US20050119160A1 - Methods and reagents for the treatment of immunoinflammatory disorders - Google Patents

Methods and reagents for the treatment of immunoinflammatory disorders Download PDF

Info

Publication number
US20050119160A1
US20050119160A1 US10/966,228 US96622804A US2005119160A1 US 20050119160 A1 US20050119160 A1 US 20050119160A1 US 96622804 A US96622804 A US 96622804A US 2005119160 A1 US2005119160 A1 US 2005119160A1
Authority
US
United States
Prior art keywords
tetra
composition
peg
substituted pyrimidopyrimidine
drug
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/966,228
Other languages
English (en)
Inventor
Curtis Keith
Alexis Borisy
Grant Zimmermann
Edward Jost-Price
Palaniyandi Manivasakam
Nicole Hurst
Michael Foley
Michael Slavonic
Brendan Smith
Benjamin Auspitz
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.)
COMINATORX Inc
Zalicus Inc
Original Assignee
COMINATORX 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 COMINATORX Inc filed Critical COMINATORX Inc
Priority to US10/966,228 priority Critical patent/US20050119160A1/en
Assigned to COMINATORX, INCORPORATED reassignment COMINATORX, INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BORISY, ALEXIS, FOLEY, MICHAEL A., KEITH, CURTIS, SLAVONIC, MICHAEL S., HURST, NICOLE, ZIMMERMANN, GRANT R., AUSPITZ, BENJAMIN A., JOST-PRICE, EDWARD ROYDON, MANIVASAKAM, PALANIYANDI, SMITH, BRENDAN
Publication of US20050119160A1 publication Critical patent/US20050119160A1/en
Priority to US11/517,593 priority patent/US8080553B2/en
Assigned to ZALICUS INC. reassignment ZALICUS INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: COMBINATORX, INCORPORATED
Priority to US13/292,669 priority patent/US20120058979A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • A61K9/209Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat containing drug in at least two layers or in the core and in at least one outer layer

