WO2009117481A1 - Use of protease-activated receptor 2 antagonists - Google Patents

Use of protease-activated receptor 2 antagonists Download PDF

Info

Publication number
WO2009117481A1
WO2009117481A1 PCT/US2009/037504 US2009037504W WO2009117481A1 WO 2009117481 A1 WO2009117481 A1 WO 2009117481A1 US 2009037504 W US2009037504 W US 2009037504W WO 2009117481 A1 WO2009117481 A1 WO 2009117481A1
Authority
WO
WIPO (PCT)
Prior art keywords
pain
par2
antagonist
antibody
fracture
Prior art date
Application number
PCT/US2009/037504
Other languages
French (fr)
Inventor
Lynn Macdonald
Richard Torres
Original Assignee
Regeneron Pharmaceuticals, 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 Regeneron Pharmaceuticals, Inc. filed Critical Regeneron Pharmaceuticals, Inc.
Publication of WO2009117481A1 publication Critical patent/WO2009117481A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention is related to therapeutic methods of treating a human protease-activated receptor 2 (PAR2)-related inflammatory condition, including inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, and gout joint pain.
  • PAR2 human protease-activated receptor 2
  • Protease-activated receptor 2 (PAR2, also known as C140 receptor) is a member of a family of GPCRs that become activated by trypsin and the mast cell protease tryptase through proteolytic cleavage of the amino termini, exposing a tethered ligand that binds to the receptors and leads to activation.
  • PAR1 , PAR3 and PAR4 are thrombin receptors.
  • PAR4 is also activated by trypsin. Short synthetic peptides that resemble the tethered ligand sequence have been successfully used to activate all of the PAR receptors, except PAR3.
  • Nucleic acid and amino acid sequences of human PAR2 are described in, for example, US 5,629,174; 5,716,789; 6,297,026; and US Patent Publication No. 2003/0018184.
  • Antibodies to PAR2 are described in, for example, US 5,874,400 and EP 0 804 452 B1. Methods of using a PAR2 antibody are described in US Patent Publication No. 2007/0237759.
  • the invention features methods for inhibiting PAR2 activity with a PAR2 antagonist capable of inhibiting PAR2 proteolytic cleavage and blocking PAR2 activation.
  • the method comprises administering a therapeutically effective amount of a PAR2 antagonist to a subject suffering from a PAR2-related condition or a condition which may be improved or ameliorated by inhibition of PAR2.
  • Such conditions or diseases include, for example, inflammation and/or pain.
  • Inflammatory conditions include, for example, inflammation of the gastrointestinal system or of the airways, rheumatic disorders, osteoarthritis, pain associated with inflammation, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, and gout joint pain.
  • the PAR2 antagonist useful in the method of the invention is a molecule capable of blocking human PAR2 activity, including a molecule which blocks activation of PAR2 by blocking proteolytic cleavage of the N-terminus of the receptor, a molecule which blocks activation of the receptor after cleavage, or a molecule which inhibits PAR2 expression.
  • the PAR2 antagonist may be an antibody, an antigen-binding fragment of an antibody, a peptibody, a soluble "decoy" receptor, or an antisense or siRNA molecule.
  • the PAR2 antagonist is an antibody or an antigen-binding fragment of an antibody.
  • the PAR2 antibody may be a fully human or partially human antibody or antigen-binding fragment of an antibody.
  • the PAR2 antagonist is a fully human antibody or an antigen- binding fragment of an antibody.
  • the therapeutic method of the invention encompasses administration of a PAR2 antagonist as a single agent, or with a second therapeutic agent.
  • the second therapeutic agent may be, for example, one or more of an interleukin-1 (IL-1) inhibitor, an antiepileptic drug, such as gabapentain.
  • IL-1 interleukin-1
  • an antiepileptic drug such as gabapentain.
  • the invention features a therapeutic method of treating neuropathic pain in a human subject with an anti-PAR2 antibody or antigen- binding fragment of an antibody.
  • an anti-PAR2 antagonist As described in the experimental section below, significant alleviation of both established thermal hyperalgia and mechanical allodynia in an animal model of neuropathic pain was achieved with an anti-PAR2 antagonist.
  • the invention features use of a protease-activated receptor 2 (PAR2) antagonist in the manufacture of a medicament for treating, inhibiting or ameliorating an inflammatory condition or disorder, wherein the inflammatory condition or disorder is rheumatoid arthritis or osteoarthritis.
  • PAR2 protease-activated receptor 2
  • the PAR2 antagonist is an anti-human PAR2 antibody or antigen-binding fragment of an antibody; preferably the anti-PAR2 antibody or antibody fragment is fully human.
  • the invention features use of a protease-activated receptor 2 (PAR2) antagonist in the manufacture of a medicament for treating, inhibiting or ameliorating pain, wherein the pain is inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, bone cancer pain and gout joint pain.
  • the inflammatory pain treated is pain associated with irritable bowel syndrome.
  • the PAR2 antagonist is an anti-human PAR2 antibody or antigen-binding fragment of an antibody; preferably the anti-PAR2 antibody or antibody fragment is fully human.
  • the PAR2 antagonist is used as a first therapeutic agent, with one or more further therapeutic agents such as another PAR2 antagonist, a cytokine inhibitor, for example, an interleukin-1 (IL-1) inhibitor (for example, rilonacept or anakinra); an IL-18 inhibitor; an IL-6 inhibitor such as an anti-IL6 or anti-IL6 receptor antibody; an antiepileptic drug, such as gabapentain; an NGF inhibitor such as an anti-NGF antibody; low dose colchicine; aspirin; or other NSAIDs, steroids such as prednisolone, methotrexate; low dose cyclosporine A; a TNF inhibitor; and/or co- therapies such as uric acid synthesis inhibitors (allopurinol); uric acid excretion promoters (probenecid, sulfinpyrazone and/or benzbromarone); and/or corticosteroids.
  • IL-1 interleukin-1
  • IL-18 inhibitor for example,
  • PAR2 Human protease-activated receptor 2
  • PAR2 refers to the protein encoded by the nucleic acid sequence of SEQ ID NO:1 and the protein having the amino acid sequence of SEQ ID NO:2.
  • PAR2 is also called “C140 receptor”.
  • PAR2 antagonist as used herein, is intended to refer to a molecule which inhibits the activity or expression of PAR2.
  • antibody is intended to refer to immunoglobulin molecules comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementary determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementary determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • a “neutralizing” or “blocking” antibody is intended to refer to an antibody whose binding to PAR2 results in inhibition of the biological activity of PAR2. This inhibition of the biological activity of PAR2 can be assessed by measuring one or more indicators of PAR2 biological activity. These indicators of PAR2 biological activity can be assessed by one or more of several standard in vitro or in vivo assays known in the art (see examples below).
  • antigen-binding portion of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., PAR2). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full- length antibody.
  • binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH 1 CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al.
  • VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • scFv single chain Fv
  • single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
  • Other forms of single chain antibodies, such as diabodies are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger et al. (1993) Proc. Natl. Acad Sci. USA 90:6444-6448; Poljak et al. (1994) Structure 2:1121- 1123).
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • human antibody as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • the term "effective amount” is a concentration or amount of a PAR2 antagonist which results in achieving a particular stated purpose.
  • An "effective amount” of an anti- PAR2 antibody or antigen-binding fragment of an antibody thereof, for example, may be determined empirically.
  • a “therapeutically effective amount” is a concentration or amount of a PAR2 antagonist, such as an anti-PAR2 antibody or antigen-binding fragment, which is effective for achieving a stated therapeutic effect This amount may also be determined empirically.
  • the present invention provides a pharmaceutical composition comprising an antagonist of human PAR2, as described above, and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprising a PAR2 antagonist of the present invention as an active ingredient can be used as an agent for preventing and/or treating a disorder or condition that is associated with PAR2, such as, for example, inflammatory pain.
  • compositions of the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • a multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences (15th ed, Mack Publishing Company, Easton, PA).
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles, DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax.
  • Any of the foregoing mixtures may be appropriate in treatments and therapies in accordance with the present invention, provided that the active ingredient in the formulation is not inactivated by the formulation and the formulation is physiologically compatible and tolerable with the route of administration. See also Powell et al. "Compendium of excipients for parenteral formulations" PDA, 1998, J Pharm Sci Technol. 52:238-311 , and the citations therein for additional information related to excipients and carriers well known to pharmaceutical chemists.
  • the dose may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like.
  • the PAR2 antagonist is used for preventing and/or treating, for example, inflammatory pain, in an adult patient
  • it is advantageous to intravenously administer the antagonist normally at a single dose of about 0.01 to about 20 mg/kg body weight, preferably about 0.1 to about 10 mg/kg body weight, and more preferably about 0.1 to about 5 mg/kg body weight, approximately 1 to 5 times per day, preferably approximately 1 to 3 times per day.
  • the antagonist can be administered in a dose corresponding to the dose given above.
  • the dose may be increased according to the condition.
  • Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:44294432).
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the pharmaceutical composition can be delivered in a vesicle, in particular a liposome (see, for example, Langer, 1990, Science 249:1527- 1533).
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton (1987) CRC Grit. Ref. Biomed. Eng. 14:201).
  • polymeric materials can be used (see, for example, Medical Applications of Controlled Release (1974) Langer and Wise (eds.), CRC Pres., Boca Raton, Florida; Controlled Drug Bioavailability, Drug Product Design and Performance, (1984) Smolen and Ball (eds.), Wiley, New York).
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release (1984) supra, vol. 2, pp. 115-138). Other controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533).
  • compositions for oral administration include solid or liquid dosage forms, specifically, tablets (including dragees and film-coated tablets), pills, granules, powdery preparations, capsules (including soft capsules), syrup, emulsions, suspensions, etc.
  • a composition is manufactured by publicly known methods and contains a vehicle, a diluent or an excipient conventionally used in the field of pharmaceutical preparations.
  • vehicle or excipient for tablets are lactose, starch, sucrose, magnesium stearate, and the like.
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known.
  • the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antagonist or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • the oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • the injection thus prepared is preferably filled in an appropriate ampoule.
  • the suppository used for rectal administration may be prepared by blending the aforesaid antagonist molecule or its salt with conventional bases for suppositories.
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of, for example, an anti-PAR2 antibody or antibody fragment, contained is generally about 5 to 500 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the aforesaid antibody is contained in about 5 to 100 mg and in about 10 to 250 mg for the other dosage forms.
  • the present invention further provides a method for preventing or treating a disorder or condition that is associated with or involves PAR2, the method comprising administering an effective amount of at least one human PAR2 antagonists to a subject in need thereof.
  • the invention provides therapeutic methods in which a PAR2 antagonist is useful to treat a subject in need thereof.
  • the method can, for example, treat, prevent, cure, relieve, or ameliorate ("treat") a disease, disorder, condition, or illness ("a condition").
  • a condition a disease, disorder, condition, or illness
  • a condition a condition characterized by inappropriate expression or activity of PAR-2.
  • Specific medical conditions and diseases that are treatable or with a PAR2 antagonist include inflammatory conditions of the gastrointestinal system, including coeliac disease, Crohn's disease; ulcerative colitis; idiopathic gastroparesis; pancreatitis, including chronic pancreatitis; inflammatory bowel disease and ulcers, including gastric and duodenal ulcers; inflammatory conditions of the airway, such as asthma, chronic obstructive pulmonary disease, and the like; rheumatic disorders, including for example, adult and juvenile rheumatoid arthritis; scleroderma; systemic lupus erythematosus; gout; osteoarthritis; polymyalgia rheumatica; seronegative spondylarthropathies, including ankylosing spondylitis, and Reiter's disease, psoriatic arthritis and chronic Lyme arthritis; Still's disease and uveitis associated with rheumatoid arthritis; disorders resulting in inflammation of the
  • an anti-human PAR2 antibody or antibody fragment is useful to treat pain associated with a variety of conditions, include cancer pain, arthritis pain, diabetic neuropathy, post-herpetic neuralgia, migraine.
  • Combination therapeutics include cancer pain, arthritis pain, diabetic neuropathy, post-herpetic neuralgia, migraine.
  • a subject suffering from joint pain associated with gout is treated with a combination of an antagonist such as an anti-PAR2 antibody or antibody fragment, and a second therapeutic agent.
  • the second therapeutic agent is preferably an interleukin-1 (IL-1) antagonist such as rilonacept ("IL-1 trap"; Regeneron Pharmaceuticals, Inc.).
  • Suitable second therapeutic agents may be one or more agents selected from the group consisting of rilonacept, anakinra, a recombinant, nonglycosylated form of the human IL- 1 receptor antagonist (ILIRa), an anti-IL-1 antibody, or an anti-IL-18 drug such as IL- 18BP or a derivative, an IL-18 Trap, anti-IL-18, anti-IL-18R1 , or anti-IL-18Racp.
  • ILIRa human IL- 1 receptor antagonist
  • an anti-IL-18 drug such as IL- 18BP or a derivative, an IL-18 Trap, anti-IL-18, anti-IL-18R1 , or anti-IL-18Racp.
  • co-therapies which may be combined with an NGF antibody or antibody fragment and other appropriate therapeutic agents, including for example, low dose colchicine, aspirin, steroids such as prednisolone, methotrexate, low dose cyclosporine A, TNF inhibitors, other inflammatory inhibitors such as inhibitors of caspase-1 , p38, IKK1/2, CTLA-4lg, anti-IL-6 or anti-IL6Ra, etc., and/or co- therapies such as uric acid synthesis inhibitors to inhibit the accumulation of uric acid in the body, for example, allopurinol, uric acid excretion promoters to accelerate the rapid excretion of uric acid accumulated in the body, for example, probenecid, sulfinpyrazone and/or benzbromarone are examples of uric acid excretion promoters; corticosteroids; and other non-steroidal anti-inflammatory drugs (NSAIDs).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • a combination therapy method comprises administering to the subject two, three, four, five, six, or more of the PAR-2 agonists or antagonists described herein.
  • the individual molecule(s) and/or treatment(s) can be administered in any order, over any length of time, which is effective, e.g., simultaneously, consecutively, or alternately.
  • the method of treatment comprises completing a first course of treatment with one molecule or other treatment before beginning a second course of treatment.
  • the length of time between the end of the first course of treatment and beginning of the second course of treatment can be any length of time that allows the total course of therapy to be effective, e.g., seconds, minutes, hours, days, weeks or months.
  • PAR2 has been found to augment pain transmission and to mediate visceral pain in irritable bowel syndrome (IBS). PAR2 is expressed in several tissues including nociceptive primary afferent neurons. PAR2 expression in these sensory neurons colocalizes with calcitonin gene related peptide (CGRP) and substance P (SP), two known modulators of nociception. PAR2 agonists have been shown to stimulate release of SP and CGRP in tissues, induce a prolonged thermal and mechanical hyperalgesia, and elevate spinal FOS protein (an established marker of nociceptive neuron activation).
  • CGRP calcitonin gene related peptide
  • SP substance P
  • mice were tested at day 4 and day 7 post-surgery to confirm that the pain had developed. Then, at day 7 post- surgery following the behavioral pain testing the mice were injected s.c. with the anti- PAR2 antibody (50mg/kg; Molino et al. (1998) Arterioscler Thromb Vase Biol 18:825- 832 or IgG negative control antibody (100 mg/kg). The results are shown in Figs. 1 and 2.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Use of a protease-activated receptor 2 (PAR2) antagonist for treatment of pain, including post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, bone cancer pain or gout joint pain; inflammatory pain associated with irritable bowel syndrome; and inflammation related to rheumatoid arthritis or osteoarthritis. Preferably, the PAR2 antagonist is an anti-human PAR2 antibody or antigen-binding fragment of an antibody.

