US20040166123A1 - Allergen dosage form - Google Patents

Allergen dosage form Download PDF

Info

Publication number
US20040166123A1
US20040166123A1 US10/723,308 US72330803A US2004166123A1 US 20040166123 A1 US20040166123 A1 US 20040166123A1 US 72330803 A US72330803 A US 72330803A US 2004166123 A1 US2004166123 A1 US 2004166123A1
Authority
US
United States
Prior art keywords
allergen
pharmaceutical product
dosage form
product according
solid dosage
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
US10/723,308
Other languages
English (en)
Inventor
Henrik Jacobi
Annette Lundegaard
Christian Houghton
Stig Aasmul-Olsen
Lise Maerkedahl
Jan Andersen
Kerry Mallindine
Owen Murray
Susan Banbury
Desmond Wong
Lisa Garrett
Michael Hall
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.)
ALK Abello AS
RP Scherer Technologies LLC
Original Assignee
ALK Abello AS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US10/723,308 priority Critical patent/US20040166123A1/en
Application filed by ALK Abello AS filed Critical ALK Abello AS
Assigned to ALK-ABELLO A/S reassignment ALK-ABELLO A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MAERKEDAHL, LISE L., HOUGHTON, CHRISTIAN G., ANDERSEN, JAN S., JACOBI, HENRIK H., LUNDEGAARD, ANNETTE R., AASMUL-OLSEN, STIG
Publication of US20040166123A1 publication Critical patent/US20040166123A1/en
Priority to US12/098,191 priority patent/US8329196B2/en
Priority to US12/098,150 priority patent/US9415015B2/en
Assigned to R. P. SCHERER TECHNOLOGIES, INC. reassignment R. P. SCHERER TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MALLINDINE, KERRY, WONG, DESMOND YIK TENG, HALL, MICHAEL, BANBURY, SUSAN GERRARD, GARRETT, LISA MARIE
Assigned to R.P. SCHERER TECHNOLOGIES, INC. reassignment R.P. SCHERER TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MURRAY, OWEN J.
Assigned to R.P. SCHERER TECHNOLOGIES, LLC reassignment R.P. SCHERER TECHNOLOGIES, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: R.P. SCHERER TECHNOLOGIES, INC
Priority to US14/287,920 priority patent/US9408805B2/en
Assigned to R.P. SCHERER TECHNOLOGIES, INC. reassignment R.P. SCHERER TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MURRAY, OWEN J.
Assigned to R.P. SCHERER TECHNOLOGIES, LLC reassignment R.P. SCHERER TECHNOLOGIES, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: R.P. SCHERER TECHNOLOGIES, INC.
Assigned to R. P. SCHERER TECHNOLOGIES, INC.. reassignment R. P. SCHERER TECHNOLOGIES, INC.. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MALLINDINE, KERRY, WONG, DESMOND YIK TENG, HALL, MICHAEL, BANBURY, SUSAN GERRARD, GARRETT, LISA MARIE
Assigned to ALK-ABELLO A/S reassignment ALK-ABELLO A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MAERKEDAHL, LISE LUND, HOUGHTON, CHRISTIAN GAUGUIN, ANDERSEN, JAN SONDERGAARD, JACOBI, HENRIK HUGO, LUNDEGAARD, ANNETTE ROMMELMAYER, AASMUL-OLSEN, STIG
Priority to US15/202,722 priority patent/US10080719B2/en
Priority to US15/699,116 priority patent/US10471008B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61JCONTAINERS SPECIALLY ADAPTED FOR MEDICAL OR PHARMACEUTICAL PURPOSES; DEVICES OR METHODS SPECIALLY ADAPTED FOR BRINGING PHARMACEUTICAL PRODUCTS INTO PARTICULAR PHYSICAL OR ADMINISTERING FORMS; DEVICES FOR ADMINISTERING FOOD OR MEDICINES ORALLY; BABY COMFORTERS; DEVICES FOR RECEIVING SPITTLE
    • A61J1/00Containers specially adapted for medical or pharmaceutical purposes
    • A61J1/03Containers specially adapted for medical or pharmaceutical purposes for pills or tablets
    • A61J1/035Blister-type containers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/35Allergens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/35Allergens
    • A61K39/36Allergens from pollen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2063Proteins, e.g. gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7007Drug-containing films, membranes or sheets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B65CONVEYING; PACKING; STORING; HANDLING THIN OR FILAMENTARY MATERIAL
    • B65DCONTAINERS FOR STORAGE OR TRANSPORT OF ARTICLES OR MATERIALS, e.g. BAGS, BARRELS, BOTTLES, BOXES, CANS, CARTONS, CRATES, DRUMS, JARS, TANKS, HOPPERS, FORWARDING CONTAINERS; ACCESSORIES, CLOSURES, OR FITTINGS THEREFOR; PACKAGING ELEMENTS; PACKAGES
    • B65D75/00Packages comprising articles or materials partially or wholly enclosed in strips, sheets, blanks, tubes, or webs of flexible sheet material, e.g. in folded wrappers
    • B65D75/28Articles or materials wholly enclosed in composite wrappers, i.e. wrappers formed by associating or interconnecting two or more sheets or blanks
    • B65D75/30Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding
    • B65D75/32Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding one or both sheets or blanks being recessed to accommodate contents
    • B65D75/325Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding one or both sheets or blanks being recessed to accommodate contents one sheet being recessed, and the other being a flat not- rigid sheet, e.g. puncturable or peelable foil
    • B65D75/327Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding one or both sheets or blanks being recessed to accommodate contents one sheet being recessed, and the other being a flat not- rigid sheet, e.g. puncturable or peelable foil and forming several compartments
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/15Medicinal preparations ; Physical properties thereof, e.g. dissolubility
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/542Mucosal route oral/gastrointestinal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/24Immunology or allergic disorders

