US20140166027A1 - Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit - Google Patents

Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit Download PDF

Info

Publication number
US20140166027A1
US20140166027A1 US13/715,028 US201213715028A US2014166027A1 US 20140166027 A1 US20140166027 A1 US 20140166027A1 US 201213715028 A US201213715028 A US 201213715028A US 2014166027 A1 US2014166027 A1 US 2014166027A1
Authority
US
United States
Prior art keywords
conditioning unit
nicotine
absorption
agent
absorption conditioning
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/715,028
Inventor
Richard C. Fuisz
Joseph M. Fuisz
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US13/715,028 priority Critical patent/US20140166027A1/en
Priority to US13/871,213 priority patent/US20140166028A1/en
Priority to PCT/US2013/073346 priority patent/WO2014093127A2/en
Publication of US20140166027A1 publication Critical patent/US20140166027A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • A24F47/002
    • 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
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61K31/465Nicotine; Derivatives thereof
    • AHUMAN NECESSITIES
    • A24TOBACCO; CIGARS; CIGARETTES; SIMULATED SMOKING DEVICES; SMOKERS' REQUISITES
    • A24FSMOKERS' REQUISITES; MATCH BOXES; SIMULATED SMOKING DEVICES
    • A24F40/00Electrically operated smoking devices; Component parts thereof; Manufacture thereof; Maintenance or testing thereof; Charging means specially adapted therefor
    • A24F40/10Devices using liquid inhalable precursors
    • 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
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals

Definitions

  • Electronic vapor devices are increasingly popular with consumers. Such devices comprise electronic cigarettes, electronic pipes, electronic hookahs, personal vaporizers and other inhalator embodiments, both electronic and mechanical.
  • the level of toxicants from the electronic cigarette is a small fraction as compared with a conventional cigarette. See, e.g., John H. Lauterbach, Murray Laugesen, James D. Ross, “Suggested protocol for estimation of harmful and potentially harmful constituents in mainstream aerosals generated by electronic delivery systems (ENDS)”, SOT, San Francisco, Calif., Mar. 10-16, 2012 (http://cigtoxdoc.ehost-services113.com/sot2012poster1860aspresented.pdf) hereby incorporated by reference).
  • the present invention relates, inter alfa, to a method for the administration of nicotine, THC, tobacco, cannabidiol or a base alkaloid to a mammal, e.g., a human.
  • the method includes administering an absorption conditioning unit, comprising at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, to the mammal, and then administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, cannabidiol and a base alkaloid.
  • the present invention also relates A method for the administration of nicotine, THC, cannabidiol or a base alkaloid to a mammal, including administering an absorption conditioning unit including at least at least one material that creates a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration; and then administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, tobacco, cannabidiol and a base alkaloid.
  • an absorption conditioning unit includes a dosage form including at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, wherein said absorption conditioning unit (i) does not have an essentially acidic character, (ii) does not contain a drug, (iii) does not contain THC or other Marijuana derived active ingredient, and (iv) does not contain tobacco except for trace amounts in flavor.
  • an absorption conditioning unit includes a dosage form including at least at least one material that creates a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration, the dosage form not containing an acid and not containing a drug.
  • the present invention relates to a method for the administration of nicotine, THC, cannabidiol or a base alkaloid to a mammal, especially to a human, and to a prior administered absorption conditioning unit useful in such a method.
  • Nicotine inhalers are known art.
  • Pfizer markets Nicotrol® in the United States.
  • the Nicotrol® inhaler consists of a mouthpiece and a plastic cartridge delivering 4 mg of nicotine from a porous plug containing 10 mg nicotine. The cartridge is inserted into the mouthpiece prior to use. Nicotine is released when air is inhaled through the inhaler. No heating element is used—the product relies on the natural propensity of nicotine to sublime at room temperature.
  • the Nicotrol® package insert describes its method of action: “Most of the nicotine released from the NICOTROL Inhaler is deposited in the mouth. Only a fraction of the dose released, less than 5%, reaches the lower respiratory tract. An intensive inhalation regimen (80 deep inhalations over 20 minutes) releases on the average 4 mg of nicotine content of each cartridge of which about 2 mg is systemically absorbed. Peak plasma concentrations are typically reached within 15 minutes of the end of inhalation . . . . Absorption of nicotine through the buccal mucosa is relatively slow and the high and rapid rise followed by the decline in nicotine arterial plasma concentrations seen with cigarette smoking are not achieved with the inhaler.
  • the arterial nicotine concentrations rise slowly to an average of 6 ng/mL in contrast to those of a cigarette, which increase rapidly and reach a mean Cmax of approximately 49 ng/mL within five minutes . . . .
  • Ad libitum use of the NICOTROL Inhaler typically produces nicotine plasma levels of 6-8 ng/mL, corresponding to about 1 ⁇ 3 of those achieved with cigarette smoking.”
  • the typical cigarette contains less than 2 mg of nicotine (http://www.bigsixsmokes.com/Camel.shtml), yet delivers a far higher peak systemic nicotine plasma concentration (Cmax), far quicker (Tmax), than Nicotrol® (see http://pharmrev.aspetjournals.org/content/57/1/79/T1.expansion.html which is incorporated by reference).
  • Cmax peak systemic nicotine plasma concentration
  • Tmax far quicker
  • Nicotrol® see http://pharmrev.aspetjournals.org/content/57/1/79/T1.expansion.html which is incorporated by reference.
  • We postulate that the more efficient delivery of nicotine from a cigarette (as compared with the Nicotrol®) is a result of enhanced motility of nicotine occurring in smoke—which results from the burning of a cigarette. This superior motility allows the nicotine to readily reach the intermediate and deep lung where the enormous surface area of the lung promotes rapid absorption (as compared with buccal delivery).
  • the effluent vapor of the electronic vapor device like an e-cigarette is heavy and slow moving. Thus, it does not reach the deep lung in substantial part, akin to the NICOTROL® device. The result is pharmacokinetic performance much closer to Nicotrol® than to a cigarette.
  • the solution will be alkaline. But if we vaporize the water, the vapor would not be 7.0. A complicated reaction is provoked and accelerated by the higher temperatures used to vaporize.
  • the present invention seeks to enhance the buccal delivery of nicotine (or Marijuana bioactives or any basic alkaloids) from an electronic (or non-electronic) vapor (or other inhalation) device. Because of the nature of vapor effluent, there is a limit to the agents and excipients that may be employed in the composition in the vapor device that will travel with the vapor into the user's buccal cavity and oral mucosa. Similarly, absorption can be enhanced for the oropharyngial mucosa.
  • the present invention contemplates an “absorption conditioning unit” that is placed in the user's mouth prior to and/or contemporaneously with, the use of the electronic vapor device (or other inhalator).
  • the absorption conditioning unit is a typically a dosage unit customarily used in the pharmaceutical and/or confection industries.
  • the absorption conditioning unit may comprise any of following non-limitative solid or liquid examples: a tablet, an orally dissolving tablet, a chewable tablet, a multi-layered tablet, a capsule, a liquid-gel, a lozenge, a thin film, a sheet, a slab, a powder, granules, a gummy, a lollipop, a suspension, a toothpaste, a chewing gum or a solution.
  • a solution or suspension may be delivered via a spray or aerosol or like method.
  • the absorption conditioning unit will preferably comprise a solid dosage unit.
  • Chewable dosage units are a preferred embodiment for their propensity to spread material around the mouth. Film dosage units are desirable because of small sized and their ability to be easily co-packaged with the inhalation device. For purposes of this invention, we would like to increase the amount of modified mucosal surface that is exposed to the agents contained in the absorption conditioning unit. Liquids are also good for this purpose but liquids are inevitably costly to distribute.
  • One exception to this, and another preferred embodiment, is a liquid that is constituted by the user whereby dry or concentrated agent-materials are provided to the user who then mixes them with a liquid like for example tap water and used as a an oral gargle and/or oral swishing agent.
  • the emission from an electronic cigarette is a vapor—formed by heating a liquid (or primarily) liquid composition that is brought into contact with the heating element of the electronic cigarette.
  • Said liquid composition typically comprises a vapor agent that creates a smoke-like vapor when heated (most commonly glycerin or propylene glycol), USP nicotine, water and a flavor system.
  • Said liquid may alternatively comprise a tobacco extract together with a vapor agent (see US 2012/0145170 A1 “METHOD FOR PREPARING TOBACCO EXTRACT FOR ELECTRONIC SMOKING DEVICES” and hereby incorporated by reference).
  • Rate of flow of the liquid composition to the heating element is typically controlled with a wick or sponge like material such that the heating element is not overwhelmed. Drawing on the e-cigarette turns on the heating element (which can be achieved through a variety of ways in including air pressure differentials, a sound-based system and other switches).
  • asthma inhalers typically employ a propellant to further help delivery the particles to the lung.
  • E-cigarettes, personal vaporizers and nicotine inhalers do not have the benefit of propellant but rather rely on the pressure differentials created by the action of the lung itself. While there is some difference of views relative to the particle size of water contained in a water vapor, some argue that droplets in the 2-5 micron range can constitute a vapor.
  • Vapors are gases, that is, consist of non-associated molecules. Generally speaking, a gas is considered a vapor when its standard state at that pressure and temperature is a liquid or a solid. So, for example, gaseous water at less than 100° C. is usually called a water vapor. The trick, of course, is that the partial pressure of the water vapor is less than one atmosphere. However more importantly, condensation occurs which results in sizes that are too large to operationally do much other than deposit in the mouth, oral pharynx and nasal pharynx.
  • a capture agent which we define as a material that can (a) temporarily bind condensate and (b) maintain the condensate in contact with mucosal surfaces for a sustained period to enable absorption. This will allow for increased absorption of nicotine (or other actives as discussed herein).
  • sustained period we mean 0.5 to 90 minutes, preferably 3 to 10 minutes.
  • the capture agent is contained in the absorption conditioning unit where it is released into the mouth. Such release is preferably controlled over a sustained period to ensure availability of the capture agent over a prolonged period.
  • any nicotine in exhaled vapor is essentially lost nicotine that was actually needed to improve the product (perhaps unlike the cigarette). This is why the capture agent is so important—to capture nicotine and maintain it in buccal or sublingual contact for effective absorption.
  • Film formers are, without limitation, useful as a capture agent because the condensate may have solvent activity on condensate, on the one hand, and two, many film agents tend to have muco-adhesive properties that will help maintain the nicotine with mucosal surfaces. Likewise, sugars are useful for this purpose. Other bioadhesive agents may be useful.
  • the film forming polymers described and/or listed in U.S. Pat. No. 7,824,588 (the contents of which are incorporated herein by reference) may be useful.
  • the polymer 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 or a cellulose derivative.
  • useful water soluble polymers include, but are not limited to, pullulan, hydroxypropylmethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, polyvinyl pyrrolidone, carboxymethyl cellulose, polyvinyl alcohol, sodium aginate, 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 refers 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. Desirably, the water soluble polymers are water soluble or water swellable having at least 20 percent by weight water uptake. Water swellable polymers having a 25 or greater percent by weight water uptake are also useful. Films or dosage forms of the present invention formed from such water soluble polymers are desirably sufficiently water soluble to be dissolvable upon contact with bodily fluids.
  • polymers useful as capture agents include biodegradable polymers, copolymers, block polymers and combinations thereof.
  • PGA poly(glycolic acid)
  • PLA poly(lactic acid)
  • PLA polydioxanoes
  • polyoxalates poly(.alpha.-esters)
  • polyanhydrides polyacetates
  • polycaprolactones poly(orthoesters)
  • polyamino acids polyaminocarbonates, polyurethanes, polycarbonates, polyamides, poly(alkyl cyanoacrylates), and mixtures and copolymers thereof.
  • 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 of polyurethane and (poly(lactic acid), 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 and mixtures thereof. Binary and ternary systems are contemplated.
  • Suitable specific polymers useful include those marketed under the Medisorb and Biodel trademarks.
  • the Medisorb materials are marketed by the Dupont Company of Wilmington, Del. and are generically identified as a “lactide/glycolide co-polymer” containing “propanoic acid, 2-hydroxy-polymer with hydroxy-polymer with hydroxyacetic acid.”
  • Four such polymers include lactide/glycolide 100 L, believed to be 100% lactide having a melting point within the range of 338-347° F. (170-175° C.); lactide/glycolide 100 L, believed to be 100%, glycolide having a melting point within the range of 437-455° F.
  • lactide/glycolide 85/15 believed to be 85% lactide and 15% glycolide with a melting point within the range of 338-347° F. (170-175° C.); and lactide/glycolide 50/50, believed to be a copolymer of 50% lactide and 50% glycolide with a melting point within the range of 338-347° F. (170-175° C.).
  • Biodel materials represent a family of various polyanhydrides which differ chemically.
  • sugars and preferred combinations of/with sugars described and/or listed at U.S. Pat. No. 5,935,600 may, without limitation, be useful as capture agents.
  • “sugars” are those substances which are based on simple crystalline mono- and di-saccharide structures, i.e., based on C 5 and C 6 sugar structures.
  • “Sugars” include simple sugars, e.g., glucose, sucrose, maltose, lactose, arabinose, xylose, ribose, fructose, mannose, pentose, galactose sorbose, dextrose, and sugar alcohols, e.g., sorbitol, xylitol, mannitol, pentatol, maltitol, isomalt, sucralose and mixtures of any of these.
  • Maltodextrins may be used Maltodextrins include those mixtures of carbohydrates resulting from hydrolysis of a saccharide feedstock which are described as solids having a DE of up to and including 65.
  • Maltooligosaccharides produced by selective hydrolysis of cornstarch followed by removal of high and low molecular weight compounds may be used.
  • the general description of maltooligosaccharides as contemplated herein is set forth in U.S. Pat. No. 5,387,431 (the contents of which are incorporated herein by reference).
  • Polydextrose is also contemplated for use as a capture agent.
  • Polydextrose is a non-sucrose, essentially non-nutritive carbohydrate substitute. It can be prepared through polymerization of glucose in the presence of polycarboxylic acid catalyst and polyols.
  • polydextrose is known to be commercially available in three forms: polydextrose A and polydextrose K, which are powdered solids, and polydextrose N supplied as a 70% solution. Each of these products also contain some low molecular weight components, such as glucose, sorbitol and certain oligomers.
  • polydextrose Applicants incorporate herein the contents of U.S. Pat. No. 5,279,849 (the contents of which are incorporated herein by reference).
  • bioadhesive (and other) agents discussed and/or listed in Pranshu Tangri et al, International Journal of Biopharmaceutics, 2011, 2(1): 36-46 (the contents of which are incorporated herein by reference) may be used as capture agents.
  • Glycerin and/or propylene glycol solubility is desirable so that the capture agent will better bind the vapor condensate.
  • materials used as capture agents are also water soluble because that they naturally wash out of the mouth without leaving unpleasant residue. The term preferably is pointedly used since non-water soluble polymers will ultimately be washed out of the mouth and swallowed—just less readily.
  • the absorption conditioning unit may comprise a pH buffer agent (a “buffer agent”).
  • the buffer agent is used to raise the pH of the mouth in order to increase nicotine absorption in the buccal cavity in a manner which is based on pka and the Henderson Hasselbach equation.
  • the pH of the mouth is increased to 7 to 10, preferably 7.8 to 10, most preferably from 8.5 to 9.5.
  • the buffer agent increases the pH of the oral cavity for a period of ten minutes or more after administration
  • Buffering agents may be used to control pH, including without limitation, sodium bicarbonate, potassium bicarbonate, sodium carbonate, potassium carbonate, calcium carbonate, dipotassium phosphate, potassium citrate, sodium phosphate and any other such buffer system.
  • the buffer system may be designed to dynamically control the pH of the product taking into consideration the effect of saliva during use, i.e., a dynamic buffer system.
  • buffer systems to obtain the preferred pH include dibasic sodium phosphate and monobasic sodium phosphate. Both are FDA accepted buffer materials used and listed in the inactive ingredients list.
  • the ratio of monobasic/dibasic can be 4.6/8.6; for a pH of 7.5 the ratio of monobasic/dibasic can be 1.9/11.9; and for a pH of 8.0 the ratio of monobasic/dibasic can be 0.6/13.4.
  • Alkaline buffer agents are easier to work with for sustained release pH control. Acidic agents tend to increase salivary flow which must be addressed in the formulation and practically makes formulation more difficult.
  • the absorption conditioning unit of the present invention will generally not have an acidic character, i.e. lower the pH of water when fully dissolved therein.
  • the buffer agent may comprise a substantial portion of the absorption conditioning unit, including from 1 to 99% by mass of the dosage unit itself.
  • the absorption conditioning unit may comprise a penetration agent, i.e., a substance that enhances absorption through the mucosa, mucosal coating and epithelium (otherwise known (see U.S. Patent Application Publication No. 2006/0257463 A1, the content of which is incorporated herein by reference) as a “penetration enhancer” or “permeability enhancer”).
  • a penetration agent i.e., a substance that enhances absorption through the mucosa, mucosal coating and epithelium (otherwise known (see U.S. Patent Application Publication No. 2006/0257463 A1, the content of which is incorporated herein by reference) as a “penetration enhancer” or “permeability enhancer”).
  • the penetration agent may comprise but is not limited to polyethylene glycol (PEG), diethylene glycol monoethyl ether (Transcutol), 23-lauryl ether, aprotinin, azone, benzalkomin chloride, cetylperidium chloride, cetylmethylammonium bromide, dextran sulfate, lauric acid, lauric acid/propylene glycol, lysophosphatilcholine, menthol, methoxysalicylate, oleic acid, phosphaidylcholine, polyoxyethylene, polysorbate 80, sodium EDTA, sodium glycholated, sodium glycodeoxycholate, sodium lauryl sulfate, sodium salicylate, sodium taurocholate, sodium taurodeoxycholate, sulfoxides, and various alkyl glycosides or, as described in U.S.
  • PEG polyethylene glycol
  • Transcutol diethylene glycol monoethyl ether
  • bile salts such as sodium deoxycholate, sodium glycodeoxycholate, sodium taurocholate and sodium glycocholate
  • surfactants such as sodium lauryl sulfate, polysorbate 80, laureth-9, benzalkonium chloride, cetylpyridinium chloride and polyoxyethylene monoalkyl ethers such as the BRIJ® and MYRJ® series
  • benzoic acids such as sodium salicylate and methoxy salicylate
  • fatty acids such as lauric acid, oleic acid, undecanoic acid and methyl oleate
  • fatty alcohols such as octanol and nonanol, laurocapram
  • the polyols propylene glycol and glycerin, cyclodextrins
  • the sulfoxides such as dimethyl sulfoxide and dodecyl methyl sulfoxide
  • the terpenes such as ment
  • the penetration agent is a polyol, e.g., polyethylene glycol (PEG), glycerin, maltitol, sorbitol etc. or diethylene glycol monoethyl ether (Transcutol).
  • PEG polyethylene glycol
  • glycerin glycerin
  • maltitol glycerin
  • sorbitol glycerin
  • Diethylene glycol monoethyl ether Transcutol
  • Thermal agents may also be employed. Heat increases blood flow which can in turn increase absorption.
  • Physical agents may be employed—for example, in a liquid format the liquid may be pre-heated (like adding agents to water pre-heated for tea).
  • Chemical thermal agents may be employed to provoke an exothermic reaction.
  • the absorption conditioning unit further might, but not necessarily, comprise at least one flavor for more pleasant administration.
  • Some care must go to flavor selection so that the absorption conditioning unit is pleasant yet not disruptive to the flavor delivered by the inhalation device.
  • Tobacco flavor may be desirable as such and not require any other flavor.
  • Appropriate flavors can be sourced from Tobacco Technologies in Eldersburg, Md. In many examples the more neutral the flavor the better so as not to interfere with the flavor of the vapor. Trace amounts of tobacco may present in flavorings, particularly for a tobacco flavored-product.
  • the absorption conditioning unit may further comprise effervescent agents to effect effervescence to enhance or improve absorption. Care must be taken that the acidic component of most effervescent systems do not interfere with the foal of a basic pH range.
  • the absorption conditioning unit may be built into the end of an electronic smoking device (or similar non-electronic inhalator) such that the user can remove it either with hands or mouth immediately prior to use. In one embodiment, this can be akin to a cigar smoker “biting off” the end of a cigar, except here the user removes the absorption conditioning unit and retains it in the mouth. It may also be possible to use a flavored liquid that is dispensed, akin to the menthol “crush” systems that are currently sold but adapted for this purpose. It may also be a dissolvable tip which conditions the pH of the saliva.
  • the present invention can be used to enhance the delivery of bioactives such nicotine, THC, tobacco, cannabidiol or a base alkaloid, or any combination thereof.
  • bioactives such nicotine, THC, tobacco, cannabidiol or a base alkaloid, or any combination thereof.
  • the present invention can be adapted for use with marijuana/tobacco combinations. Such combinations can involve the vaporization of tobacco and marijuana leaf, or a mixture of extracts from both materials.
  • the absorption conditioning unit of the present invention may be employed with suitable adaptation (e.g. optimize target buccal pH) to enhance the absorption of THC (tetrahydrocannabinol), cannabidiols and other active ingredients from cannabis.
  • suitable adaptation e.g. optimize target buccal pH
  • “Vaporization of cannabis has been explored as an alternative to smoking.
  • the advantages of vaporization apparently include the formation of a smaller quantity of toxic by-products such as carbon monoxide, polycyclic aromatic hydrocarbons (PAHs) and tar, as well as a more efficient extraction of THC from the cannabis material (47, 48, 49, 43, 50).
  • the subjective effects and plasma concentrations of THC are comparable to those of smoked cannabis with absorption being somewhat faster with the vaporizer (43).
  • the vaporizer is well-tolerated, with no reported adverse effects, and is generally preferred over smoking by most subjects (43).
  • While vaporization is amenable to self-titration (49, 43), the proper use of the vaporizer for optimal administration of medicinal cannabis has to be established in more detail (50).
  • the amount and type of cannabis placed in the vaporizer, the vaporizing temperature and duration of vaporization, and the balloon volume are some of the parameters that can affect the delivery of THC (49). Bioequivalence of vaporization
  • THC can be absorbed orally by ingestion of foods containing cannabis (butters, oils, brownies, cookies), teas prepared from leaves and flowering tops, or through ingestion of capsules containing THC or THC analogues. Absorption from an oral dose of 20 mg THC in a chocolate cookie was described as slow and unreliable (42), with a systemic availability of only 4 to 12% (46). While most subjects displayed peak plasma THC concentrations between 1 to 2 h, some of the 11 subjects in the study only peaked at 6 h and many had more than one peak. Consumption of cannabis-laced brownies containing 2.8% THC was associated with changes in behaviour although the effects were slow to appear and variable (51). Peak effects occurred 2.5 to 3.5 h after dosing.
  • the dynamic of marijuana vaporization is functionally comparable to tobacco/nicotine.
  • the actives are not effectively delivered to the deep lung but tend to be absorbed buccally.
  • Marijuana is however different in that swallowed condensate is more apt to be absorbed in the GI tract than nicotine.
  • vapor devices can also be “analog” like for example the butane-fired Ploom® device or the butane fired “click-a-toke.” Similarly, vapor can be generated from hookah-like devices powered by hot coals and the like. And, it is possible to have an inhaler—like Nicotrol®—that does not use any heat source. It should be noted that it is expressly contemplated that the present invention can be used with any inhaler-like device (and not solely with a vapor device).
  • the absorption conditioning unit does not contain tobacco or nicotine, it will generally be exempt from regulation, except where it is part of an approved pharmaceutical product (i.e. a “combination product”).
  • references to nicotine herein include any nicotine salt, nicotine that is naturally or synthetically derived, including from tobacco or any other material.
  • the present invention i.e. the absorption conditioning unit
  • the absorption conditioning unit can be readily adapted to assist with the absorption of other bioactive agents generally, and in particular, alkaloids, especially the absorption of inhaled base alkaloids.
  • alkaloids especially the absorption of inhaled base alkaloids.
  • base alkaloids in the form of those with pka of pH 6 or more.
  • Alkaloids are produced by a large variety of organisms, including bacteria, fungi, plants, and animals, and are part of the group of natural products (also called secondary metabolites). Many alkaloids can be purified from crude extracts by acid-base extraction. Many alkaloids are toxic to other organisms. They often have pharmacological effects and are used as medications, as recreational drugs, or in entheogenic rituals.
  • Examples are the local anesthetic and stimulant cocaine; the psychedelic psilocin; the stimulant caffeine; nicotine; the analgesic morphine; the antibacterial berberine; the anticancer compound vincristine; the antihypertension agent reserpine; the cholinomimeric galatamine; the spasmolysis agent atropine; the vasodilator vincamine; the anti-arhythmia compound quinidine; the anti-asthma therapeutic ephedrine; and the antimalarial drug quinine.
  • alkaloids act on a diversity of metabolic systems in humans and other animals, they almost uniformly invoke a bitter taste.
  • Alkaloids are often divided into the following major groups: “True alkaloids”, which contain nitrogen in the heterocycle and originate from amino acids. Their characteristic examples are atropine, nicotine, and morphine. This group also includes some alkaloids that besides nitrogen heterocycle contain terpene (e.g., evonine) or peptide fragments (e.g. ergotamine). This group also includes piperidine alkaloids coniine and coniceine although they do not originate from amino acids; “Protoalkaloids”, which contain nitrogen and also originate from amino acids.
  • Examples include mescaline, adrenaline and ephedrine; “Polyamine alkaloids”-derivatives of putrescine, spermidine, and spermine; “Peptide and cyclopeptide alkaloids; and “Pseudalkaloids”—alkaloid-like compounds that do not originate from amino acids. This group includes, terpene-like and steroid-like alkaloids, as well as purine-like alkaloids such as caffeine, theobromine and theophylline. Some authors classify as pseudoalkaloids such compounds such as ephedrine and cathinone. Those originate from the amino acid phenylalanine, but acquire their nitrogen atom not from the amino acid but through transamination.
  • the absorption conditioning unit does not contain a “drug” as defined hereinafter.
  • the absorption conditioning unit does not contain drugs, antigenics, antibodies, tobacco, or cosmaceuticals.
  • the absorption conditioning unit does not contain any bioactive agent.
  • bioactive agent shall mean any pharmaceutical, antigenic, antibody, botanical, tobacco, food, nutraceutical, or cosmaceutical.
  • Examples of pharmaceutical bioactive agents include, but are not limited to ace inhibitors, such as Benazepril, Captopril, Enalapril, Lisinopril, Moexipril, Perindopril, Quinapril, Ramipril and Trandolapril; acne treatments, such as adapalene, azelaic acid, BenzaClin, Benzamycin, Benzoyl Peroxide, clindamycin, Duac, Erythromycin, Glycolic Acid, Isotretinoin, Sulfacetamide with sulfur, Tazarotene and Tretinoin; actinic keratosis, such as declofenac, fluorouracil; addiction aids, such as buprenorphine, Disulfiram, Naltrexone, Suboxone and varenicline; aldosterone antagonists, such as eplerenone and spironolactone; alpha-1 adrenergic blockers,
  • Examples of food or nutraceutical bioactive agents include, but are not limited to, constituents in foods or dietary supplements that are responsible for changes in health status, such as components of plants, especially fruits and vegetables, e.g., soy which contains isoflavones and phytoestrogens, tomatoes which contain lycopene that may have anticancer properties, berries such as blueberries and raspberries which contain flavonoids like anthocyanins that may act as antioxidants, green tea which contains epigallocatechin gallate (EGCG) that may have anticancer properties, resveratrol from red grape products as an antioxidant, soluble dietary fiber products, such as psyllium seed husk for reducing hypercholesterolemia, broccoli (sulforaphane) as a cancer preventative, and soy or clover (isoflavonoids) to improve arterial health.
  • constituents in foods or dietary supplements that are responsible for changes in health status, such as components of plants, especially fruits and vegetables, e.g., soy which contains isofla
  • antigen bioactive agents include, but are not limited to exogenous antigens, endogenous antigens, autoantigens and tumor antigens.
  • Exogenous antigens are antigens that have entered the body from the outside, for example by inhalation, ingestion, or injection. By endocytosis or phagocytosis, these antigens are taken into the antigen-presenting cells (APCs) and processed into fragments. APCs then present the fragments to T helper cells (CD4 + ) by the use of class II histocompatibility molecules on their surface.
  • APCs antigen-presenting cells
  • T helper cells CD4 +
  • Some T cells are specific for the peptide:MHC complex. They become activated and start to secrete cytokines.
  • Cytokines are substances that can activate cytotoxic T lymphocytes (CTL), antibody-secreting B cells, macrophages, and other particles. Endogenous antigens are antigens that have been generated within the cell, as a result of normal cell metabolism, or because of viral or intracellular bacterial infection. The fragments are then presented on the cell surface in the complex with MHC class I molecules. If activated cytotoxic CD8 + T cells recognize them, the T cells begin to secrete various toxins that cause the lysis or apoptosis of the infected cell. In order to keep the cytotoxic cells from killing cells just for presenting self-proteins, self-reactive T cells are deleted from the repertoire as a result of tolerance (also known as negative selection).
  • An autoantigen is usually a normal protein or complex of proteins (and sometimes DNA or RNA) that is recognized by the immune system of patients suffering from a specific autoimmune disease. These antigens should, under normal conditions, not be the target of the immune system, but, due to mainly genetic and environmental factors, the normal immunological tolerance for such an antigen has been lost in these patients.
  • Tumor antigens or Neoantigens are those antigens that are presented by MHC I or MHC II molecules on the surface of tumor cells. These antigens can sometimes be presented by tumor cells and never by the normal ones.
  • TSAs tumor-specific antigens
  • TAAs tumor-associated antigens
  • Cytotoxic T lymphocytes that recognize these antigens may be able to destroy the tumor cells before they proliferate or metastasize.
  • Tumor antigens can also be on the surface of the tumor in the form of, for example, a mutated receptor, in which case they will be recognized by B cells.
  • Examples of botanical bioactive agents include, but are not limited to PMI-004 (advanced botanical formulation for type II diabetes that represents a multi-mechanism bioactive that: 1) in adipocytes increases adiponectin secretion, 2) in the liver lowers PEPCK expression, and 3) in muscle cells increases cellular signaling through the insulin receptor pathway, increasing glucose uptake, glycogen synthase, and glycogen accumulation), PMI-005 (botanical bioactive, derived from a common vegetable, that inhibits gene expression of a variety of pro-inflammatory cytokines (including a-TNF, i-NOS, IL-1b, and COX-2) that are currently undergoing a human clinical trial in osteoarthritis may also may have utility in the management of severe/life threatening inflammatory conditions, such as in the management of the septic patient), PMI-006 (botanical bioactive, derived from a spice, that inhibits a range of inflammation-related enzymes (including a-TNF and COX-
  • drug as it is defined in The Food, Drug and Cosmetic Act, which defines a drug as follows.
  • drug means (A) articles recognized in the official United States Pharmacopoeia, official Homoeopathic Pharmacopoeia of the United States, or official National Formulary, or any supplement to any of them; and (B) articles intended for use in the diagnosis, cure, mitigation, treatment, or prevention of disease in man or other animals; and (C) articles (other than food) intended to affect the structure or any function of the body of man or other animals; and (D) articles intended for use as a component of any article specified in clause (A), (B), or (C).
  • a food or dietary supplement for which a claim, subject to sections 343(r)(1)(B) and 343(r)(3) of this title or sections 343(r)(1)(B) and 343(r)(5)(D) of this title, is made in accordance with the requirements of section 343(r) of this title is not a drug solely because the label or the labeling contains such a claim.
  • a food, dietary ingredient, or dietary supplement for which a truthful and not misleading statement is made in accordance with section 343(r) (6) of this title is not a drug under clause (C) solely because the label or the labeling contains such a statement.”
  • the definitions contained in the Food, Drug and Cosmetic Act as amended and in effect from time to time are hereby incorporated by reference as if fully set forth herein.
  • confection formulations are not suitable in connection with the present invention.
  • confection formulations are typically acidic to provoke additional salivary response. This is good for a juicy candy but is the opposite pH direction to promote nicotine absorption.
  • the absorption conditioning unit of the present invention can be packaged and sold in a kit together with vapor devices and vapor compositions.
  • Vapor compositions can be any form of nicotine, tobacco, marijuana or base alkaloid suitable for vaporization.
  • the vapor device can be components useful for doing so—as simple as a candle or other basic heat source, conventional e-cigarette components, or other personal vaporizer.
  • a soft lozenge was made comprising 300 mg of sodium bicarbonate together with carbowax sentry PEG 1450.
  • the resulting lozenge weighted approximately one gram, had harmless taste and was easily chewable.
  • the lozenge of this example was provided to a healthy female volunteer, and the pH of her saliva was measured at intervals over a period of ten minutes.
  • t0 saliva pH was 6.8; t1 was 8.3; t3 was 8.3; t5 was 8.3 and t10 was 8.1.
  • pH measurements were taken using a Horiba compact portable meter.
  • a healthy male volunteer experienced with nicotine placed approximately 400 mg of sodium bicarbonate in a hot tea cup, and stirred the liquid.
  • a liquid was prepared by adding the following ingredients in the following order: Sodium bicarbonate (NaHCO 3 ) 3.35 g; tablet salt 0.315 g; tap water 16.11 g; and glycerin 11.38 g of glycerin.
  • NaHCO 3 Sodium bicarbonate
  • tablet salt 0.315 g
  • tap water 16.11 g
  • glycerin 11.38 g of glycerin.
  • the liquid of this example was provided to a healthy female volunteer (one 1 ⁇ 2 teaspoon was swirled in the mouth without swallowing), and the pH of her saliva was measured at intervals over a period of ten minutes.
  • T0 pH was 6.5; T1 was 8.5; T3 was 8.7; T4 was 8.2; T8 was 7.5 and T10 was 7.6.
  • Example D employed the same ingredients as Example C but they were mixed in a different order. Glycerin was added first, then solids, mixed together and then water added. The solution was mixed and 1 ⁇ 2 teaspoon swirled in the mouth without swallowing, and the pH of her saliva was measured at intervals over a period of ten minutes.
  • T0 pH was 6.5; T1 was 8.4; T2 was 8.5; T3 was 7.9; T5 was 7.9; T7 was 7.5 and T10 was 7.3.
  • This example used the same ingredients as the preceding Examples C and D but eliminated water.
  • a solution was made with three grams of sodium bicarbonate, 6 grams of glycerin and no water. To test the solution, a health female volunteer swished it in the mouth and expectorated.
  • T1 was 8.2; T2 was 8.7; T3 was 8.6; T4 was 8.6; T5 was 8.6; T7 was 8.0 and T10 was 7.2.
  • T0 pH was 6.5 and T1 as 7.2. Because of disagreeable taste, the test was ended after T1 timepoint.
  • Xantham was selected as a capture agent in view of its mucoadhesive properties. As an additional benefit it is relatively insensitive to pH.
  • xantham/glycerin/water gel were blended with baking soda by volume at 2:1 liquid/powder. A 1 ⁇ 2 teaspoon was taken orally, without swallowing and swirled in mouth for several seconds. The following pH measurements were taken over a ten minute period.
  • T0 was 6.5; T1 was 8.4; T2 was 8.4; T3 was 8.6; T4 was 8.4; T5 was 8.4; T6 was 8.4; T7 was 8.4; T8 was 8.4; T9 was 8.1 and T10 was 8.2.
  • T0 was 6.6; T1 was 8.1; T2 was 8.3; T3 was 8.3; T4 was 8.5; T5 was 8.5; T6 was 8.5; T7 was 8.4; T8 was 8.3; T9 was 8.3 and T10 was 8.1.
  • T0 was 6.2; T1 was 6.3; T2 was 6.6; T8 was 6.7 and T10 was 6.5.
  • Listerine® film pouch with 0.2 g sodium bicarbonate was tested by a healthy female volunteer (115 lbs).
  • T0 was 6.5; T1 was 8.5; T3 was 8.7; T5 was 8.7; T8 was 8.5 and T10 was 8.5.
  • the product was pleasant to the taste.
  • the sodium bicarbonate was reduced to 0.1 grams and used by second tester (180 lb male).
  • T0 was at 6.4; T1 was at 8.1; T2 was at 8.6; T3 was at 8.6; T4 was at 8.4; T7 was at 8.4; T9 was at 8.1 and T10 was at 7.7.
  • T0 was 7.35; T1 was 6.23; T2 was 6.35; T4 was 6.14 and T5 was 6.57.
  • Briosch® was used with 200 ml of water at the recommended dosage and the pH was measured over ten minutes as follows.
  • T0 was 6.81; T(10 s) was 4.41; T(30 s) was 4.95; T1 was 5.45; T4 was 5.47; T6 was 5.52 and T10 was 5.52.
  • the Briosch® was more acidic than the Alka Seltzer®.
  • T0 was at 6.4; t(30 s) was at 2.9; T1 was at 3.8; T2 was at 5.4; T3 was at 6.5 and T4 was at 6.5. While a very low initial pH was achieved, washout was very rapid and the mix generated a high volume of saliva.
  • a soft chew was prepared with the following composition: 6 grams of sodium bicarbonate (29%); 11 grams of Carbowax 1450 (54%); 1 gram of HPMC (5%); 1 gram of powdered sugar (5%); 1 gram of glycerin (5%); 0.5 ml mint extract (2%); and 1 drop of red food coloring.
  • the resulting soft chew was tested together with Nicotrol® by a healthy volunteer and found to be quite effective for improving nicotine delivery.
  • a gummy was prepared with the following composition: 6 grams of sodium bicarbonate (14%); 20 grams of glycerin (47%); 12 grams of water (28%); 4 grams of unflavored gelatin (9%); 1 g of corn syrup (2%) and 1 drop of red food coloring.