Description

Use of Protease-Activated Receptor 2 Antagonists
Field of the invention
[0001] The present invention is related to therapeutic methods of treating a human protease-activated receptor 2 (PAR2)-related inflammatory condition, including inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, and gout joint pain.
Statement of Related Art
[0002] Protease-activated receptor 2 (PAR2, also known as C140 receptor) is a member of a family of GPCRs that become activated by trypsin and the mast cell protease tryptase through proteolytic cleavage of the amino termini, exposing a tethered ligand that binds to the receptors and leads to activation. There are four family members, three of which, PAR1 , PAR3 and PAR4, are thrombin receptors. PAR4 is also activated by trypsin. Short synthetic peptides that resemble the tethered ligand sequence have been successfully used to activate all of the PAR receptors, except PAR3. [0003] The genes encoding the four GPCR family members are conserved across species and have a similar structure with a single intron interrupting the sequence encoding the receptor amino terminus. Mouse and human PAR2 are 83% homologous, but highly conserved within the trypsin cleavage site. PARs are widely expressed and have been implicated in important processes such as homeostasis, tissue repair, angiogenesis and inflammation.
[0004] Nucleic acid and amino acid sequences of human PAR2 (SEQ ID NO: 1-2) are described in, for example, US 5,629,174; 5,716,789; 6,297,026; and US Patent Publication No. 2003/0018184. Antibodies to PAR2 are described in, for example, US 5,874,400 and EP 0 804 452 B1. Methods of using a PAR2 antibody are described in US Patent Publication No. 2007/0237759.
BRIEF SUMMARY OF THE INVENTION
[0005] In a first aspect, the invention features methods for inhibiting PAR2 activity with a PAR2 antagonist capable of inhibiting PAR2 proteolytic cleavage and blocking PAR2 activation. In one embodiment, the method comprises administering a therapeutically effective amount of a PAR2 antagonist to a subject suffering from a PAR2-related condition or a condition which may be improved or ameliorated by inhibition of PAR2.
Such conditions or diseases include, for example, inflammation and/or pain. Inflammatory conditions include, for example, inflammation of the gastrointestinal system or of the airways, rheumatic disorders, osteoarthritis, pain associated with inflammation, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, and gout joint pain.
[0006] The PAR2 antagonist useful in the method of the invention is a molecule capable of blocking human PAR2 activity, including a molecule which blocks activation of PAR2 by blocking proteolytic cleavage of the N-terminus of the receptor, a molecule which blocks activation of the receptor after cleavage, or a molecule which inhibits PAR2 expression. The PAR2 antagonist may be an antibody, an antigen-binding fragment of an antibody, a peptibody, a soluble "decoy" receptor, or an antisense or siRNA molecule. In a preferred embodiment, the PAR2 antagonist is an antibody or an antigen-binding fragment of an antibody. The PAR2 antibody may be a fully human or partially human antibody or antigen-binding fragment of an antibody. In an even more preferred embodiment, the PAR2 antagonist is a fully human antibody or an antigen- binding fragment of an antibody.
[0007] The therapeutic method of the invention encompasses administration of a PAR2 antagonist as a single agent, or with a second therapeutic agent. The second therapeutic agent may be, for example, one or more of an interleukin-1 (IL-1) inhibitor, an antiepileptic drug, such as gabapentain.
[0008] In a preferred embodiment, the invention features a therapeutic method of treating neuropathic pain in a human subject with an anti-PAR2 antibody or antigen- binding fragment of an antibody. As described in the experimental section below, significant alleviation of both established thermal hyperalgia and mechanical allodynia in an animal model of neuropathic pain was achieved with an anti-PAR2 antagonist. [0009] In a second aspect, the invention features use of a protease-activated receptor 2 (PAR2) antagonist in the manufacture of a medicament for treating, inhibiting or ameliorating an inflammatory condition or disorder, wherein the inflammatory condition or disorder is rheumatoid arthritis or osteoarthritis. In a preferred embodiment, the PAR2 antagonist is an anti-human PAR2 antibody or antigen-binding fragment of an antibody; preferably the anti-PAR2 antibody or antibody fragment is fully human. [0010] In a third aspect, the invention features use of a protease-activated receptor 2 (PAR2) antagonist in the manufacture of a medicament for treating, inhibiting or ameliorating pain, wherein the pain is inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, bone cancer pain and gout joint pain. In a preferred embodiment, the inflammatory pain treated is pain associated with irritable bowel syndrome. In a preferred embodiment, the PAR2 antagonist is an anti-human PAR2 antibody or antigen-binding fragment of an antibody; preferably the anti-PAR2 antibody or antibody fragment is fully human.
[0011] In specific embodiments of the use of the PAR2 antagonist, the PAR2 antagonist is used as a first therapeutic agent, with one or more further therapeutic agents such as another PAR2 antagonist, a cytokine inhibitor, for example, an interleukin-1 (IL-1) inhibitor (for example, rilonacept or anakinra); an IL-18 inhibitor; an IL-6 inhibitor such as an anti-IL6 or anti-IL6 receptor antibody; an antiepileptic drug, such as gabapentain; an NGF inhibitor such as an anti-NGF antibody; low dose colchicine; aspirin; or other NSAIDs, steroids such as prednisolone, methotrexate; low dose cyclosporine A; a TNF inhibitor; and/or co- therapies such as uric acid synthesis inhibitors (allopurinol); uric acid excretion promoters (probenecid, sulfinpyrazone and/or benzbromarone); and/or corticosteroids.
BRIEF SUWtMARY OF THE FIGURES
[0012] Fig. 1. Results of Hargreaves' test. • = 100 mg/kg IgG control mAb (n=6); O= 50 mg/kg anti-PAR2 mAb (n=5); 2(treatment) X 11 (time) mixed Factorial ANOVA with Bonferroni post hoc tests; effect of treatment significant, p=0.0340 (* p<0.05; *** p<0.001 vs. IgG control).
[0013] Fig. 2. Results of von Frey test. # = 100 mg/kg IgG control mAb (n=6); O= 50 mg/kg anti-PAR2 mAb (n=6); 2(treatment) X 10(time) mixed Factorial ANOVA with Bonferroni post hoc tests; effect of treatment significant, p=0.0057.
DETAILED DESCRIPTION
[0014] Before the present methods are described, it is to be understood that this invention is not limited to particular methods, and experimental conditions described, as such methods and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
[0015] As used in this specification and the appended claims, the singular forms "a", "an", and "the" include plural references unless the context clearly dictates otherwise. Thus for example, a reference to "a method" includes one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure. [0016] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described.
Definitions
[0017] "Human protease-activated receptor 2" ("PAR2) refers to the protein encoded by the nucleic acid sequence of SEQ ID NO:1 and the protein having the amino acid sequence of SEQ ID NO:2. PAR2 is also called "C140 receptor". [0018] The term "PAR2 antagonist", as used herein, is intended to refer to a molecule which inhibits the activity or expression of PAR2.
[0019] The term "antibody", as used herein, is intended to refer to immunoglobulin molecules comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementary determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
[0020] A "neutralizing" or "blocking" antibody, is intended to refer to an antibody whose binding to PAR2 results in inhibition of the biological activity of PAR2. This inhibition of the biological activity of PAR2 can be assessed by measuring one or more indicators of PAR2 biological activity. These indicators of PAR2 biological activity can be assessed by one or more of several standard in vitro or in vivo assays known in the art (see examples below).
[0021] The term "antigen-binding portion" of an antibody (or simply "antibody portion" or "antibody fragment"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., PAR2). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full- length antibody. Examples of binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH1 CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al. (1989) Nature 241 :544-546), which consists of a VH domain; and (vi) an isolated CDR. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger et al. (1993) Proc. Natl. Acad Sci. USA 90:6444-6448; Poljak et al. (1994) Structure 2:1121- 1123).
[0022] The term "human antibody", as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
[0023] The term "effective amount" is a concentration or amount of a PAR2 antagonist which results in achieving a particular stated purpose. An "effective amount" of an anti- PAR2 antibody or antigen-binding fragment of an antibody thereof, for example, may be determined empirically. Furthermore, a "therapeutically effective amount" is a concentration or amount of a PAR2 antagonist, such as an anti-PAR2 antibody or antigen-binding fragment, which is effective for achieving a stated therapeutic effect This amount may also be determined empirically.
Pharmaceutical Compositions and Therapeutic Uses
[0024] The present invention provides a pharmaceutical composition comprising an antagonist of human PAR2, as described above, and a pharmaceutically acceptable carrier. The pharmaceutical composition comprising a PAR2 antagonist of the present invention as an active ingredient can be used as an agent for preventing and/or treating a disorder or condition that is associated with PAR2, such as, for example, inflammatory pain.
[002S] The pharmaceutical compositions of the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences (15th ed, Mack Publishing Company, Easton, PA). These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles, DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. Any of the foregoing mixtures may be appropriate in treatments and therapies in accordance with the present invention, provided that the active ingredient in the formulation is not inactivated by the formulation and the formulation is physiologically compatible and tolerable with the route of administration. See also Powell et al. "Compendium of excipients for parenteral formulations" PDA, 1998, J Pharm Sci Technol. 52:238-311 , and the citations therein for additional information related to excipients and carriers well known to pharmaceutical chemists.
[0026] The dose may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like. When the PAR2 antagonist is used for preventing and/or treating, for example, inflammatory pain, in an adult patient, it is advantageous to intravenously administer the antagonist normally at a single dose of about 0.01 to about 20 mg/kg body weight, preferably about 0.1 to about 10 mg/kg body weight, and more preferably about 0.1 to about 5 mg/kg body weight, approximately 1 to 5 times per day, preferably approximately 1 to 3 times per day. In other parenteral administration and oral administration, the antagonist can be administered in a dose corresponding to the dose given above. When the condition is especially severe, the dose may be increased according to the condition. [0027] Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:44294432). Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. [0028] In another embodiment, the pharmaceutical composition can be delivered in a vesicle, in particular a liposome (see, for example, Langer, 1990, Science 249:1527- 1533). In yet another embodiment, the pharmaceutical composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton (1987) CRC Grit. Ref. Biomed. Eng. 14:201). In another embodiment, polymeric materials can be used (see, for example, Medical Applications of Controlled Release (1974) Langer and Wise (eds.), CRC Pres., Boca Raton, Florida; Controlled Drug Bioavailability, Drug Product Design and Performance, (1984) Smolen and Ball (eds.), Wiley, New York). In yet another embodiment, a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release (1984) supra, vol. 2, pp. 115-138). Other controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533).
[0029] Examples of the composition for oral administration include solid or liquid dosage forms, specifically, tablets (including dragees and film-coated tablets), pills, granules, powdery preparations, capsules (including soft capsules), syrup, emulsions, suspensions, etc. Such a composition is manufactured by publicly known methods and contains a vehicle, a diluent or an excipient conventionally used in the field of pharmaceutical preparations. Examples of the vehicle or excipient for tablets are lactose, starch, sucrose, magnesium stearate, and the like. [0030] The injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. The injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antagonist or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections. As the aqueous medium for injections, there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared is preferably filled in an appropriate ampoule. The suppository used for rectal administration may be prepared by blending the aforesaid antagonist molecule or its salt with conventional bases for suppositories.
