US20090087459A1 - Methods for treating eye disorders - Google Patents

Methods for treating eye disorders Download PDF

Info

Publication number
US20090087459A1
US20090087459A1 US12/271,573 US27157308A US2009087459A1 US 20090087459 A1 US20090087459 A1 US 20090087459A1 US 27157308 A US27157308 A US 27157308A US 2009087459 A1 US2009087459 A1 US 2009087459A1
Authority
US
United States
Prior art keywords
eye
toxin
botulinum toxin
botulinum
clostridial
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
US12/271,573
Inventor
Eric R. First
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.)
Allergan Inc
Original Assignee
Allergan 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 Allergan Inc filed Critical Allergan Inc
Priority to US12/271,573 priority Critical patent/US20090087459A1/en
Publication of US20090087459A1 publication Critical patent/US20090087459A1/en
Priority to US12/648,260 priority patent/US20100098727A1/en
Abandoned legal-status Critical Current

Links

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • the present invention relates to methods for treating eye disorders and to compositions comprising a Clostridium toxin for treating said disorders.
  • the methods are related to treating eye disorders associated with inflammation.
  • Inflammation, or reddening, of the superficial tissues of the eye is a relatively common affliction.
  • Eye disorders associated with inflammation include, for example, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • the present invention provides such compositions and methods for treating eye disorders.
  • the methods and compositions of the present invention involve the use of a Clostridium toxin.
  • the present invention is, in part, based upon a surprising discovery that a Clostridial toxin may be administered to an eye of a patient, e.g., a mammal, to treat eye disorders.
  • botulinum toxin A Clostridial toxin that is commonly used clinically to treat various muscular conditions is botulinum toxin.
  • botulinum toxins have been used in clinical settings for the treatment of neuromuscular disorders characterized by hyperactive skeletal muscles.
  • a botulinum toxin type A complex was approved by the U.S. Food and Drug Administration for the treatment of blepharospasm, strabismus and hemifacial spasm.
  • a botulinum toxin type A was also approved by the FDA for the treatment of cervical dystonia and for the treatment of glabellar lines
  • a botulinum toxin type B was approved for the treatment of cervical dystonia.
  • Non-type A botulinum toxin serotypes apparently have a lower potency and/or a shorter duration of activity as compared to botulinum toxin type A.
  • Clinical effects of peripheral intramuscular botulinum toxin type A are usually seen within one week of injection.
  • the typical duration of symptomatic relief from a single intramuscular injection of botulinum toxin type A averages about three months, although significantly longer periods of therapeutic activity have been reported.
  • botulinum toxin type A has been used in clinical settings as follows:
  • extraocular muscles have been injected intramuscularly with between about 1-5 units of BOTOX®, the amount injected varying based upon both the size of the muscle to be injected and the extent of muscle paralysis desired (i.e. amount of diopter correction desired).
  • biceps brachii 50 U to 200 U.
  • Each of the five indicated muscles has been injected at the same treatment session, so that the patient receives from 90 U to 360 U of upper limb flexor muscle BOTOX® by intramuscular injection at each treatment session.
  • pericranial injected injection of 25 U of BOTOX® has showed significant benefit as a prophylactic treatment of migraine compared to vehicle as measured by decreased measures of migraine frequency, maximal severity, associated vomiting and acute medication use over the three month period following the 25 U injection.
  • intramuscular botulinum toxin has been used in the treatment of tremor in patients with Parkinson's disease, although it has been reported that results have not been impressive.
  • Marjama-Jyons, J., et al. Tremor - Predominant Parkinson's Disease, Drugs & Aging 16(4); 273-278:2000.
  • botulinum toxins may also have inhibitory effects in the central nervous system.
  • Work by Weigand et al, Naunyn - Schmiedeberg's Arch. Pharmacol. 1976; 292, 161-165, and Habermann, Naunyn - Schmiedeberg's Arch. Pharmacol. 1974; 281, 47-56 showed that botulinum toxin is able to ascend to the spinal area by retrograde transport.
  • a botulinum toxin injected at a peripheral location for example intramuscularly, may be retrograde transported to the spinal cord.
  • U.S. Pat. No. 5,989,545 discloses that a modified clostridial neurotoxin or fragment thereof, preferably a botulinum toxin, chemically conjugated or recombinantly fused to a particular targeting moiety can be used to treat pain by administration of the agent to the spinal cord.
  • a botulinum toxin has also been proposed for the treatment of rhinorrhea, hyperhydrosis and other disorders mediated by the autonomic nervous system (U.S. Pat. No. 5,766,605), tension headache, (U.S. Pat. No. 6,458,365), migraine headache (U.S. Pat. No. 5,714,468), post-operative pain and visceral pain (U.S. Pat. No. 6,464,986), pain treatment by intraspinal toxin administration (U.S. Pat. No. 6,113,915), Parkinson's disease and other diseases with a motor disorder component, by intracranial toxin administration (U.S. Pat. No. 6,306,403), hair growth and hair retention (U.S. Pat. No.
  • the present invention provides for methods of treating an eye disorder.
  • the methods comprise a step of locally administering a Clostridial toxin to the eye of a mammal to treat the disorder.
  • the methods comprise a step of locally administering a Clostridial toxin to a cornea of a mammal to treat the disorder.
  • a Clostridial toxin may be administered topically to the cornea to treat the eye disorder.
  • the Clostridial toxin is administered with a vasoconstrictor.
  • the eye disorder is associated with an inflammation of the eye.
  • eye disorders associated with an inflammation include, but are not limited to, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • compositions that may be employed for treating an eye disorder.
  • the compositions comprise an ophthalmically acceptable carrier, a Clostridial toxin in an amount effective to treat an eye disorder when the composition is administered to an eye, and a polyanionic component in an amount effective to provide lubrication to an eye when the composition is administered to an eye.
  • the composition is a solution.
  • the Clostridial toxin may be a toxin produced by a Clostridial beratti, a Clostridia butyricum, a Clostridial tetani bacterium or a Clostridial botulinum .
  • the Clostridial toxin may be a botulinum toxin type A, B, C 1 , D, E, F, G and/or mixtures thereof. In some embodiments, the Clostridial toxin is a botulinum toxin type A.
  • the polyanionic component comprises an anionic cellulosic derivative (e.g., carboxy methyl celluloses). In some embodiments, the compositions further comprise a vasoconstrictor.
  • the present invention is directed to treating an eye disorder by administering a Clostridial toxin to the eye of a mammal.
  • a “mammal” as used herein includes, for example, humans, rats, rabbits, mice and dogs. Any of the Clostridial toxins or compositions described below can be used in the methods described herein.
  • the eye disorder is associated with an inflammation of the eye.
  • eye disorders associated with an inflammation include, but are not limited to, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • an eye inflammation for example inflammation of the cornea
  • CGRP Calcitonin Gene-Related Peptide
  • neutrophil infiltration of the clear corneal surface is a hallmark of corneal inflammation in the human eye.
  • HCEC human corneal epithelial cells
  • IL-8 neutrophil chemotactic protein
  • HCEC bound CGRP in a saturable manner with a Kd of 2.0 ⁇ 10-9 M. Exposure of HCEC to CGRP induced a significant increase in intracellular cAMP levels and enhanced IL-8 synthesis nearly 4-fold. Also, the capacity of CGRP to stimulate cAMP and IL-8 synthesis was abrogated in the presence of the CGRP receptor antagonist CGRP8-37. CGRP stimulation had no effect on the half-life of IL-8 mRNA while increasing IL-8 pre-mRNA synthesis more than two fold.
  • CGRP did not induce monocyte chemotactic protein-1 or RANTES synthesis, nor did the neuropeptide enhance detectable increases in steady state levels of mRNA specific for these two beta-chemokines.
  • HCEC possess CGRP receptors capable of initiating a signal transduction cascade that differentially activates expression of the IL-8 gene but not the genes for monocyte chemotactic protein-1 or RANTES.
  • Tran et al. concluded that the capacity of CGRP to stimulate IL-8 synthesis in HCEC shows that sensory neurons are involved in induction of acute inflammation at the eye surface.
  • inhibiting the release of CGRP from sensory neurons in the eye may be effective in treating inflammation, preferably acute inflammation, of the eye. It is further believed that CGRP within a neuron is packaged in vesicles, and the inhibition of release of these vesicles may prevent the release of CGRP from the nerve terminals. Clostridial toxin may be effectively employed in inhibiting the release of CGRP from the nerve terminals in the eye.
  • a Clostridial toxin is locally administered to the eye to treat the eye disorder.
  • the Clostridial toxin may be locally administered to the cornea of the eye.
  • the Clostridial toxin is administered topically to treat the eye disorder.
  • the Clostridial toxin may be administered topically to the cornea of the eye.
  • a botulinum toxin type A is locally administered to the eye to treat an eye disorder that is associated with an inflammation. In some embodiments, the botulinum toxin type A is administered to the cornea of the eye. In some embodiments a botulinum toxin type A is administered topically to an eye to treat an eye disorder that is associated with an inflammation.
  • a botulinum toxin type A may be administered topically to the cornea of the eye to treat an eye disorder associated with an inflammation, wherein the eye disorders may include bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and/or inflammation response after intra-ocular lens implantation.
  • the Clostridial toxin is administered with a vasoconstrictor to treat the eye disorder.
  • vasoconstrictors are tetrahydrozoline, ephedrine, naphazoline, phenylephrine, and/or mixtures thereof.
  • the vasoconstrictor may be administered with the Clostridial toxin in a composition, as described below, or may be administered separately before or after the toxin.
  • the methods of treatment herein advantageously allow for the disordered (afflicted) eye to be treated with a reduced amount of preservatives coming into contact with the eye.
  • the Clostridial toxin that is administered to the eye according to this invention may or may not be associated with a preservative.
  • the Clostridial toxin is associated with a preservative when, for example, the toxin is one of the ingredients in an ophthalmic composition having preservatives.
  • the administration of these compositions to the eye for effective treatment may be less frequent, as compared to the frequency of administration of an existing commercial eye formulation. Because the present composition is administered less frequently than commercially available eye formulations to achieve the same therapeutic effect, less preservatives come into contact with the eye when the composition of the present invention is administered.
  • an ordinarily skilled medical provider can determine the appropriate dose and frequency of administration(s) to achieve an optimum clinical result. That is, one of ordinary skill in medicine would be able to administer the appropriate amount of the Clostridial toxin, for example botulinum toxin type A, at the appropriate time(s) to effectively treat the eye disorder.
  • the dose of the neurotoxin to be administered depends upon a variety of factors, including the severity of the eye disorder.
  • the dose of the Clostridial toxin administered is effective to treat, e.g. reduce inflammation, of the afflicted eye.
  • the dose of the Clostridial toxins employed in accordance with this invention may be equivalent to the dose of BOTOX® used in accordance with the present invention described herein.
  • a BOTOX® e.g. botulinum toxin type A
  • a BOTOX® e.g. botulinum toxin type A
  • a BOTOX® e.g. botulinum toxin type A
  • BOTOX® e.g. botulinum toxin type A
  • a BOTOX® e.g. botulinum toxin type A
  • BOTOX® e.g. botulinum toxin type A
  • BOTOX® e.g. botulinum toxin type A
  • Use of from about 0.1 U/kg to about 30 U/kg of a BOTOX® is within the scope of a method practiced according to the present disclosed invention.
  • about 0.1 U/kg to about 150 U/kg botulinum toxin may be administered to the eye to treat an eye disorder, e.g., bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • an eye disorder e.g., bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • a Clostridialtoxin e.g., botulinum toxin type A
  • a Clostridialtoxin is administered to the afflicted eye about every six days to provide for effective treatment of the eye.
  • Clostridial toxin e.g., botulinum toxin type A
  • the mode of administration of the present compositions depends on the form of the composition.
  • drops of the composition may be applied to the eye, e.g., from a conventional eye dropper.
  • the present compositions may be applied to the surface of the eye in substantially the same way as conventional ophthalmic compositions are applied.
  • Such administration of the present compositions does provide substantial and unexpected benefits, as described elsewhere herein.
  • the present invention also provides compositions for treating an eye disorder in a mammal.
  • the composition comprises an ophthalmically acceptable carrier, a Clostridial toxin in an amount effective to treat an eye disorder when the composition is administered to an eye, and a polyanionic component in an amount effective to provide lubrication to an eye when the composition is administered to an eye.
  • the Clostridial toxin may be produced from Clostridial beratti, a Clostridia butyricum, a Clostridial tetani bacterium or a Clostridial botulinum.
  • the composition is a solution.
  • the Clostridial toxins include, but are not limited to, tetanus toxins and botulinum toxin types A, B, C 1 , D, E, F, G and/or any mixtures thereof.
  • Clostridium botulinum produces a potent polypeptide neurotoxin, botulinum toxin, which causes a neuroparalytic illness in humans and animals referred to as botulism.
  • the spores of Clostridium botulinum are found in soil and can grow in improperly sterilized and sealed food containers of home based canneries, which are the cause of many of the cases of botulism.
  • the effects of botulism typically appear 18 to 36 hours after eating the foodstuffs infected with a Clostridium botulinum culture or spores.
  • botulinum toxin can apparently pass unattenuated through the lining of the gut and attack peripheral motor neurons. Symptoms of botulinum toxin intoxication can progress from difficulty walking, swallowing, and speaking to paralysis of the respiratory muscles and death.
  • Botulinum toxin type A is the most lethal natural biological agent known to man. About 50 picograms of a commercially available botulinum toxin type A (purified neurotoxin complex) (available from Allergan, Inc., of Irvine, Calif. under the tradename BOTOX® in 100 unit vials)) is a LD 50 in mice (i.e. 1 unit). One unit of BOTOX® contains about 50 picograms (about 56 attomoles) of botulinum toxin type A complex.
  • BOTOX® purified neurotoxin complex
  • botulinum toxin type A is about 1.8 billion times more lethal than diphtheria, about 600 million times more lethal than sodium cyanide, about 30 million times more lethal than cobra toxin and about 12 million times more lethal than cholera.
  • Singh Critical Aspects of Bacterial Protein Toxins, pages 63-84 (chapter 4) of Natural Toxins II, edited by B. R. Singh et al., Plenum Press, New York (1976) (where the stated LD 50 of botulinum toxin type A of 0.3 ng equals 1 U is corrected for the fact that about 0.05 ng of BOTOX® equals 1 unit).
  • One unit (U) of botulinum toxin is defined as the LD 50 upon intraperitoneal injection into female Swiss Webster mice weighing 18 to 20 grams each.
  • botulinum neurotoxins Seven generally immunologically distinct botulinum neurotoxins have been characterized, these being respectively botulinum neurotoxin serotypes A, B, C 1 , D, E, F and G each of which is distinguished by neutralization with type-specific antibodies.
  • the different serotypes of botulinum toxin vary in the animal species that they affect and in the severity and duration of the paralysis they evoke. For example, it has been determined that botulinum toxin type A is 500 times more potent, as measured by the rate of paralysis produced in the rat, than is botulinum toxin type B.
  • botulinum toxin type B has been determined to be non-toxic in primates at a dose of 480 U/kg which is about 12 times the primate LD 50 for botulinum toxin type A.
  • Botulinum toxin Type B Experimental and Clinical Experience, being chapter 6, pages 71-85 of “Therapy With Botulinum Toxin”, edited by Jankovic, J. et al. (1994), Marcel Dekker, Inc.
  • Botulinum toxin apparently binds with high affinity to cholinergic motor neurons, is translocated into the neuron and blocks the release of acetylcholine.
  • the molecular mechanism of toxin intoxication appears to be similar and to involve at least three steps or stages.
  • the toxin binds to the presynaptic membrane of the target neuron through a specific interaction between the heavy chain, H chain, and a cell surface receptor; the receptor is thought to be different for each type of botulinum toxin and for tetanus toxin.
  • the carboxyl end segment of the H chain, H C appears to be important for targeting of the toxin to the cell surface.
  • the toxin crosses the plasma membrane of the poisoned cell.
  • the toxin is first engulfed by the cell through receptor-mediated endocytosis, and an endosome containing the toxin is formed.
  • the toxin escapes the endosome into the cytoplasm of the cell.
  • This step is thought to be mediated by the amino end segment of the H chain, H N , which triggers a conformational change of the toxin in response to a pH of about 5.5 or lower.
  • Endosomes are known to possess a proton pump which decreases intra-endosomal pH.
  • the conformational shift exposes hydrophobic residues in the toxin, which permits the toxin to embed itself in the endosomal membrane.
  • the toxin (or at a minimum the light chain) then translocates through the endosomal membrane into the cytoplasm.
  • the last step of the mechanism of botulinum toxin activity appears to involve reduction of the disulfide bond joining the heavy chain, H chain, and the light chain, L chain.
  • the entire toxic activity of botulinum and tetanus toxins is contained in the L chain of the holotoxin; the L chain is a zinc (Zn++) endopeptidase which selectively cleaves proteins essential for recognition and docking of neurotransmitter-containing vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane.
  • VAMP vesicle-associated membrane protein
  • Botulinum toxin serotype A and E cleave SNAP-25.
  • Each of the botulinum toxins specifically cleaves a different bond, except botulinum toxin type B (and tetanus toxin) which cleave the same bond.
  • botulinum toxins serotypes Although all the botulinum toxins serotypes apparently inhibit release of the neurotransmitter acetylcholine at the neuromuscular junction, they do so by affecting different neurosecretory proteins and/or cleaving these proteins at different sites.
  • botulinum types A and E both cleave the 25 kiloDalton (kD) synaptosomal associated protein (SNAP-25), but they target different amino acid sequences within this protein.
  • Botulinum toxin types B, D, F and G act on vesicle-associated protein (VAMP, also called synaptobrevin), with each serotype cleaving the protein at a different site.
  • VAMP vesicle-associated protein
  • botulinum toxin type C 1 has been shown to cleave both syntaxin and SNAP-25. These differences in mechanism of action may affect the relative potency and/or duration of action of the various botulinum toxin serotypes.
  • a substrate for a botulinum toxin can be found in a variety of different cell types. See e.g. Biochem, J 1;339 (pt 1):159-65:1999, and Mov Disord, 10(3):376:1995 (pancreatic islet B cells contains at least SNAP-25 and synaptobrevin).
  • the botulinum toxins are released by Clostridial bacterium as complexes comprising the 150 kD botulinum toxin protein molecule along with associated non-toxin proteins.
  • the botulinum toxin type A complex can be produced by Clostridial bacterium as 900 kD, 500 kD and 300 kD forms.
  • Botulinum toxin types B and C 1 is apparently produced as only a 700 kD or 500 kD complex.
  • Botulinum toxin type D is produced as both 300 kD and 500 kD complexes.
  • botulinum toxin types E and F are produced as only approximately 300 kD complexes.
  • the complexes i.e. molecular weight greater than about 150 kD
  • the complexes are believed to contain a non-toxin hemaglutinin protein and a non-toxin and non-toxic nonhemaglutinin protein.
  • These two non-toxin proteins may act to provide stability against denaturation to the botulinum toxin molecule and protection against digestive acids when toxin is ingested.
  • botulinum toxin complexes may result in a slower rate of diffusion of the botulinum toxin away from a site of intramuscular injection of a botulinum toxin complex.
  • botulinum toxin inhibits potassium cation induced release of both acetylcholine and norepinephrine from primary cell cultures of brainstem tissue. Additionally, it has been reported that botulinum toxin inhibits the evoked release of both glycine and glutamate in primary cultures of spinal cord neurons and that in brain synaptosome preparations botulinum toxin inhibits the release of each of the neurotransmitters acetylcholine, dopamine, norepinephrine (Habermann E., et al., Tetanus Toxin and Botulinum A and C Neurotoxins Inhibit Noradrenaline Release From Cultured Mouse Brain, J Neurochem 51(2);522-527:1988) CGRP, substance P and glutamate (Sanchez-Prieto, J., et al., Botulinum Toxin A Blocks Glutamate Exocytosis From Guinea Pig Cerebral Cortical Synaptosome
  • Botulinum toxin type A can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures. All the botulinum toxin serotypes are initially synthesized as inactive single chain proteins which must be cleaved or nicked by proteases to become neuroactive. The bacterial strains that make botulinum toxin serotypes A and G possess endogenous proteases and serotypes A and G can therefore be recovered from bacterial cultures in predominantly their active form. In contrast, botulinum toxin serotypes C 1 , D and E are synthesized by nonproteolytic strains and are therefore typically unactivated when recovered from culture.
  • Serotypes B and F are produced by both proteolytic and nonproteolytic strains and therefore can be recovered in either the active or inactive form.
  • the proteolytic strains that produce, for example, the botulinum toxin type B serotype only cleave a portion of the toxin produced.
  • the exact proportion of nicked to unnicked molecules depends on the length of incubation and the temperature of the culture. Therefore, a certain percentage of any preparation of, for example, the botulinum toxin type B toxin is likely to be inactive, possibly accounting for the known significantly lower potency of botulinum toxin type B as compared to botulinum toxin type A.
  • botulinum toxin type B has, upon intramuscular injection, a shorter duration of activity and is also less potent than botulinum toxin type A at the same dose level.