Definitions

  • This invention relates to an allergen containing pharmaceutical product and in particular to fast-dispersing solid allergen dosage forms and a method for preparing such dosage forms.
  • Allergy is a complex disease. Many factors contribute to the sensitisation event. Among these is the susceptibility of the individual defined by an as yet insufficiently understood interplay between several genes. Another important factor is allergen exposure above certain thresholds. Several environmental factors may be important in the sensitisation process including pollution, childhood infections, parasite infections, intestinal microorganisms, etc. Once an individual is sensitised and the allergic immune response established, the presence of only minute amounts of allergen is efficiently translated into symptoms.
  • the most important allergen sources are found among the most prevalent particles of a certain size in the air we breathe. These sources are remarkably universal and include grass pollens and house dust mite faecal particles, which together are responsible for approximately 50% of all allergies. Of global importance are also animal dander, i.e. cat and dog dander, other pollens, such as mugwort pollens, and micro-fungi, such as Alternaria. On a regional basis other pollens may dominate, such as birch pollen in Northern and Central Europe, ragweed in the Eastern and Central United States, and Japanese cedar pollen in Japan. Insects, i.e. bee and wasp venoms, and foods each account for approximately 2% of all allergies.
  • Allergy i.e. type I hypersensitivity, is caused by an inappropriate immunological reaction to foreign non-pathogenic substances.
  • Important clinical manifestations of allergy include asthma, hay fever, eczema, and gastro intestinal disorders.
  • the allergic reaction is prompt and peaks within 20 minutes upon contact with the offending allergen.
  • the allergic reaction is specific in the sense that a particular individual is sensitised to particular allergen(s), whereas the individual does not necessarily show an allergic reaction to other substances known to cause allergic disease.
  • the allergic phenotype is characterized by a pronounced inflammation of the mucosa of the target organ and by the presence of allergen specific antibody of the IgE class in the circulation and on the surfaced of mast-cells and basophils.
  • An allergic attack is initiated by the reaction of the foreign allergen with allergen specific IgE antibodies, when the antibodies are bound to high affinity IgE specific receptors on the surface of mast-cells and basophils.
  • the mast-cells and basophils contain preformed mediators, i.e. histamine, tryptase, and other substances, which are released upon cross-linking of two or more receptor-bound IgE antibodies.
  • IgE antibodies are cross-linked by the simultaneous binding of one allergen molecule.
  • the cross-linking of receptor bound IgE on the surface of mast-cells also leads to release of signalling molecules responsible for the attraction of eosinophils, allergen specific T-cells, and other types of cells to the site of the allergic response.
  • Allergy disease management comprises diagnosis and treatment including prophylactic treatments.
  • Diagnosis of allergy is concerned with the demonstration of allergen specific IgE and identification of the allergen source. In many cases a careful anamnesis may be sufficient for the diagnosis of allergy and for the identification of the offending allergen source material. Most often, however, the diagnosis is supported by objective measures, such as skin prick test, blood test, or provocation test.
  • the therapeutic options fall in three major categories.
  • the first opportunity is allergen avoidance or reduction of the exposure. Whereas allergen avoidance is obvious e.g. in the case of food allergens, it may be difficult or expensive, as for house dust mite allergens, or it may be impossible, as for pollen allergens.
  • the second and most widely used therapeutic option is the prescription of classical symptomatic drugs like anti-histamines and steroids. Symptomatic drugs are safe and efficient; however, they do not alter the natural cause of the disease, and they do not control the disease dissemination.
  • the third therapeutic alternative is specific allergy vaccination that in most cases reduces or alleviates the allergic symptoms caused by the allergen in question.
  • a specific immune response such as the production of antibodies against a particular pathogen
  • an adaptive immune response is known as an adaptive immune response. This response can be distinguished from the innate immune response, which is an unspecific reaction towards pathogens.
  • An allergy vaccine is bound to address the adaptive immune response, which includes cells and molecules with antigen specificity, such as T-cells and the antibody producing B-cells. B-cells cannot mature into antibody producing cells without help from T-cells of the corresponding specificity. T-cells that participate in the stimulation of allergic immune responses are primarily of the Th2 type. Establishment of a new balance between Th1 and Th2 cells has been proposed to be beneficial and central to the immunological mechanism of specific allergy vaccination.
  • Th2 cells Whether this is brought about by a reduction in Th2 cells, a shift from Th2 to Th1 cells, or an up-regulation of Th1 cells is controversial.
  • regulatory T-cells have been proposed to be important for the mechanism of allergy vaccination. According to this model regulatory T-cells, i.e. Th3 or Tr1 cells, down-regulate both Th1 and Th2 cells of the corresponding antigen specificity.
  • an active vaccine must have the capacity to stimulate allergen specific T-cells, preferably TH1 cells.
  • the immune system is accessible through the oral cavity and oromucosal, e.g. sublingual administration, of allergens is a known route of administration.
  • allergy vaccine using the oromucosal route consists of the periodic dosing of a solution of the allergen at intervals spaced apart by at least one day.
  • the therapeutic (accumulated) maintenance doses given exceeded the maintenance of the comparable subcutaneous dose by a factor of 5-500.
  • Obvious drawbacks of this dosage form and route of administration are the problems associated with accurate and uniform self administration of the correct dose by the patient (several drops may have to be given, uniformity of the individual drops, application site accuracy, etc.). Additionally, there is a need to refrigerate the drug and include preservatives in the formulation.
  • Netien et al. (“Galenica 16-Médicaments homéopathiques” ed. 2, 1986, pages 77-99) discloses a liquid solution impregnated onto a solid particulate (granules) or conventional compressed tablets of lactose, saccharose or a mixtures of these for sublingual administration of medicaments such as allergens.
  • DD-A.0 107 208 discloses a process for preparing a conventional compressed tablet containing an allergen. Upon administration the tablet is dissolved by the saliva and the allergen is then absorbed through the mucosa of the oral cavity.
  • the formulation contains a water insoluble excipient, namely talcum as well as paraffin and fatty acids which is not desirable because it will leave an unpleasant remnant in the mouth of the patient. Moreover, the friction produced during the tabletting process may be detrimental to the physical stability of the allergens
  • EP 278 877 discloses a pharmaceutical composition for sublingual use, where a solid support is coated with a solution of an allergen when spraying the solution onto solid support globules.
  • the resulting formulation is alleged to disintegrate rapidly, but not instantaneously.
  • the formulation contains reducing sugars in the form of lactose, which are prone to react with allergens.
  • U.S. Pat. No. 4,371,516 discloses pharmaceutical dosage forms containing active ingredients, which disintegrate rapidly in water.
  • the pharmaceutical dosage forms comprise an open matrix network of carrier material, which disintegrate within 10 seconds.
  • a freeze-dried fish gelatine based carrier as disclosed in WO 00/61117 is designed to release the active ingredient instantaneously upon contact with saliva when administered in the oral cavity.
  • a freeze-dried modified starch carrier as disclosed in WO 00/44351 is designed to release the active ingredient instantaneously upon contact with saliva when administered in the oral cavity.
  • WO 99/21579 discloses a fast-dispersing dosage form comprising a vaccine and an adjuvant for oral use.
  • WO 02/13858 discloses fast dissolving pharmaceutical composition containing vaccines in the form of a fast dissolving “cake” for oral use.
  • the object of WO 02/13858 appears to be to provide viral or bacterial vaccines that will stay intact in the gastrointestinal tract. This is achieved by protecting the antigen against the acidic content of the stomach by incorporating antacids such as calcium carbonate into the cake.
  • WO 00/51568 discloses a fast-disintegrating compressed low friability tablet that is designed to dissolve in the mouth in contact with saliva in less than 30 seconds forming an easy-to-swallow suspension.
  • the present invention concerns a pharmaceutical product suitable for oromucosal administration of an allergen comprising at least one allergen and a matrix in form of a fast-dispersing non-compressed solid dosage form characterized in that the allergen containing dosage form is stable, sufficiently robust and does not release hazardous amounts of allergen residues upon handling by the patient.
  • a fast-dispersing non-compressed solid dosage form suitable for oromucosal administration comprising a matrix forming agent and an allergen wherein the allergen is stable and the dosage form has a low friability with respect to allergen release, the dosage form dissolves quickly and does not require an adjuvant.
  • the invention concerns a pharmaceutical product suitable for administration of allergen comprising
  • a fast-dispersing, non-compressed solid dosage form suitable for oromucosal administration including:
  • the loss of the allergen content in said dosage form is less than 50% of the initial allergen content after being held for 3 months at 25° C. and 60% relative humidity, and
  • the loss of allergen from said solid dosage form is less than about 0.5 ⁇ g allergen extract or less than about 0.05 ⁇ g major allergen when subjected to a friability test.
  • the solid dosage form comprises fish gelatine and mannitol as matrix-forming agents.
  • the solid dosage form comprises starch and mannitol as matrix forming agents.
  • the present invention also provides methods of producing these solid dosage forms and methods of treating allergy by administration of these solid dosage forms.
  • the present invention is based on a number of surprising findings, none of which could with a reasonable expectation of success be predicted a priori.
  • the present invention has provided the relevant dose levels for obtaining a therapeutic effect without unacceptable side effects.
  • the invention is based on the finding that an effective treatment can be obtained by the use of a fast-dispersing, non-compressed dosage form without the use of an adjuvant.
  • the invention is based on the finding that it is possible to formulate an allergen as a fast-dispersing, non-compressed dosage form while meeting the special demands required by an allergen with respect to stability and low friability.
  • Allergen proteins are susceptible to degradation that is influenced by a number of factors of the environments they are contained in. It is sought that it is pertinent for allergy treatment and in particular to allergy vaccination that the allergen is delivered intact to the immune system in therapeutically relevant doses. Thus, the allergen must remain stable during production, storage and use.
  • the present work has shown that it is in fact possible to formulate allergen proteins in a fast-dispersing, non-compressed dosage form which is stable in respect to allergen doses and allergen activity. Moreover, it has surprisingly been found that these formulations are indeed stable at room temperature. This finding has significant importance for the handling procedures of the final product.
  • the dosage form can be stored at room temperature.
  • allergen proteins in a fast-dispersing, non-compressed dosage form being therapeutically effective without the need for an adjuvant. Further such allergen containing solid dosage forms are furthermore stable at advantageous conditions.
  • allergen proteins are formulated in a fast-dispersing, non-compressed dosage form
  • it is further advantageous that the resulting dosage form does not substantially release the allergens to the surrounding environment or to a person handling the dosage form upon direct contact.
  • a priori non-compressed fast-dispersing solid dosage forms are characterized by a low mechanical strength compared to compressed tablets due to the inherent nature of the non-compressed matrix, which is fragile and brittle, almost wafer-like. During e.g. packing, storage, transport and handling of the dosage form by the patient, residual particles containing the allergen may be released to the environment and patient.
  • Non-compressed fast-dispersing solid dosage forms which are manufactured by removal of a liquid from a solidified system comprising matrix forming agents, active ingredient and other optional agents, preferably are manufactured in situ.
  • the in situ manufacturing process generally involves removal of solvent from a solidified system of the active ingredient and the matrix forming excipients within the final container such as a blister pack.
  • the in situ technique used commercially does not allow for conventional coating of the dosage form.
  • fast-dispersing dosage form refers to dosage forms which disintegrate in less than about 90 seconds, preferably in less than about 60 seconds, preferably in less than about 30 seconds, more preferably in less than about 20, even more preferably in less than about 10 seconds in the oral cavity, even more preferred in less than about 5 seconds, and most preferably in less than about 2 seconds after being received in the oral cavity.
  • the solid dosage form of the invention may be in the form of tablets, capsules, lozenges or caplets.
  • non-compressed refers to a solid dosage form, which is manufactured by removal of a liquid from a solidified system comprising matrix forming agents, active ingredient and other suitable ingredients resulting in an allergen comprised solid matrix.
  • solid dosage form refers to a unit dosage form that is not a liquid, or a powder when it is administered in the oral cavity, thus “solid dosage forms” refers to e.g. tablets containing a unit dose of the active ingredient.
  • tablets, solid dosage and vaccine form are used interchangeably herein.
  • matrix forming agent refers to any pharmaceutically acceptable water-soluble or water-dispersible excipient that will serve as a carrier for the active ingredient in the solid dosage form.
  • excipient refers to any ingredient that may be added to the formulation besides the active ingredient.
  • the term “loss of the allergen content in said dosage form” refers to for example degradation or inactivation of the allergen in the dosage form during for example storage, transportation and use.
  • the loss may be determined as either the loss in biological acticivity/potency or as the loss in the actual content of the allergen.
  • the loss of the allergen content is measured as the loss of at least one major allergen.
  • the loss may for example be measured in an ELISA method as described in Obispo et al. (Allergy, 1997, 52, pg. 806-813 using allergen specific reagents).
  • the term “loss of the allergen content from said dosage form” refers to for example release of the allergen in the dosage form during, for example storage, transportation and use.
  • the loss may be determined as either the loss in biological acticivity/potency or as the loss in the actual content of the allergen.
  • the loss of the allergen content is measured as the loss of at least one major allergen.
  • the loss may for example be measured in an ELISA method as described in Obispo et al. (Allergy, 1997, 52, pg. 806-813 using allergen specific reagents).
  • stable refers to dosage forms where the loss in allergen content is less than 50% of the initial content after being stored for 3 months at 25° C. and 60% relative humidity in the final container either measured as the loss in biological acticivity/potency or as the loss in content of at least one major allergen.
  • the loss may for example be measured in an ELISA method as described above.
  • the term “low friability” refers to the amount of allergen containing material that is lost from the dosage form when it is subjected to an external force.
  • the solid dosage form has a sufficient friability and robustness to be transported, stored and handled if the allergen containing material lost contains less than 0.5 ⁇ g allergen extract or 0.05 ⁇ g major allergen per solid dosage form.
  • the friability may be measured by a method according to the present invention.
  • Peak load to fracture means the peak force required to fracture a unit in a three point bend test using an appropriate instrument (e.g. CT5, Engineering Systems, 1 Loach Court, Radford Bridge Road, Nottingham NG8 1NA, UK).
  • oromucosal administration refers to a route of administration where the dosage form is placed under the tongue or anywhere else in the oral cavity to allow the active ingredient to come in contact with the mucosa of the oral cavity or the pharynx of the patient in order to obtain a local or systemic effect of the active ingredient.
  • An example of an oromucosal administration route is sublingual administration.
  • sublingual administration refers to a route of administration, where a dosage form is placed underneath the tongue in order to obtain a local or systemic effect of the active ingredient.
  • allergen refers to any naturally occurring protein or mixtures of proteins that have been reported to induce allergic, i.e. IgE mediated reactions upon their repeated exposure to an individual.
  • naturally occurring allergens include pollen allergens (tree, weed, herb and grass pollen allergens), mite allergens (from e.g. house dust mites and storage mites), insect allergens (inhalant, saliva- and venom origin allergens), animal allergens from e.g. saliva, hair and dander from e.g. dog, cat, horse, rat, mouse, etc., fungi allergens and food allergens.
  • the allergen may be used in the form of an allergen extract, a purified allergen, a modified allergen or a recombinant allergen or a recombinant mutant allergen, any allergen fragment above 30 amino acids or any combination thereof.
  • allergen extract refers to an extract obtained by extraction of a biological allergen source material as generally described in “Allergenic extracts”, H. Ipsen et al, chapter 20 in Allergy, principle and practise (Ed. S. Manning) 1993, Mosby-Year Book, St. Louis.
  • Such extract may be obtained by aqueous extraction of water soluble material followed by purification steps like filtration to obtain the solution i.e. the extract.
  • the extract may then be subjected to further purification and/or processing like freeze-drying removing substantially all the water.
  • an allergen extract comprises a mixture of proteins and other molecules.
  • Allergen proteins are often classified as a major allergen, an intermediate allergen, a minor allergen or no classification.
  • An allergen extract generally comprises both major and minor allergens.
  • Major allergens will generally constitute approximately 5-15% of an average allergen extract, more often about 10%. Classification of an allergen is based on an assessment of the clinical importance of the particular allergen and is given below. Examples of important major allergen found in an extract include grass group 1 and 5 and 6 allergens (e.g. Phl p 1, 5, and 6), dust mite group 1 and 2 allergens (e.g. Der p 1, Der p 2), tree pollen allergen 1 (Bet v 1), cedar pollen allergen 1 and 2 (e.g.
  • the average allergic person will be sensitised to and react to one or more major allergens and further may also be sensitised and react to minor allergens.
  • Amounts of allergen extract referred to herein refers to the dry matter content of such allergen extracts.
  • the water content of the dry matter does not exceed 10%, more preferably 5% by weight.
  • biological allergen source material refers to any biological material comprising one or more allergens.
  • examples of such materials are acarids PMB (Pure Mite Body) or WMC (Whole Mite Culture), defatted or non-defatted pollens from e.g. grasses, herbs, weeds and trees, animal hair and dander, pelt, fungi mycelia and spores, insect bodies, venom or saliva and foods.
  • Biological allergen source materials may comprise contaminating materials, such as foreign pollen and plant and flower debris for an allergen pollen source material.
  • the degree of contamination should be minimised.
  • the content of contaminants should not exceed 10% (W/W) of the biological source material.
  • an allergen extract contains at least 10% protein of the dry matter content of the allergen extract as determined in a standard protein assay such as BCA or Lowry and the remainder consists of other “non-protein material,” which may be components such as lipids, carbohydrates, or bound water which originate from the biological allergen source.
  • An allergen extract may be formulated and stored in form of a freeze-dried material obtainable by freeze-drying a liquid allergen extract at a pressure of below 800 micro bar and for a period of up till 100 hours removing the water.
  • bio-potency i.e. the in vivo allergenic activity
  • the bio-potency, i.e. the in vivo allergenic activity, of a given extract depends on a number of factors, the most important being the content of major allergens in the extract, which varies with the composition of the biological source material.
  • the amount of allergen extract in grams to be used for obtaining a desired bio-potency varies with the type of extract in question, and for a given type of extract the amount of allergen extract varies from one batch to another with the actual bio-potency of the extract.
  • the amount of allergen extract in grams to be used for obtaining a desired bio-potency may be determined using the following procedure:
  • bio-potency of various amounts of a reference extract is determined using one or more immunological in vivo tests to establish a relationship between bio-potency and amount of reference extract.
  • immunological in vivo tests are Skin Prick Test (SPT), Conjunctival Provocation Test (CPT), Bronchial Challenge with Allergen (BCA) and various clinical trials in which one or more allergy symptoms is monitored, see for example e.g. Haugaard et al., J Allergy Clin Immunol, Vol. 91, No. 3, pp 709-722, March 1993.
  • the bio-potency of one or more relevant doses for use in the dosage forms of the invention is selected with due consideration to a balance of the factors of i) the effect of treating or alleviating symptoms of allergy, ii) side effects recorded in the immunological in vivo tests, and iii) the variability of i) and ii) from one individual to another.
  • the balancing is done to obtain a maximal adequate therapeutic effect without experiencing an unacceptable level of side effect.
  • the way of balancing the factors are well known to those skilled in the art
  • bio-potency of the one or more relevant doses found may be expressed in any biopotency unit available, such as SQ units, BAU, IR units, IU, cf. above.
  • bio-potency unit values of the reference standard extracts are calculated on the basis of the bio-potency unit value allocated to the one or more relevant doses, e.g. such a standard for BAU can be obtained from FDA as illustrated below.
  • a number of parameters for evaluating the bio-potency of extracts are selected.
  • evaluation parameters are total allergenic activity, the amount of defined major allergens and overall molecular composition of the extract.
  • the total allergenic activity may be measured using an in vitro competitive immunoassay, such as ELISA and MagicLite® luminescence immunoassay (LIA), using a standardised antibody mixture raised against the extract obtained using standard methods, e.g. antibodies raised in mouse or rabbit, or a pool of allergic patients sera.
  • the content of major allergens may e.g. be quantified by rocket immuno-electrophoresis (RIE) and compared to the reference standards.
  • the overall molecular composition may be examined using e.g. crossed immunoelectrophoresis (CIE) and sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE).
  • CIE crossed immunoelectrophoresis
  • SDS-PAGE sodium dodecyl sulphate polyacrylamide gel electrophores
  • the amount of extract to be used for obtaining a desired bio-potency level may be determined as follows: For each evaluation parameter selected, the test extract is compared with the reference standard extracts using the relevant measurement methods as described above, and on the basis of the measurement results the amount of extract having the desired bio-potency is calculated.
  • SQ-Unit The SQ-Unit is determined in accordance with ALK-Abelló A/S's “SQ biopotency”-standardisation method, where 100,000 SQ units equal the standard subcutaneous maintenance dose. Normally 1 mg of extract contains between 100,000 and 1,000,000 SQ-Units, depending on the allergen source from which they originate and the manufacturing process used. The precise allergen amount can be determined by means of immunoassay i.e. total major allergen content and total allergen activity.
  • BAU Biological Allergen Units
  • BAU Bactet al.
  • a dose of 100,000 SQ-Units containing grass extract equals a content of 2600-4700 BAU according to the method above.
  • other extracts can be assessed according to the method above.
  • an allergen for desensitization shall mean a dose which when taken once or repeatedly in a monodose or in incremental doses results in, for example, an adaptive immune response and thus serves as means to desensitise allergic patients.
  • the term shall mean the amount of allergen in each dosage form necessary to induce an adaptive immune response after repeated administration of said solid dosage forms in accordance with a treatment regimen (over a period ranging from a few applications to at least one daily application over several months).
  • desensitization includes the alleviation of allergic symptoms upon administration of the dose.
  • Clinical allergy symptoms include rhinitis, conjunctivitis, asthma, urticaria, eczema, which includes reactions in the skin, eyes, nose, upper and lower airways with common symptoms such as redness and itching of eyes and nose, itching and runny nose, coaching, weezing, shortness of breathe, itching, and swelling of tissue.
  • Water content refers to the content of residual water in a solid dosage unit determined quantitatively using the Karl Fischer titration principle. This method is based on the principle that a given amount of I 2 leads to a transformation of an equivalent amount of water (European Pharmacopoeia (EP) 3 rd edition, 2.5.12).
  • Water activity a w is the effective water in a sample. Water activity measurements are carried out using methods known to the person skilled in the art, for example chilled mirror dew point technology, relative humidity with sensors that change electrical resistance or capacitance or using a lithium chloride electrode:
  • a w can be calculated according to the following equation:
  • ps saturation pressure, or the partial pressure of water vapor above pure
  • solid dosage form according to the invention provides an oromucosal pharmaceutical allergen product, which provides effective allergen doses giving an allergen specific immune response in a dose-response manner and having acceptable side effects.
  • Allergens are in a varying degree particularly susceptible to degradation in an aqueous environment, such as in an aqueous allergen solution or in a product with high water content and/or high water activity.
  • Water activity is one important factor contributing to the shelf life of a product. It is well known that the water activity of a product affects growth of bacteria as well as the stability, the potency and consistency of pharmaceuticals. Also protein stability is influenced significantly by water activity due to the relatively fragile nature of proteins. Most proteins must maintain conformation to remain active. Maintaining low water activity levels helps to prevent or entice conformational changes, which subsequently is important to ensure that a protein in the form of an allergen is stable. Also hydrolytic degradation of proteins, whether caused by enzymes or not, is affected by the water activity.
  • Water activity measurements are carried out by using methods known to the person skilled in the art for example chilled mirror dew point technology, relative humidity with sensors that change electrical resistance or capacitance or using a lithium chloride electrode.
  • the water activity of a solid dosage form preferable does not exceed 0.70 and preferably is between 0.1-0.7, more preferably is between 0.2-0.6, more preferably is between 0.3-0.5, and most preferably is between 0.4-0.5.
  • the water content of a solid dosage form determined according to the method described in Example 1 preferably does not exceed 25% and preferably is between 0.1%-20%, more preferably is between 0.5-15%, more preferably is between 2-8%, more preferably is between 4-7%, most preferably between 4.5-6% water.
  • an allergen pharmaceutical product is provided in a fast-dispersing solid dosage form, which rapidly dissolves in the oral cavity on contact with saliva, hence bringing the allergen in close contact with the immunological relevant tissue of the mucosa and allowing the allergen to address these.
  • naturally occurring allergens include pollen allergens (tree, herb, weed, and grass pollen allergens), insect allergens (inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenoptera venom allergens), animal hair and dander allergens (from e.g. dog, cat, horse, rat, mouse, etc.), and food allergens.
  • Important pollen allergens from trees, grasses and herbs are such originating from the taxonomic orders of Fagales, Oleales, Pinales and Platanaceae including for example birch (Betula), alder (Alnus), hazel (Corylus), hornbeam (Carpinus) and olive (Olea), cedar (Cryptomeria and Juniperus), Plane tree (Platanus), the order of Poales including for example grasses of the genera Lolium, Phleum, Poa, Cynodon, Dactylis, Holcus, Phalaris, Secale, and Sorghum, the orders of Asterales and Urticales including for example herbs of the genera Ambrosia, Artemisia, and Parietaria.
  • Important inhalation allergens from fungi are, for example, those originating from the genera Alternaria and Cladosporium.
  • the allergen is Bet v 1, Aln g 1, Cor a 1 and Car b 1, Que a 1, Cry j 1, Cry j 2, Cup a 1, Cup s 1, Jun a 1, Jun a 2, jun a 3, Ole e 1, Lig v 1, Pla l 1, Pla a 2, Amb a 1, Amb a 2, Amb t 5, Art v 1, Art v 2 Par j 1, Par j 2, Par j 3, Sal k 1, Ave e 1, Cyn d 1, Cyn d 7, Dac g 1, Fes p 1, Hol l 1, Lol p 1 and 5, Pha a 1, Pas n 1, Phl p 1, Phl p 5, Phl p 6, Poa p 1, Poa p 5, Sec c 1, Sec c 5, Sor h 1, Der f 1, Der f 2, Der p 1, Der p 2, Der p 7, Der m 1, Eur m 2, Gly d 1, Lep d 2, Blo t 1, Tyr p
  • the allergen is grass pollen allergen or a dust mite allergen or a ragweed allergen or a cedar pollen or a cat allergen or birch allergen.
  • the fast-dispersing solid dosage form comprises at least two different types of allergens either originating from the same allergenic source or originating from different allergenic sources.
  • the fast-dispersing solid dosage form comprises grass group 1, grass group 2/3, grass group 5 and grass group 6 allergens or mite group 1 and group 2 allergens from different mite and grass species respectively, weed antigens like short and giant ragweed allergens, different fungi allergens like alternaria and cladosporium, tree allergens like birch, hazel, hornbeam, oak and alder allergens, food allergens like peanut, soybean and milk allergens.
  • the allergen incorporated into the fast-dispersing solid dosage form may be in the form of an extract, a purified allergen, a modified allergen, a recombinant allergen or a mutant of a recombinant allergen.
  • An allergenic extract may naturally contain one or more isoforms of the same allergen, whereas a recombinant allergen typically only represents one isoform of an allergen.
  • the allergen is in the form of an extract.
  • the allergen is a recombinant allergen.
  • the allergen is a naturally occurring low IgE-binding mutant or a recombinant low IgE-binding mutant.
  • Allergens may be present in equi-molar amounts or the ratio of the allergens present may vary preferably up to 1:20.
  • the low IgE binding allergen is an allergen according to WO 99/47680 or WO 02/40676 or PCT/DK03/00322 (“Allergen mutants”).
  • the classification of an allergen as a major allergen can be subject to several tests.
  • An allergen is commonly classified as a major allergen if at least 25% of the patients shows strong IgE binding (score 3) and at least moderate binding (score 2) from 50% of the patients, the binding being determined by an CRIE (Crossed Radio Immune Electrophoresis) (CRIE Strong binding, i.e. visible IgE-binding on an X-ray film after one day; CRIE Moderate binding, i.e. binding after 3 days; CRIE Weak binding, i.e. binding after 10 days).
  • CRIE Corrossed Radio Immune Electrophoresis
  • the preferred potency of a unit dose of the dosage form is from 150-1,000,000 SQ-U/dosage form, more preferred the potency is from 500-500,000 SQ-U/dosage form, more preferred the potency is from 500-375,000 SQ-U/dosage form, more preferably the potency is from 2500-375,000 SQ-U/dosage form, more preferred the potency is from 2500-250,000 SQ-U/dosage form, more preferred 25,000-250,000 SQ-U/dosage form, more preferably 25,000-125,000 SQ-U/dosage form, more preferred 25,000-100,000 SQ-U/dosage form and most preferable 25,000-75,000 SQ-U/dosage form.
  • the solid dosage form is a repeated mono-dose, preferably within the range of from 2500-375,000 SQ-U/dosage form, more preferably 2500-250,000 SQ-U/dosage form, more preferably 25,000-250,000 SQ-U/dosage form, more preferably from 25,000-125,000 SQ-U/dosage, even more preferably from 25,000-100,000 SQ-U/dosage form, and most preferably from 25,000-75,000 SQ-U/dosage form.
  • the solid dosage from comprises an grass allergen extract wherein the potency is from 150-1,000,000 SQ-U/dosage form, more preferred the potency is from 500-500,000 SQ-U/dosage form, more preferred the potency is from 500-375,000 SQ-U/dosage form, more preferably the potency is from 2500-375,000 SQ-U/dosage form, more preferred the potency is from 2500-250,000 SQ-U/dosage form, more preferred 25,000-250,000 SQ-U/dosage form, more preferably 25,000-125,000 SQ-U/dosage form, more preferred 25,000-100,000 SQ-U/dosage form and most preferable 25,000-75,000 SQ-U/dosage form.
  • the potency of a solid dosage form according to the invention is from about 5-50,000 BAU/dosage form, more preferred the potency is from 15-25,000 BAU/dosage form, more preferably the potency is from about 15-17,600 BAU/dosage form, more preferably the potency is from about 65-17,600 BAU/dosage form, more preferably the potency is from about 65-15,000 BAU/dosage form, more preferably the potency is from about 650-15,000 BAU/dosage form, more preferred 650-6,000 BAU/dosage form, more preferred 650-4,700 BAU/dosage form, most preferable 650-3,500 BAU/dosage form.
  • the solid dosage form is a repeated mono-dose, preferably within the range of about 65-17,600 BAU/dosage form, more preferably about 65-15,000 BAU/dosage form, more preferably about 650-15,000 BAU/dosage form, more preferred 650-6,000 BAU/dosage form, even more preferred 650-4,700 BAU/dosage form, most preferable 650-3,500 BAU/dosage form.
  • the solid dosage from comprises an grass allergen extract wherein the potency is from about 5-50,000 BAU/dosage form, more preferred the potency is from 15-25,000 BAU/dosage form, more preferably the potency is from about 15-17,600 BAU/dosage form, more preferably the potency is from about 65-17,600 BAU/dosage form, more preferably the potency is from about 65-15,000 BAU/dosage form, more preferably the potency is from about 650-15,000 BAU/dosage form, more preferred 650-6,000 BAU/dosage form, more preferred 650-4,700 BAU/dosage form, most preferable 650-3,500 BAU/dosage form
  • 1 mg allergen extract normally contains between 100,000 and 1,000,000 SQ-unit. This means that 1,000,000 SQ are contained in from 1 mg extract to 10 mg allergen extract, and that 100,000 SQ are contained in from 0.1 mg extract to 1 mg allergen extract.
  • any SQ dose may be transformed into an allergen extract dose range.
  • the above dose ranges given in SQ may be recalculated into dose ranges in mg or ⁇ g allergen extract, wherein for the lower SQ limit of a range, the lower limit of the corresponding allergen extract range is used, and wherein for the upper SQ limit of a range, the upper limit of the corresponding allergen extract range is used.
  • a solid dosage form according to the invention has an allergen extract content of about 0.15 ⁇ g-10 mg/dosage form, more preferred an allergen extract content of about 0.5 ⁇ g-5 mg/dosage form, more preferably an allergen extract content of about 0.5 ⁇ g-3.75 mg/dosage form, more preferably an allergen extract content of about 2.5 ⁇ g-3.75 mg/dosage form, more preferably an allergen extract content of about 2.5 ⁇ g-2.5 mg/dosage form, more preferably an allergen extract content of about 25 ⁇ g-2.5 mg/dosage form, more preferred about 25 ⁇ g-1.25 mg/dosage form, even more preferred about 25 ⁇ g-1 mg/dosage form, most preferable about 25 ⁇ g-0.75 mg/dosage form.
  • the solid dosage form is a repeated mono-dose, preferably within the range of about 2.5 ⁇ g-3.75 mg/dosage form, more preferably 2.5 ⁇ g-2.5 mg/dosage form, more preferably of about 25 ⁇ g-2.5 mg/dosage form, more preferred of about 1.5 ⁇ g-1.25 mg/dosage form, even more of about preferred 25 ⁇ g-1 mg/dosage form, most preferable of about 25 ⁇ g-0.75 mg/dosage form.
  • a solid dosage form according to the invention has a major allergen content of about 0.015 ⁇ g-1 mg/dosage form, more preferred of about 0.05 ⁇ g-500 ⁇ g/dosage form, more preferably of about 0.05 ⁇ g-375 ⁇ g/dosage form, more preferably of about 0.25 ⁇ g-375 ⁇ g/dosage form, more preferably of about 0.25 ⁇ g-250 ⁇ g/dosage form, more preferably of about 2.5 ⁇ g-250 ⁇ g/dosage form, more preferred about 2.5 ⁇ g-125 ⁇ g/dosage form, even more preferred about 2.5 ⁇ g-100 ⁇ g/dosage form, most preferable about 2.5 ⁇ g-75 ⁇ g/dosage form.
  • the solid dosage form is a repeated mono-dose, preferably within the range of 0.25 ⁇ g-375 ⁇ g/dosage form, more preferably of about 0.25 ⁇ g-250 ⁇ g/dosage form, more preferably of about 2.5 ⁇ g-250 ⁇ g/dosage form, more preferred about 2.5 ⁇ g-125 ⁇ g/dosage form, even more preferred about 2.5 ⁇ g-100 ⁇ g/dosage form, most preferable about 2.5 ⁇ g-75 ⁇ g/dosage form.
  • the content of major allergens may be accounted for by several major allergens depending on the allergen source in question. Normally the number of major allergens is in the range of 1-10, mostly 1-5.
  • the major allergen may be comprised in an allergen extract or be recombinantly produced.
  • Recombinant major allergens may be used in the same amount as in allergen extracts comprising such major allergen or in higher doses. Higher doses are believed to be more effective, but are also believed to be associated with a risk of potentially more frequent or more severe side effects.
  • major allergens include grass group 1 allergen e.g. phl p 1, lol p 1, sor h 1, dac g 1, cyn d 1, hol 11, pha a 1, grass group 2/3 allergen e.g. phi p 2/3, lol p 2/3, grass group 5 allergen e.g. phl p 5, lol p 5, dac g 5, poa p 5, grass group 6 allergen e.g. phi p 6, poa p 6, tree pollen group 1 allergen e.g. bet v1, aln g 1, cor a 1, car b 1, mite group 1 allergen e.g.
  • grass group 1 allergen e.g. phl p 1, lol p 1, sor h 1, dac g 1, cyn d 1, hol 11, pha a 1, grass group 2/3 allergen e.g. phi p 2/3, lol p 2/3, grass group 5 allergen e.g. phl
  • a dose response effect is found cf. example 6 for solid allergen dosage forms administered oromucosally.
  • effective doses may vary. Some patients can tolerate larger doses without experiencing unacceptable side effects, while others are hypersensitive. In some cases escalating doses may be given to reach high dose levels as it is generally believed that larger doses are believed to be more effective doses. It is believed that for the majority of the average allergic population an effective dose of allergen according to the invention will preferably be between 65 BAU/solid dosage form—17,600 BAU/solid dosage form, but a dose as low as 4 and up to 47,000 BAU may be applicable for other allergic patients.
  • a dose of allergen extract of 0.5 ⁇ g-3.75 mg/dosage form, or a dose with a major allergen content of 0.05 ⁇ g-375 ⁇ g/dosage form may be suitable for an average allergic individual.
  • a dosage form according to the invention preferably contains 10-100 times more major allergen per dosage form.
  • Such hypoallergenic variants may be of recombinant or natural origin.
  • the allergen content of a solid dosage form according to the invention can be determined by routine immune assays such as CIE (Cross Immune Electrophoresis), RIE (Radio Immune Electrophoresis) and SDS-PAGE (Sodium Dodecyl Sulphate Poly Acrylamide Gel Electrophoresis) and immune assays such as ELISA and Magic Like Specific IgE assay (LIA) against extract components such as major allergens.
  • CIE Cross Immune Electrophoresis
  • RIE Radio Immune Electrophoresis
  • SDS-PAGE Sodium Dodecyl Sulphate Poly Acrylamide Gel Electrophoresis
  • immune assays such as ELISA and Magic Like Specific IgE assay (LIA) against extract components such as major allergens.
  • dosage forms which do not significantly change after manufacture with respect to physical and chemical properties, e.g. potency and content of the allergen, mechanical robustness and organoleptical properties, are preferred.
  • the stability of the active ingredient i.e. the allergen
  • the stability is assessed by means of major allergen content. Further to that stability is preferably also assessed by means of potency measurements of the allergen such as total allergen activity.
  • the “initial allergenic activity” or the “initial content of at least one major allergen” of a solid dosage form means the value of the “allergenic activity” or the “content of at least one major allergen” in the final dosage form after the manufacturing process has ended.
  • the “theoretical allergenic activity” or the “theoretical content of at least one major allergen” of a solid dosage form means the value of the “allergenic activity” or the “content of at least one major allergen” of a dose of the added allergen, e.g. in the form of an extract, before being formulated into a solid dosage form.
  • Loss in the allergen content of at least one major allergen is preferably less than 50% of the total initial content, more preferably less than 30% of the total initial content, more preferably less than 20% of the total initial content, more preferably less than 15% of the initial content, more preferably less than 10% of the initial content, more preferably less than 5% of the initial content, more preferably less than 2% of the initial content.
  • Further loss in total allergen activity according the method described in Example 1 should preferably be less than 50% of the total initial activity, more preferably less than 30% of the total initial activity, more preferably less than 20% of the total initial activity, more preferably less than 15% of the total initial activity.
  • ICH e.g. ICH guideline ICQ/Q1AR2 (Adopted by CPMP, March 2003, issued as CPMP/ICH/2736/99)) and FDA.
  • Conditions for stability testing are often referred to as zone 1-4 conditions. Zone 1 and 2 represent climatic conditions in EC, Japan and US.
  • solid dosage forms according to the present invention in their final container should preferable be stable for a least 3 months, more preferably for at least 6 months, more preferably for at least 12 months, even more preferred for at least 18 months, most preferred at least for 2 years at “long term” conditions 25° C./60 RH, more preferable at intermediate condition 30° C./65 RH, even more preferable at “accelerated conditions” 40° C./75 RH.
  • the dosage form needs to have a certain resistance to external force, but at the same time the solid dosage form needs to disintegrate quickly in the mouth.
  • the stability of the active ingredient i.e. the allergen solid dosage forms can also be assessed by additional parameters, such as mechanical robustness like friability, tensile strength, and peak load to fracture.
  • the stability of the solid dosage forms can be assessed by physical properties such as the dispersion time and organoleptical properties like visual appearance of the dosage form.
  • the solid dosage form has a Peak load to Fracture not less than 0.05 Kgf and below 0.9 KgF.
  • the solid dosage form has a tensile strength less than 1.0 N/mm2, more preferred below 0.9 N/mm2.
  • fast-dispersing dosage form disintegrates instantaneously or quickly in the mouth upon contact with the saliva in order to ensure maximum exposure of allergen to immune competent tissue of the mucosa before swallowing.
  • the solid dosage form disintegrates in less than about 90 seconds, preferably in less than about 60 seconds, preferably in less than about 30 seconds, more preferably in less than about 20, more preferably in less than about 15 seconds, even more preferably in less than about 10 seconds in the oral cavity, even more preferably in less than about 5 seconds, most preferably in less than about 2 seconds in the oral cavity.
  • the compositions of the invention are fast-dispersing solid dosage forms comprising a solid network of the allergen and any water-soluble or water-dispersible matrix.
  • the network is obtained by subliming solvent from a composition in the solid state, the composition comprising a solution of the allergen and the matrix. More preferably the network is obtained by lyophilization.
  • compositions forming part of the matrix in the fast-dispersing solid dosage form according to invention are matrix forming agents and additionally other suitable excipients such as antacids, diluents, enhancers, mucoadhesive agents, flavouring agents, taste masking agents, preservatives, antioxidants, surfactants, viscosity enhancers, colouring agents, pH modifiers, sweeteners etc.
  • suitable excipients such as antacids, diluents, enhancers, mucoadhesive agents, flavouring agents, taste masking agents, preservatives, antioxidants, surfactants, viscosity enhancers, colouring agents, pH modifiers, sweeteners etc.
  • Matrix forming agents suitable for use according to the present invention include excipients derived from animal or vegetable proteins such as gelatines, dextrins and soy, wheat and psyllium seed proteins; gums such as acacia, guar, agar and xanthan; polysaccharides; starch and modified starch, alignates; carboxymethylcellulose; carrageenans; dextrans; pectins; synthetic polymers such as polyvinylpyrrolidone; and polypeptide/protein or polysaccharide complexes such as gelatine-acacia complexes.
  • Gelatines are a heterogeneous mixture of water soluble colloid macromolecules.
  • Such heterogeneous mixtures of average molecular weights distribution may be obtained from hydrolytic action on collagen rich material of animal origin such as bone, skin, tendons, ligaments etc.
  • Gelatines may be derived from mammal e.g. cattle, pig or non-mammals e.g. warm or cold-water fish. Gelatines can be hydrolysed or non-hydrolysed, cross-linked or non-cross-linked. They can further be of a gelling or non-gelling type, the non-gelling type typically being derived from coldwater fish. In another particular embodiment starch is used. Starches are complex mixtures of carbohydrate polymers.
  • Other matrix forming agents suitable for use according to the present invention include sugars such as mannitol, dextrose, lactose, galactose and trehalose; cyclic sugars such as cyclodextrin; inorganic salts such as sodium phosphate, sodium chloride and aluminium silicates; and amino acids having from 2 to 12 carbon atoms such as a glycine, L-alanine, L-aspartic acid, L-glutamic acid, L-hydroxyproline, L-isoleucine, L-leucine and L-phenylalanine.
  • sugars such as mannitol, dextrose, lactose, galactose and trehalose
  • cyclic sugars such as cyclodextrin
  • inorganic salts such as sodium phosphate, sodium chloride and aluminium silicates
  • amino acids having from 2 to 12 carbon atoms such as a glycine, L-alanine, L-as
  • the solid dosage form preferably comprises at least about 50% W/W of at least one matrix forming agent of the dosing solution.
  • dosing solution as used in this context means a non-solid volume of a formulation of the matrix forming agents, the allergen and other optional excipients that is prepared before the solidification step.
  • the dosing solution for forming the solid dosage form comprises about 5-30% W/W, more preferably about 5-20% W/W, even more preferred between about 5-12% W/W of at least one matrix forming agent.
  • the need for dry matter content of the dosing solution will also depend on the dimensions of the tablet.
  • the solid dosage forms according to the present invention have a diameter between about 3 to about 30 mm, more preferably between about 5 to about 20 mm.
  • the solid dosage forms according to the present invention have a weight between about 1 to about 100 mg, more preferably between about 10 to about 50 mg, most preferably between about 25 to about 35 mg.
  • the solid dosage forms according to the invention have a height between about 0.5 to about 7.5 mm, more preferably between about 1 to about 5 mm.
  • a fast-dispersing solid dosage form comprising fish gelatine and mannitol as matrix-forming excipients has been found to be especially advantageous with respect to stability, visual appearance, low friability, tensile strength, peak load to fracture and mouth feel.
  • the fast-dispersing solid dosage forms comprises a solid network of the allergen and matrix form agents in the form of fish gelatine and mannitol.
  • the ratio of fish gelatine to mannitol should be controlled.
  • the ratio of fish gelatine to mannitol is from about 2:20 to about 20:1, more preferably from about 2:10 to about 10:1, most preferably from about 3:5.5 to about 6.5:3.
  • the ratio of fish gelatine to mannitol is 4:3.
  • the ratio of fish gelatine to mannitol is 6.5:5.5.
  • the ratio of fish gelatine to mannitol is 6.0:5.08.
  • the solid dosage form according to the present invention may be manufactured from a dosing solution, which is first frozen and then freeze dried.
  • the content of fish gelatine is between about 2-20% W/W of the dosing solution and the content of mannitol is between about 1-20% W/W of the dosing solution.
  • the content of fish gelatine is between about 2-10% W/W of the dosing solution and the content of mannitol is between about 1-10% W/W of the dosing solution.
  • the content of fish gelatine is between about 3-6.5% W/W of the dosing solution and the mannitol is between about 3-5.5% W/W of the dosing solution.
  • the matrix comprises about 4% W/W of the dosing solution fish gelatine and about 3% mannitol W/W of the dosing solution
  • the matrix comprises about 6.5% W/W fish gelatine of the dosing solution and about 5.5% W/W mannitol of the dosing solution.
  • the matrix comprises 6.0% W/W fish gelatine of the dosing solution and 5.08% W/W mannitol of the dosing solution.
  • a fast-dispersing solid dosage form comprising starch and mannitol as matrix-forming excipients has also been found to be especially advantageous with respect to stability, visual appearance, low friability, tensile strength, peak load to fracture and mouth feel.
  • the fast-dispersing solid dosage forms comprises a solid network of the allergen and matrix form agents in the form of starch preferably pre-gelatinised from e.g. potato, wheat, maize, corn or rice and mannitol.
  • the ratio of starch to mannitol should be controlled.
  • the ratio of starch to mannitol is from about 2:20 to about 20:1, more preferably from about 2:10 to about 10:1, most preferably from about 3:5.5 to about 6.5:3.
  • the ratio of starch to mannitol is 1:1.
  • the solid dosage form according to the present invention is manufactured from a dosing solution, which is first frozen and then freeze dried.
  • the content of starch is between about 2-20% W/W of the dosing solution and the content of mannitol is between about 1-20% W/W of the dosing solution.
  • the content of starch is between about 2-10% W/W of the dosing solution and the content of mannitol is between about 1-10% W/W of the dosing solution.
  • the content of starch is between about 3-6.5% W/W of the dosing solution and the mannitol is between about 3-5.5% W/W of the dosing solution
  • the matrix comprises about 4.4.% WAN starch of the dosing solution and about 4.4% W/W mannitol of the dosing solution.
  • pH is adjusted prior to solidification of the allergen and matrix containing solution to avoid denaturation of the allergen, precipitation and assure a stable product.
  • the optimum pH for different allergens in solution span almost the entire pH range as does their isoelectric point (pI).
  • Mixtures of allergens like extracts equally have optimum pH for solubility and stability determined by factors like the concentration of the individual allergens in the extract. Therefore an individual determination of a feasible range of pH for a formulation according to this invention may be envisaged.
  • the optimum pH for the allergen in question is determined by carrying out accelerated stability studies with formulations with different pH. The design of such studies is known to the person skilled in the art.
  • matrix compositions containing an allergen extract should be adjusted to pH between 3.5-10, more preferably 4-9, most preferably 6-9.
  • ionic strength may be a parameter affecting the stability of a freeze-dried solid dosage form primarily through its effect of the freeze-drying processes. Also it is known that high ionic strengths affect precipitation. Accordingly, an optimum must be established by measurements well known to one skilled in the art.
  • Solid dosage forms according to the invention may further comprise colouring agents, flavours, pH modifiers or taste-masking agents.
  • Suitable colouring agents include red, black and yellow iron oxides and FD & C dyes such as FD & C blue No. 2 and FD & C red No. 40.
  • Suitable flavouring agents include mint, raspberry, liquorice, orange, lemon, grapefruit, caramel, vanilla, cherry and grape flavours and combination of these.
  • Suitable pH modifiers include citric acid, tartaric acid, phosphoric acid, hydrochloric acid and maleic acid.
  • Suitable sweeteners include aspartame, acesulfame K and thaumatic.
  • Suitable taste-masking agents include sodium bicarbonate, ion-exchange resins, cyclodextrin inclusion compounds, adsorbates or microencapsulated actives.
  • Adjuvants are normally used to enhance the absorption of the allergen as well as to enhance the immune-stimulating properties of the allergen.
  • the fast-dispersing solid dosage form according to the invention does not comprise an adjuvant.
  • At least one adjuvant is incorporated into the dosage form according to the invention.
  • suitable adjuvants are aluminium salts, aluminium hydroxide such as Alhydrogel®, non-toxic bacterial fragments, cytokines, cholera toxin (and detoxified fractions thereof), cholera toxin subunit b, chitosan, homologous heat-labile fragments of E.
  • coli and detoxified fractions thereof
  • saponins bacterial products such as lipopoly-saccharides (LPS) and muramyl dipeptide (MDP), liposomes, CpG (immunostimulatory DNA sequences), lactide/glycolide homo ⁇ copolymers in the form of microparticular polymers etc.
  • LPS lipopoly-saccharides
  • MDP muramyl dipeptide
  • CpG immunonostimulatory DNA sequences
  • lactide/glycolide homo ⁇ copolymers in the form of microparticular polymers etc.
  • the use of adjuvants in allergen pharmaceutical product e.g. vaccines are often reasoned by the fact the allergens in question are not able to penetrate the barrier to be passed.
  • the adjuvants thus may serve as absorption enhancing agents or they may act as immunostimulants.
  • adjuvants may, however, be associated with serious draw backs such as unintended stimulation of various mechanisms of the immune response, systemic lupus erythematosus or affecting the barrier capabilities of the mucosal membranes and thus allowing the passage of hazardous substances. Further from an industrial point of view addition of an adjuvant further constitute further manufacturing and material cost besides the large demand for documentation in respect to drug registration.
  • a non-compressed fast-dispersing solid dosage form may be mucoadhesive to some extent. However in a preferred embodiment of the invention, it may be necessary to further add mucoadhesive excipients to said dosage form in order to increase the contact time of the dosage form with the mucosa of the oral cavity.
  • Suitable mucoadhesive excipients are polyacrylic polymers such as carbomer and carbomer derivatives; cellulose derivatives such as hydroxypropylmethylcellulose, hydroxyethylcellulose, hydroxypropylcelllulose and sodium carboxymethylcellulose; natural polymers such as gelatine, sodium alginate, pectin and glycerol.
  • the allergen dosage form dissolved in saliva is not swallowed until 3 min after administration in order to allow sufficient contact time for e.g. absorption over the mucosal membrane in the mouth.
  • the allergen dosage form is not diluted in the oral cavity e.g. by intake of a fluid like water until after 5 min.
  • Adverse events or side effects are known to exist also in connection with allergy treatment.
  • treatment aiming at modulating an ongoing response in a sensitised individual may pose a risk for inducing side effect upon administration of allergen.
  • Normally side effects seen in connection with oromucosal treatment are reported in the eye, nose, mouth, the upper and lower airway and depending on degree found acceptable. Most common are itching phenomena.
  • Adverse reactions such as anaphylactic shock, swelling of upper and lower airways, difficulty breathing, blood pressure drop, cardiac arrest would not be acceptable.
  • the fast-dispersing solid dosage form according to the invention may be manufactured and packed in disposable containers containing a multiplicity of solid dosage forms i.e. multi dosage containers.
  • the methods and materials as described in U.S. Pat. Nos. 5,729,958 and 5,343,762 are particularly favoured.
  • suitable multi dosage containers are All Aluminium Blister packs, blister packs made of polymers e.g. polypropylene, blister packs of PVC and blister packs formed from PVC/PVdC laminate and sealed with e.g. aluminium laminated to calendered kraft paper, Aclar® or Triplex®.
  • the fast-dispersing dosage form is manufactured and packed in blister packs formed from PVC/PVdC laminate and sealed with aluminium laminated to calendered kraft paper.
  • the blister pack are enclosed in an aluminium sachet of suitable size, composed of aluminium laminated to calendered kraft paper.
  • the fast-dispersing dosage form is packed in blister packs formed from aluminium and sealed with aluminium laminated to calendered kraft paper.
  • the fast-dispersing dosage form is packed in multilamilar blister packs formed from e.g. five layer aluminium laminate and sealed with aluminium laminated to calendered kraft paper.
  • the fast-dispersing dosage form is packed in blister packs formed from aluminium laminate and sealed with aluminium laminated to calendered kraft paper in such a way that is difficult for children to open the blister pack e.g. child resistant packs.
  • a solid dosage form of this type generally may be characterized by a low mechanical strength compared to compressed tablets, because of the inherent nature of such a non-compressed dosage form. This may result in the release of residual particles containing the allergen on removal from the blister pocket and during handling of the dosage form by the patient. In most situations this is of no or mainly cosmetic importance. However, this is especially detrimental when the active ingredient is an allergen, because low amounts allergen can elicit an allergic reaction in a disposed person or sensitise a previously non-sensitized individual. Normally exposure is in the range of 10 ⁇ g/year to major allergen protein accumulated for e.g. pollen allergens or dust mite allergens, which is adequate to give sensitisation or symptoms.
  • allergens may come in contact with target organs like the airways or the eye and elicit a response in an allergic person.
  • One dosage form may contain as much allergen as a person is exposed to over one year or more depending upon the nature of the exposure. It is possible to induce eye symptoms in allergic patients using a conjunctival allergen challenge. Based on such challenge studies it can be estimated how much allergen extract is needed to induce conjunctival symptoms.
  • less than about 500 SQ-U may be released from each solid dosage form during manual handling, more preferably less than about 250 SQ-U, more preferably less than about 150 SQ-U, more preferably less than about 75 SQ-U, more preferably less than about 25 SQ-U, most preferably less than about 10 SQ-U.
  • less than about 13 BAU may be released from each solid dosage form during manual handling, more preferably less than about 7 BAU, most preferably less than about 5 BAU, more preferably less than about 1.95 BAU, more preferably less than about 0.65 BAU, and most preferably less than about 0.26 BAU.
  • less than about 0.5 ⁇ g allergen extract may be released from each solid dosage form during manual handling, more preferably less than about 0.25 ⁇ g allergen extract, most preferably less than about 0.15 ⁇ g allergen extract, more preferably less than about 0.075 ⁇ g allergen extract, more preferably less than about 0.025 ⁇ g allergen extract, and most preferably less than about 0.01 ⁇ g allergen extract.
  • less than about 0.05 ⁇ g major allergen may be released from each solid dosage form during manual handling, more preferably less than about 0.025 ⁇ g major allergen, most preferably less than about 0.015 ⁇ g major allergen, more preferably less than about 0.0075 ⁇ g major allergen, more preferably less than about 0.0025 ⁇ g major allergen, most preferably less than about 0.001 ⁇ g major allergen.
  • the residual content of dust in the multi dosage container after removing the dosage form do not exceed about 2% of total allergen content, more preferred about 0.5% of total allergen content of a solid dosage form and more preferably about 0.2% of total allergen content of a solid dosage form and most preferably about 0.1% of total allergen content of a solid dosage form, more preferably about 0.01%, more preferred about 0.005%, more preferred about 0.003% of total allergen content of a solid dosage form, most preferred about 0.001% of the total allergen content of a solid dosage form.
  • friability test shall refer to any suitable test that measures the ease with which a solid dosage form crumbles, falls to pieces, are reduced to powder. Suitable friability test to use in the current invention are illustrated below and include European Pharmacopoeia 3rd edition (EP 3rd ed.) Pharmaceutical technical procedures 2.9.7. Normally friability testing of tablets is performed as set out in the EP 3rd ed. 2.9.7. and USP ⁇ 1216>, wherein loss of weight is assessed as a parameter of an intact dosage form. The EP 3rd ed. 2.9.7. friability test uses a drum having a diameter of 286 mm and about 39 mm in depth.
  • a sample of the tablets are placed on a sieve no. 100 and any loose residuals are removed by use of air pressure or a soft brush.
  • the tablets are weighed and thereafter placed in the drum.
  • the tablets are rotated for 100 times in the drum.
  • Loose residuals are then removed as just described and the tablets are weighed again.
  • the result is then expressed as the loss of mass and is calculated as a percentage of the initial mass.
  • a drum with a diameter between 283 and 291 and a depth between 36 and 40 mm may be used and a rotation of 25 ⁇ 1 rpm. Accordingly, the intactness of the current dosage form may be assessed by visual inspection and measurement of tablet weight upon having been subject to such a method.
  • the weighing can be replaced with an immune assay specific for the allergen in question.
  • the friability of said solid dosage form measured as the amount of allergen released is less than about 500 SQ-U per solid dosage form, more preferably less than about 250 SQ-U per solid dosage form, more preferably less than about 150 SQ-U per solid dosage form, more preferably less than about 75 SQ-U per solid dosage form, more preferably less than about 50 SQ-U per solid dosage form, more preferably less than about 25 SQ-U per solid dosage form, most preferably less than about 10 SQ-U per solid dosage form in any suitable friability test that exerts a sufficient external force on the compositions to be tested.
  • the friability measured as the amount of allergen released is less than about 500 SQ-U per solid dosage form, more preferably less than about 250 SQ-U per solid dosage form, more preferably less than about 150 SQ-U per solid dosage form, more preferably less than about 75 SQ-U per solid dosage form, more preferably less than about 50 SQ-U per solid dosage form, and more preferably less than about 25 SQ-U per solid dosage form, most preferably less than about 10 SQ-U per solid dosage form in a friability test performed according to the Pharmacopoeia EP 3 rd ed.
  • the friability measured as the amount of allergen released is less than 500 SQ-U per solid dosage form, more preferably less than 250 SQ-U per solid dosage form, more preferably less than about 150 SQ-U per solid dosage form, more preferably less than about 75 SQ-U per solid dosage form, more preferably less than about 50 SQ-U per solid dosage form, and more preferably less than about 25 SQ-U per solid dosage form, most preferably less than about 10 SQ-U per solid dosage form in an method comprising the following steps;
  • the friability of said solid dosage form measured as the amount of allergen released is less about 0.5 ⁇ g allergen extract per solid dosage form during manual handling, more preferably less than about 0.25 ⁇ g allergen extract per solid dosage form, more preferably less than about 0.15 ⁇ g allergen extract per solid dosage form, more preferably less than about 0.075 ⁇ g allergen extract per solid dosage form, more preferably less than about 0.025 ⁇ g allergen extract per solid dosage form, most preferably less than about 0.01 ⁇ g allergen extract per solid dosage form in an method comprising the following steps;
  • the friability measured as the amount of allergen released is less than about 0.05 ⁇ g major allergen, more preferably less than about 0.025 ⁇ g major allergen, more preferably less than about 0.015 ⁇ g major allergen per solid dosage form, more preferably less than about 0.0075 ⁇ g major allergen per solid dosage form, more preferably less than about 0.0025 ⁇ g major allergen per solid dosage form, most preferably less than about 0.001 ⁇ g major allergen per solid dosage form in an method comprising the following steps;
  • the solid dosage forms are rotated for 100 turns at 25 ⁇ 1 rpm in step b), and the allergen specific assay is an immunochemical allergen specific assay in step f).
  • the allergen content is determined by an ELISA assay.
  • the oral dosage form must have an appealing appearance.
  • the fast-dispersing solid dosage forms according to the invention are preferably subjected to visual inspection e.g. colour, shape, irregularities and defects.
  • the dosage form may also be tested for mouth feel. For example, the patients perceive the dosage form as being pleasant when it is placed in the mouth and allowed to disintegrate.
  • allergens are very bio-potent for the allergic person i.e. even small amount may trigger a response
  • uniformity of content is an important parameter during treatment to ensure, for example, that a pattern experienced by a patient is reproducible when taking the same dose.
  • the variation of content of allergen of units within a multi dosage container is within ⁇ 10%, preferably within ⁇ 7%, most preferable within ⁇ 5% compared to the set dose.
  • a multi dosage container may contain any conceivable number of fast-dispersing solid dosage forms.
  • solid allergen dosage forms are packaged and used as a group.
  • Individual solid dosage forms are packaged by dispensing as a liquid mixture into individual containers, followed by removal of water.
  • Multiple blisters may be arranged in larger sheets and multiple sheets may be packaged and sold together.
  • a container such as a blister pack may comprise a multiplicity of solid dosage forms preferably 1-100 solid dosage forms, more preferably 1-35, and most preferably 1-10 solid dosage forms per blister pack.
  • Another embodiment provides economy of manufacture, distribution and storage via the use of multiple blister packs that may, for example provided 2, 3, 4, 5, 6 or more for a single intended course of treatment use.
  • This particular packaging scheme particularly is useful for mono-dose treatments, and is made possible by dosing regimens, as described herein, that lack an up-dosing step.
  • the treatment pack comprises a sealed package of multiple solid dosage forms, each of which comprises an effective amount of an allergen.
  • the treatment pack may contain for example, at least 2, 4, 6, 7, 10, 14, 30, 60, 90, 100, 120, 200, 240, or more solid dosage forms.
  • the treatment pack may contain enough dosage forms for an entire treatment, or enough for a portion of a treatment.
  • the treatment pack contains at least one month's supply of unit doses.
  • all solid dosage forms of a treatment pack contain the same allergen dosage, thus removing the necessity for manufacturing, distributing, and storing multiple dosage units for treatment of a single allergy in an individual.
  • a kit comprising a treatment pack may further comprise instructions concerning use of the solid dosage form.
  • the instructions may comprise a warning, dosage regimen instruction, or any other information of value to the user.
  • the instructions physically may comprise, for example, a separate instruction pamphlet, a paper, a placard, and/or one or more notices printed on a container such as a box that holds the unit doses.
  • the instructions also may be supplied as a compact disk or other computer readable medium, or a video cassette.
  • the multiple solid dosage forms of a treatment pack have been formed from a liquid mixture by an in situ process that removes a suspension and/or solvation liquid, which may comprise water and/or other organic solvents and are fast dispersing.
  • a suspension and/or solvation liquid which may comprise water and/or other organic solvents and are fast dispersing.
  • each of the solid dosage forms of a treatment pack are located in individually sealed blisters in a multiple blister pack.
  • solid dosage forms of a treatment pack comprise gelatine, more preferably fish gelatine.
  • solid dosage forms of a treatment pack further comprise mannitol.
  • the effective amount of solid dosage form is between about 2.5 ⁇ g—about 3.75 mg extract/solid dosage form.
  • Clinical allergy manifestation and symptoms are several and may vary depending on the sensitized individual and the allergy inflicted. Common are symptoms like edema, itching, redness and running of the eyes and nose (rhinitis and conjunctivitis) and symptoms from upper and lower airway like wheezing, coughing, shortness of breath, skin condition like eczema, urticaria and itching. Other symptoms like fatigue are also experienced. Symptomatic treatment aims at reducing or affecting severity of the symptoms or reducing the need for other drugs given in parallel.
  • Symptomatic drug includes antihistamines like H 1 and H 2 receptor antagonists, intranasal and systemic corticosteroids, non-steroid anti-inflammatory drugs, nasal decongestants like adrenoceptor agonists. Treatment and relief of one or more allergic symptom or the reduction in the need for other medication is a further object of this invention.
  • a further object of the invention is to provide a method for treatment of allergy or alleviating symptoms of allergy in mammals comprising oromucosal administration of an effective amount of an allergen vaccine dosage form comprising (a) a matrix, and (b) at least one allergen in any of the above described embodiments.
  • Treatment of in particular seasonal allergies such as hay fever is normally associated a particular time of year were exposure to the offending allergen is present or elevated.
  • the allergen season is will vary with the allergen source e.g. the pollen and the climatic conditions for the allergen source in the particular territory.
  • the season for an allergen will differ in one part of the world from another part of the world depending on the climate, but will normally fall within the same period of the year for the same territory varying with the actual conditions of that year (see for instance “Aerobiology and inhalant allergies”, Chapter 19, T. A. E. Platts-Mills & W. R. Solomon (Ed. S. Manning) 1993, Mosby-Year Book, St. Louis).
  • This will be well known to a skilled person when a season is normally expected to start for a particular allergen in a particular region.
  • a methods of treatment including a pre-seasonal treatment i.e. an administration of solid dosage forms according to the invention before the allergen season.
  • the pre-seasonal treatment period comprises administration of solid dosage according to the invention for a period of more than 2 weeks prior to the allergen season, more preferably between 4-20 weeks, most preferably between 8-12 weeks.
  • Another object of the invention is to provide a method for treatment of allergy or allergic symptoms comprising oromucosal administration of an effective amount of an allergen vaccine dosage form comprising (a) a matrix, and (b) at least one allergen further comprising at least one anti-allergic drug e.g. antihistamines and histamine synthesis inhibitors in any of the above described embodiments.
  • an allergen vaccine dosage form comprising (a) a matrix, and (b) at least one allergen further comprising at least one anti-allergic drug e.g. antihistamines and histamine synthesis inhibitors in any of the above described embodiments.
  • anti-allergic drugs include bromapheniramine, cetirizine, fexofenadine, cyproheptadine, dexchlorpheniramine, hydroxizine, ketofene, mequitazine, oxotomide, mizolastine, ebastine, astemizole, carbinoxamide, alimemazine, buclizine, cyclizine, hydrochlorate, doxylamine, tritoqualine.
  • the invention also includes use of a pharmaceutical product comprising a non-compressed, fast dispersing solid dosage form comprising an allergen and at least one matrix forming agent for oromucosal treatment of allergy or alleviation of allergy symptoms.
  • a pharmaceutical product comprising a stable, fast-dispersing, non-compressed allergen solid dosage form with low friability comprising (a) a matrix, and (b) at least one allergen further comprising an antihistamine is used for oromucosal treatment of allergy or allergic symptoms.
  • Another embodiment provides a method for treating allergy or alleviating symptoms of allergy, comprising providing a a) treatment pack and b) repeatedly adminstering oromucosally one or more dosage forms of from the treatment pack until symptoms are alleviated.
  • a further embodiment provides a treatment regimen that utilizes a single dose for completing an entire sensitization treatment, without having to updose i.e. increase with different level(s) of allergen till reaching the particular dose.
  • This embodiment is advantageous because it simplifies and economizes on dosage form manufacture, distribution and storage by not requiring multiple dose quantities for a single treatment. Moreover, by simplifying a treatment course, patient compliance is improved, which directly leads to greater clinical effectiveness.
  • Another embodiment of the invention is pharmaceutical product comprising an orally administerable solid dosage form comprising a matrix formed of at least one pharmaceutically acceptable material, an effective amount of an allergen for desensitizing a human to said allergen, said dosage form having an allergen content at least about 50% of the initial allergen content after being held for 3 months at 25° C. and 60% relative humidity.
  • said pharmaceutical product is in form of a lozenge, tablet, a capsule or a caplet.
  • the fast-dispersing solid dosage form according to the invention can be prepared by a sublimation process according to the process disclosed in U.S. Pat. No. 4,371,516. Accordingly, a solidified solution of the allergen and the matrix forming excipients is subjected to sublimation.
  • the sublimation process is preferably carried out by freeze-drying the solution.
  • the solution is contained in a depression of the multi dosage container during the freeze-drying step to produce a solid form in any desired shape.
  • the multi dosage container can be cooled using liquid nitrogen or solid carbon dioxide. After the freezing step the frozen solution in the multi dosage container is subjected to reduced pressure and, if desired, controlled application of heat to aid the sublimation of the solvent.
  • the invention further includes the use of an allergen for the manufacture of a stable, fast-dispersing, non-compressed allergen vaccine solid dosage form with low friability comprising (a) a matrix, and (b) at least one allergen form for use in the treatment of allergy or alleviating symptoms of allergy.
  • a method is provided of producing a fast-dispersing, non-compressed solid and stable dosage form suitable for oromucosal administration having low friability comprising at least one matrix forming agent and an effective dose for desensitizing an individual to at least one allergen, comprising the steps of
  • a method is provided of producing a fast-dispersing, non-compressed solid and stable dosage form suitable for oromucosal administration having low friability comprising at least one matrix forming agent and an effective dose for desensitizing an individual to at least one allergen, comprising the steps of
  • the blister sheet is an all aluminium blister sheet.
  • the blister sheet is a multilayered, all aluminium blister sheet.
  • a method for obtaining a pharmaceutical product comprising a fast-dispersing, non-compressed solid and stable dosage form suitable for oromucosal administration comprising at least one matrix forming agent and an effective dose for desensitizing an individual to at least one allergen, said dosage form having low friability comprising
  • Oromucosal immunotherapy can be regarded as a way of inducing tolerance and inducing mucosal vaccination.
  • the mucosa of the mouth is rich in dendritic cells with a strong potential for antigen presentation.
  • the dendritic cells are believed to process the allergens and then migrate to the local lymph nodes where they present allergen derived peptides to allergen specific T cells.
  • this dendritic cell—T cell interaction is believed to induce T cells with regulatory potential or to increase the ratio of allergen specific Th1 cells to allergen specific Th2 cells.
  • a number of immunological parameters monitored during the allergy vaccination may be suitable markers for effects or efficacy of the treatment, alone or in combination respectively.
  • cytokine levels e.g. INFgamma, IL-2, IL-4, IL-5, IL-10, IL-12 and TNF alpha in blood or mucosal secretions
  • cell surface marker expression such as CD (cluster of differentiation) markers e.g.
  • effector cell function such as total histamine content of basophils; eosinophil, basophil, lymphocyte, monocyte numbers in blood, tissue and secretions; eosinophil, basophil, lymphocyte, monocyte mediator release, cytokine production, activation, chemotaxis, proliferation, signalling and other responses.
  • a vaccine according to the present invention has a profile where one or more of the following immunological changes can be found; an increased allergen specific IgG response, an increased allergen specific IgA response, reduced allergen specific IgE response, few local side effects; reduced allergen specific effector responses of eosinophils, basophils, lymphocytes and/or monocytes; induction of T cells with regulatory potential, increased ratio of allergen specific Th1 cells to allergen specific Th2 cells, induction of other cells with regulatory potential, reduced allergen specific Th2 response.
  • allergens for animal vaccines include allergens originating or transferred from parasites like ectoparasites (e.g. fleas, ticks, mosquitoes, flies), parasitic helminth venom (like heart worm e.g. Dirofilaria or onchocerciasis e.g.
  • Onchocerca and house dust mite. More preferred are saliva allergens from fleas like Ctenocephalides e.g. C. canis and C. felis , hard ticks likes Ixodes, Arnblyomma, soft ticks like Ornithodoros and from midges like Culicoides.
  • API Active Protein Ingredient
  • ELISA Enzyme Linked Immuno Sorbent Assay
  • HRP Horse Radish Peroxidase
  • LITE-reagent Luminescence labelled anti-IgE
  • TMB Tertametylbenzidine
  • composition TABLE 1 Dosage Dosage Dosage Ingredients Unit form 1 form 2 form 3
  • Function Drug substance Phleum pratense SQ-U 2500 25000 125000 API mg 0.0047 0.047 0.235 extract Other ingredients Purified water mg q.s to 250 mg q.s to 250 mg q.s to 250 mg solvent Gelatine (standard molecular mg 10 10 10 Matrix weight fish gelatin, Croda UK) Mannitol mg 7.5 7.5 7.5 Matrix Sodium hydroxide mg q.s q.s q.s pH adjustment to 7.5
  • Grass pollen extract was prepared according to the method describes in Ipsen and L ⁇ wensten (1983) Jour. Allergy. Clin. Immunol. 72:2, page 150-159.
  • grass pollen was extracted in ammonium hydrogen carbonate, for 20 hours at 5° C. Particulate matter was removed by centrifugation and the supernatant was dialysed against water (3 times), lyophilised and stored cold until reconstitution.
  • the solid dosage form had an average weight of 18 mg and average diameter of 11 mm.
  • Identity (ID), protein profile The protein profile was determined by SDS-PAGE on a Novex Mini Cell Xcell II system (Invitrogen) according to manufacturers instructions. In short, samples are diluted with sample buffer added reducing agent (0.5 M DDT), and subjected to 70° C. for 10 min and let to cool for 5 min. Sample, in-house reference and standard low-range size marker (by BIO-RAD) per well are applied on a NuPAGE 4-12% Bis-Tris gradient gel. Electrophoresis is performed at 200 V for approximately 35 min. Subsequently the gel is stained with silver stained. The protein pattern should be similar to that of the In house reference.
  • Water content The residual water was determined using a Karl Fischer titration principle. The method gives a quantitative determination of the water content in a sample based on the principle that a given amount of I 2 leads to transformation of an equivalent amount of H 2 O.
  • Total allergenic activity The test was performed using LIA (described in Eiken et al., Allergy 1992, 47:495-497), which is a competitive immunoassay. 100 ⁇ l anti human IgE monoclonal antibody bound to paramagnetic particles (PMP) (ADVIA Centaur PMP, ALK-Abelló A/S, Denmark) were washed ⁇ 3 and 100 ⁇ l of a pool of patient sera with specific Phleum pratense IgE antibodies were added and were incubated on a shaker for 2 hours at 2-8° C., whereby specific IgE binds to the PMP. The PMP were washed to remove IgG antibodies ⁇ 3 with gelatine buffer.
  • PMP paramagnetic particles
  • the ELISA method measures the concentration of Phleum pratense major allergen 5(Phl p 5).
  • Two monoclonal antibodies (ALK-Abelló A/S, DK) reacting with different epitopes on the Phl p 5 molecule were coated to the microtiterplate the night over at 4° C. After washing (4 times with washing buffer, 0.1 M PBS, 0.05% Tween-20) and blocking the plate with blocking buffer (2% Caseinbuffer), samples/references, which then binds to the antibodies, were applied. After washing again (4 times with washing buffer) biotinylated rabbit polyclonal antibodies (ALK-Abelló A/S, DK) against Phleum pratense antigens were applied to the wells and allowed to react.
  • the individual blisters were removed, opened and the solid dosage form was transferred to a suitable container.
  • the solid dosage form was then removed from the container, leaving any loose residues in said container.
  • An immunochemical allergen specific assay (ELISA) was carried out to detect the amount of allergen content in the residues (see above).
  • composition TABLE 5 Dosage Dosage Dosage Ingredients Unit form 1 form 2 form 3
  • Function Drug substance Phleum pratense SQ-U 2500 25000 125000 API mg 0.0047 0.047 0.235 extract Other ingredients Purified water Mg q.s. to 250 mg q.s. to 250 mg q.s. to 250 mg solvent Pre-gelatinised starch Mg 8 mg 9 mg 11 mg Matrix Mannitol Mg 8 mg 9 mg 11 mg Matrix Sodium hydroxide Mg q.s. q.s. q.s. pH adjustment to 7.5
  • pre-gelatinised starch was added instead of gelatine (fish source).
  • the solid dosage form had an average weight of 19 mg and average diameter of 11 mm.
  • Residues 99 Storage condition: 40° C./75% RH Tests: Friability % loss of Total total Water allergenic Sampling content of extract Disintegration content Visual activity (months): (API) (sec.) ID (%) inspection (%) 1 n.m. 58 Comply 2.5 Residues 96 2 n.m. 25 Comply 3.1 Residues 121 3 0.033 25 Comply 3.5 Residues 97 6 n.m. n.m. n.m. n.m. n.m. n.m. 9 n.m. 70 n.m. 2.7 Residues 110
  • Dosage form 1 Dosage form 2
  • Dosage form 3 Function Active substance: Phleum pratense SQ-U 2500 25000 75000 Active substance mg 0.0047 0.047 0.141 extract Other ingredients Purified water mg q.s. to 250 mg q.s. to 250 mg q.s. to 250 mg Solvent Gelatine (fish mg 16 16 16 Matrix source)* Mannitol mg 14 14 14 Matrix Sodium hydroxide mg q.s. q.s. q.s. pH adjustment 7.5
  • the solid dosage form had an average weight of 30 mg and average diameter of 12 mm.
  • the results provide an embodiment of an allergen containing solid dosage form comprising three different doses of grass allergen extract in a matrix forming agent consisting of 6.5% fish gelatin and 5.5% mannitol.
  • the allergen content and allergenic activity in the dosage forms were within the acceptable limits (see below) following manufacture. Further, the results shows that all dosage forms have a water content in the preferred range of 4-7%.
  • the stability data show that the formulations are stable at room temperature and at elevated temperature and humidity for nine months.
  • Solid allergen vaccine dosage forms were prepared containing varying ratios of matrix forming agents. TABLE 11 Solid dosage form containing 75,000 SQ-U Phleum pratense grass pollen extract prepared in different packs Load to Disintegration % gelatin % mannitol Pack type Fracture (Kgf) times (sec.) 4.00 3.00 5 layer foil 0.158 ⁇ 2 4.00 3.00 PVC/PVdC 0.199 ⁇ 2 5.00 3.75 5 layer foil 0.296 ⁇ 2 5.00 3.75 PVC/PVdC 0.264 ⁇ 2 6.00 4.50 5 layer foil 0.342 ⁇ 2 6.00 4.50 PVC/PVdC 0.386 ⁇ 2 7.00 5.25 5 layer foil 0.491 ⁇ 2 7.00 5.25 PVC/PVdC 0.421 ⁇ 2
  • Dogs were equally distributed in respect to sex within each study group and were dosed following according to table 15 TABLE 15 Group assignment dose level Number of Number of Group (in SQ-units) dosage forms a Number of dogs recovery dog 1 0 1 8 4 2 25000 1 8 — 3 500000 4 8 4
  • the dogs were administered doses as indicated in Table 15 sublingually.
  • the dosage form was placed under the tongue and the snout was held closed to allow dissolution of the dosage form.
  • the animals were dosed once per day for a period of 4 consecutive weeks. Blood samples were drawn for all dogs in every group after the completion of the treatment period. 4 dogs in the placebo and the high dose group respectively continued through a recovery period of 4 weeks where after further blood sample were drawn.
  • Phleum pratense (Phl p) specific IgG in either serum or plasma was determined as follows: ELISA plates (Costar) were coated with 10 ⁇ g/ml Phl p extract over night at 4° C. The plates were washed 4 times with 1 min soak in between and blocked against unspecific binding with 2% Casein buffer for one hour at room temperature. Individual serum or plasma samples were diluted in polypropylene plates, transferred to the ELISA plates and incubated for two hours at room temperature. After washing, HRP marked anti-dog IgG (ICN) was added to the ELISA plates and incubated for one hour at room temperature. After another wash, TMB was added to the ELISA plates, covered and incubated for 20 min at room temperature. The reaction was stopped with 0.5M sulphuric acid. The absorbance (OD) was measured in a spectrophotometer at 450 nm.
  • the dosage form containing the doses were found to be well tolerated up to and including 125,000 SQ-U as adverse events were predominantly mild in severity and limited to ‘itching’ phenomena in the mouth and throat. Adverse events were also reported in the placebo group. ‘Itching mouth’ was reported more frequently with increased dose for example, a progression of adverse events correlated with progressing doses.
  • Solid dosage forms manufactured according to example 1 were stored for 18 month at 25° C./60% RH and evaluated by visual inspection of the dosage form, i.e. tablets, and by determining the disintegration time, water content, uniformity of mass, identity (protein profile), major allergen content and total allergenic activity. All tests were performed as described in example 1. An average of a determination made in duplicate on a pool of 10 tablets is shown unless otherwise stated. TABLE 18 Storage conditions: 25° C./60% RH Product 2500 SQ-U/tablets Test: Total Major allergenic allergen Water Visual Uniformity Disintegration activity. content.
  • the solid dosage form were found to be stable, i.e. allergen content and total allergen activity remained unchanged (within assay variation and according to EP 3 rd ed. monograph for Allergen Products; total allergen activity 50-200% of theoretical value major allergen content 65-135% of theoretical value), after storage for 18 months at 25° C./60% RH. Further, there were no changes in the appearance of the solid dosage forms for all the tested allergen doses during the study. Further, there were no changes in the uniformity of mass of the dosage forms tested during the study. As to the water content, no noticeable changes were and within the preferred range of 4-7%.
  • composition of the solid dosage forms is shown in table 9 of example 1.
  • n.m n.m reference 3 n.m Similar to n.m n.m n.m n.m reference 6 n.m Similar to n.m n.m n.m reference Storage condition: 40° C./75% RH 1 Comply Similar to n.m n.m n.m n.m reference 3 Comply Similar to n.m n.m n.m n.m reference 6 Comply Similar to n.m n.m n.m n.m reference
  • the quality of the dosage forms were judged by visual inspection of the dosage form (i.e. tablets) and by determining the disintegration time, water content, uniformity of mass, identity (protein profile), major allergen content and total allergenic activity (not all data are shown).
  • the solid dosage form in all doses were found to be stable, i.e. allergen content and total allergen activity remained unchanged (within assay variation and according to EMEA 3 rd ed. monograph for Allergen Products; total allergen activity 50-200% of theoretical value, major allergen content 65-135% of theoretical value), after storage for 6 months at 25° C./60% RH and 40° C./75% RH. Further, there were no changes in the appearance of the solid dosage forms for all the tested allergen doses during the study. Further, there were no changes in the uniformity of mass of the dosage forms tested during the study. As to the water content, no noticeable changes were found during the study. A slight tendency to an increase in water content during storage at 40° C./75% RH in all doses was seen, though all the test samples were within the preferred range of 4-7%.
  • Formulation F Name of ingredient (5.5% fish gelatine) (4% fish gelatine) Function Purified water q.s. to 250.00 mg q.s. to 250.00 mg Solvent Phleum pretense 2500, 25000, or 75000 SQ-U 2500, 25000, or 125000 SQ-U Active substance 0.0047 mg, 0.047 mg, 0.141 mg 0.0047 mg, 0.047 mg, 0.235 mg Gelatine 16 mg 10 mg Matrix (fishsource) Mannitol 14 mg 7.5 mg Matrix Sodium Hydroxide q.s. to pH 7.5 q.s. to pH 7.5 pH modifier
  • a w is the water activity of a sample
  • P w is the partial water vapor pressure above a sample
  • P w * is the water vapor pressure above pure water
  • RH equ (ERH:, relative humidity equilibrium) is the relative humidity of a sample.
  • a 756 Karl Fischer Coulometer with a 774 Oven Sample Processor (Metrohm, Herisau, Switzerland) was used for the determination of the water content in the tablets. 1-3 tablets were placed in glass vials and sealed with PTFE-coated caps (Metrohm, Herisau, Switzerland). The sample vials were then placed in the 774 Oven Sample Processor (Metrohm, Herisau, Switzerland), and any moisture present in the samples was evaporated at a temperature of 130° C.
  • the results provide a further embodiment of an allergen containing solid dosage form comprising two different doses of grass allergen extract in and matrix forming agent consisting of 6.0% fish gelatin and 5.08% mannitol.
  • the dosage forms were tested in a friability test according to the methods described in example 1. The results show that the formulations are stable in respect of friability as allergen release was detected.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Mechanical Engineering (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Composite Materials (AREA)
  • Pulmonology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
US10/723,308 2002-11-26 2003-11-26 Allergen dosage form Abandoned US20040166123A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US10/723,308 US20040166123A1 (en) 2002-11-26 2003-11-26 Allergen dosage form
US12/098,191 US8329196B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US12/098,150 US9415015B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US14/287,920 US9408805B2 (en) 2002-11-26 2014-05-27 Allergen dosage form
US15/202,722 US10080719B2 (en) 2002-11-26 2016-07-06 Allergen dosage form
US15/699,116 US10471008B2 (en) 2002-11-26 2017-09-08 Allergen dosage form

