WO2003099809A1 - HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY α4 INTEGRINS - Google Patents

HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY α4 INTEGRINS Download PDF

Info

Publication number
WO2003099809A1
WO2003099809A1 PCT/US2003/016804 US0316804W WO03099809A1 WO 2003099809 A1 WO2003099809 A1 WO 2003099809A1 US 0316804 W US0316804 W US 0316804W WO 03099809 A1 WO03099809 A1 WO 03099809A1
Authority
WO
WIPO (PCT)
Prior art keywords
ylcarbonyloxy
pyrimidin
diethylamino
phenylalanine
compound
Prior art date
Application number
PCT/US2003/016804
Other languages
French (fr)
Inventor
Andrei W. Konradi
Christopher M. Semko
Ying-Zi Xu
Frank Stappenbeck
Brian P. Stupi
Jenifer Smith
Eugene D. Thorsett
Michael A. Pleiss
Original Assignee
Elan Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MXPA04010995A priority Critical patent/MXPA04010995A/en
Priority to BR0308881-2A priority patent/BR0308881A/en
Priority to IL16422503A priority patent/IL164225A0/en
Priority to EP03731419A priority patent/EP1507775B1/en
Priority to EA200401562A priority patent/EA008256B1/en
Priority to JP2004507466A priority patent/JP4469715B2/en
Priority to CA2481926A priority patent/CA2481926C/en
Application filed by Elan Pharmaceuticals, Inc. filed Critical Elan Pharmaceuticals, Inc.
Priority to DE60325583T priority patent/DE60325583D1/en
Priority to DK03731419T priority patent/DK1507775T3/en
Priority to UA20041008397A priority patent/UA76339C2/en
Priority to KR1020047018173A priority patent/KR100978832B1/en
Priority to US10/494,790 priority patent/US7135477B2/en
Priority to NZ535504A priority patent/NZ535504A/en
Priority to AU2003240823A priority patent/AU2003240823C1/en
Publication of WO2003099809A1 publication Critical patent/WO2003099809A1/en
Priority to NO20043900A priority patent/NO329933B1/en
Priority to IL164225A priority patent/IL164225A/en
Priority to HK05106028.8A priority patent/HK1072608A1/en
Priority to US11/527,901 priority patent/US7427628B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/50Three nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates to compounds wliich inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated by ⁇ 4 integrins where the ⁇ 4 integrin is preferably VLA-4.
  • VLA-4 (also referred to as ⁇ 4 ⁇ j integrin and CD49d/CD29), first identified by Hemler and Takada, 1 is a member of the ⁇ l integrin family of cell surface receptors, each of which comprises two subunits, an ⁇ chain and a ⁇ chain. VLA-4 contains an ⁇ 4 chain and a ⁇ l chain. There are at least nine ⁇ l integrins, all sharing the same ⁇ l chain and each having a distinct ⁇ chain.
  • VLA-4 binds to fibronectin.
  • VLA-4 also binds non-matrix molecules that are expressed by endothehal and other cells. These non-matrix molecules include VC AM- 1 , which is expressed on cytokine-activated human umbilical vein endothehal cells in culture. Distinct epitopes of VLA-4 are responsible for the fibronectin and VCAM-1 binding activities and each activity has been shown to be inhibited independently. 2
  • Intercellular adhesion mediated by VLA-4 and other cell surface receptors is associated with a number of inflammatory responses.
  • activated vascular endothehal cells express molecules that are adhesive for leukocytes.
  • the mechanics of leukocyte adhesion to endothehal cells involves, in part, the recognition and binding of cell surface receptors on leukocytes to the corresponding cell surface molecules on endothehal cells. Once bound, the leukocytes migrate across the blood vessel wall to enter the injured site and release chemical mediators to combat infection.
  • adhesion receptors of the immune system see, for example, Springer 3 and Osborn.
  • Inflammatory brain disorders such as experimental autoimmune encephalomyelitis (EAE), multiple sclerosis (MS) and meningitis, are examples of central nervous system disorders in which the endothelium leukocyte adhesion mechanism results in destruction to otherwise healthy brain tissue.
  • EAE experimental autoimmune encephalomyelitis
  • MS multiple sclerosis
  • M multiple sclerosis
  • meningitis are examples of central nervous system disorders in which the endothelium leukocyte adhesion mechanism results in destruction to otherwise healthy brain tissue.
  • BBB blood brain barrier
  • the leukocytes release toxic mediators that cause extensive tissue damage resulting in impaired nerve conduction and paralysis.
  • tissue damage also occurs via an adhesion mechanism resulting in migration or activation of leukocytes.
  • tissue damage also occurs via an adhesion mechanism resulting in migration or activation of leukocytes.
  • the initial insult following myocardial ischemia to heart tissue can be further complicated by leukocyte entry to the injured tissue causing still further insult (Vedder et al.).
  • inflammatory or medical conditions mediated by an adhesion mechanism include, by way of example, asthma 6"8 , Alzheimer's disease, atherosclerosis, 9"10 AIDS dementia, 11 diabetes 12" 14 (including acute juvenile onset diabetes), inflammatory bowel disease 15 (including ulcerative colitis and Crohn's disease), multiple sclerosis, 16"17 rheumatoid arthritis, 18"21 tissue transplantation, 22 tumor metastasis, 23"28 meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte- mediated lung injury such as that which occurs in adult respiratory distress syndrome.
  • Substituted aminopyrimidines as a class, have been disclosed as inhibiting binding of VLA-4 to VCAM-1 and, accordingly, exhibit anti- inflammatory properties. 29 While these compounds possess antagonist properties to such binding, enhanced bioavailability of these compounds would augment their efficacy.
  • This invention is directed to the discovery that certain N-[2-N',N'- diethylamino-5-aminosulfonylphenylpyrimidin-4-yl]-p-carbomyloxy- phenylala ine compounds possess unexpectedly superior bioavailabihty, as measured by their AUC, as compared to other substituted aminopyrimidine compounds previously disclosed.
  • this invention is directed to a compound of Formula (I) :
  • each X is independently fluoro, chloro or bromo; p is an integer from 0 to 3;
  • R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, pyrrolyl, 2,5-dihydopyrrol-l-yl, piperidinyl, or 1,2,3,6-tetrahydro ⁇ yridin-l-yl;
  • R 2 is selected from the group consisting of lower alkyl, lower alkenyl, and lower alkylenecycloalkyl; and pharmaceutically acceptable salts thereof.
  • R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group.
  • this invention provides compounds of Formula (II):
  • each X is independently selected from the group consisting of fluoro and chloro; m is an integer equal to 1 or 2;
  • R 2 is selected from the group consisting of lower alkyl, lower alkenyl, and lower alkylenecycloalkyl; R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
  • this invention provides compounds of Formula (III)
  • each X is independently fluoro or chloro; n is zero or one;
  • R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
  • this invention is directed to a compound of Fo ⁇ nula (IN):
  • each X is independently fluoro, chloro or bromo;
  • p is an integer from 0 to 3;
  • R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, pyrrolyl, 2,5-dihydopyrrol-l-yl, piperidinyl, or 1,2,3,6-tetrahydropyridin-l-yl;
  • R 2 is lower alkynyl; and pharmaceutically acceptable salts thereof.
  • R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group and R 2 is propargyl.
  • this invention provides compounds of Formula (V):
  • each X is independently selected from the group consisting of fluoro and chloro; m is an integer equal to 1 or 2;
  • R 2 is lower alkynyl
  • this invention provides compounds of Formula (VI)
  • each X is independently fluoro or chloro; n is zero or one; R 2 is lower alkynyl;
  • R 1 and R 3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
  • N- [2-N ' ,N ' -diethylamino-5 -aminosulfonylphenylpyrimidin-4-yl] -p- carbomyloxy-phenylalanine compounds within the scope of this invention include those set forth in Table I as follows:
  • Specific compounds within the scope of this invention include the following compounds. As used below, these compounds are named based on phenylalanine derivatives but, alternatively, these compounds could have been named based on N-[2-N',N'-diethylamino-5-aminosulfoiiylphenyl-pyrimidin- 4-yl]- -carbomyloxyphenylalanine derivatives or 2- ⁇ 2-diethylamino-5- [(benzenesulfonyl)methylamino]- ⁇ yrimidin-4-ylamino ⁇ -p -carbamoyloxy- phenyl)propionic acid derivatives.
  • this invention provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the compounds defined herein.
  • this invention is directed to a method for treating a disease mediated at least in part by ⁇ 4 integrins, preferably VLA-4, in a patient, which method comprises administering a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of this invention.
  • the compounds and pharmaceutical compositions of this invention are useful for treating disease conditions mediated at least in part by ⁇ 4 integrins, preferably VLA-4, or leucocyte adhesion.
  • disease conditions include, by way of example, asthma, Alzheimer's disease, atherosclerosis, ALDS dementia, diabetes (including acute juvenile onset diabetes), inflammatory bowel disease
  • Other disease conditions include, but are not limited to, inflammatory conditions such as erythema nodosum, allergic conjunctivitis, optic neuritis, uveitis, allergic rhinitis, Ankylosing spondylitis, psoriatic arthritis, vasculitis, Reiter's syndrome, systemic lupus erythematosus, progressive systemic sclerosis, polymyositis, dermatomyositis, Wegner's granulomatosis, aortitis, sarcoidosis, lymphocytopenia, temporal arteritis, pericarditis, myocarditis, congestive heart failure, polyarteritis nodosa, hypersensitivity syndromes, allergy, hypereosinophilic syndromes, Churg-Strauss syndrome, chronic obstructive pulmonary disease, hypersensitivity pneumonitis, chronic active hepatitis, interstitial cystitis, autoimmune endocrine failure, primary
  • the disease condition is an inflammatory disease.
  • this invention relates to compounds which inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated at least in part by ⁇ 4 integrins, preferably VLA-4.
  • ⁇ 4 integrins preferably VLA-4.
  • lower alkyl refers to monovalent alkyl groups having from 1 to 5 carbon atoms including straight and branched chain alkyl groups. This term is exemplified by groups such as methyl, ethyl, wo-propyl, n-propyl, ra-butyl, iso-hutyl, -.ec-butyl, t-butyl, ⁇ .-pentyl and the like.
  • lower alkylene refers to divalent alkylene groups of from 1 to 4 carbon atoms including straight and branched chain alkylene groups. This term is exemplified by groups such as methylene, ethylene, /.-propylene, iso- propylene (-CH 2 CH(CH 3 )- and -CH(CH 3 )CH 2 -) and the like.
  • lower alkynyl refers to an alkynyl group preferably having from 2 to 6 carbon atoms and having at least 1 site and preferably only 1 site of alkynyl unsaturation (i.e., -C ⁇ C-). This term is exemplified by groups such as acetyl (-C ⁇ CH), propargyl (-CH 2 -C ⁇ CH), 3-butynyl
  • lower cycloalkyl refers to cyclic alkyl groups of from 3 to 6 carbon atoms having a single cyclic ring including, by way of example, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • lower alkylenecycloalkyl refers to the group consisting of a lower alkylene-lower cycloalkyl, as defined herein. Such groups are exemplified by methylenecyclopropyl (-CH 2 -cyclopropyl), ethylenecyclopropyl and the like.
  • “Pharmaceutically acceptable carrier” means a carrier that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier that is acceptable for veterinary use as well as human pharmaceutical use.
  • “A pharmaceutically acceptable carrier” as used in the specification and claims includes both one and more than one such carrier.
  • Treating" or “treatment” of a disease includes:
  • a “therapeutically effective amount” means the amount of a compound that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • “Pharmaceutically acceptable salt” refers to pharmaceutically acceptable salts of a compound of Formula I which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
  • Integrins are a large family of homologous transmembrane linker proteins that are the principal receptors on animal cells for binding most extracellular matrix proteins, such as collagen, fibronectin, and laminin.
  • the integrins are heterodimers comprised of an ⁇ chain and a ⁇ chain.
  • ⁇ 4 integrins refers to the class of heterodimer, enzyme-linked cell-surface receptors that contain the ⁇ 4 subunit paired with any of the ⁇ subunits.
  • VLA-4 is an example of an ⁇ 4 integrin, and is a heterodimer of the ⁇ 4 and ⁇ j subunits, and is also referred to as ⁇ 4 ⁇ ! integrin.
  • the compounds of this invention can be prepared from readily available starting materials using the methods and procedures set forth in the examples below. These methods and procedures outline specific reaction protocols for preparing N-[2-N',N'-diethylamino-5-aminosulfonylphenyl- yrimidin-4-yl]-j-)-carbomyloxy-phenylalanme compounds. Compounds within the scope not exemplified in these examples and methods are readily prepared by appropriate substitution of starting materials which are either commercially available or well known in the art. [0087] Other procedures and reaction conditions for preparing the compounds of this invention are described in the examples set forth below. Additionally, other procedures for preparing compounds useful in certain aspects of this invention are disclosed in U.S. Patent 6,492,372, issued December 10, 2002; the disclosure of which is incorporated herein by reference in its entirety.
  • compositions When employed as pharmaceuticals, the compounds of this invention are usually administered in the form of pharmaceutical compositions. These compositions can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. These compositions are effective by both injectable and oral delivery. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • compositions which contain, as the active ingredient, one or more of the compounds of Formula I above associated with pharmaceutically acceptable carriers.
  • the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container.
  • the excipient employed is typically an excipient suitable for administration to human subjects or other mammals.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, macrocrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy- benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • compositions are preferably formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It, will be understood, however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • 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. Preferably the 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 breathed directly from the nebulizing device or the nebulizing device may be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • Hard gelatin capsules containing the following ingredients are prepared:
  • Quantity Ingredient (mg/capsule)
  • a tablet formula is prepared using the ingredients below:
  • Quantity Ingredient (mg/tablet)
  • the components are blended and compressed to form tablets, each weighing 240 mg.
  • a dry powder inhaler formulation is prepared containing the following components:
  • the active mixture is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
  • Example 4 Tablets, each containing 30 mg of active ingredient, are prepared as follows:
  • the active ingredient, starch and cellulose are passed through a No. 20 mesh U.S. sieve and mixed thoroughly.
  • the solution of polyvinylpyrrolidone is mixed with the resultant powders, which are then passed through a 16 mesh U.S. sieve.
  • the granules so produced are dried at 50° to 60 °C and passed through a 16 mesh U.S. sieve.
  • the sodium carboxymethyl starch, magnesium stearate, and talc previously passed through a No. 30 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 120 mg.
  • Suppositories each containing 25 mg of active ingredient are made as follows:
  • the active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed to cool.
  • the medicament, sucrose and xanthan gum are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water.
  • the sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
  • Quantity Ingredient (mg/capsule)
  • the active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 425 mg quantities.
  • An intravenous formulation may be prepared as follows:
  • a topical formulation may be prepared as follows:
  • White Soft Paraffin to 100 g The white soft paraffin is heated until molten. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The active ingredient is added and stirring is continued until dispersed. The mixture is then cooled until solid.
  • transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent 5,023,252, issued June 11, 1991, herein incorporated by reference.
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Direct or indirect placement techniques may be used when it is desirable or necessary to introduce the pharmaceutical composition to the brain.
  • Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier.
  • a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier.
  • One such implantable delivery system used for the transport of biological factors to specific anatomical regions of the body is described in U.S. Patent 5,011,472 which is herein incorporated by reference.
  • Indirect techniques which are generally preferred, usually involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs. Latentiation is generally achieved through blocking of the hydroxy, carbonyl, sulfate, and primary amine groups present on the drug to render the drug more lipid soluble and amenable to transportation across the blood-brain barrier.
  • the delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blood-brain barrier.
  • the compounds of this invention inhibit, in vivo, adhesion of leukocytes to endothehal cells mediated at least in part by ⁇ 4 integrins, preferably VLA-4, by competitive binding to ⁇ 4 integrins, preferably VLA-4.
  • the compounds of this invention can be used in the treatment of mammalian diseases mediated at least in part by ⁇ 4 integrins, preferably VLA- 4, or leucocyte adhesion.
  • diseases include inflammatory diseases in mammalian patients such as asthma, Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes (including acute juvenile onset diabetes), inflammatory bowel disease (including ulcerative colitis and Crohn's disease), multiple sclerosis, rheumatoid arthritis, tissue transplantation, tumor metastasis, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs in adult respiratory distress syndrome.
  • compositions are administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications.
  • An amount adequate to accomplish this is defined as "therapeutically effective dose.” Amounts effective for this use will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the inflammation, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient are in the form of pharmaceutical compositions described above. These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to
