WO2011159550A2 - Lysophosphatidic acid receptor antagonist and uses thereof - Google Patents

Lysophosphatidic acid receptor antagonist and uses thereof Download PDF

Info

Publication number
WO2011159550A2
WO2011159550A2 PCT/US2011/039872 US2011039872W WO2011159550A2 WO 2011159550 A2 WO2011159550 A2 WO 2011159550A2 US 2011039872 W US2011039872 W US 2011039872W WO 2011159550 A2 WO2011159550 A2 WO 2011159550A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
mammal
fibrosis
Prior art date
Application number
PCT/US2011/039872
Other languages
French (fr)
Other versions
WO2011159550A3 (en
Inventor
Jason Edward Brittain
Thomas Jon Seiders
Christopher David King
Original Assignee
Amidra Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amidra Pharmaceuticals, Inc. filed Critical Amidra Pharmaceuticals, Inc.
Publication of WO2011159550A2 publication Critical patent/WO2011159550A2/en
Publication of WO2011159550A3 publication Critical patent/WO2011159550A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/0095Drinks; Beverages; Syrups; Compositions for reconstitution thereof, e.g. powders or tablets to be dispersed in a glass of water; Veterinary drenches
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • A61K9/2846Poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system

Definitions

  • LPA receptor antagonist ⁇ 4'-[3-methyl-4-((R)-l -phenyl - ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid (Compound 1), pharmaceutically acceptable salts, polymorphs, amorphous phases, metabolites thereof, as well as pharmaceutical compositions thereof, and methods of use thereof in the treatment or prevention or diagnosis of diseases or conditions associated with the activity of one or more of the lysophosphatidic acid (LPA) receptors.
  • LPA lysophosphatidic acid
  • Lysophospholipids are membrane-derived bioactive lipid mediators. Lysophospholipids affect fundamental cellular functions that include proliferation, differentiation, survival, migration, adhesion, invasion, and morphogensis. These functions influence many biological processes that include, but are not limited to, neurogensis, angiogenesis, wound healing, fibrosis, immunity, and carcinogenesis.
  • Lysophosphatidic acid is a lysophospholipid that has been shown to act through sets of specific G protein-coupled receptors (GPCRs) in an autocrine and paracrine fashion. LPA binding to its cognate GPCRs (LPAi, LPA 2 , LPA 3 , LPA 4 , LPA 5 , LPA 6 ) activates intracellular signaling pathways to produce a variety of biological responses. Antagonists of the LPA receptors find use in the treatment of diseases, disorders or conditions in which LPA plays a role.
  • GPCRs G protein-coupled receptors
  • Compound 1 is ⁇ 4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl ⁇ -acetic acid (Compound 1), including all pharmaceutically acceptable solvates (including hydrates), prodrugs, polymorphs, amorphous phases and metabolites thereof or a pharmaceutically acceptable salt of Compound 1 including (including hydrates), prodrugs, polymorphs, amorphous phases and metabolites thereof, and methods of uses thereof.
  • Compound 1, as well as the pharmaceutically acceptable salts thereof are used in the manufacture of medicaments for the treatment or prevention of LPA mediated and/or LPA dependent diseases, disorders, or conditions.
  • Compound 1 is a LPAi antagonist.
  • pharmaceutical compositions comprising Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. the sodium salt) as the active ingredient in the pharmaceutical composition.
  • Compound 1 has the following structure:
  • the crystalline form of Compound 1 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1 ; a differential scanning calorimetry (DSC) thermogram with endotherms at about 157°C and about 198°C; a differential scanning calorimetry (DSC) thermogram
  • the crystalline form of Compound 1 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least four of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least five of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having all of the preceding properties.
  • the crystalline form of Compound 1 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 1 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.2° 2-Theta, 9.2° 2-Theta, 10.6° 2-Theta, 12.6° 2- Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 1 has a DSC thermogram with endotherms at about 157°C and about 198°C.
  • the crystalline form of Compound 1 has substantially the same X-ray powder diffraction (XRPD) pattern post storage at 40 C/75% RH for one week.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 1 is characterized as having a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 4.
  • DSC differential scanning calorimetry
  • the crystalline form of Compound 1 is characterized as having a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 4.
  • TGA thermogravimetric analysis
  • the crystalline form of Compound 1 is substantially free of the S- isomer.
  • the crystalline form of Compound 1 is crystallized from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, nitromethane, tetrahydrofuran, dioxane, methyl ethyl ketone, acetone, 2-methoxyethanol, or methanol.
  • samples of Compound 1 include a detectable amount of palladium that is less than 20 ppm. In some embodiments, samples of Compound 1 include a detectable amount of palladium that is less than 15 ppm. In some embodiments, samples of Compound 1 include a detectable amount of palladium that is less than 10 ppm. In some embodiments, samples of Compound 1 do not include a detectable amount of palladium.
  • Compound 2 has the following structure:
  • the crystalline form of Compound 2 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2-Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C; a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7; or combinations thereof.
  • XRPD X- ray powder diffraction
  • the crystalline form of Compound 2 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least four of the preceding properties. In some
  • the crystalline form of Compound 2 is characterized as having all of the preceding properties.
  • the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta and 15.8° 2-Theta.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2- Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 2 is characterized as having a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C.
  • DSC differential scanning calorimetry
  • the crystalline form of Compound 2 is characterized as having a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7.
  • the crystalline form of Compound 2 is characterized as having a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7.
  • TGA thermogravimetric analysis
  • the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 8.5° 2-Theta, 9.3° 2-Theta, 16.6° 2-Theta, 17.1° 2-Theta and 21.4° 2-Theta.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 3.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 2 is substantially free of the S- isomer.
  • the crystalline form of Compound 2 is crystallized from isopropanol.
  • samples of Compound 2 include a detectable amount of palladium that is less than 20 ppm. In some embodiments, samples of Compound 2 include a detectable amount of palladium that is less than 15 ppm. In some embodiments, samples of Compound 2 do not include a detectable amount of palladium.
  • described herein is amorphous ⁇ 4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid, sodium salt (Compound 2). In some embodiments, amorphous Compound 2 is substantially free of the S-isomer.
  • a pharmaceutically acceptable salt of ⁇ 4'-[3-methyl-4- ((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid (Compound 1), wherein the pharmaceutically acceptable salt is a sodium salt, calcium salt, potassium salt, ammonium salt, L-arginine salt, L-lysine salt, or N-methyl-D-glucamine salt. In some embodiments, the pharmaceutically acceptable salt is a sodium salt.
  • the pharmaceutically acceptable salt is ⁇ 4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4- yl ⁇ -acetic acid, sodium salt (Compound 2).
  • the pharmaceutically acceptable salt is amorphous.
  • the pharmaceutically acceptable salt is crystalline.
  • the pharmaceutically acceptable salt is Compound 2 and Compound 2 is crystalline.
  • the pharmaceutically acceptable salt is Compound 2 and
  • Compound 2 is amorphous.
  • Compound 2 is crystalline and: (a) has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta and 15.8° 2-Theta; (b) has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; (c) has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 8.5° 2-Theta, 9.3° 2-Theta, 16.6° 2- Theta, 17.1° 2-Theta and 21.4° 2-Theta; (d) has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 3; (e) was obtained from isopropanol; or (f) combinations thereof.
  • Compound 2 is crystalline and has at least two of the properties selected from (a), (b), (c), (d) and (e).
  • the pharmaceutically acceptable salt of Compound 1 is substantially free of the S-isomer.
  • the pharmaceutically acceptable salt of Compound 1 includes a detectable amount of palladium that is less than 20 ppm. In some embodiments, the
  • pharmaceutically acceptable salt of Compound 1 includes a detectable amount of palladium that is less than 15 ppm. In some embodiments, the pharmaceutically acceptable salt of Compound 1 does not include a detectable amount of palladium.
  • composition comprising
  • Compound 1, or a pharmaceutically acceptable salt thereof is amorphous. In some embodiments, Compound 1 , or the pharmaceutically acceptable salt thereof is crystalline.
  • compositions comprising a crystalline form of Compound 1 or a pharmaceutically acceptable salt. In some embodiments, described herein are pharmaceutical compositions comprising a crystalline form of Compound 2. [0039] In some embodiments, the pharmaceutical composition comprises at least inactive ingredient selected from pharmaceutically acceptable carriers, diluents and excipients.
  • the pharmaceutical composition comprises Compound 2.
  • the pharmaceutical composition comprises crystalline Compound 2.
  • Compound 2 is greater than 96% pure. In some embodiments, Compound 2 is greater than 97%> pure. In some embodiments, Compound 2 is greater than 98%) pure.
  • the pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, inhalation, nasal administration, topical administration, ophthalmic administration or otic administration.
  • the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a suspension, a gel, a colloid, a dispersion, a suspension, a solution, an emulsion, an ointment, a lotion, an eye drop or an ear drop.
  • the pharmaceutical composition is in a form suitable for oral administration to a mammal.
  • the pharmaceutical composition is in the form of a pill, capsule, tablet, aqueous solution, aqueous suspension, non-aqueous solution, or non-aqueous suspension.
  • the pharmaceutical composition is in the form of a capsule.
  • the pharmaceutical composition is in the form of an immediate release capsule or an enteric coated capsule.
  • the capsule is a hard gelatine capsule or hypromellose (HPMC) capsule.
  • HPMC hypromellose
  • the capsule comprises at least one excipient in addition to the hard gelatine capsule or hypromellose (HPMC) capsule.
  • HPMC hypromellose
  • the pharmaceutical composition is in the form of a tablet.
  • the pharmaceutical composition is in the form of an immediate release tablet, an enteric coated tablet, or a sustained release tablet.
  • the pharmaceutical composition is in the form of a moisture barrier coated tablet.
  • the pharmaceutical composition is in the form of an aqueous solution or aqueous suspension.
  • a single dose of the pharmaceutical composition comprises about 1 Omg to about 1 OOOmg of Compound 1 , or a pharmaceutically acceptable salt thereof.
  • a single dose of the pharmaceutical composition comprises about 50mg, about lOOmg, about 150mg, about 200mg, about 250mg, about 300mg, about 350mg, about 400mg, about 450mg, about 500mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg or about 1000 mg of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • a single dose of the pharmaceutical composition comprises about lOmg to about lOOOmg of Compound 2.
  • a single dose of the pharmaceutical composition comprises about 50mg, about lOOmg, about 150mg, about 200mg, about 250mg, about 300mg, about 350mg, about 400mg, about 450mg, about 500mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg or about 1000 mg of Compound 2.
  • described herein is a pharmaceutical composition that provides at least one metabolite of Compound 1 after administration to a mammal.
  • the at least one metabolite is selected from among:
  • M7 M4
  • M8 glucuronidation of M7
  • described herein is a method of inhibiting the physiological activity of LPA in a mammal comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • described herein is a method for treating or preventing a LPA- dependent or LPA-mediated disease or condition in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • a pharmaceutically acceptable salt thereof e.g. Compound 2
  • the LPA-dependent or LPA-mediated disease or condition is selected from lung fibrosis, asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud's phenomenom, or neuropathic pain.
  • COPD chronic obstructive pulmonary disease
  • renal fibrosis acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut
  • lung fibrosis asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut, peritoneal fibrosis, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud's phenomenom, neuropathic pain, or spinal cord injury in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2)
  • described herein is a method of controlling the activation of LPA receptors in a tissue in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • the activation of LPA receptors in a tissue in a mammal results in fibrosis.
  • fibrosis comprises lung fibrosis, renal fibrosis, hepatic fibrosis, cutaneous fibrosis, or peritoneal fibrosis.
  • the fibrosis comprises lung fibrosis, renal fibrosis, hepatic fibrosis or cutaneous fibrosis.
  • described herein is a method of improving lung function in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 or a pharmaceutical composition thereof to the mammal in need thereof.
  • the mammal has been diagnosed as having lung fibrosis.
  • described herein is a method of treating idopathic pulmonary fibrosis in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • described herein is a method of controlling an abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in a tissue of a mammal comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • the abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in the tissue results in fibrosis.
  • described herein is a method for the treatment or prevention of scleroderma in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • described herein is a method for the treatment or prevention of cutaneous scleroderma in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • described herein is a method for the treatment or prevention of systemic scleroderma in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • described herein is a method for reducing undesired or abnormal dermal thickening in a mammal comprising administering to mammal in need thereof Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof.
  • the dermal thickening is associated with scleroderma.
  • described herein is a method of controlling an abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in dermal tissues of a mammal comprising administering to mammal in need thereof Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof.
  • the abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in the dermal tissues results in dermal fibrosis.
  • described herein is a method of reducing hydroxyproline content in dermal tissues of a mammal with cutaneous fibrosis comprising administering to mammal in need thereof Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof.
  • described herein is a method for the treatment or prevention of Raynaud's phenomenom in a mammal comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
  • the pharmaceutical composition is administered daily to the mammal. In some embodiments, the pharmaceutical composition is administered once-daily to the mammal. In some embodiments, the pharmaceutical composition is administered twice-daily to the mammal.
  • the mammal is a human.
  • the mammal in any of the method of treatments involving a mammal, is administered one or more additional therapeutically active agents in addition to Compound 1 , or a pharmaceutically acceptable salt thereof.
  • the mammal in any of the method of treatments involving a mammal, is administered one or more additional therapeutically active agents selected from: corticosteroids, immunosuppresant, analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchodilators, leukotriene receptor antagonists, leukotriene formation inhibitors, monoacylglycerol kinase inhibitors, phospholipase Ai inhibitors, phospholipase A 2 inhibitors, and lysophospho lipase D (lysoPLD) inhibitors, autotaxin inhibitors, decongestants, antihistamines, mucolytics, anticholinergics, antitussives, expectorants, and ⁇ -2 agonists.
  • additional therapeutically active agents selected from: corticosteroids, immunosuppresant, analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchod
  • a method comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to a human with a LPA-dependent or LPA-mediated disease or condition.
  • the human is already being administered one or more additional therapeutically active agents other than Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • the method further comprises administering one or more additional therapeutically active agents other than Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • the one or more additional therapeutically active agents other than Compound 1, or a pharmaceutically acceptable salt thereof are selected from: corticosteroids, immunosuppresants, analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchodilators, leukotriene receptor antagonists, leukotriene formation inhibitors, monoacylglycerol kinase inhibitors, phospholipase Ai inhibitors, phospholipase A 2 inhibitors, and lysophospholipase D (lysoPLD) inhibitors, autotaxin inhibitors, decongestants, antihistamines, mucolytics, anticholinergics, antitussives, expectorants, and ⁇ -2 agonists.
  • the LPA receptor is selected from LPAi, LPA 2 , LPA 3 , LPA 4 , LPA 5 and LPA 6 .
  • the LPA receptor is LPAi or LPA or LPA 3 .
  • the disease or condition is any of the diseases or conditions specified herein.
  • a method of inhibiting the physiological activity of LPA in a mammal comprising administering a therapuetically effective amount of a compound of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
  • a method for treating or preventing a LPA-dependent or LPA-mediated disease or condition in a mammal comprising administering a therapuetically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • LPA-dependent or LPA-mediated diseases or conditions include, but are not limited to, fibrosis of organs or tissues, scarring, liver diseases, dermatological conditions, cancer, cardiovascular disease, respiratory diseases or conditions, inflammatory disease, gastrointestinal tract disease, renal disease, urinary tract-associated disease, inflammatory disease of lower urinary tract, dysuria, frequent urination, pancreas disease, arterial obstruction, cerebral infarction, cerebral hemorrhage, pain, peripheral neuropathy, and fibromyalgia.
  • the LPA-dependent or LPA-mediated disease or condition is selected from idiopathic pulmonary fibrosis; other diffuse parenchymal lung diseases of different etiologies including iatrogenic drug-induced fibrosis, occupational and/or environmental induced fibrosis, granulomatous diseases (sarcoidosis, hypersensitivity pneumonia), collagen vascular disease, alveolar proteinosis, langerhans cell granulomatosis, lymphangioleiomyomatosis, inherited diseases (Hermansky-Pudlak Syndrome, tuberous sclerosis, neurofibromatosis, metabolic storage disorders, familial interstitial lung disease); radiation induced fibrosis; chronic obstructive pulmonary disease (COPD); scleroderma; bleomycin induced pulmonary fibrosis; chronic asthma; silicosis; asbestos induced pulmonary fibrosis; acute respiratory distress syndrome (ARDS); kidney fibrosis;
  • COPD chronic obstructive pulmonary disease
  • tubulointerstitium fibrosis tubulointerstitium fibrosis; glomerular nephritis; focal segmental glomerular sclerosis; IgA nephropathy; hypertension; Alport; gut fibrosis; liver fibrosis; cirrhosis; alcohol induced liver fibrosis; toxic/drug induced liver fibrosis; hemochromatosis; nonalcoholic steatohepatitis (NASH); biliary duct injury; primary biliary cirrhosis; infection induced liver fibrosis; viral induced liver fibrosis; and autoimmune hepatitis; corneal scarring; hypertrophic scarring; Duputren disease, keloids, cutaneous fibrosis; cutaneous scleroderma; spinal cord injury/fibrosis; myelofibrosis;
  • NASH nonalcoholic steatohepatitis
  • vascular restenosis atherosclerosis; arteriosclerosis; Wegener's granulomatosis; Peyronie's disease, chronic lymphocytic leukemia, tumor metastasis, transplant organ rejection, endometreosis, neonatal respiratory distress syndrome and neuropathic pain.
  • a method for treating or preventing cancer in a mammal comprising administering a therapeutically effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
  • a pharmaceutically acceptable salt thereof e.g. Compound 2
  • a method for treating or preventing fibrosis in a mammal comprising administering a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
  • a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • a method for the treatment or prevention of organ fibrosis in a mammal comprising administering a therapeutically effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to a mammal in need thereof.
  • the organ fibrosis comprises lung fibrosis, renal fibrosis, or hepatic fibrosis.
  • a method of improving lung function in a mammal comprising administering a therapeutically effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
  • the mammal has been diagnosed as having lung fibrosis.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat idiopathic pulmonary fibrosis (usual interstitial pneumonia) in a mammal.
  • Compound 2 is used to treat idiopathic pulmonary fibrosis (usual interstitial pneumonia) in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat Raynaud's phenomenon.
  • Raynaud's phenomenon comprises both Raynaud's disease (where the phenomenon is idiopathic) and Raynaud's syndrome, where it is caused by some other instigating factor.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g.
  • Compound 2 is used to treat diffuse parenchymal interstitial lung diseases in mammal: iatrogenic drug induced, occupational/environmental (Farmer lung), granulomatous diseases (sarcoidosis, hypersensitivity pneumonia), collagen vascular disease (scleroderma and others), alveolar proteinosis, langerhans cell granulonmatosis, lymphangioleiomyomatosis, Hermansky-Pudlak Syndrome, Tuberous sclerosis, neurofibromatosis, metabolic storage disorders, familial interstitial lung disease.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat post-transplant fibrosis associated with chronic rejection in a mammal (e.g. Bronchiolitis obliterans for lung transplant).
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat cutaneous fibrosis in a mammal (e.g. cutaneous scleroderma, Dupuytren disease, keloids).
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat hepatic fibrosis with or without cirrhosis in a mammal: toxic/drug induced (hemochromatosis), alcoholic liver disease, viral hepatitis (hepatitis B virus, hepatitis C virus, HCV), nonalcoholic liver disease (NASH), metabolic and auto-immune.
  • toxic/drug induced hemochromatosis
  • alcoholic liver disease alcoholic liver disease
  • viral hepatitis hepatitis B virus, hepatitis C virus, HCV
  • NASH nonalcoholic liver disease
  • metabolic and auto-immune is used to treat hepatic fibrosis with or without cirrhosis in a mammal: toxic/drug induced (hemochromatosis), alcoholic liver disease, viral hepatitis (hepatitis B virus, hepatitis C virus, HCV), nonalcoholic liver disease (NASH), metabolic and auto-immune.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat renal fibrosis in a mammal: tubulointerstitium fibrosis, glomerular sclerosis.
  • any of the aforementioned aspects involving the treatment of LPA dependent diseases or conditions are further embodiments comprising administering at least one additional agent in addition to the administration of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • each agent is administered in any order, including simultaneously.
  • the mammal is a human.
  • compounds provided herein are administered to a human. In some embodiments, compounds provided herein are orally administered to a human.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g.
  • Compound 2 is used for inhibiting the activity of at least one LPA receptor or for the treatment of a disease or condition that would benefit from inhibition of the activity of at least one LPA receptor.
  • the LPA receptor is LPA ⁇
  • Compound 1, or a pharmaceutically acceptable salt thereof is used for the formulation of a medicament for the inhibition of LPAi activity.
  • an article of manufacture comprising multiple unit doses of an oral solid dosage form pharmaceutical composition described herein in a high-density polyethylene (HDPE) bottle equipped with a high-density polyethylene (HDPE) cap.
  • HDPE high-density polyethylene
  • high-density polyethylene (HDPE) bottle further comprises an aluminum foil induction seal and silica gel desiccant.
  • any of the aforementioned embodiments are further embodiments comprising single administrations of the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), including further embodiments in which Compound 1, or a
  • pharmaceutically acceptable salt thereof (e.g. Compound 2) is (i) administered once-a-day; (ii) is administered twice-a-day; or (iii) is administered multiple times over the span of one day.
  • any of the aforementioned embodiments are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the time between multiple administrations is every 8 hours; (iv) the time between multiple administrations is every 12 hours.
  • the pharmaceutical composition is administered daily to the mammal.
  • the pharmaceutical composition is administered in treatment cycles comprising: (a) a first period during which Compound 2 is administered daily to the mammal; and (b) a second period during which the Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered to the mammal in a reduced amount as compared to (a) or not administered.
  • treatment cycles comprising: (a) a first period during which Compound 2 is administered daily to the mammal; and (b) a second period during which the Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered to the mammal in a reduced amount as compared to (a) or not administered.
  • the methods of treatment or prevention disclosed herein comprise a drug holiday, wherein the administration of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is temporarily suspended or the dose being administered is temporarily reduced; at the end of the drug holiday dosing is resumed.
  • the length of the drug holiday varies from 2 days to 1 year.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) for treating any of the diseases or conditions disclosed herein.
  • Compound 1 is crystalline.
  • Compound 2 is crystalline.
  • Compound 2 is amorphous.
  • a pharmaceutical composition comprising Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) for use in any of the uses and methods disclosed herein.
  • the pharmaceutically acceptable salt of Compound 1 is the sodium salt (Compound 2).
  • a process for preparing crystalline ⁇ 4'-[3-methyl-4-((R)-l- phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid comprising isolating Compound 1 from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, or nitromethane.
  • R is Ci-C 6 alkyl
  • B is a boronic acid or boronate ester
  • B is a boronic acid or boronate ester
  • the coupling catalyst is a palladium catalyst.
  • the palladium catalyst is tetrakis(triphenylphosphine)palladium or (1,1 '-bis(diphenylphosphino)ferrocene)-dichloropalladium(II).
  • R is -CH 3 or -CH 2 CH 3 .
  • the suitable base is triethylamine, diisopropylethylamine, 1,2,2,6,6- pentamethylpiperidine, tributylamine, sodium bicarbonate, Na 2 C03, K2CO 3 , CS2CO 3 , NaOAc, KOAc, Na 3 P0 4 or K 3 P0 4 .
  • the suitable solvent is tetrahydrofuran, dioxane, water, or combinations thereof.
  • X is selected from CI, Br, I, -OS0 2 CF 3 , -OS0 2 (4-methylphenyl), - OS0 2 (phenyl) and -OS0 2 CH 3 . , or
  • step (1) further comprises isolating the compound of Formula VI prior to step (2).
  • step (1) further comprises a purification step for reducing the amount of palladium to less than 20ppm.
  • the purification step for reducing the amount of palladium comprises treatment of the compound of Formula VI with thiol derivatized silica gel.
  • the reaction of step (1) is heated to a temperature greater than 50°C.
  • step (2) comprises treatment of the compound of Formula VI with sodium hydroxide in a suitable solvent followed by a pH adjustment.
  • a process for the preparation of Compound 1 comprising the steps of: (1) treatment of a compound of Formula XII with diphenylphosphoryl azide in the presence of (R)-(+)- 1 -phenylethanol:
  • R is C1-C6 alkyl
  • step (2) comprises treatment of the compound of Formula VI with sodium hydroxide in a suitable solvent followed by a pH adjustment
  • the disclosed processes provide for the synthesis of Compound 1 and pharmaceutically acceptable salts thereof (e.g. Compound 2).
  • the processes disclosed herein are particularly applicable to large scale chemical production of Compound 1 and pharmaceutically acceptable salts thereof.
  • Compound 1, or a pharmaceutically acceptable salt thereof is replaced with: a) Compound 1 , or a pharmaceutically acceptable salt thereof, of lower chiral purity; b) (4'-[3-methyl-4-((S)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid, or a pharmaceutically acceptable salt thereof of any optical purity; or c) racemic ⁇ 4'-[3-methyl- 4-(l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid, or a pharmaceutically acceptable salt thereof.
  • amorphous Compound 1 is used.
  • crystalline Compound 1 is used.
  • amorphous Compound 2 is used.
  • crystalline Compound 2 is used.
  • Compound 1, or a pharmaceutically acceptable salt thereof is replaced with an active metabolite of Compound 1.
  • the active metabolite is in a crystalline form.
  • the active metabolite is in an amorphous phase.
  • Compound 1, or a pharmaceutically acceptable salt thereof is replaced with a prodrug of Compound 1, or a deuterated analog of Compound 1, or a
  • FIGURE 1 illustrates the XRPD of Pattern 1 of Compound 1.
  • FIGURE 2 illustrates the XRPD of Pattern 1 of Compound 2.
  • FIGURE 3 illustrates the XRPD of Pattern 2 of Compound 2.
  • FIGURE 4 illustrates the TGA and DSC of Crystalline Compound 1.
  • FIGURE 5 illustrates the VT-XRPD Diffractograms of crystalline Compound 1.
  • FIGURE 6 illustrates the cycling DSC thermogram of Crystalline Compound 1.
  • FIGURE 7 illustrates the TGA and DSC of Crystalline Compound 2.
  • FIGURE 8 illustrates the results of Compound 1 on dermal thickness in a mouse model of bleomycin-induced scleroderma.
  • FIGURE 9 illustrates the results of Compound 1 on collagen content in a mouse model of bleomycin-induced scleroderma.
  • Lysophospholipids such as lysophosphatidic acid (LP A)
  • LP A lysophosphatidic acid
  • LPA acts through sets of specific G protein-coupled receptors (GPCRs) in an autocrine and paracrine fashion. LPA binding to its cognate GPCRs (LPAi, LPA 2 , LPA 3 , LPA 4 , LPA 5 , LPA 6 ) activates intracellular signaling pathways to produce a variety of biological responses.
  • GPCRs G protein-coupled receptors
  • LPA has a role as a biological effector molecule, and has a diverse range of physiological actions such as, but not limited to, effects on blood pressure, platelet activation, and smooth muscle contraction, and a variety of cellular effects, which include cell growth, cell rounding, neurite retraction, and actin stress fiber formation and cell migration.
  • the effects of LPA are predominantly receptor mediated.
  • LPA receptors Activation of the LPA receptors with LPA mediates a range of downstream signaling cascades.
  • the actual pathway and realized end point are dependent on a range of variables that include receptor usage, cell type, expression level of a receptor or signaling protein, and LPA concentration.
  • LPAi, LPA 2 , and LPA 3 share high amino acid sequence similarity.
  • LPA regulates many important functions of fibroblasts in wound healing, including proliferation, migration, differentiation and contraction. Fibroblast proliferation is required in wound healing in order to fill an open wound. In contrast, fibrosis is characterized by intense proliferation and accumulation of myofibroblasts that actively synthesize ECM and proinflammatory cytokines. LPA can either increase or suppress the proliferation of cell types important in wound healing.
  • Tissue injury initiates a complex series of host wound-healing responses; if successful, these responses restore normal tissue structure and function. If not, these responses can lead to tissue fibrosis and loss of function.
  • CTGF connective tissue growth factor
  • LPA and LPAi play key pathogenic roles in pulmonary fibrosis. Fibroblast chemoattractant activity plays an important role in the lungs in patients with pulmonary fibrosis. Profibrotic effects of LPAi-receptor stimulation is explained by LPAi -receptor-mediated vascular leakage and increased fibroblast recruitment, both profibrotic events.
  • the LP A- LPAi pathway has a role in mediating fibroblast migration and vascular leakage in IPF. The end result is the aberrant healing process that characterises this fibrotic condition.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat pulmonary fibrosis in a mammal.
  • the LPA-LPA2 pathway contributes to the activation of the TGF- ⁇ pathway in pulmonary fibrosis.
  • compounds that inhibit LPA2 show efficacy in the treatment of lung fibrosis.
  • compounds that inhibit both LPAI and LPA2 show improved efficacy in the treatment of lung fibrosis compared to compounds which inhibit only LPAI or LPA2.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • LPA and LPAi are involved in the etiology of kidney fibrosis.
  • LPAi LPAi receptor
  • UUO animal model of renal fibrosis
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat kidney fibrosis in a mammal.
  • LPA is implicated in liver disease and fibrosis. Plasma LPA levels and serum autotoxin are elevated in hepatitis patients and animal models of liver injury in correlation with increased fibrosis. LPA also regulates liver cell function. LPAi and LPA 2 receptors are expressed by mouse hepatic stellate cells and LPA stimulates migration of hepatic myofibroblasts.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • LPA is in involved in wound healing in the eye.
  • LPAi and LP A3 receptors are detectable in the normal rabbit corneal epithelial cells, keratocytes and endothelial cells and LPAi and LPA 3 expression are increased in corneal epithelial cells following injury.
  • LPA is present in the aqueous humor and the lacrimal gland fluid of the rabbit eye and these levels are increased in a rabbit corneal injury model.
  • LPA induces actin stress fiber formation in rabbit corneal endothelial and epithelial cells and promotes contraction corneal fibroblasts. LPA also stimulates proliferation of human retinal pigmented epithelial cells.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat fibrosis involving tissues in the eye of a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat ocular fibrosis in a mammal.
  • LPA is implicated in myocardial infarction and cardiac fibrosis. Serum LPA levels are increased in patients following mycocardial infarction (MI) and LPA stimulates proliferation and collagen production (fibrosis) by rat cardiac fibroblasts. Both LPAI and LP A3 receptors are highly expressed in human heart tissue.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2 is used to treat cardiac fibrosis in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat or prevent fibrosis in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • compositions are used to treat or prevent fibrosis of an organ or tissue in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat or prevent fibrosis as described herein.
  • fibrosis refers to conditions that are associated with the abnormal accumulation of cells and/or fibronectin and/or collagen and/or increased fibroblast recruitment and include but are not limited to fibrosis of individual organs or tissues such as the heart, kidney, liver, joints, lung, pleural tissue, peritoneal tissue, skin, cornea, retina, musculoskeletal and digestive tract.
  • Exemplary diseases, disorders, or conditions that involve fibrosis include, but are not limited to: Lung diseases associated with fibrosis, e.g., idiopathic pulmonary fibrosis, pulmonary fibrosis secondary to systemic inflammatory disease such as rheumatoid arthritis, scleroderma, lupus, cryptogenic fibrosing alveolitis, radiation induced fibrosis, chronic obstructive pulmonary disease (COPD), chronic asthma, silicosis, asbestos induced pulmonary or pleural fibrosis, acute lung injury and acute respiratory distress (including bacterial pneumonia induced, trauma induced, viral pneumonia induced, ventilator induced, non-pulmonary sepsis induced, and aspiration induced); Chronic nephropathies associated with injury/fibrosis (kidney fibrosis), e.g., glomerulonephritis secondary to systemic inflammatory diseases such as lupus and scleroderma, diabetes, glomerular
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat skin fibrosis in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat peritoneal fibrosis in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat spinal cord injury in a mammal.
  • a mammal suffering from one of the following non-limiting exemplary diseases, disorders, or conditions will benefit from therapy with Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2): atherosclerosis, thrombosis, heart disease, vasculitis, formation of scar tissue, restenosis, phlobitis, COPD (chronic obstructive pulmonary disease), pulmonary hypertension, pulmonary fibrosis, pulmonary inflammation, bowel adhesions, bladder fibrosis and cystitis, fibrosis of the nasal passages, sinusitis, inflammation mediated by neutrophils, and fibrosis mediated by fibroblasts.
  • Compound 1 e.g. Compound 2
  • atherosclerosis thrombosis
  • heart disease vasculitis
  • formation of scar tissue e.g. Compound 2
  • COPD chronic obstructive pulmonary disease
  • pulmonary hypertension pulmonary fibrosis
  • pulmonary inflammation pulmonary inflammation
  • bowel adhesions
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat a dermatological disorders in a mammal.
  • Dermatological disorders include, but are not limited to, proliferative or inflammatory disorders of the skin such as, atopic dermatitis, bullous disorders, collagenoses, psoriasis, psoriatic lesions, scleroderma, dermatitis, contact dermatitis, eczema, urticaria, rosacea, scleroderma, wound healing, scarring, hypertrophic scarring, keloids, Kawasaki Disease, rosacea, Sjogren-Larsso Syndrome, urticaria.
  • LPA is released following tissue injury. LPAi plays a role in the initiation of neuropathic pain.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • the pain is acute pain or chronic pain.
  • the pain is neuropathic pain.
  • the pain is cancer pain.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Lysophospholipid receptor signaling plays a role in the etiology of cancer.
  • Lysophosphatidic acid (LPA) and its G protein-coupled receptors (GPCRs) LPAi, LPA 2 , and/or LPA 3 play a role in the development of several types of cancers.
  • LPA contributes to tumorigenesis by increasing motility and invasiveness of cells.
  • LPA has been implicated in the initiation or progression of ovarian cancer.
  • LPA is present at significant concentrations (2-80 ⁇ ) in the ascitic fluid of ovarian cancer patients.
  • LPA receptors (LPA2 and LPA3) are also overexpressed in ovarian cancer cells as compared to normal ovarian surface epithelial cells.
  • LPA has also been implicated in the initiation or progression of prostate cancer, breast cancer, melanoma, head and neck cancer, bowel cancer (colorectal cancer), thyroid cancer, glioblastoma, and other cancers.
  • LPA receptors mediate both migration of and invasion by pancreatic cancer cell lines:
  • Ki 16425 and LPA specific siRNA effectively blocked in vitro migration in response to LPA and peritoneal fluid (ascites) from pancreatic cancer patients; in addition, Ki 16425 blocked the LPA- induced and ascites-induced invasion activity of a highly peritoneal metastatic pancreatic cancer cell line (Yamada et al, J. Biol. Chem., 279, 6595-6605, 2004).
  • Colorectal carcinoma cell lines show significant expression of LPAi mRNA and respond to LPA by cell migration and production of angiogenic factors. Overexpression of LPA receptors has a role in the pathogenesis of thyroid cancer.
  • LP A3 was originally cloned from prostate cancer cells, concordant with the ability of LPA to induce autocrine proliferation of prostate cancer cells.
  • LPA has stimulatory roles in cancer progression in many types of cancer.
  • LPA is produced from and induces proliferation of prostate cancer cell lines.
  • LPA induces human colon carcinoma DLD 1 cell proliferation, migration, adhesion, and secretion of angiogenic factors through LPAi signalling.
  • LPA enhances cell proliferation and secretion of angiogenic factors.
  • LPA 2 and LPA 3 receptor activation results in proliferation of the cells.
  • LPAi is implicated in bone metastasis (Boucharaba et al, Proc. Natl. Acad. Sci USA , 103, 9643-9648, 2006).
  • Compound 1 is used in the treatment of cancer in a mammal.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of cancer in a mammal.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of cancer in a mammal.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of cancer in a mammal.
  • Compound 1 , or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Compound 1 is used in the treatment of malignant and benign proliferative disease in a mammal.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to prevent or reduce proliferation of tumor cells, invasion and metastasis of carcinomas, pleural mesothelioma or peritoneal mesothelioma, cancer pain, bone metastases.
  • a method of treating cancer in a mammal comprising administering to the mammal Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), and a second therapeutic agent, wherein the second therapeutic agent is an anticancer agent.
  • radiation therapy is also used.
  • the types of cancer include, but is not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias) at any stage of the disease with or without metastases.
  • solid tumors such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias) at any stage of the disease with or without metastases.
  • cancers include, acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, anal cancer, appendix cancer, astrocytomas, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (osteosarcoma and malignant fibrous histiocytoma), brain stem glioma, brain tumors, brain and spinal cord tumors, breast cancer, bronchial tumors, Burkitt lymphoma, cervical cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-Cell lymphoma, embryonal tumors, endometrial cancer, ependymoblastoma, ependymoma, esophageal cancer, ewing sarcoma family of tumors, eye cancer, retinoblastoma, gallbladder cancer
  • neuroblastoma non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma, malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, papillomatosis, parathyroid cancer, penile cancer, pharyngeal cancer, pineal parenchymal tumors of intermediate differentiation, pineoblastoma and supratentorial primitive neuroectodermal tumors, pituitary tumor, plasma cell neoplasm/multiple myeloma, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell (kidney) cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, Ewing sarcoma family of tumors, sarcoma, kaposi
  • Compound 1, or a pharmaceutically acceptable salt thereof is used in the treatment of breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, leukemia, lymphoma or tumor metastasis in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used in the treatment of breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, Hodgkin lymphoma, and follicular lymphoma, multiple myeloma, chronic lymphocytic leukemia, or tumor metastasis in a mammal.
  • LPA is a contributor to the pathogenesis of respiratory diseases.
  • LPA lipoprotein kinase
  • IL-8 is found in increased concentrations in BAL fluids from patients with asthma, chronic obstructive lung disease, pulmonary sarcoidosis and acute respiratory distress syndrome and 11-8 has been shown to exacerbate airway inflammation and airway remodeling of asthmatics.
  • LPA1, LPA2 and LP A3 receptors have all been shown to contribute to the LPA-induced IL-8 production.
  • LPA also induces histamine release from mouse and rat mast cells.
  • the effects of LPA are mediated through LPAi and/or LPA 3 .
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g.
  • Compound 2 is used in the treatment of asthma in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • Respiratory disease refers to diseases affecting the organs that are involved in breathing, such as the nose, throat, larynx, eustachian tubes, trachea, bronchi, lungs, related muscles (e.g., diaphram and intercostals), and nerves.
  • Respiratory diseases include, but are not limited to, asthma, adult respiratory distress syndrome and allergic (extrinsic) asthma, non- allergic (intrinsic) asthma, acute severe asthma, chronic asthma, clinical asthma, nocturnal asthma, allergen-induced asthma, aspirin-sensitive asthma, exercise-induced asthma, isocapnic
  • chronic obstructive pulmonary disease includes, but is not limited to, chronic bronchitis or emphysema, pulmonary hypertension, interstitial lung fibrosis and/or airway inflammation, and cystic fibrosis.
  • the nervous system is a major locus for LPAi expression.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), for use in the treatment or prevention of a nervous system disorder in a mammal.
  • the term "nervous system disorder,” as used herein includes, but is not limited to, Alzheimer's Disease, cerebral edema, cerebral ischemia, stroke, multiple sclerosis, neuropathies, Parkinson's Disease, multiple sclerosis, retinal ischemia, post-surgical cognitive dysfunction, migraine, peripheral neuropathy/neuropathic pain, spinal cord injury, cerebral edema and head injury.
  • Angiogenesis the formation of new capillary networks from pre-existing vasculature, is normally invoked in wound healing, tissue growth and myocardial angiogenesis after ischemic injury.
  • Peptide growth factors and lysophospholipids control coordinated proliferation, migration, adhesion, differentiation and assembly of vascular endothelial cells (VECs) and surrounding vascular smooth-muscle cells (VSMCs).
  • VECs vascular endothelial cells
  • VSMCs vascular smooth-muscle cells
  • dysregulation of the processes mediating angiogenesis leads to atherosclerosis, hypertension, tumor growth, rheumatoid arthritis and diabetic retinopathy.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat or prevent cardiovascular disease in mammal, including but not limited to:
  • arrhythmia atrial or ventricular or both
  • atherosclerosis and its sequelae angina; cardiac rhythm disturbances; myocardial ischemia; myocardial infarction; cardiac or vascular aneurysm; vasculitis, stroke; peripheral obstructive arteriopathy of a limb, an organ, or a tissue; reperfusion injury following ischemia of the brain, heart, kidney or other organ or tissue; endotoxic, surgical, or traumatic shock; hypertension, valvular heart disease, heart failure, abnormal blood pressure; shock; vasoconstriction (including that associated with migraines); vascular abnormality, inflammation, insufficiency limited to a single organ or tissue.
  • kits for preventing or treating vasoconstriction, atherosclerosis and its sequelae myocardial ischemia, myocardial infarction, aortic aneurysm, vasculitis and stroke comprising administering at least once to the mammal an effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • the LPA antagonist disclosed herein is used to treat Raynaud's phenomenon.
  • Raynaud's phenomenon comprises both Raynaud's disease (where the phenomenon is idiopathic) and Raynaud's syndrome, where it is caused by some other instigating factor.
  • kits for reducing cardiac reperfusion injury following myocardial ischemia and/or endotoxic shock comprising administering at least once to the mammal an effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) ⁇
  • kits for reducing the constriction of blood vessels in a mammal comprising administering at least once to the mammal an effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • LPA is associated with various inflammatory/immune diseases.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • antagonists of LPA i and/or LP A 3 find use in the treatment or prevention of inflammatory/immune disorders in a mammal.
  • inflammatory/immune disorders include psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, asthma, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, eczema, allogeneic or xenogeneic transplantation (organ, bone marrow, stem cells and other cells and tissues) graft rejection, graft- versus-host disease, lupus erythematosus, inflammatory disease, type I diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis (e.g., Hashimoto's and autoimmune thyroiditis), myasthenia gravis, autoimmune hemolytic anemia, multiple sclerosis, cystic fibrosis, chronic relapsing hepatitis, primary biliary cirrhosis, allergic conjun
  • [00194] are methods for treating, preventing, reversing, halting or slowing the progression of LPA-dependent or LPA-mediated diseases or conditions once it becomes clinically evident, or treating the symptoms associated with or related to LPA-dependent or LPA- mediated diseases or conditions, by administering to the mammal Compound 1 , or a
  • the subject already has a LPA-dependent or LPA-mediated disease or condition at the time of administration, or is at risk of developing a LPA-dependent or LPA-mediated disease or condition.
  • eosinophil and/or basophil and/or dendritic cell and/or neutrophil and/or monocyte and/or T-cell recruitment comprising administering at least once to the mammal an effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • cystitis including, e.g., interstitial cystitis, comprising administering at least once to the mammal a therapeutically effective amount of
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • methods described herein include the diagnosis or determination of whether or not a patient is suffering from a LPA-dependent or LPA-mediated disease or condition by administering to the subject a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), and determining whether or not the patient responds to the treatment.
  • a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • LPA-dependent conditions or diseases include those wherein an absolute or relative excess of LPA is present and/or observed.
  • the LPA-dependent or LPA-mediated diseases or conditions include, but are not limited to, organ fibrosis, tissue fibrosis, asthma, allergic disorders, chronic obstructive pulmonary disease, pulmonary hypertension, lung or pleural fibrosis, peritoneal fibrosis, arthritis, allergy, cancer, cardiovascular disease, aldult respiratory distress syndrome, myocardial infarction, aneurysm, stroke, and cancer.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to improve the corneal sensitivity decrease caused by corneal operations such as laser- assisted in situ keratomileusis (LASIK) or cataract operation, corneal sensitivity decrease caused by corneal degeneration, and dry eye symptom caused thereby.
  • corneal operations such as laser- assisted in situ keratomileusis (LASIK) or cataract operation
  • corneal sensitivity decrease caused by corneal degeneration e.g. Compound 2
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of ocular inflammation and allergic conjunctivitis, vernal keratoconjunctivitis, and papillary conjunctivitis in a mammal.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of Sjogren disease or inflammatory disease with dry eyes in a mammal.
  • LPA and LPA receptors are involved in the pathogenesis of osteoarthritis.
  • LPAi LPA receptors
  • presented herein is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of osteoarthritis in a mammal.
  • LPA receptors e.g. LPAi, LPA 3
  • LPAi LPAi
  • LPA 3 LPA 3
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of rheumatoid arthritis in a mammal.
  • LPA receptors contribute to adipogenesis.
  • LPAi LPA receptors
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the promotion of adipose tissue formation in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat Raynaud's phenomenon in a mammal.
  • Raynaud's phenomenon comprises both Raynaud's disease (where the phenomenon is idiopathic) and Raynaud's syndrome, where it is caused by some instigating factor.
  • compositions, pharmaceutical compositions, methods for treating, methods for formulating, methods for producing, methods for manufacturing, treatment strategies, pharmacokinetic strategies using Compound 1 , or pharmaceutically acceptable salts thereof are Described herein are compositions, pharmaceutical compositions, methods for treating, methods for formulating, methods for producing, methods for manufacturing, treatment strategies, pharmacokinetic strategies using Compound 1 , or pharmaceutically acceptable salts thereof.
  • Compound 1 is substantially free of the S-isomer.
  • substantially free with respect to an enantiomer, means that the referenced enantiomer is not present or there is less than 5%, less than 4%, less than 3%, less than 2% or less than 1% of the referenced enantiomer.
  • samples of Compound 1 were found to have greater than 85% e.e., greater than 90% e.e., greater than 91% e.e., greater than 92% e.e., greater than 93% e.e., greater than 94% e.e., greater than 95% e.e., greater than 96% e.e., greater than 97% e.e., greater than 98%) e.e., or greater than 99%> e.e.
  • Compound 2 or " ⁇ 4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl ⁇ -acetic acid, sodium salt” or "sodium ⁇ 4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetate” or any other similar name refers to the compound with the following structure:
  • Compound 2 is substantially free of the S-isomer.
  • samples of Compound 2 were found to have greater than 85% e.e., greater than 90% e.e., greater than 91% e.e., greater than 92% e.e., greater than 93 %> e.e., greater than 94% e.e., greater than 95% e.e., greater than 96% e.e., greater than 97% e.e., greater than 98%) e.e., or greater than 99% e.e.
  • a wide variety of pharmaceutically acceptable salts are formed from Compound 1 and include:
  • a metal ion such as for example, an alkali metal ion (e.g. lithium, sodium, potassium), an alkaline earth ion (e.g. magnesium, or calcium), or an aluminum ion, or is replaced by an ammonium cation ( H 4 + ).
  • - salts formed by reacting Compound 1 with a pharmaceutically acceptable organic base which includes alkylamines, such as choline, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine, and salts with amino acids, such as arginine, lysine, and the like.
  • a pharmaceutically acceptable organic base which includes alkylamines, such as choline, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine, and salts with amino acids, such as arginine, lysine, and the like.
  • Salts of Compound 1 include, but are not limited to, a lithium salt, a sodium salt, a potassium salt, a magnesium salt, a calcium salt, an aluminum salt, an ammonium salt, a choline, an ethanolamine salt, a diethanolamine salt, a triethanolamine salt, a tromethamine salt, a N- methylglucamine salt, a dicyclohexylamine salt, a tris(hydroxymethyl)methylamine salt, an arginine salt, or a lysine salt.
  • Compound 1 is treated with an amino acid to form a salt.
  • Compound 1 is treated with choline, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, arginine, lysine, ammonium hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like to form a salt.
  • compositions which do not cause significant irritation to a mammal to which it is administered and does not substantially abrogate the biological activity and properties of the compound.
  • a reference to a pharmaceutically acceptable salt includes the solvent addition forms (solvates).
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are formed during the process of product formation or isolation with pharmaceutically acceptable solvents such as water, ethanol, methyl tert-butyl ether, isopropanol, acetonitrile, heptane, and the like.
  • solvents such as water, ethanol, methyl tert-butyl ether, isopropanol, acetonitrile, heptane, and the like.
  • solvates are formed using, but not limited to, Class 3 solvent(s).
  • solvates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol.
  • solvates of Compound 1 , or salts thereof are conveniently prepared or formed during the processes described herein.
  • Compound 1, or salts thereof exist in unsolvated form.
  • Compound 1, or a pharmaceutically acceptable salt thereof is prepared in various forms, including but not limited to, amorphous phase, milled forms and nano-particulate forms.
  • Compound 1 is amorphous. In some embodiments, Amorphous Phase of Compound 1 has an XRPD pattern showing a lack of crystallinity.
  • Compound 1 is crystalline. In some embodiments, Compound 1 is crystalline Pattern 1.
  • the crystalline form of Compound 1 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1 ; a differential scanning calorimetry (DSC) thermogram with endotherms at about 157°C and about 198°C; a differential scanning calorimetry (DSC) thermogram
  • the crystalline form of Compound 1 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least four of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least five of the preceding properties. In some
  • the crystalline form of Compound 1 is characterized as having at all of the preceding properties.
  • crystalline Pattern 1 of Compound 1 is characterized as having:
  • Crystalline Pattern 1 of Compound 1 is obtained from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, nitromethane, tetrahydrofuran, dioxane, methyl ethyl ketone, acetone, 2-methoxyethanol, or methanol.
  • Crystalline Pattern 1 of Compound 1 is characterized as having at least two of the properties selected from (a) to (f). In some embodiments, Crystalline Pattern 1 of Compound 1 is characterized as having at least three of the properties selected from (a) to (f). In some embodiments, Crystalline Pattern 1 of Compound 1 is characterized as having at least four of the properties selected from (a) to (f).
  • Compound 2 is amorphous.
  • Amorphous Phase of Compound 2 has an XRPD pattern showing a lack of crystallinity.
  • Compound 2 is crystalline. In some embodiments, Compound 2 is crystalline Pattern 1.
  • crystalline Pattern 1 of Compound 2 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2-Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C; a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7; or combinations thereof.
  • XRPD X-ray powder diffraction