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US42908602P 2002-11-26 2002-11-26
DK200201825 2002-11-26
DKPA200201825 2002-11-26
DK200300279 2003-02-24
DKPA200300279 2003-02-24
US10/723,308 US20040166123A1 (en) 2002-11-26 2003-11-26 Allergen dosage form

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/098,150 Division US9415015B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US12/098,191 Division US8329196B2 (en) 2002-11-26 2008-04-04 Allergen dosage form

Publications (1)

Publication Number Publication Date
US20040166123A1 true US20040166123A1 (en) 2004-08-26

Family

ID=32397517

Family Applications (6)

Application Number Title Priority Date Filing Date
US10/723,308 Abandoned US20040166123A1 (en) 2002-11-26 2003-11-26 Allergen dosage form
US12/098,150 Active 2026-07-11 US9415015B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US12/098,191 Active 2026-08-23 US8329196B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US14/287,920 Expired - Lifetime US9408805B2 (en) 2002-11-26 2014-05-27 Allergen dosage form
US15/202,722 Expired - Lifetime US10080719B2 (en) 2002-11-26 2016-07-06 Allergen dosage form
US15/699,116 Expired - Lifetime US10471008B2 (en) 2002-11-26 2017-09-08 Allergen dosage form

Family Applications After (5)

Application Number Title Priority Date Filing Date
US12/098,150 Active 2026-07-11 US9415015B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US12/098,191 Active 2026-08-23 US8329196B2 (en) 2002-11-26 2008-04-04 Allergen dosage form
US14/287,920 Expired - Lifetime US9408805B2 (en) 2002-11-26 2014-05-27 Allergen dosage form
US15/202,722 Expired - Lifetime US10080719B2 (en) 2002-11-26 2016-07-06 Allergen dosage form
US15/699,116 Expired - Lifetime US10471008B2 (en) 2002-11-26 2017-09-08 Allergen dosage form

Country Status (17)

Country Link
US (6) US20040166123A1 (fr)
EP (2) EP2436395B1 (fr)
JP (2) JP5442924B2 (fr)
KR (2) KR20050103187A (fr)
CN (1) CN101816787A (fr)
AU (3) AU2003280324A1 (fr)
BR (1) BR0316564A (fr)
CA (1) CA2507551A1 (fr)
CO (1) CO5690534A2 (fr)
HK (1) HK1087008A1 (fr)
IL (1) IL168733A (fr)
IS (1) IS7894A (fr)
MX (2) MXPA05005528A (fr)
NO (1) NO20053142L (fr)
NZ (1) NZ540850A (fr)
PL (1) PL218350B1 (fr)
WO (2) WO2004047793A1 (fr)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050175638A1 (en) * 2004-02-06 2005-08-11 Esch Robert E. Methods and compositions for dosing of allergens
US20060210590A1 (en) * 2005-02-03 2006-09-21 Alk-Abello A/S Minor allergen control to increase safety of immunotherapy
US20070003580A1 (en) * 2001-03-30 2007-01-04 Emile Loria Anti-allergic pharmaceutical composition containing at least one allergen and at least one antihistamine compound
WO2007051476A1 (fr) * 2005-11-04 2007-05-10 Alk-Abelló A/S Utilisation d’une formulation liquide de vaccin antiallergique pour administration oromuqueuse
WO2008152067A1 (fr) * 2007-06-12 2008-12-18 Alk-Abelló A/S Forme de dosage d'allergène contenant une antihistamine
US20110159497A1 (en) * 2007-06-16 2011-06-30 Enigma Diagnostics Limited Freeze-dried compositions for carrying out pcr and other biochemical reactions
EP2371353A1 (fr) * 2010-03-30 2011-10-05 Nitto Denko Corporation Composition pharmaceutique stabilisée, préparation de liquide de composition pharmaceutique stabilisée, préparation de formation de film et procédé de production de préparation de formation de film
WO2011137420A1 (fr) * 2010-04-30 2011-11-03 Apellis Pharmaceuticals, Inc. Procédés, articles et trousses pour la désensibilisation allergique par l'intermédiaire des muqueuses orales
US20120076825A1 (en) * 2010-06-03 2012-03-29 Alk-Abello A/S Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US20120294894A1 (en) * 2011-05-20 2012-11-22 Nitto Denko Corporation Pharmaceutical composition and method for producing the same
US20130064854A1 (en) * 2010-03-12 2013-03-14 Antigen Laboratories, Inc. Methods of treating allergies with substantially phenol-free carriers
US20130218132A1 (en) * 2010-04-30 2013-08-22 Apellis Pharmaceuticals, Inc. Methods and Articles for Preventing or Reducing Risk of Developing a Hyperallergenic Immune System
US20140093541A1 (en) * 2011-03-17 2014-04-03 Cambridge University Hospitals Nhs Foundation Treatment for peanut allergy
US20140127298A1 (en) * 2008-04-11 2014-05-08 Alk-Abello A/S Mucosomal allergen-specific immunotherapy with initial dosing after start of pollen season
CN103933562A (zh) * 2005-11-04 2014-07-23 爱尔开-阿贝优公司 用于口腔粘膜给药的液体变态反应性疫苗制剂的用途
US9956169B2 (en) 2010-10-08 2018-05-01 R.P. Scherer Technologies, Llc Oral vaccine fast-dissolving dosage form using starch
US10359292B2 (en) 2017-04-05 2019-07-23 International Business Machines Corporation Deep learning allergen mapping
US10517834B2 (en) * 2017-05-16 2019-12-31 Owen Murray Fast dissolving tablet formulations and methods of making thereof
WO2020037151A1 (fr) * 2018-08-16 2020-02-20 Aimmune Therapeutics, Inc. Immunothérapie orale de désensibilisation à l'arachide avec dose d'entretien
US10967059B2 (en) 2013-09-19 2021-04-06 Allovate, Llc Toothpaste for delivery of allergens to oral mucosa
US11229673B2 (en) 2019-05-10 2022-01-25 Société des Produits Nestlé S.A. Methods for improving the quality of life of a patient with a peanut allergy
US12016917B2 (en) 2020-04-07 2024-06-25 Société des Produits Nestlé S.A. Peanut formulations and uses thereof

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2116257T3 (da) 2000-08-09 2013-02-04 Alk Abello As Parenterale vaccineformuleringer og anvendelser deraf
MXPA05005528A (es) 2002-11-26 2006-04-05 Alk Abello As Producto farmaceutico de alergeno.
US20060115499A1 (en) * 2004-09-27 2006-06-01 Alk-Abello A/S Liquid allergy vaccine formulation for oromucosal administration
ATE415148T1 (de) * 2004-10-19 2008-12-15 Krka Tovarna Zdravil D D Novo Feste pharmazeutische zusammensetzung mit donepezilhydrochlorid
WO2006050729A2 (fr) * 2004-11-10 2006-05-18 Alk-Abellò A/S Methode de traitement preventif contre les allergies par administration par les muqueuses d'un vaccin correspondant
US7767212B2 (en) 2005-03-18 2010-08-03 Cytos Biotechnology Ag CAT allergen conjugates and uses thereof
EP1933867A1 (fr) 2005-10-04 2008-06-25 Alk-Abello A/S Formulation vaccinale solide
CN101361000B (zh) 2005-12-23 2013-04-17 爱尔开-阿贝优公司 用于药用产品溶出试验的方法
EP1872792A1 (fr) * 2006-06-29 2008-01-02 Biotech Tools S.A. Méthode pour la production d'un allergène hydrolysé
DK2187926T3 (da) 2007-09-11 2012-04-02 Univ Koebenhavn Forebyggelse af diabetes ved administration af gliadin
WO2010086989A1 (fr) 2009-01-29 2010-08-05 日東電工株式会社 Base intra-orale en forme de film et préparation
JP5588688B2 (ja) 2010-01-28 2014-09-10 日東電工株式会社 フィルム状製剤
JP5751868B2 (ja) 2010-03-30 2015-07-22 日東電工株式会社 フィルム状製剤及びその製造方法
JP5555148B2 (ja) * 2010-12-10 2014-07-23 日東電工株式会社 シート状製剤及びシート状製剤の製造方法
JP5875246B2 (ja) * 2010-12-10 2016-03-02 日東電工株式会社 シート状製剤及びシート状製剤の製造方法
FI20115375A0 (fi) 2011-04-18 2011-04-18 Teknologian Tutkimuskeskus Vtt Oy Uudet hypoallergeenit
FI20115374A0 (fi) 2011-04-18 2011-04-18 Teknologian Tutkimuskeskus Vtt Oy Uudet hypoallergeenit
JP5952634B2 (ja) * 2011-05-20 2016-07-13 日東電工株式会社 医薬組成物およびその製造方法
JP5841433B2 (ja) 2012-01-11 2016-01-13 日東電工株式会社 口腔内フィルム状基剤及び製剤
JP5931449B2 (ja) 2012-01-11 2016-06-08 日東電工株式会社 医薬組成物及びその製造方法
IN2014MN01513A (fr) * 2012-02-07 2015-09-11 Jolla Inst Allergy Immunolog
US9539334B2 (en) 2012-05-17 2017-01-10 John Hopkins University Orally dissolving thin films containing allergens and methods of making and use
JP2014034530A (ja) * 2012-08-07 2014-02-24 Nitto Denko Corp アレルゲン含有シート状製剤及びその製造方法
US9574004B2 (en) 2012-08-21 2017-02-21 Sanofi Biotechnology Methods for treating or preventing asthma by administering an IL-4R antagonist
CN103215236A (zh) * 2013-01-29 2013-07-24 中国科学院昆明动物研究所 尘螨过敏原Derf8和Derf20及其基因和应用
ES2503516B1 (es) * 2013-04-03 2015-09-09 Inmunotek, S.L. Complejo inmunogénico para vacunación y método de obtención
TWI633891B (zh) * 2013-06-04 2018-09-01 再生元醫藥公司 藉由投與il-4r抑制劑以治療過敏及增強過敏原-特異之免疫療法的方法
EP2952200A1 (fr) 2014-06-04 2015-12-09 Alk-Abelló A/S Allergène pour le traitement prophylactique d'une allergie
EP3193843A1 (fr) 2014-09-17 2017-07-26 Steerlife India Private Limited Composition effervescente et son procédé de fabrication
CA2967602A1 (fr) 2014-11-14 2016-05-19 Sanofi Biotechnology Methodes de traitement de la sinusite chronique avec polypose nasale par administration d'un antagoniste d'il-4r
WO2017098481A1 (fr) 2015-12-12 2017-06-15 Steerlife India Private Limited Compositions effervescentes de metformine et leurs procédés de préparation
KR20240065318A (ko) 2016-09-01 2024-05-14 리제너론 파아마슈티컬스, 인크. Il-4r 길항제를 투여함에 의해 알레르기를 예방하거나 치료하기 위한 방법
US10485844B2 (en) 2016-09-22 2019-11-26 Regeneron Pharmaceuticals, Inc. Methods for treating severe atopic dermatitis by administering an IL-4R inhibitor
US11524019B2 (en) 2017-08-21 2022-12-13 Glycom A/S Synthetic composition for reducing allergy symptoms
CA3079946A1 (fr) 2017-10-30 2019-05-09 Sanofi Biotechnology Methodes de traitement ou de prevention de l'asthme par administration d'un antagoniste d'il-4r
FI128016B (en) 2018-01-04 2019-07-31 Desentum Oy Recombinant hypoallergenic Ecu c1 polypeptides for use in horse allergy immunotherapy
EP3829632A4 (fr) 2018-07-27 2022-05-04 Société des Produits Nestlé S.A. Systèmes et procédés de distribution de dose unitaire d'immunothérapie orale
CN112584831A (zh) * 2018-08-16 2021-03-30 拜奥海芬治疗学有限公司 利鲁唑口腔崩解片用于治疗疾病的用途
JP7496356B2 (ja) * 2018-11-30 2024-06-06 ジェネンテック, インコーポレイテッド 凍結乾燥薬剤内の夾雑物を同定するためのレーザー誘起破壊分光法
KR20210143246A (ko) 2019-03-21 2021-11-26 리제너론 파아마슈티컬스, 인크. 알레르기 치료용 il-4/il-13 경로 억제제 및 형질 세포 절제의 병용
BR112022000581A2 (pt) 2019-08-05 2022-03-03 Regeneron Pharma Métodos para tratamento de alergia e intensificação de imunoterapia específica dos alergénos através da administração de um antagonista de il-4r
JP7050734B2 (ja) * 2019-09-30 2022-04-08 鳥居薬品株式会社 舌下アレルゲン免疫療法におけるフィルム製剤
US11672761B2 (en) 2020-11-16 2023-06-13 Orcosa Inc. Rapidly infusing platform and compositions for therapeutic treatment in humans

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3816314A (en) * 1972-05-31 1974-06-11 Exxon Research Engineering Co Block copolymers of unsaturated ester and a nitrogen containing monomer as v.i.improving and dispersant additives for oils
US4371516A (en) * 1976-10-06 1983-02-01 John Wyeth & Brother Limited Articles for carrying chemicals
US4371513A (en) * 1977-03-25 1983-02-01 W. R. Grace & Co. Alumina compositions
US5244663A (en) * 1987-01-28 1993-09-14 Medibrevex Therapeutic method against allergy
US5343672A (en) * 1992-12-01 1994-09-06 Scherer Ltd R P Method for manufacturing freeze dried dosages in a multilaminate blister pack
US5762961A (en) * 1996-02-09 1998-06-09 Quadrant Holdings Cambridge Ltd. Rapidly soluble oral solid dosage forms, methods of making same, and compositions thereof
US5811128A (en) * 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US20020114833A1 (en) * 2000-12-06 2002-08-22 Abu-Izza Khawla A. Fast dissolving tablet
US6458968B2 (en) * 2000-06-09 2002-10-01 Rensselaer Polytechnic Institute Dithiocarboxylic ester synthetic process
US6471992B1 (en) * 1997-02-20 2002-10-29 Therics, Inc. Dosage form exhibiting rapid disperse properties, methods of use and process for the manufacture of same
US20020197321A1 (en) * 1997-10-27 2002-12-26 Harry Seager Solid dispersing vaccine composition for oral delivery
US6919086B1 (en) * 1997-03-27 2005-07-19 Merck Patent Gesellschaft Mit Graminae pollen allergen mutants for specific immunotherapy, and production and use of the same
US8012505B2 (en) * 2003-02-28 2011-09-06 Alk-Abello A/S Dosage form having a saccharide matrix

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US57856A (en) * 1866-09-11 Improvement in cultivators
US278877A (en) * 1883-06-05 Ribbon or label holder for hats
US346622A (en) * 1886-08-03 Bedstead
US271079A (en) * 1883-01-23 Grain weigher and tally
US44351A (en) * 1864-09-20 Gang-plow
US107208A (en) * 1870-09-06 Improvement in apparatus for the manufacture of illuminating-gas
US2378383A (en) * 1942-10-17 1945-06-19 Brush Dev Co Transient signal recordingreproducing device
DD107208A1 (fr) * 1973-06-04 1974-07-20
US4822611A (en) 1986-12-09 1989-04-18 Immunotec, Inc. Oral hyposensitization in the treatment of allergies
FR2609895B1 (fr) 1987-01-28 1991-06-28 Bruttmann Georges Nouvelles formes galeniques d'allergenes pour administration par voie per- et sub-linguale
IT1217628B (it) * 1988-05-18 1990-03-30 Neo Abello Spa Forma farmaceutica e supporto per la somministrazione di estratti ellergenici
US5215756A (en) * 1989-12-22 1993-06-01 Gole Dilip J Preparation of pharmaceutical and other matrix systems by solid-state dissolution
JPH0537070A (ja) 1991-07-26 1993-02-12 Sharp Corp 分布帰還型半導体レーザ素子の製造方法
JPH0597672A (ja) * 1991-10-08 1993-04-20 Terumo Corp アミド誘導体含有固形製剤およびその製造方法
WO1993019741A1 (fr) * 1992-03-31 1993-10-14 Benzon Pharma A/S Formulation pharmaceutique
IL105553A (en) 1992-05-06 1998-01-04 Janssen Pharmaceutica Inc Solid dosage forms consisting of a porous network of matrix that releases a substance that dissipates rapidly in water
US5343762A (en) 1992-10-05 1994-09-06 Rosemount Inc. Vortex flowmeter
US6074673A (en) 1996-04-22 2000-06-13 Guillen; Manuel Slow-release, self-absorbing, drug delivery system
JP2832885B2 (ja) 1996-09-02 1998-12-09 株式会社林原生物化学研究所 スギ花粉アレルゲン
JP4218987B2 (ja) 1996-11-13 2009-02-04 明治乳業株式会社 ペプチド免疫療法治療剤
GB9722682D0 (en) 1997-10-27 1997-12-24 Scherer Ltd R P Pharmaceutical products
CN1332029C (zh) 1998-03-16 2007-08-15 阿尔克-阿贝洛有限公司 突变的重组过敏原
GB9819484D0 (en) 1998-09-07 1998-10-28 Univ Bristol Therapeutic agents
GB9901819D0 (en) * 1999-01-27 1999-03-17 Scherer Corp R P Pharmaceutical compositions
FR2790387B1 (fr) 1999-03-01 2001-05-18 Prographarm Laboratoires Comprime orodispersible presentant une faible friabilite et son procede de preparation
GB9904629D0 (en) * 1999-03-02 1999-04-21 Danbiosyst Uk Oral drug delivery system
FR2791569B1 (fr) 1999-03-31 2003-05-09 Pf Medicament Composition ou structure microporeuse expansee isotrope a dissolution rapide a usage pharmaceutique, veterinaire, dietetique, alimentaire ou cosmetique et son procede d'obtention
GB9908014D0 (en) * 1999-04-08 1999-06-02 Scherer Corp R P Pharmaceutical compositions
SE9901857D0 (sv) * 1999-05-21 1999-05-21 Anders Cedergren Karl Fischer reagent
WO2001039800A2 (fr) 1999-12-06 2001-06-07 The Board Of Trustees Of The University Of Arkansas Controlled delivery of antigens
GB0000891D0 (en) 2000-01-14 2000-03-08 Allergy Therapeutics Ltd Formulation
GB0002386D0 (en) * 2000-02-02 2000-03-22 Novartis Nutrition Ag Therapeutic composition
GB0020089D0 (en) * 2000-08-15 2000-10-04 Smithkline Beecham Biolog Vaccine Composition
NZ525820A (en) 2000-11-16 2005-01-28 Alk Abello As Mutant allergens
GB2378383A (en) 2001-06-06 2003-02-12 Gursharan Moonga Nasal delivery of pharmaceutical compositions in powder form
DE10137232A1 (de) * 2001-07-30 2003-02-20 Pierre Foss Allergenträger
EP1504089A2 (fr) 2002-05-16 2005-02-09 Alk Abell A/S Mutants allergenes
MXPA05005528A (es) 2002-11-26 2006-04-05 Alk Abello As Producto farmaceutico de alergeno.
DE202008006598U1 (de) * 2008-04-11 2008-10-02 Alk-Abelló A/S Allergie-Impfstoff-Formulierung zur mucosalen Verabreichung

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3816314A (en) * 1972-05-31 1974-06-11 Exxon Research Engineering Co Block copolymers of unsaturated ester and a nitrogen containing monomer as v.i.improving and dispersant additives for oils
US4371516A (en) * 1976-10-06 1983-02-01 John Wyeth & Brother Limited Articles for carrying chemicals
US4371513A (en) * 1977-03-25 1983-02-01 W. R. Grace & Co. Alumina compositions
US5811128A (en) * 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US5244663A (en) * 1987-01-28 1993-09-14 Medibrevex Therapeutic method against allergy
US5343672A (en) * 1992-12-01 1994-09-06 Scherer Ltd R P Method for manufacturing freeze dried dosages in a multilaminate blister pack
US5729958A (en) * 1992-12-01 1998-03-24 R. P. Scherer Corporation Method for manufacturing freeze dried dosages in a multilaminate blister pack
US5762961A (en) * 1996-02-09 1998-06-09 Quadrant Holdings Cambridge Ltd. Rapidly soluble oral solid dosage forms, methods of making same, and compositions thereof
US6471992B1 (en) * 1997-02-20 2002-10-29 Therics, Inc. Dosage form exhibiting rapid disperse properties, methods of use and process for the manufacture of same
US6919086B1 (en) * 1997-03-27 2005-07-19 Merck Patent Gesellschaft Mit Graminae pollen allergen mutants for specific immunotherapy, and production and use of the same
US20020197321A1 (en) * 1997-10-27 2002-12-26 Harry Seager Solid dispersing vaccine composition for oral delivery
US6458968B2 (en) * 2000-06-09 2002-10-01 Rensselaer Polytechnic Institute Dithiocarboxylic ester synthetic process
US20020114833A1 (en) * 2000-12-06 2002-08-22 Abu-Izza Khawla A. Fast dissolving tablet
US8012505B2 (en) * 2003-02-28 2011-09-06 Alk-Abello A/S Dosage form having a saccharide matrix

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070003580A1 (en) * 2001-03-30 2007-01-04 Emile Loria Anti-allergic pharmaceutical composition containing at least one allergen and at least one antihistamine compound
US7927600B2 (en) * 2001-03-30 2011-04-19 Emile Loria Anti-allergic pharmaceutical composition containing at least one allergen and at least one antihistamine compound
US8491909B2 (en) * 2004-02-06 2013-07-23 Greer Laboratories, Inc. Methods and compositions for dosing of allergens
US9345761B2 (en) 2004-02-06 2016-05-24 Greer Laboratories, Inc. Methods and compositions for dosing of allergens
US20050175638A1 (en) * 2004-02-06 2005-08-11 Esch Robert E. Methods and compositions for dosing of allergens
US20110200641A1 (en) * 2005-02-03 2011-08-18 Alk-Abelló A/S Minor allergen control to increase safety of immunotherapy
US20060210590A1 (en) * 2005-02-03 2006-09-21 Alk-Abello A/S Minor allergen control to increase safety of immunotherapy
AU2006310881B2 (en) * 2005-11-04 2012-07-19 Alk-Abello A/S Use of a liquid allergy vaccine formulation for oromucosal administration
US20090297564A1 (en) * 2005-11-04 2009-12-03 Domingo Barber Hernandez Use of a Liquid Allergy Vaccine Formulation for Oromucosal Administration
WO2007051476A1 (fr) * 2005-11-04 2007-05-10 Alk-Abelló A/S Utilisation d’une formulation liquide de vaccin antiallergique pour administration oromuqueuse
CN103933562A (zh) * 2005-11-04 2014-07-23 爱尔开-阿贝优公司 用于口腔粘膜给药的液体变态反应性疫苗制剂的用途
WO2008152067A1 (fr) * 2007-06-12 2008-12-18 Alk-Abelló A/S Forme de dosage d'allergène contenant une antihistamine
US20110159497A1 (en) * 2007-06-16 2011-06-30 Enigma Diagnostics Limited Freeze-dried compositions for carrying out pcr and other biochemical reactions
US20140127298A1 (en) * 2008-04-11 2014-05-08 Alk-Abello A/S Mucosomal allergen-specific immunotherapy with initial dosing after start of pollen season
US20130064854A1 (en) * 2010-03-12 2013-03-14 Antigen Laboratories, Inc. Methods of treating allergies with substantially phenol-free carriers
EP2371353A1 (fr) * 2010-03-30 2011-10-05 Nitto Denko Corporation Composition pharmaceutique stabilisée, préparation de liquide de composition pharmaceutique stabilisée, préparation de formation de film et procédé de production de préparation de formation de film
WO2011137420A1 (fr) * 2010-04-30 2011-11-03 Apellis Pharmaceuticals, Inc. Procédés, articles et trousses pour la désensibilisation allergique par l'intermédiaire des muqueuses orales
AU2011245142B2 (en) * 2010-04-30 2016-10-06 Allovate, Llc Methods, articles and kits for allergic desensitization via the oral mucosa
EP4011362A1 (fr) * 2010-04-30 2022-06-15 Allovate, LLC Procédés, articles et kits de désensibilisation allergique par l'intermédiaire de la muqueuse orale
US20130218132A1 (en) * 2010-04-30 2013-08-22 Apellis Pharmaceuticals, Inc. Methods and Articles for Preventing or Reducing Risk of Developing a Hyperallergenic Immune System
JP2013527846A (ja) * 2010-04-30 2013-07-04 アロヴェイト・エルエルシー 口腔粘膜を介してアレルギーを減感作するための方法、物品及びキット
AU2017200098B2 (en) * 2010-04-30 2018-05-17 Allovate, Llc Methods, articles and kits for allergic desensitization via the oral mucosa
CN107184971A (zh) * 2010-04-30 2017-09-22 艾洛维特有限责任公司 经由口腔粘膜变应性脱敏的方法、制品和试剂盒
US9724271B2 (en) * 2010-04-30 2017-08-08 Allovate, Llc Methods and articles for preventing or reducing risk of developing a hyperallergenic immune system
US10842866B2 (en) 2010-06-03 2020-11-24 Alk-Abelló A/S Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US9265824B2 (en) * 2010-06-03 2016-02-23 Alk-Abello A/S Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US9259464B2 (en) * 2010-06-03 2016-02-16 Alk-Abelló A/S Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US20130280302A1 (en) * 2010-06-03 2013-10-24 Heather Michelle Webster Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US20120076825A1 (en) * 2010-06-03 2012-03-29 Alk-Abello A/S Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US10874736B2 (en) 2010-06-03 2020-12-29 Alk-Abello A/S Pharmaceutical product comprising mite allergen extract(s) and a method for the manufacture thereof
US9956169B2 (en) 2010-10-08 2018-05-01 R.P. Scherer Technologies, Llc Oral vaccine fast-dissolving dosage form using starch
US20140093541A1 (en) * 2011-03-17 2014-04-03 Cambridge University Hospitals Nhs Foundation Treatment for peanut allergy
US9481716B2 (en) * 2011-03-17 2016-11-01 Cambridge Univeristy Hospitals NHS Foundation Trust Treatment for peanut allergy
US20120294894A1 (en) * 2011-05-20 2012-11-22 Nitto Denko Corporation Pharmaceutical composition and method for producing the same
US10967059B2 (en) 2013-09-19 2021-04-06 Allovate, Llc Toothpaste for delivery of allergens to oral mucosa
US11980664B2 (en) 2013-09-19 2024-05-14 Allovate, Llc Toothpaste for delivering allergens to oral mucosa
US10359292B2 (en) 2017-04-05 2019-07-23 International Business Machines Corporation Deep learning allergen mapping
US10828261B2 (en) * 2017-05-16 2020-11-10 Owen Murray Fast dissolving tablet formulations and methods of making thereof
US10517834B2 (en) * 2017-05-16 2019-12-31 Owen Murray Fast dissolving tablet formulations and methods of making thereof
WO2020037151A1 (fr) * 2018-08-16 2020-02-20 Aimmune Therapeutics, Inc. Immunothérapie orale de désensibilisation à l'arachide avec dose d'entretien
US11229673B2 (en) 2019-05-10 2022-01-25 Société des Produits Nestlé S.A. Methods for improving the quality of life of a patient with a peanut allergy
US12016917B2 (en) 2020-04-07 2024-06-25 Société des Produits Nestlé S.A. Peanut formulations and uses thereof

Also Published As

Publication number Publication date
AU2003283207A1 (en) 2004-06-18
US20140271722A1 (en) 2014-09-18
CO5690534A2 (es) 2006-10-31
NZ540850A (en) 2008-11-28
AU2003283207B2 (en) 2009-12-03
BR0316564A (pt) 2005-10-04
EP2436395A2 (fr) 2012-04-04
WO2004047794A2 (fr) 2004-06-10
CN101816787A (zh) 2010-09-01
NO20053142L (no) 2005-06-27
US20080187558A1 (en) 2008-08-07
PL218350B1 (pl) 2014-11-28
KR20050103187A (ko) 2005-10-27
JP5622594B2 (ja) 2014-11-12
AU2003283207C1 (en) 2010-07-29
WO2004047794A3 (fr) 2004-09-16
US9408805B2 (en) 2016-08-09
AU2010200798A1 (en) 2010-03-25
PL377745A1 (pl) 2006-02-20
US9415015B2 (en) 2016-08-16
EP2436395B1 (fr) 2017-04-26
EP1567123A2 (fr) 2005-08-31
JP2011093926A (ja) 2011-05-12
MXPA05005527A (es) 2006-04-05
US8329196B2 (en) 2012-12-11
US20160324767A1 (en) 2016-11-10
US10080719B2 (en) 2018-09-25
EP2436395A3 (fr) 2012-06-20
JP5442924B2 (ja) 2014-03-19
US10471008B2 (en) 2019-11-12
AU2003280324A1 (en) 2004-06-18
US20170367970A1 (en) 2017-12-28
CA2507551A1 (fr) 2004-06-10
KR20120034767A (ko) 2012-04-12
KR101281147B1 (ko) 2013-07-02
WO2004047793A1 (fr) 2004-06-10
MXPA05005528A (es) 2006-04-05
US20080193535A1 (en) 2008-08-14
JP2006513269A (ja) 2006-04-20
AU2010200798B2 (en) 2012-11-15
HK1087008A1 (en) 2006-10-06
IL168733A (en) 2011-01-31
IS7894A (is) 2005-06-14

Similar Documents

Publication Publication Date Title
US10471008B2 (en) Allergen dosage form
US8012505B2 (en) Dosage form having a saccharide matrix
CA2516291C (fr) Forme de dosage presentant une matrice en saccharide
ZA200505111B (en) An allergen dosage form
EP2167084B1 (fr) Forme de dosage d'allergène contenant une antihistamine
RU2372074C2 (ru) Лекарственная форма, содержащая аллерген

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALK-ABELLO A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JACOBI, HENRIK H.;LUNDEGAARD, ANNETTE R.;HOUGHTON, CHRISTIAN G.;AND OTHERS;REEL/FRAME:015261/0212;SIGNING DATES FROM 20040317 TO 20040323

AS Assignment

Owner name: R. P. SCHERER TECHNOLOGIES, INC., NEVADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MALLINDINE, KERRY;BANBURY, SUSAN GERRARD;WONG, DESMOND YIK TENG;AND OTHERS;REEL/FRAME:021058/0004;SIGNING DATES FROM 20080311 TO 20080409

AS Assignment

Owner name: R.P. SCHERER TECHNOLOGIES, INC., NEVADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MURRAY, OWEN J.;REEL/FRAME:021664/0534

Effective date: 20080715

AS Assignment

Owner name: R.P. SCHERER TECHNOLOGIES, LLC, NEVADA

Free format text: CHANGE OF NAME;ASSIGNOR:R.P. SCHERER TECHNOLOGIES, INC;REEL/FRAME:028572/0172

Effective date: 20090902

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: R. P. SCHERER TECHNOLOGIES, INC.., NEVADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MALLINDINE, KERRY;WONG, DESMOND YIK TENG;GARRETT, LISA MARIE;AND OTHERS;SIGNING DATES FROM 20080311 TO 20080409;REEL/FRAME:039027/0152

Owner name: ALK-ABELLO A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HOUGHTON, CHRISTIAN GAUGUIN;LUNDEGAARD, ANNETTE ROMMELMAYER;JACOBI, HENRIK HUGO;AND OTHERS;SIGNING DATES FROM 20040317 TO 20040323;REEL/FRAME:039026/0785

Owner name: R.P. SCHERER TECHNOLOGIES, INC., NEVADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MURRAY, OWEN J.;REEL/FRAME:039027/0402

Effective date: 20080715

Owner name: R.P. SCHERER TECHNOLOGIES, LLC, NEVADA

Free format text: CHANGE OF NAME;ASSIGNOR:R.P. SCHERER TECHNOLOGIES, INC.;REEL/FRAME:039186/0479

Effective date: 20090902