EP3687510A1 - Enhanced delivery epinephrine and prodrug compositions - Google Patents

Enhanced delivery epinephrine and prodrug compositions

Info

Publication number
EP3687510A1
EP3687510A1 EP18786593.6A EP18786593A EP3687510A1 EP 3687510 A1 EP3687510 A1 EP 3687510A1 EP 18786593 A EP18786593 A EP 18786593A EP 3687510 A1 EP3687510 A1 EP 3687510A1
Authority
EP
European Patent Office
Prior art keywords
pharmaceutical composition
composition according
polymeric matrix
epinephrine
film
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.)
Pending
Application number
EP18786593.6A
Other languages
German (de)
English (en)
French (fr)
Inventor
Alexander Mark Schobel
Stephanie Marie VARJAN
Stephen Paul WARGACKI
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.)
Aquestive Therapeutics Inc
Original Assignee
Aquestive Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US15/717,859 external-priority patent/US20180125977A1/en
Priority claimed from US15/791,249 external-priority patent/US20180104195A1/en
Application filed by Aquestive Therapeutics Inc filed Critical Aquestive Therapeutics Inc
Publication of EP3687510A1 publication Critical patent/EP3687510A1/en
Pending 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/222Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin with compounds having aromatic groups, e.g. dipivefrine, ibopamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/235Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/46Ingredients of undetermined constitution or reaction products thereof, e.g. skin, bone, milk, cotton fibre, eggshell, oxgall or plant extracts
    • 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
    • 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
    • A61K31/49Cinchonan derivatives, e.g. quinine

Definitions

  • This invention relates to pharmaceutical compositions.
  • Active ingredients such as drugs or pharmaceuticals
  • transmucosally can require that the drug or pharmaceutical permeate or otherwise cross a biological membrane in an effective and efficient manner.
  • a pharmaceutical composition can include a polymeric matrix, epinephrine in the polymeric matrix, and an adrenergic receptor interacter.
  • Epinephrine can be provided in the form of a prodrug, such as a lipophilic prodrug, for example, dipifevrin.
  • the pharmaceutical composition can further include a permeation enhancer.
  • an adrenergic receptor interacter can be an adrenergic receptor blocker.
  • the adrenergic receptor interacter can also be a flavonoid, or used in combination with a flavonoid.
  • the adrenergic receptor interacter can be a terpenoid, terpene or a C3-C22 alcohol or acid.
  • the adrenergic receptor interacter can be a sesquiterpene.
  • the adrenergic receptor interacter can include farnesol, linoleic acid, arachidonic acid, docosahexanoic acid, eicosapentanoic acid, or docosapentanoic acid, or combinations thereof.
  • a pharmaceutical composition can include a polymeric matrix, a pharmaceutically active component in the polymeric matrix, and an aporphine alkaloid interacter.
  • a pharmaceutical composition can include a polymeric matrix, a pharmaceutically active component in the polymeric matrix, and a vasodilator interacter.
  • the pharmaceutical composition can be a film further comprising a polymeric matrix, the pharmaceutically active component being contained in the polymeric matrix.
  • the adrenergic receptor interacter can be a phytoextract.
  • the permeation enhancer can be a phytoextract.
  • the permeation enhancer can include a phenylpropanoid.
  • the pharmaceutical composition can include a fungal extract.
  • the pharmaceutical composition can include saturated or unsaturated alcohol.
  • the alcohol can be benzyl alcohol.
  • the flavonoid, phytoextract, phenylpropanoid, eugenol, or fungal extract can be used as a solubilizer.
  • the phenylpropanoid can be eugenol. In other embodiments, the phenylpropanoid can be eugenol acetate. In certain embodiments, the phenylpropanoid can be a cinnamic acid. In other embodiments, the phenylpropanoid can be a cinnamic acid ester. In other embodiments, phenylpropanoid can be a cinnamic aldehyde.
  • the phenylpropanoid can be a hydrocinnamic acid. In certain embodiments, the phenylpropanoid can be chavicol. In other embodiments, the phenylpropanoid can be safrole.
  • the phytoextract can be an essential oil extract of a clove plant.
  • the phytoextract can be an essential oil extract of a leaf of a clove plant.
  • the phytoextract can be an essential oil extract of a flower bud of a clove plant. In other embodiments, the phytoextract can be an essential oil extract of a stem of a clove plant.
  • the phytoextract can be synthetic. In certain embodiments, the phytoextract can include 20-95% eugenol, including 40-95%) eugenol, and including 60-95%) eugenol. In certain embodiments, the phytoextract can include 80-95%) eugenol.
  • the polymer matrix can include a polymer. The polymer can include a water soluble polymer.
  • the polymer can be a polyethylene oxide.
  • the polymer can be a cellulosic polymer.
  • the cellulosic polymer can be hydroxypropylmethyl cellulose, hydroxyethyl cellulose, hydroxyethylmethyl cellulose, hydroxypropyl cellulose, methylcellulose,
  • carboxymethyl cellulose and/or sodium carboxymethylcellulose.
  • the polymer can include hydroxypropyl methylcellulose.
  • the polymer can include polyethylene oxide and hydroxypropyl methylcellulose.
  • the polymer can include polyethylene oxide and/or polyvinyl pyrrolidone.
  • the polymeric matrix can include polyethylene oxide and/or a polysaccharide.
  • the polymeric matrix can include polyethylene oxide, hydroxypropyl methylcellulose and/or a polysaccharide.
  • the polymeric matrix can include polyethylene oxide, a cellulosic polymer, polysaccharide and/or polyvinylpyrrolidone.
  • the polymeric matrix can include at least one polymer selected from the group of: pullulan, polyvinyl pyrrolidone, polyvinyl alcohol, sodium alginate, polyethylene glycol, xanthan gum, tragancanth gum, guar gum, acacia gum, arabic gum, polyacrylic acid, methylmethacrylate copolymer, carboxyvinyl copolymers, starch, gelatin, ethylene oxide, propylene oxide co-polymers, collagen, albumin, poly-amino acids,
  • polyphosphazenes polysaccharides, chitin, chitosan, and derivatives thereof.
  • the pharmaceutical composition can further include a stabilizer.
  • Stabilizers can include antioxidants, which can prevent unwanted oxidation of materials, sequestrants, which can form chelate complexes and inactivating traces of metal ions that would otherwise act as catalysts, emulsifiers and surfactants, which can stabilize emulsions, ultraviolet stabilizers, which can protect materials from harmful effects of ultraviolet radiation, UV absorbers, chemicals absorbing ultraviolet radiation and preventing it from penetrating the composition, quenchers, which can dissipate the radiation energy as heat instead of letting it break chemical bonds, or scavengers which can eliminate free radicals formed by ultraviolet radiation.
  • the pharmaceutical composition has a suitable nontoxic, nonionic alkyl glycoside having a hydrophobic alkyl group joined by a linkage to a hydrophilic saccharide in combination with a mucosal delivery-enhancing agent selected from: (a) an aggregation inhibitory agent; (b) a charge-modifying agent; (c) a pH control agent; (d) a degradative enzyme inhibitory agent; (e) a mucolytic or mucus clearing agent; (f) a ciliostatic agent; (g) a membrane penetration-enhancing agent selected from: (i) a surfactant; (ii) a bile salt; (ii) a phospholipid additive, mixed micelle, liposome, or carrier; (iii) an alcohol; (iv) an enamine; (v) a nitric oxide donor compound; (vi) a long chain amphipathic molecule; (vii) a small hydrophobic penetration enhancer; (viii) sodium or a sal
  • a method of making a pharmaceutical composition can include combining an adrenergic receptor interacter with a pharmaceutically active component including epinephrine or its prodrug, and forming a pharmaceutical composition including the adrenergic receptor interacter and the pharmaceutically active component.
  • a pharmaceutical composition can be dispensed from a device.
  • a device can include a housing that holds an amount of a pharmaceutical composition, including a polymeric matrix; a pharmaceutically active component including epinephrine in the polymeric matrix; and an adrenergic receptor interacter; and an opening that dispenses a predetermined amount, such as a predetermined dose, of the pharmaceutical composition.
  • the device can also dispense a pharmaceutical composition including a permeation enhancer including a phenylpropanoid and/or a phytoextract.
  • a pharmaceutical composition can include a polymeric matrix; a pharmaceutically active component in the polymeric matrix; and an interacter that creates increased blood flow or enables a flushing of the tissue to modify transmucosal uptake of the pharmaceutically active component.
  • a pharmaceutical composition can include a polymeric matrix; a pharmaceutically active component in the polymeric matrix; and an interacter that has a positive or negative heat of solution which are used as aids to modify (increase or decrease) transmucosal uptake.
  • a pharmaceutical composition in other embodiments, includes a polymeric matrix, a pharmaceutically active component in the polymeric matrix, and an interacter, the composition contained in a multilayer film having at least one side where the edges are coterminous.
  • a method of treating a medical condition can include administering an effective amount of a pharmaceutical composition including a polymeric matrix, pharmaceutically active component including epinephrine in the polymeric matrix, and an adrenergic receptor interacter.
  • the epinephrine can be administered as a prodrug, such as dipifevrin.
  • a method of treating a medical condition can include administering an effective amount of a pharmaceutical composition including a polymeric matrix, pharmaceutically active component including dipifevrin in the polymeric matrix, and an adrenergic receptor interacter.
  • the medical condition can include hypotension, cardiac arrest, heart failure, anaphylaxis, mydriasis, asystole, pulseless electrical activity, ventricular fibrillation, pulseless ventricular tachycardia, bradycardia, arrhythmia, or asthma exacerbation.
  • a pharmaceutical film can include a polymeric matrix, a pharmaceutically active component including epinephrine or its prodrug contained in the polymeric matrix and an adrenergic receptor interacter.
  • the pharmaceutical film can have a Tmax of 5-60 minutes, and a Cmax of 0.1 ng/ml-2 ng/ml. In certain embodiments, the Tmax is 40 minutes or less and wherein the Cmax is 0.1 ng/ml or greater. In certain embodiments, the Tmax is 35 minutes or less and wherein the Cmax is 0.15 ng/ml or greater. In certain embodiments, Tmax is 30 minutes or less and wherein the Cmax is 0.2 ng/ml or greater.
  • the Cmax can be 0.1 ng/ml- 2 ng/ml, 0.15 ng/ml-25 ng/ml, 0.2 ng/ml- 1.0 ng/ml, 0.2 ng/ml -1.2 ng/ml, and 0.2 ng/ml - 1.3 ng/ml.
  • the Cmax can be greater than 0.1 ng/ml, greater than 0.15 ng/ml, greater than 0.2 ng/ml , greater than 0.4ng/ml, greater than 0.5 ng/ml, greater than l .Ong/ml, greater than 1.2 ng/ml.
  • the Cmax can be less than 3 ng/ml, less than 2 ng/ml and less than 1.5 ng/ml.
  • the Tmax can be 0-240 minutes, 10-60 minutes, 20-40 minutes, 12-15 minutes, and 5-10 minutes.
  • the Tmax can be less than 25 minutes, less than 20 minutes, 15 minutes, less than 12 minutes, and less than 10 minutes.
  • a Franz diffusion cell 100 includes a donor compound 101, a donor chamber 102, a membrane 103, sampling port 104, receptor chamber 105, stir bar 106, and a heater/circulator 107.
  • a pharmaceutical composition is a film 100 comprising a polymeric matrix 200, the pharmaceutically active component 300 being contained in the polymeric matrix.
  • the film can include a permeation enhancer 400.
  • the graphs show the permeation of an active material from a composition.
  • this graph shows average amount of active material permeated vs. time, with 8.00 mg/mL epinephrine bitartrate and 4.4 mg/mL epinephrine base solubilized.
  • this graph shows average flux vs. time, with 8.00 mg/mL epinephrine bitartrate and 4.4 mg/mL epinephrine base solubilized.
  • this graph shows ex-vivo permeation of epinephrine bitartrate as a function of concentration.
  • this graph shows permeation of epinephrine bitartrate as a function of solution pH.
  • this graph shows the influence of enhancers on permeation of epinephrine, indicated as amount permeated as a function of time.
  • these graphs show the release of epinephrine on polymer platforms (6A) and the effect of enhancers on its release (6B), indicated as amount permeated (in ⁇ g) vs. time.
  • this graph shows a pharmacokinetic model in the male Yucatan, miniature swine.
  • the study compares a 0.3 mg Epipen, a 0.12 mg Epinephrine IV and a placebo film.
  • this graph shows the impact of no enhancer on the concentration profiles of a 40 mg epinephrine film vs 0.3 mg Epipen.
  • this graph shows the impact of Enhancer A (Labrasol) on the concentration profiles of a 40 mg epinephrine film vs 0.3 mg Epipen.
  • this graph shows the impact of Enhancer L (clove oil) on the concentration profiles of two 40 mg Epinephrine films (10-1-1) and (11-1-1) vs. a 0.3 mg Epipen.
  • this graph shows the impact of Enhancer L (clove oil) and film dimension (10-1-1 thinner, bigger film and 11-1-1 thicker, smaller film) on the concentration profiles of 40 mg Epinephrine films vs. a 0.3 mg Epipen.
  • this graph shows the concentration profiles for varying doses of Epinephrine films in a constant matrix for Enhancer L (clove oil) vs. a 0.3 mg Epipen.
  • this graph shows the concentration profiles for varying doses of epinephrine films in a constant matrix for Enhancer L (clove oil) vs. a 0.3 mg Epipen.
  • this graph shows the concentration profiles for varying doses of epinephrine films in a constant matrix for Enhancer A (Labrasol) vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol and Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40mg Epinephrine Films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol on plasma concentration profiles of 40 mg epinephrine films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40 mg epinephrine films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol and Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40 mg Epinephrine Films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Enhancer A (Labrasol) in combination with Enhancer L (clove oil) on the concentration profiles of a 40 mg Epinephrine film (also shown in Fig. 20), in logarithmic view.
  • this graph shows the impact of Enhancer A (Labrasol) in combination with Enhancer L (clove oil) on the concentration profiles of a 40 mg Epinephrine film vs. the average data collected from 0.3 mg Epipens.
  • this graph shows the impact of Enhancer A (Labrasol) in combination with Enhancer L (clove oil) on the concentration profiles of a 40 mg Epinephrine films, shown as separate animal subjects.
  • the data shows average epinephrine plasma concentration vs. time.
  • the data shows average dipifevrin plasma concentration vs. time.
  • the data shows average plasma concentration of epinephrine administered as dipivefrin.
  • the graph shows the conversion of dipivefrin to epinephrine.
  • the data shows average average epinephrine profiles across all matrix studies.
  • the data shows additional data comparing plasma concentration of dipifevrin and epinephrine.
  • this graph shows an epinephrine concentration vs. time profile of a film treated with phentolamine.
  • Mucosal surfaces such as the oral mucosa
  • the buccal and sublingual tissues offer advantageous sites for drug delivery because they are highly permeable regions of the oral mucosa, allowing drugs diffusing from the oral mucosa to have direct access to systemic circulation. This also offers increased convenience and therefore increased compliance in patients.
  • a permeation enhancer can help to overcome the mucosal barrier and improve permeability.
  • Permeation enhancers reversibly modulate the penetrability of the barrier layer in favor of drug absorption. Permeation enhancers facilitate transport of molecules through the epithelium. Absorption profiles and their rates can be controlled and modulated by a variety of parameters, such as but not limited to film size, drug loading, enhancer type/loading, polymer matrix release rate and mucosal residence time.
  • a pharmaceutical composition can be designed to deliver a pharmaceutically active component in a deliberate and tailored way.
  • solubility and permeability of the pharmaceutically active component in vivo, in particular, in the mouth of a subject can vary tremendously.
  • a particular class of permeation enhancer can improve the uptake and
  • the permeation enhancer when delivered to the mouth via a film, can improve the permeability of the pharmaceutically active component
  • the permeation enhancer can improve absorption rate and amount of the pharmaceutically active component by more than 5%, more than 10%, more than 20%, more than 30%, more than 40%, more than 50%, more than 60%, more than 70%, more than 80%, more than 90%, more than 100%, more than 150%, about 200% or more, or less than 200%, less than 150%, less than 100%, less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, or less than 5%, or a combination of these ranges, depending on the other components in the composition.
  • a pharmaceutical composition has a suitable nontoxic, nonionic alkyl glycoside having a hydrophobic alkyl group joined by a linkage to a hydrophilic saccharide in combination with a mucosal delivery-enhancing agent selected from: (a) an aggregation inhibitory agent; (b) a charge-modifying agent; (c) a pH control agent; (d) a degradative enzyme inhibitory agent; (e) a mucolytic or mucus clearing agent; (f) a ciliostatic agent; (g) a membrane penetration-enhancing agent selected from: (i) 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
  • the oral mucosa might be an attractive site for the delivery of therapeutic agents into the systemic circulation. Due to the direct drainage of blood from the buccal epithelium into the internal jugular vein first-pass metabolism in the liver and intestine may be avoided. First-pass effect can be a major reason for the poor bioavailability of some compounds when administered orally. Additionally, the mucosa lining the oral cavity is easily accessible, which ensures that a dosage form can be applied to the required site and can be removed easily in the case of an emergency. However, like the skin, the buccal mucosa acts as a barrier to the absorption of xenobiotics, which can hinder the permeation of compounds across this tissue. Consequently, the identification of safe and effective penetration enhancers has become a major goal in the quest to improve oral mucosal drug delivery.
  • Chemical penetration enhancers are substances that control the permeation rate of a coadministered drug through a biological membrane. While extensive research has focused on obtaining an improved understanding of how penetration enhancers might alter intestinal and transdermal permeability, far less is known about the mechanisms involved in buccal and sublingual penetration enhancement.
  • the buccal mucosa delineates the inside lining of the cheek as well as the area between the gums and upper and lower lips and it has an average surface area of 100 cm 2 .
  • the surface of the buccal mucosa consists of a stratified squamous epithelium which is separated from the underlying connective tissue (lamina limba and submucosa) by an undulating basement membrane (a continuous layer of extracellular material approximately 1-2 ⁇ in thickness).
  • This stratified squamous epithelium consists of differentiating layers of cells which change in size, shape, and content as they travel from the basal region to the superficial region, where the cells are shed. There are approximately 40-50 cell layers, resulting in a buccal mucosa which is 500- 600 ⁇ thick.
  • the sublingual mucosa is comparable to the buccal mucosa but the thickness of this epithelium is 100-200 ⁇ .
  • This membrane is also non-keratinised and being relatively thinner has been demonstrated to be more permeable than buccal mucosa.
  • Blood flow to the sublingual mucosal is slower compared with the buccal mucosa and is of the order of 1.0 ml/ min-l/cm-2.
  • the permeability of the buccal mucosa is greater than that of the skin, but less than that of the intestine.
  • the differences in permeability are the result of structural differences between each of the tissues.
  • the absence of organized lipid lamellae in the intercellular spaces of the buccal mucosa results in greater permeability of exogenous compounds, compared to keratinized epithelia of the skin; while the increased thickness and lack of tight junctions results in the buccal mucosa being less permeable than intestinal tissue.
  • the primary barrier properties of the buccal mucosa have been attributed to the upper one-third to one-quarter of the buccal epithelium.
  • researchers have learned that beyond the surface epithelium, the permeability barrier of nonkeratinized oral mucosa could also be attributed to contents extruded from the membrane-coating granules into the epithelial intercellular spaces.
  • the intercellular lipids of the nonkeratinized regions of the oral cavity are of a more polar nature than the lipids of the epidermis, palate, and gingiva, and this difference in the chemical nature of the lipids may contribute to the differences in permeability observed between these tissues. Consequently, it appears that it is not only the greater degree of intercellular lipid packing in the stratum corneum of keratinized epithelia that creates a more effective barrier, but also the chemical nature of the lipids present within that barrier.
  • hydrophilic and lipophilic regions in the oral mucosa has led researchers to postulate the existence of two routes of drug transport through the buccal mucosa paracellular (between the cells) and transcellular (across the cells).
  • a chemical penetration enhancer, or absorption promoter is a substance added to a pharmaceutical formulation in order to increase the membrane permeation or absorption rate of the coadministered drug, without damaging the membrane and/or causing toxicity.
  • chemical penetration enhancers have been many studies investigating the effect of chemical penetration enhancers on the delivery of compounds across the skin, nasal mucosa, and intestine. In recent years, more attention has been given to the effect of these agents on the permeability of the buccal mucosa. Since permeability across the buccal mucosa is considered to be a passive diffusion process the steady state flux (Jss) should increase with increasing donor chamber concentration (CD) according to Fick's first law of diffusion.
  • Fatty acids have been shown to enhance the permeation of a number of drugs through the skin, and this has been shown by differential scanning calorimetry and Fourier transform infrared spectroscopy to be related to an increase in the fluidity of intercellular lipids.
  • OTDD transmucosal drug delivery
  • buccal penetration can be improved by using various classes of transmucosal and transdermal penetration enhancers such as bile salts, surfactants, fatty acids and their derivatives, chelators, cyclodextrins and chitosan.
  • transmucosal and transdermal penetration enhancers such as bile salts, surfactants, fatty acids and their derivatives, chelators, cyclodextrins and chitosan.
  • bile salts are the most common.
  • the permeation enhancer can be a phytoextract.
  • a phytoextract can be an essential oil or composition including essential oils extracted by distillation of the plant material.
  • the phytoextract can include synthetic analogues of the compounds extracted from the plant material (i.e., compounds made by organic synthesis).
  • the phytoextract can include a phenylpropanoid, for example, phenyl alanine, eugenol, eugenol acetate, a cinnamic acid, a cinnamic acid ester, a cinnamic aldehyde, a hydrocinnamic acid, chavicol, or safrole, or a combination thereof.
  • the phytoextract can be an essential oil extract of a clove plant, for example, from the leaf, stem or flower bud of a clove plant.
  • the clove plant can be Syzygium aromaticum.
  • the phytoextract can include 20-95% eugenol, including 40-95% eugenol, including 60-95%> eugenol, and for example, 80-95%> eugenol.
  • the extract can also include 5% to 15%) eugenol acetate.
  • the extract can also include caryophyllene.
  • the extract can also include up to 2.1%) a-humulen.
  • Other volatile compounds included in lower concentrations in clove essential oil can be ⁇ -pinene, limonene, farnesol, benzaldehyde, 2-heptanone and ethyl hexanoate.
  • Other permeation enhancers may be added to the composition to improve absorption of the drug.
  • Suitable permeation enhancers include natural or synthetic bile salts such as sodium fusidate; glycocholate or deoxycholate and their salts; fatty acids and derivatives such as sodium laurate, oleic acid, oleyl alcohol, monoolein, and palmitoyl carnitine; chelators such as disodium EDTA, sodium citrate and sodium laurylsulfate, azone, sodium cholate, sodium 5- methoxysalicylate, sorbitan laurate, glyceryl monolaurate, octoxynonyl-9, laureth-9,
  • natural or synthetic bile salts such as sodium fusidate; glycocholate or deoxycholate and their salts
  • fatty acids and derivatives such as sodium laurate, oleic acid, oleyl alcohol, monoolein, and palmitoyl carnitine
  • chelators such as disodium EDTA, sodium citrate and sodium laurylsulf
  • the permeation enhancer can include phytoextract derivatives and/or monolignols.
  • the permeation enhancer can also be a fungal extract.
  • vasodilatory effect Some natural products of plant origin have been known to have a vasodilatory effect. There are several mechanisms or modes by which plant-based products can evoke vasodilation. For review, see McNeill J.R. and Jurgens, T.M., Can. J. Physiol. Pharmacol. 84:803-821 (2006), which is incorporated by reference herein. Specifically, vasorelaxant effects of eugenol have been reported in a number of animal studies. See, e.g., Lahlou, S., et al., J. Cardiovasc.
  • Fatty acids can be used as inactive ingredients in drug preparations or drug vehicles. Fatty acids can also be used as formulation ingredients due to their certain functional effects and their biocompatible nature. Fatty acid, both free and as part of complex lipids, are major metabolic fuel (storage and transport energy), essential components of all membranes and gene regulators. For review, see Rustan A.C. and Drevon, C.A., Fatty Acids: Structures and
  • PUFAs polyunsaturated fatty acids
  • Linoleic acid which is a co-6 fatty acid, is metabolized to ⁇ -linolenic acid, dihomo- ⁇ - linolinic acid, arachidonic acid, adrenic acid, tetracosatetraenoic acid, tetracosapentaenoic acid and docosapentaenoic acid, a-linolenic acid, which is a co-3 fatty acid is metabolized to octadecatetraenoic acid, eicosatetraenoic acid, eicosapentaenoic acid (EPA), docosapentaenoic acid, tetracosapentaenoic acid, tetracosahexaenoic acid and docosahexaenoic acid (DHA).
  • EPA eicosatetraenoic acid
  • DHA docosapentaenoic acid
  • DHA docosap
  • fatty acids such as palmitic acid, oleic acid, linoleic acid and eicosapentaenoic acid
  • fatty acids such as palmitic acid, oleic acid, linoleic acid and eicosapentaenoic acid
  • fatty acids such as palmitic acid, oleic acid, linoleic acid and eicosapentaenoic acid
  • the pulmonary vascular response to arachidonic acid can be either vasoconstrictive or vasodilative, depending on the dose, animal species, the mode of arachidonic acid administration, and the tones of the pulmonary circulation.
  • arachidonic acid has been reported to cause cyclooxygenase-dependent and -independent pulmonary vasodilation. See, Feddersen, CO. et al., J. Appl. Physiol. 68(5): 1799-808 (1990); and see, Spannhake, E.W., et al., J .Appl. Physiol.
  • the adrenergic receptors are a class of G protein-coupled receptors that are a target of catecholamines, especially norepinephrine (noradrenaline) and epinephrine (adrenaline).
  • Epinephrine adrenaline
  • a receptors are less sensitive to epinephrine, when activated, they override the vasodilation mediated by ⁇ -adrenoceptors because there are more peripheral al receptors than ⁇ -adrenoceptors.
  • the al-adrenoreceptor is known for smooth muscle contraction, mydriasis, vasoconstriction in the skin, mucosa and abdominal vicera and sphincter contraction of the gastrointestinal (GI) tract and urinary bladder.
  • the al -adrenergic receptors are member of the G q protein-coupled receptor superfamily. Upon activation, a heterotrimeric G protein, G q , activates phospholipase C (PLC).
  • PLC phospholipase C
  • al -adrenergic receptors can be a main receptor for fatty acids.
  • saw palmetto extract (SPE) widely used for the treatment of benign prostatic hyperplasia (BPH)
  • BPH benign prostatic hyperplasia
  • SPE saw palmetto extract
  • BPH benign prostatic hyperplasia
  • SPE includes a variety of fatty acids including lauric acid, oleic acid, myristic acid, palmitic acid and linoleic acid. Lauric acid and oleic acid can bind noncompetitively to al- adrenergic, muscarinic and 1,4-DHP calcium channel antagonist receptors.
  • a permeation enhancer can be an adrenergic receptor interacter.
  • An adrenergic receptor interacter refers to a compound or substance that modifies and/or otherwise alters the action of an adrenergic receptor.
  • an adrenergic receptor interacter can prevent stimulation of the receptor by increasing, or decreasing their ability to bind.
  • Such interacters can be provided in either short-acting or long-acting forms. Certain short- acting interacters can work quickly, but their effects last only a few hours. Certain long-acting interacters can take longer to work, but their effects can last longer.
  • the interacter can be selected and/or designed based on, e.g., on one or more of the desired delivery and dose, active pharmaceutical ingredient, permeation modifier, permeation enhancer, matrix, and the condition being treated.
  • An adrenergic receptor interacter can be an adrenergic receptor blocker.
  • the adrenergic receptor interacter can be a terpene (e.g. volatile unsaturated hydrocarbons found in the essential oils of plants, derived from units of isoprenes) or a C3-C22 alcohol or acid, preferably a C7-C18 alcohol or acid.
  • the adrenergic receptor interacter can include farnesol, linoleic acid, arachidonic acid, docosahexanoic acid, eicosapentanoic acid, and/or docosapentanoic acid.
  • the acid can be a carboxylic acid, phosphoric acid, sulfuric acid, hydroxamic acid, or derivatives thereof.
  • the derivative can be an ester or amide.
  • the adrenergic receptor interacter can be a fatty acid or fatty alcohol.
  • the C3-C22 alcohol or acid can be an alcohol or acid having a straight C3-C22 hydrocarbon chain, for example a C3-C22 hydrocarbon chain optionally containing at least one double bond, at least one triple bond, or at least one double bond and one triple bond; said hydrocarbon chain being optionally substituted with Ci-4 alkyl, C 2- 4 alkenyl, C 2-4 alkynyl, Ci -4 alkoxy, hydroxyl, halo, amino, nitro, cyano, C3-5 cycloalkyl, 3-5 membered heterocycloalkyl, monocyclic aryl, 5-6 membered heteroaryl, Ci -4 alkylcarbonyloxy, Ci -4 alkyloxycarbonyl, Ci -4 alkylcarbonyl, or formyl; and further being optionally interrupted by -0-, -N(R a )-, -N(R a )-C(0)- 0-, -0-C(0)-N(R a )-, -N
  • Fatty acids with a higher degree of unsaturation are effective candidates to enhance the permeation of drugs. Unsaturated fatty acids showed higher enhancement than saturated fatty acids, and the enhancement increased with the number of double bonds. See, A. Mittal, et al. Status of Fatty Acids as Skin Penetration Enhancers - A Review, Current Drug Delivery, 2009, 6, pp. 274-279, which is incorporated by reference herein. Position of double bond also affects the enhancing activity of fatty acids. Differences in the physicochemical properties of fatty acid which originate from differences in the double bond position most likely determine the efficacy of these compounds as skin penetration enhancers. Skin distribution increases as the position of the double bond is shifted towards the hydrophilic end.
  • an adrenergic receptor interacter can be a terpene. Hypotensive activity of terpenes in essential oils has been reported. See, Menezes LA. et al., Z. Naturforsch. 65c:652-66 (2010), which is incorporated by reference herein.
  • the permeation enhancer can be a sesquiterpene. Sesquiterpenes are a class of terpenes that consist of three isoprene units and have the empirical formula C15H24. Like monoterpenes, sesquiterpenes may be acyclic or contain rings, including many unique combinations. Biochemical modifications such as oxidation or rearrangement produce the related sesquiterpenoids.
  • An adrenergic receptor interacter can be an unsaturated fatty acid such as linoleic acid.
  • the permeation enhancer can be farnesol.
  • Farnesol is a 15-carbon organic compound which is an acyclic sesquiterpene alcohol, which is a natural
  • farnesyl pyrophosphate Under standard conditions, it is a colorless liquid. It is hydrophobic, and thus insoluble in water, but miscible with oils.
  • Farnesol can be extracted from oils of plants such as citronella, neroli, cyclamen, and tuberose. It is an intermediate step in the biological synthesis of cholesterol from mevalonic acid in vertebrates. It has a delicate floral or weak citrus-lime odor and is used in perfumes and flavors. It has been reported that farnesol selectively kills acute myeloid leukemia blasts and leukemic cell lines in preference to primary hemopoietic cells. See, Rioja A.
  • an interacter can be an aporphine alkaloid.
  • an interacter can be a dicentrine.
  • an interacter can also be a vasodilator or a therapeutic vasodilator.
  • Vasodilators are drugs that open or widen blood vessels. They are typically used to treat hypertension, heart failure and angina, but can be used to treat other conditions as well, including glaucoma for example. Some vasodilators that act primarily on resistance vessels (arterial dilators) are used for hypertension, and heart failure, and angina; however, reflex cardiac stimulation makes some arterial dilators unsuitable for angina. Venous dilators are very effective for angina, and sometimes used for heart failure, but are not used as primary therapy for hypertension. Vasodilator drugs can be mixed (or balanced) vasodilators in that they dilate both arteries and veins and therefore can have wide application in hypertension, heart failure and angina.
  • vasodilators because of their mechanism of action, also have other important actions that can in some cases enhance their therapeutic utility or provide some additional therapeutic benefit.
  • some calcium channel blockers not only dilate blood vessels, but also depress cardiac mechanical and electrical function, which can enhance their
  • Vasodilator drugs can be classified based on their site of action (arterial versus venous) or by mechanism of action. Some drugs primarily dilate resistance vessels (arterial dilators; e.g., hydralazine), while others primarily affect venous capacitance vessels (venous dilators; e.g., nitroglycerine). Many vasodilator drugs have mixed arterial and venous dilator properties (mixed dilators; e.g., alpha-adrenoceptor antagonists, angiotensin converting enzyme inhibitors), such as phentolamine.
  • mixed d dilators e.g., alpha-adrenoceptor antagonists, angiotensin converting enzyme inhibitors
  • vasodilator drugs based on their primary mechanism of action.
  • the figure to the right depicts important mechanistic classes of vasodilator drugs.
  • These classes of drugs, as well as other classes that produce vasodilation include: alpha- adrenoceptor antagonists (alpha-blockers); Angiotensin converting enzyme (ACE) inhibitors; Angiotensin receptor blockers (ARBs); beta 2 -adrenoceptor agonists (p 2 -agonists); calcium- channel blockers (CCBs); centrally acting sympatholytics; direct acting vasodilators; endothelin receptor antagonists; ganglionic blockers; nitrodilators; phosphodiesterase inhibitors; potassium- channel openers; renin inhibitors.
  • alpha-blockers Angiotensin converting enzyme (ACE) inhibitors
  • Angiotensin receptor blockers (ARBs) Angiotensin receptor blockers
  • the active or inactive components or ingredients can be substances or compounds that create an increased blood flow or flushing of the tissue to enable a modification or difference (increase or decrease) in transmucosal uptake of the API(s), and/or have a positive or negative heat of solution which are used as aids to modify (increase or decrease) transmucosal uptake.
  • pharmaceutical ingredient(s)(API(s)) delivered to the desired mucosal surface can vary in order to deliver a desired pharmacokinetic profile.
  • the sequence can be reversed or modified, for example, by applying the API(s) first by film, by swab, or by a first layer of a film, and then applying the permeation enhancer(s) by a film, by swab, spray, gel, rinse or by a second layer of a film.
  • the penetration enhancer(s) can be used as a pretreatment alone or in combination with at least one API to precondition the mucosa for further absorption of the API(s).
  • the treatment can be followed by another treatment with neat penetration enhancer(s) to follow the at least one API mucosal application.
  • the pretreatment can be applied as a separate treatment (film, gel, solution, swab etc.) or as a layer within a multilayered film construction of one or more layers.
  • the pretreatment may be contained within a distinct domain of a single film, designed to dissolve and release to the mucosa prior to release of the secondary domains with or without penetration enhancer(s) or API(s).
  • the active ingredient may then be delivered from a second treatment, alone or in combination with additional penetration enhancer(s).
  • additional penetration enhancer(s) There may also be a tertiary treatment or domain that delivers additional penetration enhancer(s) and/or at least one API(s) or prodrug(s), either at a different ratio relative to each other or relative to the overall loading of the other treatments.
  • This allows a custom pharmacokinetic profile to be obtained.
  • the product may have single or multiple domains, with penetration enhancer(s) and API(s) that can vary in mucosal application order, composition, concentration, or overall loading that leads to the desired absorption amounts and/or rates that achieve the intended pharmacokinetic profile and/or pharmacodynamic effect.
  • the film format can be oriented such that no distinct sides, or such that the film has at least one side of a multiple layer film where the edges are co-terminus (having or meeting at a shared border or limit).
  • the pharmaceutical composition can be a chewable or gelatin based dosage form, spray, gum, gel, cream, tablet, liquid or film.
  • the composition can include textures, for example, at the surface, such as microneedles or micro-protrusions.
  • microneedles or micro-protrusions have been shown to significantly increase transdermal delivery, including and especially for macromolecules.
  • solid microneedles which have been shown to increase skin permeability to a broad range of molecules and nanoparticles in vitro. In vivo studies have demonstrated delivery of
  • oligonucleotides reduction of blood glucose level by insulin, and induction of immune responses from protein and DNA vaccines.
  • needle arrays have been used to pierce holes into skin to increase transport by diffusion or iontophoresis or as drug carriers that release drug into the skin from a microneedle surface coating. Hollow microneedles have also been developed and shown to microinject insulin to diabetic rats.
  • microneedles the ratio of microneedle fracture force to skin insertion force (i.e. margin of safety) was found to be optimal for needles with small tip radius and large wall thickness.
  • Microneedles inserted into the skin of human subjects were reported as painless. Together, these results suggest that microneedles represent a promising technology to deliver therapeutic compounds into the skin for a range of possible applications. Using the tools of the
  • microneedles have been fabricated with a range of sizes, shapes and materials.
  • Microneedles can be, for example, polymeric, microscopic needles that deliver encapsulated drugs in a minimally invasive manner, but other suitable materials can be used.
  • Applicants have found that microneedles could be used to enhance the delivery of drugs through the oral mucosa, particularly with the claimed compositions.
  • the microneedles create micron sized pores in the oral mucosa which can enhance the delivery of drugs across the mucosa.
  • Solid, hollow or dissolving microneedles can be fabricated out of suitable materials including, but not limited to, metal, polymer, glass and ceramics.
  • the microfabrication process can include photolithography, silicon etching, laser cutting, metal electroplating, metal electro polishing and molding.
  • Microneedles could be solid which is used to pretreat the tissue and are removed before applying the film.
  • the drug loaded polymer film described in this application can be used as the matrix material of the microneedles itself. These films can have microneedles or micro protrusions fabricated on their surface which will dissolve after forming microchannels in the mucosa through which drugs can permeate.
  • film can include films and sheets, in any shape, including rectangular, square, or other desired shape.
  • a film can be any desired thickness and size.
  • a film can have a thickness and size such that it can be administered to a user, for example, placed into the oral cavity of the user.
  • a film can have a relatively thin thickness of from about 0.0025mm to about 0.250mm, or a film can have a somewhat thicker thickness of from about 0.250mm to aboutl .Omm. For some films, the thickness may be even larger, i.e., greater than about 1.0mm or thinner, i.e., less than about 0.0025mm.
  • a film can be a single layer or a film can be multi-layered, including laminated or multiple cast films.
  • a permeation enhancer and pharmaceutically active component can be combined in a single layer, each contained in separate layers, or can each be otherwise contained in discrete regions of the same dosage form.
  • the pharmaceutically active component contained in the polymeric matrix can be dispersed in the matrix.
  • the permeation enhancer being contained in the polymeric matrix can be dispersed in the matrix.
  • Oral dissolving films can fall into three main classes: fast dissolving, moderate dissolving and slow dissolving. Oral dissolving films can also include a combination of any of the above categories.
  • Fast dissolving films can dissolve in about 1 second to about 30 seconds in the mouth, including more than 1 second, more than 5 seconds, more than 10 seconds, more than 20 seconds, and less than 30 seconds.
  • Moderate dissolving films can dissolve in about 1 to about 30 minutes in the mouth including more than 1 minute, more than 5 minutes, more than 10 minutes, more than 20 minutes or less than 30 minutes, and slow dissolving films can dissolve in more than 30 minutes in the mouth.
  • fast dissolving films can include (or consist of) low molecular weight hydrophilic polymers (e.g., polymers having a molecular weight between about 1,000 to 9,000 daltons, or polymers having a molecular weight up to 200,000 daltons).
  • slow dissolving films generally include high molecular weight polymers (e.g., having a molecular weight in millions). Moderate dissolving films can tend to fall in between the fast and slow dissolving films.
  • Moderate dissolving films can dissolve rather quickly, but also have a good level of mucoadhesion.
  • Moderate dissolving films can also be flexible, quickly wettable, and are typically non-irritating to the user.
  • Such moderate dissolving films can provide a quick enough dissolution rate, most desirably between about 1 minute and about 20 minutes, while providing an acceptable mucoadhesion level such that the film is not easily removable once it is placed in the oral cavity of the user. This can ensure delivery of a pharmaceutically active component to a user.
  • a pharmaceutical composition can include one or more pharmaceutically active components.
  • the pharmaceutically active component can be a single pharmaceutical component or a combination of pharmaceutical components.
  • the pharmaceutically active component can be an anti-inflammatory analgesic agent, a steroidal anti-inflammatory agent, an antihistamine, a local anesthetic, a bactericide, a disinfectant, a vasoconstrictor, a hemostatic, a chemotherapeutic drug, an antibiotic, a keratolytic, a cauterizing agent, an antiviral drug, an antirheumatic, an antihypertensive, a bronchodilator, an anticholinergic, an anti-anxiety drug, an antiemetic compound, a hormone, a peptide, a protein or a vaccine.
  • the pharmaceutically active component can be the compound, pharmaceutically acceptable salt of a drug, a prodrug, a derivative, a drug complex or analog of a drug.
  • prodrug refers to a biologically inactive compound that can be metabolized in the body to produce a biologically active drug.
  • the pharmaceutaically active component can be an ester of epinephirine, for example, dipivefrin.
  • more than one pharmaceutically active component may be included in the film.
  • the pharmaceutically active components can be ace-inhibitors, anti-anginal drugs, anti-arrhythmias, anti-asthmatics, anti-cholesterolemics, analgesics, anesthetics, anti- convulsants, anti-depressants, anti-diabetic agents, anti-diarrhea preparations, antidotes, antihistamines, anti-hypertensive drugs, anti-inflammatory agents, anti-lipid agents, anti-manics, anti-nauseants, anti-stroke agents, anti-thyroid preparations, amphetamines, anti-tumor drugs, anti-viral agents, acne drugs, alkaloids, amino acid preparations, anti-tussives, anti-uricemic drugs, anti-viral drugs, anabolic preparations, systemic and non-systemic anti-infective agents, anti-neoplastics, anti-parkinsonian agents, anti-rhe
  • hypocalcemia management agents immunomodulators, immunosuppressives, migraine preparations, motion sickness treatments, muscle relaxants, obesity management agents, osteoporosis preparations, oxytocics, parasympatholytics, parasympathomimetics,
  • prostaglandins psychotherapeutic agents, respiratory agents, sedatives, smoking cessation aids, sympatholytics, tremor preparations, urinary tract agents, vasodilators, laxatives, antacids, ion exchange resins, anti-pyretics, appetite suppressants, expectorants, anti-anxiety agents, anti-ulcer agents, anti-inflammatory substances, coronary dilators, cerebral dilators, peripheral
  • vasodilators psycho-tropics, stimulants, anti-hypertensive drugs, vasoconstrictors, migraine treatments, antibiotics, tranquilizers, anti-psychotics, anti-tumor drugs, anti-coagulants, anti- thrombotic drugs, hypnotics, anti-emetics, anti-nauseants, anti-convulsants, neuromuscular drugs, hyper- and hypo-glycemic agents, thyroid and anti-thyroid preparations, diuretics, antispasmodics, uterine relaxants, anti-obesity drugs, erythropoietic drugs, anti-asthmatics, cough suppressants, mucolytics, DNA and genetic modifying drugs, diagnostic agents, imaging agents, dyes, or tracers, and combinations thereof.
  • the pharmaceutically active component can be buprenorphine, naloxone, acetaminophen, riluzole, clobazam, Rizatriptan, propofol, methyl salicylate, monoglycol salicylate, aspirin, mefenamic acid, flufenamic acid, indomethacin, diclofenac, alclofenac, diclofenac sodium, ibuprofen, ketoprofen, naproxen, pranoprofen, fenoprofen, sulindac, fenclofenac, clidanac, flurbiprofen, fentiazac, bufexamac, piroxicam, phenylbutazone, oxyphenbutazone, clofezone, pentazocine, mepirizole, tiaramide hydrochloride, hydrocortisone, predonisolone, dexarnethasone, triamcinolone, methyl
  • a composition including epinephrine or its salts or esters can have a biodelivery profile similar to that of epinephrine administered by injection, for example, using an EpiPen.
  • Epinephrine or its prodrug can be present in an amount of from about .01 mg to about 100 mg per dosage, for example, at a 0.1 mg, 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg or 100 mg dosage, including greater than 0.1 mg, more than 5 mg, more than 20 mg, more than 30 mg, more than 40 mg, more than 50 mg, more than 60 mg, more than 70 mg, more than 80 mg, more than 90 mg, or less than 100 mg, less than 90 mg, less than 80 mg, less than 70 mg, less than 60 mg, less than 50 mg, less than 40 mg, less than 30 mg, less than 20 mg, less than 10 mg, or less than 5 mg, or any combination thereof.
  • Epinephrine or its prodrug can be
  • Dipifevrin can be present in an amount of from about 0.5 mg to about 100 mg per dosage, for example, at a 0.5mg, 1 mg, 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg or 100 mg dosage including greater than 1 mg, more than 5 mg, more than 20 mg, more than 30 mg, more than 40 mg, more than 50 mg, more than 60 mg, more than 70 mg, more than 80 mg, more than 90 mg, or less than 100 mg, less than 90 mg, less than 80 mg, less than 70 mg, less than 60 mg, less than 50 mg, less than 40 mg, less than 30 mg, less than 20 mg, less than 10 mg, or less than 5 mg, or any combination thereof.
  • a composition e.g., including epinephrine
  • a mucosal delivery-enhancing agent selected from: (a) an aggregation inhibitory agent; (b) a charge-modifying agent; (c) a pH control agent; (d) a degradative enzyme inhibitory agent; (e) a mucolytic or mucus clearing agent; (f) a ciliostatic agent; (g) a membrane penetration-enhancing agent selected from: (i) 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 hydrophobic penetration enhancer;
  • dipifevrin is lipophilic and therefore has a higher permeation through a mucosa. It also has a longer plasma half life due to higher protein binding. It is capable of sustained blood levels, and does not interact with a-receptors, therefore minimizing or eliminating unwanted or harmful vasoconstriction.
  • Pivalic Add Dipifeverin can be provided as sublingual film in a similar manner as with epinephrine.
  • a film and/or its components can be water-soluble, water swellable or water-insoluble.
  • the term "water-soluble” can refer to substances that are at least partially dissolvable in an aqueous solvent, including but not limited to water.
  • the term “water-soluble” may not necessarily mean that the substance is 100% dissolvable in the aqueous solvent.
  • water-insoluble refers to substances that are not dissolvable in an aqueous solvent, including but not limited to water.
  • a solvent can include water, or alternatively can include other solvents (preferably, polar solvents) by themselves or in combination with water.
  • the composition can include a polymeric matrix. Any desired polymeric matrix may be used, provided that it is orally dissolvable or erodible.
  • the dosage should have enough bioadhesion to not be easily removed and it should form a gel like structure when administered. They can be moderate-dissolving in the oral cavity and particularly suitable for delivery of pharmaceutically active components, although both fast release, delayed release, controlled release and sustained release compositions are also among the various embodiments
  • the pharmaceutical composition film can include dendritic polymers which can include highly branched macromolecules with various structural architectures.
  • the dendritic polymers can include dendrimers, dendronised polymers (dendrigrafted polymers), linear dendritic hybrids, multi-arm star polymers, or hyperbranched polymers.
  • Hyperbranched polymers are highly branched polymers with imperfections in their structure. However they can be synthesized in a single step reaction which can be an advantage over other dendritic structures and are therefore suitable for bulk volume applications. The properties of these polymers apart from their globular structure are the abundant functional groups, intramolecular cavities, low viscosity and high solubility. Dendritic polymers have been used in several drug delivery applications. See, e.g., Dendrimers as Drug Carriers: Applications in Different Routes of Drug Administration. J Pharm Sci, VOL. 97, 2008, 123-143, which is incorporated by reference herein.
  • the dendritic polymers can have internal cavities which can encapsulate drugs.
  • the steric hindrance caused by the highly dense polymer chains might prevent the crystallization of the drugs.
  • branched polymers can provide additional advantages in formulating crystallizable drugs in a polymer matrix.
  • Suitable dendritic polymers include poly(ether) based dendrons, dendrimers and hyperbranched polymers, poly(ester) based dendrons, dendrimers and hyperbranched polymers, poly(thioether) based dendrons, dendrimers and hyperbranched polymers, poly(amino acid) based dendrons dendrimers and hyperbranched polymers, poly(arylalkylene ether) based dendrons, dendrimers and hyperbranched polymers, poly(alkyleneimine) based dendrons, dendrimers and hyperbranched polymers, poly(amidoamine) based dendrons, dendrimers or hyperbranched polymers.
  • hyperbranched polymers include poly(amines)s, polycarbonates, poly(ether ketone)s, polyurethanes, polycarbosilanes, polysiloxanes, poly(ester amine)s, poly(sulfone amine)s, poly(urea urethane)s and polyether polyols such as polyglycerols.
  • a film can be produced by a combination of at least one polymer and a solvent, optionally including other components.
  • the solvent may be water, a polar organic solvent including, but not limited to, ethanol, isopropanol, acetone, or any combination thereof.
  • the solvent may be a non-polar organic solvent, such as methylene chloride.
  • the film may be prepared by utilizing a selected casting or deposition method and a controlled drying process. For example, the film may be prepared through a controlled drying processes, which include application of heat and/or radiation energy to the wet film matrix to form a visco-elastic structure, thereby controlling the uniformity of content of the film.
  • the controlled drying processes can include air alone, heat alone or heat and air together contacting the top of the film or bottom of the film or the substrate supporting the cast or deposited or extruded film or contacting more than one surface at the same time or at different times during the drying process.
  • the films may be extruded as described in U.S. Patent Publication No. 2005/0037055 Al, which is incorporated by reference herein.
  • a polymer included in the films may be water-soluble, water-swellable, water-insoluble, or a combination of one or more either water-soluble, water-swellable or water-insoluble polymers.
  • the polymer may include cellulose, cellulose derivatives or gums.
  • Specific examples of useful water-soluble polymers include, but are not limited to, polyethylene oxide, pullulan, hydroxypropylmethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, polyvinyl pyrrolidone, carboxymethyl cellulose, polyvinyl alcohol, sodium alginate, polyethylene glycol, xanthan gum, tragancanth gum, guar gum, acacia gum, arabic gum, polyacrylic acid,
  • methylmethacrylate copolymer carboxyvinyl copolymers, starch, gelatin, and combinations thereof.
  • useful water-insoluble polymers include, but are not limited to, ethyl cellulose, hydroxypropyl ethyl cellulose, cellulose acetate phthalate, hydroxypropyl methyl cellulose phthalate and combinations thereof.
  • water-soluble polymer and variants thereof refer to a polymer that is at least partially soluble in water, and desirably fully or predominantly soluble in water, or absorbs water. Polymers that absorb water are often referred to as being water-swellable polymers.
  • the materials useful with the present invention may be water-soluble or water- swellable at room temperature and other temperatures, such as temperatures exceeding room temperature. Moreover, the materials may be water-soluble or water-swellable at pressures less than atmospheric pressure. In some embodiments, films formed from such water-soluble polymers may be sufficiently water-soluble to be dissolvable upon contact with bodily fluids.
  • polymers useful for incorporation into the films include biodegradable polymers, copolymers, block polymers or combinations thereof. It is understood that the term
  • biodegradable is intended to include materials that chemically degrade, as opposed to materials that physically break apart (i.e., bioerodable materials).
  • the polymers incorporated in the films can also include a combination of biodegradable or bioerodable materials.
  • useful polymers or polymer classes which meet the above criteria are: poly(glycolic acid) (PGA), poly(lactic acid) (PLA), polydioxanes, polyoxalates, poly(alpha-esters), polyanhydrides, polyacetates, polycaprolactones, poly(orthoesters), polyamino acids, polyaminocarbonates, polyurethanes, polycarbonates, polyamides, poly(alkyl cyanoacrylates), and mixtures and copolymers thereof.
  • PGA poly(glycolic acid)
  • PLA poly(lactic acid)
  • PDA polydioxanes
  • polyoxalates poly(alpha-esters)
  • polyanhydrides polyacetates
  • Additional useful polymers include, stereopolymers of L- and D-lactic acid, copolymers of bis(p-carboxyphenoxy)propane acid and sebacic acid, sebacic acid copolymers, copolymers of caprolactone, poly(lactic acid)/poly(glycolic acid)/polyethyleneglycol
  • copolymers copolymers, copolymers of polyurethane and (poly(lactic acid), copolymers of alpha-amino acids, copolymers of alpha-amino acids and caproic acid, copolymers of alpha-benzyl glutamate and polyethylene glycol, copolymers of succinate and poly(glycols), polyphosphazene, polyhydroxy-alkanoates or mixtures thereof.
  • the polymer matrix can include one, two, three, four or more components.
  • one or more water-soluble polymers may be used to form the film.
  • biodegradable may provide films with slower dissolution or disintegration rates than films formed from water-soluble polymers alone. As such, the film may adhere to the mucosal tissue for longer periods of time, such as up to several hours, which may be desirable for delivery of certain pharmaceutically active components.
  • an individual film dosage of the pharmaceutical film can have a suitable thickness, and small size, which is between about 0.0625-3 inch by about 0.0625-3 inch.
  • the film size can also be greater than 0.0625 inch, greater than 0.5 inch, greater than 1 inch, greater than 2 inches, about 3 inches, and greater than 3 inches, less than 3 inches, less than 2 inches, less than 1 inch, less than 0.5 inch, less than 0.0625 inch in at least one aspect, or greater than 0.0625 inch, greater than 0.5 inch, greater than 1 inch, greater than 2 inches, or greater than 3 inches, about 3 inches, less than 3 inches, less than 2 inches, less than 1 inch, less than 0.5 inch, less than 0.0625 inch in another aspect.
  • the aspect ratio including thickness, length, and width can be optimized by a person of ordinary skill in the art based on the chemical and physical properties of the polymeric matrix, the active pharmaceutical ingredient, dosage, enhancer, and other additives involved as well as the dimensions of the desired dispensing unit.
  • the film dosage should have good adhesion when placed in the buccal cavity or in the sublingual region of the user. Further, the film dosage should disperse and dissolve at a moderate rate, most desirably dispersing within about 1 minute and dissolving within about 3 minutes.
  • the film dosage may be capable of dispersing and dissolving at a rate of between about 1 to about 30 minutes, for example, about 1 to about 20 minutes, or more than 1 minute, more than 5 minutes, more than 7 minutes, more than 10 minutes, more than 12 minutes, more than 15 minutes, more than 20 minutes, more than 30 minutes, about 30 minutes, or less than 30 minutes, less than 20 minutes, less than 15 minutes, less than 12 minutes, less than 10 minutes, less than 7 minutes, less than 5 minutes, or less than 1 minute.
  • Sublingual dispersion rates may be shorter than buccal dispersion rates.
  • the films may include polyethylene oxide alone or in combination with a second polymer component.
  • the second polymer may be another water- soluble polymer, a water-swellable polymer, a water-insoluble polymer, a biodegradable polymer or any combination thereof.
  • Suitable water-soluble polymers include, without limitation, any of those provided above.
  • the water-soluble polymer may include hydrophilic cellulosic polymers, such as hydroxypropyl cellulose and/or
  • one or more water-swellable, water- insoluble and/or biodegradable polymers also may be included in the polyethylene oxide-based film. Any of the water-swellable, water-insoluble or biodegradable polymers provided above may be employed.
  • the second polymer component may be employed in amounts of about 0% to about 80% by weight in the polymer component, more specifically about 30% to about 70% by weight, and even more specifically about 40% to about 60% by weight, including greater than 5%), greater than 10%, greater than 15%, greater than 20%, greater than 30%, greater than 40%, greater than 50%, greater than 60%, and greater than 70%, about 70%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10% or less than 5% by weight.
  • Additives may be included in the films.
  • classes of additives include preservatives, antimicrobials, excipients, lubricants, buffering agents, stabilizers, blowing agents, pigments, coloring agents, fillers, bulking agents, sweetening agents, flavoring agents, fragrances, release modifiers, adjuvants, plasticizers, flow accelerators, mold release agents, polyols, granulating agents, diluents, binders, buffers, absorbents, glidants, adhesives, anti- adherents, acidulants, softeners, resins, demulcents, solvents, surfactants, emulsifiers, elastomers, anti-tacking agents, anti-static agents and mixtures thereof.
  • additives may be added with the pharmaceutically active component(s).
  • stabilizer means an excipient capable of preventing aggregation or other physical degradation, as well as chemical degradation, of the active pharmaceutical ingredient, another excipient, or the combination thereof.
  • Stabilizers may also be classified as antioxidants, sequestrants, pH modifiers, emulsifiers and/or surfactants, and UV stabilizers.
  • Antioxidants include, in particular, the following substances: tocopherols and the esters thereof, sesamol of sesame oil, coniferyl benzoate of benzoin resin, nordihydroguaietic resin and nordihydroguaiaretic acid (NDGA), gallates (among others, methyl, ethyl, propyl, amyl, butyl, lauryl gallates), butylated hydroxyanisole (BHA/BHT, also butyl-p-cresol); ascorbic acid and salts and esters thereof (for example, acorbyl palmitate), erythorbinic acid (isoascorbinic acid) and salts and esters thereof, monothioglycerol, sodium formaldehyde sulfoxylate, sodium metabi sulfite, sodium bisulfite, sodium sulfit
  • Typical antioxidants are tocopherol such as, for example, a-tocopherol and the esters thereof, butylated hydroxytoluene and butylated hydroxyanisole.
  • tocopherol also includes esters of tocopherol.
  • a known tocopherol is a-tocopherol.
  • a-tocopherol includes esters of a-tocopherol (for example, ⁇ -tocopherol acetate).
  • Sequestrants i.e., any compounds which can engage in host-guest complex formation with another compound, such as the active ingredient or another excipient; also referred to as a sequestering agent
  • a sequestering agent include calcium chloride, calcium disodium ethylene diamine tetra-acetate, glucono delta-lactone, sodium gluconate, potassium gluconate, sodium tripolyphosphate, sodium hexametaphosphate, and combinations thereof.
  • Sequestrants also include cyclic oligosaccharides, such as cyclodextrins, cyclomannins (5 or more a-D-mannopyranose units linked at the 1,4 positions by a linkages), cyclogalactins (5 or more ⁇ -D-galactopyranose units linked at the 1,4 positions by ⁇ linkages), cycloaltnns (5 or more a-D-altropyranose units linked at the 1,4 positions by a linkages), and combinations thereof.
  • cyclic oligosaccharides such as cyclodextrins, cyclomannins (5 or more a-D-mannopyranose units linked at the 1,4 positions by a linkages), cyclogalactins (5 or more ⁇ -D-galactopyranose units linked at the 1,4 positions by ⁇ linkages), cycloaltnns (5 or more a-D-altropyranose units linked at the 1,4 positions by a linkages
  • pH modifiers include acids (e.g., tartaric acid, citric acid, lactic acid, fumaric acid, phosphoric acid, ascorbic acid, acetic acid, succininc acid, adipic acid and maleic acid), acidic amino acids (e.g., glutamic acid, aspartic acid, etc.), inorganic salts (alkali metal salt, alkaline earth metal salt, ammonium salt, etc.) of such acidic substances, a salt of such acidic substance with an organic base (e.g., basic amino acid such as lysine, arginine and the like, meglumine and the like), and a solvate (e.g., hydrate) thereof.
  • acids e.g., tartaric acid, citric acid, lactic acid, fumaric acid, phosphoric acid, ascorbic acid, acetic acid, succininc acid, adipic acid and maleic acid
  • acidic amino acids e.g., glutamic acid, aspartic acid
  • pH modifiers include silicified microcrystalline cellulose, magnesium aluminometasilicate, calcium salts of phosphoric acid (e.g., calcium hydrogen phosphate anhydrous or hydrate, calcium, sodium or potassium carbonate or hydrogencarbonate and calcium lactate or mixtures thereof), sodium and/or calcium salts of carboxymethyl cellulose, cross-linked carboxymethylcellulose (e.g., croscarmellose sodium and/or calcium), polacrilin potassium, sodium and or/calcium alginate, docusate sodium, magnesium calcium, aluminium or zinc stearate, magnesium palmitate and magnesium oleate, sodium stearyl fumarate, and combinations thereof.
  • phosphoric acid e.g., calcium hydrogen phosphate anhydrous or hydrate, calcium, sodium or potassium carbonate or hydrogencarbonate and calcium lactate or mixtures thereof
  • carboxymethyl cellulose e.g., croscarmellose sodium and/or calcium
  • polacrilin potassium sodium and or/calcium alginate
  • emulsifiers and/or surfactants include poloxamers or pluronics, polyethylene glycols, polyethylene glycol monostearate, polysorbates, sodium lauryl sulfate, polyethoxylated and hydrogenated castor oil, alkyl polyoside, a grafted water soluble protein on a hydrophobic backbone, lecithin, glyceryl monostearate, glyceryl monostearate/polyoxyethylene stearate, ketostearyl alcohol/sodium lauryl sulfate, carbomer, phospholipids, (Cio-C2o)-alkyl and alkylene carboxylates, alkyl ether carboxylates, fatty alcohol sulfates, fatty alcohol ether sulfates, alkylamide sulfates and sulfonates, fatty acid alkylamide polyglycol ether sulfates, alkanesulfonates and hydroxyalkane
  • UV stabilizers examples include UV absorbers (e.g., benzophenones), UV quenchers (i.e., any compound that dissipates UV energy as heat, rather than allowing the energy to have a degradation effect), scavengers (i.e., any compound that eliminates free radicals resulting from exposure to UV radiation), and combinations thereof.
  • UV absorbers e.g., benzophenones
  • UV quenchers i.e., any compound that dissipates UV energy as heat, rather than allowing the energy to have a degradation effect
  • scavengers i.e., any compound that eliminates free radicals resulting from exposure to UV radiation
  • stabilizers include ascorbyl palmitate, ascorbic acid, alpha tocopherol, butylated hydroxytoluene, buthylated hydroxyanisole, cysteine HC1, citric acid, ethylenediamine tetra acetic acid (EDTA), methionine, sodium citrate, sodium ascorbate, sodium thiosulfate, sodium metabi sulfite, sodium bisulfite, propyl gallate, glutathione, thioglycerol, singlet oxygen quenchers, hydroxyl radical scavengers, hydroperoxide removing agents, reducing agents, metal chelators, detergents, chaotropes, and combinations thereof.
  • EDTA ethylenediamine tetra acetic acid
  • “Singlet oxygen quenchers” include, but are not limited to, alkyl imidazoles (e.g., histidine, L-camosine, histamine, imidazole 4-acetic acid), indoles (e.g., tryptophan and derivatives thereof, such as N- acetyl-5-methoxytryptamine, N-acetyl serotonin, 6-methoxy-l,2,3,4-tetrahydro-beta-carboline), sulfur-containing amino acids (e.g., methionine, ethionine, djenkolic acid, lanthionine, N-formyl methionine, felinine, S-allyl cysteine, S-aminoethyl-L-cysteine), phenolic compounds (e.g., tyrosine and derivatives thereof), aromatic acids (e.g., ascorbate, salicylic acid, and derivatives thereof), azide (e.g., sodium azi
  • Hydroxyl radical scavengers include, but are not limited to azide, dimethyl sulfoxide, histidine, mannitol, sucrose, glucose, salicylate, and L-cysteine.
  • Hydrodroperoxide removing agents include, but are not limited to catalase, pyruvate, glutathione, and glutathione peroxidases.
  • Reducing agents include, but are not limited to, cysteine and mercaptoethylene.
  • Methodal chelators include, but are not limited to, EDTA, EGTA, o- phenanthroline, and citrate.
  • Detergents include, but are not limited to, SDS and sodium lauroyl sarcosyl.
  • Chaotropes include, but are not limited to guandinium hydrochloride, isothiocyanate, urea, and formamide.
  • stabilizers can be present in 0.0001%-50% by weight, including greater than 0.0001%, greater than 0.001%), greater than 0.01%>, greater than 0.1%), greater than 1%, greater than 5%, greater than 10%>, greater than 20%, greater than 30%>, greater than 40%, greater than 50%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 1%, less than 0.1%, less than 0.01%, less than 0.001%, or less than 0.0001% by weight.
  • Useful additives can include, for example, gelatin, vegetable proteins such as sunflower protein, soybean proteins, cotton seed proteins, peanut proteins, grape seed proteins, whey proteins, whey protein isolates, blood proteins, egg proteins, acrylated proteins, water-soluble polysaccharides such as alginates, carrageenans, guar gum, agar-agar, xanthan gum, gellan gum, gum arabic and related gums (gum ghatti, gum karaya, gum tragancanth), pectin, water-soluble derivatives of cellulose: alkylcelluloses hydroxyalkylcelluloses and hydroxyalkylalkylcelluloses, such as methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose,
  • hydroxypropyl cellulose, hydroxyethylmethylcellulose, hydroxypropylmethyl cellulose, hydroxybutylmethylcellulose, cellulose esters and hydroxyalkylcellulose esters such as cellulose acetate phthalate (CAP), hydroxypropylmethylcellulose (HPMC); carboxyalkylcelluloses, carboxyalkylalkylcelluloses, carboxyalkyl cellulose esters such as carboxymethylcellulose and their alkali metal salts; water-soluble synthetic polymers such as polyacrylic acids and polyacrylic acid esters, polymethacrylic acids and polymethacrylic acid esters, polyvinylacetates, polyvinylalcohols, polyvinylacetatephthalates (PVAP), polyvinylpyrrolidone (PVP), PVA/vinyl acetate copolymer, and polycrotonic acids; also suitable are phthalated gelatin, gelatin succinate, crosslinked gelatin, shellac, water-soluble chemical derivatives of starch, cationically modified acrylates and
  • the additional components can range up to about 80%, desirably about 0.005%> to 50% and more desirably within the range of 1% to 20% based on the weight of all composition components, including greater than 1%, greater than 5%, greater than 10%, greater than 20%, greater than 30%, greater than 40%, greater than 50%, greater than 60%, greater than 70%, about 80%, greater than 80%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, about 3%, or less than 1%.
  • Other additives can include anti-tacking, flow agents and opacifiers, such as the oxides of magnesium aluminum, silicon, titanium, etc.
  • the composition can include plasticizers, which can include polyalkylene oxides, such as polyethylene glycols, polypropylene glycols, polyethylene- propylene glycols, organic plasticizers with low molecular weights, such as glycerol, glycerol monoacetate, diacetate or triacetate, triacetin, polysorbate, cetyl alcohol, propylene glycol, sugar alcohols sorbitol, sodium diethylsulfosuccinate, tri ethyl citrate, tributyl citrate, phytoextracts, fatty acid esters, fatty acids, oils and the like, added in concentrations ranging from about 0.1% to about 40%), and desirably ranging from about 0.5% to about 20% based on the weight of the composition including greater than 0.5%, greater than 1%, greater than 1.5%, greater than 2%, greater than 4%, greater than 5%, greater than 10%, greater than 15%, about 20%, greater than 20%, less than 20%, less than 15%
  • composition can further be added compounds to improve the texture properties of the film material such as animal or vegetable fats, desirably in their hydrogenated form.
  • the composition can also include compounds to improve the textural properties of the product.
  • Other ingredients can include binders which contribute to the ease of formation and general quality of the films.
  • binders include starches, natural gums, pregelatinized starches, gelatin, polyvinylpyrrolidone, methyl cellulose, sodium
  • carboxymethylcellulose ethylcellulose, polyacrylamides, polyvinyloxoazolidone, or
  • Such agents include solubility enhancing agents, such as substances that form inclusion compounds with active components. Such agents may be useful in improving the properties of very insoluble and/or unstable actives.
  • these substances are doughnut- shaped molecules with hydrophobic internal cavities and hydrophilic exteriors. Insoluble and/or instable pharmaceutically active components may fit within the hydrophobic cavity, thereby producing an inclusion complex, which is soluble in water. Accordingly, the formation of the inclusion complex permits very insoluble and/or unstable pharmaceutically active components to be dissolved in water.
  • a particularly desirable example of such agents are cyclodextrins, which are cyclic carbohydrates derived from starch. Other similar substances, however, are considered well within the scope of the present invention.
  • Suitable coloring agents include food, drug and cosmetic colors (FD&C), drug and cosmetic colors (D&C), or external drug and cosmetic colors (Ext. D&C). These colors are dyes, their corresponding lakes, and certain natural and derived colorants. Lakes are dyes absorbed on aluminum hydroxide. Other examples of coloring agents include known azo dyes, organic or inorganic pigments, or coloring agents of natural origin.
  • Inorganic pigments are preferred, such as the oxides or iron or titanium, these oxides, being added in concentrations ranging from about 0.001 to about 10%, and preferably about 0.5 to about 3%, including greater than 0.001%, greater than 0.01%, greater than 0.1%, greater than 0.5%, greater than 1%, greater than 2%, greater than 5%, about 10%, greater than 10%, less than 10%, less than 5%, less than 2%, less than 1%), less than 0.5%, less than 0.1%, less than 0.01%, or less than 0.001%, based on the weight of all the components.
  • Flavors may be chosen from natural and synthetic flavoring liquids.