  • High quality crystalline botulinum toxin type A can be produced from the Hall A strain of Clostridium botulinum with characteristics of ⁇ 3 ⁇ 10 7 U/mg, an A 260 /A 278 of less than 0.60 and a distinct pattern of banding on gel electrophoresis.
  • the known Shantz process can be used to obtain crystalline botulinum toxin type A, as set forth in Shantz, E. J., et al, Properties and use of Botulinum toxin and Other Microbial Neurotoxins in Medicine, Microbiol Rev. 56;80-99:1992.
  • the botulinum toxin type A complex can be isolated and purified from an anaerobic fermentation by cultivating Clostridium botulinum type A in a suitable medium.
  • the known process can also be used, upon separation out of the non-toxin proteins, to obtain pure botulinum toxins, such as for example: purified botulinum toxin type A with an approximately 150 kD molecular weight with a specific potency of 1-2 ⁇ 10 8 LD 50 U/mg or greater; purified botulinum toxin type B with an approximately 156 kD molecular weight with a specific potency of 1-2 ⁇ 10 8 LD 50 U/mg or greater, and; purified botulinum toxin type F with an approximately 155 kD molecular weight with a specific potency of 1-2 ⁇ 10 7 LD 50 U/mg or greater.
  • Botulinum toxins and/or botulinum toxin complexes can be obtained from List Biological Laboratories, Inc., Campbell, Calif.; the Centre for Applied Microbiology and Research, Porton Down, U.K.; Wako (Osaka, Japan), Metabiologics (Madison, Wis.) as well as from Sigma Chemicals of St Louis, Mo. Pure botulinum toxin can also be used to prepare a pharmaceutical composition.
  • botulinum toxin type A is detoxified by heat, various chemicals surface stretching and surface drying. Additionally, it is known that dilution of the toxin complex obtained by the known culturing, fermentation and purification to the much, much lower toxin concentrations used for pharmaceutical composition formulation results in rapid detoxification of the toxin unless a suitable stabilizing agent is present. Dilution of the toxin from milligram quantities to a solution containing nanograms per milliliter presents significant difficulties because of the rapid loss of specific toxicity upon such great dilution. Since the toxin may be used months or years after the toxin containing pharmaceutical composition is formulated, the toxin can stabilized with a stabilizing agent such as albumin and gelatin.
  • a stabilizing agent such as albumin and gelatin.
  • a commercially available botulinum toxin containing pharmaceutical composition is sold under the trademark BOTOX® (available from Allergan, Inc., of Irvine, Calif.).
  • BOTOX® consists of a purified botulinum toxin type A complex, albumin and sodium chloride packaged in sterile, vacuum-dried form.
  • the botulinum toxin type A is made from a culture of the Hall strain of Clostridium botulinum grown in a medium containing N-Z amine and yeast extract.
  • the botulinum toxin type A complex is purified from the culture solution by a series of acid precipitations to a crystalline complex consisting of the active high molecular weight toxin protein and an associated hemagglutinin protein.
  • BOTOX® can be reconstituted with sterile, non-preserved saline prior to intramuscular injection.
  • Each vial of BOTOX® contains about 100 units (U) of Clostridium botulinum toxin type A purified neurotoxin complex, 0.5 milligrams of human serum albumin and 0.9 milligrams of sodium chloride in a sterile, vacuum-dried form without a preservative.
  • BOTOX® sterile normal saline without a preservative; (0.9% Sodium Chloride Injection) is used by drawing up the proper amount of diluent in the appropriate size syringe. Since BOTOX® may be denatured by bubbling or similar violent agitation, the diluent is gently injected into the vial. For sterility reasons BOTOX® is preferably administered within four hours after the vial is removed from the freezer and reconstituted. During these four hours, reconstituted BOTOX® can be stored in a refrigerator at about 2° C. to about 8° C. Reconstituted, refrigerated BOTOX® has been reported to retain its potency for at least about two weeks. Neurology, 48:249-53:1997.
  • botulinum toxin type A can have an efficacy for up to 12 months ( European J. Neurology 6 (Supp 4): S111-S1150:1999), and in some circumstances for as long as 27 months.
  • the usual duration of an intramuscular injection of Botox® is typically about 3 to 4 months.
  • botulinum toxin type A to treat a variety of clinical conditions has led to interest in other botulinum toxin serotypes.
  • Two commercially available botulinum type A preparations for use in humans are BOTOX® available from Allergan, Inc., of Irvine, Calif., and Dysport® available from Beaufour Ipsen, Porton Down, England.
  • a botulinum toxin type B preparation (MyoBloc®) is available from Elan Pharmaceuticals of San Francisco, Calif.
  • Tetanus toxin as wells as derivatives (i.e. with a non-native targeting moiety), fragments, hybrids and chimeras thereof can also have therapeutic utility.
  • the tetanus toxin bears many similarities to the botulinum toxins.
  • both the tetanus toxin and the botulinum toxins are polypeptides made by closely related species of Clostridium ( Clostridium tetani and Clostridium botulinum, respectively).
  • both the tetanus toxin and the botulinum toxins are dichain proteins composed of a light chain (molecular weight about 50 kD) covalently bound by a single disulfide bond to a heavy chain (molecular weight about 100 kD).
  • the molecular weight of tetanus toxin and of each of the seven botulinum toxins (non-complexed) is about 150 kD.
  • the light chain bears the domain which exhibits intracellular biological (protease) activity, while the heavy chain comprises the receptor binding (immunogenic) and cell membrane translocational domains.
  • both the tetanus toxin and the botulinum toxins exhibit a high, specific affinity for gangliocide receptors on the surface of presynaptic cholinergic neurons.
  • Receptor mediated endocytosis of tetanus toxin by peripheral cholinergic neurons results in retrograde axonal transport, blocking of the release of inhibitory neurotransmifters from central synapses and a spastic paralysis.
  • receptor mediated endocytosis of botulinum toxin by peripheral cholinergic neurons results in little if any retrograde transport, inhibition of acetylcholine exocytosis from the intoxicated peripheral motor neurons and a flaccid paralysis.
  • tetanus toxin and the botulinum toxins resemble each other in both biosynthesis and molecular architecture.
  • Binz T. et al. The Complete Sequence of Botulinum Neurotoxin Type A and Comparison with Other Clostridial Neurotoxins, J Biological Chemistry 265(16);9153-9158:1 990.
  • Each type of neuron in the mammalian nervous system may release only a single type or multiple types of neurotransmifters.
  • the neurotransmitter acetylcholine is secreted by neurons in many areas of the brain, but specifically by the large pyramidal cells of the motor cortex, by several different neurons in the basal ganglia, by the motor neurons that innervate the skeletal muscles, by the preganglionic neurons of the autonomic nervous system (both sympathetic and parasympathetic), by the postganglionic neurons of the parasympathetic nervous system, and by some of the postganglionic neurons of the sympathetic nervous system.
  • acetylcholine has an excitatory effect.
  • acetylcholine is known to have inhibitory effects at some of the peripheral parasympathetic nerve endings, such as inhibition of heart rate by the vagal nerve.
  • the efferent signals of the autonomic nervous system are transmitted to the body through either the sympathetic nervous system or the parasympathetic nervous system.
  • the preganglionic neurons of the sympathetic nervous system extend from preganglionic sympathetic neuron cell bodies located in the intermediolateral horn of the spinal cord.
  • the preganglionic sympathetic nerve fibers, extending from the cell body synapse with postganglionic neurons located in either a paravertebral sympathetic ganglion or in a prevertebral ganglion. Since, the preganglionic neurons of both the sympathetic and parasympathetic nervous system are cholinergic, application of acetylcholine to the ganglia will excite both sympathetic and parasympathetic postganglionic neurons.
  • Acetylcholine activates two types of receptors, muscarinic and nicotinic receptors.
  • the muscarinic receptors are found in all effector cells stimulated by the postganglionic, neurons of the parasympathetic nervous system as well as in those stimulated by the postganglionic cholinergic neurons of the sympathetic nervous system.
  • the nicotinic receptors are found in the adrenal medulla, as well as within the autonomic ganglia, that is on the cell surface of the postganglionic neuron at the synapse between the preganglionic and postganglionic neurons of both the sympathetic and parasympathetic systems. Nicotinic receptors are also found in many nonautonomic nerve endings, for example in the membranes of skeletal muscle fibers at the neuromuscular junction.
  • Acetylcholine is released from cholinergic neurons when small, clear, intracellular vesicles fuse with the presynaptic neuronal cell membrane.
  • a wide variety of non-neuronal secretory cells such as, adrenal medulla (as well as the PC12 cell line) and pancreatic islet cells release catecholamines and parathyroid hormone, respectively, from large dense-core vesicles.
  • the PC12 cell line is a clone of rat pheochromocytoma cells extensively used as a tissue culture model for studies of sympathoadrenal development.
  • Botulinum toxin inhibits the release of both types of compounds from both types of cells in vitro, permeabilized (as by electroporation) or by direct injection of the toxin into the denervated cell. Botulinum toxin is also known to block release of the neurotransmitter glutamate from cortical synaptosomes cell cultures.
  • a neuromuscular junction is formed in skeletal muscle by the proximity of axons to muscle cells.
  • a signal transmitted through the nervous system results in an action potential at the terminal axon, with activation of ion channels and resulting release of the neurotransmitter acetylcholine from intraneuronal synaptic vesicles, for example at the motor endplate of the neuromuscular junction.
  • the acetylcholine crosses the extracellular space to bind with acetylcholine receptor proteins on the surface of the muscle end plate. Once sufficient binding has occurred, an action potential of the muscle cell causes specific membrane ion channel changes, resulting in muscle cell contraction.
  • the acetylcholine is then released from the muscle cells and metabolized by cholinesterases in the extracellular space. The metabolites are recycled back into the terminal axon for reprocessing into further acetylcholine.
  • a composition, carrier component or other material is “ophthalmically acceptable” when it is compatible with ocular tissue such that it does not cause significant or undue detrimental effects when brought into contact with ocular tissue.
  • the ophthalmically acceptable material is also compatible with other components of the present compositions.
  • an ophthalmically acceptable carrier comprises water and has a pH in a range of about 6.7 to about 7.4, or about 6.8 to about 7.2.
  • the carrier component comprises an electrolyte, for example, calcium, magnesium and/or mixtures thereof, in an amount which is ophthalmically acceptable.
  • the polyanionic component comprises an anionic cellulosic derivative.
  • the polyanionic component may comprise carboxy methyl celluloses, anionic homopolymers and copolymers comprising units of one or more of acrylic acid, methacrylic acid, metal acrylates and metal methacrylates, and/or mixture thereof.
  • the present compositions may comprise about 0.05% to about 5% (w/v), or 0.3% to about 2% of a polyanionic component.
  • the compositions further comprise a vasoconstrictor.
  • vasoconstrictors that may be used in accordance with the present invention include: tetrahydrozoline, ephedrine, naphazoline, phenylephrine, and/or mixtures thereof.
  • the present composition comprises about 0.001% to about 0.5% (w/v), or about 0.005% to about 0.2% (w/v) of the vasoconstrictor.
  • the pH of the compositions is about 6 to about 8, about 6.8 to about 7.5, more preferably about 6.8 to about 7.2, or about 7 to about 7.2.
  • compositions of the present invention may also include a suitable tonicity adjusting components.
  • tonicity adjusting components include, but are not limited to, sodium borate, boric acid, sodium chloride, potassium chloride, mannitol, dextrose, glycerin, propylene glycol, and the like and/or mixtures thereof.
  • compositions may be prepared using conventional procedures and techniques, which are well known to the skilled artisan.
  • the present compositions can be prepared by blending the components together, such as in one bulk.
  • compositions may be effectively used, as needed, by methods which comprise administering an effective amount of the compositions to an eye in need of treatment for an eye disorder.
  • the administering step may be repeated as needed to provide effective reduction of inflammation of such eye.
  • Conjunctivitis is an inflammation of the conjunctiva characterized by hyperemia (“red eye”), discharge, foreign-body sensation, and/or stuck eye lids often during sleep. It can be caused by infection of the conjunctiva by bacteria and fungi.
  • a bacteria infection is determined by, for example, colorful discharge, papillae and corneal staining pattern.
  • a fungal infection is determined by, for example, a feathery border at the corneal infection site. Both may be confirmed by culture and sensitivity lab testing. If there is a history of trauma, particularly with vegetable matter (e.g., a tree branch), fungal conjunctivitis is often involved. The following presents a typical treatment protocol that may be carried out for bacterial and fungal conjunctivitis.
  • a 20-year-old woman presents with a “red eye”, foreign-body sensation, and yellowish discharge from the right eye which has lasted for 3 days. She also reports that her lids are stuck together in the morning. This patient is diagnosed with bacterial conjunctivitis in the right eye.
  • the doctor treats the patient by, for example, topically administering one drop (comprising about 1 unit of a botulinum toxin type A or alternately comprising about 50 units of a botulinum toxin type B) of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A. Additionally, the treatment may be supplemented with topical antibiotic drops (e.g. Polytrim 4 ⁇ a day for 5 to 7 days) at the discretion of the doctor.
  • topical antibiotic drops e.g. Polytrim 4 ⁇ a day for 5 to 7 days
  • the patient would be treated by topically applying one drop (comprising about 1 unit of a botulinum toxin type A or alternately comprising about 50 units of a botulinum toxin type B) of the present composition.
  • the treatment may be supplemented with topical antifungal drops—5% Natamycin drops every 1-2 hours while awake, and every 2 hours at night.
  • Clostridial toxin e.g. botulinum toxin type A
  • the patient may return to the optometrist for a follow up visit.
  • the patient's condition is improved by at least 50%, such that the inflammation of the eye subsides and the redness is substantially cleared.
  • Viral conjunctivitis tends to occur with a history of a recent upper respiratory tract infection or contact with someone with conjunctivitis. It usually starts in one eye and then involves the other eye a few days later.
  • the viral infection is determined by mild hyperemia (pink eye), excessive tearing, foreign body sensation, follicular conjunctival reaction and may involve lymph node tenderness.
  • mild hyperemia pink eye
  • excessive tearing foreign body sensation
  • follicular conjunctival reaction may involve lymph node tenderness.
  • lymph node tenderness The following presents a typical treatment protocol that may be carried out for viral conjunctivitis.
  • a 34-year-old man presents with a “red eye” that started about 3 days ago in the left eye and then spread to the right eye.
  • the eyes are watery, swollen, and mildly red. He reports having a flu for the past week. This patient is diagnosed with viral conjunctivitis.
  • the doctor treats the patient by topically administering to the patient's eye one drop (comprising about 1 unit of a botulinum toxin type A or alternately comprising about 50 units of a botulinum toxin type B) of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A. Treatment may further include the use of artificial tears, cool compresses and antihistamine drops (if itchiness is severe).
  • the patient may return to the optometrist for a follow up visit.
  • the patient's condition may be improved by at least 40%, such that the inflammation of the eye subsided and the redness is substantially cleared.
  • Uveitis is a general term referring to inflammation of the uveal tract (iris, ciliary body, and choroid). Although it refers primarily to inflammation of this vascular structure, adjacent structures such as retina, vitreous, sclera, and cornea are also frequently involved. Patients most afflicted are 20-50 years of age, with a marked decrease after the age of 70.
  • Anterior uveitis occurs more frequently than posterior uveitis and affects the iris and/or ciliary body.
  • Anterior uveitis especially acute uveitis, is usually marked by eye pain, redness, photophobia (light sensitivity), mildly decreased vision, and tearing, and may be unilateral or bilateral depending on the etiology.
  • the critical sign of uveitis is cells and flare (white blood cells and protein leakage) in the anterior chamber. Many cases of acute, non-recurrent, anterior uveitis tend to be idiopathic and are treated primarily with anti-inflammatory/steroid drops.
  • ocular trauma causes of acute anterior uveitis (which can be recurrent) include: ocular trauma, post-surgical inflammation, medications, contact lens-related complications, HLA-B27 antigen, and inflammatory/autoimmune conditions (ankylosing spondylitis, inflammatory bowel disease, Reiter's syndrome, etc.)
  • inflammatory/autoimmune conditions ankylosing spondylitis, inflammatory bowel disease, Reiter's syndrome, etc.
  • the etiology is usually due to other systemic conditions such as juvenile rheumatoid arthritis, sarcoidosis, herpes simplex/herpes zoster/varicella, tuberculosis, and Fuch's heterochromia iridocyclitis.
  • Posterior uveitis involves the posterior segment of the eye (with corresponding retinal/choroidal inflammation and lesions). The onset may be acute but most often is insidious with little pain and minimum photophobia and blurred vision. Diseases with associated posterior uveitis include Lyme disease, toxoplasmosis, toxocariasis, histoplasmosis, and syphilis. The following presents a typical treatment protocol that may be carried out for uveitis.
  • the doctor treats the patient by topically administering two drops (comprising about 2 units of a botulinum toxin type A or alternately comprising about 100 units of a botulinum toxin type B) of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A. Additionally, if it is deemed necessary, the doctor may supplement the treatment with topical steroid drops (1% prednisolone acetate every 2 hours for 2 days, then taper to 4 ⁇ a day for 4 days, then 2 ⁇ a day for 4 more days).
  • the patient After one week, the patient returns to the doctor's office for a follow up visit.
  • the symptoms of uveitis appears to have subsided.
  • the inflammation of the uveal tract iris, ciliary body, and choroid
  • Keratic precipitates are associated with acute or chronic anterior uveitis (inflammation of the uveal tract).
  • the critical sign seen in the eyes is cellular infiltrates of lymphocytes and plasma cells (cells and flare) and also the presence of precipitates on the corneal endothelium and/or pupillary border known as keratic precipitates (KPs).
  • KPs keratic precipitates
  • a 29-year-old man presents with complaints of red eyes and extreme photophobia, onset 2 days ago.
  • Exam reveals presence of moderate cells and flare and a few large KPs along the pupillary border of both eyes.
  • the patient reports a history of Reiter's syndrome and has had recurrent episodes of uveitis in the past 3 years.
  • the patient is diagnosed with acute, anterior chamber uveitis secondary to Reiter's syndrome.
  • the doctor treats the patient by topically administering one drop of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A, to the patient's eye.
  • a composition comprising a Clostridial toxin, e.g. botulinum toxin type A
  • she can additionally place the patient on topical steroid drops (1% prednisolone acetate every 1 hour for 1-2 days, then taper).
  • topical steroid drops 1% prednisolone acetate every 1 hour for 1-2 days, then taper.
  • other treatment options may call for medical and rheumatological consultation.
  • a Clostridial toxin e.g. botulinum toxin type A
  • the patient returns the doctor's office for a follow up.
  • the patient shows signs of improvement, with reduced symptoms of photophobia, along with a mild presence of cells and flare, and the keratic precipitates are resolving.
  • Retinal edema is characterized by swelling of the retinal tissue due to serous leakage.
  • retinal edema occurs after any type of ocular surgery (i.e. cataract surgery), and is associated with inflammation.
  • Other etiologies include diabetic retinopathy, uveitis, and age-related macular degeneration. The following presents a typical treatment protocol that may be carried out for macular edema.
  • a 73-year-old man presents with decreased vision about 6 weeks after cataract surgery with surgical complication of vitreous loss.
  • the patient has been compliant with all post-surgical regimen.
  • the macula appears to be edematous, with the macular tissue being slightly raised in comparison to the surrounding retina. Accordingly, this patient's visual acuities were moderately reduced.
  • This patient is diagnosed with cystoid macular edema.
  • the doctor treats the patient by topically administering to the patient's eye a composition comprising a Clostridial toxin, e.g. about 1 unit of a botulinum toxin type A or alternately about 50 units of a botulinum toxin type B is administered.
  • a composition comprising a Clostridial toxin, e.g. about 1 unit of a botulinum toxin type A or alternately about 50 units of a botulinum toxin type B is administered.
  • the doctor may additionally put the patient on topical non-steroidal anti-inflammatory medication (ketorolac 4 ⁇ a day) for 6 weeks.
  • a Clostridial toxin e.g. botulinum toxin type A
  • the patient returns the doctor's office for a follow up.
  • the patient shows signs of improvement.
  • the macula is flat and the patient is regaining his vision.
  • Inflammation after ocular surgeries can involve the retina, resulting in cystoid macular edema.
  • the incidence increases with surgical complications such as iris prolapse and vitreous loss.
  • the following presents a typical treatment protocol that may be carried out for inflammation response after intra-ocular lens implantation.
  • a 69-year-old woman presents with decreased vision about 6 weeks after cataract surgery with surgical complication of vitreous loss.
  • the patient has been compliant with all post-surgical regimen.
  • This patient is diagnosed with cystoid macular edema (a condition associated with an inflammatory response).
  • the doctor treats the patient by topically administering to the patient's eye a formulation comprising A Clostridial toxin, e.g. about 1 unit of a botulinum toxin type A or alternately about 50 units of a botulinum toxin type B is administered.
  • the doctor may additionally put the patient on topical non-steroidal anti-inflammatory medication (ketorolac 4 ⁇ a day) for 6 weeks.
  • Clostridial toxin e.g. botulinum toxin type A
  • the patient returns the doctor's office for a follow up.
  • the patient shows signs of improvement, including less macular edema and improved vision.