  • the therapeutic dosage of the compounds of the present invention will vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the dose will typically be in the range of about 20 ⁇ g to about 500 ⁇ g per kilogram body weight, preferably about 100 g to about 300 ⁇ g per kilogram body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.1 pg to 1 mg per kilogram body weight.
  • Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Hct hematocrit or measurement of packed red blood cells obtained by centrifugation in a volume of a blood sample
  • IgG Fc a binding domain of the immunoglobulin
  • MCHC mean corpuscular hemoglobin count expressed as a percentage
  • MCV mean corpuscular volume; the avg. volume of erythrocytes, conventionally expressed in cubic
  • Step 1 Preparation of 2,4-Dichloro-5-nitropyrimidine (2).
  • Nitrouracil, (1) was treated with phosphorous oxychloride (POCl 3 ) and N,N- dimethylaniline (PhNMe 2 ), according to the procedure of Whittaker (J. Chem. Soc. 1951, 1565), to give compound 2.
  • Compound 2 is also available from City Chemical (West Haven, CT).
  • Step 2 Preparation of N-(2-[N , ,N'-diethylamino]-5- nitropyrimidin-4-yl)-L-tyrosine tert-butyl ester (3).
  • L- tyrosine tert-butyl ester H-Tyr(OH)-OtBu
  • 2,4-dichloro-5-nitropyrimidine 25g, 0.129 mol
  • N,N-diiso ⁇ ro ⁇ ylethylamine (EtiPr 2 N) (33.7 mL, 0.194 mol) was added dropwise. After stirring for 1 h at -10 °C, diethylamine (Et 2 NH) (66.73 mL, 0.645 mol) was added slowly, and then the reaction mixture was warmed to room temperature overnight. The reaction mixture was diluted with diethyl ether (500 mL), and the organic layer was washed with 0.2 N citric acid (3 x
  • Step 3 Preparation of N-(2-[N',N , -diethylamino]-5- nitropyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-phenyIaIanine tert-butyl ester (4).
  • N-(2-[N',N'-diethylamino]-5- nitropyrimidin-4-yl)-L-tyrosine tert-butyl ester 37.39 g, 0.087 mol
  • Step 4 Preparation of N-(2-[N',N'-diethylamino]-5- aminopyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-p- ⁇ enylalanine tert-butyl ester (5).
  • Step 5 Preparation of N-(2-[N',N'-diethylamino]-5-[N M -(4- chlorophenyl-sulf onyl) amino] pyrimidin-4-yI)-4 '-(py r rolidin-l - ylcarbonyloxy) -L-phenylalanine tert-butyl ester (6).
  • Step 6 Preparation of N-(2-[N',N'-diethyIamino]-5-[N"-(4- chlorophenyl-sulfonyl)-N M -ethylammo]pyrimidin-4-yl)-4'-(pyrrolidin-l- ylcarbonyloxy)-L-phenyIaIanine tert-butyl ester (7).
  • Step 7 Preparation of N-(2-[N',N'-diethylamino]-5-[N"-(4- chlorophenylsulfonyl)-N"-ethylamino]pyrimidin-4-yl)-4 , -(pyrrolidin-l- ylcarbonyloxy)-L-phenylalanine hydrochloride (8).
  • N"-ethylamino] pyrimidin-4-yl)-4'-(pynolidin- l-ylcarbonyloxy)-L- phenylalanine tert-butyl ester (36.21 g, 0.052 mol) was heated to 70 °C for 2 h and then concentrated in vacuo. The residue was dissolved again in formic acid (500 mL) and heated again at 70 °C for 2 h. The solution was reduced in volume by 80% and then treated with at least 1 eq. of 1.0 N HC1 (52 mL,
  • Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 1.
  • Step 5 was performed using 4-fluorobenzenesulfonyl chloride in place of 4- chlorobenzenesulfonyl chloride.
  • ⁇ NMR (CD 3 OD) ⁇ 8.17 (bs, 1H), 7.90-7.87 (m, 2H), 7.40-7.34 (m, 2H),
  • Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 2.
  • Step 6 was performed using dimethyl sulfate in place of ethyl iodide.
  • Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 1.
  • Step 6 was performed using dimethyl sulfate in place of ethyl iodide.
  • ⁇ NMR (CD 3 OD) ⁇ 8.20 (bs, IH), 7.83-7.80 (m, 2H), 7.67-7.64 (m, 2H),
  • Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 3.
  • Step 3 was perfonned using 1-piperidinecarbonyl chloride in place of 1- pyrrolidinecarbonyl chloride.
  • Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 2.
  • Step 3 was performed using 1-piperidinecarbonyl chloride in place of 1- pyrrolidinecarbonyl chloride.
  • 1H NMR (CD 3 OD) ⁇ 8.17 (bs, IH), 7.91-7.85 (m, 2H), 7.39-7.31 (m, 3H),
  • Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 2.
  • Step 3 was performed according to the following procedure.
  • azetidine (3.30 g, 0.0578 mmol) was added dropwise, and the reaction mixtures was warmed to room temperature and stirred overnight.
  • the reaction mixture was diluted with EtOAc (100 mL) and hexanes (100 mL), and then was extracted repeatedly with 10% aqueous K 2 CO 3 , until no yellow color (4- nitrophenol) was seen in the aqueous phase.
  • Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 7.
  • Step 6 was performed using dimethyl sulfate in place of ethyl iodide.
  • Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 8.
  • Step 5 was performed using 4-chlorobenzenesulfonyl chloride in place of 4- fluorobenzenesulfonyl chloride.
  • Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 7.
  • Step 5 was performed using 4-chlorobenzenesulfonyl chloride in place of 4- fluorobenzenesulfonyl chloride.
  • Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 3.
  • Step 5 was performed using 2,4-difluorobenzenesulfonyl chloride in place of 4- fluorobenzenesulfonyl chloride.
  • Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 2.
  • Step 5 was performed using 2,4-difluorobenzenesulfonyl chloride in place of 4- fluorobenzensulfonyl chloride.
  • Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 11.
  • Step 3 was performed as for Example 7.
  • Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 12. Step 3 was performed as for Example 7.
  • Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 2.
  • Step 6 was performed using propargyl bromide in place of ethyl iodide. !H NMR (CDC1 3 ) ⁇ 1.18 (m, 6H), 1.93 (bs, 4H), 2.37 (s, IH), 3.00-3.70 (m,
  • Steps 1 , 2, 3, 4, 5 and 7 were perfonned as for Example 11. Step 6 was performed using propargyl bromide in place of dimethyl sulfate.
  • Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 16.
  • Step 3 was performed as for Example 7.
  • iH NMR (CDC1 3 ) ⁇ 1.18 (m, 6H), 2.34 (m, 3H), 3.00-3.75 (m, 6H), 3.80-4.25
  • Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 7.
  • Step 6 was performed using propargyl bromide in place of ethyl iodide.
  • Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 1.
  • Step 6 was performed using propargyl bromide in place of ethyl iodide.
  • Example A ⁇ 4 ⁇ ' Integrin Adhesion Assay JurkatTM Cell Adhesion to Human Plasma Fibronectin
  • 96 well plates (Costar 3590 EIA plates) were coated with human fibronectin (Gibco/BRL, cat #33016-023) at a concentration of 10 ⁇ g/mL overnight at 4°C. The plates were then blocked with a solution of bovine serum albumin (BSA; 0.3%) in saline.
  • BSA bovine serum albumin
  • JurkatTM cells (maintained in log phase growth) were labeled with Calcein AM according to the manufacturer's instructions, and suspended at a concentration of 2 x 10 6 cells/mL in Hepes/Saline/BSA. The cells were then exposed to test and control compounds for 30 minutes at room temperature before transfer to individual wells of the fibronectin coated plate. Adhesion was allowed to occur for 35 minutes at 37°C. The wells were then washed by gentle aspiration and pipetting with fresh saline. Fluorescence associated with the remaining adherent cells was quantified using a fluorescence plate reader at EX 485/EM 530.
  • JurkatTM cells at 1 : 10 on day one, and 1 :2 on day two to perform assay on day 3.
  • the cells split 1 : 10 on day one were split 1 :4 on day 3 for a day 4 assay.
  • the assay plates were prepared by first making a working solution of Gibco/BRL Human Fibronectin (cat # 33016-023) in PB S++, at 10 ⁇ g/mL.
  • a Costai" 3590 EIA plate was then coated with 50 ⁇ L/well for 2 hours at room temperature (thought it can also be left overnight at 4 °C). Finally the plate was asperated and blocked with Hepes/Saline Buffer, 100 ⁇ L/well, for 1 hour at RT followed by washing 3X with 150 ⁇ L of PBS++.
  • Compound dilutions were accomplished by preparing 1 :3 serial dilutions of compounds as follows. For each plate (4 compounds/plate) 600 ⁇ L were added to 4 Bio-Rad Titertubes in a Titertube rack. Enough compound was added to each appropriate tube to give a 2X concentration using methods well known in the art. Using Falcon Flexiplates, 100 ⁇ L of Hepes/Saline buffer or human serum were added to rows B through G. A multi-channel pipetter set to 180 ⁇ L was used to with four tips spaced evenly the pipetter.
  • Each set of four tubes was mixed 5 times and 180 ⁇ L of 2X compound was transferred to the first column of each compound dilution in Row B, leaving Row A empty. 180 ⁇ L were added to the other wells in Row A. Serial dilutions were performed down the plate by transferring 50 ⁇ L to the next dilution and mixing 5 times, changing tips each time after mixing. Dilutions were stopped at Row F. Row G had no compound present.
  • a 20 ⁇ g/mL solution in Hepes/Saline buffer or human serum, of 21/6 antibody was the positive control and was set aside in a reagent trough to add to cell suspension plate.
  • the cell staining was accomplished by first harvesting the log-phase JurkatTM cells by centrifugation in 50 mL tubes (1100 rpm for 5 minutes). The cells were resuspended in 50 mL PBS++, spun, and resuspend in 20 mL PBS++. The cells were stained by adding 20 ⁇ L of Calcein AM for 30 minutes RT. The volume was brought to 50 mL with Hepes/Saline buffer and the cells were counted, spun, and resuspend to 2 x 10 6 cells/mL in Hepes/Saline buffer or human serum.
  • Each plate was washed, row by row, by gently pipetting 100 ⁇ L of RT. PBS++ down the sides of the wells and turning the plate 90 degrees to aspirate. This procedure was repeated for a total of 3 washes. Each well was filled with 100 ⁇ L after washing by pipetting down the side of the well.
  • IC 50 value was calculated for each compound, both in the presence of the human serum and in the absence of human serum. IC 50 is concentration at which the growth or activity is inhibited by 50% .
  • the data is presented in the following tables.
  • EAE Experimental Autoimmune Encephalomyehtis
  • the Jurkat cells are diluted two-fold into either normal plasma samples containing known candidate compound amounts in various concentrations ranging from 66 ⁇ M to 0.01 ⁇ M, using a standard 12 point serial dilution for a standard curve, or into plasma samples obtained from the peripheral blood of candidate compound-treated animals.
  • Cells are then incubated for 30 minutes at room temperature, washed twice with phosphate-buffered saline ("PBS") containing 2% fetal bovine serum and lmM each of calcium chloride and magnesium chloride (assay medium) to remove unbound 15/7 antibody.
  • PBS phosphate-buffered saline
  • the cells are then exposed to phycoerythrin-conjugated goat F(ab') 2 anti-mouse IgG Fc (Immunotech, Westbrook, ME), which has been adsorbed for any non-specific cross-reactivity by co-incubation with 5% serum from the animal species being studied, at 1 :200 and incubated in the dark at 4 °C for 30 minutes.
  • the data is then graphed as fluorescence versus dose, e.g., in a normal dose-response fashion.
  • the dose levels that result in the upper plateau of the curve represent the levels needed to obtain efficacy in an in vivo model.
  • the oral bioavailabihty was screened by dosing rats with a cassette, i.e. mixture of 6 compounds per dosing solution.
  • the cassette included 5 test articles and a standard compound, for a total dose of 10 mg/kg.
  • Each compound/test article was converted to the sodium salt with equimolar 1 N
  • the cassette was prepared by mixing equal volumes of each of the six solutions.
  • the cassette dosing solution was mixed well and then the pH was adjusted to 7.5-9.
  • the dosing solution was prepared the day before the study and stirred overnight at room temperature.
  • Male Sprague Dawley (SD) rats from Charles River Laboratories, 6-8 weeks old were used in this screen. Rats were quarantined for at least one day and had continuous access to food and water. On the night before the administration of the cassette, the rats were fasted for approximately 16 h.
  • Blood samples were kept in heparinized microtainer tubes under sub- ambient temperature (4 °C) before they were processed. Blood samples were centrifuged (10000 rpm for 5 minutes) and plasma samples were removed and stored in a -20 °C freezer until analyzed for drug levels. Drug levels in the plasma were analyzed using the following protocol for direct plasma precipitation.
  • the in vivo plasma samples were prepared in a 1.5 mL 96-well plate, by adding, in order, 100 ⁇ L of the test plasma, 150 ⁇ L of methanol, followed by vortexing for 10-20 seconds. 150 ⁇ L of 0.05 ng/ ⁇ L of an Internal Standard in acetonitrile were added and vortexed for 30 seconds.
  • the standard curve samples were prepared in a 1.5 mL 96-well plate, by adding, in order, 100 ⁇ L of control mouse plasma, followed by 150 ⁇ L of methanol and vortexing for 10-20 seconds. 150 ⁇ L of 0.05 ng/ ⁇ L of an internal Standard in acetonitrile were added and vortexed for 30 seconds. The samples were spiked with 0-200 ng (10 concentrations) of the compound of interest in 50% methanol to obtain a standard curve range of 0.5 ng/mL - 2,000 ng/mL. Again, the sample was vortexed for 30 seconds.
  • LC/MS/MS was then run using a PE-Sciex API-3000 triple quadurpole mass spectrometer (SN0749707), Perkin-Elmer, Series200auto-sampler, and shimadzu 10A pump. Acquisition was done with PE-Sciex Analyst (vl.l) and data analysis and quantification were accomplished using PE-Sciex Analyst (vl.l).
  • a 5-50 ⁇ L sample volume was injected onto a reverse phase ThermoHypersil DASH- 18 column (Keystone 2.0 x 20 mm, 5 ⁇ m, PN: 8823025-701) using a mobile phase of 25% CH 3 OH, 0.1% TFA-100% CH 3 OH, 0.1% TFA.
  • the run time was about 8 minutes at a flow rate of about 300 ⁇ L/minutes.
  • AUC Area Under the Curve
  • AUC 0 -* S((C n + C n+1 )/2)) • (t n+] - t n ) [( ⁇ g/mL)h]
  • Inflammatory conditions mediated by ⁇ 4 ⁇ j integrin include, for example, eosinophil influx, airway hyper-responsiveness and occlusion that occurs with chronic asthma.
  • the following describes animal models of asthma that were used to study the in vivo effects of the compounds of this invention for use in treating asthma. Rat Asthma Model
  • the lungs were lavaged via a tracheal cannula using 3 aliquots (4 mL) of Hank's solution (HBSS x 10, 100 mL; EDTA 100 mM, 100 mL; HEPES 1 M, 25 mL; made up to 1 L with H 2 O); recovered cells were pooled and the total volume of recovered fluid adjusted to 12 mL by addition of Hank's solution. Total cells were counted (Sysmex microcell counter F-500, TOA Medical
  • smears were made by diluting recovered fluid (to approximately 10 6 cells/mL) and pipetting an aliquot (100 ⁇ L) into a centrifuge (Cytospin, Shandon, U.K.). Smears were air dried, fixed using a solution of fast green in methanol (2 mg/mL) for 5 seconds and stained with eosin G (5 seconds) and thiazine (5 seconds) (Diff-Quick, Browne Ltd. U.K.) in order to differentiate eosinophils, neutrophils, macrophages and lymphocytes. A total of 500 cells per smear were counted by light microscopy under oil immersion (x 100).
  • mice are challenged on days 28 and 29 with aerosolized 1% ova (in 0.9% saline) for 20 minutes. Mice are euthanized and bronchaveolar lavage samples (3 mL) are collected on day 30, 48 hours post first challenge. Eosinopliils were quantified by a FACs/FITC staining method. Compounds of this invention were formulated into a 0.5% carboxymethylcellulose and 2% Tween ⁇ O suspension and administered orally to mice which had been sensitized to the allergen, ovalbumin. Compounds which inhibited allergen-induced leucocyte accumulation in the airways of actively sensitized C57BL/6 mice were considered to be active in this model.
  • Allergic sheep which are shown to develop both early and late bronchial responses to inhaled Ascaris suum antigen were used to study the airway effects of the candidate compounds. Following topical anesthesia of the nasal passages with 2% lidocaine, a balloon catheter was advanced through one nostril into the lower esophagus. The animals were then incubated with a cuffed endotracheal tube through the other nostril with a flexible fiberoptic bronchoscope as a guide.
  • Pleural pressure was estimated according to Abraham (1994). Aerosols (see formulation below) were generated using a disposable medical nebulizer that provided an aerosol with a mass median aerodynamic diameter of 3.2 ⁇ m as determined with an Andersen cascade impactor. The nebulizer was connected to a dosimeter system consisting of a solenoid valve and a source of compressed air (20 psi). The output of the nebulizer was directed into a plastic T-piece, one end of which was connected to the inspiratory port of a piston respirator. The solenoid valve was activated for 1 second at the beginning of the inspiratory cycle of the respirator. Aerosols were delivered at V ⁇ of 500 mL and a rate of 20 breaths/minute. A 0.5% sodium bicarbonate solution only was used as a control.
  • a solution of the candidate compound in 0.5% sodium bicarbonate/saline (w/v) at a concentration of 30.0 mg/mL is prepared using the following procedure:
  • the compound was administered by gavage at five dose levels, 0 (vehicle control), 10, 30, 100, 300 and 1000 mg/kg (mpk), with five mice in each dose level.
  • the dose volume for all levels was 10 mL/kg.
  • Dose solutions or suspensions were prepared in 2%> Tween 80 in 0.5%) carboxymethyl cellulose (CMC) and new dose solutions or suspensions were prepared every two - three days.
  • EDTA blood samples were collected by cardiac puncture for clinical pathology (hematology and clinical chemistry) and drug levels.
  • the EDTA blood samples were analyzed for total white blood cell count, red blood cell count, hemoglobin, hematocrit, erythrocyte indices (MCN, MCH, MCHC), platelets and a WBC five part differential (neutrophil, lymphocytes, monocytes, eosinophils and basophils).
  • Heparinized plasma samples were analyzed for alanine transaminase, aspartate transaminase, alkaline phosphatase, total bilirubin, albumin, protein, calcium, glucose, urea nitrogen, creatinine, cholesterol and triglycerides.
  • Adjuvant-Induced Arthritis in Rats Adjuvant-Induced Arthritis in Rats
  • AIA Adjuvant induced arthritis
  • RA rheumatoid arthritis
  • RA animal model of RA
  • Using an animal model of RA such as AIA, enables one to study the cellular events involved in the early stages of the disease.
  • CD44 expression on macrophages and lymphocytes is up-regulated during the early development of adjuvant arthritis, whereas LFA-1 expression is up-regulated later in the development of the disease. Understanding the interactions between adhesion molecules and endothelium at the earliest stages of adjuvant arthritis could lead to significant advances in the methods used m the treatment of RA.

Abstract

Disclosed are compounds which bind α4 integrins, preferably VLA-4. Certain of these compounds also inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated by α4 integrins, preferably VLA-4. Such compounds are useful in the treatment of inflammatory diseases in a mammalian patient, e.g., human, such as asthma, Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes, inflammatory bowel disease, rheumatoid arthritis, tissue transplantation, tumor metastasis and myocardial ischemia. The compounds can also be administered for the treatment of inflammatory brain diseases such as multiple sclerosis.

Description

HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY α4 INTEGRINS
BACKGROUND OF THE INVENTION
Field of the Invention
[0001] This invention relates to compounds wliich inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated by α 4 integrins where the α 4 integrin is preferably VLA-4.
References
[0002] The following publications, patents and patent applications are cited in this application as superscript numbers:
[0003] 1 Hemler and Takada, European Patent Application Publication
No. 330,506, published August 30, 1989
[0004] 2 Elices, et al., Cell, 6-0:577-584 (1990)
[0005] 3 Springer, Nature, 246:425-434 (1990)
[0006] Osborne, Cell, 62:3-6 (1990)
[0007] 5 Vedder, et al, Surgey, I :509 (1989)
[0008] 6 Pretolani, et al., J Exp. Med., 1S-Ω:795 (1994)
[0009] 7 Abraham, et al, J Clin. Invest, 9-1:776 (1994)
[0010] 8 Mulligan, et al., J. Immunology, 15ϋ:2407 (1993)
[0011] 9 Cybulsky, et al., Science, 25-1:788 (1991) [0012] 10 Li, et al., Arterioscler. Thromb. , 11: 197 (1993)
[0013] n Sasseville, et al., Am. J. Path., 1AA:21 (1994)
[0014] 12 Yang, et al., Proc. Nat. Acad. Science (USA), 9-11:10494 (1993)
[0015] 13 Burkly, et al., Diabetes, 42:529 (1994)
[0016] 14 Baron, et al., J Clin. Invest., 22:1700 (1994)
[0017] 15 Hamann, et al., J Immunology, 15-2:3283 (1994)
[0018] 16 Yednock, et al, Nature, 3-56:63 (1992) [0019] 17 Baron, et al., J. Exp. Med., 172:57 (1993)
[0020] 18 van Dinther-Janssen, et al, J Immunology, 142:4207 (1991)
[0021] !9 van Dinther-Janssen, et al., Annals. Rheumatic Dis., 52:672 (1993)
[0022] 20 Elices, et al, J. Clin. Invest. , -21:405 (1994)
[0023] 21 Postigo, et al., J. Clin. Invest, &9_: 1445 (1991)
[0024] 22 Paul, et al., Transpl. Proceed., 25:813 (1993)
[0025] 23 Okarhara, et al., Can. Res., 54:3233 (1994)
[0026] 24 Paavonen, et al, Int. J. Can., 5£:298 (1994)
[0027] 25 Schadendorf, et al., J. Path. , 120:429 (1993)
[0028] 26 Bao, et al., Diff, 52:239 (1993)
[0029] 27 Lauri, et al, British J. Cancer, 6-8-: 862 (1993)
[0030] 28 Kawaguchi, et al., Japanese J. Cancer Res. , S : 1304 (1992)
[0031] 29 Konradi, et al., PCT/USOO/01686, filed January 21, 2000.
[0032] All of the above publications are herein incorporated by reference in their entirety to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference in its entirety.
State of the Art
[0033] VLA-4 (also referred to as α4βj integrin and CD49d/CD29), first identified by Hemler and Takada,1 is a member of the βl integrin family of cell surface receptors, each of which comprises two subunits, an α chain and a β chain. VLA-4 contains an α4 chain and a βl chain. There are at least nine βl integrins, all sharing the same βl chain and each having a distinct α chain.
These nine receptors all bind a different complement of the various cell matrix molecules, such as fibronectin, laminin, and collagen. VLA-4, for example, binds to fibronectin. VLA-4 also binds non-matrix molecules that are expressed by endothehal and other cells. These non-matrix molecules include VC AM- 1 , which is expressed on cytokine-activated human umbilical vein endothehal cells in culture. Distinct epitopes of VLA-4 are responsible for the fibronectin and VCAM-1 binding activities and each activity has been shown to be inhibited independently.2
[0034] Intercellular adhesion mediated by VLA-4 and other cell surface receptors is associated with a number of inflammatory responses. At the site of an injury or other inflammatory stimulus, activated vascular endothehal cells express molecules that are adhesive for leukocytes. The mechanics of leukocyte adhesion to endothehal cells involves, in part, the recognition and binding of cell surface receptors on leukocytes to the corresponding cell surface molecules on endothehal cells. Once bound, the leukocytes migrate across the blood vessel wall to enter the injured site and release chemical mediators to combat infection. For reviews of adhesion receptors of the immune system, see, for example, Springer3 and Osborn.4 [0035] Inflammatory brain disorders, such as experimental autoimmune encephalomyelitis (EAE), multiple sclerosis (MS) and meningitis, are examples of central nervous system disorders in which the endothelium leukocyte adhesion mechanism results in destruction to otherwise healthy brain tissue. Large numbers of leukocytes migrate across the blood brain barrier (BBB) in subjects with these inflammatory diseases. The leukocytes release toxic mediators that cause extensive tissue damage resulting in impaired nerve conduction and paralysis.
[0036] In other organ systems, tissue damage also occurs via an adhesion mechanism resulting in migration or activation of leukocytes. For example, it has been shown that the initial insult following myocardial ischemia to heart tissue can be further complicated by leukocyte entry to the injured tissue causing still further insult (Vedder et al.).5 Other inflammatory or medical conditions mediated by an adhesion mechanism include, by way of example, asthma6"8, Alzheimer's disease, atherosclerosis,9"10 AIDS dementia,11 diabetes12" 14 (including acute juvenile onset diabetes), inflammatory bowel disease15 (including ulcerative colitis and Crohn's disease), multiple sclerosis,16"17 rheumatoid arthritis,18"21 tissue transplantation,22 tumor metastasis,23"28 meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte- mediated lung injury such as that which occurs in adult respiratory distress syndrome.
[0037] Substituted aminopyrimidines, as a class, have been disclosed as inhibiting binding of VLA-4 to VCAM-1 and, accordingly, exhibit anti- inflammatory properties.29 While these compounds possess antagonist properties to such binding, enhanced bioavailability of these compounds would augment their efficacy. SUMMARY OF THE INVENTION
[0038] This invention is directed to the discovery that certain N-[2-N',N'- diethylamino-5-aminosulfonylphenylpyrimidin-4-yl]-p-carbomyloxy- phenylala ine compounds possess unexpectedly superior bioavailabihty, as measured by their AUC, as compared to other substituted aminopyrimidine compounds previously disclosed.
[0039] In one of its composition aspects, this invention is directed to a compound of Formula (I) :
Figure imgf000006_0001
wherein each X is independently fluoro, chloro or bromo; p is an integer from 0 to 3;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, pyrrolyl, 2,5-dihydopyrrol-l-yl, piperidinyl, or 1,2,3,6-tetrahydroρyridin-l-yl;
R2 is selected from the group consisting of lower alkyl, lower alkenyl, and lower alkylenecycloalkyl; and pharmaceutically acceptable salts thereof. [0040] In a preferred embodiment, R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group.
[0041] In a preferred embodiment, this invention provides compounds of Formula (II):
Figure imgf000007_0001
wherein each X is independently selected from the group consisting of fluoro and chloro; m is an integer equal to 1 or 2;
R2 is selected from the group consisting of lower alkyl, lower alkenyl, and lower alkylenecycloalkyl; R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
[0042] In a particularly preferred embodiment, this invention provides compounds of Formula (III)
Figure imgf000008_0001
wherein each X is independently fluoro or chloro; n is zero or one;
R >2 is -CH2-R' where R' is selected from the group consisting of hydrogen, methyl or -CH=CH2;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
[0043] In another of its composition aspects, this invention is directed to a compound of Foπnula (IN):
Figure imgf000008_0002
(IN) wherein each X is independently fluoro, chloro or bromo; p is an integer from 0 to 3; R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, pyrrolyl, 2,5-dihydopyrrol-l-yl, piperidinyl, or 1,2,3,6-tetrahydropyridin-l-yl;
R2 is lower alkynyl; and pharmaceutically acceptable salts thereof.
[0044] In a preferred embodiment, R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group and R2 is propargyl.
[0045] In a preferred embodiment, this invention provides compounds of Formula (V):
Figure imgf000009_0001
wherein each X is independently selected from the group consisting of fluoro and chloro; m is an integer equal to 1 or 2;
R2 is lower alkynyl;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof. [0046] In a particularly preferred embodiment, this invention provides compounds of Formula (VI)
Figure imgf000010_0001
wherein each X is independently fluoro or chloro; n is zero or one; R2 is lower alkynyl;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
[0047] N- [2-N ' ,N ' -diethylamino-5 -aminosulfonylphenylpyrimidin-4-yl] -p- carbomyloxy-phenylalanine compounds within the scope of this invention include those set forth in Table I as follows:
Table T
/ N
Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000012_0001
[0048] Specific compounds within the scope of this invention include the following compounds. As used below, these compounds are named based on phenylalanine derivatives but, alternatively, these compounds could have been named based on N-[2-N',N'-diethylamino-5-aminosulfoiiylphenyl-pyrimidin- 4-yl]- -carbomyloxyphenylalanine derivatives or 2-{2-diethylamino-5- [(benzenesulfonyl)methylamino]-ρyrimidin-4-ylamino } -p -carbamoyloxy- phenyl)propionic acid derivatives.
[0049] N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-phenylalanine;
[0050] N-(2-[N,,N'-diethylamino]-5-[N"-(4-fluorophenylsulfbnyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-phenylalanine;
[0051] N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-NM- methylamino]pyrimidin-4-yl)-4'-(pyrrolidin - 1 -ylcarbonyloxy)-L- phenylalanine;
[0052] N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(pyrrolidin- 1 -ylcarbonyloxy)-L- phenylalanine; [0053] N-(2-rN,,N,-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(piperidin-l-ylcarbonyloxy)-L-phenylalanine;
[0054] N-(2-[N',N'-diethylammo]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(piperidin- 1 -ylcarbonyloxy)-L-phenylalanine;
[0055] N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4l-(azetidin-l-ylcarbonyloxy)-L-phenylalanine;
[0056] N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylc-trbonyloxy)-L-phenylalarιine;
[0057] N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- memylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenylalanine;
[0058] N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenylalanine;
[0059] N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4,-(pyrrolidin-l-ylcarbonyloxy)-L- phenylalanine;
[0060] N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-phenylalanine;
[0061] N-(2-[N',N'-diethyl--mino]-5-[N"-(2,4-difluorophenylsulfbnyl)-N"- methylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenylalanine;
[0062] N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- ethyl--mino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L-phenylalanine; [0063] N-(2-[N',N,-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(pyrrolidin- 1 -ylcarbonyloxy)-L- phenylalanine;
[0064] N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(pyrrolidin- 1 -ylcarbonyloxy)-L- phenylalanine;
[0065] N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L- phenylalanine;
[0066] N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L- phenylalanine;
[0067] N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(pyrrolidin- 1 -ylcarbonyloxy)-L- phenylalanine; and pharmaceutically acceptable salts thereof.
[0068] In another aspect, this invention provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the compounds defined herein.
[0069] In one of its method aspects, this invention is directed to a method for treating a disease mediated at least in part by α 4 integrins, preferably VLA-4, in a patient, which method comprises administering a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of this invention. [0070] The compounds and pharmaceutical compositions of this invention are useful for treating disease conditions mediated at least in part by α4 integrins, preferably VLA-4, or leucocyte adhesion. Such disease conditions include, by way of example, asthma, Alzheimer's disease, atherosclerosis, ALDS dementia, diabetes (including acute juvenile onset diabetes), inflammatory bowel disease
(including ulcerative colitis and Crohn's disease), multiple sclerosis, rheumatoid arthritis, tissue transplantation, tumor metastasis, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs in adult respiratory distress syndrome.
[0071] Other disease conditions include, but are not limited to, inflammatory conditions such as erythema nodosum, allergic conjunctivitis, optic neuritis, uveitis, allergic rhinitis, Ankylosing spondylitis, psoriatic arthritis, vasculitis, Reiter's syndrome, systemic lupus erythematosus, progressive systemic sclerosis, polymyositis, dermatomyositis, Wegner's granulomatosis, aortitis, sarcoidosis, lymphocytopenia, temporal arteritis, pericarditis, myocarditis, congestive heart failure, polyarteritis nodosa, hypersensitivity syndromes, allergy, hypereosinophilic syndromes, Churg-Strauss syndrome, chronic obstructive pulmonary disease, hypersensitivity pneumonitis, chronic active hepatitis, interstitial cystitis, autoimmune endocrine failure, primary biliary cirrhosis, autoimmune aplastic anemia, chronic persistent hepatitis and thyroiditis.
[0072] In a preferred embodiment, the disease condition is an inflammatory disease. DETAILED DESCRIPTION OF THE INVENTION
[0073] As above, this invention relates to compounds which inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated at least in part by α4 integrins, preferably VLA-4. However, prior to describing this invention in further detail, the following terms will first be defined.
Definitions
[0074] Unless otherwise stated, the following terms used in the specification and claims have the meanings given below:
[0075] As used herein, "lower alkyl" refers to monovalent alkyl groups having from 1 to 5 carbon atoms including straight and branched chain alkyl groups. This term is exemplified by groups such as methyl, ethyl, wo-propyl, n-propyl, ra-butyl, iso-hutyl, -.ec-butyl, t-butyl, τ.-pentyl and the like.
[0076] The term "lower alkylene" refers to divalent alkylene groups of from 1 to 4 carbon atoms including straight and branched chain alkylene groups. This term is exemplified by groups such as methylene, ethylene, /.-propylene, iso- propylene (-CH2CH(CH3)- and -CH(CH3)CH2-) and the like.
[0077] The term "lower alkenyl" refers to an alkenyl group preferably having from 2 to 6 carbon atoms and having at least 1 site and preferably only 1 site of alkenyl unsaturation (i.e., >C=C<). This term is exemplified by groups such as allyl, ethenyl, propenyl, butenyl, and the like.
[0078] The term "lower alkynyl" refers to an alkynyl group preferably having from 2 to 6 carbon atoms and having at least 1 site and preferably only 1 site of alkynyl unsaturation (i.e., -C≡C-). This term is exemplified by groups such as acetyl (-C≡CH), propargyl (-CH2-C≡CH), 3-butynyl
(-CH2CH2C≡CH3) and the like. [0079] The term "lower cycloalkyl" refers to cyclic alkyl groups of from 3 to 6 carbon atoms having a single cyclic ring including, by way of example, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
[0080] The term "lower alkylenecycloalkyl" refers to the group consisting of a lower alkylene-lower cycloalkyl, as defined herein. Such groups are exemplified by methylenecyclopropyl (-CH2-cyclopropyl), ethylenecyclopropyl and the like.
[0081] "Pharmaceutically acceptable carrier" means a carrier that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier that is acceptable for veterinary use as well as human pharmaceutical use. "A pharmaceutically acceptable carrier" as used in the specification and claims includes both one and more than one such carrier.
[0082] "Treating" or "treatment" of a disease includes:
(1) preventing the disease, i.e., causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease,
(2) inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms, or
(3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
[0083] A "therapeutically effective amount" means the amount of a compound that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically effective amount" will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated. [0084] "Pharmaceutically acceptable salt" refers to pharmaceutically acceptable salts of a compound of Formula I which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
[0085] Integrins are a large family of homologous transmembrane linker proteins that are the principal receptors on animal cells for binding most extracellular matrix proteins, such as collagen, fibronectin, and laminin. The integrins are heterodimers comprised of an α chain and a β chain. To date, twenty different integrin heterodimers, made from 9 different α subunits and 14 different β subunits, have been identified. The term " α4 integrins" refers to the class of heterodimer, enzyme-linked cell-surface receptors that contain the α 4 subunit paired with any of the β subunits. VLA-4 is an example of an α4 integrin, and is a heterodimer of the α4 and βj subunits, and is also referred to as α4 β! integrin.
Compound Preparation
[0086] The compounds of this invention can be prepared from readily available starting materials using the methods and procedures set forth in the examples below. These methods and procedures outline specific reaction protocols for preparing N-[2-N',N'-diethylamino-5-aminosulfonylphenyl- yrimidin-4-yl]-j-)-carbomyloxy-phenylalanme compounds. Compounds within the scope not exemplified in these examples and methods are readily prepared by appropriate substitution of starting materials which are either commercially available or well known in the art. [0087] Other procedures and reaction conditions for preparing the compounds of this invention are described in the examples set forth below. Additionally, other procedures for preparing compounds useful in certain aspects of this invention are disclosed in U.S. Patent 6,492,372, issued December 10, 2002; the disclosure of which is incorporated herein by reference in its entirety.
Pharmaceutical Formulations
[0088] When employed as pharmaceuticals, the compounds of this invention are usually administered in the form of pharmaceutical compositions. These compositions can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. These compositions are effective by both injectable and oral delivery. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
[0089] This invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of Formula I above associated with pharmaceutically acceptable carriers. In making the compositions of this invention, the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container. The excipient employed is typically an excipient suitable for administration to human subjects or other mammals. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders. [0090] In preparing a formulation, it may be necessary to mill the active compound to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
[0091] Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, macrocrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy- benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
[0092] The compositions are preferably formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
[0093] The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It, will be understood, however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[0094] For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.
[0095] The tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
[0096] The liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles. [0097] 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. Preferably the 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 breathed directly from the nebulizing device or the nebulizing device may be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
[0098] The following formulation examples illustrate the pharmaceutical compositions of the present invention.
Formulation Example 1
[0099] Hard gelatin capsules containing the following ingredients are prepared:
Quantity Ingredient (mg/capsule)
Active Ingredient 30.0 Starch 305.0
Magnesium stearate 5.0
[00100] The above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities. Formulation Example 2
[00101] A tablet formula is prepared using the ingredients below:
Quantity Ingredient (mg/tablet)
Active Ingredient 25.0
Cellulose, microcrystalline 200.0
Colloidal silicon dioxide 10.0
Stearic acid 5.0
The components are blended and compressed to form tablets, each weighing 240 mg.
Formulation Example 3 [00102] A dry powder inhaler formulation is prepared containing the following components:
Ingredient Weight % Active Ingredient 5
Lactose 95
The active mixture is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
Formulation. Example 4 [00103] Tablets, each containing 30 mg of active ingredient, are prepared as follows:
Quantity
Ingredient (mg/tablet)
Active Ingredient 30.0 mg
Starch 45.0 mg
Microcrystalline cellulose 35.0 mg
Polyvinylpyrrolidone
(as 10% solution in water) 4.0 mg
Sodium carboxymethyl starch 4.5 mg
Magnesium stearate 0.5 mg
Talc 1.0 mg
Total 120 mg
[00104] The active ingredient, starch and cellulose are passed through a No. 20 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with the resultant powders, which are then passed through a 16 mesh U.S. sieve. The granules so produced are dried at 50° to 60 °C and passed through a 16 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 30 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 120 mg.
Formulation Example 5 [00105] Capsules, each containing 40 mg of medicament are made as follows:
Quantity
Ingredient (mg/capsule) Active Ingredient 40.0 mg
Starch 109.0 mg
Magnesium stearate 1.0 mg
Total 150.0 mg [00106] The active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg quantities.
Formulation Example 6
[00107] Suppositories, each containing 25 mg of active ingredient are made as follows:
Ingredient Amount
Active Ingredient 25 mg
Saturated fatty acid glycerides 2,000 mg
[00108] The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed to cool.
Formulation Example 7 [00109] Suspensions, each containing 50 mg of medicament per 5.0 ml dose are made as follows:
Ingredient Amount
Active Ingredient 50.0 mg
Xanthan gum 4.0 mg
Sodium carboxymethyl cellulose (11%)
Microcrystalline cellulose (89%) 50.0 mg
Sucrose 1.75 g
Sodium benzoate 10.0 mg
Flavor and Color q.v.
Purified water to 5.0 ml
[00110] The medicament, sucrose and xanthan gum are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water. The sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
Formulation Example 8
Quantity Ingredient (mg/capsule)
Active Ingredient 15.0 mg
Starch 407.0 mg
Magnesium stearate 3.0 mg Total 425.0 mg
[00111] The active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 425 mg quantities.
Formulation Example 9 [00112] An intravenous formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 250.0 mg
Isotonic saline 1000 ml
Formulation Example 10 [00113] A topical formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 1 - 10 g Emulsifying Wax 30 g
Liquid Paraffin 20 g
White Soft Paraffin to 100 g [00114] The white soft paraffin is heated until molten. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The active ingredient is added and stirring is continued until dispersed. The mixture is then cooled until solid.
[00115] Another preferred formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent 5,023,252, issued June 11, 1991, herein incorporated by reference. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
[00116] Direct or indirect placement techniques may be used when it is desirable or necessary to introduce the pharmaceutical composition to the brain. Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier. One such implantable delivery system used for the transport of biological factors to specific anatomical regions of the body is described in U.S. Patent 5,011,472 which is herein incorporated by reference.
[00117] Indirect techniques, which are generally preferred, usually involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs. Latentiation is generally achieved through blocking of the hydroxy, carbonyl, sulfate, and primary amine groups present on the drug to render the drug more lipid soluble and amenable to transportation across the blood-brain barrier. Alternatively, the delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blood-brain barrier. Utility
[00118] The compounds of this invention inhibit, in vivo, adhesion of leukocytes to endothehal cells mediated at least in part by α 4 integrins, preferably VLA-4, by competitive binding to α 4 integrins, preferably VLA-4.
Accordingly, the compounds of this invention can be used in the treatment of mammalian diseases mediated at least in part by α4 integrins, preferably VLA- 4, or leucocyte adhesion. Such diseases include inflammatory diseases in mammalian patients such as asthma, Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes (including acute juvenile onset diabetes), inflammatory bowel disease (including ulcerative colitis and Crohn's disease), multiple sclerosis, rheumatoid arthritis, tissue transplantation, tumor metastasis, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs in adult respiratory distress syndrome.
[00119] The amount administered to the mammalian patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions are administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as "therapeutically effective dose." Amounts effective for this use will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the inflammation, the age, weight and general condition of the patient, and the like.
[00120] The compositions administered to a patient are in the form of pharmaceutical compositions described above. These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to
8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
[00121] The therapeutic dosage of the compounds of the present invention will vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. For example, for intravenous administration, the dose will typically be in the range of about 20 μg to about 500 μg per kilogram body weight, preferably about 100 g to about 300 μg per kilogram body weight. Suitable dosage ranges for intranasal administration are generally about 0.1 pg to 1 mg per kilogram body weight. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
[00122] The following synthetic and biological examples are offered to illustrate this invention and are not to be construed in any way as limiting the scope of this invention. Unless otherwise stated, all temperatures are in degrees Celsius.
EXAMPLES
[00123] In the examples below, the following abbreviations have the following meanings. If an abbreviation is not defined, it has its generally accepted meaning.
AUC = Area under the curve bd = broad doublet bs = broad singlet BSA bovine serum albumin d doublet
DMAP 4-N,N-dimethylaminopyridine ethylcarbodiimide hydrochloride
EDTA Ethylenediamine tetraacetic acid
EtOAc ethyl acetate
EtOH ethanol eq. equivalent
FACS Fluorescence activated Cell
10 Sorter
FITC Fluorescein isothiocyanate
S grams i.p. intraperitoneal h hour
15 HBSS Hank's Balanced Saline Solution
Hct hematocrit, or measurement of packed red blood cells obtained by centrifugation in a volume of a blood sample
20 HB or Hb hemoglobin HEPES 4-(2-hydroxyethyl)- 1 -piperazine- ethanesulfonic acid
IgG Fc a binding domain of the immunoglobulin
25 kg killogram
L liter m multiplet (when used with ΝMR data)
M Molar
30 MCH Mean Corpusular Hemoglobin;
Hb/RBC
MCHC mean corpuscular hemoglobin count expressed as a percentage;
35 Hb/Hct.
MCV mean corpuscular volume; the avg. volume of erythrocytes, conventionally expressed in cubic
40 micrometers per red cell.
MeOH methanol mg milligram mL milliliter mm millimeter
45 mM millimolar mol moles mmol millimol mpk milligrams per killogram
N nonnal ng nanograms
PBS++ Phosphate buffered saline psi pounds per square inch q.s. or Q.S. bring to volume
Rfs or Rf retention factor rpm rotations per minute rt or RT room temperature s singlet t triplet
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC or tic thin layer chromatography μL microliter μg microgram μm microns
Vt Total volume WBC White Blood Cells w/v weight to volume
[00124] Compounds of the present invention may be prepared as illustrated in Scheme 1 and as described in the methods below:
Figure imgf000032_0001
Example 1
Preparation of N-(2-[N'.N'-diethy1amino]-5-rN"-(4-ch1orophenylsu1fonyl)-N''- etby1amino]pyrimidin-4-y1)-4'-(pyrro1idin-l-ylcarbony1oxy)-L-pheny1alanine
[00125] Step 1: Preparation of 2,4-Dichloro-5-nitropyrimidine (2). 5-
Nitrouracil, (1), was treated with phosphorous oxychloride (POCl3) and N,N- dimethylaniline (PhNMe2), according to the procedure of Whittaker (J. Chem. Soc. 1951, 1565), to give compound 2. Compound 2 is also available from City Chemical (West Haven, CT).
[00126] Step 2: Preparation of N-(2-[N,,N'-diethylamino]-5- nitropyrimidin-4-yl)-L-tyrosine tert-butyl ester (3). To a solution of L- tyrosine tert-butyl ester (H-Tyr(OH)-OtBu) (30.6 g, 0.129 mol) in THF (250 mL) at -10 °C was added 2,4-dichloro-5-nitropyrimidine (25g, 0.129 mol), keeping the temperature below 5 °C during the addition. Once the addition was complete, N,N-diisoρroρylethylamine (EtiPr2N) (33.7 mL, 0.194 mol) was added dropwise. After stirring for 1 h at -10 °C, diethylamine (Et2NH) (66.73 mL, 0.645 mol) was added slowly, and then the reaction mixture was warmed to room temperature overnight. The reaction mixture was diluted with diethyl ether (500 mL), and the organic layer was washed with 0.2 N citric acid (3 x
150 mL), water (1 x 150 mL), and 10% K2CO3 (3 x 150 mL). The organic phase was dried (Na^SO^, filtered, and concentrated in vacuo to yield a yellow residue. The residue was purified by flash chromatography (20% EtOAc/hexanes on silica gel) to yield 37.39 g (67%) of compound 3 as a yellow foam. R O.21 (25% EtOAc/hexanes on silica gel).
[00127] Step 3: Preparation of N-(2-[N',N,-diethylamino]-5- nitropyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-phenyIaIanine tert-butyl ester (4). To a solution of N-(2-[N',N'-diethylamino]-5- nitropyrimidin-4-yl)-L-tyrosine tert-butyl ester (37.39 g, 0.087 mol) in CH2C12
(150 mL) was added DMAP (10.59 g, 0.087 mol). After 5 minutes triethylamine (TEA) (18.19 mL, 0.131 mol) was added dropwise. 1-Pyrrolidinecarbamoyl chloride (14.42 mL, 0.131 mol) was added dropwise, and the reaction was heated to reflux (40 °C) overnight. The reaction mixture was concentrated in vacuo and taken up in EtOAc (300 mL). The organic phase was washed with 0.2 N citric acid (3 x 150 mL), water (1 x 150 mL), sat. NaHCO3 (3 x 150 mL), brine (1 x 150 mL), dried (N-^SO^, filtered, and concentrated in vacuo to yield 43.07 g (94%) of compound 4 as a yellow solid. Rf= 0.5 (50% EtOAc/hexanes on silica gel).
[00128] Step 4: Preparation of N-(2-[N',N'-diethylamino]-5- aminopyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-p-ιenylalanine tert-butyl ester (5). A mixture of N-(2-[N',N'-diethylamino]-5- nitropyrimidin-4-yl)-4'-(pyrrolidin- 1 -ylcarbonyloxy)-L-phenylalanine tert- butyl ester (43.07 g, 0.081 mol) and 10% Pd/C (4.3 g, 10 wt% Pd) in EtOH (200 mL) was shaken under 45 psi hydrogen until TLC (50% EtOAc/hexanes on silica gel) showed 100% conversion to product (48 hours). The reaction mixture was then filtered through a Celite plug and concentrated in vacuo to yield 40.29 g (100%) of compound 5 as a purple foam. Rf= 0.11 (6:1 EtOAc/hexanes on silica gel).
[00129] Step 5: Preparation of N-(2-[N',N'-diethylamino]-5-[NM-(4- chlorophenyl-sulf onyl) amino] pyrimidin-4-yI)-4 '-(py r rolidin-l - ylcarbonyloxy) -L-phenylalanine tert-butyl ester (6). A pyridine (160 mL) solution of N-(2-[N',N'-diethylamino]-5-aminopyrimidin-4-yl)-4'-(pynolidin- l-ylcarbonyloxy)-L-phenylalanine tert-butyl ester (40.29 g, 0.081 mol) was cooled to -20 °C with a dry ice/CH3CN bath. The mixture stireed for 30 minutes, and then 4-chlorobenzenesulfonyl chloride (17.06 g, 0.081 mol) was added slowly. The reaction was stirred at -20 °C to -15°C for 4 h and then allowed to warm to room temperature overnight. The reaction was diluted with EtOAc (400 mL), and the organic phase was washed with 0.2 N citric acid (3 x 150 mL), water (1 x 150 mL), sat. NaHCO3 (3 x 150 mL), brine (1 x 150 mL), dried (N-t-SO.,,), filtered, and concentrated in vacuo to yield a brown residue. The residue was purified by flash chromatography (50% EtOAc/hexanes on silica gel) to yield 43.49 g (80%) of compound 6 as a yellow foam. R 0.35 (50% EtOAc/hexanes on silica gel).
[00130] Step 6: Preparation of N-(2-[N',N'-diethyIamino]-5-[N"-(4- chlorophenyl-sulfonyl)-NM-ethylammo]pyrimidin-4-yl)-4'-(pyrrolidin-l- ylcarbonyloxy)-L-phenyIaIanine tert-butyl ester (7). To a solution of N-(2- [N',N'-diethyl-ιmino]-5-|^N"-(4-chlorophenyl-sulfonyl)amino]pyrimidin-4-yl)- 4'-(pyrrolidin-l -ylcarbonyloxy) -L-phenylalanine tert-butyl ester (42.92 g,
0.064 mol) in acetone (Me2CO) (600 mL) was added K2CO3 (12.75 g, 0.096 mol), and the mixture was stirred for 1 h at room temperature. Iodoethane (Etl) (7.73 mL, 0.096 mol) was then added slowly, and the reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated in vacuo, and the residue was taken up in EtOAc (300 mL). The organic phase was washed with water (2 x 300 mL), brine (1 x 100 mL), dried (NajSO^), filtered, and concentrated in vacuo. The residue was purified by flash chromatography (2:1 hexanes/EtOAc on silica gel) to yield 37.36 g (85%) of compound 7 as a white solid. Rf= 0.53 (50% EtOAc/hexanes on silica gel).
[00131] Step 7: Preparation of N-(2-[N',N'-diethylamino]-5-[N"-(4- chlorophenylsulfonyl)-N"-ethylamino]pyrimidin-4-yl)-4,-(pyrrolidin-l- ylcarbonyloxy)-L-phenylalanine hydrochloride (8). A formic acid (500 mL) solution of N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenyl-sulfonyl)-
N"-ethylamino] pyrimidin-4-yl)-4'-(pynolidin- l-ylcarbonyloxy)-L- phenylalanine tert-butyl ester (36.21 g, 0.052 mol) was heated to 70 °C for 2 h and then concentrated in vacuo. The residue was dissolved again in formic acid (500 mL) and heated again at 70 °C for 2 h. The solution was reduced in volume by 80% and then treated with at least 1 eq. of 1.0 N HC1 (52 mL,
0.052 mol) followed by distilled water (100 mL). The resulting heterogeneous mixture was concentrated in vacuo. Distilled water (100 mL) was added, and the heterogeneous mixture was concentrated in vacuo. The latter steps were repeated twice to yield a wet white product. This was dried by placing under high vacuum at 40 °C (7 days) to yield 32.8 g (93%) of compound 8, as a free- flowing white solid. R = 0.25 (7/3 MeOH/H2O + 0.1% TFA, reverse phase).
'H NMR (CD3OD) δ 8.22 (bs, 1H), 7.82-7.79 (m, 1H), 7.64-7.60 (m, 2H), 7.36-7.33 (m, 1H), 7.22-7.13 (m, 2H), 7.07-6.98 (m, 2H), 4.91-4.90 (m, 1H), 4.80-4.79 (m, 1H), 4.12-4.10 (m, 1H), 3.87-3.75 (m, 1H), 3.55-3.53 (m, 4H), 3.41-3.40 (m, 3H), 3.26-3.19 (m, 2H), 2.03 (bs, 1H), 1.97-1.89 (m, 3H), 1.27- 1.15 (m, 6H), 1.10-1.05 (t, 1.5H), 0.97-0.92 (t, 1.5H)
13C NMR (CD3OD) δ 175.8, 175.7, 166.5, 162.7, 162.2, 155.8, 155.7, 155.7, 152.6, 148.1, 147.7, 142.0, 138.5, 136.2, 132.6, 132.3, 131.9, 131.7, 123.7, 111.8, 111.5, 62.3, 57.8, 44.9, 38.7, 38.0, 27.4, 26.6, 15.3, 14.9, 14.7, 14.0, 13.9
Example 2
Preparation ofN-(2-[N'IN'-diethy1a.mino]-5-[N"-(4-fluoropheny1su1fonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-j)yrrolidin-l-ylcarbonyloxy)-L-pheny1a1anine
[00132] Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 1. Step 5 was performed using 4-fluorobenzenesulfonyl chloride in place of 4- chlorobenzenesulfonyl chloride. Η NMR (CD3OD) δ 8.17 (bs, 1H), 7.90-7.87 (m, 2H), 7.40-7.34 (m, 2H),
7.20-7.16 (m, 1H), 7.08-7.00 (m, 3H), 5.52-5.51 (m, 1H), 4.96-4.93 (m, 2H), 5.78-5.70 (m, 1H), 3.85-3.75 (m, 1H), 3.59-3.53 (m, 4H), 4.47-4.43 (m, 2H), 3.44-3.24 (m, 2H), 2.02-1.94 (m, 3H), 1.24-1.16 (m, 6H), 1.10-1.05 (t, 1.5H), 0.99-0.94 (t, 1.5H)
13C NMR (CD3OD) δ 133.0, 132.9, 132.5, 132.2, 123.7, 123.6, 118.6, 57.1, 44.3, 38.3, 27.3, 26.6, 14.7, 14.1
MS m/z 629.5 (MH+) Example 3
Preparation of N-(2-[N',N,-diethy1amino]-5-[N"-(4-fluorophenylsu1fonyl)-N"- methy1amino]pyrimidin-4-yl)-4'-(pynolidin -1 -ylcarbonyloxy)-L- ptienylalanine
[00133] Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 2. Step 6 was performed using dimethyl sulfate in place of ethyl iodide.
Η NMR (CD3OD) δ 8.16 (bs, IH), 7.89-7.88 (m, IH), 7.39-7.35 (m, 3H), 7.20-7.13 (m, IH), 7.05-7.00 (m, 2H), 4.85-4.84 (m, IH), 4.14-4.12 (m, IH), 3.59-3.54 (m, 5H), 3.45-3.44 (m, 2H), 3.45-3.33 (m, 3H), 3.13-3.12 (m, IH),
3.02-3.01 (m, IH), 2.04-1.95 (m, 4H), 1.29-1.18 (m, 6H)
13C NMR (CD3OD) δ 176.5, 169.8, 166.9, 166.4, 156.2, 152.7, 151.8, 150.4, 136.8, 133.3, 133.2, 132.5, 123.7, 118.8, 118.5, 57.8, 57.1, 48.3, 44.5, 41.0, 38.8, 27.5, 26.7, 14.1
MS m/z 615.2 (MH+)
Example 4 Preparation of N-(2-[N',N'-diethy1amino]-5-[N"-(4-chlorophenylsulfonyl)-N"- methylamino]pyrimidιn-4-y1)-4'-(pyrrolidin-l-ylcarbonyloxy)-L-phenylalanine
[00134] Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 1. Step 6 was performed using dimethyl sulfate in place of ethyl iodide. Η NMR (CD3OD) δ 8.20 (bs, IH), 7.83-7.80 (m, 2H), 7.67-7.64 (m, 2H),
7.37-7.34 (m, IH), 7.21-7.18 (m, IH), 7.10-7.03 (m, 2H), 4.88-4.87 (m, IH), 4.13-4.10 (m, IH), 3.55-3.45 (m, 6H), 3.42-3.40 (m, 2H), 3.24-3.23 (m, 2H), 3.11-3.10 (m, IH), 3.02-3.01 (m, IH), 2.04-2.03 (m, IH), 1.98-1.90 (m, 3H), 1.28-1.18 (m, 6H)
13C NMR (CD3OD) δ 176.0, 166.4, 161.8, 155.9, 155.4, 152.6, 146.5, 142.2, 137.6, 137.4, 136.4, 132.5, 131.9, 123.7, 114.6, 62.4, 58.1, 57.7, 45.0, 40.8, 38.6, 38.3, 27.4, 26.6, 15.3, 13.9 Example 5.
Preparation of N-(2-[ J'JN'-diethy1amino]-5-[N''-(4-fluorophenylsu1fony1)-N''- methylamino]pyrimidin-4-yl)-4'-(piperidin-l-ylcarbonylQxy)-L-phenylalanine
[00135] Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 3. Step 3 was perfonned using 1-piperidinecarbonyl chloride in place of 1- pyrrolidinecarbonyl chloride.
Η MR (CD3OD) δ 8.16 (bs, IH), 7.90-7.88 (m, 2H), 7.40-7.35 (m, 2H), 7.21-7.20 (m, IH), 7.14-7.13 (m, IH), 7.02-7.01 (m, 2H), 5.51 (bs, IH), 4.83- 4.77 (m, IH), 3.64-3.53 (m, 6H), 3.34-3.33 (m, 2H), 3.20-3.17 (m, IH), 3.12-
3.11 (m, 2H), 3.02-3.01 (m, IH), 1.68-1.65 (m, 6H), 1.19-1.17 (m, 6H)
13C NMR (CD3OD) δ 185.0, 169.7, 166.3, 152.7, 136.6, 135.0, 133.2, 133.0, 132.5, 131.8, 126.3, 123.6, 121.7, 118.6, 118.3, 57.6, 54.5, 46.9, 44.3, 39.6, 38.7, 27.6, 25.9, 14.0
Example 6
Preparation of N-(2-rN',N'-diethy1amino]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(piperidin-l-ylcarbonyloxy)-T .-phenylalanine
[00136] Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 2. Step 3 was performed using 1-piperidinecarbonyl chloride in place of 1- pyrrolidinecarbonyl chloride. 1H NMR (CD3OD) δ 8.17 (bs, IH), 7.91-7.85 (m, 2H), 7.39-7.31 (m, 3H),
7.20-7.16 (m, IH), 7.05-6.97 (m, 2H), 4.88-4.69 (m, 2H), 4.71-4.69 (m, IH), 3.80-3.75 (m, IH), 3.62-3.39 (m, 6H), 3.34-3.32 (m, 2H), 3.30-3.16 (m, 3H), 1.68-1.65 (m, 4H), 1.23-1.17 (m, 6H), 1.10-1.05 (t, 1.5H), 0.99-0.94 (t, 1.5H)
13C NMR (CD3OD) δ 199.9, 187.6, 183.1, 176.2, 169.7, 166.3, 163.0, 162.7,
153.9, 152.9, 136.5, 133.1, 133.0, 132.7, 132.4, 123.8, 118.8, 118.4, 111.1, 110.6, 102.8, 79.4, 57.3, 55.4, 44.4, 38.9, 38.4, 27.7, 26.1, 15.1, 14.8, 14.3, 14.2 Example 7
Preparation ofN-(2-[N'JN'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethyl a mo jpyrimi din-4-yl)-4'-(azeti din- 1 -yl carbony1oxy)-L-phenylal anine
[00137] Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 2. Step 3 was performed according to the following procedure.
Η MR (CD3OD) δ 7.92-7.86 (m, 2H), 7.41-7.32 (m, 3H), 7.22 (d, IH), 7.04- 6.91 (m, 3H), 4.29-3.98 (m, 4H), 3.88-3.72 (m, IH), 3.69-3.37 (m, 4H), 2.40- 2.24 (m, 2H), 1.28-1.11 (m, 6H), 1.10-1.00 (t, 1.5H), 1.01-0.89 (t, 1.5H)
13C NMR (CD3OD) δ 174.2, 169.7, 166.4, 163.2, 162.8, 157.0, 153.3, 153.2, 152.4, 144.3, 143.8, 136.1, 135.6, 135.5, 133.2, 133.1, 132.5, 132.2, 123.7, 118.9, 118.6, 112.9, 112.6, 57.5, 38.1, 37.7, 17.4, 14.7, 14.5, 13.8, 13.7 MS m/z 615 (MH+)
[00138] Alternative Preparation of N-(2-[N',N'-diethylamino]-5- nitropyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-plιenylalanine tert- butyl ester. To a-15°C stirred solution of compound 3 (24.9 g, 0.0578 mol) and 4-nitroρhenyl chloroforaiate (11.7 g, 0.0578 mmol) in CH2C12 (300 mL) was added triethylamine (24.2 mL, 0.173 mol), at a rate such that the temperature of the reaction mixture did not exceed -10°C. After stirring for 20 min, azetidine (3.30 g, 0.0578 mmol) was added dropwise, and the reaction mixtures was warmed to room temperature and stirred overnight. The reaction mixture was diluted with EtOAc (100 mL) and hexanes (100 mL), and then was extracted repeatedly with 10% aqueous K2CO3, until no yellow color (4- nitrophenol) was seen in the aqueous phase. The organic layer was washed with brine (75 mL), dried with MgSO4, filtered, and evaporated to yield 28.5 g (96%)) ofN-(2-[N',N'-diethylamino]-5-nitropyrimidin-4-yl)-4'-(azetidin-l- ylcarbonyloxy)-L-phenylalanine tert-butyl ester as a yellow solid, which was used without purification. Rf= 0.17 (2:5 EtOAc/hexanes on silica gel). Example 8
Preparation of N-(2-[N'TN'-diethy1amino]-5-[N''-(4-fluoropheny1su1fony1)-N"- methy1amino]pyrimιdin-4-y1)-4'-(azetidin-l-ylcarbonyloxy)-L-phenyla1anine
[00139] Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 7. Step 6 was performed using dimethyl sulfate in place of ethyl iodide.
'H NMR (CD3OD) δ 7.95-7.76 (m, 2H), 7.44-7.11 (m, 4H), 7.01-6.83 (m, 3H), 4.30-3.93 (m, 4H), 3.66-3.41 (m, 4H), 3.14-2.92 (m, 3H), 2.42-2.21 (m, 2H), 1.32-1.01 (m, 6H)
13C NMR (CD3OD) δ 152.3, 136.3, 133.4, 133.2, 132.4, 123.6, 118.8, 118.5, 38.2, 17.4, 13.8
MS m/z 601 (MH1")
Example 9
Preparation of N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenyla1anine
[00140] Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 8. Step 5 was performed using 4-chlorobenzenesulfonyl chloride in place of 4- fluorobenzenesulfonyl chloride.
1H NMR (CD3OD) δ 7.83 (d, 2H), 7.67 (d, 2H), 7.36-7.18 (m, 2H), 7.06-6.86 (m, 3H), 4.29-3.97 (m, 4H), 3.66-3.34 (m, 5H), 3.15-2.95 (m, 4H), 2.41-2.22 (m, 2H) 1.26-1.06 (m, 6H)
13C MR (CD3OD) δ 157.2, 153.0, 152.5, 142.9, 142.5, 136.4, 132.5, 132.1, 132.0, 123.8, 57.9, 52.2, 40.7, 38.0, 17.4, 13.6 MS m/z 617 (Mlf)
Example 10
Preparation of N-(2-[N',N'-diethy1amino]-5-[N"-(4-ch1orophenylsu1fony1)-N"- ethy1amino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenyla1anine
[00141] Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 7. Step 5 was performed using 4-chlorobenzenesulfonyl chloride in place of 4- fluorobenzenesulfonyl chloride.
Η NMR (CD3OD) δ 7.86-7.76 (m, 2H), 7.70-7.60 (m, 2H), 7.32 (bd, IH), 7.21 (bd, IH), 7.03-6.97 (m, 2H), 6.90 (bs, IH), 4.29-4.00 (m, 4H), 3.89-3.72 (m, IH), 3.70-3.36 (m, 5H), 3.28-3.10 (m, 2H), 2.42-2.24 (m, 2H), 1.28-1.13
(m, 6H), 1.11-1.02 (t, 1.5H), 1.01-0.90 (t, 1.5H)
MS m/z 631 (MH^>
Example 11
Preparation of N-(2-[N'.N'-diethy1amino]-5-[N"-(2;4-dιfluoropheny1su1fony1)- N"-methylamino]pyrimidin-4-yl)-4'-(pyrrolidin-1 -y1carbony1oxy)-L- phenylalanine
[00142] Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 3. Step 5 was performed using 2,4-difluorobenzenesulfonyl chloride in place of 4- fluorobenzenesulfonyl chloride.
1H NMR (CDC13) δ 1.1 (bs, 6H), 1.93 (bs, 4H), 2.50-3.75 (m, 13H), 4.83 (bs. IH), 6.60-7.40 (m, 7H), 7.60 (bs, IH), 7.77 (m, IH), 9.41 (bs, IH)
Example 12
Preparation of N-(2-[N',N'-diethy1amino]-5-[N"-(2,4-difluoropheny1su1fony1)- N"-ethylamino]pyrimidin-4-yl)-4'-(pyrrolidin-l-ylcarbonyloxy)-L- phenylalanine
[00143] Steps 1, 2, 3, 4, 6 and 7 were performed as for Example 2. Step 5 was performed using 2,4-difluorobenzenesulfonyl chloride in place of 4- fluorobenzensulfonyl chloride. lK NMR (CDC13) δ 0.91 (t, J= 6.9, 1.8H), 1.12 (m, 7.2H), 1.92 (bs, 4H), 2.50-4.00 (m, 13H), 4.78 (m, 0.6H), 4.88 (m, 0.4H), 6.55 (d, J= 6.9, 0.4H), 6.77 (d, J= 6.3, 0.6H), 6.80-7.38 (m, 6H), 7.51 (s, 0.4H), 7.58 (s, 0.6H), 7.74 (m, IH), 9.33 (m, IH) Example 13
Preparation of N-(2-[N'.N'-diethy1amino]-5-[N"-(2.4-difluoropheny1sulfony1)- N"-methylamino]pyrimidin-4-yl)-4'-(azetidin-1 -y1carbonyloxy)-L- phenylalanine
[00144] Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 11. Step 3 was performed as for Example 7.
1H NMR (CDC13) δ 1.14 (t, J=6.6, 6H), 2.32 (m, 2H), 2.50-3.80 (m, 9H), 4.13 (m, 4H), 4.62 (m, 0.6H), 4.81 (m, 0.4H), 5.81 (bd, 0.6H), 5.90 (bd, 0.4H),
6.90-7.40 (m, 7H), 7.77 (m, IH)
MS m z 619.2 (MH+)
Example 14
Preparation of N-(2-[N',N'-diethy1amino]-5-[N"-(2,4-difluorophenylsulfonyl)- N"-ethylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-pheny1alanine
[00145] Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 12. Step 3 was performed as for Example 7.
!H NMR (CDC13) δ 0.89 (t, J=6.7, 1.8H), 1.16 (m, 7.2H), 2.28 (m, 2H), 3.00- 4.00 (m, 8H), 4.09 (bs, 4H), 4.79 (m, 0.6H), 4.88 (m, 0.4H), 6.80-7.30 (m, 7H), 7.57 (s, 0.4H), 7.62 (s, 0.6H), 7.75 (m, IH), 11.9 (bs, IH)
MS m/z 633.2 (MET)
Example 15
Preparation of N-(2-rN',N'-diethy1amino]-5-[N"-(4-fluorophenylsulfony1)-N"- propargylamino]pyrimidin-4-yl)-4'-(pyrrolidin-1 -y1carbony1oxy)-L- phenylalanine
[00146] Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 2. Step 6 was performed using propargyl bromide in place of ethyl iodide. !H NMR (CDC13) δ 1.18 (m, 6H), 1.93 (bs, 4H), 2.37 (s, IH), 3.00-3.70 (m,
10H), 3.80 (d, J=21.3, 0.6H), 3.98 (d, J= 18.3, 0.4H), 4.51 (m, IH), 4.88 (m, IH), 6.75-7.35 (m, 7H), 7.58 (s, 0.6H), 7.63 (s, 0.4H), 7.86 (m, 2H), 9.71 (bs, IH) Example 16
Preparation o -(2-[N,,N'-diethylamino]-5-[N"-(2,4-difluoropheny1sulfony1)- N"-prQpargylaminQ]pyrimidin-4-yl)-4'-(ρyrro]idin-1-ylcarbonyloxy)-L- phenylalanine
[00147] Steps 1 , 2, 3, 4, 5 and 7 were perfonned as for Example 11. Step 6 was performed using propargyl bromide in place of dimethyl sulfate.
IHNMR (CDCI3) δ 1.17 (m, 6H), 1.94 (m, 4H), 2.40 (m, IH), 3.00-3.75 (m, 10H), 3.99 (d, J=18.0, 0.6H), 4.18 (d, J= 18.0, 0.4H), 4.50 (m, IH), 4.90 (m,
IH), 6.75-7.35 (m, 7H), 7.81 (m, 2H), 10.0 (bs, IH)
Example 17
Preparation of N-(2-[N',N'-diethy1amin ]-5-[N"-(2.4-difluoropheny1su1fony1)- N"-propargylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L- pbenylalanine
[00148] Steps 1, 2, 4, 5, 6 and 7 were performed as for Example 16. Step 3 was performed as for Example 7. iH NMR (CDC13) δ 1.18 (m, 6H), 2.34 (m, 3H), 3.00-3.75 (m, 6H), 3.80-4.25
(m, 5H), 4.47 (m, IH), 4.89 (m, IH), 6.75-7.35 (m, 7H), 7.79 (m, 2H), 10.3 (bs, IH)
MS m/z 643.2 (MH+)
Example 18
Preparation of N-(2-[N'1N'-diethylamino]-5-[N"-(4-fluoropbenylsulfonyl)-N"- propargylarnino]pyrimidin-4-yl)-4'-(azetidm-l-ylcarbonyloxy)-L- phenylalanine
[00149] Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 7. Step 6 was performed using propargyl bromide in place of ethyl iodide.
!H NMR (CDC13) δ 1.25 (m, 6H), 2.28 (m, 3H), 3.00-3.75 ( , 6H), 3.80-4.25 (m, 5H), 4.47 (m, IH), 4.89 (m, IH), 6.75-7.35 (m, 7H), 7.57 (s, 0.6H), 7.62
(s, 0.4H), 7.79 (m, 2H), 10.6 (bs, IH)
MS m/z 625.2 (MH+) Example 19
Preparation of N-(2-[ J,.N'-dietby1amino]-5-[N''-(4-cb1oropbeny1su1fony1)-N"- propargylamino]pyrimi din-4-y1)-4'-(pyrroli din- 1 -yl carbonyl oxy)-L- p enylalanine
[00150] Steps 1, 2, 3, 4, 5 and 7 were performed as for Example 1. Step 6 was performed using propargyl bromide in place of ethyl iodide.
Η NMR (CD3OD) δ 8.13 (s, IH), 7.86-7.82 (m, 2H), 7.62-7.58 (m, 2H), 7.32- 7.28 (m, 2H), 7.19-7.17 (m, IH), 7.04-6.98 (m, 2H), 4.83-4.5 (m, 2H), 4.12- 3.82 (m, IH), 3.63-3.37 (m, 8H), 3.27-3.08 (m, 2H), 2.72 (bs, IH), 2.04-1.86
(m, 4H), 1.24-1.07 (m, 6H)
13C NMR (CD3OD) δ 177.2, 176.5, 162.7, 156.7, 155.7, 154.5, 153.2, 142.6, 140.3, 137.4, 137.3, 133.1, 132.9, 132.8, 132.7, 132.2, 132.1, 124.3, 111.3, 80.5, 80.3, 77.7, 58.2, 57.7, 44.9, 43.4, 28.1, 27.3, 14.8, 14.7
MS m/z 655 (MH1")
[00151] The following methods may be used to test compounds of this invention.
Example A α4β' Integrin Adhesion Assay: Jurkat™ Cell Adhesion to Human Plasma Fibronectin
Procedure:
[00152] 96 well plates (Costar 3590 EIA plates) were coated with human fibronectin (Gibco/BRL, cat #33016-023) at a concentration of 10 μg/mL overnight at 4°C. The plates were then blocked with a solution of bovine serum albumin (BSA; 0.3%) in saline. Jurkat™ cells (maintained in log phase growth) were labeled with Calcein AM according to the manufacturer's instructions, and suspended at a concentration of 2 x 106 cells/mL in Hepes/Saline/BSA. The cells were then exposed to test and control compounds for 30 minutes at room temperature before transfer to individual wells of the fibronectin coated plate. Adhesion was allowed to occur for 35 minutes at 37°C. The wells were then washed by gentle aspiration and pipetting with fresh saline. Fluorescence associated with the remaining adherent cells was quantified using a fluorescence plate reader at EX 485/EM 530.
[00153] Cell cultures were prepared by first splitting the stationary phase
Jurkat™ cells at 1 : 10 on day one, and 1 :2 on day two to perform assay on day 3. The cells split 1 : 10 on day one were split 1 :4 on day 3 for a day 4 assay.
[00154] The assay plates were prepared by first making a working solution of Gibco/BRL Human Fibronectin (cat # 33016-023) in PB S++, at 10 μg/mL.
A Costai" 3590 EIA plate was then coated with 50 μL/well for 2 hours at room temperature (thought it can also be left overnight at 4 °C). Finally the plate was asperated and blocked with Hepes/Saline Buffer, 100 μL/well, for 1 hour at RT followed by washing 3X with 150 μL of PBS++.
[00155] Compound dilutions were accomplished by preparing 1 :3 serial dilutions of compounds as follows. For each plate (4 compounds/plate) 600 μL were added to 4 Bio-Rad Titertubes in a Titertube rack. Enough compound was added to each appropriate tube to give a 2X concentration using methods well known in the art. Using Falcon Flexiplates, 100 μL of Hepes/Saline buffer or human serum were added to rows B through G. A multi-channel pipetter set to 180 μL was used to with four tips spaced evenly the pipetter.
Each set of four tubes was mixed 5 times and 180 μL of 2X compound was transferred to the first column of each compound dilution in Row B, leaving Row A empty. 180 μL were added to the other wells in Row A. Serial dilutions were performed down the plate by transferring 50 μL to the next dilution and mixing 5 times, changing tips each time after mixing. Dilutions were stopped at Row F. Row G had no compound present.
[00156] A 20 μg/mL solution in Hepes/Saline buffer or human serum, of 21/6 antibody was the positive control and was set aside in a reagent trough to add to cell suspension plate.
[00157] The cell staining was accomplished by first harvesting the log-phase Jurkat™ cells by centrifugation in 50 mL tubes (1100 rpm for 5 minutes). The cells were resuspended in 50 mL PBS++, spun, and resuspend in 20 mL PBS++. The cells were stained by adding 20 μL of Calcein AM for 30 minutes RT. The volume was brought to 50 mL with Hepes/Saline buffer and the cells were counted, spun, and resuspend to 2 x 106 cells/mL in Hepes/Saline buffer or human serum.
[00158] The compounds were incubated using the following procedure. In a new flexiplate, 65 μL of stained cells were added to Rows B through H. Then 65 μL of 2X compounds were added to the appropriate rows following the plate setup and mixed three times. 65 μL of 2X-21/6 antibody were added to Row H and mixed 3X. Finally the plate was incubated at room temperature for 30 minutes. [00159] Fibronectin adhesion was measured using a fluorescent plate reader at EX 485/EM 530 after the following work up procedure. After incubation, the cells were mixed three times and 100 μL were transfered to the Fibronectin coated plates and incubated at 37 °C for about 35 minutes. Each plate was washed, row by row, by gently pipetting 100 μL of RT. PBS++ down the sides of the wells and turning the plate 90 degrees to aspirate. This procedure was repeated for a total of 3 washes. Each well was filled with 100 μL after washing by pipetting down the side of the well.
[00160] An IC50 value was calculated for each compound, both in the presence of the human serum and in the absence of human serum. IC50 is concentration at which the growth or activity is inhibited by 50% . The data is presented in the following tables.
Cell Adhesion to Human Plasma Fibronectin
(Without the human serum)
[00161]
Figure imgf000047_0001
Figure imgf000048_0001
Cell Adhesion to Human Plasma Fibronectin
(Containing human serum)
[00162]
Figure imgf000048_0002
Figure imgf000049_0001
Example B
In vitro Saturation Assay For Determining Binding of Candidate Compounds to α4β2
[00163] The following describes an in vitro assay to determine the plasma levels needed for a compound to be active in the Experimental Autoimmune Encephalomyehtis ("EAE") model, described in the next example, or in other in vivo models.
[00164] Log-growth Jurkat cells are washed and resuspended in normal animal plasma containing 20 μg/mL of the 15/7 antibody (Yednock, et al, J. Biol. Chem., (1995) 270(48):28740).
[00165] The Jurkat cells are diluted two-fold into either normal plasma samples containing known candidate compound amounts in various concentrations ranging from 66 μM to 0.01 μM, using a standard 12 point serial dilution for a standard curve, or into plasma samples obtained from the peripheral blood of candidate compound-treated animals.
[00166] Cells are then incubated for 30 minutes at room temperature, washed twice with phosphate-buffered saline ("PBS") containing 2% fetal bovine serum and lmM each of calcium chloride and magnesium chloride (assay medium) to remove unbound 15/7 antibody. [00167] The cells are then exposed to phycoerythrin-conjugated goat F(ab')2 anti-mouse IgG Fc (Immunotech, Westbrook, ME), which has been adsorbed for any non-specific cross-reactivity by co-incubation with 5% serum from the animal species being studied, at 1 :200 and incubated in the dark at 4 °C for 30 minutes.
[00168] Cells are washed twice with assay medium and resuspended in the same. They are then analyzed with a standard fluorescence activated cell sorter ("FACS") analysis as described in Yednock et al. J. Biol. Chem., 1995,
270:28740.
[00169] The data is then graphed as fluorescence versus dose, e.g., in a normal dose-response fashion. The dose levels that result in the upper plateau of the curve represent the levels needed to obtain efficacy in an in vivo model.
Example C
Cassette Dosing and Serum Analysis for detennination of Bioavailabihty
[00170] The oral bioavailabihty was screened by dosing rats with a cassette, i.e. mixture of 6 compounds per dosing solution. The cassette included 5 test articles and a standard compound, for a total dose of 10 mg/kg. Each compound/test article was converted to the sodium salt with equimolar 1 N
NaOH and dissolved in water at 2 mg/mL. The cassette was prepared by mixing equal volumes of each of the six solutions. The cassette dosing solution was mixed well and then the pH was adjusted to 7.5-9. The dosing solution was prepared the day before the study and stirred overnight at room temperature. [00171] Male Sprague Dawley (SD) rats from Charles River Laboratories, 6-8 weeks old were used in this screen. Rats were quarantined for at least one day and had continuous access to food and water. On the night before the administration of the cassette, the rats were fasted for approximately 16 h.
[00172] Four SD rats were assigned in each cassette. A single dose of the dosing solution was administered orally to each rat. The dosing volume (5 mL/kg) and time were recorded and rats were fed 2 h after dosing.
[00173] Blood samples were collected via cardiac puncture at the following time points: 4 h, 8 h and 12 h. hnmediately prior to blood collection, rats were anesthetized with CO2 gas within 10-20 seconds. After the 12-h samples were collected, the rats were euthanized via CO2 asphyxiation followed by cervical dislocation.
[00174] Blood samples were kept in heparinized microtainer tubes under sub- ambient temperature (4 °C) before they were processed. Blood samples were centrifuged (10000 rpm for 5 minutes) and plasma samples were removed and stored in a -20 °C freezer until analyzed for drug levels. Drug levels in the plasma were analyzed using the following protocol for direct plasma precipitation.
[00175] The in vivo plasma samples were prepared in a 1.5 mL 96-well plate, by adding, in order, 100 μL of the test plasma, 150 μL of methanol, followed by vortexing for 10-20 seconds. 150 μL of 0.05 ng/μL of an Internal Standard in acetonitrile were added and vortexed for 30 seconds.
[00176] The standard curve samples were prepared in a 1.5 mL 96-well plate, by adding, in order, 100 μL of control mouse plasma, followed by 150 μL of methanol and vortexing for 10-20 seconds. 150 μL of 0.05 ng/μL of an internal Standard in acetonitrile were added and vortexed for 30 seconds. The samples were spiked with 0-200 ng (10 concentrations) of the compound of interest in 50% methanol to obtain a standard curve range of 0.5 ng/mL - 2,000 ng/mL. Again, the sample was vortexed for 30 seconds.
[00177] The samples were then spun for 20-30 minutes at 3000 rpm in an
Eppendorf microfuge before 80-90% of supernatant was transferred into a clean 96-well plate. The organic solvent was then evaporated until the samples were dry (under N2 at 40°C/ 30-60 min (ZymarkTurbovap)).
[00178] The residue was then dissolved in 200 - 600 L mobile phase (50%
CH3OH/0.1% TFA). LC/MS/MS was then run using a PE-Sciex API-3000 triple quadurpole mass spectrometer (SN0749707), Perkin-Elmer, Series200auto-sampler, and shimadzu 10A pump. Acquisition was done with PE-Sciex Analyst (vl.l) and data analysis and quantification were accomplished using PE-Sciex Analyst (vl.l). A 5-50 μL sample volume was injected onto a reverse phase ThermoHypersil DASH- 18 column (Keystone 2.0 x 20 mm, 5 μm, PN: 8823025-701) using a mobile phase of 25% CH3OH, 0.1% TFA-100% CH3OH, 0.1% TFA. The run time was about 8 minutes at a flow rate of about 300 μL/minutes.
[00179] The Area Under the Curve (AUC) was calculated using the linear trapezoidal rule from t=0 to the last sampling time tx (see Handbook of Basic Pharmacokinetics, Wolfgang A. Ritschel and Gregory L. Kearns, 5th ed, 1999).
AUC0-* = S((Cn + Cn+1)/2)) (tn+] - tn) [(μg/mL)h]
[00180] In the case of the cassette dosing paradigm, samples at 4, 8 and 12 h post extravascular dosing, the AUC was calculated from t = 0 to t = 12 h. The AUC0→12 h values were calculated for each individual animal and the average AUC0→12 h are reported in the table below.
[00181]
Figure imgf000053_0001
Example D
Asthma Models
[00182] Inflammatory conditions mediated by α4βj integrin include, for example, eosinophil influx, airway hyper-responsiveness and occlusion that occurs with chronic asthma. The following describes animal models of asthma that were used to study the in vivo effects of the compounds of this invention for use in treating asthma. Rat Asthma Model
[00183] This model follows the procedures described by Chapman et al, Am J. Resp. Crit. Care Med,. 153 4, A219 (1996) and Chapman et al, Am. J. Resp. Crit Care Med 155:4, A881 (1997), both of which are incorporated by reference in their entirety. Ovalbumin (OA; lOmg/mL) were mixed with aluminum hydroxide (10 mg/mL) and injected (i.p.) in Brown Norway rats on day 0. Injections of OA, together with adjuvant, were repeated on days 7 and 14. On day 21, sensitized animals were restrained in plastic tubes and exposed (60 minutes) to an aerosol of OA (10 mg/kg) in a nose-only exposure system. Animals will be sacraficed 72 hours later with pentobarbital (250 mg/kg, i.p.).
The lungs were lavaged via a tracheal cannula using 3 aliquots (4 mL) of Hank's solution (HBSS x 10, 100 mL; EDTA 100 mM, 100 mL; HEPES 1 M, 25 mL; made up to 1 L with H2O); recovered cells were pooled and the total volume of recovered fluid adjusted to 12 mL by addition of Hank's solution. Total cells were counted (Sysmex microcell counter F-500, TOA Medical
Electronics Otd., Japan) and smears were made by diluting recovered fluid (to approximately 106 cells/mL) and pipetting an aliquot (100 μL) into a centrifuge (Cytospin, Shandon, U.K.). Smears were air dried, fixed using a solution of fast green in methanol (2 mg/mL) for 5 seconds and stained with eosin G (5 seconds) and thiazine (5 seconds) (Diff-Quick, Browne Ltd. U.K.) in order to differentiate eosinophils, neutrophils, macrophages and lymphocytes. A total of 500 cells per smear were counted by light microscopy under oil immersion (x 100). Compounds of this invention were formulated into a 0.5% carboxymethylcellulose and 2% Tween80 suspension and administered orally to rats which had been sensitized to the allergen, ovalbumin. Compounds which inhibited allergen-induced leucocyte accumulation in the airways of actively sensitized Brown Norway rats were considered to be active in this model. Mouse Asthma Model
[00184] Compounds were also evaluated in a mouse model of acute pulmonary inflammation following the procedures described by, Kung et al., Am J. Respir. Cell Mol. Biol. 13:360-365, (1995) and Schneider et al., (1999). Am J. Respir. Cell Mol. Biol. 20:448-457, (1999), which are each incorporated by reference in their entirety. Female Black/6 mice (8-12 weeks of age) were sensitized on day 1 by an intraperitoneal injection (i.p.) of 0.2 mL ova alum mixture containing 20 μg of ova (Grade 4, Sigma) and 2 mg inject Alum (Pierce). A booster injection was administered on day 14. Mice are challenged on days 28 and 29 with aerosolized 1% ova (in 0.9% saline) for 20 minutes. Mice are euthanized and bronchaveolar lavage samples (3 mL) are collected on day 30, 48 hours post first challenge. Eosinopliils were quantified by a FACs/FITC staining method. Compounds of this invention were formulated into a 0.5% carboxymethylcellulose and 2% TweenδO suspension and administered orally to mice which had been sensitized to the allergen, ovalbumin. Compounds which inhibited allergen-induced leucocyte accumulation in the airways of actively sensitized C57BL/6 mice were considered to be active in this model.
Sheep Asthma Model
[00185] This model follows the procedures described by Abraham et al, J.Clin, Invest, 93:776-787 (1994) and Abraham et al, Am J. Respir Crit Care Med 156:696-703 (1997), both of which are incorporated by reference in their entirety. Compounds of this invention have been evaluated by intravenous (saline aqueous solution), oral (2 % TweenδO, 0.5% carboxymethylcellulose), and aerosol administration to sheep which are hypersensitive to Ascaris suum antigen. Compounds wliich decrease the early antigen-induced bronchial response and/or block the late-phase airway response, e.g. have a protective effect against antigen-induced late responses and airway hyper-responsiveness ("AHR"), are considered to be active in this model. [00186] Allergic sheep which are shown to develop both early and late bronchial responses to inhaled Ascaris suum antigen were used to study the airway effects of the candidate compounds. Following topical anesthesia of the nasal passages with 2% lidocaine, a balloon catheter was advanced through one nostril into the lower esophagus. The animals were then incubated with a cuffed endotracheal tube through the other nostril with a flexible fiberoptic bronchoscope as a guide.
[00187] Pleural pressure was estimated according to Abraham (1994). Aerosols (see formulation below) were generated using a disposable medical nebulizer that provided an aerosol with a mass median aerodynamic diameter of 3.2 μm as determined with an Andersen cascade impactor. The nebulizer was connected to a dosimeter system consisting of a solenoid valve and a source of compressed air (20 psi). The output of the nebulizer was directed into a plastic T-piece, one end of which was connected to the inspiratory port of a piston respirator. The solenoid valve was activated for 1 second at the beginning of the inspiratory cycle of the respirator. Aerosols were delivered at Vτ of 500 mL and a rate of 20 breaths/minute. A 0.5% sodium bicarbonate solution only was used as a control.
[00188] To assess bronchial responsiveness, cumulative concentration- response curves to carbachol was generated according to Abraham (1994). Bronchial biopsies were taken prior to and following the initiation of treatment and 24 hours after antigen challenge. Bronchial biopsies were preformed according to Abraham (1994).
[00189] An in vitro adhesion study of alveolar macrophages were also performed according to Abraham (1994), and a percentage of adherent cells calculated. Aerosol Formulation
[00190] A solution of the candidate compound in 0.5% sodium bicarbonate/saline (w/v) at a concentration of 30.0 mg/mL is prepared using the following procedure:
[00191] A. Preparation of 0.5% Sodium Bicarbonate / Saline Stock Solution: ! 00.0 mL
Figure imgf000057_0001
Procedure:
1. Add 0.5g sodium bicarbonate into a 100 mL volumetric flask.
2. Add approximately 90.0 mL saline and sonicate until dissolved. 3. Q.S. to 100.0 mL with saline and mix thoroughly.
[00192] B. Preparation of 30.0 mg/mL Candidate Compound: 1 .0 L
Figure imgf000057_0002
Procedure:
1. Add 0.300 g of the candidate compound into a 10.0 mL volumetric flask.
2. Add approximately 9.7 mL of 0.5%o sodium bicarbonate / saline stock solution.
3. Sonicate until the candidate compound is completely dissolved.
4. Q.S. to 10.0 mL with 0.5% sodium bicarbonate / saline stock solution and mix thoroughly. Example E
10-Day Toxicity Study on C57B6 Mice
[00193] A 10-day study was conducted to evaluate the toxicity of compounds of the present invention to female C57B6 mice. The compound was administered by gavage at five dose levels, 0 (vehicle control), 10, 30, 100, 300 and 1000 mg/kg (mpk), with five mice in each dose level. The dose volume for all levels was 10 mL/kg. Dose solutions or suspensions were prepared in 2%> Tween 80 in 0.5%) carboxymethyl cellulose (CMC) and new dose solutions or suspensions were prepared every two - three days. In-life observations included body weights (study day 1, 2, 3, 5, 7, 8 and 11), daily cageside clinical observations (1-2/day) and periodic (study day -1, 2 and 9) functional observation battery.
[00194] At termination, blood samples were collected by cardiac puncture for clinical pathology (hematology and clinical chemistry) and drug levels. The EDTA blood samples were analyzed for total white blood cell count, red blood cell count, hemoglobin, hematocrit, erythrocyte indices (MCN, MCH, MCHC), platelets and a WBC five part differential (neutrophil, lymphocytes, monocytes, eosinophils and basophils). Heparinized plasma samples were analyzed for alanine transaminase, aspartate transaminase, alkaline phosphatase, total bilirubin, albumin, protein, calcium, glucose, urea nitrogen, creatinine, cholesterol and triglycerides.
[00195] After blood collection, the carcass was necropsied and organs (liver, spleen, kidneys, heart and thymus) were weighed. Tissue samples; brain, salivary glands, thymus, heart, lung, liver, kidney, adrenal spleen, stomach, duodenum, ileum, colon and uterus/ovary, were collected and formalin fixed. Tissues from the vehicle control and 300 and 1000 mpk group animals were processed to H & E stained glass slides and evaluated for histopathological lesions. [00196] Body weight changes, absolute and relative organ weights and clinical pathology results were analyzed for statistical significant differences compared to the vehicle controls by Dunnet's multiple comparison test using Prism software. The functional observation battery results were analyzed for differences using the Dunnet's, Fisher's exact tests and dose trend effects by the Cochran-Mantel-Haenszel correlation test using SAS software.
[00197] Using a conventional oral formulation, compounds of this invention would be active in this model.
Example F
Adjuvant-Induced Arthritis in Rats [00198] Adjuvant induced arthritis ("AIA") is an animal model useful in the study of rheumatoid arthritis (RA), which is induced by injecting M. tuberculosis in the base of the tail of Lewis rats. Between 10 and 15 days following injection, animals develop a severe, progressive arthritis.
[00199] Generally, compounds are tested for their ability to alter hind paw swelling and bone damage resulting from adjuvant-induced edema in rats. To quantitate the inhibition of hind paw swelling resulting from AIA, two phases of inflammation have been defined: (1) the primary and secondary injected hind paw, and (2) the secondary uninjected hind paw, which generally begins developing about eleven days from the induction of inflammation in the injected paw. Reduction of the latter type of inflammation is an indication of immunosuppressive activity. Cf. Chang, Arth. Rheum., 20, 1135-1141 (1977).
[00200] Using an animal model of RA, such as AIA, enables one to study the cellular events involved in the early stages of the disease. CD44 expression on macrophages and lymphocytes is up-regulated during the early development of adjuvant arthritis, whereas LFA-1 expression is up-regulated later in the development of the disease. Understanding the interactions between adhesion molecules and endothelium at the earliest stages of adjuvant arthritis could lead to significant advances in the methods used m the treatment of RA.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula (I) :
Figure imgf000061_0001
wherein each X is independently fluoro, chloro or bromo; p is an integer from 0 to 3;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, pyrrolyl, 2,5-dihydopynol-l-yl, piperidinyl, or 1,2,3,6-tetrahydro-pyridin-l-yl;
R2 is selected from the group consisting of lower alkyl, lower alkenyl, and lower alkylenecycloalkyl; and pharmaceutically acceptable salts thereof.
2. A compound of Formula (II):
Figure imgf000062_0001
(II)
wherein each X is independently selected from the group consisting of fluoro and chloro; m is an integer equal to 1 or 2;
R2 is selected from the group consisting of lower alkyl, lower alkenyl, and lower alkylenecycloalkyl;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
A compound of Formula (III)
Figure imgf000063_0001
wherein each X is independently fluoro or chloro; n is zero or one;
Rz is -CH2-R' where R' is selected from the group consisting of hydrogen, methyl or -CH=CH2;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyreolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
4. A compound of claim 1, wherein R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pynolidinyl, or piperidinyl group .
5. A compound of any one of claims 1, 2, or 3, wherein R2 is CH3.
6. A compound of claim 3, wherein X is F or Cl and n is 0.
7. A compound of claim 1 selected from the group consisting of:
N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(pynolidin-l-ylcarbonyloxy)-L-phenylalanine; N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(pynolidin-l-ylcarbonyloxy)-L-phenylalanine
ELN;
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(pynolidin -l-ylcarbonyloxy)-L- phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(pynolidin- 1 -ylcarbonyloxy)-L- phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(piperidin-l-ylcarbonyloxy)-L-phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(piperidin-l-ylcarbonyloxy)-L-phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(azetidin-l-ylcarbonyloxy)-L-phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L-phenylalanine ; N-(2-rN',N'-diethylamino]-5-[N"-(4-chlorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L-phenylalanine ;
N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfbnyl)-N"- methylamino]pyrimidin-4-yl)-4'-(pynolidin- 1 -ylcarbonyloxy)-L- phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfbnyl)-N"- ethyl-ιmino]pyrimidin-4-yl)-4'-(ρyrrolidin- 1 -ylcarbonyloxy)-L-phenylalanine ; N-(2-[N,,N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- methylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L-phenylalanine ;
N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- ethylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L-phenylalanine ; and pharmaceutically acceptable salts thereof.
8. A pharmaceutical composition comprising a phannaceutically acceptable carrier and a therapeutically effective amount of a compound from any one of claims 1-4, 6, or 7.
9. A method for treating a disease mediated by α 4 integrins in a patient, which method comprises administering a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound from any one of claims 1-4, 6, or 7.
10. A method of claim 9, wherein the disease is mediated by NLA-
11. A method of claim 9, wherein the disease is an inflammatory disease.
12. A compound of Formula (IN):
Figure imgf000066_0001
wherein each X is independently fluoro, chloro or bromo; p is an integer from 0 to 3;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, pyrrolyl, 2,5-dihydopyrrol-l-yl, piperidinyl, or 1,2,3,6-tetrahydropyridin-l-yl;
R2 is lower alkynyl; and pharmaceutically acceptable salts thereof.
13. A compound of Formula (V) :
Figure imgf000067_0001
wherein each X is independently selected from the group consisting of fluoro and chloro; m is an integer equal to 1 or 2;
R2 is lower alkynyl;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
14. A compound of Formula (NI)
Figure imgf000067_0002
wherein each X is independently fluoro or chloro; n is zero or one; R2 is lower alkynyl;
R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group; and pharmaceutically acceptable salts thereof.
15. A compound of claim 12, wherein R1 and R3 together with the nitrogen atom to which they are bound form an azetidinyl, pyrrolidinyl, or piperidinyl group.
16. A compound of any one of claims 12, 13, or 14, wherein R2 is propargyl.
17. A compound of claim 15, wherein X is F or Cl and n is 0.
18. A compound of claim 12 selected from the group consisting of:
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(pynolidin- 1 -ylcarbonyloxy)-L- phenylalanine;
N-(2-[N,,N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(pynolidin- 1 -ylcarbonyloxy)-L- phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(2,4-difluorophenylsulfonyl)-N - proρargylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L- phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-fluorophenylsulfonyl)-N"- proρargylamino]pyrimidin-4-yl)-4'-(azetidin- 1 -ylcarbonyloxy)-L- phenylalanine;
N-(2-[N',N'-diethylamino]-5-[N"-(4-chloroρhenylsulfonyl)-N"- propargylamino]pyrimidin-4-yl)-4'-(pynolidin- 1 -ylcarbonyloxy)-L- phenylalanine; and
pharmaceutically acceptable salts thereof.
19. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound from any one of claims 12-15, 17, or 18.
20. A method for treating a disease mediated by α 4 integrins in a patient, which method comprises administering a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound from any one of claims 12-15, 17, or 18.
21. The method of claim 20, wherein the disease is mediated by VLA-4.
22. The method of claim 20, wherein the disease is an inflammatory disease.
23. The method of claim 20, wherein the disease is rheumatoid arthritis.
PCT/US2003/016804 2002-05-24 2003-05-27 HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY α4 INTEGRINS WO2003099809A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
NZ535504A NZ535504A (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins and their use to treat inflammatory disorders
IL16422503A IL164225A0 (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by 4 integrins
EP03731419A EP1507775B1 (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins
EA200401562A EA008256B1 (en) 2002-05-24 2003-05-27 HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY αINTEGRINS
JP2004507466A JP4469715B2 (en) 2002-05-24 2003-05-27 Heterocyclic compounds that inhibit leukocyte adhesion mediated by α4 integrin
CA2481926A CA2481926C (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by .alpha.4 integrins
UA20041008397A UA76339C2 (en) 2002-05-24 2003-05-27 Heterocyclic compounds inhigiting leucocyte adhesion mediated by integrin
DE60325583T DE60325583D1 (en) 2002-05-24 2003-05-27 HETEROCYCLIC COMPOUNDS INHIBITING THE LEUKOCYTE ADHESION MEDIATED BY ALPHA4 INTEGRINS
DK03731419T DK1507775T3 (en) 2002-05-24 2003-05-27 Heterocyclic Compounds That Inhibit Leukocyte Adhesion Mediated by Alpha-4 Integrins
MXPA04010995A MXPA04010995A (en) 2002-05-24 2003-05-27 HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY alpha4.
KR1020047018173A KR100978832B1 (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha-4 integrins
US10/494,790 US7135477B2 (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by α4 integrins
BR0308881-2A BR0308881A (en) 2002-05-24 2003-05-27 Heterocyclic compounds that inhibit α4 integrin-mediated leukocyte adhesion
AU2003240823A AU2003240823C1 (en) 2002-05-24 2003-05-27 Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins
NO20043900A NO329933B1 (en) 2002-05-24 2004-09-17 Heterocyclic compounds, pharmaceutical compositions and uses thereof
IL164225A IL164225A (en) 2002-05-24 2004-09-22 Derivatives of benzensulfonyl - methyl - amino-pyrimidin - 4 - ylamino, pharmaceutical compositions comprising them and their use for the manufacture of medicaments for treating diseases mediated by alpha 4 integrin and a process for their preparation
HK05106028.8A HK1072608A1 (en) 2002-05-24 2005-07-15 Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins
US11/527,901 US7427628B2 (en) 2002-05-24 2006-09-26 Heterocyclic compounds which inhibit leukocyte adhesion mediated by α4 integrins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38302002P 2002-05-24 2002-05-24
US60/383,020 2002-05-24

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10494790 A-371-Of-International 2003-05-27
US11/527,901 Continuation US7427628B2 (en) 2002-05-24 2006-09-26 Heterocyclic compounds which inhibit leukocyte adhesion mediated by α4 integrins

Publications (1)

Publication Number Publication Date
WO2003099809A1 true WO2003099809A1 (en) 2003-12-04

Family

ID=29584493

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/016804 WO2003099809A1 (en) 2002-05-24 2003-05-27 HETEROCYCLIC COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY α4 INTEGRINS

Country Status (23)

Country Link
US (3) US7026328B2 (en)
EP (1) EP1507775B1 (en)
JP (1) JP4469715B2 (en)
KR (1) KR100978832B1 (en)
CN (1) CN1314682C (en)
AT (1) ATE419243T1 (en)
AU (1) AU2003240823C1 (en)
BR (1) BR0308881A (en)
CA (1) CA2481926C (en)
DE (1) DE60325583D1 (en)
DK (1) DK1507775T3 (en)
EA (1) EA008256B1 (en)
EC (1) ECSP045425A (en)
ES (1) ES2320436T3 (en)
HK (1) HK1072608A1 (en)
IL (2) IL164225A0 (en)
MX (1) MXPA04010995A (en)
NO (1) NO329933B1 (en)
NZ (1) NZ535504A (en)
TW (1) TWI281470B (en)
UA (1) UA76339C2 (en)
WO (1) WO2003099809A1 (en)
ZA (1) ZA200407590B (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005111020A2 (en) * 2004-04-30 2005-11-24 Elan Pharmaceuticals, Inc. Pyrimidine hydantoin analogues which inhibit leukocyte adhesion mediated by vla-4
WO2007041270A1 (en) 2005-09-29 2007-04-12 Elan Pharmaceuticals, Inc. Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by vla-4
WO2007041324A1 (en) 2005-09-29 2007-04-12 Elan Pharmaceuticals, Inc. Carbamate compounds which inhibit leukocyte adhesion mediated by vla-4
WO2007101165A1 (en) * 2006-02-27 2007-09-07 Elan Pharmaceuticals, Inc. Pyrimidinyl sulfonamide compounds which inhibit leukocyte adhesion mediated by vla-4
JP2008505927A (en) * 2004-07-08 2008-02-28 イーラン ファーマスーティカルズ、インコーポレイテッド Multivalent VLA-4 antagonist containing a polymer moiety
EP2085407A1 (en) 2008-02-04 2009-08-05 Sahltech I Göteborg AB Treatment of idiopathic thrombocytopenic purpura
EP2231185A1 (en) * 2007-12-07 2010-09-29 Elan Pharmaceuticals Inc. Methods and compositions for treating liquid tumors
US8030328B2 (en) 2005-05-20 2011-10-04 Elan Pharmaceuticals, Inc. Imidazolone phenylalanine derivatives
US11116760B2 (en) 2018-10-30 2021-09-14 Gilead Sciences, Inc. Quinoline derivatives
US11174256B2 (en) 2018-10-30 2021-11-16 Gilead Sciences, Inc. Imidazopyridine derivatives
US11179383B2 (en) 2018-10-30 2021-11-23 Gilead Sciences, Inc. Compounds for inhibition of α4β7 integrin
US11224600B2 (en) 2018-10-30 2022-01-18 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin
US11578069B2 (en) 2019-08-14 2023-02-14 Gilead Sciences, Inc. Compounds for inhibition of α4 β7 integrin

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE455106T1 (en) * 1999-01-22 2010-01-15 Elan Pharm Inc ACYL DERIVATIVES FOR THE TREATMENT OF VLA-4 RELATED DISEASES
TWI281470B (en) * 2002-05-24 2007-05-21 Elan Pharm Inc Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins
EP1635822A4 (en) * 2003-06-25 2009-06-24 Elan Pharm Inc Methods and compositions for treating rheumatoid arthritis
EP2007392A4 (en) * 2006-02-28 2010-04-07 Elan Pharm Inc Methods of treating inflammatory and autoimmune diseases with alpha-4 inhibitory compounds
JP2009528359A (en) 2006-02-28 2009-08-06 エラン ファーマシューティカルズ,インコーポレイテッド Methods of treating inflammatory and autoimmune diseases with natalizumab
CA2644110A1 (en) 2006-03-03 2007-09-13 Elan Pharmaceuticals, Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
EP2271634B1 (en) * 2008-04-29 2014-02-12 Integrative Research Laboratories Sweden AB Modulators of dopamine neurotransmission
CA2760151A1 (en) * 2009-04-27 2010-11-04 Elan Pharmaceuticals, Inc. Pyridinone antagonists of alpha-4 integrins
RS63744B1 (en) 2010-01-11 2022-12-30 Biogen Ma Inc Assay for jc virus antibodies
US11287423B2 (en) 2010-01-11 2022-03-29 Biogen Ma Inc. Assay for JC virus antibodies
JP6080521B2 (en) * 2012-11-30 2017-02-15 インターナショナル・ビジネス・マシーンズ・コーポレーションInternational Business Machines Corporation Data management mechanism of wide-area distributed medical information network
US10119976B2 (en) 2013-05-28 2018-11-06 Biogen Ma Inc. Method of assessing risk of PML
EP3294716B1 (en) * 2015-12-30 2020-04-15 Saint Louis University Meta-azacyclic amino benzoic acid derivatives as pan integrin antagonists
CN109541237A (en) * 2018-12-28 2019-03-29 吴江近岸蛋白质科技有限公司 The Determination of biological activity method of fibronectin

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6492372B1 (en) * 1999-01-22 2002-12-10 Elan Pharmaceuticals, Inc. Heteroaryl, heterocyclic and aryl compounds which inhibit leukocyte adhesion mediated by VLA-4

Family Cites Families (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2525656A1 (en) 1974-06-19 1976-01-15 Sandoz Ag PROCESS FOR PRODUCING NEW HETEROCYCLIC COMPOUNDS
US4046876A (en) * 1974-11-08 1977-09-06 Mitsubishi Chemical Industries Limited N2 -alkoxynaphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
US4104392A (en) * 1974-11-08 1978-08-01 Mitsubishi Chemical Industries Ltd. N2 -naphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof, and antithrombotic compositions and methods employing them
US4055636A (en) * 1974-11-08 1977-10-25 Mitsubishi Chemical Industries Ltd. N2 -alkoxynaphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
US4018915A (en) * 1976-01-05 1977-04-19 Mitsubishi Chemical Industries Ltd. N2 -alkoxynaphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
US4096255A (en) 1974-11-08 1978-06-20 Mitsubishi Chemical Industries Limited N2 -naphthalenesulfonyl-L-argininamides, and pharmaceutical salts, compositions and methods
US4073914A (en) 1974-11-08 1978-02-14 Mitsubishi Chemical Industries Limited N2 -naphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
US4055651A (en) 1974-11-08 1977-10-25 Mitsubishi Chemical Industries Ltd. N2 -alkoxynaphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
US4070457A (en) 1974-11-08 1978-01-24 Mitsubishi Chemical Industries Ltd. N2 -naphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
US4041156A (en) * 1974-11-08 1977-08-09 Mitsubishi Chemical Industries Limited N2 -alkoxynaphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
JPS5727454B2 (en) 1975-02-21 1982-06-10
US4018913A (en) 1976-01-14 1977-04-19 Mitsubishi Chemical Industries Ltd. N2 -alkoxynaphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
CA1102316A (en) 1975-12-09 1981-06-02 Shosuke Okamoto N su2 xx-arylsulfonyl-l-argininamides and the pharmaceutically acceptable salts thereof
US4036955A (en) * 1976-07-22 1977-07-19 Mitsubishi Chemical Industries Ltd. N2 -naphthalenesulfonyl-L-argininamides and the pharmaceutically acceptable salts thereof
DE2742173A1 (en) 1977-09-20 1979-03-29 Bayer Ag PHENOXY PYRIDINYL (PYRIMIDINYL) ALKANOLS, THE METHOD FOR THEIR MANUFACTURING AND THEIR USE AS FUNGICIDES
US4235871A (en) * 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
IT1211096B (en) 1981-08-20 1989-09-29 Lpb Ist Farm PYRIMIDINES AND S.TRIAZINICS HYPOLIPIDEMIZING ADAPTITY.
US4672065A (en) * 1982-11-19 1987-06-09 Chevron Research Company N-substituted phenoxyacetamide fungicides
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
CA1218655A (en) 1983-01-28 1987-03-03 Kathleen Biziere Process for the preparation of pyridazine derivatives having a psychotropic action
JPS59212480A (en) 1983-05-17 1984-12-01 Nippon Soda Co Ltd Pyridazine derivative and herbicide
DE3322720A1 (en) * 1983-06-24 1985-01-03 Chemische Werke Hüls AG, 4370 Marl USE OF 4-DL-ALKYLESTER- (ALPHA) -ALANINYL-6-CHLORINE-S-TRIAZINES SUBSTITUTED IN (2-POSITIONED) AMINO GROUPS AS HERBICIDES, ESPECIALLY AGAINST AIRPORTS
US4505910A (en) * 1983-06-30 1985-03-19 American Home Products Corporation Amino-pyrimidine derivatives, compositions and use
NZ210669A (en) 1983-12-27 1988-05-30 Syntex Inc Benzoxazin-4-one derivatives and pharmaceutical compositions
US4595364A (en) * 1984-02-15 1986-06-17 Molten Corp. Dental prosthesis and process for preparing the same
PH22520A (en) * 1984-11-12 1988-10-17 Yamanouchi Pharma Co Ltd Heterocyclic compounds having 4-lover alkyl-3-hydroxy-2-lower alkyl phenoxy-lower alkylene-y-group, and process of producing them
US4959364A (en) 1985-02-04 1990-09-25 G. D. Searle & Co. Method of treating inflammation, allergy, asthma and proliferative skin disease using heterocyclic amides
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
JPH0784424B2 (en) 1987-04-15 1995-09-13 味の素株式会社 Tyrosine derivative and its use
EP0330506A3 (en) * 1988-02-26 1990-06-20 Dana Farber Cancer Institute Vla proteins
DE3904931A1 (en) * 1989-02-17 1990-08-23 Bayer Ag PYRIDYL-SUBSTITUTED ACRYLIC ACID ESTERS
US5030644A (en) * 1989-07-31 1991-07-09 Merck & Co., Inc. Imidazole compounds and their use as transglutaminase inhibitors
US5260210A (en) 1989-09-27 1993-11-09 Rubin Lee L Blood-brain barrier model
US4992439A (en) 1990-02-13 1991-02-12 Bristol-Myers Squibb Company Pyridazine carboxylic acids and esters
FR2679903B1 (en) 1991-08-02 1993-12-03 Elf Sanofi DERIVATIVES OF N-SULFONYL INDOLINE CARRYING AN AMIDIC FUNCTION, THEIR PREPARATION, THE PHARMACEUTICAL COMPOSITIONS CONTAINING SAME.
NZ239846A (en) 1990-09-27 1994-11-25 Merck & Co Inc Sulphonamide derivatives and pharmaceutical compositions thereof
DE4108029A1 (en) 1991-03-13 1992-09-17 Bayer Ag TRIAZINYL-SUBSTITUTED ACRYLIC ACID ESTERS
WO1992016549A1 (en) 1991-03-18 1992-10-01 Pentapharm Ag Parasubstituted phenylalanine derivates
IT1247509B (en) 1991-04-19 1994-12-17 Univ Cagliari SYNTHESIS COMPOUNDS FOR USE IN THE TREATMENT OF RHINOVIRUS INFECTIONS
US5296486A (en) 1991-09-24 1994-03-22 Boehringer Ingelheim Pharmaceuticals, Inc. Leukotriene biosynthesis inhibitors
WO1993012809A1 (en) 1991-12-24 1993-07-08 Fred Hutchinson Cancer Research Center Competitive inhibition of high-avidity alpha4-beta1 receptor using tripeptide ldv
CA2130754C (en) 1992-03-11 2005-02-08 Damian W. Grobelny Amine derivatives of oxo- and hydroxy-substituted hydrocarbons
DE4227748A1 (en) 1992-08-21 1994-02-24 Bayer Ag Pyridyloxy-acrylic acid ester
JP2848232B2 (en) * 1993-02-19 1999-01-20 武田薬品工業株式会社 Aldehyde derivatives
US5770573A (en) * 1993-12-06 1998-06-23 Cytel Corporation CS-1 peptidomimetics, compositions and methods of using the same
TW530047B (en) 1994-06-08 2003-05-01 Pfizer Corticotropin releasing factor antagonists
US5510332A (en) 1994-07-07 1996-04-23 Texas Biotechnology Corporation Process to inhibit binding of the integrin α4 62 1 to VCAM-1 or fibronectin and linear peptides therefor
CA2193508A1 (en) 1994-07-11 1996-01-25 Eugene D. Thorsett Inhibitors of leukocyte adhesion
US6306840B1 (en) 1995-01-23 2001-10-23 Biogen, Inc. Cell adhesion inhibitors
IL117659A (en) 1995-04-13 2000-12-06 Dainippon Pharmaceutical Co Substituted 2-phenyl pyrimidino amino acetamide derivative process for preparing the same and a pharmaceutical composition containing same
NZ318926A (en) * 1995-08-30 1999-04-29 Searle & Co Alpha v beta 3 integrin (vitronectin) inhibitors or anatagonists derived from benzoic acid for treating diseases of cell adhesion
PT765879E (en) 1995-09-29 2001-05-31 Sankyo Co 13-SUBSTITUTED DERIVATIVES OF MILBEMYCIN-5-OXYME PREPARATION AND USE AGAINST INSECTS AND OTHER PLAGUES
DE19536891A1 (en) 1995-10-04 1997-04-10 Basf Ag New amino acid derivatives, their production and use
DE19548709A1 (en) 1995-12-23 1997-07-03 Merck Patent Gmbh Tyrosine derivatives
CN1129607C (en) 1996-06-21 2003-12-03 武田药品工业株式会社 Method for producing peptides
PL330915A1 (en) 1996-06-28 1999-06-07 Merck Patent Gmbh Phenylaniline derivatives as inhibitors of integrin
DE19629817A1 (en) * 1996-07-24 1998-01-29 Hoechst Ag New imino derivatives as inhibitors of bone resorption and vitronectin receptor antagonists
DE19647317A1 (en) * 1996-11-15 1998-05-20 Hoechst Schering Agrevo Gmbh Substituted nitrogen heterocycles, processes for their preparation and their use as pesticides
DE19647381A1 (en) * 1996-11-15 1998-05-20 Hoechst Ag New heterocycles as leukocyte adhesion inhibitors and VLA-4 antagonists
AU739035B2 (en) 1996-11-22 2001-10-04 Elan Pharmaceuticals, Inc. N-(aryl/heteroarylacetyl) amino acid esters, pharmaceutical compositions comprising same, and methods for inhibiting beta-amyloid peptide release and/or its synthesis by use of such compounds
WO1998033783A1 (en) 1997-02-04 1998-08-06 Versicor, Inc. Solid phase and combinatorial library syntheses of 3,1-benzoxazine-4-ones
DE19713000A1 (en) 1997-03-27 1998-10-01 Merck Patent Gmbh New heterocyclic compounds are adhesion receptor antagonists
WO1998053814A1 (en) 1997-05-29 1998-12-03 Merck & Co., Inc. Heterocyclic amide compounds as cell adhesion inhibitors
DE69833654T2 (en) 1997-05-29 2006-12-14 Merck & Co., Inc. (A New Jersey Corp.) BIARYL ALKANIC ACIDS IN USE AS CELL ADHESION INHIBITORS
AR016133A1 (en) 1997-07-31 2001-06-20 Wyeth Corp CARBAMILOXI COMPOUND INHIBITING THE ADHESION OF LEUKOCYTES THROUGH VLA-4, COMPOUNDS THAT ARE DRUGS OF THESE COMPOUNDS, PHARMACEUTICAL COMPOSITION, METHOD FOR SETTING VLA-4 TO A BIOLOGICAL SAMPLE, METHOD FOR THE TREATMENT OF A TREATMENT
ZA986827B (en) 1997-07-31 2000-05-02 Athena Neurosciences Inc Substituted phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4.
CN1265670A (en) 1997-07-31 2000-09-06 伊兰药品公司 Dipeptide and related compounds which inhibit leukocyte adhesion medicated by VLA-4
PL338506A1 (en) 1997-07-31 2000-11-06 Elan Pharm Inc Compounds inhibiting adhesion of leucocytes occuring through the mediation of vla-4
RU2220950C2 (en) 1997-08-22 2004-01-10 Ф.Хоффманн-Ля Рош Аг N-alkanoylphenylalanine derivatives
AU742928C (en) 1997-08-22 2003-02-20 F. Hoffmann-La Roche Ag N-alkanoylphenylalanine derivatives
EP1049665A1 (en) 1998-01-23 2000-11-08 Novartis AG (Novartis SA) (Novartis Inc.) Vla-4 antagonists
US6329372B1 (en) 1998-01-27 2001-12-11 Celltech Therapeutics Limited Phenylalanine derivatives
WO1999052493A2 (en) 1998-04-16 1999-10-21 Texas Biotechnology Corporation Compounds that inhibit the binding of integrins to their receptors
GB9821061D0 (en) 1998-09-28 1998-11-18 Celltech Therapeutics Ltd Chemical compounds
GB9825652D0 (en) 1998-11-23 1999-01-13 Celltech Therapeutics Ltd Chemical compounds
WO2000043369A1 (en) 1999-01-22 2000-07-27 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by vla-4
PE20020384A1 (en) 2000-07-21 2002-05-28 Schering Corp PEPTIDES AS INHIBITORS OF THE HEPATITIS C VIRUS SERINE NS3 / NS4a PROTEASE
TWI281470B (en) * 2002-05-24 2007-05-21 Elan Pharm Inc Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6492372B1 (en) * 1999-01-22 2002-12-10 Elan Pharmaceuticals, Inc. Heteroaryl, heterocyclic and aryl compounds which inhibit leukocyte adhesion mediated by VLA-4

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
ABRAHAM ET AL., AM J. RESPIR CRIT CARE MED, vol. 156, 1997, pages 696 - 703
ABRAHAM ET AL., J.CLIN, INVEST, vol. 93, 1994, pages 776 - 787
BAO ET AL., DIFF., vol. 52, 1993, pages 239
BARON ET AL., J. CLIN. INVEST., vol. 93, 1994, pages 1700
BARON ET AL., J. EXP. MED., vol. 177, 1993, pages 57
BURKLY ET AL., DIABETES, vol. 43, 1994, pages 529
ELICES ET AL., J. CLIN. INVEST., vol. 93, 1994, pages 405
HAMANN ET AL., J. IMMUNOLOGY, vol. 152, 1994, pages 3283
KAWAGUCHI ET AL., JAPANESE J. CANCER RES., vol. 83, 1992, pages 1304
LAURI ET AL., J. CANCER, vol. 68, 1993, pages 862
LI ET AL., ARTERIOSCLER. THROMB., vol. 13, 1993, pages 197
OKARHARA ET AL., CAN. RES., vol. 54, 1994, pages 3233
PAAVONEN ET AL., INT. J. CAN., vol. 58, 1994, pages 298
PAUL ET AL., TRANSPL. PROCEED., vol. 25, 1993, pages 813
POSTIGO ET AL., J: CLIN. INVEST., vol. 89, 1991, pages 1445
SASSEVILLE ET AL., ANI. J. PATH., vol. 144, 1994, pages 27
SCHADENDORF ET AL., J. PATH., vol. 170, 1993, pages 429
VAN DINTHER-JANSSEN ET AL., ANNALS. RHEUMATIC DIS., vol. 52, 1993, pages 672
VAN DINTHER-JANSSEN ET AL., J. IMMUNOLOGY, vol. L41, 1991, pages 4207
YANG ET AL., PROC. NAT. ACAD. SCIENCE, vol. 9Q, 1993, pages 10494
YEDNOCK ET AL., NATURE, vol. 356, 1992, pages 63

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005111020A3 (en) * 2004-04-30 2006-01-26 Elan Pharm Inc Pyrimidine hydantoin analogues which inhibit leukocyte adhesion mediated by vla-4
US7205310B2 (en) 2004-04-30 2007-04-17 Elan Pharmaceuticals, Inc. Pyrimidine hydantoin analogues which inhibit leukocyte adhesion mediated by VLA-4
WO2005111020A2 (en) * 2004-04-30 2005-11-24 Elan Pharmaceuticals, Inc. Pyrimidine hydantoin analogues which inhibit leukocyte adhesion mediated by vla-4
US7794700B2 (en) 2004-07-08 2010-09-14 Elan Pharmaceuticals Inc. Multimeric VLA-4 antagonists comprising polymer moieties
US8263063B2 (en) 2004-07-08 2012-09-11 Elan Pharmaceuticals, Inc. Multimeric VLA-4 antagonists comprising polymer moieties
EP2298356A3 (en) * 2004-07-08 2011-08-10 Elan Pharmaceuticals, Inc. Multivalent VLA-4 antagonists comprising polymer moieties
JP2008505927A (en) * 2004-07-08 2008-02-28 イーラン ファーマスーティカルズ、インコーポレイテッド Multivalent VLA-4 antagonist containing a polymer moiety
EP2258399A3 (en) * 2004-07-08 2011-08-10 Elan Pharmaceuticals Inc. Multivalent VLA-4 antagonists comprising polymer moieties
US8030328B2 (en) 2005-05-20 2011-10-04 Elan Pharmaceuticals, Inc. Imidazolone phenylalanine derivatives
US7511052B2 (en) 2005-09-29 2009-03-31 Elan Pharmaceuticals, Inc. Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by VLA-4
AU2006297220B2 (en) * 2005-09-29 2012-04-26 Elan Pharmaceuticals, Inc. Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by VLA-4
AU2006297220B8 (en) * 2005-09-29 2013-01-31 Elan Pharmaceuticals, Inc. Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by VLA-4
CN101273034B (en) * 2005-09-29 2011-07-27 伊兰制药公司 Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by VLA-4
AU2006297220C1 (en) * 2005-09-29 2012-11-29 Elan Pharmaceuticals, Inc. Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by VLA-4
WO2007041270A1 (en) 2005-09-29 2007-04-12 Elan Pharmaceuticals, Inc. Pyrimidinyl amide compounds which inhibit leukocyte adhesion mediated by vla-4
EA015388B1 (en) * 2005-09-29 2011-08-30 Элан Фамэсьютикэлс, Инк. PYRIMIDINYL AMIDE COMPOUNDS (VARIANTS), PHARMACEUTICAL COMPOSITION COMPRISING THEM, AND METHOD OF TREATMENT OF A DISEASE MEDIATED BY α4-INTEGRINS
WO2007041324A1 (en) 2005-09-29 2007-04-12 Elan Pharmaceuticals, Inc. Carbamate compounds which inhibit leukocyte adhesion mediated by vla-4
AU2007220068B2 (en) * 2006-02-27 2012-09-13 Elan Pharmaceuticals, Inc. Pyrimidinyl sulfonamide compounds which inhibit leukocyte adhesion mediated by VLA-4
WO2007101165A1 (en) * 2006-02-27 2007-09-07 Elan Pharmaceuticals, Inc. Pyrimidinyl sulfonamide compounds which inhibit leukocyte adhesion mediated by vla-4
JP2009528296A (en) * 2006-02-27 2009-08-06 エラン ファーマシューティカルズ,インコーポレイテッド Pyrimidinylsulfonamide compounds that inhibit leukocyte adhesion mediated by VAL-4
EA017110B1 (en) * 2006-02-27 2012-09-28 Элан Фамэсьютикэлс, Инк. PYRIMIDINYLSULFONAMIDE COMPOUNDS (VARIANTS), METHOD FOR PREPARING SAME (VARIANTS), PHARMACEUTICAL COMPOSITION, METHOD FOR TREATING A DISEASE MEDIATED BY α4 INTEGRIN, METHOD FOR REDUCING AND/OR PREVENTING AN INFLAMMATORY COMPONENT OF A DISEASE OR AN AUTOIMMUNE RESPONSE
EP2231185A4 (en) * 2007-12-07 2012-06-27 Elan Pharm Inc Methods and compositions for treating liquid tumors
US8269009B2 (en) 2007-12-07 2012-09-18 Elan Pharmaceuticals, Inc. Methods and compositions for treating liquid tumors
EP2231185A1 (en) * 2007-12-07 2010-09-29 Elan Pharmaceuticals Inc. Methods and compositions for treating liquid tumors
EP2085407A1 (en) 2008-02-04 2009-08-05 Sahltech I Göteborg AB Treatment of idiopathic thrombocytopenic purpura
US11116760B2 (en) 2018-10-30 2021-09-14 Gilead Sciences, Inc. Quinoline derivatives
US11174256B2 (en) 2018-10-30 2021-11-16 Gilead Sciences, Inc. Imidazopyridine derivatives
US11179383B2 (en) 2018-10-30 2021-11-23 Gilead Sciences, Inc. Compounds for inhibition of α4β7 integrin
US11224600B2 (en) 2018-10-30 2022-01-18 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin
US11578069B2 (en) 2019-08-14 2023-02-14 Gilead Sciences, Inc. Compounds for inhibition of α4 β7 integrin

Also Published As

Publication number Publication date
JP2005527628A (en) 2005-09-15
AU2003240823C1 (en) 2009-05-28
MXPA04010995A (en) 2005-09-08
DE60325583D1 (en) 2009-02-12
US20070027131A1 (en) 2007-02-01
US20050119290A1 (en) 2005-06-02
EP1507775B1 (en) 2008-12-31
US20040138243A1 (en) 2004-07-15
HK1072608A1 (en) 2005-09-02
KR20050003434A (en) 2005-01-10
KR100978832B1 (en) 2010-08-30
TW200400946A (en) 2004-01-16
JP4469715B2 (en) 2010-05-26
DK1507775T3 (en) 2009-05-11
EA200401562A1 (en) 2005-06-30
IL164225A0 (en) 2005-12-18
CN1314682C (en) 2007-05-09
US7427628B2 (en) 2008-09-23
EA008256B1 (en) 2007-04-27
UA76339C2 (en) 2006-07-17
AU2003240823A1 (en) 2003-12-12
CN1656090A (en) 2005-08-17
TWI281470B (en) 2007-05-21
CA2481926A1 (en) 2003-12-04
ATE419243T1 (en) 2009-01-15
IL164225A (en) 2010-11-30
NO20043900L (en) 2004-11-10
ECSP045425A (en) 2005-03-10
ES2320436T3 (en) 2009-05-22
CA2481926C (en) 2012-04-24
EP1507775A1 (en) 2005-02-23
US7135477B2 (en) 2006-11-14
NZ535504A (en) 2005-11-25
ZA200407590B (en) 2006-10-25
EP1507775A4 (en) 2006-05-24
US7026328B2 (en) 2006-04-11
NO329933B1 (en) 2011-01-24
BR0308881A (en) 2005-01-11
AU2003240823B2 (en) 2008-11-06

Similar Documents

Publication Publication Date Title
US7427628B2 (en) Heterocyclic compounds which inhibit leukocyte adhesion mediated by α4 integrins
EP1507533B1 (en) Heteroaryl compounds which inhibit leukocyte adhesion mediated by alpha-4 integrins
US20100016345A1 (en) Pyrimidinyl sulfonamide compounds which inhibit leukocyte adhesion mediated by vla-4
US7205310B2 (en) Pyrimidine hydantoin analogues which inhibit leukocyte adhesion mediated by VLA-4
KONRADI et al. Patent 2481926 Summary
KR20050018680A (en) Heteroaryl compounds which inhibit leukocyte adhesion mediated by alpha-4 integrins

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 1200400949

Country of ref document: VN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2003240823

Country of ref document: AU

Ref document number: 2003731419

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2004/07590

Country of ref document: ZA

Ref document number: 535504

Country of ref document: NZ

Ref document number: 200407590

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 164225

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2004507466

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1-2004-501523

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2481926

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1535/KOLNP/2004

Country of ref document: IN

Ref document number: 01535/KOLNP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2004/010995

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020047018173

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 20038115484

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 10494790

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 200401562

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 1020047018173

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2003731419

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 535504

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 535504

Country of ref document: NZ