  • An illustrative list of such agents includes volatile oils, synthetic flavor oils, flavoring aromatics, oils, liquids, oleoresins or extracts derived from plants, leaves, flowers, fruits, stems and combinations thereof.
  • a non-limiting representative list of examples includes mint oils, cocoa, and citrus oils such as lemon, orange, lime and grapefruit and fruit essences including apple, pear, peach, grape, strawberry, raspberry, cherry, plum, pineapple, apricot or other fruit flavors.
  • aldehydes and esters such as benzaldehyde (cherry, almond), citral i.e., alphacitral (lemon, lime), neral, i.e., beta-citral (lemon, lime), decanal (orange, lemon), aldehyde C-8 (citrus fruits), aldehyde C-9 (citrus fruits), aldehyde C-12 (citrus fruits), tolyl aldehyde (cherry, almond), 2,6-dimethyloctanol (green fruit), and 2-dodecenal (citrus, mandarin), combinations thereof and the like.
  • aldehydes and esters such as benzaldehyde (cherry, almond), citral i.e., alphacitral (lemon, lime), neral, i.e., beta-citral (lemon, lime), decanal (orange, lemon), aldehyde C-8 (citrus fruits), aldeh
  • the sweeteners may be chosen from the following non-limiting list: glucose (corn syrup), dextrose, invert sugar, fructose, and combinations thereof, saccharin and its various salts such as the sodium salt; dipeptide based sweeteners such as aspartame, neotame, advantame;
  • dihydrochalcone compounds glycyrrhizin; Stevia Rebaudiana (Stevioside); chloro derivatives of sucrose such as sucralose; sugar alcohols such as sorbitol, mannitol, xylitol, and the like.
  • hydrogenated starch hydrolysates and the synthetic sweetener 3,6-dihydro-6- methyl-l-l-l,2,3-oxathiazin-4-one-2,2-dioxide particularly the potassium salt (acesulfame-K), and sodium and calcium salts thereof, and natural intensive sweeteners, such as Lo Han Kuo. Other sweeteners may also be used.
  • Anti-foaming and/or de-foaming components may also be used with the films. These components aid in the removal of air, such as entrapped air, from the film-forming compositions. Such entrapped air may lead to non-uniform films.
  • Simethicone is one particularly useful anti- foaming and/or de-foaming agent.
  • the present invention is not so limited and other suitable anti-foam and/or de-foaming agents may be used.
  • Simethicone and related agents may be employed for densification purposes. More specifically, such agents may facilitate the removal of voids, air, moisture, and similar undesired components, thereby providing denser and thus more uniform films. Agents or components which perform this function can be referred to as densification or densifying agents. As described above, entrapped air or undesired components may lead to non-uniform films.
  • the film compositions further desirably contains a buffer so as to control the pH of the film composition.
  • a buffer so as to control the pH of the film composition.
  • Any desired level of buffer may be incorporated into the film composition so as to provide the desired pH level encountered as the pharmaceutically active component is released from the composition.
  • the buffer is preferably provided in an amount sufficient to control the release from the film and/or the absorption into the body of the pharmaceutically active component.
  • the buffer may include sodium citrate, citric acid, bitartrate salt and combinations thereof.
  • the pharmaceutical films described herein may be formed via any desired process.
  • the film dosage composition is formed by first preparing a wet composition, the wet composition including a polymeric carrier matrix and a therapeutically effective amount of a pharmaceutically active component.
  • the wet composition is cast into a film and then sufficiently dried to form a self-supporting film composition.
  • the wet composition may be cast into individual dosages, or it may be cast into a sheet, where the sheet is then cut into individual dosages.
  • the pharmaceutical composition can adhere to a mucosal surface.
  • the present invention finds particular use in the localized treatment of body tissues, diseases, or wounds which may have moist surfaces and which are susceptible to bodily fluids, such as the mouth, the vagina, organs, or other types of mucosal surfaces.
  • the composition carries a pharmaceutical, and upon application and adherence to the mucosal surface, offers a layer of protection and delivers the pharmaceutical to the treatment site, the surrounding tissues, and other bodily fluids.
  • the composition provides an appropriate residence time for effective drug delivery at the treatment site, given the control of erosion in aqueous solution or bodily fluids such as saliva, and the slow, natural erosion of the film concomitant or subsequent to the delivery.
  • the residence time of the composition depends on the erosion rate of the water erodable polymers used in the formulation and their respective concentrations.
  • the erosion rate may be adjusted, for example, by mixing together components with different solubility characteristics or chemically different polymers, such as hydroxyethyl cellulose and hydroxypropyl cellulose; by using different molecular weight grades of the same polymer, such as mixing low and medium molecular weight hydroxyethyl cellulose; by using excipients or plasticizers of various lipophilic values or water solubility characteristics (including essentially insoluble components); by using water soluble organic and inorganic salts; by using crosslinking agents such as glyoxal with polymers such as hydroxyethyl cellulose for partial crosslinking; or by post-treatment irradiation or curing, which may alter the physical state of the film, including its crystallinity or phase transition, once obtained.
  • the pharmaceutical composition film adheres to the mucosal surface and is held in place. Water absorption softens the composition, thereby diminishing the foreign body sensation.
  • delivery of the drug occurs. Residence times may be adjusted over a wide range depending upon the desired timing of the delivery of the chosen pharmaceutical and the desired lifespan of the carrier. Generally, however, the residence time is modulated between about a few seconds to about a few days. Preferably, the residence time for most pharmaceuticals is adjusted from about 5 seconds to about 24 hours. More preferably, the residence time is adjusted from about 5 seconds to about 30 minutes.
  • the composition adheres to the mucosal surface, it also provides protection to the treatment site, acting as an erodable bandage. Lipophilic agents can be designed to slow down erodability to decrease disintegration and dissolution.
  • excipients which are sensitive to enzymes such as amylase, very soluble in water such as water soluble organic and inorganic salts.
  • Suitable excipients may include the sodium and potassium salts of chloride, carbonate, bicarbonate, citrate, trifluoroacetate, benzoate, phosphate, fluoride, sulfate, or tartrate.
  • the amount added can vary depending upon how much the erosion kinetics is to be altered as well as the amount and nature of the other components in the composition.
  • Emulsifiers typically used in the water-based emulsions described above are, preferably, either obtained in situ if selected from the linoleic, palmitic, myristoleic, lauric, stearic, cetoleic or oleic acids and sodium or potassium hydroxide, or selected from the laurate, palmitate, stearate, or oleate esters of sorbitol and sorbitol anhydrides, polyoxyethylene derivatives including monooleate, monostearate, monopalmitate, monolaurate, fatty alcohols, alkyl phenols, allyl ethers, alkyl aryl ethers, sorbitan monostearate, sorbitan monooleate and/or sorbitan monopalmitate.
  • the amount of pharmaceutically active component to be used depends on the desired treatment strength and the composition of the layers, although preferably, the pharmaceutical component comprises from about 0.001% to about 99%, more preferably from about 0.003 to about 75%o, and most preferably from about 0.005%> to about 50% by weight of the composition, including, more than 0.005%, more than 0.05%, more than 0.5%, more than 1%, more than 5%, more than 10%, more than 15%, more than 20%, more than 30%, about 50%, more than 50%, less than 50%, less than 30%>, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%), less than 0.5%, less than 0.05%, or less than 0.005%.
  • the amounts of other components may vary depending on the drug or other components but typically these components comprise no more than 50%, preferably no more than 30%, and most preferably no more than 15% by total weight of the composition.
  • the thickness of the film may vary, depending on the thickness of each of the layers and the number of layers. As stated above, both the thickness and amount of layers may be adjusted in order to vary the erosion kinetics.
  • the thickness ranges from 0.005 mm to 2 mm, preferably from 0.01 to 1 mm, and more preferably from 0.1 to 0.5 mm, including greater than 0.1 mm, greater than 0.2 mm, about 0.5 mm, greater than 0.5 mm, less than 0.5 mm, less than 0.2 mm, or less than 0.1 mm.
  • the thickness of each layer may vary from 10 to 90% of the overall thickness of the layered composition, and preferably varies from 30 to 60%, including greater than 10%, greater than 20%, greater than 30%), greater than 40%, greater than 50%, greater than 70%, greater than 90%, about 90%, less than 90%, less than 70%, less than 50%, less than 40%, less than 30%, less than 20%, or less than 10%.
  • the preferred thickness of each layer may vary from 0.01 mm to 0.9 mm, or from 0.03 to 0.5 mm.
  • the treatment site may include any area in which the film is capable of delivery and/or maintaining a desired level of pharmaceutical in the blood, lymph, or other bodily fluid.
  • such treatment sites include the oral, esophageal, aural, ocular, anal, nasal, and vaginal mucosal tissue, as well as, the skin. If the skin is to be employed as the treatment site, then usually larger areas of the skin wherein movement will not disrupt the adhesion of the film, such as the upper arm or thigh, are preferred.
  • the pharmaceutical composition can also be used as a wound dressing.
  • the film can not only protect a wound but also deliver a pharmaceutical in order to promote healing, aseptic, scarification, to ease the pain or to improve globally the condition of the sufferer.
  • Some of the: examples given below are well suited for an application to the skin or a wound.
  • the formulation might require incorporating a specific hydrophilic/hygroscopic excipient which would help in maintaining good adhesion on dry skin over an extended period of time.
  • Another advantage of the present invention when utilized in this manner is that if one does not wish that the film be noticeable on the skin, then no dyes or colored substances need be used. If, on the other hand, one desires that the film be noticeable, a dye or colored substance may be employed.
  • the pharmaceutical composition can adhere to mucosal tissues, which are wet tissues by nature, it can also be used on other surfaces such as skin or wounds.
  • a Franz diffusion cell is an in vitro skin permeation assay used in formulation development.
  • the Franz diffusion cell apparatus ( Figure 1A) consists of two chambers separated by a membrane of, for example, animal or human tissue. The test product is applied to the membrane via the top chamber. The bottom chamber contains fluid from which samples are taken at regular intervals for analysis to determine the amount of active that has permeated the membrane.
  • a Franz diffusion cell 100 includes a donor compound 101, a donor chamber 102, a membrane 103, sampling port 104, receptor chamber 105, stir bar 106, and a heater/circulator 107.
  • a pharmaceutical composition is a film 100 comprising a polymeric matrix 200, the pharmaceutically active component 300 being contained in the polymeric matrix.
  • the film can include a permeation enhancer 400.
  • the graphs show the permeation of an active material from a composition.
  • the graph shows that for the epinephrine base - solubilized in-situ vs. the inherently soluble epinephrine bitartrate, no meaningful differences were observed.
  • Epinephrine bitartrate was selected for further development based on ease of processing. Flux is derived as slope of the amount permeated as a function of time. Steady state flux is taken from the plateau of flux vs time curve multiplied by the volume of receiver media and normalized for permeation area.
  • this graph shows average amount of active material permeated vs. time, with 8.00 mg/mL epinephrine bitartrate and 4.4 mg/mL epinephrine base solubilized.
  • this graph shows average flux vs. time, with 8.00 mg/mL epinephrine bitartrate and 4.4 mg/mL epinephrine base solubilized.
  • this graph shows ex -vivo permeation of epinephrine bitartrate as a function of concentration.
  • concentrations 4 mg/mL, 8 mg/mL, 16 mg/mL and 100 mg/mL. Results showed that increasing concentration resulted in increased permeation, and level of enhancement diminishes at higher loading.
  • this graph shows permeation of epinephrine bitartrate as a function of solution pH. Acidic conditions explored to promote stability. The results compared epinephrine bitartrate pH 3 buffer and epinephrine bitartrate pH 5 buffer, and found that the epinephrine bitartrate pH 5 buffer was slightly favorable.
  • this graph shows the influence of enhancers on permeation of epinephrine, indicated as amount permeated as a function of time. Multiple enhancers were screened, including Labrasol, capryol 90, Plurol Oleique, Labrafil, TDM, SGDC, Gelucire 44/14 and clove oil. Significant impact on time to onset and steady state flux was achieved, and surprisingly enhanced permeation was achieved for clove oil and Labrasol.
  • FIGS. 6A and 6B show the release of epinephrine on polymer platforms and the effect of enhancers on its release, indicated as amount permeated (in ⁇ g) vs. time.
  • Figure 6A shows the epinephrine release from different polymer platforms.
  • Figure 6B shows the impact of enhancers on epinephrine release.
  • this graph shows a pharmacokinetic model in the male Yucatan, miniature swine.
  • the study compares a 0.3 mg Epipen, a 0.12 mg Epinephrine IV and a placebo film.
  • this graphs shows the impact of no enhancer on the concentration profiles of a 40 mg epinephrine film vs, a 0.3 mg Epipen.
  • this graph shows the impact of Enhancer A (Labrasol) on the concentration profiles of a 40 mg epinephrine film vs, a 0.3 mg Epipen.
  • Enhancer L clove oil
  • This graph shows the impact of Enhancer L(clove oil) and film dimension (10-1-1 thinner bigger film and 11-1-1 thicker smaller film) on the concentration profiles of 40 mg Epinephrine films vs. a 0.3 mg Epipen.
  • this graph shows the concentration profiles for varying doses of Epinephrine films in a constant matrix for Enhancer L (clove oil) vs. a 0.3 mg Epipen.
  • the graph shows the concentration profiles for varying doses of Epinephrine films in a constant matrix for Enhancer L (clove oil) vs. a 0.3 mg Epipen.
  • the graph shows the concentration profiles for varying doses of Epinephrine films in a constant matrix for Enhancer A (Labrasol) vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol and Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40 mg Epinephrine Films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol and Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40 mg Epinephrine Films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40 mg Epinephrine Films vs. a 0.3 mg Epipen.
  • this graph shows the impact of Farnesol and Farnesol in combination with Linoleic Acid on plasma concentration profiles of 40 mg Epinephrine Films vs. a 0.3 mg Epipen.
  • the following examples are provided to illustrate pharmaceutical compositions, as well as, methods of making and using, pharmaceutical compositions and devices described herein.
  • Permeation enhancement was studied using a number of permeation enhancers with Epinephrine Bitartrate 16.00 mg/mL concentration. The results show flux enhancement represented in the data below. For 100% Eugenol and 100% Clove Oil, the results showed steady state flux reached significantly earlier along with an unexpectedly heightened % flux enhancement.
  • clove oil was obtained from clove leaf. Similar results may be obtained from clove oil from clove bud and/or clove stem. Based on this data, similar permeability enhancement results can be expected from pharmaceutical compounds structurally similar to epinephrine.
  • a permeation procedure is conducted as follows.
  • a temperature bath is set to 37° C, and receiver media is placed in a water bath to adjust the temperature and begin degassing.
  • a franz diffusion cell is obtained and prepared.
  • the franz diffusion cell includes a donor compound, a donor chamber, a membrane, sampling port, receptor chamber, stir bar, and a heater/circulator.
  • a stir bar is inserted into a franz diffusion cell.
  • Tissue is placed over the franz diffusion cell, and it is ensured that the tissue covers the entire area with an overlap onto a glass joint.
  • the top of a diffusion cell is placed over the tissue, and the top of the cell is clamped to the bottom.
  • If testing films one can perform the following next steps: (1) weigh films, punch to match diffusion area (or smaller), reweigh, record pre- and post-punching weight; (2) wet a donor area with approximately 100 ⁇ L of phosphate buffer; (3) place film on a donor surface, top with 400 ⁇ L of phosphate buffer, and start timers.
  • one can perform the following steps: (1) using a micropipette, dispense 500 ⁇ L of the solution into each donor cell, start the timers; (2) sample 200 ⁇ L at the following time points (time 0 min, 20 min, 40 min, 60 min, 120 min, 180 min, 240 min,300 min, 360 min), and place in labelled HPLC vials, ensure no air is trapped in the bottom of the vial by tapping the closed vials; (3) replace each sample time with 200 ⁇ L of receptor media (to maintain 5 mL); (4) When all time points completed, disassemble the cells and dispose of all materials properly.
  • Tissue is freshly excised and shipped (e.g. overnight) at 4° C.
  • the tissue is processed and frozen at -20° C for up to three weeks prior to use.
  • the tissue is dermatomed to precise thickness.
  • the tissue is placed in a franz diffusion cell, which includes a donor compound, a donor chamber, a membrane, sampling port, receptor chamber, stir bar, and a
  • Samples are taken from the receiving chamber at given intervals and replaced with fresh media.
  • the studies were conducted under a protocol approved by the Animal Experimentation Ethics Committee of the University of Barcelona (Spain) and the Committee of Animal Experimentation of the regional autonomous government of Catalonia (Spain).
  • Female pigs 3-4- months-old were used.
  • the porcine buccal mucosa from the cheek region was excised immediately after the pigs were sacrificed in the animal facility at Bellvitge Campus (University of Barcelona, Spain) using an overdose of sodium thiopental anesthesia.
  • the fresh buccal tissues were transferred from the hospital to the laboratory in containers filled with Hank's solution.
  • the remaining tissue specimens were stored at -80° C in containers with a PBS mixture containing 4% albumin and 10% DMSO as cryoprotective agents.
  • porcine buccal mucosa was cut to 500 +/- 50 ⁇ thick sheets, which contributes to the diffusional barrier (Buccal bioadhesive drug delivery— A promising option for orally less efficient drugs Sudhakar et al., Journal of Controlled Release 114 (2006) 15-40), using an electric dermatome (GA 630, Aesculap, Tuttlingen, Germany) and trimmed with surgical scissors in adequate pieces. The majority of the underlying connective tissue was removed with a scalpel. Membranes were then mounted in specially designed membrane holders with a permeation orifice diameter of 9 mm (diffusion area 0.636 cm 2 ).
  • each porcine buccal membrane was mounted between the donor (1.5 mL) and the receptor (6 mL) compartments with the epithelium side facing the donor chamber and the connective tissue region facing the receiver of static Franz-type diffusion cells (Vidra Foe Barcelona, Spain) avoiding bubbles formation.
  • Porcine oral mucosal tissue has similar histological characteristics to human oral mucosal tissue (Heaney TG, Jones RS, Histological investigation of the influence of adult porcine alveolar mucosal connective tissues on epithelial differentiation. Arch Oral Biol 23 (1978) 713— 717; Squier CA, and Collins P, The relationship between soft tissue attachment, epithelial downgrowth and surface porosity. Journal of Periodontal Research 16 (1981) 434-440). Lesch et al.
  • Tissue integrity was monitored by the absorption of the fluorescein isothiocyanate (FITC)-labeled dextran 20 kDa (FD20) and tissue viability was assessed using an MTT (3-[4,5- dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) biochemical assay and histological evaluation.
  • Permeability through the buccal epithelium was 1.8-fold greater for caffeine and 16.7-fold greater for oestradiol compared with full thickness buccal tissue. Flux values for both compounds were comparable for fresh and frozen buccal epithelium although histological evaluation demonstrated signs of cellular death in frozen tissue. The tissue appeared to remain viable for up to 12 h postmortem using the MTT viability assay which was also confirmed by histological evaluation.
  • Kulkarni et al. investigated the relative contributions of the epithelium and connective tissue to the barrier properties of porcine buccal tissue. In vitro permeation studies were conducted with antipyrine, buspirone, bupivacaine and caffeine as model permeants. The permeability of the model diffusants across buccal mucosa with thickness of 250, 400, 500, 600, and 700 ⁇ was determined. A bilayer membrane model was developed to delineate the relative contribution to the barrier function of the epithelium and the connective tissue. The relative contribution of the connective tissue region as a permeability barrier significantly increased with increasing mucosal tissue thickness.
  • a mucosal tissue thickness of approximately 500 ⁇ was recommended by the authors for in vitro transbuccal permeation studies as the epithelium represented the major permeability barrier for all diffusants at this thickness.
  • the authors also investigated the effects of a number of biological and experimental variables on the permeability of the same group of model permeants in porcine buccal mucosa (Porcine buccal mucosa as in vitro model: effect of biological and experimental variables, Kulkarni et al., J P harm Sci. 2010 99(3): 1265-77). Significantly, higher permeability of the permeants was observed for the thinner region behind the lip (170-220 ⁇ ) compared with the thicker cheek (250-280 ⁇ ) region.
  • Porcine buccal mucosa retained its integrity in Kreb's bicarbonate ringer solution at 4 °C for 24 h. Heat treatment to separate the epithelium from underlying connective tissue did not adversely affect its permeability and integrity characteristics compared with surgical separation.
  • porcine buccal mucosa Different areas of porcine buccal mucosa have different pattern of permeability, there is significantly higher permeability in the region behind the lips in comparison to cheek region, because in porcine buccal mucosa, the epithelium acts as a permeability barrier, and the thickness of the cheek epithelium is greater than that of the region behind the lips (Harris and Robinson, 1992).
  • the fresh or frozen porcine buccal mucosa from the same area was cut to 500 ⁇ 50 ⁇ thick sheets, which contributes to the diffusional barrier (Sudhakar et al., 2006), were obtained using an electric dermatome (model GA 630, Aesculap, Tuttlingen, Germany) and trimmed with surgical scissors in adequate pieces.
  • each porcine buccal membrane was mounted between the donor (1.5 mL) and the receptor (6 mL) compartments with the epithelium facing the donor chamber and the connective tissue region facing the receiver of static Franz-type diffusion cells (Vidra Foe, Barcelona, Spain) avoiding bubbles formation.
  • the diffusion cells Prior to conducting the experiments, the diffusion cells were incubated for 1 h in a water bath to equilibrate the temperature in all cells (37° ⁇ 1 C). Each cell contained a small Teflon coated magnetic stir bar which was used to ensure that the fluid in the receptor compartment remained homogenous during the experiments. Sink conditions were ensured in all experiments after initial testing of PP saturation concentration in the receptor medium.
  • Samples (300 ⁇ .) were drawn via syringe from the center of the receptor compartment at the following time intervals: 0.25, 0.5, 1, 2, 3, 4, 5 and 6 h.
  • the removed sample volume was immediately replaced with the same volume of fresh receptor medium (PBS; pH 7.4) with great care to avoid trapping air beneath the dermis.
  • Cumulative amounts of the drug ⁇ g) penetrating the unit surface area of the mucosa membrane (cm 2 ) were corrected for sample removal and plotted versus time (h).
  • the diffusion experiments were carried out 27 times for the fresh and 22 times for the frozen buccal mucosa.
  • porcine oral mucosa is a suitable model for human oral mucosa. Permeability across the porcine oral mucosa is not metabolically linked therefore it is not important for the tissue to be viable.
  • porcine floor of mouth and ventral (underside) tongue mucosa membranes were excised by blunt dissection using a scalpel.
  • the excised mucosa were cut into approximately 1 cm squares and frozen on aluminium foil at -20°C until used ( ⁇ 2 weeks).
  • the mucosa was used in the permeation studies within 3 h of excision.
  • the permeability of the membranes to quinine was determined using all-glass Franz diffusion cells with a nominal receptor volume of 3.6mL and diffusional area of 0.2 cm 2 .
  • the cell flanges were greased with high performance vacuum grease and the membranes mounted between the receptor and donor compartments, with the mucosal surface uppermost. Clamps were used to hold the membranes into position before the receptor compartments were filled with degassed phosphate buffered saline (PBS), pH 7.4.
  • PBS degassed phosphate buffered saline
  • Micromagnetic stirrer bars were added to the receptor compartments and the complete cells were placed in a water bath at 37°C.
  • the membranes were equilibrated with PBS applied to the donor compartments for 20 min before being aspirated with a pipette.
  • the receptor phases were withdrawn from the sampling ports and aliquots of lmL samples were transferred to FIPLC autosampler vials, before being replaced with fresh PBS stored at 37°C.
  • 5 ⁇ _ of the respective quinine solution was reapplied to the donor phase up to 10 h. The purpose of this was to represent a hypothetical in-use finite dosing regimen based upon an interval of 2 h between doses. At least 3 replicates were carried out for each study.
  • Epinephrine Base was tested - solubilized in situ vs. the inherently soluble Epinephrine Bitartrate and no differences were found.
  • Epinephrine Bitartrate was selected for further development based on ease of processing. Flux was derived as slope of the amount permeated as a function of time. Steady state flux extrapolated from the plateau of flux vs time curve multiplied by the volume of receiver media.
  • the graph in Figure 2A shows average amount permeated vs. time, with 8.00 mg/mL Epinephrine bitartrate and 4.4 mg/mL Epinephrine base solubilized.
  • the graph in Figure 2B shows average flux vs. time, with 8.00 mg/mL Epinephrine bitartrate and 4.4 mg/mL Epinephrine base solubilized.
  • Figure 4 shows permeation of epinephrine bitartrate as a function of solution pH. Acidic conditions were explored to promote stability. The results compared Epinephrine Bitartrate pH 3 buffer and Epinephrine Bitartrate pH 5 buffer, and found that the Epinephrine Bitartrate pH 5 buffer was slightly favorable.
  • Enhancers were selected and designed with functionality influencing different barriers in the mucosa. While all tested enhancers did improve the amout permeated over time, clove oil and Labrasol, in particular have shown significantly and unexpectedly high enhancement of permeation.
  • TDM set 2 (A,B,C) steady std dev set set 2 (D,E,F)
  • a pharmacokinetic model in the male Yucatan, miniature swine was studied.
  • the graph on Figure 7 shows the results of a pharmacokinetic model in the male Yucatan, miniature swine.
  • the study compares a 0.3 mg Epipen, a 0.12 mg Epinephrine IV and a placebo.
  • FIG. 9 shows the impact of Enhancer A (Labrasol) on the concentration profiles of a 40 mg Epinephrine film vs. a 0.3 mg Epipen.
  • Figure 10 shows the impact of Enhancer L (clove oil) on the concentration profiles of two 40 mg Epinephrine films (10-1-1) and (11-1-1) vs. a 0.3 mg Epipen.
  • a pharmacokinetic model in the male miniature swine was studied to determine the impact of an enhancer (farnesol) on epinephrine concentration over time.
  • the graph on Figure 15 shows the epinephrine plasma concentration (in ng/mL) as a function of time (in minutes) following sublingual or intramuscular administration of a farnesol permeation enhancer.
  • the 31-1-1 film demonstrates enhanced stability of epinephrine concentration starting at about 30-40 minutes until approximately 130 minutes.
  • this graph shows a pharmacokinetic model in the male miniature swine was studied to determine the impact of an enhancer (farnesol) on epinephrine concentration over time following sublingual or intramuscular administration.
  • the epinephrine plasma concentration (in ng/mL) is shown as a function of time (in minutes) following sublingual or intramuscular administration of a farnesol permeation enhancer in Epinephrine films.
  • the study compared data from three 0.3 mg Epipens against five 30 mg Epinephrine films (32-1-1). The data shows the Epinephrine films film having enhanced stability of epinephrine concentration starting at about 20-30 minutes until approximately 130 minutes.
  • an epinephrine pharmaceutical composition film can be made with the following formulation:
  • compositions were made with the following formulation:
  • compositions were made with the following formulation:
  • this graph shows a pharmacokinetic model (logarithmic scale) in the male miniature swine studied to determine the impact of an enhancer (6% clove oil and 6% Labrasol) on epinephrine plasma concentration over time following sublingual or intramuscular administration.
  • the epinephrine plasma concentration (in ng/mL) is shown as a function of time (in minutes) following sublingual or intramuscular administration of a farnesol permeation enhancer in Epinephrine films.
  • the data shows the Epinephrine films film having enhanced stability of epinephrine concentration starting at just after the 10 minute time point through about 30 minutes, and until approximately 100 minutes.
  • this graph shows a pharmacokinetic model of the Epinephrine film formulation in the male miniature swine as referenced in Figure 19 compared against the average data collected from a 0.3 mg Epipen (indicated in diamond data points).
  • the average plasma concentration for the 0.3 mg Epipen peaked between 0.5 and 1 ng/mL.
  • the Epinephrine film formulation peaked between 4 and 4.5 ng/mL.
  • this graph shows a pharmacokinetic model in the male miniature swine studied to determine the impact of an enhancer (9% clove + 3% Labrasol) on epinephrine concentration over time following sublingual or intramuscular administration across 7 animal models.
  • the general peak concentration was achieved between 10-30 minutes.
  • a dipifevrin pharmaceutical composition film can be made with the following formulation:
  • a dipifevrin pharmaceutical composition film can be made with the following formulation:
  • a dipifevrin pharmaceutical composition film can be made with the following formulation:
  • the investigators compared the pharmacokinetics of epinephrine after Dipivefnn SL administration to other routes of administrations (Oral, SC and IV), specifically analyzing epinephrine bioavailability and conversion of dipivefnn to epinephrine.
  • the study compared various dose routes: sublingual (SL), per oral tablet (PO), subcutaneous (SC) and intravenous (IV).
  • the PK Time-points (for All Groups): 0 (pre-dose), 2, 5, 10, 12, 15, 17, 20, 25, 30, 40, 60, 90 and 120 minutes and 3 hr, 4 hr, 6 hr and 8 hr post dose.
  • An Oral Irritation assessment was performed (SL film group): Predose (0) and 24 hr postdose by Draize scoring.
  • the dipifevrin film (DF) is an exemplary embodiment of the claimed pharmaceutical composition.
  • the data shows average epinephrine plasma concentration vs. time in 60 minutes.
  • the data shows average epinephrine plasma concentration vs. time in 480 minutes.
  • the dipifevrin film showed ability to meet target Cmax of 0.2 ng/ml- 1.5 ng/ml in less than 30 minutes.
  • the Cmax was shown to be 0.1 ng/ml- 2 ng/ml, 0.15 ng/ml-1.5 ng/ml, and 0.2 ng/ml- 1.0 ng/ml.
  • the Cmax was shown to be greater than 0.1 ng/ml, greater than 0.15 ng/ml, greater than 0.2 ng/ml, greater than 0.4ng/ml, greater than 0.5ng/ml, greater than 0.6 ng/ml, greater than 0.7 ng/ml and less than 2 ng/ml and less than 1.5 ng/ml.
  • the Tmax is shown in the range of 0-480 minutes, including 10-60 minutes, 20-40 minutes, 12-15 minutes, and 5-10 minutes.
  • the Tmax has been shown to be less than 25 minutes, less than 20 minutes, 15 minutes, less than 12 minutes, and less than 10 minutes.
  • the data shows average dipifevrin plasma concentration vs. time in 60 minutes.
  • the data shows average dipifevrin plasma concentration vs. time in 480 minutes.
  • the amount of dipifevrin is shown to decrease as epinephrine is being formed.
  • This data shows a surprising ability of the subject film to meet a target Cmax of 0.2 ng/ml- 1.5 ng/ml and a Tmax of less than 35 minutes.
  • the Cmax is shown to be 0.1 ng/ml- 2 ng/ml, 0.15 ng/ml -25 ng/ml, 0.2 ng/ml- 1.0 ng/ml, 0.2 ng/ml -1.2 ng/ml, and 0.2 ng/ml - 1.3 ng/ml.
  • the Cmax has been shown to be greater than 0.1 ng/ml, greater than 0.15 ng/ml, greater than 0.2 ng/ml, greater than 0.4ng/ml, greater than 0.5ng/ml, greater than l .Ong/ml, greater than 1.2 ng/ml, and less than 3 ng/ml, less than 2 ng/ml and less than 1.5 ng/ml.
  • the Tmax is shown in the range of 0-480 minutes, including 10-60 minutes, 20-40 minutes, 12-15 minutes, and 5-10 minutes.
  • the Tmax has been shown to be less than 25 minutes, less than 20 minutes, 15 minutes, less than 12 minutes, and less than 10 minutes.
  • the data shows that average plasma concentration of epinephrine (dipivefrin SL 16-1-1 E) in round data points and dipivefrin (dipifevrin SL-Dip 16-1-1 D) in square data points. This shows the amount of dipifevrin decreases as epinephrine is being formed.
  • the graph shows the conversion of dipivefrin to epinephrine (SC and IV groups).
  • the dashed lines indicate dipifevrin, and the solid lines indicate epinephrine.
  • the data shows average average epinephrine profiles across all matrix studies.
  • the time to initial peak average is within about 30 minutes, 25 minutes, and 20 minutes.
  • the 6mg dipivefrin/6% clove film (dip 10-1-1) achieved the fastest average results.
  • the data shows additional data comparing plasma concentration of dipifevrin and epinephrine (ng/ml) vs. time in hours.
  • a mean therapeautic window has shown to be achieved within 30 minutes or less, including individual averages in 20 minutes or less, and 15 minutes or less.
  • this graph shows the epinephrine plasma concentration vs. time profile in a follow-up study that examined the loading of phentolamine and how it influences the absorption of epinephrine.
  • 30 mg epinephrine sublingual films (ESF) were used. The results show that the loading is dose dependent. The results are shown in the graph and summarized below:
  • a therapeutic window can be seen for ESF within 30 minutes or less, 25 minutes or less, and 20 minutes or less.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Emergency Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Inorganic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Botany (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Plant Substances (AREA)
EP18786593.6A 2017-09-27 2018-09-27 Enhanced delivery epinephrine and prodrug compositions Pending EP3687510A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US15/717,859 US20180125977A1 (en) 2016-05-05 2017-09-27 Enhanced delivery epinephrine compositions
US15/791,249 US20180104195A1 (en) 2016-05-05 2017-10-23 Enhanced delivery epinephrine compositions
PCT/US2018/053042 WO2019067670A1 (en) 2017-09-27 2018-09-27 EPINEPHRINE AND ENHANCED DELIVERY COMPOSITIONS

