarterial, intracardiac, intraspinal, intraosseous, intrarticular, intrasynovial, intracutaneous, subcutaneous or intramuscular.
[0019] The term “injection” means administration to a discrete site through the skin or into tissue of a human or animal.
[0020] The term “implantation” means embedding a device and/or pharmaceutical composition into the skin, tissue, muscles, tendons, joints, or other body parts of a human or animal. In certain embodiments, the term refers to a surgical procedure where a device and/or pharmaceutical composition is put into the body and the skin is closed over it.
[0021] The term “aqueous formulation” refers to a water base formulation or compositionwithout a lipid material.
[0022] The term formulation and composition are used interchangeably.
[0023] Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to illuminate certain materials and methods and does not pose a limitation on scope. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosed materials and methods.
BRIEF DESCRIPTION OF THE DRAWINGS
[0024] FIG. 1 is a graphic illustration of Mean (SD) Plasma Concentrations (pg/mL) of Iloprost versus Time Post Start of Infusion in Female Dogs Following a Single Continuous Intravenous Infusion of Aqueous Formulation or Aqueous lipid-based Formulation for up to 24
Hours at 5760 ng/kg of the Pharmokinetic (PK) study. [0025] FIGS. 2 to 6 are a graphic illustration of Individual PK Profiles of Log-transformed
Concentration Values vs Time and Linear Regression to Estimate the Elimination Rate in Dog Plasma after IV Infusion of Iloprost Aqueous Formulation for up to 24 Hours at 5760 ng/kg of the Pharmokinetic (PK) study.
[0026] FIGS. 7 to 11 are a graphic illustration of Individual PK Profiles of Log-transformed Concentration Values vs Time and Linear Regression to Estimate the Elimination Rate in Dog Plasma after IV Infusion of Iloprost Aqueous lipid-based for up to 24 Hours at 5760 ng/kg of the Pharmokinetic (PK) study.
[0027] FIG. 12 is an image of the optical microscopcy of the blank aqueous lipid mixture of the Preparation example.
[0028] FIG. 13 is an image of the optical microscopy of the Manufacturing Example.
[0029] FIG. 14 is a bargraph illustrating intracellular cAMP levels in isolated hPASMCs upon treatment with Iloprost in an aqueous lipid formulation and aqueous lipid dispersion.
[0030] FIG. 15 is a bargraph illustrating intracellular cAMP levels in isolated hPASMCs upon treatment with Iloprost Trometamol solution.
[0031] FIG. 16 is a line graph illustrating intracellular cAMP levels in isolated hPASMCs upon treatment with Iloprost in an aquous lipid formulation with Iloprost Trometamol formulation.
DETAILED DESCRIPTION
[0032] In certain embodiments, the invention is directed to a pharmaceutical composition comprising a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) that can be administered (e.g., by implantable pump) to facilitate the treatment of relevant disease states.
[0033] In certain embodiments, the pharmaceutical composition comprises iloprost or a pharmaceutically acceptable salt thereof and a lipid material (e.g., a lyotropic liquid crystal material). In certain embodments, the pharmaceutical composition is an aqueous lipid solution). In certain embodiments, the lyotropic liquid crystal material comprises both polar and non-polar nano-domains. In other embodiments, the material comprises nanostructured nonlamellar liquid crystalline phases. In alternative embodiments, the material comprises reversed nanostructured liquid crystalline phases. In certain embodiments, the material comprises distinct nanostructured nonlamellar liquid crystalline material and a liquid phase embedded within said distinct nanostructured nonlamellar liquid crystalline material, said liquid phase being selected from the group consisting of a hydrophobe-rich phase and a polar solvent-rich phase.
[0034] In certain embodiments, the pharmaceutical composition disclosed herein is contained in an implantable pump that can deliver the active agent for a time period of, e.g., at least 3 days, at least 7 days, at least 10 days, at least 14 days, at least 21 days, at least 28 days, at least 30 days, at least 45 days, at least 60 days, at least 90 days, at least 100 days, at least 120 days, at least 150 days, at least 180 days, at least 200 days, at least 210 days, at least 240 days, at least 270 days, at least 300 days, or at least one year.
[0035] In certain embodiments, the pharmaceutical composition disclosed herein is contained in an implantable pump that can deliver the active agent for a time period of, e.g., about 3 days, about 7 days, about 10 days, about 14 days, about 21 days, about 28 days, about 30 days, about 45 days, about 60 days, about 90 days, about 100 days, about 120 days, about 150 days, about 180 days, about 200 days, about 210 days, about 240 days, about 270 days, about 300 days, or about one year.
[0036] In certain embodiments, the pharmaceutical composition disclosed herein is contained in an implantable pump that can deliver the active agent for a time period of, e.g., from about 3 days to about 400 days, from about 7 day to about 200 days, or from about 30 days to about 180 days.
[0037] In certain embodiments, the formulation (e.g., a lipid formulation such as a lyotropic liquid crystal formulation) of the vasodilator (e.g., iloprost or pharmaceutically acceptable salt thereof) prolongs the half-life of the drug. This provides a longer duration of action that may decrease the frequency of dosing and improve patient compliance. This may be beneficial with implantable pumps that provide intermittent or pulsed dosing. In such embodiments, the pulses can be spread apart and there can be a prolonged time between refills.
[0038] In other embodiments, the formulation disclosed herein increases the potency of the drug. For example, it may increase the potency of iloprost or pharmaceutically acceptable salt thereof on prostaglandin receptors. This may decrease the minimum effective dose and result in less drug being administered and/or decrease the frequency of dosing.
[0039] In some embodiments, the formuation dislcosed herein may have a pH of about 6 to about 10, about 7 to about 9 or about 7.5 to 8.5.
[0040] In some embodiments, the formulation disclosed herein may have a density of about 0.8 to about 1.2 g/mL, about 0.9 to about 1.1 g/mL or about 1 g/mL.
[0041] In some embodiments, the formulation disclosed herein may have a particle size distribution (D90) of the drug of about 0.1 nm to about 10 nm, about 0.5 nm to about 5 nm about 1 nm to about 3 nm, about 1.25 nm to about 2.75 nm, about 1.5 nm to about 2.5 nm, or about 1.75 nm to about 2.25 nm.
[0042] In some embodiments, the formulation disclosed herein provides iloprost or pharmaceutically acceptable salt thereof to the patient in a daily amount from about 1000 ng/kg to about 10,000 ng/kg, from about 2000 ng/kg to about 8000 ng/kg, from about 5000 ng/kg to about 6500 ng/kg, from about 5250 ng/kg to about 6250 ng/kg, from about 5500 ng/kg to about 6000 ng/kg, or from about 5700 ng/kg to about 5800 ng/kg.
[0043] In some embodiments, the formulation disclosed herein is administered daily, twice daily, three times daily, 4 times daily, 6 times daily, once weekly, twice weekly, three times weekly, 4 times weekly or any oyjer suitable disong regimin. In other embodiments, the administration is continuous (e.g., by intravenous infusion) over a time period of about 30 minutes to about 7 days, about 1 hour to about 3 days, about 2 hours to about 2 days, or about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 12 hours or about 24 hours. [0044] In some embodiments, formulation disclosed herein provides a maximum plama concentration (Cmax ) of iloprost that is higher when compared to the adminsitartiion of a comparable aqueous formulation. In some embodiments, the Cmax provided by the present formulation is up to about 100% higher, up to about about 90% higher, up to about 80% higher, up to about 75% higher, up to about 65% higher, up to about 50% higher, up to about 40% higher, up to about 25% higher, up to about 15% higher, up to about 10% higher or up to about 5% higher than a comparable aqueuous formulation.
[0045] In some embodiments, the formulation disclosed herein provides a half-life of iloprost that decreases as compared to a comparable aqueous formulation. In certain embodiments, the decrease is about 75% or less, about 50% or less, about 35% or less, about 25% or less, about 10% or less or about 5% or less as compared to an aqueous formulation.