Abstract

A method for the administration of nicotine, THC, tobacco, cannabidiol or a base alkaloid includes administering an absorption conditioning unit having at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, to the mammal, and then administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, cannabidiol and a base alkaloid. The absorption conditioning unit may be in a dosage form not containing a drug. The absorption conditioning unit may create a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration, the dosage form not containing an acid and not containing a drug.

Description

    BACKGROUND
  • Electronic vapor devices are increasingly popular with consumers. Such devices comprise electronic cigarettes, electronic pipes, electronic hookahs, personal vaporizers and other inhalator embodiments, both electronic and mechanical.
  • UBS estimates that US e-cigarette sales will reach $500 million by the end of 2012, representing 100% growth from the preceding year (See UBS Tobacco Analyst Nik Modi's May 14, 2012 report entitled “Clearing the Smoke on E-Cigarettes,” the entirety of which is incorporated herein by reference). Arguably, the most well-respected tobacco analyst, Bonnie Herzog of Wells Fargo, has gone so far as to suggest in discussing Lorillard: “We remain very encouraged by blu [Lorillard's e-cigarette brand] and see huge upside potential given our belief ecigs could overtake traditional cigarettes in 10 years.” See Wells Fargo Lorillard Equity Research dated Oct. 24, 2012, the entirely of which is incorporated herein by reference. British American Tobacco Chief Financial Officer Nicandro Durante stated in an interview with the Financial Times in September 2012 that the size of the market for tobacco alternatives like the e-cigarette could account for as much as 40% of BAT's revenues (which were £15bn in 2011) in 20 years' time. “It will be sizeable in 20 years' time . . . it's going to grow,” he said.
  • Despite this rapid sales growth, it is understood that electronic cigarettes fail to deliver a steeply peaked blood plasma delivery of nicotine like the conventional cigarette does.
  • Still, there are good reasons for using the e-cigarette as compared with cigarettes. The level of toxicants from the electronic cigarette is a small fraction as compared with a conventional cigarette. See, e.g., John H. Lauterbach, Murray Laugesen, James D. Ross, “Suggested protocol for estimation of harmful and potentially harmful constituents in mainstream aerosals generated by electronic delivery systems (ENDS)”, SOT, San Francisco, Calif., Mar. 10-16, 2012 (http://cigtoxdoc.ehost-services113.com/sot2012poster1860aspresented.pdf) hereby incorporated by reference).
  • It is typically considered in the industry that electronic vapor device adoption by consumers would be enhanced by faster, improved nicotine delivery from an electronic vapor device. Thus, there is a need to improve nicotine delivery from an electronic vapor device to its user. There is in addition need for delivery improvement (faster and more efficient) in connection with the use of all vapor devices (mechanical and electronic), of any nicotine, tobacco, or marijuana actives.
  • SUMMARY OF THE INVENTION
  • The present invention relates, inter alfa, to a method for the administration of nicotine, THC, tobacco, cannabidiol or a base alkaloid to a mammal, e.g., a human. The method includes administering an absorption conditioning unit, comprising at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, to the mammal, and then administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, cannabidiol and a base alkaloid.
  • The present invention also relates A method for the administration of nicotine, THC, cannabidiol or a base alkaloid to a mammal, including administering an absorption conditioning unit including at least at least one material that creates a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration; and then administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, tobacco, cannabidiol and a base alkaloid.
  • In one aspect of the present invention, an absorption conditioning unit includes a dosage form including at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, wherein said absorption conditioning unit (i) does not have an essentially acidic character, (ii) does not contain a drug, (iii) does not contain THC or other Marijuana derived active ingredient, and (iv) does not contain tobacco except for trace amounts in flavor.
  • In another aspect of the present invention, an absorption conditioning unit, includes a dosage form including at least at least one material that creates a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration, the dosage form not containing an acid and not containing a drug.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to a method for the administration of nicotine, THC, cannabidiol or a base alkaloid to a mammal, especially to a human, and to a prior administered absorption conditioning unit useful in such a method.
  • Nicotine inhalers are known art. For example, Pfizer markets Nicotrol® in the United States. The Nicotrol® inhaler consists of a mouthpiece and a plastic cartridge delivering 4 mg of nicotine from a porous plug containing 10 mg nicotine. The cartridge is inserted into the mouthpiece prior to use. Nicotine is released when air is inhaled through the inhaler. No heating element is used—the product relies on the natural propensity of nicotine to sublime at room temperature.
  • Despite the inhalation-route of administration by the user of Nicotrol® a buccal route of absorption is described in the Nicotrol® package insert despite the user's inhalation of nicotine from the product (the entire package insert for Nicotrol® is incorporated herein by reference as if fully set forth).
  • The Nicotrol® package insert describes its method of action: “Most of the nicotine released from the NICOTROL Inhaler is deposited in the mouth. Only a fraction of the dose released, less than 5%, reaches the lower respiratory tract. An intensive inhalation regimen (80 deep inhalations over 20 minutes) releases on the average 4 mg of nicotine content of each cartridge of which about 2 mg is systemically absorbed. Peak plasma concentrations are typically reached within 15 minutes of the end of inhalation . . . . Absorption of nicotine through the buccal mucosa is relatively slow and the high and rapid rise followed by the decline in nicotine arterial plasma concentrations seen with cigarette smoking are not achieved with the inhaler. After the use of the single inhaler the arterial nicotine concentrations rise slowly to an average of 6 ng/mL in contrast to those of a cigarette, which increase rapidly and reach a mean Cmax of approximately 49 ng/mL within five minutes . . . . Ad libitum use of the NICOTROL Inhaler typically produces nicotine plasma levels of 6-8 ng/mL, corresponding to about ⅓ of those achieved with cigarette smoking.”
  • The discrepancy between 4 mg of nicotine leaving the cartridge with just 2 mg being absorbed is noted. One source of this discrepancy is that much of the nicotine is not buccally absorbed and is swallowed where nicotine is only poorly absorbed in the GI tract.
  • Now, the typical cigarette contains less than 2 mg of nicotine (http://www.bigsixsmokes.com/Camel.shtml), yet delivers a far higher peak systemic nicotine plasma concentration (Cmax), far quicker (Tmax), than Nicotrol® (see http://pharmrev.aspetjournals.org/content/57/1/79/T1.expansion.html which is incorporated by reference). We postulate that the more efficient delivery of nicotine from a cigarette (as compared with the Nicotrol®) is a result of enhanced motility of nicotine occurring in smoke—which results from the burning of a cigarette. This superior motility allows the nicotine to readily reach the intermediate and deep lung where the enormous surface area of the lung promotes rapid absorption (as compared with buccal delivery).
  • The aerosol composition from a smoked cigarette is described by British American Tobacco at http://www.bat-science.com/groupms/sites/bat7awfh3.nsf/vwPagesWebLive/DO7AXGHN?opendocument&SKN=1 (the entirety of which is incorporated by reference, including all links).
  • Now, the obvious solution to improve nicotine delivery to a human would simply be to smoke a cigarette but for the fact that it is understood that burning tobacco results in carcinogens, and it is not yet known how to effectively filter such carcinogens prior to inhalation. Such filtration is likely not possible.
  • Others, such as Professor Jed Rose have tried to improve nicotine delivery from inhalers by using new nicotine salts (nicotine pyruvate) and thereby obtaining faster nicotine absorption than the Nicotrol®—though still slower (Tmax) and not as high (Cmax) as a cigarette. See http://www.eurekalert.org/pub releases/2010-02/dumc-nsc022210.php, http://tobaccofreeaz.wordpress.com/2011/05/27/new-nicotine-cigarette-gives-rapid-lung-delivery-of-nicotine/ and US Patent Application Publication No. 2012/0006342 A1 (“Tobacco-based aerosol generation system), each of which (together with references therein) is fully incorporated herein by reference as if fully set forth.
  • While US Patent Application Publication No. 2012/0006342 A1 suggests the use of a gaseous carrier, the poster presentation appears to suggest no additional carrier is used. We believe nicotine pyruvate has enhanced motility that allows the nicotine to more readily reach the lung as compared with the active ingredient of NICOTROL®.
  • However, Professor Rose's innovation is not directed towards use with existing electronic vapor devices such as an electronic cigarette, and poses its own challenges for commercialization and pharmacokinetic performance.
  • Now let us discuss the nicotine based electronic cigarette. The effluent vapor of the electronic vapor device like an e-cigarette is heavy and slow moving. Thus, it does not reach the deep lung in substantial part, akin to the NICOTROL® device. The result is pharmacokinetic performance much closer to Nicotrol® than to a cigarette.
  • Published studies on nicotine update from electronic cigarettes similarly show a substantially lower Cmax and slower Tmax for nicotine delivery than a cigarette does (See e.g. Electronic Cigarettes: Effective Nicotine Delivery After Acute Administration by Andrea Vansickel and Thomas Eissenberg, Nicotine and Tobacco Research Adances Access published Feb. 6, 2012; Clinical Laboratory Assessment of the Abuse Liablity of an Electronic Cigarette” by Andrea Vansickel, Michael Weaver and Thomas Eissenberg submitted 14 Oct. 2011 to the Society for the Study of Addiction, and available online; each of which is specifically incorporated herein by reference).
  • Now, it has been suggested on electronic cigarette forums (www.e-cigarette-forum.com) to enhance nicotine delivery by chewing a TUMS® antacid by well know e-cigarette blogger “Tropical Bob” but he states that this method does not work (“I concur that e-liquid is NOT alkaline enough. I've even tried chewing Tums to lower mouth acidity and help absorption. Didn't help and messed up the flavor of the vapor. The only solution is less-acid liquid. This stuff is mostly absorbed by the mouth, according to the Health New Zealand studies, and virtually none is absorbed from the lungs. So get to work, e-liquid makers.”) We demonstrate in the examples below why a TUMS® is ineffective. However, Tropical Bob's suggestion to focus on the e-cigarette's liquid composition is misguided.
  • What is however correct is that nicotine is more readily absorbed across the oral mucosa at a basic pH. The question is how to achieve this, and modifying the pH of the liquid composition in an e-cigarette will not produce substantial results.
  • It is of course possible to make a less-acidic (or alkaline) liquid for use in a vapor device—however, the delivery of a less-acidic (or alkaline) condensate (i.e. the condensate that forms when the vapor cools in the mouth of the user) is challenging. Simply stated, placing buffer agents in a liquid and then vaporizing them offers little control over the pH of the condensate that forms in the mouth as well as the pH of the saliva in the mouth, which bathes the mucosa and is very important in this process. If we add NaHCO3 (sodium bicarbonate) to water: HCO3-(aq)+H2O CO32-(aq)+H3O+(aq) Ka=4.7×10-11; HCO3-(aq)+H2O H2CO3 (aq)+OH-(aq) Kb=2.3×10-8. The solution will be alkaline. But if we vaporize the water, the vapor would not be 7.0. A complicated reaction is provoked and accelerated by the higher temperatures used to vaporize.
  • Similarly, it is not practicable to deliver sufficient amounts of capture agents (described below) or penetration enhancers (described below) or thermal agents (described below) via a vapor condensate (or comparable effluent from a non-vapor inhalator).
  • The present invention seeks to enhance the buccal delivery of nicotine (or Marijuana bioactives or any basic alkaloids) from an electronic (or non-electronic) vapor (or other inhalation) device. Because of the nature of vapor effluent, there is a limit to the agents and excipients that may be employed in the composition in the vapor device that will travel with the vapor into the user's buccal cavity and oral mucosa. Similarly, absorption can be enhanced for the oropharyngial mucosa.
  • The present invention contemplates an “absorption conditioning unit” that is placed in the user's mouth prior to and/or contemporaneously with, the use of the electronic vapor device (or other inhalator).
  • The absorption conditioning unit is a typically a dosage unit customarily used in the pharmaceutical and/or confection industries. The absorption conditioning unit may comprise any of following non-limitative solid or liquid examples: a tablet, an orally dissolving tablet, a chewable tablet, a multi-layered tablet, a capsule, a liquid-gel, a lozenge, a thin film, a sheet, a slab, a powder, granules, a gummy, a lollipop, a suspension, a toothpaste, a chewing gum or a solution. A solution or suspension may be delivered via a spray or aerosol or like method. However, the absorption conditioning unit will preferably comprise a solid dosage unit. Chewable dosage units are a preferred embodiment for their propensity to spread material around the mouth. Film dosage units are desirable because of small sized and their ability to be easily co-packaged with the inhalation device. For purposes of this invention, we would like to increase the amount of modified mucosal surface that is exposed to the agents contained in the absorption conditioning unit. Liquids are also good for this purpose but liquids are inevitably costly to distribute. One exception to this, and another preferred embodiment, is a liquid that is constituted by the user whereby dry or concentrated agent-materials are provided to the user who then mixes them with a liquid like for example tap water and used as a an oral gargle and/or oral swishing agent.
  • The emission from an electronic cigarette is a vapor—formed by heating a liquid (or primarily) liquid composition that is brought into contact with the heating element of the electronic cigarette. Said liquid composition typically comprises a vapor agent that creates a smoke-like vapor when heated (most commonly glycerin or propylene glycol), USP nicotine, water and a flavor system. Said liquid may alternatively comprise a tobacco extract together with a vapor agent (see US 2012/0145170 A1 “METHOD FOR PREPARING TOBACCO EXTRACT FOR ELECTRONIC SMOKING DEVICES” and hereby incorporated by reference). Rate of flow of the liquid composition to the heating element is typically controlled with a wick or sponge like material such that the heating element is not overwhelmed. Drawing on the e-cigarette turns on the heating element (which can be achieved through a variety of ways in including air pressure differentials, a sound-based system and other switches).
  • The effluent vapor in large part forms a condensate in the mouth—it is too heavy to readily travel to the middle or deep lung. It is understood that nicotine is absorbed across mucosal surfaces (and even the skin) but absorbed very poorly in the GI tract. Thus, to the extent condensate forms in the mouth that is in turn swallowed, the nicotine so swallowed will not reach the blood but rather will be excreted. This explains much of the discrepancy noted above for Nicotrol® whereby 4 mg of nicotine leaves the cartridge and only 2 mg is absorbed. Loss of nicotine to salivary flow is discussed in Fuisz U.S. Patent Application Publication No. 2009/0098192 A1 and Fuisz U.S. Pat. No. 8,241,661 discussing salivary flow, the content of each of which is hereby incorporated by reference in its entirety.
  • The ability of particles to transit to the lung has been considered in the context of pulmonary drug delivery for asthma and related conditions. “Depending on their particle size, inhaled drug particles will deposit in different regions of the lung. Particles <1 μm are likely to reach the peripheral airways and alveoli or be exhaled, particles 1-5 μm will deposit in the large and conducting airways, while particles >5 μm will predominately deposit in the mouth and oropharynx.” (Inhaler Technique and Training in People With Chronic Obstructive Pulmonary Disease and Asthma: Effects of Particle Size on Lung Deposition (CME) Toby G. D. Capstick, MRPharmS; Ian J. Clifton, MD). Of course, asthma inhalers typically employ a propellant to further help delivery the particles to the lung. E-cigarettes, personal vaporizers and nicotine inhalers do not have the benefit of propellant but rather rely on the pressure differentials created by the action of the lung itself. While there is some difference of views relative to the particle size of water contained in a water vapor, some argue that droplets in the 2-5 micron range can constitute a vapor. There is not rigid distinction between vapors and gases. Vapors are gases, that is, consist of non-associated molecules. Generally speaking, a gas is considered a vapor when its standard state at that pressure and temperature is a liquid or a solid. So, for example, gaseous water at less than 100° C. is usually called a water vapor. The trick, of course, is that the partial pressure of the water vapor is less than one atmosphere. However more importantly, condensation occurs which results in sizes that are too large to operationally do much other than deposit in the mouth, oral pharynx and nasal pharynx.
  • We teach herein the use of a capture agent, which we define as a material that can (a) temporarily bind condensate and (b) maintain the condensate in contact with mucosal surfaces for a sustained period to enable absorption. This will allow for increased absorption of nicotine (or other actives as discussed herein). By “sustained period,” we mean 0.5 to 90 minutes, preferably 3 to 10 minutes.
  • The capture agent is contained in the absorption conditioning unit where it is released into the mouth. Such release is preferably controlled over a sustained period to ensure availability of the capture agent over a prolonged period.
  • An additional observation is important here. When a smoker smokes a cigarette, the nicotine uptake in the lung is so rapid and efficient that the loss of nicotine in effluent smoke that is exhaled cannot be seen as a negative aspect of the cigarettes essential pharmacological function. Indeed, the presence of nicotine in such exhaled smoke is has been well studied particularly in research around real or perceived second-hand smoke issues.
  • There is little doubt that the smoke-like vapor of the e-cigarette (and other vaporizers) is a critical component to consumer acceptance. The inventors are aware of no commercially marketed e-cigarette that does not comprise a vapor agent (glycerin is rapidly becoming the vapor agent of choice). What is interesting here is the dramatic commercial success of the e-cigarette as compared with Nicotrol®—a product which has a comparable pharmacokinetic performance for the user. The inventors postulate that the cigarette-like experience of the e-cigarette makes the user more relaxed and patient for the slower nicotine delivery of the e-cigarette (as compared with a cigarette). Now, that observation notwithstanding, the e-cigarette would no doubt be even more successful with better nicotine delivery.
  • In the context of the e-cigarette and its relatively slow nicotine delivery, any nicotine in exhaled vapor is essentially lost nicotine that was actually needed to improve the product (perhaps unlike the cigarette). This is why the capture agent is so important—to capture nicotine and maintain it in buccal or sublingual contact for effective absorption.
  • Film formers are, without limitation, useful as a capture agent because the condensate may have solvent activity on condensate, on the one hand, and two, many film agents tend to have muco-adhesive properties that will help maintain the nicotine with mucosal surfaces. Likewise, sugars are useful for this purpose. Other bioadhesive agents may be useful. The film forming polymers described and/or listed in U.S. Pat. No. 7,824,588 (the contents of which are incorporated herein by reference) may be useful. The polymer 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 or a cellulose derivative. Specific examples of useful water soluble polymers include, but are not limited to, pullulan, hydroxypropylmethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, polyvinyl pyrrolidone, carboxymethyl cellulose, polyvinyl alcohol, sodium aginate, polyethylene glycol, xanthan gum, tragancanth gum, guar gum, acacia gum, arabic gum, polyacrylic acid, methylmethacrylate copolymer, carboxyvinyl copolymers, starch, gelatin, and combinations thereof. Specific examples of 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.
  • As used herein the phrase “water soluble polymer” and variants thereof refers 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. Desirably, the water soluble polymers are water soluble or water swellable having at least 20 percent by weight water uptake. Water swellable polymers having a 25 or greater percent by weight water uptake are also useful. Films or dosage forms of the present invention formed from such water soluble polymers are desirably sufficiently water soluble to be dissolvable upon contact with bodily fluids.
  • Other polymers useful as capture agents include biodegradable polymers, copolymers, block polymers and combinations thereof. Among the known useful polymers or polymer classes which meet the above criteria are: poly(glycolic acid) (PGA), poly(lactic acid) (PLA), polydioxanoes, polyoxalates, poly(.alpha.-esters), polyanhydrides, polyacetates, polycaprolactones, poly(orthoesters), polyamino acids, polyaminocarbonates, polyurethanes, polycarbonates, polyamides, poly(alkyl cyanoacrylates), and mixtures and copolymers thereof. 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 of polyurethane and (poly(lactic acid), 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 and mixtures thereof. Binary and ternary systems are contemplated.
  • Other specific polymers useful include those marketed under the Medisorb and Biodel trademarks. The Medisorb materials are marketed by the Dupont Company of Wilmington, Del. and are generically identified as a “lactide/glycolide co-polymer” containing “propanoic acid, 2-hydroxy-polymer with hydroxy-polymer with hydroxyacetic acid.” Four such polymers include lactide/glycolide 100 L, believed to be 100% lactide having a melting point within the range of 338-347° F. (170-175° C.); lactide/glycolide 100 L, believed to be 100%, glycolide having a melting point within the range of 437-455° F. (225-235° C.); lactide/glycolide 85/15, believed to be 85% lactide and 15% glycolide with a melting point within the range of 338-347° F. (170-175° C.); and lactide/glycolide 50/50, believed to be a copolymer of 50% lactide and 50% glycolide with a melting point within the range of 338-347° F. (170-175° C.).
  • The Biodel materials represent a family of various polyanhydrides which differ chemically.
  • The sugars and preferred combinations of/with sugars described and/or listed at U.S. Pat. No. 5,935,600 (the contents of which are incorporated herein by reference), may, without limitation, be useful as capture agents. For example, “sugars” are those substances which are based on simple crystalline mono- and di-saccharide structures, i.e., based on C5 and C6 sugar structures. “Sugars” include simple sugars, e.g., glucose, sucrose, maltose, lactose, arabinose, xylose, ribose, fructose, mannose, pentose, galactose sorbose, dextrose, and sugar alcohols, e.g., sorbitol, xylitol, mannitol, pentatol, maltitol, isomalt, sucralose and mixtures of any of these. Maltodextrins may be used Maltodextrins include those mixtures of carbohydrates resulting from hydrolysis of a saccharide feedstock which are described as solids having a DE of up to and including 65. Maltooligosaccharides produced by selective hydrolysis of cornstarch followed by removal of high and low molecular weight compounds may be used. The general description of maltooligosaccharides as contemplated herein is set forth in U.S. Pat. No. 5,387,431 (the contents of which are incorporated herein by reference). Polydextrose is also contemplated for use as a capture agent. Polydextrose is a non-sucrose, essentially non-nutritive carbohydrate substitute. It can be prepared through polymerization of glucose in the presence of polycarboxylic acid catalyst and polyols. Generally, polydextrose is known to be commercially available in three forms: polydextrose A and polydextrose K, which are powdered solids, and polydextrose N supplied as a 70% solution. Each of these products also contain some low molecular weight components, such as glucose, sorbitol and certain oligomers. Regarding polydextrose, Applicants incorporate herein the contents of U.S. Pat. No. 5,279,849 (the contents of which are incorporated herein by reference).
  • The bioadhesive (and other) agents discussed and/or listed in Pranshu Tangri et al, International Journal of Biopharmaceutics, 2011, 2(1): 36-46 (the contents of which are incorporated herein by reference) may be used as capture agents.
  • Glycerin and/or propylene glycol solubility is desirable so that the capture agent will better bind the vapor condensate. Preferably, materials used as capture agents are also water soluble because that they naturally wash out of the mouth without leaving unpleasant residue. The term preferably is pointedly used since non-water soluble polymers will ultimately be washed out of the mouth and swallowed—just less readily.
  • The absorption conditioning unit may comprise a pH buffer agent (a “buffer agent”). Preferably, the buffer agent is used to raise the pH of the mouth in order to increase nicotine absorption in the buccal cavity in a manner which is based on pka and the Henderson Hasselbach equation. Preferably, the pH of the mouth is increased to 7 to 10, preferably 7.8 to 10, most preferably from 8.5 to 9.5. Preferably, the buffer agent increases the pH of the oral cavity for a period of ten minutes or more after administration
  • Buffering agents may be used to control pH, including without limitation, sodium bicarbonate, potassium bicarbonate, sodium carbonate, potassium carbonate, calcium carbonate, dipotassium phosphate, potassium citrate, sodium phosphate and any other such buffer system. The buffer system may be designed to dynamically control the pH of the product taking into consideration the effect of saliva during use, i.e., a dynamic buffer system. Examples of buffer systems to obtain the preferred pH include dibasic sodium phosphate and monobasic sodium phosphate. Both are FDA accepted buffer materials used and listed in the inactive ingredients list. For example, for a pH of 7, the ratio of monobasic/dibasic can be 4.6/8.6; for a pH of 7.5 the ratio of monobasic/dibasic can be 1.9/11.9; and for a pH of 8.0 the ratio of monobasic/dibasic can be 0.6/13.4. These are mathematically calculated buffer numbers and will need to be adjusted according to the other ingredients added to the formula. They also need to be adjusted for the length of time designed for the dissolution of the dosage unit on the buccal mucosa since saliva can be of a ph of about 6.8 but as it is made in larger amounts in the mouth the ph of saliva can sometimes become more basic. Thus this dynamic buffer range is adjusted in the dosage unit by the amounts of the buffer system since saliva is freshly renewable in the mouth. See Fuisz U.S. Patent Application Publication Nos. 2009/0098192 A1 and US 2011/0318390 A1 discussing dynamic buffering and incorporated herein by reference.
  • Alkaline buffer agents are easier to work with for sustained release pH control. Acidic agents tend to increase salivary flow which must be addressed in the formulation and practically makes formulation more difficult. The absorption conditioning unit of the present invention will generally not have an acidic character, i.e. lower the pH of water when fully dissolved therein.
  • The buffer agent may comprise a substantial portion of the absorption conditioning unit, including from 1 to 99% by mass of the dosage unit itself.
  • The absorption conditioning unit may comprise a penetration agent, i.e., a substance that enhances absorption through the mucosa, mucosal coating and epithelium (otherwise known (see U.S. Patent Application Publication No. 2006/0257463 A1, the content of which is incorporated herein by reference) as a “penetration enhancer” or “permeability enhancer”). The penetration agent may comprise but is not limited to polyethylene glycol (PEG), diethylene glycol monoethyl ether (Transcutol), 23-lauryl ether, aprotinin, azone, benzalkomin chloride, cetylperidium chloride, cetylmethylammonium bromide, dextran sulfate, lauric acid, lauric acid/propylene glycol, lysophosphatilcholine, menthol, methoxysalicylate, oleic acid, phosphaidylcholine, polyoxyethylene, polysorbate 80, sodium EDTA, sodium glycholated, sodium glycodeoxycholate, sodium lauryl sulfate, sodium salicylate, sodium taurocholate, sodium taurodeoxycholate, sulfoxides, and various alkyl glycosides or, as described in U.S. Patent Application Publication No. 2006/0257463, bile salts, such as sodium deoxycholate, sodium glycodeoxycholate, sodium taurocholate and sodium glycocholate, surfactants such as sodium lauryl sulfate, polysorbate 80, laureth-9, benzalkonium chloride, cetylpyridinium chloride and polyoxyethylene monoalkyl ethers such as the BRIJ® and MYRJ® series, benzoic acids, such as sodium salicylate and methoxy salicylate, fatty acids, such as lauric acid, oleic acid, undecanoic acid and methyl oleate, fatty alcohols, such as octanol and nonanol, laurocapram, the polyols, propylene glycol and glycerin, cyclodextrins, the sulfoxides, such as dimethyl sulfoxide and dodecyl methyl sulfoxide, the terpenes, such as menthol, thymol and limonene, urea, chitosan and other natural and synthetic polymers. Preferably, the penetration agent is a polyol, e.g., polyethylene glycol (PEG), glycerin, maltitol, sorbitol etc. or diethylene glycol monoethyl ether (Transcutol).
  • Thermal agents may also be employed. Heat increases blood flow which can in turn increase absorption. Physical agents may be employed—for example, in a liquid format the liquid may be pre-heated (like adding agents to water pre-heated for tea). Chemical thermal agents may be employed to provoke an exothermic reaction.
  • Desirably, the absorption conditioning unit further might, but not necessarily, comprise at least one flavor for more pleasant administration. Some care must go to flavor selection so that the absorption conditioning unit is pleasant yet not disruptive to the flavor delivered by the inhalation device. Tobacco flavor may be desirable as such and not require any other flavor. Appropriate flavors can be sourced from Tobacco Technologies in Eldersburg, Md. In many examples the more neutral the flavor the better so as not to interfere with the flavor of the vapor. Trace amounts of tobacco may present in flavorings, particularly for a tobacco flavored-product.
  • The absorption conditioning unit may further comprise effervescent agents to effect effervescence to enhance or improve absorption. Care must be taken that the acidic component of most effervescent systems do not interfere with the foal of a basic pH range.
  • The absorption conditioning unit may be built into the end of an electronic smoking device (or similar non-electronic inhalator) such that the user can remove it either with hands or mouth immediately prior to use. In one embodiment, this can be akin to a cigar smoker “biting off” the end of a cigar, except here the user removes the absorption conditioning unit and retains it in the mouth. It may also be possible to use a flavored liquid that is dispensed, akin to the menthol “crush” systems that are currently sold but adapted for this purpose. It may also be a dissolvable tip which conditions the pH of the saliva.
  • It is expressly contemplated that the present invention can be used to enhance the delivery of bioactives such nicotine, THC, tobacco, cannabidiol or a base alkaloid, or any combination thereof. For example, the present invention can be adapted for use with marijuana/tobacco combinations. Such combinations can involve the vaporization of tobacco and marijuana leaf, or a mixture of extracts from both materials.
  • The absorption conditioning unit of the present invention may be employed with suitable adaptation (e.g. optimize target buccal pH) to enhance the absorption of THC (tetrahydrocannabinol), cannabidiols and other active ingredients from cannabis. While the use of marijuana remains controversial, the general trend towards decriminalization and even legalization is clear. Moreover, the use of medical marijuana is growing rapidly. This takes the form of marijuana leaf, marijuana extract, and marijuana active components that are delivered via approved pharmaceuticals, like GW Pharma's Sativex®, an oral mucosa spray.
  • The vaporization of marijuana has also become popular but is less visible given the grey market status of marijuana. However, vaporization of marijuana is an increasingly common and accepted form of marijuana use. Indeed, Health Canada's September 2010 document on Marijuana entitled “Information for Health Care Professionals,” notes the benefits of vaporization of marijuana as compared with smoking marijuana:
  • “Vaporization of cannabis has been explored as an alternative to smoking. The advantages of vaporization apparently include the formation of a smaller quantity of toxic by-products such as carbon monoxide, polycyclic aromatic hydrocarbons (PAHs) and tar, as well as a more efficient extraction of THC from the cannabis material (47, 48, 49, 43, 50). The subjective effects and plasma concentrations of THC are comparable to those of smoked cannabis with absorption being somewhat faster with the vaporizer (43). The vaporizer is well-tolerated, with no reported adverse effects, and is generally preferred over smoking by most subjects (43). While vaporization is amenable to self-titration (49, 43), the proper use of the vaporizer for optimal administration of medicinal cannabis has to be established in more detail (50). The amount and type of cannabis placed in the vaporizer, the vaporizing temperature and duration of vaporization, and the balloon volume are some of the parameters that can affect the delivery of THC (49). Bioequivalence of vaporization compared to smoking has not been established.”
  • The reference to THC absorption being faster with a vaporizer is curious in view of the same document's discussion of buccal and oral routes which show that buccal absorption is relatively slow. This may be explained by the fact that the cited study employed pure THC as opposed to vaporizing marijuana plant material (or an extract therefrom).
  • “Following a single buccal administration of Sativex® (four sprays of Δ9-THC 27 mg/mL and CBD 25 mg/mL, totalling 10.8 mg Δ9-THC and 10 mg CBD), peak plasma concentrations of both THC and CBD typically occur within 2-4 h (55). When administered bucally, blood levels of THC and other cannabinoids are lower than those achieved by inhalation of the same dose of smoked cannabis because absorption is slower, redistribution into fatty tissue is rapid and some of the THC undergoes hepatic first-pass metabolism to 11-hydroxy-THC (55).”
  • “THC can be absorbed orally by ingestion of foods containing cannabis (butters, oils, brownies, cookies), teas prepared from leaves and flowering tops, or through ingestion of capsules containing THC or THC analogues. Absorption from an oral dose of 20 mg THC in a chocolate cookie was described as slow and unreliable (42), with a systemic availability of only 4 to 12% (46). While most subjects displayed peak plasma THC concentrations between 1 to 2 h, some of the 11 subjects in the study only peaked at 6 h and many had more than one peak. Consumption of cannabis-laced brownies containing 2.8% THC was associated with changes in behaviour although the effects were slow to appear and variable (51). Peak effects occurred 2.5 to 3.5 h after dosing. Modest changes in pulse and blood pressure were also noted. Tea made from dried cannabis flowering tops (19.1% THCA, 0.6% THC) has been documented, but the bioavailability of THC from such teas is likely to be smaller than that achieved by smoking (52). Only 10-20% of synthetic THC (dronabinol, Marinol®) administered in capsules with sesame oil enters the systemic circulation indicating extensive first-pass metabolism (53). The psychotropic effect or “high” occurs more quickly by the smoking than the oral route, which has been characterized by Iversen (54) as the reason “smoking is the preferred route of cannabis for many people”.”
  • The buccal and/or sublingual use of tinctures is known (See http://patients4medicalmarijuana.wordpress.com/medical-use-of-cannabis-video/marijuana-tincture/, incorporated by reference hereby).
  • Thus, the dynamic of marijuana vaporization is functionally comparable to tobacco/nicotine. In each case the actives are not effectively delivered to the deep lung but tend to be absorbed buccally. Marijuana is however different in that swallowed condensate is more apt to be absorbed in the GI tract than nicotine.
  • It is important to note that while the disclosure above discussed electronic vapor devices, it will appreciated that the present invention can equally be employed with non-electronic vapor devices. Electronic—and specifically electrical devices are the most common type of vapor device. However, vapor devices can also be “analog” like for example the butane-fired Ploom® device or the butane fired “click-a-toke.” Similarly, vapor can be generated from hookah-like devices powered by hot coals and the like. And, it is possible to have an inhaler—like Nicotrol®—that does not use any heat source. It should be noted that it is expressly contemplated that the present invention can be used with any inhaler-like device (and not solely with a vapor device).
  • Because the absorption conditioning unit does not contain tobacco or nicotine, it will generally be exempt from regulation, except where it is part of an approved pharmaceutical product (i.e. a “combination product”).
  • References to nicotine herein include any nicotine salt, nicotine that is naturally or synthetically derived, including from tobacco or any other material.
  • In addition to nicotine, tobacco and marijuana, derivatives, the present invention (i.e. the absorption conditioning unit) can be readily adapted to assist with the absorption of other bioactive agents generally, and in particular, alkaloids, especially the absorption of inhaled base alkaloids. Preferably, base alkaloids in the form of those with pka of pH 6 or more.
  • Alkaloids are produced by a large variety of organisms, including bacteria, fungi, plants, and animals, and are part of the group of natural products (also called secondary metabolites). Many alkaloids can be purified from crude extracts by acid-base extraction. Many alkaloids are toxic to other organisms. They often have pharmacological effects and are used as medications, as recreational drugs, or in entheogenic rituals. Examples are the local anesthetic and stimulant cocaine; the psychedelic psilocin; the stimulant caffeine; nicotine; the analgesic morphine; the antibacterial berberine; the anticancer compound vincristine; the antihypertension agent reserpine; the cholinomimeric galatamine; the spasmolysis agent atropine; the vasodilator vincamine; the anti-arhythmia compound quinidine; the anti-asthma therapeutic ephedrine; and the antimalarial drug quinine. Although alkaloids act on a diversity of metabolic systems in humans and other animals, they almost uniformly invoke a bitter taste.
  • Alkaloids are often divided into the following major groups: “True alkaloids”, which contain nitrogen in the heterocycle and originate from amino acids. Their characteristic examples are atropine, nicotine, and morphine. This group also includes some alkaloids that besides nitrogen heterocycle contain terpene (e.g., evonine) or peptide fragments (e.g. ergotamine). This group also includes piperidine alkaloids coniine and coniceine although they do not originate from amino acids; “Protoalkaloids”, which contain nitrogen and also originate from amino acids. Examples include mescaline, adrenaline and ephedrine; “Polyamine alkaloids”-derivatives of putrescine, spermidine, and spermine; “Peptide and cyclopeptide alkaloids; and “Pseudalkaloids”—alkaloid-like compounds that do not originate from amino acids. This group includes, terpene-like and steroid-like alkaloids, as well as purine-like alkaloids such as caffeine, theobromine and theophylline. Some authors classify as pseudoalkaloids such compounds such as ephedrine and cathinone. Those originate from the amino acid phenylalanine, but acquire their nitrogen atom not from the amino acid but through transamination.
  • In a preferred aspect of the invention, the absorption conditioning unit does not contain a “drug” as defined hereinafter.
  • In another preferred aspect of the invention, the absorption conditioning unit does not contain drugs, antigenics, antibodies, tobacco, or cosmaceuticals.
  • In another preferred aspect of the invention, the absorption conditioning unit does not contain any bioactive agent.
  • As used herein, the term “bioactive agent” shall mean any pharmaceutical, antigenic, antibody, botanical, tobacco, food, nutraceutical, or cosmaceutical.
  • Examples of pharmaceutical bioactive agents include, but are not limited to ace inhibitors, such as Benazepril, Captopril, Enalapril, Lisinopril, Moexipril, Perindopril, Quinapril, Ramipril and Trandolapril; acne treatments, such as adapalene, azelaic acid, BenzaClin, Benzamycin, Benzoyl Peroxide, clindamycin, Duac, Erythromycin, Glycolic Acid, Isotretinoin, Sulfacetamide with sulfur, Tazarotene and Tretinoin; actinic keratosis, such as declofenac, fluorouracil; addiction aids, such as buprenorphine, Disulfiram, Naltrexone, Suboxone and varenicline; aldosterone antagonists, such as eplerenone and spironolactone; alpha-1 adrenergic blockers, such as alfuzosin, doxazosin, prazosin, tamsulosin and terazosin; ALS agents, such as riluzole; Alzheimer's Disease medications, such as donepezil, Galantamine, rivastigmine, tacrine and memantine; anesthetics, such as dexmedetomidine, etomidate, ketamine, methohexital, pentobarbital, propofol and thiopental; angiotensin II receptor blockers, such as candesartan, eprosartan mesylate, irbesartan, losartan, olmesartan, telmisartin and valsartan; antacids, such as Aluminum hydroxide, AIOH and magnesium trisilicate; anti-arrhythmics, such as adenosine, amiodarone, Atropine, Bretylium, digoxin-Immune Fab, disopyramide, dofetilide, epinephrine, Esmolol, flecainide, ibutilide, isoproterenol, lidocaine, mexiletine, moricizine, procainamide, propafenone, quinidine, sotalol, tocainide and verapamil; antibiotics, such as Aztreonam, TMP/SMX, Chloramphenicol, Clindamycin, Dapsone, Daptomycin, Ertapenem, Imipenem/cilastatin, Linezolid, Meropenem, Metronidazole, Nitrofurantoin, Quinupristin/Dalfopristin, Rifaximin, Tigecycline, Telithromycin and Tinidazole; anticholinergic acids, such as Dicyclomine, Donnatal, Flavoxate, Glycopyrrolate, Hyoscyamine, Oxybutynin, Propantheline and Tolterodine; anticonvulsants, such as carbamazepine, clonazepam, diazepam, ethosuximide, felbamate, fosphenytoin, gabapentin, levetiracetam, lamotrigine, lorazepam, Oxcarbazepine, Phenobarbital, phenytoin, pregabalin, primidone, tiagabine, topiramate and valproic acid; antidepressants, such as amitriptyline, buproprion, citalopram, desipramine, doxepin, duloxetine, escitalopram, fluoxetine, fluvoxamine, imipramine, mirtazapine, nefazodone, nortriptyline, nortriptyline, sertraline, trazodone and venlafaxine; anti-diarrheals, such as dephenoxylate+atropine, Imodium and bismuth subsalicylate; anti-emetics, such as Aprepitant, dolasetron, droperidol, granisetron, metoclopramide, ondansetron, prochlorperazine, scopolamine and trimethobenzamide; antifungals, such as Ampho B, Ampho B lipid, anidulafungin, caspofungin, Clotrimazole fluconazole, flucytosine, Griseofulvin, Itraconazole, ketoconazole, Micafungin, nystatin, Posaconazole, terbinafine, voriconazole, butenafine, ciclopirox, clotrimazole, enconazole, ketoconazole, Miconazole, naftifine, nystatin, oxiconazole terbinafine and Tolnaftate; anti-hepatitis, such as adefovir, entecavir, lamivudine, peginterferon alfa-2a, peginterferon alfa-2b, Rebetron and ribavirin; anti-herpetic agents, such as Acyclovir, famciclovir, valacyclovir, acyclovir, docosanol and penciclovir; antihistamines, such as cetirizine, desloratadine, fexofenadine, loratadine, chlorpheniramine, clemastine, cyproheptadine, dimenhydrinate, diphenhydramine, hydroxzine and promethazine; anti-hypertension, such as Benazepril & HCTZ, Captopril & HCTZ, Enalapril & HCTZ, Lisinopril & HCTZ, Moexipril & HCTZ, Losartan & HCTZ, Valsartan & HCTZ, Atenolol & chlorthalidone, Bisoprolol & HCTZ, Metoprolol & HCTZ, Nadolol & bendroflumethazide, Propranolol & HCTZ, Timolol & HCTZ, Amlodipine & benazepril, Verapamil & trandolapril, Amiloride & HCTZ, Spironolactone & HCTZ, Triamterene & HCTZ, Clonidine & chlorthalidone, Hydralazine & HCTZ, Methyldopa & HCTZ and Prazosin & polythiazide; anti-hypertensives, such as Aliskiren, Aliskiren, epoprostenol, fenoldopam, hydralazine, minoxidil, nitroprusside, phentolamine and treprostinil; anti-influenza agents, such as amantadine, oseltamivir phosphate, rimantadine and zanamivir; anti-malarials/anti-protozoals/amebicides, such as Atovaquone, Chloroquine, Iodoquinol, Mefloquine, Primaquine, Pyrimethamine, Pyrimethamine-Sulfadoxine and Quinine Sulfate; anti-platelet agents, such as abciximab, dipyridamole/ASA, anagrelide, cilostazol, clopidogrel, dipyridamole, eptifabatide, ticlopidine and tirofiban; antipsychotics, such as aripiprazole, chlorpromazine, Clozapine, fluphenazine, haloperidol, loxapine, molindone, olanzepine, perphenazine, pimozide, quetiapine, risperidone, thioridazine, thiothixine, trifluoperazine, ziprasidone and Lithium; antispasmotics, such as Dicyclomine, Donnatal Extentabs, Propantheline, Simethicone, hyoscyamine, Librax, tegaserod and Bellergal-S; anti-tussives/expectorants, such as Benzonatate and guaifenesin; atopic dermatitis medications, such as pimecrolimus and tacrolimus; benzodiazepines and non-benzodiazepine sedatives, such as alprazolam, buspirone, chlordiazepoxide, chlorazepate, clonazepam, diazepam, estazolam, eszcpiclone, flurazepam, lorazepam, midazolam, Oxazepam, ramelteon, temazepam, triazolam, zaleplon and zolpidem; beta blockers, such as atenolol, betaxolol, bisoprolol, carvedilol, esmolol, labetalol, metoprolol, nadolol, pindolol, propranolol, sotalol and timolol; bile acid sequestrants, such as cholestyramine, colesevelam and colestipol; bisphosphonates, such as alendronate, etidronate, pamidronate, risedronate, tiludronate and Zoledronic acid, Raloxifene and Teriparatide; bladder spasm medications, such as flavoxate, hyoscyamine, darifenacin, oxybutynin, solifenacin, tolterodine and trospium; benign prostatic hypertrophy medications, such as alfuzosin, doxazosin, dutasteride, finasteride, tamsulosin and terazosin; burn preparations, such as mafenide acetate and silver sulfadiazine; calcium channel blockers, such as amlodipine, bepridil, diltiazem, felodipine, isradipine, nicardipine, nifedipine and nisoldipine; calcium supplements, such as Calcium and Hypocalcemia; cephalosporins, such as Cefadroxil, Cefazolin, Cephradine, Cephalexin, Cefaclor, Cefotetan, Cefoxitin, Cefprozil, Cefuroxime, Cefuroxime, loracarbef, Cefdinir, Cefixime, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxime and Cefepime; colony stimulating factors, such as darbepoietin alfa, erythropoietin, filgrastim, oprelvekin, pegfilgrastim and sargramostim; corticosteroids, such as Budesonide, cortisone acetate, dexamethasone, fludrocortisones, hydrocortisone, methylprednisolone and prednisone; corticosteroids Intra-articular, such as Depo-Medrol and Triamcinolone Acetonide; cystitis, such as pentosan polysulfate, Bethanecol and Alum irrigation; decongestants, such as Phenylephrine and Pseudoephedrine; anti-diabetic agents, such as acarbose, Miglitol and metformin, Avandamet®, Glucovance, Metaglip, Metaglip, rosiglitazone, osiglitazone, repaglinide, Chlorpropamide, glimepiride, glyburide, glipizide, Tolazamide, Tolbutamide, Glucagon, extenatide and pramlintide; direct thrombin inhibitors, such as argatroban, Bivalirudin and lepirudin; disease modifying agents, such as adalimumab, anakinra, auranofin, azathioprine, etanercept, hydroxychloroquine, infliximab, leflunomide, methotrexate and sulfasalazine; diuretics, such as Acetazolamide, Amiloride, Amiloride and HCTZ Bendroflumethiazide, Bumetanide, Chlorothiazide, Chlorthalidone, Dichlorphenamide, Eplenerone, Ethacrynic acid, Furosemide, Hydrochlorothiazide, HCTZ/Triampterene, Hydroflumethiazide, Indapamide, Methazolamide, Methyclothiazide, Methyclothiazide, Metolazone, Polythiazide, Spironolactone, Spironolactone, HCTZ Torsemide, Trichlormethiazide and Triamterene; endocrine agents, such as bromoc cinacalcet cosyntropin, riptine, cabergoline, calcitonin, desmopressin, Leuprolide, octreotide and vasopressin; erectile dysfunction agents, such as Sildenafil, tadalafil, vardenafil; fever medications, such as allopurinol, antihistamines, azathioprine, barbiturates, carbamazepine, cephalosporins, cimetidine, folic acid, hydralazine, hydroxyurea, ibuprofen, isoniazid, methyldopa, nitrofurantoin, penicillins, phenytoin, phenytoin, procainamide, prophylthiouracil, quinidine, streptomycin sulfonamides, sulindac, triamterene and vancomycin; fibrates, such as clofibrate, fenofibrat and gemfibrozil; fluoroquinolones, such as Ciprofloxacin, Gatifloxacin, Levofloxacin, Moxifloxacin, Norfloxacin and Ofloxacin; gastrointestinal agents, such as Alosetron, infliximab, Mesalamine, misoprostol, Neomycin, octreotidev, osalazine, Orlistat, sucralafate, Sulfasalazine and vasopressin; gout treatments, such as allopurinol, colchicine, probenecid, Rasburicase and sulfinpyrazone; H2 receptor blockers, such as cimetidine, famotidine, nizatidine and ranitidine; aAnti-herpetic agents, such as Acyclovir, famciclovir, valacyclovir, acyclovir, docosanol and penciclovir; hypertensive urgency, such as Captopril, Clonidine and Labetalol; hypertensive emergency, such as Enalaprilat, Esmolol, Fenoldopam mesylate, Hydralazine, Labetalol, Nicardipine, Nitroglycerin and Sodium nitroprusside; hemorrhoidal preparations, such as Anusol HC, Anusol Suppository, Dibucaine, pramoxine 1%, Proctofoam-HC and Analpram-HC; inflammatory bowel disease agents, such as balsalazide, budesonide, infliximab, mesalamine, olsalazine and sulfasalazine; Interferon, such as Interferon Alfa-2A, Interferon Alfa-2b, Interferon Alfa-2b and Ribavirin combo Pack, Interferon Alfa-N3, Interferon Beta-1A, Interferon Beta-1B (Betaseron); intermittent claudication, such as cilostazol and pentoxifylline; immunizations, such as Comvax, diphtheria-tetanus toxoid, Hepatitis A vaccine, Hepatitis B vaccine, Influenza vaccine, Fluzone, Lyme disease vaccine, PNEUMOVAX* 23; laxatives, such as Bisacodyl, Cascara, Docusate, Fleet Phospho-Soda, Glycerin, Lacalutose, lubiprostone, Magnesium citrate, Magnesium hydroxide—MOM, Mineral Oil, Pericolace, Psyllium and Senna; low molecular weight heparins, such as dalteparin, danaparoid, enoxaparin, tinzaparin, fondaparinux; macrolides, such as Azithromycin, Clarithromycin and Erythromycin; magnesium, such as magnesium salt; migraine treatments, such as almotriptan, eletriptan, frovatriptan, naratriptan, rizatriptan, sumatriptan, zolmitriptan, Cafergot®, Cafergot®, dihydroergotamine and Midrin®; mouth and lip treatments, such as amlexanox, Benzocaine, carbamide, peroxide, Kenalog in Orabase®, Phenol, chlorhexidine gluconate, clotrimazole, Nystatin, Penciclovir, docosanol, Gelclair, lidocaine viscous, BMX Cocktail, Pilocarpine and Artificial saliva; multiple sclerosis treatments, such as glatiramer, interferon beta-1A and interferon beta-1B; muscle relaxants, such as baclofen, carisprodol, cyclobenzaprine, cyclobenzaprine, Diazepam, Metaxalone, Methocarbamol, Orphenadrine; nasal preparations, such as azelastine, beclomethasone, budesonide, cromolyn, desmopressin acetate, flunisolide, fluticasone, Ipratropium bromide, mometasone, oxymetazoline, phenylephrine, Saline nasal spray, Sumatriptan, triamcinolone and Zolmitriptan; urology treatments, such as Belladonna and opium, flavoxate, hyoscyamine, hyoscyamine, oxybutynin, solifenacin, tolterodine and trospium; neuromuscular blockers, such as Atracurium, Cisatracurium, doxacurium, mivacurium, pancuronium, Rocuronium, Succinylcholine, vecuronium, Mivacurium, Rapacuronium, Rocuronium, Succinylcholine, Atracurium, Cisatracurium, Pancuronium, Vecuronium, Doxacurium, Pipecuronium and Tubocurarine; nitrates, such as Isosorbide dinitrate, Isosorbide mononitrate, Nitroglycerin ointment, Nitrobid and Nitroglycerin transdermal; NSAIDs, such as Arthrotec, diclofenac, Etodolac, indomethacin, Ketorolac, Sulindac, Tolmentin Diflunisal Salsalate Meloxicam, piroxicam, Nabumetone Flurbiprofen, Ibupropen, Ketoprofen, Naproxen, Oxaprozin, celecoxib, Rofecoxib and Valdecoxib; ophthalmic agents, such as, proparacaine, tetracaine, Ciprofloxacin, Erythromycin, Gentamcyin, levofloxacin, levofloxacin, norfloxacin, Ofloxacin, Polysporin®, Polytrim, Sulfacetamide, Tobramycin, Blephamide®, Blephamide®, Maxitrol®, Pred G® and TobraDex®, Dexamethasone, Fluorometholone, Loteprednol, Prednisone, Rimexolone, azelastine, Cromolyn sodium, emedastine, Epinastine, Ketotifen Fumarate Ophthalmic Solution 0.025%, Levocabastine, Lodoxamide tromethamine, Naphazoline, Naphcon-A®, nedocromil, Olopatadine, pemirolast, Betaxolol, Betaxolol, Levobunolol, Timolol, Brinzolamide, Dorzolamide, Pilocarpine, bimatoprost, Latanoprost, travoprost, unoprostone, Apraclonidine, Brimonidine, Cosopt® and Cosopt®, Atropine, Cyclopentolate, Homatropine, Phenylephrine, Phenylephrine, Diclofenac, Flurbiprofen and Ketorolac; ear (otic) preparations, such as Auralgan®, carbamide peroxide, CIPRODEX®, Ciprofloxacin and hydrocortisone, Cortisporin®, Ofloxacin, Triethanolamine and Vosol Otic®; opiates, such as Codeine Fentanyl Hydrocodone Hydrocodone, Meperidine Methadone, morhphine, xycodone, Propoxyphene, Darvon®, Fioricet, Fiorinal, Soma compound, Tramadol, Anexsia, Darvocet, Darvon Compound, Lorcet, Lortab, Percocet, Percodan, Roxicet, Tylenol with Codeine, Tylox, Vicodin, Wygesic, Buprenorphene, Butorphanol, Dezocine, Nalbuphine, Pentazocine, Nalmefene Naloxone, Suboxone® and Ziconotide; parkinson's disease treatments, such as amantadine, benztropine, bromocriptine, entacapone, pergolide, pramipexole, ropinirole, selegiline, Sinemet®, tolcapone and trihexyphenidyl; PCA—Patient Controlled Analgesia, such as Fentanyl, Hydromorphone, Meperidine and Morphine; penicillin's, such as Ampicillin, Ampicillin/sulbactam, Amoxicillin, Amoxicillin/Clavulanate, Cloxacillin, Dicloxacillin, Nafcillin, Penicillin G, Penicillin VK, Piperacillin, Piperacillin/Tazobactamm, Ticarcillin, and Ticarcillin/Clavulanate; phosphate supplementation, such as, K-Phos® Neutral Tablets, K-PHOS® ORIGINAL, Neutra-Phos®; potassium supplementation, such as K-LOR, Klor-Con®, Potassium depletion; prostate cancer medications, such as bicalutamide, flutamide, goserelin, leuprolide and nilutamide; proton pump inhibitor's, such as esomeprazole, Lansoprazole, Omeprazole, Pantoprazole and Rabeprazole Sodium; psoriasis medications, such as acitretin, alefacept, Anthralin, Calcipotriene, efalizumab and Tazarotene; renal failure medications, such as Aluminum Hydroxide, Calcium acetate, Calcitriol, Doxercalciferol, Ferric Sodium Gluconate, paricalcitol and sevelamer; pulmonary medications, such as ipratropium, tiotropium, albuterol, bitolterol, levalbuterol, pirbuterol, metaproterenol, formoterol, salmeterol, Advair®, Symbicort®, beclomethasone, budesonide, flunisolide, fluticasone, Mometasone furoate, triamcinolone, montelukast Singulair®, zafirlukast, cromolyn sodium, nedocromil, acetylcysteine and aminophylline/theophylline; disease modifying agents, such as adalimumab, anakinra, auranofin, azathioprine, etanercept, hydroxychloroquine, infliximab, leflunomide, methotrexate and sulfasalazine; HMG COA reductase inhibitors, such as Atorvastatin, Fluvastatin, Lovastatin, Pravastatin, Rosuvastatin, Simvastatin, Advicor®, Vytorin® and ezetimibe; stimulants, such as atomoxetine, benzphetamine, Caffeine, dexmethylphenidate, Dextroamphetamine, diethylpropion, Methylphenidate, Modafinil, Pemoline, phendimetrizine, phentermine and sibutramine; tetracyclines such as Doxycycline, Minocycline and Tetracycline; thrombolytic agents such as Alteplase; anti-thyroid agents such as methimazole and propylthiouracil; toxicology related medications such as acetylcysteine, Charcoal, deferoxamine, digoxin immune fab, flumazenil, fomepizole, methylene blue, naloxone, sodium polystyrene sulfonate and Sorbitol; anti-mycobacterial agents such as Ethambutol, Isoniazid, Pyrazinamide, rifabutin, Rifamate, Rifampin, Rifapentine and Rifater; topical products such as Alitretinoin, Becaplermin, Calamine, Capsaicin, Doxepin, lidocaine/prilocaine, fluorouracil, Masoprocol, Pimecrolimus, Selenium sulfide and Tacrolimus; topical anti-viral agents such as acyclovir, docosanol, imiquimod, penciclovir, podofilox and podophyllin; topical antibacterials such as bacitracin, metronidazole, mupirocin, bacitracin/neomycin/polymyxin, bacitracin/polymyxin and silver sulfadiazine; topical antifungals such as butenafine, ciclopirox, clotrimazole, econazole, ketoconazole, miconazole, naftifine, nystatin, oxiconazole, terbinafine and tolnaftate; topical anti-parasitic agents such as Crotamiton, Lindane, Permethrin, pyrethrins and piperonyl butoxide; topical burn preparations such as mafenide acetate and silver sulfadiazine; topical corticosteroids such as Aclometasone diproprionate, Desonide, Flucinolone acetonide, Hydrocortisone, Betamethasone dipropionate, betamethasone valerate, clocortolone pivalate, desoximetasone, fluocinolone acetonide, flurandrenolide, fluticasone propionate, Chydrocortisone butyrate, hydrocortisone valerate, mometasone furoate, prednicarbate, triamcinolone, amcinonide, augmented betamethasone dipropionate, betamethasone dipropionate, desoximetasone, diflorasone diacetate, fluocinolone acetonide, fluocinonide, halcinonide, clobetasol propionate, diflorasone diacetate and halobetasol propionate; urology medications such as pentosan polysulfate, Bethanecol and phenazopyridine; vaginal preparations such as clindamycin, metronidazole, butoconazole, clotrimazole, miconazole, terconazole and tioconazole; vasodilators such as Fenoldopam mesylate, Hydralazine, Nesiritide, Nicardipine, Nitroglycerin, and Sodium Nitroprusside; and vasopressors and inotropes such as Dobutamine, Dopamine, Epinephrine, inamrinone, Milrinone, Norepinephrine, Phenylephrine, and Vasopressin.
  • Examples of food or nutraceutical bioactive agents include, but are not limited to, constituents in foods or dietary supplements that are responsible for changes in health status, such as components of plants, especially fruits and vegetables, e.g., soy which contains isoflavones and phytoestrogens, tomatoes which contain lycopene that may have anticancer properties, berries such as blueberries and raspberries which contain flavonoids like anthocyanins that may act as antioxidants, green tea which contains epigallocatechin gallate (EGCG) that may have anticancer properties, resveratrol from red grape products as an antioxidant, soluble dietary fiber products, such as psyllium seed husk for reducing hypercholesterolemia, broccoli (sulforaphane) as a cancer preventative, and soy or clover (isoflavonoids) to improve arterial health.
  • Examples of antigen bioactive agents include, but are not limited to exogenous antigens, endogenous antigens, autoantigens and tumor antigens. Exogenous antigens are antigens that have entered the body from the outside, for example by inhalation, ingestion, or injection. By endocytosis or phagocytosis, these antigens are taken into the antigen-presenting cells (APCs) and processed into fragments. APCs then present the fragments to T helper cells (CD4+) by the use of class II histocompatibility molecules on their surface. Some T cells are specific for the peptide:MHC complex. They become activated and start to secrete cytokines. Cytokines are substances that can activate cytotoxic T lymphocytes (CTL), antibody-secreting B cells, macrophages, and other particles. Endogenous antigens are antigens that have been generated within the cell, as a result of normal cell metabolism, or because of viral or intracellular bacterial infection. The fragments are then presented on the cell surface in the complex with MHC class I molecules. If activated cytotoxic CD8+T cells recognize them, the T cells begin to secrete various toxins that cause the lysis or apoptosis of the infected cell. In order to keep the cytotoxic cells from killing cells just for presenting self-proteins, self-reactive T cells are deleted from the repertoire as a result of tolerance (also known as negative selection). They include xenogenic (heterologous), autologous and idiotypic or allogenic (homologous) antigens. An autoantigen is usually a normal protein or complex of proteins (and sometimes DNA or RNA) that is recognized by the immune system of patients suffering from a specific autoimmune disease. These antigens should, under normal conditions, not be the target of the immune system, but, due to mainly genetic and environmental factors, the normal immunological tolerance for such an antigen has been lost in these patients. Tumor antigens or Neoantigens are those antigens that are presented by MHC I or MHC II molecules on the surface of tumor cells. These antigens can sometimes be presented by tumor cells and never by the normal ones. In this case, they are called tumor-specific antigens (TSAs) and, in general, result from a tumor-specific mutation. More common are antigens that are presented by tumor cells and normal cells, and they are called tumor-associated antigens (TAAs). Cytotoxic T lymphocytes that recognize these antigens may be able to destroy the tumor cells before they proliferate or metastasize. Tumor antigens can also be on the surface of the tumor in the form of, for example, a mutated receptor, in which case they will be recognized by B cells.
  • Examples of botanical bioactive agents include, but are not limited to PMI-004 (advanced botanical formulation for type II diabetes that represents a multi-mechanism bioactive that: 1) in adipocytes increases adiponectin secretion, 2) in the liver lowers PEPCK expression, and 3) in muscle cells increases cellular signaling through the insulin receptor pathway, increasing glucose uptake, glycogen synthase, and glycogen accumulation), PMI-005 (botanical bioactive, derived from a common vegetable, that inhibits gene expression of a variety of pro-inflammatory cytokines (including a-TNF, i-NOS, IL-1b, and COX-2) that are currently undergoing a human clinical trial in osteoarthritis may also may have utility in the management of severe/life threatening inflammatory conditions, such as in the management of the septic patient), PMI-006 (botanical bioactive, derived from a spice, that inhibits a range of inflammation-related enzymes (including a-TNF and COX-2) and also possesses range of novel bioactivities related to both lipid and glucose metabolism (RXR receptors)), PMI-007 (a powerful, centrally acting, botanical appetite suppressor which acts via a unique central pathway in the nutrient-sensing hypothalamic neurons by increasing ATP content/production, possesses potent anorectic activity without typical CNS appetite suppressor side effects and pre-clinical data for which has shown that the agent suppresses both appetite and reduces weight in animal models, while there is supporting clinical evidence of human efficacy), PMI-008 (botanical bioactive, derived from an agricultural waste processing stream, that blocks fat accumulation/absorption and promotes weight loss via interaction with a variety of lipases including PL, LPL, and HSL), and PMI-016 (a powerful, plant-derived anabolic/ergogenic agent, with no androgenic side effects; could be used in a range of human muscle wasting disorders, including those associated with both cancer and AIDS, as well as general aging (sarcopenia); has been shown to induce protein synthesis in muscle cells (similar to IGF) and promote a reduction in protein degradation, while it has also been shown to increase growth hormone gene transcription and decrease in ubiquitin protein ligase gene transcription; and shows no binding to testosterone receptor in contrast to anabolic steroids). Examples of botanical bioactive agents also include tobacco and all tobacco extracts, as well as nicotine itself.
  • In the present application, we define “drug” as it is defined in The Food, Drug and Cosmetic Act, which defines a drug as follows. “The term “drug” means (A) articles recognized in the official United States Pharmacopoeia, official Homoeopathic Pharmacopoeia of the United States, or official National Formulary, or any supplement to any of them; and (B) articles intended for use in the diagnosis, cure, mitigation, treatment, or prevention of disease in man or other animals; and (C) articles (other than food) intended to affect the structure or any function of the body of man or other animals; and (D) articles intended for use as a component of any article specified in clause (A), (B), or (C). A food or dietary supplement for which a claim, subject to sections 343(r)(1)(B) and 343(r)(3) of this title or sections 343(r)(1)(B) and 343(r)(5)(D) of this title, is made in accordance with the requirements of section 343(r) of this title is not a drug solely because the label or the labeling contains such a claim. A food, dietary ingredient, or dietary supplement for which a truthful and not misleading statement is made in accordance with section 343(r) (6) of this title is not a drug under clause (C) solely because the label or the labeling contains such a statement.” The definitions contained in the Food, Drug and Cosmetic Act as amended and in effect from time to time are hereby incorporated by reference as if fully set forth herein.
  • Typical confection formulations are not suitable in connection with the present invention. Foremost, confection formulations are typically acidic to provoke additional salivary response. This is good for a juicy candy but is the opposite pH direction to promote nicotine absorption.
  • The absorption conditioning unit of the present invention can be packaged and sold in a kit together with vapor devices and vapor compositions. Vapor compositions can be any form of nicotine, tobacco, marijuana or base alkaloid suitable for vaporization. The vapor device can be components useful for doing so—as simple as a candle or other basic heat source, conventional e-cigarette components, or other personal vaporizer.
  • The advantageous results of the present invention are evident from the following examples.
  • Example A Soft Lozenge
  • A soft lozenge was made comprising 300 mg of sodium bicarbonate together with carbowax sentry PEG 1450. The resulting lozenge weighted approximately one gram, had harmless taste and was easily chewable.
  • The lozenge of this example was provided to a healthy female volunteer, and the pH of her saliva was measured at intervals over a period of ten minutes.
  • The t0 saliva pH was 6.8; t1 was 8.3; t3 was 8.3; t5 was 8.3 and t10 was 8.1.
  • For this and the other examples, pH measurements were taken using a Horiba compact portable meter.
  • Example B Liquid and Nicotine Delivery
  • A healthy male volunteer experienced with nicotine placed approximately 400 mg of sodium bicarbonate in a hot tea cup, and stirred the liquid.
  • The volunteer took several strong draws on a commercially available Nicotrol® inhaler (10 mg/cartridge, marketed by Pfizer). The volunteer noted a mild throat burn sensation—attributable to nicotine absorption.
  • The volunteer then took three sips of the hot beverage, and used the same Nicotrol® inhaler again. The volunteer noted a burn sensation throughout the mouth including the roof of the mouth. The user noted the sensation was comparable to the throat sensation. The user felt the nicotine delivery was much stronger.
  • The same health male volunteer repeated the same experiment this time, except that a tobacco-based e-cigarette from Discreet®. The volunteer noted a similar improvement in nicotine delivery.
  • This same experiment was repeated with room temperature water (and Nicotrol®) While still being highly effective, it was observed that the heated water seemed to provoke stronger nicotine absorption which may be attributed to thermal effects.
  • Example C Liquid
  • A liquid was prepared by adding the following ingredients in the following order: Sodium bicarbonate (NaHCO3) 3.35 g; tablet salt 0.315 g; tap water 16.11 g; and glycerin 11.38 g of glycerin.
  • The liquid of this example was provided to a healthy female volunteer (one ½ teaspoon was swirled in the mouth without swallowing), and the pH of her saliva was measured at intervals over a period of ten minutes.
  • The T0 pH was 6.5; T1 was 8.5; T3 was 8.7; T4 was 8.2; T8 was 7.5 and T10 was 7.6.
  • Example D Liquid
  • Example D employed the same ingredients as Example C but they were mixed in a different order. Glycerin was added first, then solids, mixed together and then water added. The solution was mixed and ½ teaspoon swirled in the mouth without swallowing, and the pH of her saliva was measured at intervals over a period of ten minutes.
  • The T0 pH was 6.5; T1 was 8.4; T2 was 8.5; T3 was 7.9; T5 was 7.9; T7 was 7.5 and T10 was 7.3.
  • Example E Non Aqueous Liquid
  • This example used the same ingredients as the preceding Examples C and D but eliminated water. A solution was made with three grams of sodium bicarbonate, 6 grams of glycerin and no water. To test the solution, a health female volunteer swished it in the mouth and expectorated.
  • pH was measured over ten minutes with the following results.
  • T1 was 8.2; T2 was 8.7; T3 was 8.6; T4 was 8.6; T5 was 8.6; T7 was 8.0 and T10 was 7.2.
  • Example F Propylene Glycol
  • The following formulation as made: 3 grams glycerin; 1 gram propylene glycol; and 1.5 grams sodium bicarbonate. The solution was mixed and ½ teaspoon was swirled in mouth without swallowing.
  • T0 pH was 6.5 and T1 as 7.2. Because of disagreeable taste, the test was ended after T1 timepoint.
  • Example G Xantham
  • Xantham was selected as a capture agent in view of its mucoadhesive properties. As an additional benefit it is relatively insensitive to pH.
  • Prepared a high viscosity gum by dispersing xantham in glycerin first and then introducing the xantham/glycerin blend to water to hydrate. The composition was 60 ml glyverin, 120 ml water and 1 gram modified xantham. All components were mixed at room temperature yielding a consistent viscosity. One drop 100% pure wintergreen essential oil was added.
  • To test the xantham composition orally, xantham/glycerin/water gel were blended with baking soda by volume at 2:1 liquid/powder. A ½ teaspoon was taken orally, without swallowing and swirled in mouth for several seconds. The following pH measurements were taken over a ten minute period.
  • T0 was 6.5; T1 was 8.4; T2 was 8.4; T3 was 8.6; T4 was 8.4; T5 was 8.4; T6 was 8.4; T7 was 8.4; T8 was 8.4; T9 was 8.1 and T10 was 8.2.
  • The experiment was repeated by the same volunteer after thirty minutes and several mouth rinses to demonstrate reproducibility.
  • T0 was 6.6; T1 was 8.1; T2 was 8.3; T3 was 8.3; T4 was 8.5; T5 was 8.5; T6 was 8.5; T7 was 8.4; T8 was 8.3; T9 was 8.3 and T10 was 8.1.
  • Xantham seems to have delayed the release of the sodium bicarbonate increasing the timescale for pH>8.0. This suggests a film delivery of sodium bicarbonate.
  • Example H Listerine® Film
  • Sandwich pouches of Listerine® film as a capture agent with 200 mg of sodium bicarbonate were made and tested.
  • As a baseline, pH was measured over a ten minute period with a Listerine® film only.
  • T0 was 6.2; T1 was 6.3; T2 was 6.6; T8 was 6.7 and T10 was 6.5.
  • Listerine® film pouch with 0.2 g sodium bicarbonate was tested by a healthy female volunteer (115 lbs).
  • T0 was 6.5; T1 was 8.5; T3 was 8.7; T5 was 8.7; T8 was 8.5 and T10 was 8.5.
  • The product was pleasant to the taste.
  • The sodium bicarbonate was reduced to 0.1 grams and used by second tester (180 lb male).
  • The pH results over six minutes were as follows.
  • T0 was 6.5; T1 was 8.0; T2 was 8.2; T4 was 8.0 and T6 was 7.5.
  • The experiment was repeated with a the same 115 lb female as tester and 0.2 grams of sodium bicarbonate in a single Listerine® film (the single film was wrapped around the sodium bicarbonate). The pH results over ten minutes were was follows.
  • T0 was 6.2; T1 was 8.0; T2 was 8.7; T3 was 8.7; T5 was 8.7 and T10 was 8.4.
  • The process was repeated for the 1151b female with the same formulation (i.e., 0.2 grams baking soda in a single Listerine® film). The pH results over ten minutes were as follows:
  • T0 was 6.2; T1 was 8.0; T2 was 8.7; T3 was 8.7; T5 was 8.6; and T10 was 8.2.
  • Thus, results appear to be dependent on subject weight. We then made 0.3 gram sodium bicarbonate/2 Listerine® film pouches for the 180 lb male.
  • The pH results over ten minutes were as follows.
  • T0 was at 6.4; T1 was at 8.1; T2 was at 8.6; T3 was at 8.6; T4 was at 8.4; T7 was at 8.4; T9 was at 8.1 and T10 was at 7.7.
  • Thus the male subjected required 0.3 grams for 180 lbs of body mass as compared with 0.2 grams for 115 lbs.
  • Example I
  • In considering acid/base for CO2 generation in the mouth, we considered two commercial products were considered—Alka Seltzer® (anhydrous citric acid) and Briosch® (tartaric acid).
  • We added Alka Seltzer® Original, two tabs, to a 200 ml bottle of water and measured pH over give minutes.
  • T0 was 7.35; T1 was 6.23; T2 was 6.35; T4 was 6.14 and T5 was 6.57.
  • Briosch® was used with 200 ml of water at the recommended dosage and the pH was measured over ten minutes as follows.
  • T0 was 6.81; T(10 s) was 4.41; T(30 s) was 4.95; T1 was 5.45; T4 was 5.47; T6 was 5.52 and T10 was 5.52.
  • The Briosch® was more acidic than the Alka Seltzer®.
  • Next a dry powder blend of 1:2 citiric acid/sodium bicarbonate blend was tested orally by our 115 lb female volunteer. pH was measured as follows.
  • T0 was at 6.4; t(30 s) was at 2.9; T1 was at 3.8; T2 was at 5.4; T3 was at 6.5 and T4 was at 6.5. While a very low initial pH was achieved, washout was very rapid and the mix generated a high volume of saliva.
  • Example J Other Commercial Products Including TUMS®
  • Several commercial products were tested using pH papers and a simple procedure as follows. First, rinse mouth with water. Second, wait for 1 minute. Third, take pH of saliva. Fourth, put tablet in mouth for one minute and discard it. Fifth, measure pH as a function of time.
  • With Tums®, pH was normal at one minute and the test was discontinued. With CVS® Chewable Antacid tablets (childrens) (testing two tabs), pH at one minute was 7.
  • Example K Capture of Condensate Using a Polymer
  • Two squares were cut of identical surface area (1 by 1 inch)—one from paper and one from an HPC-extruded film sourced from Fuisz LLC of Miami, Fla. (containing approximately ⅔'s hpc by weight). The two squares were weighted, and then were held above a steaming cup of tap water for approximately forty-five seconds. After thirty seconds the samples were weighed again, demonstrating that HPC film captured approximately 20 mg of water whereas the paper captured just 5 mg.
  • Example L Soft Chew
  • A soft chew was prepared with the following composition: 6 grams of sodium bicarbonate (29%); 11 grams of Carbowax 1450 (54%); 1 gram of HPMC (5%); 1 gram of powdered sugar (5%); 1 gram of glycerin (5%); 0.5 ml mint extract (2%); and 1 drop of red food coloring.
  • The resulting soft chew was tested together with Nicotrol® by a healthy volunteer and found to be quite effective for improving nicotine delivery.
  • Example M Gummy
  • A gummy was prepared with the following composition: 6 grams of sodium bicarbonate (14%); 20 grams of glycerin (47%); 12 grams of water (28%); 4 grams of unflavored gelatin (9%); 1 g of corn syrup (2%) and 1 drop of red food coloring.

Claims (21)

We claim:
1. A method for the administration of nicotine, THC, cannabidiol or a base alkaloid to a mammal, comprising:
administering an absorption conditioning unit, comprising at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, to the mammal; and then
administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, tobacco, cannabidiol and a base alkaloid.
2. The method according to claim 1, wherein the absorption conditioning unit does not contain a bioactive.
3. The method according to claim 1, wherein the absorption conditioning unit does not contain a drug.
4. The method according to claim 3, wherein the absorption conditioning unit does not contain an acid.
5. The method according to claim 1, wherein the absorption conditioning unit does not contain an acid.
6. The method according to claim 1, wherein the absorption conditioning unit includes at least the buffer agent.
7. The method according to claim 1, wherein the absorption conditioning unit includes at least the capturing agent, and the capturing agent has mucoadhesive properties.
8. A method for the administration of nicotine, THC, cannabidiol or a base alkaloid to a mammal, comprising:
administering an absorption conditioning unit including at least at least one material that creates a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration; and then
administering by inhalation a bioactive agent selected from the group consisting of nicotine, THC, tobacco, cannabidiol and a base alkaloid.
9. The method according to claim 8, wherein the absorption conditioning unit does not contain a bioactive.
10. The method according to claim 8, wherein the absorption conditioning unit does not contain an acid.
11. The method according to claim 8, wherein the absorption conditioning unit does not contain a drug.
12. The method according to claim 8, wherein the absorption conditioning unit includes at least a buffer agent.
13. The method according to claim 1, wherein the absorption conditioning unit includes at least a capturing agent having mucoadhesive properties.
14. An absorption conditioning unit, comprising a dosage form including at least two agents selected from the group consisting of (a) a buffer agent, (b) a capturing agent, (c) a penetration agent, and (d) a thermal agent, wherein said absorption conditioning unit (i) does not have an essentially acidic character, (ii) does not contain a drug, (iii) does not contain THC or other Marijuana derived active ingredient, and (iv) does not contain tobacco except for trace amounts in flavor.
15. The absorption conditioning unit according to claim 14, wherein the absorption conditioning unit does not contain an acid.
16. The absorption conditioning unit according to claim 14, wherein the absorption conditioning unit includes at least the buffer agent.
17. The absorption conditioning unit according to claim 16, wherein the buffer agent is configured to create a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration
18. The absorption conditioning unit according to claim 14, wherein the absorption conditioning unit includes at least the capturing agent, and the capturing agent has mucoadhesive properties.
19. An absorption conditioning unit, comprising a dosage form including at least at least one material that creates a pH in the oral cavity of 7.8-10 for a period of ten minutes or more after administration, the dosage form not containing an acid and not containing a drug.
20. The absorption conditioning unit according to claim 19, wherein the absorption conditioning unit includes at least a capturing agent.
21. The absorption conditioning unit according to claim 20, wherein the capturing agent has mucoadhesive properties.
US13/715,028 2012-12-14 2012-12-14 Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit Abandoned US20140166027A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/715,028 US20140166027A1 (en) 2012-12-14 2012-12-14 Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit
US13/871,213 US20140166028A1 (en) 2012-12-14 2013-04-26 Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor or Smoke Producing Device Through Use of an Absorption Conditioning Unit
PCT/US2013/073346 WO2014093127A2 (en) 2012-12-14 2013-12-05 Enhanced delivery of nicotine, thc, tobacco, cannabidiol or base alkaloid from an electronic cigarette or other vapor or smoke producing device through use of an absorption conditioning unit

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US13/715,028 US20140166027A1 (en) 2012-12-14 2012-12-14 Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/871,213 Continuation-In-Part US20140166028A1 (en) 2012-12-14 2013-04-26 Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor or Smoke Producing Device Through Use of an Absorption Conditioning Unit

Publications (1)

Publication Number Publication Date
US20140166027A1 true US20140166027A1 (en) 2014-06-19

Family

ID=50929508

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/715,028 Abandoned US20140166027A1 (en) 2012-12-14 2012-12-14 Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit

Country Status (1)

Country Link
US (1) US20140166027A1 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9095175B2 (en) 2010-05-15 2015-08-04 R. J. Reynolds Tobacco Company Data logging personal vaporizing inhaler
WO2015200864A1 (en) * 2014-06-27 2015-12-30 Crowley Kenton L Buccal and sublingual cannabinoid formulations and method of making the same
WO2016005709A1 (en) * 2014-07-10 2016-01-14 Laboratoires Ceres Use of a composition containing a long-chain polyol as a base for e-liquids
US9259035B2 (en) 2010-05-15 2016-02-16 R. J. Reynolds Tobacco Company Solderless personal vaporizing inhaler
US20160136213A1 (en) * 2014-11-14 2016-05-19 Charles William Paul Process to suspend cannabis oil in glycerin
US9352288B2 (en) 2010-05-15 2016-05-31 Rai Strategic Holdings, Inc. Vaporizer assembly and cartridge
US20170027230A1 (en) * 2015-07-31 2017-02-02 Avanzato Technology Corp. Disposable assembly for vaporizing e-liquid and a method of using the same
US9585835B1 (en) 2015-09-16 2017-03-07 Sansa Corporation (Barbados) Inc. Inhalable nicotine formulations and methods of making and using the same
US9743691B2 (en) 2010-05-15 2017-08-29 Rai Strategic Holdings, Inc. Vaporizer configuration, control, and reporting
US9968125B2 (en) 2015-01-09 2018-05-15 Philip Morris Products S.A. Nicotine—diketopiperazine microparticle formulations and methods of making the same
US9999250B2 (en) 2010-05-15 2018-06-19 Rai Strategic Holdings, Inc. Vaporizer related systems, methods, and apparatus
WO2018142403A1 (en) * 2017-02-02 2018-08-09 Panaxia Pharmaceutical Industries Ltd. Composition for buccal or sublingual administration of cannabis extract and methods for making same
WO2018172429A1 (en) * 2017-03-21 2018-09-27 Philip Morris Products S.A. Flavor delivery system
US10092713B2 (en) 2010-05-15 2018-10-09 Rai Strategic Holdings, Inc. Personal vaporizing inhaler with translucent window
US10136672B2 (en) 2010-05-15 2018-11-27 Rai Strategic Holdings, Inc. Solderless directly written heating elements
US10149844B2 (en) 2015-09-16 2018-12-11 Philip Morris Products S.A. Inhalable nicotine formulations, and methods of making and using thereof
US10159278B2 (en) 2010-05-15 2018-12-25 Rai Strategic Holdings, Inc. Assembly directed airflow
US20190022338A1 (en) * 2016-01-11 2019-01-24 Arizona Board Of Regents On Behalf Of Arizona State University Ereptiospiration device for medicinal waxes, solids, biopolymers, or highly viscous oils, and cannabinoids
US10188628B1 (en) 2016-10-27 2019-01-29 Alvin Kershman Release composition for derivatives of Cannabaceae
WO2019030561A1 (en) 2017-08-08 2019-02-14 Creso Pharma Switzerland Gmbh Composition containing cannabinoids with improved bioavailability
US10639439B2 (en) 2013-01-30 2020-05-05 Midwest Pharmaceuticals, Llc Smokeless THC and administration method thereof
US11044937B2 (en) 2014-11-07 2021-06-29 Nicoventures Trading Limited Solution comprising nicotine in unprotonated form and protonated form
US11224594B2 (en) 2015-09-16 2022-01-18 Philip Morris Products S.A. Nicotine formulations and methods of making and using the same
US20220152323A1 (en) * 2020-11-13 2022-05-19 Aari Ruben Medical therapy using cigarettes
US11383050B2 (en) * 2010-05-15 2022-07-12 Rai Strategic Holdings, Inc. Vaporizer with a pH sensor
US20230063500A1 (en) * 2014-12-12 2023-03-02 Ojai Energetics Pbc Methods and systems for forming stable droplets
US11707432B2 (en) 2015-09-16 2023-07-25 Philip Morris Products S.A. System and method for controlling the harshness of nicotine-based dry powder formulations

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060198873A1 (en) * 2003-07-24 2006-09-07 Chan Shing Y Orally dissolving films
US20110293535A1 (en) * 2009-02-11 2011-12-01 Heglund, A.S. Composition for buccal absorption of nicotine for the purpose of smoking cessation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060198873A1 (en) * 2003-07-24 2006-09-07 Chan Shing Y Orally dissolving films
US20110293535A1 (en) * 2009-02-11 2011-12-01 Heglund, A.S. Composition for buccal absorption of nicotine for the purpose of smoking cessation

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
"Vaping Weed and Nicotine Juice Together" Internet thread; http://fuckcombustion.com/threads/vaping-weed-and-nicotine-juice-together.3790/, printed from the Internet on 4/13/2015. *
Robert Glatter, MD "Are Electronic Cigarette a Safe Alternative for Smokers?"; http://www.forbes.com/sites/robertglatter/2012/10/30/are-electronic-cigarettes-a-safe-alternative-for-smokers/, October 12, 2012. *

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9352288B2 (en) 2010-05-15 2016-05-31 Rai Strategic Holdings, Inc. Vaporizer assembly and cartridge
US9861772B2 (en) 2010-05-15 2018-01-09 Rai Strategic Holdings, Inc. Personal vaporizing inhaler cartridge
US10159278B2 (en) 2010-05-15 2018-12-25 Rai Strategic Holdings, Inc. Assembly directed airflow
US10136672B2 (en) 2010-05-15 2018-11-27 Rai Strategic Holdings, Inc. Solderless directly written heating elements
US9259035B2 (en) 2010-05-15 2016-02-16 R. J. Reynolds Tobacco Company Solderless personal vaporizing inhaler
US10092713B2 (en) 2010-05-15 2018-10-09 Rai Strategic Holdings, Inc. Personal vaporizing inhaler with translucent window
US9743691B2 (en) 2010-05-15 2017-08-29 Rai Strategic Holdings, Inc. Vaporizer configuration, control, and reporting
US9427711B2 (en) 2010-05-15 2016-08-30 Rai Strategic Holdings, Inc. Distal end inserted personal vaporizing inhaler cartridge
US9095175B2 (en) 2010-05-15 2015-08-04 R. J. Reynolds Tobacco Company Data logging personal vaporizing inhaler
US11383050B2 (en) * 2010-05-15 2022-07-12 Rai Strategic Holdings, Inc. Vaporizer with a pH sensor
US9555203B2 (en) 2010-05-15 2017-01-31 Rai Strategic Holdings, Inc. Personal vaporizing inhaler assembly
US9999250B2 (en) 2010-05-15 2018-06-19 Rai Strategic Holdings, Inc. Vaporizer related systems, methods, and apparatus
US9861773B2 (en) 2010-05-15 2018-01-09 Rai Strategic Holdings, Inc. Communication between personal vaporizing inhaler assemblies
US10639439B2 (en) 2013-01-30 2020-05-05 Midwest Pharmaceuticals, Llc Smokeless THC and administration method thereof
US9675656B2 (en) 2014-06-27 2017-06-13 Kenneth L Crowley Buccal and sublingual cannabinoid formulations and method of making the same
US10420809B2 (en) 2014-06-27 2019-09-24 Farm To Farma, Inc. Buccal and sublingual cannabinoid formulations and method of making the same
WO2015200864A1 (en) * 2014-06-27 2015-12-30 Crowley Kenton L Buccal and sublingual cannabinoid formulations and method of making the same
CN106572694A (en) * 2014-07-10 2017-04-19 塞雷斯医疗用品公司 Use of a composition containing a long-chain polyol as a base for e-liquids
FR3023453A1 (en) * 2014-07-10 2016-01-15 Antoine Piccirilli USE OF A COMPOSITION COMPRISING A LONG CHAIN POLYOL AS A BASE OF E-LIQUIDS
WO2016005709A1 (en) * 2014-07-10 2016-01-14 Laboratoires Ceres Use of a composition containing a long-chain polyol as a base for e-liquids
US11044937B2 (en) 2014-11-07 2021-06-29 Nicoventures Trading Limited Solution comprising nicotine in unprotonated form and protonated form
US20160136213A1 (en) * 2014-11-14 2016-05-19 Charles William Paul Process to suspend cannabis oil in glycerin
US20230063500A1 (en) * 2014-12-12 2023-03-02 Ojai Energetics Pbc Methods and systems for forming stable droplets
US9968125B2 (en) 2015-01-09 2018-05-15 Philip Morris Products S.A. Nicotine—diketopiperazine microparticle formulations and methods of making the same
US20170027230A1 (en) * 2015-07-31 2017-02-02 Avanzato Technology Corp. Disposable assembly for vaporizing e-liquid and a method of using the same
US10869502B2 (en) * 2015-07-31 2020-12-22 14Th Round Inc. Disposable assembly for vaporizing e-liquid and a method of using the same
US9585835B1 (en) 2015-09-16 2017-03-07 Sansa Corporation (Barbados) Inc. Inhalable nicotine formulations and methods of making and using the same
US10149844B2 (en) 2015-09-16 2018-12-11 Philip Morris Products S.A. Inhalable nicotine formulations, and methods of making and using thereof
US11707432B2 (en) 2015-09-16 2023-07-25 Philip Morris Products S.A. System and method for controlling the harshness of nicotine-based dry powder formulations
US11224594B2 (en) 2015-09-16 2022-01-18 Philip Morris Products S.A. Nicotine formulations and methods of making and using the same
US10660883B2 (en) 2015-09-16 2020-05-26 Philip Morris Products S.A. Inhalable nicotine formulations, and methods of making and using thereof
US20190022338A1 (en) * 2016-01-11 2019-01-24 Arizona Board Of Regents On Behalf Of Arizona State University Ereptiospiration device for medicinal waxes, solids, biopolymers, or highly viscous oils, and cannabinoids
US11471619B2 (en) * 2016-01-11 2022-10-18 Arizona Board Of Regents On Behalf Of Arizona State University Ereptiospiration device for medicinal waxes, solids, biopolymers, or highly viscous oils, and cannabinoids
US10188628B1 (en) 2016-10-27 2019-01-29 Alvin Kershman Release composition for derivatives of Cannabaceae
US11007170B2 (en) 2017-02-02 2021-05-18 Panaxia Pharmaceutical Industries Ltd. Composition for buccal or sublingual administration of cannabis extract and methods for making same
US11590100B2 (en) 2017-02-02 2023-02-28 Panaxia Pharmaceutical Industries Ltd. Composition for buccal or sublingual administration of cannabis extract and methods for making same
WO2018142403A1 (en) * 2017-02-02 2018-08-09 Panaxia Pharmaceutical Industries Ltd. Composition for buccal or sublingual administration of cannabis extract and methods for making same
US10342259B2 (en) 2017-03-21 2019-07-09 Altria Client Services Llc Flavor delivery system
WO2018172429A1 (en) * 2017-03-21 2018-09-27 Philip Morris Products S.A. Flavor delivery system
WO2019030561A1 (en) 2017-08-08 2019-02-14 Creso Pharma Switzerland Gmbh Composition containing cannabinoids with improved bioavailability
US20220152323A1 (en) * 2020-11-13 2022-05-19 Aari Ruben Medical therapy using cigarettes

Similar Documents

Publication Publication Date Title
US20140166027A1 (en) Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor Producing Device Through Use of an Absorption Conditioning Unit
US20140166028A1 (en) Enhanced Delivery of Nicotine, THC, Tobacco, Cannabidiol or Base Alkaloid from an Electronic Cigarette or Other Vapor or Smoke Producing Device Through Use of an Absorption Conditioning Unit
US11517039B2 (en) Water-based vaporizable liquids, methods and systems for vaporizing same
ES2938328T3 (en) Extrudable and extruded compositions for the delivery of bioactive agents, method of manufacturing the same and methods of using the same
AU2008311252B2 (en) Extrudable and extruded compositions for delivery of bioactive agents, method of making the same and method of using same
US20090098192A1 (en) Extrudable and Extruded Compositions for Delivery of Bioactive Agents, Method of Making Same and Method of Using Same
US8529914B2 (en) Bioactive dose having containing a material for modulating pH of a bodily fluid to help or hinder absorption of a bioactive
US8617589B2 (en) Biocompatible film with variable cross-sectional properties
US20110318390A1 (en) Bioactive Dose Having a Rapid Release Coating or Rapid Release Layer Containing a Material for Modulating pH of a Bodily Fluid to Help or Hinder Absorption of a Bioactive
US20100256197A1 (en) Nicotine Dissolving Film With Or Without Menthol
AU2014202357B2 (en) Nonaqueous extrudable composition

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION