WO2022109077A1 - Compositions pour une meilleure administration d'inhibiteurs de cgrp - Google Patents

Compositions pour une meilleure administration d'inhibiteurs de cgrp Download PDF

Info

Publication number
WO2022109077A1
WO2022109077A1 PCT/US2021/059801 US2021059801W WO2022109077A1 WO 2022109077 A1 WO2022109077 A1 WO 2022109077A1 US 2021059801 W US2021059801 W US 2021059801W WO 2022109077 A1 WO2022109077 A1 WO 2022109077A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
composition according
cgrp
weight
pain
Prior art date
Application number
PCT/US2021/059801
Other languages
English (en)
Inventor
Charles M. CONWAY
Gene M. Dubowchik
Rajesh Kumar
Original Assignee
Biohaven Pharmaceutical Holding Company Ltd.
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 Biohaven Pharmaceutical Holding Company Ltd. filed Critical Biohaven Pharmaceutical Holding Company Ltd.
Priority to MX2023005846A priority Critical patent/MX2023005846A/es
Priority to EP21895549.0A priority patent/EP4247364A1/fr
Priority to JP2023529881A priority patent/JP2023551146A/ja
Priority to AU2021385052A priority patent/AU2021385052A1/en
Priority to KR1020237016343A priority patent/KR20230088442A/ko
Priority to CA3202137A priority patent/CA3202137A1/fr
Priority to CN202180077330.9A priority patent/CN116456981A/zh
Priority to IL302948A priority patent/IL302948A/en
Publication of WO2022109077A1 publication Critical patent/WO2022109077A1/fr
Priority to ZA2023/04727A priority patent/ZA202304727B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2063Proteins, e.g. gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents

Definitions

  • the present invention relates to non-irritating, non-toxic compositions providing enhanced bioavailability of an active therapeutic ingredient. Specifically, the present invention relates to compositions containing carbohydrate surfactants for delivery of calcitonin gene-related peptide (CGRP) inhibitors to a subject and methods of their use.
  • CGRP calcitonin gene-related peptide
  • CGRP inhibitors are often combined with various surfactants in pharmaceutical compositions.
  • some compositions do not provide optimal delivery of the CGRP inhibitor due to its low oral bioavailability.
  • some surfactants may be irritating to mucosal membranes.
  • An ideal bioavailability enhancing surfactant will be non-toxic and non-irritable to the skin or mucosal surfaces, and enhance the passage or absorption of the CGRP inhibitor through membrane barriers without damaging the structural integrity and biological function of the membrane and increase bioavailability of the active therapeutic ingredient.
  • fast-dispersing dosage forms Upon disintegration in the oral cavity, the drug substance is swallowed resulting in pre-gastric absorption and ultimately gastric absorption.
  • Previously described fast-disperse dosage forms provide for the dosage form to disintegrate or dissolve when placed in the mouth in order to promote pre-gastric or gastric absorption of the active ingredient.
  • fast dispersing dosage forms that provide improved characteristics, such as speeding the onset of drug action and reducing the first-pass effect drug metabolism.
  • the present invention is directed to pharmaceutical compositions and methods for increasing bioavailability of a calcitonin gene-related peptide (CGRP) inhibitors.
  • CGRP calcitonin gene-related peptide
  • An embodiment provides a pharmaceutical composition including a CGRP inhibitor and an absorption increasing amount of a carbohydrate surfactant.
  • Another embodiment provides a pharmaceutical composition including a CGRP inhibitor and an absorption increasing amount of a carbohydrate surfactant, wherein the pharmaceutical composition is in a form of an oral solid molded fast-dispersing dosage form.
  • Another embodiment provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of zavegepant, a solvate thereof, or a pharmaceutically acceptable salt thereof, wherein the pharmaceutical composition is in a form of an oral solid molded fast-dispersing dosage form.
  • Another embodiment provides a method for increasing bioavailability of a CGRP inhibitor in a subject, including orally administering any of the above pharmaceutical compositions.
  • Another embodiment provides a method for treating migraine in a subject in need thereof, including orally administering to the subject any of the above pharmaceutical compositions.
  • Another embodiment provides a method for providing rapid onset of migraine pain relief in a subject in need thereof, including orally administering to the subject any of the above pharmaceutical compositions.
  • Another embodiment provides a method for providing a reduced incidence of migraine pain recurrence in a subject in need thereof, including orally administering to the subject any of the above pharmaceutical compositions.
  • Another embodiment provides a method for treatment or prevention of a condition associated with aberrant levels of CGRP in a subject in need thereof, wherein the method comprises administering to the subject any of the above pharmaceutical compositions.
  • FIG. 1 is a graph of plasma concentration (nanograms per milliliter, ng/mL) versus time (hours, hr) showing average BHV-3500 concentration in dog plasma (ng/mL) after a single 50 milligram (mg) sublingual tablet administration.
  • the embodiments are merely described below, by referring to structures and schemes, to explain aspects of the present description.
  • the term “and/or” includes any and all combinations of one or more of the associated listed items.
  • the term “or” means “and/or.” Expressions such as "at least one of,” when preceding a list of elements, modify the entire list of elements and do not modify the individual elements of the list.
  • first, second, third etc. may be used herein to describe various elements, components, regions, layers, and/or sections, these elements, components, regions, layers, and/or sections should not be limited by these terms. These terms are only used to distinguish one element, component, region, layer, or section from another element, component, region, layer, or section. Thus, a first element, component, region, layer, or section discussed below could be termed a second element, component, region, layer, or section without departing from the teachings of the present embodiments.
  • substituted refers to a group substituted with deuterium, a halogen (-F, -Cl, -Br, -I), a hydroxy group (-OH), an amino group (-NH 2 ), a carboxyl group (-CO 2 H), a substituted or unsubstituted C1-C10 amine group, a nitro group (- NO 2 ), a C1-C10 alkyl group, a C3-C10 cycloalkyl group, a C6-C12 aryl group, a C1-C10 alkoxy group, a Cl to CIO trifluoroalkyl group such as a trifluoromethyl group (-CF 3 ) and the like, or a cyano group (-CN) instead of at least one hydrogen of a substituting group or compound.
  • Embodiment of the present invention are directed to compositions for improved delivery of CGRP inhibitors to the subjects and methods of their use.
  • An embodiment provides a pharmaceutical composition including a calcitonin gene-related peptide (CGRP) receptor antagonist, and an absorption increasing amount of a carbohydrate surfactant.
  • CGRP INHIBITORS a calcitonin gene-related peptide (CGRP) receptor antagonist, and an absorption increasing amount of a carbohydrate surfactant.
  • composition according to embodiments of the present invention includes a calcitonin gene- related peptide (CGRP) inhibitor.
  • CGRP inhibitor refers to a chemical entity that may be an inhibitor of a CGRP ligand or CGRP receptor.
  • CGRP inhibitor encompasses CGRP receptor inhibitors.
  • CGRP calcitonin gene-related peptide
  • CGRP is a 37 amino acid neuropeptide, which belongs to a family of peptides that includes calcitonin, adrenomedullin, and amylin.
  • Substantial evidence has been collected to show that CGRP is implicated in pathophysiology of migraine.
  • Clinical trials were carried out to prove that CGRP inhibitors are effective in treating migraine. Following clinical trials, several CGRP inhibitors have been approved by regulatory authorities and marketed for treatment of acute migraine and migraine prevention.
  • the CGRP inhibitor may be a CGRP antibody, a CGRP receptor antibody, an antigen-binding fragment from a CGRP antibody or a CGRP receptor antibody, a CGRP infusion inhibitory protein, a CGRP bio-neutralizing agent, a small molecule CGRP receptor antagonist, a small molecule CGRP inhibitor, or a polypeptide CGRP inhibitor.
  • the CGRP inhibitor may be a CGRP receptor antagonist.
  • the CGRP receptor antagonists in accordance with the present invention are preferably non-biologic CGRP antagonists. That is, the nonbiologic CGRP receptor antagonists of the present invention preferably do not contain antibodies, antibody fragments, or peptides.
  • the CGRP receptor antagonist may be a small molecule receptor antagonist.
  • the small molecule CGRP receptor antagonists in accordance with the present invention contain molecules with a mass of less than about 900 Daltons, for example, less than about 800 Daltons, less than about 700 Daltons, less than about 600 Daltons, less than about 500 Daltons, less than about 400 Daltons, or less than about 300 Daltons.
  • non-biologic CGRP antagonists include, rimegepant, zavegepant, ubrogepant, atogepant, telcagepant, and olcegepant.
  • the small molecule CGRP receptor antagonist may be (R)-N-(3-(7-methyl-lH-indazol-5- yl)-l-(4-(l-methylpiperidin-4-yl)piperazin-l-yl)-l-oxopropan-2-yl)-4-(2-oxo-l,2-dihydroquinolin-3- yl)piperidine-l-carboxamide (zavegepant).
  • Rimegepant has the chemical formula, CzsF sFzNgOs and the IUPAC name [(5S,6S,9R)-5-amino-6- (2,3-difluorophenyl)-6,7,8,9-tetrahydro-5H-cyclohepta[b]pyridin-9-yl] 4-(2-oxo-3H-imidazo[4,5- b]pyridin-l-yl)piperidine-l-carboxylate. Rimegepant is also known as and referred to herein as BHV- 3000.
  • Rimegepant is described, for example, in WO 2011/046997 published April 21, 2011.
  • rimegepant is present in the form of a hemisulfate sesquihydrate salt. This preferred salt form is described in WO 2013/130402 published September 6, 2013.
  • CGRP antagonist is zavegepant (also known as BHV-3500), which is described in WO
  • Another CGRP antagonist is ubrogepant, which has the following structure:
  • Another CGRP antagonist is atogepant, which has the following structure:
  • telcagepant which has the following structure:
  • the CGRP inhibitor may have low oral bioavailability.
  • the low oral bioavailability may be 80% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, or 5% or less.
  • compositions described herein may include 1-1000 mg of the CGRP inhibitor.
  • the compositions may include about 1, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, 250, 300, 400, 500, 600, 700, 800, or 900 mg of the CGRP inhibitor.
  • the amount of the CGRP inhibitor may range between any of the above values.
  • the CGRP inhibitor may be administered at a dose of about 1-1000 mg per day. In another aspect, the CGRP inhibitor is administered at a dose of about 1, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, 250, 300, 400, 500, 600, 700, 800, or 900 mg per day.
  • the daily dose of the CGRP inhibitor may range between any of the above values.
  • the daily dose of the CGRP inhibitor may range between any of the above values.
  • the composition including a CGRP inhibitor may be administered as a single dose.
  • the CGRP inhibitor may be administered for at least one week and for as long as needed.
  • the CGRP inhibitor may be administered for one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, or twelve weeks.
  • the composition further includes a carbohydrate surfactant in an absorption increasing amount.
  • a "carbohydrate” is inclusive of monocarbohydrates, oligocarbohydrates, or polycarbohydrates in straight chain or ring forms, or a combination thereof to form a carbohydrate chain.
  • Oligocarbohydrates are carbohydrates having two or more but less than 100 monohydrate residues.
  • Polycarbohydrates include 100 or more monohydrates residues.
  • the carbohydrate may be chosen, for example, from any currently commercially available monocarbohydrate species or may be synthesized.
  • Some examples of the many possible carbohydrates to use include glucose, maltose, maltotriose, maltotetraose, sucrose, and trehalose.
  • Preferable carbohydrates include maltose, sucrose, and glucose.
  • the carbohydrate surfactant may be an alkyl glycoside.
  • alkyl glycoside refers to any carbohydrate joined by a linkage to any hydrophobic alkyl, as is known in the art.
  • the alkyl glycoside is preferably non-toxic and non-ionic, and increases the absorption of a CGRP inhibitor when it is administered with the compound via the oral, ocular, intranasal, nasolacrimal, nose-to-brain, inhalation or pulmonary, oral cavity (sublingual or Buccal cell), or cerebrospinal fluid (CSF) delivery route.
  • Suitable compounds can be determined using the methods set forth herein.
  • Alkyl glycosides disclosed herein may be synthesized by known procedures, i.e., chemically, as described, for example, in Rosevear et al., Biochemistry 19:4108-4115 (1980) or Koeltzow and Urfer, J. Am. Oil Chem. Soc., 61:1651-1655 (1984), U.S. Pat. No. 3,219,656 and U.S. Pat. No. 3,839,318 or enzymatically, as described, for example, in Li et al., J. Biol. Chem., 266:10723-10726 (1991) or Gopalan et al., J. Biol. Chem. 267:9629-9638 (1992).
  • Alkyl glycosides of the present invention may include, but are not limited to: alkyl glycosides, such as octyl-, nonyl-, decyl-, undecyl-, dodecyl-, tridecyl-, tetradecyl-, pentadecyl-, hexadecyl-, heptadecyl-, and octadecyl-a- or p-D-maltoside, -glucoside or -sucroside (synthesized according to Koeltzow and Urfer; Anatrace Inc., Maumee, Ohio; Calbiochem, San Diego, Calif.; Fluka Chemie, Switzerland); alkyl thiomaltosides, such as heptyl, octyl, dodecyl-, tridecyl-, and tetradecyl-p-D- thiomaltoside (synthe
  • alkyl thiosucroses (synthesized according to, for example, Binder, T. P. and Robyt, J. F., Carbohydr. Res. 140:9-20 (1985)); alkyl maltotriosides (synthesized according to Koeltzow and Urfer); long chain aliphatic carbonic acid amides of sucrose p-amino-alkyl ethers; (synthesized according to Austrian Patent 382,381 (1987); Chem. Abstr., 108:114719 (1988) and Gruber and Greber pp.
  • the carbohydrate surfactant may be a carbohydrate ester.
  • carbohydrate ester refers to a carbohydrate ester of any fatty acids. Carbohydrate esters may take many forms because of several hydroxyl groups in the carbohydrate are available for reaction and the many fatty acid groups, from acetate on up to larger, more bulky fatty acids that may be reacted with the carbohydrate. This flexibility means that many products and functionalities may be tailored, based on the fatty acid moiety used. Carbohydrate esters have food and non-food uses, especially as surfactants and emulsifiers, with growing applications in pharmaceuticals, cosmetics, detergents and food additives. They are biodegradable, non-toxic, and mild to the skin.
  • the carbohydrate ester may be a sucrose ester.
  • the carbohydrate surfactants disclosed herein may have a hydrophobic alkyl group linked to a hydrophilic carbohydrate.
  • the linkage between the hydrophobic alkyl group and the hydrophilic carbohydrate may include, among other possibilities, a glycosidic, thioglycosidic (Horton), amide (Carbohydrates as Organic Raw Materials, F. W. Lichtenthaler ed., VCH Publishers, New York, 1991), ureide (Austrian Patent 386,414 (1988); Chem. Abstr. 110: 137536p (1989); see Gruber, H. and Greber, G., "Reactive Sucrose Derivatives" in Carbohydrates as Organic Raw Materials, pp.
  • glycosides may include maltose, sucrose, and glucose linked by glycosidic linkage to an alkyl chain of about 9-16 carbon atoms, e.g., nonyl-, decyl-, dodecyl- and tetradecyl sucroside, glucoside, and maltoside, but are not limited thereto. These compositions are amphipathic and non-toxic, because they degrade to an alcohol and an oligocarbohydrate.
  • carbohydrate surfactants may include an alkyl glycoside and/or a carbohydrate ester having characteristic hydrophile-lipophile balance (HLB) numbers, which may be calculated or determined empirically (Schick, M. J. Nonionic Surfactants, p. 607 (New York: Marcel Dekker, Inc. (1967)).
  • HLB number is a direct reflection of the hydrophilic character of the surfactant, i.e., the larger the HLB number, the more hydrophilic the compound.
  • the HLB number is a direct expression of the hydrophilic character of the surfactant, i.e., the larger the HLB number, the more hydrophilic the compound.
  • the carbohydrate surfactant may have an HLB number of from about 10 to 20, for example, from about 11 to 15.
  • the hydrophobic alkyl may be chosen of any desired size, depending on the hydrophobicity desired and the hydrophilicity of the carbohydrate moiety.
  • a range of alkyl chains may be from about 9 to about 24 carbon atoms, for example, from about 9 to about 16 or about 14 carbon atoms.
  • Some glycosides may include maltose, sucrose, and glucose linked by glycosidic linkage to an alkyl chain of 9, 10, 12, 13, 14, 16, 18, 20, 22, or 24 carbon atoms, e.g., nonyl-, decyl-, dodecyl- and tetradecyl sucroside, glucoside, and maltoside, but are not limited thereto.
  • These compositions are non-toxic, since they are degraded to an alcohol and an oligocarbohydrate, and amphipathic.
  • compositions may be administered as a tablet, a capsule, a suppository, a drop, a spray, or an aerosol.
  • the compositions may be administered as oral fast-disintegrating tablets.
  • the compositions may also be administered in a sustained release or delayed burst format.
  • the spray and the aerosol administration may be achieved through use of an appropriate dispenser.
  • the sustained release format may be an ocular insert, erodible microparticulates, swelling mucoadhesive particulates, pH sensitive microparticulates, nanoparticles/latex systems, ion-exchange resins and other polymeric gels and implants (Ocusert, Alza Corp., California; Joshi, A., S. Ping and K. J. Himmelstein, Patent Application ⁇ NO 91/19481). These systems maintain prolonged drug contact with the absorptive surface preventing washout and nonproductive drug loss. The prolonged drug contact is non-toxic to the skin and mucosal surfaces.
  • compositions disclosed herein are stable.
  • Baudys et al. in U.S. Patent No. 5,726,154 show that calcitonin in an aqueous liquid composition including SDS (sodium dodecyl sulfate, a surfactant) and an organic acid is stable for at least 6 months.
  • the surfactant compositions of the present invention have improved stabilizing characteristics when admixed with a CGRP inhibitor. No organic acid is required in these formulations.
  • the composition may maintain the stability of CGRP inhibitor for about 6 months, or more, when maintained at about 4° C to 25° C.
  • NOAEL no observable adverse effect level
  • EPA Environmental Protection Agency
  • NOAEL no observable adverse effect level
  • NOAEL no observable adverse effect level
  • the NOAEL for sucrose dodecanoate is about 20-30 grams/kilogram/day, e.g., a 70 kilogram person (about 154 pounds) can consume about 1400-2100 grams (or about 3 to 4.6 pounds) of sucrose dodecanoate per day without any observable adverse effect.
  • an acceptable daily intake for humans is about 1% of the NOAEL, which translates to about 14-21 grams, or 14 million micrograms to 21 million micrograms, per day, indefinitely. Definitions of NOAELs and other related definitions may be found on the world wide web at epa.gov/OCEPAterms. Thus, although some effects may be produced with alkyl glycoside levels anticipated in the present invention, the levels are not considered adverse, or precursors to adverse effects.
  • a subject treated with the compositions, according to embodiments of the invention having at least one alkyl glycoside, e.g., tetradecylmaltoside (TDM; or Intravail A), at a concentration of about 0.125% by weight of alkyl glycoside two times per day, or three times per day, or more depending on the treatment regimen consumes about 200 to 300 micrograms per day total of TDM.
  • the effective dose of the TDM is at least 100-fold lower than (/.e., 1/1000) of the NOAEL, and falls far below 1% of the NOAEL, which is the acceptable daily intake; or in this case about 1/50,000 of the acceptable daily intake.
  • alkyl glycosides disclosed herein have a high NOAEL, such that the amount or concentration of alkyl glycosides used do not cause an adverse effect and can be safely consumed without any adverse effect.
  • compositions are also stable because they are physiologically non-toxic and non-irritants.
  • non-toxic means that the alkyl glycoside molecule has a sufficiently low toxicity to be suitable for human administration and consumption.
  • Preferred alkyl glycosides are non-irritating to the tissues to which they are applied. Any alkyl glycoside used should be of minimal or no toxicity to the cell, such that it does not cause damage to the cell. Yet, toxicity for any given alkyl glycoside may vary with the concentration of alkyl glycoside used.
  • non-irritant means that the agent does not cause inflammation following immediate, prolonged or repeated contact with the skin surface or mucous membranes.
  • compositions disclosed herein are typically present in an amount from about 0.01% to 20% by weight based on 100% of the composition weight.
  • the compositions may be present in an amount from about 0.01% to 5% by weight, from about 0.01% to 2% by weight, from about 0.01% to 1%, or from about 0.01% to 0.125% by weight based on 100% of the composition weight.
  • the carbohydrate surfactant may be formulated to be compatible with other components present in the composition. In liquid, or gel, or capsule, or injectable, or spray compositions the carbohydrate surfactant may be formulated such that it promotes, or at least does not degrade, the stability of the CGRP inhibitor. Further, the compositions optimize the concentration by keeping the concentration of absorption enhancer as low as possible, while still maintaining the desired effect.
  • compositions disclosed herein when administered to the subject, yield enhanced mucosal delivery of the CGRP inhibitor with a peak concentration (or C max ) in a tissue, or fluid, or in a blood plasma of the subject that is about 1.15%, 1.25%, 1.50%, 1.75%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% or greater as compared to a C max of the compound(s) in a tissue (e.g., central nervous system or CNS), or fluid, or blood plasma following intramuscular injection of an equivalent concentration of the compound(s) to the subject.
  • a tissue e.g., central nervous system or CNS
  • CNS central nervous system
  • the measure of how much of the CGRP inhibitor reaches the bloodstream in a set period of time can also be calculated by plotting drug blood concentration at various times during a 24-hour or longer period and then measuring the area under the curve (AUC) between 0 and 24 hours.
  • a measure of drug efficacy may also be determined from a time to maximal concentration (T max ) of the biologically active compound(s) in a tissue (e.g., CNS) or fluid or in the blood plasma of the subject between about 0.1 to 1.0 hours.
  • the therapeutic compositions disclosed herein increase the speed of onset of drug action (i.e., reduce T max ) by a factor of about 1.5-fold or greater, for example, about 1.5 to about 5-fold, about 1.5 to about 4-fold, about 1.5 to about 3-fold, or about 1.5 to about 2- fold.
  • the therapeutic compositions or formulations can be administered or delivered to a subject in need systemically or locally.
  • Suitable routes may, for example, include oral, ocular, nasal, nose-to-brain, nasolacrimal, inhalation or pulmonary, oral cavity (sublingual or Buccal cell), transmucosal administration, vaginal, rectal, parenteral delivery, including intramuscular, subcutaneous, intravenous, intraperitoneal, or CSF delivery.
  • the mode of delivery e.g., liquid, gel, tablet, spray, etc. will also depend on the method of delivery to the subject.
  • the therapeutic compositions may include a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to an aqueous or non-aqueous agent, for example alcoholic or oleaginous, or a mixture thereof, which may contain a surfactant, emollient, lubricant, stabilizer, dye, perfume, preservative, acid or base for adjustment of pH, a solvent, emulsifier, gelling agent, moisturizer, stabilizer, wetting agent, time release agent, humectant, or other component commonly included in a particular form of pharmaceutical composition.
  • Pharmaceutically acceptable carriers include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, and oils such as olive oil or injectable organic esters.
  • a pharmaceutically acceptable carrier may contain physiologically acceptable compounds that act, for example, to stabilize or to increase the absorption of the specific inhibitor, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • a pharmaceutically acceptable carrier may also be selected from substances such as distilled water, benzyl alcohol, lactose, starches, talc, magnesium stearate, polyvinylpyrrolidone, alginic acid, colloidal silica, titanium dioxide, and flavoring agents.
  • alkyl carbohydrates or carbohydrate alkyl esters to hydrolytic cleavage of the CGRP inhibitor, various oxygen atoms within the alkyl carbohydrate or the carbohydrate alkyl ester may be substituted for by sulfur (Defaye, J. and Gelas, J. in Studies in Natural Product Chemistry (Atta-ur-Rahman, ed.) Vol. 8, pp. 315-357, Elsevier, Amsterdam, 1991).
  • the heteroatom of the carbohydrate ring may be either oxygen or sulfur, or the linkage between monocarbohydrate residues in an oligocarbohydrate may be oxygen or sulfur (Horton, D. and Wander, J.
  • Oligocarbohydrates may have either a (alpha) or (beta) anomeric configuration (see Pacsu, E., et al. in Methods in Carbohydrate Chemistry (R. L. Whistler, et al., eds.) Vol. 2, pp. 376-385, Academic Press, New York 1963).
  • a composition may be prepared in tablet form by mixing a CGRP inhibitor and one alkyl glycoside and/or carbohydrate alkyl ester, and an appropriate pharmaceutical carrier or excipient, for example mannitol, corn starch, polyvinylpyrrolidone or the like, granulating the mixture and finally compressing it in the presence of a pharmaceutical carrier such as corn starch, magnesium stearate or the like.
  • an appropriate pharmaceutical carrier or excipient for example mannitol, corn starch, polyvinylpyrrolidone or the like
  • the formulation thus prepared may include a sugar-coating or enteric coating or covered in such a way that the active principle is released gradually, for example, in the appropriate pH medium.
  • enteric coating is a polymer encasing, surrounding, or forming a layer, or membrane around the therapeutic composition or core.
  • the enteric coating may contain a CGRP inhibitor which is compatible or incompatible with the coating.
  • a tablet composition may include an enteric coating polymer with a compatible CGRP inhibitor which dissolves or releases the inhibitor at higher pH levels (e.g., pH greater than 4.0, greater than 4.5, greater than 5.0 or higher) and not at low pH levels (e.g., pH 4 or less); or the reverse.