[0046] In some embodiments, the formulation disclosed herein provides a total clearance of iloprost that increases when compared to an aqueous formulation. In certain embodiments, the total clearance provided by the present formulation is up to about 100% higher, up to about about 90% higher, up to about 80% higher, up to about 75% higher, up to about 65% higher, up to about 50% higher, up to about 40% higher, up to about 25% higher, up to about 15% higher, up to about 10% higher or up to about 5% higher than a comparable aqueuous formulation
[0047] In some embodiments, the formulation disclosed herein has a relative mean plasma exposure (AUC0-48) that increases as compared to a comparable aqueous formulation. In certain embodiments, the (AUC0-48) provided by the present formulation is up to about 100% higher, up to about about 90% higher, up to about 80% higher, up to about 75% higher, up to about 65% higher, up to about 50% higher, up to about 40% higher, up to about 25% higher, up to about 15% higher, up to about 10% higher or up to about 5% higher than a comparable aqueuous formulation. [0048] The formulation may be used to treat any condition to provide vasodilator therapy or iloprost therapy such as pulmonary arterial hypertension, or hypertension. [0049] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 10 nM to about 40 nM, or about 20 nM to about 30 nM at a concentration of about 1950 nM iloprost.
[0050] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 15 nM, or about 4 nM to about 12 nM at a concentration of about 650 nM iloprost.
[0051] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 15 nM, or about 4 nM to about 12 nM at a concentration of about 195 nM iloprost.
[0052] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 15 nM, or about 4 nM to about 12 nM at a concentration of about 65 nM iloprost. In some embodiments, the formulation disclosed herein provides an increase in cAMP in human PASMCs when compared to an aqueous formulation iloprost.
[0053] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 5 nM to about 30 nM, or about 10 nM to about 20 nM at a concentration of about 2310nm iloprost.
[0054] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 20 nM, or about 5 nM to about 15 nM at a concentration of about 770 nM iloprost.
[0055] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 12 nM, or about 5 nM to about 10 nM at a concentration of about 231 nM iloprost.
[0056] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of from about 2 nM to about 12 nM, or about 5 nM to about 10 nM at a concentration of about 77 nM iloprost. [0057] In certain embodiments, the iloprost lipid formulation of the present invention provides intracellular cAMP levels in isolated hPASMCs of at least about 15 nM, at at least about 20 nM, from about 15 nM to about 50 nM, from about 20 nM to about 30 nM, or from about 22 nM to about 28 nM at a concentration of about 1000 nM to about 3000 nM, about 1500 nM to about 2500 nM, about 1900 nM to about 2400 nM or about 1950 nM or about 2310 nM iloprost.
Implantable Pump
[0058] In certain embodimemnts, the composition disclosed herein can be contained in an implantable pump. The pump may provide a continuous release or a pulsed release of the drug. The pulses may be, e.g., about every 2 hours, about every 4 hours, about every 6 hours, about every 8 hours, about every 12 hours, about every 24 hours, about every 48 hours, about every 72 hours or about every 168 hours.
[0059] The implantable pump may be implanted in any suitable body space such as the abdomen, neck area (e.g., under the collarbone), buttock, thigh or upper arm. In one embodiment, the composition is released from the pump into the surrounding tissue and the drug (e.g., iloprost or pharmaceutically acceptable salt thereof) is subsequently absorbed into the circulatory system. In another embodiment, a catheter delivers the composition from the device (e.g., implanted in the abdomen or under the collarbone) directly into the circulatory system (e.g., to the venous system such as subclavian vein or the subjugular vein).
[0060] In one embodiment, the pump is in the form of an implantable infusion device that comprises a lightweight, inert housing that contains a pharmaceutical composition disclosed herein for administration to a patient, and an enclosed, gas-impermeable variable volume chamber which contains a pressure source, such as a propellant or mechanical spring. The variable volume chamber may be in the form of a rigid-walled expandable bellows structure or a nonstretchable flexible bag and is attached to the housing so as not to obstruct the entry port. A recharging fluid flow path may be provided adjacent to where the variable volume chamber is attached to the housing. The composition is delivered from the device via a catheter in response to expansion of the variable volume chamber against the volume of infusate in the housing. A separate bolus injection port is provided which allows infusate to be safely introduced directly into the catheter and overrides the controlled pressure-driven delivery. The lightweight housing may include integrally formed needle stops and suture fastening loops.
[0061] In another embodiment, the device is in the form of an implantable infusion device comprising a housing including an inner wall and having an interior region defined at least in part by the inner wall, at least a portion of the interior region defining an pharmaceutical composition reservoir adapted to store a pharmaceutical composition as disclosed herein for delivery to a patient; a closed variable volume chamber having an outer wall, the variable volume chamber disposed within the interior region of the housing with the outer wall of the variable volume chamber being spaced from the inner wall of the housing, the variable volume chamber being supported within the interior region from an internal portion of the inner wall of the housing; a pressure source contained within the variable volume chamber; a pharmaceutical composition recharging fluid flow path adapted to recharge the infusate reservoir by carrying infusate to the infusate reservoir through the internal portion of the housing from which the variable volume chamber is supported within the interior region; and an exit fluid flow path adapted to deliver infusate from the infusate reservoir out of the housing, e.g., directly into the circulatory system.
[0062] In one embodiment, the implantable pump comprises a filter for microbial agents. The filter can comprise a material such as, e.g., plastic, ceramic, glass or fiber. In certain embodiments, the filter comprises plastic and does not comprise polyvinylidene fluoride. In other embodiments, the filter does not comprise plastic.
[0063] Exemplary infusion pumps that can be adapted for use in the devices, systems (device/pharmaceutical composition combinations) and methods disclosed herein are described in US2003/0208184; US2014/0228765; US2014/0194851; US2013/0023857; US3, 840,009; US5,167,633; US5, 514103; US3,951,147; US5,045,064; US5,395,324; US5,769,823; US5,575,770; US10, 173,004; US10, 010670; US9,968,734; US9,700,669, US9, 180,282; US9, 125,982; US8,551,044; US8,545,477 and US 8,273,058, the disclosures of which are hereby incorporated by reference for all purposes.
An Alternative Embodiment
[0064] In certain embodiments, the pharmaceutical formulation comprising iloprost or a pharmaceutically acceptable salt thereof; a carrier; and a particle or material comprising a distinct nanostructured nonlamellar liquid crystalline material; and one or more pockets or droplets of a liquid phase embedded within said distinct nanostructured nonlamellar liquid crystalline material, said liquid phase being selected from the group consisting of an oil-rich liquid phase and a polar solvent-rich liquid phase.
[0065] In certain embodiments, the distinct nanostructured nonlamellar liquid crystalline material is a reversed phase nonlamellar liquid crystalline material.
[0066] In certain embodiments, the distinct nanostructured nonlamellar liquid crystalline material comprises a reversed hexagonal phase material, a reversed bicontinuous cubic phase material, a reversed discrete cubic phase material, or a reversed intermediate phase material.
[0067] In certain embodiments, the distinct nanostructured nonlamellar liquid crystalline material is polymerized.
[0068] In certain embodiments, the pharmaceutical formulation further comprises a stabilizing layer exterior to said particle or material. In certain embodiments, the stabilizing layer is selected from the group consisting of a charged moiety, a polymer, and a surfactant.
[0069] In certain embodiments, the particle or material further comprises a coating comprising the vasodilator (e.g., iloprost or pharmaceutically acceptable salt thereof).
[0070] In certain embodiments, the liquid phase is an oil and said oil is selected from the group consisting of benzyl benzoate, estragole, eugenol, isoeugenol, linalool, and the essential oils of basil, bay, bois de rose (rosewood), carrot seed, clovebud, eucalyptus, ginger, grapefruit, hyssop, lemon, balsam of Peru, mugwort, myrrh gum, biter orange, oregano, palmarosa, patchouly, peppermint, petitgrain, rosemary, santalwood oil, spearmint, thuja (cedar leaf), thyme, vanilla, and ylang ylang (cananga).
[0071] In certain embodiments, the liquid phase is a polar solvent and said polar solvent is selected from the group consisting of water, glycerol, and N,N-dimethylacetamide.
[0072] In certain embodiments, the vasodilator (e.g., iloprost or pharmaceutically acceptable salt thereof) is dissolved or dispersed in the liquid phase or in the distinct nanostructured nonlamellar liquid crystalline material.
[0073] In certain embodiments, the one or more pockets or droplets have a diameter of 50 nm or greater.
[0074] In certain embodiments, the liquid phase includes at least one of an oil and a polar solvent.
[0075] In certain embodiments, the liquid phase is a hydrophobe-rich phase or a polar solvent- rich phase.
[0076] In certain embodiments, the distinct nanostructured nonlamellar liquid crystalline material consists essentially of a reversed bicontinuous cubic phase material.
[0077] In certain embodiments, the distinct nanostructured nonlamellar liquid crystalline material comprises or consists essentially of a reversed discrete cubic phase material.
[0078] In certain embodiments, the distinct nanostructured nonlamellar liquid crystalline material comprises a water insoluble lipid or surfactant.
A Further Alternative Embodiment
[0079] In certain embodiments, the pharmaceutical formulation comprises a vasodilator (e.g., iloprost or a pharmaceutically acceptable salt thereof) and a coated particle comprising a) an interior core comprising a matrix comprising of i) at least one nanostructured liquid phase or a dehydrated variant thereof, ii) at least one nanostructured liquid crystalline phase or a dehydrated variant thereof or iii) a combination of (1) at least one nanostructured liquid phase or a dehydrated variant thereof and (2) at least one nanostructured liquid crystalline phase or a dehydrated variant thereof and b) an exterior coating comprising nonlamellar domains wherein the iloprost or pharmaceutically acceptable salt thereof is in a), b) or a combination thereof.
[0080] In certain embodiments, the nanostructured liquid phase material comprises a nanostructured LI phase material, a nanostructured L2 phase material, a microemulsion that is nanostructured, or a nanostructured L3 phase material.
[0081] In certain embodiments, the nanostructured liquid phase material comprises a nanostructured normal or reversed cubic phase material, a nanostructured normal or reversed hexagonal phase material, a nanostructured normal or reversed intermediate phase material, or nanostructured lamellar phase material.
[0082] In certain embodiments, the nanostructured liquid phase material comprises a polar solvent and a surfactant or a lipid.
[0083] In certain embodiments, the nanostructured liquid phase material comprises a polar solvent, a surfactant or a lipid and an amphiphile or hydrophobe.
[0084] In certain embodiments, the nanostructured liquid phase material comprises a block copolymer.
[0085] In certain embodiments, the nanostructured liquid phase material comprises a block copolymer and a solvent.
[0086] In certain embodiments, the nanostructured liquid phase material comprises a polar solvent and a surfactant.
[0087] In certain embodiments, the nanostructured liquid phase material comprises a polar solvent, a surfactant and an amphiphile or hydrophobe.
[0088] In certain embodiments, the interior core comprises the vasodilator (e.g., iloprost or pharmaceutically acceptable salt thereof) disposed within said matrix.
[0089] In certain embodiments, the interior core comprises a reversed cubic phase material.
[0090] In certain embodiments, the nonlamellar domain is amorphous. [0091] In certain embodiments, the nonlamellar domain is a polymer such as polylactic glycolic acid.
[0092] In certain embodiments, the nonlamellar domain comprises a sugar such as trehalose.
[0093] In certain embodiments, the exterior coating comprises a semi-crystalline nonlamellar material.
[0094] In certain embodiments, the exterior coating comprises at least 2% nonlamellar domains, at least 10% nonlamellar domains or at least 50% nonlamellar domains.
[0095] In certain embodiments, the exterior coating comprises the vasodilator (iloprost or pharmaceutically acceptable salt thereof).
Active Agents
[0096] The pharmaceutical compositions and implants disclosed herein include iloprost or a pharmaceutically acceptable salt thereof. Pharmaceutically acceptable salts include, but are not limited to, inorganic acid salts such as hydrochloride, hydrobromide, sulfate, phosphate and the like; organic acid salts such as formate, acetate, trifluoroacetate, maleate, tartrate and the like; sulfonates such as methanesulfonate, benzenesulfonate, p-toluenesulfonate, and the like; amino acid salts such as arginate, asparginate, glutamate and the like, and metal salts such as sodium salt, potassium salt, cesium salt and the like; alkaline earth metals such as calcium salt, magnesium salt and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, tromethamine salt, triethanolamine salt, dicyclohexylamine salt, N,N'- dibenzylethylenediamine salt and the like. In one embodiment, the iloprost is iloprost base.
[0097] Other vasodilators that can be utilized in the present invention (alone or in combination with iloprost) include an alpha-adrenergic receptor antagonist (a-blocker), an angiotensin converting enzyme (ACE) inhibitor, an angiotensin receptor blocker (ARB), a beta 2 -adrenergic receptor agonist ( 2 -agonist), Calcium channel blocker (CCB), centrally acting sympathetic blocker, direct acting vasodilator, endothelin receptor antagonist, ganglion blocker, nitrodilator, phosphodiesterase inhibitor, potassium channel opener, renin inhibitor or combination thereof. [0098] Particular agents include prazosin, terazosin, doxazosin, trimazosin, phentolamine, phenoxybenzamine, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, quinapril, ramipril, candesartan, eprosartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, irbesartan, epinephrine, norepinephrine, dopamine, dobutamine, isoproterenol, amlodipine, felodipine, isradipine, nicardipine, nifedipine, nimodipine, nitrendipine, clonidine, guanabenz, guanfacine, α-methyldopa, hydralazine, bosentan, trimethophan disilicate, isosorbide mononitrate, nitroglycerin, erythritol tetranitrate, pentaerythritol tetranitrate, sodium nitroprusside, milrinone, inamrinone, cilostazol, sildenafil, tadalafil, minoxidil, aliskiren, pharmaceutically acceptable salts thereof and mixtures thereof.
Pharmaceutically Acceptable Excipients
[0099] The pharmaceutical compositions according to the present invention may comprise one or more pharmaceutically acceptable carriers and excipients appropriate for parenteral administration. Examples of possible pharmaceutically acceptable carriers and excipients are described in the Handbook of Pharmaceutical Excipients, American Pharmaceutical Association (6th Edition, 2009 Publication), which is incorporated by reference herein. Carriers and excipients suitable for parenteral compositions include antioxidants, buffering agents, diluents, surfactants, solubilizers, stabilizers, hydrophilic polymers, additional absorption or permeability enhancers, preservatives, osmotic agents, isotonicity agents, pH adjusting agents, solvents, co-solvents, viscosity agents, gelling agents, suspending agents or combinations thereof.
[0100] Suitable surfactants for the formulations disclosed herein include, but are not limited to Polysorbate 80 NF, polyoxyethylene 20 sorbitan monolaurate, polyoxyethylene (4) sorbitan monolaurate, polyoxyethylene 20 sorbitan monopalmitate, polyoxyethylene 20 sorbitan monostearate, polyoxyethylene (4) sorbitan monostearate, polyoxyethylene 20 sorbitan tristearate, polyoxyethylene (5) sorbitan monooleate, polyoxyethylene 20 sorbitan trioleate, polyoxyethylene 20 sorbitan monoisostearate, sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monostearate, sorbitan trilaurate, sorbitan trioleate, sorbitan tristearate, and the like, and combinations thereof.
Implantable Formulations
[0101] In certain embodiments, the pharmaceutical composition is used in an implantable pump, implantable depot or similar implantable device that can provide administration of the active agent over an extended period of time. In such embodiments, it may be necessary to have a concentrated formulation in order to have the a suitable amount of active agent in a practical size for implant.
[0102] In certain iloprost formulations, the iloprost is in a concentration from about 5 mg/mL to about 50 mg/mL, about 6 mg/mL to about 40 mg/mL, about 7 mg/mL to about 40 mg/mL, about 8 mg/mL to about 30 mg/mL, about 10 mg/mL to about 20 mg/mL, about 5 mg/mL to about 15 mg/mL or about 15 mg/mL to about 25 mg/mL. In other embodiments, the concentration is about 5 mg/mL, about 6 mg/mL, about 7 mg/mL, about 8 mg/mL, about 9 mg/mL, about 10 mg/mL, about 11 mg/mL about 12 mg/mL.
[0103] In certain embodiments, the device for implant can contain from about 5 mL to about 30 mL, about 10 mL to about 20 mL or about 15 mL to about 25 mL. In other embodiments the device for implant can contain about 10 mL, about 15 mL, about 20 m, about 25 mL, about 30 mL, about 40 mL or about 50 mL.
[0104] In certain embodiments, it may be necessary to include a solubilizing agent such as an alcohol (e.g., benzyl alcohol or ethyl alcohol), a polyethylene glycol or dimethylsulfoxide (DMSO).
[0105] The disclosure of US Patent Publication Nos. 2003/0022242 and 2003/0108743 are hereby incorporated by reference for all purposes.
Prepraration of Lipid Mixture
[0106] In some embodiments, the lipid mixture of the present disclosure may be prepared according to the following method. To prepare the lipid mixture, a cubic phase is first prepared. In some embodiments, the cubic phase is prepared by mixing one or more of the following ingredients: vitamin E, a phospholipid and/or a lecithin. The vitamin E may be a tocopherol, such as D,L-a-tocopherol. The phospholipid may be derived from soybean, rapeseed (canola), egg or sunflower. The lecithin may be naturally occurring and/or hydrogenated lecitihin fractions. In certain embodiments, the phospholipid and/or lecithin may be Phospolipon ®, such as Phospholipon 90G, or Phospholipon 85G, LIPOID SI 00 or LIPOID E80. In some embodiments, the phospholipid may be from LIPOID ®, such as LIPOID SI 00 or LIPOID E80. In some embodiments, the phospholipid and/or lecithin may include one or more of the following ingredients: phosphatidylcholine (PC), lyso-phosphati-dycholine (LPC) and phosphatidylethanolamine (PE). In some embodiments, the phospholipid and/or lecithin may include about 80 to 85% phosphatidylcholine (PC) + lyso-phosphati-dycholine (LPC), about 7 to about 9.5% phosphatidylethanolamine (PE), and about 2 to about 3 wt% sphingomyelin (SPM). In some embodiments, the phospholipid and/or lechithin may include greater than 95% phosphatidylcholine (PC). In some embodiments, the ratio of vitamin E 90 (e.g., D,L-a- tocopherol) to one or both of the phospholipid and lecithin may be from about 1:5 to about 5:1, from about 1:4 to about 4:1, from about 1:3 to about 3:1, from about 1:2 to about 2:1, or about 1: 1.
[0107] The mixture may be heated to fully dissolve one or both of the phospholipid and lecithinat a temperature of about 60°C to about 250°C, or any one of about about 70°C, about 80°C, about 90°C, about 100°C, or about 120°C to any one of about 130°C, about 140°C, about 150°C, about 160°C, about 170°C, about 180°C, about 190°C, or about 200°C. In certain embodiments, the temperature should be set to facilitate lipid dissolution without burning.
[0108] In certain embodiments, a deoxycholic acid may be added to the above mixture before preparation, after preparation or during preparation. In one embodiment, the deoxycholic acid is dissolved in water and added to the mixture above. The deoxycholic acid may be sodium deoxycholate. The resultant composition may be mixed to homogenize the composition, e.g., from about 1 minute to about 30 minutes. After homogenization, the mixture may be hydrated, e.g., from about 15 minutes to about 24 hours, or any of about 1 hour, 2 hours, 4 hours, 6 hours, 8 hours or 10 hours to any of about 12 hours, about 14 hours, about 16 hours, about 18 hours, about 20 hours, about 22 hours, or about 24 hours, to form the cubic phase.
After formation of the cubic phase, a separate mixture of a deoxycholic acid may be incorporated. In some embodiments, a deoxycholic acid is dissolved in water and incorporated with the cubic phase and mixed until no lamellar phase is observed (e.g. for about 30 minutes to about 8 hours). Once no lamellar phase is observed, glycine and/or water may be added. The final mixture may be microfluidized to form a nano-dispersion.
Examples
[0109] Specific embodiments of the disclosure will now be demonstrated by reference to the following examples. It should be understood that these examples are disclosed solely by way of illustrating the disclosure and should not be taken in any way to limit the scope of the present disclosure.
Pharmacokinetic (PK) Profile Study
[0110] A study was performed to evaluate and compare the pharmacokinetic (PK) profiles of iloprost in two different formulations, (1) an aqueous iloprost and (2) an aqueuous lipid iloprost, following a single intravenous (IV) dose in beagle dogs.
Study Design
[0111] Ten female beagle dogs were assigned to one of two treatment groups using a standard, by weight, randomization procedure. Iloprost formulated as Aqueous formulation or Aqueuous lipid-based formulation was administered by a single IV infusion to two groups of female beagle dogs (5 animals per group), Group 1 administered with Aqueous formulation, and Group 2 administered with Aqueous lipid-based formulation. Each formulation was administered to animals via a surgically implanted catheter into the femoral vein by a single continuous intravenous infusion for up to 24 hours at a dose level of 5760 ng/kg and a dose volume of 3.6 mL/kg, see Table 1.
Table 1 Grouping and Dosing Information
Figure imgf000021_0001
No. = Number; Cone. = Concentration a Based on the most recent practical body weight measurement. b Dose is expressed as free base equivalent (0.1 mg/mL iloprost as free acid). c Dose is expressed as free base equivalent (8.1 mg/mL iloprost).
[0112] On Day 1, blood samples (2.5 mL) were collected via the jugular, or other suitable vein from 5 animals/group for Groups 1 and 2 pre-dose (0 hour) and at 23.5 and 23.83 hours (just 30 and 10 minutes prior to the end of the 24 hour infusion) after the start of infusion, and further plasma samples were taken at 2, 5, 10, 15, 25, 45 minutes, and 1, 1.25, 1.5, 2, 2.5, 3 hours post- infusion, presented in Table 2. K2EDTA was used as the anticoagulant. The whole blood was centrifuged under refrigerated (2 to 8°C) conditions to prepare plasma. The plasma was placed into 2 aliquots (the primary aliquot contained a target volume of 600 μL; the secondary aliquot contained any residual plasma remaining). Samples were flash frozen on dry ice and transferred to a freezer set to maintain -60 to -90°C. The primary aliquot were shipped on dry ice to MBL for analysis, and the remaining samples (the secondary aliquot) were retained at the Testing Facility.
Table 2 Blood Sampling Schedule for Pharmacokinetic Assessments
Figure imgf000022_0001
a. A single continuous intravenous infusion for up to 24 hours; b. Time prior to the end of infusion Bioanalysis
[0113] Plasma samples were analyzed for Iloprost using the validated analytical method based on a liquid-liquid extraction, followed by LC-MS/MS analysis. The bioanalytical assay provided a linear range from 20.0 to 20,000 pg/mL (LLOQ: 20.0 pg/mL) for plasma based on a 200 pL sample volume. Plasma concentrations were reported to three significant figures and exported from the Watson® LIMS.
[0114] Pharmacokinetic Analysis
[0115] Non-compartmental methods were used to calculate the plasma PK parameters of Iloprost using the Phoenix™ WinNonlin software (version 8.1, Pharsight Corp., Mountain View, CA).
[0116] PK analysis for individual animals in Groups 1 and 2 were performed using Iloprost plasma concentration data at nominal time points. Mean plasma concentration for each time point was calculated using Watson® LIMS (version 7.4.2). For data below the limit of quantitation (BLQ) before dosing at time zero, a value of zero was assigned for mean calculations. Of note, one sample (animal 2501 at 27-hour point) was not received by MBL, and one sample (animal 2504 at 25.5 hour) was not processed for analysis due to quantity not sufficient for testing. Therefore, these two time points were omitted from PK calculations.
[0117] The following parameters were derived: maximum concentration (Cmax ), time of maximum concentration (Tmax), terminal half-life (ti/2), total clearance (CL), apparent volume of distribution (Vd), and area under the plasma concentration versus time curve from time zero to 48 hours (AUC0-48), to the last quantifiable concentration (AUCiast) and to infinity ( AUC0-∞ ) using the linear trapezoidal rule. For a few cases (animals 1502, 2501 and 2503), ti/2, AUC0-∞, CL and Vd were not estimated due to lack of measureable data points or the last quantifiable concentration is Cmax . Reliability of AUC0-∞ were evaluated by the coverage of AUC0-48 in relation to AUC0-∞ for individual animals and the number of samples used to fit a good regression line in the terminal log-linear phase of the individual concentration-time profiles by Phoenix™ WinNonlin software. The relative bioavailability of Iloprost between the Aqueous formulation and Aqueous lipid-based formulation was evaluated by comparing the ratios for mean AUC0-48 and mean Cmax .
[0118] Mean, standard deviation, %CV, minimum, median, maximum and geometric mean values were obtained by the Phoenix™ WinNonlin software and/or Excel where applicable (Microsoft Office 365 version 1908). Data and descriptive statistics (e.g., mean, SD and %CV) are displayed in tables and figures as appropriate, t1/2, Cmax, CL, Vd and AUC were displayed with no decimal place or with 3 significant figures, Tmax was displayed with the designated number of decimal places.
[0119] The numerical data presented in this report were computer-generated. Due to rounding, recalculation of derived values from individual data presented in this report may, in some instances, yield minor variations.
Results
[0120] Analysis results showed that there were no detectable levels of Iloprost in any pre- dose plasma samples (Table 3 and Table 4).
Table 3 Plasma Iloprost Concentrations (pg/mL) in Female Dogs Following a Single Continuous Intravenous Infusion of Aqueous Formulation at 5760 ng/kg (Group 1, Day 1)
Iloprost Aqueous formulation , 5760 ng/kg, IV Infusion for up to 24 hours
Figure imgf000024_0001
Figure imgf000025_0001
a. Time post start of infusion
BLQ: Below the limit of quantitation (Iloprost <20.0 pg/mL)
Table 4 Plasma Iloprost Concentrations (pg/mL) in Female Dogs Following a Single Continuous Intravenous Infusion of Aqueous lipid-based Formulation at 5760 ng/kg (Group 2, Day 1)
Iloprost Aqueous lipid-based formulation , 5760 ng/kg, IV Infusion for up to 24 hours
Figure imgf000025_0002
a. Time post start of infusion;
NC: No sample received for analysis;
QNS: Quantity not sufficient for testing
BLQ: Below the limit of quantitation (Iloprost <20.0 pg/mL) [0121] The individual and mean plasma concentrations of Iloprost with proposed sampling times following a single continuous intravenous infusion of Aqueous formulation or Aqueous lipid-based formulation at 5760 ng/kg on Day 1 are provided in Table 3 and Table 4. Mean plasma Iloprost concentration versus proposed sampling time plots are shown in Error! Reference source not found..
[0122] A summary of mean Iloprost plasma PK parameters derived using non- compartmental methods are shown in Table 5. PK parameters for individual animals in plasma are presented in Error! Reference source not found, through Error! Reference source not found.. The reliability of AUC0-∞ of Iloprost in plasma was evaluated by the coverage of AUC0-
48 in relation to AUC0-∞ in
[0123] Table 1, and individual PK profiles of log-transformed concentration values vs nominal sample collection time and linear regression to estimate the elimination rate in dog plasma after IV infusion of Aqueous formulation or Aqueous lipid-based formulation are presented in Error! Reference source not found, to 6 and Figures 7 to 11, respectively.
Table 5 Summary of Mean Pharmacokinetic Parameters for Iloprost in Plasma from Female Dogs Following a Single Continuous Intravenous Infusion of Aqueous Formulation or Aqueous lipid-based Formulation for up to 24 Hours at 5760 ng/kg on Day 1
Figure imgf000026_0001
Figure imgf000027_0002
a: Median (Min-Max) was used for Tmax b: Dose normalized Cmax, is calculated as Cmax, + Dose c: Dose normalized is calculated as AUC0-48 ÷Dose d: Ratio of Mean C (Aqueous) + Mean C (Aqueous lipid-based) x 100% e: Ratio of Mean (Aqueous lipid-based) + Mean (Aqueous) x 100%
Table 6 PK Parameters for Iloprost in Dog Plasma after IV Infusion of Iloprost Aqueous Formulation at 5760 ng/kg (Group 1, Day 1)
Iloprost Aqueous formulation , 5760 ng/kg, IV Infusion for up to 24 hours
Figure imgf000027_0001
Figure imgf000028_0001
a: Dose normalized Cmax is calculated as Cmax Dose b: Dose normalized AUC0-48 is calculated as AUCo-rs Dose
N/A: Not applicable due to lack of measureable data points or the last quantifiable concentration is Cmax
Table 7 PK Parameters for Iloprost in Dog Plasma after IV Infusion of Iloprost Aqueous lipid- based Formulation at 5760 ng/kg (Group 2, Day 1)
Aqueous lipid-based formulation, 5760 ng/kg, IV Infusion for up to 24 hours
Figure imgf000028_0002
Figure imgf000029_0001
a: Dose normalized Cmax is calculated as Cmax Dose b: Dose normalized AUCo-rs is calculated as AUCo-rs Dose
N/A: Not applicable due to lack of measureable data points or the last quantifiable concentration is Cmax
Pharmacokinetics of Iloprost in Dog Plasma
[0124] Iloprost formulated as Aqueous formulation and Aqueous lipid-based formulation was administrated in dogs by IV infusion for up to 24 hours. Following an initial of infusion, the plasma Cmax was observed at 24.03 hours (median Tmax) for Group 1 (Aqueous formulation) and at 24.08 hours for Group 2 (Aqueous lipid-based formulation). Mean plasma exposure ( AUC0-48) was 4286 hr*pg/mL for Group 1 and 5359 hr*pg/mL for Group 2. Mean Cmax was 2602 pg/mL for Group 1 and 3969 pg/mL for Group 2. Estimated elimination half-life (ti/2) in plasma ranged from 0.0303 to 0.797 hour for Group 1 and from 0.0289 to 0.437 hour for Group 2. Total clearance (CL) ranged from 292 to 184111 mL/hr/kg for Group 1 and from 7708 to 292462 mL/hr/kg for Group 2. Apparent volume of distribution (Vd) ranged from 335 to 8038 mL/kg for Group 1 and from 322 to 89971 mL/kg for Group 2. The relative mean plasma exposure ( AUC0-48) and mean Cmax of Group 2 to Group 1 were 125% and 153%, respectively (Table 5), indicating improved pharmacokinetics characterized by approximate 25% higher exposure (AUC0-48) and 53% higher Cmax in Aqueous lipid-based formulation compared with Aqueous formulation. It should be noted that small numbers of animals or insufficient qunatificable data points for some animals caused large variations (%CVs of AUC0-48 and Cmax ranging from 191.4% to 218.8% in the two groups) and impacted the results.
[0125] The individual ratios of AUC0-48 to AUC0-∞ of Iloprost for animals ranged from 0.960 to 1.00, except for animal 2502 (0.741) (
[0126] Table 1). Two to three time points were used to fit a linear regression in terminal elimination phase in animals of Groups 1 and 2, except for some cases (animals 1502, 2501 and 2503) due to lack of measureable data points or the last quantifiable concentration is Cmax (Error!
Reference source not found.-6 and Figures 7-11).
Table 1 Reliability of Iloprost AUC0-∞ in Dog Plasma after IV Infusion of Aqueous Formulation or Aqueous lipid-based Formulation at 5760 ng/kg AUC0-48 AUC0-∞ Ratio
Group Animal ID Formulation
(hr*pg/mL) (hr*pg/mL) ( AUC0-48/AUC0-∞
1 1501 Aqueous 18939 19733 0.960
1 1502 Aqueous 544 N/A N/A
1 1503 Aqueous 31.3 31.3 1.00
Figure imgf000031_0001
Summary
[0127] Following an initial of IV infusion of Iloprost for up to 24 hours in female dogs, Iloprost peak or near peak concentrations were achieved at 24.03 hours (median Tmax) for Group 1 (Aqueous formulation) and at 24.08 hours for Group 2 (Aqueous lipid-based formulation). Mean plasma exposure ( AUC0-48) was 4286 hr*pg/mL for Group 1 and 5359 hr*pg/mL for Group 2. Mean Cmax was 2602 pg/mL for Group 1 and 3969 pg/mL for Group 2. Estimated elimination half-life (ti/2) in plasma ranged from 0.0303 to 0.797 hour for Group 1 and from 0.0289 to 0.437 hour for Group 2. Total clearance (CL) ranged from 292 to 184111 mL/hr/kg for Group 1 and from 7708 to 292462 mL/hr/kg for Group 2, and apparent volume of distribution (Vd) ranged from 335 to 8038 mL/kg for Group 1 and from 322 to 89971 mL/kg for Group 2. The relative mean plasma exposure (AUC0-48) and mean Cmax of Group 2 to Group 1 were 125% and 153%, respectively. The results suggested improved pharmacokinetics characterized by approximate 25% higher exposure (AUC0-48) and 53% higher Cmax in Aqueous lipid-based formulation compared with Aqueous formulation.
Preparation of Blank Aqueous Lipid Mixutre
[0128] To prepare a blank aqueous lipid mixture a cubic phase was first prepared. To prepare the cubic phase, 47.24 g of D, L-a-tocopherol and 34.25 g of Phospholipon 90G were mixed to form a lipid mix. The lipid mix was heated on a hot plate set to 90°C to fully dissolve the Phospholipon 90G. 2.68 g of sodium deoxy cholate was dissolved in 27.42 g of sterile water for injection into the lipid mix. Once the Phospholipon 90G was fully dissolved in the lipid mix, the sodium deoxy cholate injection was added to the lipid mix, which was vigorously mixed to form a cubic gel. The cubic gel was then observed under polarized light under a microscope and images were recorded. If lamellar phase is observed, then continue homogenization or kneading using mortar-pestle until most of the lamellar phase disappears. An image of the final cubic gel was recorded before hydration start time and after hydratation completion if necessary. The adjustments should be recorded. The beaker was then covered with parafilm and hydrated overnight for about 18 hours.
[0129] After preparing the cubic phase, a blank aqueous lipid dispersion was prepared. In a 4L glass beaker, 1432.0 g of WFI water and 4.0 g of sodium deoxy cholate was added. The beaker was transferred under a Silverson homogenizer (L5M-A) using a square slot high shear screen, and the stirring speed was set to 5,000 rpm. The solution was mixed until the sodium deoxy cholate was dissolved, about 4-5 minutes.
[0130] Once dissolved, 85.4 g of the cubic phase was slowly added to the 4 L glass beaker while mixing at 5,000 rpm. This addition took about 14 minutes. After the addition of the cubic phase, the mixing speed was increased to 6,700 rpm and the solution was mixed for a minium of 5 hours. After 2 hours of mixing, 2.0 g of sodium deoxy cholate was added to the solution. The solution was mixed until no lamellar phase was observed under polarized light. Once the mixing was complete, 32.2 g of glycine was added to the beaker and the solution was mixed for a minmum of 30 minutes. After this mixing step, the net weight of the dispersion was measured and found to be 1,439 g.
[0131] The amount of WFI water to add to the beaker was calculated according to the following Formula:
Amount of water = Sum of the formulation component added to the beaker (amount of cubic gel + amount of DI water + amount of Sodium deoxycholate + amount of glycine) - amount of dispersion
Amount of water = (85.4 g +1432.0 g+ 6.0 g +32.2 g) - 1,439g = 116.6 g
[0132] Thus, 116.6 g of WFI water was added to the beaker and the dispersion was stirred at 6,700 rpm for 5 minutes. After 5 minutes, foam was observed so the solution was mixed for an additional 30 minutes. The pH was then measured to be 7.54. The dispersion was microfluidized (M-l 10EH-30) to form a blank nano-dispersion for 9 passes at 25000 psi.
[0133] The blank aqueous lipid mixture was filtered through a Meissner 0.2 p polypropylene vanguard filter. The pH, particle size, optical microscope, density and viscosity were measured. The results of which are presented below.
[0134] Table 9
Figure imgf000033_0001
Manufacturing Example [0135] An aqueous lipid mixture is prepared accoding to the following process, where the aqueous lipid mixture has a concentration of about 2 mg/mL. In a 400 mL beaker, 1.103 g of iloprost and 125 g of an aqueous lipid mixture was added. The beaker was secured under a Silverson homogenizer (L5M-A). After being secured, the small-scale square hole screen homogenizing head was lowered into the solution in the beaker and the solution was mixed at 5,000 rpm. This was performed until all the iloprost was dissolved, i.e. until the iloprost was no longer visible. If iloprost is not dissolving, a magnetic stir bar should be used instead of the homogenizer and the solution should be mixed for a longer interval, at least 2 days, but at a lower speed then was used with the homogenizer.
[0136] After the iloprost was dissolved, the solution was filter using a Meissner 0.2 pm polypropylene vanguard filter into a 100 mL beaker. A 2mL sample was then prepared for assay to test the concentration of the solution. The sample was found to have a concentration of 6.4 mg/mL. Thus, 60.31 g of the 6.4 mg/mL of the solution was diluted to 193.0 g by adding 132.69 g of the aqueous lipid mixture to prepare a 2 mg/mL solution. This was then mixed for about 30 minutes.
[0137] The pH of the solution was measured in triplicate, where the pH was 7.12, 7.11 and 7. 11. The pH, assay (concentration), related substance, particle size, density, viscosity and optical microscopy were measured and the results are presented in Tables 10 and 11 below.
Table 10. Results of Batch Analysis
Figure imgf000034_0001
Figure imgf000035_0002
Table 11. Results of Characterization Test
Figure imgf000035_0001
Effect on cyclic adenosinse monophosphate (cAMP) generation in human PASMCs
[0138] A study was run to examine the cAMP dose-response relationship in primary human pulmonary arterial smooth muscle cells (hPASMC) when increasing concentrations of Iloprost in an aqueous lipid formulation when compared to Iloprost Trometamol solution.
[0139] In this study, hPASMCs were isolated from small-resistance arteries obtained from nonimplanted human Caucasian donor lungs. These cells were plated in 96-well plates the day before the experiment, and the cells were serum-starved overnight. The hPASMCs were treated with 3-isobutyl-l -methylxanthine (IBMX lOOpM; an inhibitor of both extracellular and intracellular phosphodiesterases) prior to the exposure with the different Iloprost formulations and respective vehicle controls. The Iloprost in an aqueous lipid formulation was used in the concentration range of 1.9nM - 1950nM, while the Iloprost Trometamol solution was used in the concentration range of 2.3nM - 2310 nM. Forskolin (5pL), a permanent cAMP activator, was used as a positive control. The cells were treated, and after 15 minutes, the hPASMCs were rinsed twice with ice-cold PBS (200 μl), and then IX lysis buffer was added (100 μl/well). The samples were then stored at -80°C until the cAMP levels were measured using the Cyclic AMP XP® Assay Kit. [0140] In the presence of Iloprost in an aqueous lipid formulation, there was a significant elevation of cAMP over basal values in this acute setting. The treatment exerted a dose-dependent cAMP accumulation in primary human PASMC. The results of this test can be seen in the bar graph of FIGS. 14 and 15. A line graph of this test is also produced in FIG. 16 showing the intracellular cAMP levels in isolated human PASMC upon treatment, which showed an increase in cAMP. From this study, it showed that the Iloprost in an aqueous lipid formulation significantly increases the cAMP production in native human pulmonary arterial smooth muscle cells in the acute setting. Thus, it appears that the dose-response of the Ilorost in an aqueous lipid formulation is more potent than the Iloprost Trometamol formulation in higher concentrations.
Prophetic Example 1
[0141] An amount of sodium hydroxide is dissolved in glycerol using heating and stirring to aid in dissolution. An equimolar amount of methyl paraben is then dissolved with heating. From this solution, an aliquot is taken out and mixed with lecithin and oleyl alcohol in a test tube. Iloprost is incorporated and the solution is thoroughly mixed to form a nanostructured liquid crystalline phase material with the iloprost disposed therein. An “upper solution”, is obtained by dissolving Pluronic F-68 (a polvpropyleneoxide-polvethyleneoxide block copolymer surfactant commercially available from BASF), and acetic acid together and adding to the test tube as a layer of solution above the previous solution that includes the iloprost. Immediately the test tube containing the liquid crystalline mixture and the upper solution is shaken and sonicated for in a small, table-top ultrasonicator (Model FS6, manufactured by Fisher Scientific) to form a dispersion.

Claims

CLAIMS We claim:
1. A parenteral formulation comprising a therapeutically effective amount of iloprost or a pharmaceutically acceptable salt thereof and a lyotropic liquid crystal material.
2. The parenteral formulation of claim 1, wherein the lyotropic liquid crystal material comprises both polar and non-polar nano-domains.
3. The parenteral formulation of claim 1, wherein the material comprises nanostructured nonlamellar liquid crystalline phases
4. The parenteral formulation of claim 1, wherein the material comprises reversed nanostructured liquid crystalline phases.
5. The parenteral formulation of claim 1, wherein the material comprises distinct nanostructured nonlamellar liquid crystalline material and a liquid phase embedded within said distinct nanostructured nonlamellar liquid crystalline material, said liquid phase being selected from the group consisting of a hydrophobe-rich phase and a polar solvent-rich phase.
6. An implantable pump comprising a formulation of any of claims 1-5.
7. A method of treating pulmonary arterial hypertension comprising administering the formulation of any of claims 1-6.
8. A pharmaceutical formulation comprising: iloprost or a pharmaceutically acceptable salt thereof; a carrier; and a particle or material comprising a distinct nanostructured nonlamellar liquid crystalline material; and one or more pockets or droplets of a liquid phase embedded within said distinct nanostructured nonlamellar liquid crystalline material, said liquid phase being selected from the group consisting of an oil-rich liquid phase and a polar solvent-rich liquid phase.
9. The pharmaceutical formulation of claim 8 wherein said distinct nanostructured nonlamellar liquid crystalline material is a reversed phase nonlamellar liquid crystalline material.
10. The pharmaceutical formulation of claim 8 wherein said distinct nanostructured nonlamellar liquid crystalline material comprises a reversed hexagonal phase material, a reversed bicontinuous cubic phase material, a reversed discrete cubic phase material, or a reversed intermediate phase material.
11. The pharmaceutical formulation of claim 8 wherein said distinct nanostructured nonlamellar liquid crystalline material is polymerized.
12. The pharmaceutical formulation of claim 8 further comprising a stabilizing layer exterior to said particle or material.
13. The pharmaceutical formulation of claim 12, wherein said stabilizing layer is selected from the group consisting of a charged moiety, a polymer, and a surfactant.
14. The pharmaceutical formulation of claim 8, wherein said particle or material further comprises a coating comprising the iloprost or pharmaceutically acceptable salt thereof .
15. The pharmaceutical formulation of claim 8 wherein said liquid phase is an oil and said oil is selected from the group consisting of benzyl benzoate, estragole, eugenol, isoeugenol, linalool, and the essential oils of basil, bay, bois de rose (rosewood), carrot seed, clovebud, eucalyptus, ginger, grapefruit, hyssop, lemon, balsam of Peru, mugwort, myrrh gum, bitter orange, oregano, palmarosa, patchouly, peppermint, petitgrain, rosemary, santalwood oil, spearmint, thuja (cedar leaf), thyme, vanilla, and ylang ylang (cananga).
16. The pharmaceutical formulation of claim 8 wherein said liquid phase is a polar solvent and said polar solvent is selected from the group consisting of water, glycerol, and N,N- dimethylacetamide.
17. The pharmaceutical formulation of claim 8, wherein the iloprost or pharmaceutically acceptable salt thereof is dissolved or dispersed in said liquid phase or in said distinct nanostructured nonlamellar liquid crystalline material.
18. The pharmaceutical formulation of claim 8, wherein said particle or material is pharmaceutically acceptable for injection.
19. The pharmaceutical formulation of claim 18 wherein said particle or material is pharmaceutically acceptable for oral delivery.
20. The pharmaceutical formulation of claim 8, wherein said one or more pockets or droplets have a diameter of 50 nm or greater.
21. The pharmaceutical formulation of claim 8, wherein said liquid phase includes at least one of an oil and a polar solvent.
22. The pharmaceutical formulation of claim 8, wherein said liquid phase is a hydrophobe-rich phase or a polar solvent-rich phase.
23. The pharmaceutical formulation of claim 8 wherein said distinct nanostructured nonlamellar liquid crystalline material consists essentially of a reversed bicontinuous cubic phase material.
24. The pharmaceutical formulation of claim 8 wherein said distinct nanostructured nonlamellar liquid crystalline material consists essentially of a reversed discrete cubic phase material.
25. The pharmaceutical formulation of claim 8 wherein said distinct nanostructured nonlamellar liquid crystalline material comprises a water insoluble lipid or surfactant.
26. An implantable pump comprising a formulation of any of claims 8-25.
27. A method of treating pulmonary arterial hypertension comprising administering the formulation of any of claims 8-26.
28. A pharmaceutical formulation comprising iloprost or a pharmaceutically acceptable salt thereof and a coated particle comprising a. an interior core comprising a matrix comprising of i. at least one nanostructured liquid phase or a dehydrated variant thereof, ii. at least one nanostructured liquid crystalline phase or a dehydrated variant thereof or iii. a combination of
(1) at least one nanostructured liquid phase or a dehydrated variant thereof and
(2) at least one nanostructured liquid crystalline phase or a dehydrated variant thereof and b. an exterior coating comprising nonlamellar domains; wherein the iloprost or pharmaceutically acceptable salt thereof is in a.), b.) or a combination thereof.
29. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a nanostructured LI phase material, b. a nanostructured L2 phase material, c. a microemulsion that is nanostructured, or d. a nanostructured L3 phase material.
30. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a nanostructured normal or reversed cubic phase material, b. a nanostructured normal or reversed hexagonal phase material, c. a nanostructured normal or reversed intermediate phase material, or d. a nanostructured lamellar phase material.
31. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a polar solvent and b. a surfactant or a lipid.
32. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a polar solvent, b. a surfactant or a lipid and c. an amphiphile or hydrophobe.
33. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a block copolymer.
34. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a block copolymer and b. a solvent.
35. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a polar solvent and b. a surfactant.
36. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a polar solvent, b. a surfactant and c. an amphiphile or hydrophobe.
37. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a block copolymer.
38. The coated particle of claim 28, wherein said nanostructured liquid phase material comprises a. a block copolymer and b. a solvent.
39. The coated particle of claim 28, wherein said interior core comprises the iloprost or pharmaceutically acceptable salt thereof disposed within said matrix.
40. The coated particle of claim 28, wherein said interior core comprises a reversed cubic phase material.
41. The coated particle of claim 28 wherein said nonlamellar domain is amorphous.
42. The coated particle of claim 28 wherein said nonlamellar domain is a polymer.
43. The coated particle of claim 42 wherein said polymer is polylactic glycolic acid.
44. The coated particle of claim 41 wherein said nonlamellar domain comprises a sugar.
45. The coated particle of claim 44 wherein said sugar is trehalose.
46. The coated particle of claim 28 wherein said exterior coating comprises a semi-crystalline nonlamellar material.
47. The coated particle of claim 28 wherein said exterior coating comprises at least 2% nonlamellar domains.
48. The coated particle of claim 28 wherein said exterior coating comprises at least 10% nonlamellar domains.
49. The coated particle of claim 28 wherein said exterior coating comprises at least 50% nonlamellar domains.
50. The coated particle of claim 28 wherein said exterior coating comprises the iloprost or pharmaceutically acceptable salt thereof.
51. An implantable pump comprising a formulation of any of claims 28-50.
52. A method of treating pulmonary arterial hypertension comprising administering the formulation of any of claims 28-51.
53. A parenteral formulation comprising a therapeutically effective amount of iloprost or a pharmaceutically acceptable salt thereof and an aqueous lipid material, wherein the formulation provides intracellular cAMP levels in isolated hPASMCs from about 20 nm to about 30 nM at a concentration of about 1950 nM iloprost.
PCT/US2021/071589 2020-09-25 2021-09-24 Compositions and methods for vasodilator delivery WO2022067336A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2023519289A JP2023543037A (en) 2020-09-25 2021-09-24 Compositions and methods for vasodilator delivery
MX2023003506A MX2023003506A (en) 2020-09-25 2021-09-24 Compositions and methods for vasodilator delivery.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063083150P 2020-09-25 2020-09-25
US63/083,150 2020-09-25

Publications (1)

Publication Number Publication Date
WO2022067336A1 true WO2022067336A1 (en) 2022-03-31

Family

ID=80845891

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/071589 WO2022067336A1 (en) 2020-09-25 2021-09-24 Compositions and methods for vasodilator delivery

Country Status (3)

Country Link
JP (1) JP2023543037A (en)
MX (1) MX2023003506A (en)
WO (1) WO2022067336A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6638621B2 (en) * 2000-08-16 2003-10-28 Lyotropic Therapeutics, Inc. Coated particles, methods of making and using
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
US6991809B2 (en) * 2001-06-23 2006-01-31 Lyotropic Therapeutics, Inc. Particles with improved solubilization capacity
WO2014152795A2 (en) * 2013-03-14 2014-09-25 Schentag Jerome J Cholestosome vesicles for incorporation of molecules into chylomicrons
WO2018050864A1 (en) * 2016-09-15 2018-03-22 Camurus Ab Prostacyclin analogue formulations
US20200397782A1 (en) * 2019-06-11 2020-12-24 Chdi Foundation, Inc. Solid forms of a kynurenine-3-monooxygenase inhibitor

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6638621B2 (en) * 2000-08-16 2003-10-28 Lyotropic Therapeutics, Inc. Coated particles, methods of making and using
US6991809B2 (en) * 2001-06-23 2006-01-31 Lyotropic Therapeutics, Inc. Particles with improved solubilization capacity
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
WO2014152795A2 (en) * 2013-03-14 2014-09-25 Schentag Jerome J Cholestosome vesicles for incorporation of molecules into chylomicrons
WO2018050864A1 (en) * 2016-09-15 2018-03-22 Camurus Ab Prostacyclin analogue formulations
US20200397782A1 (en) * 2019-06-11 2020-12-24 Chdi Foundation, Inc. Solid forms of a kynurenine-3-monooxygenase inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SEGURA-IBARRA VICTOR, WU SUHONG, HASSAN NIDA, MORAN-GUERRERO JOSE A., FERRARI MAURO, GUHA ASHRITH, KARMOUTY-QUINTANA HARRY, BLANCO: "Nanotherapeutics for Treatment of Pulmonary Arterial Hypertension", FRONTIERS IN PHYSIOLOGY, vol. 9, 1 July 2018 (2018-07-01), pages 1 - 16, XP055929279, DOI: 10.3389/fphys.2018.00890 *

Also Published As

Publication number Publication date
JP2023543037A (en) 2023-10-12
MX2023003506A (en) 2023-06-28

Similar Documents

Publication Publication Date Title
ES2309766T3 (en) SLOW RELEASE LIQUID FORMULATIONS.
US20200375979A1 (en) Opioid formulations
AU2012348641B2 (en) Robust controlled-release formulations
CN101014319B (en) Liquid depot formulation
EP2938332B1 (en) Sustained-release lipid pre-concentrate of gnrh analogues and pharmaceutical composition comprising the same
CN107049932A (en) A kind of small-molecule drug phase change gel slow-released system in situ and preparation method thereof
US9211274B2 (en) TRPV1 compounds in a biodegradable polymer carrier
US20220071988A1 (en) Sustained release buprenorphine microspheres (srbm) and methods of use thereof
US20240075024A1 (en) Opioid formulations
TWI700087B (en) Pharmaceutical formulations for sustained release of sebacoyl dinalbuphine ester
CN109715138B (en) Prostacyclin analogue formulations
US20130217673A1 (en) Mixed monoamine reuptake inhibitor in a biodegradable polymer carrier
WO2022067336A1 (en) Compositions and methods for vasodilator delivery
WO2018164121A1 (en) Acidic emulsion composition containing local anesthetic agent
US11357756B2 (en) Anesthetic compositions and methods comprising imidazoline compounds
JP2022548616A (en) drug delivery formulation
US11400061B2 (en) Onychomycosis treatment compositions and methods
KR20230124561A (en) Compositions and methods for treating solid tumors
WO2021221774A1 (en) Compositions and methods for emergency rescue
KR20020088181A (en) Injectable Gel Type Lipid Composition And Preparation Method Thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21873674

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2023519289

Country of ref document: JP

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023005522

Country of ref document: BR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 112023005522

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20230324

122 Ep: pct application non-entry in european phase

Ref document number: 21873674

Country of ref document: EP

Kind code of ref document: A1