US20090155176A1 - Compositions and methods for treatment of diabetic retinopathy - Google Patents

Compositions and methods for treatment of diabetic retinopathy Download PDF

Info

Publication number
US20090155176A1
US20090155176A1 US12/288,330 US28833008A US2009155176A1 US 20090155176 A1 US20090155176 A1 US 20090155176A1 US 28833008 A US28833008 A US 28833008A US 2009155176 A1 US2009155176 A1 US 2009155176A1
Authority
US
United States
Prior art keywords
alkyl
aryl
lfa
substituted
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/288,330
Other languages
English (en)
Inventor
John Burnier
Thomas Gadek
Charles Semba
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.)
Novartis AG
Original Assignee
Sarcode Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US12/288,330 priority Critical patent/US20090155176A1/en
Application filed by Sarcode Corp filed Critical Sarcode Corp
Assigned to SARCODE CORPORATION reassignment SARCODE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BURNIER, JOHN, GADEK, THOMAS, SEMBA, CHARLES
Publication of US20090155176A1 publication Critical patent/US20090155176A1/en
Assigned to SARCODE BIOSCIENCE INC. reassignment SARCODE BIOSCIENCE INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SARCODE CORPORATION
Assigned to SILICON VALLEY BANK reassignment SILICON VALLEY BANK SECURITY AGREEMENT Assignors: SARCODE BIOSCIENCE INC.
Assigned to SARCODE BIOSCIENCE INC. reassignment SARCODE BIOSCIENCE INC. RELEASE OF SECURITY INTEREST Assignors: SILICON VALLEY BANK
Priority to US14/491,333 priority patent/US9447077B2/en
Priority to US14/659,789 priority patent/US10960087B2/en
Priority to US15/235,572 priority patent/US11028077B2/en
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SARCODE BIOSCIENCE INC.
Priority to US17/181,187 priority patent/US20210338839A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F9/00Methods or devices for treatment of the eyes; Devices for putting-in contact lenses; Devices to correct squinting; Apparatus to guide the blind; Protective devices for the eyes, carried on the body or in the hand
    • A61F9/0008Introducing ophthalmic products into the ocular cavity or retaining products therein
    • A61F9/0017Introducing ophthalmic products into the ocular cavity or retaining products therein implantable in, or in contact with, the eye, e.g. ocular inserts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/14Quaternary ammonium compounds, e.g. edrophonium, choline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • 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
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2821Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against ICAM molecules, e.g. CD50, CD54, CD102
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70525ICAM molecules, e.g. CD50, CD54, CD102
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/042Disorders of carbohydrate metabolism, e.g. diabetes, glucose metabolism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/16Ophthalmology
    • G01N2800/164Retinal disorders, e.g. retinopathy
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • DR diabetic retinopathy
  • DME diabetic macular edema
  • the loss of workplace and personal function subsequent to such loss of visual function can have devastating impact upon the individual and on the community surrounding that individual as a whole.
  • Nearly all individuals with diabetes demonstrate some degree of diabetic retinopathy, and the numbers of diabetic patients are increasing, therefore there is need for more effective treatments for vision loss and the symptoms of DR and associated macular edema.
  • the present invention provides methods of treating a subject suffering from diabetic retinopathy comprising administering to said subject in need thereof a therapeutically effective amount of a therapeutic agent which inhibits the interaction of LFA-1 and an ICAM.
  • a method is provided of treating a subject suffering from macular edema comprising administering to said subject in need thereof a therapeutically effective amount of a therapeutic agent which inhibits the interaction of LFA-1 and an ICAM, thereby reducing and/or preventing macular edema in an eye of said subject.
  • a method to treat diabetic retinopathy in a subject comprising performing a diabetic retinopathy diagnostic test on said subject; determining whether said subject suffers from diabetic retinopathy based on the results of said diagnostic test; and upon diagnosis of said diabetic retinopathy, administering to said subject an effective amount of a (LFA-1) antagonist in a pharmaceutically acceptable formulation.
  • a method for reducing and/or preventing post-operative ocular inflammation in a subject suffering from diabetes comprising administering to said subject in need thereof a therapeutically effective amount of a LFA-1 antagonist, thereby reducing and/or preventing post-operative inflammation in an eye of said subject.
  • post-operative inflammation is the result of vitrectomy, laser photocoagulation therapy, photodynamic therapy, or LASIK.
  • the diagnostic step is performed by imaging an eye of said subject or analysis of a biological sample of an eye of said subject.
  • the ICAM is ICAM-1, ICAM-2, or ICAM-3. In some embodiments, the ICAM is ICAM-1.
  • the therapeutic agent is an LFA-1 antagonist. In some of the embodiments of the invention, the LFA-1 antagonist binds to a high affinity binding site in the ⁇ L subunit of LFA-1 overlapping the ICAM-1 binding site. In other embodiments of the invention, the LFA-1 antagonist is directly competitive with the binding of ICAM-1 at the ⁇ L subunit of LFA-1. In some embodiments of the invention, the LFA-1 antagonist is a competitive inhibitor of the interaction between LFA-1 and ICAM-1. In some embodiments of the invention, the LFA-1 antagonist is an allosteric antagonist of the binding of ICAM-1 at the ⁇ L subunit of LFA-1.
  • the diabetic retinopathy is non-proliferative. In some of the inventions, the diabetic retinopathy is proliferative. In some embodiments of the invention, damage resulting from diabetic retinopathy is macular edema, retinal neovascularization, fibrovascular growth over a retina, loss of vision, basement membrane thickening, retinal edema, or retinal ischemia.
  • an LFA antagonist is provided which is an antibody. In some of the embodiments of the invention, an LFA antagonist is provided which is a compound of Formula I, II, III, IV, V or VI.
  • the compound of Formula II is provided which contains a stereochemistry as in Formula II′.
  • the compound of Formula III is provided which contains a stereochemistry as in Formula III′.
  • an LFA antagonist is provided which is a compound of Formula IA, IIA or IIB.
  • the LFA-1 antagonist is a compound with one of the following structures:
  • the LFA-1 antagonist is a compound of Formula VII, VIII, IX, X, or XI or enantiomers, pharmaceutically-acceptable salts, or solvates, thereof.
  • the therapeutic agent is administered topically, orally, periocularly, intraocularly, via injection, nasally, via an aerosol, via an insert, via an implanted device, or via a drop.
  • the therapeutic agent is administered in a carrier vehicle which is liquid drops, liquid wash, nebulized liquid, gel, ointment, aerosol, spray, polymer micro and nanoparticles, solution, suspension, solid, biodegradable matrix, powder, crystals, foam, or liposomes.
  • a therapeutically effective amount of said therapeutic agent is delivered to an eye of said subject via local or systemic delivery.
  • an injectable administration is performed intraocularly or periocularly.
  • administration is accomplished by administering an intra-ocular instillation of a gel, cream, powder, foam, crystals, liposomes, spray, polymer micro or nanospheres, or liquid suspension form of said compound.
  • polymer micro or nanospheres are used to deliver the therapeutic agent via periocular or intraocular injection or implantation.
  • a therapeutically effective amount of the therapeutic agent is delivered to an eye of the subject via local or systemic delivery.
  • the therapeutic agent is administered in a carrier vehicle which is liquid drops, liquid wash, nebulized liquid, gel, ointment, aerosol, spray, polymer micro and nanoparticles, solution, suspension, solid, biodegradable matrix, powder, crystals, foam, or liposomes.
  • topical administration comprises infusion of said compound to said eyes via a device selected from the group consisting of a pump-catheter system, an insert, a continuous or selective release device, a bioabsorbable implant, a continuous or sustained release formulation, and a contact lens.
  • injectable administration is performed intraocularly, intravitreally, periocularly, subcutaneously, subconjunctivally, retrobulbarly, or intracamerally.
  • Controlled release formulations are also provided for in some embodiments of the invention.
  • the compounds of the invention are formulated as prodrugs.
  • the formulation of the therapeutic agent includes no preservative.
  • the formulation of the therapeutic agent includes at least one preservative.
  • the formulation of the therapeutic agent includes a thickening agent.
  • the formulation of the therapeutic agent uses PLGA micro- or nanoparticles.
  • the compound is administered to the subject in an amount sufficient to achieve intraocular or retinal concentrations of from about 1 ⁇ 10 ⁇ 8 to about 1 ⁇ 10 ⁇ 1 moles/liter. In some embodiments of the invention, the compound is administered at least once a year. In other embodiments of the invention, the compound is administered at least once a day. In other embodiments of the invention, the compound is administered at least once a week. In some embodiments of the invention, the compound is administered at least once a month.
  • a second therapeutic agent is administered prior to, in combination with, at the same time, or after administration of the LFA-1 antagonist.
  • the second therapeutic agent is selected from the group consisting of antioxidants, antiinflammatory agents, antimicrobials, steroids, protein kinase C inhibitors, angiotensin converting enzyme inhibitors, antiangiogenic agents, complement inhibitors, and anti-apoptotic agents.
  • the second therapeutic agent is an anti-adhesion therapeutic agent that binds to an allosteric binding site on LFA-1.
  • the second therapeutic agent is an anti-adhesion therapeutic antibody or antibody fragment.
  • a diagnostic test is included in a method of treatment with an LFA-1 antagonist.
  • a diagnostic test for diabetic retinopathy is performed and after a diagnosis of the disease is made, the subject is administered an LFA-1 antagonist as described herein.
  • the diagnostic test is performed by imaging an eye of the subject or analysis of a biological sample of an eye of the subject.
  • the invention provides a pharmaceutical composition formulated for ocular delivery comprising a therapeutic agent which inhibits the interaction of LFA-1 and an ICAM and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises a therapeutic agent which inhibits the interaction of LFA-1 and an ICAM, which is a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, or XI.
  • the pharmaceutical composition is suitable for topical administration.
  • the pharmaceutical composition is suitable for administration via injection.
  • the pharmaceutical composition is suitable for administration suitable for administration as an implant.
  • compounds are provided for use in the methods of the invention.
  • Compounds that are useful in the methods of the invention include antibodies, fragments of antibodies, polypeptides, peptides, polymers, and organic small molecules.
  • the antibody Raptiva is used in an ocular formulation to treat diabetic retinopathy.
  • FIG. 1 depicts rolling, adhesion of leukocytes and transendothelial migration resulting from LFA-1:ICAM-1 interaction.
  • FIG. 2 depicts antigen activation of the LFA-1:ICAM-1 interaction.
  • FIG. 3 depicts co-stimulatory function of the LFA-1:ICAM-1 interaction.
  • FIG. 4 depicts small molecule antagonists useful in the methods of identification.
  • FIG. 5 depicts SDS-PAGE analysis of compound 5 crosslinked LFA-1.
  • FIG. 6 depicts binding of compound 2B and ICAM-1-Ig to 293 cells expressing wild type LFA-1 or LFA-1 lacking the I domain.
  • FIG. 7 depicts antagonist competition by compounds 2A, 3, A-286982 and sICAM-1 in the LFA-1/ICAM-1 and LFA-1/small molecule ELISAs.
  • FIG. 8 depicts correlation of IC50 values from antagonist competition in the LFA-1/ICAM-1 and LFA-1/small molecule ELISAs.
  • FIG. 9 depicts the effect of antagonists on ligand binding in the LFA-1/ICAM-1 and LFA-1/small molecule ELISAs.
  • FIG. 10 depicts Schild regressions of sICAM-1 and compound 3 antagonism.
  • FIG. 11 is a graphical representation of the effect of a directly competitive LFA-1 antagonist of the invention upon the release of inflammatory cytokines, in human mononucleocytes (PBMC) stimulated with staphylococcal enterotoxin B (SEB) as compared to the effect of cyclosporine-A (CsA).
  • PBMC human mononucleocytes
  • SEB staphylococcal enterotoxin B
  • CsA cyclosporine-A
  • FIG. 12 is a graphical representation of the distribution into the eye via topical application of a 14 C labeled directly competitive LFA-1 antagonist of the invention at a 30 minute timepoint and a 4 hour timepoint after administration, as measured by detection of the radiolabel.
  • DR Diabetic Retinopathy
  • LFA-1 Antagonists the Use of LFA-1 Antagonists in Treatments for DR
  • Diabetes is often described as a global disease leading to deleterious effects observed throughout the body of an individual suffering from this disease, which may increase significantly as the individual ages. Ocular complications of diabetes is a leading cause of visual loss and blindness worldwide.
  • DR diabetic retinopathy
  • Diabetic retinopathy is often divided into two categories for clinical disease management: non-proliferative (or background stage) and a later, proliferative stage.
  • Non-proliferative diabetic retinopathy demonstrates, at its outset, abnormalities of the normal microvascular architecture characterized by degeneration of retinal capillaries, formation of saccular capillary microaneurysms, pericyte deficient capillaries, and capillary occlusion and obliteration.
  • Mechanisms of action include diabetes-induced vascular inflammation leading to occlusion of the vascular lumen by leukocytes and platelets followed by the eventual death of both pericytes and endothelial cells.
  • Attraction and adhesion of leukocytes to the vascular wall by the inflammatory process cause leukocytes to adhere temporarily to the endothelium (leukostasis), release cytotoxic factors, and injure or kill the endothelial cell.
  • the damaged endothelial surface initiates platelet adherence, aggregation, microthrombi formation, vascular occlusion and ischemia.
  • Another consequence of endothelial injury is alteration in the Blood-Retinal Barrier (BRB) causing increased vascular permeability. This can be evidenced by fluorescein leakage during fluorescein angiography or retinal thickening assessed by optical coherence tomography (OCT).
  • Consequences of this leakage can be clinically significant macular edema and deposition of lipoproteins in the retina (hard exudates) contributing to retinal thickening.
  • retinal ganglion cells are lost leading towards visual loss or blindness.
  • the disrupted autoregulation and decreased retinal blood flow resulting from the changes in vasculature in endothelial cells, pericyte death, and capillary obliteration are markers for progression of DR, and leads to development of retinal ischemia, which enables development of the more severe, proliferative stage of DR.
  • Proliferative DR involves neovascularization or angiogenesis, induced by retinal ischemia of the disc or other locations of the retina. This new vasculature can cause hemorrhage of the vitreous humour and retinal detachments from accompanying contractile fibrous tissue.
  • macular edema or diabetic macular edema can develop, with severe impact on vision function. Progression of this associated disorder is predicted by retinal vascular leakage and leads to photocoagulation treatment in order to reduce the risk of vision loss. Since a large proportion of patients with diabetic retinopathy suffer from this disorder as well, it is a relevant clinical intervention target. All of these injuries or degenerative insults may lead to impairment or even complete loss of visual acuity and offer targets for therapeutic intervention. No efficient therapeutic options currently are available. Laser photocoagulation involves administering laser burns to various areas of the eye and is used in the treatment of many neovascularization-linked disorders.
  • Neovascularization in particular, is commonly treated with scatter or panretinal photocoagulation.
  • laser treatment may cause permanent blind spots corresponding to the treated areas.
  • Laser treatment may also cause persistent or recurrent hemorrhage, increase the risk of retinal detachment, or induce neovascularization or fibrosis.
  • Other treatment options for ocular-related disorders include thermotherapy, vitrectomy, photodynamic therapy, radiation therapy, surgery, e.g., removal of excess ocular tissue, and the like.
  • all available treatment options have limited therapeutic effect, require repeated, costly procedures, and/or are associated with dangerous side-effects.
  • Hyperglycemic control has not proved to be sufficient to end this progression.
  • a number of processes have been identified as contributing to retinal capillary occlusion and capillary obliteration in DR, including microthrombosis, apoptosis, and proinflammatory changes, which may be useful intervention points to prevent progression of DR and/or reverse damage already incurred.
  • an early event in initiation of the breakdown of the BRB and capillary nonperfusion appears to be leukocyte adhesion to the diabetic retinal vasculature.
  • Adherent leukocytes are temporally and spatially associated with retinal endothelial cell injury and death within one week of streptozotocin-induced experimental diabetes in rats.
  • Antibody based neutralization of ICAM-1 and CD18 has been shown to prevent both leukocyte adhesion and retinal endothelial cell injury and death.
  • LFA-1 antagonists of the invention may be useful in therapies against one or more of the pathological symptoms observed in this disease.
  • the methods of the present invention involve the inhibition of initiation and progression of diabetic retinopathy (DR) by inhibiting the interaction between LFA-1 and ICAM-1.
  • LFA-1 and ICAM-1 are molecules with extracellular receptor domains which are involved in the process of lymphocyte/leukocyte adhesion, migration and proliferation, leading to a cascade of inflammatory responses.
  • such methods provide anti-inflammatory effects in-vitro and in-vivo, e.g., as described in more detail below, and are useful in the treatment of DR.
  • LFA-1 is one of a group of leucointegrins which are expressed on most leucocytes, and is considered to be the lymphoid integrin which interacts with a number of ICAMs as ligands. Disrupting these interactions, and thus the immune/inflammatory response provides for reduction of inflammation, in particular, inflammation of the eye.
  • ICAM-1 CD54
  • ICAM-1 CD54
  • ICAM-2 ICAM-2
  • ICAM-3 ICAM-4
  • ICAM-4 adhesion receptors
  • ICAMs Intercellular Adhesion Molecules
  • LFA-1 is also referred to as ⁇ L ⁇ 2 and CD11a/CD18
  • ⁇ L ⁇ 2 and CD11a/CD18 The interaction of ICAM-1 and LFA-1 (LFA-1 is also referred to as ⁇ L ⁇ 2 and CD11a/CD18) has been shown to be involved in the processes of adhesion, leukocyte transendothelial migration, migration to sites of injury, and proliferation of lymphocytes at the activated target site, as shown in FIG. 1 .
  • cytokines/chemokines activate integrins constitutively expressed on leukocytes.
  • Blood vessel endothelial cells also upregulate ICAM-1 in response to the presence of the same cytokines/chemokines.
  • LFA-1 plays a role in creating and maintaining the immunological synapse, which may be defined as the physical structure of the interacting surfaces of T cells and Antigen Presenting Cells (APCs), as shown in FIG. 2 .
  • LFA-1 stabilizes T-cell engagement with the APC, and thus leads to activation of T cells.
  • the interaction of LFA-1 and ICAM-1 also appears to provide co-stimulatory signals to resting T cells, as shown in FIG. 3 .
  • CD4+ T-cell proliferation and cytokine synthesis are mediated by this interaction as part of the inflammatory response.
  • ICAM-1 and LFA-1 Given the role that the interaction of ICAM-1 and LFA-1 plays in immune/inflammatory response, it is desirable to modulate these interactions to achieve a desired therapeutic result (e.g., inhibition of the interaction in the event of an overactive inflammatory response). Also, since LFA-1 has several ligand partners within the ICAM family (ICAM-1, ICAM-2 and ICAM-3), involving a number of signaling pathways, in some embodiments of the invention, it is desirable to modulate these interactions selectively. In some embodiments of the invention, therapeutic agents are provided that will interfere with the association of LFA-1 with ICAM-1, ICAM-2, and/or ICAM-3 to thus modulate the respective signaling pathways for each pair of interactions.
  • the methods and compositions described herein can modulate one or more components of the pathways described herein.
  • the methods and compositions of the present invention may also intervene in either earlier or later portions of the inflammatory process as well.
  • upregulation of ICAM-1 or LFA-1 (activation) on endothelial cells or leukocytes, prior to tethering and transendothelial migration may be modulated by the methods and compositions described herein.
  • the present invention may be useful in modulating the expression of cytokines or chemokines that activate ICAM-1 and LFA-1 in the course of leukocyte trafficking, in modulating the transport of the cytokines or chemokines, in preventing transmigration of the arrested leukocyte, in modulating signaling via other mechanisms that are involved in leukocyte proliferation at the site of injury or inflammation, and the like.
  • the invention provides therapeutic agents that interfere with the association of LFA-1 with ICAM-1, which can block the adhesion, migration, proliferation, and release of inflammatory signals to surrounding tissue by immune system cells.
  • the invention provides methods of administering a therapeutic agent which inhibits the interaction between LFA-1 and an ICAM,
  • the therapeutic agent binds to either LFA-1 or binds to an ICAM.
  • the invention provides therapeutic agents that bind to LFA-1 to inhibit the association of LFA-1 with ICAM-1, ICAM-2, and/or ICAM-3 thus acting as LFA-1 antagonists.
  • the therapeutic agent provided by the invention binds at the high affinity binding site in the ⁇ L subunit overlapping the ICAM-1 binding site, which is a directly competitive antagonist of LFA-1. In some embodiments, the therapeutic agent provided by the invention inhibits the interaction between LFA-1 and ICAM-1 but does not completely block the high affinity binding site in the ⁇ L subunit overlapping the ICAM-1 binding site, and is a competitive but not a directly competitive antagonist of LFA-1. In some embodiments, the therapeutic agent provided by the invention binds at a site outside the high affinity binding site in the ⁇ L subunit overlapping the ICAM-1 binding site, and is an allosteric antagonist.
  • the therapeutic agent provided by the invention is an allosteric antagonist and is a competitive but not directly competitive antagonist of LFA-1.
  • the therapeutic agents are useful in treating diabetic retinopathy and disorders associated with that condition.
  • aliphatic includes both saturated and unsaturated, straight chain (unbranched) or branched aliphatic hydrocarbons, which are optionally substituted with one or more functional groups.
  • aliphatic is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl moieties.
  • alkyl includes straight and branched alkyl groups.
  • alkyl encompass both substituted and unsubstituted groups.
  • lower alkyl is used to indicate those alkyl groups (substituted, unsubstituted, branched or unbranched) having about 1-6 carbon atoms.
  • the alkyl, alkenyl and alkynyl groups employed in the invention contain about 1-20 aliphatic carbon atoms. In certain other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-10 aliphatic carbon atoms. In yet other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-8 aliphatic carbon atoms. In still other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-6 aliphatic carbon atoms.
  • the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-4 carbon atoms.
  • Illustrative aliphatic groups thus include, but are not limited to, for example, methyl, ethyl, n-propyl, isopropyl, allyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, sec-pentyl, isopentyl, tert-pentyl, n-hexyl, sec-hexyl, moieties and the like, which again, may bear one or more substituents.
  • Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, and the like.
  • Representative alkynyl groups include, but are not limited to, ethynyl, 2-propynyl and the like.
  • lower alkylene refers to a hydrocarbon chain which links together two other groups, i.e. is bonded to another group at either end, for example methylene, ethylene, butylene and the like.
  • a substituent is preferably from 1 to 10 carbons and more preferably from 1 to 5 carbons.
  • groups may be substituted, preferably with an amino, acetylamino (a lower alkylcarbonyl group bonded via a nitrogen atom), or cyclo lower alkyl group.
  • a saturated hydrocarbon ring preferably with a total of 3 to 10 methylenes (inclusive of the attachment carbons), more preferably 3 to 6.
  • alicyclic refers to compounds which combine the properties of aliphatic and cyclic compounds and include but are not limited to monocyclic, or polycyclic aliphatic hydrocarbons and bridged cycloalkyl compounds, which are optionally substituted with one or more functional groups.
  • alicyclic is intended herein to include, but is not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties, which are optionally substituted with one or more functional groups.
  • Illustrative alicyclic groups thus include, but are not limited to, for example, cyclopropyl, —CH 2 -cyclopropyl, cyclobutyl, —CH 2 -cyclobutyl, cyclopentyl, —CH 2 — cyclopentyl, cyclohexyl, —CH 2 -cyclohexyl, cyclohexenylethyl, cyclohexanylethyl, norbornyl moieties and the like, which again, may bear one or more substituents.
  • alkoxy refers to a saturated or unsaturated parent molecular moiety through an oxygen atom.
  • the alkyl group contains about 1-20 aliphatic carbon atoms. In certain other embodiments, the alkyl group contains about 1-10 aliphatic carbon atoms. In yet other embodiments, the alkyl group employed in the invention contains about 1-8 aliphatic carbon atoms. In still other embodiments, the alkyl group contains about 1-6 aliphatic carbon atoms. In yet other embodiments, the alkyl group contains about 1-4 aliphatic carbon atoms.
  • alkoxy examples include but are not limited to, methoxy, ethoxy, isopropoxy, n-butoxy, i-butoxy, sec-butoxy, tert-butoxy, neopentoxy, n-hexyloxy and the like.
  • lower alkoxy refers to a lower alkyl as defined above which may be branched or unbranched as also defined above and which is bonded by an oxygen to another group (i.e. alkyl ethers).
  • thioalkyl refers to a saturated or unsaturated (i.e., S-alkenyl and S-alkynyl) group attached to the parent molecular moiety through a sulfur atom.
  • the alkyl group contains about 1-20 aliphatic carbon atoms.
  • the alkyl group contains about 1-10 aliphatic carbon atoms.
  • the alkyl group employed in the invention contains about 1-8 aliphatic carbon atoms.
  • the alkyl group contains about 1-6 aliphatic carbon atoms.
  • the alkyl group contains about 1-4 aliphatic carbon atoms.
  • Examples of thioalkyl include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like.
  • lower alkylthio refers to a lower alkyl group bonded through a divalent sulfur atom, for example, a methylmercapto or an isopropylmercapto group.
  • lower alkylenethio is meant such a group which is bonded at each end.
  • alkylamino refers to a group having the structure —NHR′ wherein R′ is alkyl, as defined herein.
  • aminoalkyl refers to a group having the structure NH 2 R′—, wherein as defined herein.
  • the alkyl group contains about 1-20 aliphatic carbon atoms.
  • the alkyl group contains about 1-10 aliphatic carbon atoms.
  • the alkyl group employed in the invention contains about aliphatic carbon atoms.
  • the alkyl group contains about 1-6 aliphatic carbon atoms.
  • the alkyl group contains about 1-4 aliphatic carbon atoms.
  • alkylamino include, but are not limited to, methylamino, and the like.
  • substituents of the above-described aliphatic (and other) moieties of compounds of the invention include, but are not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkylthio; R x independently includes, but is not limited to, aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl, wherein any of the aliphatic, alicyclic, heteroaliphatic, heterocyclic, alkylaryl, or alkylheteroaryl substituents described above and herein may be substituted
  • aromatic moiety refers to a stable mono- or polycyclic, unsaturated moiety having preferably 3-14 carbon atoms, each of which may be substituted or unsubstituted.
  • aromatic moiety refers to a planar ring having p-orbitals perpendicular to the plane of the ring at each ring atom and satisfying the Huckel rule where the number of pi electrons in the ring is (4n+2) wherein n is an integer.
  • a mono- or polycyclic, unsaturated moiety that does not satisfy one or all of these criteria for aromaticity is defined herein as “non-aromatic”, and is encompassed by the term “alicyclic”.
  • heteromatic moiety refers to a stable mono- or polycyclic, unsaturated moiety having preferably 3-14 carbon atoms, each of which may be substituted or unsubstituted; and comprising at least one heteroatom selected from O, S, and N within the ring in place of a ring carbon atom).
  • heteromatic moiety refers to a planar ring comprising at least one heteroatom, having p-orbitals perpendicular to the plane of the ring at each ring atom, and satisfying the Huckel rule where the number of pi electrons in the ring is (4n+2) wherein n is an integer.
  • aromatic and heteroaromatic moieties may be attached via an alkyl or heteroalkyl moiety and thus also include -(alkyl) aromatic, -(heteroalkyl) aromatic, -(heteroalkyl) heteroaromatic, and -(heteroalkyl) heteroaromatic moieties.
  • aromatic or heteroaromatic moieties and “aromatic, (heteroalkyl) aromatic, -(heteroalkyl) heteroaromatic, and (heteroalkyl) heteroaromatic” are interchangeable.
  • Substituents include, but are not limited to, any of the previously mentioned substituents, e., the substituents recited for aliphatic moieties, or for other moieties as disclosed herein, resulting in the formation of a stable compound.
  • aryl does not differ significantly from the common meaning of the term in the art, and refers to an unsaturated cyclic moiety comprising at least one aromatic ring.
  • aryl refers to a mono- or bicyclic carbocyclic ring system having one or two aromatic rings including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, indenyl and the like.
