US20110288107A1 - Topical formulation for a jak inhibitor - Google Patents

Topical formulation for a jak inhibitor Download PDF

Info

Publication number
US20110288107A1
US20110288107A1 US13/112,370 US201113112370A US2011288107A1 US 20110288107 A1 US20110288107 A1 US 20110288107A1 US 201113112370 A US201113112370 A US 201113112370A US 2011288107 A1 US2011288107 A1 US 2011288107A1
Authority
US
United States
Prior art keywords
formulation
weight
component
pharmaceutical formulation
independently selected
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/112,370
Inventor
Bhavnish Parikh
Bhavesh Shah
Krishnaswamy Yeleswaram
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Incyte Holdings Corp
Original Assignee
Incyte Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=44201091&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110288107(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US13/112,370 priority Critical patent/US20110288107A1/en
Application filed by Incyte Corp filed Critical Incyte Corp
Assigned to INCYTE CORPORATION reassignment INCYTE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PARIKH, BHAVNISH, YELESWARAM, KRISHNASWAMY, SHAH, BHAVESH
Publication of US20110288107A1 publication Critical patent/US20110288107A1/en
Priority to US14/714,820 priority patent/US20150250790A1/en
Assigned to INCYTE HOLDINGS CORPORATION AND INCYTE CORPORATION reassignment INCYTE HOLDINGS CORPORATION AND INCYTE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INCYTE CORPORATION
Assigned to INCYTE CORPORATION, INCYTE HOLDINGS CORPORATION reassignment INCYTE CORPORATION CORRECTIVE ASSIGNMENT TO CORRECT THE OMMISSION OF SECOND RECEIVING PARTY NAME PREVIOUSLY RECORDED AT REEL: 035292 FRAME: 0004. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: INCYTE CORPORATION
Priority to US16/566,625 priority patent/US10758543B2/en
Priority to US16/947,735 priority patent/US10869870B2/en
Priority to US16/948,408 priority patent/US11219624B2/en
Priority to US17/541,439 priority patent/US20220211707A1/en
Priority to US17/704,155 priority patent/US20220370455A1/en
Priority to US17/704,168 priority patent/US11571425B2/en
Priority to US17/704,180 priority patent/US11590136B2/en
Priority to US18/089,651 priority patent/US20230277541A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • 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
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to pharmaceutical formulations for topical skin application comprising (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, and use in the treatment of skin disorders.
  • Protein kinases regulate diverse biological processes including cell growth, survival, differentiation, organ formation, morphogenesis, neovascularization, tissue repair, and regeneration, among others. Protein kinases also play specialized roles in a host of human diseases including cancer. Cytokines, low-molecular weight polypeptides or glycoproteins, regulate many pathways involved in the host inflammatory response to sepsis. Cytokines influence cell differentiation, proliferation and activation, and can modulate both pro-inflammatory and anti-inflammatory responses to allow the host to react appropriately to pathogens.
  • JAKs Janus kinase family
  • JAK2 Janus kinase-1
  • JAK2 JAK2
  • JAK3 also known as Janus kinase, leukocyte
  • JAKL protein-tyrosine kinase 2
  • TYK2 protein-tyrosine kinase 2
  • Cytokine-stimulated immune and inflammatory responses contribute to pathogenesis of diseases: pathologies such as severe combined immunodeficiency (SCID) arise from suppression of the immune system, while a hyperactive or inappropriate immune/inflammatory response contributes to the pathology of autoimmune diseases (e.g., asthma, systemic lupus erythematosus, thyroiditis, myocarditis), and illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T. Cheng, et al. (2000) Arthritis Res 2(1): 16-32).
  • SCID severe combined immunodeficiency
  • Jak1 ⁇ / ⁇ mice are runted at birth, fail to nurse, and die perinatally (Rodig, S. J., M. A. Meraz, et al. (1998) Cell 93(3): 373-83). Jak2 ⁇ / ⁇ mouse embryos are anemic and die around day 12.5 postcoitum due to the absence of definitive erythropoiesis.
  • the JAK/STAT pathway and in particular all four JAKs, are believed to play a role in the pathogenesis of asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract.
  • Multiple cytokines that signal through JAKs have been linked to inflammatory diseases/conditions of the upper respiratory tract, such as those affecting the nose and sinuses (e.g., rhinitis and sinusitis) whether classically allergic reactions or not.
  • the JAK/STAT pathway has also been implicated in inflammatory diseases/conditions of the eye and chronic allergic responses.
  • Activation of JAK/STAT in cancers may occur by cytokine stimulation (e.g. IL-6 or GM-CSF) or by a reduction in the endogenous suppressors of JAK signaling such as SOCS (suppressor or cytokine signaling) or PIAS (protein inhibitor of activated STAT) (Boudny, V., and Kovarik, J., Neoplasm. 49:349-355, 2002).
  • Activation of STAT signaling, as well as other pathways downstream of JAKs e.g., Akt
  • Elevated levels of circulating cytokines that signal through JAK/STAT play a causal role in cachexia and/or chronic fatigue. As such, JAK inhibition may be beneficial to cancer patients for reasons that extend beyond potential anti-tumor activity.
  • Inhibition of the JAK kinases is also envisioned to have therapeutic benefits in patients suffering from skin immune disorders such as psoriasis, and skin sensitization.
  • skin immune disorders such as psoriasis, and skin sensitization.
  • psoriasis vulgaris the most common form of psoriasis, it has been generally accepted that activated T lymphocytes are important for the maintenance of the disease and its associated psoriatic plaques (Gott Kunststoff, A. B., et al, Nat Rev Drug Disc., 4:19-34).
  • Psoriatic plaques contain a significant immune infiltrate, including leukocytes and monocytes, as well as multiple epidermal layers with increased keratinocyte proliferation.
  • JAK inhibitors Given the usefulness of JAK inhibitors in the treatment of skin disorders, there is a need for improved topical formulations of JAK inhibitors. In particular, there is a need for stable, easily applied formulations for JAK inhibitors with good skin permeation characteristics.
  • the formulations of the invention, as well the methods described herein are directed toward this need and other ends.
  • a potent JAK1/JAK2 inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, and its pharmaceutically acceptable salts, has previously been described in U.S. Pat. No. 7,598,257, U.S. Patent Publ. No. 2009/0181959, and U.S. Patent Publ. No. 2008/0312259, each of which is incorporated herein by reference in its entirety.
  • the present invention describes an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile suitable for topical administration and treatment of skin disorders.
  • a pharmaceutical formulation for topical skin application comprising:
  • a therapeutically effective amount of a therapeutic agent which is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides a method of treating a skin disorder, comprising applying a pharmaceutical formulation described herein to an area of skin of the patient.
  • the present invention also provides a pharmaceutical formulation described herein for use in treatment of a skin disorder in a patient in need thereof.
  • the present invention also provides use of a pharmaceutical formulation described herein for the preparation of a medicament for use in treatment of a skin disorder in a patient in need thereof.
  • FIG. 1 depicts a flowchart describing the manufacturing process for an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt.
  • FIG. 2 depicts the change in lesion score for subjects with chronic plaque psoriasis treated with 0.5%, 1.0%, and 1.5% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis) as compared to treatment with placebo over a 12-week period (the dashed line is baseline).
  • FIG. 3 shows photographs of subjects with chronic plaque psoriasis before ( FIG. 3( a )) and after 84 days ( FIG. 3( b )) of treatment with 1.0% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 4 shows photographs of subjects with chronic plaque psoriasis before ( FIG. 4( a )) and after 84 days ( FIG. 4( b )) of treatment with 1.0% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 5 shows photographs of subjects with chronic plaque psoriasis before ( FIG. 5( a )) and after 84 days ( FIG. 5( b )) of treatment with 1.5% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 6 shows photographs of subjects with chronic plaque psoriasis before ( FIG. 6( a )) and after 84 days ( FIG. 6( b )) of treatment with 0.5% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 7 shows photographs of subjects with chronic plaque psoriasis before ( FIG. 7( a )) and after 84 days ( FIG. 7( b )) of treatment with 1.0% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • the present invention provides, inter alia, a pharmaceutical formulation for topical skin application, comprising a therapeutically effective amount of
  • the pharmaceutical formulation comprises:
  • the emulsion comprises water, an oil component, and an emulsifier component.
  • the term “emulsifier component” refers, in one aspect, to a substance, or mixtures of substances that maintains an element or particle in suspension within a fluid medium.
  • the emulsifier component allows an oil phase to form an emulsion when combined with water.
  • the emulsifier component refers to one or more non-ionic surfactants.
  • the oil-in-water formulations were found to have better appearance, spreadability and stability as compared with other formulations.
  • the formulations have a thick, creamy appearance which allows for good spreadability of the formulation on skin. This good spreadability leads to better skin permeation than comparable anhydrous formulations.
  • the oil-in-water formulations showed higher cumulative amounts in studies of transport across human cadaver skin over 24 hours when compared with an anhydrous ointment. While not wishing to be bound by any particular theory, the higher cumulative amounts are believed to be due to better spreadability of the oil-in-water formulation as compared to the anhydrous ointment, resulting in increased surface area for transport.
  • a higher viscosity for the oil-in-water formulations also appeared to be preferred with respect to skin permeation as higher viscosity cream formulations had better transport across human cadaver skin as compared with oil-in-water lotions of lower viscosity.
  • oil-in-water formulations described herein were found to have good stability over a three-month period when stored at 25° C./60% RH and 40° C./75% RH in aluminum tubes and maintain reasonable viscosity over time.
  • water-in-oil formulations displayed syneresis when stored at 40° C. (syneresis means separation of liquid from the emulsion).
  • the water-in-oil formulation was also less desirable than the formulations of the invention, because the API dissolved in the base over time, leading to highly variable skin permeation in in vitro studies as well as a lack of an increase in permeability with increasing strength of the formulation.
  • formulations described herein are relatively simple to manufacture with a repeatable process of formulation.
  • the resultant product is easily packaged.
  • the formulations appear to have good stability and relatively consistent permeation profiles.
  • the oil component is present in an amount of about 10% to about 40% by weight of the formulation.
  • the oil component is present in an amount of about 17% to about 27% by weight of the formulation.
  • the oil component is present in an amount of about 20% to about 27% by weight of the formulation.
  • the oil component comprises one or more substances independently selected from petrolatums, fatty alcohols, mineral oils, triglycerides, and silicone oils.
  • the oil component comprises one or more substances independently selected from white petrolatum, cetyl alcohol, stearyl alcohol, light mineral oil, medium chain triglycerides, and dimethicone.
  • the oil component comprises an occlusive agent component.
  • the occlusive agent component is present in an amount of about 2% to about 15% by weight of the formulation.
  • the occlusive agent component is present in an amount of about 5% to about 10% by weight of the formulation.
  • occlusive agent component refers to a hydrophobic agent or mixtures of hydrophobic agents that form an occlusive film on skin that reduces transepidermal water loss (TEWL) by preventing evaporation of water from the stratum corneum.
  • TEWL transepidermal water loss
  • the occlusive agent component comprises one or more substances selected from fatty acids (e.g., lanolin acid), fatty alcohols (e.g., lanolin alcohol), hydrocarbon oils & waxes (e.g., petrolatum), polyhydric alcohols (e.g., propylene glycol), silicones (e.g., dimethicone), sterols (e.g., cholesterol).
  • fatty acids e.g., lanolin acid
  • fatty alcohols e.g., lanolin alcohol
  • hydrocarbon oils & waxes e.g., petrolatum
  • polyhydric alcohols e.g., propylene glycol
  • silicones e.g., dimethicone
  • sterols e.g., cholesterol
  • vegetable or animal fat e.g., cocoa butter
  • vegetable wax e.g., Carnauba wax
  • wax ester e.g., bees wax
  • the occlusive agent component comprises one or more substances selected from lanolin acid fatty alcohols, lanolin alcohol, petrolatum, propylene glycol, dimethicone, cholesterol, cocoa butter, Carnauba wax, and bees wax.
  • the occlusive agent component comprises petrolatum.
  • the occlusive agent component comprises white petrolatum.
  • the oil component comprises a stiffening agent component.
  • the stiffening agent component is present in an amount of about 2% to about 8% by weight of the formulation.
  • the stiffening agent component is present in an amount of about 3% to about 6% by weight of the formulation.
  • the stiffening agent component is present in an amount of about 4% to about 7% by weight of the formulation.
  • the term “stiffening agent component” refers to a substance or mixture of substances that increases the viscosity and/or consistency of the formulation or improves the rheology of the formulation.
  • the stiffening agent component comprises one or more substances independently selected from fatty alcohols.
  • the stiffening agent component comprises one or more substances independently selected from C 12-20 fatty alcohols.
  • the stiffening agent component comprises one or more substances independently selected from C 16-18 fatty alcohols.
  • the stiffening agent component comprises one or more substances independently selected from cetyl alcohol and stearyl alcohol.
  • the oil component comprises an emollient component.
  • the emollient component is present in an amount of about 5% to about 15% by weight of the formulation.
  • the emollient component is present in an amount of about 7% to about 13% by weight of the formulation.
  • the term “emollient component” refers to an agent that softens or soothes the skin or soothes an irritated internal surface.
  • the emollient component comprises one or more substances independently selected from mineral oils and triglycerides.
  • the emollient component comprises one or more substances independently selected from light mineral oil and medium chain triglycerides.
  • the emollient component comprises one or more substances independently selected from light mineral oil, medium chain triglycerides, and dimethicone.
  • the water is present in an amount of about 35% to about 65% by weight of the formulation.
  • the water is present in an amount of about 40% to about 60% by weight of the formulation.
  • the water is present in an amount of about 45% to about 55% by weight of the formulation.
  • the emulsifier component is present in an amount of about 1% to about 9% by weight of the formulation.
  • the emulsifier component is present in an amount of about 2% to about 6% by weight of the formulation.
  • the emulsifier component is present in an amount of about 3% to about 5% by weight of the formulation.
  • the emulsifier component is present in an amount of about 4% to about 7% by weight of the formulation.
  • the pharmaceutical formulation comprises an emulsifier component and a stiffening agent component, wherein the combined amount of emulsifier component and stiffening agent component is at least about 8% by weight of the formulation.
  • the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters.
  • the emulsifier component comprises one or more substances independently selected from glyceryl stearate, and polysorbate 20.
  • the pharmaceutical formulation further comprises a stabilizing agent component.
  • the stabilizing agent component is present in an amount of about 0.05% to about 5% by weight of the formulation.
  • the stabilizing agent component is present in an amount of about 0.1% to about 2% by weight of the formulation.
  • the stabilizing agent component is present in an amount of about 0.3 to about 0.5% by weight of the formulation.
  • the term “stabilizing agent component” refers to a substance or mixture of substances that improves the stability of the pharmaceutical formulation and/or the compatibility of the components in the formulation. In some embodiments, the stabilizing agent component prevents agglomeration of the emulsion and stabilizes the droplets in the oil-in-water emulsion.
  • the stabilizing agent component comprises one or more substances independently selected from polysaccharides.
  • the stabilizing agent component comprises xanthan gum.
  • the pharmaceutical formulation further comprises a solvent component.
  • the solvent component is present in an amount of about 10% to about 35% by weight of the formulation.
  • the solvent component is present in an amount of about 15% to about 30% by weight of the formulation.
  • the solvent component is present in an amount of about 20% to about 25% by weight of the formulation.
  • solvent component is a liquid substance or mixture of liquid substances capable of dissolving (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile or other substances in the formulation.
  • the solvent component is a liquid substance or mixture of liquid substances in which (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, or its pharmaceutically acceptable salt, has reasonable solubility.
  • solubilities of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile (free base) or its phosphate salt are reported in Table 21.
  • a solvent is a substance or mixture thereof, in which (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, or its pharmaceutically acceptable salt (whichever is used), has a solubility of at least about 10 mg/mL or greater, at least about 15 mg/mL or greater, or at least about 20 mg/mL or greater, when measured as described in Example 4.
  • the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols.
  • the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol.
  • the therapeutic agent is present in an amount of about 0.5% to about 1.5% by weight of the formulation on a free base basis.
  • the therapeutic agent is present in an amount of about 0.5% by weight of the formulation on a free base basis.
  • the therapeutic agent is present in an amount of about 1% by weight of the formulation on a free base basis.
  • the therapeutic agent is present in an amount of about 1.5% by weight of the formulation on a free base basis.
  • the therapeutic agent is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile phosphate.
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the combined amount of the stiffening agent component and the emulsifier component is at least about 8% by weight of the formulation.
  • the occlusive agent component comprises a petrolatum
  • the stiffening agent component comprises one or more substances independently selected from one or more fatty alcohols
  • the emollient component comprises one or more substances independently selected from mineral oils and triglycerides;
  • the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters;
  • the stabilizing agent component comprises one or more substances independently
  • the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols.
  • the occlusive agent component comprises white petrolatum
  • the stiffening agent component comprises one or more substances independently selected from cetyl alcohol and stearyl alcohol;
  • the emollient component comprises one or more substances independently selected from light mineral oil, medium chain triglycerides, and dimethicone;
  • the emulsifier component comprises one or more substances independently selected from glyceryl stearate and polysorbate 20;
  • the stabilizing agent component comprises xanthan gum
  • the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol.
  • the pharmaceutical formulation further comprises an antimicrobial preservative component.
  • the antimicrobial preservative component is present in an amount of about 0.05% to about 3% by weight of the formulation.
  • the antimicrobial preservative component is present in an amount of about 0.1% to about 1% by weight of the formulation.
  • antimicrobial preservative component is a substance or mixtures of substances which inhibits microbial growth in the formulation.
  • the antimicrobial preservative component comprises one or more substances independently selected from alkyl parabens and phenoxyethanol.
  • the antimicrobial preservative component comprises one or more substances independently selected from methyl paraben, propyl paraben, and phenoxyethanol.
  • the pharmaceutical formulation further comprises a chelating agent component.
  • chelating agent component refers to a compound or mixtures of compounds that has the ability to bind strongly with metal ions.
  • the chelating agent component comprises edetate disodium.
  • (R)-3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile can be prepared as described in U.S. Pat. No. 7,598,257 and U.S. Patent Publ. No. 2009/0181959, each of which is incorporated herein by reference in its entirety.
  • the 1:1 phosphate salt of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile can be prepared as described in U.S. Patent Publ. No. 2008/0312259, which is incorporated herein by reference in its entirety.
  • the compounds of the present invention also include pharmaceutically acceptable salts of the compounds disclosed herein.
  • pharmaceutically acceptable salt refers to a salt formed by the addition of a pharmaceutically acceptable acid or base to a compound disclosed herein.
  • pharmaceutically acceptable refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient.
  • Pharmaceutically acceptable salts include, but are not limited to, those derived from organic and inorganic acids such as, but not limited to, acetic, lactic, citric, cinnamic, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, oxalic, propionic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, glycolic, pyruvic, methanesulfonic, ethanesulfonic, toluenesulfonic, salicylic, benzoic, and similarly known acceptable acids.
  • organic and inorganic acids such as, but not limited to, acetic, lactic, citric, cinnamic, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, oxalic, propionic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, glycolic, pyruvic, methanes
  • % by weight of the formulation on a free base basis of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, or pharmaceutically acceptable salt thereof” means that the % w/w is calculated based on the weight of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile in the total formulation.
  • “0.5% w/w on a free base basis” of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate means that for 100 grams of total formulation, there are 0.66 grams of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate in the formulation (which equates to 0.5 grams of the free base, (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile).
  • the components are present in exactly the ranges specified (e.g., the term “about” is not present). In some embodiments, “about” means plus or minus 10% of the value.
  • each component of the formulation comprises a different substance or mixture of substances.
  • component can mean one substance or a mixture of substances.
  • fatty acid refers to an aliphatic acid that is saturated or unsaturated. In some embodiments, the fatty acid is in a mixture of different fatty acids. In some embodiments, the fatty acid has between about eight to about thirty carbons on average. In some embodiments, the fatty acid has about 12 to 20, 14-20, or 16-18 carbons on average.
  • Suitable fatty acids include, but are not limited to, cetyl acid, stearic acid, lauric acid, myristic acid, erucic acid, palmitic acid, palmitoleic acid, capric acid, caprylic acid, oleic acid, linoleic acid, linolenic acid, hydroxystearic acid, 12-hydroxystearic acid, cetostearic acid, isostearic acid, sesquioleic acid, sesqui-9-octadecanoic acid, sesquiisooctadecanoic acid, behenic acid, isobehenic acid, and arachidonic acid, or mixtures thereof.
  • fatty alcohol refers to an aliphatic alcohol that is saturated or unsaturated. In some embodiments, the fatty alcohol is in a mixture of different fatty alcohols. In some embodiments, the fatty alcohol has between about 12 to about 20, about 14 to about 20, or about 16 to about 18 carbons on average. Suitable fatty alcohols include, but are not limited to, stearyl alcohol, lauryl alcohol, palmityl alcohol, cetyl alcohol, capryl alcohol, caprylyl alcohol, oleyl alcohol, linolenyl alcohol, arachidonic alcohol, behenyl alcohol, isobehenyl alcohol, selachyl alcohol, chimyl alcohol, and linoleyl alcohol, or mixtures thereof.
  • polyalkylene glycol employed alone or in combination with other terms, refers to a polymer containing oxyalkylene monomer units, or copolymer of different oxyalkylene monomer units, wherein the alkylene group has 2 to 6, 2 to 4, or 2 to 3 carbon atoms.
  • oxyalkylene employed alone or in combination with other terms, refers to a group of formula —O-alkylene-.
  • the polyalkylene glycol is polyethylene glycol.
  • sorbitan fatty ester includes products derived from sorbitan or sorbitol and fatty acids and, optionally, poly(ethylene glycol) units, including sorbitan esters and polyethoxylated sorbitan esters.
  • the sorbitan fatty ester is a polyethoxylated sorbitan ester.
  • sorbitan ester refers to a compound, or mixture of compounds, derived from the esterification of sorbitol and at least one fatty acid.
  • Fatty acids useful for deriving the sorbitan esters include, but are not limited to, those described herein.
  • Suitable sorbitan esters include, but are not limited to, the SpanTM series (available from Uniqema), which includes Span 20 (sorbitan monolaurate), 40 (sorbitan monopalmitate), 60 (sorbitan monostearate), 65 (sorbitan tristearate), 80 (sorbitan monooleate), and 85 (sorbitan trioleate).
  • Other suitable sorbitan esters include those listed in R. C. Rowe and P. J. Shesky, Handbook of pharmaceutical excipients, (2006), 5th ed., which is incorporated herein by reference in its entirety.
  • polyethoxylated sorbitan ester refers to a compound, or mixture thereof, derived from the ethoxylation of a sorbitan ester.
  • the polyoxethylene portion of the compound can be between the fatty ester and the sorbitan moiety.
  • sorbitan ester refers to a compound, or mixture of compounds, derived from the esterification of sorbitol and at least one fatty acid.
  • Fatty acids useful for deriving the polyethoyxlated sorbitan esters include, but are not limited to, those described herein.
  • the polyoxyethylene portion of the compound or mixture has about 2 to about 200 oxyethylene units.
  • the polyoxyethylene portion of the compound or mixture has about 2 to about 100 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 4 to about 80 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 4 to about 40 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 4 to about 20 oxyethylene units.
  • Suitable polyethoxylated sorbitan esters include, but are not limited to the TweenTM series (available from Uniqema), which includes Tween 20 (POE(20) sorbitan monolaurate), 21 (POE(4) sorbitan monolaurate), 40 (POE(20) sorbitan monopalmitate), 60 (POE(20) sorbitan monostearate), 60K (POE(20) sorbitan monostearate), 61 (POE(4) sorbitan monostearate), 65 (POE(20) sorbitan tristearate), 80 (POE(20) sorbitan monooleate), 80K (POE(20) sorbitan monooleate), 81 (POE(5) sorbitan monooleate), and 85 (POE(20) sorbitan trioleate).
  • TweenTM series available from Uniqema
  • Tween 20 POE(20) sorbitan monolaurate
  • 21 POE(4)
  • POE polyoxyethylene
  • the number following the POE abbreviation refers to the number of oxyethylene repeat units in the compound.
  • Other suitable polyethoxylated sorbitan esters include the polyoxyethylene sorbitan fatty acid esters listed in R. C. Rowe and P. J. Shesky, Handbook of pharmaceutical excipients, (2006), 5th ed., which is incorporated herein by reference in its entirety.
  • the polyethoxylated sorbitan ester is a polysorbate.
  • the polyethoxylated sorbitan ester is polysorbate 20.
  • the term “glyceryl fatty esters” refers to mono-, di- or triglycerides of fatty acids.
  • the glyceryl fatty esters may be optionally substituted with sulfonic acid groups, or pharmaceutically acceptable salts thereof.
  • Suitable fatty acids for deriving glycerides of fatty acids include, but are not limited to, those described herein.
  • the glyceryl fatty ester is a mono-glyceride of a fatty acid having 12 to 18 carbon atoms.
  • the glyceryl fatty ester is glyceryl stearate.
  • triglycerides refers to a triglyceride of a fatty acid. In some embodiments, the triglyceride is medium chain triglycerides.
  • alkylene glycol refers to a group of formula —O-alkylene-, wherein the alkylene group has 2 to 6, 2 to 4, or 2 to 3 carbon atoms.
  • the alkylene glycol is propylene glycol (1,2-propanediol).
  • polyethylene glycol refers to a polymer containing ethylene glycol monomer units of formula —O—CH 2 —CH 2 —.
  • Suitable polyethylene glycols may have a free hydroxyl group at each end of the polymer molecule, or may have one or more hydroxyl groups etherified with a lower alkyl, e.g., a methyl group.
  • derivatives of polyethylene glycols having esterifiable carboxy groups are also suitable.
  • Polyethylene glycols useful in the present invention can be polymers of any chain length or molecular weight, and can include branching In some embodiments, the average molecular weight of the polyethylene glycol is from about 200 to about 9000.
  • the average molecular weight of the polyethylene glycol is from about 200 to about 5000. In some embodiments, the average molecular weight of the polyethylene glycol is from about 200 to about 900. In some embodiments, the average molecular weight of the polyethylene glycol is about 400.
  • Suitable polyethylene glycols include, but are not limited to polyethylene glycol-200, polyethylene glycol-300, polyethylene glycol-400, polyethylene glycol-600, and polyethylene glycol-900. The number following the dash in the name refers to the average molecular weight of the polymer.
  • the skin disorder is an autoimmune bullous skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
  • the skin disorder is psoriasis (for example, psoriasis vulgaris), atopic dermatitis, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis).
  • psoriasis for example, psoriasis vulgaris
  • atopic dermatitis for example, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis).
  • certain substances including some pharmaceuticals when topically applied can cause skin sensitization.
  • co-administration or sequential administration of the topical formulations of the invention together with the agent causing unwanted sensitization can be helpful in treating such unwanted sensitization or dermatitis.
  • the present invention further provides a method of treating dermatological side effects of other pharmaceuticals by administration of the compound of the invention.
  • numerous pharmaceutical agents result in unwanted allergic reactions which can manifest as acneiform rash or related dermatitis.
  • Example pharmaceutical agents that have such undesirable side effects include anti-cancer drugs such as gefitinib, cetuximab, erlotinib, and the like.
  • the formulations of the invention can be administered systemically or topically (e.g., localized to the vicinity of the dermatitis) in combination with (e.g., simultaneously or sequentially) the pharmaceutical agent having the undesirable dermatological side effect.
  • the formulation of the invention can be administered topically together with one or more other pharmaceuticals, where the other pharmaceuticals when topically applied in the absence of a formulation of the invention cause contact dermatitis, allergic contact sensitization, or similar skin disorder.
  • formulation of the invention include topical formulations further comprising an additional pharmaceutical agent which can cause dermatitis, skin disorders, or related side effects.
  • the term “individual” or “patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the phrase “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician.
  • the term “treating” or “treatment” refers to one or more of (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
  • preventing the disease for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of
  • One or more additional pharmaceutical agents such as, for example, chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors such as, for example, those described in WO 2006/056399, or other agents can be used in combination with the formulations of the present invention for treatment of JAK-associated diseases, disorders or conditions.
  • the one or more additional pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • Example chemotherapeutic include proteosome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • proteosome inhibitors e.g., bortezomib
  • thalidomide thalidomide
  • revlimid thalidomide
  • DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • Example steroids include corticosteroids such as dexamethasone or prednisone.
  • Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521,184, WO 04/005281, and U.S. Ser. No. 60/578,491.
  • Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120.
  • Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444.
  • Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402.
  • the formulations of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • a corticosteroid such as dexamethasone is administered to a patient in combination with the compound of the invention where the dexamethasone is administered intermittently as opposed to continuously.
  • Another aspect of the present invention relates to formulations comprising a labeled active compound (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating JAK in tissue samples, including human, and for identifying JAK ligands by inhibition binding of a labeled compound. Accordingly, the present invention includes JAK assays that contain such labeled compounds.
  • a labeled active compound radio-labeled, fluorescent-labeled, etc.
  • the present invention further includes formulations of an isotopically-labeled compound.
  • An “isotopically” or “radio-labeled” compound is a compound where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2 H (also written as D for deuterium), 3 H (also written as T for tritium), 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 18 F, 35 S, 36 Cl, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 124 I and 131 I.
  • the radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro JAK labeling and competition assays, compounds that incorporate 3 H 14 C, 82 Br, 125 I, 131 I, 35 S or will generally be most useful.
  • radio-labeled or “labeled compound” is a compound that has incorporated at least one radionuclide.
  • the radionuclide is selected from the group consisting of 3 H, 14 C, 125 I, 35 S and 82 Br.
  • Kits The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of JAK-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical formulation of the invention.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • the present invention provides pharmaceutical formulations comprising the components specified in the example formulations (e.g., Example 3), wherein the components are present in about the amounts in Tables 2-5.
  • the aqueous layer was back-extracted with three portions of ethyl acetate.
  • the combined organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated.
  • the crude product was purified by silica gel chromatography (gradient of ethyl acetate/hexanes) to yield a viscous clear syrup, which was dissolved in ethanol and evaporated several times to remove ethyl acetate, to afford 19.4 g of racemic adduct (93%).
  • the enantiomers were separated by preparative-HPLC, (OD-H, 15% ethanol/hexanes) and used separately in the next step to generate their corresponding final product.
  • the phosphoric acid salt was shown to be a 1:1 salt by 1 H NMR and crystallinity was confirmed by X-ray powder diffraction (XRPD). Differential scanning calorimetry (DSC) gave a sharp melting peak at about 198.66° C. The product showed little weight loss up to 200° C. by TGA.
  • An oil-in-water cream formulation was prepared for (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (Example 2) at 0.5, 1.0 and 1.5% by weight of the formulation (free base equivalent).
  • the compositions for a 15 gram tube are provided in Table 2 below.
  • the formulation for three strengths were identical except for adjustments to the purified water quantity based on the amount of active ingredient. All excipients used in the formulation were compendial grade (ie, USP/NF or BP) or are approved for use in topical products.
  • FIG. 1 shows a flowchart representation of the process for making the oil-in-water formulation.
  • the (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile is referred to as “API” throughout this application.
  • a paraben phase was prepared by mixing methyl and propyl parabens with a portion of the propylene glycol (see % in Tables 2-5).
  • a xanthan gum phase was prepared by mixing xanthan gum with propylene glycol (see % in Table 2-5).
  • An oil phase was then prepared by mixing light mineral oil, glyceryl stearate, polysorbate 20, white petrolatum, cetyl alcohol, stearyl alcohol, dimethicone and medium chain triglycerides. The phase is heated to 70-80° C. to melt and form a uniform mixture.
  • the aqueous phase was next prepared by mixing purified water, polyethylene glycol, and disodium EDTA. The phase is heated to 70-80° C.
  • step 7 The oil phase from step 3 was then combined under high shear mixing with the mixture from step 6 to form an emulsion.
  • Phenoxyethanol was then added to the emulsion from step 7. Mixing was continued, and then the product was cooled under low shear mixing.
  • More consistent batches at larger scales could be obtained by adding Example 2 gradually to the aqueous phase and then combining with the other phases.
  • more consistent batches could be obtained by slower cooling (e.g., by using room temperature water in the outer jacket of the reactor, rather than lower temperature water.
  • the appearance of the cream was visually inspected. Viscosity was measured using a Brookfield viscometer at 25° C. The pH was measured on the final cream formulation. The microbial limit testing is performed as per USP. The fill weight is analyzed as an in-process test during filling of the cream into tubes.
  • Results are shown below for a 3.5 kg batches at 0.5%, 1% and 1.5% strength of Example 2 (free base basis (API)) (Table 6).
  • the stability data from batches of the cream formulation at 0.5, 1.0 and 1.5% w/w strength stored in 15 gram aluminum tubes is provided in Tables 7-10 and 19-20. Further, stability data from batches of the cream formulation at 0.5, 1.0 and 1.5% w/w strength packaged in amber glass jars (2 oz. with teflon cap) is provided in Tables 13-17, while longer stability data for the 1.0% w/w formulation packaged in 16 oz. amber glass jars is provided in Tables 11-12.
  • the preliminary stability data for the drug product did not show any chemical instability after 3 months of storage at 25° C./60% RH and 40° C./75% RH in either packaging configuration. A change in viscosity is seen following 3 months at 40° C./75% RH for formulation stored in amber glass jars. However, physical inspection of the product did not indicate any phase separation.
  • Aeruginosa Absent/1 g Absent/1 g NA Absent/1 g MLT ( S. Aureus ) Absent/1 g Absent/1 g NA Absent/1 g MLT (Total Aerobic) ⁇ 10 ⁇ 10 NA ⁇ 10 MLT (Total Yeast ⁇ 10 ⁇ 10 NA ⁇ 10 and Molds)
  • Aeruginosa Absent/1 g Absent/1 g NA Absent/1 g MLT ( S. Aureus ) Absent/1 g Absent/1 g NA Absent/1 g MLT (Total Aerobic) ⁇ 10 ⁇ 10 NA ⁇ 10 MLT (Total Yeast and ⁇ 10 ⁇ 10 NA ⁇ 10 Molds)
  • the lower viscosity was believed to be due to electrolytic nature of the phosphate salt.
  • Viscosities of the formulations and placebo over time are shown in Table 23.
  • the 1% dispersed cream (water-in-oil formulation) showed syneresis after two and four weeks of aging at 40° C., while the 1% lotion and 1% solubilized cream formulations (oil-in-water formulations) did not show syneresis.
  • the 1% solubilized cream formulation was generally higher in viscosity than the 1% lotion.
  • Example 5 The three different topical formulations in Example 5 (Table 20) and the cream formulation in Example 3 (Table 4) were evaluated for transport across human cadaver skin.
  • the skin permeation data are summarized in Table 24.
  • Significant variability was observed in the transport among the three replicates for each formulation.
  • the variability in transport may be due in part to differences in skin samples (donor, region of the body, thickness, etc.).
  • the two cream formulations showed higher flux compared to the lotion or ointment.
  • the cumulative amount of API transported for the ointment formulation was particularly low in comparison to the other three formulations and this, at least in part, could be due to poor spreadability of the ointment leading to decreased surface area for transport.
  • the two cream formulations were selected for further development, one as an oil-in-water (see Example 3 above) and the other as a water-in-oil emulsion base.
  • strengths containing 1.0, 1.5, and 2.0% w/w of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate salt were developed for the oil-in water base cream (solubilized cream) and 1.0, 2.0, and 3.0% w/w were developed for the water-in oil base cream (dispersed cream). Procedures for the skin permeation studies are described below.
  • the permeability of the API in topical formulations was studied using cadaver human skin samples and Franz diffusion cells. Dermatomed human cadaver skin was obtained from tissue banks while the Franz diffusion cells were custom made. The human cadaver skin samples, sized to fit between the donor and the receiver compartments, were positioned on the Franz diffusion cells. Topical formulations were weighed (20 mg) onto glassine paper, placed formulation side toward the skin and clamped into place. The dosing chamber was covered with parafilm. The reservoir side was filled using saline with 4% albumin. The reservoir was stirred and maintained at 37° C. using a dry block heater (Aungst B. Fatty Acid Skin Penetration Enhancers. Pharm. Res. 1989; 6(3):244-247).
  • the permeability of the API in topical formulations was studied using freshly excised mouse skin samples mounted in Franz diffusion cells. Balb/c mice were depilated using a waxing technique four days before the experiment. The morning of the experiment the mice were euthanized and as much of the depilated skin as possible was removed, rinsed and kept moist with 37° C. saline until use. The mouse skin samples, sized to fit between the donor and the receiver compartments, were positioned between the donor and the receiver compartments of the Franz diffusion cells. The opening of the Franz cell was 1 cm 2 . Topical formulations were weighed (20 mg) on to glassine paper, placed formulation side toward the skin and clamped into place. The dosing chamber was covered with parafilm.
  • the reservoir side was filled using saline with 4% albumin
  • the reservoir was stirred and maintained at 37° C. using a dry block heater (Aungst 1989 (above). At 4 hours, a 1 mL sample was removed and replaced with 1 mL of saline +4% albumin. At 24 hours, the entire reservoir was collected. The tissue was examined visually for any hole or tear. The reservoir side samples were analyzed for concentrations of the API by a LC/MS assay.
  • photos were obtained from subjects who signed an informed consent for the photos. Pictures were obtained at baseline (prior to the first application of study treatment) and on day 84 (the last application day for study treatment) (see FIG. 3-7 ). These photos are representative of a subset of the subjects who were treated with the oil-in-water formulations.
  • the formulations described herein can also be tested for their efficacies (of inhibiting JAK targets) in the T-cell driven murine delayed hypersensitivity test model.
  • the murine skin contact delayed-type hypersensitivity (DTH) response is considered to be a valid model of clinical contact dermatitis, and other T-lymphocyte mediated immune disorders of the skin, such as psoriasis ( Immunol Today. 1998 January; 19(1):37-44).
  • Murine DTH shares multiple characteristics with psoriasis, including the immune infiltrate, the accompanying increase in inflammatory cytokines, and keratinocyte hyperproliferation.
  • many classes of agents that are efficacious in treating psoriasis in the clinic are also effective inhibitors of the DTH response in mice (Agents Actions. 1993 January; 38(1-2):116-21).
  • mice On Day 0 and 1, Balb/c mice are sensitized with a topical application, to their shaved abdomen with the antigen 2,4,dinitro-fluorobenzene (DNFB). On day 5, ears are measured for thickness using an engineer's micrometer. This measurement is recorded and used as a baseline. Both of the animals' ears are then challenged by a topical application of DNFB in a total of 20 ⁇ L (10 ⁇ L on the internal pinna and 10 ⁇ L on the external pinna) at a concentration of 0.2%. Twenty-four to seventy-two hours after the challenge, ears are measured again.
  • DNFB 2,4,dinitro-fluorobenzene
  • Treatment with the test formulations is given throughout the sensitization and challenge phases (day ⁇ 1 to day 7) or prior to and throughout the challenge phase (usually afternoon of day 4 to day 7). Treatment of the test compounds (in different concentration) is administered topically (topical application of the treatment to the ears). Efficacies of the test formulations are indicated by a reduction in ear swelling comparing to the situation without the treatment. Compounds causing a reduction of 20% or more are considered efficacious. In some experiments, the mice are challenged but not sensitized (negative control).
  • the inhibitive effect (inhibiting activation of the JAK-STAT pathways) of the test formulations can be confirmed by immunohistochemical analysis.
  • Activation of the JAK-STAT pathway(s) results in the formation and translocation of functional transcription factors.
  • the influx of immune cells and the increased proliferation of keratinocytes should also provide unique expression profile changes in the ear that can be investigated and quantified.
  • Formalin fixed and paraffin embedded ear sections (harvested after the challenge phase in the DTH model) are subjected to immunohistochemical analysis using an antibody that specifically interacts with phosphorylated STAT3 (clone 58E12, Cell Signaling Technologies).
  • test formulations a clinically efficacious treatment for psoriasis
  • dexamethasone a clinically efficacious treatment for psoriasis
  • Test formulations and the dexamethasone can produce similar transcriptional changes both qualitatively and quantitatively, and both the test formulations and dexamethasone can reduce the number of infiltrating cells.
  • Topical administration of the test compounds can produce inhibitive effects, i.e., reduction in the number of infiltrating cells and inhibition of the transcriptional changes.