  • the crystalline form of Compound 2 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least four of the preceding properties. In some
  • the crystalline form of Compound 2 is characterized as having at all of the preceding properties.
  • crystalline Pattern 1 of Compound 2 is characterized as having at least one of the following properties:
  • Crystalline Pattern 1 of Compound 2 is characterized as having at least two of the properties selected from (a) to (c). In some embodiments, Crystalline Pattern 1 of Compound 2 is characterized as having properties (a), (b) and (c).
  • Crystalline Pattern 1 of Compound 2 is obtained from isopropanol.
  • Compound 2 is crystalline. In some embodiments, Compound 2 is crystalline Pattern 2. Crystalline Pattern 2 of Compound 2 is characterized as having at least one of the following properties:
  • Crystalline Pattern 2 of Compound 2 is obtained from isopropanol.
  • Compound 1 is prepared as a prodrug.
  • a "prodrug of Compound 1” refers to a compound that is converted into Compound 1 in vivo.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not.
  • the prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • prodrugs facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • An example, without limitation, of a prodrug would be an ester of Compound 1 (the "prodrug").
  • a further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. In certain embodiments, the prodrug of Compound 1 increases the bioavailability of
  • a prodrug of Compound 1 is an alkyl ester of Compound 1, such as, for example, methyl ester, ethyl ester, n-propyl ester, iso-propyl ester, n-butyl ester, sec-butyl ester, or tert-butyl ester.
  • Non- limiting examples of prodrugs of Compound 1 include:
  • sites on Compound 1 are susceptible to various metabolic reactions. Therefore incorporation of appropriate substituents on Compound 1 will reduce, minimize or eliminate this metabolic pathway.
  • the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, deuterium or an alkyl group (e.g. methyl, ethyl).
  • Compound 1 is isotopically labeled (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • Compound 1 is isotopically-labeled, which is identical to Compound 1 but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • one or more hydrogen atoms are replaced with deuterium.
  • metabolic sites on Compound 1 are deuterated.
  • substitution with deuterium affords certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements.
  • each R is independently selected from hydrogen or deutrium
  • the pharmaceutically acceptable salt of the compound is a sodium salt.
  • Compound 1, and pharmaceutically acceptable salts thereof are synthesized as described herein.
  • solvents, temperatures and other reaction conditions presented herein may vary.
  • the starting materials used for the synthesis are either synthesized or obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Fluka, Acros Organics, Alfa Aesar, VWR Scientific, and the like. General methods for the preparation of compounds can be modified by the use of appropriate reagents and conditions for the introduction of the various moieties found in the formulae as provided herein.
  • the synthesis of compound 1 begins with the reaction of an alkyl acetoacetate, such as methyl acetoacetate, with methylamine to provide 3-methylamino-but-2-enoic acid methyl ester, which is then reacted with benzoyl chlorides, such as 4-bromobenzoyl chloride, to provide compound I.
  • X is halide, triflate or any other suitable leaving group for use in a Suzuki coupling reaction.
  • X is -CI, -Br, -I, -OSO 2 CF 3 , -OS0 2 (4-methylphenyl), and - OSO 2 CH 3 .
  • X is a halide.
  • X is -Br.
  • R 1 is an alkyl or benzyl. In some embodiments, R 1 is methyl, ethyl, propyl, or benzyl.
  • Other alkyl acetoacetates contemplated include, ethyl acetoacetate, isopropyl acetoacetate, benzyl acetoacetate.
  • Treatment of compounds of structure I with hydroxyl amine and acetic acid provides isoxazoles of structure II. Hydrolysis of the ester group of isoxazoles of structure II provides carboxylic acid III. Hydrolysis can also be accomplished with the use of other bases, such as sodium hydroxide.
  • a Suzuki reaction between carbamate compound IV and compounds of structure V is used to provide compounds of structure VI.
  • B is boronic embodiments
  • X is , and In some embodiments, .
  • X is -CI, -Br, -I, -OSO 2 CF 3 , -OS0 2 (4-methylphenyl), and -OS0 2 CH 3 .
  • X is a halide.
  • X is -Br.
  • the Suzuki reaction includes the use of a palladium catalyst, a suitable base and a suitable solvent.
  • the palladium catalyst is a phosphine containing palladium catalyst. In some embodiments, the palladium catalyst is Pd(PPh 3 ) 4 or Pd(dppf)Cl 2 .
  • the suitable base for the Suzuki reaction is an inorganic base. In some embodiments, the suitable base for the Suzuki reaction is triethylamine, diisopropylethylamine, 1,2,2,6,6-pentamethylpiperidine, tributylamine, sodium bicarbonate, Na 2 C0 3 , K 2 C0 3 , Cs 2 C0 3 , NaOAc, KOAc, Na 3 P0 4 or K 3 P0 4 . Other metal mediated coupling reactions are known for the preparation of compounds of structure VI.
  • biaryls include, but are not limited to Suzuki reactions, Stille cross couplings, Negishi couplings, Kumada couplings, Ullmann reactions, Hiyama Coupling, and variants thereof (Metal-Catalyzed Cross-Coupling Reactions, Armin de Meijere (Editor), Francois Diederich (Editor), John Wiley & Sons; 2nd edition, 2004; Ozdemir, et al, Tetrahedron, 2005, 61, 9791-9798; Ackermann, et al, Org. Lett., 2006, 8, 3457-3460; Blakey, et al, J. Am. Chem.
  • compounds of structure VII are reacted with a borylating agent using transition metal mediated reaction conditions to form boronate compounds of structure IX.
  • the borylating reaction to form IX includes the use of a palladium catalyst, such as Pd(PPh 3 ) 4 or Pd(dppf)Cl 2 , in the presence of a suitable base, such as potassium acetate.
  • the borylation reagent is selected from among pinacolborane, catecholborane, bis(neopentyl glycolato)diboron, bis(pinacolato)diboron, bis(hexylene glycolato)diboron, and bis(catecholato)diboron. In some embodiments, the borylation reagent is bis(pinacolato)diboron. In some embodiments, the borylation reaction is performed with heating. Boronate compounds of structure VII are reacted with compounds of structure VIII under palladium mediated coupling conditions (Suzuki reaction conditions) to form compounds of structure VI.
  • biphenyl compounds of structure X are elaborated into compound VI as shown in Scheme 4.
  • R is ethyl.
  • R is isopropyl.
  • Biphenyl compounds of structure X are treated with acetyl chloride in the presence of a suitable Lewis acid, follow by conversion of the acetyl group to the carboxylic acid and treatment of the carboxylic acid with thionyl chloride to provide acid chlorides of structure XI. Acid chlorides of structure XI are then used to prepare isoxazoles of structure XII as described in Scheme 1.
  • R 1 is an alkyl group.
  • R 1 is methyl and R 1 is removed from the isoxazoles under hydrolysis conditions to provide compounds of structure XII.
  • R 1 is benzyl and R 1 is removed from the isoxazoles under hydrogenation conditions (e.g. H 2 , Pd/C) to provide compounds of structure XII.
  • H 2 , Pd/C hydrogenation conditions
  • Suitable bases for the hydrolysis include, but are not limited to, lithium hydroxide and sodium hydroxide.
  • Suitable solvents for the hydrolysis include, but are not limited to, water, methanol, ethanol, tetrahydrofuran, or combinations thereof. Compound 1 is then treated with sodium hydroxide in ethanol to furnish Compound 2.
  • Compound 2 is prepared from compound VI by performing a one- step hydrolysis and salt forming reaction.
  • the one-step hydrolysis and salt forming reaction includes treatment of compound VI with sodium hydroxide in a suitable solvent.
  • Compound 1 is treated with potassium hydroxide in a solvent to form Compound 1, potassium salt. In some embodiments, Compound 1 is treated with lithium hydroxide in a solvent to form Compound 1, lithium salt. In some embodiments, Compound 1 is treated with calcium hydroxide in a solvent to form Compound 1, calcium salt.
  • Compound 1 is treated with dicyclohexylamine in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with N-methyl-D-glucamine in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with choline in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with tris(hydroxymethyl)methylamine in a solvent to form the corresponding salt.
  • Compound 1 is treated with arginine in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with lysine in a solvent to form the corresponding salt.
  • purification steps to reduce the amount of palladium in a product includes, but is not limited to, treatment with solid trimercaptotriazine (TMT), polystyrene-bound TMT, mercapto- porous polystyrene-bound TMT, polystyrene-bound ethylenediamine, activated carbon, glass bead sponges, SmopexTM, silica bound scavengers, thiol-derivatized silica gel, N-acetylcysteine, n-Bu 3 P, crystallization, extraction, 1-cysteine, n-Bu3P/lactic acid.
  • TMT solid trimercaptotriazine
  • polystyrene-bound TMT polystyrene-bound TMT
  • mercapto- porous polystyrene-bound TMT polystyrene-bound ethylenediamine
  • activated carbon glass bead sponges
  • SmopexTM silica bound
  • activated carbon includes but is not limited to DARCO ® KB-G, ® KB-WJ.
  • silica bound scavengers include but are not limited to
  • the purification steps to reduce the amount of palladium include the use of activated carbon, derivatized silica gel (e.g. thiol derivatized silica gel), or combinations thereof.
  • GMP Good Manufacturing Practice
  • Preferred solvents are those that are suitable for use in GMP facilities and consistent with industrial safety concerns. Categories of solvents are defined in, for example, the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), "Impurities: Guidelines for Residual Solvents, Q3C(R3), (November 2005).
  • Solvents are categorized into three classes. Class 1 solvents are toxic and are to be avoided. Class 2 solvents are solvents to be limited in use during the manufacture of the therapeutic agent. Class 3 solvents are solvents with low toxic potential and of lower risk to human health. Data for Class 3 solvents indicate that they are less toxic in acute or short-term studies and negative in genotoxicity studies.
  • Class 1 solvents which are to be avoided, include: benzene; carbon tetrachloride; 1,2- dichloroethane; 1 , 1 -dichloroethene; and 1 , 1 , 1 -trichloroethane.
  • Class 2 solvents are: acetonitrile, chlorobenzene, chloroform, cyclohexane, 1,2- dichloroethene, dichloromethane, 1 ,2-dimethoxyethane, ⁇ , ⁇ -dimethylacetamide, N,N- dimethylformamide, 1,4-dioxane, 2-ethoxyethanol, ethyleneglycol, formamide, hexane, methanol, 2- methoxyethanol, methylbutyl ketone, methylcyclohexane, N-methylpyrrolidine, nitromethane, pyridine, sulfolane, tetralin, toluene, 1 , 1 ,2-trichloroethene and xylene.
  • Class 3 solvents which possess low toxicity, include: acetic acid, acetone, anisole, 1- butanol, 2-butanol, butyl acetate, tert-butylmethyl ether (MTBE), cumene, dimethyl sulfoxide, ethanol, ethyl acetate, ethyl ether, ethyl formate, formic acid, heptane, isobutyl acetate, isopropyl acetate, methyl acetate, 3-methyl-l-butanol, methylethyl ketone, methylisobutyl ketone, 2-methyl-l- propanol, pentane, 1-pentanol, 1-propanol, 2-propanol, propyl acetate, and tetrahydrofuran.
  • acetic acid acetone
  • anisole 1- butanol
  • 2-butanol butyl acetate
  • MTBE tert-but
  • Residual solvents in active pharmaceutical ingredients originate from the manufacture of API. In some cases, the solvents are not completely removed by practical manufacturing techniques. Appropriate selection of the solvent for the synthesis of APIs may enhance the yield, or determine characteristics such as crystal form, purity, and solubility. Therefore, the solvent is a critical parameter in the synthetic process.
  • compositions comprising salts of Compound 1 comprise an organic solvent(s). In some embodiments, compositions comprising salts of Compound 1 comprise a residual amount of an organic solvent(s). In some embodiments, compositions comprising salts of Compound 1 comprise a residual amount of a Class 3 solvent. In some embodiments, the organic solvent is a Class 3 solvent.
  • the Class 3 solvent is selected from the group consisting of acetic acid, acetone, anisole, 1 -butanol, 2-butanol, butyl acetate, tert-butylmethyl ether, cumene, dimethyl sulfoxide, ethanol, ethyl acetate, ethyl ether, ethyl formate, formic acid, heptane, isobutyl acetate, isopropyl acetate, methyl acetate, 3 -methyl- 1 -butanol, methyl ethyl ketone, methylisobutyl ketone, 2-methyl- l -propanol, pentane, 1 -pentanol, 1 -propanol, 2-propanol, propyl acetate, and tetrahydrofuran.
  • the Class 3 solvent is selected from ethyl acetate, isopropyl acetate, isopropyl
  • compositions comprising a salt of Compound 1 include a detectable amount of an organic solvent.
  • the salt of Compound 1 is a sodium salt (i.e. Compound 2).
  • the organic solvent is a Class 3 solvent.
  • the salt of Compound 1 is a sodium salt, potassium salt, lithium salt, calcium salt, magnesium salt, ammonium salt, choline salt, protonated dicyclohexylamine salt, protonated N- methyl-D-glucamine salt, protonated tris(hydroxymethyl)methylamine salt, arginine salt, or lysine salt.
  • the salt of Compound 1 is a sodium salt.
  • compositions comprising Compound 2, wherein the composition comprises a detectable amount of solvent that is less than about 1 %, wherein the solvent is selected from acetone, 1 ,2-dimethoxyethane, acetonitrile, ethyl acetate, tetrahydrofuran, methanol, ethanol, heptane, and 2-propanol.
  • the composition comprises a detectable amount of solvent which is less than about 5000 ppm.
  • compositions comprising Compound 2, wherein the detectable amount of solvent is less than about 5000 ppm, less than about 4000 ppm, less than about 3000 ppm, less than about 2000 ppm, less than about 1000 ppm, less than about 500 ppm, or less than about 100 ppm.
  • pharmaceutically acceptable excipient refers to a material, such as a carrier, diluent, stabilizer, dispersing agent, suspending agent, thickening agent, etc. which allows processing the active pharmaceutical ingredient (API) into a form suitable for administration to a mammal.
  • the mammal is a human.
  • Pharmaceutically acceptable excipients refer to materials which do not substantially abrogate the desired biological activity or desired properties of the compound (i.e. API), and is relatively nontoxic, i.e., the material is administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • an API refers to a compound that possesses a desired biological activity or desired properties.
  • an API is Compound 1.
  • an API is Compound 2.
  • an active pharmaceutical ingredient Compound 2 with a purity of greater than 80%>, greater than 85%>, greater than 90%>, greater than 95%>, greater than 96%>, greater than 97%>, greater than 98%, or greater than 99%.
  • the term "pharmaceutical combination” as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients.
  • the term "fixed combination” means that the active ingredients, e.g. Compound 1 or a pharmaceutically acceptable salt, and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non- fixed combination means that the active ingredients, e.g. Compound 1 or a pharmaceutically acceptable salt, and a co-agent, are administered to a patient as separate entities either
  • cocktail therapy e.g. the administration of three or more active ingredients.
  • composition refers to a mixture of Compound 1 , or
  • composition facilitates administration of the compound to a mammal.
  • Administration of a combination of agents includes administration of the agents described in a single composition or in a combination therapy wherein one or more agent is administered separately from at least one other agent.
  • alkyl refers to an aliphatic hydrocarbon group.
  • the alkyl moiety is branched, straight chain, or cyclic.
  • the alkyl group may be designated as "Ci-C 6 alkyl".
  • an alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, ethenyl, propenyl, allyl, butenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • Detectable amount refers to an amount that is measurable using standard analytic methods (e.g. ion chromatography, mass spectrometry, NMR, HPLC, gas chromatography, elemental analysis, IR spectroscopy, inductively coupled plasma atomic emission spectrometry,
  • an “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms.
  • therapeutically effective amount includes, for example, a
  • prophylactically effective amount The effective amount will be selected based on the particular patient and the disease level. It is understood that “an effect amount” or “a therapeutically effective amount” varies from subject to subject, due to variation in metabolism of drug, age, weight, general condition of the subject, the condition being treated, the severity of the condition being treated, and the judgment of the prescribing physician. In one embodiment, an appropriate "effective" amount in any individual case is determined using techniques, such as a dose escalation study
  • co-administration or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • the terms “enhance” or “enhancing,” as used herein, means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
  • a "metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized.
  • active metabolite refers to a biologically active derivative of a compound that is formed when the compound is metabolized (biotransformed).
  • metabolized refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound.
  • cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases (UGT) catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups (e.g. conjugation reactions).
  • UGT uridine diphosphate glucuronyltransferases
  • compounds disclosed herein are metabolite to provide taurine metabolites. Further information on metabolism is available in The Pharmacological Basis of Therapeutics, 9th Edition, McGraw-Hill (1996).
  • metabolites of the compounds disclosed herein are identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
  • module means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
  • modulator refers to a molecule that interacts with a target either directly or indirectly.
  • the interactions include, but are not limited to, the interactions of an agonist and an antagonist.
  • agonist refers to a molecule such as a compound, a drug, an enzyme activator or a hormone modulator that binds to a specific receptor and triggers a response in the cell.
  • An agonist mimics the action of an endogenous ligand (such as prostaglandin, hormone or neurotransmitter) that binds to the same receptor.
  • antagonist refers to a molecule such as a compound, which diminishes, inhibits, or prevents the action of another molecule or the activity of a receptor site.
  • LPA-dependent refers to conditions or disorders that would not occur, or would not occur to the same extent, in the absence of LPA.
  • LPA-mediated refers to refers to conditions or disorders that might occur in the absence of LPA but can occur in the presence of LPA.
  • subject or "patient” encompasses mammals.
  • the mammal is a human.
  • the mammal is a non-human primate such as chimpanzee, and other apes and monkey species.
  • the mammal is a farm animal such as cattle, horse, sheep, goat, or swine.
  • the mammal is a domestic animal such as rabbit, dog, or cat.
  • the mammal is a laboratory animal, including rodents, such as rats, mice and guinea pigs, and the like.
  • Bioavailability refers to the percentage of the weight of Compound 1, or a
  • AUC(o - ⁇ ) The total exposure (AUC(o - ⁇ )) of a drug when administered intravenously is usually defined as 100% Bioavailable (F%).
  • Oral bioavailability refers to the extent to which Compound 1, or a pharmaceutically acceptable salt and/or solvate thereof, is absorbed into the general circulation when the pharmaceutical composition is taken orally as compared to intravenous injection.
  • Blood plasma concentration refers to the concentration Compound 1, in the plasma component of blood of a mammal. It is understood that the plasma concentration of Compound 1 may vary significantly between subjects, due to variability with respect to metabolism and/or interactions with other therapeutic agents. In one aspect, the blood plasma concentration of
  • Compound 1 varies from subject to subject. Likewise, values such as maximum plasma
  • C max concentration or time to reach maximum plasma concentration (T max ), or total area under the plasma concentration time curve (AUC(o- ⁇ )) vary from subject to subject. Due to this variability, in one embodiment, the amount necessary to constitute "a therapeutically effective amount" of Compound 1 varies from subject to subject.
  • Drug absorption typically refers to the process of movement of drug from site of administration of a drug across a barrier into a blood vessel or the site of action, e.g., a drug moving from the gastrointestinal tract into the portal vein or lymphatic system.
  • Plasma concentration or “Plasma concentration” describes the blood serum or blood plasma concentration, typically measured in mg, ⁇ g, or ng of therapeutic agent per ml, dl, or 1 of blood serum, absorbed into the bloodstream after administration. Plasma concentrations are typically measured in ng/ml or ⁇ g/ml.
  • Pharmacokinetics refers to the factors which determine the attainment and maintenance of the appropriate concentration of drug at a site of action.
  • Step state is when the amount of drug administered is equal to the amount of drug eliminated within one dosing interval resulting in a plateau or constant plasma drug exposure.
  • Treatment refers to any treatment of a disorder or disease, such as preventing the disorder or disease from occurring in a subject predisposed to the disorder or disease, but has not yet been diagnosed as having the disorder or disease; inhibiting the disorder or disease, e.g., arresting the development of the disorder or disease, relieving the disorder or disease, causing regression of the disorder or disease, relieving a condition caused by the disease or disorder, or stopping the symptoms of the disease or disorder either prophylactically and/or therapeutically.
  • the term “treat” is used synonymously with the term “prevent.”
  • compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which are used pharmaceutically.
  • suitable techniques, carriers, and excipients include those found within, for example, Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington 's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975;
  • a pharmaceutical composition refers to a mixture of a compound of Formula (I) with other chemical components (i.e. pharmaceutically acceptable inactive ingredients), such as carriers, excipients, binders, filling agents, suspending agents, flavoring agents, sweetening agents, disintegrating agents, dispersing agents, surfactants, lubricants, colorants, diluents, solubilizers, moistening agents, plasticizers, stabilizers, penetration enhancers, wetting agents, anti-foaming agents, antioxidants, preservatives, or one or more combination thereof.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • compositions described herein are administerable to a subject in a variety of ways by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular, intramedullary injections, intrathecal, direct
  • the pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered orally.
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered topically.
  • Compound 1, or a pharmaceutically acceptable salt thereof is formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, shampoos, scrubs, rubs, smears, medicated sticks, medicated bandages, balms, creams or ointments.
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered topically to the skin.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 is administered by inhalation.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 is formulated for intranasal adminstration.
  • Such formulations include nasal sprays, nasal mists, and the like.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 is formulated as eye drops.
  • the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by inhalation to the mammal; and/or (e) administered by nasal administration to the mammal; or and/or (f) administered by injection to the mammal; and/or (g) administered topically to the mammal; and/or (h) administered by ophthalmic administration; and/or (i) administered rectally to the mammal; and/or (j) adminstered non- systemically or locally to the mammal.
  • Compound 1 e.g. Compound 2
  • administrations of the effective amount of the compound including further embodiments in which (i) the compound is administered once; (ii) the compound is administered to the mammal multiple times over the span of one day; (iii) continually; or (iv) continuously.
  • any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours.
  • the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed.
  • the length of the drug holiday varies from 2 days to 1 year.
  • a compound as described herein is administered in a local rather than systemic manner.
  • the compound described herein is administered topically. In some embodiments, the compound described herein is administered systemically.
  • Compound 1, or a pharmaceutically acceptably salt thereof are formulated by combining the active compound with pharmaceutically acceptable carriers or excipients.
  • Such carriers enable Compound 1, or a pharmaceutically acceptably salt thereof (e.g. Compound 2) to be formulated as tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Compound 1, or a pharmaceutically acceptably salt thereof is formulated without combining the active compound with pharmaceutically acceptable carriers or excipients and is placed directly into a capsule for administration to a mammal.
  • the pharmaceutical compositions will include at least one
  • compositions will include at least one pharmaceutically acceptable carrier, diluent or excipient and Compound 2.
  • compositions described herein include Compound 1 , or a
  • compositions described herein include Compound 1. In some embodiments, the pharmaceutical compositions described herein include amorphous Compound 1. In some embodiments, the pharmaceutical compositions described herein include crystalline Compound 1.
  • the pharmaceutical compositions described herein include Compound 2. In some embodiments, the pharmaceutical compositions described herein include amorphous
  • compositions described herein include crystalline Compound 2.
  • the pharmaceutical compositions described herein include: (a) Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2); and one or more of the following: (b) binders; (c) disintegrants; (d) fillers (diluents); (e) lubricants; (f) glidants (flow enhancers); (g) compression aids; (h) colors; (i) sweeteners; (j) preservatives; (k)
  • suspensing/dispersing agents (1) film formers/coatings; (m) flavors; (o) printing inks; (p) solubilizers; (q) alkalizing agents; (r) buffering agents; (s) antioxidants; (t) effervsescent agents.
  • the pharmaceutical compositions described herein include: (a) Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2); and (b) a capsule shell.
  • pharmaceutical compositions described herein include one or more of the following in addition to Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 (a) magnesium stearate; (b) lactose; (c) microcrystallme cellulose; (d) silicified microcrystallme cellulose; (e) mannitol; (f) starch (corn); (g) silicon dioxide; (h) titanium dioxide; (i) stearic acid; (j) sodium starch glycolate; (k) gelatin; (1) talc; (m) sucrose; (n) aspartame; (o) calcium stearate; (p) povidone; (q) pregelatinized starch; (r) hydroxy propyl methylcellulose; (s) OPA products (coatings & inks); (t) croscarmellose; (u) hydroxy propyl cellulose; (v) ethylcellulose; (w) calcium phosphate (dibasic); (x) crospovidone; (y) shellac (and glaze); (z) sodium carbonate; (aa) hypromellose.
  • pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, silicified microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents are added, such as the cross-linked croscarmellose sodium,
  • polyvinylpyrrolidone agar, or alginic acid or a salt thereof such as sodium alginate.
  • the pharmaceutical compositions described herein are formulated into any suitable dosage form, including but not limited to, aqueous oral dispersions, solid oral dosage forms, fast melt formulations, effervescent formulations, lyophilized formulations, tablets, capsules, pills, controlled release formulations, enteric coated tablets, inhaled powder, inhaled dispersion, IV formulations.
  • the pharmaceutical compositions provided herein may be provided as compressed tablets, tablet triturates, rapidly dissolving tablets, multiple compressed tablets, or enteric-coated tablets, sugar-coated, or film-coated tablets.
  • Pharmaceutical dosage forms can be formulated in a variety of methods and can provide a variety of drug release profiles, including immediate release, sustained release, and delayed release. In some cases it may be desirable to prevent drug release after drug administration until a certain amount of time has passed (i.e. timed release), to provide substantially continuous release over a predetermined time period (i.e. sustained release) or to provide release immediately following drug administration (i.e., immediate release).
  • formulations provide a therapeutically effective amount of
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), enabling, for example, once a week, twice a week, three times a week, four times a week, five times a week, once every other day, once-a-day, twice-a-day (b.i.d.), or three times a day (t.i.d.) administration if desired.
  • the formulation provides a therapeutically effective amount of
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) enabling once-a-day administration.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g.
  • Compound 2 is formulated into an immediate release form that provides for once-a-day
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) that is effective to achieve a plasma level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit a therapeutic effect.
  • Compound 2 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) that is effective to achieve a plasma level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit a therapeutic effect.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2 and one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 10 minutes, less than about 15 minutes, less than about 20 minutes, less than about 25 minutes, less than about 30 minutes, less than about 35 minutes, or less than about 40 minutes, after oral administration, thereby releasing the Compound 1, or a
  • the pharmaceutical compositions provided herein in an immediate release dosage form are capable of releasing not less than 75 % of the therapeutically active ingredient or combination and/or meet the disintegration or dissolution requirements for immediate release tablets of the particular therapeutic agents or combination included in the tablet core, as set forth in USP XXII, 1990 (The United States Pharmacopeia.).
  • Immediate release pharmaceutical compositions include capsules, tablets, pills, oral solutions, powders, beads, pellets, particles, and the like.
  • Excipients used in pharmaceutical compositions should be selected on the basis of compatibility with Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the release profile properties of the desired dosage form.
  • exemplary excipients include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like.
  • Binders impart cohesiveness to solid oral dosage form formulations: for powder filled capsule formulation, they aid in plug formation that is filled into soft or hard shell capsules and for tablet formulation, they ensure the tablet remaining intact after compression and help assure blend uniformity prior to a compression or fill step.
  • the binder(s) are selected from starches, sugars, povidone, cellulose or modified cellulose such as microcrystalline cellulose, hydroxypropyl methyl cellulose, lactose, or sugar alcohols like xylitol, sorbitol or maltitol.
  • the binder is hydroxypropyl methyl cellulose.
  • the binder is hypromellose (e.g., Methocel E5).
  • binder levels of 20-70% are used in powder- filled gelatin capsule formulations. Binder usage level in tablet formulations varies whether direct compression, wet granulation, roller compaction, or usage of other excipients such as fillers which itself acts as moderate binder.
  • Dispersing agents, and/or viscosity modulating agents include materials that control the diffusion and homogeneity of a drug through liquid media or a granulation method or blend method. In some embodiments, these agents also facilitate the effectiveness of a coating or eroding matrix.
  • Diluents increase bulk of the composition to facilitate compression or create sufficient bulk for homogenous blend for capsule filling.
  • disintegrate includes both the dissolution and dispersion of the dosage form when contacted with gastrointestinal fluid.
  • disintegration agents or disintegrants facilitate the breakup or disintegration of a substance.
  • solid oral dosage forms include up to 15% w/w of disintegrant.
  • the disintegrant is croscarmellose sodium.
  • the disintegrant is sodium starch glycolate or crospovidone.
  • Filling agents include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • the filler is lactose (e.g. monohydrate). In another aspect, the filler is mannitol, or dicalcium phosphate. In another aspect, the filler is mannitol, microcrystalline cellulose, dicalcium phosphate or sorbitol.
  • Gastrointestinal fluid is the fluid of stomach secretions of a subject or the saliva of a subject after oral administration of a composition described herein, or the equivalent thereof.
  • An "equivalent of stomach secretion” includes, e.g., an in vitro fluid having similar content and/or pH as stomach secretions such as a 1% sodium dodecyl sulfate solution or 0.1N HC1 solution in water.
  • simulated intestinal fluid USP is an aqueous phosphate buffer system at pH 6.8.
  • Solid oral dosage forms include about 0.25% w/w to about 2.5% w/w of lubricant. In another aspect solid oral dosage forms include about 0.5%) w/w to about 1.5% w/w of lubricant.
  • the solid dosage forms described herein are in the form of a tablet, (including an immediate release tablet, an extended release tablet, a sustained release tablet, a enteric coated tablet, a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder), a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or "sprinkle capsules”), solid dispersion, multiparticulate dosage forms, pellets, or granules.
  • a tablet including an immediate release tablet, an extended release tablet, a sustained release tablet, a enteric coated tablet, a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent
  • the pharmaceutical formulation is in the form of a powder. In still other embodiments, the pharmaceutical formulation is in the form of a tablet, including but not limited to, an immediate release tablet. Additionally, pharmaceutical formulations described herein are administered as a single dosage or in multiple dosages. In some embodiments, the
  • pharmaceutical formulation is administered in two, or three, or four tablets.
  • solid dosage forms e.g., tablets, effervescent tablets, and capsules
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • one or more pharmaceutical excipients e.g. Compound 2
  • compositions are dispersed evenly throughout the composition so that the composition is capable of being readily subdivided into equally effective unit dosage forms, such as tablets, pills, or capsules.
  • unit dosage forms such as tablets, pills, or capsules.
  • the individual unit dosages also include film coatings, which disintegrate upon oral ingestion or upon contact with diluent.
  • these formulations are manufactured by conventional techniques.
  • Conventional techniques include, e.g., one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion. See, e.g., Lachman et al., The Theory and Practice of Industrial Pharmacy (1986).
  • Other methods include, e.g., spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., wurster coating), tangential coating, top spraying, tableting, extruding and the like.
  • Compressed tablets are solid dosage forms prepared by compacting the bulk blend formulations described above.
  • compressed tablets which are designed to dissolve in the mouth will include one or more flavoring agents.
  • the compressed tablets will include a film surrounding the final compressed tablet.
  • the film coating aids in patient compliance (e.g., Opadry ® coatings or sugar coating). Film coatings comprising Opadry ® typically range from about 1% to about 5% of the tablet weight. In other embodiments, the compressed tablets include one or more excipients.
  • compositions in film-coated dosage forms which comprise a combination of an active ingredient, or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more tabletting excipients to form a tablet core using conventional tabletting processes and subsequently coating the core.
  • the tablet cores can be produced using conventional granulation methods, for example wet or dry granulation, with optional comminution of the granules and with subsequent compression and coating.
  • pharmaceutical compositions in enteric coated dosage forms which comprise a combination of an active ingredient, or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more release controlling excipients for use in an enteric coated dosage form.
  • the pharmaceutical compositions also comprise non-release controlling excipients.
  • Enteric-coatings are coatings that resist the action of stomach acid but dissolve or disintegrate in the intestine.
  • the oral solid dosage form disclosed herein include an enteric coating(s).
  • Enteric coatings include one or more of the following: cellulose acetate phthalate; methyl acrylate- methacrylic acid copolymers; cellulose acetate succinate; hydroxy propyl methyl cellulose phthalate; hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate);
  • PVAP polyvinyl acetate phthalate
  • methyl methacrylate-methacrylic acid copolymers methacrylic acid copolymers, cellulose acetate (and its succinate and phthalate version); styrol maleic acid copolymers; polymethacrylic acid/acrylic acid copolymer; hydroxyethyl ethyl cellulose phthalate; hydroxypropyl methyl cellulose acetate succinate; cellulose acetate tetrahydrophtalate; acrylic resin; shellac.
  • An enteric coating is a coating put on a tablet, pill, capsule, pellet, bead, granule, particle, etc. so that it doesn't dissolve until it reaches the small intestine.
  • Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
  • Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
  • Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating.
  • Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press- coated or dry-coated tablets.
  • tablets are coated with water soluble, pH independent film coating which allows for immediate disintegration for fast, active release (e.g. Opadry products).
  • the pharmaceutical compositions provided herein are in the form of a controlled release dosage form.
  • controlled release refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when orally administered.
  • Controlled release dosage forms include delayed-, extended- , prolonged-, sustained-, pulsatile-, modified -, targeted-, programmed-release.
  • the pharmaceutical compositions in controlled release dosage forms are prepared using a variety of modified release devices and methods including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, and combinations thereof.
  • the release rate of the active ingredient(s) can also be modified by varying the particle sizes.
  • controlled release compositions allow delivery of an agent to a human over an extended period of time according to a predetermined profile. Such release rates can provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response. Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding immediate release preparations.
  • controlled release compositions of Compound 1 , or a pharmaceutically acceptable salt thereof provide therapeutically effective levels of Compound 1 for an extended period of time and thereby provide a longer period of pharmacologic response.
  • Delayed release refers to the delivery so that the release can be accomplished at some generally predictable location in the intestinal tract more distal to that which would have been accomplished if there had been no delayed release alterations.
  • the method for delay of release is coating. Any coatings should be applied to a sufficient thickness such that the entire coating does not dissolve in the gastrointestinal fluids at pH below about 5, but does dissolve at pH about 5 and above.
  • the pharmaceutical compositions provided herein is in a modified release dosage form that is fabricated using a matrix controlled release device (see, Takada et al in "Encyclopedia of Controlled Drug Delivery,” Vol. 2, Mathiowitz ed., Wiley, 1999).
  • the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water-swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • an erodible matrix device which is water-swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • a matrix controlled release system includes an enteric coating so that no drug is released in the stomach.
  • the pharmaceutical compositions provided herein may be provided in unit-dosage forms or multiple-dosage forms.
  • Unit-dosage forms refer to physically discrete units suitable for administration to human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of unit-dosage forms include individually packaged tablets and capsules. Unit-dosage forms may be administered in fractions or multiples thereof.
  • a multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit- dosage form. Examples of multiple-dosage forms include bottles of tablets or capsules.
  • a powder comprising the Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) formulations described herein are formulated to include one or more pharmaceutical excipients and flavors. Additional embodiments also comprise a suspending agent and/or a wetting agent.
  • This bulk blend is uniformly subdivided into unit dosage packaging or multi-dosage packaging units. The term "uniform" means the homogeneity of the bulk blend is substantially maintained during the packaging process.
  • Effervescent powders are prepared.
  • Effervescent salts have been used to disperse medicines in water for oral administration.
  • Effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate, citric acid and/or tartaric acid.
  • the method of preparation of the effervescent granules described herein employs three basic processes: wet granulation, dry granulation and fusion.
  • the fusion method is used for the preparation of most commercial effervescent powders. It should be noted that, although these methods are intended for the preparation of granules, the formulations of effervescent salts described herein, in one embodiment, are also prepared as tablets, according to technology for tablet preparation.
  • pharmaceutical preparations which are used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the push-fit capsules contain the active ingredient only without additional inactive ingredients.
  • the active compounds are dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers are added.
  • the formulation is placed in a sprinkle capsule, wherein the capsule is swallowed whole or the capsule is opened and the contents sprinkled on food prior to eating.
  • compositions comprising Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and at least one dispersing agent or suspending agent for oral administration to a subject.
  • the formulation is a powder and/or granules for suspension, and upon admixture with water, a substantially uniform suspension is obtained.
  • a suspension is "substantially uniform" when it is mostly homogenous, that is, when the suspension is composed of approximately the same concentration of Compound 1 , or a
  • Liquid formulation dosage forms for oral administration are aqueous suspensions or nonaqueous suspensions.
  • Liquid formulation dosage forms for oral administration are aqueous suspensions selected from, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, and syrups. See, e.g., Singh et ah, Encyclopedia of Pharmaceutical Technology, 2 nd Ed., pp. 754-757 (2002).
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g.
  • the liquid dosage forms include additives, such as: (a) disintegrating agents; (b) dispersing agents; (c) wetting agents; (d) preservatives; (e) viscosity enhancing agents; (f) sweetening agents; (g) flavoring agents; (h) solibizing agents (bioavailability enhancers).
  • additives such as: (a) disintegrating agents; (b) dispersing agents; (c) wetting agents; (d) preservatives; (e) viscosity enhancing agents; (f) sweetening agents; (g) flavoring agents; (h) solibizing agents (bioavailability enhancers).
  • the aqueous suspensions and dispersions described herein remain in a homogenous state, as defined above by USP Chapter 905, for at least 4 hours.
  • Liquid compositions illustratively take the form of a liquid where the agent (e.g. Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2)) is present in solution, in suspension or both.
  • the liquid composition is aqueous.
  • Liquid compositions illustratively take the form of a liquid where the agent (e.g. Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2)) is present in solution, in suspension or both.
  • the liquid composition is non-aqueous.
  • the aqueous suspension also contains one or more polymers as suspending agents.
  • Useful polymers include water-soluble polymers such as cellulosic polymers, e.g. , hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl- containing polymers.
  • useful compositions also comprise an mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
  • compositions also include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium carbonate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium carbonate, sodium bicarbonate and ammonium chloride.
  • acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium carbonate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium carbonate, sodium bicarbonate and ammonium chloride.
  • acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • liquid pharmaceutical compositions also include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • compositions also include one or more preservatives to inhibit microbial activity.
  • Still other compositions include one or more surfactants to enhance physical stability or for other purposes. Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40.
  • compositions include one or more antioxidants to enhance chemical stability where required.
  • Suitable antioxidants include, by way of example only, ascorbic acid, tocopherol, and sodium metabisulfite.
  • aqueous compositions are packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
  • aqueous pharmaceutical compositions do not include a preservative and are used within 24 hours of preparation.
  • aqueous pharmaceutical compositions include one or more solubilizers which aid in enhancing the bioavailability of the active pharmaceutical ingredient.
  • the solubilizer is selected from Labrasol, Lutrol (macrogels, poloxamers), and others known in the art.
  • oral pharmaceutical solutions described herein are beneficial for the administration to infants (less than 2 years old), children under 10 years of age and any patient group that is unable to swallow or ingest solid oral dosage forms.
  • the compositions take the form of tablets, lozenges, or gels formulated in a conventional manner (see e.g. U.S. Pat. Nos. 4,229,447, 4,596,795, 4,755,386, and 5,739,136).
  • dragee cores are prepared with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions are used, which optionally contain gum arabic, talc,
  • polyvinylpyrrolidone carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • dyestuffs or pigments are added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Compound 1, or a pharmaceutically acceptable salt thereof is formulated in the form of a pharmaceutical composition that is suitable for inhalation/nasal delivery.
  • the pharmaceutical composition is in the form of a solution, suspension, emulsion, colloidal dispersion, spray, dry powder, aerosol, or combinations thereof.
  • the pharmaceutical composition comprises at least one
  • the pharmaceutical composition is administered with an atomizer, an insufflator, a nebulizer, a vaporizer, or a metered dose inhaler.
  • the pharmaceutical composition is inhaled nasally or orally.
  • crystalline Compound 1 is used in the pharmaceutical composition.
  • Compound 2 is used in the pharmaceutical composition.
  • Compound 1 is used in the pharmaceutical composition.
  • amorphous Compound 2 is used in the pharmaceutical composition.
  • nasal/inhalation formulations are described in, for example, Ansel, H. C. et ah, Pharmaceutical Dosage Forms and Drug Delivery Systems, Sixth Ed. (1995); REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY, 21 st edition, 2005.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is formulated in the form of a nasal spray, nasal mist, and the like.
  • Compound 1 for administration by inhalation, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is formulated for use as an aerosol, a mist or a powder.
  • a pharmaceutically acceptable salt thereof e.g. Compound 2
  • compositions suitable for nasal/inhalation administration are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant.
  • Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of the compound described herein and a suitable powder base such as lactose or starch.
  • the pharmaceutical composition is in the form of a powder for nasal/inhalation delivery to a mammal.
  • powders comprise micronized and/or nano-sized particles of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), blended with larger carrier particles that prevent aggregation.
  • a dry powder formulation is prepared as follows: Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is jet milled. Lactose is jet milled and the two ingredients are mixed and the final mixture is packaged in sterile insufflators.
  • powder inhalable e.g. Compound 2
  • formulations described herein comprise crystalline particles of Compound 1. In some instances powder inhalable formulations described herein comprise crystalline particles of Compound 2. In some embodiments, powder inhalable formulations described herein comprise amorphous particles of Compound 1. In some embodiments, powder inhalable formulations described herein comprise amorphous particles of Compound 2.
  • Compound 1 is prepared as transdermal dosage forms.
  • the transdermal formulations described herein include at least three components: (1) a formulation of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2); (2) a penetration enhancer; and (3) an aqueous adjuvant.
  • the transdermal formulations include additional components such as, but not limited to, gelling agents, creams and ointment bases, and the like.
  • the transdermal formulation further include a woven or non-woven backing material to enhance absorption and prevent the removal of the transdermal formulation from the skin.
  • the transdermal formulations described herein can maintain a saturated or supersaturated state to promote diffusion into the skin.
  • formulations suitable for transdermal administration of compounds described herein employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive.
  • patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • transdermal delivery of the compounds described herein can be accomplished by means of iontophoretic patches and the like.
  • transdermal patches provide controlled delivery of the active compound.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • Compound 1, or a pharmaceutically acceptable salt thereof is formulated into a pharmaceutical composition suitable for intramuscular, subcutaneous, or intravenous injection.
  • formulations suitable for intramuscular, subcutaneous, or intravenous injection include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • formulations suitable for subcutaneous injection also contain additives such as preserving, wetting, emulsifying, and dispensing agents.
  • additives such as preserving, wetting, emulsifying, and dispensing agents.
  • isotonic agents such as sugars, sodium chloride, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, such as aluminum monostearate and gelatin.
  • compounds described herein are formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • appropriate formulations include aqueous or nonaqueous solutions, preferably with physiologically compatible buffers or excipients. Such excipients are known.
  • Parenteral injections may involve bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the pharmaceutical composition described herein may be in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered topically and can be formulated into a variety of topically
  • compositions such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • Such pharmaceutical compounds can contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2
  • compositions such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof is about 0.3mg to about lg per dose, about lmg to about lg per dose, about 5mg to about 600mg per dose or about 5mg to about 500mg per dose.
  • the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof is about lmg to about 5g per day, about 5mg to about 2g per day, about 5mg to about lg per day, about 5mg to about 0.6g per day, or about 5mg to about 0.5g per day.
  • the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof is about lOmg per dose, about 25mg per dose, about 50mg per dose, about lOOmg per dose, about 150mg per dose, about 200mg per dose, about 300mg per dose, about 400mg per dose, about 500mg per dose, about 600mg per dose, or about lOOOmg per dose.
  • oral pharmaceutical solutions include about 0.015mg/ml to about 20mg/ml of Compound 2. In some embodiments, oral pharmaceutical solutions include about lmg/ml to about 20mg/ml of Compound 2.
  • immediate release tablets include about 5% w/w to about 50% w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • a pharmaceutically acceptable salt thereof e.g. Compound 2.
  • immediate release tablets include about 5%> w/w to about 40%> w/w, or about 5%> w/w to about 30%) w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • immediate release tablets include about 5%> w/w, about 10%> w/w, about 15%> w/w, about 20%) w/w, about 25%> w/w, about 30%> w/w, about 33%> w/w, about 35%> w/w, about 40%> w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • immediate release capsules include about 1.25% w/w to about 50% w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
  • a pharmaceutically acceptable salt thereof e.g. Compound 2
  • immediate release capsules include Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the capsule shell only.
  • the pharmaceutical compositions including Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), described herein is administered for prophylactic and/or therapeutic treatments.
  • the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition.
  • amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and/or the judgment of the treating physician.
  • compositions containing Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), described herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition.
  • a patient susceptible to or otherwise at risk of a particular disease, disorder or condition is defined to be a "prophylactically effective amount or dose.”
  • prophylactically effective amount or dose the precise amounts also depend on the patient's state of health, weight, and the like.
  • effective amounts for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • administration of the compound, compositions or therapies as described herein includes chronic administration.
  • chronic administration includes administration for an extended period of time, including, e.g., throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.
  • chronic administration includes daily administration.
  • administration of the compounds, compositions or therapies described herein is given continuously.
  • the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time ⁇ i.e., a "drug holiday").
  • the length of the drug holiday varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, and 365 days.
  • the dose reduction during a drug holiday is from 10%- 100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%.
  • the compounds, compositions or therapies described herein are administered in at least one priming dose, followed by at least one maintenance dose.
  • a priming dose of the agent(s) is administered until the symptoms of the disorder, disease or condition treated have been reduced (e.g., to a satisfactory level).
  • a maintenance dose of the compounds, compositions or therapies described herein is administered if desired or if necessary.
  • the maintenance dose comprises administration of the agent(s) described herein in an amount sufficient to at least partially maintain the reduction achieved by administration of the priming dose.
  • the maintenance dose compared to the priming dose, includes a decrease in dosage and/or frequency of administration of the agent or one or more of the agents administered in the method. In certain embodiments, however, intermittent treatment with increased frequency and/or dosage amounts may be necessary upon any recurrence of symptoms.
  • the amount of a given agent that corresponds to a priming or maintenance amount varies depending upon factors including, by way of non-limiting example, the specific agent(s) utilized, the disease condition and its severity, the identity (e.g., weight) of the subject or host in need of treatment, and/or the route of administration.
  • the desired dose is conveniently presented in a single dose or in divided doses administered
  • any standard pharmacokinetic protocol is used to determine blood plasma concentration profile in humans following administration of a formulation described herein (that include Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2)).
  • a randomized single-dose crossover study is performed using a group of healthy adult human subjects. The number of subjects is sufficient to provide adequate control of variation in a statistical analysis, and is typically about 10 or greater, although for certain purposes a smaller group suffices. Each subject receives administration at time zero a single dose of a formulation of
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2), normally at around 8am following an overnight fast.
  • the subjects continue to fast and remain in an upright position for about 2 hours after administration of the formulation.
  • Blood samples are collected from each subject prior to administration (e.g., 15 minutes) and at several intervals after administration. In certain instances, several samples are taken within the first hour and taken less frequently thereafter.
  • blood samples are collected at 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 4, 6, 8, 12, and 16 hours after administration and, 24, 36, 48, 60 and 72 hours after administration. If the same subjects are to be used for study of a second test formulation, a period of at least 10 days should elapse before administration of the second formulation. Plasma is separated from the blood samples by
  • Any formulation giving the desired pharmacokinetic profile is suitable for administration according to the present methods.
