US20140120077A1 - Compositions and Methods for Safe Treatment of Rhinitis - Google Patents

Compositions and Methods for Safe Treatment of Rhinitis Download PDF

Info

Publication number
US20140120077A1
US20140120077A1 US14/065,379 US201314065379A US2014120077A1 US 20140120077 A1 US20140120077 A1 US 20140120077A1 US 201314065379 A US201314065379 A US 201314065379A US 2014120077 A1 US2014120077 A1 US 2014120077A1
Authority
US
United States
Prior art keywords
botulinum toxin
botulinum
rhinitis
topical
poloxamer
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
US14/065,379
Other languages
English (en)
Inventor
Curtis L. Ruegg
Hongran Fan Stone
Jacob M. Waugh
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.)
Revance Therapeuticals Inc
Original Assignee
Revance Therapeuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Revance Therapeuticals Inc filed Critical Revance Therapeuticals Inc
Priority to US14/065,379 priority Critical patent/US20140120077A1/en
Publication of US20140120077A1 publication Critical patent/US20140120077A1/en
Assigned to REVANCE THERAPEUTICS, INC. reassignment REVANCE THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RUEGG, CURTIS L., WAUGH, JACOB M., STONE, HONGRAN F.
Priority to US15/155,551 priority patent/US10201594B2/en
Priority to US16/272,998 priority patent/US20200345819A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • A61K38/4893Botulinum neurotoxin (3.4.24.69)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; 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
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M31/00Devices for introducing or retaining media, e.g. remedies, in cavities of the body
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/24Metalloendopeptidases (3.4.24)
    • C12Y304/24069Bontoxilysin (3.4.24.69), i.e. botulinum neurotoxin

Definitions

  • This invention relates to compositions and methods for treating rhinitis using topical botulinum toxin compositions. More particularly, the invention relates to treating the symptoms of rhinitis using intranasal administration of topical botulinum toxin compositions that contain a purified botulinum neurotoxin.
  • Rhinitis is a worldwide health problem associated with nasal inflammation and characterized by symptoms of congestion, rhinorrhea, sneezing and itching. Allergic rhinitis is the most common form of rhinitis and affects up to 30% of adults and 40% of children in the United States. Symptoms of both allergic as well as non-allergic types of rhinitis can significantly impair patients' quality of life. Moreover, allergic rhinitis often coexists with other atopic conditions, such as asthma, sinusitis and sleep apnea.
  • Rhinitis is induced by overstimulation of parasympathetic innervation of the nasal mucosal tissue via release of acetylcholine and inflammatory mediators such as vasoactive intestinal peptide (VIP).
  • VIP vasoactive intestinal peptide
  • Pharmacologic therapy e.g., anti-histamines, decongestants, corticosteroids, anti-cholinergics, etc.
  • allergic rhinitis e.g., allergic rhinitis, allergic immunotherapy, require either frequent (one or more times per day) administration often with side effects or a long-term process of desensitization with limited effectiveness for many patients.
  • Botulinum toxin type A is obtained from serotype A of Clostridium botulinum .
  • Botulinum toxin type A acts to block the release of acetycholine from the presynaptic nerve terminal with consequent induction of muscular paralysis. Based on this anti-cholinergic activity, botulinum toxin type A has been used widely in the treatment of muscle spasticity disorders, and for the cosmetic treatment of frown lines and wrinkles.
  • botulinum toxin type A blocks parasympathetic cholinergic transmission and has been used for treatment of glandular hypersecretory disorders, such as hyperhidrosis, Frey's syndrome, sialorrhea, epiphora, rhinorrhea, and sialadenitis. It has also been shown that botulinum toxin type A can be used to suppress electrically stimulated rhinorrhea in a dog model, which is consistent with recent reports demonstrating the effect of muscarinic cholinergic agents on canine nasal veins. Subsequent studies in animal models and in human patients with different types of rhinitis demonstrated that local application of botulinum toxin type A effectively reduced rhinitis symptoms.
  • Some of these references report delivering topical liquid formulations containing commercially available botulinum type A complexes by saturating a sponge or gauze packing with a topical botulinum toxin formulation and inserting the sponge or gauze packing into the nasal cavity.
  • the practical difficulties associated with this methodology raises certain procedural issues as well as safety concerns.
  • clostridial neurotoxins have been used in the treatment of sinusitis (see, e.g. U.S. Pat. No. 8,092,781, WO2011/084507).
  • Botulinum toxin type A in its native form, exists as part of a protein complex that is produced by C. botulinum bacteria.
  • the native protein complex contains, in addition to the botulinum toxin type A neurotoxin molecule, stabilizing hemaglutinin and non-hemaglutinin proteins (sometimes referred to as “accessory proteins”) which are capable of protecting the botulinum neurotoxin molecule from degradation in the harsh acidic environment of the stomach.
  • botulinum toxin type A formulations are made using native botulinum toxin protein complexes.
  • the accessory proteins will enable the botulinum toxin to be passed through the stomach to the small intestine, where it is absorbed into the bloodstream. If this occurs, systemic poisoning, paralysis, and even death result.
  • treatment of rhinitis using the presently available commercial botulinum toxin formulations in topical preparations has been disfavored, even though botulinum toxin has been reported to be able to bind to mucosal epithelial cells and become trancytosed across mucosa. (See, e.g., U.S. Pat. No. 8,092,781).
  • botulinum toxin While botulinum toxin has been reported to cross mucosa in the respiratory and gastrointestinal tract, botulinum toxin does not penetrate intact skin, owing to its size. See, e.g., S. Arnon et al., “Botulinum Toxin as a Biological Weapon—Medical and Public Health Management,” JAMA, Vol. 285, No. 8, p. 1059 ff. Accordingly, unlike delivering botulinum toxin to mucosa, one must take special measures to either disrupt the skin (see, e.g., U.S. Pre-Grant Publication No. 20070088248) or to enhance penetration of botulinum toxin via use of a carrier (see, e.g., U.S. Pat. No. 7,807,780, and U.S. Pre-Grant Publication Nos. 20050196414, 20070077259) when administering botulinum toxin transdermally.
  • the invention relates to a method for treating rhinitis.
  • the method involves intranasally administering a botulinum toxin composition to a patient in need of treatment for rhinitis.
  • the botulinum toxin composition is a topical composition that includes (i) purified botulinum toxin neurotoxin (ii) at least one carrier, and (iii) at least one viscosity modifier.
  • the botulinum toxin is a 150 kDa botulinum neurotoxin type A which contains little or no toxin accessory proteins or human/animal-derived components.
  • the carrier comprises a positively charged backbone with positively charged efficiency groups covalently attached thereto, and the viscosity modifier is a preferably a poloxamer.
  • the carrier and viscosity modifier help to retain the intranasal composition at the site of administration and minimize the possibility of drainage or leakage to other sites, e.g. down the throat.
  • FIG. 1 Graph indicating performance severity assessment score (PSA) of rats (mean ⁇ SEM) on days 3, 5 and 7 after treatment with control (filled bars) or reconstituted Formulation 22 (open bars).
  • PSA performance severity assessment score
  • FIG. 2 Effect of reconstituted Formulation 22 on inflammatory pathology associated with allergic rhinitis in nasal tissue. Histological staining of corresponding region of (a-c) left turbinate and (d-f) lateral nasal wall from three animals, (a,d) normal animal, (b, e) allergic control, and (c, f) allergic reconstituted Formulation 22 treated, respectively. The thickness of nasal mucosa was greater in (b, e) control animals than in (a, d) normal and (c, f) reconstituted Formulation 22-treated allergic animals.
  • FIG. 3 Immunohistochemical localization of vasoactive intestinal peptide in nasal turbinate. Serial sections from tissues shown in FIG. 2 , a-c, were prepared and stained for vasoactive intestinal peptide (VIP) expression (indicated by dark staining) Strong VIP expression was noted in (b) control animals when compared with (a) normal animals. (c) After reconstituted Formulation 22 treatment, VIP expression was down-regulated essentially to normal levels (original magnification, ⁇ 20).
  • VIP vasoactive intestinal peptide
  • FIG. 4 Graph indicating effects of botulinum neurotoxin type A (BoNTA) complex (900 U, ⁇ ), reconstituted Formulation 27 (27,500 U, ⁇ ) and saline control ( ⁇ ) on mean daily weight of guinea pigs.
  • Animals received a single intranasal dose of reconstituted Formulation 27, 27,500 U (OE); BoNTA complex, 900 U (F); or saline control (f).
  • Each symbol represents the average of three animals with SD shown by error bars.
  • Asterisks denote values for reconstituted Formulation 27 that were significantly reduced compared with same day values for control (p ⁇ 0.05).
  • the present invention is based, at least in part, on the discovery that certain botulinum toxin compositions need not be administered by injection for the treatment of rhinitis.
  • the botulinum toxin compositions according to the invention can be administered by intranasally applying a topical botulinum toxin formulation. It is believed that the present topical botulinum toxin compositions offer at least two advantages over the injectable botulinum toxin formulations in the prior art. First, the present compositions utilize purified botulinum neurotoxin that is substantially free of the accessory proteins, rather than botulinum toxin complexes as used in conventional injectable formulations.
  • botulinum neurotoxin is susceptible to degradation in a patient's gastrointestinal tract. Accordingly, the present topical botulinum toxin compositions are far less likely to cause systemic poisoning and death in the event that accidental oral ingestion occurs.
  • the at least one carrier and at least one viscosity modifier help to prevent migration of the topical composition from the site of administration, thereby minimizing the possibility of migration and inadvertent oral ingestion.
  • the carrier contributes to the localization of the botulinum toxin by facilitating transport of the toxin across the mucosa to its site of action and minimizing unwanted diffusion of the botulinum toxin away from the area in need of treatment.
  • the viscosity modifier contributes to creating a formulation that essentially stays in the location where it is applied. Accordingly, the topical botulinum toxin compositions of the present invention provide an improved safety profile over the previous injectable botulinum toxin compositions.
  • the invention is suitable for the treatment of all forms of rhinitis, non-limiting examples of which include infectious rhinitis, vasomotor rhinitis, allergic rhinitis, rhinitis medicamentosa, atrophic rhinitis, rhinitis sicca, and polypous rhinitis.
  • infectious rhinitis vasomotor rhinitis
  • allergic rhinitis rhinitis medicamentosa
  • atrophic rhinitis rhinitis sicca
  • polypous rhinitis polypous rhinitis.
  • the compositions and methods disclosed herein can be used to treat rhinitis or characteristic symptoms of the same, e.g. nasal inflammation, nasal congestion, rhinorrhea, sneezing and/or itching.
  • the invention may be used to treat rhinitis-like symptoms, such as those that occur following certain ear-nose-throat (ENT) procedures, a non-limiting example of which is rhinoplasty.
  • ENT ear-nose-throat
  • the rhinitis can be allergic rhinitis or non-allergic rhinitis, where the latter type of rhinitis can be either inflammatory or non-inflammatory rhinitis (e.g. vasomotor rhinitis). It is also contemplated that multiple forms of rhinitis can be treated simultaneously with the compositions and methods of the present invention.
  • compositions and methods of the present invention can be used to treat a patient suffering from both allergic rhinitis and vasomotor rhinitis, a condition that is commonly referred to as “mixed rhinitis.”
  • the methods can also be used to treat rhinitis when it occurs in patients who have a condition in combination with other atopic conditions, such as asthma, sinusitis and sleep apnea.
  • the topical botulinum toxin compositions contemplated by the invention can be administered using an applicator that is inserted into a patients' nostril.
  • the topical administration may be achieved using visual guidance with the assistance of a nasal speculum and/or headlamp as necessary.
  • the topical composition is applied with the applicator to inner surfaces of the nose, non-limiting examples of which include the surfaces of the inferior turbinate, middle turbinate and the superior turbinate.
  • the topical botulinum toxin compositions may be administered via one nostril or via both nostrils, if deemed necessary.
  • Application of the topical composition may be achieved, for example, by using a custom applicator, such as the one described in U.S.
  • the topical composition may be applied first to an implement, such as a swab, which is then used to spread the topical composition over the area in need of treatment.
  • an implement such as a swab
  • topical botulinum toxin compositions of the invention are administered, they optionally may be allowed to remain in place for a certain dwell period in order to increase the amount of botulinum neurotoxin that is delivered to the nasal tissues in need of treatment.
  • the specific dwell time that is selected will depend on factors such as the severity of the rhinitis, the desired amount of botulinum toxin to be delivered, the concentration of the botulinum neurotoxin in the topical botulinum toxin composition, the concentration of the carrier in the topical botulinum toxin composition, and the viscosity of the topical botulinum toxin composition.
  • the dwell times may range from 5 seconds to 60 minutes, 30 seconds to 45 minutes, 1 minute to 30 minutes, 5 minutes to 20 minutes or 10 minutes to 15 minutes.
  • the dwell time may be 10, 20, 30, 40 or 50 minutes.
  • purified botulinum neurotoxin refers to any of the known types of purified botulinum neurotoxin, whether produced by the bacterium or by recombinant techniques, as well as any such types that may be subsequently discovered, including engineered variants or fusion proteins.
  • the purified botulinum neurotoxin may be a compound that has toxin activity but contains one or more chemical or functional alterations on any part or on any chain relative to naturally occurring or recombinant native neurotoxins.
  • the purified botulinum neurotoxin may be a modified neurotoxin that has at least one of its amino acids deleted, modified or replaced, as compared to a native neurotoxin molecule while maintaining significant neurotoxin activity.
  • modified botulinum neurotoxins obtained from a native botulinum neurotoxin with one or more of its amino acids replaced by conservative substitutions are expressly contemplated by the invention.
  • the purified botulinum neurotoxin may be one that has been modified in a way that, for instance, enhances its properties or decreases undesirable side effects, but that still retains the desired botulinum toxin activity.
  • the purified botulinum neurotoxin may be prepared using recombinant or synthetic chemical techniques (e.g.
  • a recombinant peptide, a fusion protein, or a hybrid neurotoxin as prepared from subunits or domains of different botulinum toxin serotypes (see, e.g., U.S. Pat. No. 6,444,209, the contents of which are incorporated by reference in their entirety).
  • the purified botulinum neurotoxin may also be a portion of the overall molecule that has been shown to possess the necessary botulinum toxin neurotoxin activity, and in such case may be used per se or as part of a combination or conjugate molecule, for instance a fusion protein.
  • the purified botulinum neurotoxin may be in the form of a purified botulinum neurotoxin precursor, which may itself be non-toxic, for instance a nontoxic zinc protease that becomes toxic on proteolytic cleavage.
  • the purified botulinum neurotoxin is obtained by isolating it from C. botulinum bacteria.
  • the invention expressly contemplates the use of purified botulinum neurotoxins selected from the group of clostridial neurotoxins consisting of serotypes A, B, C1, D, E, F, G and combinations thereof.
  • the purified botulinum neurotoxin is present as an isolated botulinum neurotoxin (e.g., purified botulinum toxin type A protein) that is stabilized by exogenous stabilizers.
  • the compositions are substantially free of the accessory proteins normally found in native botulinum toxin complexes.
  • the botulinum neurotoxin is sufficiently free of accessory proteins that the neurotoxin is susceptible to degradation in the gastrointestinal tract if ingested.
  • botulinum neurotoxin may be substantially free of accessory proteins if at least 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% of the accessory proteins found in the native botulinum toxin complexes have been removed, with each of the ranges from these specifically enumerated lower limits to 100% being a distinct embodiment expressly contemplated by the invention.
  • the purified botulinum neurotoxin is entirely free of the neurotoxin-associated proteins characteristic of the 900 kDa botulinum toxin complex.
  • Stabilized botulinum neurotoxin formulations containing such exogenous stabilizers have been reported, for example, in U.S. Pre-Grant Publication 20100330123 entitled “Albumin Free Botulinum Toxin Formulations,” which is hereby incorporated by reference in its entirety.
  • the purified botulinum neurotoxins contemplated by the invention are less toxic with respect to oral ingestion than native botulinum toxin, owing to the removal of some or all of the accessory proteins normally accompanying the native botulinum neurotoxin.
  • the invention contemplates removing a sufficient amount of the native accessory proteins to obtain a purified botulinum neurotoxin that is 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold or 90-fold less toxic with respect to oral ingestion than native botulinum toxin.
  • the purified botulinum neurotoxin may be 10-fold to 90-fold less toxic, 20-fold to 80-fold less toxic, 30-fold to 70-fold less toxic, or 40-fold to 60-fold less toxic with respect to oral ingestion.
  • the purified botulinum toxin may be 50-fold to 95-fold less toxic, 60-fold to 90-fold less toxic, or 70-fold to 85-fold less toxic with respect to oral ingestion than the native botulinum toxin.
  • the relative toxicities of botulinum neurotoxin-containing formulations may be determined using test animal studies, as described herein.
  • Removal of the accessory proteins may be accomplished using any methods known in the art, such as treating the native botulinum toxin complexes with red blood cells at a pH of 7.3, or by using separation methods such as chromatography (See, e.g., U.S. Pre-Grant Publication No. 20110092682, the contents of which are incorporated by reference in their entirety).
  • the purified botulinum neurotoxin is present in the topical botulinum toxin compositions of the invention in an effective amount to treat rhinitis.
  • the term “effective amount” refers to an amount of purified botulinum neurotoxin that is sufficient to ameliorate one or more symptoms of rhinitis safely.
  • the amount of purified botulinum neurotoxin may range from 250 U/mL to 50,000 U/mL, 500 U/mL to 25,000 U/mL, 2,500 U/mL to 12,500 U/mL, or 5,000 U/mL to 10,000 U/mL.
  • effective amounts for the purified botulinum neurotoxins disclosed herein may range from 50 U/mL to 450,000 U/mL, preferably from 5,000 U/mL-400,000 U/mL, more preferably from 10,000 U/mL-150,000 U/mL, and even more preferably from 20,000 U/mL to 125,000 U/mL.
  • the effective amount of purified botulinum toxin may range from 20,000 U/mL to 40,000 U/mL, 45,000 U/mL to 95,000 U/mL, or 100,000 U/mL to 200,000 U/mL.
  • the invention also contemplates topical botulinum toxin compositions that contain purified botulinum neurotoxin present in the composition a concentration that ranges from about 5 ng/mL to about 50 ng/mL, such as, for example about 5 ng/mL, about 10 ng/mL, about 15 ng/mL, 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 45 ng/mL or about 50 ng/mL.
  • the specific amount of purified botulinum neurotoxin in a given topical formulation will depend on the chosen carrier and viscosity modifier, and can be readily determined by a person of skill in the art.
  • the concentration and volume of the topical botulinum toxin composition are selected so as to provide a dose of botulinum toxin in the range of 1 ng to 50 ng, or 5 ng to 40 ng, or 10 ng to 30 ng, or 15 ng to 25 ng.
  • the dose of botulinum toxin can be in a range from 1 ng to 10 ng, 2 ng to 8 ng, or 3 ng to 6 ng.
  • the dose of botulinum toxin optionally may be in the range of 0.5 ng to 5 ng, 1 ng to 5 ng, or 2 ng to 4 ng.
  • the dose of botulinum toxin in certain preferred embodiments is 1, 2, 3, 4, or 5 ng.
  • the volume of the applied topical botulinum toxin composition is in the range of 0.02-0.5 mL, 0.05 to 0.4 m, 0.07 to 0.3 mL or 0.09 to 0.2 mL, and the concentration of the topical botulinum toxin is selected to provide a dose of 1 to 10 ng, or 1 to 5 ng.
  • the topical botulinum toxin compositions of the invention preferably contains a carrier to promote the transport of the purified botulinum neurotoxin into the nasal tissues in need of treatment.
  • a carrier to promote the transport of the purified botulinum neurotoxin into the nasal tissues in need of treatment.
  • the invention contemplates the use of carriers that promote the transport of botulinum toxin from applied topical compositions.
  • the carrier may be a liquid chemical carrier, such as dimethyl sulfoxide (DMSO).
  • DMSO dimethyl sulfoxide
  • the carrier may also be in the form of a nanoemulsion as described in WO2008/045107.
  • the carrier may comprise sphingosine and/or cerebroside as described in U.S. Pre-Grant Publication No. 20060182766.
  • the carrier may also comprise a sialoprotein, such as those described, for example in U.S. Pre-Grant Publication No. 20070116724. It is to be understood that the foregoing references, like all references cited herein, are incorporated by reference in their entirety.
  • the topical compositions according to the invention comprise a carrier that includes a positively charged carrier molecule with positively charged efficiency groups attached thereto, as is disclosed, for example, in U.S. Pat. No. 8,398,997, which is hereby incorporated by reference in its entirety.
  • a positively charged carrier molecule with positively charged efficiency groups attached thereto, as is disclosed, for example, in U.S. Pat. No. 8,398,997, which is hereby incorporated by reference in its entirety.
  • positively charged it is meant that the carrier molecule has a positive charge under at least some solution-phase conditions, more preferably under at least some physiologically compatible conditions.
  • the term “positively charged,” as used herein, embraces functionalities that are charged under all pH conditions, for instance, a quaternary amine, as well as functionalities which can acquire positive charge under certain solution-phase conditions, such as pH changes, in the case of primary amines.
  • “positively charged” as used herein refers to those groups that have the behavior of associating with anions over physiologically compatible conditions.
  • Polymers with a multiplicity of positively-charged moieties need not be homopolymers, as will be apparent to one skilled in the art.
  • Other examples of positively charged moieties are well known in the prior art and can be employed readily, as will be apparent to those skilled in the art.
  • the positively charged carriers of the invention help to electrostatically anchor the botulinum toxin to the treated mucosa, thereby reducing unwanted diffusion, reducing unwanted drainage of the topical formulation into the throat or airway, and increasing efficacy.
  • the positively charged carrier molecule comprises a “positively charged backbone,” which is typically a linear chain of atoms, either with groups in the chain carrying a positive charge at physiological pH, or with groups carrying a positive charge attached to side chains extending from the backbone.
  • the positively charged backbone itself will not have a defined enzymatic or therapeutic biologic activity.
  • the linear backbone is preferably a hydrocarbon backbone which is, in some embodiments, interrupted by heteroatoms selected from nitrogen, oxygen, sulfur, silicon and phosphorus. The majority of backbone chain atoms are usually carbon.
  • the backbone will often be a polymer of repeating units (e.g., amino acids, poly(ethyleneoxy), poly(propyleneamine), polyalkyleneimine, and the like) but can be a heteropolymer.
  • the positively charged backbone is a polypropyleneamine wherein a number of the amine nitrogen atoms are present as ammonium groups (tetra-substituted) carrying a positive charge.
  • the positively charged backbone will have a molecular weight that ranges from 100-2,500,000, more preferably, 200-2,000,000, even more preferably, 300-500,000, 400-100,000, 500-50,000, 600-20,000 or 700-8,000.
  • the positively charged backbone is a nonpeptidyl polymer, which may be a hetero- or homo-polymer such as a polyalkyleneimine, for example a polyethyleneimine or polypropyleneimine, having a molecular weight of from about 500 to about 2,500,000, preferably from about 100,000 to about 1,800,000, and most preferably from about 500,000 to about 1,400,000.
  • the nonpeptidyl polymer may have a molecular weight in the range from about 500 to about 5000, about 1000 to about 4000, about 1500 to about 3500, or about 2000 to about 3000.
  • the backbone may have attached thereto a plurality of side-chain moieties that include positively charged groups (e.g., ammonium groups, pyridinium groups, phosphonium groups, sulfonium groups, guanidinium groups, or amidinium groups).
  • the sidechain moieties in this group of embodiments can be placed at spacings along the backbone that are consistent in separations or variable. Additionally, the length of the sidechains can be similar or dissimilar.
  • the sidechains can be linear or branched hydrocarbon chains having from one to twenty carbon atoms and terminating at the distal end (away from the backbone) in one of the above-noted positively charged groups.
  • the association between the carrier and the chemodenervating agent is by non-covalent interaction, non-limiting examples of which include ionic interactions, hydrogen bonding, van der Waals forces, or combinations thereof.
  • the positively charged backbone is a polypeptide having multiple positively charged sidechain groups (e.g., lysine, arginine, ornithine, homoarginine, and the like).
  • the polypeptide may a molecular weight of from about 500 to about 1,500,000, about 10,000 to about 1,500,000, more preferably from about 25,000 to about 1,200,000, most preferably from about 100,000 to about 1,000,000.
  • the polypeptide may have a molecular weight in the range from about 500 to about 5,000, about 1,000 to about 4,000, about 1,500 to about 3,500, or about 2,000 to about 3,000.
  • the sidechains can have either the D- or L-form (R or S configuration) at the center of attachment.
  • the backbone can be an analog of a polypeptide such as a peptoid. See, for example, Kessler, Angew. Chem. Int. Ed. Engl. 32:543 (1993); Zuckermann et al. Chemtracts-Macromol. Chem. 4:80 (1992); and Simon et al. Proc. Nat'l. Acad. Sci. USA 89:9367 (1992)).
  • a peptoid is a polyglycine in which the sidechain is attached to the backbone nitrogen atoms rather than the ⁇ -carbon atoms. As above, a portion of the sidechains will typically terminate in a positively charged group to provide a positively charged backbone component. Synthesis of peptoids is described in, for example, U.S. Pat. No. 5,877,278, which is hereby incorporated by reference in its entirety. As the term is used herein, positively charged backbones that have a peptoid backbone construction are considered “non-peptide” as they are not composed of amino acids having naturally occurring sidechains at the ⁇ -carbon locations.
  • a variety of other backbones can be used employing, for example, steric or electronic mimics of polypeptides wherein the amide linkages of the peptide are replaced with surrogates such as ester linkages, thioamides (—CSNH—), reversed thioamide (—NHCS—), aminomethylene (—NHCH 2 —) or the reversed methyleneamino (—CH 2 NH—) groups, keto-methylene (—COCH 2 —) groups, phosphinate (—PO 2 RCH 2 —), phosphonamidate and phosphonamidate ester (—PO 2 RNH—), reverse peptide (—NHCO—), trans-alkene (—CR ⁇ CH—), fluoroalkene (—CF ⁇ CH—), dimethylene (—CH 2 CH 2 —), thioether (—CH 2 S—), hydroxyethylene (—CH(OH)CH 2 —), methyleneoxy (—CH 2 O—), tetrazole (CN 4 ), sulf
  • sidechain groups can be appended that carry a positively charged group, preferably at physiologic pH.
  • the sulfonamide-linked backbones (—SO 2 NH— and —NHSO 2 —) can have sidechain groups attached to the nitrogen atoms.
  • the hydroxyethylene (—CH(OH)CH 2 —) linkage can bear a sidechain group attached to the hydroxy substituent.
  • linkage chemistries to provide positively charged sidechain groups using standard synthetic methods.
  • the positively charged backbone is a polypeptide having efficiency groups.
  • an efficiency group is any agent that has the effect of promoting the translocation of the positively charged backbone through a tissue or cell membrane.
  • efficiency groups include -(gly) n1 -(arg) n2 , HIV-TAT or fragments thereof, or the protein transduction domain of Antennapedia, or a fragment thereof, in which the subscript n1 is an integer of from 0 to 20, more preferably 0 to 8, still more preferably 2 to 5, and the subscript n2 is independently an odd integer of from about 5 to about 25, more preferably about 7 to about 17, most preferably about 7 to about 13. In a preferred embodiment n1 is 3 and n2 is 7.
  • the efficiency group has the formula (gly) p -RGRDDRRQRRR-(gly) q , (gly) p -YGRKKRRQRRR-(gly) q or (gly) p -RKKRRQRRR-(gly) q wherein the subscripts p and q are each independently an integer of from 0 to 20 and the fragment is attached to the backbone via either the C-terminus or the N-terminus of the fragment.
  • Preferred efficiency groups are those in which the subscripts p and q are each independently integers of from 0 to 8, more preferably 2 to 5.
  • the carrier has the amino acid sequence selected from the group consisting of RKKRRQRRR-G-(K) 15 -G-RKKRRQRRR, RKKRRQRRR-G-(K) 20 -G-RKKRRQRRR, RKKRRQRRR-G-(K) 25 -G-RKKRRQRRR, RKKRRQRRR-G-(K) 30 -G-RKKRRQRRR, RGRDDRRQRRR-G-(K) 15 -G-RGRDDRRQRRR, RGRDDRRQRRR-G-(K) 20 -G-RGRDDRRQRRR, RGRDDRRQRRR-G-(K) 25 -G-RGRDDRRQRRR, RGRDDRRQRRR-G-(K) 30 -G-RGRDDRRQRRR, YGRKKRRQRRR-G-(K) 15 -G-YGRKKRRQRRR, YGRKKRRQRR-R-(K)
  • the positively charged efficiency group is the Antennapedia (Antp) protein transduction domain (PTD), or a fragment thereof that retains activity.
  • PTD Antennapedia protein transduction domain
  • the positively charged carrier includes side-chain positively charged efficiency groups in an amount of at least about 0.05%, as a percentage of the total carrier weight, preferably from about 0.05 to about 45 weight %, and most preferably from about 0.1 to about 30 weight %.
  • the most preferred amount is from about 0.1 to about 25%.
  • the backbone portion is a polylysine and positively charged efficiency groups are attached to the lysine sidechain amino groups.
  • the polylysine may have a molecular weight that ranges from about 10,000 to about 1,500,000, preferably from about 25,000 to about 1,200,000, and most preferably from about 100,000 to about 1,000,000.
  • the polylysine may have a molecular weight that ranges from about 500 to about 5000, about 1000 to about 4000, about 1500 to about 3500, or about 2000 to about 3000.
  • the polylysine may be any of the commercially available (Sigma Chemical Company, St.
  • polylysines such as, for example, polylysine having MW>70,000, polylysine having MW of 70,000 to 150,000, polylysine having MW 150,000 to 300,000 and polylysine having MW>300,000.
  • the selection of an appropriate polylysine will depend on the remaining components of the composition and will be sufficient to provide an overall net positive charge to the composition and, in some embodiments, provide a length that is preferably from one to four times the combined length of the negatively charged components.
  • Preferred positively charged efficiency groups or efficiency groups include, for example, -gly-gly-gly-arg-arg-arg-arg-arg (-Gly 3 Arg 7 ) or HIV-TAT fragments, as disclosed herein.
  • the positively charged backbone is a long chain polyalkyleneimine such as a polyethyleneimine or polypropyleneimine.
  • Such polyalkyleneimines for example, may have a molecular weight of about 1,000,000.
  • the carrier is a relatively short polylysine or polyethyleneimine (PEI) backbone (which may be linear or branched) and which has positively charged branching groups.
  • PI polyethyleneimine
  • Such carriers are useful for minimizing uncontrolled aggregation of the backbones and botulinum toxin in a therapeutic composition, which causes the transport efficiency to decrease dramatically.
  • the carrier is a relatively short linear polylysine or PEI backbone
  • the backbone will have a molecular weight of less than 75,000, more preferably less than 30,000, and most preferably, less than 25,000.
  • the backbone When the carrier is a relatively short branched polylysine or PEI backbone, however, the backbone will have a molecular weight less than 60,000, more preferably less than 55,000, and most preferably less than 50,000. For example, if desired, the backbone may have a molecular weight that ranges from about 500 to about 5000, about 1000 to about 4000, about 1500 to about 3500, or about 2000 to about 3000.
  • the topical botulinum toxin compositions for treating rhinitis according to the methods described herein typically also include a viscosity modifier to maintain the location of the applied compositions at the treatment area, to restrict the movement of the compositions from the intended treatment area after application, and/or to minimize or prevent diffusion and inadvertent oral and/or systemic exposure.
  • a viscosity modifier to maintain the location of the applied compositions at the treatment area, to restrict the movement of the compositions from the intended treatment area after application, and/or to minimize or prevent diffusion and inadvertent oral and/or systemic exposure.
  • the chemical identity of the viscosity modifier is not particularly limited, and may be any pharmaceutically acceptable composition with the appropriate viscosity at the body surface temperature at the area of application, so long as the viscosity modifier is compatible with other components of the topical botulinum toxin compositions, as described herein.
  • the viscosity modifier according to the invention may be chosen such that the viscosity of the topical botulinum toxin composition falls within a viscosity range of 10,000-500,000 cps, more preferably, 15,000-250,000 cps, even more preferably 20,000 to 200,000 cps and most preferably 25,000 to 100,000 cps at 25° C.
  • the viscosity modifier may be chosen such that the viscosity of the topical botulinum toxin composition is ⁇ 1000 cps at 26° C., such as, for example, from 1,000-10,000 cps, 1,500-8,000 cps, 2,000-6,000 cps, or 2,500-5,000 cps.
  • the required amount of viscosity modifying agent can be determined readily by a person of skill in the art, based on the disclosures set forth herein.
  • the viscosity modifier optionally may be a surfactant.
  • the surfactant may be selected from anionic surfactants, cationic surfactants, zwitterionic surfactants or non-ionic surfactants. In certain embodiments, one or more non-ionic surfactants serve as the viscosity modifier.
  • the non-ionic surfactant can be any commercially available non-ionic surfactant, such as, for example, polyoxyethylene glycol alkyl ethers, polyoxypropylene glycol alkyl ethers, glucoside alkyl ethers, polyoxyethylene glycol octylphenol ethers, polyoxyethylene glycol alkylphenol ethers, glycerol alkyl esters, polyoxyethylene glycol sorbitan alkyl esters, sorbitan alkyl esters, dodecyldimethylamine oxide, block copolymers of polyethylene glycol and polypropylene glycol (polyoxamers) and combinations thereof.
  • non-ionic surfactant such as, for example, polyoxyethylene glycol alkyl ethers, polyoxypropylene glycol alkyl ethers, glucoside alkyl ethers, polyoxyethylene glycol octylphenol ethers, polyoxyethylene glycol alkylphenol ethers, gly
  • the non-ionic surfactant is a polysorbate, non-limiting examples of which include polysorbate 20, polysorbate 40, polysorbate 60, and polysorbate 80.
  • the non-ionic surfactant is a sorbitan ester, non-limiting examples of which include Span 20, Span 60, Span 65, and Span 80.
  • the invention also contemplates using Triton X-100, trileucine, or NP-40 as the non-ionic surfactants.
  • the combinations of different non-ionic surfactants are contemplated.
  • the non-ionic surfactant is selected from the group consisting of polysorbates, poloxamers, and sorbitans, with polysorbates and sorbitans being particularly preferred.
  • the viscosity modifier can be a poloxamer.
  • poloxamers may be linear or branched, and include tri-blocks or tetra-blocks copolymers. They include poloxamines such as Tetronic and Pluronic.
  • the poloxamer may be chosen from poloxamer 101, poloxamer 105, poloxamer 108, poloxamer 122, poloxamer 123, poloxamer 124, poloxamer 181, poloxamer 182, poloxamer 183, poloxamer 184, poloxamer 185, poloxamer 188, poloxamer 212, poloxamer 215, poloxamer 217, poloxamer 231, poloxamer 234, poloxamer 235, poloxamer 237, poloxamer 238, poloxamer 282, poloxamer 284, poloxamer 288, poloxamer 331, poloxamer 333, poloxamer 334, poloxamer 335, poloxamer 338, poloxamer 401, poloxamer 402, poloxamer 403, poloxamer 407, and combinations thereof.
  • the poloxamer that is chosen has a tendency to form a gel with increasing temperature.
  • the amount of viscosity modifying agent that is present in the topical botulinum toxin compositions of the invention will depend on the identity of the viscosity modifying agent, as well as the desired viscosity of the topical botulinum toxin composition.
  • suitable concentrations for viscosity modifying agent in the topical botulinum toxin compositions of the invention may range from about 5% and about 70% (wt/wt), such as, for example, between about 5% and about 60%, between about 10% and about 50%, between about 15% and about 40%.
  • the viscosity modifier is present in the compositions in concentration between about 15% and about 20%, such as, for example, about 16%, about 17%, about 18%, about 19% or about 20%.
  • the viscosity modifying agent is present in a concentration of 15.0%, 15.5%, 16.0%, 16.5%, 17.0%, 17.5%, 18.0%, 18.5%, 19.0%, 19.5%, or 20% and is selected from the group consisting of poloxamer 212, poloxamer 215, poloxamer 217, poloxamer 231, poloxamer 234, poloxamer 235, poloxamer 237, poloxamer 238, poloxamer 282, poloxamer 284, poloxamer 288, poloxamer 331, poloxamer 333, poloxamer 334, poloxamer 335, poloxamer 338, poloxamer 401, poloxamer 402, poloxamer 403, poloxamer 407, and combinations thereof.
  • the amount of poloxamer present in the formulations of the invention may range from 15-25%, 15.5-24.5%, 16-23%, 16.5-22.5%, 17-22% 17.5%-21.5%, or 18%-21%.
  • the amount of poloxamer 407 may be 15.5%, 16.0%, 16.5%, 17.0%, 17.5%, 18.0%, 18.5%, 19.0%, 19.5%, 20%, 20.5%, 21% 21.5% or 22%.
  • the chosen poloxamer may be poloxamer 188, which may be present in the formulations of the invention at a concentration range of 15.5%, 16.0%, 16.5%, 17.0%, or 17.5%.
  • This invention also specifically contemplates adding more than one type of poloxamer to modify the viscosity of the formulation. For instance, if desired, both poloxamer 188 and poloxamer 407 may be added to the formulations of the invention to modify the viscosity.
  • the topical formulations according to the present invention may also comprise stabilizing agents.
  • stabilizing agents may include non-reducing sugars, non-limiting examples of which include trehalose and sucrose.
  • the non-reducing sugar has a glass transition temperature above 55° C., 57° C., or 60° C. Without wishing to be bound by theory, it is believed that such glass transition temperatures are sufficiently high to suppress undesirable molecular motions that cause the botulinum toxin to denature.
  • the non-reducing sugar is a disaccharide, non-limiting examples of which include trehalose and sucrose.
  • the non-reducing sugar is a trisaccharide, a non-limiting example of which is raffinose.
  • concentration of the non-reducing sugar in the botulinum toxin formulations of the invention are in the range of 10% to 40% (w/v), preferably 10% to 25% (w/v), more preferably 15% to 20% (w/v).
  • concentration of the non-reducing sugar is 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19% or 20% (w/v).
  • the non-reducing sugar is trehalose
  • preferably the hydrated form of trehalose i.e., trehalose-dihydrate
  • an agent may be added to increase the mechanical strength of amorphous glass solid cake that is formed when the formulation is lyophilized.
  • a non-limiting example of such an agent includes boric acid.
  • the topical botulinum toxin formulations of the invention comprise a buffer.
  • any physiologically compatible buffer capable of maintaining the pH in the range of 4.5 to 6.5, more preferably in the range of 5 to 6, and most preferably about 5.5, is suitable for the botulinum toxin formulations of the invention.
  • buffers include those involving salts of citric acid, acetic acid, succinic acid, tartaric acid, maleic acid, and histidine.
  • suitable buffer concentrations include buffer concentrations in the range of 0.400% to 0.600%; 0.450% to 0.575%, or 0.500% to 0.565%.
  • the invention also contemplates botulinum toxin formulations comprising a mixture of buffer salts, non-limiting examples of which include citrate/acetate, citrate/histidine, citrate/tartrate, maleate/histidine, or succinate/histidine.
  • the buffer is phosphate buffer.
  • the topical botulinum formulations may also comprise an anti-oxidant that acts as a preservative.
  • preservatives include butylhydroxytoluene (BHT), methionine, and propyl gallate.
  • BHT butylhydroxytoluene
  • methionine methionine
  • propyl gallate One or more of such preservatives may be present in the topical botulinum toxin formulations of the invention in a concentration that ranges from 0.3-2.0%, from 0.5%-1.5%, or from 0.75%-1.25%.
  • the methods described herein can be incorporated into various treatment regimens.
  • the methods for treating rhinitis described herein may be performed on a given patient every one week, two weeks, three weeks, month, two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months or twelve months.
  • the length of time between applications will vary depending on many factors, including the specific purified botulinum toxin chosen, the dosage (which depends on the concentration of topical composition and the dwell time) and the severity of the rhinitis.
  • purified botulinum toxin type A may be administered every three or four months for treating rhinitis, as needed.
  • compositions having a botulinum toxin concentration of 1,000-50,000 U/mL may be administered.
  • compositions having a concentration of 2,000 U/mL, 3000, U/mL, 4,000 U/mL, 5,000 U/mL, 6,000 U/mL, 7,000 U/mL, 8,000 U/mL or 9,000 U/mL may be topically applied to the nasal cavity as described herein for a dwell time of 10, 20, 30, 40, or 50 minutes.
  • topical compositions having a botulinum toxin concentration of 5,500 U/mL, 5,750 U/mL, 6,000 U/mL, 6,250 U/mL, 6,500 U/mL, 6,750 U/mL or 7,000 U/mL may be topically applied to the nasal cavity as described herein for a dwell time of 10, 20, 30, 40, or 50 minutes.
  • topical compositions having a botulinum toxin concentration of 15,500 U/mL, 15,750 U/mL, 16,000 U/mL, 16,250 U/mL, 16,500 U/mL, 16,750 U/mL or 17,000 U/mL may be topically applied to the nasal cavity as described herein for a dwell time of 10, 20, 30, 40, or 50 minutes.
  • One particularly suitable treatment involves applying 0.2 mL of a topical botulinum toxin type A composition containing 6,250 U/mL for a dwell time of 30 minutes. After the dwell time has elapsed, the topical botulinum toxin may be flushed out of the patient's nasal cavity by irrigating the nasal cavity with saline solution.
  • the resulting effluent contains botulinum toxin and is highly toxic.
  • the effluent is collected (e.g., absorbed by an absorbent article) and the botulinum toxin therein is reacted with an agent to render it no longer toxic.
  • Suitable methods and agents for inactivating botulinum toxin include those found, for example, in U.S. patent application Ser. No. 13/334,283, which is hereby incorporated by reference in its entirety.
  • the topical formulations of the invention optionally may be manufactured in a lyophilized form and then reconstituted with a diluent before administration.
  • the lyophilized form may be produced by lyophilizing a liquid composition comprising a botulinum toxin, a non-reducing disaccharide or a non-reducing trisaccharide, a non-ionic surfactant, and a physiologically compatible buffer.
  • the concentration of the non-reducing disaccharide or non-reducing trisaccharide is in the range of 10% to 40% (w/v)
  • the concentration of the non-ionic surfactant is in the range of 0.005% to 0.5% (w/v)
  • the pH of the liquid composition is in the range of 4.5 to 6.5.
  • any pharmaceutically acceptable diluent that does not undergo undesirable reactions with the components of the formulation in question may be used.
  • the formulations may be reconstituted using water, saline, or phosphate buffered saline.
  • one or more additives may be included in the diluent to control or improve certain properties of the diluent, non-limiting examples of which include viscosity enhancers (e.g., a poloxamer, such as poloxamer 188 or 407), anti-oxidants (e.g., BHT or methionine), co-solvents (e.g., an alcohol, such as ethanol), and/or tonicity adjusters (e.g., a salt, such as sodium chloride).
  • exemplary diluents that are suitable for use with the invention include those listed in Tables 3 and 3-1, as set forth herein. For lyophilized formulations that are stored in 2 ml vials, it is often convenient to reconstitute the formulations using 1 ml of diluent, non-limiting examples of which include those shown in Tables 3 and 3-1.
  • Table 1 shows 35 exemplary lyophilized topical botulinum toxin formulations that were prepared in accordance with the present invention. Each formulation was prepared by adding the respective components in the indicated amounts to a standard 2 ml lyophilization vial.
  • the column heading “Toxin (ng/vial)” refers to the amount of botulinum toxin type A neurotoxin present (in nanograms per vial), while the column heading “peptide (mg/ml)” refers to the amount of the peptide RKKRRQRRR-Q-(K) 15 -Q-RKKRRQRRR present (in milligrams per vial).
  • Table 1-1 shows the same 35 exemplary lyophilized topical botulinum toxin formulations as in Table 1, except that the numerical entries in Table 1-1 refer to the respective weight percent of the components relative to the total weight of the lyophilized formulation.
  • Table 2 shows twelve exemplary lyophilized formulations that were prepared according to the invention. Each formulation was prepared by adding the respective components in the indicated amounts to a standard 2 ml lyophilization vial.
  • the column heading “Toxin (ng/vial)” refers to the amount of botulinum toxin type A neurotoxin present (in nanograms per vial), while the column heading “peptide (mg/ml)” refers to the amount of the peptide RKKRRQRRR-Q-(K) 15 -Q-RKKRRQRRR present (in milligrams per vial).
  • Table 2-1 shows the same twelve exemplary lyophilized topical botulinum toxin formulations as in Table 2, except that the numerical entries in Table 2-1 refer to the respective weight percent of the components relative to the total weight of the lyophilized formulation.
  • the lyophilized formulations listed in Tables 1 and 2 generally are used after reconstitution with a diluent.
  • a diluent any pharmaceutically acceptable diluent that does not undergo undesirable reactions with the components contained in the respective formulation may be used.
  • the formulations listed in Tables 1 and 2 may be reconstituted using water, saline, or phosphate buffered saline.
  • one or more additives may be included in the diluent to control or improve certain properties of the diluent, non-limiting examples of which include viscosity enhancers (e.g., a poloxamer, such as poloxamer 188 or 407), anti-oxidants (e.g., BHT or methionine), co-solvents (e.g., an alcohol, such as ethanol), and/or tonicity adjusters (e.g., a salt, such as sodium chloride).
  • viscosity enhancers e.g., a poloxamer, such as poloxamer 188 or 407
  • anti-oxidants e.g., BHT or methionine
  • co-solvents e.g., an alcohol, such as ethanol
  • tonicity adjusters e.g., a salt, such as sodium chloride
  • the topical formulations of the invention contain botulinum neurotoxin, a carrier peptide, a buffer, a sugar, and a non-ionic surfactant.
  • certain useful topical formulations may contain 4.1 ⁇ 10 ⁇ 6 -0.22 wt. % botulinum neurotoxin type A, 2.5 ⁇ 10 ⁇ 5 -0.1 wt. % of the peptide RKKRRQRRR-Q-(K) 15 -Q-RKKRRQRRR, 1-1.5 wt. % histidine, 0.1-0.5 wt. % histidine hydrochloride, 97-99 wt. % sucrose, and 0.1-0.3 wt. % polysorbate 20. If desired, such formulations may be reconstituted using the diluent set forth in Table 3.
  • Formulation 22 reported in Table 1 above (reconstituted with the diluent reported in Table 3), a control gel (Revance Therapeutics, Inc., Newark, Calif.), ovalbumin, aluminum hydroxide, gelatin, sodium phosphate, diaminobenzadine (Sigma-Aldrich, St. Louis, Mo.), sheep anti-VIP antibody (Millipore, Billerica, Mass.), biotinylated rabbit anti-sheep antibody and ABC kit (Vector Labs, Burlingame, Calif.), Botulinum toxin type A complex stock solution (Metabiologics Inc., Madison, Wis.).
  • mice Female Sprague-Dawley rats weighing 200-250 g (Charles River Laboratories Inc, Hollister, Calif.) were used in the allergic rhinitis model. Guinea pigs weighing 283-325 g (Charles River, Raleigh, N.C.) were used in the comparative safety study. Animals were housed in a vivarium with a 12 h light/dark cycle and a controlled temperature. Food and water were provided ad libitum. All procedures in this study were performed in accordance with the guidelines detailed in the Guide for Care and Use of Laboratory Animals published by the National Academy of Sciences and approved by the Institutional Animal Care and Use Committee.
  • the allergy induction protocol consisted of a series of seven intraperitoneal injections of ovalbumin (0.3 mg) and aluminum hydroxide powder (30 mg) suspension in 0.9% saline (1 mL) administered every other day under anesthesia ( ⁇ 2% isoflurane in O 2 ).
  • Ovalbumin (2 mg) in 0.9% saline (20 ⁇ L) was then intranasally instilled daily for a total of seven doses under anesthesia ( ⁇ 2% isoflurane in O 2 ).
  • Sneezing and nasal itching are useful indications of allergic rhinitis in rats and represent two of the four traditional clinical symptoms (along with rhinorrhea and congestion) monitored in patients. Consequently, a performance severity assessment (PSA) scale was established to score the extent of these two nasal allergic signs following antigen challenge.
  • Clinical signs were scored prior to induction at baseline, following induction to establish maximal allergic signs on day 0 and on days 3, 5 and 7 following treatment using numerical scores: a) itching (rubbing nose): 0, none; 1, ⁇ 30 times; 2, 30-50 times; 3, ⁇ 50 times and b) sneezing: 0, none; 1, ⁇ 3 times; 2, 3-10 times; 3, ⁇ 10 times (30 minute period). The sum of scores for itching and sneezing comprised the composite PSA score.
  • reconstituted Formulation 22 0.4 ng dose per rat, equivalent to 100 U of botulinum toxin
  • control diluent total volume of 40 ⁇ L per rat.
  • Administration of reconstituted Formulation 22 and the control diluent was achieved by inserting a pipette loaded with the appropriate composition into the nasal cavity of the test animals and expelling the composition therein.
  • Administration of intranasal ovalbumin was continued every other day to maintain allergic signs.
  • the animals were evaluated prior to reconstituted Formulation 22 (or control) treatment and post-treatment on days 3, 5 and 7 consistent with the emergence of treatment effect previously reported on day 5 after Botulinum neurotoxin type A treatment.
  • Guinea pigs were randomly assigned (three per group) to each dose group with dose levels selected based on previous dose ranging studies.
  • Formulations 27-29 were reconstituted using the diluent described in Example 3 and then administered, one formulation per animal, to each three-animal dose group. Administration was achieved by using a loaded pipette as described above. Following intranasal administration, all animals were observed and weighed daily for the duration of the study.
  • NOAEL No Adverse Effect Level
  • the LD50 was calculated using nominal logistic platform in JMP® 9.0. (SAS Institute Inc.).
  • One-way ANOVA was used to assess statistical significance between PSA scores across groups and days of observation. Post-hoc Scheffe tests were performed to assess significance between specific day pairs of observation in the treated animal group. Student's t-test was used for comparison of treatments within treatment day and for comparison of body weights across treatment groups. For statistical significance, a confidence level of p ⁇ 0.05 was used.
  • the PSA score in the reconstituted Formulation 22 group was still significantly elevated on day 3 (p ⁇ 0.05), but the scores on days 5 and 7 did not significantly (p>0.05) differ from the normal baseline score indicating that the allergic symptoms were essentially resolved to the normal level by day 5 following reconstituted Formulation 22 treatment.
  • PSA scores for reconstituted Formulation 22 treatment were significantly reduced compared to same day control animals (p ⁇ 0.05).
  • PSA scores for control animals remained significantly elevated (p ⁇ 0.01) from normal baseline throughout the experiment indicating the sustained induction of allergic rhinitis clinical signs in the model throughout the course of the experiment.
  • the H&E stained sections of control animals revealed typical signs of inflammatory pathology including edema, congestion and vascular dilatation in nasal mucosa across the cavity, particularly in turbinate ( FIG. 2 b ) and lateral nasal wall ( FIG. 2 e ), as compared to normal control animals ( FIGS. 2 a and d ).
  • Reconstituted Formulation 22 treatment resulted in essentially complete resolution of inflammatory findings ( FIG. 2 ). Additionally, hyperplasia of serous glands also was found in some control animals ( FIG. 2 b ).
  • the nasal mucosa of reconstituted-Formulation 22-treated animals appeared essentially normal with only mild congestion (i.e. on lateral nasal wall of this animal specimen shown in FIG. 2 e ) on day 10 after treatment. No signs of atrophy or degeneration of serous glands were found after the reconstituted Formulation 22 treatment.
  • VIP expression was dramatically increased following ovalbumin challenge and observed in the control animals ( FIG. 3 b ) in contrast to the normal animals ( FIG. 3 a ), especially around blood vessels and serous glands. Following reconstituted Formulation 22 treatment, VIP expression in the nasal mucosa decreased markedly ( FIG. 3 c ) and appeared comparable to normal animal tissue samples ( FIG. 3 a ).
  • the safety profile of reconstituted Formulations 27-29 following intranasal dosing was compared to botulinum type A complex in guinea pigs which are highly sensitive to the toxic effects of botulinum type A complex, thus providing a conservative estimate of safety for reconstituted Formulation 27-29 ( FIG. 4 ). Death was observed in groups of animals treated with 110000 U of reconstituted Formulation 29, and 1800 U and 3600 U of botulinum type A complex.
  • the LD50 for reconstituted Formulations 27-29 and botulinum type A complex via intranasal route of administration was determined as 108350 U and 1836 U, respectively.
  • the No Adverse Effect Level (NOAEL) observed for intranasal dosing of guinea pigs was 27500 U/animal for reconstituted Formulation 27 and 900 U/animal for botulinum type A complex, indicating reconstituted Formulations 27-29 are approximately 31-fold safer compared to botulinum type A complex via the intranasal route of administration. Further, this NOAEL dose in guinea pigs is approximately 250-fold higher than the dose shown to be effective in treating allergic rhinitis in the rat model.
  • the PSA scale was shown to be sensitive and specific in tracking the onset of clinical signs of rhinitis.
  • the typical total nasal symptom score used in clinical studies of rhinitis treatments tracks sneezing, itching, rhinorrhea and congestion.
  • Rhinorrhea was difficult to quantitate in rats and thus not included, however, it generally tracks with the other symptoms when evaluated in clinical treatment of rhinitis.
  • Treatment with reconstituted Formulation 22 but not control resulted in significant reduction in PSA score by day 3 following treatment as compared to control and with essentially full resolution to normal baseline levels by day 5.
  • Allergic rhinitis is caused by the interaction of allergens with inflammatory cells, resulting in release of vasoactive and proinflammatory mediators within the nasal mucosa.
  • Regulatory peptides such as VIP, may play an important role in the hypersecretion of allergic rhinitis where a high density of VIP expression has been shown in nasal mucosa in allergic rhinitis patients and rats. It is therefore noteworthy that reconstituted Formulation 22 treatment reduced VIP expression to baseline levels and the suppression of VIP activity is associated with the reduction of congestion, vascular dilatation and edema.
  • Reconstituted Formulation 25 as described above was manufactured at Revance Therapeutics, Inc. (Newark, Calif.), and botulinum type A complex was purchased from Metabiologics (Madison, Wis.) as a 1 mg/mL solution.
  • Simulated gastric fluid (SGF) and simulated intestinal fluid (SIF) were prepared per United States Pharmacopeia. In some cases, the SGF was used without addition of pepsin to study the contribution of the enzyme.
  • Rats Male rats, weighing 200-250 g, were purchased from Charles River Laboratories (Hollister, Calif.). The use of animals in this study was approved by the local Institutional Animal Care and Use Committee (IACUC). Rats were housed in a vivarium with a 12 h light/dark cycle and controlled temperature. Food and water were provided ad libitum.
  • IACUC Institutional Animal Care and Use Committee
  • a single dose 14-day oral toxicity study in rats was conducted comparing oral toxicity of reconstituted Formulations 30-35 and botulinum type A complex.
  • Animals fed state
  • rats were dosed via oral gavage at 4 mL/kg.
  • rats were dosed with 1.03 ⁇ 10 7 up to 1.76 ⁇ 10 8 U/kg of toxin using a pipette as described in Example 1.
  • rats were dosed with 1.31 ⁇ 10 5 up to 4.20 ⁇ 10 6 U/kg.
  • the NOAEL was established as the highest dose causing no adverse clinical observation and no significant difference in mean body weight and overall body weight change compared to controls by t-test (Excel 2003, Microsoft, Washington, USA).
  • the LD 50 was calculated using the nominal logistic platform in JMP® 9.0. (SAS Institute Inc., NC, USA).
  • reconstituted Formulation 25 and botulinum type A complex were incubated in SGF at 9000 U/mL at 37° C.
  • the toxin was exposed to SGF for less than one minute before diluted for potency testing due to rapid inactivation.
  • potencies were tested on samples taken at 5 minute, 15 minutes, 30 minutes, and 60 minutes.
  • Formulation 25 and botulinum type A complex were incubated in SIF for up to 2 hours. Samples taken at 0, 30 minutes, and 2 hours were tested for potency.
  • the differences in reported safety margin of 150 kDa to botulinum type A complex may due to the differences in toxin preparations/formulation and the methods used to detect toxin biological activity.
  • the oral toxicity of botulinum type A complex was significantly greater than that observed for purified 150 kDa neurotoxin.
  • the oral LD 50 values for reconstituted Formulations 30-35 and botulinum type A complex were determined to be 1.19 ⁇ 10 8 U/kg and 5.03 ⁇ 10 6 U/kg, respectively; the NOAEL for reconstituted Formulation 30 and botulinum type A complex were 1.03 ⁇ 10 7 U/kg and 1.31 ⁇ 10 5 U/kg, respectively, indicating reconstituted Formulation 30 was approximately 80-fold less toxic compared to botulinum type A complex via the oral route of administration.
  • Reconstituted Formulation 25 bioactivity was undetectable within one minute in SGF with a half-life of less than 6 seconds whereas the half-life of the BoNTA complex was approximately 17 minutes in SGF.
  • a double-blind, randomized, placebo controlled study was conducted with 51 subjects enrolled. Subjects were enrolled in one of two (2) parallel treatment groups with 26 subjects in the test article group and 25 subjects in the control group, following satisfaction of entry criteria and screening procedures. Each subject received either a test article or placebo.
  • the test article in this Example was a topical gel containing “Formulation 24”, as listed above in Table 1.
  • Formulation 24 was used to prepare the topical gel formulation, which was composed of 25 ng/vial purified and lyophilized 150 kDa botulinum toxin type A and approximately 9 mcg/mL of the absorption enhancing peptide excipient, reconstituted with 1.0 mL of a poloxamer diluent, along with inactive ingredients, giving a concentration of 25 ng/mL or 5 ng/0.2 mL.
  • the peptide excipient used was the antimicrobial peptide having amino acid sequence, Arg-Lys-Lys-Arg-Arg-Gln-Arg-Arg-Arg-Arg-Gly-(Lys) 15 -Gly-Arg-Lys-Lys-Arg-Arg-Gln-Arg-Arg-Arg.
  • the poloxamer diluent used was 16.5% poloxamer 407 and 0.9% sodium chloride in water, with BHT and ethanol (see Table 3).
  • Other inactive ingredients included histidine, histidine hydrochloride, sucrose, and polysorbate 20, in the amounts provided above in Table 1.
  • the placebo comprised a topical gel formulation of only the inactive ingredients of Formulation 24 (histidine, histidine hydrochloride, sucrose, and polysorbate 20, in the amounts provided above in Table 1), reconstituted with the poloxamer diluent. Again approximately 0.2 mL total volume was used as the applied placebo dose, using topical administration. Accordingly, a dose of 5 ng of the test article was compared to placebo.
  • the topical gels were prepared by an unblinded, trained study staff member.
  • Formulation 24 and control were supplied in a professional, single-use reconstitution/administration apparatus, containing a vial of lyophilized botulinum toxin Type A, as the active pharmaceutical ingredient (25 ng/mL or placebo), formulated with peptide excipient (9 mcg/mL), and other inactive ingredients.
  • the apparatus also contained a cartridge of poloxamer diluent for reconstitution.
  • the placebo control was a lyophilized vial of inactive ingredients.
  • the reconstituted formulations provided the gels for topical application.
  • 0.5 mL of gel containing either Formulation 24 or placebo was divided between four swabs, with two swabs for each of the two nostrils, for application to the inferior turbinates and adjoining intranasal mucosal surfaces using a nasal speculum. Some of the product was absorbed by the swab, and left on the swab. Thus, while 0.5 mL of gel was used to wet swabs, this lead to a transfer of approximately 0.2 mL to the target dose site.
  • swab recovery studies based on mass transfer showed that when one loads gel (containing Formulation 24 or placebo) onto a swab, where approximately 0.117 g of gel was loaded on a swab, approximately 0.083 g of gel was recovered on the swab after application in accordance with study protocol, indicating that approximately 0.034 g of gel was applied intranasally. It follows then that using a total of four swabs (two per nostril), the total amount of gel transferred was approximately 0.034 g ⁇ 4, which equals approximately 0.136 g of gel. As the density of the gel was 1.027 g/mL, approximately 0.132 mL of gel was calculated to have been applied intranasally to each subject. This value was rounded up to 0.2 mL, as a conservative estimate for safety in the clinical trial.
  • TMS total nasal symptom
  • RQLQ Rhinoconjunctivitis Quality of Life Questionnaire
  • PNIF peak nasal inspiratory flow
  • PEF peak expiratory flow
  • FEV1 forced expiratory volume
  • TNS-4 score is the sum of individual scores for rhinorrhea, nasal congestion, nasal itching, and sneezing, each measured on an ordinal scale of 0, 1, 2 or 3, representing no symptoms, mild, moderate, or severe symptoms, respectively.
  • the individual components of the TNS score are recorded by the subject daily and reported at follow-up visits (pre- and post-allergen challenge where applicable).
  • RQLQ is a questionnaire measuring the functional problems (physical, emotional, social, and occupational) most troublesome to adults with either seasonal or perennial of allergic or non-allergic rhinoconjunctivitis (see, Juniper E F, et al. Development and testing of a new measure of health status for clinical trials in rhinoconjunctivitis. Clin Exper Allergy 1991; 21: 77-83., 1991, which is hereby incorporated by reference in its entirety).
  • the RQLQ includes 28 questions in 7 domains (activity limitation, sleep problems, nose symptoms, eye symptoms, non-nose/eye symptoms, practical problems, and emotional function) of which 3 are ‘patient-specific’ questions in the activity domain, allowing patients to select 3 activities where there are most limited by their rhinoconjunctivitis.
  • the overall RQLQ score is the mean of all 28 responses and the individual domain scores are the means of the items in those domains.
  • the RQLQ has been validated and used extensively throughout the world in clinical practice and clinical trials (see Juniper E F, et al. Validation of a standardised version of the Rhinoconjunctivitis Quality of Life Questionnaire. J Allergy Clin Immunol 1999; 104: 364-9).
  • allergen challenge was performed at baseline and week 4, wherein the subject was exposed to cognate allergen (subject confirmed as reactive by prior skin test; allergen extract applied to nasal septum on paper filter disk), and nasal symptoms and PNIF were recorded post-challenge.
  • a quantitative airflow measure also was employed. Specifically, due to the high comorbidity of bronchial reactivity (which would change both mouth and nasal airflow), the measure of nasal resistance to flow was measured as a pure rhinitis quantitative measure. This represents the difference between nasal and mouth air flow. The smaller the gap, the less nasal resistance was present.
  • Cranial Nerves I-VII Evaluation of cranial nerves I-VII was performed at treatment (pre-application), day 2, and weeks 2, 4, and 8 (or early discontinuation). Scores for each cranial nerve was captured as outlined in Table 10 (for examination procedures and criteria, see Bates B. A guide to physical examination and history taking J B Lippincott & Co. 6th edition (1995) Chapter 7:168-169, 172-175; Chapter 18:505-8, which is hereby incorporated by reference in its entirety).
  • Facial Nerve Grading System The Regional House-Brackmann Facial Nerve Grading System (see, Yen, T L, et al. Significance of House-Brackmann facial nerve grading global score in the setting of differential facial nerve function. 0 to 1 Neurotol 2003; 24(1):118-222, which is hereby incorporated by reference in its entirety) was designed to evaluate synkinesis and the four major branches of the facial nerve (VII) that innervates target and adjacent musculature. Functionality of the facial nerve (VII) was evaluated pre-application, day 2, and weeks 2, 4, and 8 (or early discontinuation). Examination procedures and criteria are set forth in Table 11.
  • AEs were graded based on the CTEP-CTCAE version 4.03 for severity, where applicable. Otherwise AEs were graded as mild, moderate, severe, or life-threatening. AEs were evaluated post-application at treatment (day 0), and on follow-up visits at day 2 and weeks 2, 4, and 8 (or early discontinuation).
  • the primary efficacy endpoint was based on change from baseline in total nasal symptom score at week 4 for treatment versus control. Demonstration of efficacy required trending at p ⁇ 0.20, rather than direct statistical significance at other thresholds given the sample size and study design. Comparisons were made using a 2 tailed t-test. Two interim analyses of efficacy were performed, the first when 4 week data became available for at least 30 subjects; and the second when 75% or more of subjects reached week 4 (or early study discontinuation).
  • Secondary endpoints included the following with statistical analyses, as with the primary endpoint, or by Inferential statistics based on the Fisher's exact test CMH, or Pearson Chi Square test.
  • Exploratory analyses using daily and mean weekly subject-recorded values also were conducted. Additional exploratory analyses evaluated composite endpoints based upon categories and definitions specified as primary or secondary endpoints above. These composites combined two endpoints, with response required on both simultaneously.
  • Treatment-emergent AEs All treatment-emergent AEs occurring during the study were recorded and classified on the basis of MedDRA terminology for the safety population.
  • Treatment-emergent AEs were summarized by treatment group, the number of subjects reporting treatment-emergent AEs, system organ class, preferred term, severity, relationship, and seriousness. Comparisons between treatment groups were made by tabulating the frequency of subjects with one or more treatment-emergent AEs during the study. The Fisher's exact test was used to compare the proportion of subjects in each treatment group who report any treatment-emergent AEs at a significance level of 0.05.
  • SAEs Serious adverse events
  • Sample Size Justification Approximately 70 subjects were enrolled and randomized 1:1 to either test article or placebo. Based upon an estimated 12% difference between treatment and placebo groups, based on historical data for injectable botulinum toxin type A, approximately 35 subjects per group were required to demonstrate trending toward efficacy (primary statistical analyses).
  • Table 12 represents secondary efficacy analysis results, based on PEF-PNIF differences at week 4, in the ITT population. In this population, the within-group median was substituted for missing observations.
  • Table 13 represents secondary efficacy analysis results, based on post-challenge total nasal symptom (TNS) score at baseline (screening) and week 4, in the ITT population. In this population, the within-group median was substituted for missing observations.
  • TMS total nasal symptom