Abstract

The present invention provides methods of treating an eye disorder. The methods comprise a step of locally administering a Clostridial toxin to the eye of a patient to treat the disorder. The eye disorder may be associated with an inflammation of the eye, including for example, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation. The Clostridial toxin may be produced by a Clostridial beratti, a Clostridia butyricum, a Clostridial tetani bacterium and/or a Clostridial botulinum.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of application Ser. No. 11/737,641 filed Apr. 19, 2007, which is a continuation of application Ser. No. 10/442,590 filed May 20, 2003, the entire contents of which applications is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to methods for treating eye disorders and to compositions comprising a Clostridium toxin for treating said disorders. In particular, the methods are related to treating eye disorders associated with inflammation.
  • BACKGROUND OF THE INVENTION
  • Inflammation, or reddening, of the superficial tissues of the eye is a relatively common affliction. Eye disorders associated with inflammation include, for example, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • Various types of palliative treatments have been used to treat this condition. The most common treatment includes the administration of eye drops which contain emollients and other ingredients designed to ease the discomfort due to the inflammation and to eliminate the redness associated with the condition. These treatments, however, have not been entirely satisfactory, however.
  • For example, current treatments often involve frequent applications of a medicinal eye drop. Unfortunately, however, many commercially available eye drops include preservatives, an ingredient that may be quite harmful to the eye. As such, frequent application of the commercially available eye drops may not be healthy for the eye.
  • Thus, there is a continued need to have improved methods and compositions for treating eye disorders. The present invention provides such compositions and methods for treating eye disorders. In particular, the methods and compositions of the present invention involve the use of a Clostridium toxin.
  • The present invention is, in part, based upon a surprising discovery that a Clostridial toxin may be administered to an eye of a patient, e.g., a mammal, to treat eye disorders.
  • A Clostridial toxin that is commonly used clinically to treat various muscular conditions is botulinum toxin. For example, botulinum toxins have been used in clinical settings for the treatment of neuromuscular disorders characterized by hyperactive skeletal muscles. In 1989 a botulinum toxin type A complex was approved by the U.S. Food and Drug Administration for the treatment of blepharospasm, strabismus and hemifacial spasm. Subsequently, a botulinum toxin type A was also approved by the FDA for the treatment of cervical dystonia and for the treatment of glabellar lines, and a botulinum toxin type B was approved for the treatment of cervical dystonia. Non-type A botulinum toxin serotypes apparently have a lower potency and/or a shorter duration of activity as compared to botulinum toxin type A. Clinical effects of peripheral intramuscular botulinum toxin type A are usually seen within one week of injection. The typical duration of symptomatic relief from a single intramuscular injection of botulinum toxin type A averages about three months, although significantly longer periods of therapeutic activity have been reported.
  • It has been reported that botulinum toxin type A has been used in clinical settings as follows:
  • (1) about 75-125 units of BOTOX® per intramuscular injection (multiple muscles) to treat cervical dystonia;
  • (2) 5-10 units of BOTOX® per intramuscular injection to treat glabellar lines (brow furrows) (5 units injected intramuscularly into the procerus muscle and 10 units injected intramuscularly into each corrugator supercilii muscle);
  • (3) about 30-80 units of BOTOX® to treat constipation by intrasphincter injection of the puborectalis muscle;
  • (4) about 1-5 units per muscle of intramuscularly injected BOTOX® to treat blepharospasm by injecting the lateral pre-tarsal orbicularis oculi muscle of the upper lid and the lateral pre-tarsal orbicularis oculi of the lower lid.
  • (5) to treat strabismus, extraocular muscles have been injected intramuscularly with between about 1-5 units of BOTOX®, the amount injected varying based upon both the size of the muscle to be injected and the extent of muscle paralysis desired (i.e. amount of diopter correction desired).
  • (6) to treat upper limb spasticity following stroke by intramuscular injections of BOTOX® into five different upper limb flexor muscles, as follows:
  • (a) flexor digitorum profundus: 7.5 U to 30 U
  • (b) flexor digitorum sublimus: 7.5 U to 30 U
  • (c) flexor carpi ulnaris: 10 U to 40 U
  • (d) flexor carpi radialis: 15 U to 60 U
  • (e) biceps brachii: 50 U to 200 U. Each of the five indicated muscles has been injected at the same treatment session, so that the patient receives from 90 U to 360 U of upper limb flexor muscle BOTOX® by intramuscular injection at each treatment session.
  • (7) to treat migraine, pericranial injected (injected symmetrically into glabellar, frontalis and temporalis muscles) injection of 25 U of BOTOX® has showed significant benefit as a prophylactic treatment of migraine compared to vehicle as measured by decreased measures of migraine frequency, maximal severity, associated vomiting and acute medication use over the three month period following the 25 U injection.
  • Additionally, intramuscular botulinum toxin has been used in the treatment of tremor in patients with Parkinson's disease, although it has been reported that results have not been impressive. Marjama-Jyons, J., et al., Tremor-Predominant Parkinson's Disease, Drugs & Aging 16(4); 273-278:2000.
  • In addition to having pharmacologic actions at the peripheral location, botulinum toxins may also have inhibitory effects in the central nervous system. Work by Weigand et al, Naunyn-Schmiedeberg's Arch. Pharmacol. 1976; 292, 161-165, and Habermann, Naunyn-Schmiedeberg's Arch. Pharmacol. 1974; 281, 47-56 showed that botulinum toxin is able to ascend to the spinal area by retrograde transport. As such, a botulinum toxin injected at a peripheral location, for example intramuscularly, may be retrograde transported to the spinal cord.
  • U.S. Pat. No. 5,989,545 discloses that a modified clostridial neurotoxin or fragment thereof, preferably a botulinum toxin, chemically conjugated or recombinantly fused to a particular targeting moiety can be used to treat pain by administration of the agent to the spinal cord.
  • A botulinum toxin has also been proposed for the treatment of rhinorrhea, hyperhydrosis and other disorders mediated by the autonomic nervous system (U.S. Pat. No. 5,766,605), tension headache, (U.S. Pat. No. 6,458,365), migraine headache (U.S. Pat. No. 5,714,468), post-operative pain and visceral pain (U.S. Pat. No. 6,464,986), pain treatment by intraspinal toxin administration (U.S. Pat. No. 6,113,915), Parkinson's disease and other diseases with a motor disorder component, by intracranial toxin administration (U.S. Pat. No. 6,306,403), hair growth and hair retention (U.S. Pat. No. 6,299,893), psoriasis and dermatitis (U.S. Pat. No. 5,670,484), injured muscles (U.S. Pat. No. 6,423,319, various cancers (U.S. Pat. No. 6,139,845), pancreatic disorders (U.S. Pat. No. 6,143,306), smooth muscle disorders (U.S. Pat. No. 5,437,291, including injection of a botulinum toxin into the upper and lower esophageal, pyloric and anal sphincters) ), prostate disorders (U.S. Pat. No. 6,365,164), inflammation, arthritis and gout (U.S. Pat. No. 6,063,768), juvenile cerebral palsy (U.S. Pat. No. 6,395,277), inner ear disorders (U.S. Pat. No. 6,265,379), thyroid disorders (U.S. Pat. No. 6,358,513), parathyroid disorders (U.S. Pat. No. 6,328,977). Additionally, controlled release toxin implants are known (see e.g. U.S. Pat. Nos. 6,306,423 and 6,312,708).
  • SUMMARY OF THE INVENTION
  • The present invention provides for methods of treating an eye disorder. The methods comprise a step of locally administering a Clostridial toxin to the eye of a mammal to treat the disorder. In some embodiments, the methods comprise a step of locally administering a Clostridial toxin to a cornea of a mammal to treat the disorder. For example, a Clostridial toxin may be administered topically to the cornea to treat the eye disorder. In some embodiments, the Clostridial toxin is administered with a vasoconstrictor.
  • Further in accordance with the present invention, the eye disorder is associated with an inflammation of the eye. Examples of eye disorders associated with an inflammation include, but are not limited to, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • The present invention provides for compositions that may be employed for treating an eye disorder. In accordance with the present invention, the compositions comprise an ophthalmically acceptable carrier, a Clostridial toxin in an amount effective to treat an eye disorder when the composition is administered to an eye, and a polyanionic component in an amount effective to provide lubrication to an eye when the composition is administered to an eye. In some embodiments, the composition is a solution. In some embodiments, the Clostridial toxin may be a toxin produced by a Clostridial beratti, a Clostridia butyricum, a Clostridial tetani bacterium or a Clostridial botulinum . In some embodiments, the Clostridial toxin may be a botulinum toxin type A, B, C1, D, E, F, G and/or mixtures thereof. In some embodiments, the Clostridial toxin is a botulinum toxin type A. In some embodiments, the polyanionic component comprises an anionic cellulosic derivative (e.g., carboxy methyl celluloses). In some embodiments, the compositions further comprise a vasoconstrictor.
  • Any feature or combination of features described herein are included within the scope of the present invention provided that the features included in any such combination are not mutually inconsistent as will be apparent from the context, this specification, and the knowledge of one of ordinary skill in the art.
  • Additional advantages and aspects of the present invention are apparent in the following detailed description and claims.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to treating an eye disorder by administering a Clostridial toxin to the eye of a mammal. A “mammal” as used herein includes, for example, humans, rats, rabbits, mice and dogs. Any of the Clostridial toxins or compositions described below can be used in the methods described herein.
  • In some embodiments, the eye disorder is associated with an inflammation of the eye. Examples of eye disorders associated with an inflammation include, but are not limited to, bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • Without limiting the invention to any theory or mechanism of operation, it is believed that an eye inflammation, for example inflammation of the cornea, is due in part to the release of a Calcitonin Gene-Related Peptide (CGRP). For example, neutrophil infiltration of the clear corneal surface is a hallmark of corneal inflammation in the human eye. Tran et al. showed that CGRP, a neuropeptide known to be released from the termini of corneal sensory, can bind to human corneal epithelial cells (HCEC) and induce expression of the neutrophil chemotactic protein IL-8. J. Immunol. 2000 Apr. 15; 164(8):4307-12.
  • Specifically, Tran et al. demonstrated the following: HCEC bound CGRP in a saturable manner with a Kd of 2.0×10-9 M. Exposure of HCEC to CGRP induced a significant increase in intracellular cAMP levels and enhanced IL-8 synthesis nearly 4-fold. Also, the capacity of CGRP to stimulate cAMP and IL-8 synthesis was abrogated in the presence of the CGRP receptor antagonist CGRP8-37. CGRP stimulation had no effect on the half-life of IL-8 mRNA while increasing IL-8 pre-mRNA synthesis more than two fold. Moreover, CGRP did not induce monocyte chemotactic protein-1 or RANTES synthesis, nor did the neuropeptide enhance detectable increases in steady state levels of mRNA specific for these two beta-chemokines. Based on that result, Tran et al. suggest that HCEC possess CGRP receptors capable of initiating a signal transduction cascade that differentially activates expression of the IL-8 gene but not the genes for monocyte chemotactic protein-1 or RANTES. Furthermore, Tran et al. concluded that the capacity of CGRP to stimulate IL-8 synthesis in HCEC shows that sensory neurons are involved in induction of acute inflammation at the eye surface.
  • Also, without wishing to limit the invention to any theory or mechanism of operation, it is believed that inhibiting the release of CGRP from sensory neurons in the eye may be effective in treating inflammation, preferably acute inflammation, of the eye. It is further believed that CGRP within a neuron is packaged in vesicles, and the inhibition of release of these vesicles may prevent the release of CGRP from the nerve terminals. Clostridial toxin may be effectively employed in inhibiting the release of CGRP from the nerve terminals in the eye.
  • In some embodiments, a Clostridial toxin is locally administered to the eye to treat the eye disorder. The Clostridial toxin may be locally administered to the cornea of the eye. In one embodiment, the Clostridial toxin is administered topically to treat the eye disorder. For example, the Clostridial toxin may be administered topically to the cornea of the eye.
  • In some embodiments, a botulinum toxin type A is locally administered to the eye to treat an eye disorder that is associated with an inflammation. In some embodiments, the botulinum toxin type A is administered to the cornea of the eye. In some embodiments a botulinum toxin type A is administered topically to an eye to treat an eye disorder that is associated with an inflammation. For example, a botulinum toxin type A may be administered topically to the cornea of the eye to treat an eye disorder associated with an inflammation, wherein the eye disorders may include bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and/or inflammation response after intra-ocular lens implantation.
  • In some embodiments, the Clostridial toxin is administered with a vasoconstrictor to treat the eye disorder. Examples of vasoconstrictors are tetrahydrozoline, ephedrine, naphazoline, phenylephrine, and/or mixtures thereof. The vasoconstrictor may be administered with the Clostridial toxin in a composition, as described below, or may be administered separately before or after the toxin.
  • The methods of treatment herein advantageously allow for the disordered (afflicted) eye to be treated with a reduced amount of preservatives coming into contact with the eye. For example, the Clostridial toxin that is administered to the eye according to this invention may or may not be associated with a preservative. The Clostridial toxin is associated with a preservative when, for example, the toxin is one of the ingredients in an ophthalmic composition having preservatives. However, the administration of these compositions to the eye for effective treatment may be less frequent, as compared to the frequency of administration of an existing commercial eye formulation. Because the present composition is administered less frequently than commercially available eye formulations to achieve the same therapeutic effect, less preservatives come into contact with the eye when the composition of the present invention is administered.
  • Of course, an ordinarily skilled medical provider can determine the appropriate dose and frequency of administration(s) to achieve an optimum clinical result. That is, one of ordinary skill in medicine would be able to administer the appropriate amount of the Clostridial toxin, for example botulinum toxin type A, at the appropriate time(s) to effectively treat the eye disorder. The dose of the neurotoxin to be administered depends upon a variety of factors, including the severity of the eye disorder. In some embodiments, the dose of the Clostridial toxin administered is effective to treat, e.g. reduce inflammation, of the afflicted eye. The dose of the Clostridial toxins employed in accordance with this invention may be equivalent to the dose of BOTOX® used in accordance with the present invention described herein. In the various methods of the present invention, from about 0.01 U/kg (units of botulinum toxin per kilogram of patient weight) to about 15 U/kg, of a BOTOX®, e.g. botulinum toxin type A, can be administered to the afflicted eye. In some embodiments, about 0.1 U/kg to about 20 U/kg of BOTOX® may be administered to the afflicted eye. Use of from about 0.1 U/kg to about 30 U/kg of a BOTOX®, is within the scope of a method practiced according to the present disclosed invention. In one embodiment, about 0.1 U/kg to about 150 U/kg botulinum toxin, for example type A, may be administered to the eye to treat an eye disorder, e.g., bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
  • In some embodiments, a Clostridialtoxin, e.g., botulinum toxin type A, is administered to the afflicted eye about every six days to provide for effective treatment of the eye. Clostridial toxin, e.g., botulinum toxin type A, may also be administered to the afflicted eye about every two weeks, every three weeks, or more, for example, every month or so.
  • The mode of administration of the present compositions depends on the form of the composition. For example, if the composition is a solution, drops of the composition may be applied to the eye, e.g., from a conventional eye dropper. In general, the present compositions may be applied to the surface of the eye in substantially the same way as conventional ophthalmic compositions are applied. Such administration of the present compositions does provide substantial and unexpected benefits, as described elsewhere herein.
  • The present invention also provides compositions for treating an eye disorder in a mammal. In some embodiments, the composition comprises an ophthalmically acceptable carrier, a Clostridial toxin in an amount effective to treat an eye disorder when the composition is administered to an eye, and a polyanionic component in an amount effective to provide lubrication to an eye when the composition is administered to an eye. The Clostridial toxin may be produced from Clostridial beratti, a Clostridia butyricum, a Clostridial tetani bacterium or a Clostridial botulinum. In some embodiments, the composition is a solution. The Clostridial toxins include, but are not limited to, tetanus toxins and botulinum toxin types A, B, C1, D, E, F, G and/or any mixtures thereof.
  • The genus Clostridium has more than one hundred and twenty seven species, grouped according to their morphology and functions. The anaerobic, gram positive bacterium Clostridium botulinum produces a potent polypeptide neurotoxin, botulinum toxin, which causes a neuroparalytic illness in humans and animals referred to as botulism. The spores of Clostridium botulinum are found in soil and can grow in improperly sterilized and sealed food containers of home based canneries, which are the cause of many of the cases of botulism. The effects of botulism typically appear 18 to 36 hours after eating the foodstuffs infected with a Clostridium botulinum culture or spores. The botulinum toxin can apparently pass unattenuated through the lining of the gut and attack peripheral motor neurons. Symptoms of botulinum toxin intoxication can progress from difficulty walking, swallowing, and speaking to paralysis of the respiratory muscles and death.
  • Botulinum toxin type A is the most lethal natural biological agent known to man. About 50 picograms of a commercially available botulinum toxin type A (purified neurotoxin complex) (available from Allergan, Inc., of Irvine, Calif. under the tradename BOTOX® in 100 unit vials)) is a LD50 in mice (i.e. 1 unit). One unit of BOTOX® contains about 50 picograms (about 56 attomoles) of botulinum toxin type A complex. Interestingly, on a molar basis, botulinum toxin type A is about 1.8 billion times more lethal than diphtheria, about 600 million times more lethal than sodium cyanide, about 30 million times more lethal than cobra toxin and about 12 million times more lethal than cholera. Singh, Critical Aspects of Bacterial Protein Toxins, pages 63-84 (chapter 4) of Natural Toxins II, edited by B. R. Singh et al., Plenum Press, New York (1976) (where the stated LD50 of botulinum toxin type A of 0.3 ng equals 1 U is corrected for the fact that about 0.05 ng of BOTOX® equals 1 unit). One unit (U) of botulinum toxin is defined as the LD50 upon intraperitoneal injection into female Swiss Webster mice weighing 18 to 20 grams each.
  • Seven generally immunologically distinct botulinum neurotoxins have been characterized, these being respectively botulinum neurotoxin serotypes A, B, C1, D, E, F and G each of which is distinguished by neutralization with type-specific antibodies. The different serotypes of botulinum toxin vary in the animal species that they affect and in the severity and duration of the paralysis they evoke. For example, it has been determined that botulinum toxin type A is 500 times more potent, as measured by the rate of paralysis produced in the rat, than is botulinum toxin type B. Additionally, botulinum toxin type B has been determined to be non-toxic in primates at a dose of 480 U/kg which is about 12 times the primate LD50 for botulinum toxin type A. Moyer E et al., Botulinum Toxin Type B: Experimental and Clinical Experience, being chapter 6, pages 71-85 of “Therapy With Botulinum Toxin”, edited by Jankovic, J. et al. (1994), Marcel Dekker, Inc. Botulinum toxin apparently binds with high affinity to cholinergic motor neurons, is translocated into the neuron and blocks the release of acetylcholine.
  • Regardless of serotype, the molecular mechanism of toxin intoxication appears to be similar and to involve at least three steps or stages. In the first step of the process, the toxin binds to the presynaptic membrane of the target neuron through a specific interaction between the heavy chain, H chain, and a cell surface receptor; the receptor is thought to be different for each type of botulinum toxin and for tetanus toxin. The carboxyl end segment of the H chain, HC, appears to be important for targeting of the toxin to the cell surface.
  • In the second step, the toxin crosses the plasma membrane of the poisoned cell. The toxin is first engulfed by the cell through receptor-mediated endocytosis, and an endosome containing the toxin is formed. The toxin then escapes the endosome into the cytoplasm of the cell. This step is thought to be mediated by the amino end segment of the H chain, HN, which triggers a conformational change of the toxin in response to a pH of about 5.5 or lower. Endosomes are known to possess a proton pump which decreases intra-endosomal pH. The conformational shift exposes hydrophobic residues in the toxin, which permits the toxin to embed itself in the endosomal membrane. The toxin (or at a minimum the light chain) then translocates through the endosomal membrane into the cytoplasm.
  • The last step of the mechanism of botulinum toxin activity appears to involve reduction of the disulfide bond joining the heavy chain, H chain, and the light chain, L chain. The entire toxic activity of botulinum and tetanus toxins is contained in the L chain of the holotoxin; the L chain is a zinc (Zn++) endopeptidase which selectively cleaves proteins essential for recognition and docking of neurotransmitter-containing vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane. Tetanus neurotoxin, botulinum toxin types B, D, F, and G cause degradation of synaptobrevin (also called vesicle-associated membrane protein (VAMP)), a synaptosomal membrane protein. Most of the VAMP present at the cytoplasmic surface of the synaptic vesicle is removed as a result of any one of these cleavage events. Botulinum toxin serotype A and E cleave SNAP-25. Botulinum toxin serotype C1 was originally thought to cleave syntaxin, but was found to cleave syntaxin and SNAP-25. Each of the botulinum toxins specifically cleaves a different bond, except botulinum toxin type B (and tetanus toxin) which cleave the same bond.
  • Although all the botulinum toxins serotypes apparently inhibit release of the neurotransmitter acetylcholine at the neuromuscular junction, they do so by affecting different neurosecretory proteins and/or cleaving these proteins at different sites. For example, botulinum types A and E both cleave the 25 kiloDalton (kD) synaptosomal associated protein (SNAP-25), but they target different amino acid sequences within this protein. Botulinum toxin types B, D, F and G act on vesicle-associated protein (VAMP, also called synaptobrevin), with each serotype cleaving the protein at a different site. Finally, botulinum toxin type C1 has been shown to cleave both syntaxin and SNAP-25. These differences in mechanism of action may affect the relative potency and/or duration of action of the various botulinum toxin serotypes. A substrate for a botulinum toxin can be found in a variety of different cell types. See e.g. Biochem, J 1;339 (pt 1):159-65:1999, and Mov Disord, 10(3):376:1995 (pancreatic islet B cells contains at least SNAP-25 and synaptobrevin).
  • The molecular weight of the botulinum toxin protein molecule, for all seven of the known botulinum toxin serotypes, is about 150 kD. Interestingly, the botulinum toxins are released by Clostridial bacterium as complexes comprising the 150 kD botulinum toxin protein molecule along with associated non-toxin proteins. Thus, the botulinum toxin type A complex can be produced by Clostridial bacterium as 900 kD, 500 kD and 300 kD forms. Botulinum toxin types B and C1 is apparently produced as only a 700 kD or 500 kD complex. Botulinum toxin type D is produced as both 300 kD and 500 kD complexes. Finally, botulinum toxin types E and F are produced as only approximately 300 kD complexes. The complexes (i.e. molecular weight greater than about 150 kD) are believed to contain a non-toxin hemaglutinin protein and a non-toxin and non-toxic nonhemaglutinin protein. These two non-toxin proteins (which along with the botulinum toxin molecule comprise the relevant neurotoxin complex) may act to provide stability against denaturation to the botulinum toxin molecule and protection against digestive acids when toxin is ingested. Additionally, it is possible that the larger (greater than about 150 kD molecular weight) botulinum toxin complexes may result in a slower rate of diffusion of the botulinum toxin away from a site of intramuscular injection of a botulinum toxin complex.
  • In vitro studies have indicated that botulinum toxin inhibits potassium cation induced release of both acetylcholine and norepinephrine from primary cell cultures of brainstem tissue. Additionally, it has been reported that botulinum toxin inhibits the evoked release of both glycine and glutamate in primary cultures of spinal cord neurons and that in brain synaptosome preparations botulinum toxin inhibits the release of each of the neurotransmitters acetylcholine, dopamine, norepinephrine (Habermann E., et al., Tetanus Toxin and Botulinum A and C Neurotoxins Inhibit Noradrenaline Release From Cultured Mouse Brain, J Neurochem 51(2);522-527:1988) CGRP, substance P and glutamate (Sanchez-Prieto, J., et al., Botulinum Toxin A Blocks Glutamate Exocytosis From Guinea Pig Cerebral Cortical Synaptosomes, Eur J. Biochem 165;675-681:1897. Thus, when adequate concentrations are used, stimulus-evoked release of most neurotransmifters is blocked by botulinum toxin. See e.g. Pearce, L. B., Pharmacologic Characterization of Botulinum Toxin For Basic Science and Medicine, Toxicon 35(9); 1373-1412 at 1393; Bigalke H., et al., Botulinum A Neurotoxin Inhibits Non-Cholinergic Synaptic Transmission in Mouse Spinal Cord Neurons in Culture, Brain Research 360;318-324:1985; Habermann E., Inhibition by Tetanus and Botulinum A Toxin of the release of [ 3 H]Noradrenaline and [ 3 H]GABA From Rat Brain Homogenate, Experientia 44;224-226:1988, Bigalke H., et al., Tetanus Toxin and Botulinum A Toxin Inhibit Release and Uptake of Various Transmitters, as Studied with Particulate Preparations From Rat Brain and Spinal Cord, Naunyn-Schmiedeberg's Arch Pharmacol 316;244-251:1981, and; Jankovic J. et al., Therapy With Botulinum Toxin, Marcel Dekker, Inc., (1994), page 5.
  • Botulinum toxin type A can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures. All the botulinum toxin serotypes are initially synthesized as inactive single chain proteins which must be cleaved or nicked by proteases to become neuroactive. The bacterial strains that make botulinum toxin serotypes A and G possess endogenous proteases and serotypes A and G can therefore be recovered from bacterial cultures in predominantly their active form. In contrast, botulinum toxin serotypes C1, D and E are synthesized by nonproteolytic strains and are therefore typically unactivated when recovered from culture. Serotypes B and F are produced by both proteolytic and nonproteolytic strains and therefore can be recovered in either the active or inactive form. However, even the proteolytic strains that produce, for example, the botulinum toxin type B serotype only cleave a portion of the toxin produced. The exact proportion of nicked to unnicked molecules depends on the length of incubation and the temperature of the culture. Therefore, a certain percentage of any preparation of, for example, the botulinum toxin type B toxin is likely to be inactive, possibly accounting for the known significantly lower potency of botulinum toxin type B as compared to botulinum toxin type A. The presence of inactive botulinum toxin molecules in a clinical preparation will contribute to the overall protein load of the preparation, which has been linked to increased antigenicity, without contributing to its clinical efficacy. Additionally, it is known that botulinum toxin type B has, upon intramuscular injection, a shorter duration of activity and is also less potent than botulinum toxin type A at the same dose level.
  • High quality crystalline botulinum toxin type A can be produced from the Hall A strain of Clostridium botulinum with characteristics of ≧3×107 U/mg, an A260/A278 of less than 0.60 and a distinct pattern of banding on gel electrophoresis. The known Shantz process can be used to obtain crystalline botulinum toxin type A, as set forth in Shantz, E. J., et al, Properties and use of Botulinum toxin and Other Microbial Neurotoxins in Medicine, Microbiol Rev. 56;80-99:1992. Generally, the botulinum toxin type A complex can be isolated and purified from an anaerobic fermentation by cultivating Clostridium botulinum type A in a suitable medium. The known process can also be used, upon separation out of the non-toxin proteins, to obtain pure botulinum toxins, such as for example: purified botulinum toxin type A with an approximately 150 kD molecular weight with a specific potency of 1-2×108 LD50 U/mg or greater; purified botulinum toxin type B with an approximately 156 kD molecular weight with a specific potency of 1-2×108 LD50 U/mg or greater, and; purified botulinum toxin type F with an approximately 155 kD molecular weight with a specific potency of 1-2×107 LD50 U/mg or greater.
  • Botulinum toxins and/or botulinum toxin complexes can be obtained from List Biological Laboratories, Inc., Campbell, Calif.; the Centre for Applied Microbiology and Research, Porton Down, U.K.; Wako (Osaka, Japan), Metabiologics (Madison, Wis.) as well as from Sigma Chemicals of St Louis, Mo. Pure botulinum toxin can also be used to prepare a pharmaceutical composition.
  • As with enzymes generally, the biological activities of the botulinum toxins (which are intracellular peptidases) is dependant, at least in part, upon their three dimensional conformation. Thus, botulinum toxin type A is detoxified by heat, various chemicals surface stretching and surface drying. Additionally, it is known that dilution of the toxin complex obtained by the known culturing, fermentation and purification to the much, much lower toxin concentrations used for pharmaceutical composition formulation results in rapid detoxification of the toxin unless a suitable stabilizing agent is present. Dilution of the toxin from milligram quantities to a solution containing nanograms per milliliter presents significant difficulties because of the rapid loss of specific toxicity upon such great dilution. Since the toxin may be used months or years after the toxin containing pharmaceutical composition is formulated, the toxin can stabilized with a stabilizing agent such as albumin and gelatin.
  • A commercially available botulinum toxin containing pharmaceutical composition is sold under the trademark BOTOX® (available from Allergan, Inc., of Irvine, Calif.). BOTOX® consists of a purified botulinum toxin type A complex, albumin and sodium chloride packaged in sterile, vacuum-dried form. The botulinum toxin type A is made from a culture of the Hall strain of Clostridium botulinum grown in a medium containing N-Z amine and yeast extract. The botulinum toxin type A complex is purified from the culture solution by a series of acid precipitations to a crystalline complex consisting of the active high molecular weight toxin protein and an associated hemagglutinin protein. The crystalline complex is re-dissolved in a solution containing saline and albumin and sterile filtered (0.2 microns) prior to vacuum-drying. The vacuum-dried product is stored in a freezer at or below −5° C. BOTOX® can be reconstituted with sterile, non-preserved saline prior to intramuscular injection. Each vial of BOTOX® contains about 100 units (U) of Clostridium botulinum toxin type A purified neurotoxin complex, 0.5 milligrams of human serum albumin and 0.9 milligrams of sodium chloride in a sterile, vacuum-dried form without a preservative.
  • To reconstitute vacuum-dried BOTOX®, sterile normal saline without a preservative; (0.9% Sodium Chloride Injection) is used by drawing up the proper amount of diluent in the appropriate size syringe. Since BOTOX® may be denatured by bubbling or similar violent agitation, the diluent is gently injected into the vial. For sterility reasons BOTOX® is preferably administered within four hours after the vial is removed from the freezer and reconstituted. During these four hours, reconstituted BOTOX® can be stored in a refrigerator at about 2° C. to about 8° C. Reconstituted, refrigerated BOTOX® has been reported to retain its potency for at least about two weeks. Neurology, 48:249-53:1997.
  • It is known that botulinum toxin type A can have an efficacy for up to 12 months (European J. Neurology 6 (Supp 4): S111-S1150:1999), and in some circumstances for as long as 27 months. The Laryngoscope 109:1344-1346:1999. However, the usual duration of an intramuscular injection of Botox® is typically about 3 to 4 months.
  • The success of botulinum toxin type A to treat a variety of clinical conditions has led to interest in other botulinum toxin serotypes. Two commercially available botulinum type A preparations for use in humans are BOTOX® available from Allergan, Inc., of Irvine, Calif., and Dysport® available from Beaufour Ipsen, Porton Down, England. A botulinum toxin type B preparation (MyoBloc®) is available from Elan Pharmaceuticals of San Francisco, Calif.
  • Tetanus toxin, as wells as derivatives (i.e. with a non-native targeting moiety), fragments, hybrids and chimeras thereof can also have therapeutic utility. The tetanus toxin bears many similarities to the botulinum toxins. Thus, both the tetanus toxin and the botulinum toxins are polypeptides made by closely related species of Clostridium (Clostridium tetani and Clostridium botulinum, respectively). Additionally, both the tetanus toxin and the botulinum toxins are dichain proteins composed of a light chain (molecular weight about 50 kD) covalently bound by a single disulfide bond to a heavy chain (molecular weight about 100 kD). Hence, the molecular weight of tetanus toxin and of each of the seven botulinum toxins (non-complexed) is about 150 kD. Furthermore, for both the tetanus toxin and the botulinum toxins, the light chain bears the domain which exhibits intracellular biological (protease) activity, while the heavy chain comprises the receptor binding (immunogenic) and cell membrane translocational domains.
  • Further, both the tetanus toxin and the botulinum toxins exhibit a high, specific affinity for gangliocide receptors on the surface of presynaptic cholinergic neurons. Receptor mediated endocytosis of tetanus toxin by peripheral cholinergic neurons results in retrograde axonal transport, blocking of the release of inhibitory neurotransmifters from central synapses and a spastic paralysis. Contrarily, receptor mediated endocytosis of botulinum toxin by peripheral cholinergic neurons results in little if any retrograde transport, inhibition of acetylcholine exocytosis from the intoxicated peripheral motor neurons and a flaccid paralysis.
  • Finally, the tetanus toxin and the botulinum toxins resemble each other in both biosynthesis and molecular architecture. Thus, there is an overall 34% identity between the protein sequences of tetanus toxin and botulinum toxin type A, and a sequence identity as high as 62% for some functional domains. Binz T. et al., The Complete Sequence of Botulinum Neurotoxin Type A and Comparison with Other Clostridial Neurotoxins, J Biological Chemistry 265(16);9153-9158:1 990.
  • Acetylcholine
  • Each type of neuron in the mammalian nervous system may release only a single type or multiple types of neurotransmifters. The neurotransmitter acetylcholine is secreted by neurons in many areas of the brain, but specifically by the large pyramidal cells of the motor cortex, by several different neurons in the basal ganglia, by the motor neurons that innervate the skeletal muscles, by the preganglionic neurons of the autonomic nervous system (both sympathetic and parasympathetic), by the postganglionic neurons of the parasympathetic nervous system, and by some of the postganglionic neurons of the sympathetic nervous system. Essentially, only the postganglionic sympathetic nerve fibers to the sweat glands, the piloerector muscles and a few blood vessels are cholinergic as most of the postganglionic neurons of the sympathetic nervous system secret the neurotransmitter norepinephine. In most instances acetylcholine has an excitatory effect. However, acetylcholine is known to have inhibitory effects at some of the peripheral parasympathetic nerve endings, such as inhibition of heart rate by the vagal nerve.
  • The efferent signals of the autonomic nervous system are transmitted to the body through either the sympathetic nervous system or the parasympathetic nervous system. The preganglionic neurons of the sympathetic nervous system extend from preganglionic sympathetic neuron cell bodies located in the intermediolateral horn of the spinal cord. The preganglionic sympathetic nerve fibers, extending from the cell body, synapse with postganglionic neurons located in either a paravertebral sympathetic ganglion or in a prevertebral ganglion. Since, the preganglionic neurons of both the sympathetic and parasympathetic nervous system are cholinergic, application of acetylcholine to the ganglia will excite both sympathetic and parasympathetic postganglionic neurons.
  • Acetylcholine activates two types of receptors, muscarinic and nicotinic receptors. The muscarinic receptors are found in all effector cells stimulated by the postganglionic, neurons of the parasympathetic nervous system as well as in those stimulated by the postganglionic cholinergic neurons of the sympathetic nervous system. The nicotinic receptors are found in the adrenal medulla, as well as within the autonomic ganglia, that is on the cell surface of the postganglionic neuron at the synapse between the preganglionic and postganglionic neurons of both the sympathetic and parasympathetic systems. Nicotinic receptors are also found in many nonautonomic nerve endings, for example in the membranes of skeletal muscle fibers at the neuromuscular junction.
  • Acetylcholine is released from cholinergic neurons when small, clear, intracellular vesicles fuse with the presynaptic neuronal cell membrane. A wide variety of non-neuronal secretory cells, such as, adrenal medulla (as well as the PC12 cell line) and pancreatic islet cells release catecholamines and parathyroid hormone, respectively, from large dense-core vesicles. The PC12 cell line is a clone of rat pheochromocytoma cells extensively used as a tissue culture model for studies of sympathoadrenal development. Botulinum toxin inhibits the release of both types of compounds from both types of cells in vitro, permeabilized (as by electroporation) or by direct injection of the toxin into the denervated cell. Botulinum toxin is also known to block release of the neurotransmitter glutamate from cortical synaptosomes cell cultures.
  • A neuromuscular junction is formed in skeletal muscle by the proximity of axons to muscle cells. A signal transmitted through the nervous system results in an action potential at the terminal axon, with activation of ion channels and resulting release of the neurotransmitter acetylcholine from intraneuronal synaptic vesicles, for example at the motor endplate of the neuromuscular junction. The acetylcholine crosses the extracellular space to bind with acetylcholine receptor proteins on the surface of the muscle end plate. Once sufficient binding has occurred, an action potential of the muscle cell causes specific membrane ion channel changes, resulting in muscle cell contraction. The acetylcholine is then released from the muscle cells and metabolized by cholinesterases in the extracellular space. The metabolites are recycled back into the terminal axon for reprocessing into further acetylcholine.
  • A composition, carrier component or other material is “ophthalmically acceptable” when it is compatible with ocular tissue such that it does not cause significant or undue detrimental effects when brought into contact with ocular tissue. In some embodiments, the ophthalmically acceptable material is also compatible with other components of the present compositions.
  • In some embodiment, an ophthalmically acceptable carrier comprises water and has a pH in a range of about 6.7 to about 7.4, or about 6.8 to about 7.2. In some embodiment, the carrier component comprises an electrolyte, for example, calcium, magnesium and/or mixtures thereof, in an amount which is ophthalmically acceptable.
  • In some embodiments, the polyanionic component comprises an anionic cellulosic derivative. For example, the polyanionic component may comprise carboxy methyl celluloses, anionic homopolymers and copolymers comprising units of one or more of acrylic acid, methacrylic acid, metal acrylates and metal methacrylates, and/or mixture thereof. The present compositions may comprise about 0.05% to about 5% (w/v), or 0.3% to about 2% of a polyanionic component.
  • In some embodiments, the compositions further comprise a vasoconstrictor. Non-limiting examples of vasoconstrictors that may be used in accordance with the present invention include: tetrahydrozoline, ephedrine, naphazoline, phenylephrine, and/or mixtures thereof. In one useful embodiment, the present composition comprises about 0.001% to about 0.5% (w/v), or about 0.005% to about 0.2% (w/v) of the vasoconstrictor.
  • In some embodiments, the pH of the compositions is about 6 to about 8, about 6.8 to about 7.5, more preferably about 6.8 to about 7.2, or about 7 to about 7.2.
  • The compositions of the present invention may also include a suitable tonicity adjusting components. In some embodiments, tonicity adjusting components include, but are not limited to, sodium borate, boric acid, sodium chloride, potassium chloride, mannitol, dextrose, glycerin, propylene glycol, and the like and/or mixtures thereof.
  • The present compositions may be prepared using conventional procedures and techniques, which are well known to the skilled artisan. For example, the present compositions can be prepared by blending the components together, such as in one bulk.
  • The present compositions may be effectively used, as needed, by methods which comprise administering an effective amount of the compositions to an eye in need of treatment for an eye disorder. The administering step may be repeated as needed to provide effective reduction of inflammation of such eye.
  • EXAMPLES Example 1 Method of Treating Bacterial and Fungal Conjunctivitis
  • Conjunctivitis is an inflammation of the conjunctiva characterized by hyperemia (“red eye”), discharge, foreign-body sensation, and/or stuck eye lids often during sleep. It can be caused by infection of the conjunctiva by bacteria and fungi. A bacteria infection is determined by, for example, colorful discharge, papillae and corneal staining pattern. A fungal infection is determined by, for example, a feathery border at the corneal infection site. Both may be confirmed by culture and sensitivity lab testing. If there is a history of trauma, particularly with vegetable matter (e.g., a tree branch), fungal conjunctivitis is often involved. The following presents a typical treatment protocol that may be carried out for bacterial and fungal conjunctivitis.
  • A 20-year-old woman presents with a “red eye”, foreign-body sensation, and yellowish discharge from the right eye which has lasted for 3 days. She also reports that her lids are stuck together in the morning. This patient is diagnosed with bacterial conjunctivitis in the right eye.
  • The doctor treats the patient by, for example, topically administering one drop (comprising about 1 unit of a botulinum toxin type A or alternately comprising about 50 units of a botulinum toxin type B) of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A. Additionally, the treatment may be supplemented with topical antibiotic drops (e.g. Polytrim 4× a day for 5 to 7 days) at the discretion of the doctor.
  • If the diagnosis were fungal conjunctivitis, the patient would be treated by topically applying one drop (comprising about 1 unit of a botulinum toxin type A or alternately comprising about 50 units of a botulinum toxin type B) of the present composition. Also, the treatment may be supplemented with topical antifungal drops—5% Natamycin drops every 1-2 hours while awake, and every 2 hours at night.
  • One week after the application of the Clostridial toxin, e.g. botulinum toxin type A, to the afflicted eye, the patient may return to the optometrist for a follow up visit. The patient's condition is improved by at least 50%, such that the inflammation of the eye subsides and the redness is substantially cleared.
  • Example 2 Method of Treating Viral Conjunctivitis
  • Viral conjunctivitis tends to occur with a history of a recent upper respiratory tract infection or contact with someone with conjunctivitis. It usually starts in one eye and then involves the other eye a few days later. The viral infection is determined by mild hyperemia (pink eye), excessive tearing, foreign body sensation, follicular conjunctival reaction and may involve lymph node tenderness. The following presents a typical treatment protocol that may be carried out for viral conjunctivitis.
  • A 34-year-old man presents with a “red eye” that started about 3 days ago in the left eye and then spread to the right eye. The eyes are watery, swollen, and mildly red. He reports having a flu for the past week. This patient is diagnosed with viral conjunctivitis.
  • The doctor treats the patient by topically administering to the patient's eye one drop (comprising about 1 unit of a botulinum toxin type A or alternately comprising about 50 units of a botulinum toxin type B) of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A. Treatment may further include the use of artificial tears, cool compresses and antihistamine drops (if itchiness is severe).
  • Five days after the application of the composition comprising a Clostridial toxin, e.g. botulinum toxin type A, to the afflicted eye, the patient may return to the optometrist for a follow up visit. The patient's condition may be improved by at least 40%, such that the inflammation of the eye subsided and the redness is substantially cleared.
  • Example 3 Method of Treating Uveitis
  • Uveitis is a general term referring to inflammation of the uveal tract (iris, ciliary body, and choroid). Although it refers primarily to inflammation of this vascular structure, adjacent structures such as retina, vitreous, sclera, and cornea are also frequently involved. Patients most afflicted are 20-50 years of age, with a marked decrease after the age of 70.
  • Anterior uveitis (iritis) occurs more frequently than posterior uveitis and affects the iris and/or ciliary body. Anterior uveitis, especially acute uveitis, is usually marked by eye pain, redness, photophobia (light sensitivity), mildly decreased vision, and tearing, and may be unilateral or bilateral depending on the etiology. The critical sign of uveitis is cells and flare (white blood cells and protein leakage) in the anterior chamber. Many cases of acute, non-recurrent, anterior uveitis tend to be idiopathic and are treated primarily with anti-inflammatory/steroid drops. Other causes of acute anterior uveitis (which can be recurrent) include: ocular trauma, post-surgical inflammation, medications, contact lens-related complications, HLA-B27 antigen, and inflammatory/autoimmune conditions (ankylosing spondylitis, inflammatory bowel disease, Reiter's syndrome, etc.) In cases of chronic anterior uveitis, the etiology is usually due to other systemic conditions such as juvenile rheumatoid arthritis, sarcoidosis, herpes simplex/herpes zoster/varicella, tuberculosis, and Fuch's heterochromia iridocyclitis.
  • Posterior uveitis involves the posterior segment of the eye (with corresponding retinal/choroidal inflammation and lesions). The onset may be acute but most often is insidious with little pain and minimum photophobia and blurred vision. Diseases with associated posterior uveitis include Lyme disease, toxoplasmosis, toxocariasis, histoplasmosis, and syphilis. The following presents a typical treatment protocol that may be carried out for uveitis.
  • A 25-year-old woman presents with a red and irritated left eye. She reports that it started feeling painful and tearing up 1 day ago, with increased pain when she goes out into bright sunlight. Vision is slightly blurring out of the left eye. Symptoms are reported to be moderate. She reports good health overall, is not taking any medications, and a negative history of trauma. She is diagnosed with a moderate case of acute anterior uveitis/iritis upon exam (presence of cells and flare, conjunctival injection, photophobia), which is probably idiopathic.
  • The doctor treats the patient by topically administering two drops (comprising about 2 units of a botulinum toxin type A or alternately comprising about 100 units of a botulinum toxin type B) of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A. Additionally, if it is deemed necessary, the doctor may supplement the treatment with topical steroid drops (1% prednisolone acetate every 2 hours for 2 days, then taper to 4× a day for 4 days, then 2× a day for 4 more days).
  • After one week, the patient returns to the doctor's office for a follow up visit. The symptoms of uveitis appears to have subsided. In particular, the inflammation of the uveal tract (iris, ciliary body, and choroid) appears to have subsided by about 50%.
  • Example 4 Method of Treating Keratic Precipitates
  • Keratic precipitates are associated with acute or chronic anterior uveitis (inflammation of the uveal tract). In this condition, the critical sign seen in the eyes is cellular infiltrates of lymphocytes and plasma cells (cells and flare) and also the presence of precipitates on the corneal endothelium and/or pupillary border known as keratic precipitates (KPs). Depending on the etiology of the uveitis, KPs can be fine or large. The following presents a typical treatment protocol that may be carried out for KPs.
  • A 29-year-old man presents with complaints of red eyes and extreme photophobia, onset 2 days ago. Exam reveals presence of moderate cells and flare and a few large KPs along the pupillary border of both eyes. The patient reports a history of Reiter's syndrome and has had recurrent episodes of uveitis in the past 3 years. The patient is diagnosed with acute, anterior chamber uveitis secondary to Reiter's syndrome.
  • The doctor treats the patient by topically administering one drop of a composition comprising a Clostridial toxin, e.g. botulinum toxin type A, to the patient's eye. At the doctor's discretion, she can additionally place the patient on topical steroid drops (1% prednisolone acetate every 1 hour for 1-2 days, then taper). Furthermore, due to the systemic etiology of the uveitis, other treatment options may call for medical and rheumatological consultation.
  • About 3-4 days after the administration of A Clostridial toxin, e.g. botulinum toxin type A, the patient returns the doctor's office for a follow up. The patient shows signs of improvement, with reduced symptoms of photophobia, along with a mild presence of cells and flare, and the keratic precipitates are resolving.
  • Example 5 Method of Treating Macular Edema (Cystoid Macular Edema)
  • Retinal edema is characterized by swelling of the retinal tissue due to serous leakage. Typically, retinal edema occurs after any type of ocular surgery (i.e. cataract surgery), and is associated with inflammation. Other etiologies include diabetic retinopathy, uveitis, and age-related macular degeneration. The following presents a typical treatment protocol that may be carried out for macular edema.
  • A 73-year-old man presents with decreased vision about 6 weeks after cataract surgery with surgical complication of vitreous loss. The patient has been compliant with all post-surgical regimen. Upon retinal exam, the macula appears to be edematous, with the macular tissue being slightly raised in comparison to the surrounding retina. Accordingly, this patient's visual acuities were moderately reduced. This patient is diagnosed with cystoid macular edema.
  • The doctor treats the patient by topically administering to the patient's eye a composition comprising a Clostridial toxin, e.g. about 1 unit of a botulinum toxin type A or alternately about 50 units of a botulinum toxin type B is administered. The doctor may additionally put the patient on topical non-steroidal anti-inflammatory medication (ketorolac 4× a day) for 6 weeks.
  • Two to three weeks after the administration of A Clostridial toxin, e.g. botulinum toxin type A, the patient returns the doctor's office for a follow up. The patient shows signs of improvement. On retinal exam, the macula is flat and the patient is regaining his vision.
  • Example 6 Method of Treating Inflammation Response After Intra-Ocular Lens Implantation (Cataract Surgery)
  • Inflammation after ocular surgeries (particularly cataract surgery) can involve the retina, resulting in cystoid macular edema. The incidence increases with surgical complications such as iris prolapse and vitreous loss. The following presents a typical treatment protocol that may be carried out for inflammation response after intra-ocular lens implantation.
  • A 69-year-old woman presents with decreased vision about 6 weeks after cataract surgery with surgical complication of vitreous loss. The patient has been compliant with all post-surgical regimen. This patient is diagnosed with cystoid macular edema (a condition associated with an inflammatory response).
  • The doctor treats the patient by topically administering to the patient's eye a formulation comprising A Clostridial toxin, e.g. about 1 unit of a botulinum toxin type A or alternately about 50 units of a botulinum toxin type B is administered. The doctor may additionally put the patient on topical non-steroidal anti-inflammatory medication (ketorolac 4× a day) for 6 weeks.
  • Three weeks after the administration of the Clostridial toxin, e.g. botulinum toxin type A, the patient returns the doctor's office for a follow up. The patient shows signs of improvement, including less macular edema and improved vision.
  • Various references have been cited herein. The disclosures of each these references are incorporated in their entirety herein by reference.
  • While this invention has been described with respect to various specific examples and embodiments, it is to be understood that the invention is not limited thereto and that it can be variously practiced with the scope of the following claims.

Claims (14)

1. A method for treating an eye disorder comprising:
administering an ophthalmic composition to a cornea of a mammal to treat the eye disorder, wherein the ophthalmic composition is a solution and includes an ophthalmically acceptable carrier, a Clostridial toxin in an amount effective to treat the eye disorder when the composition is administered to an eye and a polyanionic component in amount effective to provide lubrication to the eye, thereby treating the eye disorder.
2. The method of claim 1 wherein the ophthalmic composition includes a vasoconstrictor.
3. The method of claim 1 wherein the eye disorder is associated with an inflammation of the eye.
4. The method of claim 1 wherein the eye disorder is bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, or inflammation response after intra-ocular lens implantation.
5. The method of claim 1 wherein the eye disorder is conjunctivitis.
6. The method of claim 1 wherein the Clostridial toxin is administered topically to the cornea.
7. The method of claim 1 wherein the Clostridial toxin is produced by Clostridial beratti, Clostridia butyricum, Clostridial tetani bacterium or Clostridial botulinum.
8. The method of claim 1 wherein Clostridial toxin is a botulinum toxin selected from the group consisting of botulinum types A, B, C1, D, E, F, G and mixtures thereof.
9. The method of claim 1 wherein the Clostridial toxin is botulinum toxin type A.
10. The method of claim 8 wherein the eye disorder is associated with an inflammation of the eye.
11. The method of claim 8 wherein the eye disorder is bacterial conjunctivitis, fungal conjunctivitis, viral conjunctivitis, uveitis, keratic precipitates, macular edema, and inflammation response after intra-ocular lens implantation.
12. The method of claim 8 wherein the eye disorder is conjunctivitis.
13. The method of claim 8 wherein the Clostridial toxin is administered topically to the cornea.
14. The method of claim 8 wherein the Clostridial toxin is administered in conjunction with a vasoconstrictor.
US12/271,573 2003-05-20 2008-11-14 Methods for treating eye disorders Abandoned US20090087459A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/271,573 US20090087459A1 (en) 2003-05-20 2008-11-14 Methods for treating eye disorders
US12/648,260 US20100098727A1 (en) 2003-05-20 2009-12-28 Methods for treating eye disorders

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10/442,590 US7220422B2 (en) 2003-05-20 2003-05-20 Methods and compositions for treating eye disorders
US11/737,641 US7465458B2 (en) 2003-05-20 2007-04-19 Methods for treating eye disorders
US12/271,573 US20090087459A1 (en) 2003-05-20 2008-11-14 Methods for treating eye disorders

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US11/737,641 Continuation US7465458B2 (en) 2003-05-20 2007-04-19 Methods for treating eye disorders
US11/737,641 Division US7465458B2 (en) 2003-05-20 2007-04-19 Methods for treating eye disorders

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/648,260 Continuation US20100098727A1 (en) 2003-05-20 2009-12-28 Methods for treating eye disorders

Publications (1)

Publication Number Publication Date
US20090087459A1 true US20090087459A1 (en) 2009-04-02

Family

ID=33450241

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/442,590 Expired - Fee Related US7220422B2 (en) 2003-05-20 2003-05-20 Methods and compositions for treating eye disorders
US11/737,641 Expired - Fee Related US7465458B2 (en) 2003-05-20 2007-04-19 Methods for treating eye disorders
US12/271,573 Abandoned US20090087459A1 (en) 2003-05-20 2008-11-14 Methods for treating eye disorders
US12/648,260 Abandoned US20100098727A1 (en) 2003-05-20 2009-12-28 Methods for treating eye disorders

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/442,590 Expired - Fee Related US7220422B2 (en) 2003-05-20 2003-05-20 Methods and compositions for treating eye disorders
US11/737,641 Expired - Fee Related US7465458B2 (en) 2003-05-20 2007-04-19 Methods for treating eye disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/648,260 Abandoned US20100098727A1 (en) 2003-05-20 2009-12-28 Methods for treating eye disorders

Country Status (7)

Country Link
US (4) US7220422B2 (en)
EP (1) EP1628678A2 (en)
JP (2) JP2006528703A (en)
AU (1) AU2004249132A1 (en)
BR (1) BRPI0410784A (en)
CA (1) CA2526427A1 (en)
WO (1) WO2004112830A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110280909A1 (en) * 2009-01-29 2011-11-17 Kambiz Thomas Moazed Method and system for effecting changes in pigmented tissue
US20120207809A1 (en) * 2009-01-29 2012-08-16 Kambiz Thomas Moazed Method and system for effecting changes in pigmented tissue

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7341843B2 (en) * 2003-04-11 2008-03-11 Allergan, Inc. Botulinum toxin A peptides and methods of predicting and reducing immunoresistance to botulinum toxin therapy
US7220422B2 (en) * 2003-05-20 2007-05-22 Allergan, Inc. Methods and compositions for treating eye disorders
US20050244365A1 (en) * 2004-05-03 2005-11-03 Novaflux Biosciences, Inc. Methods, compositions, formulations, and uses of cellulose and acrylic-based polymers
US7811584B2 (en) * 2004-06-30 2010-10-12 Allergan, Inc. Multivalent clostridial toxins
US7514088B2 (en) * 2005-03-15 2009-04-07 Allergan, Inc. Multivalent Clostridial toxin derivatives and methods of their use
EP1982996A1 (en) 2004-09-01 2008-10-22 Allergan, Inc. Degradable clostridial toxins
US8343929B2 (en) 2004-09-23 2013-01-01 Toxcure, Inc. Treating neoplasms with neurotoxin
US8052979B2 (en) 2005-03-15 2011-11-08 Allergan, Inc. Modified clostridial toxins with altered targeting capabilities for clostridial toxin target cells
US20080063732A1 (en) * 2006-07-31 2008-03-13 Hantash Basil M Method of increasing the efficacy of neurotoxin
US9161970B2 (en) * 2007-12-12 2015-10-20 Allergan, Inc. Dermal filler
US9044477B2 (en) * 2007-12-12 2015-06-02 Allergan, Inc. Botulinum toxin formulation
ES2599033T3 (en) * 2008-11-26 2017-01-31 Toxcure, Inc. Neoplasms treatment with neurotoxin
KR101135486B1 (en) * 2011-05-25 2012-04-13 함종욱 A liquid product of botulinum a-type toxin
US9393291B2 (en) * 2012-04-12 2016-07-19 Botulinum Toxin Research Associates, Inc. Use of botulinum toxin for the treatment of cerebrovascular disease, renovascular and retinovascular circulatory beds
EP2649984A1 (en) 2012-04-13 2013-10-16 Lipotec, S.A. Compounds which inhibit neuronal exocytosis
KR102113994B1 (en) 2012-04-13 2020-05-25 루브리졸 어드밴스드 머티어리얼스, 인코포레이티드 Compounds which inhibit neuronal exocytosis(ii)
EP2649983A1 (en) 2012-04-13 2013-10-16 Lipotec, S.A. Compounds which inhibit neuronal exocytosis (II)
EP2649985A1 (en) 2012-04-13 2013-10-16 Lipotec, S.A. Compounds which inhibit neuronal exocytosis (III)
US9005628B2 (en) 2012-10-04 2015-04-14 Dublin City University Biotherapy for pain
GB201312317D0 (en) 2013-07-09 2013-08-21 Syntaxin Ltd Cationic neurotoxins
US9216210B2 (en) 2013-12-23 2015-12-22 Dublin City University Multiprotease therapeutics for chronic pain
US9901627B2 (en) 2014-07-18 2018-02-27 Revance Therapeutics, Inc. Topical ocular preparation of botulinum toxin for use in ocular surface disease
US11484580B2 (en) * 2014-07-18 2022-11-01 Revance Therapeutics, Inc. Topical ocular preparation of botulinum toxin for use in ocular surface disease
PT3242884T (en) 2015-01-09 2021-04-22 Ipsen Bioinnovation Ltd Cationic neurotoxins
GB201517450D0 (en) 2015-10-02 2015-11-18 Ipsen Biopharm Ltd Method
GB201607901D0 (en) 2016-05-05 2016-06-22 Ipsen Biopharm Ltd Chimeric neurotoxins
EP3263710A1 (en) 2016-07-01 2018-01-03 Ipsen Biopharm Limited Production of activated clostridial neurotoxins
CN109790204A (en) 2016-09-29 2019-05-21 益普生生物制药有限公司 Heterozygosis neurotoxin
US11123411B2 (en) 2016-12-08 2021-09-21 Gary E. Borodic Method of treating macular degeneration using botulinum toxin-based pharmaceuticals
US11096993B2 (en) 2016-12-08 2021-08-24 Gary E. Borodic Method of treating macular degeneration using botulinum toxin-based pharmaceuticals
EP3758611A1 (en) 2018-02-26 2021-01-06 Ipsen Biopharm Limited Use of ultrasound to guide injection of non-cytotoxic protease
GB202103372D0 (en) 2021-03-11 2021-04-28 Ipsen Biopharm Ltd Modified clostridial neurotoxins

Citations (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US638509A (en) * 1899-01-31 1899-12-05 Adolph Haenichen Self-adjusting spindle for belt-spinners.
US5061714A (en) * 1989-03-28 1991-10-29 Nisshin Flour Milling Co., Ltd. Isoquinoline composition for the treatment of glaucoma or ocular hypertension
US5188826A (en) * 1988-02-08 1993-02-23 Insite Vision Incorporated Topical ophthalmic suspensions
US5250293A (en) * 1991-04-22 1993-10-05 Gleich Gerald J Method for the treatment of hypersensitivity diseases by administration of anionic polymers
US5401243A (en) * 1990-08-21 1995-03-28 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US5437291A (en) * 1993-08-26 1995-08-01 Univ Johns Hopkins Method for treating gastrointestinal muscle disorders and other smooth muscle dysfunction
US5616122A (en) * 1986-11-04 1997-04-01 Baylor College Of Medicine Methods and compositions for preventing secondary cataracts
US5670484A (en) * 1994-05-09 1997-09-23 Binder; William J. Method for treatment of skin lesions associated with cutaneous cell-proliferative disorders
US5679713A (en) * 1991-08-13 1997-10-21 Astra Aktiebolag Pharmaceutical composition containing carbachol and other cholinergic substances
US5713381A (en) * 1996-11-08 1998-02-03 Sloane; Terri Eye care kit
US5714468A (en) * 1994-05-09 1998-02-03 Binder; William J. Method for reduction of migraine headache pain
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
US5827862A (en) * 1993-02-23 1998-10-27 Otsuka Pharmaceutical Company, Limited Agent for prophylaxis or treatment of cataract
US5863892A (en) * 1992-02-26 1999-01-26 Allergan Inc. Use of platelet derived growth factor in ophthalmic wound healing
US5942487A (en) * 1996-11-29 1999-08-24 Senju Pharmaceutical Co., Ltd. Composition for treating cornea
US5989545A (en) * 1995-04-21 1999-11-23 The Speywood Laboratory Ltd. Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US6025329A (en) * 1995-08-09 2000-02-15 Toray Industries, Inc. Method for treating ophthalmic diseases
US6024954A (en) * 1994-12-12 2000-02-15 Allergan Compositions and methods for disinfecting contact lenses and preserving contact lens care products
US6063768A (en) * 1997-09-04 2000-05-16 First; Eric R. Application of botulinum toxin to the management of neurogenic inflammatory disorders
US6133915A (en) * 1998-06-17 2000-10-17 Microsoft Corporation System and method for customizing controls on a toolbar
US6139845A (en) * 1999-12-07 2000-10-31 Allergan Sales, Inc. Method for treating cancer with a neurotoxin
US6143306A (en) * 2000-01-11 2000-11-07 Allergan Sales, Inc. Methods for treating pancreatic disorders
US6194457B1 (en) * 1997-01-29 2001-02-27 A. Glenn Braswell Liquid eye drop composition
US6265379B1 (en) * 1999-10-13 2001-07-24 Allergan Sales, Inc. Method for treating otic disorders
US6299893B1 (en) * 2000-04-17 2001-10-09 Marvin Schwartz Method to reduce hair loss and stimulate hair regrowth
US6306403B1 (en) * 2000-06-14 2001-10-23 Allergan Sales, Inc. Method for treating parkinson's disease with a botulinum toxin
US6306423B1 (en) * 2000-06-02 2001-10-23 Allergan Sales, Inc. Neurotoxin implant
US6328977B1 (en) * 2000-02-22 2001-12-11 Allergan Sales, Inc. Method for treating hyperparathyroidism
US20020025320A1 (en) * 1999-08-20 2002-02-28 Prosper Boyaka Sialidases as mucosal adjuvants
US20020025778A1 (en) * 2000-08-28 2002-02-28 Lg Electronics Inc. Integrated wireless local loop (WLL) and wireless local area network (WLAN) transceiver apparatus
US6358513B1 (en) * 2000-02-15 2002-03-19 Allergan Sales, Inc. Method for treating Hashimoto's thyroiditis
US20020035061A1 (en) * 1996-08-21 2002-03-21 Timothy J. Krieger Compositions and methods for treating infections using cationic peptides alone or in combination with antibiotics
US6365164B1 (en) * 1997-07-15 2002-04-02 University Technology Corporation Use of neurotoxin therapy for treatment of urologic and related disorders
US6395736B1 (en) * 1998-12-14 2002-05-28 Cellegy Pharmaceuticals, Inc. Compositions and methods for the treatment of anorectal disorders
US6395277B1 (en) * 1991-09-24 2002-05-28 Allergan Method and compositions for the treatment of cerebral palsy
US20020064536A1 (en) * 2000-02-08 2002-05-30 Allergan Sales, Inc. Methods of treating animals with botulinum toxin pharmaceutical compositions
US6406692B1 (en) * 1997-10-31 2002-06-18 New Horizons Diagnostics Corp Composition for treatment of an ocular bacterial infection
US6423319B1 (en) * 2000-10-04 2002-07-23 Allergan Sales, Inc. Methods for treating muscle injuries
US6429189B1 (en) * 1999-12-10 2002-08-06 Botulinum Toxin Research Associates, Inc. Cytotoxin (non-neurotoxin) for the treatment of human headache disorders and inflammatory diseases
US6440964B1 (en) * 1998-09-30 2002-08-27 Alcon Manufacturing, Ltd. Compositions and methods for treating ophthalmic and otic infections
US6458365B1 (en) * 1993-12-28 2002-10-01 Allergan, Inc. Method for treating headache
US6464986B1 (en) * 2000-04-14 2002-10-15 Allegan Sales, Inc. Method for treating pain by peripheral administration of a neurotoxin
US20020192239A1 (en) * 2001-01-09 2002-12-19 Borodic Gary E. Use of botulinum toxin for the treatment of chronic facial pain
US20030008834A1 (en) * 2000-08-21 2003-01-09 Yerxa Benjamin R. Compsoitions and methods for treating epithelia and retinal tissue diseases
US6562318B1 (en) * 1990-09-14 2003-05-13 Syngenix Limited Particular agents
US20030092640A1 (en) * 2001-05-31 2003-05-15 Pfizer Inc. Azalide antibiotic compositions
US20030114830A1 (en) * 2001-12-13 2003-06-19 Guerrero John M. Method and apparatus for treatment of amblyopia
US20040058313A1 (en) * 2002-04-24 2004-03-25 Abreu Marcio Marc Compositions, targets, methods and devices for the therapy of ocular and periocular disorders
US6743424B1 (en) * 2000-02-15 2004-06-01 Allergan, Inc. Method for treating hyperthyroidism
US20040122254A1 (en) * 1997-08-28 2004-06-24 Fujimoto Roger A. Certain 5-alkyl-2-arylaminophenylacetic acids and derivatives
US20040120956A1 (en) * 2002-06-27 2004-06-24 Song Xiao-Yu R. CNGH0004 polypeptides, antibodies, compositions, methods and uses
US20040170665A1 (en) * 2000-06-02 2004-09-02 Allergan, Inc. Intravitreal botulinum toxin implant
US20040204471A1 (en) * 2003-03-20 2004-10-14 Pharmacia Corporation Treatment and prevention of otic disorders with Cox-2 inhibitors alone or in combination with otic agents
US20040213811A1 (en) * 2003-04-25 2004-10-28 Allergan, Inc. Use of a botulinum neurotoxin to alleviate various disorders
US20040220100A1 (en) * 2000-07-21 2004-11-04 Essentia Biosystems, Inc. Multi-component biological transport systems
US20040265935A1 (en) * 2003-04-11 2004-12-30 Atassi M. Zouhair Botulinum toxin a peptides and methods of predicting and reducing immunoresistance to botulinum toxin therapy
US20050043410A1 (en) * 2002-01-23 2005-02-24 Brazzell Romulus Kimbro Pharmaceutical use of cox-2-inhibitors in angiogenesis-mediated ocular disorders
US20050059595A1 (en) * 2001-04-12 2005-03-17 Dana Lasko Treatment of macular degeneration with ADP-ribosyl transferase fusion protein therapeutic compositions
US6949241B2 (en) * 2000-05-04 2005-09-27 Ilona Molnarne-Kahan Use of lacrophyl preparation in eye drops containing therapeutically active compounds
US20060067950A1 (en) * 2004-09-27 2006-03-30 Merz Pharma Gmbh & Co. Kgaa Clostridial neurotoxins for use in wound healing
US20060106104A1 (en) * 2004-11-16 2006-05-18 Allergan, Inc. Ophthalmic compositions and methods for treating eyes
US20060182783A1 (en) * 2004-04-30 2006-08-17 Allergan, Inc. Sustained release intraocular drug delivery systems
US20060182767A1 (en) * 2002-05-28 2006-08-17 Borodic Gary E High-potency botulinum toxin formulations
US20060211752A1 (en) * 2004-03-16 2006-09-21 Kohn Leonard D Use of phenylmethimazoles, methimazole derivatives, and tautomeric cyclic thiones for the treatment of autoimmune/inflammatory diseases associated with toll-like receptor overexpression
US20070059336A1 (en) * 2004-04-30 2007-03-15 Allergan, Inc. Anti-angiogenic sustained release intraocular implants and related methods
US7211261B1 (en) * 1998-09-11 2007-05-01 Solstice Neurosciences, Inc. Stable liquid formulations of botulinum toxin
US7220422B2 (en) * 2003-05-20 2007-05-22 Allergan, Inc. Methods and compositions for treating eye disorders
US7390496B2 (en) * 2003-04-25 2008-06-24 Allergan, Inc. Therapeutic treatments for repetitive hand washing
US20090062315A1 (en) * 2007-08-29 2009-03-05 Yung Shin Pharmaceutical Industrial Co., Ltd. Pharmaceutical Acceptable Composition Containing Non-Steroidal Anti-Inflammatory Drug and Local Anesthetics
US7537773B1 (en) * 1998-08-25 2009-05-26 Botulinum Toxin Research Associates, Inc. Chemodenervating pharmaceutical as anti-inflammatory agent

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1757325B1 (en) 1993-12-28 2010-11-03 Allergan, Inc. Use of botulinum toxin for treating myofascial pain
TW498102B (en) * 1998-12-28 2002-08-11 Futaba Denshi Kogyo Kk A process for preparing GaN fluorescent substance
ITMI991453A1 (en) 1999-07-01 2001-01-01 Farmila Farma Milano OPHTHALMIC COMPOSITIONS IN THE FORM OF AQUEOUS GEL
US6113915A (en) * 1999-10-12 2000-09-05 Allergan Sales, Inc. Methods for treating pain
US7052714B1 (en) * 1999-10-13 2006-05-30 Senju Pharmaceutical Co., Ltd Ophthalmic adhesive preparations for percutaneous adsorption
AU2001270219A1 (en) * 2000-06-28 2002-01-08 Ira Sanders Methods for using tetanus toxin for benificial purposes in animals (mammals)
JP2002308764A (en) * 2001-02-09 2002-10-23 Taisho Pharmaceut Co Ltd Pharmaceutical composition for ophthalmic use
AR033151A1 (en) * 2001-04-12 2003-12-03 Sucampo Pharmaceuticals Inc AGENT FOR THE TOPICAL OPHTHALMIC TREATMENT OF OCULAR INFLAMMATORY DISEASES
AR044985A1 (en) * 2003-07-02 2005-10-12 Alza Corp IMMUNIZATION METHOD AND PATCH BY MICROPROJECTION PROVISION

Patent Citations (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US638509A (en) * 1899-01-31 1899-12-05 Adolph Haenichen Self-adjusting spindle for belt-spinners.
US5616122A (en) * 1986-11-04 1997-04-01 Baylor College Of Medicine Methods and compositions for preventing secondary cataracts
US5188826A (en) * 1988-02-08 1993-02-23 Insite Vision Incorporated Topical ophthalmic suspensions
US5061714A (en) * 1989-03-28 1991-10-29 Nisshin Flour Milling Co., Ltd. Isoquinoline composition for the treatment of glaucoma or ocular hypertension
US5401243A (en) * 1990-08-21 1995-03-28 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US6562318B1 (en) * 1990-09-14 2003-05-13 Syngenix Limited Particular agents
US5250293A (en) * 1991-04-22 1993-10-05 Gleich Gerald J Method for the treatment of hypersensitivity diseases by administration of anionic polymers
US5679713A (en) * 1991-08-13 1997-10-21 Astra Aktiebolag Pharmaceutical composition containing carbachol and other cholinergic substances
US6395277B1 (en) * 1991-09-24 2002-05-28 Allergan Method and compositions for the treatment of cerebral palsy
US5863892A (en) * 1992-02-26 1999-01-26 Allergan Inc. Use of platelet derived growth factor in ophthalmic wound healing
US5827862A (en) * 1993-02-23 1998-10-27 Otsuka Pharmaceutical Company, Limited Agent for prophylaxis or treatment of cataract
US5437291A (en) * 1993-08-26 1995-08-01 Univ Johns Hopkins Method for treating gastrointestinal muscle disorders and other smooth muscle dysfunction
US6458365B1 (en) * 1993-12-28 2002-10-01 Allergan, Inc. Method for treating headache
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
US5714468A (en) * 1994-05-09 1998-02-03 Binder; William J. Method for reduction of migraine headache pain
US5670484A (en) * 1994-05-09 1997-09-23 Binder; William J. Method for treatment of skin lesions associated with cutaneous cell-proliferative disorders
US6024954A (en) * 1994-12-12 2000-02-15 Allergan Compositions and methods for disinfecting contact lenses and preserving contact lens care products
US5989545A (en) * 1995-04-21 1999-11-23 The Speywood Laboratory Ltd. Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US6025329A (en) * 1995-08-09 2000-02-15 Toray Industries, Inc. Method for treating ophthalmic diseases
US20020035061A1 (en) * 1996-08-21 2002-03-21 Timothy J. Krieger Compositions and methods for treating infections using cationic peptides alone or in combination with antibiotics
US5713381A (en) * 1996-11-08 1998-02-03 Sloane; Terri Eye care kit
US5942487A (en) * 1996-11-29 1999-08-24 Senju Pharmaceutical Co., Ltd. Composition for treating cornea
US6194457B1 (en) * 1997-01-29 2001-02-27 A. Glenn Braswell Liquid eye drop composition
US6365164B1 (en) * 1997-07-15 2002-04-02 University Technology Corporation Use of neurotoxin therapy for treatment of urologic and related disorders
US20040122254A1 (en) * 1997-08-28 2004-06-24 Fujimoto Roger A. Certain 5-alkyl-2-arylaminophenylacetic acids and derivatives
US6063768A (en) * 1997-09-04 2000-05-16 First; Eric R. Application of botulinum toxin to the management of neurogenic inflammatory disorders
US6406692B1 (en) * 1997-10-31 2002-06-18 New Horizons Diagnostics Corp Composition for treatment of an ocular bacterial infection
US6133915A (en) * 1998-06-17 2000-10-17 Microsoft Corporation System and method for customizing controls on a toolbar
US7537773B1 (en) * 1998-08-25 2009-05-26 Botulinum Toxin Research Associates, Inc. Chemodenervating pharmaceutical as anti-inflammatory agent
US7211261B1 (en) * 1998-09-11 2007-05-01 Solstice Neurosciences, Inc. Stable liquid formulations of botulinum toxin
US6440964B1 (en) * 1998-09-30 2002-08-27 Alcon Manufacturing, Ltd. Compositions and methods for treating ophthalmic and otic infections
US6395736B1 (en) * 1998-12-14 2002-05-28 Cellegy Pharmaceuticals, Inc. Compositions and methods for the treatment of anorectal disorders
US20020025320A1 (en) * 1999-08-20 2002-02-28 Prosper Boyaka Sialidases as mucosal adjuvants
US6358926B2 (en) * 1999-10-13 2002-03-19 Allergan Sales, Inc. Neurotoxin therapy for inner ear disorders
US6265379B1 (en) * 1999-10-13 2001-07-24 Allergan Sales, Inc. Method for treating otic disorders
US6139845A (en) * 1999-12-07 2000-10-31 Allergan Sales, Inc. Method for treating cancer with a neurotoxin
US6429189B1 (en) * 1999-12-10 2002-08-06 Botulinum Toxin Research Associates, Inc. Cytotoxin (non-neurotoxin) for the treatment of human headache disorders and inflammatory diseases
US6143306A (en) * 2000-01-11 2000-11-07 Allergan Sales, Inc. Methods for treating pancreatic disorders
US20020064536A1 (en) * 2000-02-08 2002-05-30 Allergan Sales, Inc. Methods of treating animals with botulinum toxin pharmaceutical compositions
US6358513B1 (en) * 2000-02-15 2002-03-19 Allergan Sales, Inc. Method for treating Hashimoto's thyroiditis
US6743424B1 (en) * 2000-02-15 2004-06-01 Allergan, Inc. Method for treating hyperthyroidism
US6328977B1 (en) * 2000-02-22 2001-12-11 Allergan Sales, Inc. Method for treating hyperparathyroidism
US6464986B1 (en) * 2000-04-14 2002-10-15 Allegan Sales, Inc. Method for treating pain by peripheral administration of a neurotoxin
US6299893B1 (en) * 2000-04-17 2001-10-09 Marvin Schwartz Method to reduce hair loss and stimulate hair regrowth
US6949241B2 (en) * 2000-05-04 2005-09-27 Ilona Molnarne-Kahan Use of lacrophyl preparation in eye drops containing therapeutically active compounds
US6585993B2 (en) * 2000-06-02 2003-07-01 Allergan, Inc. Controlled release neurotoxin system
US20040170665A1 (en) * 2000-06-02 2004-09-02 Allergan, Inc. Intravitreal botulinum toxin implant
US6312708B1 (en) * 2000-06-02 2001-11-06 Allergan Sales, Inc. Botulinum toxin implant
US6306423B1 (en) * 2000-06-02 2001-10-23 Allergan Sales, Inc. Neurotoxin implant
US6306403B1 (en) * 2000-06-14 2001-10-23 Allergan Sales, Inc. Method for treating parkinson's disease with a botulinum toxin
US20040220100A1 (en) * 2000-07-21 2004-11-04 Essentia Biosystems, Inc. Multi-component biological transport systems
US20030008834A1 (en) * 2000-08-21 2003-01-09 Yerxa Benjamin R. Compsoitions and methods for treating epithelia and retinal tissue diseases
US20020025778A1 (en) * 2000-08-28 2002-02-28 Lg Electronics Inc. Integrated wireless local loop (WLL) and wireless local area network (WLAN) transceiver apparatus
US6423319B1 (en) * 2000-10-04 2002-07-23 Allergan Sales, Inc. Methods for treating muscle injuries
US20020192239A1 (en) * 2001-01-09 2002-12-19 Borodic Gary E. Use of botulinum toxin for the treatment of chronic facial pain
US20050059595A1 (en) * 2001-04-12 2005-03-17 Dana Lasko Treatment of macular degeneration with ADP-ribosyl transferase fusion protein therapeutic compositions
US20090053190A1 (en) * 2001-04-12 2009-02-26 Mckerracher Lisa Treatment of macular degeneration with ADP-ribosyl transferase fusion protein therapeutic compositions
US7442686B2 (en) * 2001-04-12 2008-10-28 Bioaxone Therapeutique Inc. Treatment of macular degeneration with ADP-ribosyl transferase fusion protein therapeutic compositions
US20030092640A1 (en) * 2001-05-31 2003-05-15 Pfizer Inc. Azalide antibiotic compositions
US20030114830A1 (en) * 2001-12-13 2003-06-19 Guerrero John M. Method and apparatus for treatment of amblyopia
US6692481B2 (en) * 2001-12-13 2004-02-17 John M. Guerrero Method and apparatus for treatment of amblyopia
US20050043410A1 (en) * 2002-01-23 2005-02-24 Brazzell Romulus Kimbro Pharmaceutical use of cox-2-inhibitors in angiogenesis-mediated ocular disorders
US20040058313A1 (en) * 2002-04-24 2004-03-25 Abreu Marcio Marc Compositions, targets, methods and devices for the therapy of ocular and periocular disorders
US20060182767A1 (en) * 2002-05-28 2006-08-17 Borodic Gary E High-potency botulinum toxin formulations
US20040120956A1 (en) * 2002-06-27 2004-06-24 Song Xiao-Yu R. CNGH0004 polypeptides, antibodies, compositions, methods and uses
US20040204471A1 (en) * 2003-03-20 2004-10-14 Pharmacia Corporation Treatment and prevention of otic disorders with Cox-2 inhibitors alone or in combination with otic agents
US20040265935A1 (en) * 2003-04-11 2004-12-30 Atassi M. Zouhair Botulinum toxin a peptides and methods of predicting and reducing immunoresistance to botulinum toxin therapy
US7390496B2 (en) * 2003-04-25 2008-06-24 Allergan, Inc. Therapeutic treatments for repetitive hand washing
US7393537B2 (en) * 2003-04-25 2008-07-01 Allergan, Inc. Botulinum toxin for treatment of obsessive compulsive finger biting disorder
US20040213811A1 (en) * 2003-04-25 2004-10-28 Allergan, Inc. Use of a botulinum neurotoxin to alleviate various disorders
US7220422B2 (en) * 2003-05-20 2007-05-22 Allergan, Inc. Methods and compositions for treating eye disorders
US20080199498A1 (en) * 2003-05-20 2008-08-21 Allergan, Inc. Methods for treating eye disorders
US7465458B2 (en) * 2003-05-20 2008-12-16 Allergan, Inc. Methods for treating eye disorders
US20060211752A1 (en) * 2004-03-16 2006-09-21 Kohn Leonard D Use of phenylmethimazoles, methimazole derivatives, and tautomeric cyclic thiones for the treatment of autoimmune/inflammatory diseases associated with toll-like receptor overexpression
US20070059336A1 (en) * 2004-04-30 2007-03-15 Allergan, Inc. Anti-angiogenic sustained release intraocular implants and related methods
US20060182783A1 (en) * 2004-04-30 2006-08-17 Allergan, Inc. Sustained release intraocular drug delivery systems
US20060067950A1 (en) * 2004-09-27 2006-03-30 Merz Pharma Gmbh & Co. Kgaa Clostridial neurotoxins for use in wound healing
US20060106104A1 (en) * 2004-11-16 2006-05-18 Allergan, Inc. Ophthalmic compositions and methods for treating eyes
US20090062315A1 (en) * 2007-08-29 2009-03-05 Yung Shin Pharmaceutical Industrial Co., Ltd. Pharmaceutical Acceptable Composition Containing Non-Steroidal Anti-Inflammatory Drug and Local Anesthetics

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110280909A1 (en) * 2009-01-29 2011-11-17 Kambiz Thomas Moazed Method and system for effecting changes in pigmented tissue
US20120207809A1 (en) * 2009-01-29 2012-08-16 Kambiz Thomas Moazed Method and system for effecting changes in pigmented tissue
US9744237B2 (en) * 2009-01-29 2017-08-29 Kambiz Thomas Moazed Method and system for effecting changes in pigmented tissue

Also Published As

Publication number Publication date
US20080199498A1 (en) 2008-08-21
WO2004112830A2 (en) 2004-12-29
US20100098727A1 (en) 2010-04-22
JP2012107034A (en) 2012-06-07
JP2006528703A (en) 2006-12-21
EP1628678A2 (en) 2006-03-01
BRPI0410784A (en) 2006-06-20
WO2004112830A3 (en) 2005-03-03
AU2004249132A1 (en) 2004-12-29
CA2526427A1 (en) 2004-12-29
US7220422B2 (en) 2007-05-22
US7465458B2 (en) 2008-12-16
US20040234532A1 (en) 2004-11-25

Similar Documents

Publication Publication Date Title
US7465458B2 (en) Methods for treating eye disorders
US8105611B2 (en) Treatment of autoimmune disorder with a neurotoxin
US6838434B2 (en) Methods for treating sinus headache
US8057808B2 (en) Use of a botulinum neurotoxin to alleviate various disorders
US9061025B2 (en) Methods for selecting headache patients responsive to botulinum toxin therapy
US20080118534A1 (en) Therapy for obsessive compulsive head banging
CA2489239C (en) Botulinum toxins for treating priapism
EP2012811B1 (en) Use of botulinum toxin for alleviating testicular pain
US20050152923A1 (en) Methods for treating vascular disorders
US7393538B2 (en) Clostridial toxin treatment for dermatillomania
US7422753B2 (en) Methods for treating trichotillomania

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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