Publications (1)

Publication Number Publication Date
EP3687510A1 true EP3687510A1 (en) 2020-08-05

Family

ID=63858169

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18786593.6A Pending EP3687510A1 (en) 2017-09-27 2018-09-27 Enhanced delivery epinephrine and prodrug compositions

Country Status (8)

Country Link
EP (1) EP3687510A1 (zh)
JP (2) JP2020535162A (zh)
KR (1) KR20200060748A (zh)
CN (1) CN111465391A (zh)
BR (1) BR112020005948A2 (zh)
CA (1) CA3076816A1 (zh)
IL (1) IL273485A (zh)
WO (1) WO2019067670A1 (zh)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11273131B2 (en) 2016-05-05 2022-03-15 Aquestive Therapeutics, Inc. Pharmaceutical compositions with enhanced permeation
KR20230137362A (ko) 2016-05-05 2023-10-04 어퀘스티브 테라퓨틱스, 아이엔씨. 강화된 전달 에프네프린 조성물
CN115003279A (zh) * 2019-11-01 2022-09-02 阿奎斯蒂弗医疗股份有限公司 前药组合物和治疗方法
US11571468B2 (en) * 2021-02-01 2023-02-07 Matthias W. Rath Micronutrient combination to reduce blood pressure
WO2023245069A2 (en) * 2022-06-14 2023-12-21 Aquestive Therapeutics, Inc. Enhanced delivery epinephrine and prodrug compositions
EP4302751A1 (en) * 2022-07-07 2024-01-10 LTS Lohmann Therapie-Systeme AG Oral thin films with low water activity

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7425292B2 (en) 2001-10-12 2008-09-16 Monosol Rx, Llc Thin film with non-self-aggregating uniform heterogeneity and drug delivery systems made therefrom
US7666337B2 (en) 2002-04-11 2010-02-23 Monosol Rx, Llc Polyethylene oxide-based films and drug delivery systems made therefrom
US8603514B2 (en) 2002-04-11 2013-12-10 Monosol Rx, Llc Uniform films for rapid dissolve dosage form incorporating taste-masking compositions
US7357891B2 (en) 2001-10-12 2008-04-15 Monosol Rx, Llc Process for making an ingestible film
US8765167B2 (en) 2001-10-12 2014-07-01 Monosol Rx, Llc Uniform films for rapid-dissolve dosage form incorporating anti-tacking compositions
US20030143195A1 (en) * 2002-01-30 2003-07-31 Pinsker Judy Senior Use of histamine as a drug delivery enhancing compound for use in transmucosal or transdermal delivery
US20040096569A1 (en) * 2002-11-15 2004-05-20 Barkalow David G. Edible film products and methods of making same
US20050266085A1 (en) * 2004-05-28 2005-12-01 Warner Kevin S Gelled emulsion and microemulsion formulations for dermal drug delivery
US20070202163A1 (en) * 2005-09-09 2007-08-30 Mutasem Rawas-Qalaji Fast-disintegrating epinephrine tablets for buccal or sublingual administration
IL175338A0 (en) * 2006-05-01 2006-09-05 Biota Ltd Orally administrable films and preparation thereof
US20070293581A1 (en) * 2006-06-05 2007-12-20 Malcolm Hill Methods for Buccal, Lingual or Sublingual Dosing Regimens of Epinephrine for the Treatment of Allergic Emergencies
US20070293582A1 (en) * 2006-06-05 2007-12-20 Malcolm Hill Epinephrine dosing regimens comprising buccal, lingual or sublingual and injectable dosage forms
PE20091084A1 (es) * 2007-12-07 2009-07-23 Schering Plough Healthcare Formulaciones farmaceuticas de fenilefrina y composiciones para absorcion transmucosal
MY167918A (en) * 2009-04-23 2018-09-27 Londonpharma Ltd Composition for use in the sublingual delivery of medicaments to humans
CA2794734C (en) * 2010-03-30 2017-12-12 Phosphagenics Limited Transdermal delivery patch
JP2014532482A (ja) * 2011-10-28 2014-12-08 クウォン, スン−ユン 片頭痛を処置するための可溶性固溶体穿孔器パッチ
CN107669649A (zh) * 2012-10-11 2018-02-09 Ix生物制药有限公司 固体剂型
US20160051494A1 (en) * 2014-08-21 2016-02-25 Mylan, Inc. Multi-dose medication kit for treating anaphylaxis
KR20230137362A (ko) * 2016-05-05 2023-10-04 어퀘스티브 테라퓨틱스, 아이엔씨. 강화된 전달 에프네프린 조성물
BR112018072467A2 (pt) * 2016-05-05 2019-02-19 Aquestive Therapeutics, Inc. composições farmacêuticas com permeação aumentada
WO2019067596A1 (en) * 2017-09-26 2019-04-04 Aquestive Therapeutics, Inc. ADMINISTRATIVE PHARMACEUTICAL COMPOSITIONS CONTAINING PERMEATION PROMOTERS

Also Published As

Publication number Publication date
WO2019067670A1 (en) 2019-04-04
CN111465391A (zh) 2020-07-28
CA3076816A1 (en) 2019-04-04
BR112020005948A2 (pt) 2020-12-01
JP2020535162A (ja) 2020-12-03
JP2023159150A (ja) 2023-10-31
KR20200060748A (ko) 2020-06-01
IL273485A (en) 2020-05-31

Similar Documents

Publication Publication Date Title
US20230138361A1 (en) Enhanced delivery epinephrine compositions
US20170290776A1 (en) Pharmaceutical compositions with enhanced permeation
EP3687510A1 (en) Enhanced delivery epinephrine and prodrug compositions
US20180125977A1 (en) Enhanced delivery epinephrine compositions
JP7431724B2 (ja) 増強された透過を有する医薬組成物
US20190022022A1 (en) Pharmaceutical compositions with enhanced permeation
US20190022023A1 (en) Enhanced delivery epinephrine and prodrug compositions
US11273131B2 (en) Pharmaceutical compositions with enhanced permeation
US20180104195A1 (en) Enhanced delivery epinephrine compositions
US20190091281A1 (en) Delivery pharmaceutical compositions including permeation enhancers
US20240148673A1 (en) Enhanced delivery epinephrine and prodrug compositions
US20240016734A1 (en) Enhanced delivery epinephrine and prodrug compositions
US20230130055A1 (en) Pharmaceutical compositions with enhanced stability profiles
KR20240093850A (ko) 개선된 안정성 프로파일을 갖는 약제학적 조성물

Legal Events

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

Free format text: STATUS: UNKNOWN

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: 20200416

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

RIN1 Information on inventor provided before grant (corrected)

Inventor name: VARJAN, STEPHANIE, MARIE

Inventor name: WARGACKI, STEPHEN, PAUL

Inventor name: SCHOBEL, ALEXANDER, MARK

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: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20210521

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SCHOBEL, ALEXANDER, MARK

Inventor name: VARJAN, STEPHANIE, MARIE

Inventor name: WARGACKI, STEPHEN, PAUL

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230522