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Otolaryngology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Anesthesiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
US14/065,379 2012-10-28 2013-10-28 Compositions and Methods for Safe Treatment of Rhinitis Abandoned US20140120077A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/065,379 US20140120077A1 (en) 2012-10-28 2013-10-28 Compositions and Methods for Safe Treatment of Rhinitis
US15/155,551 US10201594B2 (en) 2012-10-28 2016-05-16 Compositions and methods for safe treatment of rhinitis
US16/272,998 US20200345819A1 (en) 2012-10-28 2019-02-11 Compositions and Methods for Safe Treatment of Rhinitis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261719462P 2012-10-28 2012-10-28
US14/065,379 US20140120077A1 (en) 2012-10-28 2013-10-28 Compositions and Methods for Safe Treatment of Rhinitis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/155,551 Continuation US10201594B2 (en) 2012-10-28 2016-05-16 Compositions and methods for safe treatment of rhinitis

Publications (1)

Publication Number Publication Date
US20140120077A1 true US20140120077A1 (en) 2014-05-01

Family

ID=50545513

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/065,379 Abandoned US20140120077A1 (en) 2012-10-28 2013-10-28 Compositions and Methods for Safe Treatment of Rhinitis
US15/155,551 Active - Reinstated US10201594B2 (en) 2012-10-28 2016-05-16 Compositions and methods for safe treatment of rhinitis
US16/272,998 Abandoned US20200345819A1 (en) 2012-10-28 2019-02-11 Compositions and Methods for Safe Treatment of Rhinitis

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/155,551 Active - Reinstated US10201594B2 (en) 2012-10-28 2016-05-16 Compositions and methods for safe treatment of rhinitis
US16/272,998 Abandoned US20200345819A1 (en) 2012-10-28 2019-02-11 Compositions and Methods for Safe Treatment of Rhinitis

Country Status (4)

Country Link
US (3) US20140120077A1 (fr)
CA (1) CA2889833A1 (fr)
MX (1) MX370929B (fr)
WO (1) WO2014066916A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070077259A1 (en) * 2005-03-03 2007-04-05 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US20080038203A1 (en) * 2004-03-03 2008-02-14 Revance Therapeutics, Inc. Compositions and Methods for Topical Diagnostic and Therapeutic Transport
US20120251576A1 (en) * 2009-12-15 2012-10-04 Ira Sanders Treatment of Nasal and Sinus Disorders
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
WO2019006098A1 (fr) 2017-06-30 2019-01-03 Allergan, Inc. Film soluble pour l'administration d'un dérivé clostridial
WO2019046311A1 (fr) 2017-08-28 2019-03-07 Revance Therapeutics, Inc. Compositions transmucosales de toxine botulique, kits et procédés de traitement de troubles de la vessie
KR20190053214A (ko) * 2016-09-13 2019-05-17 알레간 인코포레이티드 안정화된 비단백질 클로스트리듐 독소 조성물
US10293034B2 (en) * 2007-07-10 2019-05-21 Medy-Tox, Inc. Pharmaceutical liquid composition of botulinum toxin with improved stability and method of use

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX366344B (es) 2009-06-25 2019-07-05 Revance Therapeutics Inc Formulaciones de toxina botulinica libres de albumina.
PT2490986T (pt) * 2009-10-21 2018-11-13 Revance Therapeutics Inc Métodos e sistemas para purificar neurotoxina botulínica não complexada
RU2609289C1 (ru) * 2015-09-21 2017-02-01 Государственное бюджетное учреждение здравоохранения города Москвы "Научно исследовательский клинический институт Оториноларингологии им. Л.И. Свержевского" Департамента здравоохранения города Москвы Способ лечения хронических ринитов селективной денервацией слизистой оболочки полости носа
WO2017075468A1 (fr) * 2015-10-29 2017-05-04 Revance Therapeutics, Inc. Formules injectables de toxine botulique et leurs procédés d'utilisation à longue durée d'effet thérapeutique ou cosmétique
JP2019535829A (ja) * 2016-11-21 2019-12-12 エイリオン セラピューティクス, インコーポレイテッド 大型薬剤の経皮送達
WO2018236873A1 (fr) 2017-06-19 2018-12-27 President And Fellows Of Harvard College Méthodes et compositions pour le traitement d'une infection microbienne
WO2019113044A1 (fr) * 2017-12-07 2019-06-13 Ps Therapies Ltd Compositions topiques et leurs procédés d'utilisation
WO2021113926A1 (fr) * 2019-12-13 2021-06-17 Francesco Barbagallo Formulations de toxine botulique et procédés d'administration intranasale de celles-ci pour le traitement de la rhinite allergique

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5766605A (en) * 1994-04-15 1998-06-16 Mount Sinai School Of Medicine Of The City University Of New York Treatment of autonomic nerve dysfunction with botulinum toxin
US20050196414A1 (en) * 2004-03-03 2005-09-08 Essentia Biosystems, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
WO2009105369A1 (fr) * 2008-02-22 2009-08-27 Allergan, Inc. Compositions pharmaceutiques contenant un poloxamère à libération retardée.
US20100168023A1 (en) * 2008-12-31 2010-07-01 Revance Therapeutics, Inc. Injectable Botulinum Toxin Formulations
US8022179B2 (en) * 2005-03-03 2011-09-20 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of an oligopeptide

Family Cites Families (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4861764A (en) 1986-11-17 1989-08-29 Macro Chem. Corp. Percutaneous absorption enhancers, compositions containing same and method of use
US5053005A (en) 1989-04-21 1991-10-01 Gary E. Borodic Chemomodulation of curvature of the juvenile spine
US5183462A (en) 1990-08-21 1993-02-02 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US5401243A (en) 1990-08-21 1995-03-28 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US5686268A (en) 1992-06-19 1997-11-11 Pfizer Inc. Fused proteins
WO1994000481A1 (fr) 1992-06-23 1994-01-06 Associated Synapse Biologics Composition pharmaceutique contenant un complexe de botulinum b
IL105600A (en) 1993-05-04 1999-12-31 Allon Blank Pharmaceutical and cosmetic compositions with increased rate of transdermal penetration and antioxidation protection against degradation containing derivatives of thioglycolic and mercaptopropionic acids some such novel compounds and their use
US5562907A (en) 1993-05-14 1996-10-08 Arnon; Stephen S. Method to prevent side-effects and insensitivity to the therapeutic uses of toxins
US5437291A (en) 1993-08-26 1995-08-01 Univ Johns Hopkins Method for treating gastrointestinal muscle disorders and other smooth muscle dysfunction
ES2319860T3 (es) 1993-12-28 2009-05-14 Allergan, Inc. Uso de toxina botulinica de tipo b para la fabricacion de un medicamento para reducir el dolor asociado a un trastorno muscular.
US20040126396A1 (en) 1993-12-28 2004-07-01 Allergan, Inc. Botulinum toxin treatment for strabismus
US6986893B2 (en) 1993-12-28 2006-01-17 Allergan, Inc. Method for treating a mucus secretion
US6974578B1 (en) 1993-12-28 2005-12-13 Allergan, Inc. Method for treating secretions and glands using botulinum toxin
US5670484A (en) 1994-05-09 1997-09-23 Binder; William J. Method for treatment of skin lesions associated with cutaneous cell-proliferative disorders
DK0758900T3 (da) 1994-05-09 2002-07-29 William J Binder Botulinumtoksin til reduktion af migræne-hovedpinesmerter
WO1996005222A1 (fr) 1994-08-08 1996-02-22 Wisconsin Alumni Research Foundation Procede de purification et compositions pharmaceutiques contenant la neurotoxine botulinique de type g
US5512547A (en) 1994-10-13 1996-04-30 Wisconsin Alumni Research Foundation Pharmaceutical composition of botulinum neurotoxin and method of preparation
DK0773788T3 (da) 1995-06-06 2003-09-01 Allergan Inc Forbedrede præparater og fremgangsmåder til kemodenervering under anvendelse af neurotoxiner
US5837265A (en) 1996-03-08 1998-11-17 The Regents Of The University Of California Chemically-modified clostridiatoxin with improved properties
US6063768A (en) 1997-09-04 2000-05-16 First; Eric R. Application of botulinum toxin to the management of neurogenic inflammatory disorders
US6221355B1 (en) 1997-12-10 2001-04-24 Washington University Anti-pathogen system and methods of use thereof
US6159944A (en) 1998-02-27 2000-12-12 Synchroneuron, Llc Method for treating painful conditions of the anal region and compositions therefor
US7537773B1 (en) 1998-08-25 2009-05-26 Botulinum Toxin Research Associates, Inc. Chemodenervating pharmaceutical as anti-inflammatory agent
TW574036B (en) 1998-09-11 2004-02-01 Elan Pharm Inc Stable liquid compositions of botulinum toxin
KR20010089347A (ko) 1998-10-27 2001-10-06 메이오 파운데이션 포 메디칼 에쥬케이션 앤드 리써치 상처 치료 증진 방법
WO2000034308A2 (fr) 1998-12-10 2000-06-15 Washington University Systeme de transduction de proteines et methodes d'utilisation
US20030236214A1 (en) 1999-06-09 2003-12-25 Wolff Jon A. Charge reversal of polyion complexes and treatment of peripheral occlusive disease
US6669951B2 (en) 1999-08-24 2003-12-30 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
EP2267010B1 (fr) 1999-08-25 2014-05-07 Allergan, Inc. Neurotoxines recombinantes activables
US6113915A (en) 1999-10-12 2000-09-05 Allergan Sales, Inc. Methods for treating pain
US20030118598A1 (en) 2000-02-08 2003-06-26 Allergan, Inc. Clostridial toxin pharmaceutical compositions
US7780967B2 (en) 2000-02-08 2010-08-24 Allergan, Inc. Reduced toxicity Clostridial toxin pharmaceutical compositions
US6670322B2 (en) 2000-06-01 2003-12-30 Wisconsin Alumni Research Foundation Method of targeting pharmaceuticals to motor neurons
US6903187B1 (en) 2000-07-21 2005-06-07 Allergan, Inc. Leucine-based motif and clostridial neurotoxins
US20030215412A1 (en) 2000-07-21 2003-11-20 Essentia Biosystems, Inc. Induction of hair growth with vascular endothelial growth factor
IL154044A0 (en) 2000-07-21 2003-07-31 Essentia Biosystems Inc Multi-component pharmaceutical compositions containing a complex of a positively charged backbone and a negatively charged backbone and methods for the preparation thereof
US20040220100A1 (en) 2000-07-21 2004-11-04 Essentia Biosystems, Inc. Multi-component biological transport systems
US20020127247A1 (en) 2000-11-17 2002-09-12 Allergen Sales, Inc. Modified clostridial neurotoxins with altered biological persistence
JP2005508832A (ja) 2001-02-16 2005-04-07 セルゲイト, インコーポレイテッド 間隔を開けてアルギニン部分を含むトランスポーター
US6677070B2 (en) 2001-04-19 2004-01-13 Hewlett-Packard Development Company, L.P. Hybrid thin film/thick film solid oxide fuel cell and method of manufacturing the same
US20120263781A1 (en) * 2001-08-13 2012-10-18 Lipella Pharmaceuticals Inc. Methods and compositions for treating rhinitis
US7060498B1 (en) 2001-11-28 2006-06-13 Genta Salus Llc Polycationic water soluble copolymer and method for transferring polyanionic macromolecules across biological barriers
JP2005538035A (ja) 2001-12-11 2005-12-15 ザ ボード オブ トラスティーズ オブ ザ リーランド スタンフォード ジュニア ユニバーシティ グアニジニウム輸送試薬および結合体
AU2003216389A1 (en) 2002-02-21 2003-09-09 Essentia Biosystems, Inc. Induction of hair growth with vascular endothelial growth factor
US20030215395A1 (en) 2002-05-14 2003-11-20 Lei Yu Controllably degradable polymeric biomolecule or drug carrier and method of synthesizing said carrier
US7691394B2 (en) 2002-05-28 2010-04-06 Botulinum Toxin Research Associates, Inc. High-potency botulinum toxin formulations
US20040009180A1 (en) 2002-07-11 2004-01-15 Allergan, Inc. Transdermal botulinum toxin compositions
EP2272340B1 (fr) 2002-08-19 2018-03-14 Ira Sanders Toxine botulique
EP1565210B1 (fr) 2002-11-21 2012-01-11 Ira Sanders Neurotoxine de Clostridia pour une utilisation dans le traitement de la congestion nasale
KR20050089827A (ko) 2002-12-19 2005-09-08 이츠차크 힐만 항균성 펩티드 억제물을 통한 질환 치료
WO2004076634A2 (fr) 2003-02-24 2004-09-10 Ira Sanders Translocation par membrane cellulaire d'inhibiteurs snare régulés, compositions associées et procédés de traitement de pathologies
EP1599221B1 (fr) 2003-03-06 2011-12-28 Botulinum Toxin Research Associates, Inc. Traitement des cephalees et des douleurs faciales chroniques associees a la sinusite au moyen de toxine botulinique
WO2004084805A2 (fr) 2003-03-19 2004-10-07 The J. David Gladstone Institutes Polypeptides tat acetyles et procedes d'utilisation de ceux-ci
WO2004084839A2 (fr) 2003-03-24 2004-10-07 Cady Roger K Methode et article pour le traitement de troubles associes a des neurones sensoriels par une application transdermique de toxine botulinique
US6838434B2 (en) 2003-05-02 2005-01-04 Allergan, Inc. Methods for treating sinus headache
JP4695342B2 (ja) 2003-09-03 2011-06-08 富士通株式会社 光スイッチ制御装置および移動体制御装置
CA3031270A1 (fr) 2004-03-03 2005-12-22 Revance Therapeutics, Inc. Compositions et methodes de diagnostic topique et de transport therapeutique
AU2005274822B2 (en) 2004-07-26 2008-10-30 Merz Pharma Gmbh & Co. Kgaa Therapeutic composition with a botulinum neurotoxin
HUE032950T2 (en) 2004-08-04 2017-11-28 Ipsen Biopharm Ltd Pharmaceutical preparation containing botulinum neurotoxin A2
WO2006013370A1 (fr) 2004-08-04 2006-02-09 Ipsen Limited Composition pharmaceutique contenant la neurotoxine botulique a2
GB2418358A (en) 2004-09-24 2006-03-29 Ipsen Ltd Pharmaceutical composition comprising botulinum neurotoxin
EP3141559A1 (fr) 2004-08-18 2017-03-15 Novabiotics Limited Peptides antimicrobiens
EP1661912A1 (fr) 2004-11-29 2006-05-31 Xigen S.A. Protéine de fusion comprenant un domaine BH3 d'une protéine BH3-only
US7749515B2 (en) 2005-02-01 2010-07-06 Allergan, Inc. Targeted delivery of botulinum toxin to the sphenopalatine ganglion
US7655244B2 (en) 2005-02-01 2010-02-02 Allergan, Inc. Targeted delivery of botulinum toxin for the treatment and prevention of trigeminal autonomic cephalgias, migraine and vascular conditions
RU2007136616A (ru) 2005-03-03 2009-04-10 Риванс Терапьютикс, Инк. (Us) Композиция и способ для местного применения и чрескожного введения ботулинового токсина
BRPI0608951A2 (pt) 2005-03-30 2010-02-17 Revance Therapeutics Inc composições e método para o tratamento de acne
JP5826450B2 (ja) 2005-07-22 2015-12-02 ザ ファウンドリー, エルエルシー 治療薬の送達のためのシステムおよび方法
US9511210B2 (en) 2006-05-19 2016-12-06 The Foundry, Llc Apparatus for toxin delivery to the nasal cavity
WO2007044809A2 (fr) 2005-10-11 2007-04-19 Botulinum Toxin Research Associates, Inc. Préparations pharmaceutiques basées sur une toxine botulique sans albumine et contenant une hyaluronidase, et méthodes d'emploi
US8518414B2 (en) 2005-11-17 2013-08-27 Revance Therapeutics, Inc. Compositions and methods of topical application and transdermal delivery of botulinum toxins with reduced non-toxin proteins
FR2902341B1 (fr) 2006-06-16 2011-02-25 Scras Utilisation therapeutique simultanee, separee ou etalee dans le temps d'au moins une neurotoxine botulique, et d'au moins un derive opiace
AR061669A1 (es) 2006-06-29 2008-09-10 Merz Pharma Gmbh & Co Kgaa Aplicacion de alta frecuencia de terapia con toxina botulinica
CN101583274A (zh) 2006-12-29 2009-11-18 雷文斯治疗公司 使用反向序列hiv-tat多肽的运输分子
AU2007340162B2 (en) 2006-12-29 2013-08-01 Revance Therapeutics, Inc. Compositions and methods of topical application and transdermal delivery of botulinum toxins stabilized with polypeptide fragments derived from HIV-TAT
CA2694046C (fr) 2007-07-26 2023-09-12 Revance Therapeutics, Inc. Peptides cationiques et compositions de ceux-ci
WO2009055350A1 (fr) 2007-10-23 2009-04-30 Allergan, Inc. Procédés de traitement d'une inflammation neurogénique chronique à l'aide de toxines clostridiennes modifiées
WO2009073253A1 (fr) 2007-12-04 2009-06-11 Ira Sanders Procédés de prévention ou de traitement des complications liées à des dispositifs de contrôle des voies respiratoires
US20120156244A1 (en) * 2008-08-01 2012-06-21 Alpha Synergy Development Inc. Nasal Compositions and Uses Thereof
AU2009286973B2 (en) 2008-08-29 2014-06-12 Merz Pharma Gmbh & Co. Kgaa Clostridial neurotoxins with altered persistency
WO2011057301A1 (fr) 2009-11-09 2011-05-12 Ira Sanders Traitement des troubles respiratoires du sommeil par une neurotoxine
EP2512507A4 (fr) 2009-12-15 2013-04-17 Ira Sanders Traitement d'affections nasales et sinusales
EP2399601A1 (fr) 2010-06-24 2011-12-28 Merz Pharma GmbH & Co. KGaA Thérapie de toxine botulique
WO2012019203A2 (fr) * 2010-08-03 2012-02-09 Dynova Laboratories, Inc. Agent thérapeutique pour administration intranasale et son procédé de fabrication et d'utilisation

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5766605A (en) * 1994-04-15 1998-06-16 Mount Sinai School Of Medicine Of The City University Of New York Treatment of autonomic nerve dysfunction with botulinum toxin
US20050196414A1 (en) * 2004-03-03 2005-09-08 Essentia Biosystems, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US8022179B2 (en) * 2005-03-03 2011-09-20 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of an oligopeptide
WO2009105369A1 (fr) * 2008-02-22 2009-08-27 Allergan, Inc. Compositions pharmaceutiques contenant un poloxamère à libération retardée.
US20100168023A1 (en) * 2008-12-31 2010-07-01 Revance Therapeutics, Inc. Injectable Botulinum Toxin Formulations

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Kim et al., The Journal of Laryngology and Otology, 1998; 112: 248-251 *
Nigam et al., Indian J Dermatol., 2010; 55(1): 8-14 *
Ozcan et al., American Journal of Otolarynology-Head and Neck Medicine and Surgery, 2006; 27: 314-318 *
Shaari et al., Otolaryngol Head Neck Surg, 1995; 112: 566-71 *
Wen et al., Neuroimmunomodulation, 2007; 14: 78-83 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US20080038203A1 (en) * 2004-03-03 2008-02-14 Revance Therapeutics, Inc. Compositions and Methods for Topical Diagnostic and Therapeutic Transport
US10172877B2 (en) 2004-03-03 2019-01-08 Revance Therapeutics, Inc. Compositions and methods for topical diagnostic and therapeutic transport
US8974774B2 (en) 2004-03-03 2015-03-10 Revance Therapeutics, Inc. Compositions and methods for topical diagnostic and therapeutic transport
US9180081B2 (en) * 2005-03-03 2015-11-10 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US10080786B2 (en) 2005-03-03 2018-09-25 Revance Therapeutics, Inc. Methods for treating pain by topical application and transdermal delivery of botulinum toxin
US10744078B2 (en) 2005-03-03 2020-08-18 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US20070077259A1 (en) * 2005-03-03 2007-04-05 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US10293034B2 (en) * 2007-07-10 2019-05-21 Medy-Tox, Inc. Pharmaceutical liquid composition of botulinum toxin with improved stability and method of use
US20120251576A1 (en) * 2009-12-15 2012-10-04 Ira Sanders Treatment of Nasal and Sinus Disorders
KR20190053214A (ko) * 2016-09-13 2019-05-17 알레간 인코포레이티드 안정화된 비단백질 클로스트리듐 독소 조성물
US10973890B2 (en) * 2016-09-13 2021-04-13 Allergan, Inc. Non-protein clostridial toxin compositions
KR102444230B1 (ko) 2016-09-13 2022-09-19 알레간 인코포레이티드 안정화된 비단백질 클로스트리듐 독소 조성물
KR20220132013A (ko) * 2016-09-13 2022-09-29 알레간 인코포레이티드 안정화된 비단백질 클로스트리듐 독소 조성물
KR102558892B1 (ko) 2016-09-13 2023-07-21 알레간 인코포레이티드 안정화된 비단백질 클로스트리듐 독소 조성물
CN110913838A (zh) * 2017-06-30 2020-03-24 阿勒根公司 用于递送梭菌衍生物的溶出膜
WO2019006098A1 (fr) 2017-06-30 2019-01-03 Allergan, Inc. Film soluble pour l'administration d'un dérivé clostridial
WO2019046311A1 (fr) 2017-08-28 2019-03-07 Revance Therapeutics, Inc. Compositions transmucosales de toxine botulique, kits et procédés de traitement de troubles de la vessie
EP3675900A4 (fr) * 2017-08-28 2021-05-05 Revance Therapeutics, Inc. Compositions transmucosales de toxine botulique, kits et procédés de traitement de troubles de la vessie

Also Published As

Publication number Publication date
WO2014066916A2 (fr) 2014-05-01
US10201594B2 (en) 2019-02-12
US20160250302A1 (en) 2016-09-01
CA2889833A1 (fr) 2014-05-01
WO2014066916A3 (fr) 2015-07-23
MX2015005417A (es) 2015-11-30
MX370929B (es) 2020-01-08
US20200345819A1 (en) 2020-11-05

Similar Documents

Publication Publication Date Title
US10201594B2 (en) Compositions and methods for safe treatment of rhinitis
JP6140445B2 (ja) 血管の過敏反応と関連した皮膚状態を治療する方法および組成物
AU2017248451B2 (en) Method of treating or reducing efp
ES2911124T3 (es) Formulación líquida de neurotoxina estabilizada con triptófano o tirosina
EP4324460A2 (fr) Bicarbonate de sodium pour utilisation dans le traitement de la goutte et de troubles associés
US11969461B2 (en) Composition comprising botulinum toxin
ES2822942T3 (es) Formulaciones de productos biológicos para instilación intravesical
JP2021505570A (ja) 高い応答率及び長い効果の持続期間を有する注射可能なボツリヌス毒素製剤及びその使用方法
ES2821731T3 (es) Formulaciones de productos biológicos para instilación intravesical
KR20230020961A (ko) 안면 주름의 고용량 및 저용적 보툴리눔 독소 치료
US20210338784A1 (en) Botulinum Toxin Pharmaceutical Composition Comprising Tannic Acid
TW201936207A (zh) 含有肉毒桿菌毒素、穩定劑以及局部麻醉劑的液體調配物及用於其的製備方法
WO2019191200A1 (fr) Procédé et formulation pour réaliser une anesthésie d'aspect interne de paroi oculaire par application topique
NZ788585A (en) Liquid neurotoxin formulation stabilized with tryptophan or tyrosine
NZ769098B2 (en) Method of treating or reducing efp
NZ624057B2 (en) Method of treating or reducing efp

Legal Events

Date Code Title Description
AS Assignment

Owner name: REVANCE THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:RUEGG, CURTIS L.;STONE, HONGRAN F.;WAUGH, JACOB M.;SIGNING DATES FROM 20140306 TO 20140320;REEL/FRAME:033425/0340

STCB Information on status: application discontinuation

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