  • heteroaryl does not differ significantly from the common meaning of the term in the art, and refers to a cyclic aromatic radical having from five to ten ring atoms of which one ring atom is selected from S, and N; zero, one or two ring atoms are additional heteroatoms independently selected from S, and N; and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms, such as, for example, pyridyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, and the like.
  • aryl and heteroaryl groups can be unsubstituted or substituted, wherein substitution includes replacement of one or more of the hydrogen atoms thereon independently with any one or more of the following moieties including, but not limited to: aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkylthio; heteroarylthio; F; Cl; Br; I; —OH; —NO 2 ; —CN; —CF 3 ; —CH 2 CF 3 ; —CHCl 2 ; —CH 2 OH; —CH 2 CH 2 OH; —CH 2 NH 2 ; —CH 2 SO 2
  • any two adjacent groups taken together may represent a 4, 5, 6, or 7-membered substituted or unsubstituted alicyclic or heterocyclic moiety. Additional examples of generally applicable substituents are illustrated by the specific embodiments shown in the Examples that are described herein.
  • cycloalkyl refers specifically to groups having three to seven, preferably three to ten carbon atoms. Suitable cycloalkyls include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like, which, as in the case of aliphatic, alicyclic, heteroaliphatic or heterocyclic moieties, may optionally be substituted with substituents including, but not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; heteroarylthio; F; Cl; Br; I; —OH; —NO 2 ; —CN; —
  • heteroaliphatic refers to aliphatic moieties in which one or more carbon atoms in the main chain have been substituted with a heteroatom.
  • a heteroaliphatic group refers to an aliphatic chain which contains one or more oxygen, sulfur, nitrogen, phosphorus or silicon atoms, e. place of carbon atoms.
  • Heteroaliphatic moieties may be linear or branched, and saturated or unsaturated.
  • heteroaliphatic moieties are substituted by independent replacement of one or more of the hydrogen atoms thereon with one or more moieties including, but not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; alkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroarylthio; F; Cl; Br; I; —OH; —NO 2 ; —CN; —CF 3 ; —CH 2 CF 3 ; —CHCl 2 ; —CH 2 OH; —CH 2 CH 2 OH; —CH 2 NH 2 ; —CH 2 SO 2 CH 3 ; —C( ⁇ O)R x ; —C( ⁇ O)N(R x ) 2 ; —OC( ⁇ O)R x ; —OCO 2 R
  • heterocycloalkyl refers to compounds which combine the properties of heteroaliphatic and cyclic compounds and include, but are not limited to, saturated and unsaturated mono- or polycyclic cyclic ring systems having 5-16 atoms wherein at least one ring atom is a heteroatom selected from S and N (wherein the nitrogen and sulfur heteroatoms may be optionally be oxidized), wherein the ring systems are optionally substituted with one or more functional groups, as defined herein.
  • heterocycloalkyl refers to a non-aromatic 5-, 6- or 7-membered ring or a polycyclic group wherein at least one ring atom heteroatom selected from S and N (wherein the nitrogen and sulfur heteroatoms may be optionally be oxidized), including, but not limited to, a bi- or tri-cyclic group, comprising fused six-membered rings having between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, wherein (i) each 5-membered ring has 0 to 2 double bonds, each 6-membered ring has 0 to 2 double bonds and each 7-membered ring has 0 to 3 double bonds, (ii) the nitrogen and sulfur heteroatoms may be optionally be oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above heterocyclic rings may be fused to an aryl or heteroaryl
  • heterocycles include, but are not limited to, heterocycles such as furanyl, pyranyl, pyrrolyl, thienyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, dioxazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, triazolyl, thiatriazolyl, thiadiazolyl, oxadiazolyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, dithiazolyl, dithiazolidinyl, tetrahydrofuryl, and benzofused derivatives thereof.
  • heterocycles such
  • a “substituted heterocycle, or heterocycloalkyl or heterocyclic” group refers to a heterocycle, or heterocycloalkyl or heterocyclic group, as defined above, substituted by the independent replacement of one, two or three of the hydrogen atoms thereon with but are not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkylthio; heteroarylthio; F; Cl; Br; I; —OH; —NO 2 ; —CN; —CF 3 ; —CH 2 CF 3 ; —CHCl 2 ; —CH 2 OH; —CH 2 CH 2
  • halo and “halogen” used herein refer to an atom selected from fluorine, chlorine, bromine and iodine.
  • haloalkyl denotes an alkyl group, as defined above, having one, two, or three halogen atoms attached thereto and is exemplified by such groups as chloromethyl, bromoethyl, trifluoromethyl, and the like.
  • amino refers to a primary (—NH 2 ), secondary (—NHR x ), tertiary (—NR x R y ), or quaternary amine (—N + R x R y R z ), where R y and R z are independently an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, as defined herein.
  • R y and R z are independently an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, as defined herein.
  • amino groups include, but are not limited to, methylamino, dimethylamino, ethylamino, diethylamino, diethylaminocarbonyl, iso-propylamino, piperidino, trimethylamino, and propylamino.
  • acyl refers to a group having the general formula —C( ⁇ O)R, where R is an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, as defined herein.
  • sulfonamido refers to a group of the general formula —SO2NRxRy where Rx and Ry are independently hydrogen, or an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic, heteroaromatic or acyl moiety, as defined herein.
  • benzamido refers to a group of the general formula PhNRx, where Rx is hydrogen, or an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic, heteroaromatic or acyl moiety, as defined herein.
  • C 1-6 alkylidene refers to a substituted or unsubstituted, linear or branched saturated divalent radical consisting solely of carbon and hydrogen atoms, having from one to six carbon atoms, having a free valence “-” at both ends of the radical.
  • C 2-6 alkylidene refers to a substituted or unsubstituted, linear or branched unsaturated divalent radical consisting solely of carbon and hydrogen atoms, having from two to six carbon atoms, having a free valence “-” at both ends of the radical, and wherein the unsaturation is present only as double bonds and wherein a double bond can exist between the first carbon of the chain and the rest of the molecule.
  • aliphatic As used herein, the terms “aliphatic”, “heteroaliphatic”, “alkyl”, “alkenyl”, “alkynyl”, “heteroalkyl”, “heteroalkenyl”, “heteroalkynyl”, and the like encompass substituted and unsubstituted, saturated and unsaturated, and linear and branched groups. Similarly, the terms, “alicyclic”, “heterocyclic”, heterocycloalkyl”, “heterocycle” and the like, encompass substituted and unsubstituted, and saturated and unsaturated groups.
  • cycloalkyl encompass both substituted and unsubstituted groups.
  • natural amino acid refers to any one of the common, naturally occurring L-amino acids found in naturally occurring proteins: glycine (Gly), alanine (Ala), valine (Val), leucine (Leu), isoleucine (Ile), lysine (Lys), arginine (Arg), histidine (His), proline (Pro), serine (Ser), threonine (Thr), phenylalanine (Phe), tyrosine (Tyr), tryptophan (Trp), aspartic acid (Asp), glutamic acid (Glu), asparagine (Asn), glutamine (Gln), cysteine (Cys) and methionine (Met).
  • unnatural amino acid refers to all amino acids which are not natural amino acids. This includes, for example, ⁇ -, ⁇ -, D-, L-amino acid residues, and compounds of the general formula:
  • side chain R is other than the amino acid side chains occurring in nature.
  • amino acid encompasses natural amino acids and unnatural amino acids.
  • bioisosteres generally refers to two or more compounds or moieties that possess similar molecular shapes and/or volumes. In certain embodiments, bioisosteres have approximately the same distribution of electrons. In certain other embodiments, bioisosteres exhibit similar biological properties. In preferred embodiments, bioisosteres possess similar molecular shapes and volumes; have approximately the same distribution of electrons; and exhibit similar biological properties.
  • pharmaceutically acceptable derivative denotes any pharmaceutically acceptable salt, ester, or salt of such ester, of such compound, or any other adduct or derivative which, upon administration to a subject, is capable of providing (directly or indirectly) a compound as otherwise described herein, or a metabolite or residue thereof.
  • Pharmaceutically acceptable derivatives thus include among others pro-drugs.
  • a pro-drug is a derivative of a compound, usually with significantly reduced pharmacological activity, which contains an additional moiety, which is susceptible to removal in vivo yielding the parent molecule as the pharmacologically active species.
  • An example of a pro-drug is an ester, which is cleaved in vivo to yield a compound of interest.
  • Pro-drugs of a variety of compounds, and materials and methods for derivatizing the parent compounds to create the pro-drugs, are known and may be adapted to the present invention. Certain exemplary pharmaceutical compositions and pharmaceutically acceptable derivatives will be discussed in more detail herein below.
  • the term pharmaceutically acceptable salt refers to those salts which are suitable for pharmaceutical use, preferably for use in the tissues of humans and lower animals without undue irritation, allergic response and the like.
  • Pharmaceutically acceptable salts of amines, carboxylic acids, and other types of compounds are well known in the art. For example, S. M. Berge, et al. describe pharmaceutically acceptable salts in detail in J Pharmaceutical Sciences, 66: 1-19 (1977), incorporated herein by reference.
  • the salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting a free base or free acid function with a suitable reagent, as described generally below. For example, a free base function can be reacted with a suitable acid.
  • suitable pharmaceutically acceptable salts thereof may, include metal salts such as alkali metal salts, e.g. sodium or potassium salts; and alkaline earth metal salts, e.g. calcium or magnesium salts.
  • suitable pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hernisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate,
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, sulfonate and aryl sulfonate.
  • ester refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic alcohol compounds, particularly alkanes, alkenes, ethylene glycol, cycloalkanes, and the like in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. These are exemplary only and in no way limit the possibilities of esters known in the art.
  • prodrugs refers to those prodrugs of the compounds of the present invention which are suitable for pharmaceutical use, preferably for use with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A. C. S. Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
  • Antagonists which are directly competitive antagonists of the LFA-1 interaction with ICAM-1, at a high affinity binding site in the ⁇ L subunit of LFA-1 overlapping the ICAM-1 binding site can be identified, for example, by performing competitive binding experiments as described in S. M. Keating, K. R. Clark, L. D. Stepanich, F. Arellano, C. P. Edwards, S. C. Bodary, S. A. Spencer, T. R. Gadek, J. C, Marsters Jr., M. H. Beresini, “Competition between intercellular adhesion molecule-1 and a small molecule antagonist for a common binding site on the ⁇ 1 subunit of lymphocyte function-associated antigen-1.” (2006) Protein Science, 15:290-303.
  • the binding site of small molecule antagonists of this class was identified by binding compound 5, a tritium-labeled, photoactivatable analogue of compound 3 to LFA-1 and then photocrosslinking ( FIG. 4 ).
  • compound 5 a tritium-labeled, photoactivatable analogue of compound 3 to LFA-1 and then photocrosslinking
  • FIG. 4 To maximize specific, high affinity crosslinking, it was necessary to gel filter the samples to remove unbound or weakly bound compound 5 prior to irradiation ( FIG. 5 , lanes e vs. f and g vs. h).
  • FIG. 5 lanes e vs. f and g vs. h
  • the site of crosslinking was further defined by fragmenting the affinity-labeled ⁇ L subunit with hydroxylamine, electrophoretically separating the fragments, and then performing N-terminal sequencing on the radiolabeled fragments to determine their locations within the protein sequence. Two sequences were identified, the first starting with residue 1 (sequence found: YNLDVRGARSFS (SEQ ID NO 1)) and the second with residue 30 (sequence found: GVIVGAPGEGNST (SEQ ID NO 2)). Both peptides were approximately 500 amino acids long as judged by their sizes on SDS-PAGE (50-60 kDa); this fragment size is consistent with the next two predicted cleavage sites (N-G) for hydroxylamine, N507 and N530. No label was incorporated into the C-terminal half of the subunit.
  • the role of the I domain in the binding of compound 2B and related analogs to LFA-1 was demonstrated by preparing a construct of the ⁇ L subunit lacking the I domain.
  • the 132 construct alone (mock) or together with the construct lacking the I domain or wild type ⁇ L was transfected into 293 cells, and the binding of compound 2B to the transfected cells was examined ( FIG. 6 ).
  • Compound 2B showed substantial binding to the wild type ⁇ L transfected cells but demonstrated no significant binding to the cells transfected with ⁇ L lacking the I domain relative to binding to mock ( ⁇ 2) transfected cells.
  • the binding site responsible for the stabilization of LFA-1 to SDS-PAGE may reside in the I-like domain of the ⁇ subunit. As shown above, this ⁇ subunit binding site is not related to the high affinity binding site in the ⁇ subunit which is responsible for the direct competitive inhibition of ICAM-1 binding. However, the ⁇ subunit binding site responsible for LFA-1 stabilization by compound 3 may be the same as the low affinity ⁇ subunit crosslinking site.
  • the first is a high affinity binding site in the ⁇ L subunit of LFA-1 through which the small molecule and LFA-1 form a complex which is stable enough (e.g. K d ⁇ 25 nM) to survive the gel filtration process.
  • the second site is a lower affinity binding site (e.g. K d >1 M) in the ⁇ subunit which is involved with stabilization of the LFA-1 heterodimer under SDS-PAGE. This site is more dynamic by nature (i.e. faster off rate) and does not survive the gel filtration/photolysis process.
  • this second low affinity site is consistent with those of an ⁇ / ⁇ I-like allosteric antagonist binding site in the I-like domain of the ⁇ subunit.
  • the low affinity binding of the ICAM-1 mimetics described herein to the ⁇ subunit of LFA-1, presumably to the I-like domain, is likely due to the sequence homology between the I and I-like domains, particularly with regard to similarities in MIDAS motifs and their affinities for the carboxylic acid moiety common to this class of antagonists.
  • the affinity of compounds for the I-like domain in the ⁇ 2 subunit must be attenuated in order to select antagonists which are specific to LFA-1.
  • FIG. 7A Typical competition curves for these inhibitors in the ELISA are shown in FIG. 7A .
  • Compound 3 potently inhibited the binding of ICAM-1-Ig to LFA-1 with a 2 nM IC 50 .
  • Compound 2A an analogue of compound 3, inhibited binding but with an approximately 10-fold higher IC 50 value.
  • A-286982 and sICAM-1 inhibited ICAM-1-Ig binding to LFA-1 but with IC 50 values that were more than 100-fold that of compound 3.
  • IC 50 values in the LFA-1/small molecule and LFA-1/ICAM-1 ELISAs was extended to a larger set of compounds including a group of kistrin-derived peptides and small molecules representing the evolution of this class of LFA-1 small molecule antagonists.
  • FIG. 8 Correlation of IC50 values from antagonist competition in the LFA-1:ICAM-1 and LFA-1:small molecules ELISAs.
  • the IC 50 values of a diverse group of compounds (4 peptides, 5 small molecules and sICAM-1) in competition with compound 2B are plotted against the IC 50 values determined in competition with ICAM-1-Ig for binding to LFA-1.
  • the common trend in potencies between the two antagonist competition ELISAs with ICAM-1-Ig and compound 2B as ligands reveals that each compound disrupts the binding of both ICAM-1 and small molecule ligands in a mechanistically similar fashion.
  • This parallel in potency of inhibition demonstrates that ICAM-1-Ig and compound 2B are binding to the same site on LFA-1.
  • the compounds of the invention are competitive antagonists of LFA-1.
  • An antagonist which inhibits through direct competition with the ligand of interest, exhibits a non-saturable rightward shift of the ligand binding curves to higher apparent EC 50 values with increasing antagonist concentration and no reduction in the maximal binding of the ligand. Inhibition will be surmountable but will require increasing amounts of ligand in the presence of increasing concentrations of a direct competitive inhibitor.
  • directly competitive compound 3 an allosteric antagonist A-286982 and sICAM-1 on the binding curves of ICAM-1-Ig and compound 2B to LFA-1 are shown in FIG. 9 as examples of antagonists displaying direct competition.
  • ICAM-1-Ig A, C, E
  • compound 2B B, D, F
  • the antagonists were added in two-fold dilutions starting at 2.4 (A) and 2.7 (B) ⁇ M sICAM-1, 0.040 (C) and 0.10 (D) ⁇ M compound 3 and 20 (E) and 50 (F) ⁇ M A-286982.
  • the order of antagonist concentrations was, (lowest added antagonist concentration), - ⁇ -, - ⁇ -, - ⁇ -, - ⁇ -, - ⁇ - to - ⁇ - (highest antagonist concentration).
  • the nonparallel slopes for the LFA-1/ICAM-1-Ig binding curves in the presence and absence of compound 3 may be due to an inability to attain complete equilibrium under the heterogeneous ligand binding ELISA conditions with this compound.
  • increasing concentrations of compound 3 also clearly shifted the compound 2B binding curves to higher EC 50 values with no reduction in maximal binding ( FIG. 9D ).
  • Increasing concentrations of sICAM-1 also showed a similar effect ( FIG. 9B ), although the extent of the shift in the curves was limited by the maximum achievable concentration of sICAM-1 at 2.7 ⁇ M.
  • the effects of both sICAM-1 and compound 3 on ICAM-1-Ig and compound 2B binding to LFA-1 are characteristic of direct competition as described above.
  • FIGS. 9E and 9F The effect of A-286982 on ICAM-1-Ig and compound 2B binding to the receptor was clearly different ( FIGS. 9E and 9F ).
  • the ICAM-1-Ig curves were shifted rightward to higher EC 50 values; however, the maximum binding of ICAM-1-Ig to LFA-1 decreased considerably with increasing concentrations of A-286982.
  • the reduction in maximal binding and rightward shift of the ligand binding curves with increasing A-286982 concentration are reflective of allosteric inhibition as described above.
  • A-286982 causes reductions in both ligand affinity and binding capacity; this demonstrates that A-286982 is an insurmountable antagonist of ICAM-1-Ig binding.
  • Schild analysis can be also used to investigate whether a compound inhibits ligand binding through direct competition for a single binding site. This model is based upon the assumptions that equiactive responses in an assay are the result of equivalent occupancy of receptor by ligand and that maximal binding is unchanged by the presence of antagonist.
  • the dose ratio is the ratio of the EC 50 values in the presence and absence of antagonist and is a measure of the ligand concentrations leading to equiactive responses. This dose ratio was determined for each concentration of antagonist and the Schild regressions were plotted as shown in FIG. 10 .
  • a linear response with a slope of 1 in a Schild regression indicates that inhibition by an antagonist is directly competitive and reversible.
  • the Schild analysis would yield a nonlinear relationship and/or a slope that deviates significantly from 1 in the case of an allosteric inhibitor that does not result in a reduction of maximal binding.
  • the Schild regressions for both sICAM-1 and compound 3 are shown in FIG. 10 with comparable slopes of 1.26 and 1.24, respectively.
  • Schild regressions of s-ICAM-1 (- ⁇ -) and compound 3 (- ⁇ -) antagonism in the LFA-1/ICAM-1 ligand binding ELISA are plotted from the data in FIGS. 5 (A) and (C), respectively.
  • Schild analysis requires a linear regression with a slope close to 1 to demonstrate direct competitive inhibition, there is no guidance in the extensive literature as to what range of Schild values are acceptable. Slopes of 1.24 and 1.26 fall within the bounds of many published Schild values used to support competitive binding conclusions, and therefore, these slope values are not considered significantly different than 1.
  • the linearity of the regression plots and the similarity in slopes of the relationships are consistent with binding of ligand (ICAM-1-Ig) and both antagonists (sICAM-1 and compound 3) to the same site in a similar manner.
  • ICAM-1 Blocking of the ICAMs, such as for example ICAM-1, or the leukointegrins, such as for example, LFA-1, by antibodies directed against either or both of these molecules can inhibit inflammatory response.
  • ICAM-1 ICAM-1
  • LFA-1 leukointegrins
  • Previous studies have investigated the effects of anti-CD 11a MAbs on many T-cell-dependent immune functions in vitro and a number of immune responses in vivo. In vitro, anti-CD11a MAbs inhibit T-cell activation (See Kuypers T. W., Roos D. 1989 “Leukocyte membrane adhesion proteins LFA-1, CR3 and p150,95: a review of functional and regulatory aspects” Res.
  • a number of antibodies which are directed against LFA-1 may be used to treat diabetic retinopathy, including efalizumab (Raptiva).
  • Peptides have been investigated for use in reducing the interaction of LFA-1 with ICAM-1.
  • Polypeptides that do not contain an Fc region of an IgG are described in U.S. Pat. No. 5,747,035, which can be used to treat LFA-1 mediated disorders, in particular diabetic retinopathy.
  • Use of dual peptides, the first a modulator of ICAM-1 and the second a blocking peptide with a sequence obtained from LFA-1 is described in U.S. Pat. No. 5,843,885 to reduce the interactions between LFA-1 and ICAM-1.
  • Cyclic peptides have been described in U.S. Pat. No. 6,630,447 as inhibitors of the LFA-1:ICAM-1 interaction.
  • Small organic molecule generally is used to refer to organic molecules of a size comparable to those organic molecules generally used in pharmaceuticals. The term typically excludes organic biopolymers (e.g., proteins, nucleic acids, etc.). Small organic molecules most often range in size up to about 5000 Da, in some embodiments, up to about 2000 Da, or in other embodiments, up to about 1000 Da.
  • compounds useful in the methods of the present invention include compounds of Formula I:
  • R 1 and R 2 are each independently hydrogen, an amino acid side chain, —(CH 2 ) m OH, —(CH2) m aryl, —(CH2) m heteroaryl, wherein m is 0-6, —CH(R 1A )(OR 1B ), —CH(R 1A )(NHR 1B ), U-T-Q, or an aliphatic, alicyclic, heteroaliphatic or heteroalicyclic moiety optionally substituted with U-T-Q; wherein U may be absent or one of the following: —O—, —S(O) 0-2 —, —SO 2 N(R 1A ), —N(R 1A )—, —N(R 1A )C( ⁇ O)—, —N(R 1A )C( ⁇ O)—O—, —N(R 1A )C( ⁇ O)—N(R 1B )—, —N(R 1A )—SO 2 —,
  • each occurrence of R 1A and R 1B is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, —C( ⁇ O)R 1C , or —C( ⁇ O)NR 1C R 1D ; wherein each occurrence of R 1C and R 1D is independently hydrogen, hydroxyl, or an aliphatic, heteroaliphatic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and R 1E is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, —CN, —OR 1C , —NR 1C R 1D or —SO2R 1C ; where R 3 is —C( ⁇ O)OR 3A , —C( ⁇ O)H, —CH 2
  • R 28 is one of the following groups:
  • R 27 is one of the following groups:
  • R 29 is hydrogen, a pharmaceutically acceptable salt or ester.
  • Some particularly preferred embodiments of compounds of the method of the present invention are compounds of Formulae IA, IIA and IIB:
  • R 17 is hydrogen, pharmaceutically acceptable salts or esters, and R 27 is as in Formula II.
  • R 17 is hydrogen, pharmaceutically acceptable salts or esters, and R 27 is as in Formula II.
  • Compounds of this class are disclosed in U.S. Pat. No. 7,314,938.
  • Cy is an aromatic carbocycle, aromatic heterocycle or a non-aromatic carbocycle or heterocycle optionally substituted with hydroxyl (—OH), mercapto (—SH), thioalkyl, halogen (e.g. F, Cl, Br, I), oxo ( ⁇ O), thio ( ⁇ S), amino, aminoalkyl, amidine (—C(NH)—NH 2 ), guanidine (—NH 2 —C(NH)—NH 2 ), nitro, alkyl or alkoxy.
  • Cy is a 3-5 member ring.
  • Cy is a 5- or 6-member non-aromatic heterocycle optionally substituted with hydroxyl, mercapto, halogen (preferably F or Cl), oxo ( ⁇ O), thio ( ⁇ S), amino, amidine, guanidine, nitro, alkyl or alkoxy.
  • Cy is a 5-member non-aromatic heterocycle optionally substituted with hydroxyl, oxo, thio, Cl, C 1-4 alkyl (preferably methyl), or C 1-4 alkanoyl (preferably acetyl, propanoyl or butanoyl).
  • the non-aromatic heterocycle comprises one or heteroatoms (N, O or S) and is optionally substituted with hydroxyl, oxo, mercapto, thio, methyl, acetyl, propanoyl or butyl.
  • the non-aromatic heterocycle comprises at least one nitrogen atom that is optionally substituted with methyl or acetyl.
  • the non-aromatic heterocycle is selected from the group consisting of piperidine, piperazine, morpholine, tetrahydrofuran, tetrahydrothiophene, oxazolidine, thiazolidine optionally substituted with hydroxy, oxo, mercapto, thio, alkyl or alkanoyl.
  • Cy is a non-aromatic heterocycle selected from the group consisting of tetrahydrofuran-2-yl, thiazolidin-5-yl, thiazolidin-2-one-5-yl, and thiazolidin-2-thione-5-yl and pyrrolidine.
  • Cy is a 5- or 6-member aromatic carbocycle or heterocycle optionally substituted with hydroxyl, mercapto, halogen (preferably F or Cl), oxo ( ⁇ O), thio ( ⁇ S), amino, amidine, guanidine, nitro, alkyl or alkoxy.
  • Cy is a 5-member aromatic carbocycle or heterocycle optionally substituted with hydroxyl, oxo, thio, Cl, C 1-4 alkyl (preferably methyl), or C 1-4 alkanoyl (preferably acetyl, propanoyl or butanoyl).
  • the aromatic or heterocycle comprises one or heteroatoms (N, O or S) and is optionally substituted with hydroxyl, oxo, mercapto, thio, methyl, acetyl, propanoyl or butyl.
  • Cy is a 3-6 member carbocycle optionally substituted with hydroxyl, mercapto, halogen, oxo, thio, amino, amidine, guanidine, alkyl, alkoxy or acyl.
  • the carbocycle is saturated or partially unsaturated.
  • Cy is a carbocycle selected from the group consisting of cyclopropyl, cyclopropenyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl.
  • X 2 is a C 1-5 divalent hydrocarbon linker optionally having one or more carbon atoms replaced with N, O, S, SO or SO 2 and optionally being substituted with hydroxyl, mercapto, halogen, amino, aminoalkyl, nitro, oxo or thio. In a preferred embodiment X 2 will have at least one carbon atom. Replacements and substitutions may form an amide moiety (—NRC( ⁇ O)— or —C( ⁇ O)NR—) within the hydrocarbon chain or at either or both ends. X is also sulfonamide (—NRSO 2 — or —SO 2 NR), acyl, ether, thioether or amine.
  • X 2 is the group —CH 2 —NR 10 —C(O)— wherein the carbonyl —C(O)— portion thereof is adjacent (i.e. covalently bound) to Cy and R 10 is alkyl i.e. methyl or more preferably H.
  • K is a carbocycle or heterocycle optionally substituted with hydroxyl, mercapto, halogen, oxo, thio, a hydrocarbon, a halo-substituted hydrocarbon, amino, amidine, guanidine, cyano, nitro, alkoxy or acyl.
  • K is aryl or heteroaryl optionally substituted with halogen or hydroxyl.
  • K is phenyl, furan-2-yl, thiophene-2-yl, phenyl substituted with a halogen (preferably Cl) or hydroxyl, preferably at the meta position.
  • L 2 is a divalent hydrocarbon optionally having one or more carbon atoms replaced with N, O, S, SO or SO 2 and optionally being substituted with hydroxyl, halogen oxo, or thio; or three carbon atoms of the hydrocarbon are replaced with an amino acid residue.
  • L 2 is less than 10 atoms in length and more preferably 5 or less and most preferably 5 or 3 atoms in length.
  • L 2 is —CH ⁇ CH—C(O)—NR 10 —CH 2 —, —CH 2 —NR 10 —C(O)—, —C(O)—NR 10 —CH 2 —, —CH(OH)—(CH 2 ) 2 —, —(CH 2 ) 2 —CH(OH)—, —(CH 2 ) 3 —, —C(O)—NR 10 —CH(R 7 )—C(O)—NR 10 —, —NR 10 —C(O)—CH(R 16 )—NR 10 —C(O)—, —CH(OH)—CH 2 —O— or —CH(OH)—CF 2 —CH 2 — wherein each R 10 is independently H or alkyl and R 16 is an amino acid side chain.
  • Preferred amino acid side chains include non-naturally occurring side chains such as phenyl or naturally occurring side chains. Preferred side chains are those from Phe, Tyr, Ala, Gln and Asn.
  • L 2 is —CH ⁇ CH—C(O)—NR 10 —CH 2 — wherein the —CH ⁇ CH— moiety thereof is adjacent (i.e. covalently bound) to K.
  • L 2 is —CH 2 —NR 10 —C(O)— wherein the methylene moiety (—CH 2 —) thereof is adjacent to K.
  • R 5 is H, OH, amino, O-carbocycle or alkoxy optionally substituted with amino, a carbocycle, a heterocycle, or a pharmaceutically acceptable salt or ester.
  • R 5 is H, phenyl or C 1-4 alkoxy optionally substituted with a carbocycle such as phenyl.
  • R 5 is H.
  • R 5 is methoxy, ethoxy, propyloxy, butyloxy, isobutyloxy, s-butyloxy, t-butyloxy, phenoxy or benzyloxy.
  • R 5 is NH 2 .
  • R 5 is ethoxy.
  • R 5 is isobutyloxy.
  • R 5 is alkoxy substituted with amino, for example 2-aminoethoxy, N-morpholinoethoxy, N,N-dialkyaminoethoxy, quaternary ammonium hydroxy alkoxy (e.g. trimethylammoniumhydroxyethoxy).
  • R 6-9 are independently H, hydroxyl, mercapto, halogen, cyano, amino, amidine, guanidine, nitro or alkoxy; or R 7 and R 5 together form a fused carbocycle or heterocycle optionally substituted with hydroxyl, halogen, oxo, thio, amino, amidine, guanidine or alkoxy.
  • R 6 and R 7 are independently H, F, Cl, Br or I.
  • R 8 and R 9 are both H.
  • one of R 6 and R 7 is a halogen while the other is hydrogen or a halogen.
  • R 7 is Cl while R 6 , R 8 and R 9 are each H.
  • R 6 and R 7 are both Cl while R 8 and R 9 are both H.
  • R 10 is H or a hydrocarbon chain optionally substituted with a carbocycle or a heterocycle.
  • R 10 is H or alkyl i.e. methyl, ethyl, propyl, butyl, i-butyl, s-butyl or t-butyl. In a particular embodiment R 10 is H.
  • R 11 is a group of the formula
  • A is hydrogen, hydroxy, amino, or halogen and B is amino, carboxy, hydrogen, hydroxy, cyano, trifluoromethyl, halogen, lower alkyl, or lower alkoxy;
  • R 12 is a group of the formula:
  • R 13 is hydrogen, carboxy, or lower alkyl; n is 0 or 1; U 2 , V 2 , and W 2 are independently hydrogen, halogen, or lower alkyl provided U 2 and V 2 are not both hydrogen; X 3 is carbonyl, phenyl-substituted lower alkylene, imino, substituted imino, or sulfonyl; Y 2 is lower alkylene which may be substituted by one or more of amino, substituted amino, lower alkyl, or cyclo lower alkyl, or Y 2 is lower alkenylene or lower alkylenethio;
  • k is 0 or 1; when k is 1, Z 2 is hydrogen, lower alkylthio, —COOH, —CONH 2 , amino; and when k is 0 or 1, Z 2 is 1-adamantyl, diphenylmethyl, 3-[[(5-chloropyridin-2-yl)amino]carbonyl]pyrazin-2-yl, hydroxy, phenylmethoxy, 2-chloro-4-[[[(3-hydroxyphenyl)methyl]amino]carbonyl] phenyl, [2,6-dichlorophenyl)methoxy]phenyl; further when k is 0 or 1, Z 2 may be cycloalkyl or aryl containing 0 to 3 heteroatoms which may be the same or different, or a fused ring system containing two or three rings which rings are independently cycloalkyl or aryl containing 0 to 3 heteroatoms which may be the same or different, any of which rings may be unsubsti
  • R 14 is a group of the formula:
  • R 15 is hydrogen, carboxy, or lower alkyl
  • U 3 , V 3 , and W 3 are independently hydrogen, halogen; or U 3 , V 3 , and W 3 are lower alkyl provided that U 3 and V 3 are not both hydrogen
  • X 4 is carbonyl, phenyl-substituted lower alkylene, imino, substituted imino which includes cyano, or sulfonyl
  • Y 3 is lower alkenylene, lower alkylenethio, or is lower alkylene which may be substituted by amino, acetylamino, or cyclo-lower alkyl;
  • k 2 is 0 or 1; when k 2 is 1, Z is hydrogen, lower alkylthio, —COOH, —CONH 2 —, or amino; when k 2 is 0 or 1, Z 3 is 1-adamantyl, diphenylmethyl, 3-[[(5-chloropyridin-2-yl)amino]carbonyl]pyrazin-2-yl; and when k 2 is 0 or 1, Z may be cycloalkyl or aryl containing 0 to 3 heteroatoms which may be the same or different, or a fused ring system containing two or three rings which rings are independently cycloalkyl or aryl containing 0 to 3 heteroatoms which may be the same or different, any of which rings may be unsubstituted, or substituted with at least one of halogen, cyano, amino, substituted amino, aminosulfonyl, nitro, oxo, hydroxy, aryl, aryloxy, unsubstituted
  • a preferred embodiment of compounds of Formula V has the stereochemistry as indicated in Formula V′:
  • D 4 is a mono-, bi-, or tricyclic saturated, unsaturated, or aromatic ring, each ring having 5-, 6- or 7 atoms in the ring where the atoms in the ring are carbon or from one to four heteroatoms selected from the group nitrogen, oxygen, and sulfur, where any carbon or sulfur ring atom may optionally be oxidized, each ring substituted with 0-3 R 31 ;
  • L 3 is a bivalent linking group having one of the following structures
  • L 1 is oxo (—O—), S(O) s , C( ⁇ O), CR 32 , R 32 , CR 32 het, NR 30 or N,
  • L 2 is oxo (—O—), S(O) s , C( ⁇ O), C( ⁇ N—O—R 33 ), CR 34 R 34 ′, CR 34 , het NR 30 or N,
  • L 3 is oxo (—O—), S(O) s , C( ⁇ O), C( ⁇ N—O—R 33 ), CR 35 R 35 ′, CR 35 , het NR 30 or N,
  • L 4 is absent, is oxo (—O—), S(O) s , C( ⁇ O), C( ⁇ N—O—R 33 ), CR 36 R 36 ′, CR 36 , NR 30 or N,
  • L 5 is absent, oxo (—O—), S(O) s , C( ⁇ O), CR 37 R 37 ′, CR 37 , NR 30 or N, provided that only one of L 1 -L 3 may be het and that when one of L 1 -L 3 is het the other L 1 -L 5 may be absent,
  • R 32 , R 32 ′, R 34 , R 34 ′, R 35 , R 35 ′, R 36 , R 36 ′, R 37 and R 37 ′ each are independently R 38 , R 39 or U-Q-V—W,
  • R 24 and R 34 ′ separately or together may form a saturated, unsaturated or aromatic fused ring with B 3 through a substituent RP on B, the fused ring containing 5, 6 or 7 atoms in the ring and optionally containing 1-3 heteroatoms selected from the group O, S and N, where any S or N may optionally be oxidized; optionally, R 35 and R 35 separately or together and R 36 and R 36 ′ separately or together may form a saturated, unsaturated or aromatic fused ring with D 3 through a substituent R 31 on D 3 , the fused ring containing 5, 6 or 7 atoms in the ring and optionally containing 1-3 heteroatoms selected from the group O, S and N, where any S or N may optionally be oxidized;
  • each R 32 -R 37 , NR 30 or N in L 1 -L 5 together with any other R 32 -R 37 , NR 30 or N in L 1 -L 1 may form a 5, 6 or 7 member homo- or heterocycle either saturated, unsaturated or aromatic optionally containing 1-3 additional heteroatoms selected from N, O and S, where any carbon or sulfur ring atom may optionally be oxidized, each cycle substituted with 0-3 R 31 ; and where s is 0-2;
  • B is selected from the group:
  • Y 3 is CH or NR 30 ; n is 0, 1, 3, or 3:
  • G 3 is hydrogen or C 1 -C 6 alkyl, optionally G taken together with T may form a C 3 -C 6 cycloalkyl optionally substituted with —V—W;
  • T 3 is one of the following
  • U 4 is an optionally substituted bivalent radical having one of the following structures:
  • Q 4 is absent or is —O—, —S(O) s —, —SO 2 —N(R 30 )—, —N(R 30 )—, —N(R 30 )—C( ⁇ O)—, —N(R 30 )—C( ⁇ O)—N(R 30 )—,
  • s 0-2 and
  • het is a mono- or bicyclic 5, 6, 7, 9 or 10 member heterocyclic ring, each ring containing 1-4 heteroatoms selected from N, O and S, where the heterocyclic ring may be saturated, partially saturated, or aromatic and any N or S being optionally oxidized, the heterocyclic ring being substituted with 0-3 R 41 ;
  • V 4 is absent or is an optionally substituted bivalent group with one of the following structures C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, C 0 -C 6 alkyl-C 6 -C 10 aryl, and C 0 -C 6 alkyl-het;
  • W 4 is hydrogen, OR 33 , SR 42 , NR 30 R 30 , NH—C( ⁇ O)—O—R 43 , NH—C( ⁇ O)—NR n R n , NH—C( ⁇ O)—R 43 , NH—SO 2 —R 37 , NH—SO 2 —NR 30 R 30 , NH—SO 2 —NH—C( ⁇ O)—R 43 , NH—C( ⁇ O)—NH—SO 2 —R 37 , C( ⁇ O)—NH—C( ⁇ O)—O—R 43 , C( ⁇ O)—NH—C( ⁇ O)—R 43 , C( ⁇ O)—NH—C( ⁇ O)—NR 30 R 30 ′, C( ⁇ O)—NH—SO 2 —R 37 , C( ⁇ O)—NH—SO 2 —NR 30 R 30 ′, C( ⁇ S)—NR 30 R 30 ′, SO 2 —R 37 , SO 2 —O—R 37 , SO
  • R 44 is C( ⁇ O)—R 45 , C( ⁇ O)—H, CH 2 (OH), or CH 2 O—C( ⁇ O)—C 1 -C 6 alkyl;
  • R 38 is R 38′ or R 38′′ substituted with 1-3 R 38 ′;
  • R 38 ′ is hydrogen, halo (F, Cl, Br, I), cyano, isocyanate, carboxy, carboxy-C 1 -C 11 alkyl, amino, amino-C 1 -C 8 alkyl, aminocarbonyl, carboxamido, carbamoyl, carbamoyloxy, formyl, formyloxy, azido, nitro, imidazoyl, ureido, thioureido, thiocyanato, hydroxy, C 1 -C 6 alkoxy, mercapto, sulfonamido, het, phenoxy, phenyl, benzamido, tosyl, morpholino, morpholinyl, piperazinyl, piperidinyl, pyrrolinyl, imidazolyl, or indolyl;
  • R 38 ′′ is C 0 -C 10 alkyl-Q-C 0 -C 6 alkyl, C 0 -C 10 alkenyl-Q-C 0 -C 6 alkyl, C 0 -C 10 alkynyl-Q-C 0 -C 6 alkyl, C 3 -C 11 cycloalkyl-Q-C 0 -C 6 alkyl, C 3 -C 10 cycloalkenyl-Q-C 0 -C 6 alkyl, C 1 -C 6 alkyl-C 6 -C 12 aryl-Q-C 0 -C 6 alkyl, C 6 -C 10 aryl-C 1 -C 6 alkyl-Q-C 0 -C 6 alkyl, C 0 -C 6 alkyl-het-Q-C 0 -C 6 alkyl, C 0 -C 6 alkyl-Q-het-C 0 -C 6 alkyl, C
  • R 43 is hydrogen and substituted or unsubstituted C 1 -C 10 alkyl, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, C 3 -C 11 cycloalkyl, C 3 -C 10 cycloalkenyl, C 1 -C 6 alkyl-C 6 -C 12 aryl, C 6 -C 10 aryl-C 1 -C 6 alkyl, C 1 -C 6 alkyl-het, het-C 1 -C 6 alkyl, C 6 -C 12 aryl or het, where the substituents on any alkyl, alkenyl or alkynyl are 1-3 R 38 and the substituents on any aryl or het are 1-3 R 31 ; R 31 is R 40 or R 41 ;
  • R 41 is OH, OCF 3 , OR 43 , SR 42 , halo(F, Cl. Br, I), CN, isocyanate, NO 2 , CF 3 , C 0 -C 6 alkyl-NR 30 R 30 ′, C 0 -C 6 alkyl-C( ⁇ O)—NR 30 R 30 ′, C 0 -C 6 alkyl-C( ⁇ O)—R 38 , C 1 -C 8 alkyl, C 1 -C 8 alkoxy, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkenyl, C 1 -C 6 alkyl-phenyl, phenyl-C 1 -C 6 alkyl, C 1 -C 6 alkyloxycarbonyl, phenyl-C 0 -C 6 alkyloxy, C 1 -C 6 alkyl-het
  • R 42 is S—C 1 -C 6 alkyl, C( ⁇ O)—C 1 -C 6 alkyl, C( ⁇ O)—NR 30 R 30 ′, C 1 -C 6 alkyl, halo(F, Cl, Br, I)—C 1 -C 6 alkyl, benzyl or phenyl;
  • R 30 is R 43 , NH—C( ⁇ O)—O—R 43 , NH—C( ⁇ O)—R 43 , NH—C( ⁇ O)—NHR 43 , NH—SO 2 —R 46 , NH—SO 2 —NH—C( ⁇ O)—R 43 , NH—C( ⁇ O)—NH—SO 2 —R 37 , C( ⁇ O)—O—R 43 , C( ⁇ O)—R 43 , C( ⁇ O)—NHR 43 , C( ⁇ O)—NH—C( ⁇ O)—O—R 43 , C( ⁇ O)—NH—C( ⁇ O)—R 43 , C( ⁇ O)—NH—SO 2 —R 46 , C( ⁇ O)—NH—SO 2 —NHR 37 , SO 2 —R 37 , SO 2 —O—R 37 , SO—N(R 43 ) 2 , SO 2 —NH—C( ⁇ O)—O—R 43 , SO 2 —NH
  • R 30 ′ is hydrogen, hydroxy and substituted or unsubstituted C 1 -C 11 alkyl, C 1 -C 11 alkoxy, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, C 3 -C 11 cycloalkyl, C 3 -C 10 cycloalkenyl, C 1 -C 6 alkyl-C 6 -C 12 aryl, C 6 -C 10 aryl-C 1 -C 6 alkyl, C 6 -C 10 aryl-C 0 -C 6 alkyloxy, C 1 -C 6 alkyl-het, het-C 1 -C 6 alkyl, C 6 -C 12 aryl, het, C 1 -C 6 alkylcarbonyl, C 1 -C 8 alkoxycarbonyl, C 3 -C 8 cycloalkylcarbonyl, C 3 -C 8 cycloalkoxycarbonyl, C 6 -
  • R 30 and R 30 ′ taken together with the common nitrogen to which they are attached may from an optionally substituted heterocycle having one of the following structures morpholinyl, piperazinyl, thiamorpholinyl, pyrrolidinyl, imidazolidinyl, indolinyl, isoindolinyl, 1,2,3,4-tetrahydro-quinolinyl, 1,2,3,4-tetrahydro-isoquinolinyl, thiazolidinyl or azabicyclononyl, where the substituents are 1-3 R 38 ;
  • R 33 is hydrogen and substituted or unsubstituted C 1 -C 6 alkyl, C 1 -C 6 alkylcarbonyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl or benzoyl, where the substituents on any alkyl are 1-3 R 38 and the substituents on any aryl are 1-3 R 40 ; R 40 is OH, halo(F, Cl.
  • R 46 is a substituted or unsubstituted C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 6 cycloalkenyl, C 0 -C 6 alkyl-phenyl, phenyl-C 0 -C 6 alkyl, C 0 -C 6 alkyl-het or het-C 0 -C 6 alkyl,
  • R 45 is a substituted or unsubstituted hydroxy, C 1 -C 11 alkoxy, C 3 -C 12 cycloalkoxy, C 8 -C 12 aralkoxy, C 8 -C 12 arcycloalkoxy, C 6 -C 10 aryloxy, C 3 -C 10 alkylcarbonyloxyalkyloxy, C 3 -C 10 alkoxycarbonyloxyalkyloxy, C 3 -C 10 alkoxycarbonylalkyloxy, C 5 -C 10 cycloalkylcarbonyloxyalkyloxy, C 5 -C 10 cycloalkoxycarbonyloxyalkyloxy, C 5 -C 10 cycloalkoxycarbonylalkyloxy, C 5 -C 10 cycloalkoxycarbonylalkyloxy, C 8 -C 12 aryloxycarbonylalkyloxy, C 8 -C 12 aryloxycarbonyloxyalkyloxy, C 8 -C 12
  • Compounds of Formulas I-VI also include pharmaceutically acceptable salts, and esters including pro-drug compounds of Formula I-VI, where R 3A , R 5 , R 10 , R 17 , R 18 , R 19 , R 20 , R 21 , R 29 , and a carboxylic ester at R 44 may be lower alkyl or —CH 2 CH 2 —R 22 where R 22 is one of the following:
  • R 23 is hydrogen or methyl and R 24 is lower alkyl or lower cycloalkyl.
  • a preferred embodiment of compounds of Formula VI has the stereochemistry indicated in Formula VI′.
  • compounds described herein may comprise one or more asymmetric centers, and thus may comprise individual stereoisomers, individual diastereomers and any mixtures therein. Further, compounds of the invention may contain geometric isomers of double bonds, comprising Z and E isomers, and may be present as pure geometric isomers or mixtures thereof.
  • the methods of the present invention are performed with the following compounds:
  • Compounds of the present invention include the following compounds:
  • a family of novel p-arylthio cinnamides can act as allosteric antagonists of LFA-1. See Liu, G.; Link, J. T.; Pei, Z.; Reilly, E. B.; Nguyen, B.; Marsh, K. C.; Okasinski, G. F.; von Geldern, T. W.; Ormes, M.; Fowler, K.; Gallatin, M. 2000 “Discovery of novel p-arylthio cinnamides as antagonists of leukocyte function-associated antigen-1/intracellular adhesion molecule-1 interaction. 1. Identification of an additional binding pocket based on an anilino diaryl sulfide lead.” J. Med. Chem. 43, 4015-4030.
  • R 1 , R 2 , R 3 , R 4 , and R 5 are each independently hydrogen, alkyl, alkenyl, alkenoxy, alkynyl, aldehyde, alkanoyl, alkoxy, amido, amino, aryl, aryloxy, carboxy, cyano, cycloalkyl, ether, ester, halogen, heterocyclyl, hydroxy, ketone, nitro, oxo, perfluoroalkyl, sulfonyl, sulfonate, thio, or other carbonyl-containing groups
  • R 6 is unsubstituted alkyls, unsubstituted saturated cycloalkyls, unsubstituted carboxyalkyls, or unsubstituted heterocyclylalkyls, wherein the unsubstituted saturated cycloalkyls, unsubstituted carboxyalkyls, and unsubstituted heterocyclyl
  • R 1 and R 3 are selected from:
  • cinnamides selected from cis-cinnamide or trans-cinnamide defined as
  • R 8 and R 9 are each independently hydrogen, aldehyde, alkyl, alkenyl, alkynyl, alkoxy, amido, amino, aryl, carboxy, cyano, cycloalkyl, ester, ether, halogen, hydroxy, ketone, nitro, sulfonate, sulfonyl, thio, or other carbonyl-containing groups;
  • D, B, Y and Z are each independently —CR 31 ⁇ , —CR 32 R 33 —, —C(O)—, —O—, —SO 2 —, —S—, —N ⁇ , or —NR 34 —;
  • n is an integer of zero to three; and R 31 , R 32 , R 33 and R 34 are each independently hydrogen, alkyl, carboxy, hydroxyalkyl, monoalkylaminocarbonylalkyl, dialkylaminocarbonylalkyl or carboxyalkyl;
  • cyclopropyl derivatives selected from cis-cyclopropanoic acid, trans-cyclopropanoic acid, cis-cyclopropanamide and trans-cyclopropanamide defined as
  • R 35 and R 36 are each independently hydrogen, alkyl, carboxy, hydroxyalkyl, or carboxyalkyl, and
  • R 37 and R 38 are each independently hydrogen, alkyl, carboxyalkyl, monoalkylaminocarbonylalkyl, or dialkylaminocarbonylalkyl;
  • R 5 and R 9 are as defined above;
  • R 8 and R 9 are as defined above;
  • R 10 and R 11 are each independently hydrogen, alkanoyl, alkyl, alkenyl, alkynyl, alkoxy, amido, aryl, arylalkyl, carboxy, cyano, cycloalkyl, ester, ether, heterocyclyl, hydroxy, ketone, nitro, sulfonyl thio, or other carbonyl-containing groups, or
  • R 10 and R 11 are taken together with N to form a heterocyclyl group comprising at least one substituent which is independently hydrogen, alkyl, alkenyl, alkenoxy, alkynyl, aldehyde, alkanoyl, alkoxy, amido, amino, aryl, aryloxy, carboxy, cyano, cycloalkyl, ether, ester, halogen, heterocyclyl, hydroxy, ketone, nitro, oxo, perfluoroalkyl, sulfonyl, sulfonate, thio, or other carbonyl-containing group, or
  • R 1 and R 2 , and/or R 4 and R 5 are joined together to form a 5- to 7-membered cycloalkyl, aryl or heterocyclyl ring when R 3 is a cinnamide, substituent of formula VII-a, substituent of formula VII-b, or cyclopropyl derivative as defined above; or R 2 and R 3 , and/or R 3 and R 4 , and/or R 4 and R 5 are joined to form a 5- to 7-membered cycloalkyl, aryl or heterocyclyl ring when R 1 is selected from cinnamides, substituents of formula VII-a, substituents of formula VII-b, and cyclopropyl derivatives as defined above; and
  • Ar is substituted aryl or substituted heteroaryl having at least one substituent which independently is hydrogen, alkyl, alkenyl, alkenoxy, alkynyl, aldehyde, alkanoyl, alkoxy, amido, amino, aryl, aryloxy, carboxy, cyano, cycloalkyl, ether, ester, halogen, heterocyclyl, hydroxy, ketone, nitro, oxo, perfluoroalkyl, sulfonyl, sulfonate, thio, or other carbonyl-containing groups.
  • R6 is not an unsubstituted heterocyclylalkyl of the following structures:
  • Some exemplary compounds of Formula VII include:
  • X is H, cycloalkyl or phenyl, which is unsubstituted or substituted with one or more substituents which are lower alkyl, hydroxy or halogen; n is 0 or 1; and Y is phenyl, furanyl, indole or pyrrole, which all may be substituted with one or more substituents which are independently lower alkyl, lower alkoxy, halogen, (3,5-dimethylphenoxy)propoxy, or phenyl, wherein phenyl may be further substituted with one or more halogen atoms, nitro, amino or carboxyl groups.
  • the present invention provides a 3a,4,5,6-tetrahydro-pyrrolo1,2-b]-isothiazole, and its pharmaceutically acceptable salts, wherein the sulphur is in the form of a dioxide which is a compound of Formula IX, which is useful in the methods of the present invention.
  • R 1 is hydrogen, optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl, heterocyclyl, hydroxy, SH, SR 5 , cyano, halogen or amino, or
  • the dotted line is no bond and R 1 is attached to the ring system via a double bond and is oxo,
  • R 2 is hydrogen, or optionally substituted cycloalkyl, aryl, or heterocyclyl,
  • R 3 is hydrogen, COOR 6 , or aminocarbonyl, or optionally substituted alkyl, alkenyl, alkynyl, aralkyl, alkoxy, cycloalkyloxy, aryloxy, or heterocyclyloxy,
  • R 4 is hydrogen, halogen, hydroxy, SH, optionally substituted alkyl, alkenyl, alkynyl, alkoxy or alkylthio, or R 4 is a silyl group such as trialkylsilyl or trialkylsilyloxy, e.g. tri(C 1-6 )alkylsilyl(oxy), N 3 , amino, or
  • R 4 is heterocyclyl comprising at least one nitrogen atom as a heteroatom and being bound via that nitrogen atom to the compound of formula IX, or
  • R 4 is attached to the ring system by a double bond and is oxo
  • R 5 and R 6 independently of each other are alkyl, alkenyl, alkynyl, cycloalkyl, aryl or heterocyclyl,
  • the compound of Formula IX is:
  • Allosteric small molecule antagonists include statins which bind to the CD11a domain of LFA-1. See Kallen, J., Welzenbach, K., Ramage, P. Geyl, D. Kriwacki, R., Legge, G., Cottens, S., Weitz-Schmidt, G., and Hommel, U. 1999. “Structural basis for LFA-1 inhibition upon lovastatin binding to the CD11a 1-domain”, J. Mol.
  • a family of allosteric LFA-1 antagonists of Formula X are disclosed which are useful in the methods of the present invention. See U.S. Pat. No. 6,818,638.
  • a compound of Formula X is provided for use in the methods of the invention wherein,
  • each of a---b and ⁇ --- ⁇ independently, is either a single bond or a double bond;
  • R 1 is
  • R a is H, C 1-6 alkyl optionally substituted by OH or C 1-4 alkoxy, C 2-6 alkenyl or aryl-C 1-4 alkyl;
  • R 2 is OH; —O—CO—R 5 ;
  • R 4 is H or OR 19 wherein R 19 is C 1-6 alkyl, hydroxy-C 1-6 alkyl, C 1-4 alkoxy-C 1-6 alkyl, aryl-C 1-4 alkyl or C 1-4 alkoxycarbonyl-C 1-4 alkyl;
  • R 5 is C 1-8 alkyl, C 3-7 cycloalkyl, C 3-7 cycloalkyl-C 1-4 alkyl, aryl or aryl-C 1-4 alkyl; or R 5 is —O—R 6 wherein R 6 is the residue of an ⁇ -amino-acid attached to O through its carbonyl residue; or —R 5 is —CHR 7 —COR. 8 wherein R 7 is H, C 1-4 alkyl, heteroC 1-4 alkyl, C 3-7 cycloalkyl, C 3-7 cycloalkyl-C 1-4 alkyl, aryl or aryl-C 1-4 alkyl and R 8 is OH, C 1-4 alkoxy or NR 9 R 10 ;
  • each of R 9 and R 10 independently is H or C 1-4 alkyl, or R 9 and R 10 form together with the nitrogen to which they are bound, a heteroaryl group;
  • R 3 is a lactam of formula X-a:
  • R 30 is C 1-8 alkyl, C 3-7 cycloalkyl, aryl, C 3-7 cycloalkyl-C 1-4 alkyl, aryl-C 1-4 alkyl, heteroaryl, or heteroaryl-Cl 4 ;
  • R 31 is OH, C 1-4 alkoxy, C 1-4 alkyl, C 1-4 alkoxy-carbonyl-C 1-4 alkyl, hydroxy-C 1-5 alkoxy, C 1-4 alkoxy-C 1-5 alkoxy, C 1-4 alkoxy-carbonyl-C 1-4 alkyl, amino-C 1-4 alkoxy, HOOC—C 1-4 alkoxy, HOOC—C 1-4 alkyl, R 9a R 10a N—C 1-5 alkoxy wherein R 9a and R 10a are independently R 9 or R 10 .
  • aryl or “aryl-C 1-4 alkyl” appears in the above definition, it is “phenyl” or “naphthyl” optionally substituted by halogen, OH, NR 11 , R 12 , COOH, CF 3 , C 1-4 alkoxy, C 1-4 alkyl, hydroxy-C 1-4 alkyl, hydroxy-C 1 alkoxy, C 1-4 alkoxy-carbonyl, cyano or CONR 11 R 12 , each of R 11 and R 12 independently being H, C 1-4 alkyl, phenyl, naphthyl, phenyl-C 1-4 alkyl or naphthyl-C 1-4 alkyl or R 11 and R 12 together with the nitrogen to which they are bound forming heteroaryl; and wherever “heteroaryl” appears, it is a 5- or 6-membered heteroaryl optionally fused to a benzene ring; in free form or in salt form.
  • the compound of Formula X is one of the following compounds:
  • hydantoin-based inhibitors can also be used as antagonists. See Kelly, T. A., Jeanfavre, D. D., McNeil, D. W., Woska, J. R. Jr., Reilly, P. L., Mainolfi, E. A., Kishimoto, K. M., Nabozny, G. H., Zinter, R., Bormann, B.-J., and Rothlein, R. 1999. “Cutting edge: a small molecule antagonist of LFA-1-mediated cell adhesion”, J. Immunol., 163: 5173-5177. These compounds are believed to be allosteric inhibitors of LFA-1.
  • each R 17 is independently —OR 18 , —NR 18 R 19 , —C( ⁇ O)R 18 , —CO 2 R 18 , —C( ⁇ O)NR 18 R 19 , —NR 18 C( ⁇ O)R 19 , —NR 18 C( ⁇ O)OR 19 , —S(O) p R 19 , —NR 18 SO 2 R 19 , or —SO 2 NR 18 R 19 ;
  • R 18 and R 19 are independently hydrogen, alkyl, substituted alkyl, cycloalkyl, or substituted cycloalkyl; q is 1, 2, or 3; and p is 1 or 2.
  • the compound of Formula XI is:
  • the compounds of the invention may be prepared by methods well known to those skilled in the art, or disclosed in the references incorporated herein and may be purified in a number of ways, including by crystallization or precipitation under varied conditions to yield one or more polymorphs.
  • the present invention encompasses the above described inventive compounds, their polymorphs, their pharmaceutically acceptable salts, their pharmaceutically acceptable solvates, and pharmaceutically acceptable compositions containing them.
  • the above examples of preferred embodiments are meant to illustrate some of the potential therapeutic agents, and are not meant to limit the invention in any way.
  • the method of the invention can be practiced with antibodies, fragments of antibodies, peptides and other synthetic molecules that is a selective, potent and directly competitive inhibitor of the interaction between LFA-1 and ICAM-1, in order to treat the symptoms of diabetic retinopathy.
  • subject as used herein includes animals, in particular humans as well as other mammals.
  • treating includes achieving a therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder.
  • the compositions may be administered to a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • the compositions may be administered to a subject to prevent progression of physiological symptoms or of the underlying disorder.
  • the therapeutic agent is a LFA-1 antagonist.
  • the LFA-1 antagonist is a directly competitive antagonist of LFA-1.
  • the LFA-1 antagonist is a competitive antagonist of LFA-1.
  • the LFA-1 antagonist is an allosteric antagonist of LFA-1.
  • the LFA-1 antagonist can modulate inflammation mediated by leukocytes. Another embodiment of the invention treats a subject by administering a therapeutically effective amount of an antagonist of LFA-1 to modulate inflammation associated with ocular inflammation.
  • An embodiment of the invention treats a subject with symptoms of diabetic retinopathy by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to treat a subject with symptoms of Type II diabetes by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to treat a subject with symptoms of Type I diabetes by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to treat a subject by administering a therapeutically effective amount of a LFA-1 antagonist to decrease retinal edema in an eye of the subject.
  • methods are provided to treat a subject by administering a therapeutically effect amount of a LFA-1 antagonist to decrease macular edema. In other embodiments, methods are provided to treat a subject by administering a therapeutically effective amount of a LFA-1 antagonist to decrease basement membrane thickening in an eye of the subject. In yet other embodiments of the invention, methods are provided to treat a subject by administering a therapeutically effective amount of a LFA-1 antagonist to decrease retinal neovascularization in an eye of the subject. In some embodiments of the invention, methods are provided to treat a subject by administering a therapeutically effective amount of a LFA-1 antagonist to retard the loss of vision due to diabetic retinopathy.
  • methods are provided to treat a subject by administering a therapeutically effective amount of a LFA-1 antagonist to decrease retinal ischemia in an eye of the subject.
  • methods are provided to decrease fibrovascular growth over a retina in an eye of a subject suffering from diabetic retinopathy, by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to reduce retinal injury or degeneration due to diabetic retinopathy in an eye of a subject suffering from diabetic retinopathy, by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to limit non-proliferative damage to a retina in an eye of a subject suffering from diabetic retinopathy, by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to slow proliferative damage to a retina in an eye of a subject suffering from diabetic retinopathy, by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to reduce or prevent adhesion of leukocytes to capillary epithelial cells in an eye of a subject in need thereof, by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided to reduce or prevent damage from ischemic reperfusion in an eye of a subject in need thereof, by administering a therapeutically effective amount of a LFA-1 antagonist.
  • methods are provided wherein the LFA-1 antagonist which is administered to the subject suffering from diabetic retinopathy, binds to a high affinity binding site in the ⁇ L subunit of LFA-1 overlapping the ICAM-1 binding site.
  • Some embodiments of the invention utilize compounds that are directly competitive inhibitors of the LFA-1/ICAM-1 interaction.
  • Some of the embodiments of the invention utilize compounds which directly compete for a common high affinity binding site for ICAM-1 on LFA-1.
  • methods are provided which administer to a subject in need of treatment a therapeutically effective amount of a LFA-1 antagonist which is a compound of Formulas I, II, II′, III, III′, IV, IV′, V, or VI. In some embodiments, methods are provided which administer to a subject in need of treatment a therapeutically effective amount of a LFA-1 antagonist which is a compound of Formulas VII, VIII, IX, X or XI.
  • vitrectomy procedures may be utilized.
  • Dexamethansone a glucocorticoid steroid
  • an LFA-1 antagonist of the invention it may be desirable to use an LFA-1 antagonist of the invention to reduce inflammation.
  • methods are provided to reduce postoperative inflammation in diabetic subjects undergoing vitrectomy procedures by administering an LFA-1 antagonist to the subject in need thereof.
  • an LFA-1 antagonist of the invention may be administered to a subject prior to a vitrectomy procedure to prophylactically reduce post-operative inflammation.
  • an LFA-1 antagonist of the invention may be used to reduce inflammation.
  • methods are provided to reduce postoperative inflammation in diabetic subjects undergoing photodynamic therapeutic (PDT) procedures by administering an LFA-1 antagonist to the subject in need thereof.
  • an LFA-1 antagonist of the invention may be administered to a subject prior to a PDT procedure to prophylactically reduce post-operative inflammation.
  • laser photocoagulation therapy may be utilized to correct occlusion or leakiness, and may cause excessive inflammation in a diabetic subject.
  • an LFA-1 antagonist of the invention it may be desirable to use an LFA-1 antagonist of the invention to reduce inflammation.
  • methods are provided to reduce postoperative inflammation in diabetic subjects undergoing laser photocoagulation therapeutic procedures by administering an LFA-1 antagonist to the subject in need thereof.
  • an LFA-1 antagonist of the invention may be administered to a subject prior to a laser photocoagulation therapy procedure to prophylactically reduce post-operative inflammation.
  • LASIK treatment of vision defects diabetic patients are at increased risk of post-operative complications from corneal epithelial inflammation and defects than non-diabetic patients, and anti-inflammatory treatment may be indicated.
  • methods are provided to reduce inflammation due to diabetic complications of LASIK treatment in an eye of a subject thereof, by administering a LFA-1 antagonist and thereby reduce such inflammation. Administration is made post-operatively or pre-operatively to prophylactically prevent or reduce such inflammation.
  • methods are provided to reduce anterior or posterior segment complications following cataract surgery in an eye of a subject with DME, by administering a LFA-1 antagonist to a subject in need thereof.
  • methods are provided to prophylactically administer a LFA-1 antagonist to a subject with DME who is at higher risk of developing cataracts compared to a healthy subject, thereby reducing or preventing developing cataracts.
  • the methods generally involve the administration of one or more drugs for the treatment of diabetic retinopathy where the drug is delivered to or is distributed subsequent to delivery to the retina or intraocular region of the eye.
  • Combinations of agents can be used to treat diabetic retinopathy or to modulate the side-effects of one or more agents in the combination. Since the pathological events in this disease state are marked by a combination of impaired autoregulation, apoptosis, ischemia, reperfused tissue, neovascularization, and inflammatory stimuli, it may be desirable to administer the LFA-1 antagonists of the invention in combination with other therapeutic agents to additionally or synergistically intervene.
  • the second therapeutic agent is an antioxidant, antiinflammatory agent, antimicrobial, antiangiogenic agent, and/or anti-apoptotic agent.
  • an additional therapeutic agent in addition to administering a compound which directly competes for binding to LFA-1, an additional therapeutic agent may be administered which is an allosteric, but not a directly competitive, antagonist of LFA-1 as discussed above, potentially resulting in synergistic efficacy.
  • An example of such allosteric antagonist is the class of hydantoin inhibitors of LFA-1.
  • Other examples of allosteric antagonists include compounds of Formula VII, VIII, IX, X, or XI.
  • Another class of therapeutic agents which my be useful to administer in combination with, prior to, after or concomitantly with the LFA-1 antagonists of the invention is an anti-adhesion therapeutic antibody or antibody fragment.
  • VEGF inhibitor may be of use in the compositions of the invention, which include, but are not limited to 1) neutralizing monoclonal antibodies against VEGF or its receptor, 2) small molecule tyrosine kinase inhibitors of VEGF receptors, 3) soluble VEGF receptors which act as decoy receptors for VEGF, 4) ribozymes which specifically target VEGF, and 5) siRNA which specifically targets VEGF signalling proteins.
  • antibodies which are active against VEGF are, for example, e.g., Lucentis (ranibizumab), and Avastin (bevacizumab).
  • An example of an oligonucleotide drug is, e.g., Macugen (pegaptanib sodium injection).
  • Small molecule tyrosine kinase inhibitors include, for example, pazopanib, sorafenib, sutent, and the like.
  • Inflammation is induced by the vascular permeability caused by DR and by the process of leukocyte adhesion and neovascularization. Therefore, other ant-inflammatory agents may be administered in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention.
  • the anti-inflammatory agents can be chosen from corticosteroid related drugs including but not limited to dexamethasone, fluoromethalone, medrysone, betamethasone, triamcinolone, triamcinolone acetonide, prednisone, prednisolone, hydrocortisone, rimexolone, and pharmaceutically acceptable salts thereof, prednicarbate, deflazacort, halomethasone, tixocortol, prednylidene, prednival, paramethasone, methylprednisolone, meprednisone, mazipredone, isoflupredone, halopredone acetate, halcinonide, formocortal, flurandrenolide, fluprednisolone, flurprednidine acetate, fluperolone acetate, fluocortolone, fluocortin butyl, fluocinonide, fluocinolone acet
  • the antiinflammatory agents can be chosen from the group of NSAIDs including but not limited to acetaminophen, acemetacin, aceclofenac, alminoprofen, amfenac, bendazac, benoxaprofen, bromfenac, bucloxic acid, butibufen, carprofen, celecoxib, cinmetacin, clopirac, diclofenac, etodolac, etoricoxib, felbinac, fenclozic acid, fenbufen, fenoprofen, fentiazac, flunoxaprofen, flurbiprofen, ibufenac, ibuprofen, indomethacin, isofezolac, isoxicam, isoxepac, indoprofen, ketoprofen, lonazolac, loxoprofen, mefenamic acid, meclofenamic acid,
  • Another group of therapeutic agents which may be useful to administer in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention is the group of drugs which are known to inhibit retinopathy by inhibiting NFK ⁇ .
  • Some of these classes of therapeutics include PARP inhibitors, benfotiamine or other agents which intervene in blockade of AGEs (advanced glycation endproducts), aldose reductase inhibitors; iNOS inhibitors, FasL inhibitors, or angiopoeitin-1.
  • Oxidative stress may be induced in cells with impaired autoregulatory and ischemic processes induced by DR. Therefore, anti-oxidants may be useful to administer in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention.
  • Suitable anti-oxidants useful in the methods of the invention include, but are not limited to, ascorbic acid, tocopherols, tocotrienols, carobinoids, glutathione, alpha-lipoic acid, ubiquinols, bioflavonoids, carnitine, and superoxide dismutase mimetics, such as, for example, 2,2,6,6-tetramethyl-1-piperidinyloxy (TEMPO), DOXYL, PROXYL nitroxide compounds; 4-hydroxy-2,2,6,6-tetramethyl-1-piperidinyloxy (Tempol), M-40401, M-40403, M-40407, M-40419, M-40484, M-40587, M-40588, and the like.
  • TEMPO 2,2,6,6-tetramethyl-1-piperidinyloxy
  • TEMPO 2,2,6,6-tetramethyl-1-piperidinyloxy
  • DOXYL DOXYL
  • anti-apoptotic therapeutic agents may be administered in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention.
  • suitable anti-apoptotic agents are, for example, inhibitors of caspases, cathepsins, and TNF- ⁇ .
  • complement inhibitors Another class of therapeutic agents which may be useful to administer in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention are complement inhibitors.
  • the LFA-1/ICAM binding event is downstream of complement activation of ICAM upregulation in tissue and on vascular/capillary epithelial cells.
  • Administration of both a complement inhibitor and LFA-1 antagonist may permit more complete modulation of LFA-1 expressing leukocytes.
  • a complement inhibitor is Eculizumab.
  • Other complement inhibitors include, but are not limited to U.S. Pat. Nos. 7,166,568, 6,319,897, 5,843,884, 5,135,916, and 5624837.
  • Another class of therapeutic agents which may be useful to administer in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention are medications used in the management of glaucoma in patients with background DM, which includes primary, open angle, angle-closure as well as neovascular glaucoma.
  • Some of the therapeutic agents used for glaucoma include but are not limited to prostaglandin analogs such as, for example, latanoprost, bimatoprost, and travaprost; topical beta-adrenergic receptor antagonists such as, for example, timolol, levobunolol, and betaxolol; alpha 2-adrenergic agonist such as, for example, brimonidine; sympathomimetics such as for example, epinephrine or dipivifrin; miotic agents, such as, for example, pilocarpine; carbonic anhydrase inhibitors, such as, for example, dorzolamide, brinzolamide, and acetozolamide; or physostigmine.
  • prostaglandin analogs such as, for example, latanoprost, bimatoprost, and travaprost
  • topical beta-adrenergic receptor antagonists such as, for example, t
  • antimicrobial agents include, but are not limited to, penicillins, such as, for example, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, meziocillin, nafcillin, penicillin, piperacillin, ticarcillin, and the like; beta-lactamase inhibitors; carbapenems, such as, for example, ertapenem, imipenem, meropenem, and the like; cephalosporins, such as, for example, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime
  • Examples of other therapeutic agents which may be useful to administer in combination, prior to, after, or concomitantly with the LFA-1 antagonists of the invention are, include, but are not limited to: (a) anti-diabetic agents such as insulin and insulin mimetics, sulfonylureas (e.g., glyburide, meglinatide), biguanides, e.g., metformin (GlucophageTM), .alpha.-glucosidase inhibitors (acarbose), insulin sensitizers, e.g., thiazolidinone compounds, rosiglitazone (AvandiaTM), troglitazone (RezulinTM), ciglitazone, pioglitazone (ActosTM) and englitazone; (b) cholesterol lowering agents such as HMG-CoA reductase inhibitors (e.g., lovastatin, simvastatin, pravastatin,
  • the LFA-1 antagonist is present in an amount sufficient to exert a therapeutic effect to reduce symptoms of diabetic retinopathy by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate symptoms of diabetic retinopathy.
  • the LFA-1 antagonist is present in an amount sufficient to reduce retinal degeneration in a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate retinal degeneration.
  • the LFA-1 antagonist is present in an amount sufficient to decrease retinal edema in a treated eye of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate retinal edema.
  • the LFA-1 antagonist is present in an amount sufficient to decrease basement membrane thickening in a treated eye of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate basement membrane thickening.
  • the LFA-1 antagonist is present in an amount sufficient to decrease retinal neovascularization in a treated eye of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate retinal neovascularization.
  • the LFA-1 antagonist is present in an amount sufficient to decrease fibrovascular growth over a retina in a treated eye of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate fibrovascular growth over the retina.
  • the LFA-1 antagonist is present in an amount sufficient to retard loss of vision in a treated eye of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate further loss of vision.
  • the LFA-1 antagonist is present in an amount sufficient to limit non-proliferative damage to a retina of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate the non-proliferative damage to the retina.
  • the LFA-1 antagonist is present in an amount sufficient to slow proliferative damage to a retina of a subject by an average of at least about 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, more than 90%, or substantially eliminate further proliferative damage to the retina.
  • an effective amount of the LFA-1 antagonist is a daily dose of about 1 ⁇ 10- 11 , 1 ⁇ 10- 10 , 1 ⁇ 10- 9 , 1 ⁇ 10 ⁇ 8 , 1 ⁇ 10 ⁇ 7 , 1 ⁇ 10 ⁇ 6 , 1 ⁇ 10 ⁇ 5 , 1 ⁇ 10 ⁇ 4 , 1 ⁇ 10 ⁇ 3 , 1 ⁇ 10 ⁇ 2 , 1 ⁇ 10 ⁇ 1 , 1 ⁇ 10 1 , 1 ⁇ 10 2 grams.
  • Administration of the therapeutic agent may be by any suitable means.
  • the therapeutic agent is administered by oral administration.
  • the therapeutic agent is administered by transdermal administration.
  • the therapeutic agent is administered by instillation.
  • the therapeutic agent is administered by injection.
  • the therapeutic agent is administered by slow release intravitreal injection.
  • the therapeutic agent is administered by slow release intraocular implantation.
  • the therapeutic agent is administered by periocular implantation.
  • the therapeutic agent is administered topically. In some embodiments, the therapeutic agent is administered topically, via an eye drop. If combinations of agents are administered as separate compositions, they may be administered by the same route or by different routes.
  • combinations of agents are administered in a single composition, they may be administered by any suitable route. In some embodiments, combinations of agents are administered as a single composition by oral administration. In some embodiments, combinations of agents are administered as a single composition by transdermal administration. In some embodiments, the combinations of agent are administered as a single composition by injection. In some embodiments, the combinations of agent are administered as a single composition topically.
  • diagnostic procedures will be employed to identify a subject in need of treatment by the method of the invention.
  • An exemplary list of procedures that may be used to diagnose symptoms of diabetic retinopathy includes, e.g., for example, complete ophthalmic examination (which may include Amsler grid examination and slit lamp examination), fundus photography, fluorescein angiography, optical coherence tomography, non-myriadic photography, and beta scan ultrasound, conventional ocular exam, optical coherence tomography, beta scan ultrasound alone, complete ophthalmic examination with fundus photography and fluorescein angiography.
  • the antagonist of the method of the invention may be an antibody, fragment of an antibody, peptide or small molecule.
  • the LFA-1 antagonist used is a peptide which is not an antibody.
  • the, LFA-1 antagonist used is a small molecule.
  • treating or preventing the symptoms of diabetic retinopathy by using LFA-1 antagonists requires chronic therapy; therefore, small molecule inhibitors of the LFA-1/ICAM-1 interaction may be utilized as they have the potential for local administration as ocular drops with a lowered cost of goods. Similarly oral administration offers advantages in lowered cost of goods.
  • Implantable devices which may be biodegradable or bioabsorbable or biodegradable slow release or sustained release formulations implanted or injected into the eye or near the eye in periocular tissue are used for chronic therapy.
  • Another embodiment is a method of treating the symptoms of diabetic retinopathy using therapeutic agents which are suitable for formulation and administration as ocular therapeutics.
  • a cream formulation of the compounds of the invention may be useful in the local delivery of a LFA-1 antagonist to the skin.
  • Compounds useful in this regard include LFA-1 antagonists and their pro-drugs which are transformed into the active drug once within the skin.
  • a skin cream applied to the outer surface of the eyelids thus delivering a LFA-1 antagonist across the eyelid to the inner lining of the eyelid and the intervening conjunctival tissue and accessory lacrimal glands and appear in tear and thus be absorbed into the eye.
  • This form of delivery may be desirable in treating LFA-1 mediated inflammation of the eye, particularly in the treatment of diabetic retinopathy.
  • the method of the present invention may draw upon many suitable modes of administration to deliver the LFA-1 antagonist of the methods described herein. Such delivery to affected regions of the body may be achieved either via local or systemic administration. Suitable formulations and additional carriers are described in Remington “The Science and Practice of Pharmacy” (20 th Ed., Lippincott Williams & Wilkins, Baltimore Md.), the teachings of which are incorporated by reference in their entirety herein.
  • the invention provides a pharmaceutical composition for administration to a subject containing: (i) an effective amount of a therapeutic agent; and (ii) a pharmaceutical excipient suitable for oral administration.
  • the composition further contains: (iii) an effective amount of a second therapeutic agent.
  • a pharmaceutical composition of the invention can comprise any of the molecules disclosed herein.
  • the pharmaceutical composition of the invention is preferably delivered to the retina, intraocular space, ocular surface, interconnecting innervation, conjunctiva, lacrimal glands, or meibomian glands. It is envisioned that effective treatment can encompass administering therapeutic agents of the present invention via oral administration, topical administration, via injection, intranasally, rectally, transdermally, via an impregnated or coated device such as an ocular insert or implant, or iontophoretically, amongst other routes of administration.
  • the pharmaceutical composition can be injected intraocularly, periocularly, intramuscularly, intra-arterially, subcutaneously, or intravenously.
  • a pump mechanism may be employed to administer the pharmaceutical composition over a preselected period.
  • injections may be made periocularly, intraocularly, intravitreally, subconjunctively, retrobulbarly, into the sclera, or intercamerally.
  • systemic delivery is preferred.
  • the compounds of the invention can be formulated for and administered orally.
  • the composition of the invention may be administered intranasally, transdermally, or via some forms of oral administration, e.g. with use of a mouthwash or lozenge incorporating a compound of the invention that is poorly absorbed from the G.I.
  • iontophoretic or topical administration may be used.
  • compositions of the present invention may be administered to the ocular surface via a pump-catheter system, or released from within a continuous or selective release device such as, e.g., membranes such as, but not limited to, those employed in the OcusertTM System (Alza Corp, Palo Alto, Calif.).
  • a continuous or selective release device such as, e.g., membranes such as, but not limited to, those employed in the OcusertTM System (Alza Corp, Palo Alto, Calif.).
  • the pharmaceutical compositions can be incorporated within, carried by or attached to contact lenses which are then worn by the subject.
  • the pharmaceutical compositions can be sprayed onto ocular surface.
  • compositions of the present invention may be administered intraocularly or periocularly via a pump-catheter system, or released from within a continuous or selective release device.
  • the pharmaceutical compositions may also comprise biodegradable sustained, slow and/or delayed release formulations such as, for example, PLGA microspheres, microparticles or nanoparticles which may be delivered via a device as described above or injected intraocularly or periocularly.
  • the LFA-1 antagonist is administered in a single dose.
  • a single dose of a LFA-1 antagonist may also be used when it is co-administered with another substance (e.g., an analgesic) for treatment of an acute condition.
  • another substance e.g., an analgesic
  • the LFA-1 antagonist (by itself or in combination with other drugs) is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, seven times, eight times, nine times, ten times or more than ten times per day. Dosing may be about once a year, twice a year, every six months, every 4 months, every 3 months, every 60 days, once a month, once every two weeks, once a week, or once every other day. In one embodiment the drug is an analgesic. In another embodiment the LFA-1 antagonist and another therapeutic substance are administered together about once per day to about 10 times per day. In another embodiment the administration of the LFA-1 antagonist and another therapeutic substance continues for less than about 7 days. In yet another embodiment the co-administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, co-administered dosing is maintained as long as necessary, e.g., dosing for chronic inflammation.
  • compositions of the invention may continue as long as necessary.
  • a composition of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days.
  • a composition of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day.
  • a composition of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic pain.
  • the daily dose can range from about 1 ⁇ 10 ⁇ 8 g to 5000 mg.
  • Daily dose range may depend on the form of LFA-1 antagonist e.g., the esters or salts used, and/or route of administration, as described herein.
  • typical daily dose ranges are, e.g.
  • the daily dose of LFA-1 antagonist is about 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 mg. In some embodiments, the daily dose of the LFA-1 antagonist is 10 mg. In some embodiments, the daily dose of the LFA-1 antagonist is 100 mg. In some embodiments, the daily dose of LFA-1 antagonist is 500 mg. In some embodiments, the daily dose of LFA-1 antagonist is 1000 mg.
  • the typical daily dose ranges are, e.g. about 1 ⁇ 10 ⁇ 8 g to 5.0 g, or about 1 ⁇ 10 ⁇ 8 g to 2.5 g, or about 1 ⁇ 10 ⁇ 8 g to 1.00 g, or about 1 ⁇ 10 ⁇ 8 g to 0.5 g, or about 1 ⁇ 10 ⁇ 8 g to 0.25 g, or about 1 ⁇ 10 ⁇ 8 g to 0.1 g, or about 1 ⁇ 10 ⁇ 8 g to 0.05 g, or about 1 ⁇ 10 ⁇ 8 g to 0.025 g, or about 1 ⁇ 10 ⁇ 8 g to 1 ⁇ 10 ⁇ 2 g, or about 1 ⁇ 10 ⁇ 8 g to 5 ⁇ 10 ⁇ 3 g, or about 1 ⁇ 10 ⁇ 8 g to 2.5 ⁇ 10 ⁇ 3 g, or about 1 ⁇ 10 ⁇ 8 g to 1 ⁇ 10 ⁇ 3 g, or about 1 ⁇ 10 ⁇ 8 g to 5 ⁇ 10 ⁇ 4 g, or about 1 ⁇
  • the daily dose of LFA-1 antagonist is about 1 ⁇ 10 ⁇ 7 , 1 ⁇ 10 ⁇ 6 , 1 ⁇ 10 ⁇ 5 , 1 ⁇ 10 ⁇ 4 , 1 ⁇ 10 ⁇ 3 g, 1 ⁇ 10 ⁇ 2 g, 1 ⁇ 10 1 g, or 1 g. In some embodiments, the daily dose of the LFA-1 antagonist is 1 ⁇ 10 ⁇ 7 g. In some embodiments, the daily dose of the LFA-1 antagonist is 1 ⁇ 10 ⁇ 5 g. In some embodiments, the daily dose of LFA-1 antagonist is 1 ⁇ 10 ⁇ 3 g. In some embodiments, the daily dose of LFA-1 antagonist is 1 ⁇ 10 ⁇ 2 g.
  • the subject dose ranges from about 1 ⁇ 10 ⁇ 8 g to 5.0 g, or about 1 ⁇ 10 ⁇ 8 g to 2.5 g, or about 1 ⁇ 10 ⁇ 8 g to 1.00 g, or about 1 ⁇ 10 ⁇ 8 g to 0.5 g, or about 1 ⁇ 10 ⁇ 8 g to 0.25 g, or about 1 ⁇ 10 ⁇ 8 g to 0.1 g, or about 1 ⁇ 10 ⁇ 8 g to 0.05 g, or about 1 ⁇ 10 ⁇ 8 to 0.025 g, or about 1 ⁇ 10 ⁇ 8 g to 1 ⁇ 10 ⁇ 2 g, or about 1 ⁇ 10 ⁇ 8 g to 5 ⁇ 10 ⁇ 3 g, or about 1 ⁇ 10 ⁇ 8 g to 2.5 ⁇ 10 ⁇ 3 g, or about 1 ⁇ 10 ⁇ 8 g to 1 ⁇ 10 ⁇ 3 g, or about 1 ⁇ 10 ⁇ 8 g to 5 ⁇ 10 ⁇ 4 g, or about 1 ⁇ 10 ⁇ 7 g to 5.0 g, or about 1 ⁇ 10 ⁇
  • the eye drop, cream, lotion or other topical formulations of the invention release sufficient therapeutic agent intraocularly or periocularly to sustain a level of LFA-1 antagonist of at least about 10 nM, about 50 nM, about 100 nM, about 150 nM, about 200 nM, about 250 nM, about 300 nM, about 350 nM, about 500 nM, about 600 nM, about 700 nM, about 800 nM, about 900 nM, about 1 mM, about 2 mM, about 3 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 15 mM, about 20 mM or about 25 mM from dose to dose.
  • an eye drop formulation of the invention release sufficient therapeutic agent intraocularly or periocularly to achieve a level of LFA-1 antagonist in the retina of at least about 10 nM, about 50 nM, about 100 nM, about 150 nM, about 200 nM, about 250 nM, about 300 nM, about 350 nM, about 500 nM, about 600 nM, about 700 nM, about 800 nM, about 900 nM, about 1 mM, about 2 mM, about 3 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 155 mM, about 20 mM or about 25 mM from dose to dose.
  • the daily dosages may range about the range described for systemic administration or may range about the range described for topical administration.
  • a typical dose range is about 0.1 mg to about 100 mg of LFA-1 antagonist released over the dosing period.
  • about 1 mg to about 50 mg, about 1 to about 25 mg, about 5 mg to about 100 mg, about 5 to about 50 mg, about 5 to about 25 mg, about 10 mg to about 100 mg, about 10 mg to about 50 mg, about 10 mg to about 25 mg, or about 15 mg to about 50 mg is released over the dosing period.
  • the dosing period for slow release intraocular or periocular devices and formulations typically range from about 10 days to about 1 year, about 30 days to about 1 year, about 60 days to about 1 year, about 3 months to about 1 year, about 4 months to about 1 year, about 5 months to about 1 year, or about 6 months to about 1 year.
  • the slow release intraocular or periocular devices and formulations release therapeutic agent over the period of about 1 month to about 9 months, about 1 month to about 8 months, about 1 month to about 7 months, about 1 month, to about 6 months, about 1 month to about 5 months, about 1 month to about 4 months, or about 1 month to about 3 months.
  • the slow release formulations and devices release therapeutic agent for up to 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 12 months, 18 months, 2 years, 30 months, or 3 years.
  • the sustained release formulation and/or implantations release sufficient therapeutic agent intraocularly or periocularly to sustain a level of LFA-1 antagonist of at least about 10 nM, about 50 nM, about 100 nM, about 150 nM, about 200 nM, about 250 nM, about 300 nM, about 350 nM, about 500 nM, about 600 nM, about 700 nM, about 800 nM, about 900 nM, about 1 mM, about 2 mM, about 3 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 15 mM, about 20 mM or about 25 mM across 1 year.
  • the sustained release formulation and/or implantations release sufficient therapeutic agent intraocularly or periocularly to sustain a level of LFA-1 antagonist of at least about 10 nM, about 50 nM, about 100 nM, about 150 nM, about 200 nM, about 250 nM, about 300 nM, about 350 nM, about 500 nM, about 600 nM, about 700 nM, about 800 nM, about 900 nM, about 1 mM, about 2 mM, about 3 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 15 mM, about 20 mM or about 25 mM across 6 months.
  • the compounds of the invention may be formulated as a sterile solution or suspension, in suitable vehicles, well known in the art. Suitable formulations and additional carriers are described in Remington “The Science and Practice of Pharmacy” (20 th Ed., Lippincott Williams & Wilkins, Baltimore Md.), the teachings of which are incorporated by reference in their entirety herein.
  • the vehicle may be chosen from those known in art to be suitable, including aqueous solutions or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
  • the formulation may also comprise polymer compositions which are biocompatible, biodegradable, such as poly(lactic-co-glycolic)acid. These materials may be made into micro or nanoparticles, loaded with drug and further coated or derivatized to provide superior sustained release performance.
  • Vehicles suitable for periocular or intraocular injection include, for example, suspensions of therapeutic agent in injection grade water, liposomes and vehicles suitable for lipophilic substances. Other vehicles for periocular or intraocular injection are well known in the art.
  • the concentration of drug may be adjusted, the pH of the solution buffered and the isotonicity adjusted to be compatible with intravenous injection, as is well known in the art.
  • any of the forms of LFA-1 may also be milled to provide more suitable properties for formulation. Milling may provide smaller particle size with greater surface area exposure, which can provide faster solubilization in-vivo or during formulation. Alternatively, milling to a smaller particle size may provide the capacity to pass through biological barriers, such as the skin or gut wall, directly, without initial solubilization, permitting use as a solid in the formulation, which may provide additional benefits of temperature stability, shelf life, ease of transport, and ease of use by the subject.
  • Oral formulations can be tablets, capsules, troches, pills, wafers, chewing gums, lozenges, aqueous solutions or suspensions, oily suspensions, syrups, elixirs, or dispersible powders or granules, and the like and may be made in any way known in the art. Oral formulations may also contain sweetening, flavoring, coloring and preservative agents. Pharmaceutically acceptable excipients for tablet forms may comprise nontoxic ingredients such as inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate, or sodium phosphate, and the like.
  • carriers which are commonly used include lactose and corn starch, and lubricating agents such as magnesium stearate are commonly added.
  • useful carriers include lactose and corn starch.
  • carriers and excipients include milk, sugar, certain types of clay, gelatin, stearic acid or salts thereof, calcium stearate, talc, vegetable fats or oils, gums and glycols.
  • Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
  • a suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10.
  • An empirical parameter used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance (“HLB” value).
  • HLB hydrophilic-lipophilic balance
  • Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
  • Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.
  • lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10.
  • HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
  • Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixture
  • preferred ionic surfactants include, by way of example: lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • Ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG-phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate, caprate,
  • Hydrophilic non-ionic surfactants may include, but not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene sterols, derivative
  • hydrophilic-non-ionic surfactants include, without limitation, PEG-10 laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG-15 stearate, PEG-32 distearate, PEG-40 stearate, PEG-100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl oleate
  • Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/vitamin derivatives; and mixtures thereof.
  • preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
  • Surfactants may be used in any formulation of the invention where its use is not otherwise contradicted. In some embodiments of the invention, the use of no surfactants or limited classes of surfactants are preferred.
  • aqueous vehicles include, but are not limited to, Ringer's solution and isotonic sodium chloride.
  • Aqueous suspensions may include suspending agents such as cellulose derivatives, sodium alginate, polyvinyl-pyrrolidone and gum tragacanth, and a wetting agent such as lecithin.
  • Suitable preservatives for aqueous suspensions include methyl and n-propyl p-hydroxybenzoate.
  • Chelating agents which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, ethylene diaminetetraacetic acid (EDTA), EDTA disodium, calcium disodium edetate, EDTA trisodium, albumin, transferrin, desferoxamine, desferal, desferoxamine mesylate, EDTA tetrasodium and EDTA dipotassium, sodium metasilicate or combinations of any of these.
  • EDTA ethylene diaminetetraacetic acid
  • EDTA disodium calcium disodium edetate
  • EDTA trisodium albumin
  • transferrin desferoxamine, desferal, desferoxamine mesylate
  • EDTA tetrasodium and EDTA dipotassium sodium metasilicate or combinations of any of these.
  • Preservatives which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, purite, peroxides, perborates, imidazolidinyl urea, diazolidinyl urea, phenoxyethanol, alkonium chlorides including benzalkonium chlorides, methylparaben, ethylparaben and propylparaben.
  • Thickening agents which can be used to form pharmaceutical compositions land dosage forms of the invention include, but are not limited to, isopropyl myristate, isopropyl palmitate, isodecyl neopentanoate, squalene, mineral oil, C 12 -C 15 benzoate and hydrogenated polyisobutene. Particularly preferred are those agents which would not disrupt other compounds of the final product, such as non-ionic thickening agents. The selection of additional thickening agents is well within the skill of one in the art.
  • Anti-oxidants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, propyl, octyl and dodecyl esters of gallic acid, butylated hydroxyanisole (BHA, usually purchased as a mixture of ortho and meta isomers), green tea extract, uric acid, cysteine, pyruvate, nordihydroguairetic acid, ascorbic acid, salts of ascorbic acid such as ascorbyl palmitate and sodium ascorbate, ascorbyl glucosamine, vitamin E (i.e., tocopherols such as a-tocopherol), derivatives of vitamin E (e.g., tocopheryl acetate), retinoids such as retinoic acid, retinol, trans-retinol, cis-retinol, mixtures of trans-retinol and cis-retinol, 3-dehydroretinol and derivatives of vitamin A (e.g
  • gastroretentive formulations When formulating compounds of the invention for oral administration, it may be desirable to utilize gastroretentive formulations to enhance absorption from the gastrointestinal (G1) tract.
  • a formulation which is retained in the stomach for several hours may release compounds of the invention slowly and provide a sustained release that may be preferred in some embodiments of the invention. Disclosure of such gastro-retentive formulations are found in Klausner, E. A.; Lavy, E.; Barta, M.; Cserepes, E.; Friedman, M.; Hoffman, A. 2003 “Novel gastroretentive dosage forms: evaluation of gastroretentivity and its effect on levodopa in humans.” Pharm. Res. 20, 1466-73, Hoffman, A.; Stepensky, D.; Lavy, E.; Eyal, S.
  • Intranasal administration may utilize an aerosol suspension of respirable particles comprised of the compounds of the invention, which the subject inhales.
  • the compound of the invention are absorbed into the bloodstream via pulmonary absorption or contact the lacrimal tissues via nasolacrimal ducts, and subsequently be delivered to the retinal tissues in a pharmaceutically effective amount.
  • the respirable particles may be solid or liquid, with suitably sized particles, as is known in the art to be effective for absorption.
  • Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.
  • any suitable formulation known in the art may be utilized, either as a solution, drop, suspension, gel, powder, cream, oil, solids, dimethylsulfoxide (DMSO)-based solutions or liposomal formulation for use in a patch or other delivery system known in the art.
  • the pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin. There are many of these penetration-enhancing molecules known to those trained in the art of topical formulation.
  • humectants e.g., urea
  • glycols e.g., propylene glycol
  • alcohols e.g., ethanol
  • fatty acids e.g., oleic acid
  • surfactants e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.glycerol monolaurate, sulfoxides, terpenes (e.g., menthol)
  • amines amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • all the formulations for topical ocular administration used in the field of ophthalmology e.g., eye drops, inserts, eye packs, impregnated contact lenses, pump delivery systems, dimethylsulfoxide (DMSO)-based solutions suspensions, liposomes, and eye ointment
  • all the formulations for external use in the fields of dermatology and otolaryngology e.g., ointment, cream, gel, powder, salve, lotion, crystalline forms, foam, and spray
  • the extraordinary solubility of some of the LFA-1 antagonists of the invention permit concentrated solution formulations which can then deliver therapeutically relevant doses to regions of the eye.
  • compositions of the present invention may be a liposomal formulation for topical or oral administration, any of which are known in the art to be suitable for the purpose of this invention.
  • Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof.
  • a lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
  • Skin protecting agents are agents that protect the skin against-chemical irritants and/or physical irritants, e.g., UV light, including sunscreens, anti-acne additives, anti-wrinkle and anti-skin atrophy agents.
  • Suitable sunscreens as skin protecting agents include 2-ethylhexyl p-methoxycinnamate, 2-ethylhexyl N,N-dimethyl-p-aminobenzoate, p-aminobenzoic acid, 2-phenylbenzimidazole-5-sulfonic acid, octocrylene, oxybenzone, homomethyl salicylate, octyl salicylate, 4,4′-methoxy-t-butyldibenzoylmethane, 4-isopropy dibenzoylmethane, 3-benzylidene camphor, 3-(4-methylbenzylidene) camphor, anthanilates, ultrafine titanium dioxide, zinc oxide, iron oxide, silica
  • Suitable anti-acne agents include salicylic acid; 5-octanoyl salicylic acid; resorcinol; retinoids such as retinoic acid and its derivatives; sulfur-containing D and L amino acids other than cysteine; lipoic acid; antibiotics and antimicrobials such as benzoyl peroxide, octopirox, tetracycline, 2,4,4′-trichloro-2′-hydroxydiphenyl ether, 3,4,4′-trichlorobanilide, azelaic acid, phenoxyethanol, phenoxypropanol, phenoxisopropanol, ethyl acetate, clindamycin and melclocycline; flavonoids; and bile salts such as scymnol sulfate, deoxycholate and cholate.
  • anti-wrinkle and anti-skin atrophy agents are retinoic acid and its derivatives, retinol, retinyl esters, salicylic acid and its derivatives, sulfur-containing D and L amino acids except cysteine, alpha-hydroxy acids (e.g., glycolic acid and lactic acid), phytic acid, lipoic acid and lysophosphatidic acid.
  • the formulations may also contain irritation-mitigating additives to minimize or eliminate the possibility of skin irritation or skin damage resulting from the other components of the composition.
  • Suitable irritation-mitigating additives include, for example: -tocopherol; monoamine oxidase inhibitors, particularly phenyl alcohols such as 2-phenyl-1-ethanol; glycerin; salicylic acids and salicylates; ascorbic acids and ascorbates; ionophores such as monensin; amphiphilic amines; ammonium chloride; N-acetylcysteine; cis-urocanic acid; capsaicin; and chloroquine.
  • the irritant-mitigating additive may be incorporated into the present formulations at a concentration effective to mitigate irritation or skin damage, typically representing not more than about 20 wt. %, more typically not more than about 5 wt. %, of the composition.
  • a dry-feel modifier is an agent which when added to an emulsion, imparts a “dry feel” to the skin when the emulsion dries.
  • Dry feel modifiers can include talc, kaolin, chalk, zinc oxide, silicone fluids, inorganic salts such as barium sulfate, surface treated silica, precipitated silica, fumed silica such as an Aerosil available from Degussa Inc. of New York, N.Y. U.S.A.
  • Another dry feel modifier is an epichlorohydrin cross-linked glyceryl starch of the type that is disclosed in U.S. Pat. No. 6,488,916.
  • antimicrobial agents to prevent spoilage upon storage, i.e., to inhibit growth of microbes such as yeasts and molds.
  • Suitable antimicrobial agents are typically selected from the group consisting of the methyl and propyl esters of p-hydroxybenzoic acid (i.e., methyl and propyl paraben), sodium benzoate, sorbic acid, imidurea, purite, peroxides, perborates and combinations thereof.
  • the formulation may also contain an aesthetic agent.
  • aesthetic agents include fragrances, pigments, colorants, essential oils, skin sensates and astringents.
  • Suitable aesthetic agents include clove oil, menthol, camphor, eucalyptus oil, eugenol, methyl lactate, bisabolol, witch hazel distillate (preferred) and green tea extract (preferred).
  • Fragrances are aromatic substances which can impart an aesthetically pleasing aroma to the sunscreen composition.
  • Typical fragrances include aromatic materials extracted from botanical sources (i.e., rose petals, gardenia blossoms, jasmine flowers, etc.) which can be used alone or in any combination to create essential oils.
  • alcoholic extracts may be prepared for compounding fragrances.
  • One or more fragrances can optionally be included in the sunscreen composition in an amount ranging from about 0.001 to about 5 weight percent, preferably about 0.01 to about 0.5 percent by weight.
  • preservatives may also be used if desired and include well known preservative compositions such as benzyl alcohol, phenyl ethyl alcohol and benzoic acid, diazolydinyl, urea, chlorphenesin, iodopropynyl and butyl carbamate, among others.
  • the compounds of the invention may be attached releasably to biocompatible polymers for use in sustained release formulations on, in or attached to inserts for topical, intraocular, periocular, or systemic administration.
  • the controlled release from a biocompatible polymer may be utilized with a water soluble polymer to form a instillable formulation, as well.
  • the controlled release from a biocompatible polymer such as for example, PLGA microspheres, microparticles or nanoparticles, may be utilized in a formulation suitable for intra ocular implantation or injection for sustained release administration, as well Any suitable biodegradable and biocompatible polymer or matrix may be used.
  • Eye drops may be prepared by dissolving the active ingredient in a sterile aqueous solution such as physiological saline, buffering solution, etc., or by combining powder compositions to be dissolved before use.
  • Other vehicles may be chosen, as is known in the art, including but not limited to: balance salt solution, saline solution, water soluble polyethers such as polyethyene glycol, polyvinyls, such as polyvinyl alcohol and povidone, cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose, petroleum derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, polymers of acrylic acid such as carboxypolymethylene gel, vegetable fats such as peanut oil and polysaccharides such as dextrans, and glycosaminoglycans such as sodium hyaluronate.
  • additives ordinarily used in the eye drops can be added.
  • Such additives include isotonizing agents (e.g., sodium chloride, etc.), buffer agent (e.g., boric acid, sodium monohydrogen phosphate, sodium dihydrogen phosphate, etc.), preservatives (e.g., benzalkonium chloride, benzethonium chloride, chlorobutanol, etc.), thickeners (e.g., saccharide such as lactose, mannitol, maltose, etc.; e.g., hyaluronic acid or its salt such as sodium hyaluronate, potassium hyaluronate, etc.; e.g., mucopolysaccharide such as chondroitin sulfate, etc.; e.g., sodium polyacrylate, carboxyvinyl polymer, crosslinked polyacrylate, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose
  • the solubility of the components of the present compositions may be enhanced by a surfactant or other appropriate co-solvent in the composition.
  • cosolvents include polysorbate 20, 60, and 80, Pluronic F68, F-84 and P-103, cyclodextrin, or other agents known to those skilled in the art.
  • co-solvents may be employed at a level of from about 0.01% to 2% by weight.
  • composition of the invention can be formulated as a sterile unit dose type containing no preservatives.
  • compositions of the invention may be packaged in multidose form.
  • Preservatives may be preferred to prevent microbial contamination during use. Suitable preservatives include: benzalkonium chloride, thimerosal, chlorobutanol, methyl paraben, propyl paraben, phenylethyl alcohol, edetate disodium, sorbic acid, sodium perborate, Onamer M, or other agents known to those skilled in the art. In the prior art ophthalmic products, such preservatives may be employed at a level of from 0.004% to 0.02%.
  • the preservative preferably benzalkonium chloride
  • the preservative may be employed at a level of from 0.001% to less than 0.01%, e.g. from 0.001% to 0.008%, preferably about 0.005% by weight. It has been found that a concentration of benzalkonium chloride of 0.005% may be sufficient to preserve the compositions of the present invention from microbial attack.
  • the amount of administration and the number of administrations of the active ingredient used in the present invention vary according to sex, age and body weight of subject, symptoms to be treated, desirable therapeutic effects, administration routes and period of treatment.
  • the formulations containing the compounds of the invention may range in concentration from about 0.0001 to 10.0 W/v %, about 0.005 to 10.0 W/v %, about 0.01 to 10.0 W/v %, about 0.05 to 10.0 W/v %, about 0.1 to 10.0 W/v %, about 0.5 to 10.0 W/v %, about 1.0 to 10.0 W/v %, about 20 to 10.0 W/V %, about 3.0 to 10.0 W/V %, about 4.0 to 10.0 W/V %, or about 5.0 to 10.0 W/V %.
  • One embodiment of the invention has a formulation of about 1.0 to 10.0 W/V % of the compounds of the invention.
  • One embodiment of the invention has a formulation of about 0.01 to 10.0 W/V % of the compounds of the invention.
  • One embodiment of the invention has a formulation of about 5.0 to 10.0 W/V % of the compounds of the invention.
  • the administration may be administered several times a day per eye, preferably one to ten times, more preferably one to four times, most preferably once a day.
  • the size of the drop administered may be in the range of about 10-100 ⁇ l, about 10-90 ⁇ l, about 10-80 ⁇ l, about 10-70 ⁇ l, about 10-60 ⁇ l, about 10-50 ⁇ l, about 10-40 ⁇ l, about 10-30 ⁇ l, about 20-100 ⁇ l, about 20-90 ⁇ l, about 20-80 ⁇ l, about 20-70 ⁇ l, about 20-60 ⁇ l, about 20-50 ⁇ l, about 20-40 ⁇ l, or about 20-30 ⁇ l.
  • One embodiment of the invention administers a drop in the range of about 10 to about 30 ⁇ l.
  • One embodiment of the invention administers a drop in the range of about 10 to about 100 ⁇ l.
  • One embodiment of the invention administers a drop in the range of about 20 to about 50 ⁇ l.
  • One embodiment of the invention administers a drop in the range of about 20 to about 40 ⁇ l.
  • One embodiment of the invention administers a drop in the range of about 10 to about 60 ⁇ l.
  • the formulations of the invention may be administered several drops per time, one to four drops, preferably one to three drops, more preferably one to two drops, and most preferably one drop per day. In one embodiment, the formulations of the invention are administered about one drop per time and one time per day.
  • the concentration of the compounds of the invention in the formulations may range about 0.0001 10.0 W/V %, about 0.005 to 10.0 W/V %, about 0.01 to 10.0 W/V %, about 0.05 to 10.0 W/V %, about 0.1 to 10.0 W/V %, about 0.5 to 10.0 W/V %, about 1.0 to 10.0 W/V %, about 20 to 10.0 W/V %, about 3.0 to 10.0 W/V %, about 4.0 to 10.0 W/V %, or about 5.0 to 10.0 W/V %.
  • One embodiment of the invention has a formulation of about 1.0 to 10.0 W/V % of the compounds of the invention.
  • One embodiment of the invention has a formulation of about 0.01 to 10.0 W/V % of the compounds of the invention.
  • One embodiment of the invention has a formulation of about 5.0 to 10.0 W/V % of the compounds of the invention.
  • These formulations may be applied or sprayed several times a day, preferably one to six times, more preferably one to four times, and most preferably once a day.
  • the compounding ratio of each ingredient may be suitably increased or decreased based on the degree of inflammations or infections.
  • the formulations of the invention can further include other pharmacological active ingredients as far as they do not contradict the purpose of the present invention.
  • their respective contents may be suitably increased or decreased in consideration of their effects and safety.
  • kits include a compound of the invention in suitable packaging, and written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like.
  • the kit may further contain another therapeutic agent that is co-administered with the LFA-1 antagonist of the invention.
  • the therapeutic agent and the LFA-1 antagonist of the invention are provided as separate compositions in separate containers within the kit.
  • the therapeutic agent and the LFA-1 antagonist of the invention are provided as a single composition within a container in the kit.
  • Suitable packaging and additional articles for use e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, dispensers, and the like) are known in the art and may be included in the kit.
  • affinities of the small molecules for LFA-1 were measured using fluorescence polarization (FP) in a competitive format with a small molecule antagonist, compound 1 ( FIG. 2 ), as previously described. All measurements were performed in buffer containing 50 mM Hepes, pH 7.2, 150 mM NaCl, 0.05% n-octyglucoside and 0.05% bovine gamma globulins (BGG) and either 1 mM MnCl 2 , or 1 mM CaCl 2 and 1 mM MgCl 2 .
  • FP fluorescence polarization
  • the affinity of compound 1 for LFA-1 was first measured by addition of 2 nM compound 1 to serial dilutions of LFA-1 starting from 1 ⁇ M in buffer containing either MnCl 2 or CaCl 2 and MgCl 2 .
  • Competition experiments were performed by addition of serial dilutions of antagonists to 2 nM compound 1 (using either 3 nM LFA-1 (in MnCl 2 ) or 40 nM LFA-1 (in CaCl 2 and MgCl 2 )).
  • the LFA-1 concentrations were reduced to 2 and 20 nM LFA-1 in the two divalent cation buffer conditions to maximize inhibition by ICAM-1-Ig.
  • the LFA-1 binding and antagonist competition data were analyzed using a non linear least squares fit of a four-parameter equation with KaleidaGraph software (Synergy Software, Reading, Pa.) to obtain the EC 50 values for the LFA-1 titration and the IC 50 values of the antagonists.
  • the data measured in both the homogeneous FP and heterogeneous ELISA formats described below contain relatively large signal to background ratios and the error estimates in the fits are typically less than 10% of the final value of the fitted parameter.
  • the equilibrium dissociation constants (K d ) of LFA-1 for compound 1 with and without A-286982 were calculated using Klotz and Hill analyses.
  • the affinities (K i ) of the antagonists for LFA-1 were calculated using the IC 50 values, the K d of compound 1/LFA-1, and the concentrations of compound 1 and LFA-1 in the competition experiments.
  • Antagonist Competition Small molecules and sICAM-1 were assayed for the ability to disrupt binding of ICAM-1-Ig or a fluorescein-labeled small molecule antagonist, compound 2B, to LFA-1 in a competitive format.
  • Compound 2B is similar to compound 1, but with a longer linker between the small molecule and fluorescein to maximize the binding of the anti-fluorescein detection antibody.
  • 96-well plates were coated with 5 ⁇ g/ml (33.3 nM) mouse anti-human ⁇ 2 integrin (a non-function blocking antibody) in phosphate-buffered saline (PBS) overnight at 4° C.
  • the plates were blocked with assay buffer (20 mM Hepes, pH 7.2, 140 mM NaCl, 1 mM MnCl 2 , 0.5% bovine serum albumin (BSA) and 0.05% Tween-20) for 1 hour at room temperature. After washing in buffer (50 mM Tris-HCl, pH 7.5, 100 mM NaCl, 1 mM MnCl 2 , and 0.05% Tween-20), 8 nM LFA-1 (LFA-1/ICAM-1 ELISA) or 2 nM LFA-1 (LFA-1/small molecule ELISA) were added, followed by incubation for 1 h at 37° C.
  • assay buffer 20 mM Hepes, pH 7.2, 140 mM NaCl, 1 mM MnCl 2 , 0.5% bovine serum albumin (BSA) and 0.05% Tween-20
  • buffer 50 mM Tris-HCl, pH 7.5, 100 mM NaCl, 1 mM M
  • the plates were washed, and for the LFA-1/ICAM-1 ELISA, serial dilutions of the small molecule antagonists or sICAM-1 were added to the plates for 30 minutes, followed by addition of 0.89 nM ICAM-1-Ig (final concentration) for 2 hour at 37° C. After an additional wash, goat anti-huIgG (Fc specific)-HRP was added and incubated for one hour at 37° C.
  • the diluted antagonists and 25 nM compound 2B were added concurrently to the plates, followed by a 2-hour incubation at 37° C. Sheep anti-fluorescein-HRP was added after a wash and incubated for one hour at 37° C.
  • the bound HRP-conjugated antibodies were detected by addition of tetramethylbenzidine (TMB) followed by measurement of the absorbance of the product at 450 nm after the addition of 1 M H 3 PO 4 to stop the reaction.
  • TMB tetramethylbenzidine
  • the IC 50 values for each curve were determined by fitting to the four-parameter equation described above using KaleidaGraph software.
  • ratio ⁇ 1) ((ligand EC 50 with antagonist)/(ligand EC 50 without antagonist)) ⁇ 1.
  • Full length human membrane-associated LFA-1 or BSA (0.35 mg/mL [1.4 and 5.3 ⁇ M, respectively] in 20 mM Hepes, 150 mM NaCl, 5 mM CaCl 2 , 5 mM MgCl 2 , 1 mM MnCl 2 , and 1% n-octylglucoside, pH 7.2) was incubated overnight at 37° C. with 4.1 ⁇ M compound 5, a tritium-labeled photoactivatable analogue of compound 3, in either the presence or absence of 290 ⁇ M compound 3. The molar ratio of compound 5 to LFA-1 was 3:1. A 96-well plate precoated with 1% BSA was used for the incubation.
  • the treated ⁇ L and ⁇ 2 subunits were separated by size exclusion chromatography in the presence of 6 M GuHCl, 20 mM Hepes, 10 mM EDTA, pH 6.8 and then chemically cleaved with 2.6 M hydroxylamine in 10% acetic acid with 7 M GuHCl for 4 h at 75° C.
  • the radiolabeled protein fragments were separated by SDS-PAGE and either visualized by autoradiography or transferred onto a polyvinylidene fluoride membrane, stained with Coomassie blue, and then identified by N-terminal protein sequencing.
  • the construct used, pLFA.huID. ⁇ p contains the sequence of the ⁇ L gene from the Nar1 restriction site 5′ of the I domain to the second PflM1 restriction site 3′ of the I domain in which the first PflM1 restriction site 3′ of the I domain was abolished (Edwards et al. 1995).
  • the following primers were made: the forward primer CACTGTGGCGCCCTGGTTTTCAGGAAGGTAGTGGATCAGGCACAAGCAAACAGGACCTGACTTC (SEQ ID NO 3), containing the sequence from the Nar1 site to the start of the I domain, a sequence of DNA encoding GSGSG (SEQ ID NO 3) and the 23 bp of the ⁇ L sequence after the end of the I domain, and the reverse primer TCTGAGCCATGTGCTGGTATCGAGGGGC (SEQ ID NO 5), which primes at the second PflM1 restriction site after the I domain.
  • PCR was performed using these primers and the pLFA.huID. ⁇ p linearized with Bgl II, which cut at a site within the I domain.
  • a DNA fragment was amplified that contained the sequence from the Nar 1 site to the second PflM1 site and in which the entire I domain, from C125 through G311, was replaced with a DNA sequence encoding GSGSG.
  • This piece of DNA was purified, digested with Nar1 and PflM1 and inserted into the human ⁇ L plasmid (pRKLFA ⁇ m) at the corresponding Nar1 and PflM1 sites. Correct insertion of the DNA sequence encoding GSGSG was confirmed by sequence analysis.
  • 293 cells were transfected with the P2 construct alone (mock) or with either the wild-type ⁇ L construct (wt) or the ⁇ L construct lacking the I domain (I-less) and allowed to recover for 3 days.
  • the cells were detached and resuspended in adhesion buffer (0.02 M HEPES, pH 7.2, 0.14 M NaCl, 0.2% glucose). Binding to plate bound ICAM-1-Ig was performed as described (Edwards et al. 1998).
  • the eyes of all animals are examined by fluorescein angiography all animals from the three groups are then sacrificed, and their eyes surgically removed and retinal tissue is isolated from them.
  • the retinal tissue is examined by micropictograph.
  • the extent to which microvasculature abnormalities develops in the corneal tissue of the diabetic control, their inhibition by administration of LFA-1 antagonist in the diabetic treatment group, and comparison to the normoglyemic control group are quantified by standardized morphometric analysis of the photomicrographs.
  • the T-cell adhesion assay was performed using the human T-lymphoid cell line HuT 78 (ATCC TIB-161).
  • Goat anti-HuIgG(Fc) was diluted to 2 ⁇ g/ml in PBS and 96-well plates were coated with 50 ⁇ l/well at 37° C. for 1 h. Plates were washed with PBS and blocked for 1 h at room temperature with 1% BSA in PBS.
  • 5 domain ICAM-Ig was diluted to 100 ng/ml in PBS and 50 ⁇ l/well was added to the plates O/N at 4° C.
  • HuT 78 cells were centrifuged at 100 g and the cell pellet was treated with 5 mM EDTA for ⁇ 5′ at 37° C.
  • a directly competitive LFA-1 antagonist of the invention was evaluated for its ability to inhibit release of inflammatory cytokines, in human mononucleocytes (PBMC) stimulated with staphylococcal enterotoxin B (SEB).
  • PBMC human mononucleocytes
  • SEB staphylococcal enterotoxin B
  • Stock solutions of antagonist, Rebamipide (a mucosal protective agent), and Cyclosporin A (CsA) were prepared in culture media and dilutions were prepared by addition of culture media to achieve the desired concentration.
  • Negative controls were prepared without SEB stimulation. SEB stimulation with vehicle (0.25% DMSO/media) was used as the positive control.
  • PBMC Human PBMC, frozen in cryopreservation media were thawed, washed with RPMI culture media containing 10% FBS in growth media and seeded onto a 96 well plate at 20,000 cells/well containing 180 ⁇ l culture media. Cells were incubated in the presence of antagonist, Rebamipide or CsA at 37 C for 1 hour prior to stimulation with SEB. SEB was added at 1 ng/ml and cell supernatants were harvested at 6, 16, and 48 hours. Cytokine levels in the assay supernatants were determined using a Luminex multiplex assay.
  • the antagonist demonstrated potent inhibition of the release of inflammatory cytokines, particularly the T-cell regulating cytokines, IL-2 and IL-4, with increasing dose.
  • the results are shown in Tables 2, 3, and 4 and graphically represented in FIG. 11 .
  • the pattern of cytokine release inhibited by more than 50% with the directly competitive LFA-1 antagonist is similar to that seen in comparison with CsA.
  • the exceptions to this similarity, IL-3, Il-6, and IL-12p40, have not been shown to be important to T-cell mediated inflammation.
  • a directly competitive LFA-1 antagonist of the invention was formulated in several compositions for administration as gels, lotions, ointments, and solutions, for administration by varying routes, including but not limited to topical, via instillation, aerosol, transdermal patch, via insert, or oral administration.
  • Formulations 3 and 4 of LFA-1 Antagonist Formulation 3 (% w/w) Formulation 4 (% w/w) 1% LFA-1 Antagonist 1% LFA-1 Antagonist 13% Dimethyl Isosorbide 13% Dimethyl Isosorbide 20% Transcutol 20% Transcutol 10% Hexylene glycol 10% Hexylene glycol 4% Propylene Glycol 4% Propylene Glycol 0.15% Methylparaben 0.15% Methylparaben 0.05% Propylparaben 0.05% Propylparaben 0.01% EDTA 0.01% EDTA 0.5% Carbopol Ultrez 10 0.3% Carbopol Ultrez 10 0.2% Penmulen TR-1 0.2% Penmulen TR-1 3% Isopropyl Myristate 2% Cetyl Alcohol 5% Olelyl Alcohol 5.5% Light Mineral Oil 5% White Petrolatum 5% Oleic Acid 0.02% Butylated Hydroxytoluene 0.02% Butylated Hydroxyto
  • Formulations 5 and 6 of LFA-1 Antagonist Formulation 5 (% w/w) Formulation 6 (% w/w) 1% LFA-1 Antagonist 1% LFA-1 Antagonist 15% PEG 400 10% Dimethyl Isosorbide 0.02% Butylated Hydroxytoluene 0.02% Butylated Hydroxytoluene 2% Span 80 2% Span 80 10% White Wax 10% White Wax 71.98% White Petrolatum 76.98% White Petrolatum
  • Formulation 9 Formulation 7 (% w/w) Formulation 8 (% w/w) (% w/w) 1% LFA-1 Antagonist 1% LFA-1 Antagonist 1% LFA-1 Antagonist 15% Dimethyl Isosorbide 15% Dimethyl Isosorbide 99% Dimethyl Sulfoxide 25% Transcutol 25% Transcutol 12% Hexylene glycol 12% Hexylene glycol 5% Propylene Glycol 5% Propylene Glycol q.s. pH 4.5 25% Trolamine q.s. pH 6.0 25% Trolamine q.s. 100 Water q.s. 100 Water
  • Formulations 1-9 were applied to dermatomed human skin tissue excised from a single donor in a single clinically relevant dose of 5 mg/cm 2 , which is equivalent to a 30-35 ⁇ g dose.
  • the thickness of the tissue ranges form 0.023 to 0.039 inches (0.584 to 0.991 mm) with a mean+/ ⁇ standard deviation in thickness of 0.030+/ ⁇ 0.004 inches (0.773+/ ⁇ 0.111 mm) and a coefficient of variation of 14.4%.
  • the tissue samples were mounted in Bronaugh flow-through diffusion cells. The cells were maintained at a constant temperature of 32° C. using recirculating water baths. The cells have a nominal diffusion area of 0.64 cm 2 .
  • PBS at pH 7.4, with 0.1% sodium azide and 4% Bovine Serum Albumin was used as the receptor phase below the mounted tissue.
  • Fresh receptor phase was continuously pumped under the tissue at a flow rate of nominally 1.0 ml/hr and collected in 6 hour intervals. The receptor phases were collected for analysis.
  • the tissue samples were exposed to Formulations 1-9 for 24 hours.
  • the excess formulation residing on the strateum corneum at that timepoint was removed by tape-stripping with CuDerm D-Squame stripping discs. The tape strips were discarded.
  • the epidermis and dermis were separated by blunt dissection. Epidermis, dermis and receptor phase were analyzed for content of LFA-1 Antagonist. The results are represented in Table 11.
  • Analytical data for the dermis fell within the linearity range established for LFA-1 Antagonist, and are quantitative.
  • Dermal deposition of LFA-1 Antagonist following a 24 hour exposure ranged from 0.66% (Formulation 6, 0.258 ⁇ g/cm 2 ) to 4.4% (Formulation 7, 34.3 ⁇ g/cm 2 ) of the applied dose.
  • the concentration of LFA-1 Antagonist in the dermis is calculated as 6.7 ⁇ M (Formulation 6) or greater (i.e., Formulation 7 provides a concentration in the dermis of 54.1 ⁇ M) for Formulations 1 to 9 in the dermis.
  • This assay is an in vitro model of lymphocyte proliferation resulting from activation, induced by engagement of the T-cell receptor and LFA-1, upon interaction with antigen presenting cells (Springer, Nature 346: 425 (1990)).
  • Microtiter plates (Nunc 96 well ELISA certified) are pre-coated overnight at 4° C. with 50 ⁇ l of 2 ⁇ g/ml of goat anti-human Fc (Caltag H 10700) and 50 ⁇ l of 0.07 ⁇ g/ml monoclonal antibody to CD3 (Immunotech 0178) in sterile PBS. The next day coat solutions are aspirated. Plates are then washed twice with PBS and 100 ⁇ l of 17 ng/ml 5d-ICAM-1-IgG is added for 4 hours at 37° C. Plates are washed twice with PBS prior to addition of CD4+ T cells. Lymphocytes from peripheral blood are separated from heparinized whole blood drawn from healthy donors.
  • An alternative method is to obtain whole blood from healthy donors through leukophoresis.
  • Blood is diluted 1:1 with saline, layered and centrifuged at 2500 ⁇ g for 30 minutes on LSM (6.2 g Ficoll and 9.4 g sodium diztrizoate per 100 ml) (Organon Technica, N.J.).
  • Monocytes are depleted using a myeloid cell depletion reagent method (Myeloclear, Cedarlane Labs, Hornby, Ontario, Canada).
  • PBLs are resuspended in 90% heat-inactivated Fetal Bovine serum and 10% DMSO, aliquoted, and stored in liquid nitrogen.
  • RPMI 1640 medium Gibco, Grand Island, N.Y.
  • Fetal Bovine serum Intergen, Purchase, N.Y.
  • 1 mM sodium pyruvate 3 mM L-glutamine
  • 1 mM nonessential amino acids 500 ⁇ g/ml penicillin, 50 ⁇ g/ml streptomycin, 50 ⁇ g/ml gentamycin (Gibco).
  • CD4+ T cells Purification of CD4+ T cells are obtained by negative selection method (Human CD4 Cell Recovery Column Kit # CL110-5 Accurate). 100,000 purified CD4+ T cells (90% purity) per microtiter plate well are cultured for 72 hours at 37° C. in 5% CO 2 in 100 ml of culture medium (RPMI 1640 (Gibco) supplemented with 10% heat inactivated FBS (Intergen), 0.1 mM non-essential amino acids, 1 nM Sodium Pyruvate, 100 units/ml Penicillin, 100 ⁇ g/ml Streptomycin, 50 ⁇ g/ml Gentamicin, 10 mM Hepes and 2 mM Glutamine). Inhibitors are added to the plate at the initiation of culture.
  • RPMI 1640 Gibco
  • FBS heat inactivated FBS
  • Inhibitors are added to the plate at the initiation of culture.
  • Proliferative responses in these cultures are measured by addition of 1 ⁇ Ci/well titrated thymidine during the last 6 hours before harvesting of cells. Incorporation of radioactive label is measured by liquid scintillation counting (Packard 96 well harvester and counter). Results are expressed in counts per minute (cpm).
  • the mixed lymphocyte culture model which is an in vitro model of transplantation (A. J. Cunningham, “Understanding Immunology, Transplantation Immunology” pages 157-159 (1978) examines the effects of various LFA-1 antagonists in both the proliferative and effector arms of the human mixed lymphocyte response.
  • PBMC peripheral blood
  • PBMCs Mononuclear cells from peripheral blood (PBMC) are separated from heparanized whole blood drawn from healthy donors. Blood is diluted 1:1 with saline, layered, and centrifuged at 2500 ⁇ g for 30 minutes on LSM (6.2 g Ficoll and 9.4 g sodium diztrizoate per 100 ml) (Organon Technica, N.J.).
  • LSM 6.2 g Ficoll and 9.4 g sodium diztrizoate per 100 ml
  • An alternative method is to obtain whole blood from healthy donors through leukophoresis.
  • PBMCs are separated as above, resuspended in 90% heat inactivated Fetal Bovine serum and 10% DMSO, aliquoted and stored in liquid nitrogen.
  • RPMI 1640 medium Gibco, Grand Island, N.Y.
  • Fetal Bovine serum Intergen, Purchase, N.Y.
  • 1 mM sodium pyruvate 3 mM L-glutamine
  • 1 mM nonessential amino acids 500 ⁇ g/ml penicillin, 50 ⁇ g/ml streptomycin, 50 ⁇ g/ml gentamycin (Gibco).
  • MLR Mixed Lymphocyte Response
  • the purpose of this study is to evaluate the anti-adhesive properties of LFA-1 Antagonists on the attachment of Jurkat cells to ICAM-1 following in vitro exposure.
  • Jurkat cells are labeled with an 8 ⁇ M solution of BCECF-AM (2′,7′-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein) in growth media at room temperature for 15 minutes.
  • Labeled cells are incubated in 70 mL of assay media in each well of a 96 well plate at 500,000 cells per well with 70 ⁇ L of LFA-1 Antagonist or positive control in assay media at 37° C. for 30 minutes.
  • a 100 ⁇ L aliquot of this fluorescently labeled Jurkat cell suspension is allowed to settle in the presence of LFA-1 antagonist or the positive control in wells of a 96 well plate coated with recombinant human ICAM-1 expressed as an Fc chimera at 37° C. for 1 hour.
  • Non-adherent cells are removed by washing and centrifugation at 100 g for 1 minute.
  • Adherent cells are determined as adherent fluorescent units on a fluorescent plate reader.
  • each animal receives a single topical application to the dorsal skin on Day 1.
  • each animal receives 3 topical applications given daily (approximately 4 hours apart) to the dorsal skin for 7 consecutive days.
  • Intradermal For each animal in Group 5 (DMSO, Intradermal), the single 200- ⁇ L intradermal dose is administered on Day 1 as two, 100- ⁇ L injections given sequentially via a syringe and needle in a shaved area of the subscapular region.
  • Blood All Groups. For the final dose administration as applicable based on study group, blood is collected from each animal predose and at 0.25, 0.5, 1, 2, and 4 hours postdose.
  • Sample Collection Application Sites (Dermal Groups Only). Following sacrifice for the terminal blood sample, the section of skin exposed to the test article formulation is excised. The stratum corneum and any unabsorbed test article formulation remaining on the surface of the skin is removed by tape stripping. The strips were combined into one sample vial for each animal, and the remaining skin section was placed into a second sample vial. Sample weights were recorded.
  • a rat model is used to measure distribution of a directly competitive LFA-1 antagonist to tissues in the eye, particularly to retina.
  • a directly competitive LFA-1 antagonist to tissues in the eye, particularly to retina.
  • Celecoxib a selective cyclooxygenase-2 inhibitor, inhibits retinal vascular endothelial growth factor expression and vascular leakage in a streptozotocin-induced diabetic rat model”, 458: 283-289.
  • FIG. 12 the concentration of radiolabeled antagonist measured in each anatomical region is indicated by the increasing grayscale of the box corresponding to the anatomical region as labeled. Numerical values for this data are shown in Table 14 and is given in nanogram equivalents of .radiolabelled—LFA-1 antagonist per gram tissue.
  • Healthy subjects are enrolled.
  • a randomized, controlled, dose escalation trial of both single and multiple administrations of LFA-1 antagonist is conducted. Cohorts of 7 subjects each (5 treatment, 2 placebo) are treated at each of 6-8 dose levels of LFA-1 antagonists formulated as sterile, neutral, isotonic, buffered aqueous solutions. Subjects receive a single instillation on Day 1. Samples are obtained for pharmacokinetic and pharmacodynamic assessments over the subsequent week. Starting Day 8, subjects receive the same dose of LFA-1 antagonist daily for a total of 14 days. PK/PD assessments, safety laboratory studies, ophthalmic exams, corneal staining and fluorescein angiographies are assessed.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Diabetes (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Vascular Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Dermatology (AREA)
  • Biophysics (AREA)
  • Pathology (AREA)
  • Mycology (AREA)
  • Genetics & Genomics (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Obesity (AREA)
US12/288,330 2007-10-19 2008-10-17 Compositions and methods for treatment of diabetic retinopathy Abandoned US20090155176A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US12/288,330 US20090155176A1 (en) 2007-10-19 2008-10-17 Compositions and methods for treatment of diabetic retinopathy
US14/491,333 US9447077B2 (en) 2008-04-15 2014-09-19 Crystalline pharmaceutical and methods of preparation and use thereof
US14/659,789 US10960087B2 (en) 2007-10-19 2015-03-17 Compositions and methods for treatment of diabetic retinopathy
US15/235,572 US11028077B2 (en) 2008-04-15 2016-08-12 Crystalline pharmaceutical and methods of preparation and use thereof
US17/181,187 US20210338839A1 (en) 2007-10-19 2021-02-22 Compositions and methods for treatment of diabetic retinopathy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US99957107P 2007-10-19 2007-10-19
US12/288,330 US20090155176A1 (en) 2007-10-19 2008-10-17 Compositions and methods for treatment of diabetic retinopathy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/659,789 Continuation US10960087B2 (en) 2007-10-19 2015-03-17 Compositions and methods for treatment of diabetic retinopathy

Publications (1)

Publication Number Publication Date
US20090155176A1 true US20090155176A1 (en) 2009-06-18

Family

ID=40579823

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/288,330 Abandoned US20090155176A1 (en) 2007-10-19 2008-10-17 Compositions and methods for treatment of diabetic retinopathy
US14/659,789 Active US10960087B2 (en) 2007-10-19 2015-03-17 Compositions and methods for treatment of diabetic retinopathy
US17/181,187 Abandoned US20210338839A1 (en) 2007-10-19 2021-02-22 Compositions and methods for treatment of diabetic retinopathy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/659,789 Active US10960087B2 (en) 2007-10-19 2015-03-17 Compositions and methods for treatment of diabetic retinopathy
US17/181,187 Abandoned US20210338839A1 (en) 2007-10-19 2021-02-22 Compositions and methods for treatment of diabetic retinopathy

Country Status (9)

Country Link
US (3) US20090155176A1 (es)
EP (3) EP2209371B1 (es)
JP (2) JP5808037B2 (es)
CN (1) CN101873797A (es)
AU (1) AU2008317473B2 (es)
CA (3) CA3105972A1 (es)
ES (2) ES2630406T3 (es)
MX (1) MX2010004281A (es)
WO (1) WO2009054914A1 (es)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060281739A1 (en) * 2005-05-17 2006-12-14 Thomas Gadek Compositions and methods for treatment of eye disorders
US20080176896A1 (en) * 2003-11-05 2008-07-24 Wang Shen Modulators of cellular adhesion
US20090258069A1 (en) * 2008-04-15 2009-10-15 John Burnier Delivery of LFA-1 antagonists to the gastrointestinal system
US20090257957A1 (en) * 2008-04-15 2009-10-15 John Burnier Aerosolized LFA-1 antagonists for use in localized treatment of immune related disorders
US20090258070A1 (en) * 2008-04-15 2009-10-15 John Burnier Topical LFA-1 antagonists for use in localized treatment of immune related disorders
US20090298869A1 (en) * 2008-04-15 2009-12-03 John Burnier Crystalline pharmaceutical and methods of preparation and use thereof
US20110092707A1 (en) * 2009-10-21 2011-04-21 Sarcode Corporation Crystalline Pharmaceutical and Methods of Preparation and Use Thereof
US20110104083A1 (en) * 2009-10-30 2011-05-05 Intratus, Inc. Methods and cosmetic preparations for the sustained delivery of therapeutic agents to the eye
US20110189174A1 (en) * 2010-02-01 2011-08-04 Afshin Shafiee Compositions and methods for treating, reducing, ameliorating, alleviating, or inhibiting progression of, pathogenic ocular neovascularization
US20110281901A1 (en) * 2010-05-05 2011-11-17 Glaxo Wellcome Manufacturing Pte Ltd. Pharmaceutical compositions and methods of making same
US20130324603A1 (en) * 2012-06-05 2013-12-05 Olatec Industries Llc Pharmaceutical composition for treating inflammation and pain
US9085553B2 (en) 2012-07-25 2015-07-21 SARcode Bioscience, Inc. LFA-1 inhibitor and methods of preparation and polymorph thereof
US10960087B2 (en) 2007-10-19 2021-03-30 Novartis Ag Compositions and methods for treatment of diabetic retinopathy
US11018173B2 (en) 2018-06-18 2021-05-25 Samsung Electronics Co., Ltd. Image sensor
US11753382B2 (en) 2019-06-25 2023-09-12 Translatum Medicus Inc. Processes of making 2-((1-benzyl-1H-indazol-3-yl)methoxy)-2-methylpropanoic acid and its derivatives