  • any of the aforementioned aspects involving the prevention or treatment of LPA-mediated diseases or conditions are further embodiments comprising identifying patients by screening for LPA receptor gene SNPs.
  • a SNP located in the promoter region of LPAi showed significant association with knee osteoarthritis in two independent populations (Mototani et ah Hum. Mol. Genetics, vol. 17, no. 12, 2008). Patients can be further selected based on increased LPA receptor expression in the tissue of interest.
  • CLL chronic lymphocytic leukemia
  • LPA can protect some CLL cells from apoptosis and the cells that are protected by LPA have high levels of LPAi mRNA.
  • CLL patients are selected based on the expression of the LPA1R. LPA receptor expression are determined by methods including, but not limited to, northern blotting, western blotting, quantitative PCR (qPCR), flow cytometry, autoradiography (using a small molecule radioligand or PET ligand).
  • patients are selected based on the concentration of serum or tissue LPA measured by mass spectrometry.
  • LPA concentrations are high in ovarian cancer ascites and in some breast cancer effusions.
  • patients are selected based on a combination of the above markers (increased LPA concentrations and increased LPA receptor expression).
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) in combination with another therapeutic agent.
  • a pharmaceutically acceptable salt thereof e.g. Compound 2
  • the therapeutic effectiveness of one of the compounds described herein is enhanced by administration of an adjuvant ⁇ i.e., by itself the adjuvant may have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
  • the benefit experienced by a patient is increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • Compound 1, or a pharmaceutically acceptable salt thereof is co-administered with a second therapeutic agent, wherein Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the second therapeutic agent modulate different aspects of the disease, disorder or condition being treated, thereby providing a greater overall benefit than administration of either therapeutic agent alone.
  • the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
  • different therapeutically-effective dosages of the compounds disclosed herein will be utilized in formulating pharmaceutical composition and/or in treatment regimens when the compounds disclosed herein are administered in combination with one or more additional agent, such as an additional therapeutically effective drug, an adjuvant or the like.
  • a combination treatment regimen encompasses treatment regimens in which administration of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is initiated prior to, during, or after treatment with a second agent described herein, and continues until any time during treatment with the second agent or after termination of treatment with the second agent. It also includes treatments in which Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the second agent being used in combination are administered simultaneously or at different times and/or at decreasing or increasing intervals during the treatment period. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
  • compositions and methods for combination therapy are provided herein.
  • the pharmaceutical compositions disclosed herein are used to treat LPA-dependent or LPA-mediated conditions.
  • the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought is modified in accordance with a variety of factors. These factors include the disease, disorder or condition from which the subject suffers, as well as the age, weight, sex, diet, and medical condition of the subject. Thus, in some instances, the dosage regimen actually employed varies and, in some embodiments, deviates from the dosage regimens set forth herein.
  • dosages of the co-administered compounds vary depending on the type of co-drug employed, on the specific drug employed, on the disease or condition being treated and so forth.
  • the compound provided herein when co-administered with one or more other therapeutic agents, is administered either simultaneously with the one or more other therapeutic agents, or sequentially.
  • the multiple therapeutic agents are administered in any order or even simultaneously. If administration is simultaneous, the multiple therapeutic agents are, by way of example only, provided in a single, unified form, or in multiple forms (e.g., as a single pill or as two separate pills).
  • one of the therapeutic agents is given in multiple doses, and in another, two (or more if present) are given as multiple doses.
  • the timing between the multiple doses vary from more than zero weeks to less than four weeks.
  • the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations is also envisioned.
  • Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and combination therapies are administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition containing a compound varies.
  • the compounds described herein are used as a prophylactic and are administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition.
  • the compounds and compositions are administered to a subject during or as soon as possible after the onset of the symptoms.
  • a compound described herein is administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease.
  • the length required for treatment varies, and the treatment length is adjusted to suit the specific needs of each subject.
  • a compound described herein or a formulation containing the compound is administered for at least 2 weeks, about 1 month to about 5 years.
  • therapies which combine Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) with inhibitors of LP A synthesis or LPA receptor antagonists, either acting at the same or other points in the LPA synthesis or signalling pathway, are encompassed herein for treating LPA-dependent or LPA-mediated diseases or conditions.
  • methods for treatment of LPA-dependent or LPA- mediated conditions or diseases comprises administration to a mammal Compound 1 , or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 in combination with at least one additional agent selected, by way of example only, alemtuzumab, arsenic trioxide, asparaginase (pegylated or non-), bevacizumab, cetuximab, platinum- based compounds such as cisplatin, cladribine, daunorubicin/doxorubicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, PaclitaxelTM, taxol, temozolomide, thioguanine, or classes of drugs including hormones (an antiestrogen, an antiandrogen, or gonadotropin releasing hormone analogues, interferons such as alpha interferon, nitrogen mustards such as busulfan or melphalan or mechlorethamine, retinoids such as tretinoin, topoisomerase inhibitors such as irinotecan or topotecan
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered or formulated in combination with one or more anti-cancer agents.
  • one or more of the anti-cancer agents are proapoptotic agents.
  • anticancer agents include, but are not limited to, any of the following: gossypol, genasense, polyphenol E, Chlorofusin, all trans-retinoic acid (ATRA), bryostatin, tumor necrosis factor-related apoptosis- inducing ligand (TRAIL), 5-aza-2'-deoxycytidine, all trans retinoic acid, doxorubicin, vincristine, etoposide, gemcitabine, imatinib, geldanamycin, 17-N-Allylamino- 17-Demethoxygeldanamycin (17-AAG), flavopiridol, LY294002, bortezomib, trastuzumab, BAY 11-7082, PKC412, or
  • TaxolTM paclitaxel
  • TaxotereTM analogs of TaxolTM, such as TaxotereTM.
  • anti-cancer agents for use in combination with Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) include inhibitors of mitogen-activated protein kinase signaling, e.g., U0126, PD98059, PD184352, PD0325901, ARRY-142886,
  • anti-cancer agents for use in combination with Compound 1, or a pharmaceutically acceptable salt thereof include one or more of the following: abiraterone;
  • abarelix adriamycin; aactinomycin; acivicin; aclarubicin; acodazole hydrochloride; acronine;
  • adozelesin aldesleukin; alemtuzumab; allopurinol; alitretinoin; altretamine; ambomycin;
  • ametantrone acetate aminoglutethimide; aminolevulinic acid; amifostine; amsacrine; anastrozole; anthramycin; aprepitant; arsenic trioxide; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; bendamustine hydrochloride; benzodepa; bevacizumab; bexarotene; bicalutamide;
  • bisantrene hydrochloride bisnafide dimesylate; bizelesin; bleomycin; bleomycin sulfate;
  • bortezomib brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide;
  • carbetimer carboplatin; carmustine; carubicin hydrochloride; carzelesin; capecitabine; cedefingol; cetuximab; chlorambucil; cirolemycin; cisplatin; cladribine; clofarabine; crisnatol mesylate;
  • cyclophosphamide cytarabine; dacarbazine; dasatinib; daunorubicin hydrochloride; dactinomycin; darbepoetin alfa; decitabine; degarelix; denileukin diftitox; dexormaplatin; dexrazoxane
  • hydrochloride dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; eltrombopag olamine; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; epoetin alfa; erbulozole; erlotinib hydrochloride; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; everolimus; exemestane; fadrozole hydrochloride; camb
  • ozogamicin goserelin acetate; histrelin acetate; hydroxyurea; idarubicin hydrochloride; ifosfamide; iimofosine; ibritumomab tiuxetan; idarubicin; ifosfamide; imatinib mesylate; imiquimod;
  • interleukin II (including recombinant interleukin II, or rlL2), interferon alfa-2a; interferon alfa-2b; interferon alfa-nl; interferon alfa-n3; interferon beta-1 a; interferon gamma-1 b; iproplatin; irinotecan hydrochloride; ixabepilone; lanreotide acetate; lapatinib; lenalidomide; letrozole; leuprolide acetate; leucovorin calcium; leuprolide acetate; levamisole; liposomal cytarabine; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril;
  • mitindomide mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin C; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nandrolone phenpropionate; nelarabine; nilotinib; nocodazoie; nofetumomab; nogalamycin; ofatumumab; oprelvekin; ormaplatin;
  • oxaliplatin oxaliplatin;oxisuran; paclitaxel; palifermin; palonosetron hydrochloride; pamidronate;
  • pegfilgrastim pemetrexed disodium
  • pentostatin panitumumab
  • romidepsin romiplostim; safingol; safingol hydrochloride; sargramostim; semustine; pumprazene; sipuleucel-T; sorafenib; sparfosate sodium; sparsomycin; spirogermanium hydrochloride;
  • spiromustine spiroplatin; streptonigrin; streptozocin; sulofenur; sunitinib malate; talisomycin;
  • tamoxifen citrate tecogalan sodium; tegafur; teloxantrone hydrochloride; temozolomide;
  • temoporfin temsirolimus; teniposide; teroxirone; testolactone; thalidomide;thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; topotecan hydrochloride; toremifene; tositumomab; tositumomab and 1 131 Iodine tositumomab; trastuzumab; trestolone acetate; tretinoin; triciribine phosphate;
  • trimetrexate trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard;
  • uredepa valrubicin; vapreotide; verteporfin; vinblastine; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorinostat; vorozole; zeniplatin;
  • anticancer agents for use in combination with a compound of Formula (I), (II), (III), (IV) or (V) include alkylating agents, antimetabolites, natural products, or hormones, e.g., nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc.), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, ete.), or triazenes (decarbazine, etc.).
  • nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc.
  • alkyl sulfonates e.g., busulfan
  • nitrosoureas e.g., carmustine, lomusitne, ete.
  • triazenes decarbazine, etc
  • antimetabolites include but are not limited to folic acid analog (e.g., methotrexate), or pyrimidine analogs (e.g., Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin).
  • folic acid analog e.g., methotrexate
  • pyrimidine analogs e.g., Cytarabine
  • purine analogs e.g., mercaptopurine, thioguanine, pentostatin.
  • pharmaceutically acceptable salt thereof include but are not limited to vinca alkaloids (e.g., vinblastin, vincristine), epipodophyllotoxins (e.g., etoposide), antibiotics (e.g., daunorubicin, doxorubicin, bleomycin), enzymes (e.g., L-asparaginase), or biological response modifiers (e.g., interferon alpha).
  • vinca alkaloids e.g., vinblastin, vincristine
  • epipodophyllotoxins e.g., etoposide
  • antibiotics e.g., daunorubicin, doxorubicin, bleomycin
  • enzymes e.g., L-asparaginase
  • biological response modifiers e.g., interferon alpha
  • alkylating agents for use in combination with Compound 1, or a
  • pharmaceutically acceptable salt thereof include, but are not limited to, nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, meiphalan, etc.), ethyl enimine and methylmelamines (e.g., hexamethlymelamine, thiotepa), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, semustine, streptozocin, etc.), or triazenes (decarbazine, ete.).
  • nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, meiphalan, etc.
  • ethyl enimine and methylmelamines e.g., hexamethlymelamine, thiotepa
  • antimetabolites include, but are not limited to folic acid analog (e.g., methotrexate), or pyrimidine analogs (e.g., fluorouracil, floxouridine, Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin.
  • folic acid analog e.g., methotrexate
  • pyrimidine analogs e.g., fluorouracil, floxouridine, Cytarabine
  • purine analogs e.g., mercaptopurine, thioguanine, pentostatin.
  • hormones and antagonists for use in combination with Compound 1, or a pharmaceutically acceptable salt thereof include, but are not limited to, adrenocorticosteroids (e.g., prednisone), progestins (e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate), estrogens (e.g., diethlystilbestrol, ethinyl estradiol), antiestrogen (e.g., tamoxifen), androgens (e.g., testosterone propionate, fluoxymesterone), antiandrogen (e.g., flutamide), gonadotropin releasing hormone analog (e.g., leuprolide).
  • adrenocorticosteroids e.g., prednisone
  • progestins e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate
  • platinum coordination complexes e.g., cisplatin, carboblatin
  • anthracenedione e.g., mitoxantrone
  • substituted urea e.g., hydroxyurea
  • methyl hydrazine derivative e.g., procarbazine
  • adrenocortical suppressant e.g., mitotane, aminoglutethimide
  • anti-cancer agents which act by arresting cells in the G2-M phases due to stabilized microtubules include without limitation the following marketed drugs and drugs in development: Erbulozole, Dolastatin 10, Mivobulin isethionate, Vincristine, NSC-639829,
  • Discodermolide ABT-751, Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C), Spongistatins (such as Spongistatin 1, Spongistatin 2, Spongistatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, Spongistatin 7, Spongistatin 8, and Spongistatin 9), Cemadotin hydrochloride, Epothilones (such as Epothilone A, Epothilone B, Epothilone C, Epothilone D, Epothilone E, Epothilone F, Epothilone B N-oxide, Epothilone A N-oxide, 16-aza-epothilone B, 21-aminoepothilone B, 21- hydroxyepothilone D, 26-fluoroepothilone, Auristatin PE, Soblidotin, Vincristine sulfate
  • Cryptophycin 52 Vitilevuamide, Tubulysin A, Canadensol, Centaureidin, Oncocidin Al Fijianolide B, Laulimalide, Narcosine, Nascapine, Hemiasterlin, Vanadocene acetylacetonate, Indanocine Eleutherobins (such as Desmethyleleutherobin, Desaetyleleutherobin, lsoeleutherobin A, and Z- Eleutherobin), Caribaeoside, Caribaeolin, Halichondrin B, Diazonamide A, Taccalonolide A, Diozostatin, (-)-Phenylahistin, Myoseverin B, Resverastatin phosphate sodium.
  • Eleutherobins such as Desmethyleleutherobin, Desaetyleleutherobin, lsoeleutherobin A, and Z- Eleutherobin
  • Caribaeoside Cariba
  • Compound 1, or a pharmaceutically acceptable salt thereof is co-administered with thrombolytic agents (e.g., alteplase anistreplase, streptokinase, urokinase, or tissue plasminogen activator), heparin, tinzaparin, warfarin, dabigatran (e.g., dabigatran etexilate), factor Xa inhibitors (e.g., fondaparinux, draparinux, rivaroxaban, DX-9065a, otamixaban,
  • thrombolytic agents e.g., alteplase anistreplase, streptokinase, urokinase, or tissue plasminogen activator
  • heparin e.g., tinzaparin
  • warfarin e.g., dabigatran etexilate
  • dabigatran e.g., dabigatran etex
  • LY517717 or YM150
  • ticlopidine clopidogrel
  • CS-747 prasugrel
  • ximelagatran or BIBPv 1048.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used in combination with anti-emetic agents to treat nausea or emesis.
  • Anti-emetic agents include, but are not limited to: neurokinin- 1 receptor antagonists, 5HT3 receptor antagonists (such as ondansetron, granisetron, tropisetron, Palonosetron, and zatisetron), GABA B receptor agonists (such as baclofen), corticosteroids (such as dexamethasone, prednisone, prednisolone, or others), dopamine antagonists (such as, but not limited to, domperidone, droperidol, haloperidol, chlorpromazine, promethazine, prochlorperazine, metoclopramide), antihistamines (HI histamine receptor antagonists, such as but not limited to, cyclizine, diphenhydramine, dimenhydrinate, meclizine
  • Compound 1, or a pharmaceutically acceptable salt thereof is used in combination with an agent useful in the treatment of anemia.
  • an agent useful in the treatment of anemia is, for example, a continuous eythropoiesis receptor activator (such as epoetin-a).
  • Compound 1, or a pharmaceutically acceptable salt thereof is used in combination with an agent useful in the treatment of neutropenia.
  • agents useful in the treatment of neutropenia include, but are not limited to, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF). Examples of a G-CSF include filgrastim.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used in combination with radiation therapy (or radiotherapy). Radiation therapy is the treatment of cancer and other diseases with ionizing radiation.
  • Radiation therapy can be used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, prostate, colon, uterus and/or cervix. It can also be used to treat leukemia and lymphoma (cancers of the blood- forming cells and lymphatic system, respectively).
  • a technique for delivering radiation to cancer cells is to place radioactive implants directly in a tumor or body cavity. This is called internal radiotherapy (brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.)
  • internal radiotherapy brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.
  • the radiation dose is concentrated in a small area, and the patient stays in the hospital for a few days.
  • radiation or “ionizing radiation” include all forms of radiation, including but not limited to ⁇ , ⁇ , and ⁇ radiation and ultraviolet light.
  • Compound 1, or a pharmaceutically acceptable salt thereof is used to treat or reduce fibrosis in a mammal.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2 is administered in combination with one or more immunosuppresants.
  • Immunosuppressive therapy is clinically used to treat or prevent the rejection of transplanted organs and tissues (e.g. bone marrow, heart, kidney, liver); treatment of autoimmune diseases or diseases that are most likely of autoimmune origin (e.g.
  • rheumatoid arthritis myasthenia gravis, systemic lupus erythematosus, Crohn's disease, and ulcerative colitis
  • treatment of some other non-autoimmune inflammatory diseases e.g. long term allergic asthma control
  • non-autoimmune inflammatory diseases e.g. long term allergic asthma control
  • Compound 1, or a pharmaceutically acceptable salt thereof is adminsitered with corticosteroids.
  • Compound 1 , or a pharmaceutically acceptable salt thereof is adminsitered with an a therapeutic agent selected from among: Calcineurin inhibitors (such as, but not limited to, cyclosporin, tacrolimus); mTOR inhibitors (such as, but not limited to, sirolimus, everolimus); anti-proliferatives (such as, but not limited to, azathioprine, mycophenolic acid); corticosteroids (such as, but not limited to, prednisone, cortisone acetate, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone,
  • Calcineurin inhibitors such as, but not limited to, cyclosporin, tacrolimus
  • Other therapeutic agents include, but are not limited to: cyclophosphamide, penicillamine, cyclosporine, nitrosoureas, cisplatin, carboplatin, oxaliplatin, methotrexate, azathioprine, mercaptopurine, pyrimidine analogues, protein synthesis inhibitors, dactinomycin, anthracyclines, mitomycin C, bleomycin, mithramycin, Atgam ⁇ , Thymoglobuline ® , OKT3 ® , basiliximab, daclizumab, cyclosporin, tacrolimus, sirolimus, Interferons (IFN- ⁇ , IFN- ⁇ ), opioids, TNF binding proteins (infliximab, etanercept, adalimumab, golimumab), leflunomide, gold thioglucose, gold thiomalate, aurofin,
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g.
  • Compound 2 is administered in combination with Cyclosporin A (CsA) or tacrolimus (FK506).
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2 is administered to a mammal in combination with an anti-inflammatory agent including, but not limited to, non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids (glucocorticoids).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • corticosteroids glucocorticoids
  • NSAIDs include, but are not limited to: aspirin, salicylic acid, gentisic acid, choline magnesium salicylate, choline salicylate, choline magnesium salicylate, choline salicylate, magnesium salicylate, sodium salicylate, diflunisal, carprofen, fenoprofen, fenoprofen calcium, flurobiprofen, ibuprofen, ketoprofen, nabutone, ketolorac, ketorolac tromethamine, naproxen, oxaprozin, diclofenac, etodolac, indomethacin, sulindac, tolmetin, meclofenamate, meclofenamate sodium, mefenamic acid, piroxicam, meloxicam, COX-2 specific inhibitors (such as, but not limited to, celecoxib, rofecoxib, valdecoxib, parecoxib, e
  • Corticosteroids include, but are not limited to: betamethasone, prednisone, alclometasone, aldosterone, amcinonide, beclometasone, betamethasone, budesonide, ciclesonide, clobetasol, clobetasone, clocortolone, cloprednol, cortisone, cortivazol, deflazacort, deoxycorticosterone, desonide, desoximetasone, desoxycortone, dexamethasone, diflorasone, diflucortolone,
  • difluprednate fluclorolone, fludrocortisone, fludroxycortide, flumetasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin, fluocortolone, fluorometholone, fluperolone, fluprednidene, fluticasone, formocortal, halcinonide, halometasone, hydrocortisone/cortisol, hydrocortisone aceponate, hydrocortisone buteprate, hydrocortisone butyrate, loteprednol, medrysone,
  • meprednisone methylprednisolone, methylprednisolone aceponate, mometasone furoate, paramethasone, prednicarbate, prednisone/prednisolone, rimexolone, tixocortol, triamcinolone, and ulobetasol.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g.
  • Compound 2 is administered in combination with leukotriene receptor antagonists.
  • methods for treatment of LPA-dependent or LPA- mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected, by way of example only, HMG-CoA reductase inhibitors (e.g., statins in their lactonized or dihydroxy open acid forms and pharmaceutically acceptable salts and esters thereof, including but not limited to lovastatin; simvastatin; dihydroxy open-acid simvastatin, particularly the ammonium or calcium salts thereof; pravastatin, particularly the sodium salt thereof; fluvastatin, particularly the sodium salt thereof; atorvastatin, particularly the calcium salt thereof; nisvastatin, also referred to as NK-104; rosuvastatin); agents that have both lipid-altering effects and other pharmaceutical activities; HMG-CoA synthase inhibitors; cholesterol absorption inhibitors such as ezetimibe;
  • HMG-CoA reductase inhibitors e.g.,
  • cholesterol ester transfer protein (CETP) inhibitors for example JTT-705 and CP529, 414; squalene epoxidase inhibitors; squalene synthetase inhibitors (also known as squalene synthase inhibitors); acyl-coenzyme A: cholesterol acyltransferase (ACAT) inhibitors including selective inhibitors of ACAT-1 or ACAT-2 as well as dual inhibitors of ACAT- 1 and-2; microsomal triglyceride transfer protein (MTP) inhibitors; probucol; niacin; bile acid sequestrants; LDL (low density lipoprotein) receptor inducers; platelet aggregation inhibitors, for example glycoprotein Ilb/IIIa fibrinogen receptor antagonists and aspirin; human peroxisome proliferator activated receptor gamma (PPARy) agonists, including the compounds commonly referred to as glitazones, for example troglitazone, pioglitazone
  • methods for treatment of LPA-dependent or LPA- mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, by way of example only, COX-2 inhibitors; nitric oxide synthase inhibitors, such as N-(3-(aminomethyl)benzyl) acetamidine; Rho kinase inhibitors, such as fasudil; angiotension II type-1 receptor antagonists, including candesartan, losartan, irbesartan, eprosartan, telmisartan and valsartan; glycogen synthase kinase 3 inhibitors; sodium or calcium channel blockers, including crobenetine; p38 MAP kinase inhibitors, including SKB 239063; thromboxane AX- synthetase inhibitors, including isbogrel, ozagrel, ridogrel and da
  • NMDA receptor antagonists including memantine; NR2B antagonists, such as traxoprodil; 5-HT1A agonists; receptor platelet fibrinogen receptor antagonists, including tirofiban and lamifiban;
  • thrombin inhibitors include argatroban; antihypertensive agents, such as enalapril; vasodilators, such as cyclandelate; nociceptin antagonists; DPIV antagonists; GABA 5 inverse agonists; and selective androgen receptor modulators.
  • methods for treatment of LPA-dependent or LPA- mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, by way of example only,
  • dimethylsulfoxide dimethylsulfoxide, omalizumab, and pentosan polysulfate.
  • methods for treating LPA-dependent or LPA- mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one agent used in the treatment of respiratory conditions.
  • respiratory disorders e.g., asthma, COPD and rhinitis
  • Agents used in the treatment of respiratory conditions include, but are not limited to, bronchodilators (e.g., sympathomimetic agents and xanthine derivatives), leukotriene receptor antagonists, leukotriene formation inhibitors, leukotriene modulators, nasal decongestants, respiratory enzymes, lung surfactants, antihistamines (e.g., mepyramine (pyrilamine), antazoline, diphenhydramine, carbinoxamine, doxylamine, clemastine, dimenhydrinate, pheniramine, chlorphenamine (chlorpheniramine), dexchlorpheniramine, brompheniramine, triprolidine, cetirizine, cyclizine, chlorcyclizine, hydroxyzine, meclizine, loratadine, desloratidine, promethazine, alimemazine (trimeprazine), cyproheptadine, azatadine, ketotifen, acrivas
  • FLAP 5-lipoxygenase-activating protein
  • methods for treating LPA-dependent or LPA-mediated conditions or diseases, such as the therapy of asthma and/or COPD comprises administration to a patient anti-inflammatory agents.
  • methods for treating LPA-dependent or LPA-mediated conditions or diseases, such as the therapy of asthma and/or COPD comprise administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, but not limited to, epinephrine, isoproterenol, orciprenaline, bronchodilators, glucocorticoids, leukotriene modifiers, mast-cell stabilizers, xanthines, anticholinergics, ⁇ -2 agonists, FLAP inhibitors, FLAP modulators or 5-LO inhibitors, ⁇ -2 agonists include, but are not limited to, short- acting ⁇ -2 agonists (e.g., salbutamol (albuterol), levalbuterol, terbuta
  • FLAP inhibitors and/or FLAP modulators include, but are not limited to, 3-[3-tert-butylsulfanyl-l-[4-(6-methoxy-pyridin-3-yl)-benzyl]-5-(pyridin-2- ylmethoxy)- lH-indol-2-yl]-2,2-dimethyl-propionic acid, 3-[3-tert-butylsulfanyl- 1 -[4-(6-ethoxy- pyridin-3-yl)-benzyl]-5-(5-methyl-pyridin-2-ylmethoxy)-lH-indol-2-yl]-2,2-dimethyl-propionic acid, MK-886, MK-0591, BAY-xl005 and compounds found in US 2007/0225285, US
  • Glucocorticoids include, but are not limited to, beclometasone, budesonide, ciclesonide, fluticasone and mometasone.
  • Anticholinergics include, but are not limited to, ipratropium and tiotropium.
  • Mast cell stabilizers include, but are not limited to, cromoglicate and nedocromil.
  • Xanthines include, but are not limited to, amminophylline, theobromine and theophylline.
  • Leukotriene antagonists include, but are not limited to, montelukast, tomelukast, pranlukast and zafirlukast.
  • 5-LO inhibitors include, but are not limited to, zileuton, VIA-2291 (ABT761), AZ-4407 and ZD-2138 and compounds found in US 2007/0149579, WO2007/016784.
  • methods for treating LPA-dependent or LPA-mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, by way of example only, antihistamines, leukotriene antagonists, corticosteroids and decongestants.
  • Leukotriene antagonists include, but are not limited to, montelukast, tomelukast, pranlukast and zafirlukast.
  • LPA receptor antagonists described herein are admistered in combination with one or more agents used to treat used to treat asthma, including, but not limited to: combination inhalers (fluticasone and salmeterol oral inhalation (e.g. Advair)); inhaled Beta-2 agonists (albuterol inhaler; albuterol nebulizer solution; formoterol; isoproterenol oral inhalation; levalbuterol;
  • combination inhalers fluticasone and salmeterol oral inhalation (e.g. Advair)
  • inhaled Beta-2 agonists albuterol inhaler; albuterol nebulizer solution; formoterol; isoproterenol oral inhalation; levalbuterol;
  • salmeterol powder inhalation terbutaline inhaler
  • inhaled corticosteroids beclomethasone oral inhalation; budesonide inhalation solution; budesonide inhaler; flunisolide oral inhalation;
  • fluticasone inhalation aerosol fluticasone inhalation aerosol; fluticasone powder for oral inhalation; mometasone inhalation powder; triamcinolone oral inhalation); leukotriene modifiers (montelukast; zafirlukast; zileuton); mast cell stabilizers (cromolyn inhaler; nedocromil oral inhalation); monoclonal antibodies
  • omalizumab oral Beta-2 agonists (albuterol oral syrup; albuterol oral tablets; metaproterenol; terbutaline); bronchodilator (aminophylline; oxtriphylline; theophylline).
  • LPA receptor anatogonists described herein are admistered in combination with one or more agents used to treat allergy, including, but not limited to: antihistamine and decongestant combinations (cetirizine and pseudoephedrine; desloratadine and pseudoephedrine ER; fexofenadine and pseudoephedrine; loratadine and pseudoephedrine); antihistamines (azelastine nasal spray; brompheniramine; brompheniramine oral suspension; carbinoxamine; cetirizine;
  • chlorpheniramine clemastine; desloratadine; dexchlorpheniramine ER; dexchlorpheniramine oral syrup; diphenhydramine oral; fexofenadine; loratadine; promethazine); decongestants
  • anticholinergics ipratropium
  • nasal corticosteroids beclomethasone nasal inhalation; budesonide nasal inhaler; flunisolide nasal inhalation; fluticasone nasal inhalation; mometasone nasal spray; triamcinolone nasal inhalation; triamcinolone nasal spray); nasal decongestants (phenylephrine); nasal mast cell stabilizers (cromolyn nasal spray).
  • LPA receptor anatogonists described herein are admistered in combination with one or more agents used to treat chronic obstructive pulmonary disease (COPD), including, but not limited to: anticholinergics - ipratropium bromide oral inhalation); combination Inhalers (albuterol and ipratropium (e.g. Combivent, DuoNeb); fluticasone and salmeterol oral inhalation (e.g.
  • COPD chronic obstructive pulmonary disease
  • corticosteroids (dexamethasone tablets; fludrocortisone acetate; hydrocortisone tablets; methylprednisolone; prednisolone liquid; prednisone oral; triamcinolone oral); inhaled Beta- 2 Agonists (albuterol inhaler; albuterol nebulizer solution; formoterol; isoproterenol oral inhalation; levalbuterol; metaproterenol inhalation; pirbuterol acetate oral inhalation; salmeterol aerosol inhalation; salmeterol powder inhalation; terbutaline inhaler); inhaled Corticosteroids
  • Beta-2 agonists albuterol oral syrup; albuterol oral tablets; metaproterenol; terbutaline; bronchodilator (aminophylline; oxtriphylline; theophylline).
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 is administered to a patient in combination with inhaled corticosteroids.
  • Compound 1 or a pharmaceutically acceptable salt thereof (e.g.
  • Compound 2 is administered to a patient in combination with beta2-adrenergic receptor agonists.
  • Compound 1 , or a pharmaceutically acceptable salt thereof e.g. Compound 2 is administered to a patient in combination with short acting beta2-adrenergic receptor agonists.
  • Compound 1, or a pharmaceutically acceptable salt thereof e.g. Compound 2 is administered to a patient in combination with long-acting beta2-adrenergic receptor agonists.
  • Compound 2 is combined with or administered in combination with one or more agents that are inhibitors of UDP-glucuronosyltransferase (UGT).
  • UGT inhibitors include those described in U.S. 2003/0215462; U.S. 2004/0014648.
  • co-administration of a UGT inhibitor allows for lower doses of Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
  • the individual compounds of such combinations are administered either sequentially or simultaneously in separate or combined pharmaceutical formulations.
  • the individual compounds will be administered simultaneously in a combined pharmaceutical formulation.
  • Appropriate doses of known therapeutic agents will be appreciated by those skilled in the art.
  • kits and articles of manufacture are also described herein.
  • Such kits include a carrier, package, or container that is
  • each of the container(s) comprising one of the separate elements to be used in a method described herein.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers are formed from a variety of materials such as glass or plastic.
  • the articles of manufacture provided herein contain packaging materials.
  • Packaging materials for use in packaging pharmaceutical products include, e.g., U.S. Patent Nos. 5,323,907, 5,052,558 and 5,033,252.
  • Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, bags, containers, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • a wide array of formulations of the compounds and compositions provided herein are contemplated as are a variety of treatments for any disease, disorder, or condition that would benefit by antagonism of LP A receptors.
  • the container(s) include Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), optionally in a composition or in combination with another agent as disclosed herein.
  • Such kits optionally include an identifying description or label or instructions relating to its use in the methods described herein.
  • a kit typically includes labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included.
  • a label is on or associated with the container.
  • a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • a label is used to indicate that the contents are to be used for a specific therapeutic application. The label also indicates directions for use of the contents, such as in the methods described herein.
  • the pharmaceutical compositions are presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein.
  • the pack for example, contains metal or plastic foil, such as a blister pack.
  • the pack or dispenser device is accompanied by instructions for administration.
  • the pack or dispenser is also accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are also prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • compositions described as comprising a pharmaceutically acceptable salt described herein encompass pharmaceutical compositions comprising the associated and/or disassociated forms of the salt.
  • a pharmaceutical composition described herein comprising an aqueous solution of Compound 2 encompasses a composition comprising a population of sodium cations and a population of ⁇ 4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ - acetate anions.
  • compositions that include a Compound 1 , or a pharmaceutically acceptable salt and/or solvate thereof, and pharmacokinetic profiles and pharmacodynamic effects thereof.
  • Compound 1 is optionally prepared as outlined in US patent application 12,638,702, or as outlined herein.
  • Step 1 3-Methylamino-but-2-enoic acid methyl ester
  • Step 2 2-(4-Bromo-benzoyl)-3-oxo-butyric acid methyl ester
  • Step 3 5-(4-Bromo-phenyl)-3-methyl-isoxazole-4-carboxylic acid methyl ester
  • Step 4 5-(4-Bromo-phenyl)-3-methyl-isoxazole-4-carboxylic acid
  • Step 1 [4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-acetic acid ethyl ester
  • Step 1 (R)-l-Phenyl-ethanol [00482] To acetophenone (19.47mL, 166.6mmol) in THF (lOOmL) was added (S)-(-)-2-methyl-CBS- oxazaborolidine (4.62g, 16.6mmol), and the reaction was cooled to 0°C. Borane methyl sulfide complex (2M in THF; 50mL, lOOmmol) was added over 15 minutes, and the reaction was stirred at room temperature. Aqueous work-up gave the title compound.
  • Step 3 ⁇ 4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ - acetic acid ethyl ester
  • Step 4 ⁇ 4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ - acetic acid
  • Mass spectrometric data (mass spec, data) is obtained on with a Shimadzu LCMS 201 OA.
  • Example 7 Large Scale Synthesis of ⁇ 4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yll-biphenyl-4-yl
  • the bottom organic layer was bag filtered to remove insoluble solids and sodium sulfate treated.
  • the organic layer was charged back to the reactor and vacuum distilled to remove -55% of the dichloromethane charged to the reactor.
  • 63.9 kg of heptane was charged to the reactor and the reactor agitated at 20 ⁇ 5°C for at least 1 hr.
  • the reactor contents were filtered and washed with 20.0 kg (1 v/2v) toluene/heptane.
  • the filter cake was dried in a vacuum oven at a temperature ⁇ 50°C.
  • a GL Nutsche Filter was setup and loaded with 35.0 kg of silica gel.
  • the silica gel was wet with 30.0 kg (9v/lv) of dichloromethane/hexanes. 26.8 kg of ⁇ 4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid ethyl ester in
  • dichloromethane/hexanes was charged to the Nutsche filter, and washed with 150.0 kg (9vlv) of dichloromethane/hexanes, 300.0 kg of dichloromethane, 391.0 kg (95v/5v) of dichloromethane/ethyl acetate, and 130.0 kg (9vlv) of dichloromethane/ethyl acetate. The fractions were monitored by
  • a GL Nutsche Filter was setup and loaded with 37.0 kg of Silica Gel.
  • the silica gel was wet with 37.8 kg (9v/lv) of dichloromethane/hexanes. 26.7 kg of ⁇ 4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl ⁇ -acetic acid ethyl ester in
  • dichloromethane/hexanes was charged to the Nutsche filter, and washed with 150.0 kg (9v/lv) of dichloromethane/hexanes, 300.0 kg of dichloromethane, 391.8 kg (95v/5v) of
  • the reactor contents were bag and polished filter to a clean HDPE drum.
  • the drum contents were charged back to the reactor with 500 g of silicycle thiol.
  • the reactor contents were agitated at a jacket temperature of 45 °C for 8 hr.
  • the reactor contents were bag and polished filter to a clean HDPE drum.
  • the reactor was rinsed with 10 kg of tetrahydrofuran and the rinse transferred through the filters to the drum.
  • the reactor contents were filtered using a Nutsche Filter.
  • the filter cake was washed with 18 kg of heptane. Dried the filter cake in a vacuum oven at a temperature ⁇ 85°C until constant weight was achieved (23 hr). Upon achieving constant weight, the oven temperature was reduced to
  • Example 10 Crystalline Compound 2 (Pattern 1)
  • the remaining salt formation experiments were allowed to evaporate and the solids obtained were analyzed.
  • the potassium salt formation experiments produced the same XRPD pattern for three experiments, all of low crystallinity.
  • the L-arginine salts were either amorphous or oils.
  • the L-lysine salts showed signs of crystallinity in IPA and THF, in EtOH and EtOAc the products were amorphous. All ammonium salts formation experiments yielded the free acid back, i.e. no salt formation had occurred.
  • the N-methylglucamine experiments yielded material which showed signs of crystallinity with three solvents yielding the same crystalline pattern (IPA, EtOH, EtOAc).
  • Bruker AXS/Siemens D5000 [00515] X-Ray Powder Diffraction patterns were collected on a Siemens D5000 diffractometer using Cu Ka radiation (40kV, 40mA), ⁇ - ⁇ goniometer, divergence of V20 and receiving slits, a graphite secondary monochromator and a scintillation counter. The instrument is performance checked using a certified Corundum standard (NIST 1976). The software used for data collection was Diffrac Plus XPvD Commander v2.3.1 and the data were analyzed and presented using Diffrac Plus EVA v 11.0.0.2 or v 13.0.0.2.
  • X-ray optics consisted of a single G5bel multilayer mirror coupled with a pinhole collimator of 0.3 mm.
  • the beam divergence i.e. the effective size of the X-ray beam on the sample, was approximately 4 mm.
  • a ⁇ - ⁇ continuous scan mode was employed with a sample - detector distance of 20 cm which gives an effective 2 ⁇ range of 3.2° - 29.7°.
  • the software used for data collection was GADDS for WNT 4.1.16 and the data were analyzed and presented using Diffrac Plus EVA v 9.0.0.2 or v 13.0.0.2.
  • Samples run under ambient conditions were prepared as flat plate specimens using powder as received without grinding. Approximately 1-2 mg of the sample was lightly pressed on a glass slide to obtain a flat surface.
  • VT-XRPD variable temperature-XRPD
  • Compound 1 pattern 1 free acid
  • Selected XRPD diffractograms from the VT-XRPD experiment are shown in Figure 5. It would appear that at a temperature above the small endotherm observed in the DSC ( ⁇ 190°C) a different XRPD pattern is observed. This endothermic transition to a different polymorph would indicate an enantiotropic relationship between the two polymorphs.
  • DSC data were collected on a TA Instruments Q2000 equipped with a 50 position auto- sampler. The calibration for thermal capacity was carried out using sapphire and the calibration for energy and temperature was carried out using certified indium. Typically 0.5-3 mg of each sample, in a pin-holed aluminum pan, was heated at 10°C min "1 from 25°C to 220°C. A purge of dry nitrogen at 50 ml ' min "1 was maintained over the sample. The instrument control software was Advantage for Q Series v2.8.0.392 and Thermal Advantage v4.8.3 and the data were analyzed using Universal Analysis v4.3A.
  • TGA data were collected on a TA Instruments Q500 TGA, equipped with a 16 position auto- sampler.
  • the instrument was temperature calibrated using certified Alumel. Typically 5-30 mg of each sample was loaded onto a pre-tared platinum crucible and aluminum DSC pan, and was heated at 10°C min "1 from ambient temperature to 350°C. A nitrogen purge at 60 ml min "1 was maintained over the sample.
  • the instrument control software was Advantage for Q Series v2.8.0.392 and Thermal Advantage v4.8.3
  • VT-XRPD variable temperature-XRPD
  • a cycling DSC experiment was performed. Selected XRPD diffractograms from the VT-XRPD experiment are shown in Figure 5.
  • the sodium salt (Compound 2) showed a gradual loss in the TGA ( Figure 7) of 5.5% before decomposition.
  • the DSC showed a single broad endotherm onset 176°C ( ⁇ 30 J ' g "1 ).
  • sorption isotherms were obtained using a Hiden IGASorp moisture sorption analyzer, controlled by CFRSorp software.
  • the sample temperature was maintained at 25°C by a Huber re-circulating water bath.
  • the humidity was controlled by mixing streams of dry and wet nitrogen, with a total flow rate of 250 ml min- 1.
  • the relative humidity was measured by a calibrated Vaisala RH probe (dynamic range of 0-95 %RH), located near the sample.
  • the weight change (mass relaxation) of the sample as a function of %RH was constantly monitored by the microbalance (accuracy ⁇ 0.001 mg).
  • 10-20 mg of sample was placed in a tared mesh stainless steel basket under ambient conditions. The sample was loaded and unloaded at 40 %RH and 25°C (typical room conditions).
  • the software uses a least squares minimization procedure together with a model of the mass relaxation, to predict an asymptotic value.
  • the measured mass relaxation value must be within 5% of that predicted by the software, before the next %RH value is selected.
  • the minimum equilibration time was set to 1 hour and the maximum to 4 hours.
  • sorption isotherms were obtained using a SMS DVS Intrinsic moisture sorption analyzer, controlled by SMS Analysis Suite software.
  • the sample temperature was maintained at 25°C by the instrument controls.
  • the humidity was controlled by mixing streams of dry and wet nitrogen, with a total flow rate of 200 ml min-1.
  • the relative humidity was measured by a calibrated Rotronic probe (dynamic range of 1.0-100% RH), located near the sample.
  • the weight change (mass relaxation) of the sample as a function of %RH was constantly monitored by the microbalance (accuracy ⁇ 0.005 mg). Typically 5-20 mg of sample was placed in a tared mesh stainless steel basket under ambient conditions.
  • the sample was loaded and unloaded at 40 %RH and 25°C (typical room conditions).
  • a moisture sorption isotherm was performed as outlined below (2 scans giving 1 complete cycle).
  • the standard isotherm was performed at 25°C at 10 %RH intervals over a 0.5-90 %RH range.
  • the sodium salt sample (Compound 2) was hygroscopic taking up 17.9%) mass on going from 40-90%) RH. There was no hysteresis and no evidence of a form change. The total uptake for 0-90%oRH was 19.1% The XRPD diffractogram post GVS showed a small increase in crystallinity.
  • Aqueous solubility was determined by suspending sufficient compound in water to give a maximum final concentration of >10 mg ml "1 of the parent free-form of the compound. The suspension was equilibrated at 25 °C for 24 hours then the pH was measured. The suspension was then filtered through a glass fiber C filter into a 96 well plate unless stated otherwise. The filtrate was then diluted by a factor of 101. Quantification was by HPLC with reference to a standard solution of approximately 0.25 mg ml "1 in DMSO. Different volumes of the standard, diluted and undiluted sample solutions were injected. The solubility was calculated using the peak areas determined by integration of the peak found at the same retention time as the principal peak in the standard injection.
  • thermodynamic aqueous solubility of both the free acid (Compound 1) and the sodium salt (Compound 2) were determined at a variety of pH levels. Analysis and observations were carried out after 2 hours. The results for the free acid (Compound 1) are shown in Table 8.
  • Timetable Time (min) % Phase A % Phase B
  • Samples of Compound 1 and Compound 2 were found to be greater than 90% pure. In some embodiments, samples of Compound 1 were found to be greater than 95%) pure, greater than 96%) pure, greater than 97%o pure, greater than 98%o pure, greater than 99%o pure. In some embodiments, samples of Compound 2 were found to be greater than 94%o pure, greater than 95%o pure, greater than 96%o pure, greater than 97%> pure, greater than 98%> pure, greater than 99%> pure.
  • Chiral purity (%> enatiomeric excess; %> e.e.) was determined.
  • samples of Compound 1 and Compound 2 were found to have greater than 98%> e.e.
  • samples of Compound 1 were found to have greater than 95%> e.e., greater than 96%> e.e., greater than 97%> e.e., greater than 98%> e.e., or greater than 99%> e.e.
  • samples of Compound 2 were found to have greater than 94%> e.e., greater than 95%> e.e., greater than 96%> e.e., greater than 97%> e.e., greater than 98%> e.e., or greater than 99%> e.e.
  • the test for Residual Solvents is performed to detect trace amounts of solvents used in the synthesis that may be present in the API.
  • the analysis is performed via headspace or direct injection analysis using a gas chromatograph equipped with a flame ionization detector (FID). All residual solvents used in the synthesis are capable of being detected by this method.
  • FID flame ionization detector
  • Potential residual solvents include acetone, ethanol, methanol, dichloromethane, methyl-tert- butyl-ether (MTBE), ethyl acetate, tetrahydrofuran, heptane, dimethoxyethane (DME), toluene.
  • MTBE methyl-tert- butyl-ether
  • DME dimethoxyethane
  • ICP-AES inductively coupled plasma atomic emission spectrometry
  • Pd content by ICP-AES is a detectable amount of palladium that is less than about 20ppm.
  • Pd content by ICP-AES is less than about 20ppm.
  • Pd content by ICP-AES is a detectable amount of palladium that is less than 20ppm, less than 15ppm, less than lOppm, or less than 5ppm.
  • Pd content by ICP-AES is less than 20 ppm, less than 15ppm, less than lOppm, or less than 5ppm.
  • samples or pharmaceutical compositions do not include a detectable amount of palladium.
  • compositions that include Compound 1 include pharmaceutically acceptable salts (e.g. Compound 2) and/or pharmaceutically acceptable solvates thereof include a variety of forms.
  • pharmaceutical compositions are in the form of oral dosage forms.
  • the oral dosage forms are formulated as: oral solutions, oral suspensions, tablets, pills, capsules, ointments, creams or gels.
  • an oral pharmaceutical composition in the form of an oral solution is prepared as outlined below.
  • an oral pharmaceutical composition is prepared with the following ingredients:
  • an oral pharmaceutical composition is prepared with the following ingredients:
  • an oral pharmaceutical composition is prepared with the following ingredients:
  • the manufacturing process for the oral solutions of Compound 1 or Compound 2 described above is as follows: weigh the required amount of sodium carbonate (if present) and transfer to the container. Add the required amount of water to make a lOmM solution and mix until dissolved. Weigh the required amount of propylene glycol and Tween 80 (if present) and add this to the solution and mix until homogenous. Weigh the required amount of Compound 1 or Compound 2 and slowly add to the solution. Mix until all Compound 1 or Compound 2 is dissolved (sonicate, warm, or stir if necessary).
  • a capsule formulation of Compound 1 or Compound 2 for administration to humans is prepared with the following ingredients:
  • Matching Placebo Capsules are prepared with the following ingredients:
  • the process to prepare Compound 1 or Compound 2 in a capsule is as follows: Weigh the required amount of Compound 1 or Compound 2, add into the appropriate size capsule, and close capsule. For example, 50-200 mg of Compound 1 or Compound 2 is placed into a Size 4 Capsule. In one embodiment, 100-500 mg of Compound 1 or Compound 2 is placed into a Size 1 Capsule.
  • the manufacturing process includes blending and compression.
  • An optional coating process can be performed.
  • Manufacturing/analytical equipment typically used in the preparation of tablets include: formulation (U.S.A. standard testing sieve; V-shell blender; ERWEKA TBH300 MD hardness tester; Vanderkamp friability tester; Manesty beta press, sixteen station); analytical (Agilent 1100 series HPLC with variable wavelength detector; VanKel model VK7000 dissolution apparatus; VanKel model VK8000 dissolution autosampler).
  • Blending Add all excipients (except lubricant) to a V-shell blender. Add Compound 1 or Compound 2 to the V-shell blender. Mix in the V-shell blender for approximately 15 minutes. Add lubricant and blend for 2 minutes.
  • Tableting Tablet cores are compressed on a Manesty Betapress sixteen station with two sets of 3/8" round standard concave tooling. During compression, individual and average tablet weight, hardness, thickness, and friability are monitored.
  • all tablets are tested for dissolution using the following parameters:
  • Immediate release tablets show release no less than (NLT) 80% of Compound 1 or Compound 2 within 10 minutes.
  • Tablets are optionally packaged in 40 count in 50 cc HDPE bottles, with CRC caps and heat induction seal.
  • Example 25 Enteric Coated tablets
  • Enteric coated tablets are prepared with the ingredients listed in Table 13.
  • the preparation of the enteric coated tablets is as follows: Weigh 388.0 g of acetone and 12. Og of purified water and mix them in a beaker with an overhead stirrer. Weigh 40 g of the Eudragit and pour into the solvent mixture slowly in portions to prevent lump formation. Stir until a clear solution is made. Then weigh 6g of triethyl citrate and add into the clear solution and keep stirring until a homogeneous solution is made. Mix around 60g of placebo tablets with about 80 of the 750 mg immediate release tablets and coat with the coating mixture.
  • the blend of the formulation is prepared in the same manner as the immediate release tablets (e.g. sieving, blending, and compression). Other preparations are acceptable, such as wet granulation, fliudized bed, high shear granulation, etc.
  • the formulation includes drug modifying release excipients. These excipients include but not limited to HPMC (hydroxy propyl)
  • the amount of drug release modifying excipient ranges from about 10% to about 80%) in the formulation.
  • the drug release profile ranges from 0 to 4 hours, 0 to 6 hours, 0 to 8 hours, 0 to 12 hours, 0 to 24 hours, 2 to 4 hours, 2 to 6 hours, etc.
  • the formulations are coated with the Opadry coatings after direct compression.
  • Example sustained release formulations are listed below. Table 14. Sustained Release Tablet
  • Rat, dog, monkey, or human hepatocytes were thawed according to the supplier's instructions. Cells were counted using the Trypan Blue method, and then diluted to 1 x 10 6 viable cells/ml with KB medium. Compound 1 was tested at 30 ⁇ and incubated for up to 2 hours in rat hepatocytes and 4 hours in dog, monkey or human hepatocytes at 37°C. Fresh human hepatocytes were from a single male donor lot Hu8080 (CellsDirect, Raleigh, NC). Reactions were terminated with addition of 3 times incubation volume of acetonitrile, centrifuged, and supernatants were transferred, nitrogen blow dried and reconstituted for LC/MS analysis.
  • Rats with surgically placed bile duct and jugular vein cannula were purchased from Charles River Laboratories and allowed to acclimate for 2 days.
  • Compound 1 was intravenously dosed (2 mg/kg) to three rats as a solution in 0.9% saline (2 mg/mL; 1 mL/kg).
  • Bile samples were collected at time-points 0-2, 2-5 and 5-8, and urine samples were collected at time-points 0-4 and 4-8 hrs post- dose in 20 mL scintillation vials and stored at -40°C until analysis.
  • the major metabolites generated in vitro appear to be a taurine conjugate, N-dealkylated amine, glycosylation of the amine, mono oxidation, and a glucuronide.
  • the major metabolites isolated from rat bile are several taurine conjugates, N-dealkylated amine, mono oxidation, and a glucuronide.
  • urine from these animals no metabolites were found.
  • Fed animals were fasted for at least 12 hours and then were given food ad libitum for 1 hour prior to dosing.
  • rats were anesthetized and Compound 1 or Compound 2 was administered directly to the duodenum or ileum at 10 mg/kg in 0.5%) methylcellulose.
  • a 1.1 kb cDNA encoding the human LPAi receptor was cloned from human lung.
  • Human lung RNA (Clontech Laboratories, Inc. USA) was reverse transcribed using the RETROscript kit (Ambion, Inc.) and the full-length cDNA for human LPAi was obtained by PCR of the reverse transcription reaction.
  • the nucleotide sequence of the cloned human LPAi was determined by sequencing and confirmed to be identical to the published human LPAi sequence (An et al.
  • the cDNA was cloned into the pCDNA5/FRT expression plasmid and transfected in CHO cells using lipofectamine 2000 (Invitrogen Corp., USA). Clones stably expressing human LPAi were selected using hygromycin and identified as cells that show Ca-flux in response to LPA.
  • An expression vector encoding the human LPA 2 cDNA was transiently transfected into B 103 cells using LipofectamineTM 2000 (Invitrogen) following the manufacturers instruction. On the day before the assay, 30,000-35,000 cells/well were seeded together with 0.2 ⁇ lipofectamine 2000 and 0.2 ⁇ g human LPA2 expression vector in 96-well Poly-D-Lysine coated black-wall clear-bottom plates (BD BioCoat) in DMEM + 10%) FBS. Following an overnight culture, cells were washed once with PBS then cultured in serum- free media for 4 hours prior to start of the calcium flux assay.
  • LipofectamineTM 2000 Invitrogen
  • Example 36 Establishment of a CHO Cell Line Stably Expressing Human LPA ⁇
  • a vector containing the human LPA 3 receptor cDNA was obtained from the Missouri S&T cDNA Resource Center (www.cdna.org).
  • the full-length cDNA fragment for human LPA 3 was obtained by PCR from the vector.
  • the nucleotide sequence of the cloned human LPA 3 was determined by sequencing and confirmed to be identical to the published human LPA 3 sequence (NCBI accession number NM_012152).
  • the cDNA was cloned into the pCDNA5/FRT expression plasmid and transfected in CHO cells using lipofectamine 2000 (Invitrogen Corp., USA). Clones stably expressing human LPA 3 were selected using hygromycin and identified as cells that show Ca- flux in response to LPA.
  • Example 37 LPA1 and LPA3 Calcium Flux Assays.
  • Human LPAi or LPA 3 expressing CHO cells are seeded at 20,000-45,000 cells per well in a 96-well poly-D-lysine coated plate one or two days before the assay. Prior to the assay, the cells are washed once with PBS and then cultured in serum- free media for at least 6 hrs and up to 24hrs . On the day of the assay, a calcium indicator dye (Calcium 5, Molecular Devices) in assay buffer (HBSS with Ca 2+ and Mg 2+ and containing 20 mM Hepes and 0.3% fatty-acid free human serum albumin) is added to each well and incubation continued for 1 hour at room temperature.
  • assay buffer HBSS with Ca 2+ and Mg 2+ and containing 20 mM Hepes and 0.3% fatty-acid free human serum albumin
  • test compounds 10 ⁇ of test compounds in 2.5% DMSO are added to the cells and incubation continued at room temperature for 30 minutes.
  • Cells are the stimulated by the addition of 10 nM LPA and intracellular Ca 2+ measured using the Flexstation 3 (Molecular Devices).
  • IC 50 s are determined using Symyx Assay Explorer analysis of drug titration curves.
  • LPA2 calcium flux is measured using at least one of two different assays.
  • BT- 20 human breast cancer cells are seeded at 25,000-35,000 cells per well in 150 ⁇ complete media on Poly-D-Lysine coated black-wall clear-bottom plates. Following an overnight culture, cells are washed once with PBS then serum starved for 4-6 hours prior to the assay.
  • a calcium indicator dye (Calcium 5, Molecular Devices) in assay buffer (HBSS with Ca 2+ and Mg 2+ and containing 20 mM Hepes and 0.3%) fatty-acid free human serum albumin) is added to each well and incubation continued for 15 minutes at 37°C.
  • test compounds in 2.5% DMSO 25 ⁇ of test compounds in 2.5% DMSO are added to the cells and incubation continued at 37°C for 15-30 minutes.
  • Cells are the stimulated by the addition of 100 nM LPA and intracellular Ca 2+ measured using the Flexstation 3 (Molecular Devices).
  • IC 50 s are determined using Symyx Assay Explorer analysis of drug titration curves
  • B 103 cells transiently expressing huma LPA2 are serum starved for 4 hours.
  • a calcium indicator dye (Calcium 4, Molecular Devices) in assay buffer (HBSS with Ca 2+ and Mg 2+ and containing 20 mM Hepes and 0.3%) fatty-acid free human serum albumin) is then added to each well and incubation continued for 1 hour at 37°C.
  • test compounds 10 ⁇ of test compounds in 2.5%o DMSO are added to the cells and incubation continued at room temperature for 30 minutes.
  • Cells are the stimulated by the addition of 10 nM LPA and intracellular Ca 2+ measured using the Flexstation 3 (Molecular Devices).
  • IC5 0 S are determined using Symyx Assay Explorer analysis of drug titration curves.