Abstract

This invention relates to pharmaceutical formulations for topical skin application comprising (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, and use in the treatment of skin disorders.

Description

  • This application claims the benefit of priority of U.S. Provisional Application 61/347,132, filed May 21, 2010, which is incorporated herein by reference in its entirety.
  • TECHNICAL FIELD
  • This invention relates to pharmaceutical formulations for topical skin application comprising (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, and use in the treatment of skin disorders.
  • BACKGROUND
  • Protein kinases (PKs) regulate diverse biological processes including cell growth, survival, differentiation, organ formation, morphogenesis, neovascularization, tissue repair, and regeneration, among others. Protein kinases also play specialized roles in a host of human diseases including cancer. Cytokines, low-molecular weight polypeptides or glycoproteins, regulate many pathways involved in the host inflammatory response to sepsis. Cytokines influence cell differentiation, proliferation and activation, and can modulate both pro-inflammatory and anti-inflammatory responses to allow the host to react appropriately to pathogens. Signaling of a wide range of cytokines involves the Janus kinase family (JAKs) of protein tyrosine kinases and Signal Transducers and Activators of Transcription (STATs). There are four known mammalian JAKs: JAK1 (Janus kinase-1), JAK2, JAK3 (also known as Janus kinase, leukocyte; JAKL; and L-JAK), and TYK2 (protein-tyrosine kinase 2).
  • Cytokine-stimulated immune and inflammatory responses contribute to pathogenesis of diseases: pathologies such as severe combined immunodeficiency (SCID) arise from suppression of the immune system, while a hyperactive or inappropriate immune/inflammatory response contributes to the pathology of autoimmune diseases (e.g., asthma, systemic lupus erythematosus, thyroiditis, myocarditis), and illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T. Cheng, et al. (2000) Arthritis Res 2(1): 16-32).
  • Deficiencies in expression of JAKs are associated with many disease states. For example, Jak1−/− mice are runted at birth, fail to nurse, and die perinatally (Rodig, S. J., M. A. Meraz, et al. (1998) Cell 93(3): 373-83). Jak2−/− mouse embryos are anemic and die around day 12.5 postcoitum due to the absence of definitive erythropoiesis.
  • The JAK/STAT pathway, and in particular all four JAKs, are believed to play a role in the pathogenesis of asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract. Multiple cytokines that signal through JAKs have been linked to inflammatory diseases/conditions of the upper respiratory tract, such as those affecting the nose and sinuses (e.g., rhinitis and sinusitis) whether classically allergic reactions or not. The JAK/STAT pathway has also been implicated in inflammatory diseases/conditions of the eye and chronic allergic responses.
  • Activation of JAK/STAT in cancers may occur by cytokine stimulation (e.g. IL-6 or GM-CSF) or by a reduction in the endogenous suppressors of JAK signaling such as SOCS (suppressor or cytokine signaling) or PIAS (protein inhibitor of activated STAT) (Boudny, V., and Kovarik, J., Neoplasm. 49:349-355, 2002). Activation of STAT signaling, as well as other pathways downstream of JAKs (e.g., Akt), has been correlated with poor prognosis in many cancer types (Bowman, T., et al. Oncogene 19:2474-2488, 2000). Elevated levels of circulating cytokines that signal through JAK/STAT play a causal role in cachexia and/or chronic fatigue. As such, JAK inhibition may be beneficial to cancer patients for reasons that extend beyond potential anti-tumor activity.
  • Inhibition of the JAK kinases is also envisioned to have therapeutic benefits in patients suffering from skin immune disorders such as psoriasis, and skin sensitization. In psoriasis vulgaris, the most common form of psoriasis, it has been generally accepted that activated T lymphocytes are important for the maintenance of the disease and its associated psoriatic plaques (Gottlieb, A. B., et al, Nat Rev Drug Disc., 4:19-34). Psoriatic plaques contain a significant immune infiltrate, including leukocytes and monocytes, as well as multiple epidermal layers with increased keratinocyte proliferation. While the initial activation of immune cells in psoriasis occurs by an ill defined mechanism, the maintenance is believed to be dependent on a number of inflammatory cytokines, in addition to various chemokines and growth factors (JCI, 113:1664-1675). Many of these, including interleukins -2,-4,-6,-7,-12,-15,-18, and -23 as well as GM-CSF and IFNg, signal through the Janus (JAK) kinases (Adv Pharmacol. 2000;47:113-74). As such, blocking signal transduction at the level of JAK kinases may result in therapeutic benefits in patients suffering from psoriasis or other immune disorders of the skin.
  • Given the usefulness of JAK inhibitors in the treatment of skin disorders, there is a need for improved topical formulations of JAK inhibitors. In particular, there is a need for stable, easily applied formulations for JAK inhibitors with good skin permeation characteristics. The formulations of the invention, as well the methods described herein are directed toward this need and other ends.
  • SUMMARY
  • A potent JAK1/JAK2 inhibitor, (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, and its pharmaceutically acceptable salts, has previously been described in U.S. Pat. No. 7,598,257, U.S. Patent Publ. No. 2009/0181959, and U.S. Patent Publ. No. 2008/0312259, each of which is incorporated herein by reference in its entirety. The present invention describes an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile suitable for topical administration and treatment of skin disorders.
  • Figure US20110288107A1-20111124-C00001
  • Accordingly, the present invention provides, inter alia, a pharmaceutical formulation for topical skin application, comprising:
  • an oil-in-water emulsion; and
  • a therapeutically effective amount of a therapeutic agent which is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof.
  • The present invention also provides a method of treating a skin disorder, comprising applying a pharmaceutical formulation described herein to an area of skin of the patient.
  • The present invention also provides a pharmaceutical formulation described herein for use in treatment of a skin disorder in a patient in need thereof.
  • The present invention also provides use of a pharmaceutical formulation described herein for the preparation of a medicament for use in treatment of a skin disorder in a patient in need thereof.
  • The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
  • DESCRIPTION OF DRAWINGS
  • FIG. 1 depicts a flowchart describing the manufacturing process for an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt.
  • FIG. 2 depicts the change in lesion score for subjects with chronic plaque psoriasis treated with 0.5%, 1.0%, and 1.5% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis) as compared to treatment with placebo over a 12-week period (the dashed line is baseline).
  • FIG. 3 shows photographs of subjects with chronic plaque psoriasis before (FIG. 3( a)) and after 84 days (FIG. 3( b)) of treatment with 1.0% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 4 shows photographs of subjects with chronic plaque psoriasis before (FIG. 4( a)) and after 84 days (FIG. 4( b)) of treatment with 1.0% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 5 shows photographs of subjects with chronic plaque psoriasis before (FIG. 5( a)) and after 84 days (FIG. 5( b)) of treatment with 1.5% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 6 shows photographs of subjects with chronic plaque psoriasis before (FIG. 6( a)) and after 84 days (FIG. 6( b)) of treatment with 0.5% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • FIG. 7 shows photographs of subjects with chronic plaque psoriasis before (FIG. 7( a)) and after 84 days (FIG. 7( b)) of treatment with 1.0% w/w of an oil-in-water formulation of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (on a free base basis).
  • DETAILED DESCRIPTION
  • Accordingly, the present invention provides, inter alia, a pharmaceutical formulation for topical skin application, comprising a therapeutically effective amount of
  • (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-y0-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the pharmaceutical formulation comprises:
      • an oil-in-water emulsion; and
      • a therapeutically effective amount of a therapeutic agent which is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the emulsion comprises water, an oil component, and an emulsifier component.
  • As used herein, the term “emulsifier component” refers, in one aspect, to a substance, or mixtures of substances that maintains an element or particle in suspension within a fluid medium. In some embodiments, the emulsifier component allows an oil phase to form an emulsion when combined with water. In some embodiments, the emulsifier component refers to one or more non-ionic surfactants.
  • The oil-in-water formulations were found to have better appearance, spreadability and stability as compared with other formulations. The formulations have a thick, creamy appearance which allows for good spreadability of the formulation on skin. This good spreadability leads to better skin permeation than comparable anhydrous formulations. For example, the oil-in-water formulations showed higher cumulative amounts in studies of transport across human cadaver skin over 24 hours when compared with an anhydrous ointment. While not wishing to be bound by any particular theory, the higher cumulative amounts are believed to be due to better spreadability of the oil-in-water formulation as compared to the anhydrous ointment, resulting in increased surface area for transport. A higher viscosity for the oil-in-water formulations also appeared to be preferred with respect to skin permeation as higher viscosity cream formulations had better transport across human cadaver skin as compared with oil-in-water lotions of lower viscosity.
  • The oil-in-water formulations described herein were found to have good stability over a three-month period when stored at 25° C./60% RH and 40° C./75% RH in aluminum tubes and maintain reasonable viscosity over time. By comparison, the water-in-oil formulations displayed syneresis when stored at 40° C. (syneresis means separation of liquid from the emulsion).
  • The water-in-oil formulation was also less desirable than the formulations of the invention, because the API dissolved in the base over time, leading to highly variable skin permeation in in vitro studies as well as a lack of an increase in permeability with increasing strength of the formulation.
  • In transport studies with freshly excised mouse skin, the oil-in-water formulations also displayed a general trend of increased permeability when the strength of the solubilized cream was increased from 0.5% w/w to 1.5% w/w, while such a trend was not seen with the water-in-oil formulations. Thus, it appears that the water-in-oil emulsions will not have any advantage in terms of providing enhanced permeation with increasing strengths.
  • Further, the formulations described herein are relatively simple to manufacture with a repeatable process of formulation. The resultant product is easily packaged. The formulations appear to have good stability and relatively consistent permeation profiles.
  • In some embodiments, the oil component is present in an amount of about 10% to about 40% by weight of the formulation.
  • In some embodiments, the oil component is present in an amount of about 17% to about 27% by weight of the formulation.
  • In some embodiments, the oil component is present in an amount of about 20% to about 27% by weight of the formulation.
  • In some embodiments, the oil component comprises one or more substances independently selected from petrolatums, fatty alcohols, mineral oils, triglycerides, and silicone oils.
  • In some embodiments, the oil component comprises one or more substances independently selected from white petrolatum, cetyl alcohol, stearyl alcohol, light mineral oil, medium chain triglycerides, and dimethicone.
  • In some embodiments, the oil component comprises an occlusive agent component.
  • In some embodiments, the occlusive agent component is present in an amount of about 2% to about 15% by weight of the formulation.
  • In some embodiments, the occlusive agent component is present in an amount of about 5% to about 10% by weight of the formulation.
  • As used herein, the term “occlusive agent component” refers to a hydrophobic agent or mixtures of hydrophobic agents that form an occlusive film on skin that reduces transepidermal water loss (TEWL) by preventing evaporation of water from the stratum corneum.
  • In some embodiments, the occlusive agent component comprises one or more substances selected from fatty acids (e.g., lanolin acid), fatty alcohols (e.g., lanolin alcohol), hydrocarbon oils & waxes (e.g., petrolatum), polyhydric alcohols (e.g., propylene glycol), silicones (e.g., dimethicone), sterols (e.g., cholesterol). vegetable or animal fat (e.g., cocoa butter), vegetable wax (e.g., Carnauba wax), and wax ester (e.g., bees wax).
  • In some embodiments, the occlusive agent component comprises one or more substances selected from lanolin acid fatty alcohols, lanolin alcohol, petrolatum, propylene glycol, dimethicone, cholesterol, cocoa butter, Carnauba wax, and bees wax.
  • In some embodiments, the occlusive agent component comprises petrolatum.
  • In some embodiments, the occlusive agent component comprises white petrolatum.
  • In some embodiments, the oil component comprises a stiffening agent component.
  • In some embodiments, the stiffening agent component is present in an amount of about 2% to about 8% by weight of the formulation.
  • In some embodiments, the stiffening agent component is present in an amount of about 3% to about 6% by weight of the formulation.
  • In some embodiments, the stiffening agent component is present in an amount of about 4% to about 7% by weight of the formulation.
  • As used herein, the term “stiffening agent component” refers to a substance or mixture of substances that increases the viscosity and/or consistency of the formulation or improves the rheology of the formulation.
  • In some embodiments, the stiffening agent component comprises one or more substances independently selected from fatty alcohols.
  • In some embodiments, the stiffening agent component comprises one or more substances independently selected from C12-20 fatty alcohols.
  • In some embodiments, the stiffening agent component comprises one or more substances independently selected from C16-18 fatty alcohols.
  • In some embodiments, the stiffening agent component comprises one or more substances independently selected from cetyl alcohol and stearyl alcohol.
  • In some embodiments, the oil component comprises an emollient component. In some embodiments, the emollient component is present in an amount of about 5% to about 15% by weight of the formulation.
  • In some embodiments, the emollient component is present in an amount of about 7% to about 13% by weight of the formulation.
  • As used herein, the term “emollient component” refers to an agent that softens or soothes the skin or soothes an irritated internal surface.
  • In some embodiments, the emollient component comprises one or more substances independently selected from mineral oils and triglycerides.
  • In some embodiments, the emollient component comprises one or more substances independently selected from light mineral oil and medium chain triglycerides.
  • In some embodiments, the emollient component comprises one or more substances independently selected from light mineral oil, medium chain triglycerides, and dimethicone.
  • In some embodiments, the water is present in an amount of about 35% to about 65% by weight of the formulation.
  • In some embodiments, the water is present in an amount of about 40% to about 60% by weight of the formulation.
  • In some embodiments, the water is present in an amount of about 45% to about 55% by weight of the formulation.
  • In some embodiments, the emulsifier component is present in an amount of about 1% to about 9% by weight of the formulation.
  • In some embodiments, the emulsifier component is present in an amount of about 2% to about 6% by weight of the formulation.
  • In some embodiments, the emulsifier component is present in an amount of about 3% to about 5% by weight of the formulation.
  • In some embodiments, the emulsifier component is present in an amount of about 4% to about 7% by weight of the formulation.
  • In some embodiments, the pharmaceutical formulation comprises an emulsifier component and a stiffening agent component, wherein the combined amount of emulsifier component and stiffening agent component is at least about 8% by weight of the formulation.
  • In some embodiments, the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters.
  • In some embodiments, the emulsifier component comprises one or more substances independently selected from glyceryl stearate, and polysorbate 20.
  • In some embodiments, the pharmaceutical formulation further comprises a stabilizing agent component.
  • In some embodiments, the stabilizing agent component is present in an amount of about 0.05% to about 5% by weight of the formulation.
  • In some embodiments, the stabilizing agent component is present in an amount of about 0.1% to about 2% by weight of the formulation.
  • In some embodiments, the stabilizing agent component is present in an amount of about 0.3 to about 0.5% by weight of the formulation.
  • As used herein, the term “stabilizing agent component” refers to a substance or mixture of substances that improves the stability of the pharmaceutical formulation and/or the compatibility of the components in the formulation. In some embodiments, the stabilizing agent component prevents agglomeration of the emulsion and stabilizes the droplets in the oil-in-water emulsion.
  • In some embodiments, the stabilizing agent component comprises one or more substances independently selected from polysaccharides.
  • In some embodiments, the stabilizing agent component comprises xanthan gum.
  • In some embodiments, the pharmaceutical formulation further comprises a solvent component.
  • In some embodiments, the solvent component is present in an amount of about 10% to about 35% by weight of the formulation.
  • In some embodiments, the solvent component is present in an amount of about 15% to about 30% by weight of the formulation.
  • In some embodiments, the solvent component is present in an amount of about 20% to about 25% by weight of the formulation.
  • As used herein, the term “solvent component” is a liquid substance or mixture of liquid substances capable of dissolving (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile or other substances in the formulation. In some embodiments, the solvent component is a liquid substance or mixture of liquid substances in which (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, or its pharmaceutically acceptable salt, has reasonable solubility. For example, solubilities of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile (free base) or its phosphate salt are reported in Table 21. In some embodiments, a solvent is a substance or mixture thereof, in which (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, or its pharmaceutically acceptable salt (whichever is used), has a solubility of at least about 10 mg/mL or greater, at least about 15 mg/mL or greater, or at least about 20 mg/mL or greater, when measured as described in Example 4.
  • In some embodiments, the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols.
  • In some embodiments, the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol.
  • In some embodiments, the therapeutic agent is present in an amount of about 0.5% to about 1.5% by weight of the formulation on a free base basis.
  • In some embodiments, the therapeutic agent is present in an amount of about 0.5% by weight of the formulation on a free base basis.
  • In some embodiments, the therapeutic agent is present in an amount of about 1% by weight of the formulation on a free base basis.
  • In some embodiments, the therapeutic agent is present in an amount of about 1.5% by weight of the formulation on a free base basis.
  • In some embodiments, the therapeutic agent is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile phosphate.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 35% to about 65% of water by weight of the formulation;
      • from about 10% to about 40% of an oil component by weight of the formulation;
      • from about 1% to about 9% of an emulsifier component by weight of the formulation;
      • from about 10% to about 35% of a solvent component by weight of the formulation;
      • from about 0.05% to about 5% of a stabilizing agent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 40% to about 60% of water by weight of the formulation;
      • from about 15% to about 30% of an oil component by weight of the formulation;
      • from about 2% to about 6% of an emulsifier component by weight of the formulation;
      • from about 15% to about 30% of a solvent component by weight of the formulation;
      • from about 0.1% to about 2% of a stabilizing agent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 45% to about 55% of water by weight of the formulation;
      • from about 17% to about 27% of an oil component by weight of the formulation;
      • from about 3% to about 5% of an emulsifier component by weight of the formulation;
      • from about 20% to about 25% of a solvent component by weight of the formulation;
      • from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 45% to about 55% of water by weight of the formulation;
      • from about 17% to about 27% of an oil component by weight of the formulation;
      • from about 4% to about 7% of an emulsifier component by weight of the formulation;
      • from about 20% to about 25% of a solvent component by weight of the formulation;
      • from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments:
      • the oil component comprises one or more substances independently selected from petrolatums, fatty alcohols, mineral oils, triglycerides, and dimethicones;
      • the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters;
      • the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols; and
      • the stabilizing agent component comprises one or more substances independently selected from polysaccharides.
  • In some embodiments:
      • the oil component comprises one or more substances independently selected from white petrolatum, cetyl alcohol, stearyl alcohol, light mineral oil, medium chain triglycerides, and dimethicone;
      • the emulsifier component comprises one or more substances independently selected from glyceryl stearate and polysorbate 20;
      • the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol; and
      • the stabilizing agent component comprises xanthan gum.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 35% to about 65% of water by weight of the formulation;
      • from about 2% to about 15% of an occlusive agent component by weight of the formulation;
      • from about 2% to about 8% of a stiffening agent component by weight of the formulation;
      • from about 5% to about 15% of an emollient component by weight of the formulation;
      • from about 1% to about 9% of an emulsifier component by weight of the formulation;
      • from about 0.05% to about 5% of a stabilizing agent component by weight of the formulation;
      • from about 10% to about 35% of a solvent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 40% to about 60% of water by weight of the formulation;
      • from about 5% to about 10% of an occlusive agent component by weight of the formulation;
      • from about 2% to about 8% of a stiffening agent component by weight of the formulation;
      • from about 7% to about 12% of an emollient component by weight of the formulation;
      • from about 2% to about 6% of an emulsifier component by weight of the formulation;
      • from about 0.1% to about 2% of a stabilizing agent by weight of the formulation;
      • from about 15% to about 30% of a solvent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 45% to about 55% of water by weight of the formulation;
      • from about 5% to about 10% of an occlusive agent component by weight of the formulation;
      • from about 3% to about 6% of a stiffening agent component by weight of the formulation;
      • from about 7% to about 13% of an emollient component by weight of the formulation;
      • from about 3% to about 5% of an emulsifier component by weight of the formulation;
      • from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation;
      • from about 20% to about 25% of a solvent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 45% to about 55% of water by weight of the formulation;
      • from about 5% to about 10% of an occlusive agent component by weight of the formulation;
      • from about 4% to about 7% of a stiffening agent component by weight of the formulation;
      • from about 7% to about 13% of an emollient component by weight of the formulation;
      • from about 4% to about 7% of an emulsifier component by weight of the formulation;
      • from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation;
      • from about 20% to about 25% of a solvent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the pharmaceutical formulation comprises:
      • from about 45% to about 55% of water by weight of the formulation;
      • about 7% of an occlusive agent component by weight of the formulation;
      • from about 4.5% to about 5% of a stiffening agent component by weight of the formulation;
      • about 10% of an emollient component by weight of the formulation;
      • from about 4% to about 4.5% of an emulsifier component by weight of the formulation;
      • about 0.4% of a stabilizing agent component by weight of the formulation;
      • about 22% of a solvent component by weight of the formulation; and
      • from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
  • In some embodiments, the combined amount of the stiffening agent component and the emulsifier component is at least about 8% by weight of the formulation.
  • In some embodiments:
  • the occlusive agent component comprises a petrolatum;
  • the stiffening agent component comprises one or more substances independently selected from one or more fatty alcohols;
  • the emollient component comprises one or more substances independently selected from mineral oils and triglycerides;
  • the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters;
  • the stabilizing agent component comprises one or more substances independently
  • selected from polysaccharides; and
  • the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols.
  • In some embodiments:
  • the occlusive agent component comprises white petrolatum;
  • the stiffening agent component comprises one or more substances independently selected from cetyl alcohol and stearyl alcohol;
  • the emollient component comprises one or more substances independently selected from light mineral oil, medium chain triglycerides, and dimethicone;
  • the emulsifier component comprises one or more substances independently selected from glyceryl stearate and polysorbate 20;
  • the stabilizing agent component comprises xanthan gum; and
  • the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol.
  • In some embodiments, the pharmaceutical formulation further comprises an antimicrobial preservative component.
  • In some embodiments, the antimicrobial preservative component is present in an amount of about 0.05% to about 3% by weight of the formulation.
  • In some embodiments, the antimicrobial preservative component is present in an amount of about 0.1% to about 1% by weight of the formulation.
  • As used herein, the phrase “antimicrobial preservative component” is a substance or mixtures of substances which inhibits microbial growth in the formulation.
  • In some embodiments, the antimicrobial preservative component comprises one or more substances independently selected from alkyl parabens and phenoxyethanol.
  • In some embodiments, the antimicrobial preservative component comprises one or more substances independently selected from methyl paraben, propyl paraben, and phenoxyethanol.
  • In some embodiments, the pharmaceutical formulation further comprises a chelating agent component.
  • As used herein, the phrase “chelating agent component” refers to a compound or mixtures of compounds that has the ability to bind strongly with metal ions.
  • In some embodiments, the chelating agent component comprises edetate disodium.
  • (R)-3-(4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile can be prepared as described in U.S. Pat. No. 7,598,257 and U.S. Patent Publ. No. 2009/0181959, each of which is incorporated herein by reference in its entirety. The 1:1 phosphate salt of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile can be prepared as described in U.S. Patent Publ. No. 2008/0312259, which is incorporated herein by reference in its entirety.
  • The compounds of the present invention also include pharmaceutically acceptable salts of the compounds disclosed herein. As used herein, the term “pharmaceutically acceptable salt” refers to a salt formed by the addition of a pharmaceutically acceptable acid or base to a compound disclosed herein. As used herein, the phrase “pharmaceutically acceptable” refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient. Pharmaceutically acceptable salts, including mono- and bi- salts, include, but are not limited to, those derived from organic and inorganic acids such as, but not limited to, acetic, lactic, citric, cinnamic, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, oxalic, propionic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, glycolic, pyruvic, methanesulfonic, ethanesulfonic, toluenesulfonic, salicylic, benzoic, and similarly known acceptable acids. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in their entireties.
  • It will also be understood that compounds described herein may exist in solvated, for example hydrated, as well as unsolvated forms. It will further be understood that the present invention encompasses all such solvated forms of the compounds.
  • As used herein, “% by weight of the formulation” means the percent concentration of the component in the formulation is on weight/weight basis. For example, 1% w/w of component A=[(mass of component A)/(total mass of the formulation)] x 100.
  • As used herein, “% by weight of the formulation on a free base basis” of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, or pharmaceutically acceptable salt thereof” means that the % w/w is calculated based on the weight of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile in the total formulation. For example, “0.5% w/w on a free base basis” of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate means that for 100 grams of total formulation, there are 0.66 grams of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate in the formulation (which equates to 0.5 grams of the free base, (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile).
  • In some embodiments, the components are present in exactly the ranges specified (e.g., the term “about” is not present). In some embodiments, “about” means plus or minus 10% of the value.
  • As will be appreciated, some components of the pharmaceutical formulations described herein can possess multiple functions. For example, a given substance may act as both an emulsifying agent component and a stabilizing agent. In some such cases, the function of a given component can be considered singular, even though its properties may allow multiple functionality. In some embodiments, each component of the formulation comprises a different substance or mixture of substances.
  • As used herein, the term “component” can mean one substance or a mixture of substances.
  • As used herein, the term “fatty acid” refers to an aliphatic acid that is saturated or unsaturated. In some embodiments, the fatty acid is in a mixture of different fatty acids. In some embodiments, the fatty acid has between about eight to about thirty carbons on average. In some embodiments, the fatty acid has about 12 to 20, 14-20, or 16-18 carbons on average. Suitable fatty acids include, but are not limited to, cetyl acid, stearic acid, lauric acid, myristic acid, erucic acid, palmitic acid, palmitoleic acid, capric acid, caprylic acid, oleic acid, linoleic acid, linolenic acid, hydroxystearic acid, 12-hydroxystearic acid, cetostearic acid, isostearic acid, sesquioleic acid, sesqui-9-octadecanoic acid, sesquiisooctadecanoic acid, behenic acid, isobehenic acid, and arachidonic acid, or mixtures thereof.
  • As used herein, the term “fatty alcohol” refers to an aliphatic alcohol that is saturated or unsaturated. In some embodiments, the fatty alcohol is in a mixture of different fatty alcohols. In some embodiments, the fatty alcohol has between about 12 to about 20, about 14 to about 20, or about 16 to about 18 carbons on average. Suitable fatty alcohols include, but are not limited to, stearyl alcohol, lauryl alcohol, palmityl alcohol, cetyl alcohol, capryl alcohol, caprylyl alcohol, oleyl alcohol, linolenyl alcohol, arachidonic alcohol, behenyl alcohol, isobehenyl alcohol, selachyl alcohol, chimyl alcohol, and linoleyl alcohol, or mixtures thereof.
  • As used herein, the term “polyalkylene glycol”, employed alone or in combination with other terms, refers to a polymer containing oxyalkylene monomer units, or copolymer of different oxyalkylene monomer units, wherein the alkylene group has 2 to 6, 2 to 4, or 2 to 3 carbon atoms. As used herein, the term “oxyalkylene”, employed alone or in combination with other terms, refers to a group of formula —O-alkylene-. In some embodiments, the polyalkylene glycol is polyethylene glycol.
  • As used herein, the term, “sorbitan fatty ester” includes products derived from sorbitan or sorbitol and fatty acids and, optionally, poly(ethylene glycol) units, including sorbitan esters and polyethoxylated sorbitan esters. In some embodiments, the sorbitan fatty ester is a polyethoxylated sorbitan ester.
  • As used herein, the term “sorbitan ester” refers to a compound, or mixture of compounds, derived from the esterification of sorbitol and at least one fatty acid. Fatty acids useful for deriving the sorbitan esters include, but are not limited to, those described herein. Suitable sorbitan esters include, but are not limited to, the Span™ series (available from Uniqema), which includes Span 20 (sorbitan monolaurate), 40 (sorbitan monopalmitate), 60 (sorbitan monostearate), 65 (sorbitan tristearate), 80 (sorbitan monooleate), and 85 (sorbitan trioleate). Other suitable sorbitan esters include those listed in R. C. Rowe and P. J. Shesky, Handbook of pharmaceutical excipients, (2006), 5th ed., which is incorporated herein by reference in its entirety.
  • As used herein, the term “polyethoxylated sorbitan ester” refers to a compound, or mixture thereof, derived from the ethoxylation of a sorbitan ester. The polyoxethylene portion of the compound can be between the fatty ester and the sorbitan moiety. As used herein, the term “sorbitan ester” refers to a compound, or mixture of compounds, derived from the esterification of sorbitol and at least one fatty acid. Fatty acids useful for deriving the polyethoyxlated sorbitan esters include, but are not limited to, those described herein. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 2 to about 200 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 2 to about 100 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 4 to about 80 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 4 to about 40 oxyethylene units. In some embodiments, the polyoxyethylene portion of the compound or mixture has about 4 to about 20 oxyethylene units. Suitable polyethoxylated sorbitan esters include, but are not limited to the Tween™ series (available from Uniqema), which includes Tween 20 (POE(20) sorbitan monolaurate), 21 (POE(4) sorbitan monolaurate), 40 (POE(20) sorbitan monopalmitate), 60 (POE(20) sorbitan monostearate), 60K (POE(20) sorbitan monostearate), 61 (POE(4) sorbitan monostearate), 65 (POE(20) sorbitan tristearate), 80 (POE(20) sorbitan monooleate), 80K (POE(20) sorbitan monooleate), 81 (POE(5) sorbitan monooleate), and 85 (POE(20) sorbitan trioleate). As used herein, the abbreviation “POE” refers to polyoxyethylene. The number following the POE abbreviation refers to the number of oxyethylene repeat units in the compound. Other suitable polyethoxylated sorbitan esters include the polyoxyethylene sorbitan fatty acid esters listed in R. C. Rowe and P. J. Shesky, Handbook of pharmaceutical excipients, (2006), 5th ed., which is incorporated herein by reference in its entirety. In some embodiments, the polyethoxylated sorbitan ester is a polysorbate. In some embodiments, the polyethoxylated sorbitan ester is polysorbate 20.
  • As used herein, the term “glyceryl fatty esters” refers to mono-, di- or triglycerides of fatty acids. The glyceryl fatty esters may be optionally substituted with sulfonic acid groups, or pharmaceutically acceptable salts thereof. Suitable fatty acids for deriving glycerides of fatty acids include, but are not limited to, those described herein. In some embodiments, the glyceryl fatty ester is a mono-glyceride of a fatty acid having 12 to 18 carbon atoms. In some embodiments, the glyceryl fatty ester is glyceryl stearate.
  • As used herein, the term “triglycerides” refers to a triglyceride of a fatty acid. In some embodiments, the triglyceride is medium chain triglycerides.
  • As used herein, the term “alkylene glycol” refers to a group of formula —O-alkylene-, wherein the alkylene group has 2 to 6, 2 to 4, or 2 to 3 carbon atoms. In some embodiments, the alkylene glycol is propylene glycol (1,2-propanediol).
  • As used herein, the term “polyethylene glycol” refers to a polymer containing ethylene glycol monomer units of formula —O—CH2—CH2—. Suitable polyethylene glycols may have a free hydroxyl group at each end of the polymer molecule, or may have one or more hydroxyl groups etherified with a lower alkyl, e.g., a methyl group. Also suitable are derivatives of polyethylene glycols having esterifiable carboxy groups. Polyethylene glycols useful in the present invention can be polymers of any chain length or molecular weight, and can include branching In some embodiments, the average molecular weight of the polyethylene glycol is from about 200 to about 9000. In some embodiments, the average molecular weight of the polyethylene glycol is from about 200 to about 5000. In some embodiments, the average molecular weight of the polyethylene glycol is from about 200 to about 900. In some embodiments, the average molecular weight of the polyethylene glycol is about 400. Suitable polyethylene glycols include, but are not limited to polyethylene glycol-200, polyethylene glycol-300, polyethylene glycol-400, polyethylene glycol-600, and polyethylene glycol-900. The number following the dash in the name refers to the average molecular weight of the polymer.
  • It is further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.
  • Methods
  • The pharmaceutical formulations of the invention are useful in treating skin disorders. In some embodiments, the skin disorder is an autoimmune bullous skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP). In some embodiments, the skin disorder is psoriasis (for example, psoriasis vulgaris), atopic dermatitis, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis). For example, certain substances including some pharmaceuticals when topically applied can cause skin sensitization. In some embodiments, co-administration or sequential administration of the topical formulations of the invention together with the agent causing unwanted sensitization can be helpful in treating such unwanted sensitization or dermatitis.
  • The present invention further provides a method of treating dermatological side effects of other pharmaceuticals by administration of the compound of the invention. For example, numerous pharmaceutical agents result in unwanted allergic reactions which can manifest as acneiform rash or related dermatitis. Example pharmaceutical agents that have such undesirable side effects include anti-cancer drugs such as gefitinib, cetuximab, erlotinib, and the like. The formulations of the invention can be administered systemically or topically (e.g., localized to the vicinity of the dermatitis) in combination with (e.g., simultaneously or sequentially) the pharmaceutical agent having the undesirable dermatological side effect. In some embodiments, the formulation of the invention can be administered topically together with one or more other pharmaceuticals, where the other pharmaceuticals when topically applied in the absence of a formulation of the invention cause contact dermatitis, allergic contact sensitization, or similar skin disorder. Accordingly, formulation of the invention include topical formulations further comprising an additional pharmaceutical agent which can cause dermatitis, skin disorders, or related side effects.
  • As used herein, the term “individual” or “patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • As used herein, the phrase “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician.
  • As used herein, the term “treating” or “treatment” refers to one or more of (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
  • Combination Therapies
  • One or more additional pharmaceutical agents such as, for example, chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors such as, for example, those described in WO 2006/056399, or other agents can be used in combination with the formulations of the present invention for treatment of JAK-associated diseases, disorders or conditions. The one or more additional pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • Example chemotherapeutic include proteosome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • Example steroids include corticosteroids such as dexamethasone or prednisone.
  • Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521,184, WO 04/005281, and U.S. Ser. No. 60/578,491.
  • Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120.
  • Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444.
  • Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402.
  • In some embodiments, the formulations of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • In some embodiments, a corticosteroid such as dexamethasone is administered to a patient in combination with the compound of the invention where the dexamethasone is administered intermittently as opposed to continuously.
  • Labeled Compounds and Assay Methods
  • Another aspect of the present invention relates to formulations comprising a labeled active compound (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating JAK in tissue samples, including human, and for identifying JAK ligands by inhibition binding of a labeled compound. Accordingly, the present invention includes JAK assays that contain such labeled compounds.
  • The present invention further includes formulations of an isotopically-labeled compound. An “isotopically” or “radio-labeled” compound is a compound where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2H (also written as D for deuterium), 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 18F, 35S, 36Cl, 82Br, 75Br, 76Br, 77Br, 123I, 124I and 131I. The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro JAK labeling and competition assays, compounds that incorporate 3H 14C, 82Br, 125I, 131I, 35S or will generally be most useful. For radio-imaging applications 11C, 18F, 125I, 123I, 124I, 131I, 75Br, 76Br or 77Br will generally be most imaging applications 11C, 18F, 125I, 123I, 124I, 131I, 75Br, 76Br or 77Br will generally be most useful.
  • It is understood that a “radio-labeled ” or “labeled compound” is a compound that has incorporated at least one radionuclide. In some embodiments the radionuclide is selected from the group consisting of 3H, 14C, 125I, 35 S and 82Br.
  • Kits The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of JAK-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical formulation of the invention. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results. In some embodiments, the present invention provides pharmaceutical formulations comprising the components specified in the example formulations (e.g., Example 3), wherein the components are present in about the amounts in Tables 2-5.
  • EXAMPLES Example 1 (3R)- and (3S)-3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile
  • Figure US20110288107A1-20111124-C00002
  • Step 1. (2E)- and (2Z)-3-Cyclopentylacrylonitrile
  • To a solution of 1.0 M potassium tert-butoxide in THF (235 mL) at 0° C. was added dropwise a solution of diethyl cyanomethylphosphonate (39.9 mL, 0.246 mol) in THF (300 mL). The cold bath was removed and the reaction was warmed to room temperature followed by recooling to 0° C., at which time a solution of cyclopentanecarbaldehyde (22.0 g, 0.224 mol) in THF (60 mL) was added dropwise. The bath was removed and the reaction warmed to ambient temperature and stirred for 64 hours. The mixture was partitioned between diethyl ether and water, the aqueous was extracted with three portions of ether, followed by two portions of ethyl acetate. The combined extracts were washed with brine, then dried over sodium sulfate, filtered and concentrated in vacuo to afford a mixture containing 24.4 g of olefin isomers which was used without further purification (89%).
  • 1H NMR (400 MHz, CDCl3): δ 6.69 (dd, 1H, trans olefin), 6.37 (t, 1H, cis olefin), 5.29 (dd, 1H, trans olefin), 5.20 (d, 1H, cis olefin), 3.07-2.95 (m, 1H, cis product), 2.64-2.52 (m, 1H, trans product), 1.98-1.26 (m, 16H).
  • Step 2. (3R)- and (3S)-3-Cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile
  • To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (15.0 g, 0.0476 mol) in ACN (300 mL) was added 3-cyclopentylacrylonitrile (15 g, 0.12 mol) (as a mixture of cis and trans isomers), followed by DBU (15 mL, 0.10 mol). The resulting mixture was stirred at room temperature overnight. The ACN was evaporated. The mixture was diluted with ethyl acetate, and the solution was washed with 1.0 N HCl. The aqueous layer was back-extracted with three portions of ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel chromatography (gradient of ethyl acetate/hexanes) to yield a viscous clear syrup, which was dissolved in ethanol and evaporated several times to remove ethyl acetate, to afford 19.4 g of racemic adduct (93%). The enantiomers were separated by preparative-HPLC, (OD-H, 15% ethanol/hexanes) and used separately in the next step to generate their corresponding final product. The final products (see Step 3) stemming from each of the separated enantiomers were found to be active JAK inhibitors; however, the final product stemming from the second peak to elute from the preparative-HPLC was more active than its enantiomer.
  • 1H NMR (300 MHz, CDCl3): δ 8.85 (s, 1H), 8.32 (s, 2H), 7.39 (d, 1H), 6.80 (d, 1H), 5.68 (s, 2H), 4.26 (dt, 1H), 3.54 (t, 2H), 3.14 (dd, 1H), 2.95 (dd, 1H), 2.67-2.50 (m, 1H), 2.03-1.88 (m, 1H), 1.80-1.15 (m, 7H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):437 (M+1).
  • Step 3. (3R)- and (3S)-3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile
  • To a solution of 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (6.5 g, 0.015 mol, R or S enantiomer as isolated above) in DCM (40 mL) was added TFA (16 mL) and this was stirred for 6 hours. The solvent and TFA were removed in vacuo. The residue was dissolved in DCM and concentrated using a rotary evaporator two further times to remove as much as possible of the TFA. Following this, the residue was stirred with ethylenediamine (4 mL, 0.06 mol) in methanol (30 mL) overnight. The solvent was removed in vacuo, water was added and the product was extracted into three portions of ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, decanted and concentrated to afford the crude product which was purified by flash column chromatography (eluting with a gradient of methanol/DCM). The resulting mixture was further purified by preparative-HPLC/MS (C18 eluting with a gradient of ACN/H2O containing 0.15% NH4OH) to afford product (2.68 g, 58%).
  • 1H NMR (400 MHz, D6-dmso): δ 12.11 (br s, 1H), 8.80 (s, 1H), 8.67 (s, 1H), 8.37 (s, 1H), 7.60 (d, 1H), 6.98 (d, 1H), 4.53 (dt, 1H), 3.27 (dd, 1H), 3.19 (dd, 1H), 2.48-2.36 (m, 1H), 1.86-1.76 (m, 1H), 1.68-1.13 (m, 7H); MS(ES):307(M+1).
  • Example 2 (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt
  • Figure US20110288107A1-20111124-C00003
  • To a test tube was added (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile (153.5 mg) and phosphoric acid (56.6 mg) followed by isopropyl alcohol (IPA) (5.75 mL). The resulting mixture was heated to clear, cooled to room temperature, and then stirred for another 2 hours. The precipitate was collected by filtration and the cake was washed with 0.6 mL of cold IPA. The cake was dried under vacuum to constant weight to provide the final salt product (171.7 mg).
  • The phosphoric acid salt was shown to be a 1:1 salt by 1H NMR and crystallinity was confirmed by X-ray powder diffraction (XRPD). Differential scanning calorimetry (DSC) gave a sharp melting peak at about 198.66° C. The product showed little weight loss up to 200° C. by TGA.
  • Example 3 Preparation of Oil-In-Water Cream Formulations of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric Acid Salt
  • An oil-in-water cream formulation was prepared for (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphoric acid salt (Example 2) at 0.5, 1.0 and 1.5% by weight of the formulation (free base equivalent). The compositions for a 15 gram tube are provided in Table 2 below. The formulation for three strengths were identical except for adjustments to the purified water quantity based on the amount of active ingredient. All excipients used in the formulation were compendial grade (ie, USP/NF or BP) or are approved for use in topical products.
  • The quantitative formulae for representative 400 kg batches of the cream formulation for Example 2 at 0.5, 1.0 and 1.5% are also provided in Tables 3, 4, and 5, respectively.
  • TABLE 2
    FORMULA Percentage of Total
    PHASE COMPONENT Function (% w/w) Grams/Tube
    Paraben Propylene Glycol Solvent 10.00 1.5
    USP
    Methyl Paraben Antimicrobial 0.10 0.015
    NF preservative
    Propyl Paraben Antimicrobial 0.05 0.0075
    NF preservative
    Xanthan Propylene Glycol Solvent 5.00 0.75
    Gum USP
    Xanthan Gum NF Suspending 0.40 0.06
    stabilizing, viscosity-
    increasing agent
    Oil Light Mineral Oil Emollient, solvent 4.00 0.6
    NF
    Glyceryl Stearate Emulsifier 3.00 0.45
    SE
    Polysorbate
    20 Emulsifying/ 1.25 0.1875
    NF stabilizing agen
    White Petrolatum Occlusive agent 7.00 1.05
    USP
    Cetyl Alcohol NF Stiffening agent, 3.00 0.45
    consistency improver
    Stearyl Alcohol Stiffening agent 1.75 0.2625
    NF
    Dimethicone
    360 Skin protectant 1.00 0.15
    NF
    Medium Chain Emollient, solvent 5.00 0.75
    Triglyceride NF
    Aqueous/ Purified Water Solvent 50.24-48.92 7.536-7.338
    Active USP
    Edetate Disodium Chelating agent 0.05 0.0075
    USP
    Polyethylene Solvent 7.00 1.05
    Glycol USP
    Example 2 * Active 0.66-1.98 0.099-0.297
    Final Phenoxyethanol Antimicrobial 0.50 0.075
    BP preservative
    Total 100.00% 15
    * 1.32% of Example 2 is equivalent to 1.0% of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile free base
  • TABLE 3
    Ingredient Kilograms Percentage (w/w)
    (R)-3-(4-(7H-pyrrolo[2,3-d] 2.64 (phosphate 0.66 (phosphate
    pyrimidin-4-yl)- salt)/2.0 (free salt)/0.5 (free
    1H-pyrazol-1-yl)-3- base) base)
    cyclopentylpropanenitrile
    phosphoric acid salt (Example 2)
    Propylene Glycol USP 40.0 10.00
    Methyl Paraben NF 0.4 0.10
    Propyl Paraben NF 0.2 0.05
    Propylene Glycol USP 20.0 5.00
    Xanthan Gum NF 1.6 0.40
    Light Mineral Oil NF 16.0 4.00
    Glyceryl Stearate SE 12.0 3.00
    Polysorbate 20 NF 5.0 1.25
    White Petrolatum USP 28.0 7.00
    Cetyl alcohol NF 12.0 3.00
    Stearyl alcohol NF 7.0 1.75
    Dimethicone 360 NF 4.0 1.00
    Medium Chain Triglycerides NF 20.0 5.00
    Purified Water USP 201 50.25
    (approximate)
    Edetate Disodium USP 0.2 0.05
    Polyethylene Glycol USP 28.0 7.00
    Phenoxyethanol BP 2.0 0.5
    Total (approximate) 400.0 100
  • TABLE 4
    Ingredient Kilograms Percentage (w/w)
    (R)-3-(4-(7H-pyrrolo[2,3-d] 5.28 (phosphate 1.32 (phosphate
    pyrimidin-4-yl)- salt)/4.0 (free salt)/1.00 (free
    1H-pyrazol-1-yl)-3- base) base)
    cyclopentylpropanenitrile
    phosphoric acid salt (Example 2)
    Propylene Glycol USP 40.0 10.00
    Methyl Paraben NF 0.4 0.10
    Propyl Paraben NF 0.2 0.05
    Propylene Glycol USP 20.0 5.00
    Xanthan Gum NF 1.6 0.40
    Light Mineral Oil NF 16.0 4.00
    Glyceryl Stearate SE 12.0 3.00
    Polysorbate 20 NF 5.0 1.25
    White Petrolatum USP 28.0 7.00
    Cetyl alcohol NF 12.0 3.00
    Stearyl alcohol NF 7.0 1.75
    Dimethicone 360 NF 4.0 1.00
    Medium Chain Triglycerides NF 20.0 5.00
    Purified Water USP 198.5 49.6
    (approximate)
    Edetate Disodium USP 0.2 0.05
    Polyethylene Glycol USP 28.0 7.00
    Phenoxyethanol BP 2.0 0.5
    Total (approximate) 400.0 100
  • TABLE 5
    Ingredient Kilograms Percentage (w/w)
    (R)-3-(4-(7H-pyrrolo[2,3- 7.92 (phosphate 1.98 (phophate
    d]pyrimidin-4-yl)- salt)/6.0 (free salt)/1.5 (free
    1H-pyrazol-1-yl)-3- base) base)
    cyclopentylpropanenitrile
    phosphoric acid salt (Example 2)
    Propylene Glycol USP 40.0 10.00
    Methyl Paraben NF 0.4 0.10
    Propyl Paraben NF 0.2 0.05
    Propylene Glycol USP 20.0 5.00
    Xanthan Gum NF 1.6 0.40
    Light Mineral Oil NF 16.0 4.00
    Glyceryl Stearate SE 12.0 3.00
    Polysorbate 20 NF 5.0 1.25
    White Petrolatum USP 28.0 7.00
    Cetyl alcohol NF 12.0 3.00
    Stearyl alcohol NF 7.0 1.75
    Dimethicone 360 NF 4.0 1.00
    Medium Chain Triglycerides NF 20.0 5.00
    Purified Water USP 195.5 48.9
    (approximate)
    Edetate Disodium USP 0.2 0.05
    Polyethylene Glycol USP 28.0 7.00
    Phenoxyethanol BP 2.0 0.5
    Total (approximate) 400.0 100
  • The oil-in-water cream formulations were synthesized according to the following procedure at either a 3.5 kg or 400 kg scale (when made at a 3.5 kg batch size, the amounts in Tables 3-5 were scaled appropriately). Some batches were subject to minor changes associated with scale-up, such as the size of mixing vessels and mixers. Generally, overhead mixer with high and low shear mixing blades are suitable for the process. FIG. 1 shows a flowchart representation of the process for making the oil-in-water formulation. The (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile is referred to as “API” throughout this application.
  • Procedure
  • 1. A paraben phase was prepared by mixing methyl and propyl parabens with a portion of the propylene glycol (see % in Tables 2-5).
  • 2. Next, a xanthan gum phase was prepared by mixing xanthan gum with propylene glycol (see % in Table 2-5).
  • 3. An oil phase was then prepared by mixing light mineral oil, glyceryl stearate, polysorbate 20, white petrolatum, cetyl alcohol, stearyl alcohol, dimethicone and medium chain triglycerides. The phase is heated to 70-80° C. to melt and form a uniform mixture.
  • 4. The aqueous phase was next prepared by mixing purified water, polyethylene glycol, and disodium EDTA. The phase is heated to 70-80° C.
  • 5. The aqueous phase of step 4, paraben phase of step 1, and Example 2 (phosphate salt of API) were combined to form a mixture.
  • 6. The xanthan gum phase from step 2 was then added to the mixture from step 5.
  • 7. The oil phase from step 3 was then combined under high shear mixing with the mixture from step 6 to form an emulsion.
  • 8. Phenoxyethanol was then added to the emulsion from step 7. Mixing was continued, and then the product was cooled under low shear mixing.
  • More consistent batches at larger scales (e.g., 140 kg) could be obtained by adding Example 2 gradually to the aqueous phase and then combining with the other phases. Similarly, more consistent batches could be obtained by slower cooling (e.g., by using room temperature water in the outer jacket of the reactor, rather than lower temperature water.
  • Analytical Results for Cream Formulations and Stability Studies
  • A. Methods
  • The appearance of the cream was visually inspected. Viscosity was measured using a Brookfield viscometer at 25° C. The pH was measured on the final cream formulation. The microbial limit testing is performed as per USP. The fill weight is analyzed as an in-process test during filling of the cream into tubes.
  • Assay, related substances, identity and content uniformity were determined in the formulation by a gradient reverse-phase HPLC with UV detection at 294 nm A Waters HPLC was used with a Zorbax SB-C18 column (3.5 μm, 4.6×150 mm) at a flow rate of 1.0 mL/minute, temperature of 40° C. using Mobile Phase A of 2 mL of TFA into 4 L of Water (0.05% TFA), or Mobile Phase B of 2 mL of TFA into 4 L of methanol (0.05% TFA).
  • B. Results
  • Results are shown below for a 3.5 kg batches at 0.5%, 1% and 1.5% strength of Example 2 (free base basis (API)) (Table 6).
  • TABLE 6
    Accep- Strength
    tance 0.5% 1.0% 1.5%
    Test Criteria Placebo w/w w/w w/w
    Appearance Smooth, Conforms Conforms Conforms Conforms
    white
    emulsion
    pH Report 6.5 3.6 3.3 3.1
    results
    Viscosity Report 96,500 66,500 64,800 72,900
    results
    API Assay 90.0- N/A 100.0 102.0 102.0
    (%) 110.0%
    API Report ND* ND* ND* ND*
    Related results
    Substances
    Content   90- Top N/A 100 101 101
    Uniformity   110% 100 101 101
    testing RSD: ≦5% Middle N/A 100 101 102
    100 102 103
    Bottom N/A 100 102 103
    100 102 102
    Avg. N/A 100 102 102
    RSD % 0.0 0.5 0.8
  • The stability data from batches of the cream formulation at 0.5, 1.0 and 1.5% w/w strength stored in 15 gram aluminum tubes is provided in Tables 7-10 and 19-20. Further, stability data from batches of the cream formulation at 0.5, 1.0 and 1.5% w/w strength packaged in amber glass jars (2 oz. with teflon cap) is provided in Tables 13-17, while longer stability data for the 1.0% w/w formulation packaged in 16 oz. amber glass jars is provided in Tables 11-12. The preliminary stability data for the drug product did not show any chemical instability after 3 months of storage at 25° C./60% RH and 40° C./75% RH in either packaging configuration. A change in viscosity is seen following 3 months at 40° C./75% RH for formulation stored in amber glass jars. However, physical inspection of the product did not indicate any phase separation.
  • Acceptance criteria are shown below.
  • Test Acceptance Criteria
    Appearance Smooth, white cream
    pH Report results
    Weight Loss Report results
    Viscosity (cps) Report results
    API Assay (%) 90.0-110.0% of label claim
    API Related Substances (RRT: Area %) Report results
    Total Related Substances (RRT: Area %) Report results
    MLT Absent/1 g
    (Objectionable organisms)
    MLT (P.Aeruginosa) Absent/1 g
    MLT (S.Aureus) Absent/1 g
    MLT (Total Aerobic) NMT 100 CFU/g
    MLT (Total Yeast and Molds) NMT 10 CFU/g
  • TABLE 7
    Stability Data for 0.5% w/w Cream at 25° C./60% RH
    (15 aluminum gram tubes)
    Time (Months)
    Test 0 1 3 6
    Appearance Conforms Conforms Conforms Conforms
    pH 3.6 3.6 3.6 3.6
    Weight Loss NA NA 0.0 0.0
    Viscosity (cps) 23400 29900 25400 24900
    API Assay (%) 103.7 107.2 102.5 105.9
    API Related ND ND 1.09:0.15 ND
    Substances 1.18:0.19
    (RRT:Area %)
    Total Related NA NA 0.34 NA
    Substances
    (RRT:Area %)
    MLT Absent/1 g NA Absent/1 g Absent/1 g
    (Objectionable
    organisms)
    MLT (P. Aeruginosa) Absent/1 g NA Absent/1 g Absent/1 g
    MLT (S. Aureus) Absent/1 g NA Absent/1 g Absent/1 g
    MLT (Total Aerobic) <10 NA <10 <10
    MLT (Total Yeast <10 NA <10 <10
    and Molds)
    Time (Months)
    Test 9 12 18 24
    Appearance Conforms Conforms Conforms Conforms
    pH 3.5 3.5 3.5 3.6
    Weight Loss 0.0 0.0 0.0 0.0
    Viscosity (cps) 26000 23000 20900 22500
    API Assay (%) 105.4 105.7 104.4 104.0
    API Related 1.10:0.10 1.09:0.14 0.95:0.18 0.11:0.24
    Substances 1.09:0.20 0.95:0.23
    (RRT:Area %) 1.11:0.08
    Total Related 0.10 0.14 0.38 0.55
    Substances
    (RRT:Area %)
    MLT Absent/1 g Absent/1 g NA Absent/1 g
    (Objectionable
    organisms)
    MLT (P. Aeruginosa) Absent/1 g Absent/1 g NA Absent/1 g
    MLT (S. Aureus) Absent/1 g Absent/1 g NA Absent/1 g
    MLT (Total Aerobic) <10 <10 NA <10
    MLT (Total Yeast <10 <10 NA <10
    and Molds)
  • TABLE 8
    Stability Data for 0.5% w/w Cream at 40° C./75% RH
    (15 aluminum gram tubes)
    Time (Months)
    Test 0 mo. 1 mo. 3 mo. 6 mo.
    Appearance Conforms Conforms Conforms Conforms
    pH 3.6 3.6 3.6 3.5
    Weight Loss N/A N/A 0.0 0.0
    Viscosity (cps) 23400 26300 19800 18600
    API Assay (%) 103.7 103.1 105.3 105.0
    API Related Substances N/D N/D 1.09:0.14 1.32:0.21
    (RRT:Area %) 1.39:0.40
    Total Related N/A N/A 0.14 0.61
    Substances
    (RRT:Area %)
    MLT Absent/1 g N/A Absent/1 g Absent/1 g
    (Objectionable
    organisms)
    MLT (P. Aeruginosa) Absent/1 g N/A Absent/1 g Absent/1 g
    MLT (S. Aureus) Absent/1 g N/A Absent/1 g Absent/1 g
    MLT (Total Aerobic) <10 N/A <10 <10
    MLT (Total Yeast and <10 N/A <10 <10
    Molds)
  • TABLE 9
    Stability Data for 1.5% w/w Cream at 25° C./60% RH
    (15 aluminum gram tubes)
    Time (Months)
    Test 0 1 3 6
    Appearance Conforms Conforms Conforms Conforms
    pH 3.2 3.1 3.2 3.1
    Weight Loss NA NA 0.0 0.0
    Viscosity (cps) 29433 35800 27400 26200
    API Assay (%) 102.7 104.9 103.9 105.0
    API Related Substances ND ND 1.09:0.14 ND
    (RRT:Area %)
    Total Related Substances NA NA 0.14 ND
    (RRT:Area %)
    MLT (Objectionable Absent/1 g NA Absent/1 g Absent/1 g
    organisms)
    MLT (P. Aeruginosa) Absent/1 g NA Absent/1 g Absent/1 g
    MLT (S. Aureus) Absent/1 g NA Absent/1 g Absent/1 g
    MLT (Total Aerobic) <10 NA <10 <10
    MLT (Total Yeast and <10 NA <10 <10
    Molds)
    Time (Months)
    Test 9 12 18 24
    Appearance Conforms Conforms Conforms Conforms
    pH 3.4 3.1 3.1 3.1
    Weight Loss 0.0 0.0 0.0 0.0
    Viscosity (cps) 25600 23800 21200 22200
    API Assay (%) 103.7 105.0 102.6 103.0
    API Related Substances 1.10:0.12 1.09:0.13 1.09:0.21 0.20:0.09
    (RRT:Area %) 0.95:0.07
    1.11:0.10
    Total Related Substances 0.12 0.13 0.21 0.26
    (RRT:Area %)
    MLT (Objectionable Absent/1 g Absent/1 g NA Absent/1 g
    organisms)
    MLT (P. Aeruginosa) Absent/1 g Absent/1 g NA Absent/1 g
    MLT (S. Aureus) Absent/1 g Absent/1 g NA Absent/1 g
    MLT (Total Aerobic) <10 <10 NA <10
    MLT (Total Yeast and <10 <10 NA <10
    Molds)
  • TABLE 10
    Stability Data for 1.5% w/w Cream at 40° C./75% RH
    (15 aluminum gram tubes)
    Time (Months)
    Test 0 mo. 1 mo. 3 mo. 6 mo.
    Appearance Conforms Conforms Conforms Conforms
    pH 3.2 3.1 3.2 3.1
    Weight Loss(g) N/A N/A 0.0 0.0
    Viscosity (cps) 29433 29800 22400 16300
    API Assay 102.7 104.9 103.0 104.4
    API Related Substances N/D N/D 1.09:0.14 1.32:0.20
    RRT:Area % 1.39:0.34
    Total Related Substance N/A N/A 0.14 0.54
    Objectionable Absent/1 g N/A Absent/1 g Absent/1 g
    organisms
    P. Aeruginosa Absent/1 g N/A Absent/1 g Absent/1 g
    S. Aureus Absent/1 g N/A Absent/1 g Absent/1 g
    Total Aerobic <10 N/A <10 <10
    Total Yeast and Molds <10 N/A <10 <10
  • TABLE 11
    Stability Data for 1.0% w/w Cream at 25° C./60% RH
    (16 oz. amber glass jars)
    Time (Months)
    Test 0 3 6 12
    Appearance Conforms Conforms Conforms Conforms
    PH 3.5 3.3 3.3 3.3
    Viscosity (cps) 35700 25600 21200 21400
    API Assay 102.5 98.6 101.5 99.2
    API Related Substances 0.89:0.08 0.11:0.20 ND 0.88:NQ
    (RRT:Area %) 1.15:0.19
    Total Related Substances 0.27 0.20 NA 0.20
    (RRT:Area %)
    MLT Absent/1 g Absent/1 g Absent/1 g Absent/1 g
    (Objectionable
    organisms)
    MLT (P. Aeruginosa) Absent/1 g Absent/1 g Absent/1 g Absent/1 g
    MLT (S. Aureus) Absent/1 g Absent/1 g Absent/1 g Absent/1 g
    MLT (Total Aerobic) <10 <10 <10 <10
    MLT (Total Yeast and <10 <10 <10 <10
    Molds)
    NA: Not applicable
    ND: Not Detected
    NQ: Not Quantifiable
  • TABLE 12
    Stability Data for 1.0% w/w Cream at 25° C./60% RH
    (16 oz. amber glass jars)
    Time (Months)
    Test 0 6
    Appearance Conforms Conforms
    pH 3.5 3.2
    API Assay 102.5 100.8
    API Related Substances 0.89:0.08 ND
    (RRT: Area %) 1.15:0.19
    Total Related Substances 0.27 ND
    (RRT: Area %)
    MLT Absent/1 g Absent/1 g
    (Objectionable
    organisms)
    MLT (P.Aeruginosa) Absent/1 g Absent/1 g
    MLT (S.Aureus) Absent/1 g Absent/1 g
    MLT (Total Aerobic) <10 <10
    MLT (Total Yeast and <10 <10
    Molds)
  • TABLE 13
    Stability Data for 0.5% w/w Cream at 25° C./60% RH
    (2 oz. amber glass jars)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, Conforms Conforms Conforms Conforms
    ance white cream
    pH Report 3.6 3.5 3.6 3.6
    result
    Viscosity Report 66500 71500 66000 56800
    (cps) results
    API Assay 90.0- 100.0 101.0 100.0 100.0
    (%) 110.0%
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • TABLE 14
    Stability Data for 0.5% w/w Cream at 40° C./75% RH
    (2 oz. amber glass jars)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, Conforms Conforms Conforms Conforms
    ance white
    cream
    pH Report 3.6 3.6 3.5 3.6
    result
    Viscosity Report 66500 63900 51900 39000
    (cps) results
    API 90.0- 100.0 99.0 98.0 102.0
    Assay (%) 110.0%
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • TABLE 15
    Stability Data for 1.0% w/w Cream at
    25° C./60% RH (2 oz. amber glass jars)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, Con- Con- Con- Con-
    ance white forms forms forms forms
    cream
    pH Report 3.3 3.2 3.2 3.3
    result
    Viscosity Report 64800 69300 61400 50500
    (cps) results
    API 90.0- 102.0 102.0 103.0 102.5
    Assay (%) 110.0%
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • TABLE 16
    Stability Data for 1.0% w/w Cream at 40° C./75% RH
    (2 oz. amber glass jars)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, Con- Con- Con- Con-
    ance white cream forms forms forms forms
    pH Report 3.3 3.2 3.2 3.3
    result
    Viscosity Report 64800 57900 55100 33500
    (cps) results
    API Assay 90.0- 102.0 102.0 101.0 103.0
    (%) 110.0%
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • TABLE 17
    Stability Data for 1.5% w/w Cream at 25° C./
    60% RH (2 oz. amber glass jars)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, Con- Con- Con- Con-
    ance white forms forms forms forms
    cream
    pH Report 3.1 2.9 3.1 3.2
    result
    Viscosity Report 72900 66600 62400 60300
    (cps) results
    API 90.0- 101.7 101.7 101.7 104.3
    Assay (%) 110.0%
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • TABLE 18
    Stability Data for 1.5% w/w Cream at 40° C./75% RH
    (2 oz. amber glass jars)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, Con- Con- Con- Con-
    ance white cream forms forms forms forms
    pH Report 3.1 3.1 3.1 3.2
    result
    Viscosity Report 72900 62500 53000 43800
    (cps) results
    Assay 90.0- 101.7 103.0 102.0 104.3
    (%) 110.0%
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • TABLE 19
    Stability Data for 1.0% w/w Cream at 25° C./
    60% RH (15 gram aluminum tubes)
    Acceptance Time (Months)
    Test Criteria 0 mo. 3 mo.
    Appear- Smooth, white Conforms Conforms
    ance emulsion
    pH Report result 3.3 3.2
    Assay (%) 90.0-110.0% 102.2 101.7
    Related Report ND* ND*
    Substances results
    *Not detected
  • TABLE 20
    Stability Data for 1.0% w/w Cream at 40° C./75% RH
    (15 gram aluminum tubes)
    Acceptance Time (Months)
    Test Criteria 0 mo. 1 mo. 2 mo. 3 mo.
    Appear- Smooth, white Conforms Conforms
    ance emulsion
    pH Report result 3.3 3.2
    API Assay (%) 90.0-110.0% 102.2 103.5 103.8 101.7
    Related Report ND* ND* ND* ND*
    Substances results
    *Not detected
  • Example 4 Solubility Studies
  • In order to determine the solubility of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile (free base) or its phosphate salt, approximately 5 mL of a potential solvent was added to approximately 50 mg of the API or its salt at room temperature. The mixtures were suspended and rotated on a wheel. If the mixtures became clear solutions, more solid material was added. The suspensions were then suspended over 24 hours. The samples were filtered through 0.2 micron filters.
  • The liquid portions were collected and diluted with 50/50 water methanol/water. The concentrations of the diluted samples were analyzed by HPLC. When the free base or salt was fairly insoluble, the results are approximate only.
  • TABLE 21
    Solubility of Phosphate Solubility of Free
    Potential Solvent Salt (mg/mL) Base (mg/mL)
    Water 2.7 2.0
    pH 4, citric buffer, 0.1M 1.5 1.1
    pH 6, citric buffer, 0.1M 0.2 0.15
    Ethanol 7.3 5.5
    Isopropanol 0.6 0.45
    Benzyl alcohol 3 2.3
    Propylene glycol 24 18.2
    PEG 200 23 17.4
    PEG 300 14 10.6
    Glycerin 11 8.3
    Transcutol 10 7.6
    Trolamine 51 38.6
    Water/PEG 200 (50/50) 23 17.4
    Water/glyercin (50/50) 21 15.9
    Water/glycerin/trolamine 18 13.6
    (40/40/20)
    Isopropyl myristate <0.1 0.08
    Isosorbide dimethyl ether 0.4 0.3
    Mineral oil <0.1 0.08
    Olelyl alcohol 0.1 0.08
    Dimethicone <0.2 0.15
    C12-15 alcohol benzoate <0.2 0.15
    Caprylic triglyceride <0.2 0.15
  • Example 5 Other Topical Formulations
  • Three different topical formulations incorporating the phosphate salt of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile were also prepared. The compositions of a 1% w/w dispersed cream (water-in-oil formulation), 1% w/w anhydrous ointment, and 1% w/w lotion are summarized in Table 22 (percentages are on a free base basis). Each of the formulations with 1% w/w of the phosphate salt of the API were lower in viscosity as compared to placebo (in the placebo, the balance is water). While not wishing to be bound by any particular theory, the lower viscosity was believed to be due to electrolytic nature of the phosphate salt. Viscosities of the formulations and placebo over time are shown in Table 23. The 1% dispersed cream (water-in-oil formulation) showed syneresis after two and four weeks of aging at 40° C., while the 1% lotion and 1% solubilized cream formulations (oil-in-water formulations) did not show syneresis. The 1% solubilized cream formulation was generally higher in viscosity than the 1% lotion.
  • TABLE 22
    1% w/w
    1% w/w dispersed 1% w/w
    Ingredient lotion cream ointment
    Purified water USP 52.03 39.48
    Polyethylene glycol 200 USP 7.00
    Example 2* 1.32 1.32 1.32
    Disodium EDTA USP 0.05 0.50
    Phenoxyethanol BP 0.50 0.50
    Propylene glycol USP 15.00 7.50
    Xanthan Gum NF 0.20
    Methylparaben NF 0.10 0.10
    Propylparaben NF 0.05 0.05
    Light mineral oil NF 4.00 6.00
    Glyceryl stearate SE FDA IIG 2.00
    Polysorbate 20 NF 1.00
    White Petrolatum USP 7.00 5.00 78.68
    Cetyl Alcohol NF 2.50
    Stearyl Alcohol NF 1.25
    Dimethicone NF 1.00 1.00
    Caprilic/capric triglycerides FDA-IIG 5.00 6.00
    Sodium Chloride 0.05
    Glycerin 99% USP 7.50
    Sorbitol solution 70% USP 5.00
    White Wax NF 1.50
    Hydrogenated castor oil NF 1.50
    Cyclomethicone NF 12.00
    Polyglycery1-3-diisostearate NF/BP 5.00
    Cyclomethicone (D5) NF 15.00
    Paraffin NF 5.00
    Total
    *1.32% of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate salt is 1% of the free base.
  • TABLE 23
    Aging Viscosity Spindle/rpm
    Type of 1% w/w 1% w/w
    Formulation Time Temp. API Placebo API Placebo
    Solubilized Initial 99,400 195,600 T-B/2.0 T-C/2.5
    cream*
    2 weeks RT 67,625  80,125 27/2.0 27/2.0
    4 weeks RT 65,875  82,750 27/2.0 27/2.0
    2 weeks  5° C. 73,125  55,250 27/2.0 27/2.0
    4 weeks  5° C. 86,000  70,125 27/2.0 27/2.0
    2 weeks 40° C. 46,375  41,875 27/2.0 27/2.0
    4 weeks 40° C. 47,500  50,125 27/2.0 27/2.0
    Lotion* Initial 24,700  70,500 T-A/4.0 27/2.0
    2 weeks RT 28,875  79,250 27/2.0 27/2.0
    4 weeks RT 32,750  73,875 27/2.0 27/2.0
    2 weeks  5° C. 31,750  70,250 27/2.0 27/2.0
    4 weeks  5° C. 34,750  75,750 27/2.0 27/2.0
    2 weeks 40° C. 28,250  44,250 27/2.0 27/2.0
    4 weeks 40° C. 29,125  53,000 27/2.0 27/2.0
    Dispersed Initial 11,400 255,500 27/5.0 28/1.0
    cream
    2 weeks RT  8,850 204,500 27/5.0 28/1.0
    4 weeks RT 12,200 208,500 27/5.0 28/1.0
    2 weeks  5° C.  9,550 226,000 27/5.0 28/1.0
    4 weeks  5° C. 11,200 238,500 27/5.0 28/1.0
    2 weeks 40° C. Syneresis 185,500 27/5.0 28/1.0
    4 weeks 40° C. Syneresis 185,000 27/5.0 28/1.0
    *No syneresis observed
  • Example 6 Skin Permeation Studies
  • The three different topical formulations in Example 5 (Table 20) and the cream formulation in Example 3 (Table 4) were evaluated for transport across human cadaver skin. The skin permeation data are summarized in Table 24. Significant variability was observed in the transport among the three replicates for each formulation. The variability in transport may be due in part to differences in skin samples (donor, region of the body, thickness, etc.). In general, the two cream formulations showed higher flux compared to the lotion or ointment. The cumulative amount of API transported for the ointment formulation was particularly low in comparison to the other three formulations and this, at least in part, could be due to poor spreadability of the ointment leading to decreased surface area for transport. As a result, the two cream formulations were selected for further development, one as an oil-in-water (see Example 3 above) and the other as a water-in-oil emulsion base. Based on the solubility of the drug substance, strengths containing 1.0, 1.5, and 2.0% w/w of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate salt were developed for the oil-in water base cream (solubilized cream) and 1.0, 2.0, and 3.0% w/w were developed for the water-in oil base cream (dispersed cream). Procedures for the skin permeation studies are described below.
  • Human Cadaver Skin Transport Studies
  • The permeability of the API in topical formulations was studied using cadaver human skin samples and Franz diffusion cells. Dermatomed human cadaver skin was obtained from tissue banks while the Franz diffusion cells were custom made. The human cadaver skin samples, sized to fit between the donor and the receiver compartments, were positioned on the Franz diffusion cells. Topical formulations were weighed (20 mg) onto glassine paper, placed formulation side toward the skin and clamped into place. The dosing chamber was covered with parafilm. The reservoir side was filled using saline with 4% albumin. The reservoir was stirred and maintained at 37° C. using a dry block heater (Aungst B. Fatty Acid Skin Penetration Enhancers. Pharm. Res. 1989; 6(3):244-247). At 4 hours, a 1 mL sample was removed and replaced with 1 mL of saline +4% albumin. At 24 hours, the entire reservoir was collected. The tissue was examined visually for any hole or tear. The reservoir side samples were analyzed for concentrations of the API by a LC/MS assay.
  • Mouse Skin Transport Studies
  • The permeability of the API in topical formulations was studied using freshly excised mouse skin samples mounted in Franz diffusion cells. Balb/c mice were depilated using a waxing technique four days before the experiment. The morning of the experiment the mice were euthanized and as much of the depilated skin as possible was removed, rinsed and kept moist with 37° C. saline until use. The mouse skin samples, sized to fit between the donor and the receiver compartments, were positioned between the donor and the receiver compartments of the Franz diffusion cells. The opening of the Franz cell was 1 cm2. Topical formulations were weighed (20 mg) on to glassine paper, placed formulation side toward the skin and clamped into place. The dosing chamber was covered with parafilm. The reservoir side was filled using saline with 4% albumin The reservoir was stirred and maintained at 37° C. using a dry block heater (Aungst 1989 (above). At 4 hours, a 1 mL sample was removed and replaced with 1 mL of saline +4% albumin. At 24 hours, the entire reservoir was collected. The tissue was examined visually for any hole or tear. The reservoir side samples were analyzed for concentrations of the API by a LC/MS assay.
  • TABLE 24
    Transport of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-
    yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile from
    Topical Formulations across Human Cadaver Skin
    Average
    Cumulative cumulative
    Strength, Type Human Cadaver amount over amount at
    of Formulation Skin Sample 24 h (μg) 24 h (μg)
    1% w/w ABS #0510038 0.77 5.16
    Dispersed Cream Asterand #52214A1 10.8
    (see Example 5, Asterand #46581A1 3.91
    Table 20, above)
    1% w/w Solubilized ABS #0510038 0.21 3.73
    Cream Asterand #52214A1 10.6
    (see Example 3, Asterand #46581A1 0.39
    Table 4, above)
    1% w/w Ointment ABS #0510038 0.06 0.06
    (Anhydrous) Asterand #52214A1 0.07
    (see Example 5, Asterand #46581A1 0.07
    Table 20, above)
    1% w/w Lotion ABS #0510038 0.10 0.83
    (see Example 5, Asterand #52214A1 1.96
    Table 20, above) Asterand #46581A1 0.42
  • The effect of strength of solubilized or dispersed cream formulation on the transport of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol- 1-yl)-3-cyclopentylpropanenitrile across human cadaver skin was also examined and the data are summarized in Table 25. Increases in strength from 1% w/w to 3% w/w of the dispersed cream formulation (water-in-oil base) and 1% w/w to 2% w/w of the solubilized cream formulation (oil-in-water base) did not result in any significant change in transport of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1H-pyrazol- 1-yl)-3-cyclopentylpropanenitrile, suggesting that the flux of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile is not limited by the rate of release from each of these formulations.
  • TABLE 25
    Transport of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-
    pyrazol-1-yl)-3-cyclopentylpropanenitrile from Increasing
    Strength Topical Formulations across Human Cadaver Skin
    Cumulative Average
    Amount Cumulative
    Strength, Type Human Cadaver over Amount at
    of Formulation Skin Sample 24 h (μg) 24 h (μg)
    1% w/w ABS #0510038 1.26 2.29
    Dispersed Cream Asterand #42996A1 3.31
    (water-in-oil base)
    2% w/w ABS #0510038 1.79 1.68
    Dispersed Cream Asterand #42996A1 1.56
    (water-in-oil base)
    3% w/w ABS #0510038 1.40 1.81
    Dispersed Cream Asterand #42996A1 2.23
    (water-in-oil base)
    1% w/w ABS #0510038 0.17 0.89
    Solubilized Asterand #42996A1 1.62
    Cream (see Ex-
    ample 3 above)
    1.5% w/w ABS #0510038 0.21 0.30
    Solubilized Asterand #42996A1 0.39
    Cream (see Ex-
    ample 3 above)
    2% w/w Solu- ABS #0510038 0.24 0.25
    bilized Cream Asterand #42996A1 0.26
  • The transport of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1H-pyrazol- 1-yl)-3-cyclopentylpropanenitrile across freshly excised mouse skin was also evaluated using formulations that were employed in rodent pharmacology studies (Table 26). There was a general trend of increased permeability when the strength of the solubilized cream was increased from 0.5 to 1.5%, while such a trend was not seen with the dispersed formulation. For the solubilized cream, the average cumulative amount of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile transported across mouse skin over 24 h was about twenty times higher than that seen with human cadaver skin studies (cumulative average of all experiments).
  • Based on the solubility of (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate, a maximum drug loading of 1.5% was possible with the oil-in-water (solubilized cream) formulation. Of the two creams formulated, the oil in water (solubilized cream) product exhibited better physical stability (see Table 21 above). It should be noted that strengths higher than 3% in the dispersed cream formulation and 2% in the solubilized cream formulation were not physically stable beyond several days of storage at controlled room temperature, as the drug substance crystallized out of solution. Based on these findings, coupled with skin permeability results, manufacturability data, and physical and chemical characterization data obtained for the early stage formulations, a solubilized cream with an oil-in water emulsion base (with a maximum strength of 1.5% w/w) was chosen for further development.
  • TABLE 26
    Transport of Various Formulations of (R)-
    3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-
    pyrazol-1-yl)-3-cyclopentylpropanenitrile
    across Freshly Excised Mouse Skin
    Cumulative Average
    Amount Cumulative
    Strength, over 24 amount at
    Formulation h (μg) 24 h (μg)
    1% w/w dispersed cream 37.1 42.0
    (water-in-oil base) 46.9
    1% w/w dispersed cream 18.0 23.1
    (water-in-oil base) 28.2
    3% w/w dispersed cream 29.6 29.8
    (water-in-oil base) 30.0
    0.5% w/w solubulized cream 26.5 23.5
    (see Example 3 above) 20.4
    1% w/w solubulized cream 40.8 32.8
    (see Example 3 above) 24.9
    1.5% w/w solubulized cream 44.6 41.8
    (see Example 3 above) 38.9
  • Example 7 Clinical Treatment of Psoriasis with Formulations
  • Approximately 200 subjects with chronic plaque psoriasis were enrolled in a double-blind, placebo-controlled study. There were four dose groups, three active treatment groups and vehicle. The active treatment groups were treated with the 0.5%, 1.0%, and 1.5% w/w oil-in-water formulations (see Example 3 supra). Approximately 50 subjects were randomized into each treatment group. A thin layer of cream was applied once per day to up to 20% body surface area of plaque psoriasis. Treatment was applied for 84 days and efficacy measured by the change in total lesion score, a measurement scale which assesses the amount of erythema, scaling and thickness of the plaques (FIG. 2). 25% of patents randomized to 1% w/w or 1.5% w/w of the API had lesions that were clear or almost clear at week 12, versus 6% on vehicle.
  • At a subset of sites, photos were obtained from subjects who signed an informed consent for the photos. Pictures were obtained at baseline (prior to the first application of study treatment) and on day 84 (the last application day for study treatment) (see FIG. 3-7). These photos are representative of a subset of the subjects who were treated with the oil-in-water formulations.
  • Example 8 Murine Skin Contact Delayed Hypersensitivity Response Test
  • The formulations described herein can also be tested for their efficacies (of inhibiting JAK targets) in the T-cell driven murine delayed hypersensitivity test model. The murine skin contact delayed-type hypersensitivity (DTH) response is considered to be a valid model of clinical contact dermatitis, and other T-lymphocyte mediated immune disorders of the skin, such as psoriasis (Immunol Today. 1998 January; 19(1):37-44). Murine DTH shares multiple characteristics with psoriasis, including the immune infiltrate, the accompanying increase in inflammatory cytokines, and keratinocyte hyperproliferation. Furthermore, many classes of agents that are efficacious in treating psoriasis in the clinic are also effective inhibitors of the DTH response in mice (Agents Actions. 1993 January; 38(1-2):116-21).
  • On Day 0 and 1, Balb/c mice are sensitized with a topical application, to their shaved abdomen with the antigen 2,4,dinitro-fluorobenzene (DNFB). On day 5, ears are measured for thickness using an engineer's micrometer. This measurement is recorded and used as a baseline. Both of the animals' ears are then challenged by a topical application of DNFB in a total of 20 μL (10 μL on the internal pinna and 10 μL on the external pinna) at a concentration of 0.2%. Twenty-four to seventy-two hours after the challenge, ears are measured again. Treatment with the test formulations is given throughout the sensitization and challenge phases (day −1 to day 7) or prior to and throughout the challenge phase (usually afternoon of day 4 to day 7). Treatment of the test compounds (in different concentration) is administered topically (topical application of the treatment to the ears). Efficacies of the test formulations are indicated by a reduction in ear swelling comparing to the situation without the treatment. Compounds causing a reduction of 20% or more are considered efficacious. In some experiments, the mice are challenged but not sensitized (negative control).
  • The inhibitive effect (inhibiting activation of the JAK-STAT pathways) of the test formulations can be confirmed by immunohistochemical analysis. Activation of the JAK-STAT pathway(s) results in the formation and translocation of functional transcription factors. Further, the influx of immune cells and the increased proliferation of keratinocytes should also provide unique expression profile changes in the ear that can be investigated and quantified. Formalin fixed and paraffin embedded ear sections (harvested after the challenge phase in the DTH model) are subjected to immunohistochemical analysis using an antibody that specifically interacts with phosphorylated STAT3 (clone 58E12, Cell Signaling Technologies). The mouse ears are treated with test formulations, vehicle, or dexamethasone (a clinically efficacious treatment for psoriasis), or without any treatment, in the DTH model for comparisons. Test formulations and the dexamethasone can produce similar transcriptional changes both qualitatively and quantitatively, and both the test formulations and dexamethasone can reduce the number of infiltrating cells. Topical administration of the test compounds can produce inhibitive effects, i.e., reduction in the number of infiltrating cells and inhibition of the transcriptional changes.
  • Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in the present application is incorporated herein by reference in its entirety.

Claims (66)

1. A pharmaceutical formulation for topical skin application, comprising:
an oil-in-water emulsion; and
a therapeutically effective amount of a therapeutic agent which is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof
2. A pharmaceutical formulation according to claim 1, wherein the emulsion comprises water, an oil component, and an emulsifier component.
3-4. (canceled)
5. A pharmaceutical formulation according to claim 2, wherein the oil component is present in an amount of about 20% to about 27% by weight of the formulation.
6. (canceled)
7. A pharmaceutical formulation according to claim 2, wherein the oil component comprises one or more substances independently selected from white petrolatum, cetyl alcohol, stearyl alcohol, light mineral oil, medium chain triglycerides, and dimethicone.
8. A pharmaceutical formulation according to claim 2, wherein the oil component comprises an occlusive agent component.
9. (canceled)
10. A pharmaceutical formulation according to claim 8, wherein the occlusive agent component is present in an amount of about 5% to about 10% by weight of the formulation.
11. (canceled)
12. A pharmaceutical formulation according to claim 8, wherein the occlusive agent component comprises white petrolatum.
13. A pharmaceutical formulation according to claim 2, wherein the oil component comprises a stiffening agent component.
14-15. (canceled)
16. A pharmaceutical formulation according to claim 13, wherein the stiffening agent component is present in an amount of about 4% to about 7% by weight of said formulation.
17-18. (canceled)
19. A pharmaceutical formulation according to claim 13, wherein the stiffening agent component comprises one or more substances independently selected from C16-18 fatty alcohols.
20. (canceled)
21. A pharmaceutical formulation according to claim 2, wherein the oil component comprises an emollient component.
22. (canceled)
23. A pharmaceutical formulation according to claim 21, wherein the emollient component is present in an amount of about 7% to about 13% by weight of the formulation.
24-25. (canceled)
26. A pharmaceutical formulation according to claim 21, wherein the emollient component comprises one or more substances independently selected from light mineral oil, medium chain triglycerides, and dimethicone.
27-28. (canceled)
29. A pharmaceutical formulation according to claim 2, wherein the water is present in an amount of about 45% to about 55% by weight of the formulation.
30-32. (canceled)
33. A pharmaceutical formulation according to claim 2, wherein the emulsifier component is present in an amount of about 4% to about 7% by weight of the formulation.
34. (canceled)
35. A pharmaceutical formulation according to claim 2, wherein the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters.
36. (canceled)
37. A pharmaceutical formulation according to claim 1, wherein said pharmaceutical formulation further comprises a stabilizing agent component.
38-39. (canceled)
40. A pharmaceutical formulation according to claim 37, wherein the stabilizing agent component is present in an amount of about 0.3 to about 0.5% by weight of the formulation.
41. (canceled)
42. A pharmaceutical formulation according to claim 37, wherein the stabilizing agent component comprises xanthan gum.
43. A pharmaceutical formulation according to claim 1, wherein said pharmaceutical formulation further comprises a solvent component.
44-45. (canceled)
46. A pharmaceutical formulation according to claim 43, wherein the solvent component is present in an amount of about 20% to about 25% by weight of the formulation.
47. A pharmaceutical formulation according to claim 43, wherein the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols.
48. (canceled)
49. A pharmaceutical formulation according to claim 1, wherein the therapeutic agent is present in an amount of about 0.5% to about 1.5% by weight of the formulation on a free base basis.
50. A pharmaceutical formulation according to claim 1, wherein said therapeutic agent is present in an amount of about 0.5% by weight of the formulation on a free base basis.
51. A pharmaceutical formulation according to claim 1, wherein said therapeutic agent is present in an amount of about 1% by weight of the formulation on a free base basis.
52. A pharmaceutical formulation according to claim 1, wherein said therapeutic agent is present in an amount of about 1.5% by weight of the formulation on a free base basis.
53. A pharmaceutical formulation according to claim 1, wherein said therapeutic agent is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile phosphoric acid salt.
54. A pharmaceutical formulation according to claim 1, comprising:
from about 35% to about 65% of water by weight of the formulation;
from about 10% to about 40% of an oil component by weight of the formulation;
from about 1% to about 9% of an emulsifier component by weight of the formulation;
from about 10% to about 35% of a solvent component by weight of the formulation;
from about 0.05% to about 5% of a stabilizing agent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
55. A pharmaceutical formulation according to claim 1, comprising:
from about 40% to about 60% of water by weight of the formulation;
from about 15% to about 30% of an oil component by weight of the formulation;
from about 2% to about 6% of an emulsifier component by weight of the formulation;
from about 15% to about 30% of a solvent component by weight of the formulation;
from about 0.1% to about 2% of a stabilizing agent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
56. A pharmaceutical formulation according to claim 1, comprising:
from about 45% to about 55% of water by weight of the formulation;
from about 17% to about 27% of an oil component by weight of the formulation;
from about 3% to about 5% of an emulsifier component by weight of the formulation;
from about 20% to about 25% of a solvent component by weight of the formulation;
from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
57. A pharmaceutical formulation according to claim 1, comprising:
from about 45% to about 55% of water by weight of the formulation;
from about 17% to about 27% of an oil component by weight of the formulation;
from about 4% to about 7% of an emulsifier component by weight of the formulation;
from about 20% to about 25% of a solvent component by weight of the formulation;
from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
58. A pharmaceutical formulation according to claim 54, wherein:
the oil component comprises one or more substances independently selected from petrolatums, fatty alcohols, mineral oils, triglycerides, and dimethicones;
the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters;
the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols; and
the stabilizing agent component comprises one or more substances independently selected from polysaccharides.
59. A pharmaceutical formulation according to claim 54, wherein:
the oil component comprises one or more substances independently selected from white petrolatum, cetyl alcohol, stearyl alcohol, light mineral oil, medium chain triglycerides, and dimethicone;
the emulsifier component comprises one or more substances independently selected from glyceryl stearate and polysorbate 20;
the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol; and
the stabilizing agent component comprises xanthan gum.
60. A pharmaceutical formulation according to claim 1, comprising:
from about 35% to about 65% of water by weight of the formulation;
from about 2% to about 15% of an occlusive agent component by weight of the formulation;
from about 2% to about 8% of a stiffening agent component by weight of said formulation;
from about 5% to about 15% of an emollient component by weight of the formulation;
from about 1% to about 9% of an emulsifier component by weight of the formulation;
from about 0.05% to about 5% of a stabilizing agent component by weight of the formulation;
from about 10% to about 35% of a solvent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
61. A pharmaceutical formulation according to claim 1, comprising:
from about 40% to about 60% of water by weight of the formulation;
from about 5% to about 10% of an occlusive agent component by weight of the formulation;
from about 2% to about 8% of a stiffening agent component by weight of said formulation;
from about 7% to about 12% of an emollient component by weight of the formulation;
from about 2% to about 6% of an emulsifier component by weight of the formulation;
from about 0.1% to about 2% of a stabilizing agent by weight of the formulation;
from about 15% to about 30% of a solvent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
62. A pharmaceutical formulation according to claim 1, comprising:
from about 45% to about 55% of water by weight of the formulation;
from about 5% to about 10% of an occlusive agent component by weight of the formulation;
from about 3% to about 6% of a stiffening agent component by weight of said formulation;
from about 7% to about 13% of an emollient component by weight of the formulation;
from about 3% to about 5% of an emulsifier component by weight of the formulation;
from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation;
from about 20% to about 25% of a solvent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
63. A pharmaceutical formulation according to claim 1, comprising:
from about 45% to about 55% of water by weight of the formulation;
from about 5% to about 10% of an occlusive agent component by weight of the formulation;
from about 4% to about 7% of a stiffening agent component by weight of said formulation;
from about 7% to about 13% of an emollient component by weight of the formulation;
from about 4% to about 7% of an emulsifier component by weight of the formulation;
from about 0.3% to about 0.5% of a stabilizing agent component by weight of the formulation;
from about 20% to about 25% of a solvent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
64. A pharmaceutical formulation according to claim 1, comprising:
from about 45% to about 55% of water by weight of the formulation;
about 7% of an occlusive agent component by weight of the formulation;
from about 4.5% to about 5% of a stiffening agent component by weight of said formulation;
about 10% of an emollient component by weight of the formulation;
from about 4% to about 4.5% of an emulsifier component by weight of the formulation;
about 0.4% of a stabilizing agent component by weight of the formulation;
about 22% of a solvent component by weight of the formulation; and
from about 0.5% to about 1.5% of (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, or a pharmaceutically acceptable salt thereof, by weight of the formulation on a free base basis.
65. A pharmaceutical formulation according to claim 60, wherein the combined amount of the stiffening agent component and the emulsifier component is at least about 8% by weight of the formulation.
66. A pharmaceutical formulation according to claim 60, wherein:
the occlusive agent component comprises a petrolatum;
the stiffening agent component comprises one or more substances independently selected from one or more fatty alcohols;
the emollient component comprises one or more substances independently selected from mineral oils and triglycerides;
the emulsifier component comprises one or more substances independently selected from glyceryl fatty esters and sorbitan fatty esters;
the stabilizing agent component comprises one or more substances independently selected from polysaccharides; and
the solvent component comprises one or more substances independently selected from alkylene glycols and polyalkylene glycols.
67. A pharmaceutical formulation according to claim 60, wherein:
the occlusive agent component comprises white petrolatum;
the stiffening agent component comprises one or more substances independently selected from cetyl alcohol and stearyl alcohol;
the emollient component comprises one or more substances independently selected from light mineral oil, medium chain triglycerides, and dimethicone;
the emulsifier component comprises one or more substances independently selected from glyceryl stearate and polysorbate 20;
the stabilizing agent component comprises xanthan gum; and
the solvent component comprises one or more substances independently selected from propylene glycol and polyethylene glycol.
68-74. (canceled)
75. A method of treating a skin disorder in a patient in need thereof, comprising applying a pharmaceutical formulation according to claim 1 to an area of skin of said patient.
76. The method of claim 75, wherein the skin disorder is a topic dermatitis or psoriasis.
77. The method of claim 75, wherein the skin disorder is psoriasis.
78. The method of claim 75, wherein said skin disorder is skin sensitization, skin irritation, skin rash, contact dermatitis or allergic contact sensitization.
79. The method of claim 75, wherein said skin disorder is a bullous skin disorder.
80. The method of claim 79, wherein said bullous skin disorder is pemphigus vulgaris (PV) or bullous pemphigoid (BP).
81-82. (canceled)
US13/112,370 2010-05-21 2011-05-20 Topical formulation for a jak inhibitor Abandoned US20110288107A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US13/112,370 US20110288107A1 (en) 2010-05-21 2011-05-20 Topical formulation for a jak inhibitor
US14/714,820 US20150250790A1 (en) 2010-05-21 2015-05-18 Topical formulation for a jak inhibitor
US16/566,625 US10758543B2 (en) 2010-05-21 2019-09-10 Topical formulation for a JAK inhibitor
US16/947,735 US10869870B2 (en) 2010-05-21 2020-08-14 Topical formulation for a JAK inhibitor
US16/948,408 US11219624B2 (en) 2010-05-21 2020-09-17 Topical formulation for a JAK inhibitor
US17/541,439 US20220211707A1 (en) 2010-05-21 2021-12-03 Topical formulation for a jak inhibitor
US17/704,180 US11590136B2 (en) 2010-05-21 2022-03-25 Topical formulation for a JAK inhibitor
US17/704,168 US11571425B2 (en) 2010-05-21 2022-03-25 Topical formulation for a JAK inhibitor
US17/704,155 US20220370455A1 (en) 2010-05-21 2022-03-25 Topical formulation for a jak inhibitor
US18/089,651 US20230277541A1 (en) 2010-05-21 2022-12-28 Topical formulation for a jak inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US34713210P 2010-05-21 2010-05-21
US13/112,370 US20110288107A1 (en) 2010-05-21 2011-05-20 Topical formulation for a jak inhibitor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/714,820 Continuation US20150250790A1 (en) 2010-05-21 2015-05-18 Topical formulation for a jak inhibitor

Publications (1)

Publication Number Publication Date
US20110288107A1 true US20110288107A1 (en) 2011-11-24

Family

ID=44201091

Family Applications (10)

Application Number Title Priority Date Filing Date
US13/112,370 Abandoned US20110288107A1 (en) 2010-05-21 2011-05-20 Topical formulation for a jak inhibitor
US14/714,820 Abandoned US20150250790A1 (en) 2010-05-21 2015-05-18 Topical formulation for a jak inhibitor
US16/566,625 Active US10758543B2 (en) 2010-05-21 2019-09-10 Topical formulation for a JAK inhibitor
US16/947,735 Active US10869870B2 (en) 2010-05-21 2020-08-14 Topical formulation for a JAK inhibitor
US16/948,408 Active US11219624B2 (en) 2010-05-21 2020-09-17 Topical formulation for a JAK inhibitor
US17/541,439 Pending US20220211707A1 (en) 2010-05-21 2021-12-03 Topical formulation for a jak inhibitor
US17/704,155 Pending US20220370455A1 (en) 2010-05-21 2022-03-25 Topical formulation for a jak inhibitor
US17/704,180 Active US11590136B2 (en) 2010-05-21 2022-03-25 Topical formulation for a JAK inhibitor
US17/704,168 Active US11571425B2 (en) 2010-05-21 2022-03-25 Topical formulation for a JAK inhibitor
US18/089,651 Pending US20230277541A1 (en) 2010-05-21 2022-12-28 Topical formulation for a jak inhibitor

Family Applications After (9)

Application Number Title Priority Date Filing Date
US14/714,820 Abandoned US20150250790A1 (en) 2010-05-21 2015-05-18 Topical formulation for a jak inhibitor
US16/566,625 Active US10758543B2 (en) 2010-05-21 2019-09-10 Topical formulation for a JAK inhibitor
US16/947,735 Active US10869870B2 (en) 2010-05-21 2020-08-14 Topical formulation for a JAK inhibitor
US16/948,408 Active US11219624B2 (en) 2010-05-21 2020-09-17 Topical formulation for a JAK inhibitor
US17/541,439 Pending US20220211707A1 (en) 2010-05-21 2021-12-03 Topical formulation for a jak inhibitor
US17/704,155 Pending US20220370455A1 (en) 2010-05-21 2022-03-25 Topical formulation for a jak inhibitor
US17/704,180 Active US11590136B2 (en) 2010-05-21 2022-03-25 Topical formulation for a JAK inhibitor
US17/704,168 Active US11571425B2 (en) 2010-05-21 2022-03-25 Topical formulation for a JAK inhibitor
US18/089,651 Pending US20230277541A1 (en) 2010-05-21 2022-12-28 Topical formulation for a jak inhibitor

Country Status (35)

Country Link
US (10) US20110288107A1 (en)
EP (2) EP2574168B9 (en)
JP (7) JP5849312B2 (en)
KR (5) KR102303885B1 (en)
CN (2) CN105853356B (en)
AR (2) AR084691A1 (en)
AU (6) AU2011255443B2 (en)
BR (1) BR112012029653B1 (en)
CA (1) CA2799928C (en)
CL (1) CL2012003229A1 (en)
CO (1) CO6640250A2 (en)
CR (1) CR20120605A (en)
CY (1) CY1117815T1 (en)
DK (1) DK2574168T3 (en)
EA (2) EA035981B1 (en)
EC (1) ECSP13012546A (en)
ES (1) ES2581834T3 (en)
HK (1) HK1182313A1 (en)
HR (1) HRP20160841T1 (en)
HU (1) HUE029035T2 (en)
IL (1) IL223084A (en)
ME (1) ME02445B (en)
MX (1) MX338228B (en)
MY (2) MY161078A (en)
NZ (1) NZ603686A (en)
PE (1) PE20130216A1 (en)
PL (1) PL2574168T3 (en)
RS (1) RS54824B1 (en)
SG (3) SG10201503983QA (en)
SI (1) SI2574168T1 (en)
SM (1) SMT201600172B (en)
TW (1) TWI499421B (en)
UA (1) UA111588C2 (en)
WO (1) WO2011146808A2 (en)
ZA (1) ZA202001999B (en)

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8415362B2 (en) 2005-12-13 2013-04-09 Incyte Corporation Pyrazolyl substituted pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US8604043B2 (en) 2009-05-22 2013-12-10 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as jak inhibitors
US8691807B2 (en) 2011-06-20 2014-04-08 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US8716303B2 (en) 2009-05-22 2014-05-06 Incyte Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8765734B2 (en) 2010-03-10 2014-07-01 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US8883806B2 (en) 2009-01-15 2014-11-11 Incyte Corporation Processes for preparing JAK inhibitors and related intermediate compounds
US8933085B2 (en) 2010-11-19 2015-01-13 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US8987443B2 (en) 2013-03-06 2015-03-24 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
US9181271B2 (en) 2012-11-01 2015-11-10 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9359358B2 (en) 2011-08-18 2016-06-07 Incyte Holdings Corporation Cyclohexyl azetidine derivatives as JAK inhibitors
US9358229B2 (en) 2011-08-10 2016-06-07 Novartis Pharma Ag JAK PI3K/mTOR combination therapy
US9382231B2 (en) 2013-05-17 2016-07-05 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9802957B2 (en) 2014-04-30 2017-10-31 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
US9993480B2 (en) 2011-02-18 2018-06-12 Novartis Pharma Ag mTOR/JAK inhibitor combination therapy
US10064866B2 (en) 2014-04-08 2018-09-04 Incyte Corporation Treatment of B-cell malignancies by a combination JAK and PI3K inhibitors
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10463667B2 (en) 2007-06-13 2019-11-05 Incyte Incorporation Metabolites of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US11103510B2 (en) 2018-02-16 2021-08-31 Incyte Corporation JAK1 pathway inhibitors for the treatment of cytokine-related disorders
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11324749B2 (en) 2018-10-31 2022-05-10 Incyte Corporation Combination therapy for treatment of hematological diseases
WO2022125670A1 (en) 2020-12-08 2022-06-16 Incyte Corporation Jak1 pathway inhibitors for the treatment of vitiligo
US11372003B2 (en) 2018-04-13 2022-06-28 Incyte Corporation Biomarkers for graft-versus-host disease
US11584961B2 (en) 2018-03-30 2023-02-21 Incyte Corporation Biomarkers for inflammatory skin disease
US11685731B2 (en) 2020-06-02 2023-06-27 Incyte Corporation Processes of preparing a JAK1 inhibitor
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140343034A1 (en) * 2013-04-25 2014-11-20 Japan Tobacco Inc. Skin barrier function improving agent
SG10201900648SA (en) * 2014-07-25 2019-02-27 Novartis Ag Tablet formulation of 2-fluoro-n-methyl-4-[7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide
CZ2014773A3 (en) * 2014-11-10 2016-05-18 Zentiva, K.S. Salts of (3R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]propanenitrile
CZ201629A3 (en) * 2016-01-22 2017-08-02 Zentiva, K.S. Crystalline modifications of the (3R)-3-cyclopentyl-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4yl) pyrazol-1yl] propanenitrile salts and the method of their preparation
CN110913862B (en) * 2017-11-20 2022-12-30 江苏恒瑞医药股份有限公司 A topical pharmaceutical composition and its preparation method
US10370361B2 (en) 2017-11-21 2019-08-06 Denali Therapeutics Inc. Polymorphs and solid forms of a pyrimidinylamino-pyrazole compound, and methods of production
BR112020012611A2 (en) 2017-12-20 2020-12-08 Denali Therapeutics Inc. PROCESS FOR THE PREPARATION OF PYRIMIDINYL-4-AMINOPYRAZOLE COMPOUNDS
CN111818910A (en) * 2018-01-09 2020-10-23 德玛万科学有限公司 Topical skin pharmaceutical composition containing cerdulatinib and application thereof
JP6830460B2 (en) * 2018-07-05 2021-02-17 コンサート ファーマシューティカルズ インコーポレイテッド Deuterated derivative of ruxolitinib
CA3150975A1 (en) 2019-06-10 2020-12-17 Incyte Corporation Topical treatment of vitiligo by a jak inhibitor
WO2021046350A1 (en) 2019-09-05 2021-03-11 Incyte Corporation Ruxolitinib formulation for reduction of itch in atopic dermatitis
US11738026B2 (en) 2019-11-22 2023-08-29 Incyte Corporation Combination therapy comprising an ALK2 inhibitor and a JAK2 inhibitor
AU2021308340A1 (en) * 2020-07-17 2023-02-23 Pfizer Inc. Stable pharmaceutical topical formulation containing immunosuppressant for treating dermatological conditions
US11905292B2 (en) 2020-08-18 2024-02-20 Incyte Corporation Process and intermediates for preparing a JAK inhibitor
US11897889B2 (en) 2020-08-18 2024-02-13 Incyte Corporation Process and intermediates for preparing a JAK1 inhibitor
CN117440814A (en) 2020-10-02 2024-01-23 因赛特公司 External ruxotinib for treating lichen planus
US20220202834A1 (en) 2020-12-04 2022-06-30 Incyte Corporation Jak inhibitor with a vitamin d analog for treatment of skin diseases
US20220347179A1 (en) 2021-05-03 2022-11-03 Incyte Corporation Ruxolitinib for the treatment of prurigo nodularis
CN113264936B (en) * 2021-05-25 2022-08-09 常州制药厂有限公司 JAK inhibitor key intermediate and preparation method thereof
CN115702936A (en) * 2021-08-13 2023-02-17 杭州中美华东制药有限公司 Lucotinib composition and application thereof
WO2023020567A1 (en) * 2021-08-19 2023-02-23 珠海联邦制药股份有限公司 Local topical formulation containing jak inhibitor or salt thereof or crystal form thereof, preparation method therefor and application thereof
CN114870016B (en) * 2022-04-21 2023-05-26 上海博悦生物科技有限公司 Micro-emulsion foaming agent of JAK inhibitor and application thereof
WO2023245053A1 (en) 2022-06-14 2023-12-21 Incyte Corporation Solid forms of a jak inhibitor and process of preparing the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6075056A (en) * 1997-10-03 2000-06-13 Penederm, Inc. Antifungal/steroid topical compositions
WO2009158687A1 (en) * 2008-06-26 2009-12-30 Anterios, Inc. Dermal delivery

Family Cites Families (279)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2985589A (en) 1957-05-22 1961-05-23 Universal Oil Prod Co Continuous sorption process employing fixed bed of sorbent and moving inlets and outlets
US3832460A (en) 1971-03-19 1974-08-27 C Kosti Anesthetic-vasoconstrictor-antihistamine composition for the treatment of hypertrophied oral tissue
US4140755A (en) 1976-02-13 1979-02-20 Hoffmann-La Roche Inc. Sustained release tablet formulations
DE3036390A1 (en) 1980-09-26 1982-05-13 Troponwerke GmbH & Co KG, 5000 Köln Antiinflammatory intermediate 7H-pyrrolo-(2,3-D)-pyrimidine derivs. - prepd. by dealkylation of 7-phenyl:ethyl derivs. by reaction with hydrochloric, phosphoric or poly:phosphoric acid
DE3220113A1 (en) 1982-05-28 1983-12-01 Basf Ag, 6700 Ludwigshafen DIFLUORMETHOXIPHENYLTHIOPHOSPHORSAEUREESTER
US4402832A (en) 1982-08-12 1983-09-06 Uop Inc. High efficiency continuous separation process
US4548990A (en) 1983-08-15 1985-10-22 Ciba-Geigy Corporation Crosslinked, porous polymers for controlled drug delivery
US4498991A (en) 1984-06-18 1985-02-12 Uop Inc. Serial flow continuous separation process
NL8403224A (en) 1984-10-24 1986-05-16 Oce Andeno Bv DIOXAPHOSPHORINANS, THEIR PREPARATION AND THE USE FOR SPLITTING OF OPTICALLY ACTIVE COMPOUNDS.
CA1306260C (en) 1985-10-18 1992-08-11 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
US5702688A (en) * 1986-12-23 1997-12-30 Tristrata Technology, Inc. Amphoteric compositions and polymeric forms of alpha hydroxyacids, and their therapeutic use
KR0155955B1 (en) 1989-10-11 1998-11-16 이따가끼 히로시 Bicyclic pyrimidine derivatives, method of producing the same and pharmaceutical preparation containing the same as active ingredient
IT1258781B (en) 1992-01-16 1996-02-29 Zambon Spa OPHTHALMIC PHARMACEUTICAL COMPOSITION CONTAINING N-ACETYLCISTEIN AND POLYVINYL ALCOHOL
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
AU671491B2 (en) 1992-12-18 1996-08-29 F. Hoffmann-La Roche Ag N-oxycarbonyl substituted 5'-deoxy-5-fluorcytidines
JPH0710876A (en) 1993-06-24 1995-01-13 Teijin Ltd Pyrrolo(2,3-d)pyrimidine having cyclic amino group at 4-position
EP0727217A3 (en) 1995-02-10 1997-01-15 Suntory Ltd Pharmaceutical composition containing god-type ellagitannin as active ingredient
IL117580A0 (en) 1995-03-29 1996-07-23 Merck & Co Inc Inhibitors of farnesyl-protein transferase and pharmaceutical compositions containing them
US5856326A (en) 1995-03-29 1999-01-05 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
JP4010563B2 (en) 1995-07-05 2007-11-21 イー・アイ・デユポン・ドウ・ヌムール・アンド・カンパニー Bactericidal and fungicidal pyrimidinones
US5630943A (en) 1995-11-30 1997-05-20 Merck Patent Gesellschaft Mit Beschrankter Haftung Discontinuous countercurrent chromatographic process and apparatus
GB9604361D0 (en) 1996-02-29 1996-05-01 Pharmacia Spa 4-Substituted pyrrolopyrimidine compounds as tyrosine kinase inhibitors
AU727939B2 (en) 1996-04-03 2001-01-04 Merck & Co., Inc. A method of treating cancer
WO1997038664A2 (en) 1996-04-18 1997-10-23 Merck & Co., Inc. A method of treating cancer
US5795909A (en) 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
WO1997045412A1 (en) 1996-05-30 1997-12-04 Merck & Co., Inc. A method of treating cancer
US6624138B1 (en) 2001-09-27 2003-09-23 Gp Medical Drug-loaded biological material chemically treated with genipin
WO1998044797A1 (en) 1997-04-07 1998-10-15 Merck & Co., Inc. A method of treating cancer
US6060038A (en) 1997-05-15 2000-05-09 Merck & Co., Inc. Radiolabeled farnesyl-protein transferase inhibitors
US6063284A (en) 1997-05-15 2000-05-16 Em Industries, Inc. Single column closed-loop recycling with periodic intra-profile injection
WO1999007379A1 (en) 1997-08-11 1999-02-18 Boehringer Ingelheim Pharmaceuticals, Inc. 5,6-HETEROARYL-DIPYRIDO[2,3-b:3',2'-f]AZEPINES AND THEIR USE IN THE PREVENTION OR TREATMENT OF HIV INFECTION
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6232320B1 (en) 1998-06-04 2001-05-15 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory compounds
SK18542000A3 (en) 1998-06-04 2001-12-03 Abbott Laboratories Cell adhesion-inhibiting anti-inflammatory compounds
PA8474101A1 (en) 1998-06-19 2000-09-29 Pfizer Prod Inc PYROLEUM [2,3-D] PIRIMIDINE COMPOUNDS
CA2339961C (en) 1998-08-11 2009-01-20 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
JP2000119271A (en) 1998-08-12 2000-04-25 Hokuriku Seiyaku Co Ltd 1h-imidazopyridine derivative
DK1109534T3 (en) 1998-09-10 2003-06-10 Nycomed Danmark As Pharmaceuticals with quick release of the active substance
US6413419B1 (en) 1998-10-29 2002-07-02 Institut Francais Du Petrole Process and device for separation with variable-length chromatographic
US6375839B1 (en) 1998-10-29 2002-04-23 Institut Francais Du Petrole Process and device for separation with variable-length chromatographic zones
FR2785196B1 (en) 1998-10-29 2000-12-15 Inst Francais Du Petrole METHOD AND DEVICE FOR SEPARATION WITH VARIABLE LENGTH CHROMATOGRAPHIC AREAS
US6333384B1 (en) 1998-11-02 2001-12-25 Gil Technologies Vinyl-terminated polybutadiene and butadiene-styrene copolymers containing urethane and/or ester residues, and the electrical laminates obtained therefrom
US6133031A (en) 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
JP2002538121A (en) 1999-03-03 2002-11-12 メルク エンド カムパニー インコーポレーテッド Inhibitors of prenyl protein transferase
GB9905075D0 (en) 1999-03-06 1999-04-28 Zeneca Ltd Chemical compounds
US6217895B1 (en) 1999-03-22 2001-04-17 Control Delivery Systems Method for treating and/or preventing retinal diseases with sustained release corticosteroids
US6239113B1 (en) 1999-03-31 2001-05-29 Insite Vision, Incorporated Topical treatment or prevention of ocular infections
WO2000063168A1 (en) 1999-04-16 2000-10-26 Coelacanth Chemical Corporation Synthesis of azetidine derivatives
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
AU766233B2 (en) 1999-10-13 2003-10-09 Banyu Pharmaceutical Co., Ltd. Substituted imidazolidinone derivatives
EP1235830B1 (en) 1999-12-10 2004-01-02 Pfizer Products Inc. PYRROLO[2,3-d]PYRIMIDINE COMPOUNDS AS PROTEIN KINASES INHIBITORS
AP1917A (en) 1999-12-24 2008-11-12 Aventis Pharma Ltd Azaindoles
GB0004890D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
US7235551B2 (en) 2000-03-02 2007-06-26 Smithkline Beecham Corporation 1,5-disubstituted-3,4-dihydro-1h-pyrimido[4,5-d]pyrimidin-2-one compounds and their use in treating csbp/p38 kinase mediated diseases
ATE250924T1 (en) 2000-04-07 2003-10-15 Medidom Lab EYE MEDICINAL CONTAINING CYCLOSPORINE, HYALURONIC ACID AND POLYSORBATES
AU4878601A (en) 2000-04-20 2001-11-07 Mitsubishi Corporation Aromatic amide compounds
ES2567260T3 (en) 2000-04-25 2016-04-21 Icos Corporation Human phosphatidylinositol 3-kinase delta inhibitors
IL153115A0 (en) 2000-06-16 2003-06-24 Curis Inc Angiogenesis-modulating compositions and uses
US7498304B2 (en) 2000-06-16 2009-03-03 Curis, Inc. Angiogenesis-modulating compositions and uses
US6335342B1 (en) 2000-06-19 2002-01-01 Pharmacia & Upjohn S.P.A. Azaindole derivatives, process for their preparation, and their use as antitumor agents
CN100548375C (en) 2000-06-23 2009-10-14 田边三菱制药株式会社 Antitumor effect potentiators
EA006153B1 (en) 2000-06-26 2005-10-27 Пфайзер Продактс Инк. PYRROLO[2,3-d]PYRIMIDINE COMPOUNDS AS IMMUNOSUPPRESSIVE AGENTS
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
AU2002228783A1 (en) 2000-12-05 2002-06-18 Vertex Pharmaceuticals Incorporated Inhibitors of c-jun n-terminal kinases (jnk) and other protein kinases
GB0100622D0 (en) 2001-01-10 2001-02-21 Vernalis Res Ltd Chemical compounds V111
US20040077654A1 (en) 2001-01-15 2004-04-22 Bouillot Anne Marie Jeanne Aryl piperidine and piperazine derivatives as inducers of ldl-receptor expression
EP1363702A4 (en) 2001-01-30 2007-08-22 Cytopia Pty Ltd Methods of inhibiting kinases
WO2002092573A2 (en) 2001-05-16 2002-11-21 Vertex Pharmaceuticals Incorporated Heterocyclic substituted pyrazoles as inhibitors of src and other protein kinases
US7301023B2 (en) 2001-05-31 2007-11-27 Pfizer Inc. Chiral salt resolution
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
GB0115393D0 (en) 2001-06-23 2001-08-15 Aventis Pharma Ltd Chemical compounds
DE60216115T2 (en) 2001-08-01 2007-05-31 Merck & Co., Inc. BENZIMIDAZO 4,5-föISOCHINOLINONE DERIVATIVES
JP4456365B2 (en) 2001-09-19 2010-04-28 アベンティス・ファーマ・ソシエテ・アノニム Compound
US6429231B1 (en) 2001-09-24 2002-08-06 Bradley Pharmaceuticals, Inc. Compositions containing antimicrobials and urea for the treatment of dermatological disorders and methods for their use
DE60213842T2 (en) 2001-10-30 2007-09-06 Novartis Ag STAUROSPORINE DERIVATIVES AS INHIBITORS OF THE FLT3 RECEPTOR TYROSINE KINASE EFFECT
JP2003155285A (en) 2001-11-19 2003-05-27 Toray Ind Inc Cyclic nitrogen-containing derivative
CA2468942A1 (en) 2001-11-30 2003-06-12 Teijin Limited Process for producing 5-(3-cyanophenyl)-3-formylbenzoic acid compound
GT200200234A (en) 2001-12-06 2003-06-27 NEW CRYSTAL COMPOUNDS
US6995144B2 (en) 2002-03-14 2006-02-07 Eisai Co., Ltd. Nitrogen containing heterocyclic compounds and medicines containing the same
TW200403058A (en) 2002-04-19 2004-03-01 Bristol Myers Squibb Co Heterocyclo inhibitors of potassium channel function
AU2003237121A1 (en) 2002-04-26 2003-11-10 Vertex Pharmaceuticals Incorporated Pyrrole derivatives as inhibitors of erk2 and uses thereof
JP2005530745A (en) 2002-05-02 2005-10-13 メルク エンド カムパニー インコーポレーテッド Tyrosine kinase inhibitor
JP2005532313A (en) 2002-05-07 2005-10-27 コントロール・デリバリー・システムズ・インコーポレイテッド Method for forming drug delivery device
CA2486183C (en) 2002-05-23 2012-01-10 Cytopia Pty Ltd. Protein kinase inhibitors
TW200406374A (en) 2002-05-29 2004-05-01 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
EP1535934A4 (en) 2002-06-26 2005-11-02 Idemitsu Kosan Co Hydrogenated copolymer, process for producing the same, and hot-melt adhesive composition containing the same
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
GB0215844D0 (en) 2002-07-09 2002-08-14 Novartis Ag Organic compounds
AU2003252478A1 (en) 2002-07-10 2004-02-02 Ono Pharmaceutical Co., Ltd. Ccr4 antagonist and medicinal use thereof
AU2003278727A1 (en) 2002-09-20 2004-04-08 Alcon, Inc. Use of cytokine synthesis inhibitors for the treatment of dry eye disorders
US20040204404A1 (en) 2002-09-30 2004-10-14 Robert Zelle Human N-type calcium channel blockers
EP1562938B1 (en) 2002-11-04 2007-08-29 Vertex Pharmaceuticals Incorporated Heteroaryl-pyrimidine derivatives as jak inhibitors
TWI335913B (en) 2002-11-15 2011-01-11 Vertex Pharma Diaminotriazoles useful as inhibitors of protein kinases
US20040099204A1 (en) 2002-11-25 2004-05-27 Nestor John J. Sheet, page, line, position marker
PL378246A1 (en) 2002-11-26 2006-03-20 Pfizer Products Inc. Method of treatment of transplant rejection
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
TWI335819B (en) 2002-12-24 2011-01-11 Alcon Inc Use of oculosurface selective glucocorticoid in the treatment of dry eye
TW200418806A (en) 2003-01-13 2004-10-01 Fujisawa Pharmaceutical Co HDAC inhibitor
US7444183B2 (en) 2003-02-03 2008-10-28 Enteromedics, Inc. Intraluminal electrode apparatus and method
EP1611125A1 (en) 2003-02-07 2006-01-04 Vertex Pharmaceuticals Incorporated Heteroaryl substituted pyrolls useful as inhibitors of protein kinases
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
CA2522595A1 (en) 2003-04-03 2004-10-28 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
SE0301373D0 (en) 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
SE0301372D0 (en) 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
FR2857454B1 (en) 2003-07-08 2006-08-11 Aventis Pasteur DOSAGE OF TECHIC ACIDS OF BACTERIA GRAM +
US20050043346A1 (en) 2003-08-08 2005-02-24 Pharmacia Italia S.P.A. Pyridylpyrrole derivatives active as kinase inhibitors
AU2004268621C1 (en) 2003-08-29 2011-08-18 Exelixis, Inc. c-Kit modulators and methods of use
WO2005026129A1 (en) 2003-09-15 2005-03-24 Gpc Biotech Ag Pharmaceutically active 4,6-disubstituted aminopyrimidine derivatives as modulators of protein kinases
PE20050952A1 (en) 2003-09-24 2005-12-19 Novartis Ag DERIVATIVES OF ISOQUINOLINE AS INHIBITORS OF B-RAF
US7358255B2 (en) 2003-10-24 2008-04-15 Santen Pharmaceutical Co., Ltd. Therapeutic agent for keratoconjunctival disorder
MY141220A (en) 2003-11-17 2010-03-31 Astrazeneca Ab Pyrazole derivatives as inhibitors of receptor tyrosine kinases
JP2007512316A (en) 2003-11-25 2007-05-17 ファイザー・プロダクツ・インク Method for treating atherosclerosis
KR20060096153A (en) 2003-12-17 2006-09-07 화이자 프로덕츠 인코포레이티드 Pyrrolo[2,3-d]pyrimidine compounds for treating transplant rejection
DE602004016211D1 (en) 2003-12-19 2008-10-09 Schering Corp Thiadiazole als cxc- und cc-chemokinrezeptorliganden
SI1696920T1 (en) 2003-12-19 2015-02-27 Plexxikon Inc. Compounds and methods for development of ret modulators
NZ547696A (en) 2003-12-23 2009-12-24 Astex Therapeutics Ltd Pyrazole derivatives as protein kinase modulators
WO2005069865A2 (en) 2004-01-13 2005-08-04 Ambit Biosciences Corporation Pyrrolopyrimidine derivatives and analogs and their use in the treatment and prevention of diseases
EP1744751A4 (en) 2004-03-18 2010-03-10 Brigham & Womens Hospital Methods for the treatment of synucleinopathies
RS51830B (en) 2004-03-30 2012-02-29 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of jak and other protein kinases
WO2005117909A2 (en) 2004-04-23 2005-12-15 Exelixis, Inc. Kinase modulators and methods of use
WO2005105814A1 (en) 2004-04-28 2005-11-10 Incyte Corporation Tetracyclic inhibitors of janus kinases
US7558717B2 (en) 2004-04-28 2009-07-07 Vertex Pharmaceuticals Incorporated Crystal structure of human JAK3 kinase domain complex and binding pockets thereof
JP2007536310A (en) 2004-05-03 2007-12-13 ノバルティス アクチエンゲゼルシャフト Combination comprising S1P receptor agonist and JAK3 kinase inhibitor
JP2007537296A (en) 2004-05-14 2007-12-20 アボット・ラボラトリーズ Kinase inhibitors as therapeutic agents
PE20060426A1 (en) 2004-06-02 2006-06-28 Schering Corp TARTARIC ACID DERIVATIVES AS INHIBITORS OF MMPs, ADAMs, TACE AND TNF-alpha
AR049511A1 (en) 2004-06-10 2006-08-09 Irm Llc COMPOUNDS AND COMPOSITIONS AS PROTEIN QUINASE INHIBITORS
US8039674B2 (en) 2004-06-23 2011-10-18 Ono Pharmaceutical Co., Ltd. Compound having S1P receptor binding potency and use thereof
AU2005260689B2 (en) 2004-06-30 2012-05-10 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of protein kinases
US7138423B2 (en) 2004-07-20 2006-11-21 Bristol-Myers Squibb Company Arylpyrrolidine derivatives as NK-1 /SSRI antagonists
FR2873691B1 (en) 2004-07-29 2006-10-06 Sanofi Synthelabo AMINO-PIPERIDINE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
WO2006013114A1 (en) 2004-08-06 2006-02-09 Develogen Aktiengesellschaft Use of a timp-2 secreted protein product for preventing and treating pancreatic diseases and/or obesity and/or metabolic syndrome
WO2006022459A1 (en) 2004-08-23 2006-03-02 Mogam Biotechnology Institute Primer and probe for detection of sars coronavirus, kit comprising the primer and/or the probe, and detection method thereof
US20070054916A1 (en) 2004-10-01 2007-03-08 Amgen Inc. Aryl nitrogen-containing bicyclic compounds and methods of use
PL1802625T3 (en) 2004-10-13 2008-12-31 Hoffmann La Roche Disubstituted pyrazolobenzodiazepines useful as inhibitors for cdk2 and angiogesis, and for the treatment of breast, colon, lung and prostate cancer
MY179032A (en) 2004-10-25 2020-10-26 Cancer Research Tech Ltd Ortho-condensed pyridine and pyrimidine derivatives (e.g.purines) as protein kinase inhibitors
UY29177A1 (en) 2004-10-25 2006-05-31 Astex Therapeutics Ltd SUBSTITUTED DERIVATIVES OF PURINA, PURINONA AND DEAZAPURINA, COMPOSITIONS THAT CONTAIN METHODS FOR THEIR PREPARATION AND ITS USES
WO2006052913A1 (en) 2004-11-04 2006-05-18 Vertex Pharmaceuticals Incorporated PYRAZOLO[1,5-a]PYRIMIDINES USEFUL AS INHIBITORS OF PROTEIN KINASES
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
US7517870B2 (en) 2004-12-03 2009-04-14 Fondazione Telethon Use of compounds that interfere with the hedgehog signaling pathway for the manufacture of a medicament for preventing, inhibiting, and/or reversing ocular diseases related with ocular neovascularization
WO2006065916A1 (en) 2004-12-14 2006-06-22 Alcon, Inc. Method of treating dry eye disorders using 13(s)-hode and its analogs
AR054416A1 (en) 2004-12-22 2007-06-27 Incyte Corp PIRROLO [2,3-B] PIRIDIN-4-IL-AMINAS AND PIRROLO [2,3-B] PIRIMIDIN-4-IL-AMINAS AS INHIBITORS OF THE JANUS KINASES. PHARMACEUTICAL COMPOSITIONS.
WO2006067445A2 (en) 2004-12-22 2006-06-29 Astrazeneca Ab Csf-1r kinase inhibitors
WO2006077499A1 (en) 2005-01-20 2006-07-27 Pfizer Limited Chemical compounds
US8633205B2 (en) 2005-02-03 2014-01-21 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-d]pyrimidines as inhibitors of protein kinases
WO2007044050A2 (en) 2005-02-04 2007-04-19 Bristol-Myers Squibb Company 1h-imidazo[4,5-d]thieno[3,2-b]pyridine based tricyclic compounds and pharmaceutical compositions comprising same
US20080188483A1 (en) 2005-03-15 2008-08-07 Irm Llc Compounds and Compositions as Protein Kinase Inhibitors
US20070021443A1 (en) 2005-04-05 2007-01-25 Ohlmeyer Michael J Purine and imidazopyridine derivatives for immunosuppression
GB0510139D0 (en) 2005-05-18 2005-06-22 Addex Pharmaceuticals Sa Novel compounds B1
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
ZA200710379B (en) 2005-05-20 2009-05-27 Vertex Pharma Pyrrolopyridines useful as inhibitors of protein kinase
WO2006133426A2 (en) 2005-06-08 2006-12-14 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
WO2006136823A1 (en) 2005-06-21 2006-12-28 Astex Therapeutics Limited Heterocyclic containing amines as kinase b inhibitors
US7863288B2 (en) 2005-06-22 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
EP2251341A1 (en) 2005-07-14 2010-11-17 Astellas Pharma Inc. Heterocyclic Janus kinase 3 inhibitors
FR2889662B1 (en) * 2005-08-11 2011-01-14 Galderma Res & Dev OIL-IN-WATER EMULSION FOR TOPICAL APPLICATION IN DERMATOLOGY
WO2007025090A2 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Heterobicyclic and - tricyclic inhibitors of mapk/erk kinase
JP5119154B2 (en) 2005-09-22 2013-01-16 インサイト・コーポレイション Tetracyclic inhibitors of JANUS kinase
CA2623032A1 (en) 2005-09-30 2007-04-12 Vertex Pharmaceuticals Incorporated Deazapurines useful as inhibitors of janus kinases
US20070128633A1 (en) 2005-10-11 2007-06-07 Chembridge Research Laboratories, Inc. Cell-free protein expression systems and methods of use thereof
US7867949B2 (en) 2005-10-14 2011-01-11 Sumitomo Chemical Company, Limited Hydrazide compound and pesticidal use of the same
EP1945631B8 (en) 2005-10-28 2013-01-02 AstraZeneca AB 4- (3-aminopyrazole) pyrimidine derivatives for use as tyrosine kinase inhibitors in the treatment of cancer
RU2448959C2 (en) 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
WO2007062459A1 (en) 2005-11-29 2007-06-07 Cytopia Research Pty Ltd Selective kinase inhibitors based on pyridine scaffold
AU2006326548B2 (en) 2005-12-13 2012-04-05 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US20130137681A1 (en) 2005-12-13 2013-05-30 Incyte Corporation HETEROARYL SUBSTITUTED PYRROLO[2,3-b]PYRIDINES AND PYRROLO[2,3-b]PYRIMIDINES AS JANUS KINASE INHIBITORS
WO2007076423A2 (en) 2005-12-22 2007-07-05 Smithkline Beecham Corporation INHIBITORS OF Akt ACTIVITY
EA200870117A1 (en) 2005-12-23 2008-12-30 Смитклайн Бичам Корпорейшн AZAINDOL INHIBITORS AURORA-KINAZ
CN104650077A (en) 2006-01-17 2015-05-27 沃泰克斯药物股份有限公司 Azaindoles useful as inhibitors of janus kinases
WO2007084667A2 (en) 2006-01-19 2007-07-26 Osi Pharmaceutical, Inc. Fused heterobicyclic kinase inhibitors
EP1981887A2 (en) 2006-02-01 2008-10-22 SmithKline Beecham Corporation Pyrrolo[2,3,b]pyridine derivatives useful as raf kinase inhibitors
US7745477B2 (en) 2006-02-07 2010-06-29 Hoffman-La Roche Inc. Heteroaryl and benzyl amide compounds
US8003642B2 (en) 2006-03-10 2011-08-23 Ono Pharmaceutical Co., Ltd. Nitrogenated heterocyclic derivative, and pharmaceutical agent comprising the derivative as active ingredient
FR2898498B1 (en) * 2006-03-15 2008-11-28 Galderma Sa NOVEL TOPIC COMPOSITIONS IN THE FORM OF O / W EMULSION COMPRISING PRO-PENETRANT GLYCOL
TWI382984B (en) 2006-04-03 2013-01-21 Astellas Pharma Inc Hetero compound
CN101460499A (en) 2006-04-05 2009-06-17 沃泰克斯药物股份有限公司 Deazapurines useful as inhibitors of JANUS kinases
US20090124636A1 (en) 2006-04-12 2009-05-14 Pfizer Inc. Chemical compounds
WO2007129195A2 (en) 2006-05-04 2007-11-15 Pfizer Products Inc. 4-pyrimidine-5-amino-pyrazole compounds
AU2007252994A1 (en) 2006-05-18 2007-11-29 Bayer Healthcare Ag Pharmaceutical compositions comprising implitapide and methods of using same
US7691811B2 (en) 2006-05-25 2010-04-06 Bodor Nicholas S Transporter-enhanced corticosteroid activity and methods and compositions for treating dry eye
JO3235B1 (en) 2006-05-26 2018-03-08 Astex Therapeutics Ltd Pyrrolopyrimidine compounds and their uses
US20080021026A1 (en) 2006-07-20 2008-01-24 Mehmet Kahraman Benzothiophene inhibitors of rho kinase
US8715700B2 (en) * 2006-07-21 2014-05-06 Dow Pharmaceutical Sciences, Inc. Alpha hydroxy acid sustained release formulation
WO2008013622A2 (en) 2006-07-27 2008-01-31 E. I. Du Pont De Nemours And Company Fungicidal azocyclic amides
WO2008016123A1 (en) 2006-08-03 2008-02-07 Takeda Pharmaceutical Company Limited GSK-3β INHIBITOR
EP2061762B1 (en) 2006-08-16 2011-07-27 Boehringer Ingelheim International GmbH Pyrazine compounds, their use and methods of preparation
RU2009112719A (en) 2006-09-08 2010-10-20 Новартис АГ (CH) N-BIARIL (HETERO) ARILSULFONAMIDE DERIVATIVES APPLICABLE IN THE TREATMENT OF DISEASES MEDIATED BY THE INTERACTION OF LYMPHOCYTES
WO2008035376A2 (en) 2006-09-19 2008-03-27 Council Of Scientific & Industrial Research A novel bio-erodible insert for ophthalmic applications and a process for the preparation thereof
CL2007002867A1 (en) 2006-10-04 2008-06-27 Pharmacopeia Inc COMPOUNDS DERIVED FROM 2- (BENCIMIDAZOLIL) PURINA, INHIBITORS OF JANUS QUINASA 3; PHARMACEUTICAL COMPOSITION THAT CONTAINS THEM; AND ITS USE TO TREAT AUTOIMMUNE, INFLAMMATORY, CARDIOVASCULAR DISEASES, IMPLANT REJECTION, AMONG OTHERS.
WO2008043031A1 (en) 2006-10-04 2008-04-10 Pharmacopeia, Inc. 6-substituted 2-(benzimidazolyl)purine and purinone derivatives for immunosuppression
US20120225057A1 (en) 2006-10-11 2012-09-06 Deciphera Pharmaceuticals, Llc Methods and compositions for the treatment of myeloproliferative diseases and other proliferative diseases
MY146474A (en) 2006-11-06 2012-08-15 Supergen Inc Imidazo[1,2-b]pyridazine and pyrazolo[1,5-a]pyrimidine derivatives and their use as protein kinase inhibitors
US20080119496A1 (en) 2006-11-16 2008-05-22 Pharmacopeia Drug Discovery, Inc. 7-Substituted Purine Derivatives for Immunosuppression
PT2099447E (en) 2006-11-22 2013-02-27 Incyte Corp Imidazotriazines and imidazopyrimidines as kinase inhibitors
WO2008067119A2 (en) 2006-11-27 2008-06-05 Smithkline Beecham Corporation Novel compounds
JP2010513283A (en) 2006-12-15 2010-04-30 アボット・ラボラトリーズ Novel oxadiazole compounds
WO2008079291A2 (en) 2006-12-20 2008-07-03 Amgen Inc. Substituted heterocycles and methods of use
EP2118088B1 (en) 2006-12-20 2012-05-30 Amgen Inc. Heterocyclic compounds and their use in treating inflammation, angiogenesis and cancer
JP5492565B2 (en) 2006-12-22 2014-05-14 インサイト・コーポレイション Substituted heterocycles as JANUS kinase inhibitors
EA016795B1 (en) 2006-12-22 2012-07-30 Сигма-Тау Индустрие Фармасьютике Риуните С.П.А. Gel useful for the delivery of ophthalmic drugs
WO2008082839A2 (en) 2006-12-29 2008-07-10 Abbott Laboratories Pim kinase inhibitors as cancer chemotherapeutics
KR20080062876A (en) 2006-12-29 2008-07-03 주식회사 대웅제약 Novel antifungal triazole derivatives
WO2008082840A1 (en) 2006-12-29 2008-07-10 Abbott Laboratories Pim kinase inhibitors as cancer chemotherapeutics
BRPI0808523A2 (en) 2007-03-01 2014-08-19 Novartis Vaccines & Diagnostic PIM KINASE INHIBITORS AND METHODS OF USE
MY157724A (en) 2007-04-03 2016-07-15 Array Biopharma Inc IMIDAZO[1,2-a]PYRIDINE COMPOUNDS AS RECEPTOR TYROSINE KINASE INHIBITORS
GB0709031D0 (en) 2007-05-10 2007-06-20 Sareum Ltd Pharmaceutical compounds
EP2155689B1 (en) 2007-05-31 2015-07-08 Boehringer Ingelheim International GmbH Ccr2 receptor antagonists and uses thereof
GB0710528D0 (en) 2007-06-01 2007-07-11 Glaxo Group Ltd Novel compounds
EP2173752B2 (en) 2007-06-13 2022-07-13 Incyte Holdings Corporation Salts of the janus kinase inhibitor (r)-3-(4-(7h-pyrrolo(2,3-d)pyrimidin-4-yl)-1h-pyrazol-1-yl)-3-cyclopentylpropanenitrile
CL2008001709A1 (en) * 2007-06-13 2008-11-03 Incyte Corp Compounds derived from pyrrolo [2,3-b] pyrimidine, jak kinase modulators; pharmaceutical composition; and use in the treatment of diseases such as cancer, psoriasis, rheumatoid arthritis, among others.
BRPI0813553A2 (en) 2007-07-11 2017-05-09 Pfizer pharmaceutical compositions and methods for treating dry eye disorders
AP2010005167A0 (en) 2007-08-01 2010-02-28 Pfizer Pyrazole compounds and their use as RAF inhibitors
WO2009049028A1 (en) 2007-10-09 2009-04-16 Targegen Inc. Pyrrolopyrimidine compounds and their use as janus kinase modulators
CA2743756A1 (en) 2007-11-15 2009-05-22 Musc Foundation For Research Development Inhibitors of pim protein kinases, compositions, and methods for treating cancer
CN101910152B (en) 2007-11-16 2014-08-06 因塞特公司 4-pyrazolyl-N-arylpyrimidin-2-amines and 4-pyrazolyl-N-heteroarylpyrimidin-2-amines as janus kinase inhibitors
GB0723815D0 (en) 2007-12-05 2008-01-16 Glaxo Group Ltd Compounds
PL2231689T3 (en) 2008-01-18 2017-01-31 Inst Of Organic Chemistry And Biochemistry Of The Acad Of Sciences Of The Czech Republic Novel cytostatic 7-deazapurine nucleosides
EP2240016A4 (en) 2008-02-04 2011-11-16 Mercury Therapeutics Inc Ampk modulators
AR070531A1 (en) 2008-03-03 2010-04-14 Novartis Ag PIM KINASE INHIBITORS AND METHODS FOR USE
ES2602577T3 (en) 2008-03-11 2017-02-21 Incyte Holdings Corporation Azetidine and cyclobutane derivatives as JAK inhibitors
CA2718936A1 (en) 2008-03-21 2009-09-24 Novartis Ag Novel heterocyclic compounds and uses therof
TWI461423B (en) 2008-07-02 2014-11-21 Astrazeneca Ab Thiazolidinedione compounds useful in the treatment of pim kinase related conditions and diseases
FR2933409B1 (en) 2008-07-03 2010-08-27 Centre Nat Rech Scient NEW PYRROLO ° 2,3-a! CARBAZOLES AND THEIR USE AS INHIBITORS OF PIM KINASES
US8557809B2 (en) 2008-08-19 2013-10-15 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
TWI496779B (en) 2008-08-19 2015-08-21 Array Biopharma Inc Triazolopyridine compounds as pim kinase inhibitors
WO2010020905A1 (en) 2008-08-20 2010-02-25 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
CN102197032B (en) 2008-09-02 2014-07-23 诺华股份有限公司 Heterocyclic PIM-kinase inhibitors
EA201100426A1 (en) 2008-09-02 2011-10-31 Новартис Аг BICYCLIC KINASE INHIBITORS
KR101345920B1 (en) 2008-09-02 2014-02-06 노파르티스 아게 Picolinamide derivatives as kinase inhibitors
CL2009001884A1 (en) 2008-10-02 2010-05-14 Incyte Holdings Corp Use of 3-cyclopentyl-3- [4- (7h-pyrrolo [2,3-d] pyrimidin-4-yl) -1h-pyrazol-1-yl) propanonitrile, janus kinase inhibitor, and use of a composition that understands it for the treatment of dry eye.
US20110183958A1 (en) 2008-10-17 2011-07-28 Merck Frosst Canada Ltd. Azetidine derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
JOP20190230A1 (en) 2009-01-15 2017-06-16 Incyte Corp Processes for preparing jak inhibitors and related intermediate compounds
EP2210890A1 (en) 2009-01-19 2010-07-28 Almirall, S.A. Oxadiazole derivatives as S1P1 receptor agonists
US8263601B2 (en) 2009-02-27 2012-09-11 Concert Pharmaceuticals, Inc. Deuterium substituted xanthine derivatives
JP5775070B2 (en) 2009-05-22 2015-09-09 インサイト・コーポレイションIncyte Corporation N- (hetero) aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo [2,3-d] pyrimidine and pyrrol-3-yl-pyrrolo [2,3-d] pyrimidine as Janus kinase inhibitors
CN106967070A (en) 2009-05-22 2017-07-21 因塞特控股公司 It is used as the compound of JAK inhibitor
UA110324C2 (en) 2009-07-02 2015-12-25 Genentech Inc Jak inhibitory compounds based on pyrazolo pyrimidine
CN101958119B (en) 2009-07-16 2012-02-29 中兴通讯股份有限公司 Audio-frequency drop-frame compensator and compensation method for modified discrete cosine transform domain
US20120157500A1 (en) 2009-08-24 2012-06-21 Weikang Tao Jak inhibition blocks rna interference associated toxicities
TW201111385A (en) 2009-08-27 2011-04-01 Biocryst Pharm Inc Heterocyclic compounds as janus kinase inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
SG10201405568UA (en) 2009-09-08 2014-11-27 Hoffmann La Roche 4-substituted pyridin-3-yl-carboxamide compounds and methods of use
EP2305660A1 (en) 2009-09-25 2011-04-06 Almirall, S.A. New thiadiazole derivatives
WO2011044481A1 (en) 2009-10-09 2011-04-14 Incyte Corporation Hydroxyl, keto, and glucuronide derivatives of 3-(4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl)-3-cyclopentylpropanenitrile
JP5744887B2 (en) 2009-10-20 2015-07-08 セルゾーム リミティッド Heterocyclylpyrazolopyrimidine analogs as JAK inhibitors
EP2332917B1 (en) 2009-11-11 2012-08-01 Sygnis Bioscience GmbH & Co. KG Compounds for PIM kinase inhibition and for treating malignancy
EP2504032A4 (en) 2009-11-24 2013-06-19 Alderbio Holdings Llc Antagonists of il-6 to prevent or treat thrombosis
US20130129675A1 (en) 2009-12-04 2013-05-23 Board Of Regents, The University Of Texas System Interferon therapies in combination with blockade of stat3 activation
RU2012132278A (en) 2010-01-12 2014-02-20 Ф. Хоффманн-Ля Рош Аг TRICYCLIC HETEROCYCLIC COMPOUNDS CONTAINING THEIR COMPOSITIONS AND WAYS OF THEIR APPLICATION
SA111320200B1 (en) 2010-02-17 2014-02-16 ديبيوفارم اس ايه Bicyclic Compounds and their Uses as Dual C-SRC / JAK Inhibitors
AU2011217961B2 (en) 2010-02-18 2016-05-05 Incyte Holdings Corporation Cyclobutane and methylcyclobutane derivatives as Janus kinase inhibitors
PT3354652T (en) 2010-03-10 2020-07-20 Incyte Holdings Corp Piperidin-4-yl azetidine derivatives as jak1 inhibitors
CN102985424B (en) 2010-04-14 2015-03-11 阵列生物制药公司 5, 7-substituted-imidazo [1, 2-c] pyrimidines as inhibitors of jak kinases
EP2390252A1 (en) 2010-05-19 2011-11-30 Almirall, S.A. New pyrazole derivatives
KR102303885B1 (en) * 2010-05-21 2021-09-24 인사이트 홀딩스 코포레이션 Topical formulation for a jak inhibitor
WO2011156698A2 (en) 2010-06-11 2011-12-15 Abbott Laboratories NOVEL PYRAZOLO[3,4-d]PYRIMIDINE COMPOUNDS
US9351943B2 (en) 2010-07-01 2016-05-31 Matthew T. McLeay Anti-fibroblastic fluorochemical emulsion therapies
EP2621489A1 (en) 2010-09-30 2013-08-07 Portola Pharmaceuticals, Inc. Combinations of 4-(cyclopropylamino)-2-(4-(4-(ethylsulfonyl)piperazin-1-yl)phenylamino)pyrimidine-5-carboxamide and fludarabine
ES2536415T3 (en) 2010-11-19 2015-05-25 Incyte Corporation Pyrrolopyridines and heterocyclic substituted pyrrolopyrimidines as JAK inhibitors
WO2012068450A1 (en) 2010-11-19 2012-05-24 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
CA2819560A1 (en) 2010-12-03 2012-06-07 Ym Biosciences Australia Pty Ltd Treatment of jak2-mediated conditions
ES2547916T3 (en) 2011-02-18 2015-10-09 Novartis Pharma Ag MTOR / JAK inhibitor combination therapy
BR112013032720A2 (en) 2011-06-20 2016-09-13 Incyte Corp "azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors, composition and use of said derivatives"
EP2741747A1 (en) 2011-08-10 2014-06-18 Novartis Pharma AG JAK P13K/mTOR COMBINATION THERAPY
TW201313721A (en) 2011-08-18 2013-04-01 Incyte Corp Cyclohexyl azetidine derivatives as JAK inhibitors
UA111854C2 (en) 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн METHODS AND INTERMEDIATE COMPOUNDS FOR JAK INHIBITORS
WO2013173720A1 (en) 2012-05-18 2013-11-21 Incyte Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US10155987B2 (en) 2012-06-12 2018-12-18 Dana-Farber Cancer Institute, Inc. Methods of predicting resistance to JAK inhibitor therapy
US9573958B2 (en) 2012-08-31 2017-02-21 Principia Biopharma, Inc. Benzimidazole derivatives as ITK inhibitors
TWI646099B (en) 2012-11-01 2019-01-01 英塞特控股公司 Tricyclic fused thiophene derivatives as JAK inhibitors
MY191357A (en) 2012-11-15 2022-06-19 Incyte Holdings Corp Sustained-release dosage forms of ruxolitinib
ES2900492T3 (en) 2013-03-06 2022-03-17 Incyte Holdings Corp Processes and intermediates to make a JAK inhibitor
SI2997023T1 (en) 2013-05-17 2017-07-31 Incyte Corporation Bipyrazole derivatives as jak inhibitors
KR102419714B1 (en) 2013-08-07 2022-07-13 인사이트 코포레이션 Sustained release dosage forms for a jak1 inhibitor
KR20160044570A (en) 2013-08-20 2016-04-25 인사이트 코포레이션 Survival benefit in patients with solid tumors with elevated c-reactive protein levels
CN112494652A (en) 2014-02-28 2021-03-16 因赛特公司 JAK1 inhibitors for the treatment of myelodysplastic syndrome
SG10201808870TA (en) 2014-04-08 2018-11-29 Incyte Corp Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
CA2947418A1 (en) 2014-04-30 2015-11-05 Incyte Corporation Processes of preparing a jak1 inhibitor and new forms thereto
ES2946179T3 (en) 2014-05-28 2023-07-13 Onco Tracker Inc Antineoplastic effects of JAK2 inhibitors in combination with thalidomide derivatives and glucocorticoids
WO2015184305A1 (en) 2014-05-30 2015-12-03 Incyte Corporation TREATMENT OF CHRONIC NEUTROPHILIC LEUKEMIA (CNL) AND ATYPICAL CHRONIC MYELOID LEUKEMIA (aCML) BY INHIBITORS OF JAK1

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6075056A (en) * 1997-10-03 2000-06-13 Penederm, Inc. Antifungal/steroid topical compositions
WO2009158687A1 (en) * 2008-06-26 2009-12-30 Anterios, Inc. Dermal delivery

Cited By (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9974790B2 (en) 2005-12-13 2018-05-22 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US11331320B2 (en) 2005-12-13 2022-05-17 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US8933086B2 (en) 2005-12-13 2015-01-13 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B]pyridines and pyrrolo[2,3-B]pyrimidines as Janus kinase inhibitors
US9662335B2 (en) 2005-12-13 2017-05-30 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US11744832B2 (en) 2005-12-13 2023-09-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9814722B2 (en) 2005-12-13 2017-11-14 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US8530485B2 (en) 2005-12-13 2013-09-10 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US8415362B2 (en) 2005-12-13 2013-04-09 Incyte Corporation Pyrazolyl substituted pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9206187B2 (en) 2005-12-13 2015-12-08 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase
US9079912B2 (en) 2005-12-13 2015-07-14 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase inhibitors
US10398699B2 (en) 2005-12-13 2019-09-03 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US8541425B2 (en) 2005-12-13 2013-09-24 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US10639310B2 (en) 2005-12-13 2020-05-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US8946245B2 (en) 2005-12-13 2015-02-03 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9376439B2 (en) 2007-06-13 2016-06-28 Incyte Corporation Salts of the janus kinase inhibitor (R)-3(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10016429B2 (en) 2007-06-13 2018-07-10 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8822481B1 (en) 2007-06-13 2014-09-02 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d] pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10610530B2 (en) 2007-06-13 2020-04-07 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8829013B1 (en) 2007-06-13 2014-09-09 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10463667B2 (en) 2007-06-13 2019-11-05 Incyte Incorporation Metabolites of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US11213528B2 (en) 2007-06-13 2022-01-04 Incyte Holdings Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9000161B2 (en) 2009-01-15 2015-04-07 Incyte Corporation Processes for preparing JAK inhibitors and related intermediate compounds
US10364248B2 (en) 2009-01-15 2019-07-30 Incyte Corporation Processes for preparing 4-chloro-7H-pyrrolo[2,3-d]pyrimidine
US9290506B2 (en) 2009-01-15 2016-03-22 Incyte Corporation Processes for preparing JAK inhibitors and related intermediate compounds
US8993582B2 (en) 2009-01-15 2015-03-31 Incyte Corporation Processes for preparing JAK inhibitors and related intermediate compounds
US10975085B2 (en) 2009-01-15 2021-04-13 Incyte Holdings Corporation Process for preparing a composition comprising an enantiomeric excess of greater than or equal to 90% of the (R)-enantiomer of a compound of formula III
US8883806B2 (en) 2009-01-15 2014-11-11 Incyte Corporation Processes for preparing JAK inhibitors and related intermediate compounds
US9908888B2 (en) 2009-01-15 2018-03-06 Incyte Corporation Processes for preparing pyrazolyl-substituted pyrrolo[2,3-d]pyrimidines
US9216984B2 (en) 2009-05-22 2015-12-22 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane—or heptane-nitrile as JAK inhibitors
US9334274B2 (en) 2009-05-22 2016-05-10 Incyte Holdings Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
US8716303B2 (en) 2009-05-22 2014-05-06 Incyte Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US8604043B2 (en) 2009-05-22 2013-12-10 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as jak inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9999619B2 (en) 2010-03-10 2018-06-19 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US11285140B2 (en) 2010-03-10 2022-03-29 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US8765734B2 (en) 2010-03-10 2014-07-01 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10695337B2 (en) 2010-03-10 2020-06-30 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US9464088B2 (en) 2010-03-10 2016-10-11 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
US10869870B2 (en) 2010-05-21 2020-12-22 Incyte Corporation Topical formulation for a JAK inhibitor
US11219624B2 (en) 2010-05-21 2022-01-11 Incyte Holdings Corporation Topical formulation for a JAK inhibitor
US11590136B2 (en) 2010-05-21 2023-02-28 Incyte Corporation Topical formulation for a JAK inhibitor
US11571425B2 (en) 2010-05-21 2023-02-07 Incyte Corporation Topical formulation for a JAK inhibitor
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
US8933085B2 (en) 2010-11-19 2015-01-13 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US9993480B2 (en) 2011-02-18 2018-06-12 Novartis Pharma Ag mTOR/JAK inhibitor combination therapy
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US11214573B2 (en) 2011-06-20 2022-01-04 Incyte Holdings Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US8691807B2 (en) 2011-06-20 2014-04-08 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9023840B2 (en) 2011-06-20 2015-05-05 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US10513522B2 (en) 2011-06-20 2019-12-24 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9358229B2 (en) 2011-08-10 2016-06-07 Novartis Pharma Ag JAK PI3K/mTOR combination therapy
US9359358B2 (en) 2011-08-18 2016-06-07 Incyte Holdings Corporation Cyclohexyl azetidine derivatives as JAK inhibitors
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US11161855B2 (en) 2012-11-01 2021-11-02 Incyte Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9181271B2 (en) 2012-11-01 2015-11-10 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US10370387B2 (en) 2012-11-01 2019-08-06 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9908895B2 (en) 2012-11-01 2018-03-06 Incyte Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9777017B2 (en) 2012-11-01 2017-10-03 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US11851442B2 (en) 2012-11-01 2023-12-26 Incyte Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US11337927B2 (en) 2012-11-15 2022-05-24 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US11576864B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10874616B2 (en) 2012-11-15 2020-12-29 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11576865B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11896717B2 (en) 2012-11-15 2024-02-13 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US9221845B2 (en) 2013-03-06 2015-12-29 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9714233B2 (en) 2013-03-06 2017-07-25 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US8987443B2 (en) 2013-03-06 2015-03-24 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US11001571B2 (en) 2013-05-17 2021-05-11 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US10435392B2 (en) 2013-05-17 2019-10-08 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US11591318B2 (en) 2013-05-17 2023-02-28 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US11905275B2 (en) 2013-05-17 2024-02-20 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US9382231B2 (en) 2013-05-17 2016-07-05 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US9926301B2 (en) 2013-05-17 2018-03-27 Incyte Corporation Bipyrazole derivatives as JAK inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US11045421B2 (en) 2013-08-07 2021-06-29 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10561616B2 (en) 2013-08-07 2020-02-18 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10675284B2 (en) 2014-04-08 2020-06-09 Incyte Corporation Treatment of B-cell malignancies by a combination JAK and PI3K inhibitors
US10064866B2 (en) 2014-04-08 2018-09-04 Incyte Corporation Treatment of B-cell malignancies by a combination JAK and PI3K inhibitors
US9802957B2 (en) 2014-04-30 2017-10-31 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
US10450325B2 (en) 2014-04-30 2019-10-22 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US11833152B2 (en) 2018-02-16 2023-12-05 Incyte Corporation JAK1 pathway inhibitors for the treatment of cytokine-related disorders
US11103510B2 (en) 2018-02-16 2021-08-31 Incyte Corporation JAK1 pathway inhibitors for the treatment of cytokine-related disorders
US11584961B2 (en) 2018-03-30 2023-02-21 Incyte Corporation Biomarkers for inflammatory skin disease
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11372003B2 (en) 2018-04-13 2022-06-28 Incyte Corporation Biomarkers for graft-versus-host disease
US11324749B2 (en) 2018-10-31 2022-05-10 Incyte Corporation Combination therapy for treatment of hematological diseases
US11685731B2 (en) 2020-06-02 2023-06-27 Incyte Corporation Processes of preparing a JAK1 inhibitor
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
WO2022125670A1 (en) 2020-12-08 2022-06-16 Incyte Corporation Jak1 pathway inhibitors for the treatment of vitiligo

Also Published As

Publication number Publication date
US20200368240A1 (en) 2020-11-26
AU2020201151A1 (en) 2020-03-05
MX2012013400A (en) 2013-02-26
KR20210118207A (en) 2021-09-29
KR20130109012A (en) 2013-10-07
JP2020079281A (en) 2020-05-28
JP5849312B2 (en) 2016-01-27
CR20120605A (en) 2013-03-11
ES2581834T3 (en) 2016-09-07
CO6640250A2 (en) 2013-03-22
MY178634A (en) 2020-10-19
CN103002875B (en) 2016-05-04
CL2012003229A1 (en) 2013-01-25
JP2013529214A (en) 2013-07-18
US20200046707A1 (en) 2020-02-13
KR20190125531A (en) 2019-11-06
SMT201600172B (en) 2016-08-31
US20220211707A1 (en) 2022-07-07
KR101921466B1 (en) 2018-11-26
JP2016053069A (en) 2016-04-14
JP2021193140A (en) 2021-12-23
AU2011255443A1 (en) 2012-12-06
US11590136B2 (en) 2023-02-28
AU2011255443B2 (en) 2014-07-03
US20220378792A1 (en) 2022-12-01
SG10201910912TA (en) 2020-01-30
CN105853356A (en) 2016-08-17
EP2574168B1 (en) 2016-04-20
US20230277541A1 (en) 2023-09-07
MY161078A (en) 2017-04-14
US20220370455A1 (en) 2022-11-24
CA2799928C (en) 2020-03-31
SG10201503983QA (en) 2015-06-29
CN105853356B (en) 2019-07-16
EA201291310A1 (en) 2013-06-28
US11571425B2 (en) 2023-02-07
RS54824B1 (en) 2016-10-31
US20150250790A1 (en) 2015-09-10
HRP20160841T1 (en) 2016-09-23
MX338228B (en) 2016-04-08
EA202091303A2 (en) 2021-03-31
KR102402137B1 (en) 2022-05-30
AU2020201151B2 (en) 2022-04-07
EP2574168A2 (en) 2013-04-03
AR084691A1 (en) 2013-06-05
WO2011146808A3 (en) 2012-06-07
CA2799928A1 (en) 2011-11-24
EP3087972A1 (en) 2016-11-02
CY1117815T1 (en) 2017-05-17
KR102635013B1 (en) 2024-02-13
DK2574168T3 (en) 2016-05-09
IL223084A (en) 2017-03-30
JP6479877B2 (en) 2019-03-06
JP6657441B2 (en) 2020-03-04
KR20180101617A (en) 2018-09-12
BR112012029653A2 (en) 2016-08-02
KR102040479B1 (en) 2019-11-06
US11219624B2 (en) 2022-01-11
AU2022204807A1 (en) 2022-07-28
KR102303885B1 (en) 2021-09-24
HK1182313A1 (en) 2013-11-29
IL223084A0 (en) 2013-02-03
HUE029035T2 (en) 2017-01-30
EA035981B1 (en) 2020-09-09
EA202091303A3 (en) 2021-05-31
AU2018201889B2 (en) 2020-03-05
ZA202001999B (en) 2022-03-30
JP2023002758A (en) 2023-01-10
NZ603686A (en) 2014-11-28
UA111588C2 (en) 2016-05-25
BR112012029653B1 (en) 2021-01-12
ME02445B (en) 2016-09-20
PE20130216A1 (en) 2013-02-27
KR20220104166A (en) 2022-07-26
SG185567A1 (en) 2012-12-28
US20220211712A1 (en) 2022-07-07
JP7167280B2 (en) 2022-11-08
JP2017149739A (en) 2017-08-31
WO2011146808A2 (en) 2011-11-24
JP6952143B2 (en) 2021-10-20
US10869870B2 (en) 2020-12-22
AR124134A2 (en) 2023-02-15
ECSP13012546A (en) 2013-06-28
CN103002875A (en) 2013-03-27
EP2574168B9 (en) 2016-10-05
PL2574168T3 (en) 2016-10-31
AU2016204689A1 (en) 2016-07-21
TWI499421B (en) 2015-09-11
SI2574168T1 (en) 2016-07-29
AU2014202896A1 (en) 2014-06-19
US10758543B2 (en) 2020-09-01
US20210000832A1 (en) 2021-01-07
AU2018201889A1 (en) 2018-04-12
TW201201809A (en) 2012-01-16
JP2019081783A (en) 2019-05-30

Similar Documents

Publication Publication Date Title
US11571425B2 (en) Topical formulation for a JAK inhibitor

Legal Events

Date Code Title Description
AS Assignment

Owner name: INCYTE CORPORATION, DELAWARE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PARIKH, BHAVNISH;SHAH, BHAVESH;YELESWARAM, KRISHNASWAMY;SIGNING DATES FROM 20110715 TO 20110720;REEL/FRAME:026641/0350

AS Assignment

Owner name: INCYTE HOLDINGS CORPORATION AND INCYTE CORPORATION

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INCYTE CORPORATION;REEL/FRAME:035924/0004

Effective date: 20150107

AS Assignment

Owner name: INCYTE HOLDINGS CORPORATION, DELAWARE

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE OMMISSION OF SECOND RECEIVING PARTY NAME PREVIOUSLY RECORDED AT REEL: 035292 FRAME: 0004. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNOR:INCYTE CORPORATION;REEL/FRAME:036054/0696

Effective date: 20150107

Owner name: INCYTE CORPORATION, DELAWARE

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE OMMISSION OF SECOND RECEIVING PARTY NAME PREVIOUSLY RECORDED AT REEL: 035292 FRAME: 0004. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNOR:INCYTE CORPORATION;REEL/FRAME:036054/0696

Effective date: 20150107

STCB Information on status: application discontinuation

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