  • the dependent release form of the invention may be a tablet including:
  • a surfactant including at least one alkyl glycoside and/or carbohydrate alkyl ester
  • membrane coating surrounding the core, wherein the coating is an impermeable, permeable, semi-permeable, or porous coating, and becomes more permeable or porous upon contacting an aqueous environment of a defined pH.
  • membrane is synonymous with "coating,” or equivalents thereof. The terms are used to identify a region of a medicament, for example, a tablet, that is impermeable, permeable, semi-permeable or porous to an aqueous solution(s) or bodily fluid(s), and/or to the therapeutic agent(s) or drug(s) encapsulated therein.
  • the inhibitor may be released through the openings or pores of the membrane in solution or in vivo.
  • the porous membrane may be manufactured mechanically (e.g., drilling microscopic holes or pores in the membrane layer using a laser), or it may be imparted due to the physiochemical properties of the coating polymer(s).
  • Membrane or coating polymers of the invention are well-known in the art, and include cellulose esters, cellulose diesters, cellulose triesters, cellulose ethers, cellulose ester-ether, cellulose acylate, cellulose diacylate, cellulose triacylate, cellulose acetate, cellulose diacetate, cellulose triacetate, cellulose acetate propionate, and cellulose acetate butyrate.
  • Other suitable polymers are described in U.S. Patent Nos. 3,845,770, 3,916,899, 4,008,719, and 4,036,228.
  • the enteric coating described herein may include a plasticizer, and a sufficient amount of sodium hydroxide (NaOH) to effect or adjust the pH of the suspension in solution or in vivo.
  • plasticizers include triethyl citrate, triacetin, tributyl sebecate, or polyethylene glycol.
  • alkalizing agents including potassium hydroxide, calcium carbonate, sodium carboxymethylcellulose, magnesium oxide, and magnesium hydroxide can also be used to effect or adjust the pH of the suspension in solution or in vivo.
  • an enteric coating may be designed to release a certain percentage of a CGRP inhibitor in certain mediums with a certain pH or pH range.
  • the composition may include at least one enteric coating encasing or protecting at least one CGRP inhibitor, which is chemically unstable in an acidic environment (e.g., the stomach).
  • the enteric coating protects the CGRP inhibitor from the acidic environment (e.g., pH ⁇ 3), while releasing the inhibitor in locations which are less acidic, for example, regions of the small and large intestine where the pH is 3, or 4, or 5, or greater.
  • a medicament of this nature will travel from one region of the gastrointestinal tract to the other, for example, it takes about 2 to about 4 hours for a CGRP inhibitor to move from the stomach to the small intestine (duodenum, jejunum and ileum).
  • the pH changes from about 3 (e.g., stomach) to 4, or 5, or to about a pH of 6 or 7 or greater.
  • the enteric coating allows the core containing the CGRP inhibitor to remain substantially intact, and prevents premature release of the inhibitor or the acid from penetrating and de-stabilizing the CGRP inhibitor.
  • enteric polymers include, but are not limited to, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, polyvinylacetate phthalate, methacrylic acid copolymer, shellac, cellulose acetate trimellitate, hydroxypropylmethylcellulose acetate succinate, hydroxypropylmethylcellulose phthalate, cellulose acetate phthalate, cellulose acetate succinate, cellulose acetate malate, cellulose benzoate phthalate, cellulose propionate phthalate, methylcellulose phthalate, carboxymethylethylcellulose, ethylhydroxyethylcellulose phthalate, shellac, styrene-acrylic acid copolymer, methyl acrylate-acrylic acid copolymer, methyl acrylate-methacrylic acid copolymer, butyl acrylate-styrene-acrylic acid copolymer, methacrylic acid-methyl methacrylate copolymer,
  • the therapeutic compositions of the invention in the form of a tablet may have a plurality of coatings, for example, a hydrophilic coating (e.g., hydroxypropylmethylcellulose), and/or a hydrophobic coating (e.g., alkylcelluloses), and/or an enteric coating.
  • a hydrophilic coating e.g., hydroxypropylmethylcellulose
  • a hydrophobic coating e.g., alkylcelluloses
  • an enteric coating e.g., the tablet core may be encases by a plurality of the same type of coating, or a plurality of different types of coating selected from a hydrophilic, hydrophobic or enteric coating.
  • a tablet may be designed having at least one, but can have more than one layer consisting of the same or different coatings dependent on the target tissue or purpose of the CGRP inhibitor.
  • the tablet core layer may have a first composition enclosed by a first coating layer (e.g., hydrophilic, hydrophobic, or enteric coating), and a second same or different composition or CGRP inhibitor having the same or different dosage may be enclosed in second coating layer, etc.
  • first coating layer e.g., hydrophilic, hydrophobic, or enteric coating
  • second same or different composition or CGRP inhibitor having the same or different dosage may be enclosed in second coating layer, etc.
  • This layering of various coatings provides for a first, second, third, or more gradual or dose dependent release of the same or different CGRP inhibitor containing composition.
  • a first dosage of a first composition of the invention is contained in a tablet core and with an enteric-coating, such that the enteric-coating protects and prevents the composition contained therein from breaking down or being released into the stomach.
  • the first loading dose of the therapeutic composition is included in the first layer and includes from about 10% to about 40% of the total amount of the total composition included in the formulation or tablet.
  • a second loading dose another percentage of the total dose of the composition is released.
  • the invention contemplates as many time release doses as desired in a treatment regimen.
  • a single coating or plurality of coating layers may be in an amount ranging from about 2% to 6% by weight, for example, about 2% to about 5%, for example, from about 2% to about 3% by weight of the coated unit dosage form.
  • the formulations according to embodiments of the invention make it possible for contents of a hard capsule or tablet to be selectively released at a desired site the more distal parts of the gastro-intestinal tract (e.g., small and large intestine) by selecting a suitable pH-soluble polymer for a specific region.
  • Mechanical expulsion of the composition preparations may also be achieved by inclusion of a water absorbing polymer that expands upon water absorption within a hard semi- permeable capsule, thus expelling composition through an opening in the hard capsule.
  • the specific dose level and frequency of dosage for any particular subject in need of treatment may be varied and will depend upon a variety of factors including the activity of the specific CGRP inhibitor employed, the metabolic stability, and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • Alkyl glycosides for example, alkylmaltosides, for example, dodecyhnaltoside (DDM) and tetradecylmaltoside (TDM), may stabilize the CGRP inhibitor in solution and prevent its aggregation.
  • an aspect of the invention is to provide therapeutic compositions having at least one CGRP inhibitor and one carbohydrate surfactant, wherein the surfactant further includes at least one alkyl glycoside and/or carbohydrate alkyl ester formulation, which enhances the bioavailability of the CGRP inhibitor. Determining the bioavailability of drug formulations is described herein.
  • bioavailability is the rate and extent to which the active substance, or moiety, which reaches the systemic circulation as an intact drug. The bioavailability of any drug will depend on how well it is adsorbed and how much of it escapes removal by the liver.
  • the tested drug and mode of administration is measured against an intravenous reference dose.
  • the bioavailability of the intravenous dose is 100% by definition.
  • animals or volunteering humans are given an intravenous injections and corresponding oral doses of a drug.
  • Urinary or plasma samples are taken over a period of time and levels of the drug over that period of time are determined.
  • AUC areas under the curve
  • C max peak concentration of the plasma drug concentration is also measured to the peak concentration (C max ) of the plasma drug concentration following intramuscular (IM) injection of an equivalent concentration the drug.
  • the time to maximal concentration (or T max ) of the plasma drug is about 0.1 to 1.0 hours.
  • bioavailability of the formulations are assessed against each other as one or both drugs could be subject to first pass clearance (discussed more below), and thus, undetected.
  • a first oral formulation is assessed against a second oral formulation.
  • the second formulation is used as a reference to assess the bioavailability of the first. This type of study provides a measure of the relative performance of two formulations in getting a drug absorbed.
  • the alkyl glycosides or carbohydrate include any compounds now known or later discovered.
  • CGRP inhibitors which are particularly well- suited for admixture with the alkyl glycosides and/or carbohydrate alkyl esters are those that are difficult to administer by other methods, e.g., compounds that are degraded in the gastrointestinal (Gl) tract or those that are not absorbed well from the Gl tract, or compounds that can be self-administered via the oral, ocular, nasal, nasolacrimal, inhalation, sublingual, or CSF delivery route instead of traditional methods such as injection.
  • Gl gastrointestinal
  • bioavailability of a CGRP inhibitor can be determined by measuring the levels of the drug's first pass clearance by the liver.
  • Alkyl glycosides and/or carbohydrate alkyl ester compositions of the invention administered intranasally or via oral cavity (sublingual or Buccal cell) do not enter the hepatic portal blood system, thereby avoiding first pass clearance by the liver. Avoiding first past clearance of these formulations by the liver is described herein.
  • first pass liver clearance is the extent to which the drug is removed by the liver during its first passage in the portal blood through the liver to the systemic circulation. This is also called first pass metabolism or first pass extraction.
  • the two major routes of drug elimination from the body are excretion by the kidneys whereby the drug is unchanged; and elimination by the liver, whereby the drug is metabolized.
  • the balance between these two routes depends on the relative efficiency of the two processes.
  • the present invention describes herein elimination by the liver or liver clearance.
  • First pass liver clearance is described by Birkett et al (1990 and 1991), which is incorporated by reference in its entirety. Birkett et al., Aust Prescr, 13 (1990):88-9; and Birkett et al., Austra Prescr, 14:14-16 (1991).
  • Blood carrying drug from the systemic circulation enter the liver via the portal vein, and the liver in turn extracts a certain percentage or ratio (/.e., 0.5 or 50%) of that drug.
  • the remainder left over (/.e., 0.2 or 20%) re-enters the systemic circulation via the hepatic vein.
  • This rate of clearance of the drug is called the hepatic extraction ratio. It is the fraction of the drug in the blood which is irreversibly removed (or extracted) during the first pass of the blood through the liver. If no drug is extracted, the hepatic extraction ratio is zero. Conversely, if the drug is highly extracted in the first pass through the liver, the hepatic extraction ratio may be as high as 100% or 1.0.
  • the "unbound fraction" of drug is dependent on how tightly the drug is bound to proteins and cells in the blood. In general, it is only this unbound (or free) drug which is available for diffusion from the blood into the liver cell. In the absence of hepatic blood flow and protein binding, the "intrinsic clearance” is the ability of the liver to remove (or metabolize) that drug. In biochemical terms, it is a measure of liver enzyme activity for a particular drug substrate. Again, although intrinsic clearance can be high, drugs cannot be cleared more rapidly than that presented to the liver. In simple terms, there are two situations: where liver enzyme activity is very high or very low (/.e., high extraction ratio or low extraction ratio).
  • Clearance then is independent of blood flow, but instead depends directly on the degree of protein binding in the blood and the activity of drug metabolizing enzymes towards that drug.
  • the liver removes most of the drug presented to it and the extraction ratio is high.
  • the only factor determining the actual hepatic clearance is the rate of supply of drug to the liver (or hepatic blood flow).
  • First pass liver clearance is important because even small changes in the extraction of drugs can cause large changes in bioavailability. For example, if the bioavailability of drug A by oral administration is 20% by the time it reaches the systemic circulation, and the same drug A by intravenous administration is 100%, absent no other complicating factors, the oral dose will therefore have to be 5 times the intravenous dose to achieve similar plasma concentrations.
  • drug formulations should be designed to have the drug pass directly through to the systemic circulation and avoid first pass liver clearance all together.
  • drugs administered intranasally, sublingual, buccal, rectal, vagina, etc. directly enter the systemic circulation and do not enter the hepatic portal blood circulation to be partially or fully extracted by the liver.
  • a tablet with at least one enteric-coating layer to prevent release of the drug in the stomach is provided.
  • an objective of the invention is to administer drugs using these alternative routes.
  • first pass liver clearance is an important factor because many patients are on more than one drug regimen, and this may cause drug interactions which increase or decrease liver enzyme activity; thereby increasing or decreasing metabolism (increasing or decreasing the hepatic extraction ratio) of the drug of interest.
  • compositions of the invention may be administered directly to the systemic circulatory system and avoid first pass liver clearance. Avoiding first pass clearance assures that more of the drug will be available to the system. Stated another way, by avoiding first pass liver clearance, the bioavailability of the drug is increased.
  • Embodiments of the present invention also relate to methods of increasing absorption of a low molecular weight CGRP inhibitor into the circulatory system of a subject including administering via the oral, ocular, nasal, nasolacrimal, inhalation, or the CSF delivery route the compound and an absorption increasing amount of a suitable non-toxic, non-ionic alkyl glycoside having a hydrophobic alkyl joined by a linkage to a hydrophilic carbohydrate.
  • composition formulation is appropriately selected according to the administration route, such as oral administration (oral preparation), external administration (e.g., ointment), injection (preparations for injection), and mucosal administration (e.g., buccal and suppository) etc.
  • oral administration oral preparation
  • external administration e.g., ointment
  • injection preparations for injection
  • mucosal administration e.g., buccal and suppository
  • excipients e.g., starch, lactose, crystalline cellulose, calcium lactate, magnesium aluminometasilicate and anhydrous silicate
  • disintegrators e.g., carboxymethylcellulose and calcium carboxymethylcellulose
  • lubricants e.g., magnesium stearate and talc
  • coating agents e.g., hydroxyethylcellulose
  • flavoring agents can be used for oral and mucosal formulations
  • solubilizers and auxiliary solubilizers capable of forming aqueous injections (e.g., distilled water for injection, physiological saline and propylene glycol), suspending agents (e.g., surfactant such as polysorbate 80), pH regulators (e.g., organic acid and metal salt thereof) and stabilizers are used for injections; and aqueous or oily solubilizers and auxiliary solubilizers (e.g., alcohols and fatty acid esters), tack
  • the CGRP inhibitor and the alkyl glycoside can be admixed, mixed, or blended along with the above excipients, disintegrators, coating polymers, solubilizers, suspending agents, etc., prior to administration, or they can be administered sequentially, in either order. It is preferred that they be mixed prior to administration.
  • mucosal delivery-enhancing agent includes agents which enhance the release or solubility (e.g., from a formulation delivery vehicle), diffusion rate, penetration capacity and timing, uptake, residence time, stability, effective half-life, peak or sustained concentration levels, clearance and other desired mucosal delivery characteristics (e.g., as measured at the site of delivery, or at a selected target site of activity such as the bloodstream or central nervous system) of a compound(s) (e.g., biologically active compound).
  • Enhancement of mucosal delivery can occur by any of a variety of mechanisms, including, for example, by increasing the diffusion, transport, persistence or stability of the compound, increasing membrane fluidity, modulating the availability or action of calcium and other ions that regulate intracellular or paracellular permeation, solubilizing mucosal membrane components (e.g., lipids), changing non-protein and protein sulfhydryl levels in mucosal tissues, increasing water flux across the mucosal surface, modulating epithelial junction physiology, reducing the viscosity of mucus overlying the mucosal epithelium, reducing mucociliary clearance rates, and other mechanisms.
  • mucosal membrane components e.g., lipids
  • mucosal membrane components e.g., lipids
  • changing non-protein and protein sulfhydryl levels in mucosal tissues increasing water flux across the mucosal surface
  • modulating epithelial junction physiology reducing the viscosity of mucu
  • Exemplary mucosal delivery enhancing agents include the following agents and any combinations thereof: o (a) an aggregation inhibitory agent; o (b) a charge-modifying agent; o (c) a pH control agent; o (d) a degradative enzyme inhibitory agent; o (e) a mucolytic or mucus clearing agent; o (f) a ciliostatic agent; o (g) a membrane penetration-enhancing agent selected from:
  • a surfactant e.g. a surfactant; (ii) a bile salt; (ii) a phospholipid additive, mixed micelle, liposome, or carrier; (iii) an alcohol; (iv) an enamine; (v) an NO donor compound; (vi) a long-chain amphipathic molecule; (vii) a small hydrophobic penetration enhancer; (viii) sodium or a salicylic acid derivative; (ix) a glycerol ester of acetoacetic acid; (x) a cyclodextrin or beta-cyclodextrin derivative; (xi) a medium-chain fatty acid; (xii) a chelating agent; (xiii) an amino acid or salt thereof; (xiv) an N-acetylamino acid or salt thereof; (xv) an enzyme degradative to a selected membrane component; (ix) an inhibitor of fatty acid synthesis; (x) an inhibitor of cholesterol synthesis;
  • Additional mucosal delivery-enhancing agents include, for example, citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid (e.g., L-ascorbic acid), sodium metabisulfite, ethylenediaminetetraacetic acid (EDTA) disodium, benzalkonium chloride, sodium hydroxide, and mixtures thereof.
  • EDTA or its salts e.g., sodium or potassium
  • Compounds whose absorption may be increased by the method described herein include any CGRP inhibitor now known or later discovered, in particular compounds that are difficult to administer by other methods, for example, compounds that are degraded in the gastrointestinal (Gl) tract or that are not absorbed well from the Gl tract, or compounds that subjects could administer to themselves more readily via the oral, ocular, nasal, nose-to-brain, nasolacrimal, inhalation or pulmonary, oral cavity (sublingual or Buccal cell), or CSF delivery route than by traditional self-administration methods such as injection.
  • Gl gastrointestinal
  • varying amounts of a CGRP inhibitor may be absorbed as the CGRP inhibitor passes through the buccal, sublingual, oropharyngeal and oesophageal pregastric portions of the alimentary canal.
  • the bulk of the CGRP inhibitor passes into the stomach and is absorbed in the usual mode in which enteric dosage forms such as tablets, capsules, or liquids are absorbed.
  • enteric dosage forms such as tablets, capsules, or liquids are absorbed.
  • the compound is brought directly into the liver, where, depending upon its specific chemical structure, it may be metabolized and eliminated by enzymes that perform the normal detoxifying processes in liver cells. This elimination is referred to as "first-pass" metabolism or the "first-pass" effect in the liver as previously discussed.
  • resulting metabolites are often found circulating in the blood stream and subsequently eliminated in the urine and/or feces.
  • Aspects of the present invention are based on the finding that addition of certain alkyl carbohydrates, when included in fast-dispersing dosage forms, modulate the proportion of the CGRP inhibitor that is subject to the first-pass effect, thus allowing a fixed amount of the CGRP inhibitor to exert greater clinical benefit, or allowing a smaller amount of the CGRP inhibitor to achieve similar clinical benefit compared to an otherwise larger dose.
  • the pharmaceutical compositions are prepared in oral solid molded fast-dispersing dosage form, such as described in U.S. Patent No. 9,192,580, issued November 24, 2015.
  • the phrase "fast-dispersing dosage form” refers to compositions which disintegrate or disperse within 1 to 60 seconds, for example, 1 to 50 seconds, 1 to 40 seconds, 1 to 30 seconds, 1 to 20 seconds, 1 to 10 seconds, or 2 to 8 seconds, after being placed in contact with a fluid.
  • the fluid is preferably that found in the oral cavity, i.e., saliva, as with oral administration.
  • the compositions described herein are solid fast dispersing dosage forms including a solid network of the active ingredient, for example, zavegepant, and a water- soluble or water-dispersible carrier containing fish gelatin. Accordingly, the carrier is inert towards the active ingredient.
  • the network is obtained by subliming solvent from a composition in the solid state, the composition including the active ingredient and a solution of the carrier in the solvent.
  • the dosage forms according to the invention can be prepared according to the process disclosed in Gregory et al., U.K. Patent No. 1,548,022 using fish gelatin as the carrier. Accordingly, an initial composition (or admixture) including the active ingredient and a solution of the fish gelatin carrier in a solvent is prepared followed by sublimation.
  • the sublimation is preferably carried out by freeze drying the composition.
  • the composition can be contained in a mold during the freeze- drying process to produce a solid form in any desired shape.
  • the mold can be cooled using liquid nitrogen or solid carbon dioxide in a preliminary step prior to the deposition of the composition therein.
  • the reduced pressure applied in the process can be below about 4 mm Hg, preferably below about 0.3 mm Hg.
  • the freeze dried compositions can then be removed from the mold if desired or stored therein until later use.
  • a solid fastdispersing dosage form is produced having the advantages associated with the use of fish gelatin described herein.
  • fish gelatin is categorized as being from cold water and warm water fish sources and as being of the gelling or non-gelling variety.
  • the non-gelling variety of fish gelatin in comparison to gelling fish gelatin and bovine gelatin, contains lower proline and hydroxyproline amino acid content, which are known to be associated with cross-linking properties and gelling ability.
  • Non-gelling fish gelatin can remain at solution concentrations of up to about 40% as well as in temperatures as low as 20° C.
  • the fish gelatin used in accordance with the invention is preferably obtained from cold water fish sources and is the non-gelling type of fish gelatin. More preferably, in an aspect of the invention, the non-hydrolyzed form of non-gelling fish gelatin is used. In an alternative embodiment, spray-dried non-hydrolyzed non-gelling fish gelatin may be used. Fish gelatins suitable for use in the compositions described herein are commercially available.
  • compositions can also contain, in addition to the active ingredient arid fish gelatin carrier, other matrix forming agents and secondary components.
  • Matrix forming agents suitable for use include materials derived from animal or vegetable proteins, such as other gelatins, dextrins and soy, wheat and psyllium seed proteins; gums such as acacia, guar, agar, and 10 xanthan; polycarbohydrates; alginates; carboxymethylcelluloses; carrageenans; dextrans; pectins; synthetic polymers such as polyvinylpyrrolidone; and polypeptide/protein or polycarbohydrate complexes such as gelatinacacia complexes.
  • Other materials which may also be incorporated into the fast-dissolving compositions, according to embodiments of the present invention, include sugars such as mannitol, dextrose, lactose, galactose, and trehalose; cyclic sugars such as cyclodextrin; inorganic salts such as sodium phosphate, sodium chloride and aluminum silicates; and amino acids having from 2 to 12 carbon atoms such as glycine, L-alanine, L-aspartic acid, L-glutamic acid, L- hydroxyproline, L-isoleucine, L-leucine and L-phenylalanine.
  • sugars such as mannitol, dextrose, lactose, galactose, and trehalose
  • cyclic sugars such as cyclodextrin
  • inorganic salts such as sodium phosphate, sodium chloride and aluminum silicates
  • amino acids having from 2 to 12 carbon atoms such as gly
  • One or more matrix forming agents may be incorporated into the solution or suspension prior to solidification (freezing).
  • the matrix forming agent may be present in addition to a surfactant or to the exclusion of a surfactant.
  • the matrix forming agent may aid in maintaining the dispersion of any active ingredient within the solution of suspension. This is especially helpful in the case of active agents that are not sufficiently soluble in water and must, therefore, be suspended rather than dissolved.
  • Secondary components such as preservatives, antioxidants, surfactants, viscosity enhancers, coloring agents, flavoring agents, pH modifiers, sweeteners or taste-masking agents may also be incorporated into the fast-dissolving compositions.
  • Suitable coloring agents include red, black and yellow iron oxides and FD & C dyes such as FD&C Blue No. 2 and FD&C Red No. 40 available from Ellis & Everard.
  • Suitable flavoring agents include mint, raspberry, licorice, orange, lemon, grapefruit, caramel, vanilla, cherry and grape flavors and combinations of these.
  • Suitable pH modifiers include the edible acids and bases, such as citric acid, tartaric acid, phosphoric acid, hydrochloric acid, maleic acid and sodium hydroxide.
  • Suitable sweeteners include, for example, sucralose, aspartame, acesulfame K and thaumatin.
  • Suitable taste-masking agents include, for example, sodium bicarbonate, ion exchange resins, cyclodextrin inclusion compounds, adsorbates or microencapsulated actives.
  • Increasing or decreasing the amount of specific alkyl carbohydrates included in fast-dispersing dosage forms may alter or modulate the site of absorption of the CGRP inhibitor, increasing or decreasing, respectively, that a proportion of the CGRP inhibitor that is absorbed through buccal tissue compared to other portions of the alimentary canal.
  • the alkyl glycoside content can be reduced to attenuate buccal absorption so that a portion of the drug is immediately absorbed buccally for rapid onset, but the rest is absorbed through the slower gastric absorption process.
  • addition of certain alkyl glycosides having specific alkyl chain lengths to the fast-dispersing tablets may alter the pharmacokinetics of pre-gastric drug absorption in beneficial ways.
  • incorporation of from between about 0.2%-0.3%, 0.3%- 0.4%, 0.4%-0.5%, 0.5%-1.0%, 1.0%-2.0%, 2.0%-3.0%, 3.0%-4.0%, 4.0%-5.0%, 5.0%-6.0%, 6.0%-7.0%, 7.0%-8.0%, 9.0%-10.0%, and greater than 10% of alkyl glycoside may alter the pharmacokinetics of pre- gastric drug absorption in beneficial ways.
  • the alkyl glycoside is dodecyl maltoside, tetradecyl maltoside and/or sucrose dodecanoate, which when incorporated into a fastdispersing tablet format increases the drug that enters into systemic circulation and decreases the drug that is eliminated by the "first-pass" effect in the liver. Additionally, the time to maximum drug levels may be dramatically reduced, for example, from one to six hours, to approximately 15 to 45 minutes. For use in treating patients undergoing acute migraine episodes, this more rapid absorption of the CGRP inhibitor, resulting in more rapid onset of action, may be of great benefit.
  • the term "fast-dispersing dosage form” is intended to encompass all the types of dosage forms capable of dissolving, entirely or in part, within the mouth.
  • the fast-dispersing dosage form is a solid, fast-dispersing network of the active ingredient and a water-soluble or water-dispersible carrier matrix, which is inert towards the active ingredient and excipients.
  • the network may be obtained by lyophilizing or subliming solvent from a composition in the solid state, which composition includes the active ingredient, an alkyl glycoside, and a solution of the carrier in a solvent.
  • one solvent particularly well suited for use with the present invention is water.
  • Water- alcohol mixtures may also be employed where solubility of the CGRP inhibitor in the mixed solvent is enhanced.
  • dispersions of small drug particles can be suspended in an aqueous gel that maintains uniform distribution of the substantially insoluble drug during the lyophilization or subliming process.
  • the dosage form may include the CGRP antagonist in an amount from about 10-80 weight%, for example, about 20-80 weight%, about 30-80 weight%, about 40-80 weight%, or about 50-80 weight% based on the total weight of the dosage form.
  • the dosage form may further include the alkyl glycoside in an amount of about 0.01-50 weight%, for example, about 0.1-50 weight%, about 1-50 weight%, about 5-50 weight%, or about 10-50 weight% based on the total weight of the dosage form.
  • the dosage form may further include fish gelatin in an amount of about 10-30 weight%, for example, about 15-30 weight% or about 20-30 weight% based on the total weight of the dosage form.
  • the dosage form may further include about 10-25 weight% of a filler.
  • the aqueous gel may be the self-assembling hydrogels described in U.S. Patent Application No. 60/957,960, formed using selected alkyl glycosides such as sucrose mono- and distearate and/or tetradecylmaltoside. 1
  • Matrix forming agents suitable for use in fast-dissolve formulations of the present invention are describe throughout this application.
  • Such agents include materials derived from animal or vegetable proteins, such as the gelatins, collagens, dextrins and soy, wheat and psyllium seed proteins; gums such as acacia, guar, agar, and xanthan; polycarbohydrates; alginates; carrageenans; dextrans; carboxymethylcelluloses; pectins; synthetic polymers such as polyvinylpyrrolidone; and polypeptide/protein or polycarbohydrate complexes such as gelatin-acacia complexes.
  • gelatin particularly fish gelatin or porcine gelatin is used.
  • CGRP antagonists may be incorporated into a fast-dissolve dosage formulation as described herein, particularly well suited CGRP antagonists have low oral bioavailability (for example, less than 80%), such as BHV-3500.
  • the pharmaceutical composition may include a pharmaceutically acceptable carrier and a therapeutically effective amount of zavegepant, a solvate thereof, or a pharmaceutically acceptable salt thereof, wherein the pharmaceutical composition is in a form of an oral solid molded fast-dispersing dosage form.
  • a pharmaceutical composition may be formulated, for example, as orally disintegrating tablet (ODT).
  • ODT orally disintegrating tablet
  • An oral solid molded fast-dispersing dosage form of a CGRP inhibitor is described, for example, in International Application No. PCT/US2019/023940 filed on March 25, 2019 and published as WO 2019/191008 Al on October 3, 2019, which is incorporated herein in its entirety by reference.
  • the soft gel pharmaceutical formulation may include: a synthetic or natural poorly permeable calcitonin gene-related peptide (CGRP) inhibitor or salt or solvate thereof in an amount 0.01-20 weight% of the total weight of the formulation; a lipophilic phase comprising triglycerides of fatty acids in an amount of 50-80 weight% of the total weight of the formulation; and at least one lipophilic surfactant comprising partial esters of polyol and fatty acids in an amount of 10-50 weight% of the total weight of the formulation.
  • CGRP poorly permeable calcitonin gene-related peptide
  • An embodiment provides a method for treatment or prevention of a condition associated with aberrant levels of CGRP in a subject in need thereof, wherein the method comprises administering to the subject any of the above pharmaceutical compositions and formulations.
  • the CGRP inhibitor may be a CGRP antibody, a CGRP receptor antibody, an antigen-binding fragment from a CGRP antibody or a CGRP receptor antibody, a CGRP infusion inhibitory protein, a CGRP bio-neutralizing agent, a small molecule CGRP receptor antagonist, a small molecule CGRP inhibitor, or a polypeptide CGRP inhibitor.
  • the small molecule CGRP receptor antagonist is zavegepant, rimegepant, ubrogepant, atogepant, telcagepant, or olcegepant, a solvate thereof, or a pharmaceutically acceptable salt thereof.
  • the condition may be a disorder selected from acute migraine, chronic migraine, cluster headache, chronic tension type headache, medication overuse headache, post- traumatic headache, post-concussion syndrome, brain trauma, and vertigo.
  • the condition may be a disorder selected from chronic pain, neurogenic vasodilation, neurogenic inflammation, inflammatory pain, neuropathic pain, diabetic peripheral neuropathic pain, small fiber neuropathic pain, Morton's neuroma, chronic knee pain, chronic back pain, chronic hip pain, chronic finger pain, exercise-induced muscle pain, cancer pain, chronic inflammatory skin pain, pain from burns, pain from scars, complex regional pain syndrome, burning mouth syndrome, alcoholic polyneuropathy, chronic inflammatory demyelinating polyradiculoneuropathy, human immunodeficiency virus (HIV) or acquired immunodeficiency syndrome (AIDS)-associated neuropathy, drug-induced neuropathy, industrial neuropathy, lymphomatous neuropathy, myelomatous neuropathy, multi-focal motor neuropathy, chronic idiopathic sensory neuropathy, carcinomatous, neuropathy, acute pain autonomic neuropathy, compressive neuropathy, vasculitic/ischaemic neuropathy, temporomandibular joint pain, post-
  • HIV human immunodefic
  • the condition may be medication overuse headache (MOH), and the subject having the condition may be undergoing treatment for pain, wherein the treatment for pain may include a medicament selected from acute pain medications and chronic pain medications.
  • the treatment for pain includes a medicament selected from triptans, ergot alkaloids, analgesics and opioids.
  • the triptans may be selected from rizatriptan, sumatriptan, naratriptan, eletriptan, donitriptan, almotriptan, frovatriptan, avitriptan, and zolmitriptan.
  • the ergot alkaloids may be selected from clavines, lysergic acid amides and ergopeptines.
  • the ergot alkaloid may also be selected from ergonovine, methylergonovine, methysergide, ergotamine, dihydroergotamine, bromocriptine, ergoloid mesylates and lysergic acid diethylamide, or a combination thereof.
  • the MOH may result from the chronic use of one or more pain medications.
  • the subject may have a primary headache disorder selected from migraine, cluster-type headache, or tension-type headache.
  • the subject may be currently undergoing treatment or may have received treatment for the primary headache disorder.
  • the treatment for pain may include a medicament selected from aspirin, diclofenac; dif lunisal, etodolac, fenoprofen, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, mefenamic acid, meloxicam, nabumetone, naproxen, oxaprozin, piroxicam, salsalate, sulindac, tolmetin, celecoxib, rofecoxib, etoricoxib, valdecoxib, parecoxib, meloxicam, lumiracoxib, or a combination thereof.
  • a medicament selected from aspirin, diclofenac; dif lunisal, etodolac, fenoprofen, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofena
  • the MOH may result from treatment with a medicament selected from ketamine, esketamine, alfentanil, alimemazine, alprazolam, amphetamine, buprenorphine, butorphanol, clonazepam, codeine, cyclobenzaprine, diazepam, dihydrocodeine, dihydromorphine, dronabinol, estazolam, ezopiclone, fentanyl, flurazepam, hydrocodone, hydromorphone, lorazepam, methobarbital, methylphenidate, methadone, morphine, oxycodone, oxymorphone, phenobarbital, secobarbital, tempazepam, tramadol, triazolam, zaleplon, zopiclone, and zolpidem.
  • a medicament selected from ketamine, esketamine, alfentanil, alimemazine, alprazol
  • the MOH may result from the chronic use of a medicament selected from alimemazine, alprazolam, amphetamine, buprenorphine, butorphanol, clonazepam, codeine, cyclobenzaprine, diazepam, dihydrocodeine, dihydromorphine, dronabinol, estazolam, ezopiclone, fentanyl, flurazepam, hydrocodone, hydromorphone, lorazepam, methobarbital, methylphenidate, methadone, morphine, oxycodone, oxymorphone, phenobarbital, secobarbital, tempazepam, tramadol, triazolam, zaleplon, zopiclone, and zolpidem.
  • a medicament selected from alimemazine, alprazolam, amphetamine, buprenorphine, butorphanol, clonazepam, codeine
  • the MOH may result from the chronic use of a medicament selected from aspirin, ibuprofen, naproxen, acetaminophen, diclofenac, flurbiprofen, meclofenamate, isometheptene, indomethacin; codeine, morphine, hydrocodone, acetyldihydrocodeine, oxycodone, oxymorphone, papaverine, fentanyl, alfentanil, sufentanil, remifentanyl, tramadol, prochlorperazine, celecoxib, rofecoxib, meloxicam, piroxicam, JTE-522, L-745,337, NS388, deracoxib, valdecoxib, iumiracoxib, etoricoxib, parecoxib, 4-(4-cyclohexyl- 2-methyloxazol-5-yl)-2 fluorobenzene