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8598156B2 (en) 2010-03-25 2013-12-03 Glaxosmithkline Llc Chemical compounds
EP3078384A1 (en) * 2012-02-22 2016-10-12 Stealth Peptides International, Inc. Methods and compositions for preventing or treating ophthalmic conditions
SG11201407534PA (en) 2012-06-13 2014-12-30 Hoffmann La Roche New diazaspirocycloalkane and azaspirocycloalkane
IN2015DN00960A (es) 2012-09-25 2015-06-12 Hoffmann La Roche
AR095079A1 (es) 2013-03-12 2015-09-16 Hoffmann La Roche Derivados de octahidro-pirrolo[3,4-c]-pirrol y piridina-fenilo
MY187449A (en) 2013-11-26 2021-09-22 Hoffmann La Roche New octahydro-cyclobuta [1,2-c;3,4-c']dipyrrol-2-yl
JP6272604B2 (ja) * 2013-12-27 2018-01-31 日本サプリメント株式会社 糖取り込み促進剤
JP6554481B2 (ja) 2014-03-26 2019-07-31 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft オートタキシン(atx)及びリゾホスファチジン酸(lpa)産生の阻害剤としての縮合[1,4]ジアゼピン化合物
HUE046820T2 (hu) 2014-03-26 2020-03-30 Hoffmann La Roche Biciklusos vegyületek autotaxin (ATX) és lizofoszfatidsav (LPA) termelésgátlókként
TW201637648A (zh) * 2015-04-03 2016-11-01 參天製藥股份有限公司 淚腺藥物遞輸系統
CR20180058A (es) 2015-09-04 2018-02-26 Hoffmann La Roche Nuevos derivados de fenoximetilo
MX2018001890A (es) 2015-09-24 2018-06-20 Hoffmann La Roche Compuestos biciclicos como inhibidores de autotaxina (atx).
MX2017015034A (es) 2015-09-24 2018-04-13 Hoffmann La Roche Nuevos compuestos biciclicos como inhibidores duales de autotaxina (atx)/anhidrasa carbonica (ca).
CN107922415B (zh) 2015-09-24 2022-04-15 豪夫迈·罗氏有限公司 新的作为双重atx/ca抑制剂的二环化合物
KR20180054830A (ko) 2015-09-24 2018-05-24 에프. 호프만-라 로슈 아게 오토탁신(atx) 억제제로서의 이환형 화합물
CR20190423A (es) 2017-03-16 2019-11-01 Hoffmann La Roche Nuevos compuestos biciclícos como inhibidores dobles de atx/ca
WO2018167113A1 (en) * 2017-03-16 2018-09-20 F. Hoffmann-La Roche Ag New bicyclic compounds as atx inhibitors
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors
FI3697785T3 (fi) 2017-10-18 2023-04-03 Jubilant Epipad LLC Imidatsopyridiiniyhdisteitä pad:n estäjinä
EP3707135A1 (en) 2017-11-06 2020-09-16 Jubilant Prodel LLC Pyrimidine derivatives as inhibitors of pd1/pd-l1 activation
JP7368369B2 (ja) 2017-11-24 2023-10-24 ジュビラント・エピスクライブ・エルエルシー Prmt5阻害剤としてのヘテロ環式化合物
JP7279063B6 (ja) 2018-03-13 2024-02-15 ジュビラント プローデル エルエルシー Pd1/pd-l1相互作用/活性化の阻害剤としての二環式化合物
CA3098428A1 (en) 2018-04-18 2019-10-24 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
CA3100977A1 (en) 2018-05-21 2019-11-28 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
MX2022015327A (es) * 2020-06-05 2023-02-22 Occurx Pty Ltd Compuestos y metodos para el tratamiento de trastornos oculares.
CN116635032A (zh) * 2020-10-21 2023-08-22 阿祖拉眼科有限公司 用于治疗眼部病症的化合物和方法
JP2024042677A (ja) * 2022-09-15 2024-03-28 日油株式会社 コンタクトレンズ用溶液

Citations (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4316893A (en) * 1975-06-19 1982-02-23 Nelson Research & Development Co. Vehicle composition containing 1-substituted azacycloalkan-2-ones
US4568343A (en) * 1984-10-09 1986-02-04 Alza Corporation Skin permeation enhancer compositions
US4668506A (en) * 1985-08-16 1987-05-26 Bausch & Lomb Incorporated Sustained-release formulation containing and amino acid polymer
US4908202A (en) * 1988-02-02 1990-03-13 Cassella Aktiengesellschaft Use of 2-oxo-1-pyrrolidineacetamide for the determination of the glomerular filtration rate in humans
US4931279A (en) * 1985-08-16 1990-06-05 Bausch & Lomb Incorporated Sustained release formulation containing an ion-exchange resin
US4992445A (en) * 1987-06-12 1991-02-12 American Cyanamid Co. Transdermal delivery of pharmaceuticals
US5001139A (en) * 1987-06-12 1991-03-19 American Cyanamid Company Enchancers for the transdermal flux of nivadipine
US5023252A (en) * 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5134122A (en) * 1989-07-28 1992-07-28 Debiopharm S.A. Method for preparing a pharmaceutical composition in the form of microparticles
US5192741A (en) * 1987-09-21 1993-03-09 Debiopharm S.A. Sustained and controlled release of water insoluble polypeptides
US5288854A (en) * 1990-11-28 1994-02-22 Center For Blood Research, Inc. Functional derivatives of ICAM-1 which are substantially capable of binding to LFA-1 but are substantially incapable of binding to MAC-1
US5298492A (en) * 1992-08-04 1994-03-29 Schering Corporation Diamino acid derivatives as antihypertensives
US5397791A (en) * 1993-08-09 1995-03-14 Merck & Co., Inc. Fibrinogen receptor antagonists
US5424289A (en) * 1993-07-30 1995-06-13 Alza Corporation Solid formulations of therapeutic proteins for gastrointestinal delivery
US5424399A (en) * 1988-06-28 1995-06-13 The Children's Medical Center Corporation Human CR3α/β heterodimers
US5597567A (en) * 1991-10-04 1997-01-28 The United States Of America As Represented By The Department Of Health And Human Services Blocking cell adhesion molecules and treating animals with ocular inflammation
US5612052A (en) * 1995-04-13 1997-03-18 Poly-Med, Inc. Hydrogel-forming, self-solvating absorbable polyester copolymers, and methods for use thereof
US5622700A (en) * 1992-08-21 1997-04-22 Genentech, Inc. Method for treating a LFA-1-mediated disorder
US5624837A (en) * 1994-03-03 1997-04-29 Alexion Pharmaceuticals, Inc. Nucleic acid encoding chimeric complement inhibitor proteins
US5747035A (en) * 1995-04-14 1998-05-05 Genentech, Inc. Polypeptides with increased half-life for use in treating disorders involving the LFA-1 receptor
US5863910A (en) * 1996-01-12 1999-01-26 Bolonick; Joel Treatment of chronic inflammatory disorders of the gastrointestinal tract
US5877224A (en) * 1995-07-28 1999-03-02 Rutgers, The State University Of New Jersey Polymeric drug formulations
US5893985A (en) * 1997-03-14 1999-04-13 The Lincoln Electric Company Plasma arc torch
US5922356A (en) * 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US6011011A (en) * 1992-09-21 2000-01-04 Pharmacia & Upjohn Company Sustained-release protein formulations
US6180608B1 (en) * 1996-12-11 2001-01-30 Praecis Pharmaceuticals, Inc. Pharmaceutical formulations for sustained drug delivery
US6204280B1 (en) * 1994-11-02 2001-03-20 Merck Patent Gesellschaft Mit Berschrankter Haftung Adhesion receptor antagonists
US6203793B1 (en) * 1992-02-28 2001-03-20 Board Of Regents Of The University Of Texas System Compositions and methods for treating thermal injury
US20010006656A1 (en) * 1999-02-17 2001-07-05 University Of Washington Methods and compositions for inhibiting inflammation associated with pulmonary disease
US6340679B1 (en) * 1999-02-13 2002-01-22 Aventis Pharma Deutschland Gmbh Guanidine derivatives as inhibitors of cell adhesion
US6358976B1 (en) * 1996-11-27 2002-03-19 John Wityak Integrin receptor antagonists
US20020045582A1 (en) * 1997-12-31 2002-04-18 Alexey L. Margolin Stabilized protein crystals formulations containing them and methods of making them
US6515124B2 (en) * 2000-02-09 2003-02-04 Hoffman-La Roche Inc. Dehydroamino acids
US6521619B2 (en) * 2000-06-29 2003-02-18 Icos Corporation Aryl phenylcyclopropyl sulfide derivatives and their use as cell adhesion inhibiting anti-inflammatory and immune suppressive agents
US6524581B1 (en) * 1998-12-30 2003-02-25 The Children's Medical Center Corporation Prevention and treatment of retinal ischemia and edema
US20030044406A1 (en) * 2001-03-02 2003-03-06 Christine Dingivan Methods of preventing or treating inflammatory or autoimmune disorders by administering CD2 antagonists in combination with other prophylactic or therapeutic agents
US20030064105A1 (en) * 2000-08-25 2003-04-03 Myung-Jin Kim Lipophilic-coated microparticle containing a protein drug and formulation comprising same
US20030069560A1 (en) * 2001-05-03 2003-04-10 Massachusetts Eye And Ear Infirmary Implantable drug delivery device and use thereof
US20030138488A1 (en) * 1999-12-31 2003-07-24 Kohn Joachim B Pharmaceutical formulation for regulating the timed release of biologically active compounds based on a polymer matrix
US20040006236A1 (en) * 1998-10-13 2004-01-08 Nader Fotouhi Diaminopropionic acid derivatives
US20040009998A1 (en) * 2001-10-01 2004-01-15 Dhar T. G. Murali Spiro-hydantoin compounds useful as anti-inflammatory agents
US20040038973A1 (en) * 2002-08-13 2004-02-26 Joe Nahra Phthalimide derivatives as matrix metalloproteinase inhibitors
US6710064B2 (en) * 2000-12-01 2004-03-23 Bristol-Myers Squibb Co. Hydantoin compounds useful as anti-inflammatory agents
US20040058968A1 (en) * 2000-11-28 2004-03-25 Genentech, Inc. LFA-1 antagonist compounds
US20040122047A1 (en) * 2001-06-06 2004-06-24 Aventis Pharma Limited Substituted tetrahydroisoquinolines
US6764681B2 (en) * 1991-10-07 2004-07-20 Biogen, Inc. Method of prophylaxis or treatment of antigen presenting cell driven skin conditions using inhibitors of the CD2/LFA-3 interaction
US6867203B2 (en) * 1998-12-29 2005-03-15 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory and immune-suppressive compounds
US6872382B1 (en) * 2001-05-21 2005-03-29 Alcon, Inc. Use of selective PDE IV inhibitors to treat dry eye disorders
US20050107399A1 (en) * 2003-09-11 2005-05-19 Kemia, Inc. Cytokine inhibitors
US20050119279A1 (en) * 2003-10-02 2005-06-02 Dhar T. G. M. Spiro-cyclic compounds useful as anti-inflammatory agents
US20050142066A1 (en) * 2002-06-06 2005-06-30 Dana Farber Cancer Institute, Inc Compounds or agents that inhibit and induce the formation of focal microvessel dilatations
US20060052434A1 (en) * 2004-08-18 2006-03-09 Dhar T G M Pyrrolizine compounds useful as anti-inflammatory agents
US20060074099A1 (en) * 2004-10-01 2006-04-06 Delmonte Albert J Crystalline forms and process for preparing spiro-hydantoin compounds
US20060142319A1 (en) * 2004-12-14 2006-06-29 Bang-Chi Chen Pyridyl-substituted spiro-hydantoin crystalline forms and process
US20060148836A1 (en) * 2004-12-14 2006-07-06 Dhar T G M Pyridyl-substituted spiro-hydantoin compounds and use thereof
US7166568B1 (en) * 1998-02-09 2007-01-23 Oklahoma Medical Research Foundation Compositions and methods to inhibit formation of the C5b-9 complex of complement
US20070027101A1 (en) * 2001-11-09 2007-02-01 Guyer David R Methods for treating ocular neovascular diseases
US20070048216A1 (en) * 2005-08-23 2007-03-01 Norenberg Jeffrey P Non-invasive diagnostic agents of cancer and methods of diagnosing cancer, especially leukemia and lymphoma
US20070066585A1 (en) * 2000-06-29 2007-03-22 Wang Gary T Aryl phenylheterocyclyl sulfide derivatives and their use as cell adhesion-inhibiting anti-inflammatory and immune-suppressive agents
US20070090205A1 (en) * 2005-06-24 2007-04-26 Boehringer Ingelheim International Gmbh Nebuliser and container
US20070105824A1 (en) * 2003-04-29 2007-05-10 Erickson-Miller Connie L Methods for treating degenerative diseases/injuries
US20070142317A1 (en) * 2003-12-29 2007-06-21 Qlt Usa, Inc. Topical composition for treatment of skin disorders
US20070149593A1 (en) * 2005-12-23 2007-06-28 Alcon, Inc. PHARMACEUTICAL FORMULATION FOR DELIVERY OF RECEPTOR TYROSINE KINASE INHIBITING (RTKi) COMPOUNDS TO THE EYE
US7314938B2 (en) * 2003-11-05 2008-01-01 Sunesis Pharmaceuticals, Inc. Modulators of cellular adhesion
US20080108677A1 (en) * 2004-08-30 2008-05-08 Karyon-Ctt Ltd Thioxothiazolidinone Compounds For Use As Pharmaceuticals
US7396530B2 (en) * 2004-06-09 2008-07-08 Genentech, Inc. Method of treating granuloma annulare or sarcoid
US20100092541A1 (en) * 2005-05-17 2010-04-15 John Burnier Compositions and methods for treatment of eye disorders
US20110092707A1 (en) * 2009-10-21 2011-04-21 Sarcode Corporation Crystalline Pharmaceutical and Methods of Preparation and Use Thereof
US20120107404A1 (en) * 2008-04-15 2012-05-03 Sarcode Bioscience Inc. Crystalline Pharmaceutical and Methods of Preparation and Use Thereof

Family Cites Families (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6872A (en) 1849-11-13 Means of changing the combination in bevolving tumbler-locks
US735A (en) 1838-05-10 Rtjftjg porter
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US3989816A (en) 1975-06-19 1976-11-02 Nelson Research & Development Company Vehicle composition containing 1-substituted azacycloheptan-2-ones
US4405616A (en) 1975-06-19 1983-09-20 Nelson Research & Development Company Penetration enhancers for transdermal drug delivery of systemic agents
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US4557934A (en) 1983-06-21 1985-12-10 The Procter & Gamble Company Penetrating topical pharmaceutical compositions containing 1-dodecyl-azacycloheptan-2-one
US4713244A (en) 1985-08-16 1987-12-15 Bausch & Lomb Incorporated Sustained-release formulation containing an amino acid polymer with a lower alkyl (C1 -C4) polar solvent
ES2064327T3 (es) 1987-11-02 1995-02-01 Baylor College Medicine Uso de icam-1 o de sus derivados funcionales para el tratamiento de una inflamacion no especifica.
US5236704A (en) 1988-01-28 1993-08-17 Sumitomo Pharmaceuticals Co., Ltd. Controlled release formulation
DE68923048T2 (de) 1988-08-23 1995-11-16 Dana Farber Cancer Inst Inc Alpha-Subeinheit des LFA-1-Leukocyt-Adhäsions-Rezeptors.
DE68929096T2 (de) 1988-09-01 2000-05-11 Bayer Ag Menschliches Rhinovirusrezeptorprotein, das die Virusinfektionsanfälligkeit hemmt
KR900701846A (ko) 1988-09-28 1990-12-04 원본미기재 세포간 점착분자 및 이의 결합 리간드
US5149780A (en) 1988-10-03 1992-09-22 The Scripps Research Institute Peptides and antibodies that inhibit integrin-ligand binding
CA2050329A1 (en) 1989-03-09 1990-09-10 Timothy A. Springer Method of treating viral infections using lfa-1
ZA903223B (en) 1989-04-28 1991-02-27 Baylor College Medicine Dissemination of hiv-1 infected cells
US5135916A (en) 1989-06-12 1992-08-04 Oklahoma Medical Research Foundation Inhibition of complement mediated inflammatory response
WO1991019511A1 (en) 1990-06-18 1991-12-26 The General Hospital Corporation CONTROLLING CELLULAR IMMUNE/INFLAMMATORY RESPONSES WITH β2 INTEGRINS
CA2046830C (en) 1990-07-19 1999-12-14 Patrick P. Deluca Drug delivery system involving inter-action between protein or polypeptide and hydrophobic biodegradable polymer
US5318965A (en) 1990-08-24 1994-06-07 Abbott Laboratories Quinobenzoxazine, antineoplastic agents
CA2090427A1 (en) 1990-08-27 1992-02-28 Cetus Oncology Corporation Cd18 peptide medicaments for the treatment of disease
CH683149A5 (fr) 1991-07-22 1994-01-31 Debio Rech Pharma Sa Procédé pour la préparation de microsphères en matériau polymère biodégradable.
US6162432A (en) 1991-10-07 2000-12-19 Biogen, Inc. Method of prophylaxis or treatment of antigen presenting cell driven skin conditions using inhibitors of the CD2/LFA-3 interaction
GB9211268D0 (en) 1992-05-28 1992-07-15 Ici Plc Salts of basic peptides with carboxyterminated polyesters
WO1994011400A1 (en) 1992-11-18 1994-05-26 Helsinki University Licensing Ltd. Oy Peptides from human icam-2 and from human icam-1 and their analogs for use in therapy and diagnosis
US5672659A (en) 1993-01-06 1997-09-30 Kinerton Limited Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
PL174772B1 (pl) 1993-01-06 1998-09-30 Kinerton Ltd Kompozycja farmaceutyczna zawierająca jonowy konjugat cząsteczkowy
DE59409322D1 (de) 1993-12-03 2000-06-08 Hoffmann La Roche Essigsäurederivate als Arzneimittel
US5470953A (en) 1993-12-23 1995-11-28 Icos Corporation Human β2 integrin α subunit
JPH09512021A (ja) 1994-04-19 1997-12-02 ザ ユニバーシティ オブ カンサス Icam−1/lfa−1 短鎖ペプチドおよび該ペプチドの使用方法
US5849327A (en) 1994-07-29 1998-12-15 Advanced Polymer Systems, Inc. Delivery of drugs to the lower gastrointestinal tract
US5585359A (en) 1994-09-29 1996-12-17 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US20020028798A1 (en) 1995-12-12 2002-03-07 Omeros Medical Systems Irrigation solution and method for inhibition of pain and inflammation
MX9709278A (es) 1995-05-29 1998-03-31 Pfizer Dipeptidos que promueven la liberacion de la hormona de crecimiento, composiciones que los contienen y uso de los mismos.
US5843884A (en) 1995-11-15 1998-12-01 Oklahoma Medical Research Foundation C9 complement inhibitor
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US5840332A (en) 1996-01-18 1998-11-24 Perio Products Ltd. Gastrointestinal drug delivery system
WO1997033603A1 (en) 1996-03-13 1997-09-18 Trustees Of The University Of Pennsylvania Novel peptides which inhibit complement activation
CA2250981C (en) 1996-04-23 2002-07-02 Kinerton Limited Acidic polylactic polymers
HRP970493A2 (en) 1996-09-23 1998-08-31 Wienman E. Phlips Oral delayed immediate release medical formulation and method for preparing the same
AR012443A1 (es) 1997-04-16 2000-10-18 Uriach & Cia Sa J Nuevas carboxamidas como inhibidores de la agregacion plaquetaria, procedimiento para su preparacion, composiciones farmaceuticas que loscontienen y uso de los mismos en la manufactura de medicamentos
PT1007033E (pt) 1997-08-28 2008-07-22 Novartis Ag Antagonistas do antigénio-1 da função de linfócitos
PL343555A1 (en) 1998-03-27 2001-08-27 Genentech Inc Antagonists for treatment of cd11/cd18 adhesion receptor mediated disorders
ATE279914T1 (de) 1998-11-27 2004-11-15 Kanji Takada Oral anzuwendendes arzneimittel zur wirkstoffverabreichung im magendarmtrakt
IT1304152B1 (it) 1998-12-10 2001-03-08 Mediolanum Farmaceutici Srl Composizioni comprendenti un peptide ed acido polilattico-glicolicoatte alla preparazione di impianti sottocutanei aventi un prolungato
WO2000040262A1 (en) 1999-01-05 2000-07-13 The Flinders University Of South Australia Novel agents and methods for treatment and diagnosis of ocular disorders
WO2000044731A1 (en) 1999-01-27 2000-08-03 G.D. Searle & Co. Novel hydroxyamidino carboxylate derivatives useful as nitric oxide synthase inhibitors
CO5140104A1 (es) 1999-02-16 2002-03-22 Novartis Ag Derivados de mevinolina y preparacion farmaceuticas que los contienen
IL145661A0 (en) 1999-03-31 2002-06-30 Janssen Pharmaceutica Nv Pregelatinized starch in a controlled release formulation
WO2000072883A2 (en) 1999-06-02 2000-12-07 Aviana Biopharm Pharmaceutical transdermal compositions
AU1186401A (en) 1999-06-23 2001-01-22 Sedum Laboratories, Inc. Ionically formulated biomolecule microcarriers
WO2001012233A2 (en) 1999-08-18 2001-02-22 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Sustained release formulation of a peptide
ECSP003707A (es) 1999-10-13 2002-05-23 Novartis Ag Diazepanes
US6294522B1 (en) 1999-12-03 2001-09-25 Cv Therapeutics, Inc. N6 heterocyclic 8-modified adenosine derivatives
US6605597B1 (en) 1999-12-03 2003-08-12 Cv Therapeutics, Inc. Partial or full A1agonists-N-6 heterocyclic 5′-thio substituted adenosine derivatives
EP1237575B1 (en) 1999-12-14 2008-08-06 Genentech, Inc. Tnf-alpha antagonist and lfa-1 antagonist for treating rheumatoid arthritis
JP4917726B2 (ja) 1999-12-31 2012-04-18 ルトガーズ、ザ ステイト ユニバーシティ オブ ニュージャージー ポリマー混合物および活性化合物からなる徐放用医薬製剤
AU784226B2 (en) 1999-12-31 2006-02-23 Rutgers, The State University Pharmaceutical formulation for regulating the timed release of biologically active compounds based on a polymer matrix
ATE294188T1 (de) 2000-01-14 2005-05-15 Richard S Larson Peptidische inhibitoren der lfa-1/icam-1 interaktion
CA2407830A1 (fr) 2000-05-05 2001-11-15 Serge Auvin Derives d'aminoacides et leur preparation a titre de medicaments
GB0011817D0 (en) 2000-05-16 2000-07-05 Pharmacia & Upjohn Spa Antagonists of integrin receptors
AR030817A1 (es) 2000-10-02 2003-09-03 Novartis Ag Derivados de diazacicloalcanodiona
GB0025208D0 (en) 2000-10-13 2000-11-29 Euro Celtique Sa Delayed release pharmaceutical formulations
US6653478B2 (en) 2000-10-27 2003-11-25 Ortho-Mcneil Pharmaceutical, Inc. Substituted benzimidazol-2-ones as vasopressin receptor antagonists and neuropeptide Y modulators
DE10055857A1 (de) 2000-11-10 2002-08-22 Creative Peptides Sweden Ab Dj Neue pharmazeutische Depotformulierung
GB0028367D0 (en) 2000-11-21 2001-01-03 Celltech Chiroscience Ltd Chemical compounds
US20030008848A1 (en) 2000-12-19 2003-01-09 Fleck Roman Wolfgang Small molecules useful in the treatment of inflammatory disease
CA2435415A1 (en) 2001-01-26 2002-08-01 Debio Recherche Pharmaceutique S.A. Microparticles of biodegradable polymer encapsulating a biologically active substance
ES2275808T3 (es) 2001-02-06 2007-06-16 Pfizer Products Inc. Composiciones farmaceuticas para el tratamiento de trastornos del snc y otros trastornos.
EP1383376A4 (en) 2001-03-19 2006-03-08 Praecis Pharm Inc PHARMACEUTICAL FORMULATIONS WITH PROLONGED RELEASE
US6391290B1 (en) 2001-03-21 2002-05-21 Schering-Plough Healthcare Products, Inc. Skin care compositions
CA2448076A1 (en) 2001-05-24 2002-11-28 Masahiko Hayakawa 3-quinoline-2-(1h)-ylideneindolin-2-one derivatives
US7858095B2 (en) 2001-07-24 2010-12-28 Astellas Us Llc Method for treating or preventing sclerotic disorders using CD-2 binding agents
IL162293A0 (en) 2001-12-19 2005-11-20 Alza Corp Formulation and dosage form for increasing oral bioavailability of hydrophilic macromolecules
JP2005525374A (ja) 2002-03-04 2005-08-25 ソシエテ・ドゥ・コンセイユ・ドゥ・ルシェルシュ・エ・ダプリカーション・シャンティフィック・エス・ア・エス 担体ペプチドを含有する持続放出ドラッグ製剤
JP2006502183A (ja) 2002-09-20 2006-01-19 アルコン,インコーポレイテッド ドライアイ障害の処置のためのサイトカイン合成インヒビターの使用
US7785578B2 (en) 2002-10-11 2010-08-31 Aciont, Inc. Non-invasive ocular drug delivery
AU2004205895B2 (en) * 2003-01-16 2009-02-26 The Trustees Of The University Of Pennsylvania Compositions and methods for siRNA inhibition of ICAM-1
WO2004073701A1 (ja) * 2003-02-19 2004-09-02 Kuniyasu Soda Lfa-1抑制剤、及びその用途
WO2005010024A2 (en) 2003-07-22 2005-02-03 Science & Technology Corporation @ Unm Potent peptide inhibitors and methods of use
EP1660439A2 (en) 2003-08-08 2006-05-31 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions, and methods of use
WO2005014532A1 (en) 2003-08-08 2005-02-17 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions and methods of use
GB0323684D0 (en) 2003-10-09 2003-11-12 Jagotec Ag Improvements in or relating to organic compounds
WO2005042710A1 (en) 2003-10-28 2005-05-12 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Use of statin to kill ebv-transformed b cells
CN1914195A (zh) 2004-02-04 2007-02-14 辉瑞产品公司 取代的喹啉化合物
CA2555133A1 (en) * 2004-02-04 2005-09-01 Pfizer Products Inc. Substituted quinoline compounds
US20080234271A1 (en) 2004-04-28 2008-09-25 Icos Corporation Arylphenylamino- and Arylphenylether-Sulfide Derivatives, Useful For the Treatment of Inflammatory and Immune Diseases, and Pharmaceutical Compositions Containing Them
GT200500139A (es) 2004-06-08 2005-07-25 Metodo para la preparacion de acidos hidroxamicos
US8246949B2 (en) 2004-10-27 2012-08-21 Aciont, Inc. Methods and devices for sustained in-vivo release of an active agent
US7943660B2 (en) 2005-04-15 2011-05-17 Dominion Pharmakine S.L. Inhibitors of the LFA-1/ICAM-1 interaction and uses thereof
US20060286108A1 (en) 2005-06-16 2006-12-21 Bell Katherine A Topical compositions for the treatment of chronic wounds
JP2007099641A (ja) * 2005-09-30 2007-04-19 Tsumura & Co インドールキノキサリン類化合物、その製造方法およびそれを用いた医薬組成物
GB0520378D0 (en) 2005-10-06 2005-11-16 Novartis Ag Organic compounds
JP2009510150A (ja) * 2005-10-06 2009-03-12 ノバルティス アクチエンゲゼルシャフト Lfa−1メディエーターとしてのテトラヒドロ−ピロリジノン化合物
US20090155176A1 (en) 2007-10-19 2009-06-18 Sarcode Corporation Compositions and methods for treatment of diabetic retinopathy
US20090258070A1 (en) 2008-04-15 2009-10-15 John Burnier Topical LFA-1 antagonists for use in localized treatment of immune related disorders
US20090258069A1 (en) 2008-04-15 2009-10-15 John Burnier Delivery of LFA-1 antagonists to the gastrointestinal system

Patent Citations (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4316893A (en) * 1975-06-19 1982-02-23 Nelson Research & Development Co. Vehicle composition containing 1-substituted azacycloalkan-2-ones
US4568343A (en) * 1984-10-09 1986-02-04 Alza Corporation Skin permeation enhancer compositions
US4668506A (en) * 1985-08-16 1987-05-26 Bausch & Lomb Incorporated Sustained-release formulation containing and amino acid polymer
US4931279A (en) * 1985-08-16 1990-06-05 Bausch & Lomb Incorporated Sustained release formulation containing an ion-exchange resin
US5023252A (en) * 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US4992445A (en) * 1987-06-12 1991-02-12 American Cyanamid Co. Transdermal delivery of pharmaceuticals
US5001139A (en) * 1987-06-12 1991-03-19 American Cyanamid Company Enchancers for the transdermal flux of nivadipine
US5192741A (en) * 1987-09-21 1993-03-09 Debiopharm S.A. Sustained and controlled release of water insoluble polypeptides
US4908202A (en) * 1988-02-02 1990-03-13 Cassella Aktiengesellschaft Use of 2-oxo-1-pyrrolidineacetamide for the determination of the glomerular filtration rate in humans
US5424399A (en) * 1988-06-28 1995-06-13 The Children's Medical Center Corporation Human CR3α/β heterodimers
US5134122A (en) * 1989-07-28 1992-07-28 Debiopharm S.A. Method for preparing a pharmaceutical composition in the form of microparticles
US5288854A (en) * 1990-11-28 1994-02-22 Center For Blood Research, Inc. Functional derivatives of ICAM-1 which are substantially capable of binding to LFA-1 but are substantially incapable of binding to MAC-1
US5597567A (en) * 1991-10-04 1997-01-28 The United States Of America As Represented By The Department Of Health And Human Services Blocking cell adhesion molecules and treating animals with ocular inflammation
US6764681B2 (en) * 1991-10-07 2004-07-20 Biogen, Inc. Method of prophylaxis or treatment of antigen presenting cell driven skin conditions using inhibitors of the CD2/LFA-3 interaction
US7323171B2 (en) * 1991-10-07 2008-01-29 Astellas Us Llc Methods of treating skin conditions using inhibitors of the CD2/LFA-3 interaction
US6203793B1 (en) * 1992-02-28 2001-03-20 Board Of Regents Of The University Of Texas System Compositions and methods for treating thermal injury
US5298492A (en) * 1992-08-04 1994-03-29 Schering Corporation Diamino acid derivatives as antihypertensives
US5622700A (en) * 1992-08-21 1997-04-22 Genentech, Inc. Method for treating a LFA-1-mediated disorder
US20040120960A1 (en) * 1992-08-21 2004-06-24 Genentech, Inc. Method for treating multiple sclerosis
US6011011A (en) * 1992-09-21 2000-01-04 Pharmacia & Upjohn Company Sustained-release protein formulations
US5424289A (en) * 1993-07-30 1995-06-13 Alza Corporation Solid formulations of therapeutic proteins for gastrointestinal delivery
US5397791A (en) * 1993-08-09 1995-03-14 Merck & Co., Inc. Fibrinogen receptor antagonists
US5624837A (en) * 1994-03-03 1997-04-29 Alexion Pharmaceuticals, Inc. Nucleic acid encoding chimeric complement inhibitor proteins
US6204280B1 (en) * 1994-11-02 2001-03-20 Merck Patent Gesellschaft Mit Berschrankter Haftung Adhesion receptor antagonists
US5612052A (en) * 1995-04-13 1997-03-18 Poly-Med, Inc. Hydrogel-forming, self-solvating absorbable polyester copolymers, and methods for use thereof
US5747035A (en) * 1995-04-14 1998-05-05 Genentech, Inc. Polypeptides with increased half-life for use in treating disorders involving the LFA-1 receptor
US5877224A (en) * 1995-07-28 1999-03-02 Rutgers, The State University Of New Jersey Polymeric drug formulations
US20030068384A1 (en) * 1995-07-28 2003-04-10 Stephen Brocchini Polymeric drug formulations
US20020019446A1 (en) * 1995-07-28 2002-02-14 Stephen Brocchini Polymeric drug formulations
US5863910A (en) * 1996-01-12 1999-01-26 Bolonick; Joel Treatment of chronic inflammatory disorders of the gastrointestinal tract
US5922356A (en) * 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US6358976B1 (en) * 1996-11-27 2002-03-19 John Wityak Integrin receptor antagonists
US6180608B1 (en) * 1996-12-11 2001-01-30 Praecis Pharmaceuticals, Inc. Pharmaceutical formulations for sustained drug delivery
US5893985A (en) * 1997-03-14 1999-04-13 The Lincoln Electric Company Plasma arc torch
US20020045582A1 (en) * 1997-12-31 2002-04-18 Alexey L. Margolin Stabilized protein crystals formulations containing them and methods of making them
US7166568B1 (en) * 1998-02-09 2007-01-23 Oklahoma Medical Research Foundation Compositions and methods to inhibit formation of the C5b-9 complex of complement
US20070155671A1 (en) * 1998-10-13 2007-07-05 Nader Fotouhi Diaminopropionic acid derivatives
US20050080119A1 (en) * 1998-10-13 2005-04-14 Nader Fotouhi Diaminopropionic acid derivatives
US20040006236A1 (en) * 1998-10-13 2004-01-08 Nader Fotouhi Diaminopropionic acid derivatives
US7217728B2 (en) * 1998-10-13 2007-05-15 Hoffmann-La Roche Inc. Diaminopropionic acid derivatives
US6867203B2 (en) * 1998-12-29 2005-03-15 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory and immune-suppressive compounds
US6524581B1 (en) * 1998-12-30 2003-02-25 The Children's Medical Center Corporation Prevention and treatment of retinal ischemia and edema
US20080019977A1 (en) * 1998-12-30 2008-01-24 The Children's Medical Center Corporation Prevention and treatment of retinal ischemia and edema
US20040028648A1 (en) * 1998-12-30 2004-02-12 The Children's Medical Center Corporation Prevention and treatment of retinal ischemia and edema
US6340679B1 (en) * 1999-02-13 2002-01-22 Aventis Pharma Deutschland Gmbh Guanidine derivatives as inhibitors of cell adhesion
US20010006656A1 (en) * 1999-02-17 2001-07-05 University Of Washington Methods and compositions for inhibiting inflammation associated with pulmonary disease
US20030138488A1 (en) * 1999-12-31 2003-07-24 Kohn Joachim B Pharmaceutical formulation for regulating the timed release of biologically active compounds based on a polymer matrix
US6515124B2 (en) * 2000-02-09 2003-02-04 Hoffman-La Roche Inc. Dehydroamino acids
US20070066585A1 (en) * 2000-06-29 2007-03-22 Wang Gary T Aryl phenylheterocyclyl sulfide derivatives and their use as cell adhesion-inhibiting anti-inflammatory and immune-suppressive agents
US6521619B2 (en) * 2000-06-29 2003-02-18 Icos Corporation Aryl phenylcyclopropyl sulfide derivatives and their use as cell adhesion inhibiting anti-inflammatory and immune suppressive agents
US20030064105A1 (en) * 2000-08-25 2003-04-03 Myung-Jin Kim Lipophilic-coated microparticle containing a protein drug and formulation comprising same
US20040058968A1 (en) * 2000-11-28 2004-03-25 Genentech, Inc. LFA-1 antagonist compounds
US6872735B2 (en) * 2000-11-28 2005-03-29 Genentech, Inc. LFA-1 antagonist compounds
US20050148588A1 (en) * 2000-11-28 2005-07-07 Genentech, Inc. LFA-1 antagonist compounds
US6710064B2 (en) * 2000-12-01 2004-03-23 Bristol-Myers Squibb Co. Hydantoin compounds useful as anti-inflammatory agents
US20030044406A1 (en) * 2001-03-02 2003-03-06 Christine Dingivan Methods of preventing or treating inflammatory or autoimmune disorders by administering CD2 antagonists in combination with other prophylactic or therapeutic agents
US20030068320A1 (en) * 2001-03-02 2003-04-10 Christine Dingivan Methods of administering/dosing CD2 antagonists for the prevention and treatment of autoimmune disorders or inflammatory disorders
US20070025990A1 (en) * 2001-03-02 2007-02-01 Medimmune, Inc. Methods of administering/dosing CD2 antagonists for the prevention and treatment of autoimmune disorders or inflammatory disorders
US20030069560A1 (en) * 2001-05-03 2003-04-10 Massachusetts Eye And Ear Infirmary Implantable drug delivery device and use thereof
US6872382B1 (en) * 2001-05-21 2005-03-29 Alcon, Inc. Use of selective PDE IV inhibitors to treat dry eye disorders
US20040122047A1 (en) * 2001-06-06 2004-06-24 Aventis Pharma Limited Substituted tetrahydroisoquinolines
US7211586B2 (en) * 2001-06-06 2007-05-01 Aventis Pharma Limited Substituted tetrahydroisoquinolines
US20050004153A1 (en) * 2001-10-01 2005-01-06 Dhar T.G. Murali Spiro-hydantoin compounds useful as anti-inflammatory agents
US20040009998A1 (en) * 2001-10-01 2004-01-15 Dhar T. G. Murali Spiro-hydantoin compounds useful as anti-inflammatory agents
US7078420B2 (en) * 2001-10-01 2006-07-18 Bristol-Myers Squibb Company Spiro-hydantoin compounds useful as anti-inflammatory agents
US20070027101A1 (en) * 2001-11-09 2007-02-01 Guyer David R Methods for treating ocular neovascular diseases
US20050142066A1 (en) * 2002-06-06 2005-06-30 Dana Farber Cancer Institute, Inc Compounds or agents that inhibit and induce the formation of focal microvessel dilatations
US20040038973A1 (en) * 2002-08-13 2004-02-26 Joe Nahra Phthalimide derivatives as matrix metalloproteinase inhibitors
US20070105824A1 (en) * 2003-04-29 2007-05-10 Erickson-Miller Connie L Methods for treating degenerative diseases/injuries
US20050107399A1 (en) * 2003-09-11 2005-05-19 Kemia, Inc. Cytokine inhibitors
US7199125B2 (en) * 2003-10-02 2007-04-03 Bristol-Myers Squibb Company Spiro-cyclic compounds useful as anti-inflammatory agents
US20050119279A1 (en) * 2003-10-02 2005-06-02 Dhar T. G. M. Spiro-cyclic compounds useful as anti-inflammatory agents
US20080182839A1 (en) * 2003-11-05 2008-07-31 Sunesis Pharmaceuticals, Inc. Modulators of cellular adhesion
US20080176896A1 (en) * 2003-11-05 2008-07-24 Wang Shen Modulators of cellular adhesion
US20100093693A1 (en) * 2003-11-05 2010-04-15 Wang Shen Modulators of cellular adhesion
US7928122B2 (en) * 2003-11-05 2011-04-19 Sarcode Corporation Modulators of cellular adhesion
US20110124669A1 (en) * 2003-11-05 2011-05-26 Sarcode Corporation Modulators of Cellular Adhesion
US7314938B2 (en) * 2003-11-05 2008-01-01 Sunesis Pharmaceuticals, Inc. Modulators of cellular adhesion
US20070142317A1 (en) * 2003-12-29 2007-06-21 Qlt Usa, Inc. Topical composition for treatment of skin disorders
US7396530B2 (en) * 2004-06-09 2008-07-08 Genentech, Inc. Method of treating granuloma annulare or sarcoid
US20060052434A1 (en) * 2004-08-18 2006-03-09 Dhar T G M Pyrrolizine compounds useful as anti-inflammatory agents
US7375237B2 (en) * 2004-08-18 2008-05-20 Bristol-Myers Squibb Company Pyrrolizine compounds useful as anti-inflammatory agents
US20080108677A1 (en) * 2004-08-30 2008-05-08 Karyon-Ctt Ltd Thioxothiazolidinone Compounds For Use As Pharmaceuticals
US20060074099A1 (en) * 2004-10-01 2006-04-06 Delmonte Albert J Crystalline forms and process for preparing spiro-hydantoin compounds
US7381737B2 (en) * 2004-10-01 2008-06-03 Bristol-Myers Squibb Company Crystalline forms and process for preparing spiro-hydantoin compounds
US20060142319A1 (en) * 2004-12-14 2006-06-29 Bang-Chi Chen Pyridyl-substituted spiro-hydantoin crystalline forms and process
US20060148836A1 (en) * 2004-12-14 2006-07-06 Dhar T G M Pyridyl-substituted spiro-hydantoin compounds and use thereof
US7186727B2 (en) * 2004-12-14 2007-03-06 Bristol-Myers Squibb Company Pyridyl-substituted spiro-hydantoin compounds and use thereof
US20100092541A1 (en) * 2005-05-17 2010-04-15 John Burnier Compositions and methods for treatment of eye disorders
US20100092542A1 (en) * 2005-05-17 2010-04-15 John Burnier Compositions and methods for treatment of eye disorders
US8168655B2 (en) * 2005-05-17 2012-05-01 Sarcode Bioscience Inc. Compositions and methods for treatment of eye disorders
US20070090205A1 (en) * 2005-06-24 2007-04-26 Boehringer Ingelheim International Gmbh Nebuliser and container
US20070048216A1 (en) * 2005-08-23 2007-03-01 Norenberg Jeffrey P Non-invasive diagnostic agents of cancer and methods of diagnosing cancer, especially leukemia and lymphoma
US20070149593A1 (en) * 2005-12-23 2007-06-28 Alcon, Inc. PHARMACEUTICAL FORMULATION FOR DELIVERY OF RECEPTOR TYROSINE KINASE INHIBITING (RTKi) COMPOUNDS TO THE EYE
US20120107404A1 (en) * 2008-04-15 2012-05-03 Sarcode Bioscience Inc. Crystalline Pharmaceutical and Methods of Preparation and Use Thereof
US20130144063A1 (en) * 2008-04-15 2013-06-06 Sarcode Bioscience Inc. Crystalline Pharmaceutical and Methods of Preparation and Use Thereof
US20110092707A1 (en) * 2009-10-21 2011-04-21 Sarcode Corporation Crystalline Pharmaceutical and Methods of Preparation and Use Thereof
US20130150585A1 (en) * 2009-10-21 2013-06-13 SARcode Bioscience, Inc. Crystalline Pharmaceutical and Methods of Preparation and Use Thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Adamis AP. Is diabetic retinopathy an inflammatory disease? Br J Ophthalmol. 2002 Apr;86(4):363-5. *
Devine L, Lightman SL, Greenwood J. Role of LFA-1, ICAM-1, VLA-4 and VCAM-1 in lymphocyte migration across retinal pigment epithelial monolayers in vitro. Immunology. 1996 Jul;88(3):456-62. *
King, F.D. (Ed.), "Bioisosteres, conformational restriction and pro-drugs--case history: an example of a conformational restriction approach," Medical Chemistry: Principles and Practice, 1994, Chapter 14, 206-209. *
Meleth AD, Agrón E, Chan CC, Reed GF, Arora K, Byrnes G, Csaky KG, Ferris FL 3rd, Chew EY. Serum inflammatory markers Diabetologia. 2002 Dec;45(12):1617-34. in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2005 Nov;46(11):4295-301. *
Nam K1, Maiorov V, Feuston B, Kearsley S. Dynamic control of allosteric antagonism of leukocyte function antigen-1 and intercellular adhesion molecule-1 interaction. Proteins. 2006 Aug 1;64(2):376-84. *
Porta M, Bandello F. Diabetic retinopathyA clinical update.Diabetologia. 2002 Dec;45(12):1617-34. *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110124625A1 (en) * 2003-11-05 2011-05-26 Sarcode Corporation Modulators of Cellular Adhesion
US20080182839A1 (en) * 2003-11-05 2008-07-31 Sunesis Pharmaceuticals, Inc. Modulators of cellular adhesion
US20110124669A1 (en) * 2003-11-05 2011-05-26 Sarcode Corporation Modulators of Cellular Adhesion
US8071617B2 (en) 2003-11-05 2011-12-06 Sarcode Bioscience Inc. Modulators of cellular adhesion
US9216174B2 (en) 2003-11-05 2015-12-22 Sarcode Bioscience Inc. Modulators of cellular adhesion
US9248126B2 (en) 2003-11-05 2016-02-02 Sarcode Bioscience Inc. Modulators of cellular adhesion
US20080176896A1 (en) * 2003-11-05 2008-07-24 Wang Shen Modulators of cellular adhesion
US20100093693A1 (en) * 2003-11-05 2010-04-15 Wang Shen Modulators of cellular adhesion
US7745460B2 (en) 2003-11-05 2010-06-29 Sarcode Corporation Modulators of cellular adhesion
US7790743B2 (en) 2003-11-05 2010-09-07 Sarcode Corporation Modulators of cellular adhesion
US7928122B2 (en) 2003-11-05 2011-04-19 Sarcode Corporation Modulators of cellular adhesion
US7989626B2 (en) 2003-11-05 2011-08-02 SAR code Corporation Modulators of cellular adhesion
US9045458B2 (en) 2005-05-17 2015-06-02 Sarcode Bioscience Inc. Compositions and methods for treatment
US8592450B2 (en) 2005-05-17 2013-11-26 Sarcode Bioscience Inc. Compositions and methods for treatment of eye disorders
US9045457B2 (en) 2005-05-17 2015-06-02 Sarcode Bioscience Inc. Compositions and methods for treatment
US8771715B2 (en) 2005-05-17 2014-07-08 Sarcode Bioscience Inc. Compositions and methods for treatment
US8758776B2 (en) 2005-05-17 2014-06-24 Sarcode Bioscience Inc. Compositions and methods for treatment
US9051297B2 (en) 2005-05-17 2015-06-09 Sarcode Bioscience Inc. Compositions and methods for treatment
US10188641B2 (en) 2005-05-17 2019-01-29 Sarcode Bioscience Inc. Compositions and methods for treatment
US20060281739A1 (en) * 2005-05-17 2006-12-14 Thomas Gadek Compositions and methods for treatment of eye disorders
US8168655B2 (en) 2005-05-17 2012-05-01 Sarcode Bioscience Inc. Compositions and methods for treatment of eye disorders
US8084047B2 (en) 2005-05-17 2011-12-27 Sarcode Bioscience Inc. Compositions and methods for treatment of eye disorders
US10960087B2 (en) 2007-10-19 2021-03-30 Novartis Ag Compositions and methods for treatment of diabetic retinopathy
US8080562B2 (en) 2008-04-15 2011-12-20 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US8367701B2 (en) 2008-04-15 2013-02-05 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US20090258069A1 (en) * 2008-04-15 2009-10-15 John Burnier Delivery of LFA-1 antagonists to the gastrointestinal system
US20090257957A1 (en) * 2008-04-15 2009-10-15 John Burnier Aerosolized LFA-1 antagonists for use in localized treatment of immune related disorders
US20090258070A1 (en) * 2008-04-15 2009-10-15 John Burnier Topical LFA-1 antagonists for use in localized treatment of immune related disorders
US20090298869A1 (en) * 2008-04-15 2009-12-03 John Burnier Crystalline pharmaceutical and methods of preparation and use thereof
US11028077B2 (en) 2008-04-15 2021-06-08 Novartis Pharmaceuticals Corporation Crystalline pharmaceutical and methods of preparation and use thereof
US8871935B2 (en) 2008-04-15 2014-10-28 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US8378105B2 (en) 2009-10-21 2013-02-19 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US9353088B2 (en) * 2009-10-21 2016-05-31 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US8927574B2 (en) 2009-10-21 2015-01-06 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US20110092707A1 (en) * 2009-10-21 2011-04-21 Sarcode Corporation Crystalline Pharmaceutical and Methods of Preparation and Use Thereof
US9890141B2 (en) 2009-10-21 2018-02-13 Sarcode Bioscience Inc. Crystalline pharmaceutical and methods of preparation and use thereof
US20110104083A1 (en) * 2009-10-30 2011-05-05 Intratus, Inc. Methods and cosmetic preparations for the sustained delivery of therapeutic agents to the eye
US20110104206A1 (en) * 2009-10-30 2011-05-05 Intratus, Inc. Methods and compositions for sustained delivery of drugs
US9034830B2 (en) 2009-10-30 2015-05-19 Intratus, Inc. Methods and compositions for sustained delivery of drugs
US20110189174A1 (en) * 2010-02-01 2011-08-04 Afshin Shafiee Compositions and methods for treating, reducing, ameliorating, alleviating, or inhibiting progression of, pathogenic ocular neovascularization
US20110281901A1 (en) * 2010-05-05 2011-11-17 Glaxo Wellcome Manufacturing Pte Ltd. Pharmaceutical compositions and methods of making same
US10751316B2 (en) 2012-06-05 2020-08-25 Olatec Therapeutics Llc Pharmaceutical composition for treating inflammation and pain
US20130324603A1 (en) * 2012-06-05 2013-12-05 Olatec Industries Llc Pharmaceutical composition for treating inflammation and pain
US9085553B2 (en) 2012-07-25 2015-07-21 SARcode Bioscience, Inc. LFA-1 inhibitor and methods of preparation and polymorph thereof
US10214517B2 (en) 2012-07-25 2019-02-26 Sarcode Bioscience Inc. LFA-1 inhibitor and methods of preparation and polymorph thereof
US10906892B2 (en) 2012-07-25 2021-02-02 Novartis Pharmaceuticals Corporation LFA-1 inhibitor and methods of preparation and polymorph thereof
US11018173B2 (en) 2018-06-18 2021-05-25 Samsung Electronics Co., Ltd. Image sensor
US11753382B2 (en) 2019-06-25 2023-09-12 Translatum Medicus Inc. Processes of making 2-((1-benzyl-1H-indazol-3-yl)methoxy)-2-methylpropanoic acid and its derivatives

Also Published As

Publication number Publication date
JP2011500683A (ja) 2011-01-06
CN101873797A (zh) 2010-10-27
US10960087B2 (en) 2021-03-30
CA2958665C (en) 2021-03-02
JP2014221808A (ja) 2014-11-27
WO2009054914A1 (en) 2009-04-30
US20210338839A1 (en) 2021-11-04
CA2702984A1 (en) 2009-04-30
AU2008317473A1 (en) 2009-04-30
EP3797775A1 (en) 2021-03-31
JP5808037B2 (ja) 2015-11-10
CA2958665A1 (en) 2009-04-30
US20160220702A1 (en) 2016-08-04
ES2830024T3 (es) 2021-06-02
EP3167886B1 (en) 2020-08-05
EP2209371A1 (en) 2010-07-28
AU2008317473B2 (en) 2014-07-17
CA2702984C (en) 2017-04-11
ES2630406T3 (es) 2017-08-21
EP2209371B1 (en) 2017-01-04
CA3105972A1 (en) 2009-04-30
MX2010004281A (es) 2010-09-10
EP2209371A4 (en) 2011-11-02
EP3167886A1 (en) 2017-05-17

Similar Documents

Publication Publication Date Title
US20210338839A1 (en) Compositions and methods for treatment of diabetic retinopathy
US20230233551A1 (en) Compositions and methods for treatment
AU2014208186A1 (en) Compositions and methods for treatment of diabetic retinopathy

Legal Events

Date Code Title Description
AS Assignment

Owner name: SARCODE CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BURNIER, JOHN;GADEK, THOMAS;SEMBA, CHARLES;REEL/FRAME:022167/0029

Effective date: 20081201

AS Assignment

Owner name: SARCODE BIOSCIENCE INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:SARCODE CORPORATION;REEL/FRAME:026876/0312

Effective date: 20110715

AS Assignment

Owner name: SILICON VALLEY BANK, CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:SARCODE BIOSCIENCE INC.;REEL/FRAME:029778/0888

Effective date: 20130208

AS Assignment

Owner name: SARCODE BIOSCIENCE INC., CALIFORNIA

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:SILICON VALLEY BANK;REEL/FRAME:030295/0028

Effective date: 20130424

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SARCODE BIOSCIENCE INC.;REEL/FRAME:050900/0268

Effective date: 20190701