Abstract

Described herein is the LPAl antagonist {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid (Compound 1), including pharmaceutically acceptable salts thereof. Also described are methods of preparing the LPAl antagonist, or pharmaceutically acceptable salts thereof, as well as pharmaceutical compositions suitable for administration to a mammal that include the LPAl antagonist, or pharmaceutically acceptable salt thereof, and methods of using such pharmaceutical compositions for treating LPA-dependent or LPA-mediated diseases or conditions.

Description

LYSOPHOSPHATIDIC ACID RECEPTOR ANTAGONIST AND USES THEREOF
RELATED APPLICATIONS
[0001] This application claims benefit of U.S. Provisional Application No. 61/354,622, entitled "LYSOPHOSPHATIDIC ACID RECEPTOR ANTAGONIST AND USES THEREOF" filed on June 14, 2010, which is herein incorporated by reference.
FIELD OF THE INVENTION
[0002] Described herein is the LPA receptor antagonist {4'-[3-methyl-4-((R)-l -phenyl - ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid (Compound 1), pharmaceutically acceptable salts, polymorphs, amorphous phases, metabolites thereof, as well as pharmaceutical compositions thereof, and methods of use thereof in the treatment or prevention or diagnosis of diseases or conditions associated with the activity of one or more of the lysophosphatidic acid (LPA) receptors.
BACKGROUND OF THE INVENTION
[0003] Lysophospholipids are membrane-derived bioactive lipid mediators. Lysophospholipids affect fundamental cellular functions that include proliferation, differentiation, survival, migration, adhesion, invasion, and morphogensis. These functions influence many biological processes that include, but are not limited to, neurogensis, angiogenesis, wound healing, fibrosis, immunity, and carcinogenesis.
[0004] Lysophosphatidic acid (LPA) is a lysophospholipid that has been shown to act through sets of specific G protein-coupled receptors (GPCRs) in an autocrine and paracrine fashion. LPA binding to its cognate GPCRs (LPAi, LPA2, LPA3, LPA4, LPA5, LPA6) activates intracellular signaling pathways to produce a variety of biological responses. Antagonists of the LPA receptors find use in the treatment of diseases, disorders or conditions in which LPA plays a role.
SUMMARY OF THE INVENTION
[0005] Described herein is {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid (Compound 1), including all pharmaceutically acceptable solvates (including hydrates), prodrugs, polymorphs, amorphous phases and metabolites thereof or a pharmaceutically acceptable salt of Compound 1 including (including hydrates), prodrugs, polymorphs, amorphous phases and metabolites thereof, and methods of uses thereof. Compound 1, as well as the pharmaceutically acceptable salts thereof, are used in the manufacture of medicaments for the treatment or prevention of LPA mediated and/or LPA dependent diseases, disorders, or conditions. Compound 1 is a LPAi antagonist. [0006] Described herein are pharmaceutical compositions comprising Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. the sodium salt) as the active ingredient in the pharmaceutical composition.
[0007] In one aspect, described is a crystalline form of {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid (Compound 1). Compound 1 has the following structure:
Figure imgf000004_0001
[0008] In some embodiments, the crystalline form of Compound 1 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1 ; a differential scanning calorimetry (DSC) thermogram with endotherms at about 157°C and about 198°C; a differential scanning calorimetry (DSC) thermogram
substantially the same as Figure 4; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 4; substantially the same X-ray powder diffraction (XRPD) pattern post storage at 40 °C/75% RH for one week; or combinations thereof. In some embodiments, the crystalline form of Compound 1 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least four of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least five of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having all of the preceding properties.
[0009] In some embodiments, the crystalline form of Compound 1 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta.
[0010] In some embodiments, the crystalline form of Compound 1 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.2° 2-Theta, 9.2° 2-Theta, 10.6° 2-Theta, 12.6° 2- Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta. [0011] In some embodiments, the crystalline form of Compound 1 has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1.
[0012] In some embodiments, the crystalline form of Compound 1 has a DSC thermogram with endotherms at about 157°C and about 198°C.
[0013] In some embodiments, the crystalline form of Compound 1 has substantially the same X-ray powder diffraction (XRPD) pattern post storage at 40 C/75% RH for one week.
[0014] In some embodiments, the crystalline form of Compound 1 is characterized as having a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 4.
[0015] In some embodiments, the crystalline form of Compound 1 is characterized as having a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 4.
[0016] In some embodiments, the crystalline form of Compound 1 is substantially free of the S- isomer.
[0017] In some embodiments, the crystalline form of Compound 1 is crystallized from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, nitromethane, tetrahydrofuran, dioxane, methyl ethyl ketone, acetone, 2-methoxyethanol, or methanol.
[0018] In some embodiments, samples of Compound 1 include a detectable amount of palladium that is less than 20 ppm. In some embodiments, samples of Compound 1 include a detectable amount of palladium that is less than 15 ppm. In some embodiments, samples of Compound 1 include a detectable amount of palladium that is less than 10 ppm. In some embodiments, samples of Compound 1 do not include a detectable amount of palladium.
[0019] In one aspect, described herein is a crystalline form of {4'-[3-methyl-4-((R)-l -phenyl - ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid, sodium salt (Compound 2).
Compound 2 has the following structure:
Figure imgf000005_0001
[0020] In some embodiments, the crystalline form of Compound 2 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2-Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C; a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7; or combinations thereof. In some embodiments, the crystalline form of Compound 2 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least four of the preceding properties. In some
embodiments, the crystalline form of Compound 2 is characterized as having all of the preceding properties.
[0021] In some embodiments, the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta and 15.8° 2-Theta.
[0022] In some embodiments, the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2- Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta.
[0023] In some embodiments, the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2.
[0024] In some embodiments, the crystalline form of Compound 2 is characterized as having a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C.
[0025] In some embodiments, the crystalline form of Compound 2 is characterized as having a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7.
[0026] In some embodiments, the crystalline form of Compound 2 is characterized as having a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7.
[0027] In some embodiments, the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 8.5° 2-Theta, 9.3° 2-Theta, 16.6° 2-Theta, 17.1° 2-Theta and 21.4° 2-Theta.
[0028] In some embodiments, the crystalline form of Compound 2 has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 3.
[0029] In some embodiments, the crystalline form of Compound 2 is substantially free of the S- isomer.
[0030] In some embodiments, the crystalline form of Compound 2 is crystallized from isopropanol.
[0031] In some embodiments, samples of Compound 2 include a detectable amount of palladium that is less than 20 ppm. In some embodiments, samples of Compound 2 include a detectable amount of palladium that is less than 15 ppm. In some embodiments, samples of Compound 2 do not include a detectable amount of palladium. [0032] In one aspect, described herein is amorphous {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid, sodium salt (Compound 2). In some embodiments, amorphous Compound 2 is substantially free of the S-isomer.
[0033] In some embodiments, described is a pharmaceutically acceptable salt of {4'-[3-methyl-4- ((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1), wherein the pharmaceutically acceptable salt is a sodium salt, calcium salt, potassium salt, ammonium salt, L-arginine salt, L-lysine salt, or N-methyl-D-glucamine salt. In some embodiments, the pharmaceutically acceptable salt is a sodium salt. In some embodiments, the pharmaceutically acceptable salt is {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4- yl} -acetic acid, sodium salt (Compound 2). In some embodiments, the pharmaceutically acceptable salt is amorphous. In some embodiments, the pharmaceutically acceptable salt is crystalline. In some embodiments, the pharmaceutically acceptable salt is Compound 2 and Compound 2 is crystalline. In some embodiments, the pharmaceutically acceptable salt is Compound 2 and
Compound 2 is amorphous.
[0034] In some embodiments, Compound 2 is crystalline and: (a) has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta and 15.8° 2-Theta; (b) has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; (c) has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 8.5° 2-Theta, 9.3° 2-Theta, 16.6° 2- Theta, 17.1° 2-Theta and 21.4° 2-Theta; (d) has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 3; (e) was obtained from isopropanol; or (f) combinations thereof. In some embodiments, Compound 2 is crystalline and has at least two of the properties selected from (a), (b), (c), (d) and (e).
[0035] In some embodiments, the pharmaceutically acceptable salt of Compound 1 is substantially free of the S-isomer.
[0036] In some embodiments, the pharmaceutically acceptable salt of Compound 1 includes a detectable amount of palladium that is less than 20 ppm. In some embodiments, the
pharmaceutically acceptable salt of Compound 1 includes a detectable amount of palladium that is less than 15 ppm. In some embodiments, the pharmaceutically acceptable salt of Compound 1 does not include a detectable amount of palladium.
[0037] In some embodiments, described herein is a pharmaceutical composition comprising
Compound 1, or a pharmaceutically acceptable salt thereof. In some embodiments, Compound 1, or the pharmaceutically acceptable salt thereof is amorphous. In some embodiments, Compound 1 , or the pharmaceutically acceptable salt thereof is crystalline.
[0038] In some embodiments, described herein are pharmaceutical compositions comprising a crystalline form of Compound 1 or a pharmaceutically acceptable salt. In some embodiments, described herein are pharmaceutical compositions comprising a crystalline form of Compound 2. [0039] In some embodiments, the pharmaceutical composition comprises at least inactive ingredient selected from pharmaceutically acceptable carriers, diluents and excipients.
[0040] In some embodiments, the pharmaceutical composition comprises Compound 2.
[0041] In some embodiments, the pharmaceutical composition comprises crystalline Compound 2.
[0042] In some embodiments, Compound 2 is greater than 96% pure. In some embodiments, Compound 2 is greater than 97%> pure. In some embodiments, Compound 2 is greater than 98%) pure.
[0043] In some embodiments, the pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, inhalation, nasal administration, topical administration, ophthalmic administration or otic administration.
[0044] In some embodiments, the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a suspension, a gel, a colloid, a dispersion, a suspension, a solution, an emulsion, an ointment, a lotion, an eye drop or an ear drop.
[0045] In some embodiments, the pharmaceutical composition is in a form suitable for oral administration to a mammal.
[0046] In some embodiments, the pharmaceutical composition is in the form of a pill, capsule, tablet, aqueous solution, aqueous suspension, non-aqueous solution, or non-aqueous suspension.
[0047] In some embodiments, the pharmaceutical composition is in the form of a capsule.
[0048] In some embodiments, the pharmaceutical composition is in the form of an immediate release capsule or an enteric coated capsule.
[0049] In some embodiments, the capsule is a hard gelatine capsule or hypromellose (HPMC) capsule.
[0050] In some embodiments, the capsule comprises at least one excipient in addition to the hard gelatine capsule or hypromellose (HPMC) capsule.
[0051] In some embodiments, the pharmaceutical composition is in the form of a tablet.
[0052] In some embodiments, the pharmaceutical composition is in the form of an immediate release tablet, an enteric coated tablet, or a sustained release tablet.
[0053] In some embodiments, the pharmaceutical composition is in the form of a moisture barrier coated tablet.
[0054] In some embodiments, the pharmaceutical composition is in the form of an aqueous solution or aqueous suspension.
[0055] In some embodiments, a single dose of the pharmaceutical composition comprises about 1 Omg to about 1 OOOmg of Compound 1 , or a pharmaceutically acceptable salt thereof.
[0056] In some embodiments, a single dose of the pharmaceutical composition comprises about 50mg, about lOOmg, about 150mg, about 200mg, about 250mg, about 300mg, about 350mg, about 400mg, about 450mg, about 500mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg or about 1000 mg of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
[0057] In some embodiments, a single dose of the pharmaceutical composition comprises about lOmg to about lOOOmg of Compound 2.
[0058] In some embodiments, a single dose of the pharmaceutical composition comprises about 50mg, about lOOmg, about 150mg, about 200mg, about 250mg, about 300mg, about 350mg, about 400mg, about 450mg, about 500mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg or about 1000 mg of Compound 2.
[0059] In some embodiments, described herein is a pharmaceutical composition that provides at least one metabolite of Compound 1 after administration to a mammal.
0060] In some embodiments, the at least one metabolite is selected from among:
Figure imgf000009_0001
of M4 (M7); or glucuronidation of M7 (M8).
[0061] In some embodiments, described herein is a method of inhibiting the physiological activity of LPA in a mammal comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
[0062] In some embodiments, described herein is a method for treating or preventing a LPA- dependent or LPA-mediated disease or condition in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
[0063] In some embodiments, the LPA-dependent or LPA-mediated disease or condition is selected from lung fibrosis, asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud's phenomenom, or neuropathic pain.
[0064] In some embodiments, described herein is a method for treating or preventing lung fibrosis, asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut, peritoneal fibrosis, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud's phenomenom, neuropathic pain, or spinal cord injury in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
[0065] In some embodiments, described herein is a method of controlling the activation of LPA receptors in a tissue in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof. In some embodiments, the activation of LPA receptors in a tissue in a mammal results in fibrosis.
[0066] In some embodiments, described herein is a method for the treatment or prevention of fibrosis in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof. In some embodiments, the fibrosis comprises lung fibrosis, renal fibrosis, hepatic fibrosis, cutaneous fibrosis, or peritoneal fibrosis. In some embodiments, the fibrosis comprises lung fibrosis, renal fibrosis, hepatic fibrosis or cutaneous fibrosis.
[0067] In some embodiments, described herein is a method of improving lung function in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof. In some embodiments, the mammal has been diagnosed as having lung fibrosis. [0068] In some embodiments, described herein is a method of treating idopathic pulmonary fibrosis in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
[0069] In some embodiments, described herein is a method of controlling an abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in a tissue of a mammal comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
[0070] In some embodiments, the abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in the tissue results in fibrosis.
[0071] In some embodiments, described herein is a method for the treatment or prevention of scleroderma in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof. In some embodiments, described herein is a method for the treatment or prevention of cutaneous scleroderma in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof. In some embodiments, described herein is a method for the treatment or prevention of systemic scleroderma in a mammal comprising administering Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
[0072] In some embodiments, described herein is a method for reducing undesired or abnormal dermal thickening in a mammal comprising administering to mammal in need thereof Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof. In some embodiments, the dermal thickening is associated with scleroderma.
[0073] In some embodiments, described herein is a method of controlling an abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in dermal tissues of a mammal comprising administering to mammal in need thereof Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof. In some embodiments, the abnormal accumulation or activation of cells, fibronectin, collagen or increased fibroblast recruitment in the dermal tissues results in dermal fibrosis. In some embodiments, described herein is a method of reducing hydroxyproline content in dermal tissues of a mammal with cutaneous fibrosis comprising administering to mammal in need thereof Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof.
[0074] In some embodiments, described herein is a method for the treatment or prevention of Raynaud's phenomenom in a mammal comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), or a pharmaceutical composition thereof to the mammal in need thereof.
[0075] In some embodiments, the pharmaceutical composition is administered daily to the mammal. In some embodiments, the pharmaceutical composition is administered once-daily to the mammal. In some embodiments, the pharmaceutical composition is administered twice-daily to the mammal.
[0076] In some embodiments, the mammal is a human.
[0077] In some embodiments, in any of the method of treatments involving a mammal, the mammal is administered one or more additional therapeutically active agents in addition to Compound 1 , or a pharmaceutically acceptable salt thereof.
[0078] In some embodiments, in any of the method of treatments involving a mammal, the mammal is administered one or more additional therapeutically active agents selected from: corticosteroids, immunosuppresant, analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchodilators, leukotriene receptor antagonists, leukotriene formation inhibitors, monoacylglycerol kinase inhibitors, phospholipase Ai inhibitors, phospholipase A2 inhibitors, and lysophospho lipase D (lysoPLD) inhibitors, autotaxin inhibitors, decongestants, antihistamines, mucolytics, anticholinergics, antitussives, expectorants, and β-2 agonists.
[0079] In some embodiments, provided is a method comprising administering Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to a human with a LPA-dependent or LPA-mediated disease or condition. In some embodiments, the human is already being administered one or more additional therapeutically active agents other than Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In some embodiments, the method further comprises administering one or more additional therapeutically active agents other than Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
[0080] In some embodiments, the one or more additional therapeutically active agents other than Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), are selected from: corticosteroids, immunosuppresants, analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchodilators, leukotriene receptor antagonists, leukotriene formation inhibitors, monoacylglycerol kinase inhibitors, phospholipase Ai inhibitors, phospholipase A2 inhibitors, and lysophospholipase D (lysoPLD) inhibitors, autotaxin inhibitors, decongestants, antihistamines, mucolytics, anticholinergics, antitussives, expectorants, and β-2 agonists.
[0081] In another aspect is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment of a disease, disorder or condition in which the activity of at least one LPA receptor contributes to the pathology and/or symptoms of the disease or condition. In one embodiment of this aspect, the LPA receptor is selected from LPAi, LPA2, LPA3, LPA4, LPA5 and LPA6. In some embodiments, the LPA receptor is LPAi or LPA or LPA3. In some
embodiments, the disease or condition is any of the diseases or conditions specified herein. [0082] Also provided is a method of inhibiting the physiological activity of LPA in a mammal comprising administering a therapuetically effective amount of a compound of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
[0083] In one aspect, is a method for treating or preventing a LPA-dependent or LPA-mediated disease or condition in a mammal comprising administering a therapuetically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
[0084] In one aspect, LPA-dependent or LPA-mediated diseases or conditions include, but are not limited to, fibrosis of organs or tissues, scarring, liver diseases, dermatological conditions, cancer, cardiovascular disease, respiratory diseases or conditions, inflammatory disease, gastrointestinal tract disease, renal disease, urinary tract-associated disease, inflammatory disease of lower urinary tract, dysuria, frequent urination, pancreas disease, arterial obstruction, cerebral infarction, cerebral hemorrhage, pain, peripheral neuropathy, and fibromyalgia.
[0085] In some embodiments, the LPA-dependent or LPA-mediated disease or condition is selected from idiopathic pulmonary fibrosis; other diffuse parenchymal lung diseases of different etiologies including iatrogenic drug-induced fibrosis, occupational and/or environmental induced fibrosis, granulomatous diseases (sarcoidosis, hypersensitivity pneumonia), collagen vascular disease, alveolar proteinosis, langerhans cell granulomatosis, lymphangioleiomyomatosis, inherited diseases (Hermansky-Pudlak Syndrome, tuberous sclerosis, neurofibromatosis, metabolic storage disorders, familial interstitial lung disease); radiation induced fibrosis; chronic obstructive pulmonary disease (COPD); scleroderma; bleomycin induced pulmonary fibrosis; chronic asthma; silicosis; asbestos induced pulmonary fibrosis; acute respiratory distress syndrome (ARDS); kidney fibrosis;
tubulointerstitium fibrosis; glomerular nephritis; focal segmental glomerular sclerosis; IgA nephropathy; hypertension; Alport; gut fibrosis; liver fibrosis; cirrhosis; alcohol induced liver fibrosis; toxic/drug induced liver fibrosis; hemochromatosis; nonalcoholic steatohepatitis (NASH); biliary duct injury; primary biliary cirrhosis; infection induced liver fibrosis; viral induced liver fibrosis; and autoimmune hepatitis; corneal scarring; hypertrophic scarring; Duputren disease, keloids, cutaneous fibrosis; cutaneous scleroderma; spinal cord injury/fibrosis; myelofibrosis;
vascular restenosis; atherosclerosis; arteriosclerosis; Wegener's granulomatosis; Peyronie's disease, chronic lymphocytic leukemia, tumor metastasis, transplant organ rejection, endometreosis, neonatal respiratory distress syndrome and neuropathic pain.
[0086] In one aspect, is a method for treating or preventing cancer in a mammal comprising administering a therapeutically effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
[0087] In one aspect, is a method for treating or preventing fibrosis in a mammal comprising administering a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof. [0088] In one aspect, is a method for treating or preventing lung fibrosis, asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud's phenomenom, or neuropathic pain in a mammal comprising administering a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof.
[0089] In one aspect, provided is a method for the treatment or prevention of organ fibrosis in a mammal comprising administering a therapeutically effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to a mammal in need thereof. In some embodiments, the organ fibrosis comprises lung fibrosis, renal fibrosis, or hepatic fibrosis.
[0090] In one aspect, provided is a method of improving lung function in a mammal comprising administering a therapeutically effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), to the mammal in need thereof. In one aspect, the mammal has been diagnosed as having lung fibrosis.
[0091] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat idiopathic pulmonary fibrosis (usual interstitial pneumonia) in a mammal.
[0092] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat Raynaud's phenomenon. Raynaud's phenomenon comprises both Raynaud's disease (where the phenomenon is idiopathic) and Raynaud's syndrome, where it is caused by some other instigating factor.
[0093] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), is used to treat diffuse parenchymal interstitial lung diseases in mammal: iatrogenic drug induced, occupational/environmental (Farmer lung), granulomatous diseases (sarcoidosis, hypersensitivity pneumonia), collagen vascular disease (scleroderma and others), alveolar proteinosis, langerhans cell granulonmatosis, lymphangioleiomyomatosis, Hermansky-Pudlak Syndrome, Tuberous sclerosis, neurofibromatosis, metabolic storage disorders, familial interstitial lung disease.
[0094] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat post-transplant fibrosis associated with chronic rejection in a mammal (e.g. Bronchiolitis obliterans for lung transplant). [0095] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat cutaneous fibrosis in a mammal (e.g. cutaneous scleroderma, Dupuytren disease, keloids).
[0096] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat hepatic fibrosis with or without cirrhosis in a mammal: toxic/drug induced (hemochromatosis), alcoholic liver disease, viral hepatitis (hepatitis B virus, hepatitis C virus, HCV), nonalcoholic liver disease (NASH), metabolic and auto-immune.
[0097] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat renal fibrosis in a mammal: tubulointerstitium fibrosis, glomerular sclerosis.
[0098] In any of the aforementioned aspects involving the treatment of LPA dependent diseases or conditions are further embodiments comprising administering at least one additional agent in addition to the administration of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In various embodiments, each agent is administered in any order, including simultaneously.
[0099] In any of the embodiments disclosed herein, the mammal is a human.
[00100] In some embodiments, compounds provided herein are administered to a human. In some embodiments, compounds provided herein are orally administered to a human.
[00101] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), is used for inhibiting the activity of at least one LPA receptor or for the treatment of a disease or condition that would benefit from inhibition of the activity of at least one LPA receptor. In one aspect, the LPA receptor is LPA^
[00102] In other embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used for the formulation of a medicament for the inhibition of LPAi activity.
[00103] Also provided is an article of manufacture comprising multiple unit doses of an oral solid dosage form pharmaceutical composition described herein in a high-density polyethylene (HDPE) bottle equipped with a high-density polyethylene (HDPE) cap.
[00104] In some embodiments, high-density polyethylene (HDPE) bottle further comprises an aluminum foil induction seal and silica gel desiccant.
[00105] In any of the aforementioned embodiments are further embodiments comprising single administrations of the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), including further embodiments in which Compound 1, or a
pharmaceutically acceptable salt thereof (e.g. Compound 2) is (i) administered once-a-day; (ii) is administered twice-a-day; or (iii) is administered multiple times over the span of one day.
[00106] In any of the aforementioned embodiments are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the time between multiple administrations is every 8 hours; (iv) the time between multiple administrations is every 12 hours.
[00107] In some embodiments, the pharmaceutical composition is administered daily to the mammal.
[00108] In some embodiments, the pharmaceutical composition is administered in treatment cycles comprising: (a) a first period during which Compound 2 is administered daily to the mammal; and (b) a second period during which the Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered to the mammal in a reduced amount as compared to (a) or not administered.
[00109] In some embodiments, the methods of treatment or prevention disclosed herein comprise a drug holiday, wherein the administration of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is temporarily suspended or the dose being administered is temporarily reduced; at the end of the drug holiday dosing is resumed. In some embodiments, the length of the drug holiday varies from 2 days to 1 year.
[00110] Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) for treating any of the diseases or conditions disclosed herein. In some embodiments, Compound 1 is crystalline. In some embodiments, Compound 2 is crystalline. In some embodiments, Compound 2 is amorphous.
[00111] A pharmaceutical composition comprising Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) for use in any of the uses and methods disclosed herein.
[00112] Also described herein are process for the preparation of Compound 1 and pharmaceutically acceptable salts thereof. In one aspect, the pharmaceutically acceptable salt of Compound 1 is the sodium salt (Compound 2).
[00113] In one embodiment provided is a process for preparing crystalline {4'-[3-methyl-4-((R)-l- phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid (Compound 1) comprising isolating Compound 1 from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, or nitromethane.
[00114] In one embodiment provided is a process for preparing crystalline {4'-[3-methyl-4-((R)-l- phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid, sodium salt (Compound 2) comprising isolating Compound 2 from isopropanol.
[00115] In one embodiment provided is a process for the preparation of Compound 1 comprising the steps of: (1) reacting a compound of Formula IV:
Figure imgf000016_0001
IV wherein, X is a leaving group;
with a compound of Formula V:
Figure imgf000017_0001
wherein, R is Ci-C6 alkyl; and B is a boronic acid or boronate ester;
in the presence of a coupling catalyst, a suitable base, and in a suitable solvent, to provide a compound of Formula VI:
Figure imgf000017_0002
(2) hydrolysis of the ester moiety of the compound of Formula VI to provide Compound 1.
[00116] In one embodiment provided is a process for the preparation of Compound 1 comprising the steps of: (1) reacting a compound of Formula VII:
Figure imgf000017_0003
wherein, B is a boronic acid or boronate ester;
with a compound of Formula VIII:
Figure imgf000017_0004
wherein, R is C1-C6 alkyl; and X is a leaving group;
in the presence of a coupling catalyst, a suitable base, and in a suitable solvent, to provide a compound of Formula VI:
Figure imgf000018_0001
(2) hydrolysis of the ester moiety of the compound of Formula VI to provide Compound 1.
[00117] In some embodiments, the coupling catalyst is a palladium catalyst.
[00118] In some embodiments, the palladium catalyst is tetrakis(triphenylphosphine)palladium or (1,1 '-bis(diphenylphosphino)ferrocene)-dichloropalladium(II).
[00119] In some embodiments, R is -CH3 or -CH2CH3.
[00120] In some embodiments, the suitable base is triethylamine, diisopropylethylamine, 1,2,2,6,6- pentamethylpiperidine, tributylamine, sodium bicarbonate, Na2C03, K2CO3, CS2CO3, NaOAc, KOAc, Na3P04 or K3P04.
[00121] In some embodiments, the suitable solvent is tetrahydrofuran, dioxane, water, or combinations thereof.
[00122] In some embodiments, X is selected from CI, Br, I, -OS02CF3, -OS02(4-methylphenyl), - OS02(phenyl) and -OS02CH3. , or
Figure imgf000018_0002
[00124] In some embodiments, step (1) further comprises isolating the compound of Formula VI prior to step (2).
[00125] In some embodiments, step (1) further comprises a purification step for reducing the amount of palladium to less than 20ppm.
[00126] In some embodiments, the purification step for reducing the amount of palladium comprises treatment of the compound of Formula VI with thiol derivatized silica gel.
[00127] In some embodiments, the reaction of step (1) is heated to a temperature greater than 50°C.
[00128] In some embodiments, step (2) comprises treatment of the compound of Formula VI with sodium hydroxide in a suitable solvent followed by a pH adjustment. [00129] In one embodiments, provided is a process for the preparation of Compound 1 comprising the steps of: (1) treatment of a compound of Formula XII with diphenylphosphoryl azide in the presence of (R)-(+)- 1 -phenylethanol:
Figure imgf000019_0001
wherein, R is C1-C6 alkyl;
to provide a compound of Formula VI:
Figure imgf000019_0002
(2) hydrolysis of the ester moiety of the compound of Formula VI to provide Compound 1.
[00130] In some embodiments, step (2) comprises treatment of the compound of Formula VI with sodium hydroxide in a suitable solvent followed by a pH adjustment
[00131] The disclosed processes provide for the synthesis of Compound 1 and pharmaceutically acceptable salts thereof (e.g. Compound 2). The processes disclosed herein are particularly applicable to large scale chemical production of Compound 1 and pharmaceutically acceptable salts thereof.
[00132] In some embodiments, in any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), Compound 1, or a pharmaceutically acceptable salt thereof, is replaced with: a) Compound 1 , or a pharmaceutically acceptable salt thereof, of lower chiral purity; b) (4'-[3-methyl-4-((S)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid, or a pharmaceutically acceptable salt thereof of any optical purity; or c) racemic {4'-[3-methyl- 4-(l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid, or a pharmaceutically acceptable salt thereof.
[00133] In any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), amorphous Compound 1 is used. In any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), crystalline Compound 1 is used.
[00134] In any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), amorphous Compound 2 is used. In any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), crystalline Compound 2 is used.
[00135] In some embodiments, in any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), Compound 1, or a pharmaceutically acceptable salt thereof, is replaced with an active metabolite of Compound 1. In some embodiments, the active metabolite is in a crystalline form. In some embodiments, the active metabolite is in an amorphous phase. In some embodiments, in any of the embodiments disclosed herein (including methods, uses, formulations, combination therapy, etc.), Compound 1, or a pharmaceutically acceptable salt thereof, is replaced with a prodrug of Compound 1, or a deuterated analog of Compound 1, or a
pharmaceutically acceptable salt thereof.
[00136] Other objects, features and advantages of the methods and compositions described herein will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. BRIEF DESCRIPTION OF THE FIGURES
[00137] FIGURE 1 illustrates the XRPD of Pattern 1 of Compound 1.
[00138] FIGURE 2 illustrates the XRPD of Pattern 1 of Compound 2.
[00139] FIGURE 3 illustrates the XRPD of Pattern 2 of Compound 2.
[00140] FIGURE 4 illustrates the TGA and DSC of Crystalline Compound 1.
[00141] FIGURE 5 illustrates the VT-XRPD Diffractograms of crystalline Compound 1.
[00142] FIGURE 6 illustrates the cycling DSC thermogram of Crystalline Compound 1.
[00143] FIGURE 7 illustrates the TGA and DSC of Crystalline Compound 2.
[00144] FIGURE 8 illustrates the results of Compound 1 on dermal thickness in a mouse model of bleomycin-induced scleroderma.
[00145] FIGURE 9 illustrates the results of Compound 1 on collagen content in a mouse model of bleomycin-induced scleroderma.
DETAILED DESCRIPTION OF THE INVENTION
[00146] Lysophospholipids (such as lysophosphatidic acid (LP A)) affect fundamental cellular functions that include cellular proliferation, differentiation, survival, migration, adhesion, invasion, and morphogensis. These functions influence many biological processes that include neurogensis, angiogenesis, wound healing, immunity, and carcinogenesis.
[00147] LPA acts through sets of specific G protein-coupled receptors (GPCRs) in an autocrine and paracrine fashion. LPA binding to its cognate GPCRs (LPAi, LPA2, LPA3, LPA4, LPA5, LPA6) activates intracellular signaling pathways to produce a variety of biological responses.
[00148] LPA has a role as a biological effector molecule, and has a diverse range of physiological actions such as, but not limited to, effects on blood pressure, platelet activation, and smooth muscle contraction, and a variety of cellular effects, which include cell growth, cell rounding, neurite retraction, and actin stress fiber formation and cell migration. The effects of LPA are predominantly receptor mediated.
[00149] Activation of the LPA receptors with LPA mediates a range of downstream signaling cascades. The actual pathway and realized end point are dependent on a range of variables that include receptor usage, cell type, expression level of a receptor or signaling protein, and LPA concentration. Nearly all mammalian cells, tissues and organs co-express several LPA-receptor subtypes, which indicates that LPA receptors signal in a cooperative manner. LPAi, LPA2, and LPA3 share high amino acid sequence similarity.
[00150] LPA regulates many important functions of fibroblasts in wound healing, including proliferation, migration, differentiation and contraction. Fibroblast proliferation is required in wound healing in order to fill an open wound. In contrast, fibrosis is characterized by intense proliferation and accumulation of myofibroblasts that actively synthesize ECM and proinflammatory cytokines. LPA can either increase or suppress the proliferation of cell types important in wound healing.
[00151] Tissue injury initiates a complex series of host wound-healing responses; if successful, these responses restore normal tissue structure and function. If not, these responses can lead to tissue fibrosis and loss of function.
[00152] A number of muscular dystrophies are characterized by a progressive weakness and wasting of musculature, and by extensive fibrosis. It has been shown that LPA treatment of cultured myoblasts induced significant expression of connective tissue growth factor (CTGF). CTGF subsequently induces collagen, fibronectin and integrin expression and induces dedifferentiation of these myoblasts. Treatment of a variety of cell types with LPA induces reproducible and high level induction of CTGF. CTGF is a profibrotic cytokine, signaling down-stream and in parallel with TGF .
[00153] LPA and LPAi play key pathogenic roles in pulmonary fibrosis. Fibroblast chemoattractant activity plays an important role in the lungs in patients with pulmonary fibrosis. Profibrotic effects of LPAi-receptor stimulation is explained by LPAi -receptor-mediated vascular leakage and increased fibroblast recruitment, both profibrotic events. The LP A- LPAi pathway has a role in mediating fibroblast migration and vascular leakage in IPF. The end result is the aberrant healing process that characterises this fibrotic condition. In one aspect, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat pulmonary fibrosis in a mammal.
[00154] The LPA-LPA2 pathway contributes to the activation of the TGF-β pathway in pulmonary fibrosis. In some embodiments, compounds that inhibit LPA2 show efficacy in the treatment of lung fibrosis. In some embodiments, compounds that inhibit both LPAI and LPA2 show improved efficacy in the treatment of lung fibrosis compared to compounds which inhibit only LPAI or LPA2. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat lung fibrosis in a mammal.
[00155] LPA and LPAi are involved in the etiology of kidney fibrosis. In mice invalidated for the LPAi receptor (LPAi (~/~), the development of renal fibrosis was significantly attenuated. Unilateral ureteral obstruction (UUO; animal model of renal fibrosis) mice treated with the LPA receptor antagonist Ki 16425 closely resembled the LPAi (~/~) mice. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat kidney fibrosis in a mammal.
[00156] LPA is implicated in liver disease and fibrosis. Plasma LPA levels and serum autotoxin are elevated in hepatitis patients and animal models of liver injury in correlation with increased fibrosis. LPA also regulates liver cell function. LPAi and LPA2 receptors are expressed by mouse hepatic stellate cells and LPA stimulates migration of hepatic myofibroblasts. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat liver fibrosis in a mammal.
[00157] LPA is in involved in wound healing in the eye. LPAi and LP A3 receptors are detectable in the normal rabbit corneal epithelial cells, keratocytes and endothelial cells and LPAi and LPA3 expression are increased in corneal epithelial cells following injury.
[00158] LPA is present in the aqueous humor and the lacrimal gland fluid of the rabbit eye and these levels are increased in a rabbit corneal injury model.
[00159] LPA induces actin stress fiber formation in rabbit corneal endothelial and epithelial cells and promotes contraction corneal fibroblasts. LPA also stimulates proliferation of human retinal pigmented epithelial cells.
[00160] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat fibrosis involving tissues in the eye of a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat ocular fibrosis in a mammal.
[00161] LPA is implicated in myocardial infarction and cardiac fibrosis. Serum LPA levels are increased in patients following mycocardial infarction (MI) and LPA stimulates proliferation and collagen production (fibrosis) by rat cardiac fibroblasts. Both LPAI and LP A3 receptors are highly expressed in human heart tissue. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat cardiac fibrosis in a mammal.
[00162] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat or prevent fibrosis in a mammal. In one aspect, Compound 1, or a
pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat or prevent fibrosis of an organ or tissue in a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat or prevent fibrosis as described herein.
[00163] The terms "fibrosis" or "fibrosing disorder," as used herein, refers to conditions that are associated with the abnormal accumulation of cells and/or fibronectin and/or collagen and/or increased fibroblast recruitment and include but are not limited to fibrosis of individual organs or tissues such as the heart, kidney, liver, joints, lung, pleural tissue, peritoneal tissue, skin, cornea, retina, musculoskeletal and digestive tract.
[00164] Exemplary diseases, disorders, or conditions that involve fibrosis include, but are not limited to: Lung diseases associated with fibrosis, e.g., idiopathic pulmonary fibrosis, pulmonary fibrosis secondary to systemic inflammatory disease such as rheumatoid arthritis, scleroderma, lupus, cryptogenic fibrosing alveolitis, radiation induced fibrosis, chronic obstructive pulmonary disease (COPD), chronic asthma, silicosis, asbestos induced pulmonary or pleural fibrosis, acute lung injury and acute respiratory distress (including bacterial pneumonia induced, trauma induced, viral pneumonia induced, ventilator induced, non-pulmonary sepsis induced, and aspiration induced); Chronic nephropathies associated with injury/fibrosis (kidney fibrosis), e.g., glomerulonephritis secondary to systemic inflammatory diseases such as lupus and scleroderma, diabetes, glomerular nephritis, focal segmental glomerular sclerosis, IgA nephropathy, hypertension, allograft and Alport; Gut fibrosis, e.g., scleroderma, and radiation induced gut fibrosis; Liver fibrosis, e.g., cirrhosis, alcohol induced liver fibrosis, nonalcoholic steatohepatitis (NASH), biliary duct injury, primary biliary cirrhosis, infection or viral induced liver fibrosis (e.g., chronic HCV infection), and autoimmune hepatitis; Head and neck fibrosis, e.g., radiation induced; Corneal scarring, e.g., LASIK (laser-assisted in situ keratomileusis), corneal transplant, and trabeculectomy; Hypertrophic scarring and keloids, e.g., burn induced or surgical; and other fibrotic diseases, e.g., sarcoidosis, scleroderma, spinal cord injury/fibrosis, myelofibrosis, peritoneal fibrosis, vascular restenosis, atherosclerosis, arteriosclerosis, Wegener's granulomatosis, mixed connective tissue disease, and Peyronie's disease.
[00165] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat skin fibrosis in a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat scleroderma in a mammal.
[00166] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat peritoneal fibrosis in a mammal. [00167] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat spinal cord injury in a mammal.
[00168] In one aspect, a mammal suffering from one of the following non-limiting exemplary diseases, disorders, or conditions will benefit from therapy with Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2): atherosclerosis, thrombosis, heart disease, vasculitis, formation of scar tissue, restenosis, phlobitis, COPD (chronic obstructive pulmonary disease), pulmonary hypertension, pulmonary fibrosis, pulmonary inflammation, bowel adhesions, bladder fibrosis and cystitis, fibrosis of the nasal passages, sinusitis, inflammation mediated by neutrophils, and fibrosis mediated by fibroblasts.
[00169] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat a dermatological disorders in a mammal. Dermatological disorders include, but are not limited to, proliferative or inflammatory disorders of the skin such as, atopic dermatitis, bullous disorders, collagenoses, psoriasis, psoriatic lesions, scleroderma, dermatitis, contact dermatitis, eczema, urticaria, rosacea, scleroderma, wound healing, scarring, hypertrophic scarring, keloids, Kawasaki Disease, rosacea, Sjogren-Larsso Syndrome, urticaria.
[00170] LPA is released following tissue injury. LPAi plays a role in the initiation of neuropathic pain. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of pain in a mammal. In one aspect, the pain is acute pain or chronic pain. In another aspect, the pain is neuropathic pain. In another aspect, the pain is cancer pain. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of fibromylagia.
[00171] Lysophospholipid receptor signaling plays a role in the etiology of cancer. Lysophosphatidic acid (LPA) and its G protein-coupled receptors (GPCRs) LPAi, LPA2, and/or LPA3 play a role in the development of several types of cancers.
[00172] LPA contributes to tumorigenesis by increasing motility and invasiveness of cells. LPA has been implicated in the initiation or progression of ovarian cancer. LPA is present at significant concentrations (2-80 μΜ) in the ascitic fluid of ovarian cancer patients. LPA receptors (LPA2 and LPA3) are also overexpressed in ovarian cancer cells as compared to normal ovarian surface epithelial cells. LPA has also been implicated in the initiation or progression of prostate cancer, breast cancer, melanoma, head and neck cancer, bowel cancer (colorectal cancer), thyroid cancer, glioblastoma, and other cancers.
[00173] LPA receptors mediate both migration of and invasion by pancreatic cancer cell lines:
Ki 16425 and LPA specific siRNA effectively blocked in vitro migration in response to LPA and peritoneal fluid (ascites) from pancreatic cancer patients; in addition, Ki 16425 blocked the LPA- induced and ascites-induced invasion activity of a highly peritoneal metastatic pancreatic cancer cell line (Yamada et al, J. Biol. Chem., 279, 6595-6605, 2004). [00174] Colorectal carcinoma cell lines show significant expression of LPAi mRNA and respond to LPA by cell migration and production of angiogenic factors. Overexpression of LPA receptors has a role in the pathogenesis of thyroid cancer. LP A3 was originally cloned from prostate cancer cells, concordant with the ability of LPA to induce autocrine proliferation of prostate cancer cells.
[00175] LPA has stimulatory roles in cancer progression in many types of cancer. LPA is produced from and induces proliferation of prostate cancer cell lines. LPA induces human colon carcinoma DLD 1 cell proliferation, migration, adhesion, and secretion of angiogenic factors through LPAi signalling. In other human colon carcinoma cells lines (HT29 and WiDR), LPA enhances cell proliferation and secretion of angiogenic factors. In other colon cancer cell lines, LPA2 and LPA3 receptor activation results in proliferation of the cells. LPAi is implicated in bone metastasis (Boucharaba et al, Proc. Natl. Acad. Sci USA , 103, 9643-9648, 2006).
[00176] In one aspect, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of cancer in a mammal. In one aspect, Compound 1 , or a
pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of malignant and benign proliferative disease in a mammal. In one aspect, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to prevent or reduce proliferation of tumor cells, invasion and metastasis of carcinomas, pleural mesothelioma or peritoneal mesothelioma, cancer pain, bone metastases. In one aspect is a method of treating cancer in a mammal, the method comprising administering to the mammal Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), and a second therapeutic agent, wherein the second therapeutic agent is an anticancer agent. In some embodiments, radiation therapy is also used.
[00177] The types of cancer include, but is not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias) at any stage of the disease with or without metastases.
[00178] Additional non-limiting examples of cancers include, acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, anal cancer, appendix cancer, astrocytomas, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (osteosarcoma and malignant fibrous histiocytoma), brain stem glioma, brain tumors, brain and spinal cord tumors, breast cancer, bronchial tumors, Burkitt lymphoma, cervical cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-Cell lymphoma, embryonal tumors, endometrial cancer, ependymoblastoma, ependymoma, esophageal cancer, ewing sarcoma family of tumors, eye cancer, retinoblastoma, gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), gastrointestinal stromal cell tumor, germ cell tumor, glioma, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors (endocrine pancreas), Kaposi sarcoma, kidney cancer, Langerhans cell histiocytosis, laryngeal cancer, leukemia, Acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, liver cancer, non-small cell lung cancer, small cell lung cancer, Burkitt lymphoma, cutaneous T-cell lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, lymphoma, Waldenstrom macroglobulinemia, medulloblastoma, medulloepithelioma, melanoma, mesothelioma, mouth cancer, chronic myelogenous leukemia, myeloid leukemia, multiple myeloma, nasopharyngeal cancer,
neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma, malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, papillomatosis, parathyroid cancer, penile cancer, pharyngeal cancer, pineal parenchymal tumors of intermediate differentiation, pineoblastoma and supratentorial primitive neuroectodermal tumors, pituitary tumor, plasma cell neoplasm/multiple myeloma, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell (kidney) cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, Ewing sarcoma family of tumors, sarcoma, kaposi, Sezary syndrome, skin cancer, small cell Lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, stomach (gastric) cancer, supratentorial primitive neuroectodermal tumors, T-cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, urethral cancer, uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, Wilms tumor.
[00179] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, leukemia, lymphoma or tumor metastasis in a mammal.
[00180] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, Hodgkin lymphoma, and follicular lymphoma, multiple myeloma, chronic lymphocytic leukemia, or tumor metastasis in a mammal.
[00181] In one aspect, LPA is a contributor to the pathogenesis of respiratory diseases.
Proinflammatory effects of LPA include degranulation of mast cells, contraction of smooth-muscle cells and release of cytokines from dendritic cells. LPA induces the secretion of IL-8 from human bronchial epithelial cells. IL-8 is found in increased concentrations in BAL fluids from patients with asthma, chronic obstructive lung disease, pulmonary sarcoidosis and acute respiratory distress syndrome and 11-8 has been shown to exacerbate airway inflammation and airway remodeling of asthmatics. LPA1, LPA2 and LP A3 receptors have all been shown to contribute to the LPA-induced IL-8 production.
[00182] Administration of LP A in vivo induces airway hyper-responsiveness, itch-scratch responses, infiltration and activation of eosinophils and neutrophils, vascular remodeling, and nociceptive flexor responses. LPA also induces histamine release from mouse and rat mast cells. In one aspect, the effects of LPA are mediated through LPAi and/or LPA3. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of various allergic disorders in a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of respiratory diseases, disorders or conditions in a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), is used in the treatment of asthma in a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used in the treatment of chronic asthma in a mammal.
[00183] The term "respiratory disease," as used herein, refers to diseases affecting the organs that are involved in breathing, such as the nose, throat, larynx, eustachian tubes, trachea, bronchi, lungs, related muscles (e.g., diaphram and intercostals), and nerves. Respiratory diseases include, but are not limited to, asthma, adult respiratory distress syndrome and allergic (extrinsic) asthma, non- allergic (intrinsic) asthma, acute severe asthma, chronic asthma, clinical asthma, nocturnal asthma, allergen-induced asthma, aspirin-sensitive asthma, exercise-induced asthma, isocapnic
hyperventilation, child-onset asthma, adult-onset asthma, cough- variant asthma, occupational asthma, steroid-resistant asthma, seasonal asthma, seasonal allergic rhinitis, perennial allergic rhinitis, chronic obstructive pulmonary disease, including chronic bronchitis or emphysema, pulmonary hypertension, interstitial lung fibrosis and/or airway inflammation and cystic fibrosis, and hypoxia.
[00184] In one aspect, presented herein is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of chronic obstructive pulmonary disease in a mammal comprising administering to the mammal at least once an effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In addition, chronic obstructive pulmonary disease includes, but is not limited to, chronic bronchitis or emphysema, pulmonary hypertension, interstitial lung fibrosis and/or airway inflammation, and cystic fibrosis.
[00185] The nervous system is a major locus for LPAi expression. In one aspect, provided is Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), for use in the treatment or prevention of a nervous system disorder in a mammal. The term "nervous system disorder," as used herein includes, but is not limited to, Alzheimer's Disease, cerebral edema, cerebral ischemia, stroke, multiple sclerosis, neuropathies, Parkinson's Disease, multiple sclerosis, retinal ischemia, post-surgical cognitive dysfunction, migraine, peripheral neuropathy/neuropathic pain, spinal cord injury, cerebral edema and head injury.
[00186] Angiogenesis, the formation of new capillary networks from pre-existing vasculature, is normally invoked in wound healing, tissue growth and myocardial angiogenesis after ischemic injury. Peptide growth factors and lysophospholipids control coordinated proliferation, migration, adhesion, differentiation and assembly of vascular endothelial cells (VECs) and surrounding vascular smooth-muscle cells (VSMCs). In one aspect, dysregulation of the processes mediating angiogenesis leads to atherosclerosis, hypertension, tumor growth, rheumatoid arthritis and diabetic retinopathy.
[00187] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat or prevent cardiovascular disease in mammal, including but not limited to:
arrhythmia (atrial or ventricular or both); atherosclerosis and its sequelae; angina; cardiac rhythm disturbances; myocardial ischemia; myocardial infarction; cardiac or vascular aneurysm; vasculitis, stroke; peripheral obstructive arteriopathy of a limb, an organ, or a tissue; reperfusion injury following ischemia of the brain, heart, kidney or other organ or tissue; endotoxic, surgical, or traumatic shock; hypertension, valvular heart disease, heart failure, abnormal blood pressure; shock; vasoconstriction (including that associated with migraines); vascular abnormality, inflammation, insufficiency limited to a single organ or tissue.
[00188] In one aspect, provided herein are methods for preventing or treating vasoconstriction, atherosclerosis and its sequelae myocardial ischemia, myocardial infarction, aortic aneurysm, vasculitis and stroke comprising administering at least once to the mammal an effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In some
embodiments, the LPA antagonist disclosed herein is used to treat Raynaud's phenomenon.
Raynaud's phenomenon comprises both Raynaud's disease (where the phenomenon is idiopathic) and Raynaud's syndrome, where it is caused by some other instigating factor.
[00189] In one aspect, provided herein are methods for reducing cardiac reperfusion injury following myocardial ischemia and/or endotoxic shock comprising administering at least once to the mammal an effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2)·
[00190] In one aspect, provided herein are methods for reducing the constriction of blood vessels in a mammal comprising administering at least once to the mammal an effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
[00191] In one aspect, provided herein are methods for lowering or preventing an increase in blood pressure of a mammal comprising administering at least once to the mammal an effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). [00192] LPA is associated with various inflammatory/immune diseases. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat or prevent inflammation in a mammal. In one aspect, antagonists of LPA i and/or LP A3 find use in the treatment or prevention of inflammatory/immune disorders in a mammal.
[00193] Examples of inflammatory/immune disorders include psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, asthma, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, eczema, allogeneic or xenogeneic transplantation (organ, bone marrow, stem cells and other cells and tissues) graft rejection, graft- versus-host disease, lupus erythematosus, inflammatory disease, type I diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis (e.g., Hashimoto's and autoimmune thyroiditis), myasthenia gravis, autoimmune hemolytic anemia, multiple sclerosis, cystic fibrosis, chronic relapsing hepatitis, primary biliary cirrhosis, allergic conjunctivitis and atopic dermatitis.
[00194] In accordance with one aspect, are methods for treating, preventing, reversing, halting or slowing the progression of LPA-dependent or LPA-mediated diseases or conditions once it becomes clinically evident, or treating the symptoms associated with or related to LPA-dependent or LPA- mediated diseases or conditions, by administering to the mammal Compound 1 , or a
pharmaceutically acceptable salt thereof (e.g. Compound 2). In certain embodiments, the subject already has a LPA-dependent or LPA-mediated disease or condition at the time of administration, or is at risk of developing a LPA-dependent or LPA-mediated disease or condition.
[00195] In certain aspects, are methods for preventing or treating eosinophil and/or basophil and/or dendritic cell and/or neutrophil and/or monocyte and/or T-cell recruitment comprising administering at least once to the mammal an effective amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
[00196] In certain aspects, are methods for the treatment of cystitis, including, e.g., interstitial cystitis, comprising administering at least once to the mammal a therapeutically effective amount of
Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2).
[00197] In accordance with one aspect, methods described herein include the diagnosis or determination of whether or not a patient is suffering from a LPA-dependent or LPA-mediated disease or condition by administering to the subject a therapeutically effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), and determining whether or not the patient responds to the treatment.
[00198] In one aspect provided herein is Compound 1, pharmaceutically acceptable salts, pharmaceutically acceptable prodrugs, and pharmaceutically acceptable solvates thereof, which is an antagonist of at least one LPA receptor (e.g. LPAi, LPA2, LP A3) and is used to treat patients suffering from one or more LPA-dependent or LPA-mediated conditions or diseases, including, but not limited to, lung fibrosis, kindney fibrosis, liver fibrosis, scarring, scleroderma, asthma, rhinitis, chronic obstructive pulmonary disease, pulmonary hypertension, interstitial lung fibrosis, arthritis, allergy, psoriasis, inflammatory bowel disease, adult respiratory distress syndrome, myocardial infarction, aneurysm, stroke, cancer, pain, proliferative disorders and inflammatory conditions. In some embodiments, LPA-dependent conditions or diseases include those wherein an absolute or relative excess of LPA is present and/or observed.
[00199] In any of the aforementioned aspects the LPA-dependent or LPA-mediated diseases or conditions include, but are not limited to, organ fibrosis, tissue fibrosis, asthma, allergic disorders, chronic obstructive pulmonary disease, pulmonary hypertension, lung or pleural fibrosis, peritoneal fibrosis, arthritis, allergy, cancer, cardiovascular disease, aldult respiratory distress syndrome, myocardial infarction, aneurysm, stroke, and cancer.
[00200] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to improve the corneal sensitivity decrease caused by corneal operations such as laser- assisted in situ keratomileusis (LASIK) or cataract operation, corneal sensitivity decrease caused by corneal degeneration, and dry eye symptom caused thereby.
[00201] In one aspect, presented herein is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of ocular inflammation and allergic conjunctivitis, vernal keratoconjunctivitis, and papillary conjunctivitis in a mammal.
[00202] In one aspect, presented herein is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of Sjogren disease or inflammatory disease with dry eyes in a mammal.
[00203] In one aspect, LPA and LPA receptors (e.g. LPAi) are involved in the pathogenesis of osteoarthritis. In one aspect, presented herein is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of osteoarthritis in a mammal.
[00204] In one aspect, LPA receptors (e.g. LPAi, LPA3) contribute to the pathogenesis of rheumatoid arthritis. In one aspect, presented herein is the use of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the treatment or prevention of rheumatoid arthritis in a mammal.
[00205] In one aspect, LPA receptors (e.g. LPAi) contribute to adipogenesis. In one aspect, presented herein is the use of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), in the promotion of adipose tissue formation in a mammal.
[00206] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is used to treat Raynaud's phenomenon in a mammal. Raynaud's phenomenon comprises both Raynaud's disease (where the phenomenon is idiopathic) and Raynaud's syndrome, where it is caused by some instigating factor. [00207] Described herein are compositions, pharmaceutical compositions, methods for treating, methods for formulating, methods for producing, methods for manufacturing, treatment strategies, pharmacokinetic strategies using Compound 1 , or pharmaceutically acceptable salts thereof.
Compound 1, and Pharmaceutically Acceptable Salts Thereof
[00208] "Compound 1" or "{4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid" or "(R)- 1 -phenyl ethyl 5-(4-biphenyl acetic acid)-3-methylisoxazole-4- ylcarbamate" or any other similar name refers to the compound with the following structure:
Figure imgf000031_0001
[00209] In some embodiments, Compound 1 is substantially free of the S-isomer.
[00210] "Substantially free" with respect to an enantiomer, means that the referenced enantiomer is not present or there is less than 5%, less than 4%, less than 3%, less than 2% or less than 1% of the referenced enantiomer.
[00211] In some embodiments, samples of Compound 1 were found to have greater than 85% e.e., greater than 90% e.e., greater than 91% e.e., greater than 92% e.e., greater than 93% e.e., greater than 94% e.e., greater than 95% e.e., greater than 96% e.e., greater than 97% e.e., greater than 98%) e.e., or greater than 99%> e.e.
[00212] "Compound 2" or "{4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid, sodium salt" or "sodium {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetate" or any other similar name refers to the compound with the following structure:
Figure imgf000031_0002
[00213] In some embodiments, Compound 2 is substantially free of the S-isomer.
[00214] In some embodiments, samples of Compound 2 were found to have greater than 85% e.e., greater than 90% e.e., greater than 91% e.e., greater than 92% e.e., greater than 93 %> e.e., greater than 94% e.e., greater than 95% e.e., greater than 96% e.e., greater than 97% e.e., greater than 98%) e.e., or greater than 99% e.e.
[00215] A wide variety of pharmaceutically acceptable salts are formed from Compound 1 and include:
[00216] - salts formed when the acidic proton of the carboxylic acid of Compound 1 is replaced by a metal ion, such as for example, an alkali metal ion (e.g. lithium, sodium, potassium), an alkaline earth ion (e.g. magnesium, or calcium), or an aluminum ion, or is replaced by an ammonium cation ( H4 +).
[00217] - salts formed by reacting Compound 1 with a pharmaceutically acceptable organic base, which includes alkylamines, such as choline, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine, and salts with amino acids, such as arginine, lysine, and the like.
[00218] Salts of Compound 1 include, but are not limited to, a lithium salt, a sodium salt, a potassium salt, a magnesium salt, a calcium salt, an aluminum salt, an ammonium salt, a choline, an ethanolamine salt, a diethanolamine salt, a triethanolamine salt, a tromethamine salt, a N- methylglucamine salt, a dicyclohexylamine salt, a tris(hydroxymethyl)methylamine salt, an arginine salt, or a lysine salt.
[00219] In some embodiments, Compound 1 is treated with an amino acid to form a salt.
[00220] In other embodiments, Compound 1 is treated with choline, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, arginine, lysine, ammonium hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like to form a salt.
[00221] The term "pharmaceutically acceptable salt" in reference to Compound 1 refers to a salt of Compound 1 , which does not cause significant irritation to a mammal to which it is administered and does not substantially abrogate the biological activity and properties of the compound.
[00222] It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms (solvates). Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are formed during the process of product formation or isolation with pharmaceutically acceptable solvents such as water, ethanol, methyl tert-butyl ether, isopropanol, acetonitrile, heptane, and the like. In one aspect, solvates are formed using, but not limited to, Class 3 solvent(s). Categories of solvents are defined in, for example, the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), "Impurities: Guidelines for Residual Solvents, Q3C(R3), (November 2005). Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. In one embodiment, solvates of Compound 1 , or salts thereof, are conveniently prepared or formed during the processes described herein. In addition, Compound 1, or salts thereof, exist in unsolvated form.
[00223] In yet other embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is prepared in various forms, including but not limited to, amorphous phase, milled forms and nano-particulate forms.
Amorphous Compound 1
[00224] In some embodiments, Compound 1 is amorphous. In some embodiments, Amorphous Phase of Compound 1 has an XRPD pattern showing a lack of crystallinity.
Compound 1 - Pattern 1
[00225] In some embodiments, Compound 1 is crystalline. In some embodiments, Compound 1 is crystalline Pattern 1.
[00226] In some embodiments, the crystalline form of Compound 1 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1 ; a differential scanning calorimetry (DSC) thermogram with endotherms at about 157°C and about 198°C; a differential scanning calorimetry (DSC) thermogram
substantially the same as Figure 4; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 4; substantially the same X-ray powder diffraction (XRPD) pattern post storage at 40 °C/75% RH for one week; or combinations thereof. In some embodiments, the crystalline form of Compound 1 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least four of the preceding properties. In some embodiments, the crystalline form of Compound 1 is characterized as having at least five of the preceding properties. In some
embodiments, the crystalline form of Compound 1 is characterized as having at all of the preceding properties.
[00227] In some embodiments, crystalline Pattern 1 of Compound 1 is characterized as having:
(a) an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2- Theta, 21.7° 2-Theta, and 23.0° 2-Theta;
(b) an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.2° 2-Theta, 9.2° 2- Theta, 10.6° 2-Theta, 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta;
(c) an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1 ;
(d) a DSC thermogram with endotherms at about 165°C and about 199°C.
(e) a DSC thermogram with endotherms at about 157°C and about 198°C. (f) substantially the same X-ray powder diffraction (XRPD) pattern post storage at 40 C/75% RH for one week; or
(g) combinations thereof.
[00228] In some embodiments, Crystalline Pattern 1 of Compound 1 is obtained from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, nitromethane, tetrahydrofuran, dioxane, methyl ethyl ketone, acetone, 2-methoxyethanol, or methanol.
[00229] In some embodiments, Crystalline Pattern 1 of Compound 1 is characterized as having at least two of the properties selected from (a) to (f). In some embodiments, Crystalline Pattern 1 of Compound 1 is characterized as having at least three of the properties selected from (a) to (f). In some embodiments, Crystalline Pattern 1 of Compound 1 is characterized as having at least four of the properties selected from (a) to (f).
Amorphous Compound 2
[00230] In some embodiments, Compound 2 is amorphous. In some embodiments, Amorphous Phase of Compound 2 has an XRPD pattern showing a lack of crystallinity.
Compound 2 - Pattern 1
[00231] In some embodiments, Compound 2 is crystalline. In some embodiments, Compound 2 is crystalline Pattern 1.
[00232] In some embodiments, crystalline Pattern 1 of Compound 2 is characterized as having: an X- ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2-Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C; a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7; or combinations thereof. In some embodiments, the crystalline form of Compound 2 is characterized as having at least two of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least three of the preceding properties. In some embodiments, the crystalline form of Compound 2 is characterized as having at least four of the preceding properties. In some
embodiments, the crystalline form of Compound 2 is characterized as having at all of the preceding properties.
[00233] In some embodiments, crystalline Pattern 1 of Compound 2 is characterized as having at least one of the following properties:
(a) an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta and 15.8° 2-Theta. (b) an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2- Theta, 15.8° 2-Theta, 17.1° 2-Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta.
(c) an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2.
[00234] In some embodiments, Crystalline Pattern 1 of Compound 2 is characterized as having at least two of the properties selected from (a) to (c). In some embodiments, Crystalline Pattern 1 of Compound 2 is characterized as having properties (a), (b) and (c).
[00235] In some embodiments, Crystalline Pattern 1 of Compound 2 is obtained from isopropanol.
Compound 2 - Pattern 2
[00236] In some embodiments, Compound 2 is crystalline. In some embodiments, Compound 2 is crystalline Pattern 2. Crystalline Pattern 2 of Compound 2 is characterized as having at least one of the following properties:
(a) an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 8.5° 2-Theta, 9.3° 2- Theta, 16.6° 2-Theta, 17.1° 2-Theta and 21.4° 2-Theta;
(b) an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 3.
[00237] In some embodiments, Crystalline Pattern 2 of Compound 2 is obtained from isopropanol.
Prodrugs of Compound 1
[00238] In some embodiments, Compound 1 is prepared as a prodrug.
[00239] A "prodrug of Compound 1" refers to a compound that is converted into Compound 1 in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. In some embodiments, prodrugs facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial. An example, without limitation, of a prodrug would be an ester of Compound 1 (the "prodrug"). A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
[00240] Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. In certain embodiments, the prodrug of Compound 1 increases the bioavailability of
Compound 1 when orally administered. In some embodiments, the prodrug of Compound 1 has improved solubility in pharmaceutical compositions over Compound 1. [00241] In some embodiments, a prodrug of Compound 1 is an alkyl ester of Compound 1, such as, for example, methyl ester, ethyl ester, n-propyl ester, iso-propyl ester, n-butyl ester, sec-butyl ester, or tert-butyl ester.
242] Non- limiting examples of prodrugs of Compound 1 include:
Figure imgf000036_0001
Metabolites of Compound 1
[00243] Compound 1 metabolites formed during incubation of Compound 1 with rat, dog, and human liver microsomes, rat and human hepatocytes, as well as those generated in vivo and isolated from rat bile and rat and dog plasma have been investigated. Authentic standards of the majority of the metabolites have been chemically synthesized. The identity of the in vitro and in vivo metabolites were confirmed by comparison with the authentic standard and/or by the fragmentation pattern observed following LC-MS/MS analysis. [00244] The following metabolites of Compound 1 were observed both in vitro and in vivo:
Ml - Taurine conjugate of Compound 1 (2-(2-{4'-[3-Methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetylamino)-ethanesulfonic acid);
M2 - Oxidation of phenyl ring of benzyl group of Compound 1.
M3 - M4 + taurine conjugation
M4 - [4'-(4-Amino-3-methyl-isoxazol-5-yl)-biphenyl-4-yl]-acetic acid
M5 - Oxidation of biphenyl of Compound 1
M6 - Glucuronidation of Compound 1
M7 - M4 + glycosylation
M8 - Glucuronidation of M7
[00245] In some embodiments, sites on Compound 1 are susceptible to various metabolic reactions. Therefore incorporation of appropriate substituents on Compound 1 will reduce, minimize or eliminate this metabolic pathway. In specific embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, deuterium or an alkyl group (e.g. methyl, ethyl).
[00246] In some embodiments, Compound 1 is isotopically labeled (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels. In some embodiments, Compound 1 is isotopically-labeled, which is identical to Compound 1 but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. In some embodiments, one or more hydrogen atoms are replaced with deuterium. In some embodiments, metabolic sites on Compound 1 are deuterated. In some embodiments, substitution with deuterium affords certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements.
[00247] In one aspect, described is a compound with the following structure:
Figure imgf000037_0001
wherein, each R is independently selected from hydrogen or deutrium,
or a pharmaceutically acceptable salt thereof.
[00248] In some embodiments, the pharmaceutically acceptable salt of the compound is a sodium salt.
Synthesis of Compound 1, and Pharmaceutically Acceptable Salts Thereof
[00249] Compound 1, and pharmaceutically acceptable salts thereof (e.g. Compound 2), are synthesized as described herein. In additions, solvents, temperatures and other reaction conditions presented herein may vary.
[00250] The starting materials used for the synthesis are either synthesized or obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Fluka, Acros Organics, Alfa Aesar, VWR Scientific, and the like. General methods for the preparation of compounds can be modified by the use of appropriate reagents and conditions for the introduction of the various moieties found in the formulae as provided herein.
[00251] In one aspect, the preparation of Compound 1, or pharmaceutically acceptable salts thereof (e.g. sodium salt) begins with the steps outlined in Scheme 1.
Scheme 1.
Figure imgf000038_0001
pyridine, THF
[00252] In one aspect, the synthesis of compound 1 begins with the reaction of an alkyl acetoacetate, such as methyl acetoacetate, with methylamine to provide 3-methylamino-but-2-enoic acid methyl ester, which is then reacted with benzoyl chlorides, such as 4-bromobenzoyl chloride, to provide compound I. X is halide, triflate or any other suitable leaving group for use in a Suzuki coupling reaction. In some embodiments, X is -CI, -Br, -I, -OSO2CF3, -OS02(4-methylphenyl), and - OSO2CH3. In some embodiments, X is a halide. In some embodiments, X is -Br. In some embodiments, R1 is an alkyl or benzyl. In some embodiments, R1 is methyl, ethyl, propyl, or benzyl. Other alkyl acetoacetates contemplated include, ethyl acetoacetate, isopropyl acetoacetate, benzyl acetoacetate. Treatment of compounds of structure I with hydroxyl amine and acetic acid provides isoxazoles of structure II. Hydrolysis of the ester group of isoxazoles of structure II provides carboxylic acid III. Hydrolysis can also be accomplished with the use of other bases, such as sodium hydroxide.
Scheme 2.
Figure imgf000039_0001
[00253] A Curtius rearrangement of carboxylic acid III in the presence of (R)- 1 -phenyl ethyl alcohol provides carbamate compound IV.
Scheme 3.
Figure imgf000039_0002
[00254] In some embodiments, a Suzuki reaction between carbamate compound IV and compounds of structure V is used to provide compounds of structure VI. In some embodiments, B is boronic embodiments, X is
Figure imgf000039_0003
, and In some embodiments, . In some embodiments, X is -CI, -Br, -I, -OSO2CF3, -OS02(4-methylphenyl), and -OS02CH3. In some embodiments, X is a halide. In some embodiments, X is -Br. In some embodiments, the Suzuki reaction includes the use of a palladium catalyst, a suitable base and a suitable solvent. In some embodiments, the palladium catalyst is a phosphine containing palladium catalyst. In some embodiments, the palladium catalyst is Pd(PPh3)4 or Pd(dppf)Cl2. In some embodiments, the suitable base for the Suzuki reaction is an inorganic base. In some embodiments, the suitable base for the Suzuki reaction is triethylamine, diisopropylethylamine, 1,2,2,6,6-pentamethylpiperidine, tributylamine, sodium bicarbonate, Na2C03, K2C03, Cs2C03, NaOAc, KOAc, Na3P04 or K3P04. Other metal mediated coupling reactions are known for the preparation of compounds of structure VI.
[00255] Other metal mediated coupling reactions to form biaryls include, but are not limited to Suzuki reactions, Stille cross couplings, Negishi couplings, Kumada couplings, Ullmann reactions, Hiyama Coupling, and variants thereof (Metal-Catalyzed Cross-Coupling Reactions, Armin de Meijere (Editor), Francois Diederich (Editor), John Wiley & Sons; 2nd edition, 2004; Ozdemir, et al, Tetrahedron, 2005, 61, 9791-9798; Ackermann, et al, Org. Lett., 2006, 8, 3457-3460; Blakey, et al, J. Am. Chem. Soc, 2003, 125, 6046-6047; Dai, et al, Org. Lett., 2004, 6, 221-224; Yoshikai, et al, J. Am. Chem. Soc, 2005, 127, 17978-17979; Tang, et al, J. Org. Chem., 2006, 71, 2167-2169; Murata, et al, Synthesis, 2001, 2231-2233).
Scheme 4.
Figure imgf000040_0001
[00256] In some embodiments, compounds of structure VII are reacted with a borylating agent using transition metal mediated reaction conditions to form boronate compounds of structure IX. In some embodiments, the borylating reaction to form IX includes the use of a palladium catalyst, such as Pd(PPh3)4 or Pd(dppf)Cl2, in the presence of a suitable base, such as potassium acetate. In some embodiments, the borylation reagent is selected from among pinacolborane, catecholborane, bis(neopentyl glycolato)diboron, bis(pinacolato)diboron, bis(hexylene glycolato)diboron, and bis(catecholato)diboron. In some embodiments, the borylation reagent is bis(pinacolato)diboron. In some embodiments, the borylation reaction is performed with heating. Boronate compounds of structure VII are reacted with compounds of structure VIII under palladium mediated coupling conditions (Suzuki reaction conditions) to form compounds of structure VI.
[00257] An alternative route to the synthesis of compound VI is outlined in Scheme 5.
Scheme 5.
Figure imgf000040_0002
[00258] In some embodiments, biphenyl compounds of structure X are elaborated into compound VI as shown in Scheme 4. In some embodiments, R is ethyl. In some embodiments, R is isopropyl. Biphenyl compounds of structure X are treated with acetyl chloride in the presence of a suitable Lewis acid, follow by conversion of the acetyl group to the carboxylic acid and treatment of the carboxylic acid with thionyl chloride to provide acid chlorides of structure XI. Acid chlorides of structure XI are then used to prepare isoxazoles of structure XII as described in Scheme 1. In some embodiments, R1 is an alkyl group. In some embodiments, R1 is methyl and R1 is removed from the isoxazoles under hydrolysis conditions to provide compounds of structure XII. In some embodiments, R1 is benzyl and R1 is removed from the isoxazoles under hydrogenation conditions (e.g. H2, Pd/C) to provide compounds of structure XII. A Curtius rearrangement of compounds of structure XII in the presence of 1 -phenyl ethyl alcohol provides carbamate compounds of structure VI.
[00259] An additional alternative route to the synthesis of compound VI is outlined in Scheme 6. Scheme 6
Figure imgf000041_0001
[00260] Conversion of the X group in biphenyl compound XIV to a boronic acid or boronate ester produces a coupling partner for compound XV in a Suzuki reaction that provides compound VI.
[00261] The hydrolysis of the ester group in compound VI to provide Compound 1 and Compound 2 is outlined in Scheme 7.
Scheme 7
Figure imgf000041_0002
i Compound 1 Compound 2
[00262] Hydrolysis of alkyl esters of structure VI with a suitable base in a suitable solvent yields compound 1 after pH adjustment. Suitable bases for the hydrolysis include, but are not limited to, lithium hydroxide and sodium hydroxide. Suitable solvents for the hydrolysis include, but are not limited to, water, methanol, ethanol, tetrahydrofuran, or combinations thereof. Compound 1 is then treated with sodium hydroxide in ethanol to furnish Compound 2.
[00263] In some embodiments, Compound 2 is prepared from compound VI by performing a one- step hydrolysis and salt forming reaction. In some embodiments, the one-step hydrolysis and salt forming reaction includes treatment of compound VI with sodium hydroxide in a suitable solvent.
[00264] In some embodiments, Compound 1 is treated with potassium hydroxide in a solvent to form Compound 1, potassium salt. In some embodiments, Compound 1 is treated with lithium hydroxide in a solvent to form Compound 1, lithium salt. In some embodiments, Compound 1 is treated with calcium hydroxide in a solvent to form Compound 1, calcium salt.
[00265] In some embodiments, Compound 1 is treated with dicyclohexylamine in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with N-methyl-D-glucamine in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with choline in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with tris(hydroxymethyl)methylamine in a solvent to form the corresponding salt.
[00266] In some embodiments, Compound 1 is treated with arginine in a solvent to form the corresponding salt. In some embodiments, Compound 1 is treated with lysine in a solvent to form the corresponding salt.
[00267] Due to the fact that that synthetic methods described above utilize a transition metal catalyst, purification steps are performed to reduce the amount of palladium in the product. Purification steps to reduce the amount of palladium in a product are conducted so that active pharmaceutical ingredients meet palladium specification guidelines. ("Guideline on the Specification Limits for Residues of Metal Catalysts" European Medicines Agency Pre-authorisation Evaluation of
Medicines for Human Use, London, January 2007, Doc. Ref. CPMP/SWP/QWP/4446/00 corr.). In some embodiments, purification steps to reduce the amount of palladium in a product includes, but is not limited to, treatment with solid trimercaptotriazine (TMT), polystyrene-bound TMT, mercapto- porous polystyrene-bound TMT, polystyrene-bound ethylenediamine, activated carbon, glass bead sponges, Smopex™, silica bound scavengers, thiol-derivatized silica gel, N-acetylcysteine, n-Bu3P, crystallization, extraction, 1-cysteine, n-Bu3P/lactic acid. (Garrett et ah, Adv. Synth. Catal. 2004, 346, 889-900). In some embodiments, activated carbon includes but is not limited to DARCO® KB-G, ® KB-WJ. In one aspect silica bound scavengers include but are not limited to
Figure imgf000042_0001
Figure imgf000043_0001
; where denotes silica gel. In some embodiments, the purification steps to reduce the amount of palladium include the use of activated carbon, derivatized silica gel (e.g. thiol derivatized silica gel), or combinations thereof.
[00268] Although the foregoing schemes exemplified the synthesis with (R)- 1 -phenyl ethyl alcohol, the same synthetic procedures could be performed with (S)-l -phenyl ethyl alcohol or (R/S)-l- phenyl ethyl alcohol in place of (R)- 1 -phenyl ethyl alcohol. In some embodiments, (R)- 1 -phenyl ethyl alcohol is optically pure. In some embodiments, (R)- 1 -phenylethyl alcohol has an enantiomeric excess that of at least 97%, at least 98%, or at least 99%.
Suitable Solvents
[00269] Therapeutic agents that are administrable to mammals, such as humans, must be prepared by following regulatory guidelines. Such government regulated guidelines are referred to as Good Manufacturing Practice (GMP). GMP guidelines outline acceptable contamination levels of active therapeutic agents, such as, for example, the amount of residual solvent in the final product.
Preferred solvents are those that are suitable for use in GMP facilities and consistent with industrial safety concerns. Categories of solvents are defined in, for example, the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), "Impurities: Guidelines for Residual Solvents, Q3C(R3), (November 2005).
[00270] Solvents are categorized into three classes. Class 1 solvents are toxic and are to be avoided. Class 2 solvents are solvents to be limited in use during the manufacture of the therapeutic agent. Class 3 solvents are solvents with low toxic potential and of lower risk to human health. Data for Class 3 solvents indicate that they are less toxic in acute or short-term studies and negative in genotoxicity studies.
[00271] Class 1 solvents, which are to be avoided, include: benzene; carbon tetrachloride; 1,2- dichloroethane; 1 , 1 -dichloroethene; and 1 , 1 , 1 -trichloroethane.
[00272] Examples of Class 2 solvents are: acetonitrile, chlorobenzene, chloroform, cyclohexane, 1,2- dichloroethene, dichloromethane, 1 ,2-dimethoxyethane, Ν,Ν-dimethylacetamide, N,N- dimethylformamide, 1,4-dioxane, 2-ethoxyethanol, ethyleneglycol, formamide, hexane, methanol, 2- methoxyethanol, methylbutyl ketone, methylcyclohexane, N-methylpyrrolidine, nitromethane, pyridine, sulfolane, tetralin, toluene, 1 , 1 ,2-trichloroethene and xylene.
[00273] Class 3 solvents, which possess low toxicity, include: acetic acid, acetone, anisole, 1- butanol, 2-butanol, butyl acetate, tert-butylmethyl ether (MTBE), cumene, dimethyl sulfoxide, ethanol, ethyl acetate, ethyl ether, ethyl formate, formic acid, heptane, isobutyl acetate, isopropyl acetate, methyl acetate, 3-methyl-l-butanol, methylethyl ketone, methylisobutyl ketone, 2-methyl-l- propanol, pentane, 1-pentanol, 1-propanol, 2-propanol, propyl acetate, and tetrahydrofuran. [00274] Residual solvents in active pharmaceutical ingredients (APIs) originate from the manufacture of API. In some cases, the solvents are not completely removed by practical manufacturing techniques. Appropriate selection of the solvent for the synthesis of APIs may enhance the yield, or determine characteristics such as crystal form, purity, and solubility. Therefore, the solvent is a critical parameter in the synthetic process.
[00275] In some embodiments, compositions comprising salts of Compound 1 comprise an organic solvent(s). In some embodiments, compositions comprising salts of Compound 1 comprise a residual amount of an organic solvent(s). In some embodiments, compositions comprising salts of Compound 1 comprise a residual amount of a Class 3 solvent. In some embodiments, the organic solvent is a Class 3 solvent. In some embodiments, the Class 3 solvent is selected from the group consisting of acetic acid, acetone, anisole, 1 -butanol, 2-butanol, butyl acetate, tert-butylmethyl ether, cumene, dimethyl sulfoxide, ethanol, ethyl acetate, ethyl ether, ethyl formate, formic acid, heptane, isobutyl acetate, isopropyl acetate, methyl acetate, 3 -methyl- 1 -butanol, methyl ethyl ketone, methylisobutyl ketone, 2-methyl- l -propanol, pentane, 1 -pentanol, 1 -propanol, 2-propanol, propyl acetate, and tetrahydrofuran. In some embodiments, the Class 3 solvent is selected from ethyl acetate, isopropyl acetate, tert-butylmethylether, heptane, isopropanol, and ethanol.
[00276] In some embodiments, the compositions comprising a salt of Compound 1 include a detectable amount of an organic solvent. In some embodiments, the salt of Compound 1 is a sodium salt (i.e. Compound 2). In some embodiments, the organic solvent is a Class 3 solvent.
[00277] In one aspect, the salt of Compound 1 is a sodium salt, potassium salt, lithium salt, calcium salt, magnesium salt, ammonium salt, choline salt, protonated dicyclohexylamine salt, protonated N- methyl-D-glucamine salt, protonated tris(hydroxymethyl)methylamine salt, arginine salt, or lysine salt. In one aspect, the salt of Compound 1 is a sodium salt.
[00278] In other embodiments are compositions comprising Compound 2, wherein the composition comprises a detectable amount of solvent that is less than about 1 %, wherein the solvent is selected from acetone, 1 ,2-dimethoxyethane, acetonitrile, ethyl acetate, tetrahydrofuran, methanol, ethanol, heptane, and 2-propanol. In a further embodiment are compositions comprising Compound 2, wherein the composition comprises a detectable amount of solvent which is less than about 5000 ppm. In yet a further embodiment are compositions comprising Compound 2, wherein the detectable amount of solvent is less than about 5000 ppm, less than about 4000 ppm, less than about 3000 ppm, less than about 2000 ppm, less than about 1000 ppm, less than about 500 ppm, or less than about 100 ppm.
Certain Terms
[00279] Unless otherwise stated, the following terms used in this application, including the specification and claims, have the definitions given below. It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, organic synthesis, biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art are employed. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, use of the term "including" as well as other forms, such as "include", "includes," and "included," is not limiting.
[00280] The term "pharmaceutically acceptable excipient," as used herein, refers to a material, such as a carrier, diluent, stabilizer, dispersing agent, suspending agent, thickening agent, etc. which allows processing the active pharmaceutical ingredient (API) into a form suitable for administration to a mammal. In one aspect, the mammal is a human. Pharmaceutically acceptable excipients refer to materials which do not substantially abrogate the desired biological activity or desired properties of the compound (i.e. API), and is relatively nontoxic, i.e., the material is administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
[00281] "Active pharmaceutical ingredient" or API refers to a compound that possesses a desired biological activity or desired properties. In some embodiments, an API is Compound 1. In some embodiments, an API is Compound 2. Provided herein is an active pharmaceutical ingredient (API), Compound 1 , or pharmaceutically acceptable salt thereof (e.g. Compound 2), with a purity of greater than 80%, greater than 85%, greater than 90%, greater than 95%, greater than 96%, greater than 97%), greater than 98%>, greater than 98%>, or greater than 99%>. In specific embodiments, provided herein is an active pharmaceutical ingredient (API), Compound 2, with a purity of greater than 80%>, greater than 85%>, greater than 90%>, greater than 95%>, greater than 96%>, greater than 97%>, greater than 98%, or greater than 99%.
[00282] The term "pharmaceutical combination" as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. Compound 1 or a pharmaceutically acceptable salt, and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non- fixed combination" means that the active ingredients, e.g. Compound 1 or a pharmaceutically acceptable salt, and a co-agent, are administered to a patient as separate entities either
simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.
[00283] The term "pharmaceutical composition" refers to a mixture of Compound 1 , or
pharmaceutically acceptable salt and/or solvate thereof, with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, excipients, etc. The pharmaceutical composition facilitates administration of the compound to a mammal. [00284] Administration of a combination of agents, as used herein, includes administration of the agents described in a single composition or in a combination therapy wherein one or more agent is administered separately from at least one other agent.
[00285] An "alkyl" group refers to an aliphatic hydrocarbon group. The alkyl moiety is branched, straight chain, or cyclic. The alkyl group may be designated as "Ci-C6alkyl". In one aspect, an alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, ethenyl, propenyl, allyl, butenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
[00286] "Detectable amount" refers to an amount that is measurable using standard analytic methods (e.g. ion chromatography, mass spectrometry, NMR, HPLC, gas chromatography, elemental analysis, IR spectroscopy, inductively coupled plasma atomic emission spectrometry,
USP<231>Method II, etc) (ICH guidances, Q2A Text on Validation of Analytical Procedures (March 1995) and Q2B Validation of Analytical Procedures: Methodology (November 1996)).
[00287] The term "acceptable" with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
[00288] The terms "effective amount" or "therapeutically effective amount," as used herein, refer to a sufficient amount of an agent being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. The term "therapeutically effective amount" includes, for example, a
prophylactically effective amount. The effective amount will be selected based on the particular patient and the disease level. It is understood that "an effect amount" or "a therapeutically effective amount" varies from subject to subject, due to variation in metabolism of drug, age, weight, general condition of the subject, the condition being treated, the severity of the condition being treated, and the judgment of the prescribing physician. In one embodiment, an appropriate "effective" amount in any individual case is determined using techniques, such as a dose escalation study
[00289] The terms "co-administration" or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
[00290] The terms "enhance" or "enhancing," as used herein, means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term "enhancing" refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An "enhancing-effective amount," as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
[00291] The terms "kit" and "article of manufacture" are used as synonyms.
[00292] A "metabolite" of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized. The term "active metabolite" refers to a biologically active derivative of a compound that is formed when the compound is metabolized (biotransformed). The term "metabolized," as used herein, refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound. For example, cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases (UGT) catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups (e.g. conjugation reactions). In some embodiments, compounds disclosed herein are metabolite to provide taurine metabolites. Further information on metabolism is available in The Pharmacological Basis of Therapeutics, 9th Edition, McGraw-Hill (1996). In one embodiment, metabolites of the compounds disclosed herein are identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
[00293] The term "modulate," as used herein, means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
[00294] The term "modulator," as used herein, refers to a molecule that interacts with a target either directly or indirectly. The interactions include, but are not limited to, the interactions of an agonist and an antagonist.
[00295] The term "agonist," as used herein, refers to a molecule such as a compound, a drug, an enzyme activator or a hormone modulator that binds to a specific receptor and triggers a response in the cell. An agonist mimics the action of an endogenous ligand (such as prostaglandin, hormone or neurotransmitter) that binds to the same receptor.
[00296] The term "antagonist," as used herein, refers to a molecule such as a compound, which diminishes, inhibits, or prevents the action of another molecule or the activity of a receptor site.
[00297] The term "LPA-dependent", as used herein, refers to conditions or disorders that would not occur, or would not occur to the same extent, in the absence of LPA.
[00298] The term "LPA-mediated", as used herein, refers to refers to conditions or disorders that might occur in the absence of LPA but can occur in the presence of LPA. [00299] The term "subject" or "patient" encompasses mammals. In one aspect, the mammal is a human. In another aspect, the mammal is a non-human primate such as chimpanzee, and other apes and monkey species. In one aspect, the mammal is a farm animal such as cattle, horse, sheep, goat, or swine. In one aspect, the mammal is a domestic animal such as rabbit, dog, or cat. In one aspect, the mammal is a laboratory animal, including rodents, such as rats, mice and guinea pigs, and the like.
[00300] "Bioavailability" refers to the percentage of the weight of Compound 1, or a
pharmaceutically acceptable salt and/or solvate thereof, dosed that is delivered into the general circulation of the animal or human being studied. The total exposure (AUC(o-∞)) of a drug when administered intravenously is usually defined as 100% Bioavailable (F%). "Oral bioavailability" refers to the extent to which Compound 1, or a pharmaceutically acceptable salt and/or solvate thereof, is absorbed into the general circulation when the pharmaceutical composition is taken orally as compared to intravenous injection.
[00301] "Blood plasma concentration" refers to the concentration Compound 1, in the plasma component of blood of a mammal. It is understood that the plasma concentration of Compound 1 may vary significantly between subjects, due to variability with respect to metabolism and/or interactions with other therapeutic agents. In one aspect, the blood plasma concentration of
Compound 1 varies from subject to subject. Likewise, values such as maximum plasma
concentration (Cmax) or time to reach maximum plasma concentration (Tmax), or total area under the plasma concentration time curve (AUC(o-)) vary from subject to subject. Due to this variability, in one embodiment, the amount necessary to constitute "a therapeutically effective amount" of Compound 1 varies from subject to subject.
[00302] "Drug absorption" or "absorption" typically refers to the process of movement of drug from site of administration of a drug across a barrier into a blood vessel or the site of action, e.g., a drug moving from the gastrointestinal tract into the portal vein or lymphatic system.
[00303] "Serum concentration" or "Plasma concentration" describes the blood serum or blood plasma concentration, typically measured in mg, μg, or ng of therapeutic agent per ml, dl, or 1 of blood serum, absorbed into the bloodstream after administration. Plasma concentrations are typically measured in ng/ml or μg/ml.
[00304] "Pharmacodynamics" refers to the factors which determine the biologic response observed relative to the concentration of drug at a site of action.
[00305] "Pharmacokinetics" refers to the factors which determine the attainment and maintenance of the appropriate concentration of drug at a site of action.
[00306] "Steady state," as used herein, is when the amount of drug administered is equal to the amount of drug eliminated within one dosing interval resulting in a plateau or constant plasma drug exposure. [00307] "Treat" or "treatment" as used herein refers to any treatment of a disorder or disease, such as preventing the disorder or disease from occurring in a subject predisposed to the disorder or disease, but has not yet been diagnosed as having the disorder or disease; inhibiting the disorder or disease, e.g., arresting the development of the disorder or disease, relieving the disorder or disease, causing regression of the disorder or disease, relieving a condition caused by the disease or disorder, or stopping the symptoms of the disease or disorder either prophylactically and/or therapeutically. Thus, as used herein, the term "treat" is used synonymously with the term "prevent."
Pharmaceutical Compositions/Formulations
[00308] Pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which are used pharmaceutically. Suitable techniques, carriers, and excipients include those found within, for example, Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington 's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975;
Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed.
(Lippincott Williams & Wilkinsl999), herein incorporated by reference in their entirety.
[00309] A pharmaceutical composition, as used herein, refers to a mixture of a compound of Formula (I) with other chemical components (i.e. pharmaceutically acceptable inactive ingredients), such as carriers, excipients, binders, filling agents, suspending agents, flavoring agents, sweetening agents, disintegrating agents, dispersing agents, surfactants, lubricants, colorants, diluents, solubilizers, moistening agents, plasticizers, stabilizers, penetration enhancers, wetting agents, anti-foaming agents, antioxidants, preservatives, or one or more combination thereof. The pharmaceutical composition facilitates administration of the compound to an organism.
[00310] Pharmaceutical formulations described herein are administerable to a subject in a variety of ways by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular, intramedullary injections, intrathecal, direct
intraventricular, intraperitoneal, intralymphatic, intranasal injections), intranasal, buccal, topical or transdermal administration routes. The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
[00311] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is administered orally. [00312] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is administered topically. In such embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, shampoos, scrubs, rubs, smears, medicated sticks, medicated bandages, balms, creams or ointments. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is administered topically to the skin.
[00313] In another aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), is administered by inhalation.
[00314] In another aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), is formulated for intranasal adminstration. Such formulations include nasal sprays, nasal mists, and the like.
[00315] In another aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2), is formulated as eye drops.
[00316] In any of the aforementioned aspects are further embodiments in which the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by inhalation to the mammal; and/or (e) administered by nasal administration to the mammal; or and/or (f) administered by injection to the mammal; and/or (g) administered topically to the mammal; and/or (h) administered by ophthalmic administration; and/or (i) administered rectally to the mammal; and/or (j) adminstered non- systemically or locally to the mammal.
[00317] In any of the aforementioned aspects are further embodiments comprising single
administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered once; (ii) the compound is administered to the mammal multiple times over the span of one day; (iii) continually; or (iv) continuously.
[00318] In any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours. In further or alternative embodiments, the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed. In one embodiment, the length of the drug holiday varies from 2 days to 1 year. [00319] In certain embodiments, a compound as described herein is administered in a local rather than systemic manner.
[00320] In some embodiments, the compound described herein is administered topically. In some embodiments, the compound described herein is administered systemically.
[00321] In some embodiments, for oral administration, Compound 1, or a pharmaceutically acceptably salt thereof (e.g. Compound 2), are formulated by combining the active compound with pharmaceutically acceptable carriers or excipients. Such carriers enable Compound 1, or a pharmaceutically acceptably salt thereof (e.g. Compound 2) to be formulated as tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated. In some embodiments, for oral administration, Compound 1, or a pharmaceutically acceptably salt thereof (e.g. Compound 2), is formulated without combining the active compound with pharmaceutically acceptable carriers or excipients and is placed directly into a capsule for administration to a mammal.
[00322] In some embodiments, the pharmaceutical compositions will include at least one
pharmaceutically acceptable carrier, diluent or excipient and Compound 1 as an active ingredient in free-acid or free-base form, or in a pharmaceutically acceptable salt form. In some embodiments, the pharmaceutical compositions will include at least one pharmaceutically acceptable carrier, diluent or excipient and Compound 2.
[00323] The pharmaceutical compositions described herein include Compound 1 , or a
pharmaceutically acceptable salt thereof (e.g. Compound 2). In some embodiments, the
pharmaceutical compositions described herein include Compound 1. In some embodiments, the pharmaceutical compositions described herein include amorphous Compound 1. In some embodiments, the pharmaceutical compositions described herein include crystalline Compound 1.
In some embodiments, the pharmaceutical compositions described herein include Compound 2. In some embodiments, the pharmaceutical compositions described herein include amorphous
Compound 2. In some embodiments, the pharmaceutical compositions described herein include crystalline Compound 2.
[00324] In some embodiments, the pharmaceutical compositions described herein include: (a) Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2); and one or more of the following: (b) binders; (c) disintegrants; (d) fillers (diluents); (e) lubricants; (f) glidants (flow enhancers); (g) compression aids; (h) colors; (i) sweeteners; (j) preservatives; (k)
suspensing/dispersing agents; (1) film formers/coatings; (m) flavors; (o) printing inks; (p) solubilizers; (q) alkalizing agents; (r) buffering agents; (s) antioxidants; (t) effervsescent agents.
[00325] In some embodiments, the pharmaceutical compositions described herein include: (a) Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2); and (b) a capsule shell. [00326] In some embodiments, pharmaceutical compositions described herein include one or more of the following in addition to Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2): (a) magnesium stearate; (b) lactose; (c) microcrystallme cellulose; (d) silicified microcrystallme cellulose; (e) mannitol; (f) starch (corn); (g) silicon dioxide; (h) titanium dioxide; (i) stearic acid; (j) sodium starch glycolate; (k) gelatin; (1) talc; (m) sucrose; (n) aspartame; (o) calcium stearate; (p) povidone; (q) pregelatinized starch; (r) hydroxy propyl methylcellulose; (s) OPA products (coatings & inks); (t) croscarmellose; (u) hydroxy propyl cellulose; (v) ethylcellulose; (w) calcium phosphate (dibasic); (x) crospovidone; (y) shellac (and glaze); (z) sodium carbonate; (aa) hypromellose.
[00327] In one embodiment, pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, silicified microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents are added, such as the cross-linked croscarmellose sodium,
polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[00328] In one embodiment, the pharmaceutical compositions described herein are formulated into any suitable dosage form, including but not limited to, aqueous oral dispersions, solid oral dosage forms, fast melt formulations, effervescent formulations, lyophilized formulations, tablets, capsules, pills, controlled release formulations, enteric coated tablets, inhaled powder, inhaled dispersion, IV formulations.
[00329] In further embodiments, the pharmaceutical compositions provided herein may be provided as compressed tablets, tablet triturates, rapidly dissolving tablets, multiple compressed tablets, or enteric-coated tablets, sugar-coated, or film-coated tablets.
[00330] Pharmaceutical dosage forms can be formulated in a variety of methods and can provide a variety of drug release profiles, including immediate release, sustained release, and delayed release. In some cases it may be desirable to prevent drug release after drug administration until a certain amount of time has passed (i.e. timed release), to provide substantially continuous release over a predetermined time period (i.e. sustained release) or to provide release immediately following drug administration (i.e., immediate release).
[00331] In some embodiments, formulations provide a therapeutically effective amount of
Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), enabling, for example, once a week, twice a week, three times a week, four times a week, five times a week, once every other day, once-a-day, twice-a-day (b.i.d.), or three times a day (t.i.d.) administration if desired. In one embodiment, the formulation provides a therapeutically effective amount of
Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) enabling once-a-day administration.
[00332] In one embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) is formulated into an immediate release form that provides for once-a-day
administration. Generally speaking, one will desire to administer an amount of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) that is effective to achieve a plasma level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit a therapeutic effect.
[00333] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 10 minutes, less than about 15 minutes, less than about 20 minutes, less than about 25 minutes, less than about 30 minutes, less than about 35 minutes, or less than about 40 minutes, after oral administration, thereby releasing the Compound 1, or a
pharmaceutically acceptable salt thereof (e.g. Compound 2) formulation into the gastrointestinal fluid.
[00334] In some embodiments, the pharmaceutical compositions provided herein in an immediate release dosage form are capable of releasing not less than 75 % of the therapeutically active ingredient or combination and/or meet the disintegration or dissolution requirements for immediate release tablets of the particular therapeutic agents or combination included in the tablet core, as set forth in USP XXII, 1990 (The United States Pharmacopeia.). Immediate release pharmaceutical compositions include capsules, tablets, pills, oral solutions, powders, beads, pellets, particles, and the like.
[00335] Excipients used in pharmaceutical compositions should be selected on the basis of compatibility with Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the release profile properties of the desired dosage form. Exemplary excipients include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like.
[00336] Binders impart cohesiveness to solid oral dosage form formulations: for powder filled capsule formulation, they aid in plug formation that is filled into soft or hard shell capsules and for tablet formulation, they ensure the tablet remaining intact after compression and help assure blend uniformity prior to a compression or fill step.
[00337] In some embodiments, the binder(s) are selected from starches, sugars, povidone, cellulose or modified cellulose such as microcrystalline cellulose, hydroxypropyl methyl cellulose, lactose, or sugar alcohols like xylitol, sorbitol or maltitol. In some embodiments, the binder is hydroxypropyl methyl cellulose. In some embodiments, the binder is hypromellose (e.g., Methocel E5).
[00338] In general, binder levels of 20-70% are used in powder- filled gelatin capsule formulations. Binder usage level in tablet formulations varies whether direct compression, wet granulation, roller compaction, or usage of other excipients such as fillers which itself acts as moderate binder.
[00339] Dispersing agents, and/or viscosity modulating agents include materials that control the diffusion and homogeneity of a drug through liquid media or a granulation method or blend method. In some embodiments, these agents also facilitate the effectiveness of a coating or eroding matrix.
[00340] Diluents increase bulk of the composition to facilitate compression or create sufficient bulk for homogenous blend for capsule filling.
[00341] The term "disintegrate" includes both the dissolution and dispersion of the dosage form when contacted with gastrointestinal fluid. "Disintegration agents or disintegrants" facilitate the breakup or disintegration of a substance. In some embodiments, one aspect, solid oral dosage forms include up to 15% w/w of disintegrant. In some embodiments, the disintegrant is croscarmellose sodium. In another aspect, the disintegrant is sodium starch glycolate or crospovidone.
[00342] Filling agents include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
[00343] In one aspect, the filler is lactose (e.g. monohydrate). In another aspect, the filler is mannitol, or dicalcium phosphate. In another aspect, the filler is mannitol, microcrystalline cellulose, dicalcium phosphate or sorbitol.
[00344] Gastrointestinal fluid is the fluid of stomach secretions of a subject or the saliva of a subject after oral administration of a composition described herein, or the equivalent thereof. An "equivalent of stomach secretion" includes, e.g., an in vitro fluid having similar content and/or pH as stomach secretions such as a 1% sodium dodecyl sulfate solution or 0.1N HC1 solution in water. In addition, simulated intestinal fluid (USP) is an aqueous phosphate buffer system at pH 6.8.
[00345] Lubricants and glidants are compounds that prevent, reduce or inhibit adhesion or friction of materials. In one aspect, solid oral dosage forms include about 0.25% w/w to about 2.5% w/w of lubricant. In another aspect solid oral dosage forms include about 0.5%) w/w to about 1.5% w/w of lubricant.
[00346] In some embodiments, the solid dosage forms described herein are in the form of a tablet, (including an immediate release tablet, an extended release tablet, a sustained release tablet, a enteric coated tablet, a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder), a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or "sprinkle capsules"), solid dispersion, multiparticulate dosage forms, pellets, or granules.
[00347] In other embodiments, the pharmaceutical formulation is in the form of a powder. In still other embodiments, the pharmaceutical formulation is in the form of a tablet, including but not limited to, an immediate release tablet. Additionally, pharmaceutical formulations described herein are administered as a single dosage or in multiple dosages. In some embodiments, the
pharmaceutical formulation is administered in two, or three, or four tablets.
[00348] In some embodiments, solid dosage forms, e.g., tablets, effervescent tablets, and capsules, are prepared by mixing Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) with one or more pharmaceutical excipients to form a bulk blend composition. When referring to these bulk blend compositions as homogeneous, it is meant that the Compound 1, or a
pharmaceutically acceptable salt thereof (e.g. Compound 2) particles are dispersed evenly throughout the composition so that the composition is capable of being readily subdivided into equally effective unit dosage forms, such as tablets, pills, or capsules. In one embodiment, the individual unit dosages also include film coatings, which disintegrate upon oral ingestion or upon contact with diluent. In one embodiment, these formulations are manufactured by conventional techniques.
[00349] Conventional techniques include, e.g., one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion. See, e.g., Lachman et al., The Theory and Practice of Industrial Pharmacy (1986). Other methods include, e.g., spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., wurster coating), tangential coating, top spraying, tableting, extruding and the like.
[00350] Compressed tablets are solid dosage forms prepared by compacting the bulk blend formulations described above. In various embodiments, compressed tablets which are designed to dissolve in the mouth will include one or more flavoring agents. In other embodiments, the compressed tablets will include a film surrounding the final compressed tablet. In some
embodiments, the film coating aids in patient compliance (e.g., Opadry® coatings or sugar coating). Film coatings comprising Opadry® typically range from about 1% to about 5% of the tablet weight. In other embodiments, the compressed tablets include one or more excipients.
[00351] Provided herein are pharmaceutical compositions in film-coated dosage forms, which comprise a combination of an active ingredient, or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more tabletting excipients to form a tablet core using conventional tabletting processes and subsequently coating the core. The tablet cores can be produced using conventional granulation methods, for example wet or dry granulation, with optional comminution of the granules and with subsequent compression and coating. [00352] Further provided herein are pharmaceutical compositions in enteric coated dosage forms, which comprise a combination of an active ingredient, or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more release controlling excipients for use in an enteric coated dosage form. The pharmaceutical compositions also comprise non-release controlling excipients.
[00353] Enteric-coatings are coatings that resist the action of stomach acid but dissolve or disintegrate in the intestine.
[00354] In one aspect, the oral solid dosage form disclosed herein include an enteric coating(s). Enteric coatings include one or more of the following: cellulose acetate phthalate; methyl acrylate- methacrylic acid copolymers; cellulose acetate succinate; hydroxy propyl methyl cellulose phthalate; hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate);
polyvinyl acetate phthalate (PVAP); methyl methacrylate-methacrylic acid copolymers; methacrylic acid copolymers, cellulose acetate (and its succinate and phthalate version); styrol maleic acid copolymers; polymethacrylic acid/acrylic acid copolymer; hydroxyethyl ethyl cellulose phthalate; hydroxypropyl methyl cellulose acetate succinate; cellulose acetate tetrahydrophtalate; acrylic resin; shellac.
[00355] An enteric coating is a coating put on a tablet, pill, capsule, pellet, bead, granule, particle, etc. so that it doesn't dissolve until it reaches the small intestine.
[00356] Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
[00357] Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press- coated or dry-coated tablets. In some embodiments, tablets are coated with water soluble, pH independent film coating which allows for immediate disintegration for fast, active release (e.g. Opadry products).
[00358] In some embodiments, the pharmaceutical compositions provided herein are in the form of a controlled release dosage form. As used herein, the term "controlled release" refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when orally administered. Controlled release dosage forms include delayed-, extended- , prolonged-, sustained-, pulsatile-, modified -, targeted-, programmed-release. The pharmaceutical compositions in controlled release dosage forms are prepared using a variety of modified release devices and methods including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes.
[00359] In contrast to immediate release compositions, controlled release compositions allow delivery of an agent to a human over an extended period of time according to a predetermined profile. Such release rates can provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response. Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding immediate release preparations. In one aspect, controlled release compositions of Compound 1 , or a pharmaceutically acceptable salt thereof, provide therapeutically effective levels of Compound 1 for an extended period of time and thereby provide a longer period of pharmacologic response.
[00360] Delayed release as used herein refers to the delivery so that the release can be accomplished at some generally predictable location in the intestinal tract more distal to that which would have been accomplished if there had been no delayed release alterations. In some embodiments the method for delay of release is coating. Any coatings should be applied to a sufficient thickness such that the entire coating does not dissolve in the gastrointestinal fluids at pH below about 5, but does dissolve at pH about 5 and above.
[00361] In some embodiments, the pharmaceutical compositions provided herein is in a modified release dosage form that is fabricated using a matrix controlled release device (see, Takada et al in "Encyclopedia of Controlled Drug Delivery," Vol. 2, Mathiowitz ed., Wiley, 1999).
[00362] In one embodiment, the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water-swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
[00363] In some embodiments, a matrix controlled release system includes an enteric coating so that no drug is released in the stomach.
[00364] The pharmaceutical compositions provided herein may be provided in unit-dosage forms or multiple-dosage forms. Unit-dosage forms, as used herein, refer to physically discrete units suitable for administration to human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of unit-dosage forms include individually packaged tablets and capsules. Unit-dosage forms may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit- dosage form. Examples of multiple-dosage forms include bottles of tablets or capsules.
[00365] In other embodiments a powder comprising the Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) formulations described herein are formulated to include one or more pharmaceutical excipients and flavors. Additional embodiments also comprise a suspending agent and/or a wetting agent. This bulk blend is uniformly subdivided into unit dosage packaging or multi-dosage packaging units. The term "uniform" means the homogeneity of the bulk blend is substantially maintained during the packaging process.
[00366] In still other embodiments, effervescent powders are prepared. Effervescent salts have been used to disperse medicines in water for oral administration. Effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate, citric acid and/or tartaric acid.
[00367] The method of preparation of the effervescent granules described herein employs three basic processes: wet granulation, dry granulation and fusion. The fusion method is used for the preparation of most commercial effervescent powders. It should be noted that, although these methods are intended for the preparation of granules, the formulations of effervescent salts described herein, in one embodiment, are also prepared as tablets, according to technology for tablet preparation.
[00368] In one embodiment, pharmaceutical preparations which are used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In one embodiment, the push-fit capsules contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In one embodiment, the push-fit capsules contain the active ingredient only without additional inactive ingredients. In one embodiment, in soft capsules, the active compounds are dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, in one embodiment, stabilizers are added. In other embodiments, the formulation is placed in a sprinkle capsule, wherein the capsule is swallowed whole or the capsule is opened and the contents sprinkled on food prior to eating.
[00369] All formulations for oral administration should be in dosages suitable for such
administration.
[00370] In some embodiments, pharmaceutical formulations are provided comprising Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and at least one dispersing agent or suspending agent for oral administration to a subject. In one embodiment, the formulation is a powder and/or granules for suspension, and upon admixture with water, a substantially uniform suspension is obtained.
[00371] A suspension is "substantially uniform" when it is mostly homogenous, that is, when the suspension is composed of approximately the same concentration of Compound 1 , or a
pharmaceutically acceptable salt thereof (e.g. Compound 2) at any point throughout the suspension (USP Chapter 905).
[00372] Liquid formulation dosage forms for oral administration are aqueous suspensions or nonaqueous suspensions. [00373] Liquid formulation dosage forms for oral administration are aqueous suspensions selected from, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, and syrups. See, e.g., Singh et ah, Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 754-757 (2002). In addition to including Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), the liquid dosage forms include additives, such as: (a) disintegrating agents; (b) dispersing agents; (c) wetting agents; (d) preservatives; (e) viscosity enhancing agents; (f) sweetening agents; (g) flavoring agents; (h) solibizing agents (bioavailability enhancers).
[00374] In one embodiment, the aqueous suspensions and dispersions described herein remain in a homogenous state, as defined above by USP Chapter 905, for at least 4 hours.
[00375] Liquid compositions illustratively take the form of a liquid where the agent (e.g. Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2)) is present in solution, in suspension or both. In one embodiment, the liquid composition is aqueous.
[00376] Liquid compositions illustratively take the form of a liquid where the agent (e.g. Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2)) is present in solution, in suspension or both. In one embodiment, the liquid composition is non-aqueous.
[00377] In one embodiment, the aqueous suspension also contains one or more polymers as suspending agents. Useful polymers include water-soluble polymers such as cellulosic polymers, e.g. , hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl- containing polymers. In one embodiment, useful compositions also comprise an mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
[00378] In one embodiment, pharmaceutical compositions also include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium carbonate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium carbonate, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
[00379] In one embodiment, liquid pharmaceutical compositions also include one or more salts in an amount required to bring osmolality of the composition into an acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
[00380] In one embodiment, pharmaceutical compositions also include one or more preservatives to inhibit microbial activity. [00381] Still other compositions include one or more surfactants to enhance physical stability or for other purposes. Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40.
[00382] Still other compositions include one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example only, ascorbic acid, tocopherol, and sodium metabisulfite.
[00383] In one embodiment, aqueous compositions are packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
[00384] In some embodiments, aqueous pharmaceutical compositions do not include a preservative and are used within 24 hours of preparation.
[00385] In some embodiments, aqueous pharmaceutical compositions include one or more solubilizers which aid in enhancing the bioavailability of the active pharmaceutical ingredient. In some embodiments, the solubilizer is selected from Labrasol, Lutrol (macrogels, poloxamers), and others known in the art.
[00386] The oral pharmaceutical solutions described herein are beneficial for the administration to infants (less than 2 years old), children under 10 years of age and any patient group that is unable to swallow or ingest solid oral dosage forms.
[00387] For buccal or sublingual administration, in one embodiment, the compositions take the form of tablets, lozenges, or gels formulated in a conventional manner (see e.g. U.S. Pat. Nos. 4,229,447, 4,596,795, 4,755,386, and 5,739,136).
[00388] In one embodiment, dragee cores are prepared with suitable coatings. For this purpose, concentrated sugar solutions are used, which optionally contain gum arabic, talc,
polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. In one embodiment, dyestuffs or pigments are added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[00389] It should be understood that many carriers and excipients may serve several functions, even within the same formulation.
[00390] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is formulated in the form of a pharmaceutical composition that is suitable for inhalation/nasal delivery. In some embodiments, the pharmaceutical composition is in the form of a solution, suspension, emulsion, colloidal dispersion, spray, dry powder, aerosol, or combinations thereof. In some embodiments, the pharmaceutical composition comprises at least one
pharmaceutically acceptable excipient that is commonly used in nasal/inhalable pharmaceutical compositions. In some embodiments, the pharmaceutical composition is administered with an atomizer, an insufflator, a nebulizer, a vaporizer, or a metered dose inhaler. In some embodiments, the pharmaceutical composition is inhaled nasally or orally. In some embodiments, crystalline Compound 1 is used in the pharmaceutical composition. In some embodiments, crystalline
Compound 2 is used in the pharmaceutical composition. In some embodiments, amorphous
Compound 1 is used in the pharmaceutical composition. In some embodiments, amorphous Compound 2 is used in the pharmaceutical composition.
[00391] Representative nasal/inhalation formulations are described in, for example, Ansel, H. C. et ah, Pharmaceutical Dosage Forms and Drug Delivery Systems, Sixth Ed. (1995); REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY, 21 st edition, 2005.
[00392] In some embodiments, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is formulated in the form of a nasal spray, nasal mist, and the like.
[00393] For administration by inhalation, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is formulated for use as an aerosol, a mist or a powder.
[00394] In some embodiments, pharmaceutical compositions suitable for nasal/inhalation administration are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant. Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of the compound described herein and a suitable powder base such as lactose or starch.
[00395] In some embodiments, the pharmaceutical composition is in the form of a powder for nasal/inhalation delivery to a mammal. In some embodiments, powders comprise micronized and/or nano-sized particles of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), blended with larger carrier particles that prevent aggregation. For example, in one embodiment a dry powder formulation is prepared as follows: Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is jet milled. Lactose is jet milled and the two ingredients are mixed and the final mixture is packaged in sterile insufflators. In some instances powder inhalable
formulations described herein comprise crystalline particles of Compound 1. In some instances powder inhalable formulations described herein comprise crystalline particles of Compound 2. In some embodiments, powder inhalable formulations described herein comprise amorphous particles of Compound 1. In some embodiments, powder inhalable formulations described herein comprise amorphous particles of Compound 2.
[00396] In some embodiments, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is prepared as transdermal dosage forms. In one embodiment, the transdermal formulations described herein include at least three components: (1) a formulation of Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2); (2) a penetration enhancer; and (3) an aqueous adjuvant. In some embodiments the transdermal formulations include additional components such as, but not limited to, gelling agents, creams and ointment bases, and the like. In some embodiments, the transdermal formulation further include a woven or non-woven backing material to enhance absorption and prevent the removal of the transdermal formulation from the skin. In other embodiments, the transdermal formulations described herein can maintain a saturated or supersaturated state to promote diffusion into the skin.
[00397] In one aspect, formulations suitable for transdermal administration of compounds described herein employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive. In one aspect, such patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Still further, transdermal delivery of the compounds described herein can be accomplished by means of iontophoretic patches and the like. In one aspect, transdermal patches provide controlled delivery of the active compound. In one aspect, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
[00398] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is formulated into a pharmaceutical composition suitable for intramuscular, subcutaneous, or intravenous injection. In one aspect, formulations suitable for intramuscular, subcutaneous, or intravenous injection include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. In some embodiments, formulations suitable for subcutaneous injection also contain additives such as preserving, wetting, emulsifying, and dispensing agents. In some cases it is desirable to include isotonic agents, such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, such as aluminum monostearate and gelatin.
[00399] For intravenous injections, compounds described herein are formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. For other parenteral injections, appropriate formulations include aqueous or nonaqueous solutions, preferably with physiologically compatible buffers or excipients. Such excipients are known.
[00400] Parenteral injections may involve bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The pharmaceutical composition described herein may be in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. In one aspect, the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[00401] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is administered topically and can be formulated into a variety of topically
administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. Such pharmaceutical compounds can contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
[00402] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), is administered topically and can be formulated into a variety of topically
administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
Dose Amounts of Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound }
[00403] In certain embodiments, the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is about 0.3mg to about lg per dose, about lmg to about lg per dose, about 5mg to about 600mg per dose or about 5mg to about 500mg per dose. In some embodiments, the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is about lmg to about 5g per day, about 5mg to about 2g per day, about 5mg to about lg per day, about 5mg to about 0.6g per day, or about 5mg to about 0.5g per day.
[00404] In one embodiment, the effective amount of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is about lOmg per dose, about 25mg per dose, about 50mg per dose, about lOOmg per dose, about 150mg per dose, about 200mg per dose, about 300mg per dose, about 400mg per dose, about 500mg per dose, about 600mg per dose, or about lOOOmg per dose.
[00405] In some embodiments, oral pharmaceutical solutions include about 0.015mg/ml to about 20mg/ml of Compound 2. In some embodiments, oral pharmaceutical solutions include about lmg/ml to about 20mg/ml of Compound 2.
[00406] In one aspect, immediate release tablets include about 5% w/w to about 50% w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In some
embodiments, immediate release tablets include about 5%> w/w to about 40%> w/w, or about 5%> w/w to about 30%) w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In some embodiments, immediate release tablets include about 5%> w/w, about 10%> w/w, about 15%> w/w, about 20%) w/w, about 25%> w/w, about 30%> w/w, about 33%> w/w, about 35%> w/w, about 40%> w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). [00407] In one aspect, immediate release capsules include about 1.25% w/w to about 50% w/w of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2). In some
embodiments, immediate release capsules include Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the capsule shell only.
Methods of Dosing and Treatment Regimens
[00408] In one embodiment, the pharmaceutical compositions including Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), described herein is administered for prophylactic and/or therapeutic treatments. In therapeutic applications, the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition. In certain embodiments, amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and/or the judgment of the treating physician.
[00409] In prophylactic applications, compositions containing Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), described herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition. Such an amount is defined to be a "prophylactically effective amount or dose." In this use, the precise amounts also depend on the patient's state of health, weight, and the like. When used in a patient, effective amounts for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
[00410] In certain embodiments, administration of the compound, compositions or therapies as described herein includes chronic administration. In certain embodiments, chronic administration includes administration for an extended period of time, including, e.g., throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition. In some embodiments, chronic administration includes daily administration.
[00411] In some embodiments, administration of the compounds, compositions or therapies described herein is given continuously. In alternative embodiments, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time {i.e., a "drug holiday"). The length of the drug holiday varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, and 365 days. The dose reduction during a drug holiday is from 10%- 100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%.
[00412] In some embodiments, the compounds, compositions or therapies described herein are administered in at least one priming dose, followed by at least one maintenance dose. In certain embodiments, a priming dose of the agent(s) is administered until the symptoms of the disorder, disease or condition treated have been reduced (e.g., to a satisfactory level). Upon reduction, a maintenance dose of the compounds, compositions or therapies described herein is administered if desired or if necessary. In some embodiments, the maintenance dose comprises administration of the agent(s) described herein in an amount sufficient to at least partially maintain the reduction achieved by administration of the priming dose. In various embodiments, the maintenance dose, compared to the priming dose, includes a decrease in dosage and/or frequency of administration of the agent or one or more of the agents administered in the method. In certain embodiments, however, intermittent treatment with increased frequency and/or dosage amounts may be necessary upon any recurrence of symptoms.
[00413] In certain embodiments, the amount of a given agent that corresponds to a priming or maintenance amount varies depending upon factors including, by way of non-limiting example, the specific agent(s) utilized, the disease condition and its severity, the identity (e.g., weight) of the subject or host in need of treatment, and/or the route of administration. In various embodiments, the desired dose is conveniently presented in a single dose or in divided doses administered
simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
Pharmacokinetic and Pharmacodynamic Analysis
[00414] In one embodiment, any standard pharmacokinetic protocol is used to determine blood plasma concentration profile in humans following administration of a formulation described herein (that include Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2)). For example, a randomized single-dose crossover study is performed using a group of healthy adult human subjects. The number of subjects is sufficient to provide adequate control of variation in a statistical analysis, and is typically about 10 or greater, although for certain purposes a smaller group suffices. Each subject receives administration at time zero a single dose of a formulation of
Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2), normally at around 8am following an overnight fast. The subjects continue to fast and remain in an upright position for about 2 hours after administration of the formulation. Blood samples are collected from each subject prior to administration (e.g., 15 minutes) and at several intervals after administration. In certain instances, several samples are taken within the first hour and taken less frequently thereafter.
Illustratively, blood samples are collected at 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 4, 6, 8, 12, and 16 hours after administration and, 24, 36, 48, 60 and 72 hours after administration. If the same subjects are to be used for study of a second test formulation, a period of at least 10 days should elapse before administration of the second formulation. Plasma is separated from the blood samples by
centrifugation and the separated plasma is analyzed for Compound 1 by a validated high performance liquid chromatography/tandem weight spectrometry (LC/APCI-MS/MS) procedure such as, for example, Ramu et ah, Journal of Chromatography B, 751 (2001) 49-59).
[00415] Any formulation giving the desired pharmacokinetic profile is suitable for administration according to the present methods.
Patient Selection
[00416] In any of the aforementioned aspects involving the prevention or treatment of LPA-mediated diseases or conditions are further embodiments comprising identifying patients by screening for LPA receptor gene SNPs. A SNP located in the promoter region of LPAi showed significant association with knee osteoarthritis in two independent populations (Mototani et ah Hum. Mol. Genetics, vol. 17, no. 12, 2008). Patients can be further selected based on increased LPA receptor expression in the tissue of interest. For example, chronic lymphocytic leukemia (CLL) is characterized by the accumulation of CD19+/CD5+ B -lymphocytes in the peripheral blood, bone marrow and lymphoid organs which occurs as a result of a block in B-lymphocyte apoptosis. LPA can protect some CLL cells from apoptosis and the cells that are protected by LPA have high levels of LPAi mRNA. In some embodiments, CLL patients are selected based on the expression of the LPA1R. LPA receptor expression are determined by methods including, but not limited to, northern blotting, western blotting, quantitative PCR (qPCR), flow cytometry, autoradiography (using a small molecule radioligand or PET ligand). In some embodiments, patients are selected based on the concentration of serum or tissue LPA measured by mass spectrometry. LPA concentrations are high in ovarian cancer ascites and in some breast cancer effusions. In some embodiments, patients are selected based on a combination of the above markers (increased LPA concentrations and increased LPA receptor expression).
Combination Treatments
[00417] In certain instances, it is appropriate to administer Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving one of the compounds herein is inflammation, then it may be appropriate to administer an anti-inflammatory agent in combination with the initial therapeutic agent.
[00418] Or, in one embodiment, the therapeutic effectiveness of one of the compounds described herein is enhanced by administration of an adjuvant {i.e., by itself the adjuvant may have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, in some embodiments, the benefit experienced by a patient is increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
[00419] In one specific embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is co-administered with a second therapeutic agent, wherein Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the second therapeutic agent modulate different aspects of the disease, disorder or condition being treated, thereby providing a greater overall benefit than administration of either therapeutic agent alone.
[00420] In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
[00421] In certain embodiments, different therapeutically-effective dosages of the compounds disclosed herein will be utilized in formulating pharmaceutical composition and/or in treatment regimens when the compounds disclosed herein are administered in combination with one or more additional agent, such as an additional therapeutically effective drug, an adjuvant or the like.
Therapeutically-effective dosages of drugs and other agents for use in combination treatment regimens can be determined by means similar to those set forth hereinabove for the actives themselves. Furthermore, the methods of prevention/treatment described herein encompasses the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects. In some embodiments, a combination treatment regimen encompasses treatment regimens in which administration of Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is initiated prior to, during, or after treatment with a second agent described herein, and continues until any time during treatment with the second agent or after termination of treatment with the second agent. It also includes treatments in which Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and the second agent being used in combination are administered simultaneously or at different times and/or at decreasing or increasing intervals during the treatment period. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
[00422] Compositions and methods for combination therapy are provided herein. In accordance with one aspect, the pharmaceutical compositions disclosed herein are used to treat LPA-dependent or LPA-mediated conditions.
[00423] It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is modified in accordance with a variety of factors. These factors include the disease, disorder or condition from which the subject suffers, as well as the age, weight, sex, diet, and medical condition of the subject. Thus, in some instances, the dosage regimen actually employed varies and, in some embodiments, deviates from the dosage regimens set forth herein.
[00424] For combination therapies described herein, dosages of the co-administered compounds vary depending on the type of co-drug employed, on the specific drug employed, on the disease or condition being treated and so forth. In additional embodiments, when co-administered with one or more other therapeutic agents, the compound provided herein is administered either simultaneously with the one or more other therapeutic agents, or sequentially. [00425] In combination therapies, the multiple therapeutic agents (one of which is one of the compounds described herein) are administered in any order or even simultaneously. If administration is simultaneous, the multiple therapeutic agents are, by way of example only, provided in a single, unified form, or in multiple forms (e.g., as a single pill or as two separate pills). In one embodiment, one of the therapeutic agents is given in multiple doses, and in another, two (or more if present) are given as multiple doses. In some embodiments of non-simultaneous administration, the timing between the multiple doses vary from more than zero weeks to less than four weeks. In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations is also envisioned.
[00426] Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) and combination therapies are administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition containing a compound varies. Thus, in one embodiment, the compounds described herein are used as a prophylactic and are administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition. In another embodiment, the compounds and compositions are administered to a subject during or as soon as possible after the onset of the symptoms. In specific embodiments, a compound described herein is administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease. In some embodiments, the length required for treatment varies, and the treatment length is adjusted to suit the specific needs of each subject. For example, in specific embodiments, a compound described herein or a formulation containing the compound is administered for at least 2 weeks, about 1 month to about 5 years.
[00427] By way of example, therapies which combine Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) with inhibitors of LP A synthesis or LPA receptor antagonists, either acting at the same or other points in the LPA synthesis or signalling pathway, are encompassed herein for treating LPA-dependent or LPA-mediated diseases or conditions.
Exemplary Agent for use in Combination Therapy
[00428] In another embodiment described herein, methods for treatment of LPA-dependent or LPA- mediated conditions or diseases, such as proliferative disorders, including cancer, comprises administration to a mammal Compound 1 , or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) in combination with at least one additional agent selected, by way of example only, alemtuzumab, arsenic trioxide, asparaginase (pegylated or non-), bevacizumab, cetuximab, platinum- based compounds such as cisplatin, cladribine, daunorubicin/doxorubicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, Paclitaxel™, taxol, temozolomide, thioguanine, or classes of drugs including hormones (an antiestrogen, an antiandrogen, or gonadotropin releasing hormone analogues, interferons such as alpha interferon, nitrogen mustards such as busulfan or melphalan or mechlorethamine, retinoids such as tretinoin, topoisomerase inhibitors such as irinotecan or topotecan, tyrosine kinase inhibitors such as gefinitinib or imatinib, or agents to treat signs or symptoms induced by such therapy including allopurinol, filgrastim, granisetron/ ondansetron/palonosetron, dronabinol.
[00429] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered or formulated in combination with one or more anti-cancer agents. In some embodiments, one or more of the anti-cancer agents are proapoptotic agents. Examples of anticancer agents include, but are not limited to, any of the following: gossypol, genasense, polyphenol E, Chlorofusin, all trans-retinoic acid (ATRA), bryostatin, tumor necrosis factor-related apoptosis- inducing ligand (TRAIL), 5-aza-2'-deoxycytidine, all trans retinoic acid, doxorubicin, vincristine, etoposide, gemcitabine, imatinib, geldanamycin, 17-N-Allylamino- 17-Demethoxygeldanamycin (17-AAG), flavopiridol, LY294002, bortezomib, trastuzumab, BAY 11-7082, PKC412, or
PD 184352, Taxol™ (paclitaxel), and analogs of Taxol™, such as Taxotere™. Compounds that have the basic taxane skeleton as a common structure feature, have also been shown to have the ability to arrest cells in the G2-M phases due to stabilized microtubules and may be useful for treating cancer in combination with the compounds described herein.
[00430] Further examples of anti-cancer agents for use in combination with Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) include inhibitors of mitogen-activated protein kinase signaling, e.g., U0126, PD98059, PD184352, PD0325901, ARRY-142886,
SB239063, SP600125, BAY 43-9006, wortmannin, or LY294002; Syk inhibitors; mTOR inhibitors; and antibodies (e.g., rituxan).
[00431] Other anti-cancer agents for use in combination with Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) include one or more of the following: abiraterone;
abarelix; adriamycin; aactinomycin; acivicin; aclarubicin; acodazole hydrochloride; acronine;
adozelesin; aldesleukin; alemtuzumab; allopurinol; alitretinoin; altretamine; ambomycin;
ametantrone acetate; aminoglutethimide; aminolevulinic acid; amifostine; amsacrine; anastrozole; anthramycin; aprepitant; arsenic trioxide; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; bendamustine hydrochloride; benzodepa; bevacizumab; bexarotene; bicalutamide;
bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin; bleomycin sulfate;
bortezomib; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide;
carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; capecitabine; cedefingol; cetuximab; chlorambucil; cirolemycin; cisplatin; cladribine; clofarabine; crisnatol mesylate;
cyclophosphamide; cytarabine; dacarbazine; dasatinib; daunorubicin hydrochloride; dactinomycin; darbepoetin alfa; decitabine; degarelix; denileukin diftitox; dexormaplatin; dexrazoxane
hydrochloride; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; eltrombopag olamine; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; epoetin alfa; erbulozole; erlotinib hydrochloride; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; everolimus; exemestane; fadrozole hydrochloride; fazarabine; fenretinide; filgrastim; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; fulvestrant;
gefitinib; gemcitabine; gemcitabine hydrochloride; gemcitabine -cisplatin; gemtuzumab
ozogamicin; goserelin acetate; histrelin acetate; hydroxyurea; idarubicin hydrochloride; ifosfamide; iimofosine; ibritumomab tiuxetan; idarubicin; ifosfamide; imatinib mesylate; imiquimod;
interleukin II (including recombinant interleukin II, or rlL2), interferon alfa-2a; interferon alfa-2b; interferon alfa-nl; interferon alfa-n3; interferon beta-1 a; interferon gamma-1 b; iproplatin; irinotecan hydrochloride; ixabepilone; lanreotide acetate; lapatinib; lenalidomide; letrozole; leuprolide acetate; leucovorin calcium; leuprolide acetate; levamisole; liposomal cytarabine; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; methoxsalen; metoprine; meturedepa;
mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin C; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nandrolone phenpropionate; nelarabine; nilotinib; nocodazoie; nofetumomab; nogalamycin; ofatumumab; oprelvekin; ormaplatin;
oxaliplatin;oxisuran; paclitaxel; palifermin; palonosetron hydrochloride; pamidronate;
pegfilgrastim; pemetrexed disodium; pentostatin; panitumumab; pazopanib hydrochloride;
pemetrexed disodium; plerixafor; pralatrexate; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; quinacrine; raloxifene hydrochloride; rasburicase; recombinant HPV bivalent vaccine; recombinant HPV quadrivalent vaccine; riboprine; rogletimide; rituximab;
romidepsin; romiplostim; safingol; safingol hydrochloride; sargramostim; semustine; simtrazene; sipuleucel-T; sorafenib; sparfosate sodium; sparsomycin; spirogermanium hydrochloride;
spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; sunitinib malate; talisomycin;
tamoxifen citrate; tecogalan sodium; tegafur; teloxantrone hydrochloride; temozolomide;
temoporfin; temsirolimus; teniposide; teroxirone; testolactone; thalidomide;thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; topotecan hydrochloride; toremifene; tositumomab; tositumomab and 1 131 Iodine tositumomab; trastuzumab; trestolone acetate; tretinoin; triciribine phosphate;
trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard;
uredepa; valrubicin; vapreotide; verteporfin; vinblastine; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorinostat; vorozole; zeniplatin;
zinostatin; zoledronic acid; zorubicin hydrochloride.
[00432] Yet other anticancer agents for use in combination with a compound of Formula (I), (II), (III), (IV) or (V) include alkylating agents, antimetabolites, natural products, or hormones, e.g., nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc.), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, ete.), or triazenes (decarbazine, etc.). Examples of antimetabolites include but are not limited to folic acid analog (e.g., methotrexate), or pyrimidine analogs (e.g., Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin).
[00433] Examples of natural products for use in combination with Compound 1, or a
pharmaceutically acceptable salt thereof (e.g. Compound 2) include but are not limited to vinca alkaloids (e.g., vinblastin, vincristine), epipodophyllotoxins (e.g., etoposide), antibiotics (e.g., daunorubicin, doxorubicin, bleomycin), enzymes (e.g., L-asparaginase), or biological response modifiers (e.g., interferon alpha).
[00434] Examples of alkylating agents for use in combination with Compound 1, or a
pharmaceutically acceptable salt thereof (e.g. Compound 2) include, but are not limited to, nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, meiphalan, etc.), ethyl enimine and methylmelamines (e.g., hexamethlymelamine, thiotepa), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, semustine, streptozocin, etc.), or triazenes (decarbazine, ete.). Examples of antimetabolites include, but are not limited to folic acid analog (e.g., methotrexate), or pyrimidine analogs (e.g., fluorouracil, floxouridine, Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin.
[00435] Examples of hormones and antagonists for use in combination with Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) include, but are not limited to, adrenocorticosteroids (e.g., prednisone), progestins (e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate), estrogens (e.g., diethlystilbestrol, ethinyl estradiol), antiestrogen (e.g., tamoxifen), androgens (e.g., testosterone propionate, fluoxymesterone), antiandrogen (e.g., flutamide), gonadotropin releasing hormone analog (e.g., leuprolide). Other agents that can be used in the methods and compositions described herein for the treatment or prevention of cancer include platinum coordination complexes (e.g., cisplatin, carboblatin), anthracenedione (e.g., mitoxantrone), substituted urea (e.g., hydroxyurea), methyl hydrazine derivative (e.g., procarbazine), adrenocortical suppressant (e.g., mitotane, aminoglutethimide).
[00436] Examples of anti-cancer agents which act by arresting cells in the G2-M phases due to stabilized microtubules include without limitation the following marketed drugs and drugs in development: Erbulozole, Dolastatin 10, Mivobulin isethionate, Vincristine, NSC-639829,
Discodermolide, ABT-751, Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C), Spongistatins (such as Spongistatin 1, Spongistatin 2, Spongistatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, Spongistatin 7, Spongistatin 8, and Spongistatin 9), Cemadotin hydrochloride, Epothilones (such as Epothilone A, Epothilone B, Epothilone C, Epothilone D, Epothilone E, Epothilone F, Epothilone B N-oxide, Epothilone A N-oxide, 16-aza-epothilone B, 21-aminoepothilone B, 21- hydroxyepothilone D, 26-fluoroepothilone, Auristatin PE, Soblidotin, Vincristine sulfate,
Cryptophycin 52, Vitilevuamide, Tubulysin A, Canadensol, Centaureidin, Oncocidin Al Fijianolide B, Laulimalide, Narcosine, Nascapine, Hemiasterlin, Vanadocene acetylacetonate, Indanocine Eleutherobins (such as Desmethyleleutherobin, Desaetyleleutherobin, lsoeleutherobin A, and Z- Eleutherobin), Caribaeoside, Caribaeolin, Halichondrin B, Diazonamide A, Taccalonolide A, Diozostatin, (-)-Phenylahistin, Myoseverin B, Resverastatin phosphate sodium.
[00437] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is co-administered with thrombolytic agents (e.g., alteplase anistreplase, streptokinase, urokinase, or tissue plasminogen activator), heparin, tinzaparin, warfarin, dabigatran (e.g., dabigatran etexilate), factor Xa inhibitors (e.g., fondaparinux, draparinux, rivaroxaban, DX-9065a, otamixaban,
LY517717, or YM150), ticlopidine, clopidogrel, CS-747 (prasugrel, LY640315), ximelagatran, or BIBPv 1048.
[00438] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is used in combination with anti-emetic agents to treat nausea or emesis. Anti-emetic agents include, but are not limited to: neurokinin- 1 receptor antagonists, 5HT3 receptor antagonists (such as ondansetron, granisetron, tropisetron, Palonosetron, and zatisetron), GABAB receptor agonists (such as baclofen), corticosteroids (such as dexamethasone, prednisone, prednisolone, or others), dopamine antagonists (such as, but not limited to, domperidone, droperidol, haloperidol, chlorpromazine, promethazine, prochlorperazine, metoclopramide), antihistamines (HI histamine receptor antagonists, such as but not limited to, cyclizine, diphenhydramine, dimenhydrinate, meclizine, promethazine, hydroxyzine), cannabinoids (such as but not limited to, cannabis, marinol, dronabinol), and others (such as, but not limited to, trimethobenzamide; ginger, emetrol, propofol).
[00439] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is used in combination with an agent useful in the treatment of anemia. Such an anemia treatment agent is, for example, a continuous eythropoiesis receptor activator (such as epoetin-a).
[00440] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is used in combination with an agent useful in the treatment of neutropenia. Examples of agents useful in the treatment of neutropenia include, but are not limited to, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF). Examples of a G-CSF include filgrastim. [00441] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is used in combination with radiation therapy (or radiotherapy). Radiation therapy is the treatment of cancer and other diseases with ionizing radiation. Radiation therapy can be used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, prostate, colon, uterus and/or cervix. It can also be used to treat leukemia and lymphoma (cancers of the blood- forming cells and lymphatic system, respectively).
[00442] A technique for delivering radiation to cancer cells is to place radioactive implants directly in a tumor or body cavity. This is called internal radiotherapy (brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.) Using internal radiotherapy, the radiation dose is concentrated in a small area, and the patient stays in the hospital for a few days.
Internal radiotherapy is frequently used for cancers of the tongue, uterus, prostate, colon, and cervix.
[00443] The term "radiotherapy" or "ionizing radiation" include all forms of radiation, including but not limited to α, β, and γ radiation and ultraviolet light.
[00444] In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is used to treat or reduce fibrosis in a mammal. In one aspect, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered in combination with one or more immunosuppresants. Immunosuppressive therapy is clinically used to treat or prevent the rejection of transplanted organs and tissues (e.g. bone marrow, heart, kidney, liver); treatment of autoimmune diseases or diseases that are most likely of autoimmune origin (e.g. rheumatoid arthritis, myasthenia gravis, systemic lupus erythematosus, Crohn's disease, and ulcerative colitis); and treatment of some other non-autoimmune inflammatory diseases (e.g. long term allergic asthma control), and in the treatment of fibrotic conditions.
[00445] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is adminsitered with corticosteroids. In some embodiments, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is adminsitered with an a therapeutic agent selected from among: Calcineurin inhibitors (such as, but not limited to, cyclosporin, tacrolimus); mTOR inhibitors (such as, but not limited to, sirolimus, everolimus); anti-proliferatives (such as, but not limited to, azathioprine, mycophenolic acid); corticosteroids (such as, but not limited to, prednisone, cortisone acetate, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, hydrocortisone); antibodies (such as, but not limited to, monoclonal anti-IL-2Ra receptor antibodies (basiliximab, daclizumab), polyclonal anti-T-cell antibodies (anti-thymocyte globulin (ATG), anti-lymphocyte globulin (ALG)), B-cell antagonists, rituximab, natalizumab.
[00446] Other therapeutic agents include, but are not limited to: cyclophosphamide, penicillamine, cyclosporine, nitrosoureas, cisplatin, carboplatin, oxaliplatin, methotrexate, azathioprine, mercaptopurine, pyrimidine analogues, protein synthesis inhibitors, dactinomycin, anthracyclines, mitomycin C, bleomycin, mithramycin, Atgam^, Thymoglobuline®, OKT3®, basiliximab, daclizumab, cyclosporin, tacrolimus, sirolimus, Interferons (IFN-β, IFN-γ), opioids, TNF binding proteins (infliximab, etanercept, adalimumab, golimumab), leflunomide, gold thioglucose, gold thiomalate, aurofin, sulfasalazine, hydroxychloroquinine, minocycline, rapamicin, mycophenolic acid, mycophenolate mofetil, FTY720, as well as those listed in US 7,060,697.
[00447] In one embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) is administered in combination with Cyclosporin A (CsA) or tacrolimus (FK506). In one embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered to a mammal in combination with an anti-inflammatory agent including, but not limited to, non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids (glucocorticoids).
[00448] NSAIDs include, but are not limited to: aspirin, salicylic acid, gentisic acid, choline magnesium salicylate, choline salicylate, choline magnesium salicylate, choline salicylate, magnesium salicylate, sodium salicylate, diflunisal, carprofen, fenoprofen, fenoprofen calcium, flurobiprofen, ibuprofen, ketoprofen, nabutone, ketolorac, ketorolac tromethamine, naproxen, oxaprozin, diclofenac, etodolac, indomethacin, sulindac, tolmetin, meclofenamate, meclofenamate sodium, mefenamic acid, piroxicam, meloxicam, COX-2 specific inhibitors (such as, but not limited to, celecoxib, rofecoxib, valdecoxib, parecoxib, etoricoxib, lumiracoxib, CS-502, JTE-522, L- 745,337 and NS398).
[00449] Corticosteroids, include, but are not limited to: betamethasone, prednisone, alclometasone, aldosterone, amcinonide, beclometasone, betamethasone, budesonide, ciclesonide, clobetasol, clobetasone, clocortolone, cloprednol, cortisone, cortivazol, deflazacort, deoxycorticosterone, desonide, desoximetasone, desoxycortone, dexamethasone, diflorasone, diflucortolone,
difluprednate, fluclorolone, fludrocortisone, fludroxycortide, flumetasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin, fluocortolone, fluorometholone, fluperolone, fluprednidene, fluticasone, formocortal, halcinonide, halometasone, hydrocortisone/cortisol, hydrocortisone aceponate, hydrocortisone buteprate, hydrocortisone butyrate, loteprednol, medrysone,
meprednisone, methylprednisolone, methylprednisolone aceponate, mometasone furoate, paramethasone, prednicarbate, prednisone/prednisolone, rimexolone, tixocortol, triamcinolone, and ulobetasol.
[00450] In one embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) is administered in combination with leukotriene receptor antagonists.
[00451] In another embodiment described herein, methods for treatment of LPA-dependent or LPA- mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected, by way of example only, HMG-CoA reductase inhibitors (e.g., statins in their lactonized or dihydroxy open acid forms and pharmaceutically acceptable salts and esters thereof, including but not limited to lovastatin; simvastatin; dihydroxy open-acid simvastatin, particularly the ammonium or calcium salts thereof; pravastatin, particularly the sodium salt thereof; fluvastatin, particularly the sodium salt thereof; atorvastatin, particularly the calcium salt thereof; nisvastatin, also referred to as NK-104; rosuvastatin); agents that have both lipid-altering effects and other pharmaceutical activities; HMG-CoA synthase inhibitors; cholesterol absorption inhibitors such as ezetimibe;
cholesterol ester transfer protein (CETP) inhibitors, for example JTT-705 and CP529, 414; squalene epoxidase inhibitors; squalene synthetase inhibitors (also known as squalene synthase inhibitors); acyl-coenzyme A: cholesterol acyltransferase (ACAT) inhibitors including selective inhibitors of ACAT-1 or ACAT-2 as well as dual inhibitors of ACAT- 1 and-2; microsomal triglyceride transfer protein (MTP) inhibitors; probucol; niacin; bile acid sequestrants; LDL (low density lipoprotein) receptor inducers; platelet aggregation inhibitors, for example glycoprotein Ilb/IIIa fibrinogen receptor antagonists and aspirin; human peroxisome proliferator activated receptor gamma (PPARy) agonists, including the compounds commonly referred to as glitazones, for example troglitazone, pioglitazone and rosiglitazone and including those compounds included within the structural class known as thiazolidinediones as well as those PPARy agonists outside the thiazolidinedione structural class; PPARa agonists such as clofibrate, fenofibrate including micronized fenofibrate, and gemfibrozil ; PPAR dual α/γ agonists such as 5-[(2, 4-dioxo-5-thiazolidinyl)methyl]-2-methoxy-N- [[4-(trifluoromethyl)phenyl] methyl] -benzamide, known as KRP-297; vitamin B6 (also known as pyridoxine) and the pharmaceutically acceptable salts thereof such as the HCl salt; vitamin B 12 (also known as cyanocobalamin); folic acid or a pharmaceutically acceptable salt or ester thereof such as the sodium salt and the methylglucamine salt; anti-oxidant vitamins such as vitamin C and E and beta carotene; beta-blockers; angiotensin II antagonists such as losartan; angiotensin converting enzyme inhibitors such as enalapril and captopril ; calcium channel blockers such as nifedipine and diltiazam; endothelian antagonists; agents that enhance ABC1 gene expression; FXR and LXR ligands including both inhibitors and agonists; bisphosphonate compounds such as alendronate sodium; and cyclooxygenase-2 inhibitors such as rofecoxib and celecoxib.
[00452] In another embodiment described herein, methods for treatment of LPA-dependent or LPA- mediated conditions or diseases comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, by way of example only, COX-2 inhibitors; nitric oxide synthase inhibitors, such as N-(3-(aminomethyl)benzyl) acetamidine; Rho kinase inhibitors, such as fasudil; angiotension II type-1 receptor antagonists, including candesartan, losartan, irbesartan, eprosartan, telmisartan and valsartan; glycogen synthase kinase 3 inhibitors; sodium or calcium channel blockers, including crobenetine; p38 MAP kinase inhibitors, including SKB 239063; thromboxane AX- synthetase inhibitors, including isbogrel, ozagrel, ridogrel and dazoxiben; statins (HMG CoA reductase inhibitors), including lovastatin, simvastatin, dihydroxy open-acid simvastatin, pravastatin, fluvastatin, atorvastatin, nisvastatin, and rosuvastatin; neuroprotectants, including free radical scavengers, calcium channel blockers, excitatory amino acid antagonists, growth factors, antioxidants, such as edaravone, vitamin C, TROLOX™, citicoline and minicycline, and reactive astrocyte inhibitors, such as (2R)-2-propyloctanoic acid; beta andrenergic blockers, such as propranolol, nadolol, timolol, pindolol, labetalol, metoprolol, atenolol, esmolol and acebutolol;
NMDA receptor antagonists, including memantine; NR2B antagonists, such as traxoprodil; 5-HT1A agonists; receptor platelet fibrinogen receptor antagonists, including tirofiban and lamifiban;
thrombin inhibitors; antithrombotics, such as argatroban; antihypertensive agents, such as enalapril; vasodilators, such as cyclandelate; nociceptin antagonists; DPIV antagonists; GABA 5 inverse agonists; and selective androgen receptor modulators.
[00453] In another embodiment described herein, methods for treatment of LPA-dependent or LPA- mediated conditions or diseases, such as the therapy of interstitial cystitis, comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, by way of example only,
dimethylsulfoxide, omalizumab, and pentosan polysulfate.
[00454] In yet another embodiment described herein, methods for treating LPA-dependent or LPA- mediated conditions or diseases, such as the therapy of respiratory disorders (e.g., asthma, COPD and rhinitis), comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one agent used in the treatment of respiratory conditions. Agents used in the treatment of respiratory conditions include, but are not limited to, bronchodilators (e.g., sympathomimetic agents and xanthine derivatives), leukotriene receptor antagonists, leukotriene formation inhibitors, leukotriene modulators, nasal decongestants, respiratory enzymes, lung surfactants, antihistamines (e.g., mepyramine (pyrilamine), antazoline, diphenhydramine, carbinoxamine, doxylamine, clemastine, dimenhydrinate, pheniramine, chlorphenamine (chlorpheniramine), dexchlorpheniramine, brompheniramine, triprolidine, cetirizine, cyclizine, chlorcyclizine, hydroxyzine, meclizine, loratadine, desloratidine, promethazine, alimemazine (trimeprazine), cyproheptadine, azatadine, ketotifen, acrivastine, astemizole, cetirizine, mizolastine, terfenadine, azelastine, levocabastine, olopatadine, levocetirizine, fexofenadine), mucolytics, corticosteroids, anticholinergics, antitussives, analgesics, expectorants, albuterol, ephedrine, epinephrine, fomoterol, metaproterenol, terbutaline, budesonide, ciclesonide,
dexamethasone, flunisolide, fluticasone propionate, triamcinolone acetonide, ipratropium bromide, pseudoephedrine, theophylline, montelukast, zafirlukast, ambrisentan, bosentan, enrasentan, sitaxsentan, tezosentan, iloprost, treprostinil, pirfenidone, 5-lipoxygenase-activating protein (FLAP) inhibitors, FLAP modulators and 5-LO inhibitors.
[00455] In a specific embodiment described herein, methods for treating LPA-dependent or LPA- mediated conditions or diseases, such as the therapy of asthma and/or COPD, comprises administration to a patient anti-inflammatory agents. In certain embodiments, methods for treating LPA-dependent or LPA-mediated conditions or diseases, such as the therapy of asthma and/or COPD, comprise administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, but not limited to, epinephrine, isoproterenol, orciprenaline, bronchodilators, glucocorticoids, leukotriene modifiers, mast-cell stabilizers, xanthines, anticholinergics, β-2 agonists, FLAP inhibitors, FLAP modulators or 5-LO inhibitors, β-2 agonists include, but are not limited to, short- acting β-2 agonists (e.g., salbutamol (albuterol), levalbuterol, terbutaline, pirbuterol, procaterol, metaproterenol, fenoterol and bitolterol mesylate) and long-acting β-2 agonists (e.g., salmeterol, formoterol, bambuterol and clenbuterol). FLAP inhibitors and/or FLAP modulators include, but are not limited to, 3-[3-tert-butylsulfanyl-l-[4-(6-methoxy-pyridin-3-yl)-benzyl]-5-(pyridin-2- ylmethoxy)- lH-indol-2-yl]-2,2-dimethyl-propionic acid, 3-[3-tert-butylsulfanyl- 1 -[4-(6-ethoxy- pyridin-3-yl)-benzyl]-5-(5-methyl-pyridin-2-ylmethoxy)-lH-indol-2-yl]-2,2-dimethyl-propionic acid, MK-886, MK-0591, BAY-xl005 and compounds found in US 2007/0225285, US
2007/0219206, US 2007/0173508, US 2007/0123522 and US 2007/0105866 (each of which are hereby incorporated by reference). Glucocorticoids include, but are not limited to, beclometasone, budesonide, ciclesonide, fluticasone and mometasone. Anticholinergics include, but are not limited to, ipratropium and tiotropium. Mast cell stabilizers include, but are not limited to, cromoglicate and nedocromil. Xanthines include, but are not limited to, amminophylline, theobromine and theophylline. Leukotriene antagonists include, but are not limited to, montelukast, tomelukast, pranlukast and zafirlukast. 5-LO inhibitors include, but are not limited to, zileuton, VIA-2291 (ABT761), AZ-4407 and ZD-2138 and compounds found in US 2007/0149579, WO2007/016784.
[00456] In another specific embodiment described herein, methods for treating LPA-dependent or LPA-mediated conditions or diseases, such as the therapy of allergic diseases or conditions, comprises administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from, by way of example only, antihistamines, leukotriene antagonists, corticosteroids and decongestants. Leukotriene antagonists include, but are not limited to, montelukast, tomelukast, pranlukast and zafirlukast.
[00457] In one aspect, LPA receptor antagonists described herein are admistered in combination with one or more agents used to treat used to treat asthma, including, but not limited to: combination inhalers (fluticasone and salmeterol oral inhalation (e.g. Advair)); inhaled Beta-2 agonists (albuterol inhaler; albuterol nebulizer solution; formoterol; isoproterenol oral inhalation; levalbuterol;
metaproterenol inhalation; pirbuterol acetate oral inhalation; salmeterol aerosol inhalation;
salmeterol powder inhalation; terbutaline inhaler); inhaled corticosteroids (beclomethasone oral inhalation; budesonide inhalation solution; budesonide inhaler; flunisolide oral inhalation;
fluticasone inhalation aerosol; fluticasone powder for oral inhalation; mometasone inhalation powder; triamcinolone oral inhalation); leukotriene modifiers (montelukast; zafirlukast; zileuton); mast cell stabilizers (cromolyn inhaler; nedocromil oral inhalation); monoclonal antibodies
(omalizumab); oral Beta-2 agonists (albuterol oral syrup; albuterol oral tablets; metaproterenol; terbutaline); bronchodilator (aminophylline; oxtriphylline; theophylline).
[00458] In one aspect, LPA receptor anatogonists described herein are admistered in combination with one or more agents used to treat allergy, including, but not limited to: antihistamine and decongestant combinations (cetirizine and pseudoephedrine; desloratadine and pseudoephedrine ER; fexofenadine and pseudoephedrine; loratadine and pseudoephedrine); antihistamines (azelastine nasal spray; brompheniramine; brompheniramine oral suspension; carbinoxamine; cetirizine;
chlorpheniramine; clemastine; desloratadine; dexchlorpheniramine ER; dexchlorpheniramine oral syrup; diphenhydramine oral; fexofenadine; loratadine; promethazine); decongestants
(pseudoephedrine); leukotriene modifiers (montelukast; montelukast granules); nasal
anticholinergics (ipratropium); nasal corticosteroids (beclomethasone nasal inhalation; budesonide nasal inhaler; flunisolide nasal inhalation; fluticasone nasal inhalation; mometasone nasal spray; triamcinolone nasal inhalation; triamcinolone nasal spray); nasal decongestants (phenylephrine); nasal mast cell stabilizers (cromolyn nasal spray).
[00459] In one aspect, LPA receptor anatogonists described herein are admistered in combination with one or more agents used to treat chronic obstructive pulmonary disease (COPD), including, but not limited to: anticholinergics - ipratropium bromide oral inhalation); combination Inhalers (albuterol and ipratropium (e.g. Combivent, DuoNeb); fluticasone and salmeterol oral inhalation (e.g. Advair)); corticosteroids (dexamethasone tablets; fludrocortisone acetate; hydrocortisone tablets; methylprednisolone; prednisolone liquid; prednisone oral; triamcinolone oral); inhaled Beta- 2 Agonists (albuterol inhaler; albuterol nebulizer solution; formoterol; isoproterenol oral inhalation; levalbuterol; metaproterenol inhalation; pirbuterol acetate oral inhalation; salmeterol aerosol inhalation; salmeterol powder inhalation; terbutaline inhaler); inhaled Corticosteroids
(beclomethasone oral inhalation; budesonide inhalation solution; budesonide inhaler; flunisolide oral inhalation; fluticasone inhalation aerosol; fluticasone powder for oral inhalation; triamcinolone oral inhalation); mukolytics (guaifenesin); oral Beta-2 agonists (albuterol oral syrup; albuterol oral tablets; metaproterenol; terbutaline); bronchodilator (aminophylline; oxtriphylline; theophylline).
[00460] In one embodiment, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) is administered to a patient in combination with inhaled corticosteroids.
[00461] In one embodiment, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) is administered to a patient in combination with beta2-adrenergic receptor agonists. In one embodiment, Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered to a patient in combination with short acting beta2-adrenergic receptor agonists. In one embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g. Compound 2) is administered to a patient in combination with long-acting beta2-adrenergic receptor agonists.
[00462] In one embodiment, Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) is combined with or administered in combination with one or more agents that are inhibitors of UDP-glucuronosyltransferase (UGT). UGT inhibitors include those described in U.S. 2003/0215462; U.S. 2004/0014648. In some embodiments, co-administration of a UGT inhibitor allows for lower doses of Compound 1, or a pharmaceutically acceptable salt thereof (e.g.
Compound 2) to be administered.
[00463] The individual compounds of such combinations are administered either sequentially or simultaneously in separate or combined pharmaceutical formulations. In one embodiment, the individual compounds will be administered simultaneously in a combined pharmaceutical formulation. Appropriate doses of known therapeutic agents will be appreciated by those skilled in the art.
[00464] The combinations referred to herein are conveniently presented for use in the form of a pharmaceutical compositions together with a pharmaceutically acceptable diluent(s) or carrier(s). Kits/Articles of Manufacture
[00465] For use in the therapeutic methods of use described herein, kits and articles of manufacture are also described herein. Such kits include a carrier, package, or container that is
compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein.
Suitable containers include, for example, bottles, vials, syringes, and test tubes. In one embodiment, the containers are formed from a variety of materials such as glass or plastic.
[00466] The articles of manufacture provided herein contain packaging materials. Packaging materials for use in packaging pharmaceutical products include, e.g., U.S. Patent Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, bags, containers, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment. A wide array of formulations of the compounds and compositions provided herein are contemplated as are a variety of treatments for any disease, disorder, or condition that would benefit by antagonism of LP A receptors.
[00467] For example, the container(s) include Compound 1 , or a pharmaceutically acceptable salt thereof (e.g. Compound 2), optionally in a composition or in combination with another agent as disclosed herein. Such kits optionally include an identifying description or label or instructions relating to its use in the methods described herein.
[00468] A kit typically includes labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included. [00469] In one embodiment, a label is on or associated with the container. In one embodiment, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In one embodiment, a label is used to indicate that the contents are to be used for a specific therapeutic application. The label also indicates directions for use of the contents, such as in the methods described herein.
[00470] In certain embodiments, the pharmaceutical compositions are presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein. The pack, for example, contains metal or plastic foil, such as a blister pack. In one embodiment, the pack or dispenser device is accompanied by instructions for administration. In one embodiment, the pack or dispenser is also accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. In one embodiment, compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are also prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
[00471] It is to be understood that as used herein, pharmaceutical compositions described as comprising a pharmaceutically acceptable salt described herein, e.g., liquid solutions, encompass pharmaceutical compositions comprising the associated and/or disassociated forms of the salt. Thus, for example, a pharmaceutical composition described herein comprising an aqueous solution of Compound 2 encompasses a composition comprising a population of sodium cations and a population of {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}- acetate anions.
EXAMPLES
[00472] The following ingredients, formulations, processes and procedures for practicing the methods disclosed herein correspond to that described above. The procedures below describe with particularity illustrative, non- limiting embodiment of formulations that include a Compound 1 , or a pharmaceutically acceptable salt and/or solvate thereof, and pharmacokinetic profiles and pharmacodynamic effects thereof. By way of example only, Compound 1 is optionally prepared as outlined in US patent application 12,638,702, or as outlined herein.
Example 1: Synthesis of 5-(4-Bromo-phenyl)-3-methyl-isoxazole-4-carboxylic acid
Step 1: 3-Methylamino-but-2-enoic acid methyl ester [00473] To a solution of methyl acetoacetate (29.4g, 253mmol) in Methanol (30mL) was added methylamine (33 wt% in EtOH; 48mL, 385mmol) dropwise at room temperature. The reaction was stirred for 1 hour, and then concentrated and dried to give the title compound as a white crystalline solid.
[00474] Step 2: 2-(4-Bromo-benzoyl)-3-oxo-butyric acid methyl ester
[00475] To 3-methylamino-but-2-enoic acid methyl ester (5.0g, 39.1mmol) in THF (70mL) was added pyridine (3.7mL, 47mmol) dropwise. The mixture was cooled to 0°C, and 4-bromobenzoyl chloride (8.55g, 39.1mmol) in THF (30mL) was added dropwise. The reaction was stirred at room temperature overnight, and then water was added. The mixture was extracted with EtOAc, and the combined organic layers were washed with water, dried, filtered, and concentrated to give the title compound.
[00476] Step 3: 5-(4-Bromo-phenyl)-3-methyl-isoxazole-4-carboxylic acid methyl ester
[00477] To a mixture of 2-(4-bromo-benzoyl)-3-oxo-butyric acid methyl ester (1 lg, 39mmol) in acetic acid (50mL) was added hydroxylamine hydrochloride (2.66g, 39mmol), and the reaction was stirred at 115°C for 1 hours. After cooling, saturated aqueous NaHC03 was added to the mixture to adjust to pH 8. The solution was extracted with EtOAc, and the combined organic layers were washed with brine, dried, filtered, and concentrated to give the title compound.
[00478] Step 4: 5-(4-Bromo-phenyl)-3-methyl-isoxazole-4-carboxylic acid
[00479] Lithium hydroxide (2g, 48mmol) was added to a solution of 5-(4-bromo-phenyl)-3-methyl- isoxazole-4-carboxylic acid methyl ester (39mmol) in methanol (50mL) and water (lOmL), and the reaction was stirred at 60°C for 1 hour. Acidic work-up gave the title compound.
Example 2: Synthesis of [4-( A5,5-Tetramethyl-[l,3,21dioxaborolan-2-vO-phenyll-acetic acid ethyl ester and 4-((EthoxycarbonvDmethvDphenylboronic acid
Step 1: [4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-acetic acid ethyl ester
[00480] Ethyl 4-bromophenylacetate (12g, 49mmol), bis(pinacolato)diboron (12.4g, 59mmol), 1,1'- bis(diphenylphosphino)ferrocene)-dichloropalladium(II) (2g, 2.4mmol), and potassium acetate (9.6g,
98mmol) were combined in 1,4-dioxane (lOOmL), and the reaction was heated to 80°C for 6 hours.
The mixture was worked-up to give the title compound.
Step 2: 4-((Ethoxycarbonyl)methyl)phenylboronic acid
[00481] [4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-acetic acid ethyl (0.460g,
1.58mmol), sodium periodate (0.746g, 3.49mmol), and ammonium acetate (0.309g, 4.01mmol) were combined in 1 : 1 acetone:water and stirred overnight at room temperature. Aqueous work-up provided the title compound.
Example 3: Synthesis of {4W3-Methyl-4-(YR -phenyl-ethoxycarbonylamino)-isoxazol-5-yll- biphenyl-4-yll-acetic acid (Compound 1)
Step 1: (R)-l-Phenyl-ethanol [00482] To acetophenone (19.47mL, 166.6mmol) in THF (lOOmL) was added (S)-(-)-2-methyl-CBS- oxazaborolidine (4.62g, 16.6mmol), and the reaction was cooled to 0°C. Borane methyl sulfide complex (2M in THF; 50mL, lOOmmol) was added over 15 minutes, and the reaction was stirred at room temperature. Aqueous work-up gave the title compound.
Step 2: [5-(4-Bromo-phenyl)-3-methyl-isoxazol-4-yl]-carbamic acid (R)-l-phenyl-ethyl ester
[00483] 5-(4-Bromo-phenyl)-3-methyl-isoxazole-4-carboxylic acid (25g, 88.7mmol) in toluene (500mL) was added triethylamine (18.5mL, 133mmol), followed by diphenylphosphoryl azide (22.1mL, 101.9mmol). (R)-(+)- 1 -Phenylethyl alcohol (11.9mL, 97.5mmol) was added, and the reaction was stirred at 75°C for 2 hours. The mixture was partitioned between EtOAc and H 0 and filtered through Celite. The aqueous layer was extracted with EtOAc, and the combined organic layers were dried over MgS04, filtered, and concentrated to give the title compound.
Step 3: {4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}- acetic acid ethyl ester
[00484] [5-(4-Bromo-phenyl)-3-methyl-isoxazol-4-yl]-carbamic acid (R)- 1 -phenyl-ethyl ester (39g, 97.2mmol), [4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-acetic acid ethyl ester (31g, 107mmol), and sodium bicarbonate (32.6g, 389mmol) were combined in 3: 1 DME:H20 (500mL), and the mixture was purged with N2 for 15 minutes. (l,l'-Bis(diphenylphosphino)ferrocene)- dichloropalladium(II) (2.13g, 2.91mmol) was added, and the reaction was purged with N2 for an additional 10 minutes and then stirred at 90°C overnight. The mixture was partitioned between EtOAc and H20, and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with H20, dried over MgS04, filtered, and concentrated, and the residue was purified by silica gel chromatography (EtOAc/hexane gradient) to give the title compound.
Step 4: {4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}- acetic acid
[00485] To a suspension of {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid ethyl ester (24g, 49mmol) in 3: 1 MeOH:H 0 (300mL) was added lithium hydroxide (8.3g, 198mmol), and the reaction was stirred at room temperature overnight. The mixture was acidified and partitioned between EtOAc and H20. The aqueous layer was extracted with EtOAc, and the combined organic layers were washed with H20, dried over MgS04, filtered, and concentrated to give the title compound. Mass spec, data (M+H) = 457.
Example 4: Synthesis of {4W3-Methyl-4-(YS -phenyl-ethoxycarbonylamino)-isoxazol-5-yll- biphenyl-4-yll-acetic acid
[00486] Prepared in a similar manner to the procedures described in Example 3 using (S)- 1 -phenyl- ethanol in place of (R)- 1 -phenyl-ethanol. Mass spec, data (M+H) = 457.
Example 5: Synthesis of {4'-[3-Methyl-4-(l-phenyl-ethoxycarbonylamino)-isoxazol-5-yll- biphenyl-4-yll-acetic acid [00487] Prepared in a similar manner to the procedures described in Example 3 using (R/S)-l- phenyl-ethanol in place of (R)-l-phenyl-ethanol. Mass spec, data (M+H) = 457.
Example 6: Synthesis of {4W3-Methyl-4-(Yl-phenyl-ethoxy-d9)-carbonylamino)-isoxazol-5-yll- biphenyl-4-yll-acetic acid
[00488] Prepared in a similar manner to the procedures described in Example 3 but using 1 - phenyl ethanol-d9 (obtained from Carbocore) in place of (R)-l-phenyl-ethanol. Mass spec, data (M+H) = 466.
[00489] In some embodiments, Mass spectrometric data (mass spec, data) is obtained on with a Shimadzu LCMS 201 OA.
Example 7: Large Scale Synthesis of {4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yll-biphenyl-4-yl|-acetic acid
Step 1.
Figure imgf000083_0001
[00490] To a 200L glass-lined reactor was charged 7.8 kg 5-(4-bromo-phenyl)-3-methyl-isoxazole-4- carboxylic acid and 75.0 kg of toluene. The agitator was started and 3.40kg (R)-(+)-l -phenyl ethanol was charged to the reactor. The reactor was heated to reflux (~113°C) and two thirds of the toluene was atmospherically distilled off. The reactor was cooled to 0°C. 2.5kg of molecular sieves and 5.6 kg of triethylamine were charged to the reactor. The reactor was heated to 80°C. 8.20 kg of diphenylphosphorylazide was dissolved in 6.7 kg of toluene and treated with 2.50 kg of molecular sieves was charged to the reactor using a diaphragm pump. The charge was performed over a period of at least 1 hour while maintaining an internal temperature of 85 ± 10°C and monitoring the gas evolution. The reactor was agitated for 1 hr at an internal temperature of 80 ± 5°C. The reactor was cooled to 20 ± 5°C and 124.0 kg of dichloromethane was charged. The bottom organic layer was washed with 39.0 kg of water three (3) times followed by two (2) washes with 39.0 kg 1M HC1, aqueous solution. The bottom organic layer was bag filtered to remove insoluble solids and sodium sulfate treated. The organic layer was charged back to the reactor and vacuum distilled to remove -55% of the dichloromethane charged to the reactor. 63.9 kg of heptane was charged to the reactor and the reactor agitated at 20 ± 5°C for at least 1 hr. The reactor contents were filtered and washed with 20.0 kg (1 v/2v) toluene/heptane. The filter cake was dried in a vacuum oven at a temperature < 50°C. The oven was unloaded and [5-(4-bromo-phenyl)-3-methyl-isoxazol-4-yl]-carbamic acid (R)- 1-phenyl-ethyl ester was placed in nitrogen flushed double-heavy duty polybags in secondary containment to give a final weight of 11.45 kg of [5-(4-bromo-phenyl)-3-methyl-isoxazol-4-yl]- carbamic acid (R)- 1 -phenyl-ethyl ester.
Step 2.
Figure imgf000084_0001
[00491] Charged to a 200L glass-lined reactor 6.70 kg 4-bromophenylacetate ethyl ester, 7.70 kg of bis(pinacolato)diboron, 5.90kg of potassium acetate, and 35.0 kg of 1 ,4-dioxane. Started agitator. Bubbled nitrogen into the reaction solution for at least 1 hr. Charged 400 g of [ 1 , 1 '- bis(diphenylphosphino)ferrocene]dichloropalladium(II). Bubbled nitrogen into the reaction solution for at least 10 minutes. Heated the reactor to a j acket temperature of 85 °C. Agitated at this j acket temperature for at least 3 hr. Cooled the reactor to a jacket temperature of 20 °C. Agitated at this jacket temperature for at least 1 hr. Filtered the reactor contents through a Nutsche filter which contained a bed of diatomaceous earth to a clean HDPE drum. The reactor contents were rinsed with 35 kg of 1 ,4-dioxane. The filter cake was washed with the reactor contents into the drum.
Step 3
Figure imgf000084_0002
[00492] Charged to a 200L glass-lined reactor 10.5 kg of [5-(4-bromo-phenyl)-3-methyl-isoxazol-4- yl]-carbamic acid (R)- 1 -phenyl-ethyl ester, 66.1 kg of [4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2- yl)-phenyl] -acetic acid ethyl ester in dioxane (theoretical yield of previous step: 8.0kg), and 5.0 kg of 1,4-dioxane. Started the agitator. Charged 42.0 kg of 25% potassium bicarbonate, aqueous solution. Purged nitrogen into the reactor contents for at least 1 hr. Charged 300 g of [Ι, - bis(diphenylphosphino)ferrocene]dichloropalladium(II) (1 : 1 complex with dichloromethane).
Bubbled nitrogen into the reaction solution for at least 10 minutes and then heated the reactor contents to 80 ± 5°C and agitated for at least 2 hr. Cooled the reactor contents to 20 ± 5 °C and drained the bottom layer. Vacuum distilled the reactor contents until rate of distillation decreased significantly. Charged 47.0 kg of ethyl acetate and 58.0 kg of 12% sodium chloride, aqueous solution. Agitated and drained the bottom layer. Charged 63.0 kg of 25% sodium chloride, aqueous solution. Agitated and drained the bottom layer. Drained the {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid ethyl ester in ethyl acetate to drums and rinsed the reactor with 10.0 kg of ethyl acetate. Charged 5.0 kg Sodium Sulfate to the drum.
[00493] Charged the (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl- 4-yl} -acetic acid ethyl ester in EtOAc to a 200L GL Reactor and distilled until the rate decreased significantly. The reactor was held under maximum achievable vacuum with a jacket temperature 45 °C for at least 1 hr. Charged 23 kg of dichloromethane. Agitated until a complete solution was obtained. The reactor contents were drained to a clean HDPE drum and 13 kg of dichloromethane used to rinse the reactor and drained to the same drum. 2 kg of hexanes was charged to the drum.
[00494] A GL Nutsche Filter was setup and loaded with 35.0 kg of silica gel. The silica gel was wet with 30.0 kg (9v/lv) of dichloromethane/hexanes. 26.8 kg of {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid ethyl ester in
dichloromethane/hexanes was charged to the Nutsche filter, and washed with 150.0 kg (9vlv) of dichloromethane/hexanes, 300.0 kg of dichloromethane, 391.0 kg (95v/5v) of dichloromethane/ethyl acetate, and 130.0 kg (9vlv) of dichloromethane/ethyl acetate. The fractions were monitored by
TLC using (5v/95v) EtOAc/DCM. Plates were visualized with UV, spots marked with a pencil, and then stained with PMA.
[00495] A GL Nutsche Filter was setup and loaded with 37.0 kg of Silica Gel. The silica gel was wet with 37.8 kg (9v/lv) of dichloromethane/hexanes. 26.7 kg of {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid ethyl ester in
dichloromethane/hexanes was charged to the Nutsche filter, and washed with 150.0 kg (9v/lv) of dichloromethane/hexanes, 300.0 kg of dichloromethane, 391.8 kg (95v/5v) of
dichloromethane/ethyl acetate, and 119.6 kg (9v/lv) of dichloromethane/ethyl acetate. The fractions were monitored by TLC using (5v/95v) EtOAc/DCM. Plates were visualized with UV, spots marked with a pencil, and then stained with PMA.
[00496] {4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid ethyl ester in dichloromethane/ethyl acetate/hexane/heptane from the plugs was charged to the reactor and distilled until all solvent was removed. 25.0 kg of tetrahydrofuran was charged to the reactor and distilled all solvent was removed. 500 g of silicycle thiol and 56 kg of tetrahydrofuran was charged to the reactor, agitated at a jacket temperature of 45 °C for 8 hr. The reactor contents were bag and polished filter to a clean HDPE drum. The drum contents were charged back to the reactor with 500 g of silicycle thiol. The reactor contents were agitated at a jacket temperature of 45 °C for 8 hr. The reactor contents were bag and polished filter to a clean HDPE drum. The reactor was rinsed with 10 kg of tetrahydrofuran and the rinse transferred through the filters to the drum.
[00497] {4'-[3-Methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid ethyl ester in tetrahydrofuran was distilled in a reactor until the rate of distillation decreased significantly. 35.0 kg (lv/9v) of ethyl acetate/heptane was charged to the reactor and distilled until the rate of distillation decreased significantly.
[00498] 35.0 kg (lv/9v) of ethyl acetate/heptane was charged to the reactor and the reactor heated to a jacket temperature of 45 °C and agitated for at least 2 hr to obtain a uniform slurry. The reactor jacket temperature was cooled to 45°C and agitated for at least 1 hr The reactor contents were filtered using a Nutsche filter. The filter cake was washed with 35.0 kg (1 v/9v) of ethyl
acetate/heptane. Wet (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl- 4-yl} -acetic acid ethyl ester was dried in a vacuum oven at a temperature < 50°C. The oven was unloaded to give a final weight of 8.00 kg of (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid ethyl ester.
Step 4.
Figure imgf000086_0001
[00499] Charged 7.85 kg of {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid ethyl ester, 13 kg of methanol, and 14 kg of tetrahydrofuran to a 200L GL Reactor. Started agitator. Charged 18.2 kg of 2M sodium hydroxide in water and agitated for 2 hr. Performed a vacuum distillation to remove tetrahydrofuran and methanol. Charge 31 kg of water. Heated reactor contents to 40°C. Charged 24 kg of methyl tert-butyl ether (MTBE). Agitated and separated layers. Washed the aqueous layer 2 times with 24 kg of MTBE each time. Charged the aqueous layer back to the reactor and vacuum distill until any residual solvent was removed. Heated the reactor jacket to 45°C. Setup 3 x 50L MN RBF inside heating mantles. Drain the reactor contents equally to the 50L MN RBF. Each flask heated to 45 ± 5°C. Charged 35.4 kg of 1M HC1 in water to the reactor. Transfered the contents of each flask to the reactor. After verification that the pH of the reactor contents < 4, agitate at a jacket temperature of 20°C for 1 hr. Filter the reactor contents washing the filter cake with water. Dry the filter cake in a vacuum oven at a temperature <
50°C.
[00500] Charged (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4- yl} -acetic acid (from oven) and 62 kg of tetrahydrofuran to the reactor. Agitated until a complete solution is obtained. Charged 0.6 kg of silicycle-thiol. Heated the reactor jacket to 45°C. Agitated for 16 hr. Cooled the jacket to 20°C. Polish filtered to a clean HDPE drum. Charged the solution to the reactor. Charged 0.6 kg of silicycle-thiol. Heated the reactor jacket to 45°C. Agitate for 16 hr. Cooled the jacket to 20°C. Polish filtered to a clean HDPE drum. Charged the solution to the reactor and vacuum distill to remove the THF. Charged 70 kg of water. Also charged 4 kg of 6M NaOH in water. Heated the reactor jacket to 45°C. Setup 3 x 50L MN RBF inside heating mantles. Drain the reactor contents equally to the 50L MN RBF. Each flask heated to 45 ± 5°C. Charged 39.8 kg of 0.5 ± 0.2M HC1 in water to the reactor. Transferred the contents of each flask to the reactor. After an additional charge of 4 kg 0.5 ± 0.2M HCL in water to adjust the pH of the reactor contents < 4, agitated at a jacket temperature of 20°C for 1 hr. Filtered the reactor contents, washed the filter cake with water. Dried the filter cake in a vacuum oven at a temperature < 50°C. The oven was unloaded and (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid was screened using a 10-mesh screen to give a final weight of 6.70 kg.
Step 5.
Figure imgf000087_0001
[00501] Charged to a 200L glass-lined reactor 5.30 kg of {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid and 42 kg of ethanol, absolute 200 proof. Started agitation and set up a diaphragm pump to circulate the reactor contents from the bottom of the reactor into the process liquid line of the reactor. A pH meter was placed in front of the diaphragm pump to allow constant pH monitoring of the reactor contents. While monitoring the pH, 963 g of 50% sodium hydroxide was charged to the reactor to adjust the pH of the reactor contents to a pH of 8.7 ± 1. The pH exceeded the desired range, an additional 300 g of {4'-[3- methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid was charged to the reactor to lower the pH.
[00502] Charged 72 kg of heptane was charged to the reactor and the reactor contents vacuum distilled to remove the ethanol. Charged 72 kg of heptane and vacuum distilled to remove the same amount of solvent that was distilled on the first distillation.
[00503] The reactor contents were filtered using a Nutsche Filter. The filter cake was washed with 18 kg of heptane. Dried the filter cake in a vacuum oven at a temperature < 85°C until constant weight was achieved (23 hr). Upon achieving constant weight, the oven temperature was reduced to
45°C. [00504] The oven was unloaded and the sodium salt of {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid was screened using a 10-mesh screen to give a final weight of 5.40 kg.
Example 8: Solubility Assessment of Compound 1 in Organic Solvents and Formation of Crystalline Compound 1
[00505] A solubility assessment of the free acid (Compound 1) was carried out. Twenty lots of 20mg of the free acid (Compound 1) were weighed into vials and treated with solvent and warmed. The observations made are described in Table 1.
Table 1. Solubility of the free acid (Compound 1).
Figure imgf000088_0001
[00506] These experiments were then allowed to cool to room temperature and left overnight.
Samples which had dissolved and then produced a solid upon cooling were filtered and analysed by XRPD and DSC. These samples were shown to provide the crystalline compound 1 (Pattern 1).
[00507] Samples which had dissolved but yielded a solution were allowed to slowly evaporate to produce solids that were isolated and analysed by XRPD. These samples also provided crystalline compound 1 (Pattern 1).
Example 9: Preparation of Crystalline Compound 1 from Compound 2
[00508] Amorphous Compound 2 (approximately 2g) was partitioned between 1M HC1 (150ml) and EtOAc (300ml). The aqueous phase was extracted with EtOAc (2 x 100ml) and the combined organic phases were washed with brine (100ml) dried over MgS04, filtered and concentrated in vacuo. The residue was recrystallised from EtOAc (~20ml) to yield a fine white powder. Yield = 1.012g. Example 10: Crystalline Compound 2 (Pattern 1)
[00509] lOOmg of Compound 1 (free acid) was treated with 2ml of isopropanol and warmed to approximately 70°C before addition of sodium hydroxide (1M in water; 240μ1). The solution was then cooled to 5°C at l0C.min_1. The solids formed were filtered and dried on the filter bed for 1.5 hrs.
Example 11: Crystalline Compound 2 (Pattern 2)
[00510] 500mg of the free acid (Compound 1) was treated with isopropanol (IP A) (10ml) and warmed to approximately 85°C. Sodium hydroxide (1200μ1 of 1M solution in water) was added and the reaction was cooled to 5°C at 0.5°C min 1. After approximately 30 minutes the solid produced was filtered off and dried under suction. Yield = 230mg.
Example 12: Additional Salts of Compound 1
[00511] The free acid (Compound 1) (20mg) was placed in a HPLC vial and treated with 400μ1 of isopropanol, ethanol, tetrahydrofuran, or ethyl acetate. The vial was capped, warmed to ~50°C before addition of a base solution (1.1 eq). Base solutions included 1M KOH (water); 1M NaOH (water); 0.5M L-Arginine (water); 0.5M L-Lysine (water); 2M NH4OH (EtOH); or 1M N-Me- glucamine (water). The reactions were then allowed to cool to room temperature and then to -18°C if no solids had been formed. If no solids had formed after cooling to -18°C the vial caps were removed and allowed the solvent to slowly evaporate at room temperature. Any solids formed were analyzed by XRPD.
[00512] Three experiments produced solids upon cooling - 1M NaOH (water) in IPA; 2M NH4OH (EtOH) in IPA; 2M NH4OH (EtOH) in ethanol. Both ammonium hydroxide experiments were shown to be the free acid, i.e. no salt formation had occurred. The diffractogram observed for the solid obtained with NaOH in IPA was different to the free acid (Compound 1) and not very crystalline.
[00513] The remaining salt formation experiments were allowed to evaporate and the solids obtained were analyzed. The potassium salt formation experiments produced the same XRPD pattern for three experiments, all of low crystallinity. The L-arginine salts were either amorphous or oils. The L-lysine salts showed signs of crystallinity in IPA and THF, in EtOH and EtOAc the products were amorphous. All ammonium salts formation experiments yielded the free acid back, i.e. no salt formation had occurred. The N-methylglucamine experiments yielded material which showed signs of crystallinity with three solvents yielding the same crystalline pattern (IPA, EtOH, EtOAc).
Example 13: X-Ray Powder Diffraction (XRPD)
[00514] X-Ray powder diffraction patterns were collected on a Bruker AXS/Siemens D5000 or Bruker AXS C2 GADDS or Bruker AXS D8 Advance diffractometer.
Bruker AXS/Siemens D5000 [00515] X-Ray Powder Diffraction patterns were collected on a Siemens D5000 diffractometer using Cu Ka radiation (40kV, 40mA), Θ-Θ goniometer, divergence of V20 and receiving slits, a graphite secondary monochromator and a scintillation counter. The instrument is performance checked using a certified Corundum standard (NIST 1976). The software used for data collection was Diffrac Plus XPvD Commander v2.3.1 and the data were analyzed and presented using Diffrac Plus EVA v 11.0.0.2 or v 13.0.0.2.
Ambient conditions
[00516] Samples run under ambient conditions were prepared as flat plate specimens using powder. Approximately 35 mg of the sample was gently packed into a cavity cut into polished, zero- background (510) silicon wafer. The sample was rotated in its own plane during analysis. The details of the data collection are:
• Angular range: 2 to 42 °2Θ
• Step size: 0.05 °2Θ
• Collection time: 4 s'step"1
Bruker AXS C2 GADDS
[00517] X-Ray Powder Diffraction patterns were collected on a Bruker AXS C2 GADDS
diffractometer using Cu Ka radiation (40 kV, 40 mA), automated XYZ stage, laser video microscope for auto-sample positioning and a HiStar 2-dimensional area detector. X-ray optics consisted of a single G5bel multilayer mirror coupled with a pinhole collimator of 0.3 mm. The beam divergence, i.e. the effective size of the X-ray beam on the sample, was approximately 4 mm. A Θ-Θ continuous scan mode was employed with a sample - detector distance of 20 cm which gives an effective 2Θ range of 3.2° - 29.7°. Typically the sample would be exposed to the X-ray beam for 120 seconds. The software used for data collection was GADDS for WNT 4.1.16 and the data were analyzed and presented using Diffrac Plus EVA v 9.0.0.2 or v 13.0.0.2.
Ambient conditions
[00518] Samples run under ambient conditions were prepared as flat plate specimens using powder as received without grinding. Approximately 1-2 mg of the sample was lightly pressed on a glass slide to obtain a flat surface.
Non-ambient conditions
[00519] Samples run under non-ambient conditions were mounted on a silicon wafer with heat- conducting compound. The sample was then heated to the appropriate temperature at ca. 10°C min"1 and subsequently held isothermally for ca 2 minutes before data collection was initiated.
Bruker AXS D8 Advance
[00520] X-Ray Powder Diffraction patterns were collected on a Bruker D8 diffractometer using Cu Ka radiation (40kV, 40mA), Θ-2Θ goniometer, and divergence of V4 and receiving slits, a Ge monochromator and a Lynxeye detector. The instrument was performance checked using a certified Corundum standard (NIST 1976). The software used for data collection was Diffrac Plus XRD Commander v2.5.0 and the data were analyzed and presented using Diffrac Plus EVA v 11.0.0.2 or v 13.0.0.2. Samples were run under ambient conditions as flat plate specimens using powder. Approximately 5 mg of the sample was gently packed into a cavity cut into polished, zero- background (510) silicon wafer. The sample was rotated in its own plane during analysis. The details of the data collection are:
• Angular range: 2 to 42 °2Θ
• Step size: 0.05 °2Θ
• Collection time: 0.5 s step"1
XRPD on Pattern 1 Free Acid (Compound 1)
[00521] The X-Ray powder diffraction pattern for Pattern 1 of the free acid (Compound 1) is displayed in Figure 1 and characteristic peaks are tabulated in Table 2.
Table 2. XRPD pattern peak data for Pattern 1 of Crystalline Compound 1
Figure imgf000091_0001
Pattern 2 Free Acid (Compound 1)
[00522] A variable temperature-XRPD (VT-XRPD) and a cycling DSC experiment was performed on pattern 1 free acid (Compound 1). Selected XRPD diffractograms from the VT-XRPD experiment are shown in Figure 5. It would appear that at a temperature above the small endotherm observed in the DSC (~190°C) a different XRPD pattern is observed. This endothermic transition to a different polymorph would indicate an enantiotropic relationship between the two polymorphs.
XRPD on Pattern 1 of Crystalline Compound 2
[00523] The X-Ray powder diffraction pattern for Pattern 1 of the sodium salt (Compound 2) is displayed in Figure 2 and characteristic peaks are tabulated in Table 3.
Table 3. XRPD pattern peak data for Pattern 1 of Crystalline Compound 2
Figure imgf000091_0002
Angle 2-Theta ° Intensity %
13.1 7.0
13.5 11.1
14.7 8.4
15.1 5.0
15.8 60.8
17.1 12.2
18.3 15.0
19.5 8.5
21.1 15.8
21.7 18.1
XRPD on Pattern 2 of Crystalline Compound 2
[00524] The X-Ray powder diffraction pattern for Pattern 2 is displayed in Figure 3
characteristic peaks are tabulated in Table 4.
Table 4. XRPD pattern peak data for Pattern 2 of Crystalline Compound 2
Figure imgf000092_0001
Example 14: Differential Scanning Calorimetry (DSC) and Thermogravimetric analysis
[00525] DSC data were collected on a TA Instruments Q2000 equipped with a 50 position auto- sampler. The calibration for thermal capacity was carried out using sapphire and the calibration for energy and temperature was carried out using certified indium. Typically 0.5-3 mg of each sample, in a pin-holed aluminum pan, was heated at 10°C min"1 from 25°C to 220°C. A purge of dry nitrogen at 50 ml'min"1 was maintained over the sample. The instrument control software was Advantage for Q Series v2.8.0.392 and Thermal Advantage v4.8.3 and the data were analyzed using Universal Analysis v4.3A.
[00526] TGA data were collected on a TA Instruments Q500 TGA, equipped with a 16 position auto- sampler. The instrument was temperature calibrated using certified Alumel. Typically 5-30 mg of each sample was loaded onto a pre-tared platinum crucible and aluminum DSC pan, and was heated at 10°C min"1 from ambient temperature to 350°C. A nitrogen purge at 60 ml min"1 was maintained over the sample. The instrument control software was Advantage for Q Series v2.8.0.392 and Thermal Advantage v4.8.3
Pattern 1 Free Acid (Compound 1)
[00527] A sample of the free acid (Compound 1) were analyzed by TGA and DSC and the thermogram is shown in Figure 4. The DSC shows a small endotherm, onset 157°C (ΔΗ 5.4 J'g"1) followed by a much larger endotherm onset at 198°C (ΔΗ 88.1 J'g"1). The TGA shows no losses until decomposition with 95% mass remaining at 210°C.
Pattern 2 Free Acid (Compound 1)
[00528] A variable temperature-XRPD (VT-XRPD) and a cycling DSC experiment was performed. Selected XRPD diffractograms from the VT-XRPD experiment are shown in Figure 5.
[00529] It would appear that at a temperature above the small endotherm observed in the DSC (~190°C) a different XRPD pattern is observed. This endothermic transition to a different polymorph would indicate an enantiotropic relationship between the two polymorphs. This was examined further by a cycling DSC experiment as shown in Figure 6.
[00530] In this experiment a sample of the free acid (Compound 1) was heated to 180°C at 10°C min" 'and then cooled to -80°C at 10°C min"1 before being heated to 220°C again at 10°C min"1. We see the expected small endotherm on the first heating ramp and, upon cooling see an exothermic event at approximately 120°C. Upon reheating we see the small endothermic event again before the larger endotherm due to melting proving that the two polymorphs can readily interconvert with each other depending upon the temperature. This reversibility of the endothermic event proves the enantiotropic (reversible stability according to temperature) relationship between Patterns 1 and 2.
Pattern 1 of Crystalline Compound 2 (post GVS at 25°C)
[00531] The sodium salt (Compound 2) showed a gradual loss in the TGA (Figure 7) of 5.5% before decomposition. The DSC showed a single broad endotherm onset 176°C (ΔΗ 30 J'g"1).
Example 15: Gravimetric Vapour Sorption (GVS)
[00532] In some embodiments, sorption isotherms were obtained using a Hiden IGASorp moisture sorption analyzer, controlled by CFRSorp software. The sample temperature was maintained at 25°C by a Huber re-circulating water bath. The humidity was controlled by mixing streams of dry and wet nitrogen, with a total flow rate of 250 ml min- 1. The relative humidity was measured by a calibrated Vaisala RH probe (dynamic range of 0-95 %RH), located near the sample. The weight change (mass relaxation) of the sample as a function of %RH was constantly monitored by the microbalance (accuracy ±0.001 mg). Typically 10-20 mg of sample was placed in a tared mesh stainless steel basket under ambient conditions. The sample was loaded and unloaded at 40 %RH and 25°C (typical room conditions).
[00533] A moisture sorption isotherm was performed as outlined below (2 scans giving 1 complete cycle). The standard isotherm was performed at 25 °C at 10 %RH intervals over a 0-90 %RH range. Table 5. Method Parameters for Hiden IGASorp Experiments
Figure imgf000094_0001
[00534] The software uses a least squares minimization procedure together with a model of the mass relaxation, to predict an asymptotic value. The measured mass relaxation value must be within 5% of that predicted by the software, before the next %RH value is selected. The minimum equilibration time was set to 1 hour and the maximum to 4 hours.
[00535] The sample was recovered after completion of the isotherm and re-analyzed by XRPD.
[00536] In some embodiments, sorption isotherms were obtained using a SMS DVS Intrinsic moisture sorption analyzer, controlled by SMS Analysis Suite software. The sample temperature was maintained at 25°C by the instrument controls. The humidity was controlled by mixing streams of dry and wet nitrogen, with a total flow rate of 200 ml min-1. The relative humidity was measured by a calibrated Rotronic probe (dynamic range of 1.0-100% RH), located near the sample. The weight change (mass relaxation) of the sample as a function of %RH was constantly monitored by the microbalance (accuracy ±0.005 mg). Typically 5-20 mg of sample was placed in a tared mesh stainless steel basket under ambient conditions. The sample was loaded and unloaded at 40 %RH and 25°C (typical room conditions). A moisture sorption isotherm was performed as outlined below (2 scans giving 1 complete cycle). The standard isotherm was performed at 25°C at 10 %RH intervals over a 0.5-90 %RH range.
Table 6. Method Parameters for SMS DVS Intrinsic Experiments
Figure imgf000094_0002
[00537] Samples of the free acid and sodium salt were analyzed by GVS. The isotherms are shown in Figure 8 and Figure 9, respectively. Pattern 1 of Compound 1
[00538] The free acid (Compound 1) only showed slight hygroscopicity taking up ~2% mass on going to from 40% - 90% RH. There was no significant hysteresis and no evidence of a form change The XRPD diffractogram post GVS showed no change. The total uptake from 0-90%RH was -2.4%
Pattern 1 of Compound 2
[00539] The sodium salt sample (Compound 2) was hygroscopic taking up 17.9%) mass on going from 40-90%) RH. There was no hysteresis and no evidence of a form change. The total uptake for 0-90%oRH was 19.1% The XRPD diffractogram post GVS showed a small increase in crystallinity.
Example 16: Thermodynamic Aqueous Solubility
[00540] Aqueous solubility was determined by suspending sufficient compound in water to give a maximum final concentration of >10 mg ml"1 of the parent free-form of the compound. The suspension was equilibrated at 25 °C for 24 hours then the pH was measured. The suspension was then filtered through a glass fiber C filter into a 96 well plate unless stated otherwise. The filtrate was then diluted by a factor of 101. Quantification was by HPLC with reference to a standard solution of approximately 0.25 mg ml"1 in DMSO. Different volumes of the standard, diluted and undiluted sample solutions were injected. The solubility was calculated using the peak areas determined by integration of the peak found at the same retention time as the principal peak in the standard injection.
[00541] For solubility assessment at various pH levels sufficient material to produce a maximum concentration of 10 mg ml"1 of API was treated with 0.15M NaCl solution and then the pH adjusted with HC1 or NaOH solutions to achieve the desired pH levels. The suspensions were allowed to equilibrate for 2 hours and the pH measured and adjusted if necessary. Suspensions were then filtered and the amount of dissolved API quantified by HPLC against a standard reference solution. Table 7. HPLC Method Parameters for Solubility Measurements
Figure imgf000095_0001
[00542] Analysis was performed on an Agilent HP 1100 series system equipped with a diode array detector and using ChemStation software vB.02.01-SRl.
[00543] The thermodynamic aqueous solubility of both the free acid (Compound 1) and the sodium salt (Compound 2) were determined at a variety of pH levels. Analysis and observations were carried out after 2 hours. The results for the free acid (Compound 1) are shown in Table 8.
Table 8. Thermodynamic aqueous solubility of free acid (Compound 1) at various pH levels.
Figure imgf000096_0001
ND =Not Detected. * = completely dissolved, therefore not analyzed by HPLC. Result is based on the amount of material and volume used in the test.
[00544] Due to the gelling effect noticed for the higher pH levels a maximum concentration of 5 mg ml"1 was aimed for when testing the sodium salt (Compound 2), otherwise the method was identical to that described for the free acid (Compound 1). The results for the sodium salt
(Compound 2) are shown in Table 9.
Table 9. Thermodynamic aqueous solubility of the sodium salt (Compound 2) at various pH levels.
Figure imgf000096_0002
ND =Not Detected.
Example 17. Chemical Purity Determination
[00545] Purity analysis was performed by HPLC on an Agilent HP 1100 series system equipped with a diode array detector and using ChemStation software vB.02.01-SRl.
Table 10. HPLC Method Parameters for Chemical Purity Determinations
Figure imgf000096_0003
Injection (μΐ): 5
Detection:
Wavelength, Bandwidth( nm): 255, 90
Flow Rate (nil'min 1): 2.0
Phase A: 0.1%TFA in water
Phase B: 0.085% TFA in acetonitrile
Timetable: Time (min) % Phase A % Phase B
0 95 5
6 5 95
6.2 95 5
8 95 5
[00546] Samples of Compound 1 and Compound 2 were found to be greater than 90% pure. In some embodiments, samples of Compound 1 were found to be greater than 95%) pure, greater than 96%) pure, greater than 97%o pure, greater than 98%o pure, greater than 99%o pure. In some embodiments, samples of Compound 2 were found to be greater than 94%o pure, greater than 95%o pure, greater than 96%o pure, greater than 97%> pure, greater than 98%> pure, greater than 99%> pure.
Example 18. Chiral Purity
[00547] Chiral purity analysis was performed by HPLC on an Agilent HP 1 100 series system equipped with a diode array detector and using ChemStation software vB.02.01-SRl .
Table 11. HPLC Method Parameters for Chemical Purity Determinations
Figure imgf000097_0001
[00548] Chiral purity (%> enatiomeric excess; %> e.e.) was determined. In some embodiments, samples of Compound 1 and Compound 2 were found to have greater than 98%> e.e. In some embodiments, samples of Compound 1 were found to have greater than 95%> e.e., greater than 96%> e.e., greater than 97%> e.e., greater than 98%> e.e., or greater than 99%> e.e. In some embodiments, samples of Compound 2 were found to have greater than 94%> e.e., greater than 95%> e.e., greater than 96%> e.e., greater than 97%> e.e., greater than 98%> e.e., or greater than 99%> e.e.
Example 19. Residual Solvents
[00549] The test for Residual Solvents is performed to detect trace amounts of solvents used in the synthesis that may be present in the API. The analysis is performed via headspace or direct injection analysis using a gas chromatograph equipped with a flame ionization detector (FID). All residual solvents used in the synthesis are capable of being detected by this method.
[00550] Potential residual solvents include acetone, ethanol, methanol, dichloromethane, methyl-tert- butyl-ether (MTBE), ethyl acetate, tetrahydrofuran, heptane, dimethoxyethane (DME), toluene. Table 11. Residual Solvents by GC Headspace for Crystalline Compound 1
Figure imgf000098_0001
Table 12. Residual Solvents by GC Headspace for Amorphous Compound 2
Figure imgf000098_0002
Example 20: Heavy Metals (Pd) by ICP-AES
[00551] Trace palladium (Pd) resulting from the use of catalytic amounts of Pd in the synthesis is assayed by inductively coupled plasma atomic emission spectrometry (ICP-AES). Pd content by ICP-AES is a detectable amount of palladium that is less than about 20ppm. Pd content by ICP-AES is less than about 20ppm. Pd content by ICP-AES is a detectable amount of palladium that is less than 20ppm, less than 15ppm, less than lOppm, or less than 5ppm. Pd content by ICP-AES is less than 20 ppm, less than 15ppm, less than lOppm, or less than 5ppm. In some embodiments, samples or pharmaceutical compositions do not include a detectable amount of palladium.
Example 21: Heavy Metals (as Lead)
[00552] This test is performed according to USP<231> Method II.
Pharmaceutical Compositions
[00553] Pharmaceutical compositions that include Compound 1 , including pharmaceutically acceptable salts (e.g. Compound 2) and/or pharmaceutically acceptable solvates thereof include a variety of forms. In one aspect, pharmaceutical compositions are in the form of oral dosage forms. In some embodiments, the oral dosage forms are formulated as: oral solutions, oral suspensions, tablets, pills, capsules, ointments, creams or gels.
Example 22: Oral Solutions
[00554] In one aspect, an oral pharmaceutical composition in the form of an oral solution is prepared as outlined below.
[00555]
[00556] An oal solution is prepared at 20 mg/mL of Compound 1 or Compound 2.
Oral Solution A:
[00557] In one embodiment, an oral pharmaceutical composition is prepared with the following ingredients:
- 20 mg/mL of Compound 1 or Compound 2
- aqueous 10mM Na2CO3
- 20% propylene glycol
Oral Solution B:
[00558] In one embodiment, an oral pharmaceutical composition is prepared with the following ingredients:
- 20 mg/mL of Compound 1 or Compound 2
- aqueous 10mM Na2CO3
- 20% propylene glycol
- 2% Tween 80
Oral Solution C:
[00559] In one embodiment, an oral pharmaceutical composition is prepared with the following ingredients:
- 20 mg/mL of Compound 1 or Compound 2
- 0.5%) methocel aqueous solution
[00560] The manufacturing process for the oral solutions of Compound 1 or Compound 2 described above is as follows: weigh the required amount of sodium carbonate (if present) and transfer to the container. Add the required amount of water to make a lOmM solution and mix until dissolved. Weigh the required amount of propylene glycol and Tween 80 (if present) and add this to the solution and mix until homogenous. Weigh the required amount of Compound 1 or Compound 2 and slowly add to the solution. Mix until all Compound 1 or Compound 2 is dissolved (sonicate, warm, or stir if necessary).
Example 23: Capsule Formulations
Immediate Release Capsules [00561] In one embodiment, a capsule formulation of Compound 1 or Compound 2 for administration to humans is prepared with the following ingredients:
Figure imgf000100_0001
[00562] Matching Placebo Capsules (for clinical study purposes) are prepared with the following ingredients:
Figure imgf000100_0002
[00563] The process to prepare Compound 1 or Compound 2 in a capsule (which is the same for the preparation of the placebo capsules) is as follows: Weigh the required amount of Compound 1 or Compound 2, add into the appropriate size capsule, and close capsule. For example, 50-200 mg of Compound 1 or Compound 2 is placed into a Size 4 Capsule. In one embodiment, 100-500 mg of Compound 1 or Compound 2 is placed into a Size 1 Capsule.
Example 24: Immediate Release Tablets
[00564] Non- limiting examples of immediate release tables that include either Compound 1 or Compound 2 are presented below.
Immediate release Tablet No. 1
Immediate
Figure imgf000100_0003
Immediate release Tablet No. 3
Figure imgf000100_0004
Sodium Stearyl Fumarate 7.5 1.0
Total 750 100
[00565] A non- limiting example for the preparation of immediate release tablets is described below. Other dose amounts are contemplated. In some cases, the tablets are coated with a thin film (e.g. opadry coating).
[00566] The manufacturing process includes blending and compression. An optional coating process can be performed. Manufacturing/analytical equipment typically used in the preparation of tablets include: formulation (U.S.A. standard testing sieve; V-shell blender; ERWEKA TBH300 MD hardness tester; Vanderkamp friability tester; Manesty beta press, sixteen station); analytical (Agilent 1100 series HPLC with variable wavelength detector; VanKel model VK7000 dissolution apparatus; VanKel model VK8000 dissolution autosampler).
Method of Manufacture of Immediate Release Tablets
[00567] Blending: Add all excipients (except lubricant) to a V-shell blender. Add Compound 1 or Compound 2 to the V-shell blender. Mix in the V-shell blender for approximately 15 minutes. Add lubricant and blend for 2 minutes.
[00568] Tableting: Tablet cores are compressed on a Manesty Betapress sixteen station with two sets of 3/8" round standard concave tooling. During compression, individual and average tablet weight, hardness, thickness, and friability are monitored.
Dissolution Studies
[00569] In some embodiments, all tablets are tested for dissolution using the following parameters:
Figure imgf000101_0001
[00570] Immediate release tablets show release no less than (NLT) 80% of Compound 1 or Compound 2 within 10 minutes.
[00571] Tablets are optionally packaged in 40 count in 50 cc HDPE bottles, with CRC caps and heat induction seal.
Example 25: Enteric Coated tablets
[00572] Enteric coated tablets are prepared with the ingredients listed in Table 13.
Table 13. Enteric Coated tablets
Figure imgf000101_0002
Eudragit L 100 20
Triethyl Citrate 5 5 5
Acetone
Purified Water
Total 775 775 775
[00573] The preparation of the enteric coated tablets is as follows: Weigh 388.0 g of acetone and 12. Og of purified water and mix them in a beaker with an overhead stirrer. Weigh 40 g of the Eudragit and pour into the solvent mixture slowly in portions to prevent lump formation. Stir until a clear solution is made. Then weigh 6g of triethyl citrate and add into the clear solution and keep stirring until a homogeneous solution is made. Mix around 60g of placebo tablets with about 80 of the 750 mg immediate release tablets and coat with the coating mixture.
Example 26: Sustained Release tablets
[00574] The blend of the formulation is prepared in the same manner as the immediate release tablets (e.g. sieving, blending, and compression). Other preparations are acceptable, such as wet granulation, fliudized bed, high shear granulation, etc. The formulation includes drug modifying release excipients. These excipients include but not limited to HPMC (hydroxy propyl
methylcellulose or hypromellose), methacrylic polymers, polyvinyl acetate, and povidone. The amount of drug release modifying excipient ranges from about 10% to about 80%) in the formulation. The drug release profile ranges from 0 to 4 hours, 0 to 6 hours, 0 to 8 hours, 0 to 12 hours, 0 to 24 hours, 2 to 4 hours, 2 to 6 hours, etc. In some embodiments, the formulations are coated with the Opadry coatings after direct compression. Example sustained release formulations are listed below. Table 14. Sustained Release Tablet
Figure imgf000102_0001
Table 14. Sustained Release Tablet
Figure imgf000102_0002
Example 27: Ointment Compositions
[00575] A non-limiting example of an ointment composition is presented in Table 15.
Table 15. PEG Ointment Compositions
Components (%w/w) Formula #
Figure imgf000103_0001
Example 28: Gel Compositions
[00576] A non- limiting example of a gel composition is presented in Table 16. Table 16.
Figure imgf000103_0002
Example 29: Cream Compositions
[00577] A non- limiting example of a cream composition is presented in Table 17.
Table 17.
Figure imgf000103_0003
Figure imgf000104_0001
Example 30: Identification of Metabolic Pathways
[00578] Compound 1 metabolites formed during incubation of Compound 1 with rat, dog, monkey, and human liver microsomes, rat, dog, monkey and human hepatocytes, as well as those generated in vivo and isolated from rat bile and rat and dog plasma were investigated.
Materials
[00579] Male Sprague-Dawley rat, male beagle dog, male rheses monkey and mixed pool human liver microsomes were purchased from BD Gentest (Kansas City, MO). Rat, dog, monkey and human hepatocytes and InVitroGRO HI medium were purchased from In Vitro Technologies (Gaithersburg, MD). Fresh human hepatocytes were purchased from CellzDirect, lot Hu8080. Microsomes
[00580] To determine the qualitative metabolic profile, 30 μΜ of Compound 1 was incubated aerobically with rat, dog, monkey, or human liver microsomes (1 mg/mL). The incubations were performed in phosphate buffer at pH7.4, 37°C, with the reaction initiated by the addition of β- NADPH and UDPGA (1 and 2 mM mM final concentration, respectively). The reaction was terminated by the addition of 3 times incubation volume of acetonitrile after 60 min. The sample was centrifuged and the supernatant was transferred, nitrogen blow dried and reconstituted for LC/MS analysis.
Hepatocytes
[00581] Rat, dog, monkey, or human hepatocytes were thawed according to the supplier's instructions. Cells were counted using the Trypan Blue method, and then diluted to 1 x 106 viable cells/ml with KB medium. Compound 1 was tested at 30 μΜ and incubated for up to 2 hours in rat hepatocytes and 4 hours in dog, monkey or human hepatocytes at 37°C. Fresh human hepatocytes were from a single male donor lot Hu8080 (CellsDirect, Raleigh, NC). Reactions were terminated with addition of 3 times incubation volume of acetonitrile, centrifuged, and supernatants were transferred, nitrogen blow dried and reconstituted for LC/MS analysis.
Rat Bile Duct Cannulation
[00582] Rats with surgically placed bile duct and jugular vein cannula were purchased from Charles River Laboratories and allowed to acclimate for 2 days. Compound 1 was intravenously dosed (2 mg/kg) to three rats as a solution in 0.9% saline (2 mg/mL; 1 mL/kg). Bile samples were collected at time-points 0-2, 2-5 and 5-8, and urine samples were collected at time-points 0-4 and 4-8 hrs post- dose in 20 mL scintillation vials and stored at -40°C until analysis.
LC-MS Analysis
[00583] In the positive ion mode (ESI) by multiple reaction monitoring of parent (MH+/daughter are noted in Table 1) and the internal standard, buspirone (386.2— »· 122.2). The mobile phases contained 10 mM ammonium acetate in water with 0.05% formic acid (solvent A) and lOmM ammonium acetate in 50% acetonitrile/50%) methanol with 0.05% formic acid (solvent B). For AM095, the flow rate was maintained at 1 mL/min and the total run time was 2.5 min. Analytes were separated using a linear gradient as follows:
1. mobile phase was held for 0.5 min at 5% B,
2. B was increased from 5% to 95% over then next 0.2 min,
3. B was held constant for 1.3 min at 95%, and
4. B was returned to the initial gradient conditions.
[00584] For metabolite quantification, the flow rate was maintained at 0.25 mL/min and the total run time was 30 min. Analytes were separated using a linear gradient as follows:
1. mobile phase was held for 5 min at 5% B,
2. B was increased from 5% to 95% over then next 35 min,
3. B was held constant for 15 min at 95%,
4. B was held constant for 5 min at 95%, and
5. B was returned to the initial gradient conditions
Results
[00585] The following metabolites were observed:
Table 18. Metabolites of Compound 1
Figure imgf000105_0001
Figure imgf000106_0001
[00586] In vivo metabolism shows the same metabolites as in vitro, and the taurine conjugate appears to be a major route of metabolism
[00587] The major metabolites generated in vitro appear to be a taurine conjugate, N-dealkylated amine, glycosylation of the amine, mono oxidation, and a glucuronide. Following intravenous dosing of Compound 1, the major metabolites isolated from rat bile are several taurine conjugates, N-dealkylated amine, mono oxidation, and a glucuronide. In urine from these animals, no metabolites were found. The only metabolite circulating in rat and dog plasma was the taurine conjugate.
Example 31: Preparation of Authentic Samples of Representative Metabolites
[00588] Synthesis of [4'-(4-Amino-3-methyl-isoxazol-5-vD-biphenyl-4-yll-acetic acid: [4'-(4-tert- Butoxycarbonylamino-3-methyl-isoxazol-5-yl)-biphenyl-4-yl]-acetic acid (l.Ommol) was treated with trifluoroacetic acid (5mL) for 1 hour. Work-up provided the title compound. Mass spec, data (M+H) = 309.
[00589] Synthesis of 2-(2-i4'-[3-Methyl-4- R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yll- biphenyl-4-yl}-acetylamino)-ethanesulfonic acid: Rats were dosed with {4'-[3-ethyl-4-((R)-l- phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (30 mg/kg) and the bile was collected over 6 hours. The title compound was purified from bile by reverse phase HPLC. Mass spec, data (M+H) = 564.
[00590] Synthesis of (4'-{4-[(R)-l-(3-Hydroxy-phenyl)-ethoxycarbonylamino]-3-methyl- isoxazol-5-yl}-biphenyl-4-yl)-acetic acid: Prepared in a similar manner to the procedures described in Example 3 using 3-((R)-l-hydroxy-ethyl)-phenol in place of (R)- 1 -phenyl-ethanol. Mass spec, data (M+H) = 473.
Example 32. Pharmacokinetics in Sprague-Dawley Rats
[00591] The pharmacokinetics of Compound 1 and Compound 2 was assessed in male and female Sprague-Dawley rats.
[00592] Male or female Sprague-Dawley rats surgically cannulated in their jugular vein (approximate weight 300g for male and 225g for female) were purchased from Charles River (Wilmington, MA).
[00593] Compound 1 or Compound 2 (0.2 mg/kg or 1 mg/kg) was administered intravenously (IV) to fasted rats (n = 2) as a solution in 0.9% saline via a bolus injection into the jugular vein (2 mg/mL; 1 mL/kg). Compound 1 or Compound 2 (10 mg/kg) was administered orally (PO), to fasted (for at least 12 hours) or non-fasted rats (n = 2 or 3) as a solution in 0.5% methylcellulose unless otherwise noted via an oral gavage to the stomach (3.33 mg/mL; 3 mL/kg). Fed animals were fasted for at least 12 hours and then were given food ad libitum for 1 hour prior to dosing. For dose proportionality studies, Compound 1 or Compound 2 was administered PO at 1, 30, 100 or 300 mg/kg as a solution in 0.5%) methylcellulose via an oral gavage to the stomach (0.69 mg/mL, 10 mg/mL, 33.3 mg/mL or 100 mg/mL) (n= 2 or 3 per dose group). In the gastrointestinal study, rats were anesthetized and Compound 1 or Compound 2 was administered directly to the duodenum or ileum at 10 mg/kg in 0.5%) methylcellulose.
[00594] After intravenous administration of 0.2 or 1 mg/kg of Compound 1 or Compound 2 to male and female rats, respectively, the compound showed a moderate systemic clearance value of 1.7 and 1.2 mL/min/kg, an estimated volume of distribution value to be 0.2 and 0.4 L/kg, and a 1.9 and 5.1 hr terminal-life in male and female rats, respectively. Oral administration of 1 mg/kg showed the apparent bioavailability to be 76 and 100%, with a Cmax value of 3.5 and 3.9 μg/mL, respectively in male and female rats. The AUC value of Compound 1 increases in a linear fashion with doses up to 300 mg/kg in male and female rats, with females having a 2-fold AUC value compared to males. In non-fasted male rats, orally dosed compound (1 and 10 mg/kg) had reduced AUC and Cmax values compared to fasted animals. In female rats, nonfasted animals were similar to fasted with respect to AUC and Cmax values. The pharmacokinetic results suggest that Compound 1 has systemic exposure and a half-life in rats of >1.9 hrs, and food affects its oral bioavailability in males.
Example 33. Pharmacokinetics in Male Beagle Dogs
[00595] The pharmacokinetics of Compound 1 was assessed in male Beagle dogs.
[00596] Dosing of male Beagle dogs (n =3) was performed. Compound 1 or Compound 2 was dosed intravenously at 2 mg/kg (fasted) and orally (fasted or non-fasted) at 5 mg/kg. In fasted animals, dogs were fasted a minimum of 12 hours prior to dosing and then were non- fasted 4 hours post-dose. Non- fasted animals were allowed to feed 1 hour prior to dosing ad libitum.
[00597] After intravenous administration of 2 mg/kg, the compound showed a systemic clearance value of 7.8 mL/min/kg, an estimated volume of distribution value of 0.2 L/kg, and a 1.5 hr terminal half-life. Oral administration of 5 mg/kg in solution to fasted dogs showed an apparent oral bioavailability of greater than 100%, with a Cmax value of 10.6 μg/mL. There was little change in the AUC values with compound administered in the non-fasted state or as the free acid in fasted dogs. Upon dosing compound as a powder in a capsule (5 mg/kg), the oral AUC and Cmax values decreased 2 - 3 -fold when compared to that of the solution. Pharmacokinetic results suggest that Compound 2 and Compound 1 have systemic exposure and food has little effect on the absorption in dog, while administration as powder in a capsule decreases the dog exposure.
Figure imgf000108_0001
[00598] A 1.1 kb cDNA encoding the human LPAi receptor was cloned from human lung. Human lung RNA (Clontech Laboratories, Inc. USA) was reverse transcribed using the RETROscript kit (Ambion, Inc.) and the full-length cDNA for human LPAi was obtained by PCR of the reverse transcription reaction. The nucleotide sequence of the cloned human LPAi was determined by sequencing and confirmed to be identical to the published human LPAi sequence (An et al.
Biochem. Biophys. Res. Commun. 231 :619 (1997). The cDNA was cloned into the pCDNA5/FRT expression plasmid and transfected in CHO cells using lipofectamine 2000 (Invitrogen Corp., USA). Clones stably expressing human LPAi were selected using hygromycin and identified as cells that show Ca-flux in response to LPA.
Example 35. Generation of Cells Transiently Expressing Human hLPA?
[00599] An expression vector encoding the human LPA2 cDNA was transiently transfected into B 103 cells using Lipofectamine™ 2000 (Invitrogen) following the manufacturers instruction. On the day before the assay, 30,000-35,000 cells/well were seeded together with 0.2 μΐ lipofectamine 2000 and 0.2 μg human LPA2 expression vector in 96-well Poly-D-Lysine coated black-wall clear-bottom plates (BD BioCoat) in DMEM + 10%) FBS. Following an overnight culture, cells were washed once with PBS then cultured in serum- free media for 4 hours prior to start of the calcium flux assay.
Example 36. Establishment of a CHO Cell Line Stably Expressing Human LPA^ [00600] A vector containing the human LPA3 receptor cDNA was obtained from the Missouri S&T cDNA Resource Center (www.cdna.org). The full-length cDNA fragment for human LPA3 was obtained by PCR from the vector. The nucleotide sequence of the cloned human LPA3 was determined by sequencing and confirmed to be identical to the published human LPA3 sequence (NCBI accession number NM_012152). The cDNA was cloned into the pCDNA5/FRT expression plasmid and transfected in CHO cells using lipofectamine 2000 (Invitrogen Corp., USA). Clones stably expressing human LPA3 were selected using hygromycin and identified as cells that show Ca- flux in response to LPA.
Example 37. LPA1 and LPA3 Calcium Flux Assays.
[00601] Human LPAi or LPA3 expressing CHO cells are seeded at 20,000-45,000 cells per well in a 96-well poly-D-lysine coated plate one or two days before the assay. Prior to the assay, the cells are washed once with PBS and then cultured in serum- free media for at least 6 hrs and up to 24hrs . On the day of the assay, a calcium indicator dye (Calcium 5, Molecular Devices) in assay buffer (HBSS with Ca2+ and Mg2+ and containing 20 mM Hepes and 0.3% fatty-acid free human serum albumin) is added to each well and incubation continued for 1 hour at room temperature. 10 μΐ of test compounds in 2.5% DMSO are added to the cells and incubation continued at room temperature for 30 minutes. Cells are the stimulated by the addition of 10 nM LPA and intracellular Ca2+ measured using the Flexstation 3 (Molecular Devices). IC50s are determined using Symyx Assay Explorer analysis of drug titration curves.
Example 38. LPA2 Calcium Flux Assay.
[00602] LPA2 calcium flux is measured using at least one of two different assays. In one assay, BT- 20 human breast cancer cells are seeded at 25,000-35,000 cells per well in 150 μΐ complete media on Poly-D-Lysine coated black-wall clear-bottom plates. Following an overnight culture, cells are washed once with PBS then serum starved for 4-6 hours prior to the assay. On the day of the assay, a calcium indicator dye (Calcium 5, Molecular Devices) in assay buffer (HBSS with Ca2+ and Mg2+ and containing 20 mM Hepes and 0.3%) fatty-acid free human serum albumin) is added to each well and incubation continued for 15 minutes at 37°C. 25 μΐ of test compounds in 2.5% DMSO are added to the cells and incubation continued at 37°C for 15-30 minutes. Cells are the stimulated by the addition of 100 nM LPA and intracellular Ca2+ measured using the Flexstation 3 (Molecular Devices). IC50s are determined using Symyx Assay Explorer analysis of drug titration curves In the second assay, B 103 cells transiently expressing huma LPA2 are serum starved for 4 hours. A calcium indicator dye (Calcium 4, Molecular Devices) in assay buffer (HBSS with Ca2+ and Mg2+ and containing 20 mM Hepes and 0.3%) fatty-acid free human serum albumin) is then added to each well and incubation continued for 1 hour at 37°C. 10 μΐ of test compounds in 2.5%o DMSO are added to the cells and incubation continued at room temperature for 30 minutes. Cells are the stimulated by the addition of 10 nM LPA and intracellular Ca2+ measured using the Flexstation 3 (Molecular Devices). IC50S are determined using Symyx Assay Explorer analysis of drug titration curves.
Example 39. GTPyS Binding Assay
[00603] The ability of a compound to inhibit binding of GTP to LPAi is assessed via a membrane [35S]-GTPyS binding assay. CHO cells stably expressing the recombinant human LPAi receptor are resuspended in 10 mM Hepes, 7.4 containing 1 mM DTT, lysed and centrifuged at 75,000 xg to pellet the membranes. The membranes are resuspended in 10 mM Hepes, 7.4 containing 1 mM DTT and 10% glycerol. Membranes (~25 μg per well) are incubated in 96-well plates with 0.1 nM [35S]- GTPyS, 900 nM LPA, 5 μΜ GDP, and test compound in Assay Buffer (50 mM Hepes, pH 7.4, 100 mM NaCl, 10 mM MgCl2 , 50 μg/ml saponin and 0.2% fatty-acid free human serum albumin) for 30 minutes at 30°C. The reactions are terminated by rapid filtration through Whatman GF/B glass fibre filter plates. The filter plates are washed 3 times with 1 ml cold Wash Buffer (50 mM Hepes, 7.5, 100 mM NaCl and 10 mM MgCl2) and dried. Scintillant is then added to the plates and the radioactivity retained on the filters is determined on a Packard TopCount (Perkin Elmer). Specific binding is determined as total radioactive binding minus non-specific binding in the absence of the ligand (900 nM LPA). IC50s were determined using Graphpad prism analysis of drug titration curves.
[00604] Illustrative in vitro biological data is presented in the Table below.
Figure imgf000110_0001
Example 40. LPAI Chemotaxis Assay.
[00605] Chemotaxis of the A2058 human melanoma cells was measured using the Neuroprobe
ChemoTx® System plates (8 μηι pore size, 5.7 mm diameter sites). The filter sites were coated with 0.001 ) fibronectin (Sigma) in 20 mM Hepes, pH 7.4 and allowed to dry. A2058 cells were serum- starved for 24 hours , then harvested with Cell Stripper and resuspended in DMEM containing 0.1% fatty-acid- free bovine serum albumin (BSA) to a concentration of 1 x 106/ml. Cells were mixed with an equal volume of test compound (2X) in DMEM containing 0.1% fatty- acid- free BSA and incubated at 37°C for 15 minutes. LPA (100 nM in DMEM containing 0.1% fatty-acid-free BSA) or vehicle was added to each well of the lower chamber and 50 μΐ of the cell suspension/test compound mix was applied to the upper portion of the ChemoTx plate. Plates were incubated at 37°C for three hours and then the cells removed from the upper portion by rinsing with PBS and scraping. The filter was dried then stained with HEMA 3 Staining System (Fisher Scientific). The absorbance of the filter was read at 590 nM and IC50S were determined using Symyx Assay Explorer.
[00606] Compound 1 inhibited LPA-driven chemotaxis (IC50 less than 300 nM) of human A2058 melanoma cells.
[00607] As described for the A2058 melanoma cells, human MDA-MB-231 breast cancer cells chemotax in response to LPA1 signaling and this is inhibited by LPA1 antagonists.
Example 41 : Bleomycin-induced Lung fibrosis model in mice
[00608] Female CD-I mice (Harlan, 25-30g) are housed 4 per cage, given free access to food and water and allowed to acclimate for at least 7 days prior to test initiation. After the habituation phase, mice are lightly anesthetized with isoflurane (5% in 100% O2) and administered with bleomycin sulfate (0.01-5 U/kg, Henry Schein) via intratracheal instillation (Cuzzocrea S et al. Am J Physiol Lung Cell Mol Physiol. 2007 May;292(5):L1095-104. Epub 2007 Jan 12.). Mice are returned to their cages and monitored daily for the duration of the experiment. Test compound or vehicle is delivered po, ip or sc daily. The route and frequency of dosing is based on previously determined
pharmacokinetic properties. All animals are sacrificed using inhaled isoflurane 3, 7, 14, 21 or 28 days after bleomycin instillation. Following sacrifice, mice are intubated with a 20 gauge angiocatheter attached to a 1 ml syringe. Lungs are lavaged with saline to obtain bronchoalveolar lavage fluid (BALF) and then removed and fixed in 10% neutral buffered formalin for subsequent histopathological analysis. BALF is centrifuged for 10 min at 800 x g to pellet the cells and the cell supernatant removed and frozen at -80 °C for subsequent protein analysis using the DC protein assay kit (Biorad, Hercules, CA.) and soluble collagen analysis using Sircol (Biocolor Ltd, UK). BALF is analyzed for concentrations of inflammatory, pro-fibrotic and tissue injury biomarkers including transforming growth factor β 1 , hyaluronic acid, tissue inhibitor of metalloproteinase- 1 , matrix matelloproteinase-7, connective tissue growth factor and lactate dehydrogenase activity, using commercially available ELISA. The cell pellet is re-suspended in PBS. Total cell counts are then obtained using a Hemavet hematology system (Drew Scientific, Wayne, PA.) and differential cells counts are determined using Shandon cytospin (Thermo Scientific, Waltham, MA.). Lung tissue is stained using hematoxylin and eosin (H&E) and trichrome and lung fibrosis.is determined by semiquantitative histopathological scoring (Ashcroft T. et al. J. Clin. Path. 1988;41;4, 467-470.) using light microscopy (lOx magnification) and quantitative, computer-assisted densitometry of collagen in lung tissue sections using light microscopy. The data are plotted using Graphpad prism and statistical differences between groups determined.
[00609] In the acute setting (3 day), Compound 1-88 significantly reduced total protein, lactate dehydrogenase activity (LDH; tissue injury marker) and tissue inhibitor of metalloproteinase- 1 (TIMP-1; pro-fibrotic marker) concentrations in broncheoalveolar lavage fluid (BALF). In the chronic setting (14 and 28 day) model, Compound 1-88 maintained mouse body weight and decreased inflammatory cell influx and fibrosis after a single bleomycin (1.5 units/kg) instillation and decreased pulmonary resistance and lung fibrosis following repeated bleomycin (3.0-5.0 units/kg/week) instillations.
[00610] Compound 1 reduced total protein, lactate and TIMP-1 in the BALF in the acute setting (3- day). Compound 1 decreased inflammatory cell influx and fibrosis after a single bleomycin instillation (3.0 units) in the chronic setting (14-days only).
Example 42: Mouse carbon tetrachloride (CCUHnduced liver fibrosis model
[00611] Female C57BL/6 mice (Harlan, 20-25g) housed 4/cage are given free access to food and water and allowed to acclimate for at least 7 days prior to test initiation. After the habituation phase, mice receive CC14 (0.5-1.0 ml/kg body weight) diluted in corn oil vehicle (100 μΐ^ volume) via i.p. injection twice a week for 4-6 weeks. (Higazi, A. A. et ah, Clin Exp Immunol. 2008 Apr;152(l): 163- 73. Epub 2008 Feb 14.). Control mice receive an equivalent volume of corn oil vehicle only. Test compound or vehicle is delivered po, ip or sc daily. At the end of the study (4-6 weeks after first i.p. injection of CC14), mice are sacrificed using inhaled isoflurane and blood is drawn via cardiac puncture for subsequent analysis of ALT/AST levels. The liver is harvested, and one half of the liver is frozen at -80°C and the other half is fixed in 10% neutral buffered formalin for histological assessment of liver fibrosis using light microscopy (lOx magnification). Liver tissue homogenates are analyzed for collagen levels using Sircol (Biocolor Ltd, UK). Fixed Liver tissue is stained using hematoxylin and eosin (H&E) and trichrome and liver fibrosis is determined by quantitative, computer-assisted densitometry of collagen in liver tissue sections using light microscopy. Plasma and liver tissue lysates are also analyzed for concentrations of inflammatory, pro-fibrotic and tissue injury biomarkers including transforming growth factor βΐ, hyaluronic acid, tissue inhibitor of metalloproteinase- 1 , matrix matelloproteinase-7, connective tissue growth factor and lactate dehydrogenase activity, using commercially available ELISA. The resulting data are plotted using Graphpad prism and statistical differences between groups determined.
[00612] In this experiment, Compound 1 significantly reduced collagen deposition in the liver as compared to untreated control group.
Example 43: Mouse intravenous LPA-induced histamine release
[00613] A mouse intravenous LPA-induced histamine release model is utilized to determine the in vivo potency of LPAi and LPA3 receptor antagonists. Female CD-I mice (weighing 25 - 35 grams) are administered compound (i.p., s.c. or p.o.) in a volume of lOml/kg 30 minutes to 24 hours prior to intravenous LPA challenge (300 μg/mouse in 0.1% FAF BSA). Immediately following LPA challenge mice are placed into an enclosed Plexiglas chamber and exposed to an isoflurane for a period of 2 minutes. They are removed, decapitated and trunk blood collected into tubes containing EDTA. Blood is then centrifuged at 10,000 X g for 10 minutes at 4°C. Histamine concentrations in the plasma are determined by EIA. Drug concentrations in plasma are determined by mass spectrometry. The dose to achieve 50% inhibition of blood histamine release is calculated by nonlinear regression (Graphpad Prism) and plotted as the ED50. The plasma concentration associated with this dose is plotted as the EC50.
Example 44: Mouse unilateral ureteral obstruction kidney fibrosis model
[00614] Female C57BL/6 mice (Harlan, 20-25g) housed 4/cage will be given free access to food and water and allowed to acclimate for at least 7 days prior to test initiation. After the habituation phase,, mice undergo unilateral ureteral obstruction (UUO) surgery or sham to left kidney. Briefly, a longitudinal, upper left incision is performed to expose the left kidney. The renal artery is located and 6/0 silk thread is passed between the artery and the ureter. The thread is looped around the ureter and knotted 3 times insuring full ligation of ureter. The kidney is returned to abdomen, the abdominal muscle is sutured and the skin is stapled closed. Mice are returned to their cages and monitored daily for the duration of the experiment. Test compound or vehicle is delivered po, ip or sc daily. The route and frequency of dosing is based on previously determined pharmacokinetic properties. All animals are sacrificed using inhaled isoflurane 4, 8 or 14 days after UUO surgery. Following sacrifice blood is drawn via cardiac puncture, the kidneys are harvested and one half of the kidney is frozen at -80°C and the other half is fixed in 10% neutral buffered formalin for histological assessment of kidney fibrosis using light microscopy (lOx magnification). Kidney tissue homogenates are analyzed for collagen levels using Sircol (Biocolor Ltd, UK). Fixed kidney tissue is also stained using hematoxylin and eosin (H&E) and trichrome and kidney fibrosis is determined by quantitative, computer- assisted densitometry of collagen in liver tissue sections using light microscopy. Plasma and kidney tissue lysates are also analyzed for concentrations of inflammatory, pro-fibrotic and tissue injury biomarkers including transforming growth factor βΐ , hyaluronic acid, tissue inhibitor of metalloproteinase- 1 , matrix matelloproteinase-7, connective tissue growth factor and lactate dehydrogenase activity, using commercially available ELISA. The resulting data are plotted using Graphpad prism and statistical differences between groups determined.
[00615] In this experiment, Compound 1 reduced kidney fibrosis by 55% as compared to untreated group.
Example 45: Mouse Model of Bleomycin-Induced Scleroderma
[00616] A mouse model of bleomycin-induced scleroderma was used to evaluate the effect of compounds A and B in skin fibrosis. Methods were adapted from (Yamamoto, T et al. The Journal of Investigative Dermatology, 1 12: 456-462, 1999). Female C57B1/6 mice were anesthetized with isoflurane (3.0-3.5%) in 100%) O2) and two areas shaved bilaterally on the lower dorsolateral region. BLM (1 - 10 μg in 100 μΐ) prepared in sterile filtered PBS was administered subcutaneously to each shaved region once daily for 5 to 7 days per week for a total of 4 weeks (28 days).
[00617] Compound 1 was prepared in water vehicle and delivered orally twice a day on weekdays and once daily on weekends.
I l l [00618] On day 28 all animals were sacrificed. The dorsolateral skin removed, trimmed of adherent subcutaneous fat and an 8 mm biopsy punch was used to collect two skin samples from each ,subject. One sample submerged in 10% neutral buffered formalin and submitted for histological analysis. The second sample was frozen at -80°C for further processing of collagen content using either Sircol or hydroxyproline methods. The amount of the collagen-specific amino acid hydroxyproline was determined as a measure of collagen present in lesional skin.
[00619] FIGURE 8 and 9: Results of assay on mouse model of bleomycin-induced scleroderma using Compound 1. FIGURE 8 shows dermal thickness. FIGURE 9 shows collagen content.
*P<0.05 versus vehicle group, t-test. Compound 1 reduced both the dermal thickness and collagen content.
Example 46: Mouse Spinal Cord Contusion Injury Model
[00620] Preliminary data suggest that LPA/LPA1 could be an important trigger of inflammation in the central nervous system and contribute to functional, locomotor deficits after spinal cord injury. Therefore, the spinal cord contusion injury model was used to assess the effects of LPAl antagonism on functional recovery after spinal cord trauma. Adult (8- to 10-week-old) female BALB/c mice (Charles River) were anesthetized with ketamine/xylazine/acepromazine (50:5: 1 mg/kg). After performing a laminectomy at the 11th thoracic vertebrae, the exposed spinal cord was contused using the Infinite Horizons Impactor device (Precision Scientific). Moderate injuries were made using a force of 50 kdyn and tissue displacement ranging between 400 and 600μηι as described previously (Ghasemlou et ah, (2005) Exp Neurol 196:9 -17.). Starting at 1, 6 or 24 hr after spinal cord, mice were dosed twice daily (30 mg/kg; bid) with the LPAl antagonist by oral gavage. On day 28 after injury, locomotor recovery was evaluated in an open field test using the nine-point Basso Mouse Scale (BMS) (Basso et al., (2006) J Neurotrauma 23:635- 659), which was specifically developed for locomotor testing after contusion injuries in mice. The BMS analysis of hindlimb movements and coordination was performed by two independent assessors who were trained in the Basso laboratory at Ohio State University, and the consensus score was taken. The final score is presented as mean ± SEM. The 11 -point BMS subscores, which evaluate finer aspects of locomotor control, were also assessed. Histopathological outcome and inflammatory response (using FACS analysis) were also determined.
[00621] Compound 1 significantly (p<0.05) increased locomotor recovery starting on day 3 after spinal cord injury (SCI). Compared to vehicle control, Compound 1 also increased motor evoked potential along the spinal cord by approximately 65% after SCI.
Example 47: Mouse Peritoneal Fibrosis Model
[00622] The ability of the LPAl receptor to regulate peritoneal fibrosis and myofibroblast density was examined using wild-type and LPAl KO. Pharmacological studies were also conducted using collagen-GFP mice to assess the ability of an LPAl receptor antagonist to reduce peritoneal fibrosis, fibroblast density and profibrotic protein expression (connective tissue growth factor; CTGF). To induce peritoneal fibrosis, mice (6-10 weeks old) received intraperitoneal (i.p.) injections of PBS (vehicle) or chlorhexidine gluconate (CG; 0.1%, 10 ml/kg) every other day for a period of 21 days. For pharmacological studies, mice were dosed twice daily with water (vehicle) or an LPA1 antagonist (30 mg/kg; bid) by oral gavage. On day 21 , mice were sacrificed and peritoneal collagen, serosal thickness, fibroblast/myofibroblast density and CTGF expression were measured.
[00623] Compound 1 significantly (p<0.05) reduced peritoneal fibrosis by 55% and CTGF expression by 61%. Compound 1 also dramatically reduced fibroblast density in the peritoneal serosa.
Example 48: Clinical Trial in Humans with Idiopathic Pulmonary Fibrosis (IPF)
[00624] The purposes of this study is to assess the efficacy of treatment with Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) compared with placebo in patients with idiopathic pulmonary fibrosis (IPF) and to assess the safety of treatment with Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) compared with placebo in patients with IPF.
[00625] The primary outcome variable is the absolute change in percent predicted forced vital capacity (FVC) from baseline to Week 72.
[00626] Secondary outcome measures include: composite outcomes of important IPF-related events; progression- free survival; categorical assessment of absolute change in percent predicted FVC from baseline to Week 72; change in Shortness-of-Breath from baseline to Week 72; change in percent predicted hemoglobin (Hb)-corrected carbon monoxide diffusing capacity (DLco) of the lungs from baseline to Week 72; change in oxygen saturation during the 6 minute walk test (6MWT) from baseline to Week 72; change in high-resolution computed tomography (HRCT) assessment from baseline to Week 72; change in distance walked in the 6MWT from baseline to Week 72.
[00627] Patients eligible for this study include those patients that satisfy the following inclusion criteria: diagnosis of IPF; 40 to 80 years of age; FVC > 50% predicted value; DLco > 35%> predicted value; either FVC or DLco < 90%> predicted value; no improvement in past year; able to walk 150 meters in 6 minutes and maintain saturation > 83%) while on no more than 6 L/min supplemental oxygen.
[00628] Patients are excluded from this study if they satisfy any of the following criteria: unable to undergo pulmonary function testing; evidence of significant obstructive lung disease or airway hyper-responsiveness; in the clinical opinion of the investigator, the patient is expected to need and be eligible for a lung transplant within 72 weeks of randomization; active infection; liver disease; cancer or other medical condition likely to result in death within 2 years; diabetes; pregnancy or lactation; substance abuse; personal or family history of long QT syndrome; other IPF treatment; unable to take study medication; withdrawal from other IPF trials. [00629] Patients are orally dosed with either placebo or an amount of Compound 1 or a pharmaceutically acceptable salt thereof (e.g. Compound 2) (1 mg/day - 1000 mg/day). The primary outcome variable will be the absolute change in percent predicted FVC from Baseline to Week 72. Patients will receive blinded study treatment from the time of randomization until the last patient randomized has been treated for 72 weeks. A Data Monitoring Committee (DMC) will periodically review safety and efficacy data to ensure patient safety.
[00630] After week 72, patients who meet the Progression of Disease (POD) definition, which is a > 10% absolute decrease in percent predicted FVC or a > 15% absolute decrease in percent predicted DLco, will be eligible to receive permitted IPF therapies in addition to their blinded study drug. Permitted IPF therapies include corticosteroids, azathioprine, cyclophosphamide and N-acetyl- cysteine.
[00631] The examples and embodiments described herein are illustrative and various modifications or changes suggested to persons skilled in the art are to be included within this disclosure. As will be appreciated by those skilled in the art, the specific components listed in the above examples may be replaced with other functionally equivalent components, e.g., diluents, binders, lubricants, fillers, and the like.

Claims

What is claimed is:
1. A pharmaceutically acceptable salt of (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1), wherein the pharmaceutically acceptable salt is a sodium salt, calcium salt, potassium salt, ammonium salt, L-arginine salt, L-lysine salt, or N-methyl-D-glucamine salt.
2. The pharmaceutically acceptable salt of claim 1 , wherein the pharmaceutically acceptable salt is a sodium salt.
3. The pharmaceutically acceptable salt of claim 1 or claim 2, wherein the pharmaceutically acceptable salt is amorphous.
4. The pharmaceutically acceptable salt of claim 1 or claim 2, wherein the pharmaceutically acceptable salt is crystalline.
5. The pharmaceutically acceptable salt of any one of claims 1-4, wherein the pharmaceutically acceptable salt is substantially free of the S-isomer.
6. A crystalline form of {4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid sodium salt (Compound 2):
Figure imgf000117_0001
(Compound 2).
7. The crystalline form of claim 6, wherein the crystalline form of Compound 2 is characterized as having:
an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 5.3° 2-Theta, 13.5° 2-Theta, 15.8° 2-Theta, 17.1° 2-Theta, 18.3° 2-Theta, 21.1° 2-Theta, and 21.7° 2-Theta; an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 2; a differential scanning calorimetry (DSC) thermogram with an endotherm at about 176°C; a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 7; a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 7;
or combinations thereof.
8. The crystalline form of claim 6, wherein the crystalline form of Compound 2 is characterized as having:
has an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 8.5° 2-Theta, 9.3° 2-Theta, 16.6° 2-Theta, 17.1° 2-Theta and 21.4° 2-Theta; has an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure
3;
or combinations thereof.
9. The crystalline form of any one of claims 6-8, wherein the crystalline form of Compound 2 was obtained from isopropanol.
10. The crystalline form of any one of claims 6-9, wherein the crystalline form of Compound 2 is substantially free of the S-isomer.
11. A crystalline form of (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid Compound 1):
Figure imgf000118_0001
(Compound 1).
12. The crystalline form of claim 11, wherein the crystalline form of Compound 1 is
characterized as having:
an X-ray powder diffraction (XRPD) pattern with characteristic peaks at 12.6° 2-Theta, 17.3° 2-Theta, 21.7° 2-Theta, and 23.0° 2-Theta;
an X-ray powder diffraction (XRPD) pattern substantially the same as shown in Figure 1 ; a differential scanning calorimetry (DSC) thermogram with endotherms at about 157°C and about 198°C;
a differential scanning calorimetry (DSC) thermogram substantially the same as Figure 4; has a thermogravimetric analysis (TGA) thermogram substantially the same as Figure 4; substantially the same X-ray powder diffraction (XRPD) pattern post storage at 40 °C/75% RH for one week;
or combinations thereof.
13. The crystalline form of claim 11 or claim 12, wherein the crystalline form of Compound 1 is crystallized from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, nitromethane, tetrahydrofuran, dioxane, methyl ethyl ketone, acetone, 2-methoxyethanol, or methanol.
14. The crystalline form of any one of claims 11-13, wherein the crystalline form of Compound 1 is substantially free of the S-isomer.
15. A pharmaceutical composition comprising {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 14 and at least one inactive ingredient selected from pharmaceutically acceptable carriers, diluents and excipients.
16. The pharmaceutical composition of claim 15, wherein the pharmaceutical composition comprises Compound 2.
17. The pharmaceutical composition of claim 16, wherein the pharmaceutical composition comprises crystalline Compound 2.
18. The pharmaceutical composition according to any one of claims 15-17, wherein the
pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, inhalation, nasal administration, topical administration, ophthalmic administration or otic administration.
19. The pharmaceutical composition according to any one of claims 15-17, wherein the
pharmaceutical composition is a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a suspension, a gel, a colloid, a dispersion, a suspension, a solution, an emulsion, an ointment, a lotion, an eye drop or an ear drop.
20. The pharmaceutical composition according to any one of claims 15-17, wherein the
pharmaceutical composition is in a form suitable for oral administration to a mammal.
21. The pharmaceutical composition according to claim 20, wherein the pharmaceutical
composition is in the form of a pill, capsule, tablet, aqueous solution, aqueous suspension, non-aqueous solution, or non-aqueous suspension.
22. A method for treating or preventing a LPA-dependent or LPA-mediated disease or condition in a mammal comprising administering {4'-[3-methyl-4-((R)-l-phenyl- ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl}-acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14 to the mammal in need thereof.
23. A method for treating or preventing lung fibrosis, asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis, skin fibrosis, fibrosis of the gut, peritoneal fibrosis, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease,
atherosclerosis, Raynaud's phenomenom, neuropathic pain, or spinal cord injury in a mammal comprising administering (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14 to the mammal in need thereof.
A method for the treatment or prevention of fibrosis in a mammal comprising administering (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14 to the mammal in need thereof.
25. The method of claim 73, wherein the fibrosis comprises lung fibrosis, renal fibrosis, hepatic fibrosis, cutaneous fibrosis, or peritoneal fibrosis.
26. A method of treating idopathic pulmonary fibrosis in a mammal comprising administering (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14 to the mammal in need thereof.
27. A method for the treatment or prevention of scleroderma in a mammal comprising
administering (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5-yl]- biphenyl-4-yl} -acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14 to the mammal in need thereof.
28. A method for the treatment or prevention of Raynaud's phenomenom in a mammal
comprising administering (4'-[3-methyl-4-((R)- 1 -phenyl-ethoxycarbonylamino)-isoxazol-5- yl]-biphenyl-4-yl} -acetic acid (Compound 1), or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14 to the mammal in need thereof.
29. A process for preparing crystalline (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1) comprising isolating Compound 1 from toluene, anisole, ethyl acetate, butyl acetate, butanol, iso-propanol, propanol, ethanol, acetonitrile, or nitromethane.
30. A process for preparing crystalline (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid, sodium salt (Compound 2) comprising isolating Compound 2 from isopropanol.
31. A process for the preparation of (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1) comprising the steps of:
(1) reacting a compound of Formula IV:
Figure imgf000120_0001
IV
wherein, X is a leaving group;
a compound of Formula V:
Figure imgf000121_0001
V
wherein,
R is Ci-C6 alkyl; and B is a boronic acid or boronate ester;
in the presence of a coupling catalyst, a suitable base, and in a suitable solvent, to provide compound of Formula VI:
Figure imgf000121_0002
and
(2) hydrolysis of the ester moiety of the compound of Formula VI to provide Compound 1 A process for the preparation of (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1) comprising the steps of:
(1) reacting a compound of Formula VII:
Figure imgf000121_0003
VII
wherein,
B is a boronic acid or boronate ester;
with a compound of Formula VIII:
Figure imgf000121_0004
VIII wherein,
R is C1-C6 alkyl; and X is a leaving group;
in the presence of a coupling catalyst, a suitable base, and in a suitable solvent, to provide a compound of Formula VI:
Figure imgf000122_0001
and
(2) hydrolysis of the ester moiety of the compound of Formula VI to provide Compound 1.
33. The process according to claim 31 or claim 32, wherein:
the coupling catalyst is a palladium catalyst;
the suitable base is triethylamine, diisopropylethylamine, 1,2,2,6,6-pentamethylpiperidine, tributylamine, sodium bicarbonate, Na2C03, K2C03, Cs2C03, NaOAc, KOAc, Na3P04 or K3P04;
the suitable solvent is tetrahydrofuran, dioxane, water, or combinations thereof.
34. The process according to claim 33, wherein:
R is -CH3 or -CH2CH3;
X is selected from CI, Br, I, -OS02CF3, -OS02(4-methylphenyl), -OS02(phenyl) and -
Figure imgf000122_0002
A process for the preparation of (4'-[3-methyl-4-((R)-l-phenyl-ethoxycarbonylamino)- isoxazol-5-yl]-biphenyl-4-yl} -acetic acid (Compound 1) comprising the steps of:
(1) treatment of a compound of Formula XII with diphenylphosphoryl azide in the presence of (R)-(+)- 1 -phenylethanol:
Figure imgf000123_0001
wherein,
R is Ci-Ce alkyl;
to provide a compound of Formula VI:
Figure imgf000123_0002
and
(2) hydrolysis of the ester moiety of the compound of Formula VI to provide Compound 1 The process according to claim 31, claim 32 or claim 35, wherein step (2) comprises treatment of the compound of Formula VI with sodium hydroxide in a suitable solvent followed by a pH adjustment.
PCT/US2011/039872 2010-06-14 2011-06-09 Lysophosphatidic acid receptor antagonist and uses thereof WO2011159550A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US35462210P 2010-06-14 2010-06-14
US61/354,622 2010-06-14

Publications (2)

Publication Number Publication Date
WO2011159550A2 true WO2011159550A2 (en) 2011-12-22
WO2011159550A3 WO2011159550A3 (en) 2012-04-05

Family

ID=45348798

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/039872 WO2011159550A2 (en) 2010-06-14 2011-06-09 Lysophosphatidic acid receptor antagonist and uses thereof

Country Status (1)

Country Link
WO (1) WO2011159550A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012078593A3 (en) * 2010-12-07 2012-08-02 Amira Pharmaceuticals, Inc. Lysophosphatidic acid receptor antagonists and their use in the treatment fibrosis
WO2013189864A1 (en) * 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag N-alkyltriazole compounds as lpar antagonists
WO2013189865A1 (en) * 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag N-aryltriazole compounds as lpar antagonists
WO2014058729A1 (en) * 2012-10-09 2014-04-17 California Institute Of Technology In vivo and in vitro carbene insertion and nitrene transfer reactions catalyzed by heme enzymes
US8975235B2 (en) 2011-03-20 2015-03-10 Intermune, Inc. Lysophosphatidic acid receptor antagonists
US8993262B2 (en) 2012-10-09 2015-03-31 California Institute Of Technology In vivo and in vitro olefin cyclopropanation catalyzed by heme enzymes
US9399762B2 (en) 2014-02-18 2016-07-26 California Institute Of Technology Methods and systems for sulfimidation or sulfoximidation of organic molecules
WO2016197009A1 (en) 2015-06-05 2016-12-08 Vertex Pharmaceuticals Incorporated Triazoles for the treatment of demyelinating diseases
WO2018106641A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Pyrazoles for the treatment of demyelinating diseases
WO2018106643A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Heterocyclic azoles for the treatment of demyelinating diseases
WO2018106646A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Aminotriazoles for the treatment of demyelinating diseases
CN113069460A (en) * 2021-03-24 2021-07-06 兰州大学 Application of halcinonide and derivatives thereof in preparation of drugs for treating and/or preventing cerebrovascular diseases
US11548871B2 (en) 2019-11-15 2023-01-10 Gilead Sciences, Inc. Triazole carbamate pyridyl sulfonamides as LPA receptor antagonists and uses thereof
US11584738B2 (en) 2020-06-03 2023-02-21 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof
US11702407B2 (en) 2020-06-03 2023-07-18 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof
US11939318B2 (en) 2021-12-08 2024-03-26 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040204383A1 (en) * 2003-04-11 2004-10-14 Gabor Tigyi Lysophosphatidic acid analogs and inhibition of neointima formation
US20060009507A1 (en) * 2003-10-09 2006-01-12 Miller Duane D LPA receptor agonists and antagonists and methods of use
WO2008014286A1 (en) * 2006-07-24 2008-01-31 University Of Virginia Patent Foundation Vinyl phosphonate lysophosphatidic acid receptor antagonists

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040204383A1 (en) * 2003-04-11 2004-10-14 Gabor Tigyi Lysophosphatidic acid analogs and inhibition of neointima formation
US20060009507A1 (en) * 2003-10-09 2006-01-12 Miller Duane D LPA receptor agonists and antagonists and methods of use
WO2008014286A1 (en) * 2006-07-24 2008-01-31 University Of Virginia Patent Foundation Vinyl phosphonate lysophosphatidic acid receptor antagonists

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9272990B2 (en) 2010-12-07 2016-03-01 Amira Pharmaceuticals, Inc. Lysophosphatidic acid receptor antagonists and their use in the treatment fibrosis
WO2012078593A3 (en) * 2010-12-07 2012-08-02 Amira Pharmaceuticals, Inc. Lysophosphatidic acid receptor antagonists and their use in the treatment fibrosis
US8975235B2 (en) 2011-03-20 2015-03-10 Intermune, Inc. Lysophosphatidic acid receptor antagonists
WO2013189864A1 (en) * 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag N-alkyltriazole compounds as lpar antagonists
WO2013189865A1 (en) * 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag N-aryltriazole compounds as lpar antagonists
CN104395299A (en) * 2012-06-20 2015-03-04 霍夫曼-拉罗奇有限公司 N-aryltriazole compounds as lpar antagonists
CN104411692A (en) * 2012-06-20 2015-03-11 霍夫曼-拉罗奇有限公司 N-alkyltriazole compounds as lpar antagonists
US9321738B2 (en) 2012-06-20 2016-04-26 Hoffman-La Roche Inc. N-alkyltriazole compounds as LPAR antagonists
JP2015520203A (en) * 2012-06-20 2015-07-16 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft N-aryltriazole compounds as LPAR antagonists
US9493799B2 (en) 2012-10-09 2016-11-15 California Institute Of Technology In vivo and in vitro olefin cyclopropanation catalyzed by heme enzymes
US10208322B2 (en) 2012-10-09 2019-02-19 California Institute Of Technology In vivo and in vitro olefin cyclopropanation catalyzed by heme enzymes
WO2014058729A1 (en) * 2012-10-09 2014-04-17 California Institute Of Technology In vivo and in vitro carbene insertion and nitrene transfer reactions catalyzed by heme enzymes
US8993262B2 (en) 2012-10-09 2015-03-31 California Institute Of Technology In vivo and in vitro olefin cyclopropanation catalyzed by heme enzymes
US11008596B2 (en) 2012-10-09 2021-05-18 California Institute Of Technology Cytochrome P450 BM3 enzyme variants for preparation of cyclopropanes
US9399762B2 (en) 2014-02-18 2016-07-26 California Institute Of Technology Methods and systems for sulfimidation or sulfoximidation of organic molecules
US10501762B2 (en) 2014-02-18 2019-12-10 California Institute Of Technology Methods and systems for sulfimidation or sulfoximidation of organic molecules
WO2016197009A1 (en) 2015-06-05 2016-12-08 Vertex Pharmaceuticals Incorporated Triazoles for the treatment of demyelinating diseases
WO2018106641A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Pyrazoles for the treatment of demyelinating diseases
WO2018106646A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Aminotriazoles for the treatment of demyelinating diseases
WO2018106643A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Heterocyclic azoles for the treatment of demyelinating diseases
US11548871B2 (en) 2019-11-15 2023-01-10 Gilead Sciences, Inc. Triazole carbamate pyridyl sulfonamides as LPA receptor antagonists and uses thereof
US11584738B2 (en) 2020-06-03 2023-02-21 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof
US11702407B2 (en) 2020-06-03 2023-07-18 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof
US11912686B2 (en) 2020-06-03 2024-02-27 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof
CN113069460A (en) * 2021-03-24 2021-07-06 兰州大学 Application of halcinonide and derivatives thereof in preparation of drugs for treating and/or preventing cerebrovascular diseases
US11939318B2 (en) 2021-12-08 2024-03-26 Gilead Sciences, Inc. LPA receptor antagonists and uses thereof

Also Published As

Publication number Publication date
WO2011159550A3 (en) 2012-04-05

Similar Documents

Publication Publication Date Title
AU2016238848B2 (en) Polycyclic LPA1 antagonist and uses thereof
WO2011159550A2 (en) Lysophosphatidic acid receptor antagonist and uses thereof
US9624182B2 (en) Compounds as lysophosphatidic acid receptor antagonists
EP2648714B1 (en) Lysophosphatidic acid receptor antagonists and uses thereof
EP2462128B1 (en) Compounds as lysophosphatidic acid receptor antagonists
US8541587B2 (en) Lysophosphatidic acid receptor antagonists
US8455499B2 (en) Alkyne antagonists of lysophosphatidic acid receptors
US20130072490A1 (en) Polycyclic compounds as lysophosphatidic acid receptor antagonists
WO2010141768A2 (en) Polycyclic antagonists of lysophosphatidic acid receptors

Legal Events

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

Ref document number: 11796208

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11796208

Country of ref document: EP

Kind code of ref document: A2