Definitions

  • the invention relates to the treatment of immunoinflammatory disorders.
  • Immunoinflammatory conditions are characterized by the inappropriate activation of the body's immune defenses. Rather than targeting infectious invaders, the immune response targets and damages the body's own tissues or transplanted tissues.
  • the tissue targeted by the immune system varies with the disorder. For example, in multiple sclerosis, the immune response is directed against the neuronal tissue, while in Crohn's disease the digestive tract is targeted.
  • Immunoinflammatory disorders affect millions of individuals and include conditions such as asthma, allergic intraocular inflammatory diseases, arthritis, atopic dermatitis, atopic eczema, diabetes, hemolytic anaemia, inflammatory dermatoses, inflammatory bowel or gastrointestinal disorders (e.g., Crohn's disease and ulcerative colitis), multiple sclerosis, myasthenia gravis, pruritis/inflammation, psoriasis, rheumatoid arthritis, cirrhosis, and systemic lupus erythematosus.
  • conditions such as asthma, allergic intraocular inflammatory diseases, arthritis, atopic dermatitis, atopic eczema, diabetes, hemolytic anaemia, inflammatory dermatoses, inflammatory bowel or gastrointestinal disorders (e.g., Crohn's disease and ulcerative colitis), multiple sclerosis, myasthenia gravis, pruritis/inflammation, psoriasis, rheuma
  • the invention features a method for treating an immunoinflammatory disease by administering to a patient in need thereof certain tetra-substituted pyrimidopyrimidines, either alone or in combination with any of a number of companion compounds, including an antihistamine, a corticosteroid, rolipram, ibudilast, a tricyclic or tetracyclic antidepressant, an SSRI, a non-steroidal anti-inflammatory drug, a non-steroidal immunophilin-dependent immunosuppressant, and an analog of any thereof, as described herein.
  • a number of companion compounds including an antihistamine, a corticosteroid, rolipram, ibudilast, a tricyclic or tetracyclic antidepressant, an SSRI, a non-steroidal anti-inflammatory drug, a non-steroidal immunophilin-dependent immunosuppressant, and an analog of any thereof, as described herein.
  • the invention features a method of treating a patient having an immunoinflammatory disease by administering to the patient a tetra-substituted pyrimidopyrimidine in an amount and for duration to treat the patient.
  • the invention features a method for treating a patient having an immunoinflammatory disorder by administering to the patient tetra-substituted pyrimidopyrimidine and an antihistamine simultaneously or within 14 days of each other in amounts sufficient to treat the patient.
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to the patient a tetra-substituted pyrimidopyrimidine and an antihistamine simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient.
  • the invention features a composition that includes a tetra-substituted pyrimidopyrimidine and an antihistamine.
  • a particularly desirable tetra-substituted pyrimidopyrimidine is dipyridamole.
  • the composition may be formulated for topical or systemic administration.
  • the invention features a kit that includes: (i) a composition that includes a tetra-substituted pyrimidopyrimidine and an antihistamine; and (ii) instructions for administering the composition to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a kit that includes: (i) an antihistamine; (ii) a tetra-substituted pyrimidopyrimidine; and (iii) instructions for administering the tetra-substituted pyrimidopyrimidine and the antihistamine to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a composition that includes a tetra-substituted pyrimidopyrimidine and a corticosteroid.
  • Particularly desirable corticosteroids are prednisolone, cortisone, dexamethasone, hydrocortisone, methylprednisolone, fluticasone, prednisone, triamcinolone, and diflorasone.
  • the composition may be formulated for topical or systemic administration (e.g., oral administration).
  • One or both of the drugs may be present in the composition in a low dosage or a high dosage, each of which is defined herein.
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to the patient a tetra-substituted pyrimidopyrimidine and a corticosteroid simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient.
  • the invention features a method for treating a patient diagnosed with or at risk of developing an immunoinflammatory disorder by administering to the patient a tetra-substituted pyrimidopyrimidine and a corticosteroid simultaneously or within 14 days of each other in amounts sufficient to treat the patient.
  • the invention features a kit that includes: (i) a composition that includes a tetra-substituted pyrimidopyrimidine and a corticosteroid; and (ii) instructions for administering the composition to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a kit that includes: (i) a tetra-substituted pyrimidopyrimidine; (ii) a corticosteroid; and (iii) instructions for administering the tetra-substituted pyrimidopyrimidine and the corticosteroid to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a composition that includes tetra-substituted pyrimidopyrimidine and ibudilast.
  • the composition may be formulated for topical or systemic administration.
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to the patient tetra-substituted pyrimidopyrimidine and ibudilast simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient.
  • the invention features a method for treating a patient diagnosed with or at risk of developing an immunoinflammatory disorder by administering to the patient tetra-substituted pyrimidopyrimidine and ibudilast simultaneously or within 14 days of each other in amounts sufficient to treat the patient.
  • the invention features a kit that includes: (i) a composition that includes tetra-substituted pyrimidopyrimidine and ibudilast; and (ii) instructions for administering the composition to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a kit that includes: (i) tetra-substituted pyrimidopyrimidine; (ii) ibudilast; and (iii) instructions for administering the tetra-substituted pyrimidopyrimidine and the ibudilast to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a composition that includes tetra-substituted pyrimidopyrimidine and rolipram.
  • the composition may be formulated for topical or systemic administration.
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to the patient a tetra-substituted pyrimidopyrimidine and rolipram simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient.
  • the invention features a method for treating a patient diagnosed with or at risk of developing an immunoinflammatory disorder by administering to the patient tetra-substituted pyrimidopyrimidine and rolipram simultaneously or within 14 days of each other in amounts sufficient to treat the patient.
  • the invention features a kit that includes: (i) a composition that includes tetra-substituted pyrimidopyrimidine and rolipram; and (ii) instructions for administering the composition to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a kit that includes: (i) tetra-substituted pyrimidopyrimidine; (ii) rolipram; and (iii) instructions for administering the tetra-substituted pyrimidopyrimidine and the rolipram to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a composition that includes a tetra-substituted pyrimidopyrimidine and a tricyclic or tetracyclic antidepressant.
  • a tricyclic or tetracyclic antidepressant are nortryptiline, amoxapine, and desipramine.
  • the composition may be formulated for topical or systemic administration.
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to the patient a tetra-substituted pyrimidopyrimidine and a tricyclic or tetracyclic antidepressant simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient.
  • the invention features a method for treating a patient diagnosed with or at risk of developing an immunoinflammatory disorder by administering to the patient a tetra-substituted pyrimidopyrimidine and a tricyclic or tetracyclic antidepressant simultaneously or within 14 days of each other in amounts sufficient to treat the patient.
  • the invention features a kit that includes: (i) a composition that includes a tetra-substituted pyrimidopyrimidine and a tricyclic or tetracyclic antidepressant; and (ii) instructions for administering the composition to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a kit that includes: (i) a tetra-substituted pyrimidopyrimidine; (ii) a tricyclic or tetracyclic antidepressant; and (iii) instructions for administering the tetra-substituted pyrimidopyrimidine and the tricyclic or tetracyclic antidepressant to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a composition that includes a tetra-substituted pyrimidopyrimidine and a selective serotonin reuptake inhibitor (SSRI).
  • SSRI selective serotonin reuptake inhibitor
  • Particularly desirable SSRIs are paroxetine, fluoxetine, sertraline, and citalopram.
  • the composition may be formulated for topical or systemic administration (e.g., oral administration).
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to the patient a tetra-substituted pyrimidopyrimidine and an SSRI simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient.
  • the invention features a method for treating a patient diagnosed with or at risk of developing an immunoinflammatory disorder by administering to the patient an a tetra-substituted pyrimidopyrimidine and an SSRI simultaneously or within 14 days of each other in amounts sufficient to treat the patient.
  • the invention features a kit that includes: (i) a composition that includes a tetra-substituted pyrimidopyrimidine and an SSRI; and (ii) instructions for administering the composition to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the invention features a kit that includes: (i) a tetra-substituted pyrimidopyrimidine; (ii) an SSRI; and (iii) instructions for administering the antihistamine and the SSRI to a patient diagnosed with or at risk of developing an immunoinflammatory disorder.
  • the tetra-substituted pyrimidopyrimidine and the companion compound are administered within 10 days of each other, within five days of each other, within twenty-four hours of each other, or even simultaneously.
  • the compounds may be formulated together as a single composition, or may be formulated and administered separately.
  • One or both compounds may be administered in a low dosage or in a high dosage, each of which is defined herein.
  • the composition, method, or kit may include one or more additional compounds (e.g., a glucocorticoid receptor modulator, NSAID, COX-2 inhibitor, DMARD, biologic, xanthine, small molecule immunomodulator, anticholinergic compound, beta receptor agonist, bronchodilator, non-steroidal immunophilin-dependent immunosuppressant, vitamin D analog, psoralen, retinoid, or 5-amino salicylic acid).
  • additional compounds e.g., a glucocorticoid receptor modulator, NSAID, COX-2 inhibitor, DMARD, biologic, xanthine, small molecule immunomodulator, anticholinergic compound, beta receptor agonist, bronchodilator, non-steroidal immunophilin-dependent immunosuppressant, vitamin D analog, psoralen, retinoid, or 5-amino salicylic acid.
  • additional compounds e.g., a glucocorticoid receptor modulator,
  • Combination therapies of the invention are especially useful for the treatment of immunoinflammatory disorders in combination with other anti-cytokine agents or agents that modulate the immune response to positively effect disease, such as agents that influence cell adhesion, or biologics or small molecules that block the action of IL-6, IL-1, IL-2, IL-12, IL-15 or TNF ⁇ (e.g., etanercept, adelimumab, infliximab, or CDP-870).
  • TNF ⁇ e.g., etanercept, adelimumab, infliximab, or CDP-870.
  • the combination therapy reduces the production of cytokines, etanercept or infliximab act on the remaining fraction of inflammatory cytokines, providing enhanced treatment.
  • p38 MAP kinase e.g., Doramapimod, SCIO-469, VX-702
  • ICE e.g., Pralnacasan
  • TACE e.g., BMS-561392
  • analogs of certain compounds may be employed in lieu of the compounds themselves.
  • Analogs of a tetra-substituted pyrimidopyrimidine and other compounds are described herein.
  • Structural analogs of a compound (e.g, ibudilast) or class of compound (e.g., antihistamines) do not need to have the same activity as the compound or class to which it is related.
  • an SSRI analog does not necessarily inhibit serotonin reuptake.
  • the invention features a method for identifying combinations of compounds useful for suppressing the secretion of proinflammatory cytokines in a patient in need of such treatment, said method comprising the steps of: (a) contacting cells in vitro with a combination of a tetra-substituted pyrimidopyrimidine, an antihistamine, a corticosteroid, ibudilast, rolipram, a tricyclic or tetracyclic antidepressant, or an SSRI and a candidate compound; and (b) determining whether the combination reduces cytokine levels in blood cells stimulated to secrete the cytokines relative to cells contacted with the tetra-substituted pyrimidopyrimidine, antihistamine, corticosteroid, ibudilast, rolipram, tricyclic or tetracyclic antidepressant, or SSRI but not contacted with the candidate compound or cells contacted with the candidate compound but not
  • the invention features a method for suppressing secretion of one or more proinflammatory cytokines in a cell by contacting the cell with: (i) a tetra-substituted pyrimidopyrimidine; and (ii) an antihistamine, a corticosteroid, ibudilast, rolipram, a tricyclic or tetracyclic antidepressant, or an SSRI simultaneously or within 14 days of each other in amounts sufficient to suppress secretion of one or more proinflammatory cytokines in the cell.
  • the preferred cytokines are TNF ⁇ , IL-1, IL-2 and INF- ⁇ .
  • the invention features a composition that includes an antihistamine, a corticosteroid, rolipram, ibudilast, a tricyclic or tetracyclic antidepressant, an SSRI, a non-steroidal anti-inflammatory drug, a non-steroidal immunophilin-dependent immunosuppressant, or an analog of any thereof; and (ii) a compound selected from the group consisting of a xanthine, small molecule immunomodulator, anticholinergic compound, biologic, DMARD, COX-2 inhibitor, beta-receptor agonist, bronchodilator, non-steroidal immunophilin-dependent immunosuppressant, vitamin D analog, psoralen, retinoid, or 5-amino salicylic acid.
  • the invention features a method of decreasing proinflammatory cytokine secretion or production in a patient by administering to a patient a tetra-substituted pyrimidopyrimidine or an analog thereof and an NSAID or an analog thereof simultaneously or within 14 days of each other in amounts sufficient to decrease proinflammatory cytokine secretion or production in the patient, with the proviso that when tetra-substituted pyrimidopyrimidine is dipyridamole, the NSAID is not aspirin.
  • the invention features a method for treating a patient diagnosed with or at risk of developing an immunoinflammatory disorder by administering to the patient a tetra-substituted pyrimidopyrimidine or an analog thereof and an NSAID or an analog thereof simultaneously or within 14 days of each other in amounts sufficient to treat the patient, with the proviso that when tetra-substituted pyrimidopyrimidine is dipyridamole, the NSAID is not aspirin.
  • the invention features a composition
  • a composition comprising a unit dose form of a tetra-substituted pyrimidopyrimidine and a second compound selected from an NSAID, COX-2 inhibitor, biologic, small molecule immunomodulator, DMARD, xanthine, anticholinergic compound, beta receptor agonist, bronchodilator, non-steroidal immunophilin-dependent immunosuppressant, vitamin D analog, psoralen, retinoid, and 5-amino salicylic acid with the proviso that when tetra-substituted pyrimidopyrimidine is dipyridamole, the second compound is not methotrexate or aspirin.
  • the unit dose form this composition can be oral, topical, parenteral, rectal, cutaneous, nasal, vaginal, inhalant, skin (patch), or ocular administration.
  • the invention also features a method for inhibiting proinflammatory cytokine activity in a patient suffering from or at risk of suffering from a disorder associated with at least one immunoinflammatory disorder mediated by the cytokine by administering to the patient a unit dose of a tetra-substituted pyrimidopyrimidine in an amount effective to inhibit or decrease the cytokine activity in the patient, wherein when the tetra-substituted pyrimidopyrimidine is dipyridamole, the unit dose is suitable for systemic administration.
  • the cytokine is desirably selected from TNF ⁇ , IL-1, IL-2, IL-6, IL-12, IL-15, and IFN- ⁇ , and
  • the methods and compositions of the invention desirably have increased effectiveness, safety, tolerability, or satisfaction of treatment of a patient suffering from or at risk of suffering from immunoinflammatory disorder, as compared to methods and compositions using each component of the combination individually.
  • dipyridamole also known as 2,6-bis(diethanolamino)-4,8-dipiperidinopyrimido(5,4-d)pyrimidine
  • 2,6-disubstituted 4,8-dibenzylaminopyrimido[5,4-d]pyrimidines mopidamole; dipyridamole monoacetate
  • NU3026 (2,6-di-(2,2-dimethyl-1,3-dioxolan-4-yl)-methoxy-4,8-di-piperidinopyrimidopyrimidine
  • NU3059 (2,6-bis-(2,3-dimethyoxypropoxy)-4,8-di-piperidinopyrimidopyrimidine
  • NU3060 (2,6-bis[N,N-di(2-methoxy)ethyl]-4,
  • corticosteroid any naturally occurring or synthetic compound characterized by a hydrogenated cyclopentanoperhydrophenanthrene ring system.
  • Naturally occurring corticosteroids are generally produced by the adrenal cortex.
  • Synthetic corticosteroids may be halogenated. Exemplary corticosteroids are described herein.
  • tricyclic or tetracyclic antidepressant is meant a compound having one the formulas (I), (II), (III), or (IV): wherein each X is, independently, H, Cl, F, Br, I, CH 3 , CF 3 , OH, OCH 3 , CH 2 CH 3 , or OCH 2 CH 3 ; Y is CH 2 , O, NH, S(O) 0-2 , (CH 2 ) 3 , (CH) 2 , CH 2 O, CH 2 NH, CHN, or CH 2 S; Z is C or S; A is a branched or unbranched, saturated or monounsaturated hydrocarbon chain having between 3 and 6 carbons, inclusive; each B is, independently, H, Cl, F, Br, I, CX 3 , CH 2 CH 3 , OCX 3 , or OCX 2 CX 3 ; and D is CH 2 , O, NH, S(O) 0-2 .
  • each X is, independently, H,
  • tetra-substituted pyrimidopyrimidine is meant a compound having the formula (V): wherein each Z and each Z′ is, independently, N, O, C,
  • each R is, independently, X, OH, N-alkyl (wherein the alkyl group has 1 to 20, more preferably 1-5, carbon atoms); a branched or unbranched alkyl group having 1 to 20, more preferably 1-5, carbon atoms; or a heterocycle, preferably as defined in formula (Y), below.
  • two R 1 groups from a common Z or Z′ atom, in combination with each other may represent —(CY 2 ) k — in which k is an integer between 4 and 6, inclusive.
  • Each Y is, independently, H, F, Cl, Br, or I.
  • each Z is the same moiety
  • each Z′ is the same moiety
  • Z and Z′ are different moieties
  • antihistamine is meant a compound that blocks the action of histamine.
  • Classes of antihistamines include but are not limited to ethanolamines, ethylenediamines, phenothiazines, alkylamines, piperazines, and piperidines.
  • SSRI selective serotonin reuptake inhibitor
  • SSRI any member of the class of compounds that (i) inhibit the uptake of serotonin by neurons of the central nervous system, (ii) have an inhibition constant (Ki) of 10 nM or less, and (iii) a selectivity for serotonin over norepinephrine (i.e., the ratio of Ki(norepinephrine) over Ki(serotonin)) of greater than 100.
  • Ki inhibition constant
  • Ki the ratio of Ki(norepinephrine
  • Ki(serotonin) the ratio of Ki(serotonin)
  • SSRIs are administered in dosages of greater than 10 mg per day when used as antidepressants.
  • Exemplary SSRIs for use in the invention are fluoxetine, fluvoxamine, paroxetine, sertraline, citalopram, and venlafaxine.
  • non-steroidal immunophilin-dependent immunosuppressant or “NsIDI” is meant any non-steroidal agent that decreases proinflammatory cytokine production or secretion, binds an immunophilin, or causes a down regulation of the proinflammatory reaction.
  • NsIDIs include calcineurin inhibitors, such as cyclosporine, tacrolimus, ascomycin, pimecrolimus, as well as other agents (peptides, peptide fragments, chemically modified peptides, or peptide mimetics) that inhibit the phosphatase activity of calcineurin.
  • NsIDIs also include rapamycin (sirolimus) and everolimus, which bind to an FK506-binding protein, FKBP-12, and block antigen-induced proliferation of white blood cells and cytokine secretion.
  • small molecule immunomodulator is meant a non-steroidal, non-NsIDI compound that decreases proinflammatory cytokine production or secretion, causes a down regulation of the proinflammatory reaction, or otherwise modulates the immune system in an immunophilin-independent manner.
  • Examplary small molecule immunomodulators are p38 MAP kinase inhibitors such as VX 702 (Vertex Pharmaceuticals), SCIO 469 (Scios), doramapimod (Boehringer Ingelheim), RO 30201195 (Roche), and SCIO 323 (Scios), TACE inhibitors such as DPC 333 (Bristol Myers Squibb), ICE inhibitors such as pranalcasan (Vertex Pharmaceuticals), and IMPDH inhibitors such as mycophenylate (Roche) and merimepodib (Vertex Pharamceuticals).
  • VX 702 Very Pharmaceuticals
  • SCIO 469 Scios
  • doramapimod Boehringer Ingelheim
  • RO 30201195 Roche
  • SCIO 323 Scios
  • TACE inhibitors such as DPC 333 (Bristol Myers Squibb)
  • ICE inhibitors such as pranalcasan
  • IMPDH inhibitors such as mycophenylate (Roche) and merimepodib (Vertex Ph
  • a “low dosage” is meant at least 5% less (e.g., at least 10%, 20%, 50%, 80%, 90%, or even 95%) than the lowest standard recommended dosage of a particular compound formulated for a given route of administration for treatment of any human disease or condition.
  • a low dosage of corticosteroid formulated for administration by inhalation will differ from a low dosage of corticosteroid formulated for oral administration.
  • a “high dosage” is meant at least 5% (e.g., at least 10%, 20%, 50%, 100%, 200%, or even 300%) more than the highest standard recommended dosage of a particular compound for treatment of any human disease or condition.
  • a “moderate dosage” is meant the dosage between the low dosage and the high dosage.
  • treating is meant administering or prescribing a composition for the treatment or prevention of an immunoinflammatory disease.
  • patient any animal (e.g., a human).
  • Other animals that can be treated using the methods, compositions, and kits of the invention include horses, dogs, cats, pigs, goats, rabbits, hamsters, monkeys, guinea pigs, rats, mice, lizards, snakes, sheep, cattle, fish, and birds.
  • an amount sufficient is meant the amount of a compound, in a combination of the invention, required to treat or prevent an immunoinflammatory disease in a clinically relevant manner.
  • a sufficient amount of an active compound used to practice the present invention for therapeutic treatment of conditions caused by or contributing to an immunoinflammatory disease varies depending upon the manner of administration, the age, body weight, and general health of the patient. Ultimately, the prescribers will decide the appropriate amount and dosage regimen. Additionally, an effective amount may can be that amount of compound in the combination of the invention that is safe and efficacious in the treatment of a patient having the immunoinflammatory disease over each agent alone as determined and approved by a regulary authority (such as the U.S. Food and Drug Administration).
  • Efficacy may be measured by a skilled practitioner using any standard method that is appropriate for a given indication.
  • immunoinflammatory disorder encompasses a variety of conditions, including autoimmune diseases, proliferative skin diseases, and inflammatory dermatoses. Immunoinflammatory disorders result in the destruction of healthy tissue by an inflammatory process, dysregulation of the immune system, and unwanted proliferation of cells.
  • immunoinflammatory disorders are acne vulgaris; acute respiratory distress syndrome; Addison's disease; allergic rhinitis; allergic intraocular inflammatory diseases, ANCA-associated small-vessel vasculitis; ankylosing spondylitis; arthritis, asthma; atherosclerosis; atopic dermatitis; autoimmune hemolytic anemia; autoimmune hepatitis; Behcet's disease; Bell's palsy; bullous pemphigoid; cerebral ischaemia; chronic obstructive pulmonary disease; cirrhosis; Cogan's syndrome; contact dermatitis; COPD; Crohn's disease; Cushing's syndrome; dermatomyositis; diabetes mellitus; discoid lupus erythematosus; eosinophilic fasciitis; erythema nodosum; exfoliative dermatitis; fibromyalgia; focal glomerulosclerosis; giant cell arteritis; gout; gouty,
  • Non-dermal inflammatory disorders include, for example, rheumatoid arthritis, inflammatory bowel disease, asthma, and chronic obstructive pulmonary disease.
  • “Dermal inflammatory disorders” or “inflammatory dernatoses” include, for example, psoriasis, acute febrile neutrophilic dermatosis, eczema (e.g., histotic eczema, dyshidrotic eczema, vesicular palmoplantar eczema), balanitis circumscripta plasmacellularis, balanoposthitis, Behcet's disease, erythema annulare centrifugum, erythema dyschromicum perstans, erythema multiforme, granuloma annulare, lichen nitidus, lichen planus, lichen sclerosus et atrophicus, lichen simplex chronicus, lichen spinulosus, nummular dermatitis, pyoderma gangrenosum, sarcoidosis, subcomeal pustular dermatosis, urticaria, and transient acantho
  • proliferative skin disease is meant a benign or malignant disease that is characterized by accelerated cell division in the epidermis or dermis.
  • proliferative skin diseases are psoriasis, atopic dermatitis, non-specific dermatitis, primary irritant contact dermatitis, allergic contact dermatitis, basal and squamous cell carcinomas of the skin, lamellar ichthyosis, epidermolytic hyperkeratosis, premalignant keratosis, acne, and seborrheic dermatitis.
  • a particular disease, disorder, or condition may be characterized as being both a proliferative skin disease and an inflammatory dermatosis.
  • An example of such a disease is psoriasis.
  • sustained release or “controlled release” is meant that the therapeutically active component is released from the formulation at a controlled rate such that therapeutically beneficial blood levels (but below toxic levels) of the component are maintained over an extended period of time ranging from e.g., about 12 to about 24 hours, thus, providing, for example, a 12 hour or a 24 hour dosage form.
  • the number of atoms of a particular type in a substituent group is generally given as a range, e.g., an alkyl group containing from 1 to 7 carbon atoms or C 1-7 alkyl. Reference to such a range is intended to include specific references to groups having each of the integer number of atoms within the specified range.
  • an alkyl group from 1 to 7 carbon atoms includes each of C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , and C 7 .
  • a C 1-7 heteroalkyl for example, includes from 1 to 7 carbon atoms in addition to one or more heteroatoms. Other numbers of atoms and other types of atoms may be indicated in a similar manner.
  • alkyl and the prefix “alk-” are inclusive of both straight chain and branched chain groups and of cyclic groups, i.e., cycloalkyl.
  • Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 6 ring carbon atoms, inclusive.
  • Exemplary cyclic groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups.
  • the C 1-7 alkyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C 1-7 alkyls include, without limitation, methyl; ethyl; n-propyl; isopropyl; cyclopropyl; cyclopropylmethyl; cyclopropylethyl; n-butyl; iso-butyl; sec-butyl; tert-butyl; cyclobutyl; cyclobutylmethyl; cyclobutylethyl; n-pentyl; cyclopentyl; cyclopentylmethyl; cyclopentylethyl; 1-methylbutyl; 2-methylbutyl; 3-methylbutyl; 2,2-dimethylpropyl; 1-ethylpropyl; 1,1-dimethylpropyl; 1,2-dimethylpropyl; 1-methylpentyl; 2-methylpentyl; 3-methylpentyl; 4-methylpentyl; 1,1-dimethylbutyl; 1,2-dimethylbutyl; 1,3-dimethylbut
  • C 2-7 alkenyl is meant a branched or unbranched hydrocarbon group containing one or more double bonds and having from 2 to 7 carbon atoms.
  • a C 2-7 alkenyl may optionally include monocyclic or polycyclic rings, in which each ring desirably has from three to six members.
  • the C 2-7 alkenyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C 2-7 alkenyls include, without limitation, vinyl; allyl; 2-cyclopropyl-1-ethenyl; 1-propenyl; 1-butenyl; 2-butenyl; 3-butenyl; 2-methyl-1-propenyl; 2-methyl-2-propenyl; 1-pentenyl; 2-pentenyl; 3-pentenyl; 4-pentenyl; 3-methyl-1-butenyl; 3-methyl-2-butenyl; 3-methyl-3-butenyl; 2-methyl-1-butenyl; 2-methyl-2-butenyl; 2-methyl-3-butenyl; 2-ethyl-2-propenyl; 1-methyl-1-butenyl; 1-methyl-2-butenyl; 1-methyl-3-butenyl; 2-methyl-2-pentenyl; 3-methyl-2-pentenyl; 4-methyl-2-pentenyl; 2-methyl-3-pentenyl; 3-methyl-3-pentenyl; 4-methyl-2-pentenyl; 2-methyl-3-pentenyl
  • C 2-7 alkynyl is meant a branched or unbranched hydrocarbon group containing one or more triple bonds and having from 2 to 7 carbon atoms.
  • a C 2-7 alkynyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members.
  • the C 2-7 alkynyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C 2-7 alkynyls include, without limitation, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 5-hexene-1-ynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl; 1-methyl-2-propynyl; 1-methyl-2-butynyl; 1-methyl-3-butynyl; 2-methyl-3-butynyl; 1,2-dimethyl-3-butynyl; 2,2-dimethyl-3-butynyl; 1-methyl-2-pentynyl; 2-methyl-3-pentynyl; 1-methyl-4-pentynyl; 2-methyl-4-pentynyl; and 3-methyl-4-pentynyl
  • C 2-6 heterocyclyl is meant a stable 5- to 7-membered monocyclic or 7- to 14-membered bicyclic heterocyclic ring which is saturated partially unsaturated or unsaturated (aromatic), and which consists of 2 to 6 carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • the nitrogen and sulfur heteroatoms may optionally be oxidized.
  • the heterocyclic ring may be covalently attached via any heteroatom or carbon atom that results in a stable structure, e.g., an imidazolinyl ring may be linked at either of the ring-carbon atom positions or at the nitrogen atom.
  • a nitrogen atom in the heterocycle may optionally be quaternized.
  • Heterocycles include, without limitation, 1H-indazole, 2-pyrrolidonyl, 2H,6H-1,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H-quinolizinyl, 6H-1,2,5-thiadiazinyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, b-carboliny
  • Preferred 5 to 10 membered heterocycles include, but are not limited to, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, tetrazolyl, benzofuranyl, benzothiofuranyl, indolyl, benzimidazolyl, 1H-indazolyl, oxazolidinyl, isoxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, quinolinyl, and isoquinolinyl.
  • Preferred 5 to 6 membered heterocycles include, without limitation, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, piperazinyl, piperidinyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, 1,4,5,6-tetrahydro pyridinyl, and tetrazolyl.
  • C 6-12 aryl is meant an aromatic group having a ring system comprised of carbon atoms with conjugated ⁇ electrons (e.g., phenyl).
  • the aryl group has from 6 to 12 carbon atoms.
  • Aryl groups may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members.
  • the aryl group may be substituted or unsubstituted.
  • Exemplary subsituents include alkyl, hydroxy, alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, fluoroalkyl, carboxyl, hydroxyalkyl, carboxyalkyl, amino, aminoalkyl, monosubstituted amino, disubstituted amino, and quaternary amino groups.
  • C 7-14 alkaryl is meant an alkyl substituted by an aryl group (e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl) having from 7 to 14 carbon atoms.
  • aryl group e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl
  • C 3-10 alkheterocyclyl is meant an alkyl substituted heterocyclic group having from 7 to 14 carbon atoms in addition to one or more heteroatoms (e.g., 3-furanylmethyl, 2-furanylmethyl, 3-tetrahydrofuranylmethyl, or 2-tetrahydrofuranylmethyl).
  • C 1-7 heteroalkyl is meant a branched or unbranched alkyl, alkenyl, or alkynyl group having from 1 to 7 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P.
  • Heteroalkyls include, without limitation, tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides.
  • a heteroalkyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members.
  • the heteroalkyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • acyl is meant a chemical moiety with the formula R—C(O)—, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • alkoxy is meant a chemical substituent of the formula —OR, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • aryloxy is meant a chemical substituent of the formula —OR, wherein R is a C 6-12 aryl group.
  • —NRR′ a chemical substituent of the formula —NRR′, wherein the nitrogen atom is part of an amide bond (e.g., —C(O)—NRR′) and wherein R and R′ are each, independently, selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, and C 1-7 heteroalkyl, or —NRR′ forms a C 2-6 heterocyclyl ring, as defined above, but containing at least one nitrogen atom, such as piperidino, morpholino, and azabicyclo, among others.
  • R and R′ are each, independently, selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocycly
  • halide is meant bromine, chlorine, iodine, or fluorine.
  • fluoroalkyl is meant an alkyl group that is substituted with a fluorine.
  • perfluoroalkyl is meant an alkyl group consisting of only carbon and fluorine atoms.
  • Carboxyalkyl is meant a chemical moiety with the formula —(R)—COOH, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • hydroxyalkyl is meant a chemical moiety with the formula —(R)—OH, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • alkylthio is meant a chemical substituent of the formula —SR, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-1 2 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • arylthio is meant a chemical substituent of the formula —SR, wherein R is a C 6-12 aryl group.
  • quaternary amino is meant a chemical substituent of the formula —(R)—N(R′)(R′′)(R′′′) + , wherein R, R′, R′′, and R′′′ are each independently an alkyl, alkenyl, alkynyl, or aryl group.
  • R may be an alkyl group linking the quaternary amino nitrogen atom, as a substituent, to another moiety.
  • the nitrogen atom, N is covalently attached to four carbon atoms of alkyl and/or aryl groups, resulting in a positive charge at the nitrogen atom.
  • pharmaceutically acceptable salt represents those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art.
  • the salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphersulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, isethionate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, mesylate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxa
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • Compounds useful in the invention include those described herein in any of their pharmaceutically acceptable forms, including isomers such as diastereomers and enantiomers, salts, esters, amides, thioesters, solvates, and polymorphs thereof, as well as racemic mixtures and pure isomers of the compounds described herein.
  • fexofenadine is meant the free base, as well as any pharmaceutically acceptable salt thereof (e.g., fexofenadine hydrochloride).
  • any of the foregoing conditions may be treated by administration of an effective amount of a tetra-substituted pyrimidopyrimidine or analog thereof, either alone or in combination with one or more companion compounds, including an antihistamine, a corticosteroid, rolipram, ibudilast, a tricyclic or tetracyclic antidepressant, an SSRI, a non-steroidal anti-inflammatory drug, a non-steroidal immunophilin-dependent immunosuppressant, and an analog thereof.
  • companion compounds including an antihistamine, a corticosteroid, rolipram, ibudilast, a tricyclic or tetracyclic antidepressant, an SSRI, a non-steroidal anti-inflammatory drug, a non-steroidal immunophilin-dependent immunosuppressant, and an analog thereof.
  • treatment of an immunoinflammatory disorder is performed by administering a tetra-substituted pyrimidopyrimidine (or an analog thereof) and an antihistamine to a patient in need of such treatment.
  • an immunoinflammatory disorder e.g., an inflammatory dermatosis, proliferative skin disease, organ transplant rejection, or graft versus host disease
  • treatment of an immunoinflammatory disorder is performed by administering a tetra-substituted pyrimidopyrimidine (or an analog thereof) and a tricyclic or tetracyclic antidepressant to a patient in need of such treatment.
  • treatment is performed by administering a tetra-substituted pyrimidopyrimidine (or an analog thereof) and a selective serotonin reuptake inhibitor to a patient suffering from any of the foregoing conditions.
  • treatment is performed by administering to a patient in need of such treatment, in conjunction with a tetra-substituted pyrimidopyrimidine or a tetra-substituted pyrimidopyrimidine analog, a corticosteroid, or ibudilast, or rolipram, or an analog of any of these compounds.
  • Routes of administration for the various embodiments include, but are not limited to, topical, transdermal, and systemic administration (such as, intravenous, intramuscular, subcutaneous, inhalation, rectal, buccal, vaginal, intraperitoneal, intraarticular, ophthalmic or oral administration).
  • systemic administration refers to all nondermal routes of administration, and specifically excludes topical and transdermal routes of administration.
  • Any of the foregoing therapies may be administered with conventional pharmaceuticals useful for the treatment of immunoinflammatory disorders.
  • tetra-substituted pyrimidopyrimidines are effective in treating immunoinflammatory diseases, particularly those mediated by TNF ⁇ , IL-1, or IFN- ⁇ .
  • Tetra-substituted pyrimidopyrimidines have the formula (V): wherein each Z and each Z′ is, independently, N, O, C,
  • each R 1 is, independently, X, OH, N-alkyl (wherein the alkyl group has 1 to 20, more preferably 1-5, carbon atoms); a branched or unbranched alkyl group having 1 to 20, more preferably 1-5, carbon atoms; or a heterocycle, preferably as defined in formula (Y), below.
  • two R 1 groups from a common Z or Z′ atom, in combination with each other may represent —(CY 2 ) k — in which k is an integer between 4 and 6, inclusive.
  • Each Y is, independently, H, F, Cl, Br, or I.
  • each Z is the same moiety, each Z′ is the same moiety, and Z and Z′ are different moieties.
  • Tetra-substituted pyrimidopyrimidines that are useful in the methods, compositions, and kits of this invention include 2,6-disubstituted 4,8-dibenzylaminopyrimido[5,4-d]pyrimidines.
  • dipyridamole also known as 2,6-bis(diethanolamino)-4,8-dipiperidinopyrimido(5,4-d)pyrimidine
  • mopidamole dipyridamole monoacetate
  • NU3026 (2,6-di-(2,2-dimethyl-1,3-dioxolan-4-yl)-methoxy-4,8-di-piperidinopyrimidopyrimidine
  • NU3059 (2,6-bis-(2,3-dimethyoxypropoxy)-4,8-di-piperidinopyrimidopyrimidine
  • NU3060 (2,6-bis[N,N-di(2-methoxy)ethyl]-4,6-di-piperidinopyrimidopyrimidine
  • NU3076 (2,6-bis(diethanolamino)-4,8-di-4-methoxybenzylamin
  • the standard recommended dosage for dipyridamole is 300-400 mg/day.
  • the invention relates to a method for inhibiting proinflammatory cytokine activity in a patient suffering from or at risk of suffering from a disorder associated with cytokine activity, comprising administering to the patient a unit dose of a tetra-substituted pyrimidopyrimidine in an amount effective to inhibit or decrease the cytokine activity in the patient, said cytokine selected from TNF ⁇ , IL-1, IL-2, IL-6, IL-12, IL-15, or IFN- ⁇ , and wherein when the tetra-substituted pyrimidopyrimidine is dipyridamole, the unit dose is suitable for systemic administration.
  • Antihistamines are compounds that block the action of histamine. Classes of antihistamines include:
  • non-sedating and sedating antihistamines may be employed.
  • Particularly desirable antihistamines for use in the methods, compositions, and kits of the invention are non-sedating antihistamines such as loratadine and desloratadine. Sedating antihistamines can also be used in the methods, compositions, and kits of the invention.
  • Preferred sedating antihistamines are methods, compositions, and kits of the invention are azatadine, bromodiphenhydramine; chlorpheniramine; clemizole; cyproheptadine; dimenhydrinate; diphenhydramine; doxylamine; meclizine; promethazine; pyrilamine; thiethylperazine; and tripelennamine.
  • antihistamines suitable for use in the methods and compositions of the invention are acrivastine; ahistan; antazoline; astemizole; azelastine (e.g., azelsatine hydrochloride); bamipine; bepotastine; bietanautine; brompheniramine (e.g., brompheniramine maleate); carbinoxamine (e.g., carbinoxamine maleate); cetirizine (e.g., cetirizine hydrochloride); cetoxime; chlorocyclizine; chloropyramine; chlorothen; chlorphenoxamine; cinnarizine; clemastine (e.g., clemastine fumarate); clobenzepam; clobenztropine; clocinizine; cyclizine (e.g., cyclizine hydrochloride; cyclizine lactate); deptropine; dexchlorpheniramine; de
  • Antihistamine analogs include, without limitation, 10-piperazinylpropylphenothiazine; 4-(3-(2-chlorophenothiazin-10-yl)propyl)-1-piperazineethanol dihydrochloride; 1-(10-(3-(4-methyl-1-piperazinyl)propyl)-10H-phenothiazin-2-yl)-(9CI) 1-propanone; 3-methoxycyproheptadine; 4-(3-(2-Chloro-10H-phenothiazin-10-yl)propyl)piperazine-1-ethanol hydrochloride; 10,11-dihydro-5-(3-(4-ethoxycarbonyl-4-phenylpiperidino)propylidene)-5H-dibenzo(a,d)cycloheptene; aceprometazine; acetophenazine; alimemaz
  • AD-0261 AHR-5333; alinastine; arpromidine; ATI-19000; bermastine; bilastin; Bron-12; carebastine; chlorphenamine; clofurenadine; corsym; DF-1105501; DF-11062; DF-1111301; EL-301; elbanizine; F-7946T; F-9505; HE-90481; HE-90512; hivenyl; HSR-609; icotidine; KAA-276; KY-234; lamiakast; LAS-36509; LAS-36674; levocetirizine; levoprotiline; metoclopramide; NIP-531; noberastine; oxatomide; PR-881-884A; quisultazine; rocastine; selenotifen; SK&F-94461; SODAS-HC; tagorizine; TAK-4
  • Standard recommended dosages for several exemplary antihistamines are shown in Table 1. Other standard dosages are provided, e.g., in the Merck Manual of Diagnosis & Therapy (17th Ed. MH Beers et al., Merck & Co.) and Physicians' Desk Reference 2003 (57 th Ed. Medical Economics Staff et al., Medical Economics Co., 2002).
  • Loratadine is a tricyclic piperidine that acts as a selective peripheral histamine H1-receptor antagonist.
  • loratadine and structural and functional analogs thereof such as piperidines, tricyclic piperidines, histamine H1-receptor antagonists, are useful in the anti-immunoinflammatory combination of the invention for the treatment of immunoinflammatory disorders, transplanted organ rejection, and graft versus host disease.
  • Loratadine functional and/or structural analogs include other H1-receptor antagonists, such as AHR-11325, acrivastine, antazoline, astemizole, azatadine, azelastine, bromopheniramine, carebastine, cetirizine, chlorpheniramine, chlorcyclizine, clemastine, cyproheptadine, descarboethoxyloratadine, dexchlorpheniramine, dimenhydrinate, diphenylpyraline, diphenhydramine, ebastine, fexofenadine, hydroxyzine ketotifen, lodoxamide, levocabastine, methdilazine, mequitazine, oxatomide, pheniramine pyrilamine, promethazine, pyrilamine, setastine, tazifylline, warmthlastine, terfenadine, trimeprazine, tripelennamine, trip
  • Piperidine H1-receptor antagonists include loratadine, cyproheptadine hydrochloride (PERIACTIN), and phenindiamine tartrate (NOLAHIST).
  • Piperazine H1-receptor antagonists include hydroxyzine hydrochloride (ATARAX), hydroxyzine pamoate (VISTARIL), cyclizine hydrochloride (MAREZINE), cyclizine lactate, and meclizine hydrochloride.
  • Loratadine oral formulations include tablets, redi-tabs, and syrup. Loratadine tablets contain 10 mg micronized loratadine. Loratadine syrup contains 1 mg/ml micronized loratadine, and reditabs (rapidly-disintegrating tablets) contain 10 mg micronized loratadine in tablets that disintegrate quickly in the mouth. While suggested dosages will vary with a patient's condition, standard recommended dosages are provided below. Loratadine is typically administered once daily in a 10 mg dose, although other daily dosages useful in the anti-immunoinflammatory combination of the invention include 0.01-0.05 mg, 0.05-1 mg, 1-3 mg, 3-5 mg, 5-10 mg, 10-15 mg, 15-20 mg, 20-30 mg, and 30-40 mg.
  • Loratadine is rapidly absorbed following oral administration. It is metabolized in the liver to descarboethoxyloratadine by cytochrome P450 3A4 and cytochrome P450 2D6. Loratadine metabolites are also useful in the anti-immunoinflammatory combination of the invention.
  • one or more corticosteroid may be administered in a method of the invention or may be formulated with a tetra-substituted pyrimidopyrimidine or analog thereof in a composition of the invention.
  • Our data show that dipyridamole in combination with various corticosteroids is more effective in suppressing TNF ⁇ in vitro than either agent alone. Accordingly, this combination may be more effective in treating immunoinflammatory diseases, particularly those mediated by TNF ⁇ levels, than either the tetra-substituted pyrimidopyrimidine or corticosteroid alone.
  • Suitable corticosteroids include 11-alpha, 17-alpha,21-trihydroxypregn-4-ene-3,20-dione; 11-beta,16-alpha,17,21-tetrahydroxypregn-4-ene-3,20-dione; 11-beta,16-alpha, 17,21-tetrahydroxypregn-1,4-diene-3,20-dione; 11-beta, 17-alpha,21-trihydroxy-6-alpha-methylpregn-4-ene-3,20-dione; 1-dehydrocorticosterone; 11-deoxycortisol; 11-hydroxy-1,4-androstadiene-3,17-dione; 11-ketotestosterone; 14-hydroxyandrost-4-ene-3,6,17-trione; 15,17-dihydroxyprogesterone; 16-methylhydrocortisone; 17,21-dihydroxy-16-alpha-methylpregna-1,4,9(11)-triene
  • Standard recommended dosages for various steroid/disease combinations are provided in Table 2, below.
  • TABLE 2 Standard Recommended Corticosteroid Dosages Indication Route Drug Dose Schedule Psoriasis oral prednisolone 7.5-60 mg per day or divided b.i.d. oral prednisone 7.5-60 mg per day or divided b.i.d.
  • inhaled fluticasone propionate 44, 110 or 220 ⁇ g/puff
  • puffs b.i.d. inhaled triamcinolone acetonide 100 ⁇ g/puff
  • puffs b.i.d. COPD oral prednisone 30-40 mg per day Crohn's disease oral budesonide 9 mg per day Ulcerative colitis oral prednisone 40-60 mg per day oral hydrocortisone 300 mg (IV) per day oral methylprednisolone 40-60 mg per day Rheumatoid arthritis oral prednisone 10 mg per day
  • the dosage of corticosteroid administered is a dosage equivalent to a prednisolone dosage, as defined herein.
  • a low dosage of a corticosteroid may be considered as the dosage equivalent to a low dosage of prednisolone.
  • Steroid receptor modulators e.g., antagonists and agonists
  • the invention features the combination of a tetra-substituted pyrimidopyridine and a glucocorticoid receptor modulator or other steroid receptor modulator, and methods of treating immunoinflammatory disorders therewith.
  • Glucocorticoid receptor modulators that may used in the methods, compositions, and kits of the invention include compounds described in U.S. Pat. Nos. 6,380,207, 6,380,223, 6,448,405, 6,506,766, and 6,570,020, U.S. Patent Application Publication Nos. 2003/0176478, 2003/0171585, 2003/0120081, 2003/0073703, 2002/015631, 2002/0147336, 2002/0107235, 2002/0103217, and 2001/0041802, and PCT Publication No. WO00/66522, each of which is hereby incorporated by reference.
  • Other steroid receptor modulators may also be used in the methods, compositions, and kits of the invention are described in U.S. Pat.
  • a tetra-substituted pyrimidopyrimidine or a tetra-substituted pyrimidopyrimidine analog may be administered or formulated with ibudilast or an ibudilast analog, defined by formula (VI).
  • R 1 and R 2 are each, independently, selected from H, C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, and C 1-7 heteroalkyl;
  • R 3 is selected from H, halide, alkoxy, and C 1-4 alkyl;
  • X 1 is selected from C ⁇ O, C ⁇ N—NH—R 4 , C ⁇ C(R 5 )—C(O)—R 6 , C ⁇ CH ⁇ CH—C(O)—R 6 , and C(OH)—R 7 ;
  • R 4 is selected from H and acyl;
  • R 5 is selected from H, halide, and C 1-4 alkyl;
  • R 6 is selected from OH, alkoxy and amido;
  • R 7 is selected from H, C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl
  • Compounds of formula (VI) include, the compounds described in U.S. Pat. Nos. 3,850,941; 4,097,483; 4,578,392; 4,925,849; 4,994,453; and 5,296,490.
  • Commercially available compounds of formula (VI) include ibudilast and KC-764.
  • the standard recommended dosage for the treatment of bronchial asthma is typically 10 mg of ibudilast twice daily, while in the case of cerebrovascular disorders, the standard recoomended dosage is 10 mg of ibudilast three times daily.
  • the structure of ibudilast is shown below:
  • KC-764 (CAS 94457-09-7) is reported to be a platelet aggregation inhibitor.
  • the structure of KC-764 is shown below:
  • KC-764 and other compound of formula (VI) can be prepared using the synthetic methods described in U.S. Pat. Nos. 3,850,941; 4,097,483; 4,578,392; 4,925,849; 4,994,453; and 5,296,490.
  • a tetra-substituted pyrimidopyrimidine or analog thereof is administered or formulated with rolipram (4-[3-(cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidone) or an analog of rolipram.
  • rolipram 4-[3-(cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidone
  • Rolipram analogs are described by formula (I) of U.S. Pat. No. 4,193,926, hereby incorporated by reference.
  • a tetra-substituted pyrimidopyrimidine or analog thereof is administered or formulated with a tricyclic or tetracyclic antidepressant, or an analog thereof.
  • tricyclic or tetracyclic antidepressant analog is meant a compound having one the formulas (I), (II), (III), or (IV): or a pharmaceutically acceptable salt, ester, amide, or derivative thereof, wherein each X is, independently, H, Cl, F, Br, I, CH 3 , CF 3 , OH, OCH 3 , CH 2 CH 3 , or OCH 2 CH 3 ;Y is CH 2 , O, NH, S(O) 0-2 , (CH 2 ) 3 , (CH) 2 , CH 2 O, CH 2 NH, CHN, or CH 2 S; Z is C or S; A is a branched or unbranched, saturated or monounsaturated hydrocarbon chain having between 3 and 6 carbons
  • Tricyclic or tetracyclic antidepressants include 10-(4-methylpiperazin-1-yl)pyrido(4,3-b)(1,4)benzothiazepine; 11-(4-methyl-1-piperazinyl)-5H-dibenzo(b,e)(1,4)diazepine; 5,10-dihydro-7-chloro-10-(2-(morpholino)ethyl)-11H-dibenzo(b,e)(1,4)diazepin-11-one; 2-(2-(7-hydroxy-4-dibenzo(b,f)(1,4)thiazepine-11-yl-1-piperazinyl)ethoxy)ethanol; 2-chloro-11-(4-methyl-1-piperazinyl)-5H-dibenzo(b,e)(1,4)diazepine; 4-(11H-dibenz(b,e)a
  • a tetra-substituted pyrimidopyrimidine or analog thereof is administered or formulated with an SSRI or an analog thereof.
  • Suitable SSRIs include cericlamine (e.g., cericlamine hydrochloride); citalopram (e.g., citalopram hydrobromide); clovoxamine; cyanodothiepin; dapoxetine; escitalopram (escitalopram oxalate); femoxetine (e.g., femoxetine hydrochloride); fluoxetine (e.g., fluoxetine hydrochloride); fluvoxamine (e.g., fluvoxamine maleate); ifoxetine; indalpine (e.g., indalpine hydrochloride); indeloxazine (e.g., indeloxazine hydrochloride); litoxetine; milnacipran (e.g.
  • Cericlamine has the following structure:
  • Structural analogs of cericlamine are those having the formula: as well as pharmaceutically acceptable salts thereof, wherein R 1 is a C 1 -C 4 alkyl and R 2 is H or C 1-4 alkyl, R 3 is H, C 1-4 alkyl, C 2-4 alkenyl, phenylalkyl or cycloalkylalkyl with 3 to 6 cyclic carbon atoms, alkanoyl, phenylalkanoyl or cycloalkylcarbonyl having 3 to 6 cyclic carbon atoms, or R 2 and R 3 form, together with the nitrogen atom to which they are linked, a heterocycle saturated with 5 to 7 chain links which can have, as the second heteroatom not directly connected to the nitrogen atom, an oxygen, a sulphur or a nitrogen, the latter nitrogen heteroatom possibly carrying a C 2-4 alkyl.
  • cericlamine structural analogs are 2-methyl-2-amino-3-(3,4-dichlorophenyl)-propanol, 2-pentyl-2-amino-3-(3,4-dichlorophenyl)-propanol, 2-methyl-2-methylamino-3-(3,4-dichlorophenyl)-propanol, 2-methyl-2-dimethylamino-3-(3,4-dichlorophenyl)-propanol, and pharmaceutically acceptable salts of any thereof.
  • Citalopram has the following structure:
  • Structural analogs of citalopram are those having the formula: as well as pharmaceutically acceptable salts thereof, wherein each of R 1 and R 2 is independently selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, cyano and R—CO—, wherein R is C 1-4 alkyl.
  • Exemplary citalopram structural analogs are 1-(4′-fluorophenyl)-1-(3-dimethylaminopropyl)-5-bromophthalane; 1-(4′-chlorophenyl)-1-(3-dimethylaminopropyl)-5-chlorophthalane; 1-(4′-bromophenyl)-1-(3-dimethylaminopropyl)-5-chlorophthalane; 1-(4′-fluorophenyl)-1-(3-dimethylaminopropyl)-5-chlorophthalane; 1-(4′-chlorophenyl)-1-(3-dimethylaminopropyl)-5-trifluoromethyl-phthalane; 1-(4′-bromophenyl)-1-(3-dimethylaminopropyl)-5-trifluoromethyl-phthalane; 1-(4′-fluorophenyl)-1-(3-dimethylamin
  • Clovoxamine has the following structure:
  • Structural analogs of clovoxamine are those having the formula: as well as pharmaceutically acceptable salts thereof, wherein Hal is a chloro, bromo, or fluoro group and R is a cyano, methoxy, ethoxy, methoxymethyl, ethoxymethyl, methoxyethoxy, or cyanomethyl group.
  • Exemplary clovoxamine structural analogs are 4′-chloro-5-ethoxyvalerophenone O-(2-aminoethyl)oxime; 4′-chloro-5-(2-methoxyethoxy)valerophenone O-(2-aminoethyl)oxime; 4′-chloro-6-methoxycaprophenone O-(2-aminoethyl)oxime; 4′-chloro-6-ethoxycaprophenone O-(2-aminoethyl)oxime; 4′-bromo-5-(2-methoxyethoxy)valerophenone O-(2-aminoethyl)oxime; 4′-bromo-5-methoxyvalerophenone O-(2-aminoethyl)oxime; 4′-chloro-6-cyanocaprophenone O-(2-aminoethyl)oxime; 4′-chloro-5-cyanovalerophenone O-(2-
  • Femoxetine has the following structure:
  • Structural analogs of femoxetine are those having the formula: wherein R 1 represents a C 1-4 alkyl or C 2-4 alkynyl group, or a phenyl group optionally substituted by C 1-4 alkyl, C 1-4 alkylthio, C 1-4 alkoxy, bromo, chloro, fluoro, nitro, acylamino, methylsulfonyl, methylenedioxy, or tetrahydronaphthyl, R 2 represents a C 1-4 alkyl or C 2-4 alkynyl group, and R 3 represents hydrogen, C 1-4 alkyl, C 1-4 alkoxy, trifluoroalkyl, hydroxy, bromo, chloro, fluoro, methylthio, or aralkyloxy.
  • Fluoxetine has the following structure:
  • Structural analogs of fluoxetine are those compounds having the formula: as well as pharmaceutically acceptable salts thereof, wherein each R 1 is independently hydrogen or methyl; R is naphthyl or wherein each of R 2 and R 3 is, independently, bromo, chloro, fluoro, trifluoromethyl, C 1-4 alkyl, C 1-3 alkoxy or C 3-4 alkenyl; and each of n and m is, independently, 0, 1 or 2.
  • R is naphthyl, it can be either ⁇ -naphthyl or naphthyl.
  • Exemplary fluoxetine structural analogs are 3-(p-isopropoxyphenoxy)-3-phenylpropylamine methanesulfonate, N,N-dimethyl 3-(3′,4′-dimethoxyphenoxy)-3-phenylpropylamine p-hydroxybenzoate, N,N-dimethyl 3- ⁇ -naphthoxy)-3-phenylpropylamine bromide, N,N-dimethyl 3-( ⁇ -naphthoxy)-3-phenyl-1-methylpropylamine iodide, 3-(2′-methyl-4′,5′-dichlorophenoxy)-3-phenylpropylamine nitrate, 3-(p-t-butylphenoxy)-3-phenylpropylamine glutarate, N-methyl 3-(2′-chloro-p-tolyloxy)-3-phenyl-1-methylpropylamine lactate, 3-(2′,4′-dichlorophenoxy)-3-phenyl
  • Fluvoxamine has the following structure:
  • Structural analogs of fluvoxamine are those having the formula: as well as pharmaceutically acceptable salts thereof, wherein R is cyano, cyanomethyl, methoxymethyl, or ethoxymethyl.
  • Indalpine has the following structure:
  • Structural analogs of indalpine are those having the formula: or pharmaceutically acceptable salts thereof, wherein R 1 is a hydrogen atom, a C 1 -C 4 alkyl group, or an aralkyl group of which the alkyl has 1 or 2 carbon atoms, R 2 is hydrogen, C 1-4 alkyl, C 1-4 alkoxy or C 1-4 alkylthio, chloro, bromo, fluoro, trifluoromethyl, nitro, hydroxy, or amino, the latter optionally substituted by one or two C 1-4 alkyl groups, an acyl group or a C 1-4 alkylsulfonyl group; A represents —CO or —CH 2 — group; and n is 0, 1 or 2.
  • indalpine structural analogs are indolyl-3 (piperidyl-4 methyl) ketone; (methoxy-5-indolyl-3) (piperidyl-4 methyl) ketone; (chloro-5-indolyl-3) (piperidyl-4 methyl) ketone; (indolyl-3)-1(piperidyl-4)-3 propanone, indolyl-3 piperidyl-4 ketone; (methyl-1 indolyl-3) (piperidyl-4 methyl) ketone, (benzyl-1 indolyl-3) (piperidyl-4 methyl) ketone; [(methoxy-5 indolyl-3)-2 ethyl]-piperidine, [(methyl-1 indolyl-3)-2 ethyl]-4-piperidine; [(indolyl-3)-2 ethyl]-4 piperidine; (indolyl-3 methyl)-4 piperidine, [(chloro
  • Indeloxezine has the following structure:
  • Structural analogs of indeloxazine are those having the formula: and pharmaceutically acceptable salts thereof, wherein R 1 and R 3 each represents hydrogen, C 1-4 alkyl, or phenyl; R 2 represents hydrogen, C 1-4 alkyl, C 4-7 cycloalkyl, phenyl, or benzyl; one of the dotted lines means a single bond and the other means a double bond, or the tautomeric mixtures thereof.
  • Exemplary indeloxazine structural analogs are 2-(7-indenyloxymethyl)-4-isopropylmorpholine; 4-butyl-2-(7-indenyloxymethyl)morpholine; 2-(7-indenyloxymethyl)-4-methylmorpholine; 4-ethyl-2-(7-indenyloxymethyl)morpholine, 2-(7-indenyloxymethyl)-morpholine; 2-(7-indenyloxymethyl)-4-propylmorpholine; 4-cyclohexyl-2-(7-indenyloxymethyl)morpholine; 4-benzyl-2-(7-indenyloxymethyl)-morpholine; 2-(7-indenyloxymethyl)-4-phenylmorpholine; 2-(4-indenyloxymethyl)morpholine; 2-(3-methyl-7-indenyloxymethyl)-morpholine; 4-isopropyl-2-(3-methyl-7-indenyloxymethyl)morpholine; 4-is
  • Milnacipram has the following structure:
  • Structural analogs of milnacipram are those having the formula: as well as pharmaceutically acceptable salts thereof, wherein each R, independently, represents hydrogen, bromo, chloro, fluoro, C 1-4 alkyl, C 1-4 alkoxy, hydroxy, nitro or amino; each of R 1 and R 2 , independently, represents hydrogen, C 1-4 alkyl, C 6-12 aryl or C 7-14 alkylaryl, optionally substituted, preferably in para position, by bromo, chloro, or fluoro, or R 1 and R 2 together form a heterocycle having 5 or 6 members with the adjacent nitrogen atoms; R 3 and R 4 represent hydrogen or a C 1-4 alkyl group or R 3 and R 4 form with the adjacent nitrogen atom a heterocycle having 5 or 6 members, optionally containing an additional heteroatom selected from nitrogen, sulphur, and oxygen.
  • Exemplary milnacipram structural analogs are 1-phenyl 1-aminocarbonyl 2-dimethylaminomethyl cyclopropane; 1-phenyl 1-dimethylaminocarbonyl 2-dimethylaminomethyl cyclopropane; 1-phenyl 1-ethylaminocarbonyl 2-dimethylaminomethyl cyclopropane; 1-phenyl 1-diethylaminocarbonyl 2-aminomethyl cyclopropane; 1-phenyl 2-dimethylaminomethyl N-(4′-chlorophenyl)cyclopropane carboxamide; 1-phenyl 2-dimethylaminomethyl N-(4′-chlorobenzyl)cyclopropane carboxamide; 1-phenyl 2-dimethylaminomethyl N-(2-phenylethyl)cyclopropane carboxamide; (3,4-dichloro-1-phenyl) 2-dimethylaminomethyl N,N-dimethylcyclopropan
  • Paroxetine has the following structure:
  • Structural analogs of paroxetine are those having the formula: and pharmaceutically acceptable salts thereof, wherein R 1 represents hydrogen or a C 1-4 alkyl group, and the fluorine atom may be in any of the available positions.
  • Sertraline has the following structure:
  • Structural analogs of sertraline are those having the formula: wherein R 1 is selected from the group consisting of hydrogen and C 1-4 alkyl; R 2 is C 1-4 alkyl; X and Y are each selected from the group consisting of hydrogen, fluoro, chloro, bromo, trifluoromethyl, C 1-3 alkoxy, and cyano; and W is selected from the group consisting of hydrogen, fluoro, chloro, bromo, trifluoromethyl and C 1-3 alkoxy.
  • Preferred sertraline analogs are in the cis-isomeric configuration.
  • cis-isomeric refers to the relative orientation of the NR 1 R 2 and phenyl moieties on the cyclohexene ring (i.e. they are both oriented on the same side of the ring). Because both the 1- and 4-carbons are asymmetrically substituted, each cis-compound has two optically active enantiomeric forms denoted (with reference to the 1-carbon) as the cis-(1R) and cis-(1S) enantiomers.
  • Sibutramine hydrochloride monohydrate (MERIDIATM) is an orally administered agent for the treatment of obesity.
  • Sibutramine hydrochloride is a racemic mixture of the (+) and ( ⁇ ) enantiomers of cyclobutanemethanamine, 1-(4-chlorophenyl)-N,N-dimethyl-(alpha)-(2-methylpropyl)-, hydrochloride, monohydrate.
  • Each MERIDIATM capsule contains 5 mg, 10 mg, or 15 mg of sibutramine hydrochloride monohydrate.
  • the recommended starting dose of MERIDIATM is 10 mg administered once daily with or without food. If there is inadequate weight loss, the dose may be titrated after four weeks to a total of 15 mg once daily. The 5 mg dose is typically reserved for patients who do not tolerate the 10 mg dose.
  • Zimeldine has the following structure:
  • Structural analogs of zimeldine are those compounds having the formula: and pharmaceutically acceptable salts thereof, wherein the pyridine nucleus is bound in ortho-, meta- or para-position to the adjacent carbon atom and where R 1 is selected from the group consisting of H, chloro, fluoro, and bromo.
  • Exemplary zimeldine analogs are (e)- and (z)-3-(4′-bromophenyl-3-(2′′-pyridyl)-dimethylallylamine; 3-(4′-bromophenyl)-3-(3′′-pyridyl)-dimethylallylamine; 3-(4′-bromophenyl)-3-(4′′-pyridyl)-dimethylallylamine; and pharmaceutically acceptable salts of any thereof.
  • Structural analogs of any of the above SSRIs are considered herein to be SSRI analogs and thus may be employed in any of the methods, compositions, and kits of the invention.
  • Pharmacologically active metabolites of any of the foregoing SSRIs can also be used in the methods, compositions, and kits of the invention.
  • Exemplary metabolites are didesmethylcitalopram, desmethylcitalopram, desmethylsertraline, and norfluoxetine.
  • SSRIs serotonin norepinephrine reuptake inhibitors
  • SNRIs selective serotonin norepinephrine reuptake inhibitors
  • venlafaxine venlafaxine
  • duloxetine venlafaxine
  • Venlafaxine hydrochloride is an antidepressant for oral administration. It is designated (R/S)-1-[2-(dimethylamino)-1-(4-methoxyphenyl)ethyl] cyclohexanol hydrochloride or ( ⁇ )-1-[(alpha)-[(dimethyl-amino)methyl]-p-methoxybenzyl] cyclohexanol hydrochloride.
  • Compressed tablets contain venlafaxine hydrochloride equivalent to 25 mg, 37.5 mg, 50 mg, 75 mg, or 100 mg venlafaxine. The recommended starting dose for venlafaxine is 75 mg/day, administered in two or three divided doses, taken with food.
  • the dose may be increased to 150 mg/day. If desirable, the dose can be further increased up to 225 mg/day. When increasing the dose, increments of up to 75 mg/day are typically made at intervals of no less than four days.
  • Venlafaxine has the following structure:
  • Structural analogs of venlafaxine are those compounds having the formula: as well as pharmaceutically acceptable salts thereof, wherein A is a moiety of the formula: where the dotted line represents optional unsaturation; R 1 is hydrogen or alkyl; R 2 is C 1-4 alkyl; R 4 is hydrogen, C 1-4 alkyl, formyl or alkanoyl; R 3 is hydrogen or C 1-4 alkyl; R 5 and R 6 are, independently, hydrogen, hydroxyl, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkanoyloxy, cyano, nitro, alkylmercapto, amino, C 1-4 alkylamino, dialkylamino, C 1-4 alkanamido, halo, trifluoromethyl or, taken together, methylenedioxy; and n is 0, 1, 2, 3 or 4.
  • Duloxetine has the following structure:
  • Structural analogs of duloxetine are those compounds described by the formula disclosed in U.S. Pat. No. 4,956,388, hereby incorporated by reference.
  • SSRI analogs are 4-(2-fluorophenyl)-6-methyl-2-piperazinothieno [2,3-d]pyrimidine, 1,2,3,4-tetrahydro-N-methyl-4-phenyl-1-naphthylamine hydrochloride; 1,2,3,4-tetrahydro-N-methyl-4-phenyl-(E)-1-naphthylamine hydrochloride; N,N-dimethyl-1-phenyl-1-phthalanpropylamine hydrochloride; gamma-(4-(trifluoromethyl)phenoxy)-benzenepropanamine hydrochloride; BP 554; CP 53261; O-desmethylvenlafaxine; WY 45,818; WY 45,881; N-(3-fluoropropyl)paroxetine; Lu 19005; and SNRIs described in PCT Publication No. WO04/004734.
  • Standard recommended dosages for exemplary SSRIs are provided in Table 4, below. Other standard dosages are provided, e.g., in the Merck Manual of Diagnosis & Therapy (17th Ed. MH Beers et al., Merck & Co.) and Physicians' Desk Reference 2003 (57 th Ed. Medical Economics Staff et al., Medical Economics Co., 2002). TABLE 4 Compound Standard Dose Fluoxetine 20-80 mg/day Sertraline 50-200 mg/day Paroxetine 20-50 mg/day Fluvoxamine 50-300 mg/day Citalopram 10-80 mg qid Escitalopram 10 mg qid Other Compounds
  • NSAIDs Non-Steroidal Anti-Inflammatory Drugs
  • the tetra-substituted pyrimidopyrimidines of the invention may be administered in conjunction with one or more of non-steroidal anti-inflammatory drugs (NSAIDs), such as naproxen sodium, diclofenac sodium, diclofenac potassium, aspirin, sulindac, diflunisal, piroxicam, indomethacin, ibuprofen, nabumetone, choline magnesium trisalicylate, sodium salicylate, salicylsalicylic acid (salsalate), fenoprofen, flurbiprofen, ketoprofen, meclofenamate sodium, meloxicam, oxaprozin, sulindac, and tolmetin.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • tetra-substituted pyrimidopyrimidines When tetra-substituted pyrimidopyrimidines is administered in combination with acetylsalicylic acid, it is desirable that the combination is effective in suppressing TNF ⁇ , IL-1, IL-2 or IFN- ⁇ in vitro. Accordingly, the combination of tetra-substituted pyrimidopyrimidine or tetra-substituted pyrimidopyrimidine analog in combination with acetylsalicylic acid and their analogs may be more effective in treating immunoinflammatory diseases, particulary those mediated by TNF ⁇ , IL-1, IL-2 or IFN- ⁇ than either agent alone.
  • Acetylsalicylic acid also known by trade name aspirin, is an acetyl derivative of salicylic acid and has the following structural formula.
  • Aspirin is useful in the relief of headache and muscle and joint aches. Aspirin is also effective in reducing fever, inflammation, and swelling and thus has been used for treatment of rheumatoid arthritis, rheumatic fever, and mild infection.
  • combination of a tetra-substituted pyrimidopyrimidine or analog thereof (e.g., dipyridamole) and acetylsalicylic acid (aspirin) or analog thereof can also be administered to enhance the treatment or prevention of the diseases mentioned above.
  • An NSAID may be administered in conjunction with any one of the combinations described in this application.
  • a patient suffering from immunoinflammatory disorder may be initially treated with a combination of a tetra-substituted pyrimidopyrimidine/SSRI or tetra-substituted pyrimidopyrimidine/glucocorticoid receptor modulator or tetra-substituted pyrimidopyrimidine/antihistamine or tetra-substituted pyrimidopyrimidine/ibudilast combination and then the patient may also be treated with an NSAID, such as acetylsalicylic acid, in conjunction with the combinations described above.
  • an NSAID such as acetylsalicylic acid
  • Dosage amounts of acetylsalicylic acid are known to those skilled in medical arts, and generally range from about 70 mg to about 350 mg per day.
  • a formulation containing dipyridamole and aspirin may contain 0-25 mg, 25-50 mg, 50-70 mg, 70-75 mg, 75-80 mg, 80-85 mg, 85-90 mg, 90-95 mg, 95-100 mg, 100-150 mg, 150-160 mg, 160-250 mg, 250-300 mg, 300-350 mg, or 350-1000 mg of aspirin.
  • NSAIDs e.g., acetylsalicylic acid
  • tetra-substituted pyrimidopyrimidines alone or by administering a combination of NSAIDs (e.g., acetylsalicylic acid) and tetra-substituted pyrimidopyrimidines.
  • the composition comprising tetra-substituted pyrimidopyrimidine and an NSAID has increased effectiveness, safety, tolerability, or satisfaction of treatment of a patient suffering from or at risk of suffering from immunoinflammatory disorder as compared to a composition having a tetra-substituted pyrimidopyrimidine or a NSAID alone.
  • the invention features methods, compositions, and kits employing an SSRI and a non-steroidal immunophilin-dependent immunosuppressant (NsIDI), optionally with a corticosteroid or other agent described herein.
  • NsIDI non-steroidal immunophilin-dependent immunosuppressant
  • the immune system uses cellular effectors, such as B-cells and T-cells, to target infectious microbes and abnormal cell types while leaving normal cells intact.
  • activated T-cells damage healthy tissues.
  • Calcineurin inhibitors e.g., cyclosporines, tacrolimus, pimecrolimus
  • rapamycin target many types of immunoregulatory cells, including T-cells, and suppress the immune response in organ transplantation and autoimmune disorders.
  • the NsIDI is cyclosporine, and is administered in an amount between 0.05 and 50 milligrams per kilogram per day (e.g., orally in an amount between 0.1 and 12 milligrams per kilogram per day).
  • the NsIDI is tacrolimus and is administered in an amount between 0.0001-20 milligrams per kilogram per day (e.g., orally in an amount between 0.01-0.2 milligrams per kilogram per day).
  • the NsIDI is rapamycin and is administered in an amount between 0.1-502 milligrams per day (e.g., at a single loading dose of 6 mg/day, followed by a 2 mg/day maintenance dose).
  • the NsIDI is everolimus, administered at a dosage of 0.75-8 mg/day.
  • the NsIDI is pimecrolimus, administered in an amount between 0.1 and 200 milligrams per day (e.g., as a 1% cream/twice a day to treat atopic dermatitis or 60 mg a day for the treatment of psoriasis), or the NsIDI is a calcineurin-binding peptide administered in an amount and frequency sufficient to treat the patient. Two or more NsIDIs can be administered contemporaneously.
  • the cyclosporines are fungal metabolites that comprise a class of cyclic oligopeptides that act as immunosuppressants.
  • Cyclosporine A is a hydrophobic cyclic polypeptide consisting of eleven amino acids. It binds and forms a complex with the intracellular receptor cyclophilin. The cyclosporine/cyclophilin complex binds to and inhibits calcineurin, a Ca 2+ -calmodulin-dependent serine-threonine-specific protein phosphatase. Calcineurin mediates signal transduction events required for T-cell activation (reviewed in Schreiber et al., Cell 70:365-368, 1991). Cyclosporines and their functional and structural analogs suppress the T cell-dependent immune response by inhibiting antigen-triggered signal transduction. This inhibition decreases the expression of proinflammatory cytokines, such as IL-2.
  • Cyclosporine A is a commercially available under the trade name NEORAL from Novartis.
  • Cyclosporine A structural and functional analogs include cyclosporines having one or more fluorinated amino acids (described, e.g., in U.S. Pat. No. 5,227,467); cyclosporines having modified amino acids (described, e.g., in U.S. Pat. Nos. 5,122,511 and 4,798,823); and deuterated cyclosporines, such as ISAtx247 (described in U.S. Patent Application Publication No.
  • Cyclosporine analogs include, but are not limited to, D-Sar ( ⁇ -SMe) 3 Val 2 -DH-Cs (209-825), Allo-Thr-2-Cs, Norvaline-2-Cs, D-Ala(3-acetylamino)-8-Cs, Thr-2-Cs, and D-MeSer-3-Cs, D-Ser(O—CH 2 CH 2 —OH)-8-Cs, and D-Ser-8-Cs, which are described in Cruz et al. (Antimicrob.
  • Cyclosporines are highly hydrophobic and readily precipitate in the presence of water (e.g. on contact with body fluids). Methods of providing cyclosporine formulations with improved bioavailability are described in U.S. Pat. Nos. 4,388,307, 6,468,968, 5,051,402, 5,342,625, 5,977,066, and 6,022,852. Cyclosporine microemulsion compositions are described in U.S. Pat. Nos. 5,866,159, 5,916,589, 5,962,014, 5,962,017, 6,007,840, and 6,024,978.
  • Cyclosporines can be administered either intravenously or orally, but oral administration is preferred.
  • an intravenous cyclosporine A is usually provided in an ethanol-polyoxyethylated castor oil vehicle that must be diluted prior to administration.
  • Cyclosporine A may be provided, e.g., as a microemulsion in a 25 mg or 100 mg tablets, or in a 100 mg/ml oral solution (NEORAL).
  • patient dosage of an oral cyclosporine varies according to the patient's condition, but some standard recommended dosages are provided herein.
  • Patients undergoing organ transplant typically receive an initial dose of oral cyclosporine A in amounts between 12 and 15 mg/kg/day. Dosage is then gradually decreased by 5% per week until a 7-12 mg/kg/day maintenance dose is reached.
  • For intravenous administration 2-6 mg/kg/day is preferred for most patients.
  • dosage amounts from 6-8 mg/kg/day are generally given.
  • dosage amounts from 2.2-6.0 mg/kg/day are generally given.
  • dosage amounts from 0.5-4 mg/kg/day are typical.
  • a suggested dosing schedule is shown in Table 5.
  • Other useful dosages include 0.5-5 mg/kg/day, 5-10 mg/kg/day, 10-15 mg/kg/day, 15-20 mg/kg/day, or 20-25 mg/kg/day.
  • cyclosporines are administered in combination with other immunosuppressive agents, such as glucocorticoids.
  • Tacrolimus is an immunosuppressive agent that targets T cell intracellular signal transduction pathways. Tacrolimus binds to an intracellular protein FK506 binding protein (FKBP-12) that is not structurally related to cyclophilin (Harding et al. Nature 341:758-7601, 1989; Siekienka et al. Nature 341:755-757, 1989; and Soltoff et al., J. Biol. Chem. 267:17472-17477, 1992).
  • FKBP/FK506 complex binds to calcineurin and inhibits calcineurin's phosphatase activity.
  • NFAT nuclear factor of activated T cells
  • cytokine e.g., IL-2, gamma interferon
  • Tacrolimus is a macrolide antibiotic that is produced by Streptomyces tsukubaensis . It suppresses the immune system and prolongs the survival of transplanted organs. It is currently available in oral and injectable formulations.
  • Tacrolimus capsules contain 0.5 mg, 1 mg, or 5 mg of anhydrous tacrolimus within a gelatin capsule shell.
  • the injectable formulation contains 5 mg anhydrous tacrolimus in castor oil and alcohol that is diluted with 0.9% sodium chloride or 5% dextrose prior to injection. While oral administration is preferred, patients unable to take oral capsules may receive injectable tacrolimus.
  • the initial dose should be administered no sooner than six hours after transplant by continuous intravenous infusion.
  • Tacrolimus and tacrolimus analogs are described by Tanaka et al., (J. Am. Chem. Soc., 109:5031, 1987) and in U.S. Pat. Nos. 4,894,366, 4,929,611, and 4,956,352.
  • FK506-related compounds including FR-900520, FR-900523, and FR-900525, are described in U.S. Pat. No. 5,254,562; O-aryl, O-alkyl, O-alkenyl, and O-alkynylmacrolides are described in U.S. Pat. Nos. 5,250,678, 532,248, 5,693,648; amino O-aryl macrolides are described in U.S. Pat. No.
  • alkylidene macrolides are described in U.S. Pat. No. 5,284,840; N-heteroaryl, N-alkylheteroaryl, N-alkenylheteroaryl, and N-alkynylheteroaryl macrolides are described in U.S. Pat. No. 5,208,241; aminomacrolides and derivatives thereof are described in U.S. Pat. No. 5,208,228; fluoromacrolides are described in U.S. Pat. No. 5,189,042; amino O-alkyl, O-alkenyl, and O-alkynylmacrolides are described in U.S. Pat. No. 5,162,334; and halomacrolides are described in U.S. Pat. No. 5,143,918.
  • While suggested dosages will vary with a patient's condition, standard recommended dosages are provided below.
  • patients diagnosed as having Crohn's disease or ulcerative colitis are administered 0.1-0.2 mg/kg/day oral tacrolimus.
  • Patients having a transplanted organ typically receive doses of 0.1-0.2 mg/kg/day of oral tacrolimus.
  • Patients being treated for rheumatoid arthritis typically receive 1-3 mg/day oral tacrolimus.
  • 0.01-0.15 mg/kg/day of oral tacrolimus is administered to a patient.
  • Atopic dermatitis can be treated twice a day by applying a cream having 0.03-0.1% tacrolimus to the affected area.
  • tacrolimus capsules typically receive the first dose no sooner than six hours after transplant, or eight to twelve hours after intravenous tacrolimus infusion was discontinued.
  • Other suggested tacrolimus dosages include 0.005-0.01 mg/kg/day, 0.01-0.03 mg/kg/day, 0.03-0.05 mg/kg/day, 0.05-0.07 mg/kg/day, 0.07-0.10 mg/kg/day, 0.10-0.25 mg/kg/day, or 0.25-0.5 mg/kg/day.
  • Tacrolimus is extensively metabolized by the mixed-function oxidase system, in particular, by the cytochrome P-450 system.
  • the primary mechanism of metabolism is demethylation and hydroxylation. While various tacrolimus metabolites are likely to exhibit immunosuppressive biological activity, the 13-demethyl metabolite is reported to have the same activity as tacrolimus.
  • Pimecrolimus is the 33-epi-chloro derivative of the macrolactam ascomyin. Pimecrolimus structural and functional analogs are described in U.S. Pat. No. 6,384,073. Pimecrolimus is particularly useful for the treatment of atopic dermatitis. Pimecrolimus is currently available as a 1% cream. Suggested dosing schedule for pimecrolimus is shown at Table 5. While individual dosing will vary with the patient's condition, some standard recommended dosages are provided below. Oral pimecrolimus can be given for the treatment of psoriasis or rheumatoid arthritis in amounts of 40-60 mg/day.
  • pimecrolimus For the treatment of Crohn's disease or ulcerative colitis amounts of 80-160 mg/day pimecrolimus can be given. Patients having an organ transplant can be administered 160-240 mg/day of pimecrolimus. Patients diagnosed as having systemic lupus erythamatosus can be administered 40-120 mg/day of pimecrolimus. Other useful dosages of pimecrolimus include 0.5-5 mg/day, 5-10 mg/day, 10-30 mg/day, 40-80 mg/day, 80-120 mg/day, or even 120-200 mg/day.
  • Rapamycin is a cyclic lactone produced by Streptomyces hygroscopicus . Rapamycin is an immunosuppressive agent that inhibits T cell activation and proliferation. Like cyclosporines and tacrolimus, rapamycin forms a complex with the immunophilin FKBP-12, but the rapamycin-FKBP-12 complex does not inhibit calcineurin phosphatase activity. The rapamycin immunophilin complex binds to and inhibits the mammalian kinase target of rapamycin (mTOR). mTOR is a kinase that is required for cell-cycle progression. Inhibition of mTOR kinase activity blocks T cell activation and proinflammatory cytokine secretion.
  • mTOR mammalian kinase target of rapamycin
  • Rapamycin structural and functional analogs include mono- and diacylated rapamycin derivatives (U.S. Pat. No. 4,316,885); rapamycin water-soluble prodrugs (U.S. Pat. No. 4,650,803); carboxylic acid esters (PCT Publication No. WO 92/05179); carbamates (U.S. Pat. No. 5,118,678); amide esters (U.S. Pat. No. 5,118,678); biotin esters (U.S. Pat. No. 5,504,091); fluorinated esters (U.S. Pat. No. 5,100,883); acetals (U.S. Pat. No. 5,151,413); silyl ethers (U.S.
  • Rapamycin is currently available for oral administration in liquid and tablet formulations.
  • RAPAMUNE liquid contains 1 mg/mL rapamycin that is diluted in water or orange juice prior to administration. Tablets containing 1 or 2 mg of rapamycin are also available. Rapamycin is preferably given once daily as soon as possible after transplantation. It is absorbed rapidly and completely after oral administration.
  • patient dosage of rapamycin varies according to the patient's condition, but some standard recommended dosages are provided below.
  • the initial loading dose for rapamycin is 6 mg. Subsequent maintenance doses of 0.5-2 mg/day are typical.
  • a loading dose of 3 mg, 5 mg, 10 mg, 15 mg, 20 mg, or 25 mg can be used with a 1 mg, 3 mg, 5 mg, 7 mg, or 10 mg per day maintenance dose.
  • rapamycin dosages are typically adjusted based on body surface area; generally a 3 mg/m 2 /day loading dose and a 1 mg/m 2 /day maintenance dose is used.
  • Peptides, peptide mimetics, peptide fragments, either natural, synthetic or chemically modified, that impair the calcineurin-mediated dephosphorylation and nuclear translocation of NFAT are suitable for use in practicing the invention.
  • Examples of peptides that act as calcineurin inhibitors by inhibiting the NFAT activation and the NFAT transcription factor are described, e.g., by Aramburu et al., Science 285:2129-2133, 1999) and Aramburu et al., Mol. Cell 1:627-637, 1998).
  • these agents are useful in the methods of the invention.
  • the invention features methods for suppressing secretion of proinflammatory cytokines as a means for treating an immunoinflammatory disorder, proliferative skin disease, organ transplant rejection, or graft versus host disease.
  • Therapy according to the invention may be performed alone or in conjunction with another therapy and may be provided at home, the doctor's office, a clinic, a hospital's outpatient department, or a hospital.
  • the duration of the therapy depends on the type of disease or disorder being treated, the age and condition of the patient, the stage and type of the patient's disease, and how the patient responds to the treatment. Additionally, a person having a greater risk of developing an inflammatory disease (e.g., a person who is undergoing age-related hormonal changes) may receive treatment to inhibit or delay the onset of symptoms.
  • the compounds are administered within 10 days of each other, within five days of each other, within twenty-four hours of each other, or simultaneously.
  • the compounds may be formulated together as a single composition, or may be formulated and administered separately.
  • One or both compounds may be administered in a low dosage or in a high dosage, each of which is defined herein.
  • NSAID e.g., naproxen sodium, diclofenac sodium, diclofenac potassium, aspirin, sulindac, diflunisal, piroxicam, indomethacin, ibuprofen, nabumetone, choline magnesium trisalicylate, sodium salicylate, salicylsalicylic acid, fenoprofen, flurbiprofen, ketoprofen, meclofenamate sodium, meloxicam, oxaprozin, sulindac, and tolmetin), COX-2 inhibitor (e.g., rofecoxib, celecoxib, valdecoxib, and lumiracoxib), glucocorticoid receptor modulator, or DMARD.
  • COX-2 inhibitor e.g., rofecoxib, celecoxib, valdecoxib, and lumiracoxib
  • glucocorticoid receptor modulator e.g.,
  • Combination therapies of the invention are especially useful for the treatment of immunoinflammatory disorders in combination with other agents—either biologics or small molecules—that modulate the immune response to positively affect disease.
  • agents include those that deplete key inflammatory cells, influence cell adhesion, or influence cytokines involved in immune response.
  • This last category includes both agents that mimic or increase the action of anti-inflammatory cytokines such as IL-10, as well as agents inhibit the activity of pro-inflammatory cytokines such as IL-6, IL-1, IL-2, IL-112, IL-15 or TNF ⁇ .
  • Agents that inhibit TNF ⁇ include etanercept, adelimumab, infliximab, and CDP-870.
  • the combination therapy reduces the production of cytokines, etanercept or infliximab act on the remaining fraction of inflammatory cytokines, providing enhanced treatment.
  • Small molecule immunodulators include, e.g., p38 MAP kinase inhibitors such as VX 702, SCIO 469, doramapimod, RO 30201195, SCIO 323, TACE inhibitors such as DPC 333, ICE inhibitors such as pranalcasan, and IMPDH inhibitors such as mycophenylate and merimepodib.
  • each component of the combination can be controlled independently.
  • one compound may be administered three times per day, while the second compound may be administered once per day.
  • Combination therapy may be given in on-and-off cycles that include rest periods so that the patient's body has a chance to recover from any as yet unforeseen side effects.
  • the compounds may also be formulated together such that one administration delivers both compounds.
  • the compound in question may be administered orally in the form of tablets, capsules, elixirs or syrups, or rectally in the form of suppositories.
  • Parenteral administration of a compound is suitably performed, for example, in the form of saline solutions or with the compound incorporated into liposomes.
  • a solubilizer such as ethanol can be applied.
  • the methods, compositions, and kits of the invention are more effective than other methods, compositions, and kits.
  • “more effective” is meant that a method, composition, or kit exhibits greater efficacy, is less toxic, safer, more convenient, better tolerated, or less expensive, or provides more treatment satisfaction than another method, composition, or kit with which it is being compared.
  • the methods, compositions, and kits of the invention are used for the treatment of chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • one or more agents typically used to treat COPD may be used as a substitute for or in addition to a corticosteroid in the methods, compositions, and kits of the invention.
  • Such agents include xanthines (e.g., theophylline), anticholinergic compounds (e.g., ipratropium, tiotropium), biologics, small molecule immunomodulators, and beta receptor agonists/bronchdilators (e.g., ibuterol sulfate, bitolterol mesylate, epinephrine, formoterol fumarate, isoproteronol, levalbuterol hydrochloride, metaproterenol sulfate, pirbuterol scetate, salmeterol xinafoate, and terbutaline).
  • xanthines e.g., theophylline
  • anticholinergic compounds e.g., ipratropium, tiotropium
  • biologics e.g., small molecule immunomodulators
  • beta receptor agonists/bronchdilators e.g., ibuterol sul
  • the methods, compositions, and kits of the invention may be used for the treatment of psoriasis.
  • one or more antipsoriatic agents typically used to treat psoriasis may be used as a substitute for or in addition to a corticosteroid in the methods, compositions, and kits of the invention.
  • Such agents include biologics (e.g., alefacept, inflixamab, adelimumab, efalizumab, etanercept, and CDP-870), small molecule immunomodulators (e.g., VX 702, SCIO 469, doramapimod, RO 30201195, SCIO 323, DPC 333, pranalcasan, mycophenylate, and merimepodib), non-steroidal immunophilin-dependent immunosuppressants (e.g., cyclosporine, tacrolimus, pimecrolimus, and ISAtx247), vitamin D analogs (e.g., calcipotriene, calcipotriol), psoralens (e.g., methoxsalen), retinoids (e.g., acitretin, tazoretene), DMARDs (e.g., methotrexate), and anthralin.
  • biologics e.g.,
  • the methods, compositions, and kits of the invention may be used for the treatment of inflammatory bowel disease.
  • one or more agents typically used to treat inflammatory bowel disease may be used as a substitute for or in addition to a corticosteroid in the methods, compositions, and kits of the invention.
  • Such agents include biologics (e.g., inflixamab, adelimumab, and CDP-870), small molecule immunomodulators (e.g., VX 702, SCIO 469, doramapimod, RO 30201195, SCIO 323, DPC 333, pranalcasan, mycophenylate, and merimepodib), non-steroidal immunophilin-dependent immunosuppressants (e.g., cyclosporine, tacrolimus, pimecrolimus, and ISAtx247), 5-amino salicylic acid (e.g., mesalamine, sulfasalazine, balsalazide disodium, and olsalazine sodium), DMARDs (e.g., methotrexate and azathioprine) and alosetron.
  • biologics e.g., inflixamab, adelimumab, and CDP-870
  • small molecule immunomodulators
  • the methods, compositions, and kits of the invention may be used for the treatment of rheumatoid arthritis. If desired, one or more agents typically used to treat rheumatoid arthritis may be used as a substitute for or in addition to a corticosteroid in the methods, compositions, and kits of the invention.
  • Such agents include NSAIDs (e.g., naproxen sodium, diclofenac sodium, diclofenac potassium, aspirin, sulindac, diflunisal, piroxicam, indomethacin, ibuprofen, nabumetone, choline magnesium trisalicylate, sodium salicylate, salicylsalicylic acid (salsalate), fenoprofen, flurbiprofen, ketoprofen, meclofenamate sodium, meloxicam, oxaprozin, sulindac, and tolmetin), COX-2 inhibitors (e.g., rofecoxib, celecoxib, valdecoxib, and lumiracoxib), biologics (e.g., inflixamab, adelimumab, etanercept, CDP-870, rituximab, and atlizumab), small molecule immunomodulators (e.g., VX
  • the methods, compositions, and kits of the invention may be used for the treatment of asthma.
  • agents typically used to treat asthma may be used as a substitute for or in addition to a corticosteroid in the methods, compositions, and kits of the invention.
  • agents include beta 2 agonists/bronchodilators/leukotriene modifiers (e.g., zafirlukast, montelukast, and zileuton), biologics (e.g., omalizumab), small molecule immunomodulators, anticholinergic compounds, xanthines, ephedrine, guaifenesin, cromolyn sodium, nedocromil sodium, and potassium iodide.
  • the invention features the combination of a tetra-substituted pyrimidopyrimidine and any of the foregoing agents, and methods of treating asthma therewith.
  • the administration of a combination of the invention may be by any suitable means that results in suppression of proinflammatory cytokine levels at the target region.
  • the compound may be contained in any appropriate amount in any suitable carrier substance, and is generally present in an amount of 1-95% by weight of the total weight of the composition.
  • the composition may be provided in a dosage form that is suitable for the oral, parenteral (e.g., intravenously, intramuscularly), rectal, cutaneous, nasal, vaginal, inhalant, skin (patch), or ocular administration route.
  • the composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, or aerosols.
  • the compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy, 20th edition, 2000, ed. A. R. Gennaro, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
  • each compound of the combination may be formulated in a variety of ways that are known in the art.
  • the first and second agents may be formulated together or separately.
  • the first and second agents are formulated together for the simultaneous or near simultaneous administration of the agents.
  • Such co-formulated compositions can include, for example, the tetra-substituted pyrimidopyrimidine and the antihistamine formulated together in the same pill, capsule, liquid, etc.
  • tetra-substituted pyrimidopyrimidine/antihistamine the formulation technology employed is also useful for the formulation of the individual agents of the combination, as well as other combinations of the invention (e.g., a tetra-substituted pyrimidopyrimidine/SSRI or tetra-substituted pyrimidopyrimidine/glucocorticoid receptor modulator combination).
  • a tetra-substituted pyrimidopyrimidine/SSRI or tetra-substituted pyrimidopyrimidine/glucocorticoid receptor modulator combination By using different formulation strategies for different agents, the pharmacokinetic profiles for each agent can be suitably matched.
  • kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc.
  • the kit can include optional components that aid in the administration of the unit dose to patients, such as vials for reconstituting powder forms, syringes for injection, customized IV delivery systems, inhalers, etc.
  • the unit dose kit can contain instructions for preparation and administration of the compositions.
  • the kit may be manufactured as a single use unit dose for one patient, multiple uses for a particular patient (at a constant dose or in which the individual compounds may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple patients (“bulk packaging”).
  • the kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.
  • the tetra-substituted pyrimidopyrimidine/antihistamine combination in which one or both of the active agents is formulated for controlled and/or extended release is useful where the tetra-substituted pyrimidopyrimidine or the antihistamine, has (i) a narrow therapeutic index (e.g., the difference between the plasma concentration leading to harmful side effects or toxic reactions and the plasma concentration leading to a therapeutic effect is small; generally, the therapeutic index, TI, is defined as the ratio of median lethal dose (LD 50 ) to median effective dose (ED 50 )); (ii) a narrow absorption window in the gastro-intestinal tract; (iii) a short biological half-life; or (iv) the pharmacokinetic profile of each component must be modified to maximize the contribution of each agent, when used together, to an amount of that is therapeutically effective for cytokine suppression.
  • a narrow therapeutic index e.g., the difference between the plasma concentration leading to harmful side effects or toxic reactions and the
  • a sustained release formulation may be used to avoid frequent dosing that may be required in order to sustain the plasma levels of both agents at a therapeutic level.
  • half-life and mean residency times from 10 to 20 hours for one or both agents of the combination of the invention are observed.
  • controlled release can be obtained by the appropriate selection of formulation parameters and ingredients (e.g., appropriate controlled release compositions and coatings). Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, nanoparticles, patches, and liposomes.
  • the release mechanism can be controlled such that the tetra-substituted pyrimidopyrimidine and/or companion compounds (e.g., antihistamine, corticosteroid, rolipram, ibudilast, tricyclic and tetracyclic antidepressants, SSRI, non-steroidal anti-inflammatory drugs, non-steroidal immunophilin-dependent immunosuppressants and analogs thereof, as described herein) are released at period intervals, the release could be simultaneous, or a delayed release of one of the agents of the combination can be affected, when the early release of one particular agent is preferred over the other.
  • companion compounds e.g., antihistamine, corticosteroid, rolipram, ibudilast, tricyclic and tetracyclic antidepressants, SSRI, non-steroidal anti-inflammatory drugs, non-steroidal immunophilin-dependent immunosuppressants and analogs thereof, as described herein
  • the release mechanism of additional compounds can also be controlled like that of the tetra-substituted pyrimidopyrimidine and/or companion compounds (e.g., antihistamine, corticosteroid, rolipram, ibudilast, tricyclic and tetracyclic antidepressants, SSRI, non-steroidal immunophilin-dependent immunosuppressants and analogs thereof, as described herein) and are similarly released at period intervals, the release could be simultaneous, or a delayed release of one of the agents of the combination can be affected, when the early release of one particular agent is preferred over the other.
  • additional compounds such as an NSAID, COX-2 inhibitor, biologic, small molecule immunomodulator, DMARD, xanthine, anticholinergic compound, beta receptor agonist, bronchodilator, non-steroidal immunophilin-dependent immunomodulators, vitamin D analog, psoralen, retinoid, and 5-amino salicylic acid
  • the release mechanism of additional compounds can
  • dipyridamole pellets can be processed together with the acetylsalicylic acid to form corresponding drug preparations. If it is intended that the acetylsalicylic acid should be released first, the dipyridamolepellets may be coated with a coating which delays the release of this active substance and the cores containing the acetylsalicylic acid coated with a coating that is soluble in gastric juices. In the case of dipyridamole pellets with a controlled release of the active substance it is particularly advantageous to use pellets prepared according to the instructions given in U.S. Pat. No. 4,367,217.
  • Controlled and/or extended release formulations may include a degradable or nondegradable polymer, hydrogel, organogel, or other physical construct that modifies the bioabsorption, half-life or biodegradation of the agent.
  • the controlled and/or extended release formulation can be a material that is painted or otherwise applied onto the afflicted site, either internally or externally.
  • the invention provides a biodegradable bolus or implant that is surgically inserted at or near a site of interest (for example, proximal to an arthritic joint).
  • the controlled release formulation implant can be inserted into an organ, such as in the lower intestine for the treatment inflammatory bowel disease.
  • Hydrogels can be used in controlled release formulations for any one of the combinations of this invention.
  • Such polymers are formed from macromers with a polymerizable, non-degradable, region that is separated by at least one degradable region.
  • the water soluble, non-degradable, region can form the central core of the macromer and have at least two degradable regions which are attached to the core, such that upon degradation, the non-degradable regions (in particular a polymerized gel) are separated, as described in U.S. Pat. No. 5,626,863.
  • Hydrogels can include acrylates, which can be readily polymerized by several initiating systems such as eosin dye, ultraviolet or visible light.
  • Hydrogels can also include polyethylene glycols (PEGs), which are highly hydrophilic and biocompatible. Hydrogels can also include oligoglycolic acid, which is a poly( ⁇ -hydroxy acid) that can be readily degraded by hydrolysis of the ester linkage into glycolic acid, a nontoxic metabolite. Other chain extensions can include polylactic acid, polycaprolactone, polyorthoesters, polyanhydrides or polypeptides. The entire network can be gelled into a biodegradable network that can be used to entrap and homogeneously disperse various combinations of the invention for delivery at a controlled rate.
  • PEGs polyethylene glycols
  • oligoglycolic acid which is a poly( ⁇ -hydroxy acid) that can be readily degraded by hydrolysis of the ester linkage into glycolic acid, a nontoxic metabolite.
  • Other chain extensions can include polylactic acid, polycaprolactone, polyorthoesters, polyanhydrides or polypeptides.
  • Chitosan and mixtures of chitosan with carboxymethylcellulose sodium have been used as vehicles for the sustained release of drugs, as described by Inouye et al., Drug Design and Delivery 1: 297-305, 1987. Mixtures of these compounds and agents of the any one of the combinations described above, when compressed under 200 kg/cm 2 , form a tablet from which the active agent is slowly released upon administration to a subject.
  • the release profile can be changed by varying the ratios of chitosan, CMC-Na, and active agent(s).
  • the tablets can also contain other additives, including lactose, CaHPO 4 dihydrate, sucrose, crystalline cellulose, or croscarmellose sodium. Several examples are given in Table 6.
  • Baichwal in U.S. Pat. No. 6,245,356, describes a sustained release oral solid dosage forms that includes agglomerated particles of a therapeutically active medicament in amorphous form, a gelling agent, an ionizable gel strength enhancing agent and an inert diluent.
  • the gelling agent can be a mixture of a xanthan gum and a locust bean gum capable of cross-linking with the xanthan gum when the gums are exposed to an environmental fluid.
  • the ionizable gel enhancing agent acts to enhance the strength of cross-linking between the xanthan gum and the locust bean gum and thereby prolonging the release of the medicament component of the formulation.
  • acceptable gelling agents include those gelling agents well known in the art. Examples include naturally occurring or modified naturally occurring gums such as alginates, carrageenan, pectin, guar gum, modified starch, hydroxypropylmethylcellulose, methylcellulose, and other cellulosic materials or polymers, such as, for example, sodium carboxymethylcellulose and hydroxypropyl cellulose, and mixtures of the foregoing.
  • Baichwal and Staniforth in U.S. Pat. No. 5,135,757 describe a free-flowing slow release granulation for use as a pharmaceutical excipient that includes from about 20 to about 70 percent or more by weight of a hydrophilic material that includes a heteropolysaccharide (such as, for example, xanthan gum or a derivative thereof) and a polysaccharide material capable of cross-linking the heteropolysaccharide (such as, for example, galactomannans, and most preferably locust bean gum) in the presence of aqueous solutions, and from about 30 to about 80 percent by weight of an inert pharmaceutical filler (such as, for example, lactose, dextrose, sucrose, sorbitol, xylitol, fructose or mixtures thereof).
  • an inert pharmaceutical filler such as, for example, lactose, dextrose, sucrose, sorbitol, xylitol, fructose or
  • the mixture After mixing the excipient with a combination, or combination agent, of the invention, the mixture is directly compressed into solid dosage forms such as tablets.
  • the tablets thus formed slowly release the medicament when ingested and exposed to gastric fluids.
  • a slow release profile can be attained.
  • Shell in U.S. Pat. No. 5,007,790, describes sustained-release oral drug-dosage forms that release a drug in solution at a rate controlled by the solubility of the drug.
  • the dosage form comprises a tablet or capsule that includes a plurality of particles of a dispersion of a limited solubility drug (such as, for example, prednisolone, paroxetine, or any other agent of anyone or all of the combination of the present invention) in a hydrophilic, water-swellable, crosslinked polymer that maintains its physical integrity over the dosing lifetime but thereafter rapidly dissolves.
  • a limited solubility drug such as, for example, prednisolone, paroxetine, or any other agent of anyone or all of the combination of the present invention
  • the particles swell to promote gastric retention and permit the gastric fluid to penetrate the particles, dissolve drug and leach it from the particles, assuring that drug reaches the stomach in the solution state which is less injurious to the stomach than solid-state drug.
  • the programmed eventual dissolution of the polymer depends upon the nature of the polymer and the degree of crosslinking.
  • the polymer is nonfibrillar and substantially water soluble in its uncrosslinked state, and the degree of crosslinking is sufficient to enable the polymer to remain insoluble for the desired time period, normally at least from about 4 hours to 8 hours up to 12 hours, with the choice depending upon the drug incorporated and the medical treatment involved.
  • crosslinked polymers examples include gelatin, albumin, sodium alginate, carboxymethyl cellulose, polyvinyl alcohol, and chitin.
  • crosslinking may be achieved by thermal or radiation treatment or through the use of crosslinking agents such as aldehydes, polyamino acids, metal ions and the like.
  • Silicone microspheres for pH-controlled gastrointestinal drug delivery that are useful in the formulation of anyone or all of the combinations of the invention have been described by Carelli et al., Int. J. Pharmaceutics 179: 73-83, 1999.
  • the microspheres so described are pH-sensitive semi-interpenetrating polymer hydrogels made of varying proportions of poly(methacrylic acid-co-methylmethacrylate) (Eudragit L100 or Eudragit S100) and crosslinked polyethylene glycol 8000 that are encapsulated into silicone microspheres in the 500 to 1000 ⁇ m size range.
  • Slow-release formulations may include a coating that is not readily water-soluble but is slowly attacked and removed by water, or through which water can slowly permeate.
  • a combination of the invention can be spray-coated with a solution of a binder under continuously fluidizing conditions, such as describe by Kitamori et al., U.S. Pat. No. 4,036,948.
  • Water-soluble binders include pregelatinized starch (e.g., pregelatinized corn starch, pregelatinized white potato starch), pregelatinized modified starch, water-soluble celluloses (e.g.
  • hydroxypropyl-cellulose hydroxymethyl-cellulose, hydroxypropylmethyl-cellulose, carboxymethyl-cellulose
  • polyvinylpyrrolidone polyvinyl alcohol
  • dextrin polyvinyl alcohol
  • gum arabicum and gelatin organic solvent-soluble binders, such as cellulose derivatives (e.g., cellulose acetate phthalate, hydroxypropylmethyl-cellulose phthalate, ethylcellulose).
  • Combinations of the invention, or a component thereof, with sustained release properties can also be formulated by spray drying techniques.
  • prednisolone was encapsulated in methyacrylate microparticles (Eudragit RS) using a Mini Spray Dryer, model 190 (Buchi, Laboratoriumtechnik AG, Flawil, Germany).
  • Optimal conditions for microparticle formation were found to be a feed (pump) rate of 0.5 mL/min of a solution containing 50 mg prednisolone in 10 mL of acetonitrile, a flow rate of nebulized air of 600 L/hr, dry air temperature heating at 80° C., and a flow rate of aspirated drying air of 28 m 3 /hr.
  • sustained release combinations can be prepared by microencapsulation of combination agent particles in membranes which act as microdialysis cells.
  • gastric fluid permeates the microcapsule walls and swells the microcapsule, allowing the active agent(s) to dialyze out (see, for example, Tsuei et al., U.S. Pat. No. 5,589,194).
  • One commercially available sustained-release system of this kind consists of microcapsules having membranes of acacia gum/gelatine/ethyl alcohol. This product is available from Eurand Limited (France) under the trade name DiffucapsTM. Microcapsules so formulated might be carried in a conventional gelatine capsule or tabletted.
  • Extended- and/or controlled-release formulations of combinations of this invention such as, both tetra-substituted pyrimidopyrimidine and antihistamine or SSRI are known.
  • a system for the controlled release of an active substance which is an tetra-substituted pyrimidopyrimidine such as dipyridamole comprising (a) a deposit-core comprising an effective amount of the active substance and having defined geometric form, and (b) a support-platform applied to the deposit-core, wherein the deposit-core contains at least the active substance, and at least one member selected from the group consisting of (1) a polymeric material which swells on contact with water or aqueous liquids and a gellable polymeric material wherein the ratio of the swellable polymeric material to the gellable polymeric material is in the range 1:9 to 9:1, and (2) a single polymeric material having both swelling and gelling properties, and wherein the support-platform is an elastic support, applied to said deposit-core so that it partially covers the surface of the deposit-core and follows changes due to hydration of the deposit-core and is
  • the support-platform may comprise polymers such as hydroxypropylmethylcellulose, plasticizers such as a glyceride, binders such as polyvinylpyrrolidone, hydrophilic agents such as lactose and silica, and/or hydrophobic agents such as magnesium stearate and glycerides.
  • the polymer(s) typically make up 30 to 90% by weight of the support-platform, for example about 35 to 40%.
  • Plasticizer may make up at least 2% by weight of the support-platform, for example about 15 to 20%.
  • Binder(s), hydrophilic agent(s) and hydrophobic agent(s) typically total up to about 50% by weight of the support-platform, for example about 40 to 50%.
  • a controlled-release formulation of budesonide (3 mg capsules) for the treatment of inflammatory bowel disease is available from AstraZeneca (sold as EntocortTM).
  • a sustained-release formulation useful for corticosteroids is also described in U.S. Pat. No. 5,792,476, where the formulation includes 2.5-7 mg of a glucocorticoid as active substance with a regulated sustained-release such that at least 90% by weight of the glucocorticoid is released during a period of about 40-80 min, starting about 1-3 h after the entry of said glucocorticoid into the small intestine of the patient.
  • the active substance i.e.
  • the glucocorticoid such as prednisolone or prednisone
  • the glucocorticoid is micronised, suitably mixed with known diluents, such as starch and lactose, and granulated with PVP (polyvinylpyrrolidone).
  • PVP polyvinylpyrrolidone
  • the granulate is laminated with a sustained release inner layer resistant to a pH of 6.8 and a sustained release outer layer resistant to a pH of 1.0.
  • the inner layer is made of Eudragit®RL (copolymer of acrylic and methacrylic esters with a low content of quaternary ammonium groups) and the outer layer is made of Eudragi®L (anionic polymer synthesized from methacrylic acid and methacrylic acid methyl ester).
  • a bilayer tablet can be formulated for any one of the combinations described herein in which different custom granulations are made for each agent of the combination and the two agents are compressed on a bi-layer press to form a single tablet.
  • 12.5 mg, 25 mg, 37.5 mg, or 50 mg of paroxetine formulated for a controlled release that results in a paroxetine t 1/2 of 15 to 20 hours may be combined in the same tablet with 3 mg of predinisolone, which is formulated such that the t 1/2 approximates that of paroxetine.
  • Examples of paroxetine extended-release formulations, including those used in bilayer tablets, can be found in U.S. Pat. No. 6,548,084.
  • Cyclodextrins are cyclic polysaccharides containing naturally occurring D(+)-glucopyranose units in an ⁇ -(1,4) linkage.
  • Alpha-, beta- and gamma-cyclodextrins which contain, respectively, six, seven or eight glucopyranose units, are most commonly used and suitable examples are described in WO91/11172, WO94/02518 and WO98/55148.
  • the cyclic nature of a cyclodextrin forms a torus or donut-like shape having an inner apolar or hydrophobic cavity, the secondary hydroxyl groups situated on one side of the cyclodextrin torus and the primary hydroxyl groups situated on the other.
  • the side on which the secondary hydroxyl groups are located has a wider diameter than the side on which the primary hydroxyl groups are located.
  • the hydrophobic nature of the cyclodextrin inner cavity allows for the inclusion of a variety of compounds.
  • Cyclodextrins have been used as a delivery vehicle of various therapeutic compounds by forming inclusion complexes with various drugs that can fit into the hydrophobic cavity of the cyclodextrin or by forming non-covalent association complexes with other biologically active molecules.
  • U.S. Pat. No. 4,727,064 describes pharmaceutical preparations consisting of a drug with substantially low water solubility and an amorphous, water-soluble cyclodextrin-based mixture in which the drug forms an inclusion complex with the cyclodextrins of the mixture.
  • Formation of a drug-cyclodextrin complex can modify the drug's solubility, dissolution rate, bioavailability, and/or stability properties.
  • Sulfobutylether- ⁇ -cyclodextrin can also be used as an aid in the preparation of sustained-release formulations of agents of the combinations of the present invention.
  • a sustained-release tablet has been prepared that includes prednisolone and SBE- ⁇ -CD compressed in a hydroxypropyl methylcellulose matrix (see Rao et al., J. Pharm. Sci. 90: 807-16, 2001).
  • EP 1109806 B1 describes cyclodextrin complexes of paroxetine, where ⁇ -, ⁇ -, or ⁇ -cyclodextrins [including eptakis(2-6-di-O-methyl)- ⁇ -cyclodextrin, (2,3,6-tri-O-methyl)- ⁇ -cyclodextrin, monosuccinyl eptakis(2,6-di-O-methyl)- ⁇ -cyclodextrin, or 2-hydroxypropyl- ⁇ -cyclodextrin] in anhydrous or hydrated form formed complex ratios of agent to cyclodextrin of from 1:0.25 to 1:20 can be obtained.
  • Polymeric cyclodextrins have also been prepared, as described in U.S. patent application Ser. Nos. 10/021,294 and 10/021,312.
  • the cyclodextrin polymers so formed can be useful for formulating agents of the combinations of the present invention.
  • These multifunctional polymeric cyclodextrins are commercially available from Insert Therapeutics, Inc., Pasadena, Calif.
  • cyclodextrins may be used as an auxiliary additive, e.g. as a carrier, diluent or solubiliser.
  • Formulations that include cyclodextrins and other agents of the combinations of the present invention can be prepared by methods similar to the preparations of the cyclodextrin formulations described herein.
  • the liposomal carriers are composed of three general types of vesicle-forming lipid components.
  • the first includes vesicle-forming lipids that form the bulk of the vesicle structure in the liposome.
  • these vesicle-forming lipids include any amphipathic lipids having hydrophobic and polar head group moieties, and which (a) can form spontaneously into bilayer vesicles in water, as exemplified by phospholipids, or (b) are stably incorporated into lipid bilayers, with its hydrophobic moiety in contact with the interior, hydrophobic region of the bilayer membrane, and its polar head group moiety oriented toward the exterior, polar surface of the membrane.
  • the vesicle-forming lipids of this type are preferably ones having two hydrocarbon chains, typically acyl chains, and a polar head group. Included in this class are the phospholipids, such as phosphatidylcholine (PC), PE, phosphatidic acid (PA), phosphatidylinositol (PI), and sphingomyelin (SM), where the two hydrocarbon chains are typically between about 14-22 carbon atoms in length, and have varying degrees of unsaturation.
  • PC phosphatidylcholine
  • PA phosphatidic acid
  • PI phosphatidylinositol
  • SM sphingomyelin
  • the above-described lipids and phospholipids whose acyl chains have a variety of degrees of saturation can be obtained commercially, or prepared according to published methods.
  • Other lipids that can be included in the invention are glycolipids and sterols, such as cholesterol.
  • the second general component includes a vesicle-forming lipid that is derivatized with a polymer chain which will form the polymer layer in the composition.
  • the vesicle-forming lipids thta can be used as the second general vesicle-forming lipid component are any of those described for the first general vesicle-forming lipid component.
  • Vesicle forming lipids with diacyl chains, such as phospholipids are preferred.
  • One exemplary phospholipid is phosphatidylethanolamine (PE), which provides a reactive amino group that is convenient for coupling to the activated polymers.
  • An exemplary PE is distearyl PE (DSPE).
  • the preferred polymer in the derivatized lipid is polyethyleneglycol (PEG), preferably a PEG chain having a molecular weight between 1,000-15,000 daltons, more preferably between 2,000 and 10,000 daltons, most preferably between 2,000 and 5,000 daltons.
  • PEG polyethyleneglycol
  • Other hydrophilic polymers that may be suitable include polyvinylpyrrolidone, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyl methacrylamide, polymethacrylamide and polydimethylacrylamide, polylactic acid, polyglycolic acid, and derivatized celluloses, such as hydroxymethylcellulose or hydroxyethylcellulose.
  • block copolymers or random copolymers of these polymers may be suitable.
  • Methods for preparing lipids derivatized with hydrophilic polymers, such as PEG, are well known e.g., as described in U.S. Pat. No. 5,013,556.
  • a third general vesicle-forming lipid component which is optional, is a lipid anchor by which a targeting moiety is anchored to the liposome, through a polymer chain in the anchor. Additionally, the targeting group is positioned at the distal end of the polymer chain in such a way so that the biological activity of the targeting moiety is not lost.
  • the lipid anchor has a hydrophobic moiety which serves to anchor the lipid in the outer layer of the liposome bilayer surface, a polar head group to which the interior end of the polymer is covalently attached, and a free (exterior) polymer end which is or can be activated for covalent coupling to the targeting moiety.
  • the lipids components used in forming the liposomes are preferably present in a molar ratio of about 70-90 percent vesicle forming lipids, 1-25 percent polymer derivatized lipid, and 0.1-5 percent lipid anchor.
  • One exemplary formulation includes 50-70 mole percent underivatized PE, 20-40 mole percent cholesterol, 0.1-1 mole percent of a PE-PEG (3500) polymer with a chemically reactive group at its free end for coupling to a targeting moiety, 5-10 mole percent PE derivatized with PEG 3500 polymer chains, and 1 mole percent alpha-tocopherol.
  • the liposomes are preferably prepared to have substantially homogeneous sizes in a selected size range, typically between about 0.03 to 0.5 microns.
  • One effective sizing method for REVs and MLVs involves extruding an aqueous suspension of the liposomes through a series of polycarbonate membranes having a selected uniform pore size in the range of 0.03 to 0.2 micron, typically 0.05, 0.08, 0.1, or 0.2 microns.
  • the pore size of the membrane corresponds roughly to the largest sizes of liposomes produced by extrusion through that membrane, particularly where the preparation is extruded two or more times through the same membrane. Homogenization methods are also useful for down-sizing liposomes to sizes of 100 nm or less.
  • the liposomal formulations of the present invention include at least one surface-active agent.
  • Suitable surface-active agents useful for the formulation of the various combinations described herein include compounds belonging to the following classes: polyethoxylated fatty acids, PEG-fatty acid diesters, PEG-fatty acid mono-ester and di-ester mixtures, polyethylene glycol glycerol fatty acid esters, alcohol-oil transesterification products, polyglycerized fatty acids, propylene glycol fatty acid esters, mixtures of propylene glycol esters and glycerol esters, mono- and diglycerides, sterol and sterol derivatives, polyethylene glycol sorbitan fatty acid esters, polyethylene glycol alkyl ethers, sugar esters, polyethylene glycol alkyl phenyls, polyoxyethylene-polyoxypropylene block copolymers, sorbitan fatty acid esters, lower alcohol fatty acid esters, and ionic surfactants.
  • Polyethoxylated fatty acids may be used as excipients for the formulation of any one of the combinations described herein.
  • Examples of commercially available polyethoxylated fatty acid monoester surfactants include: PEG 4-100 monolaurate (Crodet L series, Croda), PEG 4-100 monooleate (Crodet 0 series, Croda), PEG 4-100 monostearate (Crodet S series, Croda, and Myij Series, Atlas/ICI), PEG 400 distearate (Cithrol 4DS series, Croda), PEG 100, 200, or 300 monolaurate (Cithrol ML series, Croda), PEG 100, 200, or 300 monooleate (Cithrol MO series, Croda), PEG 400 dioleate (Cithrol 4DO series, Croda), PEG 400-1000 monostearate (Cithrol MS series, Croda), PEG-1 stearate (Nikkol MYS-1EX,
  • Polyethylene glycol fatty acid diesters may also be used as excipients for anyone or all of the combinations described herein.
  • Examples of commercially available polyethylene glycol fatty acid diesters include: PEG-4 dilaurate (Mapeg® 200 DL, PPG), PEG-4 dioleate (Mapeg® 200 DO, PPG), PEG-4 distearate (Kessco® 200 DS, Stepan), PEG-6 dilaurate (Kessco® PEG 300 DL, Stepan), PEG-6 dioleate (Kessco® PEG 300 DO, Stepan), PEG-6 distearate (Kessco® PEG 300 DS, Stepan), PEG-8 dilaurate (Mapeg® 400 DL, PPG), PEG-8 dioleate (Mapeg® 400 DO, PPG), PEG-8 distearate (Mapeg® 400 DS, PPG), PEG-10 dipalmitate (Polyaldo 2PKFG), PEG-12 dilau
  • PEG-fatty acid mono- and di-ester mixtures may be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available PEG-fatty acid mono- and di-ester mixtures include: PEG 4-150 mono, dilaurate (Kessco® PEG 200-6000 mono, Dilaurate, Stepan), PEG 4-150 mono, dioleate (Kessco® PEG 200-6000 mono, Dioleate, Stepan), and PEG 4-150 mono, distearate (Kessco® 200-6000 mono, Distearate, Stepan).
  • Formulations of the anyone or all of the combinations according to the invention may include one or more of the PEG-fatty acid mono- and di-ester mixtures above.
  • polyethylene glycol glycerol fatty acid esters may be used as excipients for the formulation of anyone or all of the combinations described herein.
  • examples of commercially available polyethylene glycol glycerol fatty acid esters include: PEG-20 glyceryl laurate (Tagat® L, Goldschmidt), PEG-30 glyceryl laurate (Tagat® L2, Goldschmidt), PEG-15 glyceryl laurate (Glycerox L series, Croda), PEG-40 glyceryl laurate (Glycerox L series, Croda), PEG-20 glyceryl stearate (Capmul® EMG, ABITEC), and Aldo® MS-20 KFG, Lonza), PEG-20 glyceryl oleate (Tagat® 0, Goldschmidt), and PEG-30 glyceryl oleate (Tagat® O2, Goldschmidt).
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the
  • Alcohol-oil transesterification products may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available alcohol-oil transesterification products include: PEG-3 castor oil (Nikkol CO-3, Nikko), PEG-5, 9, and 16 castor oil (ACCONON CA series, ABITEC), PEG-20 castor oil, (Emalex C-20, Nihon Emulsion), PEG-23 castor oil (Emulgante EL23), PEG-30 castor oil (Incrocas 30, Croda), PEG-35 castor oil (Incrocas-35, Croda), PEG-38 castor oil (Emulgante EL 65, Condea), PEG-40 castor oil (Emalex C-40, Nihon Emulsion), PEG-50 castor oil (Emalex C-50, Nihon Emulsion), PEG-56 castor oil (Eumulgin® PRT 56, Pulcra SA), PEG-60 castor oil (Nikkol CO-60
  • oils in this category of surfactants are oil-soluble vitamins, such as vitamins A, D, E, K, etc.
  • derivatives of these vitamins such as tocopheryl PEG-1000 succinate (TPGS, available from Eastman) are also suitable surfactants.
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the alcohol-oil transesterification products above.
  • Polyglycerized fatty acids may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available polyglycerized fatty acids include: polyglyceryl-2 stearate (Nikkol DGMS, Nikko), polyglyceryl-2 oleate (Nikkol DGMO, Nikko), polyglyceryl-2 isostearate (Nikkol DGMIS, Nikko), polyglyceryl-3 oleate (Caprol® 3GO, ABITEC), polyglyceryl-4 oleate (Nikkol Tetraglyn 1-O, Nikko), polyglyceryl-4 stearate (Nikkol Tetraglyn 1-S, Nikko), polyglyceryl-6 oleate (Drewpol 6-1-O, Stepan), polyglyceryl-10 laurate (Nikkol Decaglyn 1-L, Nikko), polyglyceryl-10 oleate (Nikkol
  • propylene glycol fatty acid esters may be used as excipients for the formulation of the tetra-substituted pyrimidopyrimidine anyone or all of the combinations described herein.
  • Examples of commercially available propylene glycol fatty acid esters include: propylene glycol monocaprylate (Capryol 90, Gattefosse), propylene glycol monolaurate (Lauroglycol 90, Gattefosse), propylene glycol oleate (Lutrol OP2000, BASF), propylene glycol myristate (Mirpyl), propylene glycol monostearate (LIPO PGMS, Lipo Chem.), propylene glycol hydroxystearate, propylene glycol ricinoleate (PROPYMULS, Henkel), propylene glycol isostearate, propylene glycol monooleate (Myverol P-O6, Eastman), propylene glycol dicaprylate dicaprate (Captex® 200,
  • propylene glycol esters and glycerol esters may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • One preferred mixture is composed of the oleic acid esters of propylene glycol and glycerol (Arlacel 186).
  • these surfactants include: oleic (ATMOS 300, ARLACEL 186, ICI), and stearic (ATMOS 150).
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the mixtures of propylene glycol esters and glycerol esters above.
  • mono- and diglycerides may be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available mono- and diglycerides include: monopalmitolein (C16:1) (Larodan), monoelaidin (C18:1) (Larodan), monocaproin (C6) (Larodan), monocaprylin (Larodan), monocaprin (Larodan), monolaurin (Larodan), glyceryl monomyristate (C14) (Nikkol MGM, Nikko), glyceryl monooleate (C18:1) (PECEOL, Gattefosse), glyceryl monooleate (Myverol, Eastman), glycerol monooleate/linoleate (OLICINE, Gattefosse), glycerol monolinoleate (Maisine, Gattefosse), glyceryl ricinoleate (Softigen® 701, Huls), glyceryl monol
  • Sterol and sterol derivatives may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available sterol and sterol derivatives include: cholesterol, sitosterol, lanosterol, PEG-24 cholesterol ether (Solulan C-24, Amerchol), PEG-30 cholestanol (Phytosterol GENEROL series, Henkel), PEG-25 phytosterol (Nikkol BPSH-25, Nikko), PEG-5 soyasterol (Nikkol BPS-5, Nikko), PEG-10 soyasterol (Nikkol BPS-10, Nikko), PEG-20 soyasterol (Nikkol BPS-20, Nikko), and PEG-30 soyasterol (Nikkol BPS-30, Nikko).
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the sterol and sterol derivatives above.
  • Polyethylene glycol sorbitan fatty acid esters may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available polyethylene glycol sorbitan fatty acid esters include: PEG-10 sorbitan laurate (Liposorb L-10, Lipo Chem.), PEG-20 sorbitan monolaurate (Tween® 20, Atlas/ICI), PEG-4 sorbitan monolaurate (Tweeng 21, Atlas/ICI), PEG-80 sorbitan monolaurate (Hodag PSML-80, Calgene), PEG-6 sorbitan monolaurate (Nikkol GL-1, Nikko), PEG-20 sorbitan monopalmitate (Tween® 40, Atlas/ICI), PEG-20 sorbitan monostearate (Tween® 60, Atlas/ICI), PEG-4 sorbitan monostearate (Tweeng 61, Atlas/ICI), PEG-8 sorbitan monostearate (DACOL
  • polyethylene glycol alkyl ethers may be used as excipients for the formulation of anyone or all of the combinations described herein.
  • examples of commercially available polyethylene glycol alkyl ethers include: PEG-2 oleyl ether, oleth-2 (Brij 92/93, Atlas/ICI), PEG-3 oleyl ether, oleth-3 (Volpo 3, Croda), PEG-5 oleyl ether, oleth-5 (Volpo 5, Croda), PEG-10 oleyl ether, oleth-10 (Volpo 10, Croda), PEG-20 oleyl ether, oleth-20 (Volpo 20, Croda), PEG-4 lauryl ether, laureth-4 (Brij 30, Atlas/ICI), PEG-9 lauryl ether, PEG-23 lauryl ether, laureth-23 (Brij 35, Atlas/ICI), PEG-2 cetyl ether (Brij 52, ICI), PEG-10 cetyl ether (B
  • Sugar esters may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • examples of commercially available sugar esters include: sucrose distearate (SUCRO ESTER 7, Gattefosse), sucrose distearate/monostearate (SUCRO ESTER 11, Gattefosse), sucrose dipalmitate, sucrose monostearate (Crodesta F-160, Croda), sucrose monopalmitate (SUCRO ESTER 15, Gattefosse), and sucrose monolaurate (Saccharose monolaurate 1695, Mitsubisbi-Kasei).
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the sugar esters above.
  • Polyethylene glycol alkyl phenyls are also useful as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially available polyethylene glycol alkyl phenyls include: PEG-10-100 nonylphenyl series (Triton X series, Rohm & Haas) and PEG-15-100 octylphenyl ether series (Triton N-series, Rohm & Haas).
  • Formulations of anyone or all of the combinations to the invention may include one or more of the polyethylene glycol alkyl phenyls above.
  • Polyoxyethylene-polyoxypropylene block copolymers may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • These surfactants are available under various trade names, including one or more of Synperonic PE series (ICI), Pluronicg series (BASF), Lutrol (BASF), Supronic, Monolan, Pluracare, and Plurodac.
  • the generic term for these copolymers is “poloxamer” (CAS 9003-11-6). These polymers have the formula shown below: HO(C 2 H 4 O) a (C 3 H 6 O) b (C 2 H 4 O) a H where “a” and “b” denote the number of polyoxyethylene and polyoxypropylene units, respectively.
  • copolymers are available in molecular weights ranging from 1000 to 15000 daltons, and with ethylene oxide/propylene oxide ratios between 0.1 and 0.8 by weight.
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the polyoxyethylene-polyoxypropylene block copolymers above.
  • Polyoxyethylenes such as PEG 300, PEG 400, and PEG 600, may be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Sorbitan fatty acid esters may also be used as excipients for the formulation of anyone or all of the combinations described herein.
  • Examples of commercially sorbitan fatty acid esters include: sorbitan monolaurate (Span-20, Atlas/ICI), sorbitan monopalmitate (Span-40, Atlas/ICI), sorbitan monooleate (Span-80, Atlas/ICI), sorbitan monostearate (Span-60, Atlas/ICI), sorbitan trioleate (Span-85, Atlas/ICI), sorbitan sesquioleate (Arlacel-C, ICI), sorbitan tristearate (Span-65, Atlas/ICI), sorbitan monoisostearate (Crill 6, Croda), and sorbitan sesquistearate (Nikkol SS-15, Nikko).
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the sorbitan fatty acid esters above.
  • Esters of lower alcohols (C 2 to C 4 ) and fatty acids (C 8 to C 18 ) are suitable surfactants for use in the invention.
  • these surfactants include: ethyl oleate (Crodamol EO, Croda), isopropyl myristate (Crodamol IPM, Croda), isopropyl palmitate (Crodamol IPP, Croda), ethyl linoleate (Nikkol VF-E, Nikko), and isopropyl linoleate (Nikkol VF-IP, Nikko).
  • Formulations of anyone or all of the combinations according to the invention may include one or more of the lower alcohol fatty acid esters above.
  • ionic surfactants may be used as excipients for the formulation of anyone or all of the combinations described herein.
  • useful ionic surfactants include: sodium caproate, sodium caprylate, sodium caprate, sodium laurate, sodium myristate, sodium myristolate, sodium palmitate, sodium palmitoleate, sodium oleate, sodium ricinoleate, sodium linoleate, sodium linolenate, sodium stearate, sodium lauryl sulfate (dodecyl), sodium tetradecyl sulfate, sodium lauryl sarcosinate, sodium dioctyl sulfosuccinate, sodium cholate, sodium taurocholate, sodium glycocholate, sodium deoxycholate, sodium taurodeoxycholate, sodium glycodeoxycholate, sodium ursodeoxycholate, sodium chenodeoxycholate, sodium taurochenodeoxycholate, sodium glyco cheno deoxycholate, sodium
  • excipients present in the formulations of the invention are present in amounts such that the carrier forms a clear, or opalescent, aqueous dispersion of the tetra-substituted pyrimidopyrimidine, or the SSRI or the antihistamine, or anyone or all of the combination sequestered within the liposome.
  • the relative amount of a surface active excipient necessary for the preparation of liposomal or solid lipid nanoparticulate formulations is determined using known methodology.
  • liposomes may be prepared by a variety of techniques, such as those detailed in Szoka et al, 1980.
  • Multilamellar vesicles (MLVs) can be formed by simple lipid-film hydration techniques.
  • lipid film hydrates to form MLVs, typically with sizes between about 0.1 to 10 microns.
  • liposomes to facilitate cellular uptake is described in U.S. Pat. Nos. 4,897,355 and 4,394,448.
  • Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients.
  • excipients may be, for example, inert diluents or fillers (e.g., sucrose and sorbitol), lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc).
  • the two compounds may be mixed together in a tablet, capsule, or other vehicle, or may be partitioned.
  • the first compound is contained on the inside of the tablet, and the second compound is on the outside, such that a substantial portion of the second compound is released prior to the release of the first compound.
  • Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium.
  • an oral vehicle e.g., a capsule
  • a tetra-substituted pyrimidopyrimidine or analog and/or additional agent preferably from between 0.01% to 10% (w/w), more preferably from between 0.05% to 4% (w/w) active agent.
  • the capsule can be taken one to four times daily, or as needed.
  • an oral vehicle will contain from between 0.01% to 5% (w/w), preferably from between 0.01% to 2% (w/w), more preferably from between 0.01% to 1% (w/w) dipyridamole.
  • the oral vehicle containing a compound of dipyridamole or dipyridamole analog, and/or the additional agent is preferably taken orally.
  • a capsule may be taken in the morning and one in the evening by a subject suffering from an immunoinflammatory disorder or an immunoinflammatory-related disorder, like anti-platelet aggregatory activity.
  • compositions can also be adapted for topical use with a topical vehicle containing from between 0.0001% to 25% (w/w) or more of tetra-substituted pyrimidopyrimidine and/or analog and between 0.001% to 25% (w/w) and more of antihistamine and/or analog.
  • a topical vehicle containing from between 0.0001% to 25% (w/w) or more of tetra-substituted pyrimidopyrimidine and/or analog and between 0.001% to 25% (w/w) and more of antihistamine and/or analog.
  • tetra-substituted pyrimidopyrimidine is subjected to an extended-release mechanism.
  • the antihistamine and/or tetra-substituted pyrimidopyrimidine are preferably from between 0.0001% to 10% (w/w), more preferably from between 0.0005% to 4% (w/w) active agent.
  • the cream can be applied one to four times daily, or as needed.
  • a topical vehicle will contain from between 0.01% to 5% (w/w), preferably from between 0.01% to 2% (w/w), more preferably from between 0.01% to 1% (w/w) prednisolone in combination with tetra-substituted pyrimidopyrimidine, which is 0.0001% to 2% (w/w), more preferably from between 0.0005% to 1% (w/w).
  • the topical vehicle containing a compound of antihistamine or antihistamine analog, and/or the tetra-substituted pyrimidopyrimidine is preferably applied to the site of discomfort on the subject.
  • a cream may be applied to the hands of a subject suffering from arthritic fingers, while topical eye drops may be applied to an eye of a subject to treat uveitis.
  • the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurized container or nebulizer may contain a solution or suspension of the active compound.
  • Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.
  • a low dosage (as defined herein) of the tetra-substituted pyrimidopyrimidine and/or the additional agents can be used. These dosages will vary depending on the health and condition of the patient. Thus, a moderate dosage or even a high dosage of one or both agents can be used.
  • Administration of each drug in the combination can, independently, be one to four times daily for one day to one year, and may even be for the life of the patient. Chronic, long-term administration will be indicated in many cases.
  • the compounds of the invention are also useful as screening tools.
  • Single agents and combinations of the invention can be employed in antiproliferative or mechanistic assays to determine whether other combinations, or single agents are as effective as the combination in inhibiting the proliferation of proinflammatory cytokines using assays generally known in the art, e.g., TNF ⁇ , IL-1, IL-2, IFN- ⁇ etc., specific, non-limiting examples of which are described in the Examples section.
  • candidate compounds are combined with a compound from with either the tetra-substituted pyrimidopyrimidine (or tetra-substituted pyrimidopyrimidine analog) or the additional agents described herein, applied to stimulate PBMCs, and at after a suitable time, the cells are examined for anitproliferative activity, TNF ⁇ or other assays for proinflammatory cytokine secretion.
  • the relative effects of the combinations versus each other, and versus the single agents are compared, and effective compounds and combinations are identified.
  • the screening method can be used for comparing the activity of novel single agents or new combinations of agents (novel or known) for relative activity in the assays.
  • the combinations of the invention are also useful tools in elucidating mechanistic information about the biological pathways involved in inflammation or novel targets. Such information can lead to the development of new combinations or single agents (mechanistic and/or structural analogs of either the tetra-substituted pyrimidopyrimidine or companion compound) for inhibiting proinflammatory cytokine secretion. Methods known in the art to determine biological pathways can be used to determine the pathway, or network of pathways affected by contacting cells stimulated to produce proinflammatory cytokines with the compounds of the invention.
  • Such methods can include, analyzing cellular constituents that are expressed or repressed after contact with the compounds of the invention as compared to untreated, positive or negative control compounds, and/or new single agents and combinations, or analyzing some other metabolic activity of the cell such as enzyme activity, nutrient uptake, and proliferation.
  • Cellular components analyzed can include gene transcripts, and protein expression.
  • Suitable methods can include standard biochemistry techniques, radiolabeling the compounds of the invention (e,.g., 14 C or 3 H labeling), and observing the compounds binding to proteins, e.g. using 2d gels, gene expression profiling. Once identified, such compounds can be used in in vivo models to further validate the tool or develop new anti-inflammatory agents.
  • test compound combinations on TNF ⁇ secretion were assayed in white blood cells from human buffy coat stimulated with LPS or phorbol 12-myristate 13-acetateand ionomycin as follows.
  • Human white blood cells from buffy coat were diluted 1:50 in media (RPMI; Gibco BRL, #11875-085), 10% fetal bovine serum (Gibco BRL, #25140-097), 2% penicillin/streptomycin (Gibco BRL, #15140-122)) and 50 ⁇ L of the diluted white blood cells was placed in each well of the assay plate. Drugs were added to the indicated concentration. After 16-18 hours of incubation at 37° C.
  • test compound combinations on IFN- ⁇ secretion were assayed in white blood cells from human buffy coat stimulated with phorbol 12-myristate 13-acetate as follows.
  • Human white blood cells from buffy coat were diluted 1:50 in media (RPMI; Gibco BRL, #11875-085), 10% fetal bovine serum (Gibco BRL, #25140-097), 2% penicillin/streptomycin (Gibco BRL, #15140-122)) and 50 ⁇ L of the diluted white blood cells was placed in each well of the final assay plate created in the above section. After 16-18 hours of incubation at 37° C.
  • test compound combinations on IL-1 secretion were assayed in white blood cells from human buffy coat stimulated with LPS, as follows.
  • Human white blood cells from buffy coat were diluted 1:50 in media (RPMI; Gibco BRL, #11875-085), 10% fetal bovine serum (Gibco BRL, #25140-097), 2% penicillin/streptomycin (Gibco BRL, #15140-122)) and 50 ⁇ L of the diluted white blood cells was placed in each well of the final assay plate created in the above section. After 16-18 hours of incubation at 37° C.
  • the plate was centrifuged and the supernatant was transferred to a white opaque 384-well plate (NalgeNunc, MAXISORB) coated with an anti-IL-1 antibody (R&D Systems, MAB601). After a two-hour incubation, the plate was washed (Tecan Powerwasher 384) with PBS containing 0.1% Tween 20 and incubated for an additional one hour with a biotin labeled anti-IL-1 antibody (R&D Systems, BAF201) and horse radish peroxidase coupled to streptavidin (PharMingen, #554066). The plate was then washed again with 0.1% Tween 20/PBS, and an HRP-luminescent substrate was added to each well. Light intensity was then measured using a plate luminometer.
  • test compound combinations on IL-2 secretion were assayed in white blood cells from human buffy coat stimulated with phorbol 12-myristate 13-acetate, as follows.
  • Human white blood cells from buffy coat were diluted 1:50 in media (RPMI; Gibco BRL, #11875-085), 10% fetal bovine serum (Gibco BRL, #25140-097), 2% penicillin/streptomycin (Gibco BRL, #15140-122)) and 50 ⁇ L of the diluted white blood cells was placed in each well of the final assay plate created in the above section. After 16-18 hours of incubation at 37° C.
  • the plate was centrifuged and the supernatant was transferred to a white opaque 384-well plate (NalgeNunc, MAXISORB) coated with an anti-IL-2 antibody (PharMingen, #555051). After a two-hour incubation, the plate was washed (Tecan Powerwasher 384) with PBS containing 0.1% Tween 20 and incubated for an additional one hour with a biotin labeled anti-IL-2 antibody (Endogen, M600B) and horse radish peroxidase coupled to streptavidin (PharMingen, #13047E). The plate was then washed again with 0.1% Tween 20/PBS, and an HRP-luminescent substrate was added to each well. Light intensity was then measured using a plate luminometer.
  • the average untreated well value is the arithmetic mean of 40 wells from the same assay plate treated with vehicle alone. Negative inhibition values result from local variations in treated wells as compared to untreated wells.
  • Tables 7-53 The results of various combinations of compounds described on the reduction of TNF ⁇ , IL-2, IL-1, or IFN- ⁇ secretion are shown below in Tables 7-53.
  • Table 8 shows the effects of varying concentrations of dipyridamole and a combination of dipyridamole and bromodiphenhydramine HCl. These results were compared to control wells. These wells were stimulated with phorbol 12-myristate 13-acetate and ionomycin, but did not receive dipyridamole or bromodiphenhydramine HCl.
  • the effects of the agents alone and in combination are shown as percent inhibition of TNF ⁇ secretion.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Pain & Pain Management (AREA)
  • Biomedical Technology (AREA)
  • Transplantation (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Neurosurgery (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US10/966,228 2003-10-15 2004-10-15 Methods and reagents for the treatment of immunoinflammatory disorders Abandoned US20050119160A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/966,228 US20050119160A1 (en) 2003-10-15 2004-10-15 Methods and reagents for the treatment of immunoinflammatory disorders
US11/517,593 US8080553B2 (en) 2003-10-15 2006-09-07 Methods and reagents for the treatment of immunoinflammatory disorders
US13/292,669 US20120058979A1 (en) 2003-10-15 2011-11-09 Methods and reagents for the treatment of immunoinflammatory disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US51241503P 2003-10-15 2003-10-15
US10/966,228 US20050119160A1 (en) 2003-10-15 2004-10-15 Methods and reagents for the treatment of immunoinflammatory disorders

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/517,593 Continuation US8080553B2 (en) 2003-10-15 2006-09-07 Methods and reagents for the treatment of immunoinflammatory disorders

Publications (1)

Publication Number Publication Date
US20050119160A1 true US20050119160A1 (en) 2005-06-02

Family

ID=34465346

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/966,228 Abandoned US20050119160A1 (en) 2003-10-15 2004-10-15 Methods and reagents for the treatment of immunoinflammatory disorders
US11/517,593 Expired - Fee Related US8080553B2 (en) 2003-10-15 2006-09-07 Methods and reagents for the treatment of immunoinflammatory disorders
US13/292,669 Abandoned US20120058979A1 (en) 2003-10-15 2011-11-09 Methods and reagents for the treatment of immunoinflammatory disorders

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/517,593 Expired - Fee Related US8080553B2 (en) 2003-10-15 2006-09-07 Methods and reagents for the treatment of immunoinflammatory disorders
US13/292,669 Abandoned US20120058979A1 (en) 2003-10-15 2011-11-09 Methods and reagents for the treatment of immunoinflammatory disorders

Country Status (17)

Country Link
US (3) US20050119160A1 (is)
EP (3) EP2301628A1 (is)
JP (1) JP2007508391A (is)
KR (1) KR20070001060A (is)
CN (1) CN1889956A (is)
AR (1) AR046291A1 (is)
AU (1) AU2004281729A1 (is)
BR (1) BRPI0415397A (is)
CA (1) CA2542074A1 (is)
IL (1) IL174918A0 (is)
IS (1) IS8451A (is)
NO (1) NO20062003L (is)
RU (1) RU2006116511A (is)
SG (1) SG147442A1 (is)
TW (1) TW200517114A (is)
WO (1) WO2005037203A2 (is)
ZA (1) ZA200603116B (is)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060234991A1 (en) * 2001-10-05 2006-10-19 Curtis Keith Combinations for the treatment of immunoinflammatory disorders
US20070010502A1 (en) * 2003-10-15 2007-01-11 Combinatorx Inc. Methods and reagents for the treatment of immunoinflammatory disorders
US20070020298A1 (en) * 2003-12-31 2007-01-25 Pipkin James D Inhalant formulation containing sulfoalkyl ether gamma-cyclodextrin and corticosteroid
WO2007139753A2 (en) * 2006-05-22 2007-12-06 Combinatorx, Incorporated Methods and compositions for the treatment of diseases or conditions associated with increased c-reactive protein, interleukin-6, or interferon-gamma levels
WO2007146012A1 (en) * 2006-06-07 2007-12-21 Combinatorx, Incorporated Treatment of periodontal disease associated with increased c-reactive protein levels
WO2007089617A3 (en) * 2006-01-26 2008-06-12 Combinatorx Inc Methods, compositions, and kits for the treatment of musculoskeletal disorders and symptoms associated therewith
US20080181876A1 (en) * 2007-01-30 2008-07-31 Johnson Kirk W Methods for treating acute and subchronic pain
US20080287402A1 (en) * 2007-05-03 2008-11-20 Johnson Kirk W Use of a glial attenuator to prevent amplified pain responses caused by glial priming
US20090018203A1 (en) * 2002-10-25 2009-01-15 Collegium Pharmaceutical, Inc. Modified release compositions of milnacipran
US20090047243A1 (en) * 2007-07-17 2009-02-19 Richard Rickles Combinations for the treatment of b-cell proliferative disorders
US20090053168A1 (en) * 2007-07-17 2009-02-26 Richard Rickles Treatments of b-cell proliferative disorders
US20090075955A1 (en) * 2007-09-19 2009-03-19 Combinatorx, Inc. Therapeutic regimens for the treatment of immunoinflammatory disorders
US20090092665A1 (en) * 2007-10-08 2009-04-09 Lux Biosciences, Inc. OPHTHALMIC COMPOSITIONS COMPRISING CALCINEURIN INHIBITORS OR mTOR INHIBITORS
US7534806B2 (en) 2004-12-06 2009-05-19 Avigen, Inc. Method for treating neuropathic pain and associated syndromes
US20090312724A1 (en) * 2007-06-28 2009-12-17 Cydex Pharmaceuticals, Inc. Nasal and Ophthalmic Delivery of Aqueous Corticosteroid Solutions
US20100009934A1 (en) * 2008-06-09 2010-01-14 Combinatorx, Incorporated Beta adrenergic receptor agonists for the treatment of b-cell proliferative disorders
US20100092479A1 (en) * 2008-08-18 2010-04-15 Combinatorx (Singapore) Pte. Ltd. Compositions and methods for treatment of viral diseases
US20100310642A1 (en) * 2009-06-09 2010-12-09 Lux Biosciences, Inc. Topical Drug Delivery Systems for Ophthalmic Use
US20110189293A1 (en) * 2007-12-17 2011-08-04 CombinatoRx, Incoporated Therapeutic regimens for the treatment of immunoinflammatory disorders
US9827324B2 (en) 2003-12-31 2017-11-28 Cydex Pharmaceuticals, Inc. Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US11622991B2 (en) 2017-05-12 2023-04-11 Aurinia Pharmaceuticals Inc. Protocol for treatment of lupus nephritis

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0416812A (pt) * 2003-11-21 2007-03-06 Combinatorx Inc métodos e reagentes para o tratamento de desordens inflamatórias
MX336033B (es) * 2005-08-03 2016-01-07 Immunogen Inc Formulaciones de inmunoconjugado.
CA2687715A1 (en) * 2007-05-09 2008-11-20 Traffick Therapeutics Inc. Screening assay to identify correctors of protein trafficking defects
US20090005358A1 (en) * 2007-06-26 2009-01-01 Jan Lessem Compositions and methods for treating medical conditions
US9168263B2 (en) * 2008-01-24 2015-10-27 Soligenix, Inc. Topically active steroids for use in interstitial pulmonary fibrosis
US9314469B2 (en) * 2008-05-05 2016-04-19 Tonix Pharma Holdings Limited Method for treating neurocognitive dysfunction
RS62297B1 (sr) 2011-11-23 2021-09-30 Therapeuticsmd Inc Prirodne kombinovane hormonske supstitucione formulacije i terapije
US9301920B2 (en) 2012-06-18 2016-04-05 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US20150196640A1 (en) 2012-06-18 2015-07-16 Therapeuticsmd, Inc. Progesterone formulations having a desirable pk profile
US10806697B2 (en) 2012-12-21 2020-10-20 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US20130338122A1 (en) 2012-06-18 2013-12-19 Therapeuticsmd, Inc. Transdermal hormone replacement therapies
US9145383B2 (en) 2012-08-10 2015-09-29 Hallstar Innovations Corp. Compositions, apparatus, systems, and methods for resolving electronic excited states
US9867800B2 (en) 2012-08-10 2018-01-16 Hallstar Innovations Corp. Method of quenching singlet and triplet excited states of pigments, such as porphyrin compounds, particularly protoporphyrin IX, with conjugated fused tricyclic compounds have electron withdrawing groups, to reduce generation of reactive oxygen species, particularly singlet oxygen
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10568891B2 (en) 2012-12-21 2020-02-25 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US20150031712A1 (en) 2013-03-12 2015-01-29 Moshe Rogosnitzky Therapeutic Compositions Containing Dipyridamole and Treatment Packs Including Such Compositions and Methods for Producing Same
GB201309379D0 (en) * 2013-05-24 2013-07-10 Chronos Therapeutics Ltd Medical methods and compounds for medical use
JP2017516768A (ja) 2014-05-22 2017-06-22 セラピューティックスエムディー インコーポレーテッドTherapeuticsmd, Inc. 天然の併用ホルモン補充療法剤及び療法
RU2016141135A (ru) 2014-07-29 2018-08-28 Терапьютиксмд, Инк. Трансдермальный крем
RU2572709C1 (ru) * 2015-03-03 2016-01-20 Государственное бюджетное образовательное учреждение высшего профессионального образования "Курский государственный медицинский университет" Министерства здравоохранения Российской Федерации Способ коррекции структурно-функциональных нарушений артериального русла у больных ревматоидным артритом
EP3302487A4 (en) 2015-06-08 2019-05-22 O.D. Ocular Discovery Ltd. THERAPEUTIC COMPOSITIONS CONTAINING DIPYRIDAMOLE, MODULES AND TREATMENT KITS COMPRISING SUCH COMPOSITIONS, AND METHODS OF PRODUCING THE SAME
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
RU2018133932A (ru) 2016-04-01 2020-05-12 Терапьютиксмд, Инк. Фармацевтическая композиция стероидного гормона
WO2017173044A1 (en) 2016-04-01 2017-10-05 Therapeuticsmd Inc. Steroid hormone compositions in medium chain oils
EP3562812A1 (en) 2016-12-28 2019-11-06 Tonix Pharma Holdings Limited Tianeptine oxalate salts and polymorphs
WO2019157428A1 (en) * 2018-02-12 2019-08-15 Medicinova, Inc. Methods of suppressing myeloid-derived suppressor cells in patients
CN109568564B (zh) * 2018-12-29 2022-04-29 武汉天天好生物制品有限公司 一种多肽组件复合物、多肽组件双层片及制备工艺和应用
US11633405B2 (en) 2020-02-07 2023-04-25 Therapeuticsmd, Inc. Steroid hormone pharmaceutical formulations
EP4236949A1 (en) * 2020-10-30 2023-09-06 The Board of Trustees of the Leland Stanford Junior University Drugs targeting inflammation for the treatment of osteoarthritis and other inflammatory diseases

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030125343A1 (en) * 1999-12-27 2003-07-03 Carlo Gambacorti-Passerini Combinations of receptor tyrosine kinase inhibitor with an a1-acidic glycoprotein binding compound

Family Cites Families (182)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US532248A (en) 1895-01-08 powell
US2129401A (en) 1937-09-09 1938-09-06 Botinger Fred Flexible utility ladder
US3031450A (en) 1959-04-30 1962-04-24 Thomae Gmbh Dr K Substituted pyrimido-[5, 4-d]-pyrimidines
JPS5229318B2 (is) 1972-03-30 1977-08-01
US4034087A (en) 1973-12-17 1977-07-05 The Regents Of The University Of Michigan Pharmaceutical composition and process of treatment
US4107306A (en) 1973-01-16 1978-08-15 The Regents Of The University Of Michigan Process for treating proliferative skin disease
GB1422263A (en) 1973-01-30 1976-01-21 Ferrosan As 4-phenyl-piperidine compounds
DD119586A5 (is) 1973-12-21 1976-05-05
US4193926A (en) 1974-03-20 1980-03-18 Schering Aktiengesellschaft 4-(Polyalkoxy phenyl)-2-pyrrolidones
US4097483A (en) 1974-11-01 1978-06-27 Kyorin Pharmaceutical Co., Ltd. Pyrazolo 1,5-a!pyridines
US3956296A (en) 1974-12-11 1976-05-11 A. H. Robins Company, Incorporated 1-Substituted-4-benzylpiperidines
US3934036A (en) 1975-01-23 1976-01-20 Kyorin Seiyaku Kabushiki Kaisha N-benzenesulfonyl-β-alanine hydrazide useful as an immunosuppressive agent
JPS58403B2 (ja) 1975-07-24 1983-01-06 武田薬品工業株式会社 L− アスコルビンサンセイザイノ セイゾウホウ
US4394448A (en) 1978-02-24 1983-07-19 Szoka Jr Francis C Method of inserting DNA into living cells
FI65914C (fi) 1978-03-07 1984-08-10 Sandoz Ag Foerfarande foer framstaellning av farmaceutiska kompositionerinnehaollande cyklosporin a
NL171194C (nl) 1978-05-23 1983-02-16 Giesen Metaalgieterij Warmwater-ketel, bijvoorbeeld een centrale verwarmingsketel.
EP0005911B1 (en) * 1978-05-26 1982-02-17 Imperial Chemical Industries Plc Analgesic 6-acylaminotetrahydro-1,3,5-triazine-2,4-dione derivatives, pharmaceutical compositions thereof, and process for their manufacture
JPS6056143B2 (ja) 1979-08-02 1985-12-09 山之内製薬株式会社 アミジン誘導体ならびにその製造法
PH16099A (en) 1979-03-06 1983-06-24 Yamanouchi Pharma Co Ltd Guanidinothiazole compounds,process for preparing them and medical composition containing them
US4254129A (en) 1979-04-10 1981-03-03 Richardson-Merrell Inc. Piperidine derivatives
US4285958A (en) 1979-04-10 1981-08-25 Richardson-Merrell Inc. 1-Piperidine-alkylene ketones, pharmaceutical compositions thereof and method of use thereof
US4285957A (en) 1979-04-10 1981-08-25 Richardson-Merrell Inc. 1-Piperidine-alkanol derivatives, pharmaceutical compositions thereof, and method of use thereof
US4254130A (en) 1979-04-10 1981-03-03 Richardson-Merrell Inc. Piperidine derivatives
FR2470599A1 (fr) 1979-12-07 1981-06-12 Panoz Donald Perfectionnements apportes aux procedes de preparation de formes galeniques a action retard et a liberation programmee et formes galeniques de medicaments ainsi obtenus
DE3000979A1 (de) 1980-01-12 1981-07-23 Dr. Karl Thomae Gmbh, 7950 Biberach Neue dipyridamol-retardformen und verfahren zu ihrer herstellung
US4394508A (en) 1980-06-07 1983-07-19 Bristol-Myers Company Chemical compounds
US4316885A (en) 1980-08-25 1982-02-23 Ayerst, Mckenna And Harrison, Inc. Acyl derivatives of rapamycin
US4384996A (en) 1981-01-09 1983-05-24 Sandoz Ltd. Novel cyclosporins
US4510309A (en) 1981-03-03 1985-04-09 Bristol-Myers Company Histamine H2 -antagonists
BG36086A1 (en) 1982-01-19 1984-09-14 Glbov Method for inducing interferon
JPS59167590A (ja) 1983-03-14 1984-09-21 Kyorin Pharmaceut Co Ltd ピラゾロ〔1,5−a〕ピリジン誘導体、その製法及びそれを含有する治療剤
US4440933A (en) 1983-03-16 1984-04-03 Bristol-Myers Company Process for preparing 1,2,5-thiadiazoles
GB8321157D0 (en) 1983-08-05 1983-09-07 Fordonal Sa Piperidine derivatives
US4879119A (en) 1984-02-21 1989-11-07 Yamanouchi Pharmaceutical Co., Ltd. Patch
JPS60174716A (ja) 1984-02-21 1985-09-09 Yamanouchi Pharmaceut Co Ltd パツチ剤
US4554271A (en) 1984-02-24 1985-11-19 The Upjohn Company Use of high doses of derivatives of 6α-methylprednisolone for the acute treatment of stroke syndrome
US4933438A (en) 1984-02-29 1990-06-12 University Of Florida Brain-specific analogues of centrally acting amines
US4894366A (en) 1984-12-03 1990-01-16 Fujisawa Pharmaceutical Company, Ltd. Tricyclo compounds, a process for their production and a pharmaceutical composition containing the same
US4727064A (en) 1984-04-25 1988-02-23 The United States Of America As Represented By The Department Of Health And Human Services Pharmaceutical preparations containing cyclodextrin derivatives
US5639724A (en) 1984-07-24 1997-06-17 Sandoz Ltd. Cyclosporin galenic forms
US5254562A (en) 1984-12-03 1993-10-19 Fujisawa Pharmaceutical Company, Ltd. Tricyclo compounds, a process for their production and a pharmaceutical composition containing the same
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
GB8509276D0 (en) 1985-04-11 1985-05-15 Smith Kline French Lab Pyridine derivatives
US4650803A (en) 1985-12-06 1987-03-17 University Of Kansas Prodrugs of rapamycin
US4742175A (en) 1986-05-07 1988-05-03 Merrell Dow Pharmaceuticals Inc. Preparation of polymorphically pure terfenadine
DE3627423A1 (de) 1986-08-13 1988-02-18 Thomae Gmbh Dr K Arzneimittel enthaltend dipyridamol oder mopidamol und o-acetylsalicylsaeure bzw. deren physiologisch vertraegliche salze, verfahren zu ihrer herstellung und ihre verwendung zur bekaempfung der thrombusbildung
US4956388A (en) 1986-12-22 1990-09-11 Eli Lilly And Company 3-aryloxy-3-substituted propanamines
AU618517B2 (en) 1986-12-23 1992-01-02 Eugene J. Van Scott Additives enhancing topical actions of therapeutic agents
AU611421B2 (en) 1987-02-23 1991-06-13 Shiseido Company Ltd. Percutaneous absorption promoter and dermatologic preparation for external use
DE3706399A1 (de) 1987-02-27 1988-09-08 Boehringer Ingelheim Kg Mittel mit antidepressiver wirkung
US5668116A (en) 1987-03-19 1997-09-16 Anthropharm Pty. Limited Anti-inflammatory compounds and compositions
US4798823A (en) 1987-06-03 1989-01-17 Merck & Co., Inc. New cyclosporin analogs with modified "C-9 amino acids"
JP2577049B2 (ja) 1987-06-04 1997-01-29 三共株式会社 シクロスポリン製剤
US4925849A (en) 1987-06-15 1990-05-15 Fujisawa Pharmaceutical Company, Ltd. Pharmaceutically useful pyrazolopyridines
US5227467A (en) 1987-08-03 1993-07-13 Merck & Co., Inc. Immunosuppressive fluorinated cyclosporin analogs
US4833138A (en) 1987-10-23 1989-05-23 Washington University Phenothiazinealkaneamines for treatment of neurotoxic injury
US5242921A (en) 1988-04-27 1993-09-07 Yale University Compositions and methods for treating cutaneous hyperproliferative disorders
WO1989010122A1 (en) 1988-04-27 1989-11-02 Yale University Compositions and methods for treating cutaneous hyperproliferative disorders
GB8813032D0 (en) * 1988-06-02 1988-07-06 Boehringer Ingelheim Int Antiviral pharmaceutical composition
GB8819626D0 (en) * 1988-08-18 1988-09-21 Boehringer Ingelheim Int Pharmaceutical composition for treatment of tumours
GB2222770B (en) 1988-09-16 1992-07-29 Sandoz Ltd Pharmaceutical compositions containing cyclosporins
US6007840A (en) 1988-09-16 1999-12-28 Novartis Ag Pharmaceutical compositions comprising cyclosporins
US5342625A (en) 1988-09-16 1994-08-30 Sandoz Ltd. Pharmaceutical compositions comprising cyclosporins
US5135757A (en) 1988-09-19 1992-08-04 Edward Mendell Co., Inc. Compressible sustained release solid dosage forms
US4908372A (en) 1988-10-13 1990-03-13 Merrell Dow Pharmaceuticals Inc. Antihistaminic piperidinyl benzimidazoles
US4963541A (en) 1989-02-22 1990-10-16 Abbott Laboratories Pyrimido-pyrimidine lipoxygenase inhibiting compounds
US5007790A (en) 1989-04-11 1991-04-16 Depomed Systems, Inc. Sustained-release oral drug dosage form
US5284826A (en) 1989-07-24 1994-02-08 Sandoz Ltd. 0-hydroxyethyl and acyloxyethyl derivatives of [ser]8 cyclosporins
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5208228A (en) 1989-11-13 1993-05-04 Merck & Co., Inc. Aminomacrolides and derivatives having immunosuppressive activity
IT1237904B (it) 1989-12-14 1993-06-18 Ubaldo Conte Compresse a rilascio a velocita' controllata delle sostanze attive
KR0166088B1 (ko) 1990-01-23 1999-01-15 . 수용해도가 증가된 시클로덱스트린 유도체 및 이의 용도
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5122511A (en) 1990-02-27 1992-06-16 Merck & Co., Inc. Immunosuppressive cyclosporin analogs with modified amino acids at position-8
PT98990A (pt) 1990-09-19 1992-08-31 American Home Prod Processo para a preparacao de esteres de acidos carboxilicos de rapamicina
US5143918A (en) 1990-10-11 1992-09-01 Merck & Co., Inc. Halomacrolides and derivatives having immunosuppressive activity
US5626863A (en) 1992-02-28 1997-05-06 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
DE4034218A1 (de) 1990-10-27 1992-04-30 Merck Patent Gmbh Verfahren zur herstellung von carebastin
WO1992016226A1 (en) 1991-03-19 1992-10-01 Smithkline Beecham Corporation Il-1 inhibitors
US5120842A (en) 1991-04-01 1992-06-09 American Home Products Corporation Silyl ethers of rapamycin
US5100883A (en) 1991-04-08 1992-03-31 American Home Products Corporation Fluorinated esters of rapamycin
US5118678A (en) 1991-04-17 1992-06-02 American Home Products Corporation Carbamates of rapamycin
US5565560A (en) 1991-05-13 1996-10-15 Merck & Co., Inc. O-Aryl,O-alkyl,O-alkenyl and O-alkynylmacrolides having immunosuppressive activity
US5162334A (en) 1991-05-13 1992-11-10 Merck & Co., Inc. Amino O-alkyl, O-alkenyl and O-alkynlmacrolides having immunosuppressive activity
US5262533A (en) 1991-05-13 1993-11-16 Merck & Co., Inc. Amino O-aryl macrolides having immunosuppressive activity
US5250678A (en) 1991-05-13 1993-10-05 Merck & Co., Inc. O-aryl, O-alkyl, O-alkenyl and O-alkynylmacrolides having immunosuppressive activity
US5120725A (en) 1991-05-29 1992-06-09 American Home Products Corporation Bicyclic rapamycins
US5120727A (en) 1991-05-29 1992-06-09 American Home Products Corporation Rapamycin dimers
US5169851A (en) 1991-08-07 1992-12-08 American Home Products Corporation Rapamycin analog as immunosuppressants and antifungals
US5202332A (en) 1991-08-07 1993-04-13 American Home Products Corporation Rapamycin analog as immunosuppressant
US5189042A (en) 1991-08-22 1993-02-23 Merck & Co. Inc. Fluoromacrolides having immunosuppressive activity
US5208241A (en) 1991-09-09 1993-05-04 Merck & Co., Inc. N-heteroaryl, n-alkylheteroaryl, n-alkenylheteroaryl and n-alkynylheteroarylmacrolides having immunosuppressive activity
US5151413A (en) 1991-11-06 1992-09-29 American Home Products Corporation Rapamycin acetals as immunosuppressant and antifungal agents
US5270047A (en) 1991-11-21 1993-12-14 Kauffman Raymond F Local delivery of dipyridamole for the treatment of proliferative diseases
US5474765A (en) 1992-03-23 1995-12-12 Ut Sw Medical Ctr At Dallas Preparation and use of steroid-polyanionic polymer-based conjugates targeted to vascular endothelial cells
US5284840A (en) 1992-06-12 1994-02-08 Merck & Co., Inc. Alkylidene macrolides having immunosuppressive activity
JP3041954B2 (ja) 1992-08-03 2000-05-15 セプラコア インコーポレーテッド アレルギー疾患治療用のテルフェナジン代謝物及びその光学的に純粋な異性体
US5258389A (en) 1992-11-09 1993-11-02 Merck & Co., Inc. O-aryl, O-alkyl, O-alkenyl and O-alkynylrapamycin derivatives
US5504091A (en) 1993-04-23 1996-04-02 American Home Products Corporation Biotin esters of rapamycin
AU670004B2 (en) 1993-06-24 1996-06-27 Albany Molecular Research, Inc. Piperidine derivatives and process for their production
EG20321A (en) 1993-07-21 1998-10-31 Otsuka Pharma Co Ltd Medical material and process for producing the same
US5428040A (en) 1993-08-31 1995-06-27 The Du Pont Merck Pharmaceutical Company Carbocyclic fused-ring quinolinecarboxylic acids useful as immunosuppressive agents
US5773025A (en) 1993-09-09 1998-06-30 Edward Mendell Co., Inc. Sustained release heterodisperse hydrogel systems--amorphous drugs
US5589194A (en) 1993-09-20 1996-12-31 Minnesota Mining And Manufacturing Company Method of encapsulation and microcapsules produced thereby
US6022852A (en) 1993-10-22 2000-02-08 Hexal Ag Pharmaceutical composition containing cyclosporin A
US5468729A (en) 1993-10-26 1995-11-21 Alpha 1 Biomedicals Method for treatment of autoimmune hepatitis
GB2292079B (en) 1994-08-12 1998-07-15 Flexpharm Ltd Coated prednisolone preparation for the treatment of inflamatory bowel disease
DE4430128A1 (de) * 1994-08-25 1996-02-29 Hoechst Ag Kombinationspräparat mit immunsuppressiven, kardiovaskulären und cerebralen Wirkungen
US5693648A (en) 1994-09-30 1997-12-02 Merck & Co., Inc. O-aryl, O-alkyl, O-alkenyl and O-alkynyl-macrolides having immunosuppressive activity
US5696127A (en) 1994-12-22 1997-12-09 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
FR2732223B1 (fr) 1995-03-30 1997-06-13 Sanofi Sa Composition pharmaceutique pour administration transdermique
AU5772196A (en) 1995-05-19 1996-11-29 Chiroscience Limited 3,4-disubstituted-phenylsulphonamides and their therapeutic use
US6265427B1 (en) 1995-06-07 2001-07-24 The Proctor & Gamble Company Pharmaceutical composition for the method of treating leukemia
US6548084B2 (en) 1995-07-20 2003-04-15 Smithkline Beecham Plc Controlled release compositions
DE19537012A1 (de) 1995-10-04 1997-04-10 Dietl Hans Cyclosporin(e) enhaltende pharmazeutische Zubereitung zur oralen Applikation und Verfahren zu ihrer Herstellung
US6153754A (en) 1995-12-21 2000-11-28 Albany Molecular Research, Inc. Process for production of piperidine derivatives
US6201124B1 (en) 1995-12-21 2001-03-13 Albany Molecular Research, Inc. Process for production of piperidine derivatives
US5709797A (en) 1996-06-05 1998-01-20 Poli Industria Chimica S.P.A. Method of isolating cyclosporins
US6235706B1 (en) 1996-09-18 2001-05-22 Merck & Co., Inc. Combination therapy for reducing the risks associated with cardiovascular disease
US5874437A (en) 1996-11-01 1999-02-23 Nitromed, Inc. Nitrosated and nitrosylated phosphodiesterase inhibitor compounds, compositions and their uses
US6331543B1 (en) 1996-11-01 2001-12-18 Nitromed, Inc. Nitrosated and nitrosylated phosphodiesterase inhibitors, compositions and methods of use
US5958926A (en) 1996-11-01 1999-09-28 Nitromed, Inc. Nitrosated and nitrosylated phosphodiesterase inhibitor compounds, compositions and their uses
CA2270118A1 (en) 1996-11-01 1998-05-14 Nitromed Inc. Nitrosated and nitrosylated phosphodiesterase inhibitor compounds, compositions and their uses
US5792476A (en) 1996-12-19 1998-08-11 Abigo Medical Ab Sustained release glucocorticoid pharmaceutical composition
US6054487A (en) 1997-03-18 2000-04-25 Basf Aktiengesellschaft Methods and compositions for modulating responsiveness to corticosteroids
EA199901112A1 (ru) 1997-06-05 2000-06-26 Эли Лилли Энд Компани Способы лечения тромботических нарушений
GB9711643D0 (en) 1997-06-05 1997-07-30 Janssen Pharmaceutica Nv Glass thermoplastic systems
US6342507B1 (en) 1997-09-05 2002-01-29 Isotechnika, Inc. Deuterated rapamycin compounds, method and uses thereof
US20030077229A1 (en) 1997-10-01 2003-04-24 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing cardiovascular or renal drugs
JP2001517689A (ja) 1997-10-01 2001-10-09 フレミントン ファーマシューティカル コーポレイション 極性または非極性の、バッカルスプレーまたはカプセル
ES2256959T3 (es) 1997-10-08 2006-07-16 Isotechnika,Inc. Analogos de ciclosporina deuterados y no deuterados y su uso como agentes inmunomoduladores.
AR014195A1 (es) 1997-12-29 2001-02-07 Ortho Mcneil Pharm Inc Compuestos de trifenilpropanamida utiles para el tratamiento de procesos inflamatorios, composiciones anti-inflamatorias que los comprenden, ymetodos para prepararlos
US6506766B1 (en) 1998-02-13 2003-01-14 Abbott Laboratories Glucocortiocoid-selective antinflammatory agents
US6380207B2 (en) 1998-02-13 2002-04-30 Abbott Laboratories Glucocortiocoid-selective antiinflammatory agents
ATE238792T1 (de) 1998-02-23 2003-05-15 Fujisawa Pharmaceutical Co Verwendung von makroliden zur behandlung von glaukom
EP1061908A4 (en) * 1998-03-13 2007-01-24 Merck & Co Inc COMBINATION THERAPY AND COMPOSITION FOR ACUTE CORONARY MIXING SYNDROME AND ASSOCIATED CONDITIONS
US6372254B1 (en) 1998-04-02 2002-04-16 Impax Pharmaceuticals Inc. Press coated, pulsatile drug delivery system suitable for oral administration
WO1999062537A1 (en) 1998-06-04 1999-12-09 The Rockefeller University Methods and agents for modulating the immune response and inflammation involving monocyte and dendritic cell membrane proteins
US6184226B1 (en) 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
CN1246335A (zh) 1998-08-28 2000-03-08 冀聪韬 一种用于治疗皮肤病药物的制作方法
US6602521B1 (en) 1998-09-29 2003-08-05 Impax Pharmaceuticals, Inc. Multiplex drug delivery system suitable for oral administration
US6177077B1 (en) * 1999-02-24 2001-01-23 Edward L. Tobinick TNT inhibitors for the treatment of neurological disorders
US6677326B2 (en) 1999-03-15 2004-01-13 Arakis, Ltd. Corticosteroid formulation comprising less than 2.5 mg prednisolone for once daily administration
US6383471B1 (en) 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
EA007483B1 (ru) 1999-04-30 2006-10-27 Пфайзер Продактс Инк. Применение модулятора рецепторов глюкокортикоидов и агониста рецепторов глюкокортикоидов при получении лекарственного средства
WO2001002393A1 (en) 1999-07-01 2001-01-11 Italfarmaco S.P.A. Complexes of paroxetine, with cyclodextrins or cyclodextrin derivatives
AU6078500A (en) * 1999-07-09 2001-01-30 Amgen, Inc. Combination therapy for conditions leading to bone loss
US6593480B2 (en) 1999-09-01 2003-07-15 Abbott Laboratories Glucocorticoid receptor antagonists for treatment of diabetes
JP2003509478A (ja) 1999-09-21 2003-03-11 エモリー・ユニバーシティ 血小板関連障害を処置する方法および組成物
EP1093814A1 (en) 1999-10-22 2001-04-25 Boehringer Ingelheim Pharma KG Use of dipyridamole or mopidamol in the manufacture of a medicament for the treatment and prevention of fibrin-dependent microcirculation disorders
EP1233768A1 (en) 1999-11-15 2002-08-28 Smithkline Beecham Carvedilol methanesulfonate
WO2001047572A2 (en) 1999-12-29 2001-07-05 Advanced Cardiovascular Systems, Inc. Device and active component for inhibiting formation of thrombus-inflammatory cell matrix
WO2001054679A2 (en) 2000-01-27 2001-08-02 Children's Hospital Research Foundation Transdermal composition containing an anesthetic and a vasodilator agent
DE10012555A1 (de) 2000-03-15 2001-09-20 Merck Patent Gmbh Glucocorticoide in einer Modified Release-Formulierung
US6290445B1 (en) 2000-04-18 2001-09-18 Avibank Mfg., Inc. Non-removable structural threaded fastener with threads of lesser outer diameter than the shank and method of forming same
WO2002022127A1 (en) 2000-09-13 2002-03-21 Isis Innovation Limited Use of phosphodiesterase inhibitors for the treatment of anorectal disorders
IL146057A (en) 2000-10-27 2007-09-20 Pfizer Prod Inc A process for the preparation of modulators of a non-steroidal glucocorticoid receptor
ATE327974T1 (de) 2000-10-28 2006-06-15 Pfizer Prod Inc Modulatoren des glucocorticoid-rezeptors
TWI321054B (en) 2000-12-19 2010-03-01 California Inst Of Techn Compositions containing inclusion complexes
US6583180B2 (en) 2001-02-14 2003-06-24 Abbott Laboratories Glucocorticoid receptor modulators
CA2439691A1 (en) * 2001-03-02 2002-09-12 Bristol-Myers Squibb Company Co-administration of melanocortin receptor agonist and phosphodiesterase inhibitor for treatment of cyclic-amp associated disorders
US6759058B1 (en) 2001-04-25 2004-07-06 Western Center For Drug Development College Of Pharmacy Western University Of Health Sciences Enteric-coated proliposomal formulations for poorly water soluble drugs
US6960357B2 (en) 2001-05-25 2005-11-01 Mistral Pharma Inc. Chemical delivery device
JP2004534841A (ja) * 2001-07-09 2004-11-18 コンビナトアールエックス インコーポレーティッド 炎症疾患の治療のための組合せ
GB0119848D0 (en) 2001-08-15 2001-10-10 Univ Sheffield Delayed and sustained drug release
CA2461946C (en) * 2001-10-05 2010-01-12 Combinatorx, Incorporated Combinations for the treatment of immunoinflammatory disorders
WO2003043603A1 (en) 2001-11-20 2003-05-30 Advanced Inhalation Research, Inc. Particulate compositions for improving solubility of poorly soluble agents
US20030157061A1 (en) * 2001-12-05 2003-08-21 Pharmacia Corporation Combinations of a cyclooxygenase-2 selective inhibitor and a TNFalpha antagonist and therapeutic uses therefor
AU2003214879B2 (en) 2002-01-22 2008-02-07 The Regents Of The University Of California Non-steroidal ligands for the glucocorticoid receptor, compositions and uses thereof
JP2005519923A (ja) * 2002-02-04 2005-07-07 ファルマシア・コーポレーション シクロオキシゲナーゼ−2選択的阻害剤ならびに風邪および咳止め薬の組み合わせを用いる風邪および咳の治療ならびにそれらの組成物
BR0312511A (pt) 2002-07-10 2005-04-12 Arachnova Therapeutics Ltd 4- (2-fluorofenil)-6-metil-2- (1-piperazinil) tieno [2,3-d] pirimidina no tratamento de distúrbio funcional dos intestinos
US20040180812A1 (en) 2002-12-13 2004-09-16 Technology Center Methods of treating and preventing proliferative disease
JP2006515320A (ja) 2002-12-31 2006-05-25 ラリー エル オーグスバーガー 活性クッション性成分を含有する医薬品剤形の製造方法
CN1747734A (zh) 2003-02-07 2006-03-15 贝林格尔·英格海姆国际有限公司 潘生丁或单哌潘生丁在治疗和预防mmp-9-依赖性疾病中的用途
US20050058688A1 (en) 2003-02-22 2005-03-17 Lars Boerger Device for the treatment and prevention of disease, and methods related thereto
WO2005020933A2 (en) 2003-09-02 2005-03-10 University Of South Florida Nanoparticles for drug-delivery
AU2004275777A1 (en) * 2003-09-24 2005-04-07 Combinatorx, Incorporated Therapeutic regimens for administering drug combinations
TW200517114A (en) 2003-10-15 2005-06-01 Combinatorx Inc Methods and reagents for the treatment of immunoinflammatory disorders
US20070213308A1 (en) 2006-01-26 2007-09-13 Lessem Jan N Methods, compositions, and kits for the treatment of musculoskeletal disorders and symptoms associated therewith
AU2007211101B2 (en) 2006-01-27 2013-05-02 Adare Pharmaceuticals, Inc. Drug delivery systems comprising weakly basic drugs and organic acids
JP2009529053A (ja) 2006-03-07 2009-08-13 コンビナトアールエックス インコーポレーティッド 免疫炎症性障害の処置のための組成物および方法
CA2737131A1 (en) 2007-09-19 2009-03-26 Zalicus Inc. Therapeutic regimens for the treatment of immunoinflammatory disorders

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030125343A1 (en) * 1999-12-27 2003-07-03 Carlo Gambacorti-Passerini Combinations of receptor tyrosine kinase inhibitor with an a1-acidic glycoprotein binding compound

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060234991A1 (en) * 2001-10-05 2006-10-19 Curtis Keith Combinations for the treatment of immunoinflammatory disorders
US7915265B2 (en) 2001-10-05 2011-03-29 Zalicus Inc. Combinations for the treatment of immunoinflammatory disorders
US20110223621A1 (en) * 2001-10-05 2011-09-15 Curtis Keith Combinations for the treatment of immunoinflammatory disorders
US7704527B2 (en) 2002-10-25 2010-04-27 Collegium Pharmaceutical, Inc. Modified release compositions of milnacipran
US8021687B2 (en) 2002-10-25 2011-09-20 Collegium Pharmaceutical, Inc. Modified release compositions of milnacipran
US20100196472A1 (en) * 2002-10-25 2010-08-05 Collegium Pharmaceutical, Inc. Modified release compositions of milnacipran
US20090018203A1 (en) * 2002-10-25 2009-01-15 Collegium Pharmaceutical, Inc. Modified release compositions of milnacipran
US8080553B2 (en) 2003-10-15 2011-12-20 Zalicus Inc. Methods and reagents for the treatment of immunoinflammatory disorders
US20070010502A1 (en) * 2003-10-15 2007-01-11 Combinatorx Inc. Methods and reagents for the treatment of immunoinflammatory disorders
US9827324B2 (en) 2003-12-31 2017-11-28 Cydex Pharmaceuticals, Inc. Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US10799599B2 (en) 2003-12-31 2020-10-13 Cydex Pharmaceuticals, Inc. Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US10207008B2 (en) 2003-12-31 2019-02-19 Cydex Pharmaceuticals, Inc. Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US10159752B2 (en) 2003-12-31 2018-12-25 Cydex Pharmaceuticals, Inc. Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US20070020298A1 (en) * 2003-12-31 2007-01-25 Pipkin James D Inhalant formulation containing sulfoalkyl ether gamma-cyclodextrin and corticosteroid
US7534806B2 (en) 2004-12-06 2009-05-19 Avigen, Inc. Method for treating neuropathic pain and associated syndromes
US20090209575A1 (en) * 2004-12-06 2009-08-20 Johnson Kirk W Method for treating neuropathic pain and associated syndromes
WO2007089617A3 (en) * 2006-01-26 2008-06-12 Combinatorx Inc Methods, compositions, and kits for the treatment of musculoskeletal disorders and symptoms associated therewith
WO2007139753A3 (en) * 2006-05-22 2008-02-07 Combinatorx Inc Methods and compositions for the treatment of diseases or conditions associated with increased c-reactive protein, interleukin-6, or interferon-gamma levels
US20080003213A1 (en) * 2006-05-22 2008-01-03 Jan Lessem Methods and compositions for the treatment of diseases or conditions associated with increased C-reactive protein, interleukin-6, or interferon-gamma levels
WO2007139753A2 (en) * 2006-05-22 2007-12-06 Combinatorx, Incorporated Methods and compositions for the treatment of diseases or conditions associated with increased c-reactive protein, interleukin-6, or interferon-gamma levels
WO2007146012A1 (en) * 2006-06-07 2007-12-21 Combinatorx, Incorporated Treatment of periodontal disease associated with increased c-reactive protein levels
US20080181876A1 (en) * 2007-01-30 2008-07-31 Johnson Kirk W Methods for treating acute and subchronic pain
US20080287402A1 (en) * 2007-05-03 2008-11-20 Johnson Kirk W Use of a glial attenuator to prevent amplified pain responses caused by glial priming
US20090312724A1 (en) * 2007-06-28 2009-12-17 Cydex Pharmaceuticals, Inc. Nasal and Ophthalmic Delivery of Aqueous Corticosteroid Solutions
US20090047243A1 (en) * 2007-07-17 2009-02-19 Richard Rickles Combinations for the treatment of b-cell proliferative disorders
US20090053168A1 (en) * 2007-07-17 2009-02-26 Richard Rickles Treatments of b-cell proliferative disorders
WO2009038708A1 (en) * 2007-09-19 2009-03-26 Combinatorx, Incorporated Therapeutic regimens for the treatment of immunoinflammatory disorders
US20090075955A1 (en) * 2007-09-19 2009-03-19 Combinatorx, Inc. Therapeutic regimens for the treatment of immunoinflammatory disorders
US10265375B2 (en) 2007-10-08 2019-04-23 Aurinia Pharmaceuticals Inc. Ophthalmic compositions
US8435544B2 (en) 2007-10-08 2013-05-07 Lux Biosciences, Inc. Ophthalmic compositions comprising calcineurin inhibitors or mTOR inhibitors
US8535694B2 (en) 2007-10-08 2013-09-17 Lux Biosciences, Inc. Ophthalmic compositions comprising calcineurin inhibitors or mTOR inhibitors
US20090092665A1 (en) * 2007-10-08 2009-04-09 Lux Biosciences, Inc. OPHTHALMIC COMPOSITIONS COMPRISING CALCINEURIN INHIBITORS OR mTOR INHIBITORS
US10973871B2 (en) 2007-10-08 2021-04-13 Aurinia Pharmaceuticals, Inc. Ophthalmic compositions
US20110189293A1 (en) * 2007-12-17 2011-08-04 CombinatoRx, Incoporated Therapeutic regimens for the treatment of immunoinflammatory disorders
US20100009934A1 (en) * 2008-06-09 2010-01-14 Combinatorx, Incorporated Beta adrenergic receptor agonists for the treatment of b-cell proliferative disorders
US20100092479A1 (en) * 2008-08-18 2010-04-15 Combinatorx (Singapore) Pte. Ltd. Compositions and methods for treatment of viral diseases
US9017725B2 (en) 2009-06-09 2015-04-28 Aurinia Pharmaceuticals Inc. Topical drug delivery systems for ophthalmic use
US20100310642A1 (en) * 2009-06-09 2010-12-09 Lux Biosciences, Inc. Topical Drug Delivery Systems for Ophthalmic Use
US11622991B2 (en) 2017-05-12 2023-04-11 Aurinia Pharmaceuticals Inc. Protocol for treatment of lupus nephritis

Also Published As

Publication number Publication date
US20070010502A1 (en) 2007-01-11
US20120058979A1 (en) 2012-03-08
AU2004281729A1 (en) 2005-04-28
ZA200603116B (en) 2008-06-25
RU2006116511A (ru) 2007-11-27
US8080553B2 (en) 2011-12-20
KR20070001060A (ko) 2007-01-03
BRPI0415397A (pt) 2006-12-19
IS8451A (is) 2006-05-11
JP2007508391A (ja) 2007-04-05
CA2542074A1 (en) 2005-04-28
CN1889956A (zh) 2007-01-03
EP2301628A1 (en) 2011-03-30
EP1680121A4 (en) 2007-12-26
WO2005037203A3 (en) 2006-03-16
IL174918A0 (en) 2008-04-13
SG147442A1 (en) 2008-11-28
EP2070550A1 (en) 2009-06-17
EP1680121A2 (en) 2006-07-19
AR046291A1 (es) 2005-11-30
NO20062003L (no) 2006-07-07
WO2005037203A2 (en) 2005-04-28
TW200517114A (en) 2005-06-01

Similar Documents

Publication Publication Date Title
US8080553B2 (en) Methods and reagents for the treatment of immunoinflammatory disorders
US20040224876A1 (en) Combination therapy for the treatment of immunoinflammatory disorders
US20100210606A1 (en) Methods and reagents for the treatment of inflammatory disorders
US20090075951A1 (en) Methods and Reagents for the Treatment of Inflammatory Disorders
WO2005079284A2 (en) Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
US20060286177A1 (en) Methods and reagents for the treatment of inflammatory disorders
US20040220153A1 (en) Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
CA2509526A1 (en) Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
RU2329037C2 (ru) Комбинированная терапия для лечения иммуновоспалительных заболеваний
MXPA06004258A (en) Methods and reagents for the treatment of immunoinflammatory disorders
ZA200502708B (en) Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
MX2007016114A (en) Combination therapy for the treatment of immunoinflammatory disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: COMINATORX, INCORPORATED, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KEITH, CURTIS;BORISY, ALEXIS;ZIMMERMANN, GRANT R.;AND OTHERS;REEL/FRAME:016271/0553;SIGNING DATES FROM 20041201 TO 20041216

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ZALICUS INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:COMBINATORX, INCORPORATED;REEL/FRAME:025150/0079

Effective date: 20100908