  • the condition may be post-traumatic headache (PTH) headache
  • the subject having the condition may experience a PTH one, two, three, four, five, six or seven days after a traumatic incident.
  • the traumatic incident may result in a concussion or loss of consciousness.
  • the subject may suffers from dizziness, insomnia, poor concentration, memory problems, photophobia, phonophobia, or fatigue, or a combination thereof.
  • the condition may be a disorder selected from non-insulin dependent diabetes mellitus, vascular disorders, inflammation, arthritis, thermal injury, circulatory shock, sepsis, alcohol withdrawal syndrome, opiate withdrawal syndrome, morphine tolerance, hot flashes in men and women, flushing associated with menopause, allergic dermatitis, psoriasis, encephalitis, ischaemia, stroke, epilepsy, neuroinflammatory disorders, neurodegenerative diseases, skin diseases, neurogenic cutaneous redness, skin rosaceousness, erythema, tinnitus, obesity, inflammatory bowel disease, irritable bowel syndrome, vulvodynia, polycystic ovarian syndrome, uterine fibroids, neurofibromatosis, hepatic fibrosis, renal fibrosis, focal segmental glomerulosclerosis, glomerulonephritis, IgA nephropathy, multiple myeloma, myasthenia gravis, Sjo
  • the skin disease are selected from recurrent herpes, contact hypersensitivity, prurigo nodularis, chronic pruritus, and uremic pruritus.
  • the condition may be a disorder selected from chronic obstructive pulmonary disease, pulmonary fibrosis, bronchial hyperreactivity, asthma, cystic fibrosis, chronic idiopathic cough, and a toxic injury.
  • the toxic injury is selected from chlorine gas injury, mustard gas injury, acrolein injury, smoke injury, ozone injury, warfare chemical exposure, and industrial chemical exposure.
  • PK pharmacokinetics
  • BHV-3500 The chemical name of BHV-3500 is (R)-N-(3-(7-methyl-lH-indazol-5-yl)-l-(4-(l-methylpiperidin- 4-yl)piperazin-l-yl)-l-oxopropan-2-yl)-4-(2-oxo-l,2-dihydroquinolin-3-yl)piperidine-l-carboxamide.
  • BHV-3500 is described, for example, in WO 03/104236 published December 18, 2003 and US 8,481,546 issued July 9, 2013, which are incorporated herein in their entireties by reference.
  • BHV-3500 has poor permeability and was selected as an object of the present study.
  • BHV-3500-d8 which is an octadeuterated analog of BHV-3500 has the following formula II:
  • test article is identified as BHV-3500.
  • the test article will be supplied as tablets.
  • Test Article Characterization A certificate of analysis (or other appropriate documentation) verifying the identity or purity of test articles will be provided.
  • the BHV-3500 tablets will be stored at room temperature. Sample Disposition and Retention. All quantities of the test articles that are dispensed will be documented. Retention samples are not required for a study of this duration.
  • test articles dose levels were selected on the basis of previous PK studies with the test articles.
  • test article will be administered sublingually.
  • test Article Disposition of Test Article. Upon completion of the study, any remaining test articles will be returned and discarded.
  • Test Animals Three (3) or 3 female beagle dogs are obtained from Ridglan Farms, Mount Horeb, Wl for use in this study. All animals are immunized against distemper, type 2 adenovirus, parainfluenza, Bordetella, rabies, papilloma virus, and parvovirus by the supplier. Dogs will be approximately one year old and weigh approximately 8 to 12 kg at the initiation of dosing. The same 3 animals will be used for all test article administrations.
  • the dog is a standard species used for non-clinical toxicity studies, and is accepted by the U.S. Food and Drug Administration as a large animal (non-rodent) model system for the safety assessment of pharmacokinetics of pharmaceutical agents.
  • Dogs will be housed individually in pens equipped with automatic watering systems. Pens will be cleaned daily. Dogs will be housed in accordance with U.S. Department of Agriculture Welfare Standards (Title 9, Code of Federal Regulation, Part 3, 1991 Revision) and standards set forth in the Guide for the Care and Use of Laboratory Animals (National Research Council, 2011).
  • Each dog will be identified by a USDA tattoo number in the right or left ear. Each dog will also be assigned a unique number within the study. All pens will be identified by the Project Number, Animal Number, and Sex. Cage cards will be color-coded according to group.
  • Temperature and relative humidity in the animal room will be recorded manually each day. A 12-hour light/dark cycle (maintained with an automatic timer) will be used. Animal rooms will be held within temperature and relative humidity ranges of approximately 20°C to 25°C and 30%-70%, respectively.
  • mice After animals have been released from quarantine, animals will be randomly assigned into groups. Prior to randomization, each dog will receive a detailed clinical observation to ensure its suitability as a test animal. Administration. Animals in groups 1 to 2 will receive a single oral capsule administration of BHV-3500 at 20 mg/dog. Animals in groups 4 to 6 will receive a single oral (capsule) administration of BHV-3500 at a dose of 50 mg/dog. Each group will be followed by a washout period of at least 48 hours prior to the next group being dosed.
  • Moribundity/Mortality Observations Prior to initiation of dosing, animals will be observed at least once daily for mortality or evidence of moribundity. Upon initiation of dosing and then throughout the remainder of the observation periods, all surviving study animals will be observed at least twice daily for mortality or evidence of moribundity and to assess their general health. Any abnormal clinical signs will be recorded. Moribundity/mortality checks will be separated by a minimum of four hours.
  • Injured or Diseased Animals Animals on test will be treated for any disease or injury in conformance with standard veterinary practice. A complete record of the circumstances and the disposition of any affected animals will be made in the study notebook. Any animal that pose a potential infectious threat to other studies will be isolated.
  • Clinical observation will be done approximately 1 hour after each dose administration.
  • Plasma Drug Level Blood samples (approximately 3 mL collected from the jugular vein) for determination of plasma levels of BHV-3500 will be obtained from each dog at six time points (pre-dose; 15, 30, and 60 minutes, 2 and 4 hours) after each dose. EDTA will be used as the anticoagulant. Plasma samples will be frozen at approximately -70°C until analyzed at the testing center for concentration of BHV-3500. Pharmacokinetic modeling will include AUC, ti/2, T max , and C max .
  • Paper data to be maintained in loose-leaf notebooks will include, but not necessarily be limited to, the following:
  • ToxData Data captured electronically using ToxData’ (e.g., dose administration, daily moribundity/mortality and environmental data, clinical observations, body weights, etc.) will be maintained within the computer system's database; electronic copies of the ToxData’. htm files will also be backed-up onto CD-ROM(s) and the disc(s) will be maintained with the raw data.
  • ToxData e.g., dose administration, daily moribundity/mortality and environmental data, clinical observations, body weights, etc.
  • Alteration of Design Alterations in the Protocol may be as the study progresses in the form of a protocol amendment. No changes in the Protocol will be made without the specific written consent of the Sponsor.
  • Protocol Approval This protocol complies with the specific documents of the Sponsor.
  • Plasma samples were obtained from of each dog at six time points (pre-dose; 15, 30, and 60 minutes and 2 and 4 hours post-dose). EDTA was used as the anticoagulant. Plasma samples were frozen at approximately -70°C until analyzed.
  • the reference standards for BHV-3500 and BHV-3500-d8 were provided by the Sponsor and stored at room temperature. The standards were used without further purification for the preparation of calibration standards and quality control (QC) samples for the determination of BHV-3500 concentrations in plasma samples collected during this study.
  • QC quality control
  • BHV-3500 in plasma a 50 pL aliquot from each sample was transferred into the appropriate well of a 96-well plate to which 10 pL of 50% acetonitrile (ACN) in water was added, followed by 250 pL of internal standard solution (10 ng/mL BHV-3500-d8 in ACN). After sealing the plate and vortexing for approximately 5 minutes, the plate was centrifuged at 4000 rpm for 10 minutes at 4 ⁇ 4°C. A portion (100 pL) of the resulting supernatant was transferred to into the appropriate well (containing 300 pL 0.15% formic acid in water) of another 96-well plate. This plate was sealed and its contents mixed prior to instrumental analysis.
  • ACN acetonitrile
  • Calibrator, QC and study samples were analyzed under LC-MS/MS instrument conditions detailed in Table 2. Calibration curves were calculated from the linear regression (weighting factor of 1/x 2 ) of the analyte to internal standard peak area ratios versus the analyte concentrations.
  • Concentrations of analyte in the samples were determined using the peak area ratios and the regression parameters of the calibration curves.
  • Elimination rate constant values were calculated by log-linear regression on data points of the terminal phase (using Phoenix WinNonlin's Best Fit Lambda Z Calculation Method option) when allowed by the data; the plasma elimination half-life (t 1/2 ) was calculated as In (2)/A z . Area under the plasma concentration-time curve values from time zero to the concentration at the 4 hour time point (AUC 0.4hr ) were calculated by the linear-up/log-down trapezoidal rule.
  • Nominal dose levels were used for PK analysis. The PK parameters listed below were evaluated (as applicable and when allowed by the data).
  • BHV-3500 concentration determinations are presented in Table 4, and are shown graphically in FIG. 1.
  • PK parameters are presented in Table 5 (BHV-3500).
  • Group 1 where BHV-3500 was administered with DDM had AUC 55.1 hr*ng/mL versus 44.4 h*ng/mL where BHV-3500 was administered without DDM (a 24% increase in AUC).
  • the purpose of the study described in this example was to evaluate the technical feasibility of formulating BHV-3500 into the Zydis® dosage form.
  • the feasibility determined the maximum dose strengths to be 50mg free base, corresponding to 52.85mg of the Hydrochloride salt (salt equivalency factor: 1.057).
  • the formulation feasibility activities consisted of five manufacturing studies in an attempt to develop a robust formulation containing BHV-3500 and Dodecyl Maltoside, with acceptable critical quality attributes, such as satisfactory finished product appearance and dispersion time.
  • Study 1 consisted of 5 batches. All batches contained a fixed concentration of Gelatin, mannitol and Dodecyl Maltoside. Study 1 investigated:
  • Popping out was also identified as a problem, particularly in units with a higher concentration of BHV-3500 and larger wet fill weight (batches Z4840/94/5a, 5b - 16.67% w/w API at 300mg/600mg wet fill weight respectively). [0160] All batches had dispersion times within the acceptable criterion of ⁇ 10 seconds.
  • Study 2 The purpose of Study 2 was to optimize formulation containing 16.67% w/w BHV-3500 to improve the physical appearance and rigidity. Study 2 investigated:
  • study 3 The purpose of study 3 was to optimize the formulation containing 16.67%w/w BHV-3500 and 0.50%w/w DDM to improve the physical appearance and rigidity. Study 3 investigated:
  • study 4 The purpose of study 4 was to range the HMW fish gelatin whilst maintaining the ratio of mannitol, as well as ranging the concentration of DDM, and also to assess the effects of fill weight and solution hold time of the finished product appearance as part of an experimental design study. A larger pocket size was also to be used to lower the concentration of BHV-3500. Study 4 investigated:
  • Batch Z4840/120/1 did however have air bubbles present in the units whereas batch Z4840/120/2 did not, this could suggest the presence of DDM causes air bubbles during dosing, which could be due to an increase in viscosity when DDM is present in the mix. However, this was not considered to be a significant defect due to the nature of the small batch size and hand dosing, this will be optimized for larger scale batches in the future.
  • Table 7 lists the packaging materials used in all studies.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pain & Pain Management (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Hydrogenated Pyridines (AREA)

Abstract

L'invention concerne une composition pharmaceutique comprenant un Inhibiteur du peptide lié au gène de la calcitonine (CGRP) et une quantité augmentant l'absorption d'une substance pharmaceutiquement acceptable, telle qu'un tensioactif de carbohydrate . L'invention concerne également un procédé pour augmenter la biodisponibilité d'un inhibiteur de peptide lié au gène de la calcitonine (CGRP) chez un sujet, comprenant l'administration par voie orale pour augmenter la biodisponibilité de l'inhibiteur de CGRP chez le sujet.
PCT/US2021/059801 2020-11-19 2021-11-18 Compositions pour une meilleure administration d'inhibiteurs de cgrp WO2022109077A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
MX2023005846A MX2023005846A (es) 2020-11-19 2021-11-18 Composiciones para la administracion mejorada de los inhibidores de cgrp.
EP21895549.0A EP4247364A1 (fr) 2020-11-19 2021-11-18 Compositions pour une meilleure administration d'inhibiteurs de cgrp
JP2023529881A JP2023551146A (ja) 2020-11-19 2021-11-18 Cgrp阻害剤の送達を向上させるための組成物
AU2021385052A AU2021385052A1 (en) 2020-11-19 2021-11-18 Compositions for improved delivery of cgrp inhibitors
KR1020237016343A KR20230088442A (ko) 2020-11-19 2021-11-18 Cgrp 억제제의 개선된 전달을 위한 조성물
CA3202137A CA3202137A1 (fr) 2020-11-19 2021-11-18 Compositions pour une meilleure administration d'inhibiteurs de cgrp
CN202180077330.9A CN116456981A (zh) 2020-11-19 2021-11-18 用于改善的grp抑制剂递送的组合物
IL302948A IL302948A (en) 2020-11-19 2021-11-18 Preparations for the administration of CGRP inhibitors
ZA2023/04727A ZA202304727B (en) 2020-11-19 2023-04-24 Compositions for improved delivery of cgrp inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063115789P 2020-11-19 2020-11-19
US63/115,789 2020-11-19
US202063126550P 2020-12-17 2020-12-17
US63/126,550 2020-12-17

Publications (1)

Publication Number Publication Date
WO2022109077A1 true WO2022109077A1 (fr) 2022-05-27

Family

ID=81709748

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/059801 WO2022109077A1 (fr) 2020-11-19 2021-11-18 Compositions pour une meilleure administration d'inhibiteurs de cgrp

Country Status (9)

Country Link
EP (1) EP4247364A1 (fr)
JP (1) JP2023551146A (fr)
KR (1) KR20230088442A (fr)
AU (1) AU2021385052A1 (fr)
CA (1) CA3202137A1 (fr)
IL (1) IL302948A (fr)
MX (1) MX2023005846A (fr)
WO (1) WO2022109077A1 (fr)
ZA (1) ZA202304727B (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115154469A (zh) * 2022-07-22 2022-10-11 中国科学院广州生物医药与健康研究院 瑞美吉泮或其药学上可接受的盐在制备预防或治疗肥胖的药物中的应用

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100112050A1 (en) * 2008-11-03 2010-05-06 Je Phil Ryoo Dosage Form For Insertion Into The Mouth
US20120059017A1 (en) * 2010-03-30 2012-03-08 Bristol-Myers Squibb Company CGRP Receptor Antagonist
US20160346198A1 (en) * 2014-02-05 2016-12-01 Merck Sharp & Dohme Corp. Novel disintegration systems for pharmaceutical dosage forms
US20170298127A1 (en) * 2016-04-15 2017-10-19 Alder Biopharmaceuticals, Inc. Anti-pacap antibodies and uses thereof
WO2020150703A1 (fr) * 2019-01-20 2020-07-23 Biohaven Pharmaceutical Holding Company Ltd. Antagonistes du cgrp pour le traitement d'une migraine intense
US20200360292A1 (en) * 2019-05-16 2020-11-19 Aegis Therapeutics, Llc Oil-soluble drug containing compositions and methods of use thereof
WO2021062282A1 (fr) * 2019-09-25 2021-04-01 Allergan Pharmaceuticals International Limited Polythérapie avec des antagonistes du cgrp
WO2021127070A1 (fr) * 2019-12-17 2021-06-24 Biohaven Pharmaceutical Holding Company Ltd. Compositions pharmaceutiques intranasales d'inhibiteurs de cgrp

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100112050A1 (en) * 2008-11-03 2010-05-06 Je Phil Ryoo Dosage Form For Insertion Into The Mouth
US20120059017A1 (en) * 2010-03-30 2012-03-08 Bristol-Myers Squibb Company CGRP Receptor Antagonist
US20160346198A1 (en) * 2014-02-05 2016-12-01 Merck Sharp & Dohme Corp. Novel disintegration systems for pharmaceutical dosage forms
US20170298127A1 (en) * 2016-04-15 2017-10-19 Alder Biopharmaceuticals, Inc. Anti-pacap antibodies and uses thereof
WO2020150703A1 (fr) * 2019-01-20 2020-07-23 Biohaven Pharmaceutical Holding Company Ltd. Antagonistes du cgrp pour le traitement d'une migraine intense
US20200360292A1 (en) * 2019-05-16 2020-11-19 Aegis Therapeutics, Llc Oil-soluble drug containing compositions and methods of use thereof
WO2021062282A1 (fr) * 2019-09-25 2021-04-01 Allergan Pharmaceuticals International Limited Polythérapie avec des antagonistes du cgrp
WO2021127070A1 (fr) * 2019-12-17 2021-06-24 Biohaven Pharmaceutical Holding Company Ltd. Compositions pharmaceutiques intranasales d'inhibiteurs de cgrp

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115154469A (zh) * 2022-07-22 2022-10-11 中国科学院广州生物医药与健康研究院 瑞美吉泮或其药学上可接受的盐在制备预防或治疗肥胖的药物中的应用

Also Published As

Publication number Publication date
CA3202137A1 (fr) 2022-05-27
JP2023551146A (ja) 2023-12-07
MX2023005846A (es) 2023-06-02
AU2021385052A1 (en) 2023-06-01
IL302948A (en) 2023-07-01
AU2021385052A9 (en) 2024-05-02
ZA202304727B (en) 2023-12-20
KR20230088442A (ko) 2023-06-19
EP4247364A1 (fr) 2023-09-27

Similar Documents

Publication Publication Date Title
US20210121573A1 (en) Absorption enhancers for drug administration
CA2234140C (fr) Formulation pharmaceutique lyophilisee stable
DK2381773T3 (en) COMPOSITIONS FOR THE ADMINISTRATION OF PHARMACEUTICALS
US9283280B2 (en) Compositions for drug administration
US20090047347A1 (en) Compositions for Drug Administration
CA2814927A1 (fr) Compositions pour l'administration de medicaments
US20120309740A1 (en) Pharmaceutical Compositions Having Improved Dissolution Profiles For Poorly Soluble Drugs
AU2021385052A1 (en) Compositions for improved delivery of cgrp inhibitors
US20240066028A1 (en) Compositions for improved delivery of cgrp inhibitors
US20110046058A1 (en) Compositions for enteral absorption and sustained action of leptin-related peptides useful in the treatment of obesity and leptin-modulated disease

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: 21895549

Country of ref document: EP

Kind code of ref document: A1

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023006623

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20237016343

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3202137

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2023529881

Country of ref document: JP

Ref document number: 202180077330.9

Country of ref document: CN

ENP Entry into the national phase

Ref document number: 112023006623

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20230410

ENP Entry into the national phase

Ref document number: 2021385052

Country of ref document: AU

Date of ref document: 20211118

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021895549

Country of ref document: EP

Effective date: 20230619

WWE Wipo information: entry into national phase

Ref document number: 523440827

Country of ref document: SA