[0031] Advantageously, the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients. Such dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc. The amount of, for example, an anti-PAR2 antibody or antibody fragment, contained is generally about 5 to 500 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the aforesaid antibody is contained in about 5 to 100 mg and in about 10 to 250 mg for the other dosage forms.
[0032] As described above, the present invention further provides a method for preventing or treating a disorder or condition that is associated with or involves PAR2, the method comprising administering an effective amount of at least one human PAR2 antagonists to a subject in need thereof.
[0033] Therapeutic Indications. The invention provides therapeutic methods in which a PAR2 antagonist is useful to treat a subject in need thereof. The method can, for example, treat, prevent, cure, relieve, or ameliorate ("treat") a disease, disorder, condition, or illness ("a condition"). Among the conditions to be treated in accordance with the present invention are conditions characterized by inappropriate expression or activity of PAR-2. Specific medical conditions and diseases that are treatable or with a PAR2 antagonist include inflammatory conditions of the gastrointestinal system, including coeliac disease, Crohn's disease; ulcerative colitis; idiopathic gastroparesis; pancreatitis, including chronic pancreatitis; inflammatory bowel disease and ulcers, including gastric and duodenal ulcers; inflammatory conditions of the airway, such as asthma, chronic obstructive pulmonary disease, and the like; rheumatic disorders, including for example, adult and juvenile rheumatoid arthritis; scleroderma; systemic lupus erythematosus; gout; osteoarthritis; polymyalgia rheumatica; seronegative spondylarthropathies, including ankylosing spondylitis, and Reiter's disease, psoriatic arthritis and chronic Lyme arthritis; Still's disease and uveitis associated with rheumatoid arthritis; disorders resulting in inflammation of the voluntary muscle and other muscles, including dermatomyositis, inclusion body myositis, polymyositis, and lymphangioleimyomatosis. Other indications include, for example, breast cancer (Adriaenssens et al. (2008) Cancer Res 68:346-51).
[0034] In a more specific therapeutic use, an anti-human PAR2 antibody or antibody fragment is useful to treat pain associated with a variety of conditions, include cancer pain, arthritis pain, diabetic neuropathy, post-herpetic neuralgia, migraine. [0035] Combination therapeutics. In specific embodiments of the therapeutic methods of the invention, a subject suffering from joint pain associated with gout is treated with a combination of an antagonist such as an anti-PAR2 antibody or antibody fragment, and a second therapeutic agent. The second therapeutic agent is preferably an interleukin-1 (IL-1) antagonist such as rilonacept ("IL-1 trap"; Regeneron Pharmaceuticals, Inc.). Suitable second therapeutic agents may be one or more agents selected from the group consisting of rilonacept, anakinra, a recombinant, nonglycosylated form of the human IL- 1 receptor antagonist (ILIRa), an anti-IL-1 antibody, or an anti-IL-18 drug such as IL- 18BP or a derivative, an IL-18 Trap, anti-IL-18, anti-IL-18R1 , or anti-IL-18Racp. Other co-therapies which may be combined with an NGF antibody or antibody fragment and other appropriate therapeutic agents, including for example, low dose colchicine, aspirin, steroids such as prednisolone, methotrexate, low dose cyclosporine A, TNF inhibitors, other inflammatory inhibitors such as inhibitors of caspase-1 , p38, IKK1/2, CTLA-4lg, anti-IL-6 or anti-IL6Ra, etc., and/or co- therapies such as uric acid synthesis inhibitors to inhibit the accumulation of uric acid in the body, for example, allopurinol, uric acid excretion promoters to accelerate the rapid excretion of uric acid accumulated in the body, for example, probenecid, sulfinpyrazone and/or benzbromarone are examples of uric acid excretion promoters; corticosteroids; and other non-steroidal anti-inflammatory drugs (NSAIDs).
[0036] In various embodiments, a combination therapy method comprises administering to the subject two, three, four, five, six, or more of the PAR-2 agonists or antagonists described herein. When a of molecules and/or other treatments is used, the individual molecule(s) and/or treatment(s) can be administered in any order, over any length of time, which is effective, e.g., simultaneously, consecutively, or alternately. In one embodiment, the method of treatment comprises completing a first course of treatment with one molecule or other treatment before beginning a second course of treatment. The length of time between the end of the first course of treatment and beginning of the second course of treatment can be any length of time that allows the total course of therapy to be effective, e.g., seconds, minutes, hours, days, weeks or months.
EXAMPLES
Example 1. Therapeutic Treatment of Neuropathic Pain. [0037] PAR2 has been found to augment pain transmission and to mediate visceral pain in irritable bowel syndrome (IBS). PAR2 is expressed in several tissues including nociceptive primary afferent neurons. PAR2 expression in these sensory neurons colocalizes with calcitonin gene related peptide (CGRP) and substance P (SP), two known modulators of nociception. PAR2 agonists have been shown to stimulate release of SP and CGRP in tissues, induce a prolonged thermal and mechanical hyperalgesia, and elevate spinal FOS protein (an established marker of nociceptive neuron activation).
[0038] Experiments were conducted to test the effect of PAR-2 inhibition on neuropathic pain. The Seltzer model of neuropathic pain was used with an anti-PAR-2 antibody in C57BL/6 male mice. This neuropathic pain model produces a partial nerve injury by tying a tight ligature with a 7-0 silk suture around approximately 1/3 to 1/2 the diameter of the sciatic nerve of one single thigh per mouse. Post-surgery, the mice are allowed to recover for at least two days and then they are studied for several weeks post-surgery for thermal hyperalgesia in the Hargreaves' test and for mechanical allodynia in the von Frey test. Following the Seltzer surgery, mice were tested at day 4 and day 7 post-surgery to confirm that the pain had developed. Then, at day 7 post- surgery following the behavioral pain testing the mice were injected s.c. with the anti- PAR2 antibody (50mg/kg; Molino et al. (1998) Arterioscler Thromb Vase Biol 18:825- 832 or IgG negative control antibody (100 mg/kg). The results are shown in Figs. 1 and 2.

Claims

Claims:
1. Use of a protease-activated receptor 2 (PAR2) antagonist in the manufacture of a medicament for treating, inhibiting or ameliorating pain, wherein the pain is inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, bone cancer pain or gout joint pain.
2. Use according to claim 1 , wherein the inflammatory pain is pain associated with irritable bowel syndrome.
3. Use of a protease-activated receptor 2 (PAR2) antagonist in the manufacture of a medicament for treating, inhibiting or ameliorating inflammation related to rheumatoid arthritis or osteoarthritis.
4. Use according to any one of claim 1 to 3, wherein the PAR2 antagonist is an anti- human PAR2 antibody or antigen-binding fragment of an antibody.
5. Use according to claim 4, wherein the anti-PAR2 antibody or antibody fragment is a fully human antibody or antibody fragment.
6. Use of a protease-activated receptor 2 (PAR2) antagonist as defined in claim 4 or 5, and one or more further therapeutic agents selected from another PAR2 antagonist, a cytokine inhibitor, an antiepileptic drug, an NGF inhibitor, colchicine, a non-steroidal anti-inflammatory drug (NSAID), a corticosteroid and a TNF inhibitor, in the manufacture of a medicament for treating, inhibiting or ameliorating pain associated with inflammation, post-operative incision, neuropathy, bone fracture, osteroporotic fracture, bone cancer or gout.
7. Use of a protease-activated receptor 2 (PAR2) antagonist as defined in claim 4 or 5, in the manufacture of a medicament for treating, inhibiting or ameliorating pain associated with inflammation, post-operative incision, neuropathy, bone fracture, osteroporotic fracture, bone cancer or gout, in combination with one or more further therapeutic agents selected from another PAR2 antagonist, a cytokine inhibitor, an antiepileptic drug, an NGF inhibitor, colchicine, a non-steroidal anti-inflammatory drug (NSAID), a corticosteroid and a TNF inhibitor.
8. Use of a therapeutic agent selected from a PAR2 antagonist other than one as defined in claim 4 or 5, a cytokine inhibitor, an antiepileptic drug, an NGF inhibitor, colchicine, a non-steroidal anti-inflammatory drug (NSAID), a corticosteroid and a TNF inhibitor, in the manufacture of a medicament for treating, inhibiting or ameliorating pain associated with inflammation, post-operative incision, neuropathy, bone fracture, osteroporotic fracture, bone cancer or gout, in combination with a protease-activated receptor 2 (PAR2) antagonist as defined in claim 4 or 5.
9. A protease-activated receptor 2 (PAR2) antagonist for use in treating, inhibiting or ameliorating pain, wherein the pain is inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, bone cancer pain or gout joint pain.
10. A PAR2 antagonist for use in treating, inhibiting or ameliorating pain according to claim 9, wherein the inflammatory pain is pain associated with irritable bowel syndrome.
11. A protease-activated receptor 2 (PAR2) antagonist for use in treating, inhibiting or ameliorating inflammation related to rheumatoid arthritis or osteoarthritis.
12. A PAR2 antagonist according to any one of claims 9 to 11 , wherein the PAR2 antagonist is an anti-human PAR2 antibody or antigen-binding fragment of an antibody.
13. A PAR2 antagonist according to claim 12, wherein the anti-PAR2 antibody or antibody fragment is a fully human antibody or antibody fragment.
14. A protease-activated receptor 2 (PAR2) antagonist as defined in claim 4 or 5 for use in treating, inhibiting or ameliorating pain associated with inflammation, postoperative incision, neuropathy, bone fracture, osteroporotic fracture, bone cancer or gout, in combination with one or more further therapeutic agents selected from another PAR2 antagonist, a cytokine inhibitor, an antiepileptic drug, an NGF inhibitor, colchicine, a non-steroidal anti-inflammatory drug (NSAID), a corticosteroid and a TNF inhibitor.
15. A method of treating, inhibiting or ameliorating pain, wherein the pain is inflammatory pain, post-operative incision pain, neuropathic pain, fracture pain, osteroporotic fracture pain, bone cancer pain or gout joint pain comprising administering an effective amount of a protease-activated receptor 2 (PAR2) antagonist.
16. A method according to claim 15 wherein the inflammatory pain is pain associated with irritable bowel syndrome.
17. A method of treating, inhibiting or ameliorating inflammation related to rheumatoid arthritis or osteoarthritis comprising administering an effective amount of a protease- activated receptor 2 (PAR2) antagonist.
18. A method according to claim 15 or 17, wherein the PAR2 antagonist is an anti- human PAR2 antibody or antigen-binding fragment of an antibody.
19. A method according to claim 18, wherein the anti-PAR2 antibody or antibody fragment is a fully human antibody or antibody fragment.
20. A method of treating, inhibiting or ameliorating pain associated with inflammation, post-operative incision, neuropathy, bone fracture, osteroporotic fracture, bone cancer or gout comprising administering an effective amount of a protease-activated receptor 2 (PAR2) antagonist as defined in claim 4 or 5, in combination with one or more further therapeutic agents selected from another PAR2 antagonist, a cytokine inhibitor, an antiepileptic drug, an NGF inhibitor, colchicine, a non-steroidal anti-inflammatory drug (NSAID), a corticosteroid and a TNF inhibitor.
PCT/US2009/037504 2008-03-19 2009-03-18 Use of protease-activated receptor 2 antagonists WO2009117481A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US3790808P 2008-03-19 2008-03-19
US61/037,908 2008-03-19
US11915508P 2008-12-02 2008-12-02
US61/119,155 2008-12-02

Publications (1)

Publication Number Publication Date
WO2009117481A1 true WO2009117481A1 (en) 2009-09-24

Family

ID=40718925

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/037504 WO2009117481A1 (en) 2008-03-19 2009-03-18 Use of protease-activated receptor 2 antagonists

Country Status (6)

Country Link
AR (1) AR070911A1 (en)
CL (1) CL2009000660A1 (en)
PE (1) PE20091693A1 (en)
TW (1) TW201002345A (en)
UY (1) UY31723A (en)
WO (1) WO2009117481A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1578769A2 (en) * 2001-05-21 2005-09-28 Johnson &amp; Johnson Consumer Companies, Inc. Peptides and the use thereof in darkening the skin
US7888482B2 (en) 2006-02-10 2011-02-15 Amgen Inc. Antibodies that bind PAR-2
US8101724B2 (en) 2009-09-09 2012-01-24 Regeneron Pharmaceuticals, Inc. High-affinity human antibodies to human protease-activated receptor-2
WO2013064583A1 (en) 2011-11-04 2013-05-10 Lipotec, S.A. Peptides which inhibit activated receptors and their use in cosmetic or pharmaceutical compositions
WO2017197463A1 (en) * 2016-05-20 2017-11-23 Monash University Treatment of pain
WO2017223427A1 (en) * 2016-06-23 2017-12-28 Mayo Foundation For Medical Education And Research Par2 modulation to alter myelination
US10030024B2 (en) 2013-09-25 2018-07-24 Vertex Pharmaceuticals Incorporated Imidazopyridazines useful as inhibitors of the PAR-2 signaling pathway
CN110446720A (en) * 2017-03-16 2019-11-12 免疫医疗有限公司 Anti- PAR2 antibody and application thereof
US11813264B2 (en) 2014-07-07 2023-11-14 Mayo Foundation For Medical Education And Research PAR1 modulation to alter myelination

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995003318A1 (en) * 1993-07-26 1995-02-02 Cor Therapeutics Recombinant c140 receptor and its agonists and antagonists
WO1996023225A1 (en) * 1995-01-25 1996-08-01 Cor Therapeutics, Inc. Recombinant c140 receptor, its agonists and antagonists, and nucleic acids encoding the receptor
WO2005123193A2 (en) * 2004-06-17 2005-12-29 Osteologix A/S Treatments comprising strontium for rheumatic and arthritic diseases and pain

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995003318A1 (en) * 1993-07-26 1995-02-02 Cor Therapeutics Recombinant c140 receptor and its agonists and antagonists
WO1996023225A1 (en) * 1995-01-25 1996-08-01 Cor Therapeutics, Inc. Recombinant c140 receptor, its agonists and antagonists, and nucleic acids encoding the receptor
WO2005123193A2 (en) * 2004-06-17 2005-12-29 Osteologix A/S Treatments comprising strontium for rheumatic and arthritic diseases and pain

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BUENO L: "Protease activated receptor 2: a new target for IBS treatment.", EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES AUG 2008, vol. 12 Suppl 1, August 2008 (2008-08-01), pages 95 - 102, XP008107526, ISSN: 1128-3602 *
BUSSO NATHALIE ET AL: "Evaluation of protease-activated receptor 2 in murine models of arthritis.", ARTHRITIS AND RHEUMATISM JAN 2007, vol. 56, no. 1, January 2007 (2007-01-01), pages 101 - 107, XP002533055, ISSN: 0004-3591 *
CENAC NICOLAS ET AL: "Role for protease activity in visceral pain in irritable bowel syndrome.", THE JOURNAL OF CLINICAL INVESTIGATION MAR 2007, vol. 117, no. 3, March 2007 (2007-03-01), pages 636 - 647, XP002534418, ISSN: 0021-9738 *
DAI YI ET AL: "Sensitization of TRPA1 by PAR2 contributes to the sensation of inflammatory pain.", THE JOURNAL OF CLINICAL INVESTIGATION JUL 2007, vol. 117, no. 7, July 2007 (2007-07-01), pages 1979 - 1987, XP002533056, ISSN: 0021-9738 *
KELSO ELIZABETH B ET AL: "Therapeutic promise of proteinase-activated receptor-2 antagonism in joint inflammation", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, AMERICAN SOCIETY FOR PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, US, vol. 316, no. 3, 31 October 2005 (2005-10-31), pages 1017 - 1024, XP002457848, ISSN: 0022-3565 *
NICKEL TIMOTHY J ET AL: "Constitutive expression of functionally active protease-activated receptors 1 and 2 in human conjunctival epithelial cells.", MEDIATORS OF INFLAMMATION 2006, vol. 2006, no. 3, 2006, pages 61359, XP002533058, ISSN: 0962-9351 *
SCHMIDLIN FABIEN ET AL: "Protease-activated receptor 2 mediates eosinophil infiltration and hyperreactivity in allergic inflammation of the airway.", JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 1 NOV 2002, vol. 169, no. 9, 1 November 2002 (2002-11-01), pages 5315 - 5321, XP002533057, ISSN: 0022-1767 *
SOREIDE KJETIL: "Proteinase-activated receptor 2 (PAR-2) in gastrointestinal and pancreatic pathophysiology, inflammation and neoplasia", SCANDINAVIAN JOURNAL OF GASTROENTEROLOGY,, vol. 43, no. 8, 1 August 2008 (2008-08-01), pages 902 - 909, XP008107531, ISSN: 0036-5521 *
WANG JUNRU ET AL: "Up-regulation and activation of proteinase-activated receptor 2 in early anddelayed radiation injury in the rat intestine: influence of biological activators of proteinase-activated receptor 2", RADIATION RESEARCH, ACADEMIC PRESS INC, US, vol. 160, no. 5, 1 November 2003 (2003-11-01), pages 524 - 535, XP009091829, ISSN: 0033-7587 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1578769A4 (en) * 2001-05-21 2010-09-29 Johnson & Johnson Consumer Peptides and the use thereof in darkening the skin
EP1578769A2 (en) * 2001-05-21 2005-09-28 Johnson &amp; Johnson Consumer Companies, Inc. Peptides and the use thereof in darkening the skin
US7888482B2 (en) 2006-02-10 2011-02-15 Amgen Inc. Antibodies that bind PAR-2
US8287872B2 (en) 2006-02-10 2012-10-16 Amgen Inc. Methods for treating inflammatory conditions using antibodies that bind PAR-2
US9028819B2 (en) 2009-09-09 2015-05-12 Regeneron Pharmaceuticals, Inc. Methods of alleviating itch by administering human antibodies to human protease-activated receptor 2
US8101724B2 (en) 2009-09-09 2012-01-24 Regeneron Pharmaceuticals, Inc. High-affinity human antibodies to human protease-activated receptor-2
US8425907B2 (en) 2009-09-09 2013-04-23 Regeneron Pharmaceuticals, Inc. Methods for treating pruritus by administering an antibody that specifically binds human PAR2
US9333152B2 (en) 2011-11-04 2016-05-10 Lipotec, S.A. Peptides which inhibit activated receptors and their use in cosmetic or pharmaceutical compositions
WO2013064583A1 (en) 2011-11-04 2013-05-10 Lipotec, S.A. Peptides which inhibit activated receptors and their use in cosmetic or pharmaceutical compositions
US10030024B2 (en) 2013-09-25 2018-07-24 Vertex Pharmaceuticals Incorporated Imidazopyridazines useful as inhibitors of the PAR-2 signaling pathway
US11813264B2 (en) 2014-07-07 2023-11-14 Mayo Foundation For Medical Education And Research PAR1 modulation to alter myelination
JP2022061998A (en) * 2016-05-20 2022-04-19 武田薬品工業株式会社 Pain treatment
WO2017197463A1 (en) * 2016-05-20 2017-11-23 Monash University Treatment of pain
CN109152777A (en) * 2016-05-20 2019-01-04 武田药品工业株式会社 The treatment of pain
JP2019516734A (en) * 2016-05-20 2019-06-20 武田薬品工業株式会社 Pain treatment
JP7441246B2 (en) 2016-05-20 2024-02-29 武田薬品工業株式会社 pain treatment
WO2017223427A1 (en) * 2016-06-23 2017-12-28 Mayo Foundation For Medical Education And Research Par2 modulation to alter myelination
JP7222901B2 (en) 2017-03-16 2023-02-15 メドイミューン・リミテッド Anti-PAR2 antibody and use thereof
US11591389B2 (en) 2017-03-16 2023-02-28 Medimmune Limited Nucleic acids encoding anti-PAR2 antibodies and uses thereof
CN110446720B (en) * 2017-03-16 2023-09-12 免疫医疗有限公司 anti-PAR 2 antibodies and uses thereof
JP2020509762A (en) * 2017-03-16 2020-04-02 メドイミューン・リミテッドMedImmune Limited Anti-PAR2 antibody and use thereof
CN110446720A (en) * 2017-03-16 2019-11-12 免疫医疗有限公司 Anti- PAR2 antibody and application thereof

Also Published As

Publication number Publication date
CL2009000660A1 (en) 2010-04-16
AR070911A1 (en) 2010-05-12
TW201002345A (en) 2010-01-16
PE20091693A1 (en) 2009-11-16
UY31723A (en) 2009-11-10

Similar Documents

Publication Publication Date Title
WO2009117481A1 (en) Use of protease-activated receptor 2 antagonists
KR102580468B1 (en) Methods for treating allergy and enhancing allergen-specific immunotherapy by administering an il-4r inhibitor
KR20190042702A (en) Methods for preventing or treating allergy by administering an IL-4R antagonist
KR20160029750A (en) Methods for treating eosinophilic esophagitis by administering an il-4r inhibitor
KR20150048887A (en) Methods for treating atopic dermatitis by administering an il-4r antagonist
KR20210010518A (en) How to treat atopic dermatitis by administering an IL-4R inhibitor
JP6554155B2 (en) Methods of treating generalized pustular psoriasis (GPP) using an IL-17 antagonist
RU2759630C2 (en) Methods for treating severe atopic dermatitis by administering an il-4r inhibitor
KR20220042217A (en) A method for treating allergy and enhancing allergen-specific immunotherapy by administering an IL-4R antagonist
KR20170045240A (en) Use of il-17 antagonists to inhibit the progression of structural damage in psoriatic arthritis patients
KR20220158821A (en) Methods for Treating Atopic Dermatitis by Administering an IL-4R Antagonist
AU2022200690B2 (en) Method of treating tendinopathy using interleukin-17 (IL-17) antagonists
KR20190090820A (en) Pharmaceutical composition to prevent opioid poisoning
US20230203149A1 (en) Treatment of atopic dermatitis
KR20210104744A (en) Compositions and methods for improving body weight and lean muscle mass using antagonists for the leptin receptor, GDF8 and activin A
KR102284244B1 (en) Methods for treating atopic dermatitis by administering an il-4r antagonist
RU2801204C2 (en) Method of treatment of atopic dermatitis through introduction of il-4r inhibitor
RU2777328C2 (en) Methods for preventing or treating allergies by introducing an il-4r antagonist
KR20230079268A (en) Anti-IL-36R Antibodies for Treatment of Chronic Inflammatory Pain
RU2789047C2 (en) Methods for the treatment of allergy and increasing the efficiency of allergen-specific immunotherapy by introducing il-4r inhibitor
JP2023504679A (en) Methods of treating lichen planus using interleukin-17 (IL-17) antagonists

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09722546

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 09722546

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE