EP3600265A1 - Behandlungsverfahren und vorrichtung zur verbesserten bioverfügbarkeit von leukotrienrezeptorantagonisten - Google Patents

Behandlungsverfahren und vorrichtung zur verbesserten bioverfügbarkeit von leukotrienrezeptorantagonisten

Info

Publication number
EP3600265A1
EP3600265A1 EP18774366.1A EP18774366A EP3600265A1 EP 3600265 A1 EP3600265 A1 EP 3600265A1 EP 18774366 A EP18774366 A EP 18774366A EP 3600265 A1 EP3600265 A1 EP 3600265A1
Authority
EP
European Patent Office
Prior art keywords
dosage form
film
oral
montelukast
mtl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP18774366.1A
Other languages
English (en)
French (fr)
Inventor
Nadine Paiement
Horst G. Zerbe
Justin W. Conway
Rodolphe Obeid
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
IntelGenx Corp
Original Assignee
IntelGenx Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by IntelGenx Corp filed Critical IntelGenx Corp
Publication of EP3600265A1 publication Critical patent/EP3600265A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7007Drug-containing films, membranes or sheets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • 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
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/18Halogen atoms or nitro radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame

Definitions

  • This disclosure concerns a formulation and method of treatment and pharmaceutical dosage form for improving the bioavailability of a leukotriene receptor antagonist or leukotriene synthesis inhibitor for the treatment of a disorder.
  • Montelukast (MTL) sodium is an orally active leukotriene receptor antagonist commonly used to treat patients suffering from chronic asthma as well as symptomatic relief of seasonal allergic rhinitis.
  • MTL functions to suppress this inflammatory response by binding to the leukotriene receptor with high affinity and selectivity, thereby blocking the pathway leading to the physiological response for extended periods.
  • neuroinflammation within the brain has been linked to age-related dementia and neurodegenerative diseases. MTL applied under these biological conditions has been shown to significantly reduce neuroinflammation, elevate hippocampal neurogenesis and improve learning and memory in old animals.
  • U.S. Patent Nos. 8,575, 194 and 9,149,472 disclose methods of improving cognitive impairments by administering Montelukast in a single tablet or capsule that comprises an extended release (ER) component and an immediate release (IR) component in a single dosage unit.
  • the method involves administering the dosage unit to provide an initial burst of IR active pharmaceutical ingredient (API) into the system, followed by the ER API over the course of 12 hours, thereby maintaining a constant effective plasma level.
  • Disclosed embodiments include a tablet with an ER core and an IR shell or a capsule containing a mixture of ER and IR beads combined in a specific ratio to achieve the desired effect.
  • the regimen in general consists of an initial high dose of 10 mg of MTL followed by 5 mg doses approximately every 2 hours afterwards over the course of 12 hours.
  • the patents discuss plasma levels as being critical for achieving cognitive improvement.
  • MTL can only exert its therapeutic effects if it crosses the blood-brain barrier (BBB) and accumulates in the cerebrospinal fluid (CSF) at sufficient concentration levels. Neither plasma nor CSF concentration levels of MTL are discussed in the patents.
  • BBB blood-brain barrier
  • CSF cerebrospinal fluid
  • Surge Dose® Montelukast tablets have been proposed in a method for improving the formulation of a tablet capable of accelerated API release. The method attempts to improve MTL solubility in the stomach.
  • the Surge Dose® product may still be limited by gastric emptying cycles and food effects similar to the Singulair® tablet and chewable.
  • the chewable tablet is also comprised of solid MTL.
  • an alkaline oral film dosage form for improving bioavailability of leukotriene antagonist inhibitor. Accordingly, the oral film dosage form deliver leukotriene antagonist inhibitor such as Montelukast in a form that renders it suitable for improved bioavailability when compared with commercially available oral dosage forms.
  • the disclosed oral film dosage form has an alkaline surface pH that is preferably greater than to 7.5, more preferably greater than to 8 and optimally greater than to 8.5.
  • a dosage form of a leukotriene receptor antagonist exhibiting an improved bioavailability as compared with existing oral dosage forms.
  • a pharmaceutical dosage form for human pharmaceutical use comprising Montelukast salt, free base, or prodrug in a unit dosage form suitable for oral administration.
  • the dosage form can be configured for enteral delivery of the active agent.
  • the Montelukast salt, free base, or prodrug according to the disclosed dosage form can be configured to reach the stomach in an amorphous form in aqueous suspension.
  • Montelukast solubilized in an oral dosage form is orally administered such as on the tongue, buccaly or sublingually. Upon contact of the dosage form with saliva, the dosage form preferably solubilizes and/or disintegrates.
  • the dissolution and/or disintegration of the oral dosage form transforms the solubilized Montelukast into a suspended and/or insoluble precipitate creating a pre-solubilized dosage form ready to be absorbed and/or swallowed in the oral cavity.
  • the pre-solubilized dosage form improves the bioavailability of the Montelukast compared with the equivalent tablet or chewable oral dosage forms.
  • the Montelukast may be delivered through the use of a film layer having an alkaline surface pH.
  • Montelukast salt, free base, or prodrug is disposed within or on a polymeric film suitable for oral administration.
  • the films can be formulated for rapid disintegration and distribution of micro- or nano-scopic particles of the active agent in the gastrointestinal tract.
  • the active agent in the film dosage form is Montelukast sodium.
  • a alkaline surface pH Montelukast oral film dosage form having an improved bioavailability when compared to swallowable and chewable oral tablet dosage forms.
  • Specific conditions that can be treated by the present disclosure include, but are not limited to, neuroinflammation, neurodegenerative diseases and cognitive impairment.
  • the present disclosure is directed to a pharmaceutical unit dosage composition comprising about 0.5 to about 25 mg of Montelukast.
  • the unit dosage form is suitable for oral administration to treat neuroinflammation.
  • the unit dosage form contains about 10 mg of the compound and is administered once or twice per day.
  • Also disclosed is a method of treating a neurodegenerative disease or neuroinflammatory disorder.
  • the method comprising the steps of enterally delivering to a person or other animal in need of treatment for a neurodegenerative disease or neuroinflammatory disorder via a film dosage form, a safe and effective amount of a leukotriene receptor antagonist, wherein the amount of Montelukast is about 0.5 mg to about 25mg per day, preferably about 1 mg to about 10 mg and wherein leukotriene receptor antagonist is enterally delivered as a precipitate suspended in an aqueous medium, wherein the precipitate is generated orally upon dissolution and/or disintegration of an oral film dosage form
  • an oral film dosage form comprising: a film layer having an alkaline surface pH; and a safe and effective amount of a leukotriene receptor antagonist incorporated into the film layer.
  • the film layer is formulated to dissolve and/or disintegrate when in contact with an aqueous solution.
  • the leukotriene receptor antagonist is preferably incorporated into the film layer in an amorphous form and most preferably solubilized in the film layer.
  • a preferred film dosage form comprises Montelukast, present in an amount of about 0.5 mg to about 25 mg, preferably about 5 mg to about 15 mg and most preferably about 10 mg.
  • an oral film dosage form having a film layer with an alkaline surface pH; and a safe and effective amount of a leukotriene receptor antagonist incorporated into the film layer wherein the film layer dissolves and/or disintegrates in contact with an aqueous solution.
  • the alkaline surface pH is preferably greater than pH 7.5, more preferably greater than pH 8.0 and optimally greater than pH 8.5.
  • an oral dosage form having an unbuffered alkaline surface pH.
  • an oral film dosage form having a film layer with an alkaline surface pH; and a safe and effective amount of a leukotriene receptor antagonist incorporated into the film layer wherein the film layer dissolves and/or disintegrates in contact with an aqueous solution, and wherein the film layer comprises a plurality of stabilizers.
  • the plurality of stabilizers may be selected from parabens, EDTA, BHT and combinations of parabens, EDTA and BHT.
  • a film dosage form comprising Montelukast, wherein the area under the curve (AUC) is between about 3120 and about 4700 ng*h/mL and/or wherein the Cmax is between about 475 and about 720 ng/mL.
  • AUC area under the curve
  • the film layer(s) is configured for enteral delivery of the active agent.
  • the film layer may also be configured for transmucosal or sublingual delivery.
  • Figure 1 is representation of the dissolution of swallowable tablets.
  • Figure 2 is an illustrative representation of the absorption or an oral film dosage form when administered to a subject.
  • Figure 3 is an illustrative representation of the behavior of the active in the stomach following administration of the oral film to a subject.
  • Figure 4 is illustrative representation of the transmucosal absorption following administration of the oral film to a subject.
  • Figure 5 is a graphical representation of the dissolution data shown in table 12.
  • Figure 6 is a graphical representation of the solubility limits of MTL in solutions containing EDTA.
  • a method and device for the improved bioavailability of leukotriene inhibitors.
  • This method and device involve an oral dosage form designed to deliver leukotriene inhibitor such as Montelukast, to the mouth and stomach in an amorphous the form of an amorphous precipitate suspended in an aqueous medium (e.g., saliva and/or gastric fluids).
  • leukotriene inhibitor such as Montelukast
  • an amorphous medium e.g., saliva and/or gastric fluids
  • methods for treating neurodegenerative diseases and/or other conditions that are at least partially induced by leukotrienes include enteral delivery or a combination of transmucosal, sublingual or both transmucosal and sublingual, along with enteral delivery of Montelukast.
  • the Montelukast is incorporated into a film layer in an amount that is safe and effective to reduce leukotriene induced neuroinflammation in patients.
  • Neurodegenerative diseases that can be treated in accordance with this disclosure include, but are not limited to, loss of memory function (long term or short term), dementia, apathy, depression, fatigue (acute or chronic), cognitive losses, loss of focus, loss of libido, and disorientation.
  • Specific disease conditions that can be treated with the disclosed methods include Huntington's disease, Parkinson's disease and Alzheimer's disease.
  • Such treatments can also be effective for treating neurological diseases, neurodegenerative diseases, neuroinflammatory disorders, traumatic or posttraumatic disorders, vascular or more precisely, neurovascular disorders, hypoxic disorders, and postinfectious central nervous system disorders.
  • neurodegenerative disease or “neurological disease” or “neuroinflammatory disorder” refers to any disease, disorder, or condition affecting the central or peripheral nervous system, including ADHD, AIDS -neurological complications, absence of the Septum Pellucidum, acquired epileptiform aphasia, acute disseminated encephalomyelitis, adrenoleukodystrophy, agenesis of the Corpus Callosum, agnosia, Aicardi Syndrome, Alexander Disease, Alpers' Disease, alternating hemiplegia, Alzheimer's Disease, amyotrophic lateral sclerosis (ALS), anencephaly, aneurysm, Angelman Syndrome, angiomatosis, anoxia, aphasia, apraxia, arachnoid cysts, arachnoiditis, Arnold-Chiari Malformation, arteriovenous malformation, aspartame, Asperger Syndrome, ataxia telangiectasia, ataxia
  • the disclosed dosage forms and methods are expected to be especially useful for treating neurodegenerative diseases and neuroinflammatory disorders selected from the group comprising or consisting of: Alzheimer's disease, Parkinson's disease, Creutzfeldt Jakob disease (CJD), new variant of Creutzfeldt Jakobs disease (nvCJD), Hallervorden Spatz disease, Huntington's disease, multisystem atrophy, dementia, frontotemporal dementia, motor neuron disorders of multiple spontaneous or genetic background, amyotrophic lateral sclerosis (ALS), spinal muscular atrophy, spinocerebellar atrophies (SCAs), schizophrenia, affective disorders, major depression, meningoencephalitis, bacterial meningoencephalitis, viral meningoencephalitis, CNS autoimmune disorders, multiple sclerosis (MS), acute ischemic/hypoxic lesions, stroke, CNS and spinal cord trauma, head and spinal trauma, brain traumatic injuries, arteriosclerosis, atherosclerosis, microangiopathic dementia, Binswanger' disease (Leukoara), Alzheimer'
  • the disclosed dosage forms and methods are expected to be especially useful for treating neurodegenerative diseases and neuroinflammatory disorders selected from the group comprising or consisting of: Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), hydrocephalus, CNS and spinal cord trauma such as spinal cord injury, head and spinal trauma, brain traumatic injuries, cochlear deafness, AIDS-related dementia, trinucleotide repeat disorders as Huntington ' s Disease, and CNS-aging.
  • neurodegenerative diseases and neuroinflammatory disorders selected from the group comprising or consisting of: Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), hydrocephalus, CNS and spinal cord trauma such as spinal cord injury, head and spinal trauma, brain traumatic injuries, cochlear deafness, AIDS-related dementia, trinucleotide repeat disorders as Huntington ' s Disease, and CNS-aging.
  • treatment refers to curing, mitigating or relieving symptoms of a disease, medical condition or injury.
  • a film layer that is "unbuffered” is a film layer that does not contain a weak acid or weak base that is effective to maintain pH near a chosen value upon addition of another acid or base. Stated differently, the unbuffered film layer does not contain a buffering agent, such as borates, citrates, or phosphates.
  • Enteral delivery refers to passing the active agent through the gastrointestinal tract, either naturally via the mouth and esophagus, or through an artificial opening (e.g., stoma) and absorbing the active agent in the intestine.
  • an artificial opening e.g., stoma
  • Leukotriene inhibitions include leukotriene receptor antagonists and/or leukotriene synthesis inhibitors that block 5 -lipoxygenase activity.
  • leukotriene inhibitors include, but are not necessarily limited to, leukotriene receptor antagonist such as Montelukast, Zafirlukast, Pranlukast, cinalukast, probilukast, iralukast and sulukast.
  • Active agents capable of existing in various forms, such as base form, salts, esters, prodrugs, etc. are, unless otherwise indicated, encompassed by reference to the base drug.
  • the term "Montelukast" is intended to encompass all forms, including salts (e.g., Montelukast sodium), esters and prodrugs.
  • amorphous refers to a non-crystalline form of the solid i.e. a state that lacks the regular crystalline organization of atoms. Amorphous solids are generally more soluble, faster dissolving, easier to absorb in the GI tract or oral cavity and less stable than their crystalline counterparts.
  • the amorphous content (amorphicity) of a solid can be accurately and precisely assessed using a number of well-established methodologies, including isothermal calorimetry, Powder X-ray diffraction (PXRD), Raman Spectroscopy, Differential Scanning Calorimetry (DSC), Continuous Relative Humidity Perfusion Microcalorimetry (cRHp), and Dynamic Vapor Sorption (DVS).
  • PXRD Powder X-ray diffraction
  • DSC Differential Scanning Calorimetry
  • CRHp Continuous Relative Humidity Perfusion Microcalorimetry
  • DVDS Dynamic Vapor Sorption
  • the term amorphous also refer
  • active agent(s) or API (active pharmaceutical ingredient) refers mainly to pharmaceutically active ingredients, but may also refer to generally any agent(s) that chemically interacts with the subject to which it is administered to cause a biological change, such as, but not limited to eliminating symptoms of disease or regulating biological functions.
  • stable refers to a product which exhibit no or very limited changes in the dissolution profile and recovery (or assay) when the product is exposed to normal stability conditions (example 25°C/60%RH and 40°C/75%RH) for extended period of time.
  • oral film dosage form generally refers to an edible composition that can be ingested by a subject (human or animal) to orally, buccally or sublingually administer a predetermined amount of an active agent(s) to the subject, wherein the composition is in the form of a film.
  • the "surface pH” is the pH measured on a surface of the film, such as the top or bottom surface of a monolayer film or on an exposed surface of the layer containing the active in a multilayer oral film.
  • the film is prepared for pH testing by slightly wetting the film (adding water as needed for a pH test - e.g. one to three drops).
  • the pH is then measured by bringing the electrode in contact with the surface of the oral film. This measurement of the surface pH is preferably performed on several films of the same formulation.
  • film and “film layer” refer to a component or layer of dosage form that is distinctly different from pills, tablets, caplets, and capsules, and in which the dosage form is a thin strip of material.
  • Such films are typically rapidly disintegrating or rapidly dissolving, but can also exhibit longer disintegration and/or dissolution time when required.
  • the films are generally sufficiently flexible to allow bending or even folding without breaking.
  • a film layer is a sheet-like material having a thickness that is much less than its length or width.
  • oral transmucosal devices typically have a thickness on the order of about 50 ⁇ to 500 ⁇ (i.e., 0.05 mm to 0.5 mm), although thicker or thin films may be suitable; and width and length dimensions typically on the order of about 5 mm to 40 mm, although larger or smaller dimensions can be used.
  • the film dosage form can comprise a single film layer, or multiple layers.
  • a biocompatible layer e.g., a bioadhesive layer
  • a non- adhesive barrier layer to prevent or reduce ingestion of the active agent(s) and ensure that all or most of the active agent crosses the mucous membrane to which the bioadhesive layer is applied.
  • bioadhesive means that the composition of the film layer is formulated to adhere to the selected mucous membrane through which delivery of the active agent is targeted, and encompasses the term “mucoadhesive.”
  • bioadhesive polymers used in formulating the film should be selected to exhibit adequate adhesion within the environment at the targeted mucous membrane to ensure that the bioadhesive layer remains in contact with the mucous membrane to which it is applied and allows the active agent to directly enter the blood stream through the mucous membrane.
  • the active agent can be combined or blended with film forming polymers and/or bioadhesive polymers to obtain a balanced combination of properties like flexibility, tensile strength, uniformity of the film and the drug, hydration speed, drug release, disintegration time, palatability (taste, smell, texture and aftertaste), mouth feel, mucoadhesion, and chemical and physical stability suitable for an oral delivery device.
  • suitable film forming polymers exhibiting bioadhesion include hydroxypropyl cellulose, hydroxymethylcellulose, natural or synthetic gum, polyvinyl alcohol, polyethylene oxide, homo- and copolymers of acrylic acid crosslinked with a polyalkenyl polyether or divinyl alcohol, polyvinylpyrrolidone, hydroxypropyl methyl cellulose, sodium alginate, pectin, gelatin maltodextrins chitosan, and poly-lysines.
  • the active agent can be combined with film forming neutral polysaccharides such as pullulan.
  • Penetration enhancing agents can also or alternatively be employed to further increase the rate and/or total amount of absorption of the active agent.
  • penetration enhancers examples include 2,3-lauryl ether, phosphatidylcholine, aprotinin, polyoxy ethylene, azone, polysorbate 80, benzalkonium chloride, polyoxyethylene, cetylpyridinium chloride, phosphatidylcholine, cetyltrimethyl ammonium bromide, sodium EDTA, cyclodextrin, chitosan, sodium glycocholate, dextran sulfate 16 sodium glycodeoxycholate.
  • penetration enhancers include surfactants, bile salts (by extracting membrane protein or lipids, by membrane fluidization, by producing reverse micellization in the membrane and creating aqueous channels), fatty acids (that act by disrupting intercellular lipid packing), azone (by creating a region of fluidity in intercellular lipids), pore forming agents (e.g., molecules, peptides, nucleic acids or particles that insert into the lipid membrane and create a hole through which the API can pass) and alcohols (by reorganizing the lipid domains and by changing protein conformation), sulphoxides (dimethylsulphoxide, decylmethyl sulfoxide), , pyrrolidones (2pyrrolidone, 2P), alcohols/alkanols (ethanol or decanol), glycols (propylene glycol), terpenes (1,8-cineole, menthol, and menthone, D- limonene), fatty acids (oleic acid
  • Anti-oxidants and chelating agents that can be advantageously employed comprise di sodium -EDT A, sodium calcium EDTA, citric acid, L-cystein, vitamin E, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, potassium metabisulfite, propyl gallate, sodium metabi sulfite, sodium thiosulfate, 3,4-dihydroxybenzoic acid.
  • surfactants examples include polysorbates (TweenTM, SpanTM), sodium dodecyl sulfate (sodium lauryl sulfate), lauryl dimethyl amine oxide, cetyltrimethylammonium bromide (CTAB), polyethoxylated alcohols, polyoxyethylene sorbitan octoxynol (Triton XI 00TM), N,N-dimethyldodecylamine-N-oxide, hexadecyltrimethylammonium bromide (HTAB), polyoxyl 10 lauryl ether, Brij 721TM, bile salts (sodium deoxycholate, sodium cholate) polyoxyl castor oil (CremophorTM), nonylphenol ethoxylate (TergitolTM), cyclodextrins, lecithin, methylbenzethonium
  • the solubility and disintegration profiles of the film can influence the bioavailability of the drug. Therefore, certain embodiments of the film platform will contain specific quantities of disintegrants to control the residence time of the film in the oral cavity. Certain forms of the drug product may contain between 0-10% by mass of a disintegrant. Examples of disintegrants that could be used are Maltodextrin, Citric acid, Sodium starch, glycolate, crosslinked polyvinylpyrrolidone (crospovidone), crosslinked sodium carboxymethyl cellulose, Calcium silicate, Alginic acid, and vinylpyrrolidone-vinyl acetate copolymers.
  • disintegrants Maltodextrin, Citric acid, Sodium starch, glycolate, crosslinked polyvinylpyrrolidone (crospovidone), crosslinked sodium carboxymethyl cellulose, Calcium silicate, Alginic acid, and vinylpyrrolidone-vinyl acetate copolymers.
  • pre-solubilized refers to a dosage form comprising an active agent that undergoes a phase transformation in the oral cavity upon administration.
  • a pre-solubilized form of MTL could be a precipitated MTL previously administered as a solubilized MTL in a film matrix.
  • the pre-solubilized precipitate is not dissolved, but is in a form (e.g., very small particles dispersed in a liquid) that is susceptible to rapid dissolution, such as upon exposure to the higher pH environment of the intestine.
  • matrix refers to the surroundings or medium constituting the film layer in which the active agent (e.g., Montelukast) is solubilized or distributed, and generally comprises a mixture of polymers and excipients.
  • the film forming matrix supporting the API within the oral film dosage form can comprise about 40.0-99.0 % dry w/w of the film layer.
  • Stability enhancing agents can be added to the film to prevent photodegradation, oxidation, and/or microbial contamination.
  • Photodegradation inhibitors include ultraviolet radiation absorbers and pigments.
  • Ultraviolet absorbers include hydroxyl benzophenones and hydroxyphenyl benzotriazoles.
  • Pigments that can be added to the film include various metal oxides, such as titanium dioxide (Ti0 2 ), ferric oxide (Fe 2 0 3 ), iron oxide (Fe 3 0 4 ), and zinc oxide (ZnO).
  • Ti0 2 titanium dioxide
  • Fe 2 0 3 ferric oxide
  • Fe 3 0 4 iron oxide
  • ZnO zinc oxide
  • the pouches are made out of laminated material, comprising some aluminum or reflective foil material preventing photodegradation of the film and products contained therein.
  • Microbial contamination may be controlled by the use of antimicrobial agent such as methyl, ethyl or propyl paraben, sodium benzoate, benzoic acid, sorbic acid, potassium sorbate, propionic acid or a combination of the above.
  • additives such as excipients or adjuvants
  • excipients or adjuvants include flavors, sweeteners, coloring agents (e.g., dyes), plasticizers, and other conventional additives that do not deleteriously affect transmucosal delivery of the active agent, oral mucoadhesivity, or their important film properties.
  • the film can be used in a monolayer, bilayer or other multilayer form.
  • the bilayer film dosage form comprises a first layer having the API and a second layer having agents such as a taste-masking agent, backing agent for protecting the first layer, and/or a permeation enhancer.
  • the second layer can also be used to favor the directed absorption through the oral mucosa (unidirectional absorption).
  • Other embodiments could have the same API or a different API present in the second layer to enterally deliver the active with a controlled release profile.
  • an active agent in the second layer could be used to modify the absorption of the active agent in the first layer.
  • a safe and effective amount generally refers to an amount that provides a beneficial or therapeutic effect, i.e., provides a curing or mitigating effect on disease or disease symptoms, but which is sufficiently low to avoid severe or life-threatening side effects when the active agent is administered and delivered transmucosally and/or enterally.
  • Montelukast solubility in aqueous media is dependent on the pH. It has been found that MTL exhibits increasing solubility at alkaline (basic) pH above 7.5 and is found to rapidly precipitate in media below pH 7.5. This has been experimentally shown by Okumu et al (Okumu, Pharm. Res, 25, 12, 2008), see Fig. 6, where MTL alone or in the presence of surfactants only displays a marked increase in solubility above pH 7.5. This study has also shown that although the impact of surfactants may marginally increase MTL solubility, it is only at alkaline (basic) pH environments that MTL readily solubilizes.
  • Fig. 1 is showing the schematic representation of the dissolution of an oral dosage form of MTL, such as a conventional tablet.
  • Fig. 1A depicts the initial disintegration of the tablet in the stomach.
  • IB depicts the tablet disintegration after 10-15 minutes, where due to slower disintegration, the tablet pieces remain concentrated in a localized cluster limiting the dissolution and potential absorption. This limiting impediment is further exacerbated due to the poor solubility of MTL in acidic environments such as the stomach. Since MTL has an especially low solubility at low pH, the high concentration of MTL following disintegration of the tablet further increases the insolubility of MTL thereby potentially further reducing the bioavailability of the API.
  • the active agent can be distributed in the film matrix in the form of micro- or nano- particles.
  • a film oral dosage form wherein a leukotriene receptor antagonist (e.g., MTL) is administered via enteric absorption (Fig. 2B) alone or in combination with oral transmucosal and/or sublingual absorption (Fig. 2A)
  • the film oral dosage form is designed to disintegrate in the mouth and allow a solubilized active agent to precipitate in the mouth and be swallowed, thereby delivering the API into the stomach as a fine precipitate suspended in aqueous medium.
  • the film layer containing the active agent dissolves and/or disintegrates in the oral cavity upon contact with saliva. While the film dissolves and/or disintegrates, the Montelukast (or other leukotriene receptor antagonist) precipitates in the saliva (Montelukast API precipitates below pH 8) thus forming an API precipitate suspension in the saliva. The suspended API is then swallowed and reaches the stomach as a dispersed precipitate, improving the bioavailability of the Montelukast API.
  • the pre-solubilized film at least mitigates the dissolution problem associated with the poor solubility of Montelukast in the patient's acidic stomach conditions.
  • the poor solubility is generally amplified by the presence of a concentrated form of MTL. Though buccal and/or sublingual absorption may arise, the drug is predominantly absorbed enterally.
  • the oral film dosage can be used to overcome the solubility problem encountered when having Montelukast sodium present in the stomach in a solid or undissolved form.
  • Montelukast film particulates reach the stomach already in a suspended/precipitated form, meaning that the Montelukast solubilized in the dosage form and precipitates in the oral cavity and/or esophagus, resulting in a suspended Montelukast precipitate being delivered to the stomach.
  • the pre-solubilized Montelukast in the dosage form has an improved bioavailability derived at least in part from the fact that the API is delivered to the stomach in a dispersed and thus less concentrated form than conventional tablets.
  • the suspended precipitate thus exhibits an improvement in bioavailability when compared with tablets which must initially be dissolved in the stomach before being absorbed.
  • the improved bioavailability can lead to increased transport of the active agent across the blood-brain barrier, allowing lower doses and/or more effective treatment.
  • the administration of a Montelukast API suspension to the stomach at least mitigates solubility related problems arising in or with other Montelukast oral dosage forms such as swallowable and chewable tablets.
  • the administration of the suspended form through a film dosage form at least mitigates stability problems typically associated with API administered through liquid medium.
  • the orally precipitated Montelukast is likely able to reach the small intestine quicker through the pylorus than other oral dosage forms of Montelukast or other Leukotriene receptor antagonist.
  • a dosage of up to a maximum of 20 mg a day of Montelukast is sufficient to alleviate symptoms or treat conditions associated with neuroinflammation.
  • An essential element of such oral film dosage form is its ability to maintain Montelukast in a condition promoting its solubility, i.e. alkaline pH.
  • the Montelukast oral film has an alkaline surface.
  • the alkaline surface pH signifies that the film maintains Montelukast under alkaline conditions favoring its solubility and preventing recrystallization of the Montelukast. Recrystallization of the Montelukast is associated with unstable oral films.
  • the Montelukast oral film preferably has a surface pH greater than to pH 7.5, preferably greater than 8.0 and more preferably greater than 8.5.
  • Another embodiment of the oral dosage form comprises a capsule dosage form (e.g., a gelatin or cellulose -base capsule) containing the leukotriene inhibitor solubilized or distributed as an amorphous precipitate in a polymer matrix that disintegrates or dissolves in an aqueous medium.
  • a capsule dosage form e.g., a gelatin or cellulose -base capsule
  • the oral dosage form of Montelukast is taken orally by the patient.
  • the capsule shell is solubilized thus delivering the solubilized or amorphous precipitate of Montelukast (or other leukotriene receptor antagonist) into the aqueous medium of the stomach.
  • Such precipitate will be rapidly distributed throughout the stomach and mitigates the shortcoming related to tablets and chewables.
  • the oral capsule dosage form effectively mitigates low dosage bioavailability problems. Therefore, the Montelukast capsule allow the Montelukast to reach the stomach as a pre-solubilized, amorphous precipitate in suspension. It is possible that the stomach conditions, unfavorable to the dissolution of Montelukast tablets and chewables, result in some precipitation of the Montelukast in the stomach.
  • the extent of precipitation should be less than the loss of efficacy associated with the need to solubilize the Montelukast tablet in the stomach.
  • Leukotriene blockers or inhibitors can function to improve cognitive impairment by reducing the neuroinflammatory response within the brain.
  • Leukotriene blockers, such as MTL must therefore cross the blood-brain barrier and accumulate in the CSF. Consequently, during clinical trials, patients were tested for CSF levels of MTL after 3 and 7 hours respectively, (see Table 1). What is most surprising about this finding is that between the 3- and 7-hour test points, the concentration of MTL continued to increase. This is particularly unexpected as the plasma levels show a Tmax value between 2-4 hours indicating that the maximum accumulated concentration is rapidly reached in the blood.
  • a disclosed method of treating neurodegenerative or neuroinflammatory disorder comprises the step of enterally delivering to a person or other animal in need of treatment for a neurodegenerative disease or neuroinflammatory disorder via a film dosage form, a safe and effective amount of solubilized Montelukast.
  • the MTL reaches the stomach in a compressed solid state and thus must solubilize in the stomach under unfavorable conditions.
  • the disclosed MTL03 oral film dosage form comprises solubilized MTL, which is placed in the mouth and allowed to dissolve before being swallowed.
  • the MTL precipitated in the oral cavity while the remainder of the dosage form disintegrated and/or dissolved, creating a MTL precipitate ultimately suspended in aqueous medium (i.e. saliva).
  • aqueous medium i.e. saliva
  • the MTL is then transferred to the small intestine via the pylori. Since the MTL is already present as a suspended precipitate, the MTL may more easily reach the small intestine through the leaking pylori. It is well known that the pylori is not leak proof and allows some liquid to flow through even in its closed position. As such, MTL of the disclosed MTL03 film dosage once in the stomach is believed to more easily traverse the pylori. Administering MTL enterally as a suspended precipitate in aqueous medium improves bioavailability.
  • the disclosed MTL03 film dosage which contains MTL solubilized in the film matrix and precipitates in the saliva once the matrix dissolves has proven to be 1.5 times more bioavailable than the tablet when comparing the area under the curve (AUC) (see Tables 3 & 4). It is believed that administering the MTL as a precipitate suspension that is free from the film or tablet matrix improves the bioavailability of the MTL when compared with the corresponding tablet and chewable. This improved bioavailability is believed to be at least in part caused by the increased contact area of the precipitate API. In addition, the MTL is delivered in the stomach in a less concentrated manner than corresponding tablet and chewable oral dosage forms (see Figs. 1 and 3).
  • the method for treating a neurodegenerative disease or neuroinflammatory disorder comprises the step of (a) enterally delivering to a person or other animal in need of treatment for a neurodegenerative disease or neuroinflammatory disorder via a film dosage form, a safe and effective amount of Montelukast.
  • the Montelukast is orally administered via an oral film dosage comprising MTL or any other suitable salt, ester or prodrug thereof.
  • the Montelukast is at least substantially solubilized in the film dosage form and administered orally with a film matrix that dissolves and/or disintegrates in contact with aqueous medium such as saliva when in the oral cavity.
  • the MTL precipitates upon dissolution of the film matrix in the saliva in the person's or animal's oral cavity. Furthermore, the pharmacokinetic data for the disclosed MTL03 MTL dosage form show that absorption is significantly higher than for the branded form Montelukast Singulair® product (tablet). Therefore administering MTL as a film dosage form having a matrix that rapidly dissolves or disintegrates (i.e.
  • a leukotriene receptor antagonist such as Montelukast, is solubilized in the oral film dosage form.
  • the leukotriene receptor antagonist is present in the film as a particulate active in an oral film dosage form.
  • this particulate API is held in the oral film matrix, in which the film matrix will dissolve and/or disintegrate when in contact with an aqueous medium (i.e. saliva).
  • an aqueous medium i.e. saliva
  • the particulate API will be present as a particulate suspension in aqueous medium.
  • the particulate API is preferably in amorphous form in the film matrix.
  • the oral film is preferably applied against the subjects' oral mucosa where it will be adhered to and enter in contact with the subject's saliva. Contact between the film and the saliva dissolves and/or disintegrates the film in the oral cavity.
  • the dissolved and/or disintegrated oral film matrix advantageously allows precipitation of the active agent in the oral cavity of a subject. The precipitate is swallowed for enteral administration as a suspended precipitate in aqueous medium.
  • a preferred amount of MTL per unit dosage form is from about 0.5 mg to about 25 mg, preferably about 1 mg to about 25 mg, more preferably about 5 mg to about 10 mg.
  • Preparation of a film product typically involves casting or otherwise thinly spreading the liquid film formulation on a substrate, drying (e.g., evaporating) all or most of the solvent(s) from the film to produce a thin, solid film sheet of material, and cutting the solid film sheet into individual unit dosage forms.
  • Fig. 5 shows an increased rate of dissolution of the present film oral dosage form of MTL when compared with the Singulair® MTL tablet.
  • the dissolution of the present film oral dosage form taking into account the buccal delivery method.
  • the "pre-dissolved film” refers to a film that is pretreated to simulates conditions typical of when the film is applied to oral mucosa of a human subject. Under such simulated conditions, the film slowly disintegrates before being subjected to the dissolution experiment. This method is used for a more representative comparison of the swallowed tablet behavior in the stomach with that of the swallowed film; the film is again much faster.
  • the dissolutions were conducted under the following conditions.
  • the dosage consists of a 10 mg unit of either film or tablet.
  • a USP dissolution apparatus was used to measure the API release profiles. Each dissolution container was filled with 900 mL of phosphate based simulated saliva buffer pH 6.8. The paddle speed was set to 50 rpm and the temperature was kept at 37°C. Each pull point consisted of 8 mL and the time points were taken at 1, 2.5, 5, 7.5, 10, 15, 20, 30, 45. Samples were analyzed using UV absorption at 273 nm. Pre-solubilized Montelukast-Film dissolution was prepared by mixing a single film unit in 2 mL of simulated saliva buffer. This volume is considered to be representative of the volume of saliva generally found in the oral cavity under normal conditions. Data is summarized in Table 12.
  • MTL03 and MTLlO-films platform releases MTL significantly more quickly than the MTL-tablet dosage. This is believed to contribute towards the observed improved bioavailability during our Phase I Clinical study.
  • the oral film of MTL (principally MTL03) exhibits improved bioavailability compared to presently marketed products available as tablets/granules or suspensions. It is believed that the increased bioavailability of the MTL is related to the state of the MTL within the oral film.
  • improved bioavailability of the oral film dosage form critically linked to the incorporation of solubilized MTL into the alkaline oral films, ensuring a rapid release of pre-solubilized therapeutic which is easily absorbed in the oral cavity and enterically.
  • the alkalinity of the oral film as measured by the surface pH of the film favors dissolution of the MTL within the film. It is believed that the MTL remain soluble to some extend within the film due in part by the presence of residual solvent.
  • Our preliminary results from manufacturing processes demonstrate the presence of between 5 to 9% dry w/w of residual solvent.
  • alkaline surface pH oral films of MTL (MTL01, MTL03, MTL05, MTL06 and MTL 07) are expected to exhibit the observed increased bioavailability of MTL03.
  • the alkaline film layer is designed to keep MTL in a favorable solubilized condition that readily forms amorphous precipitates in the saliva upon oral administration of the film.
  • Montelukast oral film formulations therefore pertains to the stability of the solubilized API during the manufacturing, processing and long term storage.
  • solubilizing API significantly improves the bioavailability of the drug, it also potentially accelerates the degradation/decomposition pathways of the API leading to unwanted impurities.
  • the present disclosure addresses why achieving a stable solubilized MTL product is unexpectedly challenging for those skilled in the art, and the process by which it can be accomplished using by using specific critical excipient to API ratios and mixing conditions.
  • Montelukast is known to degrade over time (M. M. Al Omari et al.) in a solid or liquid state when exposed to light, moisture or heat, yielding degradation products such as Montelukast sulfoxide (SO) and Montelukast cis-isomer ⁇ Journal of Pharmaceutical and Biomedical Analysis, 45, 2007, 465-471).
  • SO Montelukast sulfoxide
  • Montelukast cis-isomer Journal of Pharmaceutical and Biomedical Analysis, 45, 2007, 465-471.
  • Singulair® chewable tablets exposed to sunlight show an increased amount of the montelukast sulfoxide impurity of 2.4% after 3 weeks.
  • montelukast in 0.1 M hydrochloric acid solution exposed to a sodium vapor lamp for 6 hours leads to a 14.6% increase in the amount of montelukast m-isomer.
  • the choice of stabilizer or antioxidant ca be important.
  • the choice of antioxidant/stabilizer is limited to molecules which will not lead to, or interact with, the API in such a way as to cause precipitation. This challenge would not be encountered in tablet formulations, as MTL is kept and used in its solid state. Solubilized MTL is particularly sensitive to changes in the pH environment and precipitates at lower pH, such as below 8.
  • Solubilized MTL is also negatively charged which can lead to unwanted complexations. Therefore, the choice and amount of antioxidant is further limited and excludes highly acidic molecules or molecules which may associate with the API covalently or non-covalently to form insoluble precipitate complexes and/or aggregated material.
  • MTL is particularly susceptible when in its solubilized state to metal catalyzed degradation as well as other oxidative or photo-induced decomposition pathways.
  • Existing MTL dosage forms are predominantly found as tablets, tablet variants or suspensions in which MTL is a solid or a suspension.
  • antioxidants/stabilizers can be directly added as solid material or applied to the product indirectly (spray coatings, shells or film coating). There is no need to consider antioxidant/stabilizer interactions which would precipitate MTL in a tablet dosage form, as it is already a solid.
  • chelating agent examples include molecules such as disodium edetate (EDTA), tetra sodium edetate, calcium disodium edetate, pentetic acid (DTP A), citric acid (CA), DL-2,3-Dimercapto-l-propanesulfonic (DMPS), dimercaptosuccinic acid (DMSA), monoisoamyl DMSA (MiADMSA) alpha lipoic acid (ALA), glutathione, N-acetyl-cystein (NAC), vitamin C, (2)-2-amino-3 -methyl -3- sulfanylbutanoic acid, dithioglycerine, N-(alpha-L-arabinofuranos-
  • EDTA disodium edetate
  • DTP A pentetic acid
  • CA citric acid
  • DMPS dimercaptosuccinic acid
  • DMSA dimercaptosuccinic acid
  • MiADMSA monoisoamyl DM
  • chelators such as EDTA are offered as different salts which exhibit more alkaline pH effects on the aqueous media, however these molecules, such as tetra sodium edetate or disodium calcium edetate do not perform as well in maintaining MTL stability in long term studies.
  • chelators such as EDTA are highly effective at sequestering the metal ions responsible for catalyzing the sulphoxide impurity formation.
  • concentration of EDTA the greater the stability of the MTL API.
  • addition of chelators in an aqueous medium in general leads to deprotonation of the chelators and consequent acidification of the aqueous blend. This is problematic as MTL solubility is particularly sensitive to changes in the pH of the environment and rapidly precipitates at pH below 8. In fact as seen in Figure 1 below only a limited amount of EDTA can be added to a solution of MTL before precipitation is observed.
  • MTL solubility and stability of MTL are critical parameters to consider when formulating oral films that will generate a reproducible target bioavailability and stable product. Therefore, optimal formulations of MTL will need to balance the amount of API with EDTA in order to achieve the needed stability while maintaining a solubilized drug component. This can be achieved using several strategies: (1) balancing the ratio of EDTA to MTL (MTL itself is a basifying agent), (2) using base modifying excipients to compensate for increasing amounts of EDTA, and (3) application of alkaline buffering components.
  • a second surprising challenge for using EDTA to stabilize MTL is that regardless of the concentration of EDTA, overtime nearly 100% of the EDTA is observed to precipitate. Higher concentrations of ETDA lead to accelerated precipitation of MTL within minutes, while lower concentrations result in precipitation only after 10 days. This is of particular importance as it means that the holding time of the blend should never be longer than the observed time to precipitation.
  • These are binary mixtures in water, the blend behavior is likely to be different (yet similar) in a blend with higher viscosity and many more excipients. This is important for the wet blend holding time during manufacturing.
  • Basifying agents i.e., additives that cause pH to increase
  • Basifying agents i.e., additives that cause pH to increase
  • Comparing the Controls A and B demonstrates the threshold for the maximum amount of EDTA that can be added to the solution while maintaining MTL solubility. Adding a portion of NaOH basifying agent to these mixtures allows more EDTA to be added while maintaining solubilized MTL. However, this ratio does not scale linearly. For example, lg of a 1M NaOH is sufficient to solubilize MTL in the presence of 0.225g EDTA, however if we increase the EDTA to 0.3 OOg, even tripling the amount of NaOH does not solubilize the MTL.
  • FIG. 6 is a graphical representation of the solubility limits of MTL in solutions containing EDTA.
  • the amount of MTL and water used are kept constant and are proportional to what is found in the formula.
  • Increasing amounts of basifying agents (NaOH and TEA) and EDTA are used and we visually monitor the precipitation of MTL.
  • the arrows for each sample listed terminate when MTL precipitates from solution. It was found that in a solution containing only water and MTL, we can add up to 0.15 g of EDTA after which the MTL begins to precipitate due to acidification from the EDTA.
  • Adding the basifying agents allows more EDTA to be added to the solution without precipitating MTL; the greatest increase in amount of EDTA added was found using TEA.
  • a third possibility for basifying the blend to compensate for the acidity of EDTA and other chelators, is to actually increase the amount of dissolved MTL.
  • MTL itself contributes significantly to the basification of the solution and is freely soluble in pure water.
  • MTL is responsible for the needed basification/buffering of the blend at alkaline pH to allow incorporation of the minimum amount of EDTA required for stability.
  • the addition of too much MTL unexpectedly has a significant negative impact on the film mechanical properties and blending. As MTL % w/w increases, the film becomes increasingly brittle and sticky, leading to strong liner interactions, which prevents easy release of the product during packaging steps.
  • MTL The range and ratio of MTL with respect to EDTA is critical from this second perspective, so as to not generate a product with poor flexibility, mechanical strength and liner release, which will impede scaled up manufacturing. MTL also behaves as an amphiphilic molecule in solution, acting to stabilize bubbles and foam in the blend during mixing when present at high relative concentrations. This will slow down manufacturing as longer degassing conditions will need to be applied.
  • the ratio of MTL to EDTA is therefore quite sensitive for the development of a functional pharmaceutical product. Accordingly, stabilization of the montelukast film using
  • EDTA is preferred using a liquid or water soluble weak organic base such as TEA.
  • the film layer comprises between 0.01 to 0.04% dry w/w of BHT with between 1.6 to 2.0% dry w/w of
  • the preferred ratio of MTL to EDTA is about 1.00 MTL to about 0.15 EDTA. This preferred ratio balances MTL solubility and stability. According to the preferred embodiment, the ratio of MTL to EDTA is between 13 : 1 to 3 :2 to maintain the Montelukast soluble within the film and prevent precipitation.
  • alkaline buffering components The final strategy used to incorporate more EDTA into the blend to improve stability while maintaining MTL solubility, is the incorporation of alkaline buffering components.
  • An alkaline buffer will react with any free protons from EDTA that would normally acidify the blend, thereby allowing more EDTA to be added without a change in pH.
  • these mixtures were prepared by first making an appropriate buffer. The buffer used in our experiments was selected for use in maintaining alkaline environments; CHES.
  • Results indicate that when using the CHES buffer which maintains the pH at 9.3, MTL does not solubilize even after overnight mixing.
  • Illustrative, but non-limiting, examples of a formulation used to prepare a MTL oral films with EDTA are shown in Tables 18-24.
  • Formulations MTL01, MTL03 MTL05 MTL06 MTL07 MTL08 MTL 10 MTL 12 MTL13 and MTL14 are believed to be suitable for maintaining at least a portion of the MTL under a solubilized form within the film and improve the bioavailability of the Montelukast oral film when compared with Singulair® swallowable or chewable tablets.
  • MTL02, MTL 03, MTL 09 and MTLl l are provide an undesired dosage form in which the Montelukast precipitates and hence does not provide the desired improved bioavailability derived from alkaline surface pH.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Psychiatry (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
EP18774366.1A 2017-03-30 2018-03-29 Behandlungsverfahren und vorrichtung zur verbesserten bioverfügbarkeit von leukotrienrezeptorantagonisten Withdrawn EP3600265A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762478876P 2017-03-30 2017-03-30
PCT/CA2018/050389 WO2018176149A1 (en) 2017-03-30 2018-03-29 Method of treatment and device for the improved bioavailability of leukotriene receptor antagonists

Publications (1)

Publication Number Publication Date
EP3600265A1 true EP3600265A1 (de) 2020-02-05

Family

ID=63673863

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18774366.1A Withdrawn EP3600265A1 (de) 2017-03-30 2018-03-29 Behandlungsverfahren und vorrichtung zur verbesserten bioverfügbarkeit von leukotrienrezeptorantagonisten

Country Status (9)

Country Link
EP (1) EP3600265A1 (de)
JP (1) JP2020512309A (de)
KR (1) KR20190128637A (de)
CN (1) CN110381931A (de)
AU (1) AU2018241534A1 (de)
BR (1) BR112019018388A2 (de)
CA (1) CA3056944A1 (de)
MX (1) MX2019010573A (de)
WO (1) WO2018176149A1 (de)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020051709A1 (en) * 2018-09-14 2020-03-19 Intelgenx Corp. Method of treatment and device for the improved bio availability of montelukast, a leukotriene receptor antagonist
US11602504B2 (en) * 2018-11-05 2023-03-14 Intelgenx Corp. Lipophilic active oral film formulation and method of making the same
US20210393611A1 (en) * 2018-11-05 2021-12-23 Intelgenx Corp. Lipophilic active oral film formulation and method of making the same
CN110496124A (zh) * 2019-04-10 2019-11-26 中山大学附属第五医院 治疗脉管畸形的化合物
CN114931578A (zh) * 2022-05-16 2022-08-23 中国医学科学院皮肤病医院(中国医学科学院皮肤病研究所) 孟鲁司特在制备用于治疗***性红斑狼疮药物中的应用

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101074271B1 (ko) * 2009-06-25 2011-10-17 (주)차바이오앤디오스텍 불쾌한 맛을 효과적으로 은폐한 경구용 속용 필름
KR101077468B1 (ko) * 2011-03-04 2011-11-07 (주)차바이오앤디오스텍 안정한 경구용 속용 필름 제제
EA038558B1 (ru) * 2013-10-14 2021-09-14 Зим Лэбораториз Лимитед Водорастворимая фармацевтическая пленка с повышенной стабильностью
US9668970B2 (en) * 2013-12-02 2017-06-06 Intelgenx Corp. Film dosage form with extended release mucoadhesive particles
CN104784157B (zh) * 2015-04-04 2018-06-26 齐鲁制药有限公司 一种稳定的孟鲁司特口腔薄膜剂

Also Published As

Publication number Publication date
AU2018241534A1 (en) 2019-09-26
JP2020512309A (ja) 2020-04-23
MX2019010573A (es) 2019-10-24
CA3056944A1 (en) 2018-10-04
BR112019018388A2 (pt) 2020-04-07
KR20190128637A (ko) 2019-11-18
CN110381931A (zh) 2019-10-25
WO2018176149A1 (en) 2018-10-04

Similar Documents

Publication Publication Date Title
EP3600265A1 (de) Behandlungsverfahren und vorrichtung zur verbesserten bioverfügbarkeit von leukotrienrezeptorantagonisten
CA2998218C (en) Device and method of treating conditions associated with neuroinflammation
US20220395452A1 (en) Method of treatment and device for the improved bioavailability of leukotriene receptor antagonists
US20230201130A1 (en) Lipophilic active oral film formulation and method of making the same
TW201829365A (zh) 苯甲酸鈉之同素異形體形式及其用途
US20220362164A1 (en) Method of treatment and device for the improved bioavailability of leukotriene receptor antagonists
US10098861B1 (en) Pharmaceutical composition comprising sodium benzoate compound and clozapine, and uses thereof
EP3068396B1 (de) Schnell zerfallende formulierungen und verfahren zur verwendung
US11369579B2 (en) Polymorphic forms of sodium benzoate and uses thereof
EP3849553A1 (de) Behandlungsverfahren und vorrichtung zur verbesserten bioverfügbarkeit von montelukast, einem leukotrienrezeptorantagonist
CA3017526A1 (en) Method of treatment and device for the improved bioavailability of leukotriene receptor antagonists
WO2022155341A1 (en) Transdermal formulations for phosphodiesterase-5 inhibitors
TW202017568A (zh) 環絲胺酸化合物的製劑及其應用
US11890278B2 (en) Betel quid cessation therapy with nicotine and pilocarpine
JPWO2021062061A5 (de)

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190905

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20201001