WO2015021153A1 - Sustained release dosage forms for a jak1 inhibitor - Google Patents

Sustained release dosage forms for a jak1 inhibitor Download PDF

Info

Publication number
WO2015021153A1
WO2015021153A1 PCT/US2014/049940 US2014049940W WO2015021153A1 WO 2015021153 A1 WO2015021153 A1 WO 2015021153A1 US 2014049940 W US2014049940 W US 2014049940W WO 2015021153 A1 WO2015021153 A1 WO 2015021153A1
Authority
WO
WIPO (PCT)
Prior art keywords
dosage forms
sustained release
release dosage
isonicotinoyl
azetidin
Prior art date
Application number
PCT/US2014/049940
Other languages
French (fr)
Inventor
Krishnaswamy Yeleswaram
Bhavnish Parikh
Dilip P. MODI
Trupti SHETH
Original Assignee
Incyte Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MYPI2016000200A priority Critical patent/MY195091A/en
Priority to KR1020217018433A priority patent/KR102419714B1/en
Priority to MX2016001639A priority patent/MX2016001639A/en
Priority to EP14753182.6A priority patent/EP3030227B1/en
Priority to DK14753182.6T priority patent/DK3030227T3/en
Priority to EA201690357A priority patent/EA201690357A1/en
Priority to PL14753182T priority patent/PL3030227T3/en
Priority to CR20190343A priority patent/CR20190343A/en
Priority to KR1020167006096A priority patent/KR20160045081A/en
Priority to SG11201600815WA priority patent/SG11201600815WA/en
Priority to IL277554A priority patent/IL277554B2/en
Priority to CN202110883638.4A priority patent/CN114010611B/en
Priority to JP2016533398A priority patent/JP6334700B2/en
Priority to AU2014305989A priority patent/AU2014305989B2/en
Priority to CN202310115279.7A priority patent/CN116036089A/en
Priority to NZ717230A priority patent/NZ717230B2/en
Priority to CN201480052299.3A priority patent/CN105579032A/en
Application filed by Incyte Corporation filed Critical Incyte Corporation
Priority to UAA201602100A priority patent/UA120499C2/en
Priority to CR20230316A priority patent/CR20230316A/en
Priority to PE2021002226A priority patent/PE20220579A1/en
Priority to ES14753182T priority patent/ES2792549T3/en
Priority to LTEP14753182.6T priority patent/LT3030227T/en
Priority to EP20164849.0A priority patent/EP3721873A1/en
Priority to KR1020227023370A priority patent/KR20220103810A/en
Priority to SI201431549T priority patent/SI3030227T1/en
Priority to CA2920108A priority patent/CA2920108C/en
Priority to RS20200696A priority patent/RS60469B1/en
Priority to BR112016002571-7A priority patent/BR112016002571B1/en
Priority to BR122020001831A priority patent/BR122020001831A8/en
Publication of WO2015021153A1 publication Critical patent/WO2015021153A1/en
Priority to IL243920A priority patent/IL243920B/en
Priority to PH12016500243A priority patent/PH12016500243B1/en
Priority to CR20160102A priority patent/CR20160102A/en
Priority to PH12019502074A priority patent/PH12019502074A1/en
Priority to PH12019502072A priority patent/PH12019502072A1/en
Priority to AU2019257368A priority patent/AU2019257368B2/en
Priority to HRP20200955TT priority patent/HRP20200955T1/en
Priority to CY20201100572T priority patent/CY1123314T1/en
Priority to AU2021202916A priority patent/AU2021202916A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • This application relates to a sustained release dosage form comprising ⁇ 1- ⁇ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, and doses and methods related thereto.
  • 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 antiinflammatory 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 JAKL
  • L-JAK L-JAK
  • TYK2 protein-tyros ine 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
  • JAKs Deficiencies in expression of JAKs are associated with many disease states. For example, Jakl-/- 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).
  • cytokine stimulation e.g. IL-6 or GM-CSF
  • SOCS suppressor or cytokine signaling
  • PIAS protein inhibitor of activated STAT
  • JAK2 tyrosine kinase can be beneficial for patients with myeloproliferative disorders, e.g., polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM) (Levin, et al, Cancer Cell, vol. 7, 2005: 387- 397).
  • PV polycythemia vera
  • ET essential thrombocythemia
  • MMM myeloid metaplasia with myelofibrosis
  • Inhibition of the JAK2V617F kinase decreases proliferation of hematopoietic cells, suggesting JAK2 as a potential target for pharmacologic inhibition in patients with PV, ET, and MMM.
  • Inhibition of the JAKs may benefit patients suffering from skin immune disorders such as psoriasis, and skin sensitization.
  • JAK inhibitors are described in U.S. Serial No. 13/043,986 (US
  • 201 1/0224190 filed March 9, 201 1, which is incorporated herein by reference in its entirety, including ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3 -yl ⁇ acetonitrile, which is depicted below as Formula I.
  • sustained-release dosage forms comprising about 25 mg to about 600 mg (e.g., 25 mg, 100 mg, 200 mg, 300 mg, or 600 mg) on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present invention further provides one or more sustained release dosage forms each comprising ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said one or more sustained release dosage forms together provide a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile
  • the present invention also provides a dose, comprising one or more sustained release dosage forms each comprising ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application also provides a dose comprising one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
  • the present application further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage forms as described herein.
  • the present application also provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage as described herein.
  • the present application also provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • FIG. 1A-C depicts plasma concentrations for the compound of Formula I (Mean ⁇ SE) in healthy subjects receiving single doses of 300 mg IR capsules (1A: Cohorts 1-4, fasted), SRI, SR2, SR3, and SR4 tablets (2B: Cohorts 1-4, fasted; and 2C: Cohorts 1-4, fed a high-fat meal).
  • FIG. 2A-B depicts single-dose 300 mg SR3 PK profiles (Mean ⁇ SE) (2A: Cohort 3, SR3, fasted versus high-fat meal; and 2B: Cohort 5, SR3, fasted versus medium-fat meal).
  • FIG. 3 depicts a comparison of PK profiles (mean ⁇ SE) between the 25 mg and 100 mg SR3 tablets (treatment A vs C) and the food effect of a high- fat meal on the 25 mg SR3 tablet (treatment B vs A).
  • FIG. 4 depicts the percent change from baseline for hemoglobin for several dosing regimens for sustained release tablets versus placebo.
  • FIG. 5(a) depicts the percentage of patients having a > 50% reduction in total symptom score (TSS) at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
  • FIG. 5(b) depicts the percent change in total symptom score (TSS) from baseline at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
  • FIG. 6(a) depicts mean hemoglobin levels over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
  • FIG. 6(b) depicts mean hemoglobin levels (g/dL) over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) at 48 weeks.
  • FIG. 6(c) depicts mean hemoglobin levels (g/dL) over time by dose cohort at 48 weeks as an average for three dose cohorts as compared to individuals dosed with placebo or ruxolitinib.
  • the present application provides sustained-release dosage forms comprising ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the present application provides a sustained-release dosage form comprising about 25 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the sustained-release dosage form comprises about 300 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the sustained-release dosage form comprises about 200 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the sustained-release dosage form comprises about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the sustained-release dosage form comprises about 300 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile adipic acid salt.
  • the sustained-release dosage form comprises about 200 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile adipic acid salt.
  • the sustained-release dosage form comprises about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile adipic acid salt.
  • oral administration of three of said dosage forms to a fasted individual provides a mean peak plasma concentration (Cmax) of ⁇ 1- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 100 nM to about 1000 nM.
  • Cmax mean peak plasma concentration
  • oral administration means that a single dose is administered to the individual (in this case, 3 x 100 mg) and the PK parameter is calculated from the measurements of plasma concentration over time.
  • the PK parameter in this case, Cmax
  • the PK parameter is being used to characterize the single sustained release dosage form (i.e., the claims are directed to a single dosage form, not three dosage forms).
  • oral administration of three of said dosage forms to a fasted individual provides a mean peak plasma concentration (Cmax) of ⁇ 1- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 400 nM to about 700 nM.
  • Cmax mean peak plasma concentration
  • oral administration of three of said dosage forms to a fasted individual provides a mean time to peak plasma concentration (Tmax) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 0.5 hours to about 3 hours.
  • Tmax mean time to peak plasma concentration
  • oral administration of three of said dosage forms to a fasted individual provides a mean time to peak plasma concentration (Tmax) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of at least 0.5 hours.
  • Tmax mean time to peak plasma concentration
  • oral administration of three of said dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile of about 5 to about 50.
  • Cmax mean peak plasma concentration
  • Ca2h mean 12-hour plasma concentration
  • oral administration of three of said dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile of about 9 to about 40.
  • Cmax mean peak plasma concentration
  • Ca2h mean 12-hour plasma concentration
  • oral administration of three of said dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile of about 15 to about 30.
  • Cmax mean peak plasma concentration
  • Ca2h mean 12-hour plasma concentration
  • sustained-release dosage form comprising about 100 mg
  • oral administration of three of said dosage forms to a fasted individual provides a mean half-life (ti/2) of ⁇ l- ⁇ l-[3-fluoro-2-
  • sustained-release dosage form comprising about 100 mg
  • oral administration of three of said dosage forms to a fasted individual provides a mean half-life (ti/2) of ⁇ l- ⁇ l-[3-fluoro-2-
  • sustained-release dosage form comprising about 100 mg
  • oral administration of three of said dosage forms to a fasted individual provides a mean half-life (ti/2) of ⁇ l- ⁇ l-[3-fluoro-2-
  • oral administration of three of said dosage forms to a fasted individual provides a mean bioavailability (AUCo - ⁇ ) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidm ⁇
  • oral administration of three of said dosage forms to a fasted individual provides a mean bioavailability (AUCo - ⁇ ) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 1500 nM*h to about 3100 nM*h.
  • AUCo - ⁇ mean bioavailability
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean peak plasma concentration (Cmax) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 200 nM to about 2000 nM.
  • Cmax mean peak plasma concentration
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean peak plasma concentration (Cmax) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 500 nM to about 1500 nM.
  • Cmax mean peak plasma concentration
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of ⁇ l- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 1 hour to about 9 hours.
  • Tmax mean time to peak plasma concentration
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of ⁇ l- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of at least 1.5 hours.
  • Tmax mean time to peak plasma concentration
  • sustained-release dosage form comprising about 100 mg
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12- hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro-2-
  • sustained-release dosage form comprising about 100 mg
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12- hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro-2-
  • sustained-release dosage form comprising about 100 mg
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12- hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro-2-
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 1 hour to about 7 hours.
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 2 hours to about 5 hours.
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean bioavailability (AUCo - ⁇ ) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 2000 nM*h to about 5000 nM*h.
  • AUCo - ⁇ mean bioavailability
  • oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean bioavailability (AUCo - ⁇ ) of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 3000 nM*h to about 4000 nM*h.
  • AUCo - ⁇ mean bioavailability
  • the percent geometric mean ratio of the sustained release dosage form relative to an immediate release dosage form for Cmax is about 15% to about 30%, wherein one or more immediate release dosage forms and one or more sustained release dosage forms are independently orally administered to fasted individuals as a single dose, wherein the same size dose of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt, is administered.
  • the percent geometric mean ratio of the sustained release dosage form relative to an immediate release dosage form for Cmax is about 15% to about 30%, wherein one or more immediate release dosage forms and one or more sustained release dosage forms are independently orally administered to fasted individuals as a single dose, wherein the same size dose of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt, is administered.
  • the percent geometric mean ratio of the sustained release dosage form relative to an immediate release dosage form for AUCo - ⁇ is about 40% to about 55%, wherein one or more immediate release dosage forms and one or more sustained release dosage forms are independently orally administered to fasted individuals as a single dose, wherein the same size dose of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt, is administered.
  • the percent geometric mean ratio for Cmax of the sustained release dosage form orally administered to an individual after a high-fat meal relative to the sustained release dosage form orally administered to a fasted individual is about 150% to about 250%.
  • the percent geometric mean ratio for AUCo - ⁇ of the sustained release dosage form orally administered to an individual after a high-fat meal relative to the sustained release dosage form orally administered to a fasted individual is about 125% to about 170%.
  • the sustained-release dosage forms of the invention may include a sustained-release matrix former.
  • Example sustained-release matrix formers include cellulosic ethers such as hydroxypropyl methylcellulose (HPMC, hypromellose) which is a high viscosity polymer, and methyl celluloses.
  • Example hydroxypropyl methylcelluloses include MethocelTM K15M, MethocelTM K4M, MethocelTM K100LV, MethocelTM E3, MethocelTM E5, MethocelTM E6, MethocelTM E15, MethocelTM E50, MethocelTM E10M, MethocelTM E4M, and MethocelTM E10M.
  • the sustained release dosage form comprises one or more hypromelloses.
  • the sustained release dosage form comprises a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP and a second hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP.
  • the sustained release dosage form comprises about 8% to about 20% by weight of one or more hypromelloses.
  • the sustained release dosage form comprises about 10% to about 15% by weight of one or more hypromelloses.
  • the sustained-release dosage forms of the invention can further include one or more fillers, glidants, disintegrants, binders, or lubricants as inactive ingredients.
  • the filler comprises microcrystalline cellulose, lactose monohydrate, or both.
  • the sustained release dosage form comprises about 16% to about 22% by weight of microcrystalline cellulose.
  • the sustained release dosage form comprises about 45% to about 55% by weight of lactose monohydrate.
  • lubricants can be present in the dosage forms of the invention in an amount of 0 to about 5% by weight.
  • lubricants include magnesium stearate, stearic acid (stearin), hydrogenated oil, polyethylene glycol, sodium stearyl fumarate, and glyceryl behenate.
  • the formulations include magnesium stearate, stearic acid, or both.
  • the sustained release dosage form comprises about 0.3% to about 0.7% by weight of magnesium stearate.
  • glidants may be present in the dosage forms. In some embodiments, glidants can be present in the dosage forms of the invention in an amount of 0 to about 5% by weight.
  • glidants include talc, colloidal silicon dioxide, and cornstarch. In some embodiments, the glidant is colloidal silicon dioxide.
  • film-coating agents can be present in an amount of 0 to about 5% by weight.
  • Non-limiting illustrative examples of film-coating agents include hypromellose or polyvinyl alcohol based coating with titanium dioxide, talc and optionally colorants available in several commercially available complete coating systems.
  • the sustained release dosage form comprises pregelatinized starch.
  • the sustained release dosage form is a tablet.
  • the sustained release dosage form is prepared by process comprising wet granulation.
  • the sustained release dosage form comprises one or more excipients independently selected from hypromelloses and microcrystalline celluloses.
  • the sustained release dosage form comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, and lactose monohydrate.
  • the sustained release dosage form comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, lactose monohydrate, and pregelatinized starch.
  • the present invention further provides one or more sustained release dosage forms each comprising ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said one or more sustained release dosage forms together provide a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile
  • the present invention also provides a dose, comprising one or more sustained release dosage forms each comprising ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 500 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 400 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of ⁇ 1- ⁇ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, is provided.
  • the present application also provides a dose comprising one or more sustained release dosage forms as described herein, which provide a once-daily oral dosage of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application also provides a dose comprising one or more sustained release dosage forms as described herein, which provide a once-daily oral dosage of about 500 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the dose comprises six dosage forms of about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the dose comprises three dosage forms of about 200 mg on a free base basis of ⁇ 1- ⁇ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the dose comprises two dosage forms of about 300 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the dose comprises one dosage form of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 400 mg to about 600 mg on a free base basis of ⁇ 1- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
  • the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once- daily dose of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
  • the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once-daily dose of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 500 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
  • the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once-daily dose of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 400 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
  • the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once-daily dose of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the kit comprises six dosage forms of about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ - 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the kit comprises three dosage forms of about 200 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the kit comprises two dosage forms of about 300 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the kit comprises one dosage form of about 600 mg on a free base basis of ⁇ 1- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • sustained-release is used as generally understood in the art and refers to a formulation designed to slowly release the active ingredient into a patient after oral administration.
  • dose refers to the total amount of the compound of Formula I orally administered to the individual or patient.
  • the dose may be in a single dosage form, or a plurality of dosage forms (e.g., a 600 mg dose may be one 600 mg dosage form, two 300 mg dosage forms, three 200 mg dosage forms, six 100 mg dosage forms, etc.).
  • a dose can refer to a plurality of pills to be taken by a patient at nearly simultaneously.
  • a fasted individual means an individual who has fasted for at least 10 hours prior to administration of the dose.
  • mean when preceding a pharmacokinetic value (e.g. mean Cmax) represents the arithmetic mean value of the pharmacokinetic value taken from a population of patients unless otherwise specified.
  • Cmax means the maximum observed plasma concentration.
  • Ci2h refers to the plasma concentration measured at 12 hours from administration.
  • Tmax refers to the time at which the maximum blood plasma concentration is observed.
  • T1/2 refers to the time at which the plasma concentration is half of the observed maximum.
  • AUC refers to the area under the plasma concentration-time curve which is a measure of total bioavailability.
  • AUCo - ⁇ refers to the area under the plasma concentration- time curve extrapolated to infinity.
  • AUCo-t refers to the area under the plasma concentration- time curve from time 0 to the last time point with a quantifiable plasma concentration, usually about 12-36 hours.
  • AUCo- ⁇ refers to the area under the plasma concentration- time curve from time 0 to the time of the next dose.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • ACN acetonitrile
  • the present application further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage forms as described herein.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application further provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage as described herein.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 500 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 400 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ - 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • oral administration of one or more sustained release dosage forms to a fasted individual provides a mean time to peak plasma
  • Tmax concentration of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrileof about 0.5 hours to about 3 hours.
  • oral administration of one or more sustained release dosage forms to a fasted individual provides a mean time to peak plasma
  • Tmax concentration of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile of at least 0.5 hours.
  • oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma
  • Cmax concentration to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 5 to about 50.
  • oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma
  • Cmax concentration to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 9 to about 40.
  • oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma
  • Cmax concentration to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 15 to about 30.
  • oral administration of one or more sustained release dosage forms to a fasted individual provides a mean half-life (ti/ 2 ) of ⁇ l- ⁇ l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrileof about 1 hour to about 20 hours.
  • oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of ⁇ l- ⁇ l-[3-fluoro-2-
  • oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a mean time to peak plasma concentration (Tmax) of ⁇ l- ⁇ l-[3-fluoro-2-
  • oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of ⁇ 1- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 10 to about 70.
  • Cmax mean peak plasma concentration
  • Si2h mean 12-hour plasma concentration
  • oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h)of ⁇ l- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 15 to about 50.
  • Cmax mean peak plasma concentration
  • Ca2h mean 12-hour plasma concentration
  • oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of ⁇ l- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 25 to about 45.
  • Cmax mean peak plasma concentration
  • Ca2h mean 12-hour plasma concentration
  • oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean half-life (ti/ 2 ) of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 1 hour to about 7 hours.
  • oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean half-life (ti/ 2 ) of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile of about 2 hours to about 5 hours.
  • the one or more sustained release dosage forms are each a tablet. In some embodiments, the one or more sustained release dosage forms are prepared by process comprising wet granulation.
  • the one or more sustained release dosage forms each comprises one or more hypromelloses. In some embodiments, the one or more sustained release dosage forms each comprises one or more excipients independently selected from hypromelloses and microcrystalline celluloses. In some embodiments, the one or more sustained release dosage forms each comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, and lactose monohydrate.
  • the one or more sustained release dosage forms each comprises a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP and a second hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP.
  • the one or more sustained release dosage forms each comprises about 10% to about 15% by weight of one or more hypromelloses. In some embodiments, the one or more sustained release dosage forms each comprises about 16% to about 22% by weight of microcrystalline cellulose. In some embodiments, the one or more sustained release dosage forms each comprises about 45% to about 55% by weight of lactose monohydrate. In some embodiments, the one or more sustained release dosage forms each comprises about 0.3% to about 0.7% by weight of magnesium stearate.
  • the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-
  • the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of ⁇ 1- ⁇ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
  • TSS total symptom score
  • the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 500 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
  • TSS total symptom score
  • the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 400 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
  • TSS total symptom score
  • the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ - 3-[4-(7H ⁇ yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • total symptom score refers to the TSS derived from the modified Myelofibrosis Symptom Assessment Form (MFSAF) (e.g., v3.0) electronic diary as compared with baseline (baseline is the patient's baseline TSS before treatment).
  • MFSAF Myelofibrosis Symptom Assessment Form
  • baseline baseline is the patient's baseline TSS before treatment.
  • myelofibrosis is primary myelofibrosis (PMF), post-polycythemia vera MF, or post-essential thrombocythemia MF.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 500 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia.
  • the present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 400 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia.
  • the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of ⁇ 1- ⁇ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of ⁇ 1 - ⁇ 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, are provided.
  • the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, is provided.
  • Reduced anemia is relative to that experienced for a twice-daily dose of 200 mg on a free base basis of ⁇ l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising ⁇ l- ⁇ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable
  • the compound of Formula I is a JAK inhibitor.
  • a JAKl selective inhibitor is a compound that inhibits JAKl activity preferentially over other Janus kinases. JAKl plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states. For example, IL- 6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (Fonesca, J.E. et al, Autoimmunity Reviews, 8:538-42, 2009).
  • JAKl is mutated resulting in constitutive undesirable tumor cell growth and survival (Mullighan CG, Proc Natl Acad Sci U S A.106:9414-8, 2009; Flex E., et al.J Exp Med. 205:751-8, 2008).
  • JAK1 inhibition In other autoimmune diseases and cancers elevated systemic levels of inflammatory cytokines that activate JAK1 may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAK1 inhibition. Selective inhibitors of JAK1 may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases.
  • JAK1 erythropoietin
  • Tpo thrombopoietin
  • Epo is a key growth factor for red blood cells production; hence a paucity of Epo-dependent signaling can result in reduced numbers of red blood cells and anemia (Kaushansky K, NEJM 354:2034-45, 2006).
  • Tpo another example of a JAK2-dependent growth factor, plays a central role in controlling the proliferation and maturation of megakaryocytes - the cells from which platelets are produced (Kaushansky K, NEJM 354:2034-45, 2006). As such, reduced Tpo signaling would decrease megakaryocyte numbers (megakaryocytopenia) and lower circulating platelet counts (thrombocytopenia). This can result in undesirable and/or uncontrollable bleeding.
  • JAK3 and Tyk2 Reduced inhibition of other JAKs, such as JAK3 and Tyk2 may also be desirable as humans lacking functional version of these kinases have been shown to suffer from numerous maladies such as severe-combined immunodeficiency or hyperimmunoglobulin E syndrome (Minegishi, Y, et al.
  • JAK1 inhibitor with reduced affinity for other JAKs would have significant advantages over a less-selective inhibitor with respect to reduced side effects involving immune suppression, anemia and thrombocytopenia.
  • a JAK-associated disease can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the JAK, including overexpression and/or abnormal activity levels.
  • a JAK-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating JAK activity.
  • JAK-associated diseases include diseases involving the immune system including, for example, organ transplant rejection (e.g., allograft rejection and graft versus host disease).
  • organ transplant rejection e.g., allograft rejection and graft versus host disease.
  • JAK-associated diseases include autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, chronic obstructive pulmonary disease (COPD), and the like.
  • the autoimmune disease is an autoimmune bullous skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
  • JAK-associated diseases include allergic conditions such as asthma, food allergies, eszematous dermatitis, contact dermatitis, atopic dermatitis (atropic eczema), and rhinitis.
  • JAK-associated diseases include viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV, HTLV 1, Varicella-Zoster Virus (VZV) and Human Papilloma Virus (HPV).
  • EBV Epstein Barr Virus
  • Hepatitis B Hepatitis C
  • HIV HTLV 1
  • VZV Varicella-Zoster Virus
  • HPV Human Papilloma Virus
  • JAK-associated disease examples include diseases associated with cartilage turnover, for example, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome, costal athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease, degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, or ankylosing spondylitis.
  • diseases associated with cartilage turnover for example, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome, costal athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease, degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, or ankylosing spondylitis.
  • JAK-associated disease examples include congenital cartilage malformations, including hereditary chrondrolysis, chrondrodysplasias, and pseudochrondrodysplasias (e.g., microtia, enotia, and metaphyseal
  • JAK-associated diseases or conditions include skin disorders such as psoriasis (for example, psoriasis vulgaris), atopic dermatitis, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis).
  • skin disorders such as psoriasis (for example, psoriasis vulgaris), atopic dermatitis, 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 at least one JAK inhibitor of the invention together with the agent causing unwanted sensitization can be helpful in treating such unwanted sensitization or dermatitis.
  • the skin disorder is treated by topical administration of at least one JAK inhibitor of the invention.
  • the JAK-associated disease is cancer including those characterized by solid tumors (e.g., prostate cancer, renal cancer, hepatic cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, Castleman's disease, uterine leiomyosarcoma, melanoma etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and cutaneous B-cell lymphoma.
  • CTCLs include Sezary syndrome and mycosis fungoides.
  • the dosage forms described herein, or in combination with other JAK inhibitors can be used to treat inflammation- associated cancers.
  • the cancer is associated with inflammatory bowel disease.
  • the inflammatory bowel disease is ulcerative colitis.
  • the inflammatory bowel disease is Crohn's disease.
  • the inflammation-associated cancer is colitis-associated cancer.
  • the inflammation-associated cancer is colon cancer or colorectal cancer.
  • the cancer is gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), adenocarcinoma, small intestine cancer, or rectal cancer.
  • JAK-associated diseases can further include those characterized by expression of: JAK2 mutants such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F); JAK2 mutants having at least one mutation outside of the pseudo-kinase domain; JAK1 mutants; JAK3 mutants; erythropoietin receptor (EPOR) mutants; or deregulated expression of CRLF2.
  • JAK2 mutants such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F); JAK2 mutants having at least one mutation outside of the pseudo-kinase domain; JAK1 mutants; JAK3 mutants; erythropoietin receptor (EPOR) mutants; or deregulated expression of CRLF2.
  • JAK-associated diseases can further include myeloproliferative disorders (MPDs) such as polycythemia vera (PV), essential thrombocythemia (ET), myelofibrosis with myeloid metaplasia (MMM), primary myelofibrosis (PMF), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hypereosinophilic syndrome (HES), systemic mast cell disease (SMCD), and the like.
  • MPDs myeloproliferative disorders
  • PV polycythemia vera
  • ET essential thrombocythemia
  • MMM myelofibrosis with myeloid metaplasia
  • PMF primary myelofibrosis
  • CML chronic myelogenous leukemia
  • CMML chronic myelomonocytic leukemia
  • HES hypereosinophilic syndrome
  • SMCD systemic mast cell disease
  • the myeloproliferative disorder is myelofibrosis (e.g., primary myelofibrosis (PMF) or post polycythemia vera/essential thrombocythemia myelofibrosis (Post-PV/ET MF)).
  • the myeloproliferative disorder is post- essential thrombocythemia myelofibrosis (Post-ET).
  • the myeloproliferative disorder is post polycythemia vera myelofibrosis (Post-PV MF).
  • dosage forms described herein can be used to treat pulmonary arterial hypertension.
  • the present invention further provides a method of treating dermatological side effects of other pharmaceuticals by administration of the dosage forms 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 dosage forms of the invention can be administered systemically in combination with (e.g., simultaneously or sequentially) the pharmaceutical agent having the undesirable dermatological side effect.
  • JAK-associated diseases include inflammation and inflammatory diseases.
  • Example inflammatory diseases include sarcoidosis, inflammatory diseases of the eye (e.g., ulceris, uveitis, scleritis, conjunctivitis, or related disease),
  • the inflammation disease of the eye is blepharitis.
  • the dosage forms described herein can further be used to treat ischemia reperfusion injuries or a disease or condition related to an inflammatory ischemic event such as stroke or cardiac arrest.
  • the dosage forms described herein can further be used to treat endotoxin-driven disease state (e.g., complications after bypass surgery or chronic endotoxin states contributing to chronic cardiac failure).
  • the dosage forms described herein can further be used to treat anorexia, cachexia, or fatigue such as that resulting from or associated with cancer.
  • the dosage forms described herein can further be used to treat restenosis, sclerodermitis, or fibrosis.
  • the dosage forms described herein can further be used to treat conditions associated with hypoxia or astrogliosis such as, for example, diabetic retinopathy, cancer, or neurodegeneration. See, e.g., Dudley, A.C. et al. Biochem. J. 2005, 390(Pt 2):427-36 and Sriram, K. et al. J. Biol. Chem. 2004, 279(19): 19936-47. Epub 2004 Mar 2, both of which are incorporated herein by reference in their entirety.
  • the JAK inhibitors described herein can be used to treat Alzheimer's disease.
  • the dosage forms described herein can further be used to treat other inflammatory diseases such as systemic inflammatory response syndrome (SIRS) and septic shock.
  • SIRS systemic inflammatory response syndrome
  • the dosage forms described herein can further be used to treat gout and increased prostate size due to, e.g., benign prostatic hypertrophy or benign prostatic hyperplasia.
  • JAK-associated diseases include bone resorption diseases such as osteoporosis, osteoarthritis. Bone resorption can also be associated with other conditions such as hormonal imbalance and/or hormonal therapy, autoimmune disease (e.g. osseous sarcoidosis), or cancer (e.g. myeloma).
  • the reduction of the bone resorption due to the the compound of Formula I can be about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, or about 90%.
  • the dosage forms described herein can further be used to treat a dry eye disorder.
  • dry eye disorder is intended to encompass the disease states summarized in a recent official report of the Dry Eye Workshop (DEWS), which defined dry eye as "a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface. It is accompanied by increased osmolarity of the tear film and inflammation of the ocular surface.” Lemp, “The Definition and Classification of Dry Eye Disease: Report of the Definition and Classification Subcommittee of the International Dry Eye Workshop", The Ocular Surface, 5(2), 75-92 April 2007, which is incorporated herein by reference in its entirety.
  • the dry eye disorder is selected from aqueous tear- deficient dry eye (ADDE) or evaporative dry eye disorder, or appropriate
  • the dry eye disorder is Sjogren syndrome dry eye (SSDE). In some embodiments, the dry eye disorder is non- Sjogren syndrome dry eye (NSSDE).
  • the present invention provides a method of treating conjunctivitis, uveitis (including chronic uveitis), chorioditis, retinitis, cyclitis, sclieritis, episcleritis, or ulceris; treating inflammation or pain related to corneal transplant, LASIK (laser assisted in situ keratomileusis), photorefractive keratectomy, or LASEK (laser assisted sub-epithelial keratomileusis); inhibiting loss of visual acuity related to corneal transplant, LASIK, photorefractive keratectomy, or LASEK; or inhibiting transplant rejection in a patient in need thereof, comprising administering to the patient a dosage form of the invention.
  • the dosage forms of the invention can be used to treat respiratory dysfunction or failure associated with viral infection, such as influenza and SARS.
  • the present invention provides a dosage form as described in any of the embodiments herein, for use in a method of treating any of the diseases or disorders described herein. In some embodiments, the present invention provides the use of a dosage form as described in any of the embodiments herein, for the preparation of a medicament for use in a method of treating any of the diseases or disorders described herein.
  • the present invention provides a dosage form as described herein, or a pharmaceutically acceptable salt thereof, for use in a method of modulating JAKl .
  • the present invention also provides use of a dosage form as described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in a method of modulating JAKl.
  • the term "individual” is a human. In some embodiments, the human is an adult subject.
  • treating refers to one or more of (1) 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 (2) 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.
  • 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, which is incorporated herein by reference in its entirety, or other agents can be used in combination with the dosage forms described herein for treatment of JAK-associated diseases, disorders or conditions.
  • the one or more additional pharmaceutical agents can be administered to a patient
  • Example chemotherapeutics 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 coriticosteroids 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.
  • Example suitable Flt-3 inhibitors include compounds, and their
  • Example suitable RAF inhibitors include compounds, and their
  • Example suitable FAK inhibitors include compounds, and their
  • one or more of the dosage forms 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.
  • one or more dosage forms of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, such as multiple myeloma, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects.
  • additional pharmaceutical agents used in the treatment of multiple myeloma can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib).
  • Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. Additive or synergistic effects are desirable outcomes of combining a dosage form of the present invention with an additional agent.
  • agents such as dexamethasone may be reversible upon treatment with a dosage form of the present invention.
  • the agents can be combined with the present compounds in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • a corticosteroid such as dexamethasone is administered to a patient in combination with at the dosage form of the invention where the dexamethasone is administered intermittently as opposed to continuously.
  • combinations of one or more JAK inhibitors of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
  • the additional therapeutic agent is fluocinolone acetonide (Retisert®), or rimexolone (AL-2178, Vexol, Alcon).
  • the additional therapeutic agent is cyclosporine (Restasis®). In some embodiments, the additional therapeutic agent is a corticosteroid. In some embodiments, the corticosteroid is triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
  • the additional therapeutic agent is selected from DehydrexTM (Holies Labs), Civamide (Opko), sodium hyaluronate (Vismed,
  • the additional therapeutic agent is an anti-angiogenic agent, cholinergic agonist, TRP- 1 receptor modulator, a calcium channel blocker, a mucin secretagogue, MUC 1 stimulant, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, an mTOR inhibitor, another JAK inhibitor, Bcr-Abl kinase inhibitor, Flt-3 kinase inhibitor, RAF kinase inhibitor, and FAK kinase inhibitor such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety.
  • the additional therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycline). In some embodiments, the additional therapeutic agent binds to FKBP12. In some embodiments, the additional therapeutic agent is an alkylating agent or DNA cross-linking agent; an anti-metabolite/demethylating agent (e.g., 5- flurouracil, capecitabine or azacitidine); an anti-hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e.g.
  • an anti-metabolite/demethylating agent e.g., 5- flurouracil, capecitabine or azacitidine
  • an anti-hormone therapy e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor
  • a mitotic inhibitor e.g.
  • vincristine or paclitaxel paclitaxel
  • an topoisomerase (I or II) inhibitor e.g. mitoxantrone and irinotecan
  • an apoptotic inducers e.g. ABT-737
  • a nucleic acid therapy e.g.
  • RNAi nuclear receptor ligands
  • nuclear receptor ligands e.g., agonists and/or antagonists: all- trans retinoic acid or bexarotene
  • epigenetic targeting agents such as histone deacetylase inhibitors (e.g. vorinostat), hypomethylating agents (e.g. decitabine); regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
  • the additional therapeutic agent(s) are demulcent eye drops (also known as "artificial tears"), which include, but are not limited to, compositions containing polyvinylalcohol, hydroxypropyl methylcellulose, glycerin, polyethylene glycol (e.g. PEG400), or carboxymethyl cellulose. Artificial tears can help in the treatment of dry eye by compensating for reduced moistening and lubricating capacity of the tear film.
  • the additional therapeutic agent is a mucolytic drug, such as N-acetyl-cysteine, which can interact with the mucoproteins and, therefore, to decrease the viscosity of the tear film.
  • the additional therapeutic agent includes an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and nonsteroidal anti-inflammatories, and anti-allergic agents.
  • suitable medicaments include aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine; sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin; colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives ("rifampins"); cycloserine; beta-lactams; cephalosporins; amphoterici
  • Sustained release tablets were prepared with the excipients being in the amounts shown in the table below.
  • Protocol A was used for the SRI tablets
  • protocol B was used for the SR2 tablets
  • Protocol C was used for the SR3 tablets and the 25 mg SR tablets
  • Protocol D was used for the SR4 tablets.
  • Step 1 Individually screen the adipic acid salt of the compound of
  • Formula I microcrystalline cellulose, hypromelloses (Methocel K100 LV and Methocel K4M), and lactose monohydrate.
  • Step 2 Transfer the screened material from Step 1 to a suitable blender and mix.
  • Step 3 Transfer the blend from Step 2 to a suitable granulator and mix.
  • Step 4 Add purified water while mixing.
  • Step 5 Transfer the granules from Step 4 into a suitable dryer and dry until LOD is less than 3%.
  • Step 6 Screen the granules from Step 5.
  • Step 7 Mix screened Magnesium Stearate with granules in Step 6 in a suitable blender.
  • Step 8 Compress the final blend in Step 7 on a suitable rotary tablet press.
  • Step 1 Individually screen the adipic acid salt of the compound of
  • Formula I microcrystalline cellulose, hypromellose and pregelatinized starch.
  • Step 2 Transfer the screened material from Step 1 to a suitable blender and mix.
  • Step 3 Transfer the blend from Step 2 to a suitable granulator and mix.
  • Step 4 Add purified water while mixing.
  • Step 5 Transfer the granules from Step 4 into a suitable dryer and dry until LOD is less than 3%.
  • Step 6 Screen the granules from Step 5.
  • Step 7 Individually screened polyox, butylated hydroxytoluene and colloidal silicone dioxide.
  • Step 8 Transfer the granules from Step 6 and material from Step 7 into a suitable blender and mix.
  • Step 9 Add screened Magnesium Stearate to the material in Step 8 and continue blending.
  • Step 10 Compress the final blend in Step 9 on a suitable rotary tablet press.
  • Step 1 Individually screen lactose monohydrate, the adipic acid salt of the compound of Formula I, microcrystalline cellulose and hypromelloses through a suitable screen.
  • Step 2 Transfer the screened material from Step 1 to a suitable blender and mix.
  • Step 3 Transfer the blend from Step 2 to a suitable granulator and mix.
  • Step 4 Add purified water while mixing.
  • Step 5 Screen wet granules through a suitable screen.
  • Step 6 Transfer the granules from Step 5 into a suitable dryer and dry until LOD is less than 3%.
  • Step 7 Mill the granules from Step 6.
  • Step 8 Mix screened magnesium stearate with granules in Step 7 in a suitable blender.
  • Step 9 Compress the final blend in Step 8 on a suitable rotary tablet press.
  • Protocol D
  • Step 1 Individually screen pregelatinized starch, the adipic acid salt of the compound of Formula I, hypromellose, and a portion of required microcrystalline cellulose through a suitable screen.
  • Step 2 Transfer the screened material from Step 1 to a suitable blender and mix.
  • Step 3 Transfer the blend from Step 2 to a suitable granulator and mix.
  • Step 4 Add purified water while mixing.
  • Step 5 Screen wet granules through a suitable screen.
  • Step 6 Transfer the granules from Step 5 into a suitable dryer and dry until LOD is less than 3%.
  • Step 7 Mill the granules from Step 6.
  • Step 8 Screen the remaining portion of microcrystalline cellulose and half of the sodium bicarbonate.
  • Step 9 Transfer the milled granules from Step 7 and screened materials from Step 8 into a suitable blender and mix.
  • Step 10 Screen the remaining portion of sodium bicarbonate and mix with blend in Step 9.
  • Step 11 Screen magnesium stearate and mix with blend in Step 10.
  • Step 12 Compress the final blend in Step 1 1 on a suitable rotary tablet press.
  • the IR formulation used in the studies in Example 3 was prepared as 50 mg capsules with the composition shown in the table below according to Protocol E below.
  • Step 1 Pre-mix the required amount of the adipic acid salt of the compound of
  • Step 2 Pass the mixture in Step 1 through a suitable screen (for example 40 mesh).
  • Step 3 Screen the remaining SMCC through the same screen used in Step 2.
  • Step 4 Blend the screened SMCC from Step 3 along with mixture from Step 2 in a suitable blender (for example Turbula blender) for approximately 5 minutes.
  • Step 5. Fill the blend into capsules to desired fill weight.
  • a total of 72 healthy adult subjects were enrolled in 6 cohorts (12 subjects per cohort) and randomized to treatment sequences within each cohort according to a randomization schedule. All treatments were single-dose administrations of the compound of Formula I. There was a washout period of 7 days between the treatment periods.
  • the SRI, SR2, SR3, and SR4 formulations were evaluated in Cohort 1, Cohort 2, Cohort 3, and Cohort 4, respectively (see Example 1 for SRI, SR2, SR3, SR4, and 25 mg SR tablets used in study).
  • the subjects received the IR and SR treatments according to a 3 -way crossover design:
  • Treatment A 300 mg (6 x 50 mg capsule) IR formulation of the compound of Formula I administered orally after an overnight fast of at least 10 hours.
  • Treatment B 300 mg (3 x 100 mg tablets) SR formulation of the compound of Formula I administered orally after an overnight fast of at least 10 hours.
  • Treatment C 300 mg (3 x 100 mg tablets) SR formulation of the compound of Formula I administered orally after a high-fat meal.
  • Treatment A 300 mg (3 x 100 mg tablets of the compound of Formula I) SR3 administered orally after an overnight fast of at least 10 hours.
  • Treatment B 300 mg (3 x 100 mg tablets of the compound of Formula I) SR3 administered orally after a medium-fat meal.
  • Treatment A 50 mg (2 x 25 mg tablets of the compound of Formula I (25 mg SR tablets from Example 1)) administered orally after an overnight fast of at least 10 hours.
  • Treatment B 50 mg (2 x 25 mg tablets of the compound of Formula I (25 mg SR tablets from Example 1)) administered orally after a high-fat meal.
  • Treatment C 100 mg (1 x 100 mg tablets) SR3 administered orally after an overnight fast of at least 10 hours.
  • Blood samples for determination of plasma concentrations of the compound of Formula I were collected using lavender top (K2EDTA) Vacutainer® tubes at 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose.
  • Plasma samples were assayed by a validated, GLP, LC/MS/MS method with a linear range of 5.0 to 5000 nM.
  • Table 1 summarizes the accuracy and precision (CV %) of the assay quality control samples during the analysis of the plasma samples from this study.
  • CV% percent coefficient of variability
  • QC quality control
  • Theo theoretical or nominal concentration.
  • the actual sample collection times were used. For any sample with missing actual collection time, the scheduled time was used provided that there was no protocol deviation noted for the collection of these samples.
  • Standard noncompartmental PK methods were used to analyze the data for the plasma concentration of the compound of Formula using Phoenix WinNonlin version 6.0 (Pharsight Corporation, Mountain View, CA). Thus, Cmax and Tmax were taken directly from the observed plasma concentration data.
  • the terminal-phase disposition rate constant ( ⁇ ⁇ ) was estimated using a log-linear regression of the concentration data in the terminal disposition phase, and t1 ⁇ 2 was estimated as 1 ⁇ (2)/ ⁇ ⁇ .
  • AUCo-t was estimated using the linear trapezoidal rule for increasing concentrations and the log- trapezoidal rule for decreasing concentrations, and the total AUCo- ⁇ was calculated as AUCo-t + Ct/ ⁇ .
  • the oral-dose clearance (CL/F) was estimated as Dose/AUCo- ⁇
  • the terminal-phase volume of distribution V F) was estimated as Dose/[AUCo- ⁇ * z].
  • FIG. 1 presents plasma concentrations of the compound of Formula I (mean ⁇ SE) for the subjects in Cohorts 1 to 4 following Treatment A (300 mg IR
  • FIG. 2 compares the effect of a high-fat meal and medium- fat meal on the mean PK profile following a single-dose 300 mg (3 x lOOmg) administration of the compound of Formula I SR3 tablets.
  • FIG. 3 presents plasma concentrations of the compound of Formula I (mean ⁇ SE) for the subjects in Cohort 6 following Treatment A (2 x 25 mg SR tablet administration in fasted state), Treatment B (2 x 25 mg SR tablet with a high-fat meal), and Treatment C (1 x 100 mg SR3 administration in fasted state).
  • Table 4A and 4B summarize mean PK parameters for subjects in Cohort 5, and food effect (medium- fat meal) for the 100 mg strength SR3 tablet.
  • PK parameter values are mean ⁇ SD and geometric mean except for T where median (90%
  • PK parameter values are mean ⁇ SD and geometric mean except for T where median (90% confidence interval) is reported. Table 4A
  • Pharmacokinetic parameter values are mean ⁇ SD and geometric mean except for Tmax, where median (90% confidence interval) is reported.
  • Pharmacokinetic parameter values are mean ⁇ SD and geometric mean except for T m ax, where median
  • PK parameter values are mean ⁇ SD and geometric mean except for Tmax, where median (90% confidence interval) is reported.
  • PK parameter values are mean ⁇ SD and geometric mean except for T m ax, where median (90% confidence interval) is reported.
  • the observed plasma median Tmax values were moderately prolonged (by 0.3 to 1.5 hours) with significantly reduced mean Cmax values (the upper bounds of the 90% CI for the geometric mean Cmax ratios were ⁇ 30%), suggesting decreased absorption rate of the compound of Formula I for the SR tablets.
  • the apparent mean disposition t1 ⁇ 2 observed in the terminal phase was significantly longer, ranging from 7.3 to 1 1 hours for SR1-SR4, as compared to about 2 hours for the IR capsule, indicating that the systemic elimination of the compound of Formula I was likely rate-limited by its absorption, which was sustained in the terminal disposition phase.
  • the Cmax/ Ci2h ratios were significantly lower for the SR tablets compared to the IR capsule for the same subjects studied.
  • the geometric mean Cmax/Ci2h ratios were 1 1.6-, 8.6-, 19.3-, and 8.9-fold, respectively, for SRI, SR2, SR3, and SR4 tablets, as compared to 1 12- to 162-fold for the IR capsules administered in the fasted state.
  • the 4 SR tablets showed reduced relative bioavailability compared to the IR capsule dosed in the same subjects.
  • the percent geometric mean ratios (90% CI) of Cmax were 14.2 % (11.4%-17.5%), 8.9% (6.7%- 1 1.9%), 22.3% (17.4%-28.6%) and 9.0% (6.8%-11.9%) for SRI, SR2, SR3, and SR4, respectively.
  • the percent geometric mean ratios (90% CI) of AUCo - ⁇ were 36.1 % (33.3%-39.2%), 26.0% (21.6%-31.3%), 47.5% (41.9%-53.9%), and 28.5% (23.2%- 35.1%) for SRI, SR2, SR3, and SR4, respectively.
  • SR3 and SRI demonstrated the best and second best relative bioavailability, respectively, among the SR formulations tested.
  • the intersubject variability as measured by percent coefficient of variability (CV%) in plasma exposure was significantly higher for the gastroretentive formulation SR4, but comparable among the 3 regular SR tablets designed for intestinal release.
  • the intersubject CV% for the 100 mg SRI tablet was 39% and 33% for Cmax and respectively.
  • the intersubject CV% for the 100 mg SR2 tablet was 50% and 37% for Cmax and AUCo - ⁇ , respectively.
  • the intersubject CV% for the 100 mg SR3 tablet was 43% and 29% for Cmax and respectively.
  • the intersubject CV% for the 100 mg SR4 tablet was 83% and 73% for Cmax and AUCo-oo, respectively.
  • SR3 delivered a mean relative bioavailability of 48% and is likely to be associated with a moderate food effect.
  • the SR3 tablet (3 x 100 mg) was dosed with a medium- fat meal (which is a more typical daily diet)
  • the observed increase in geometric mean AUCo ⁇ was only 17%, suggesting that this formulation may be administered without regard to medium- or low-fat meals. From the perspective of avoiding significant food effect, SR3 is superior to the other formulations.
  • Part 2 of the study was randomized, double-blind, placebo controlled (sponsor unblinded) with treatment for 84 days.
  • the ACR scores for the 600 mg QD are unprecedented as compared to other JAK inhibitors that are approved for treatment of RA.
  • the approved product for tofacitinib citrate (5 mg BID) showed much lower ACR scores at 3 months: 59% (ACR20), 31% (ACR50), and 15% (ACR70) (Table 5 of XELJANZ® - tofacitinib citrate tablet - label).
  • the percent change from baseline for hemoglobin was also studied for each of the dosing regimens (FIG. 4).
  • the 200 mg BID dose showed a drop away from the baseline compared to the other doses which tended to stay close to the placebo levels.
  • the 600 mg QD dose did not show the same downward trend as shown for the BID dose.
  • the once-daily dosing 600 mg QD did not compromise efficacy compared with the BID doses. This indicates that the once-daily dosing (such as 600 mg QD) may achieve maximal efficacy without inducing side-effects such anemia.
  • the once-daily dosing such as 600 mg QD
  • the 600 mg QD dose has robust efficacy with trivial change in hemoglobin levels. It is believed that this efficacy/side-effect profile may be due to the QD dose achieving maximal JAKl signaling (tied to efficacy) with low JAK2 inhibition at the trough, as JAK2 signaling is tied to hematopoiesis. This hypothesis is supported by the PK derived JAKl (IL-6) and JAK2 (TPO) inhibition data for the compound of Formula at various doses (Table 7).
  • the 600 mg QD dose showed similar average IL-6 inhibition to the 200 mg BID and 400 mg BID doses (61% versus 64% and 69%), but lower trough TPO inhibition in comparison to the 200 mg BID and 400 mg BID doses (4% versus 13% and 16%).
  • the trough IL-6 inhibition for the 600 mg QD dose is also lower than the trough IL-6 inhibition for the 200 mg BID and 400 mg BID doses, which suggests that there may be a reduction in infection from the QD dose.
  • PASI 50 is Psoriasis Area and Severity Index.
  • the 5 mg tofacitinib dose is the approved dosage level for RA for safety reasons in the US.
  • FIG. 5(a)-(b) show interim results with respect to proportion of subjects with > 50% reduction in total symptom score (TSS) in each dose group per the modified Myelofibrosis Symptom Assessment Form (MFSAF) v3.0 electronic diary at week 12 compared with baseline (The modified MFSAF v3.0 comprises 19 questions assessing MF-related symptoms on a scale of 0 (absent) to 10 (worst imaginable)).
  • FIG. 5(a) depicts the percentage of patients having a > 50% reduction in TSS at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) (patients who discontinued prior to the week 12 visit were considered nonresponders).
  • FIG. 6(a) depicts mean hemoglobin levels (g/dL) over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) (interim results of study for all patients).
  • FIG. 6(b) depicts mean hemoglobin levels (g/dL) over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) at 48 weeks.
  • FIG. 6(c) depicts mean hemoglobin levels (g/dL) over time by dose cohort at 48 weeks as an average for three dose cohorts as compared to individuals dosed with placebo or ruxolitinib (ruxolitinib was dosed according to the label for Jakafi®). The data show an increase in hemoglobin levels for the 600 mg QD dose.
  • Table 9 below show interim hematology laboratory results (new and worsening) for each dose cohort.
  • Table 9a shows the hematology laboratory results (new and worsening) for each dose cohort after long exposure.
  • the compound of Formula I herein was tested for inhibitory activity of JAK targets according to the following in vitro assay described in Park et ah, Analytical Biochemistry 1999, 269, 94-104.
  • the catalytic domains of human JAKl (a.a. 837- 1 142) and JAK2 (a.a. 828-1132) with an N-terminal His tag were expressed using baculovirus in insect cells and purified.
  • the catalytic activity of JAKl and JAK2 was assayed by measuring the phosphorylation of a biotinylated peptide.
  • phosphorylated peptide was detected by homogenous time resolved fluorescence (HTRF).
  • IC50S of compounds were measured for each kinase in the 40 microL reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8) buffer with 100 mM NaCl, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA.
  • ATP concentration in the reactions was 1 mM.
  • Reactions were carried out at room temperature for 1 hr and then stopped with 20 ⁇ ⁇ 45 mM EDTA, 300 nM SA-APC, 6 nM Eu-Py20 in assay buffer (Perkin Elmer, Boston, MA).
  • Cancer cell lines dependent on cytokines and hence JAK/STAT signal transduction, for growth can be plated at 6000 cells per well (96 well plate format) in RPMI 1640, 10% FBS, and 1 nG/niL of appropriate cytokine.
  • Compounds can be added to the cells in DMSO/media (final concentration 0.2% DMSO) and incubated for 72 hours at 37 °C, 5% CO2.
  • the effect of compound on cell viability is assessed using the CellTiter-Glo Luminescent Cell Viability Assay (Promega) followed by TopCount (Perkin Elmer, Boston, MA) quantitation. Potential off-target effects of compounds are measured in parallel using a non-JAK driven cell line with the same assay readout. All experiments are typically performed in duplicate.
  • the above cell lines can also be used to examine the effects of compounds on phosphorylation of JAK kinases or potential downstream substrates such as STAT proteins, Akt, Shp2, or Erk. These experiments can be performed following an overnight cytokine starvation, followed by a brief preincubation with compound (2 hours or less) and cytokine stimulation of approximately 1 hour or less. Proteins are then extracted from cells and analyzed by techniques familiar to those schooled in the art including Western blotting or ELISAs using antibodies that can differentiate between phosphorylated and total protein. These experiments can utilize normal or cancer cells to investigate the activity of compounds on tumor cell survival biology or on mediators of inflammatory disease.
  • cytokines such as IL-6, IL-12, IL-23, or IFN can be used to stimulate JAK activation resulting in phosphorylation of STAT protein(s) and potentially in transcriptional profiles (assessed by array or qPCR technology) or production and/or secretion of proteins, such as IL-17.
  • the ability of compounds to inhibit these cytokine mediated effects can be measured using techniques common to those schooled in the art.
  • JAK2V617F mutation found in myeloid proliferative disorders.
  • These experiments often utilize cytokine dependent cells of hematological lineage (e.g. BaF/3) into which the wild- type or mutant JAK kinases are ectopically expressed (James, C, et al. Nature 434: 1144-1148; Staerk, J., et al. JBC 280:41893-41899).
  • Endpoints include the effects of compounds on cell survival, proliferation, and phosphorylated JAK, STAT, Akt, or Erk proteins.
  • PBMCs Peripheral blood mononuclear cells
  • Freshly isolated human T-cells can be maintained in culture medium (RPMI 1640 supplemented with 10% fetal bovine serum, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin) at a density of 2 x 10 6 cells/ml at 37 °C for up to 2 days.
  • T-cells are first treated with Phytohemagglutinin (PHA) at a final concentration of 10 ⁇ g/mL for 72h. After washing once with PBS, 6000 cells/well are plated in 96-well plates and treated with compounds at different concentrations in the culture medium in the presence of 100 U/mL human IL-2 (ProSpec-Tany TechnoGene; Rehovot, Israel). The plates are incubated at 37 °C for 72h and the proliferation index is assessed using CellTiter-Glo Luminescent reagents following the manufactory suggested protocol (Promega;
  • Example C In vivo anti-tumor efficacy
  • Compounds herein can be evaluated in human tumor xenograft models in immune compromised mice.
  • a tumorigenic variant of the ⁇ -6 plasmacytoma cell line can be used to inoculate SCID mice subcutaneous ly (Burger, R., et al. Hematol J. 2:42-53, 2001).
  • Tumor bearing animals can then be randomized into drug or vehicle treatment groups and different doses of compounds can be administered by any number of the usual routes including oral, i.p., or continuous infusion using implantable pumps. Tumor growth is followed over time using calipers.
  • tumor samples can be harvested at any time after the initiation of treatment for analysis as described above (Example B) to evaluate compound effects on JAK activity and downstream signaling pathways.
  • selectivity of the compound(s) can be assessed using xenograft tumor models that are driven by other know kinases (e.g. Bcr-Abl) such as the K562 tumor model.
  • Bcr-Abl know kinases
  • 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 Jan; 19(l):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 Jan;38(l-2): 1 16-21).
  • 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 ⁇ (10 ⁇ . on the internal pinna and 10 ⁇ . 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 compounds 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 either systemically or topically (topical application of the treatment to the ears). Efficacies of the test compounds are indicated by a reduction in ear swelling comparing to the situation without the treatment. Compounds causing a reduction of 20% or more were 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 compounds 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 compounds a clinically efficacious treatment for psoriasis
  • dexamethasone a clinically efficacious treatment for psoriasis
  • Test compounds and the dexamethasone can produce similar transcriptional changes both qualitatively and quantitatively, and both the test compounds and dexamethasone can reduce the number of infiltrating cells.
  • Both systemically and 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.
  • Example E In vivo anti-inflammatory activity
  • rodent models of arthritis can be used to evaluate the therapeutic potential of compounds dosed preventatively or therapeutically.
  • These models include but are not limited to mouse or rat collagen-induced arthritis, rat adjuvant-induced arthritis, and collagen antibody- induced arthritis.
  • Autoimmune diseases including, but not limited to, multiple sclerosis, type I-diabetes mellitus, uveoretinitis, thyroditis, myasthenia gravis, immunoglobulin nephropathies, myocarditis, airway sensitization (asthma), lupus, or colitis may also be used to evaluate the therapeutic potential of compounds herein.
  • These models are well established in the research community and are familiar to those schooled in the art (Current Protocols in Immunology, Vol 3., Coligan, J.E. et al, Wiley Press.; Methods in Molecular Biology: Vol. 225, Inflammation Protocols., Winyard, P.G. and Willoughby, D.A., Humana Press, 2003.).
  • Example F Animal Models for the Treatment of Dry Eye, Uveitis, and
  • Agents may be evaluated in one or more preclinical models of dry eye known to those schooled in the art including, but not limited to, the rabbit concanavalin A (ConA) lacrimal gland model, the scopolamine mouse model (subcutaneous or transdermal), the Botulinumn mouse lacrimal gland model, or any of a number of spontaneous rodent auto-immune models that result in ocular gland dysfunction (e.g. NOD-SCID, MRL/lpr, or ZB/NZW) (Barabino et al, Experimental Eye Research 2004, 79, 613-621 and Schrader et al, Developmental Opthalmology, Karger 2008, 41, 298-312, each of which is incorporated herein by reference in its entirety).
  • ConA rabbit concanavalin A
  • scopolamine mouse model subcutaneous or transdermal
  • Botulinumn mouse lacrimal gland model or any of a number of spontaneous rodent auto-immune models that result in ocular gland dysfunction (e
  • Endpoints in these models may include histopathology of the ocular glands and eye (cornea, etc.) and possibly the classic Schirmer test or modified versions thereof (Barabino et al.) which measure tear production. Activity may be assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists.
  • routes of administration e.g. systemic or topical
  • Agents may be evaluated in one or more preclinical models of uveitis known to those schooled in the art. These include, but are not limited to, models of experimental autoimmune uveitis (EAU) and endotoxin induced uveitis (EIU). EAU experiements may be performed in the rabbit, rat, or mouse and may involve passive or activate immunization. For instance, any of a number or retinal antigens may be used to sensitize animals to a relevant immunogen after which animals may be challenged ocuarly with the same antigen.
  • the EIU model is more acute and involves local or systemic administration of lipopolysaccaride at sublethal doses.
  • Endpoints for both the EIU and EAU models may include fundoscopic exam, histopathology amongst others. These models are reviewed by Smith et al. (Immunology and Cell Biology 1998, 76, 497-512, which is incorporated herein by reference in its entirety). Activity is assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists. Some models listed above may also develop scleritis/episcleritis, chorioditis, cyclitis, or ulceris and are therefore useful in investigating the potential activity of compounds for the therapeutic treatment of these diseases. Agents may also be evaluated in one or more preclinical models of conjunctivitis known those schooled in the art.
  • guinea-pig models include those utilizing active or passive immunization and/or immune challenge protocols with antigens such as ovalbumin or ragweed (reviewed in Groneberg, D.A., et al, Allergy 2003, 58, 1101-11 13, which is incorporated herein by reference in its entirety).
  • Rat and mouse models are similar in general design to those in the guinea- pig (also reviewed by Groneberg). Activity may be assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists. Endpoints for such studies may include, for example, histological, immunological, biochemical, or molecular analysis of ocular tissues such as the conjunctiva.
  • Example G In vivo protection of bone
  • Compounds may be evaluated in various preclinical models of osteopenia, osteoporosis, or bone resorption known to those schooled in the art.
  • ovariectomized rodents may be used to evaluate the ability of compounds to affect signs and markers of bone remodeling and/or density (W.S.S. Jee and W. Yao, J Musculoskel. Nueron. Interact., 2001, 1(3), 193-207, which is incorporated herein by reference in its entirety).
  • bone density and architecture may be evaluated in control or compound treated rodents in models of therapy (e.g.

Abstract

This invention relates to sustained release dosage forms comprising {1-{1-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, and doses and methods related thereto.

Description

SUSTAINED RELEASE DOSAGE FORMS FOR A JAK1 INHIBITOR
This application claims the benefit of priority of U.S. Prov. Appl. No.
61/863,325, filed August 7, 2013, and U.S. Prov. Appl. No. 61/913,066, filed
December 6, 2013, each of which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
This application relates to a sustained release dosage form comprising { 1- { 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, and doses and methods related thereto.
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 antiinflammatory 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-tyros ine 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, Jakl-/- 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.
JAK2 tyrosine kinase can be beneficial for patients with myeloproliferative disorders, e.g., polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM) (Levin, et al, Cancer Cell, vol. 7, 2005: 387- 397). Inhibition of the JAK2V617F kinase decreases proliferation of hematopoietic cells, suggesting JAK2 as a potential target for pharmacologic inhibition in patients with PV, ET, and MMM. Inhibition of the JAKs may benefit patients suffering from skin immune disorders such as psoriasis, and skin sensitization. The maintenance of psoriasis is believed to depend on a number of inflammatory cytokines in addition to various chemokines and growth factors (JCI, 1 13 : 1664-1675), many of which signal through JAKs (Adv Pharmacol. 2000;47: 113-74).
Due to the usefulness of compounds which inhibit JAK in targeting augmentation or suppression of the immune and inflammatory pathways (such as immunosuppressive agents for organ transplants), as well as the treatment of autoimmune diseases, diseases involving a hyperactive inflammatory response (e.g., eczema), allergies, cancer (e.g., prostate, leukemia, multiple myeloma), and some immune reactions (e.g., skin rash or contact dermatitis or diarrhea) caused by other therapeutics, there is a need for improved formulations for administering JAK kinases. The dosages forms described herein, as well as the doses and methods described supra are directed toward this need and other ends.
SUMMARY
JAK inhibitors are described in U.S. Serial No. 13/043,986 (US
201 1/0224190), filed March 9, 201 1, which is incorporated herein by reference in its entirety, including { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3 -yl} acetonitrile, which is depicted below as Formula I.
Figure imgf000005_0001
I The present application provides, inter alia, sustained-release dosage forms comprising about 25 mg to about 600 mg (e.g., 25 mg, 100 mg, 200 mg, 300 mg, or 600 mg) on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present invention further provides one or more sustained release dosage forms each comprising { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said one or more sustained release dosage forms together provide a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present invention also provides a dose, comprising one or more sustained release dosage forms each comprising { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said dose provides a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 600 mg on a free base basis of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application also provides a dose comprising one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 600 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage forms as described herein.
The present application also provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present application further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage as described herein.
The present application also provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
DESCRIPTION OF DRAWINGS
FIG. 1A-C depicts plasma concentrations for the compound of Formula I (Mean ± SE) in healthy subjects receiving single doses of 300 mg IR capsules (1A: Cohorts 1-4, fasted), SRI, SR2, SR3, and SR4 tablets (2B: Cohorts 1-4, fasted; and 2C: Cohorts 1-4, fed a high-fat meal).
FIG. 2A-B depicts single-dose 300 mg SR3 PK profiles (Mean ± SE) (2A: Cohort 3, SR3, fasted versus high-fat meal; and 2B: Cohort 5, SR3, fasted versus medium-fat meal).
FIG. 3 depicts a comparison of PK profiles (mean ± SE) between the 25 mg and 100 mg SR3 tablets (treatment A vs C) and the food effect of a high- fat meal on the 25 mg SR3 tablet (treatment B vs A).
FIG. 4 depicts the percent change from baseline for hemoglobin for several dosing regimens for sustained release tablets versus placebo.
FIG. 5(a) depicts the percentage of patients having a > 50% reduction in total symptom score (TSS) at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
FIG. 5(b) depicts the percent change in total symptom score (TSS) from baseline at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
FIG. 6(a) depicts mean hemoglobin levels over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
FIG. 6(b) depicts mean hemoglobin levels (g/dL) over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) at 48 weeks.
FIG. 6(c) depicts mean hemoglobin levels (g/dL) over time by dose cohort at 48 weeks as an average for three dose cohorts as compared to individuals dosed with placebo or ruxolitinib. DETAILED DESCRIPTION
The present application provides sustained-release dosage forms comprising { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the present application provides a sustained-release dosage form comprising about 25 mg to about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained-release dosage form comprises about 300 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained-release dosage form comprises about 200 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained-release dosage form comprises about 100 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained-release dosage form comprises about 300 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile adipic acid salt.
In some embodiments, the sustained-release dosage form comprises about 200 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile adipic acid salt.
In some embodiments, the sustained-release dosage form comprises about 100 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile adipic acid salt.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean peak plasma concentration (Cmax) of { 1- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 100 nM to about 1000 nM. As used in this context, oral administration means that a single dose is administered to the individual (in this case, 3 x 100 mg) and the PK parameter is calculated from the measurements of plasma concentration over time. In this context, the PK parameter (in this case, Cmax) is being used to characterize the single sustained release dosage form (i.e., the claims are directed to a single dosage form, not three dosage forms).
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean peak plasma concentration (Cmax) of { 1- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 400 nM to about 700 nM.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean time to peak plasma concentration (Tmax) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 0.5 hours to about 3 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean time to peak plasma concentration (Tmax) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of at least 0.5 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile of about 5 to about 50.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile of about 9 to about 40.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile of about 15 to about 30.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean half-life (ti/2) of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 5 hours to about 15 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean half-life (ti/2) of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 7 hours to about 12 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean half-life (ti/2) of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1 hour to about 20 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean bioavailability (AUCo -∞) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidm^
yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1000 nM*h to about 4000 nM*h.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to a fasted individual provides a mean bioavailability (AUCo -∞) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1500 nM*h to about 3100 nM*h.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean peak plasma concentration (Cmax) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 200 nM to about 2000 nM.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean peak plasma concentration (Cmax) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 500 nM to about 1500 nM.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of { l- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1 hour to about 9 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of { l- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of at least 1.5 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12- hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 10 to about 70.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12- hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 15 to about 50.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12- hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 25 to about 45.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1 hour to about 7 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 2 hours to about 5 hours.
In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean bioavailability (AUCo -∞) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 2000 nM*h to about 5000 nM*h. In some embodiments of the sustained-release dosage form comprising about 100 mg, oral administration of three of said dosage forms to an individual after a high-fat meal provides a mean bioavailability (AUCo -∞) of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 3000 nM*h to about 4000 nM*h.
In some embodiments, the percent geometric mean ratio of the sustained release dosage form relative to an immediate release dosage form for Cmax is about 15% to about 30%, wherein one or more immediate release dosage forms and one or more sustained release dosage forms are independently orally administered to fasted individuals as a single dose, wherein the same size dose of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt, is administered.
In some embodiments, the percent geometric mean ratio of the sustained release dosage form relative to an immediate release dosage form for Cmax is about 15% to about 30%, wherein one or more immediate release dosage forms and one or more sustained release dosage forms are independently orally administered to fasted individuals as a single dose, wherein the same size dose of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt, is administered.
In some embodiments, the percent geometric mean ratio of the sustained release dosage form relative to an immediate release dosage form for AUCo -∞ is about 40% to about 55%, wherein one or more immediate release dosage forms and one or more sustained release dosage forms are independently orally administered to fasted individuals as a single dose, wherein the same size dose of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt, is administered.
In some embodiments, the percent geometric mean ratio for Cmax of the sustained release dosage form orally administered to an individual after a high-fat meal relative to the sustained release dosage form orally administered to a fasted individual is about 150% to about 250%.
In some embodiments, the percent geometric mean ratio for AUCo -∞ of the sustained release dosage form orally administered to an individual after a high-fat meal relative to the sustained release dosage form orally administered to a fasted individual is about 125% to about 170%.
In some embodiments, the sustained-release dosage forms of the invention may include a sustained-release matrix former. Example sustained-release matrix formers include cellulosic ethers such as hydroxypropyl methylcellulose (HPMC, hypromellose) which is a high viscosity polymer, and methyl celluloses. Example hydroxypropyl methylcelluloses include Methocel™ K15M, Methocel™ K4M, Methocel™ K100LV, Methocel™ E3, Methocel™ E5, Methocel™ E6, Methocel™ E15, Methocel™ E50, Methocel™ E10M, Methocel™ E4M, and Methocel™ E10M. In some embodiments, the sustained release dosage form comprises one or more hypromelloses. In some embodiments, the sustained release dosage form comprises a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP and a second hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP. In some embodiments, the sustained release dosage form comprises about 8% to about 20% by weight of one or more hypromelloses. In some embodiments, the sustained release dosage form comprises about 10% to about 15% by weight of one or more hypromelloses.
In some embodiments, the sustained-release dosage forms of the invention can further include one or more fillers, glidants, disintegrants, binders, or lubricants as inactive ingredients. In some embodiments, the filler comprises microcrystalline cellulose, lactose monohydrate, or both. In some embodiments, the sustained release dosage form comprises about 16% to about 22% by weight of microcrystalline cellulose. In some embodiments, the sustained release dosage form comprises about 45% to about 55% by weight of lactose monohydrate.
In some embodiments, lubricants can be present in the dosage forms of the invention in an amount of 0 to about 5% by weight. Non-limiting examples of lubricants include magnesium stearate, stearic acid (stearin), hydrogenated oil, polyethylene glycol, sodium stearyl fumarate, and glyceryl behenate. In some embodiments, the formulations include magnesium stearate, stearic acid, or both. In some embodiments, the sustained release dosage form comprises about 0.3% to about 0.7% by weight of magnesium stearate.
In some embodiments, glidants may be present in the dosage forms. In some embodiments, glidants can be present in the dosage forms of the invention in an amount of 0 to about 5% by weight. Non-limiting examples of glidants include talc, colloidal silicon dioxide, and cornstarch. In some embodiments, the glidant is colloidal silicon dioxide.
In some embodiments, film-coating agents can be present in an amount of 0 to about 5% by weight. Non-limiting illustrative examples of film-coating agents include hypromellose or polyvinyl alcohol based coating with titanium dioxide, talc and optionally colorants available in several commercially available complete coating systems.
In some embodiments, the sustained release dosage form comprises pregelatinized starch.
In some embodiments, the sustained release dosage form is a tablet.
In some embodiments, the sustained release dosage form is prepared by process comprising wet granulation.
In some embodiments, the sustained release dosage form comprises one or more excipients independently selected from hypromelloses and microcrystalline celluloses.
In some embodiments, the sustained release dosage form comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, and lactose monohydrate.
In some embodiments, the sustained release dosage form comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, lactose monohydrate, and pregelatinized starch.
The present invention further provides one or more sustained release dosage forms each comprising { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said one or more sustained release dosage forms together provide a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present invention also provides a dose, comprising one or more sustained release dosage forms each comprising { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said dose provides a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 600 mg on a free base basis of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 500 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application further provides one or more sustained release dosage forms as described herein, which together provide a once-daily oral dosage of about 400 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
In some embodiments, the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of { 1-{ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, is provided.
The present application also provides a dose comprising one or more sustained release dosage forms as described herein, which provide a once-daily oral dosage of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
The present application also provides a dose comprising one or more sustained release dosage forms as described herein, which provide a once-daily oral dosage of about 500 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient. The present application also provides a dose comprising one or more sustained release dosage forms as described herein, which provide a once-daily oral dosage of about 400 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
In some embodiments, the dose comprises six dosage forms of about 100 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the dose comprises three dosage forms of about 200 mg on a free base basis of { 1- { 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the dose comprises two dosage forms of about 300 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the dose comprises one dosage form of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 400 mg to about 600 mg on a free base basis of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient. In some embodiments, the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once- daily dose of about 400 mg to about 600 mg on a free base basis of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. The present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 600 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient. In some embodiments, the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once-daily dose of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 500 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient. In some embodiments, the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once-daily dose of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present application further provides a kit comprising one or more sustained release dosage forms as described herein, which together provide a once- daily oral dosage of about 400 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient. In some embodiments, the kit further comprises an instruction to administer the one or more sustained release dosage forms as a once-daily dose of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the kit comprises six dosage forms of about 100 mg on a free base basis of { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}- 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises three dosage forms of about 200 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises two dosage forms of about 300 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises one dosage form of about 600 mg on a free base basis of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
As used herein, "sustained-release" is used as generally understood in the art and refers to a formulation designed to slowly release the active ingredient into a patient after oral administration.
As used herein, "dose" refers to the total amount of the compound of Formula I orally administered to the individual or patient. The dose may be in a single dosage form, or a plurality of dosage forms (e.g., a 600 mg dose may be one 600 mg dosage form, two 300 mg dosage forms, three 200 mg dosage forms, six 100 mg dosage forms, etc.). Hence, a dose can refer to a plurality of pills to be taken by a patient at nearly simultaneously.
As used herein, "a fasted individual" means an individual who has fasted for at least 10 hours prior to administration of the dose.
As used herein, "mean" when preceding a pharmacokinetic value (e.g. mean Cmax) represents the arithmetic mean value of the pharmacokinetic value taken from a population of patients unless otherwise specified.
As used herein, "Cmax" means the maximum observed plasma concentration. As used herein, "Ci2h" refers to the plasma concentration measured at 12 hours from administration.
As used herein, "Tmax" refers to the time at which the maximum blood plasma concentration is observed.
As used herein, "T1/2" refers to the time at which the plasma concentration is half of the observed maximum.
As used herein, "AUC" refers to the area under the plasma concentration-time curve which is a measure of total bioavailability.
As used herein, "AUCo -∞" refers to the area under the plasma concentration- time curve extrapolated to infinity.
As used herein, "AUCo-t" refers to the area under the plasma concentration- time curve from time 0 to the last time point with a quantifiable plasma concentration, usually about 12-36 hours.
As used herein, "AUCo-τ" refers to the area under the plasma concentration- time curve from time 0 to the time of the next dose.
As used herein, "Cl/F" refers to oral clearance.
The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred. 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 its entirety. In some embodiments, the compounds described herein include the N-oxide forms.
Methods
The present application further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage forms as described herein.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof. The present application further provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient one or more sustained release dosage as described herein.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 500 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 400 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}- 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, is provided.
In some embodiments, oral administration of one or more sustained release dosage forms to a fasted individual provides a mean time to peak plasma
concentration (Tmax) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrileof about 0.5 hours to about 3 hours.
In some embodiments,oral administration of one or more sustained release dosage forms to a fasted individual provides a mean time to peak plasma
concentration (Tmax) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile of at least 0.5 hours.
In some embodiments,oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma
concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 5 to about 50.
In some embodiments,oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma
concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 9 to about 40.
In some embodiments,oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma
concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 15 to about 30. In some embodiments, oral administration of one or more sustained release dosage forms to a fasted individual provides a mean half-life (ti/2) of { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrileof about 1 hour to about 20 hours.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrileof about 1 hour to about 9 hours.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a mean time to peak plasma concentration (Tmax) of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of at least 1.5 hours.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 10 to about 70.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h)of { l-{ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 15 to about 50.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high- fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l-{ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 25 to about 45.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1 hour to about 7 hours.
In some embodiments, oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 2 hours to about 5 hours.
In some embodiments, the one or more sustained release dosage forms are each a tablet. In some embodiments, the one or more sustained release dosage forms are prepared by process comprising wet granulation.
In some embodiments, the one or more sustained release dosage forms each comprises one or more hypromelloses. In some embodiments, the one or more sustained release dosage forms each comprises one or more excipients independently selected from hypromelloses and microcrystalline celluloses. In some embodiments, the one or more sustained release dosage forms each comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, and lactose monohydrate. In some embodiments, the one or more sustained release dosage forms each comprises a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP and a second hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP.
In some embodiments, the one or more sustained release dosage forms each comprises about 10% to about 15% by weight of one or more hypromelloses. In some embodiments, the one or more sustained release dosage forms each comprises about 16% to about 22% by weight of microcrystalline cellulose. In some embodiments, the one or more sustained release dosage forms each comprises about 45% to about 55% by weight of lactose monohydrate. In some embodiments, the one or more sustained release dosage forms each comprises about 0.3% to about 0.7% by weight of magnesium stearate.
In some embodiments, the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline. In some embodiments, the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of { 1- { 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
In some embodiments, the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 500 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
In some embodiments, the present application provides a method of treating myelofibrosis in a patient, comprising orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 400 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}- 3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the methods in the preceding three paragraphs, the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, is provided.
In some embodiments, "total symptom score (TSS)" refers to the TSS derived from the modified Myelofibrosis Symptom Assessment Form (MFSAF) (e.g., v3.0) electronic diary as compared with baseline (baseline is the patient's baseline TSS before treatment). In some embodiments, myelofibrosis is primary myelofibrosis (PMF), post-polycythemia vera MF, or post-essential thrombocythemia MF.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 500 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia.
The present application also provides a method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, wherein the method comprises orally administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 400 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said method results in reduced anemia. In some embodiments, the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of { 1-{ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, is provided.
Reduced anemia is relative to that experienced for a twice-daily dose of 200 mg on a free base basis of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising { l- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
The compound of Formula I is a JAK inhibitor. A JAKl selective inhibitor is a compound that inhibits JAKl activity preferentially over other Janus kinases. JAKl plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states. For example, IL- 6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (Fonesca, J.E. et al, Autoimmunity Reviews, 8:538-42, 2009). Because IL-6 signals, at least in part, through JAKl, antagonizing IL-6 directly or indirectly through JAKl inhibition is expected to provide clinical benefit (Guschin, D., N., et al Embo J 14: 1421, 1995; Smolen, J. S., et al. Lancet 371 :987, 2008). Moreover, in some cancers JAKl is mutated resulting in constitutive undesirable tumor cell growth and survival (Mullighan CG, Proc Natl Acad Sci U S A.106:9414-8, 2009; Flex E., et al.J Exp Med. 205:751-8, 2008). In other autoimmune diseases and cancers elevated systemic levels of inflammatory cytokines that activate JAK1 may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAK1 inhibition. Selective inhibitors of JAK1 may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases.
Selective inhibitors of JAK1, relative to other JAK kinases, may have multiple therapeutic advantages over less selective inhibitors. With respect to selectivity against JAK2, a number of important cytokines and growth factors signal through JAK2 including, for example, erythropoietin (Epo) and thrombopoietin (Tpo) (Parganas E, et al. Cell. 93:385-95, 1998). Epo is a key growth factor for red blood cells production; hence a paucity of Epo-dependent signaling can result in reduced numbers of red blood cells and anemia (Kaushansky K, NEJM 354:2034-45, 2006). Tpo, another example of a JAK2-dependent growth factor, plays a central role in controlling the proliferation and maturation of megakaryocytes - the cells from which platelets are produced (Kaushansky K, NEJM 354:2034-45, 2006). As such, reduced Tpo signaling would decrease megakaryocyte numbers (megakaryocytopenia) and lower circulating platelet counts (thrombocytopenia). This can result in undesirable and/or uncontrollable bleeding. Reduced inhibition of other JAKs, such as JAK3 and Tyk2, may also be desirable as humans lacking functional version of these kinases have been shown to suffer from numerous maladies such as severe-combined immunodeficiency or hyperimmunoglobulin E syndrome (Minegishi, Y, et al.
Immunity 25:745-55, 2006; Macchi P, et al. Nature. 377:65-8, 1995). Therefore a JAK1 inhibitor with reduced affinity for other JAKs would have significant advantages over a less-selective inhibitor with respect to reduced side effects involving immune suppression, anemia and thrombocytopenia.
Another aspect of the present invention pertains to methods of treating a JAK- associated disease or disorder in an individual (e.g., patient) by administering to the individual in need of such treatment a sustained-release dosage form of the invention. A JAK-associated disease can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the JAK, including overexpression and/or abnormal activity levels. A JAK-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating JAK activity.
Examples of JAK-associated diseases include diseases involving the immune system including, for example, organ transplant rejection (e.g., allograft rejection and graft versus host disease).
Further examples of JAK-associated diseases include autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, chronic obstructive pulmonary disease (COPD), and the like. In some embodiments, the autoimmune disease is an autoimmune bullous skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
Further examples of JAK-associated diseases include allergic conditions such as asthma, food allergies, eszematous dermatitis, contact dermatitis, atopic dermatitis (atropic eczema), and rhinitis. Further examples of JAK-associated diseases include viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV, HTLV 1, Varicella-Zoster Virus (VZV) and Human Papilloma Virus (HPV).
Further examples of JAK-associated disease include diseases associated with cartilage turnover, for example, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome, costal athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease, degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, or ankylosing spondylitis.
Further examples of JAK-associated disease include congenital cartilage malformations, including hereditary chrondrolysis, chrondrodysplasias, and pseudochrondrodysplasias (e.g., microtia, enotia, and metaphyseal
chrondrodysplasia).
Further examples of JAK-associated diseases or conditions include skin disorders such as 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 at least one JAK inhibitor of the invention together with the agent causing unwanted sensitization can be helpful in treating such unwanted sensitization or dermatitis. In some embodiments, the skin disorder is treated by topical administration of at least one JAK inhibitor of the invention.
In further embodiments, the JAK-associated disease is cancer including those characterized by solid tumors (e.g., prostate cancer, renal cancer, hepatic cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, Castleman's disease, uterine leiomyosarcoma, melanoma etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and cutaneous B-cell lymphoma. Example CTCLs include Sezary syndrome and mycosis fungoides.
In some embodiments, the dosage forms described herein, or in combination with other JAK inhibitors, such as those reported in U.S. Ser. No. 1 1/637,545, which is incorporated herein by reference in its entirety, can be used to treat inflammation- associated cancers. In some embodiments, the cancer is associated with inflammatory bowel disease. In some embodiments, the inflammatory bowel disease is ulcerative colitis. In some embodiments, the inflammatory bowel disease is Crohn's disease. In some embodiments, the inflammation-associated cancer is colitis-associated cancer. In some embodiments, the inflammation-associated cancer is colon cancer or colorectal cancer. In some embodiments, the cancer is gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), adenocarcinoma, small intestine cancer, or rectal cancer.
JAK-associated diseases can further include those characterized by expression of: JAK2 mutants such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F); JAK2 mutants having at least one mutation outside of the pseudo-kinase domain; JAK1 mutants; JAK3 mutants; erythropoietin receptor (EPOR) mutants; or deregulated expression of CRLF2.
JAK-associated diseases can further include myeloproliferative disorders (MPDs) such as polycythemia vera (PV), essential thrombocythemia (ET), myelofibrosis with myeloid metaplasia (MMM), primary myelofibrosis (PMF), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hypereosinophilic syndrome (HES), systemic mast cell disease (SMCD), and the like. In some embodiments, the myeloproliferative disorder is myelofibrosis (e.g., primary myelofibrosis (PMF) or post polycythemia vera/essential thrombocythemia myelofibrosis (Post-PV/ET MF)). In some embodiments, the myeloproliferative disorder is post- essential thrombocythemia myelofibrosis (Post-ET). In some embodiments, the myeloproliferative disorder is post polycythemia vera myelofibrosis (Post-PV MF).
In some embodiments, dosage forms described herein can be used to treat pulmonary arterial hypertension.
The present invention further provides a method of treating dermatological side effects of other pharmaceuticals by administration of the dosage forms 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 dosage forms of the invention can be administered systemically in combination with (e.g., simultaneously or sequentially) the pharmaceutical agent having the undesirable dermatological side effect.
Further JAK-associated diseases include inflammation and inflammatory diseases. Example inflammatory diseases include sarcoidosis, inflammatory diseases of the eye (e.g., iritis, uveitis, scleritis, conjunctivitis, or related disease),
inflammatory diseases of the respiratory tract (e.g., the upper respiratory tract including the nose and sinuses such as rhinitis or sinusitis or the lower respiratory tract including bronchitis, chronic obstructive pulmonary disease, and the like), inflammatory myopathy such as myocarditis, and other inflammatory diseases. In some embodiments, the inflammation disease of the eye is blepharitis.
The dosage forms described herein can further be used to treat ischemia reperfusion injuries or a disease or condition related to an inflammatory ischemic event such as stroke or cardiac arrest. The dosage forms described herein can further be used to treat endotoxin-driven disease state (e.g., complications after bypass surgery or chronic endotoxin states contributing to chronic cardiac failure). The dosage forms described herein can further be used to treat anorexia, cachexia, or fatigue such as that resulting from or associated with cancer. The dosage forms described herein can further be used to treat restenosis, sclerodermitis, or fibrosis. The dosage forms described herein can further be used to treat conditions associated with hypoxia or astrogliosis such as, for example, diabetic retinopathy, cancer, or neurodegeneration. See, e.g., Dudley, A.C. et al. Biochem. J. 2005, 390(Pt 2):427-36 and Sriram, K. et al. J. Biol. Chem. 2004, 279(19): 19936-47. Epub 2004 Mar 2, both of which are incorporated herein by reference in their entirety. The JAK inhibitors described herein can be used to treat Alzheimer's disease.
The dosage forms described herein can further be used to treat other inflammatory diseases such as systemic inflammatory response syndrome (SIRS) and septic shock.
The dosage forms described herein can further be used to treat gout and increased prostate size due to, e.g., benign prostatic hypertrophy or benign prostatic hyperplasia.
Further JAK-associated diseases include bone resorption diseases such as osteoporosis, osteoarthritis. Bone resorption can also be associated with other conditions such as hormonal imbalance and/or hormonal therapy, autoimmune disease (e.g. osseous sarcoidosis), or cancer (e.g. myeloma). The reduction of the bone resorption due to the the compound of Formula I can be about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, or about 90%.
In some embodiments, the dosage forms described herein can further be used to treat a dry eye disorder. As used herein, "dry eye disorder" is intended to encompass the disease states summarized in a recent official report of the Dry Eye Workshop (DEWS), which defined dry eye as "a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface. It is accompanied by increased osmolarity of the tear film and inflammation of the ocular surface." Lemp, "The Definition and Classification of Dry Eye Disease: Report of the Definition and Classification Subcommittee of the International Dry Eye Workshop", The Ocular Surface, 5(2), 75-92 April 2007, which is incorporated herein by reference in its entirety. In some embodiments, the dry eye disorder is selected from aqueous tear- deficient dry eye (ADDE) or evaporative dry eye disorder, or appropriate
combinations thereof. In some embodiments, the dry eye disorder is Sjogren syndrome dry eye (SSDE). In some embodiments, the dry eye disorder is non- Sjogren syndrome dry eye (NSSDE).
In a further aspect, the present invention provides a method of treating conjunctivitis, uveitis (including chronic uveitis), chorioditis, retinitis, cyclitis, sclieritis, episcleritis, or iritis; treating inflammation or pain related to corneal transplant, LASIK (laser assisted in situ keratomileusis), photorefractive keratectomy, or LASEK (laser assisted sub-epithelial keratomileusis); inhibiting loss of visual acuity related to corneal transplant, LASIK, photorefractive keratectomy, or LASEK; or inhibiting transplant rejection in a patient in need thereof, comprising administering to the patient a dosage form of the invention.
Additionally, the dosage forms of the invention, or in combination with other JAK inhibitors, such as those reported in U.S. Ser. No. 1 1/637,545, which is incorporated herein by reference in its entirety, can be used to treat respiratory dysfunction or failure associated with viral infection, such as influenza and SARS.
In some embodiments, the present invention provides a dosage form as described in any of the embodiments herein, for use in a method of treating any of the diseases or disorders described herein. In some embodiments, the present invention provides the use of a dosage form as described in any of the embodiments herein, for the preparation of a medicament for use in a method of treating any of the diseases or disorders described herein.
In some embodiments, the present invention provides a dosage form as described herein, or a pharmaceutically acceptable salt thereof, for use in a method of modulating JAKl . In some embodiments, the present invention also provides use of a dosage form as described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in a method of modulating JAKl.
As used herein, the term "individual" is a human. In some embodiments, the human is an adult subject.
As used herein, the term "treating" or "treatment" refers to one or more of (1) 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 (2) 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, which is incorporated herein by reference in its entirety, or other agents can be used in combination with the dosage forms described herein 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 chemotherapeutics 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 coriticosteroids 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, all of which are incorporated herein by reference in their entirety.
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, all of which are incorporated herein by reference in their entirety.
Example suitable RAF inhibitors include compounds, and their
pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444, both of which are incorporated herein by reference in their entirety. 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, all of which are incorporated herein by reference in their entirety.
In some embodiments, one or more of the dosage forms 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, one or more dosage forms of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, such as multiple myeloma, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects. Examples of additional pharmaceutical agents used in the treatment of multiple myeloma, for example, can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib). Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. Additive or synergistic effects are desirable outcomes of combining a dosage form of the present invention with an additional agent.
Furthermore, resistance of multiple myeloma cells to agents such as dexamethasone may be reversible upon treatment with a dosage form of the present invention. The agents can be combined with the present compounds in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
In some embodiments, a corticosteroid such as dexamethasone is administered to a patient in combination with at the dosage form of the invention where the dexamethasone is administered intermittently as opposed to continuously.
In some further embodiments, combinations of one or more JAK inhibitors of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
In some embodiments, the additional therapeutic agent is fluocinolone acetonide (Retisert®), or rimexolone (AL-2178, Vexol, Alcon).
In some embodiments, the additional therapeutic agent is cyclosporine (Restasis®). In some embodiments, the additional therapeutic agent is a corticosteroid. In some embodiments, the corticosteroid is triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
In some embodiments, the additional therapeutic agent is selected from Dehydrex™ (Holies Labs), Civamide (Opko), sodium hyaluronate (Vismed,
Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine, doxycycline (ALTY-0501, Alacrity), minocycline, iDestrin™ (NP50301, Nascent Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline
(Duramycin, MOLI1901, Lantibio), CF101 (2S,3S,4R,5R)-3,4-dihydroxy-5-[6-[(3- iodophenyl)methylamino]purin-9-yl]-N-methyl-oxolane-2-carbamyl, Can-Fite Biopharma), voclosporin (LX212 or LX214, Lux Biosciences), ARG103 (Agentis), RX- 10045 (synthetic resolvin analog, Resolvyx), DY 15 (Dyanmis Therapeutics), rivoglitazone (DE011, Daiichi Sanko), TB4 (RegeneRx), OPH-01 (Ophtalmis Monaco), PCS101 (Pericor Science), REV1-31 (Evolutec), Lacritin (Senju), rebamipide (Otsuka-Novartis), OT-551 (Othera), PAI-2 (University of Pennsylvania and Temple University), pilocarpine, tacrolimus, pimecrolimus (AMS981, Novartis), loteprednol etabonate, rituximab, diquafosol tetrasodium (TNS365, Inspire), KLS- 061 1 (Kissei Pharmaceuticals), dehydroepiandrosterone, anakinra, efalizumab, mycophenolate sodium, etanercept (Embrel®), hydroxychloroquine, NGX267 (TorreyPines Therapeutics), actemra, gemcitabine, oxaliplatin, L-asparaginase, or thalidomide.
In some embodiments, the additional therapeutic agent is an anti-angiogenic agent, cholinergic agonist, TRP- 1 receptor modulator, a calcium channel blocker, a mucin secretagogue, MUC 1 stimulant, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, an mTOR inhibitor, another JAK inhibitor, Bcr-Abl kinase inhibitor, Flt-3 kinase inhibitor, RAF kinase inhibitor, and FAK kinase inhibitor such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety. In some embodiments, the additional therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycline). In some embodiments, the additional therapeutic agent binds to FKBP12. In some embodiments, the additional therapeutic agent is an alkylating agent or DNA cross-linking agent; an anti-metabolite/demethylating agent (e.g., 5- flurouracil, capecitabine or azacitidine); an anti-hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e.g.
vincristine or paclitaxel); an topoisomerase (I or II) inhibitor (e.g. mitoxantrone and irinotecan); an apoptotic inducers (e.g. ABT-737); a nucleic acid therapy (e.g.
antisense or RNAi); nuclear receptor ligands (e.g., agonists and/or antagonists: all- trans retinoic acid or bexarotene); epigenetic targeting agents such as histone deacetylase inhibitors (e.g. vorinostat), hypomethylating agents (e.g. decitabine); regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
In some embodiments, the additional therapeutic agent(s) are demulcent eye drops (also known as "artificial tears"), which include, but are not limited to, compositions containing polyvinylalcohol, hydroxypropyl methylcellulose, glycerin, polyethylene glycol (e.g. PEG400), or carboxymethyl cellulose. Artificial tears can help in the treatment of dry eye by compensating for reduced moistening and lubricating capacity of the tear film. In some embodiments, the additional therapeutic agent is a mucolytic drug, such as N-acetyl-cysteine, which can interact with the mucoproteins and, therefore, to decrease the viscosity of the tear film.
In some embodiments, the additional therapeutic agent includes an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and nonsteroidal anti-inflammatories, and anti-allergic agents. Examples of suitable medicaments include aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine; sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin; colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives ("rifampins"); cycloserine; beta-lactams; cephalosporins; amphotericins; fluconazole; flucytosine; natamycin; miconazole; ketoconazole; corticosteroids; diclofenac;
flurbiprofen; ketorolac; suprofen; cromolyn; lodoxamide; levocabastin; naphazoline; antazoline; pheniramine; or azalide antibiotic. 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 (as if the embodiments of the specification are written as multiply dependent claims).
Example 1. Preparation of Sustained Release Formulations
Sustained release tablets were prepared with the excipients being in the amounts shown in the table below. Protocol A was used for the SRI tablets, protocol B was used for the SR2 tablets, Protocol C was used for the SR3 tablets and the 25 mg SR tablets, and Protocol D was used for the SR4 tablets.
Protocol A:
Step 1. Individually screen the adipic acid salt of the compound of
Formula I, microcrystalline cellulose, hypromelloses (Methocel K100 LV and Methocel K4M), and lactose monohydrate.
Step 2. Transfer the screened material from Step 1 to a suitable blender and mix.
Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Transfer the granules from Step 4 into a suitable dryer and dry until LOD is less than 3%.
Step 6. Screen the granules from Step 5.
Step 7. Mix screened Magnesium Stearate with granules in Step 6 in a suitable blender.
Step 8. Compress the final blend in Step 7 on a suitable rotary tablet press.
Protocol B:
Step 1. Individually screen the adipic acid salt of the compound of
Formula I, microcrystalline cellulose, hypromellose and pregelatinized starch.
Step 2. Transfer the screened material from Step 1 to a suitable blender and mix. Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Transfer the granules from Step 4 into a suitable dryer and dry until LOD is less than 3%.
Step 6. Screen the granules from Step 5.
Step 7. Individually screened polyox, butylated hydroxytoluene and colloidal silicone dioxide.
Step 8. Transfer the granules from Step 6 and material from Step 7 into a suitable blender and mix.
Step 9. Add screened Magnesium Stearate to the material in Step 8 and continue blending.
Step 10. Compress the final blend in Step 9 on a suitable rotary tablet press.
Protocol C:
Step 1. Individually screen lactose monohydrate, the adipic acid salt of the compound of Formula I, microcrystalline cellulose and hypromelloses through a suitable screen.
Step 2. Transfer the screened material from Step 1 to a suitable blender and mix.
Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Screen wet granules through a suitable screen.
Step 6. Transfer the granules from Step 5 into a suitable dryer and dry until LOD is less than 3%.
Step 7. Mill the granules from Step 6.
Step 8. Mix screened magnesium stearate with granules in Step 7 in a suitable blender.
Step 9. Compress the final blend in Step 8 on a suitable rotary tablet press. Protocol D:
Step 1. Individually screen pregelatinized starch, the adipic acid salt of the compound of Formula I, hypromellose, and a portion of required microcrystalline cellulose through a suitable screen.
Step 2. Transfer the screened material from Step 1 to a suitable blender and mix.
Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Screen wet granules through a suitable screen.
Step 6. Transfer the granules from Step 5 into a suitable dryer and dry until LOD is less than 3%.
Step 7. Mill the granules from Step 6.
Step 8. Screen the remaining portion of microcrystalline cellulose and half of the sodium bicarbonate.
Step 9. Transfer the milled granules from Step 7 and screened materials from Step 8 into a suitable blender and mix.
Step 10. Screen the remaining portion of sodium bicarbonate and mix with blend in Step 9.
Step 11. Screen magnesium stearate and mix with blend in Step 10.
Step 12. Compress the final blend in Step 1 1 on a suitable rotary tablet press.
SRI: Composition of 100 mg Sustained Release Tablets
Figure imgf000044_0001
Component Function Weight (mg/tablet) Composition
(wt%)
Lactose Monohydrate Filler 290.58 48.4
Magnesium Stearate b Lubricant 3.0 0.5
Purified Water c Granulating q.s. —
Liquid
Total 600.0 100 a Conversion factor for adipate salt to free base
b Added after granulation
c Removed during processing SR2: Composition of 100 mg Sustained Release Tablets
Figure imgf000045_0001
a Conversion factor for adipate salt to free base
b Added after granulation
c Removed during processing SR3 (100 mg): Composition of 100 mg Sustained Release Tablets
Figure imgf000046_0001
a Conversion factor for adipate salt to free base is 0.791 1
b Added after granulation
c Removed during processing
SR4: Composition of 100 mg Sustained Release Tablets
Excipient Function Weight (mg/tablet) Composition
(wt%)
Adipic acid salt of the Active
126.4 a 21.1 compound of Formula Ia
Microcrystalline
Filler 104.6 17.4 Cellulose d
Hypromellose
Release Control 210.0 35.0 (Methocel KlOOLV)
Pregelatinized Starch Filler 60.0 10.0
Gastric Floating
Sodium Bicarbonate b 96.0 16.0
Aid
Magnesium Stearate b Lubricant 3.0 0.5
Purified Water c Granulation Liquid q.s. —
Total 600.0 100.0 a Conversion factor for adipate salt to free base is 0.791 1
b Added after granulation
c Removed during processing
d Partial added before and partial added after granulation
25mg SR: Composition of 25 mg Sustained Release Tablets
Figure imgf000047_0001
a Conversion factor for adipate salt to free base
b Added after granulation
c Removed during processing
Example 2. Preparation of the IR Formulation of the Compound of Formula I
The IR formulation used in the studies in Example 3 was prepared as 50 mg capsules with the composition shown in the table below according to Protocol E below.
Protocol E:
Step 1. Pre-mix the required amount of the adipic acid salt of the compound of
Formula I and an approximately equal amount of silicified microcrystalline cellulose (SMCC). Step 2. Pass the mixture in Step 1 through a suitable screen (for example 40 mesh).
Step 3. Screen the remaining SMCC through the same screen used in Step 2.
Step 4. Blend the screened SMCC from Step 3 along with mixture from Step 2 in a suitable blender (for example Turbula blender) for approximately 5 minutes. Step 5. Fill the blend into capsules to desired fill weight.
Figure imgf000048_0001
* Adipic acid salt of the compound of Formula I with salt conversion factor of 0.7911
Example 3. Relative Bioavailability Study of Sustained Release Dosage Forms
A total of 72 healthy adult subjects were enrolled in 6 cohorts (12 subjects per cohort) and randomized to treatment sequences within each cohort according to a randomization schedule. All treatments were single-dose administrations of the compound of Formula I. There was a washout period of 7 days between the treatment periods.
The SRI, SR2, SR3, and SR4 formulations were evaluated in Cohort 1, Cohort 2, Cohort 3, and Cohort 4, respectively (see Example 1 for SRI, SR2, SR3, SR4, and 25 mg SR tablets used in study). The subjects received the IR and SR treatments according to a 3 -way crossover design:
Treatment A: 300 mg (6 x 50 mg capsule) IR formulation of the compound of Formula I administered orally after an overnight fast of at least 10 hours. Treatment B: 300 mg (3 x 100 mg tablets) SR formulation of the compound of Formula I administered orally after an overnight fast of at least 10 hours.
Treatment C: 300 mg (3 x 100 mg tablets) SR formulation of the compound of Formula I administered orally after a high-fat meal.
The subjects in Cohort 5 received the following treatments in a 2-way crossover design:
Treatment A: 300 mg (3 x 100 mg tablets of the compound of Formula I) SR3 administered orally after an overnight fast of at least 10 hours.
Treatment B: 300 mg (3 x 100 mg tablets of the compound of Formula I) SR3 administered orally after a medium-fat meal.
The subjects in Cohort 6 received the following treatments in a 3 -way crossover design:
Treatment A: 50 mg (2 x 25 mg tablets of the compound of Formula I (25 mg SR tablets from Example 1)) administered orally after an overnight fast of at least 10 hours.
Treatment B: 50 mg (2 x 25 mg tablets of the compound of Formula I (25 mg SR tablets from Example 1)) administered orally after a high-fat meal.
Treatment C: 100 mg (1 x 100 mg tablets) SR3 administered orally after an overnight fast of at least 10 hours.
Blood samples for determination of plasma concentrations of the compound of Formula I were collected using lavender top (K2EDTA) Vacutainer® tubes at 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose.
Plasma samples were assayed by a validated, GLP, LC/MS/MS method with a linear range of 5.0 to 5000 nM. Table 1 summarizes the accuracy and precision (CV %) of the assay quality control samples during the analysis of the plasma samples from this study.
Accuracy and Precision of the Plasma Assay Quality Control
Figure imgf000050_0001
CV% =percent coefficient of variability; QC = quality control; Theo = theoretical or nominal concentration.
For the PK analysis, the actual sample collection times were used. For any sample with missing actual collection time, the scheduled time was used provided that there was no protocol deviation noted for the collection of these samples.
Standard noncompartmental PK methods were used to analyze the data for the plasma concentration of the compound of Formula using Phoenix WinNonlin version 6.0 (Pharsight Corporation, Mountain View, CA). Thus, Cmax and Tmax were taken directly from the observed plasma concentration data. The terminal-phase disposition rate constant (λζ) was estimated using a log-linear regression of the concentration data in the terminal disposition phase, and t½ was estimated as 1η(2)/λζ. AUCo-t was estimated using the linear trapezoidal rule for increasing concentrations and the log- trapezoidal rule for decreasing concentrations, and the total AUCo-∞ was calculated as AUCo-t + Ct/λζ. The oral-dose clearance (CL/F) was estimated as Dose/AUCo-∞, and the terminal-phase volume of distribution V F) was estimated as Dose/[AUCo-∞* z].
The log-transformed Cmax and AUC values (after dose normalization, where the doses were different) were compared between the fasted and fed dosing treatments, and between the SR and IR dosing treatments, using a crossover ANOVA (fixed factor = treatment, sequence and period, random effect = subject (sequence)). The adjusted geometric mean ratios of Cmax and AUC between the treatments (reference = IR or fasted administration of SR) and the corresponding 90% confidence intervals (CIs) were determined. In addition, the correlation between the observed food effect of a high- fat meal on AUCo-∞ and the relative bioavailability of the SR formulations (with reference to the IR capsule) were explored by a quantile plot using the data from all subjects who completed Treatment A, B, and C in Cohorts 1 to 4. The statistical analysis was performed using Phoenix WinNonlin version 6.0.
FIG. 1 presents plasma concentrations of the compound of Formula I (mean ± SE) for the subjects in Cohorts 1 to 4 following Treatment A (300 mg IR
administration in fasted state), Treatment B (300 mg SR administration in fasted state), and Treatment C (300 mg SR administration with a high-fat meal). FIG. 2 compares the effect of a high-fat meal and medium- fat meal on the mean PK profile following a single-dose 300 mg (3 x lOOmg) administration of the compound of Formula I SR3 tablets. FIG. 3 presents plasma concentrations of the compound of Formula I (mean ± SE) for the subjects in Cohort 6 following Treatment A (2 x 25 mg SR tablet administration in fasted state), Treatment B (2 x 25 mg SR tablet with a high-fat meal), and Treatment C (1 x 100 mg SR3 administration in fasted state).
Tables 2A, 2B, 3 A and 3B summarize mean PK parameters for subjects in Cohorts 1 to 4, the relative bioavailability (reference = IR capsule) and food effect (high-fat meal) for the 100 mg strength SR1-SR4 tablets. Table 4A and 4B summarize mean PK parameters for subjects in Cohort 5, and food effect (medium- fat meal) for the 100 mg strength SR3 tablet. Table 5A and 5B summarizes mean PK parameters for subjects in Cohort 6, the dose-normalized relative bioavailability (reference = 100 mg SR3 tablet), and the food effect (high-fat meal) for the 25 mg SR tablet.
Table 2A
Cohort/Treatment n Cmax/C
(μΜ) 12h
(h) (h)
Cohort 1
12 2.29 ± 1.0
300 mg IR 197 ± 147 2.0 ± 0.27
0.50 (0.50- (fasted) 159 2.0
2.24 2.0)
12 0.341 ± 1.3
300 mg SRI 13.2 ± 7.8 9.2 ± 4.5
0.13 (0.50- (fasted) 11.6 8.3
0.317 3.0)
12 0.610 ±
300 mg SRI 4.0 18.0 ± 6.4 3.2 ± 1.4
0.14
(high-fat meal) (2.0-8.0) 16.8 3.0
0.595
Cohort 2
12 2.05 ± 1.0
300 mg IR 130 ± 72.9 2.1 ± 0.34
0.67 (0.50- (fasted) 112 2.1
1.92 3.0)
12 0.191 ±
300 mg SR2 2.5 11.4 ± 9.9 11 ± 8.4
0.10
(fasted) (1.0-4.0) 8.60 9.23
0.172
12 0.470 ±
300 mg SR2 6.0 11.0 ± 4.0 3.5 ±2.6
0.16
(high-fat meal) (1.5-6.0) 10.4 3.0
0.443
Cohort 3
11 2.35 ± 1.0
300 mg IR 136 ± 70.8 2.2 ± 0.53
0.41 (0.50- (fasted) 120 2.2
2.31 2.0)
11 0.553 ± 1.5
300 mg SR3 22.9 ± 13.4 9.8 ± 8.5
0.24 (0.50- (fasted) 19.3 7.2
0.502 3.0)
12 1.05 ±
300 mg SR3 4.0 34.9 ± 15.8 3.3 ± 1.2
0.47
(high-fat meal) (1.5-8.0) 30.8 3.1
0.968
Cohort 4
12 2.94 ± 1.0
300 mg IR 170 ± 58.6 2.1 ± 0.58
0.98 (0.25- (fasted) 162 2.1
2.78 1.5)
12 0.321 ±
300 mg SR4 2.0 10.3 ± 6.0 7.3 ± 5.3
0.27
(fasted) (1.5-8.1) 8.92 6.0
0.249
12 0.549 ±
300 mg SR4 4.0 12.8 ± 14.8 4.9 ± 2.6
0.28
(high-fat meal) (2.0-16) 6.06 4.4
0.481 Table 2B
AUCo-t AUC<n» CL/F
Cohort/Treatment
(μΜ*1ι) (μΜ*1ι) (L/h)
Cohort 1
300mgIR 4-4^ 4.45 ± 1.00 127 ±27.1
(fasted) " J 4.35 124
300 mg SRI ^f 1.65 ± 0.54 359 ±106
(fasted) 1.57 345
300 mg SRI ^i 2.91 ±0.65 194 ±39.9
(high-fat meal) 2.85 190
Z.oZ
Cohort 2
300mgIR 4.47 ± 1.36 134 ±50.1
(fasted) 424 4.27 127
300mgSR2 1.00 ±037 1.17 ± 0.43 510 ±148
(fasted) 0.95 1.11 488
748 ±
300mgSR2 n ° 2.52 ± 0.72 235 ± 83.5
(high-fat meal) ' " 2.42 224
.3o
Cohort 3
300mgIR 5.03 ±1.34 115 ±32.4
(fasted) 4.87 111
7 ?8 ±
300mgSR3 " ° 2.39 ± 0.70 248 ±82.8
(fasted) 2 2 9 236
300mgSR3 3'55 * 3.59 ±1.13 165 ±50.2
(high-fat meal) 3A4 158
Cohort 4
300mgIR 5.23 ± 5.25 ±2.15 117 ±39.8
(fasted) 4^ 4.90 111
300mgSR4 lfl * 1.70 ± 1.25 456 ±259
(fasted) j'^ 1.40 387
300mgSR4 3·00± 3.13 ± 1.20 200 ±80.0
(high-fat meal) ' 2.92 186 Table 3A
Cohort/Treatment (μΜ) (,hT) Cmax/C ' (h)
SRI fasted vs IR 14.2%
(11.4%-17.5%)
SRI fed vs fasted 188%
(152%-232%)
SR2 fasted vs IR 8.9%
(6.7%- 11.9%)
SR2 fed vs fasted 258%
(193%-344%)
SR3 fasted vs IR 22.3%
(17.4%-28.6%)
SR3 fed vs fasted 191%
(150%-244%)
SR4 fasted vs IR 9.0%
(6.8%-11.9%)
SR4 fed vs fasted 193%
(146%-256%)
PK parameter values are mean ± SD and geometric mean except for T where median (90%
confidence interval) is reported.
Table 3B
r, . trr t . AUCo-t AUCo-oo CL/F
Cohort/Treatment , 4 . τ. »*,^ ,t n.
(μΜ*1ι) (μΜ*1ι) (L/h)
Geometric Mean Relative Bioavailability and the 90% Confidence Intervals
SRI fasted vs IR 34.1% 36.1%
(31.3%-37.0%) (33.3%-39.2%)
SRI fed vs fasted 191% 181%
(176%-208%) (167%- 196%)
SR2 fasted vs IR 22.4% 26.0%
(18.3%-27.4%) (21.6%-31.3%)
SR2 fed vs fasted 250% 218%
(204%-306%) (181%-262%)
SR3 fasted vs IR 45.4% 47.5%
(39.6%-52.0%) (41.9%-53.9%)
SR3 fed vs fasted 151% 145%
(132%-173%) (128%-164%)
SR4 fasted vs IR 26.9% 28.5%
(21.6%-33.4%) (23.2%-35.1%)
SR4 fed vs fasted 213% 215%
(171%-264%) (172%-268%)
PK parameter values are mean ± SD and geometric mean except for T where median (90% confidence interval) is reported. Table 4A
Cohort/Tre C max Tma
atment " (μΜ) (h) max/ l2h (h)
Cohort 5
300mgSR3 12 0.619 ±0.41 1.75 22.8 ±16.7 7.7 ±5.2
(fasted) 0.523 (0.50-4.0) 17.8 6.2
300mgSR3 12 0.875 ± 0.47 2.5 40.6 ±22.7 3.6 ±2.0
(medium-fat Q M ^2 ^ meal)
Geometric Mean Relative Bioavailability and the 90% Confidence Intervals
Figure imgf000055_0001
Pharmacokinetic parameter values are mean ± SD and geometric mean except for Tmax, where median (90% confidence interval) is reported.
Table 4B
Cohort/Tre AUCo-t AUCo-o CL/F
atment (μΜ*1ι) (μΜ*1ι) (L/h)
Cohort 5
300mgSR3 2.46 ± 1.13 2.58 ± 1.12 251 ± 105
(fasted) 2.23 2.36 230
300mgSR3 2.98 ± 1.34 3.02 ± 1.35 215 ±94.2
(medium-fat ∑J2 2Jfi 1% meal)
Geometric Mean Relative Bioavailability and the 90% Confidence Intervals
Figure imgf000055_0002
Pharmacokinetic parameter values are mean ± SD and geometric mean except for Tmax, where median
(90% confidence interval) is reported.
Table 5A
Cohort/Treatment
Figure imgf000056_0001
(nM) (h) h (h)
Cohort 6
2 25 mg SR3 12 55.1 ± 30.3 1.3 4.0 ± 2.6
NR
(fasted) 48.0 (0.50-4.0) 3.4
2 25 mg SR3 12 80.3 ± 27.3 3.0 2.2 ± 0.4
NR
(high-fat meal) 76.7 (1.5-6.0) 2.2
1 x 100 mg SR3 1 1 174 ± 69.5 1.8 3.0 ± 1.3
NR
(fasted) 161 (0.50-4.0) 2.7
Geometric Mean Relative Bioavailability and the 90% Confidence
Intervals
2 x 25 mg SR3 fed 160%
vs fasted (129%- 199%)
2 x 25 mg SR3 vs 1 x 58.7%''
100 mg SR3 (fasted) (46.9%-73.5%)
NC = not calculated because of significant numbers of mismatching Tiast within the subjects between treatments; NR = not reported because significant numbers of C values were BQL. PK parameter values are mean ± SD and geometric mean except for Tmax, where median (90% confidence interval) is reported.
Statistical comparison was dose-normalized.
Table 5B
Cohort/Treatme AUCo-t AUCo-*, CL/F
nt (nM*h) (nM*h) (L/h)
Cohort 6
2 x 25 mg SR3 205 ± 103 243 ± 99.9 429 ± 167
(fasted) 183 226 400 2 x 25 mg SR3 333 ± 104 376 ± 94.6 253 ± 57.7 (high- fat meal) 319 366 247
1 x 100 mg SR3 671 ± 230 704 ± 230 280 ± 81.5 (fasted) 639 673 268
Geometric Mean Relative Bioavailability and the 90% Confidence
Intervals
2 x 25 mg SR3 fed 174% 158%
vs fasted (150%-202%) (138%-182%
2 x 25 mg SR3 vs 1 x 66.1%°
100 mg SR3 (fasted) (57.5%-75.9°/i
NC = not calculated because of significant numbers of mismatching Tiast within the subjects between treatments; NR = not reported because significant numbers of Cn values were BQL. PK parameter values are mean ± SD and geometric mean except for Tmax, where median (90% confidence interval) is reported.
Statistical comparison was dose-normalized. The mean PK profiles following the fasting single-dose administration of 300 mg IR capsules were similar among the subjects in Cohorts 1 to 4 (FIG. 1).
Compared to the IR formulation, following fasting single-dose administration of the SR1-SR4 formulations (3 x 100 mg tablets), the observed plasma median Tmax values were moderately prolonged (by 0.3 to 1.5 hours) with significantly reduced mean Cmax values (the upper bounds of the 90% CI for the geometric mean Cmax ratios were < 30%), suggesting decreased absorption rate of the compound of Formula I for the SR tablets. The apparent mean disposition t½ observed in the terminal phase was significantly longer, ranging from 7.3 to 1 1 hours for SR1-SR4, as compared to about 2 hours for the IR capsule, indicating that the systemic elimination of the compound of Formula I was likely rate-limited by its absorption, which was sustained in the terminal disposition phase. As a result of lower Cmax and longer disposition t½, the Cmax/ Ci2h ratios were significantly lower for the SR tablets compared to the IR capsule for the same subjects studied. The geometric mean Cmax/Ci2h ratios were 1 1.6-, 8.6-, 19.3-, and 8.9-fold, respectively, for SRI, SR2, SR3, and SR4 tablets, as compared to 1 12- to 162-fold for the IR capsules administered in the fasted state.
For administration in the fasted state, the 4 SR tablets showed reduced relative bioavailability compared to the IR capsule dosed in the same subjects. The percent geometric mean ratios (90% CI) of Cmax were 14.2 % (11.4%-17.5%), 8.9% (6.7%- 1 1.9%), 22.3% (17.4%-28.6%) and 9.0% (6.8%-11.9%) for SRI, SR2, SR3, and SR4, respectively. The percent geometric mean ratios (90% CI) of AUCo -∞ were 36.1 % (33.3%-39.2%), 26.0% (21.6%-31.3%), 47.5% (41.9%-53.9%), and 28.5% (23.2%- 35.1%) for SRI, SR2, SR3, and SR4, respectively. SR3 and SRI demonstrated the best and second best relative bioavailability, respectively, among the SR formulations tested.
Dosed in the fasted state, the intersubject variability as measured by percent coefficient of variability (CV%) in plasma exposure was significantly higher for the gastroretentive formulation SR4, but comparable among the 3 regular SR tablets designed for intestinal release. The intersubject CV% for the 100 mg SRI tablet was 39% and 33% for Cmax and
Figure imgf000057_0001
respectively. The intersubject CV% for the 100 mg SR2 tablet was 50% and 37% for Cmax and AUCo -∞, respectively. The intersubject CV% for the 100 mg SR3 tablet was 43% and 29% for Cmax and
Figure imgf000057_0002
respectively. The intersubject CV% for the 100 mg SR4 tablet was 83% and 73% for Cmax and AUCo-oo, respectively. Pooling all subjects in Cohorts 1-5 (n = 59) who were administered 300 mg IR in the fasted state, the intersubject CV% was 49% and 39% for Cmax and AUCo -∞, respectively, comparable to the CV% values observed for SRI, SR2, and SR3.
A positive food effect was observed for all SR formulations studied at the 300 mg (3 x 100 mg) dose level. Administered after a high-fat meal, geometric mean Cmax and AUCo-oo values increased by 88% and 81%, respectively, for SRI; by 158% and 118%, respectively; for SR2; by 91% and 45%; respectively; for SR3; and by 93% and 115%; respectively; for SR4. The food effect was moderate for a medium- fat meal as compared to a high- fat meal, as suggested by the data for SR3 in Cohort 5. For SR3, Cmax and AUCo∞ values increased by 46% and 17%, respectively, when it was administered following a standardized medium- fat meal. Administration with food did not significantly change the intersubject CV% in compound of Formula I plasma exposure for SRI, SR2, and SR3, which are SR formulations designed for intra-intestinal release. For SR4, which is a gastroretentive SR formulation, the intersubject CV% in plasma exposures appeared to be significantly reduced with a concomitant high-fat meal.
This study also explored the dose-normalized relative bioavailability of the 25 mg SR tablet in reference to the 100 mg SR3 tablet. For the subjects in Cohort 6, the dose-normalized Cmax and AUCo∞ percent geometric mean ratio for the 2 x 25 mg SR3 treatment was 59% and 66%, respectively, versus the 1 x 100 mg SR3 administration in the fasted state. However, due to the supralinear dose-exposure relationship for the compound of Formula I, the relative bioavailability of the 25 mg SR tablet may be underestimated. For the 2 x 25 mg SR dose, a high-fat meal increased compound of Formula I Cmax and AUCo∞ by 60% and 58%, respectively.
For the four SR formulations evaluated, the observed apparent disposition t½ was comparable, and the Cmax/Ci2h ratios from a fasting single-dose administration (which is used as a proxy for P/T ratio from twice-daily administration) were similar among SRI, SR2, and SR4 (~10-fold) and moderately higher for SR3 (~20-fold). Overall, all 4 SR formulations demonstrated a significantly flatter PK profile compared the IR capsule, meeting an important objective for sustained release. Bioavailability of orally administered drug products may be defined by the rate and extent of the drug absorption into systemic circulation. A reduction in drug absorption rate by limiting the drug release rate from drug products is a design requirement in sustained release formulations. Therefore, for SR formulations, the extent of the compound of Formula I absorption as measured by the plasma AUCo -∞ is used as the primary endpoint to assess the relative bioavailability. Thus, the mean relative bioavailability is similar between SR2 (26%) and SR4 (29%), which was slightly lower than that of SRI (36%). The best relative bioavailability was observed for SR3 (48%). The results are in line with the in vitro dissolution profiles obtained before conducting this study.
There was an apparent inverse correlation between the food effect and relative bioavailability for the SR formulations. On average, dosed with a high-fat meal, the food-effect measured by the increase in AUCo∞ was the greatest for SR2 (118%) and SR4 (115%), which was lower than that for SRI (81%). The smallest food effect was observed for SR3 (45%). This correlation was also apparent when the data from all the subjects were pooled together. A quantile plot using the pooled individual data (divided into 5 bins with 9 subjects per bin) suggests that the food effect was more significant (> 2-fold increase in AUC) for the subjects with relative bioavailability less than 35%, regardless of the formulation. The food effect was moderate (-50% or less increase in AUC) for the subjects with relative bioavailability greater than 40%, regardless of formulation. SR3 delivered a mean relative bioavailability of 48% and is likely to be associated with a moderate food effect. In fact, when the SR3 tablet (3 x 100 mg) was dosed with a medium- fat meal (which is a more typical daily diet), the observed increase in geometric mean AUCo∞ was only 17%, suggesting that this formulation may be administered without regard to medium- or low-fat meals. From the perspective of avoiding significant food effect, SR3 is superior to the other formulations.
Example 4. Clinical Results in Phase 2a in patients with active rheumatoid arthritis (RA)
An initial 28 day part of the study was conducted in order to select doses moving forward, guiding dose selection for the 3 month second part of the study. Part 2 of the study was randomized, double-blind, placebo controlled (sponsor unblinded) with treatment for 84 days. Sixty subjects to be randomized, using the same population as in Part 1: single cohort, five parallel treatment groups, 12 subjects each: 100 mg SR3 tablets BID; 300 mg (3 x 100 mg SR3 tablets) QD; 200 mg (2 x 100 mg SR3 tablets) BID; 600 mg (6 x 100 mg SR3 tablets) QD; and placebo. Interim data was submitted to ACR (American College of Rheumatology) 2013 (n=40 subjects who completed day 84). The ACR scores at 3 months re shown in Table 6. The ACR scores for the 600 mg QD are unprecedented as compared to other JAK inhibitors that are approved for treatment of RA. For example, the approved product for tofacitinib citrate (5 mg BID) showed much lower ACR scores at 3 months: 59% (ACR20), 31% (ACR50), and 15% (ACR70) (Table 5 of XELJANZ® - tofacitinib citrate tablet - label).
Table 6
Figure imgf000060_0001
The percent change from baseline for hemoglobin was also studied for each of the dosing regimens (FIG. 4). As can be seen in FIG. 4, the 200 mg BID dose showed a drop away from the baseline compared to the other doses which tended to stay close to the placebo levels. For example, the 600 mg QD dose did not show the same downward trend as shown for the BID dose. However, as can be seen in Table 6, the once-daily dosing (600 mg QD) did not compromise efficacy compared with the BID doses. This indicates that the once-daily dosing (such as 600 mg QD) may achieve maximal efficacy without inducing side-effects such anemia. As shown in FIG. 4 and Table 6, the 600 mg QD dose has robust efficacy with trivial change in hemoglobin levels. It is believed that this efficacy/side-effect profile may be due to the QD dose achieving maximal JAKl signaling (tied to efficacy) with low JAK2 inhibition at the trough, as JAK2 signaling is tied to hematopoiesis. This hypothesis is supported by the PK derived JAKl (IL-6) and JAK2 (TPO) inhibition data for the compound of Formula at various doses (Table 7). In particular, the 600 mg QD dose showed similar average IL-6 inhibition to the 200 mg BID and 400 mg BID doses (61% versus 64% and 69%), but lower trough TPO inhibition in comparison to the 200 mg BID and 400 mg BID doses (4% versus 13% and 16%). The trough IL-6 inhibition for the 600 mg QD dose is also lower than the trough IL-6 inhibition for the 200 mg BID and 400 mg BID doses, which suggests that there may be a reduction in infection from the QD dose.
Table 7
Figure imgf000061_0001
Example 5. Clinical Results in Patients with Plaque Psoriasis
A double-blind (sponsor unblinded), randomized, placebo controlled study was conducted in approximately 48 subjects treated for 28 days. Eligibility requirements included: active plaque psoriasis for at least 6 months at screening; body surface area (BSA) of plaque psoriasis of > 5%; psoriasis area and severity index (PASI) score of > 5; static physician's global assessment (sPGA) score of > 3; inadequate response to topical therapies; innovative design allowing rapid progress between doses, with conservative safety assessment. Four staggered dose groups of 12 subjects each (9 active and 3 PBO) progressing from 100 mg QD to 200 mg QD to 200 mg BID to 600 mg QD. Once the 4th subject (block of 3 active 1 PBO) completed 28 days administration without a Grade 3 or higher AE, the next group of 12 subjects initiated treatment with the next highest dose; while the first 4 subjects in this group are treated for 28 days, the 1st group is filled
60 subjects with moderate to severe psoriasis were randomized. There were five treatment groups: placebo, 100 mg QD, 200 mg QD, 200 mg BID and 600 mg QD. A sequential method of recruitment was used, increasing from the lowest dose to the highest, each after the completion of 28 days for the first four subjects in the previous dose. The results at 28 days are show in Table 8 (PASI 50 is Psoriasis Area and Severity Index). These PASI 50 score of 81.8% for the 600 mg QD dose are unprecedented as compared to other JAK inhibitors that are in development for treatment of psoriasis. For example, 5 mg tofacitinib (also known as tasocitinib) showed lower PASI 50 score of 65.3% at 12 weeks (published on
http://press.pfizer.com on 10/7/2010). The 5 mg tofacitinib dose is the approved dosage level for RA for safety reasons in the US.
Table 8
Figure imgf000062_0001
Example 6. Open-Label Phase II Study in Patients with Myelofibrosis
In this study, patients with age >18 years, a diagnosis of primary myelofibrosis (PMF) or post-polycythemia vera MF or post-essential thrombocythemia MF
(JAK2V617F positive or negative mutation status), platelet counts > 50 x 109/L, hemoglobin levels > 8.0 g/dL (transfusions permitted to achieve these levels), intermediate- 1 or higher per DIPSS criteria, and palpable spleen or prior splenectomy were enrolled. Three different dose cohorts were assessed: (1) 100 mg SR3 tablets BID) (2) 200 mg (2 x 100 mg SR3 tablets) BID; and (3) 600 mg (6 x 100 mg SR3 tablets) QD. FIG. 5(a)-(b) show interim results with respect to proportion of subjects with > 50% reduction in total symptom score (TSS) in each dose group per the modified Myelofibrosis Symptom Assessment Form (MFSAF) v3.0 electronic diary at week 12 compared with baseline (The modified MFSAF v3.0 comprises 19 questions assessing MF-related symptoms on a scale of 0 (absent) to 10 (worst imaginable)). FIG. 5(a) depicts the percentage of patients having a > 50% reduction in TSS at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) (patients who discontinued prior to the week 12 visit were considered nonresponders). FIG. 5(b) depicts the percent change in TSS from baseline at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) (only patients with baseline and week 12 data were included). FIG. 6(a) depicts mean hemoglobin levels (g/dL) over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) (interim results of study for all patients). FIG. 6(b) depicts mean hemoglobin levels (g/dL) over time by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) at 48 weeks. FIG. 6(c) depicts mean hemoglobin levels (g/dL) over time by dose cohort at 48 weeks as an average for three dose cohorts as compared to individuals dosed with placebo or ruxolitinib (ruxolitinib was dosed according to the label for Jakafi®). The data show an increase in hemoglobin levels for the 600 mg QD dose. Finally, Table 9 below show interim hematology laboratory results (new and worsening) for each dose cohort. Table 9a shows the hematology laboratory results (new and worsening) for each dose cohort after long exposure.
Table 9
Figure imgf000063_0001
Table 9a
Figure imgf000064_0001
Example A: In vitro JAK Kinase Assay
The compound of Formula I herein was tested for inhibitory activity of JAK targets according to the following in vitro assay described in Park et ah, Analytical Biochemistry 1999, 269, 94-104. The catalytic domains of human JAKl (a.a. 837- 1 142) and JAK2 (a.a. 828-1132) with an N-terminal His tag were expressed using baculovirus in insect cells and purified. The catalytic activity of JAKl and JAK2 was assayed by measuring the phosphorylation of a biotinylated peptide. The
phosphorylated peptide was detected by homogenous time resolved fluorescence (HTRF). IC50S of compounds were measured for each kinase in the 40 microL reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8) buffer with 100 mM NaCl, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA. For the 1 mM IC50 measurements, ATP concentration in the reactions was 1 mM. Reactions were carried out at room temperature for 1 hr and then stopped with 20 μϊ^ 45 mM EDTA, 300 nM SA-APC, 6 nM Eu-Py20 in assay buffer (Perkin Elmer, Boston, MA).
Binding to the Europium labeled antibody took place for 40 minutes and HTRF signal was measured on a Fusion plate reader (Perkin Elmer, Boston, MA). The compound of Formula I and the adipic acid salt had an IC50 at JAKl of < 5 nM (measured at 1 mM ATP) with a JAK2/JAK1 ratio of > 10 (measured at 1 mM ATP). Example B: Cellular Assays
Cancer cell lines dependent on cytokines and hence JAK/STAT signal transduction, for growth, can be plated at 6000 cells per well (96 well plate format) in RPMI 1640, 10% FBS, and 1 nG/niL of appropriate cytokine. Compounds can be added to the cells in DMSO/media (final concentration 0.2% DMSO) and incubated for 72 hours at 37 °C, 5% CO2. The effect of compound on cell viability is assessed using the CellTiter-Glo Luminescent Cell Viability Assay (Promega) followed by TopCount (Perkin Elmer, Boston, MA) quantitation. Potential off-target effects of compounds are measured in parallel using a non-JAK driven cell line with the same assay readout. All experiments are typically performed in duplicate.
The above cell lines can also be used to examine the effects of compounds on phosphorylation of JAK kinases or potential downstream substrates such as STAT proteins, Akt, Shp2, or Erk. These experiments can be performed following an overnight cytokine starvation, followed by a brief preincubation with compound (2 hours or less) and cytokine stimulation of approximately 1 hour or less. Proteins are then extracted from cells and analyzed by techniques familiar to those schooled in the art including Western blotting or ELISAs using antibodies that can differentiate between phosphorylated and total protein. These experiments can utilize normal or cancer cells to investigate the activity of compounds on tumor cell survival biology or on mediators of inflammatory disease. For example, with regards to the latter, cytokines such as IL-6, IL-12, IL-23, or IFN can be used to stimulate JAK activation resulting in phosphorylation of STAT protein(s) and potentially in transcriptional profiles (assessed by array or qPCR technology) or production and/or secretion of proteins, such as IL-17. The ability of compounds to inhibit these cytokine mediated effects can be measured using techniques common to those schooled in the art.
Compounds herein can also be tested in cellular models designed to evaluate their potency and activity against mutant JAKs, for example, the JAK2V617F mutation found in myeloid proliferative disorders. These experiments often utilize cytokine dependent cells of hematological lineage (e.g. BaF/3) into which the wild- type or mutant JAK kinases are ectopically expressed (James, C, et al. Nature 434: 1144-1148; Staerk, J., et al. JBC 280:41893-41899). Endpoints include the effects of compounds on cell survival, proliferation, and phosphorylated JAK, STAT, Akt, or Erk proteins.
Certain compounds herein can be evaluated for their activity inhibiting T-cell proliferation. Such as assay can be considered a second cytokine (i.e. JAK) driven proliferation assay and also a simplistic assay of immune suppression or inhibition of immune activation. The following is a brief outline of how such experiments can be performed. Peripheral blood mononuclear cells (PBMCs) are prepared from human whole blood samples using Ficoll Hypaque separation method and T-cells (fraction 2000) can be obtained from PBMCs by elutriation. Freshly isolated human T-cells can be maintained in culture medium (RPMI 1640 supplemented with 10% fetal bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin) at a density of 2 x 106 cells/ml at 37 °C for up to 2 days. For IL-2 stimulated cell proliferation analysis, T-cells are first treated with Phytohemagglutinin (PHA) at a final concentration of 10 μg/mL for 72h. After washing once with PBS, 6000 cells/well are plated in 96-well plates and treated with compounds at different concentrations in the culture medium in the presence of 100 U/mL human IL-2 (ProSpec-Tany TechnoGene; Rehovot, Israel). The plates are incubated at 37 °C for 72h and the proliferation index is assessed using CellTiter-Glo Luminescent reagents following the manufactory suggested protocol (Promega;
Madison, WI).
Example C: In vivo anti-tumor efficacy
Compounds herein can be evaluated in human tumor xenograft models in immune compromised mice. For example, a tumorigenic variant of the ΓΝΑ-6 plasmacytoma cell line can be used to inoculate SCID mice subcutaneous ly (Burger, R., et al. Hematol J. 2:42-53, 2001). Tumor bearing animals can then be randomized into drug or vehicle treatment groups and different doses of compounds can be administered by any number of the usual routes including oral, i.p., or continuous infusion using implantable pumps. Tumor growth is followed over time using calipers. Further, tumor samples can be harvested at any time after the initiation of treatment for analysis as described above (Example B) to evaluate compound effects on JAK activity and downstream signaling pathways. In addition, selectivity of the compound(s) can be assessed using xenograft tumor models that are driven by other know kinases (e.g. Bcr-Abl) such as the K562 tumor model.
Example D: Murine Skin Contact Delayed Hypersensitivity Response Test
Compounds 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 Jan; 19(l):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 Jan;38(l-2): 1 16-21).
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 μΤ (10 μΐ. on the internal pinna and 10 μΐ. 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 compounds 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 either systemically or topically (topical application of the treatment to the ears). Efficacies of the test compounds are indicated by a reduction in ear swelling comparing to the situation without the treatment. Compounds causing a reduction of 20% or more were 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 compounds 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 compounds, vehicle, or dexamethasone (a clinically efficacious treatment for psoriasis), or without any treatment, in the DTH model for comparisons. Test compounds and the dexamethasone can produce similar transcriptional changes both qualitatively and quantitatively, and both the test compounds and dexamethasone can reduce the number of infiltrating cells. Both systemically and 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.
Example E: In vivo anti-inflammatory activity
Compounds herein can be evaluated in rodent or non-rodent models designed to replicate a single or complex inflammation response. For instance, rodent models of arthritis can be used to evaluate the therapeutic potential of compounds dosed preventatively or therapeutically. These models include but are not limited to mouse or rat collagen-induced arthritis, rat adjuvant-induced arthritis, and collagen antibody- induced arthritis. Autoimmune diseases including, but not limited to, multiple sclerosis, type I-diabetes mellitus, uveoretinitis, thyroditis, myasthenia gravis, immunoglobulin nephropathies, myocarditis, airway sensitization (asthma), lupus, or colitis may also be used to evaluate the therapeutic potential of compounds herein. These models are well established in the research community and are familiar to those schooled in the art (Current Protocols in Immunology, Vol 3., Coligan, J.E. et al, Wiley Press.; Methods in Molecular Biology: Vol. 225, Inflammation Protocols., Winyard, P.G. and Willoughby, D.A., Humana Press, 2003.). Example F: Animal Models for the Treatment of Dry Eye, Uveitis, and
Conjunctivitis
Agents may be evaluated in one or more preclinical models of dry eye known to those schooled in the art including, but not limited to, the rabbit concanavalin A (ConA) lacrimal gland model, the scopolamine mouse model (subcutaneous or transdermal), the Botulinumn mouse lacrimal gland model, or any of a number of spontaneous rodent auto-immune models that result in ocular gland dysfunction (e.g. NOD-SCID, MRL/lpr, or ZB/NZW) (Barabino et al, Experimental Eye Research 2004, 79, 613-621 and Schrader et al, Developmental Opthalmology, Karger 2008, 41, 298-312, each of which is incorporated herein by reference in its entirety).
Endpoints in these models may include histopathology of the ocular glands and eye (cornea, etc.) and possibly the classic Schirmer test or modified versions thereof (Barabino et al.) which measure tear production. Activity may be assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists.
Agents may be evaluated in one or more preclinical models of uveitis known to those schooled in the art. These include, but are not limited to, models of experimental autoimmune uveitis (EAU) and endotoxin induced uveitis (EIU). EAU experiements may be performed in the rabbit, rat, or mouse and may involve passive or activate immunization. For instance, any of a number or retinal antigens may be used to sensitize animals to a relevant immunogen after which animals may be challenged ocuarly with the same antigen. The EIU model is more acute and involves local or systemic administration of lipopolysaccaride at sublethal doses. Endpoints for both the EIU and EAU models may include fundoscopic exam, histopathology amongst others. These models are reviewed by Smith et al. (Immunology and Cell Biology 1998, 76, 497-512, which is incorporated herein by reference in its entirety). Activity is assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists. Some models listed above may also develop scleritis/episcleritis, chorioditis, cyclitis, or iritis and are therefore useful in investigating the potential activity of compounds for the therapeutic treatment of these diseases. Agents may also be evaluated in one or more preclinical models of conjunctivitis known those schooled in the art. These include, but are not limited to, rodent models utilizing guinea-pig, rat, or mouse. The guinea-pig models include those utilizing active or passive immunization and/or immune challenge protocols with antigens such as ovalbumin or ragweed (reviewed in Groneberg, D.A., et al, Allergy 2003, 58, 1101-11 13, which is incorporated herein by reference in its entirety). Rat and mouse models are similar in general design to those in the guinea- pig (also reviewed by Groneberg). Activity may be assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists. Endpoints for such studies may include, for example, histological, immunological, biochemical, or molecular analysis of ocular tissues such as the conjunctiva.
Example G: In vivo protection of bone
Compounds may be evaluated in various preclinical models of osteopenia, osteoporosis, or bone resorption known to those schooled in the art. For example, ovariectomized rodents may be used to evaluate the ability of compounds to affect signs and markers of bone remodeling and/or density (W.S.S. Jee and W. Yao, J Musculoskel. Nueron. Interact., 2001, 1(3), 193-207, which is incorporated herein by reference in its entirety). Alternatively, bone density and architecture may be evaluated in control or compound treated rodents in models of therapy (e.g.
glucocorticoid) induced osteopenia (Yao, et al. Arthritis and Rheumatism, 2008, 58(6), 3485-3497; and id. 58(11), 1674-1686, both of which are incorporated herein by reference in its entirety). In addition, the effects of compounds on bone resorption and density may be evaluable in the rodent models of arthritis discussed above (Example E). Endpoints for all these models may vary but often include histological and radiological assessments as well as immunohisotology and appropriate biochemical markers of bone remodeling.

Claims

WHAT IS CLAIMED IS:
1. A method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising orally administering to said patient a once-daily dose of about 400 mg to about 600 mg on a free base basis of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose comprises one or more sustained release dosage forms each comprising { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
2. A method according to claim 1, wherein the method comprises administering to said patient the one or more sustained release dosage forms as a once-daily dosage of about 600 mg on a free base basis of { l-{ l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
3. A method according to claim 1, wherein the one or more sustained release dosage forms are six dosage forms of about 100 mg on a free base basis of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
4. A method according to claim 1, wherein the one or more sustained release dosage forms are three dosage forms of about 200 mg on a free base basis of { 1- { 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
5. A method according to claim 1, wherein the one or more sustained release dosage forms are two dosage forms of about 300 mg on a free base basis of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
6. A method according to claim 1, wherein the one or more sustained release dosage forms is one dosage form of about 600 mg on a free base basis of { 1- { l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
7. A method according to any one of claims 1 to 6, wherein said autoimmune disease is a skin disorder, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, type I diabetes, lupus, inflammatory bowel disease, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorder, atopic dermatitis, psoriasis, skin sensitization, skin irritation, skin rash, contact dermatitis or allergic contact sensitization.
8. A method according to any one of claims 1 to 6, wherein said autoimmune disease is rheumatoid arthritis.
9. A method according to claim 8, wherein the ACR70 score of said patient is greater than about 40%.
10. A method according to claim 8, wherein the ACR70 score of said patient is greater than about 50%.
11. A method according to any one of claims 1 to 6, wherein said autoimmune disease is psoriasis.
12. A method according to claim 11, wherein the PASI 50 score is greater than about 70%.
13. A method according to any one of claims 1 to 6, wherein said cancer is prostate cancer, renal cancer, hepatic cancer, breast cancer, lung cancer, thyroid cancer, Kaposi's sarcoma, Castleman's disease, pancreatic cancer, lymphoma, leukemia, multiple myeloma, polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM), primary myelofibrosis (PMF), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hypereosinophilic syndrome (HES), idiopathic myelofibrosis (IMF), or systemic mast cell disease (SMCD).
14. A method according to any one of claims 1 to 6, wherein said
myeloproliferative disorder is primary myelofibrosis (PMF).
15. A method according to claim 14, wherein the method results in a reduced total symptom score (TSS) of said patient compared with baseline.
16. A method according to claim 14, wherein said method results in reduced anemia.
17. A method according to any one of claims 1 to 6, wherein said bone resorption disease is osteoporosis, osteoarthritis, bone resorption associated with hormonal imbalance, bone resorption associated with hormonal therapy, bone resorption associated with autoimmune disease, or bone resorption associated with cancer.
18. A method according to any one of claims 1 to 17, wherein oral administration of one or more sustained release dosage forms to a fasted individual provides a mean time to peak plasma concentration (Tmax) of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrileof about 0.5 hours to about 3 hours.
19. A method according to any one of claims 1 to 17, wherein oral administration of one or more sustained release dosage forms to a fasted individual provides a mean time to peak plasma concentration (Tmax) of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of at least 0.5 hours.
20. A method according to any one of claims 1 to 19, wherein oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 5 to about 50.
21. A method according to any one of claims 1 to 19, wherein oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 9 to about 40.
22. A method according to any one of claims 1 to 19, wherein oral administration of one or more sustained release dosage forms to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3 -d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3 -yl} acetonitrile of about 15 to about 30.
23. A method according to any one of claims 1 to 22, wherein oral administration of one or more sustained release dosage forms to a fasted individual provides a mean half-life (ti/2) of { l- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrileof about 1 hour to about 20 hours.
24. A method according to any one of claims 1 to 22, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of { 1 - { 1 -[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrileof about 1 hour to about 9 hours.
25. A method according to any one of claims 1 to 22, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean time to peak plasma concentration (Tmax) of { 1 - { 1 -[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of at least 1.5 hours.
26. A method according to any one of claims 1 to 25, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3- [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 - yl}acetonitrile of about 10 to about 70.
27. A method according to any one of claims 1 to 25, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h)of { 1 - { 1 -[3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3- [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 - yl}acetonitrile of about 15 to about 50.
28. A method according to any one of claims 1 to 25, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (Ci2h) of { l-{ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3- [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 - yl}acetonitrile of about 25 to about 45.
29. A method according to any one of claims 1 to 28, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean half-life (tie) of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 1 hour to about 7 hours.
30. A method according to any one of claims 1 to 28, wherein oral administration of one or more sustained release dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of { l- { l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile of about 2 hours to about 5 hours.
31. A method according to any one of claims 1 to 30, wherein the one or more sustained release dosage forms are each a tablet.
32. A method according to any one of claims 1 to 31, wherein the one or more sustained release dosage forms are prepared by process comprising wet granulation.
33. A method according to any one of claims 1 to 32, wherein the one or more sustained release dosage forms each comprises one or more hypromelloses.
34. A method according to any one of claims 1 to 32, wherein the one or more sustained release dosage forms each comprises one or more excipients independently selected from hypromelloses and microcrystalline celluloses.
35. A method according to any one of claims 1 to 34, wherein the one or more sustained release dosage forms each comprises one or more excipients independently selected from hypromelloses, microcrystalline celluloses, magnesium stearate, lactose, and lactose monohydrate.
36. A method according to any one of claims 1 to 32, wherein the one or more sustained release dosage forms each comprises a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP and a second hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP.
37. A method according to any one of claims 1 to 32, wherein the one or more sustained release dosage forms each comprises about 10% to about 15% by weight of one or more hypromelloses.
38. A method according to any one of claims 1 to 32 and 37, wherein the one or more sustained release dosage forms each comprises about 16% to about 22% by weight of microcrystalline cellulose.
39. A method according to any one of claims 1 to 32 and 37 to 38, wherein the one or more sustained release dosage forms each comprises about 45% to about 55% by weight of lactose monohydrate.
40. A method according to any one of claims 1 to 17 and 37 to 39, wherein the one or more sustained release dosage forms each comprises about 0.3% to about 0.7% by weight of magnesium stearate.
41. A method according to any one of claims 1 to 40, wherein said salt is { 1- { 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt.
42. One or more sustained release dosage forms, which together provide a once- daily oral dosage of about 600 mg on a free base basis of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
43. A dose comprising one or more sustained release dosage forms, which provide a once-daily oral dosage of about 600 mg on a free base basis of { l-{ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
44. One or more sustained release dosage forms each comprising { l- { l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl } -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said one or more sustained release dosage forms together provide a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of { 1- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
45. A dose, comprising one or more sustained release dosage forms each comprising { 1- { l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt thereof; wherein said dose provides a once-daily oral dosage of about 400 mg to about 600 mg on a free base basis of { l- { l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof, to a patient.
PCT/US2014/049940 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor WO2015021153A1 (en)

Priority Applications (38)

Application Number Priority Date Filing Date Title
BR112016002571-7A BR112016002571B1 (en) 2013-08-07 2014-08-06 SUSTAINED RELEASE TABLETS WITH A JAK1 INHIBITOR
MX2016001639A MX2016001639A (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor.
EP14753182.6A EP3030227B1 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
DK14753182.6T DK3030227T3 (en) 2013-08-07 2014-08-06 RELEASING RELEASE DOSAGE FORMS FOR A JAK1 INHIBITOR
EA201690357A EA201690357A1 (en) 2013-08-07 2014-08-06 DOSAGE FORMS WITH Slow Release For JAK1 Inhibitor
UAA201602100A UA120499C2 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
CR20190343A CR20190343A (en) 2013-08-07 2014-08-06 PROLONGED RELEASE DOSAGE FORMS FOR A JAK 1 INHIBITOR (Exp Divisional: 2016-0102)
KR1020167006096A KR20160045081A (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
SG11201600815WA SG11201600815WA (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
IL277554A IL277554B2 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
CN202110883638.4A CN114010611B (en) 2013-08-07 2014-08-06 Sustained release dosage forms of JAK1 inhibitors
JP2016533398A JP6334700B2 (en) 2013-08-07 2014-08-06 Sustained release dosage form for JAK1 inhibitor
AU2014305989A AU2014305989B2 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a JAK1 inhibitor
CN202310115279.7A CN116036089A (en) 2013-08-07 2014-08-06 Sustained release dosage forms of JAK1 inhibitors
NZ717230A NZ717230B2 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
CN201480052299.3A CN105579032A (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a JAK1 inhibitor
PE2021002226A PE20220579A1 (en) 2013-08-07 2014-08-06 PROLONGED RELEASE DOSAGE FORMS FOR A JAK 1 INHIBITOR
PL14753182T PL3030227T3 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
CR20230316A CR20230316A (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
MYPI2016000200A MY195091A (en) 2013-08-07 2014-08-06 Sustained Release Dosage Forms for a JAK1 Inhibitor
ES14753182T ES2792549T3 (en) 2013-08-07 2014-08-06 Sustained-release dosage forms for a JAK1 inhibitor
LTEP14753182.6T LT3030227T (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
EP20164849.0A EP3721873A1 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
KR1020227023370A KR20220103810A (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
SI201431549T SI3030227T1 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
CA2920108A CA2920108C (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
RS20200696A RS60469B1 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
KR1020217018433A KR102419714B1 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor
BR122020001831A BR122020001831A8 (en) 2013-08-07 2014-08-06 {1-{1-[3-FLUORINE-2-(TRIFLUORMETHYL)ISONICOTINOL]PIPERIDIN-4-IL}-3-[4-(7H-PYROLO[2,3-D]PRIMIDIN-4-IL) -1H- PYRAZOL-1-YL]AZETIDIN-3-YL}ACETONITRIL, ITS USE, SUSTAINED RELEASE DOSAGE FORMS AND DOSES
IL243920A IL243920B (en) 2013-08-07 2016-02-03 Sustained release dosage forms for a jak1 inhibitor
PH12016500243A PH12016500243B1 (en) 2013-08-07 2016-02-04 Sustained release dosage forms for a jak1 inhibitor
CR20160102A CR20160102A (en) 2013-08-07 2016-03-01 PROLONGED RELEASE DOSAGE FORMS FOR A JAK INHIBITOR 1
PH12019502074A PH12019502074A1 (en) 2013-08-07 2019-09-12 Sustained release dosage forms for a jak1 inhibitor
PH12019502072A PH12019502072A1 (en) 2013-08-07 2019-09-12 Sustained release dosage forms for a jak1 inhibitor
AU2019257368A AU2019257368B2 (en) 2013-08-07 2019-10-28 Sustained release dosage forms for a jak1 inhibitor
HRP20200955TT HRP20200955T1 (en) 2013-08-07 2020-06-16 Sustained release dosage forms for a jak1 inhibitor
CY20201100572T CY1123314T1 (en) 2013-08-07 2020-06-23 EXTENDED RELEASE DOSAGE FORMS FOR JAK1 INHIBITOR
AU2021202916A AU2021202916A1 (en) 2013-08-07 2021-05-07 Sustained release dosage forms for a jak1 inhibitor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361863325P 2013-08-07 2013-08-07
US61/863,325 2013-08-07
US201361913066P 2013-12-06 2013-12-06
US61/913,066 2013-12-06

Publications (1)

Publication Number Publication Date
WO2015021153A1 true WO2015021153A1 (en) 2015-02-12

Family

ID=51383933

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/049940 WO2015021153A1 (en) 2013-08-07 2014-08-06 Sustained release dosage forms for a jak1 inhibitor

Country Status (30)

Country Link
US (4) US9655854B2 (en)
EP (2) EP3030227B1 (en)
JP (5) JP6334700B2 (en)
KR (3) KR102419714B1 (en)
CN (3) CN116036089A (en)
AU (3) AU2014305989B2 (en)
BR (1) BR122020001831A8 (en)
CA (2) CA2920108C (en)
CL (1) CL2016000292A1 (en)
CR (3) CR20190343A (en)
CY (1) CY1123314T1 (en)
DK (1) DK3030227T3 (en)
EA (1) EA201690357A1 (en)
ES (1) ES2792549T3 (en)
HR (1) HRP20200955T1 (en)
HU (1) HUE049345T2 (en)
IL (2) IL277554B2 (en)
LT (1) LT3030227T (en)
MX (2) MX2016001639A (en)
MY (1) MY195091A (en)
PE (2) PE20160223A1 (en)
PH (3) PH12016500243B1 (en)
PL (1) PL3030227T3 (en)
PT (1) PT3030227T (en)
RS (1) RS60469B1 (en)
SG (3) SG11201600815WA (en)
SI (1) SI3030227T1 (en)
TW (3) TWI730935B (en)
UA (1) UA120499C2 (en)
WO (1) WO2015021153A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016020901A1 (en) 2014-08-07 2016-02-11 Acerta Pharma B.V. Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on btk occupancy and btk resynthesis rate
WO2016196244A1 (en) 2015-05-29 2016-12-08 Incyte Corporation Pyridineamine compounds useful as pim kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
WO2017044730A1 (en) 2015-09-09 2017-03-16 Incyte Corporation Salts of a pim kinase inhibitor
WO2017059251A1 (en) 2015-10-02 2017-04-06 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
WO2020181034A1 (en) * 2019-03-05 2020-09-10 Incyte Corporation Jak1 pathway inhibitors for the treatment of chronic lung allograft dysfunction

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MY162590A (en) 2005-12-13 2017-06-30 Incyte Holdings Corp Heteroaryl substituted pyrrolo[2,3-b] pyridines and pyrrolo[2,3-b] pyrimidines as janus kinase inhibitors
EP3495369B1 (en) 2007-06-13 2021-10-27 Incyte Holdings Corporation Use of salts of the janus kinase inhibitor (r)-3-(4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h- pyrazol-1-yl)-3- cyclopentylpropanenitrile
EA025520B1 (en) 2009-05-22 2017-01-30 Инсайт Холдингс Корпорейшн 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
MX2011012262A (en) 2009-05-22 2012-01-25 Incyte Corp 3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl]octane- or heptane-nitrile as jak inhibitors.
SI3354652T1 (en) 2010-03-10 2020-08-31 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as jak1 inhibitors
SG10201910912TA (en) 2010-05-21 2020-01-30 Incyte Corp Topical Formulation for a JAK Inhibitor
BR112013012502A2 (en) 2010-11-19 2019-03-06 Incyte Corporation substituted cyclobutyl pyrrolopyridine and derivative pyrrolopyrimidine derivatives as jak inhibitors
MY165963A (en) 2011-06-20 2018-05-18 Incyte Holdings Corp Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
UA111854C2 (en) 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн METHODS AND INTERMEDIATE COMPOUNDS FOR JAK INHIBITORS
SG10201703533VA (en) 2012-11-01 2017-06-29 Incyte Corp Tricyclic fused thiophene derivatives as jak inhibitors
SG11201503695XA (en) 2012-11-15 2015-06-29 Incyte Corp Sustained-release dosage forms of ruxolitinib
EA030705B1 (en) 2013-03-06 2018-09-28 Инсайт Холдингс Корпорейшн Processes and intermediates for making a jak inhibitor
PE20160126A1 (en) 2013-05-17 2016-02-24 Incyte Corp DERIVATIVES OF BIPIRAZOLE AS JAK INHIBITORS
ES2792549T3 (en) 2013-08-07 2020-11-11 Incyte Corp Sustained-release dosage forms for a JAK1 inhibitor
KR20220066179A (en) 2014-04-08 2022-05-23 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
AU2015253192B2 (en) 2014-04-30 2019-05-16 Incyte Holdings Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
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
KR101836822B1 (en) * 2016-10-17 2018-03-09 주식회사 엔지켐생명과학 Composition for preventing or treating psoriasis comprising monoacetyl-diacylglycerol compound
PL3746429T3 (en) 2018-01-30 2022-06-20 Incyte Corporation Processes for preparing (1-(3-fluoro-2-(trifluoromethyl)isonicotinyl)piperidine-4-one)
SG11202007805SA (en) 2018-02-16 2020-09-29 Incyte Corp 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
WO2019191684A1 (en) 2018-03-30 2019-10-03 Incyte Corporation Treatment of hidradenitis suppurativa using jak inhibitors
MX2020010815A (en) 2018-04-13 2020-12-11 Incyte Corp Biomarkers for graft-versus-host disease.
US11324749B2 (en) 2018-10-31 2022-05-10 Incyte Corporation Combination therapy for treatment of hematological diseases
CN113692278A (en) 2018-12-19 2021-11-23 因赛特公司 JAK1 pathway inhibitors for the treatment of gastrointestinal diseases
WO2021046350A1 (en) * 2019-09-05 2021-03-11 Incyte Corporation Ruxolitinib formulation for reduction of itch in atopic dermatitis
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
CR20230129A (en) 2020-08-18 2023-07-13 Incyte Corp Process and intermediates for preparing a jak inhibitor
EP4200278A1 (en) 2020-08-18 2023-06-28 Incyte Corporation Process and intermediates for preparing a jak1 inhibitor
KR20230118118A (en) 2020-12-08 2023-08-10 인사이트 코포레이션 JAK1 pathway inhibitors for the treatment of vitiligo
KR20230157307A (en) 2021-01-11 2023-11-16 인사이트 코포레이션 Combination therapy involving JAK pathway inhibitors and ROCK inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090233903A1 (en) * 2008-03-11 2009-09-17 Incyte Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2010039939A1 (en) * 2008-10-02 2010-04-08 Incyte Corporation Janus kinase inhibitors for treatment of dry eye and other eye related diseases
WO2011112662A1 (en) * 2010-03-10 2011-09-15 Incyte Corporation Piperidin-4-yl azetidine derivatives as jak1 inhibitors

Family Cites Families (303)

* 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
US3632836A (en) 1968-10-25 1972-01-04 Dow Chemical Co Solid curable polyepoxides modified with hydrolyzed liquid polyepoxides
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
US4404335A (en) 1982-08-16 1983-09-13 The Dow Chemical Company Hydrolyzing epoxy resins in absence of solvent and in presence of oxalic acid and a phosphonium compound
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
AU645504B2 (en) 1989-10-11 1994-01-20 Teijin Limited Bicyclic pyrimidine derivative, method of producing the same, and pharmaceutical preparation containing the same as active ingredient
US5403593A (en) 1991-03-04 1995-04-04 Sandoz Ltd. Melt granulated compositions for preparing sustained release dosage forms
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
FR2695126B1 (en) 1992-08-27 1994-11-10 Sanofi Elf Thienyl or pyrrolyl carboxylic acid derivatives, their preparation and medicaments containing them.
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
USH1439H (en) 1993-10-18 1995-05-02 The Dow Chemical Company Method to increase the level of α-glycol in liquid epoxy resin
EP0727217A3 (en) 1995-02-10 1997-01-15 Suntory Ltd Pharmaceutical composition containing god-type ellagitannin as active ingredient
US5856326A (en) 1995-03-29 1999-01-05 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
IL117580A0 (en) 1995-03-29 1996-07-23 Merck & Co Inc Inhibitors of farnesyl-protein transferase and pharmaceutical compositions containing them
CN1105113C (en) 1995-07-05 2003-04-09 纳幕尔杜邦公司 Fungicidal pyrimidinones
SI9620103A (en) 1995-07-06 1998-10-31 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
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
JP2000504023A (en) 1996-04-03 2000-04-04 メルク エンド カンパニー インコーポレーテッド Cancer treatment methods
AU2802297A (en) 1996-04-18 1997-11-07 Merck & Co., Inc. A method of treating cancer
US5795909A (en) 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
JP2000508335A (en) 1996-05-30 2000-07-04 メルク エンド カンパニー インコーポレーテッド How to treat 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
JP2001513502A (en) 1997-08-11 2001-09-04 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド 5,6-Heteroaryldipyrido [2,3-b: 3 ', 2'-f] azepines and their use in the prevention and treatment of HIV infection
US7153845B2 (en) 1998-08-25 2006-12-26 Columbia Laboratories, Inc. Bioadhesive progressive hydration tablets
US6075056A (en) 1997-10-03 2000-06-13 Penederm, Inc. Antifungal/steroid topical compositions
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
SK18542000A3 (en) 1998-06-04 2001-12-03 Abbott Laboratories Cell adhesion-inhibiting anti-inflammatory compounds
US6232320B1 (en) 1998-06-04 2001-05-15 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory compounds
ID26698A (en) 1998-06-19 2001-02-01 Pfizer Prod Inc PIROLO COMPOUNDS [2,3-d] PYRIMIDINE
PA8474101A1 (en) 1998-06-19 2000-09-29 Pfizer Prod Inc PYROLEUM [2,3-D] PIRIMIDINE COMPOUNDS
ATE459616T1 (en) 1998-08-11 2010-03-15 Novartis Ag ISOCHINOLINE DERIVATIVES WITH ANGIOGENESIS-INHIBITING EFFECT
JP2000119271A (en) 1998-08-12 2000-04-25 Hokuriku Seiyaku Co Ltd 1h-imidazopyridine derivative
CA2343148C (en) 1998-09-10 2005-11-15 Nycomed Danmark A/S Quick release pharmaceutical compositions of drug substances
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
US6133031A (en) 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
WO2000051614A1 (en) 1999-03-03 2000-09-08 Merck & Co., Inc. Inhibitors of prenyl-protein transferases
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
AU3565999A (en) 1999-04-16 2000-11-02 Coelacanth Chemical Corporation Synthesis of azetidine derivatives
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
CA2387535A1 (en) 1999-10-13 2001-04-19 Banyu Pharmaceutical Co., Ltd. Substituted imidazolidinone derivatives
US7235258B1 (en) 1999-10-19 2007-06-26 Nps Pharmaceuticals, Inc. Sustained-release formulations for treating CNS-mediated disorders
DK1235830T3 (en) 1999-12-10 2004-03-29 Pfizer Prod Inc Pyrrolo [2,3-d] pyrimidine compounds as protein kinase inhibitors
CN1615873A (en) 1999-12-24 2005-05-18 阿文蒂斯药物有限公司 Azaindoles compound
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
DK1142566T3 (en) 2000-04-07 2004-02-09 Medidom Lab Ophthalmological formulations based on cyclosporine, hyaluronic acid and polysorbate
WO2001081345A1 (en) 2000-04-20 2001-11-01 Mitsubishi Pharma Corporation Aromatic amide compounds
SI2223922T1 (en) 2000-04-25 2016-04-29 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
US7498304B2 (en) 2000-06-16 2009-03-03 Curis, Inc. Angiogenesis-modulating compositions and uses
JP4954426B2 (en) 2000-06-16 2012-06-13 キュリス,インコーポレイテッド Angiogenesis regulating composition and use
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
ATE465756T1 (en) 2000-06-23 2010-05-15 Mitsubishi Tanabe Pharma Corp ANTITUMOR EFFECT AMPLIFIER
EE200200711A (en) 2000-06-26 2004-06-15 Pfizer Products Inc. Pyrrolo [2,3-d] pyrimidine compounds as immunosuppressive agents
CN1321628C (en) 2000-06-28 2007-06-20 史密斯克莱·比奇曼公司 Wet milling process
AU2001278790A1 (en) 2000-08-22 2002-03-04 Hokuriku Seiyaku Co. Ltd 1h-imidazopyridine derivatives
MXPA03005001A (en) 2000-12-05 2003-09-05 Vertex Pharma 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
JP2004520347A (en) 2001-01-15 2004-07-08 グラクソ グループ リミテッド Arylpiperidine and piperazine derivatives as inducers of LDL-receptor expression
EP1363702A4 (en) 2001-01-30 2007-08-22 Cytopia Pty Ltd Methods of inhibiting kinases
JP4316893B2 (en) 2001-05-16 2009-08-19 バーテックス ファーマシューティカルズ インコーポレイテッド 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
US6852727B2 (en) 2001-08-01 2005-02-08 Merck & Co., Inc. Benzimisazo[4,5-f]isoquinolinone derivatives
DE60230890D1 (en) 2001-09-19 2009-03-05 Aventis Pharma Sa INDOLICINE AS KINASEPROTEINHEMMER
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
IL161156A0 (en) 2001-10-30 2004-08-31 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
JP2003155285A (en) 2001-11-19 2003-05-27 Toray Ind Inc Cyclic nitrogen-containing derivative
AU2002224131A1 (en) 2001-11-30 2003-06-17 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
WO2003088952A1 (en) 2002-04-15 2003-10-30 Adams Laboratories, Inc. Sustained release of guaifenesin combination drugs
TW200403058A (en) 2002-04-19 2004-03-01 Bristol Myers Squibb Co Heterocyclo inhibitors of potassium channel function
US7304061B2 (en) 2002-04-26 2007-12-04 Vertex Pharmaceuticals Incorporated Heterocyclic inhibitors of ERK2 and uses thereof
CA2483084A1 (en) 2002-05-02 2003-11-13 Merck & Co., Inc. Tyrosine kinase inhibitors
CA2484632C (en) 2002-05-07 2012-12-11 Control Delivery Systems, Inc. Processes for forming a drug delivery device
CA2486183C (en) 2002-05-23 2012-01-10 Cytopia Pty Ltd. Protein kinase inhibitors
PE20040522A1 (en) 2002-05-29 2004-09-28 Novartis Ag DIARYLUREA DERIVATIVES DEPENDENT ON PROTEIN KINASE
US7385018B2 (en) 2002-06-26 2008-06-10 Idemitsu Kosan Co., Ltd. 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
WO2004007472A1 (en) 2002-07-10 2004-01-22 Ono Pharmaceutical Co., Ltd. Ccr4 antagonist and medicinal use thereof
JP2006502183A (en) 2002-09-20 2006-01-19 アルコン,インコーポレイテッド 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
CA2506773A1 (en) 2002-11-04 2004-05-21 Vertex Pharmaceuticals Incorporated Heteroaryl-pyramidine derivatives as jak inhibitors
US8034831B2 (en) 2002-11-06 2011-10-11 Celgene Corporation Methods for the treatment and management of myeloproliferative diseases using 4-(amino)-2-(2,6-Dioxo(3-piperidyl)-isoindoline-1,3-dione in combination with other therapies
AR042052A1 (en) 2002-11-15 2005-06-08 Vertex Pharma USEFUL DIAMINOTRIAZOLS AS INHIBITORS OF PROTEINQUINASES
US20040099204A1 (en) 2002-11-25 2004-05-27 Nestor John J. Sheet, page, line, position marker
EP1572213A1 (en) 2002-11-26 2005-09-14 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
US7167750B2 (en) 2003-02-03 2007-01-23 Enteromedics, Inc. Obesity treatment with electrically induced vagal down regulation
US7407962B2 (en) 2003-02-07 2008-08-05 Vertex Pharmaceuticals Incorporated Heteroaryl compounds useful as inhibitors or protein kinases
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
US7547794B2 (en) 2003-04-03 2009-06-16 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
SE0301372D0 (en) 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
SE0301373D0 (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
WO2005020921A2 (en) 2003-08-29 2005-03-10 Exelixis, Inc. C-kit modulators and methods of use
EP1678147B1 (en) 2003-09-15 2012-08-08 Lead Discovery Center GmbH 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
EP1679074B1 (en) 2003-10-24 2010-12-08 Santen Pharmaceutical Co., Ltd. Therapeutic agent for keratoconjunctive disorder
US7387793B2 (en) 2003-11-14 2008-06-17 Eurand, Inc. Modified release dosage forms of skeletal muscle relaxants
MY141220A (en) 2003-11-17 2010-03-31 Astrazeneca Ab Pyrazole derivatives as inhibitors of receptor tyrosine kinases
BRPI0416909A (en) 2003-11-25 2007-01-16 Pfizer Prod Inc atherosclerosis treatment method
CA2549485A1 (en) 2003-12-17 2005-07-07 Pfizer Products Inc. Pyrrolo [2,3-d] pyrimidine compounds for treating transplant rejection
CA2550189A1 (en) 2003-12-19 2005-07-21 Schering Corporation Thiadiazoles as cxc- and cc- chemokine receptor ligands
EP1696920B8 (en) 2003-12-19 2015-05-06 Plexxikon Inc. Compounds and methods for development of ret modulators
CA2548374C (en) 2003-12-23 2014-05-27 Astex Therapeutics Limited Pyrazole derivatives as protein kinase modulators
US20050165029A1 (en) 2004-01-13 2005-07-28 Ambit Biosciences Corporation Pyrrolopyrimidine derivatives and analogs and their use in the treatment and prevention of diseases
US20050277629A1 (en) 2004-03-18 2005-12-15 The Brigham And Women's Hospital, Inc. Methods for the treatment of synucleinopathies (Lansbury)
PL2332940T3 (en) 2004-03-30 2013-03-29 Vertex Pharma 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
US20060106020A1 (en) 2004-04-28 2006-05-18 Rodgers James D 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
WO2005110410A2 (en) 2004-05-14 2005-11-24 Abbott Laboratories 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
TW200610762A (en) 2004-06-10 2006-04-01 Irm Llc Compounds and compositions as protein kinase inhibitors
JP5315611B2 (en) 2004-06-23 2013-10-16 小野薬品工業株式会社 Compound having S1P receptor binding ability and use thereof
EP1765819B1 (en) 2004-06-30 2014-03-12 Vertex Pharmaceuticals Inc. 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
CA2582985A1 (en) 2004-10-13 2006-04-20 Jin-Jun Liu 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
US7528138B2 (en) 2004-11-04 2009-05-05 Vertex Pharmaceuticals Incorporated Pyrazolo[1,5-a]pyrimidines useful as inhibitors of protein kinases
AU2005309019A1 (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
US20060128803A1 (en) 2004-12-14 2006-06-15 Alcon, Inc. Method of treating dry eye disorders using 13(S)-HODE and its analogs
WO2006067445A2 (en) 2004-12-22 2006-06-29 Astrazeneca Ab Csf-1r kinase inhibitors
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.
US20090124635A1 (en) 2005-01-20 2009-05-14 Pfizer Inc. Chemical compounds
KR20070104641A (en) 2005-02-03 2007-10-26 버텍스 파마슈티칼스 인코포레이티드 Pyrrolopyrimidines useful as inhibitors of protein kinase
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
BRPI0608513A2 (en) 2005-03-15 2010-01-05 Irm Llc compounds and compositions as protein kinase inhibitors
BRPI0610514A2 (en) 2005-04-05 2016-11-16 Pharmacopeia Inc compound, pharmaceutical composition, and method of treating a disorder
GB0510139D0 (en) 2005-05-18 2005-06-22 Addex Pharmaceuticals Sa Novel compounds B1
MX2007014619A (en) 2005-05-20 2009-02-13 Vertex Pharma Pyrrolopyridines useful as inhibitors of protein kinase.
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
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
CN102603581B (en) 2005-06-22 2015-06-24 普莱希科公司 Pyrrolo[2,3-b] pyridine derivatives as protein kinase inhibitors
CN102127078A (en) 2005-07-14 2011-07-20 安斯泰来制药株式会社 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
US20070049591A1 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Inhibitors of MAPK/Erk Kinase
EP1926735A1 (en) 2005-09-22 2008-06-04 Incyte Corporation Tetracyclic inhibitors of janus kinases
AU2006297351A1 (en) 2005-09-30 2007-04-12 Vertex Pharmaceuticals Incorporated Deazapurines useful as inhibitors of janus kinases
WO2007044894A2 (en) 2005-10-11 2007-04-19 Chembridge Research Laboratories, Inc. Cell-free protein expression systems and methods of use thereof
EP1937664B1 (en) 2005-10-14 2011-06-15 Sumitomo Chemical Company, Limited Hydrazide compound and pesticidal use of the same
BRPI0618011A2 (en) 2005-10-28 2011-08-16 Astrazeneca Ab compound or a pharmaceutically acceptable salt thereof, process for the preparation thereof, pharmaceutical composition, use of a compound or a pharmaceutically acceptable salt thereof, and methods for producing an antiproliferative effect and a pro-apoptotic effect in an animal warm-blooded to treat disease and to produce a jak inhibiting effect on a warm-blooded animal
DK1951684T3 (en) 2005-11-01 2016-10-24 Targegen Inc BIARYLMETAPYRIMIDIN kinase inhibitors
WO2007062459A1 (en) 2005-11-29 2007-06-07 Cytopia Research Pty Ltd Selective kinase inhibitors based on pyridine scaffold
MY162590A (en) 2005-12-13 2017-06-30 Incyte Holdings Corp 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
JP2009521504A (en) 2005-12-22 2009-06-04 スミスクライン・ビーチャム・コーポレイション Akt activity inhibitor
KR20080083680A (en) 2005-12-23 2008-09-18 스미스클라인 비참 코포레이션 Azaindole inhibitors of aurora kinases
JP4643455B2 (en) 2006-01-12 2011-03-02 株式会社ユニバーサルエンターテインメント Game system
BRPI0706537A2 (en) 2006-01-17 2011-03-29 Vertex Pharma azaindoles useful as janus kinases inhibitors
EP1979353A2 (en) 2006-01-19 2008-10-15 OSI Pharmaceuticals, Inc. Fused heterobicyclic kinase inhibitors
JP2009525350A (en) 2006-02-01 2009-07-09 スミスクライン ビーチャム コーポレーション 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
PL1842534T3 (en) 2006-02-24 2012-05-31 Teva Pharma Metoprolol succinate extended release tablets and methods for their preparation
EP1995246A4 (en) 2006-03-10 2010-11-17 Ono Pharmaceutical Co Nitrogenated heterocyclic derivative, and pharmaceutical agent comprising the derivative as active ingredient
WO2007116866A1 (en) 2006-04-03 2007-10-18 Astellas Pharma Inc. Hetero compound
MX2008012860A (en) 2006-04-05 2009-01-07 Vertex Pharma Deazapurines useful as inhibitors of janus kinases.
WO2007116313A2 (en) 2006-04-12 2007-10-18 Pfizer Limited Pyrrolidine derivatives as modulators of chemokine ccr5 receptors
WO2007129195A2 (en) 2006-05-04 2007-11-15 Pfizer Products Inc. 4-pyrimidine-5-amino-pyrazole compounds
WO2007135461A2 (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
TWI398252B (en) 2006-05-26 2013-06-11 Novartis Ag Pyrrolopyrimidine compounds and their uses
NZ573174A (en) 2006-06-01 2012-01-12 Msd Consumer Care Inc Sustained release pharmaceutical dosage form containing phenylephrine
US20080021217A1 (en) 2006-07-20 2008-01-24 Allen Borchardt Heterocyclic inhibitors of rho kinase
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
CA2660560A1 (en) 2006-08-16 2008-02-21 Boehringer Ingelheim International Gmbh Pyrazine compounds, their use and methods of preparation
WO2008028937A1 (en) 2006-09-08 2008-03-13 Novartis Ag N-biaryl (hetero) arylsulphonamide derivatives useful in the treatment of diseases mediated by lymphocytes interactions
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
AR063142A1 (en) 2006-10-04 2008-12-30 Pharmacopeia Inc DERIVATIVES OF 2- (BENCIMIDAZOLIL) PURINE AND PURINONES 6-USEFUL SUBSTITUTES AS IMMUNOSUPPRESSORS, AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
AR063141A1 (en) 2006-10-04 2008-12-30 Pharmacopeia Inc DERIVATIVES OF 2- (BENZIMIDAZOLIL) PURINA 8- REPLACED 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
NZ576234A (en) 2006-11-06 2011-06-30 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
JP5572388B2 (en) 2006-11-22 2014-08-13 インサイト・コーポレイション Imidazotriazines and imidazopyrimidines as kinase inhibitors
WO2008067119A2 (en) 2006-11-27 2008-06-05 Smithkline Beecham Corporation Novel compounds
SG177221A1 (en) 2006-12-15 2012-01-30 Abbott Lab Novel oxadiazole compounds
CA2672903C (en) 2006-12-20 2012-10-23 Amgen Inc. Heterocyclic compounds and their use in treating inflammation, angiogenesis and cancer
AU2007338792B2 (en) 2006-12-20 2012-05-31 Amgen Inc. Substituted heterocycles and methods of use
CA2667072C (en) 2006-12-22 2015-11-24 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Gel useful for the delivery of ophthalmic drugs
US8513270B2 (en) 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
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
BRPI0809998B8 (en) 2007-04-03 2021-05-25 Array Biopharma Inc imidazo[1,2-a]pyridine compound as receptor tyrosine kinase inhibitors, their uses, their preparation processes and pharmaceutical compositions
US8188178B2 (en) 2007-05-07 2012-05-29 3M Innovative Properties Company Cold shrinkable article including an epichlorohydrin composition
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
EP3495369B1 (en) 2007-06-13 2021-10-27 Incyte Holdings Corporation Use of 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.
KR20120115413A (en) 2007-07-11 2012-10-17 화이자 인코포레이티드 Pharmaceutical compositions and methods of 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
CA2704599C (en) 2007-11-16 2015-05-12 Incyte Corporation 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
CA2711384C (en) 2008-01-18 2016-07-26 Institute Of Organic Chemistry And Biochemistry As Cr, V.V.I. Cytostatic 7-deazapurine nucleosides
EA019309B1 (en) 2008-02-04 2014-02-28 Меркьюри Терапьютикс, Инк. Ampk (amp-activated protein kinase) modulators
UY31679A1 (en) 2008-03-03 2009-09-30 PIM KINASE INHIBITORS AND METHODS FOR USE
JP5384611B2 (en) 2008-03-21 2014-01-08 ノバルティス アーゲー Novel heterocyclic compounds and their use
US8344144B2 (en) 2008-06-18 2013-01-01 Merck Sharp & Dohme Corp. Inhibitors of Janus kinases
BRPI0914630A2 (en) 2008-06-26 2019-09-24 Anterios Inc dermal release
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
WO2010022081A1 (en) 2008-08-19 2010-02-25 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
JP4884570B2 (en) 2008-08-20 2012-02-29 ファイザー・インク Pyrrolo [2,3-d] pyrimidine compound
BRPI0918846A2 (en) 2008-09-02 2019-09-24 Novartis Ag heterocyclic kinase inhibitors
EP2342190A1 (en) 2008-09-02 2011-07-13 Novartis AG Bicyclic kinase inhibitors
PT2344474E (en) 2008-09-02 2015-12-28 Novartis Ag Picolinamide derivatives as kinase inhibitors
CA2739466A1 (en) 2008-10-17 2010-04-22 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
MX2011012262A (en) 2009-05-22 2012-01-25 Incyte Corp 3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl]octane- or heptane-nitrile as jak inhibitors.
EA025520B1 (en) 2009-05-22 2017-01-30 Инсайт Холдингс Корпорейшн 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
UA110324C2 (en) * 2009-07-02 2015-12-25 Genentech Inc Jak inhibitory compounds based on pyrazolo pyrimidine
CA2767079A1 (en) 2009-07-08 2011-01-13 Leo Pharma A/S Heterocyclic compounds as jak receptor and protein tyrosine kinase inhibitors
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
TW201113285A (en) 2009-09-01 2011-04-16 Incyte Corp Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
JP5567136B2 (en) 2009-09-08 2014-08-06 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト 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
PT2486041E (en) 2009-10-09 2013-11-14 Incyte Corp Hydroxyl, keto, and glucuronide derivatives of 3-(4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl)-3-cyclopentylpropanenitrile
MX2012004020A (en) * 2009-10-20 2012-05-08 Cellzome Ltd Heterocyclyl pyrazolopyrimidine analogues as jak inhibitors.
US8671402B2 (en) * 2009-11-09 2014-03-11 Bank Of America Corporation Network-enhanced control of software updates received via removable computer-readable medium
EP2332917B1 (en) 2009-11-11 2012-08-01 Sygnis Bioscience GmbH & Co. KG Compounds for PIM kinase inhibition and for treating malignancy
CN102740888B (en) 2009-11-24 2016-10-12 奥尔德生物制药公司 IL-6 antibody and application thereof
EP2506852A4 (en) 2009-12-04 2013-06-19 Univ Texas Interferon therapies in combination with blockade of stat3 activation
JP5739446B2 (en) * 2009-12-18 2015-06-24 ファイザー・インク Pyrrolo [2,3-d] pyrimidine compound
CN102712640A (en) 2010-01-12 2012-10-03 弗·哈夫曼-拉罗切有限公司 Tricyclic heterocyclic compounds, compositions and methods of use thereof
SA111320200B1 (en) * 2010-02-17 2014-02-16 ديبيوفارم اس ايه Bicyclic Compounds and their Uses as Dual C-SRC / JAK Inhibitors
EA023444B1 (en) 2010-02-18 2016-06-30 Инсайт Холдингс Корпорейшн Cyclobutane and methylcyclobutane derivatives, composition based thereon and methods of use thereof
KR20130094710A (en) 2010-04-14 2013-08-26 어레이 바이오파마 인크. 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
SG10201910912TA (en) 2010-05-21 2020-01-30 Incyte Corp Topical Formulation for a JAK Inhibitor
US8637529B2 (en) 2010-06-11 2014-01-28 AbbYie Inc. Pyrazolo[3,4-d]pyrimidine compounds
US9351943B2 (en) 2010-07-01 2016-05-31 Matthew T. McLeay Anti-fibroblastic fluorochemical emulsion therapies
US20130237493A1 (en) 2010-09-30 2013-09-12 Portola Pharmaceuticals, Inc. Combination therapy of 4-(cyclopropylamino)-2-(4-(4-(ethylsulfonyl)piperazin-1-yl)phenylamino)pyrimidine-5-carboxamide and fludarabine
BR112013012502A2 (en) 2010-11-19 2019-03-06 Incyte Corporation substituted cyclobutyl pyrrolopyridine and derivative pyrrolopyrimidine derivatives as jak inhibitors
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
CN103370068A (en) 2010-12-03 2013-10-23 Ym生物科学澳大利亚私人有限公司 Treatment of JAK2-mediated conditions
WO2012112847A1 (en) 2011-02-18 2012-08-23 Novartis Pharma Ag mTOR/JAK INHIBITOR COMBINATION THERAPY
CN102247368B (en) 2011-05-19 2013-05-29 安徽永生堂药业有限责任公司 Compound acrivastine sustained release tablets, and preparation method thereof
CN102218042A (en) 2011-05-26 2011-10-19 青岛黄海制药有限责任公司 Sustained release tablet of quetiapine fumarate composition and preparation method of sustained release tablet
MY165963A (en) 2011-06-20 2018-05-18 Incyte Holdings Corp Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
WO2013007768A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors
WO2013007765A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Fused tricyclic compounds for use as inhibitors of janus kinases
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
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings 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
EP3450434B1 (en) 2012-06-15 2021-02-24 CoNCERT Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib
EA201590272A1 (en) 2012-07-27 2015-05-29 Рациофарм Гмбх ORAL DOSED FORMS FOR MODIFIED SURVENT CONTAINING RUXOLITINIB
CN102772384A (en) 2012-08-07 2012-11-14 四川百利药业有限责任公司 Minocycline hydrochloride sustained release tablet and preparation method thereof
JP2015526520A (en) * 2012-08-31 2015-09-10 プリンシピア バイオファーマ インコーポレイテッド Benzimidazole derivatives as ITK inhibitors
SG10201703533VA (en) 2012-11-01 2017-06-29 Incyte Corp Tricyclic fused thiophene derivatives as jak inhibitors
SG11201503695XA (en) * 2012-11-15 2015-06-29 Incyte Corp Sustained-release dosage forms of ruxolitinib
EA030705B1 (en) 2013-03-06 2018-09-28 Инсайт Холдингс Корпорейшн Processes and intermediates for making a jak inhibitor
PE20160126A1 (en) 2013-05-17 2016-02-24 Incyte Corp DERIVATIVES OF BIPIRAZOLE AS JAK INHIBITORS
ES2792549T3 (en) 2013-08-07 2020-11-11 Incyte Corp Sustained-release dosage forms for a JAK1 inhibitor
CN105555313A (en) * 2013-08-20 2016-05-04 因赛特公司 Survival benefit in patients with solid tumors with elevated c-reactive protein levels
CN106456773A (en) 2014-02-28 2017-02-22 因赛特公司 Jak1 inhibitors for the treatment of myelodysplastic syndromes
KR20220066179A (en) 2014-04-08 2022-05-23 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
AU2015253192B2 (en) 2014-04-30 2019-05-16 Incyte Holdings Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
EP4233870A3 (en) 2014-05-28 2024-01-24 Onco Tracker, Inc. Anti-cancer 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
US10766900B2 (en) 2017-12-29 2020-09-08 Formosa Laboratories, Inc. Baricitinib intermediate, method for forming Baricitinib intermediate, and method for preparing Baricitinib or pharmaceutically acceptable salt thereof
WO2020163653A1 (en) 2019-02-06 2020-08-13 Concert Pharmaceuticals, Inc. Process for preparing enantiomerically enriched jak inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090233903A1 (en) * 2008-03-11 2009-09-17 Incyte Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2010039939A1 (en) * 2008-10-02 2010-04-08 Incyte Corporation Janus kinase inhibitors for treatment of dry eye and other eye related diseases
WO2011112662A1 (en) * 2010-03-10 2011-09-15 Incyte Corporation Piperidin-4-yl azetidine derivatives as jak1 inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MANISH J CHAUHAN ET AL: "A concise review on sustained drug delivery system and its opportunities", vol. 2, no. 2, 1 March 2012 (2012-03-01), pages 227 - 238, XP002718280, ISSN: 0974-4304, Retrieved from the Internet <URL:http://www.ajptr.com/archive/volume-2/april-2012-issue-2/article-137.html> [retrieved on 20140102] *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
WO2017025814A1 (en) 2014-08-07 2017-02-16 Acerta Pharma B.V. Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on btk occupancy and btk resynthesis rate
WO2016020901A1 (en) 2014-08-07 2016-02-11 Acerta Pharma B.V. Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on btk occupancy and btk resynthesis rate
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
WO2016196244A1 (en) 2015-05-29 2016-12-08 Incyte Corporation Pyridineamine compounds useful as pim kinase inhibitors
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
WO2017044730A1 (en) 2015-09-09 2017-03-16 Incyte Corporation Salts of a pim kinase inhibitor
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
WO2017059251A1 (en) 2015-10-02 2017-04-06 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11406640B2 (en) 2019-03-05 2022-08-09 Incyte Corporation JAK1 pathway inhibitors for the treatment of chronic lung allograft dysfunction
WO2020181034A1 (en) * 2019-03-05 2020-09-10 Incyte Corporation Jak1 pathway inhibitors for the treatment of chronic lung allograft dysfunction
US11896595B2 (en) 2019-03-05 2024-02-13 Incyte Corporation JAK1 pathway inhibitors for the treatment of chronic lung allograft dysfunction

Also Published As

Publication number Publication date
PT3030227T (en) 2020-06-25
KR20220103810A (en) 2022-07-22
KR102419714B1 (en) 2022-07-13
US11045421B2 (en) 2021-06-29
PH12016500243A1 (en) 2016-05-16
TWI789516B (en) 2023-01-11
CR20160102A (en) 2016-07-28
TWI730935B (en) 2021-06-21
CA2920108A1 (en) 2015-02-12
PE20160223A1 (en) 2016-04-27
MY195091A (en) 2023-01-10
SG10201912203XA (en) 2020-02-27
TW201529099A (en) 2015-08-01
KR20210076186A (en) 2021-06-23
SI3030227T1 (en) 2020-08-31
JP2022136270A (en) 2022-09-15
US9655854B2 (en) 2017-05-23
UA120499C2 (en) 2019-12-26
BR112016002571A8 (en) 2018-01-23
HRP20200955T1 (en) 2020-10-02
BR122020001831A2 (en) 2017-08-01
US10561616B2 (en) 2020-02-18
SG11201600815WA (en) 2016-03-30
US20150065484A1 (en) 2015-03-05
JP2018131452A (en) 2018-08-23
TW202320789A (en) 2023-06-01
MX2019013381A (en) 2020-02-20
AU2014305989B2 (en) 2019-11-28
EP3030227A1 (en) 2016-06-15
RS60469B1 (en) 2020-07-31
CN105579032A (en) 2016-05-11
IL277554B2 (en) 2024-03-01
PH12016500243B1 (en) 2016-05-16
MX2016001639A (en) 2016-10-07
JP6334700B2 (en) 2018-05-30
CN114010611B (en) 2023-11-28
PL3030227T3 (en) 2020-08-24
DK3030227T3 (en) 2020-04-20
TWI822248B (en) 2023-11-11
JP2016527315A (en) 2016-09-08
CR20230316A (en) 2023-11-10
US20170319487A1 (en) 2017-11-09
JP6880266B2 (en) 2021-06-02
BR112016002571A2 (en) 2017-08-01
CY1123314T1 (en) 2021-12-31
IL277554A (en) 2020-11-30
AU2021202916A1 (en) 2021-06-03
CA2920108C (en) 2022-07-05
CA3155500A1 (en) 2015-02-12
IL243920A0 (en) 2016-04-21
CN114010611A (en) 2022-02-08
PH12019502072A1 (en) 2020-07-13
SG10201801069QA (en) 2018-03-28
JP2020079317A (en) 2020-05-28
CL2016000292A1 (en) 2016-08-05
KR20160045081A (en) 2016-04-26
AU2019257368A1 (en) 2019-11-14
NZ717230A (en) 2021-05-28
CR20190343A (en) 2019-10-02
TW201943409A (en) 2019-11-16
PE20220579A1 (en) 2022-04-20
LT3030227T (en) 2020-06-10
ES2792549T3 (en) 2020-11-11
JP7126643B2 (en) 2022-08-29
JP2021107454A (en) 2021-07-29
AU2019257368B2 (en) 2021-02-25
IL243920B (en) 2021-08-31
US20220016036A1 (en) 2022-01-20
IL277554B1 (en) 2023-11-01
CN116036089A (en) 2023-05-02
HUE049345T2 (en) 2020-09-28
PH12019502074A1 (en) 2020-02-24
EA201690357A1 (en) 2016-07-29
US20200253879A1 (en) 2020-08-13
AU2014305989A1 (en) 2016-03-10
BR122020001831A8 (en) 2023-01-24
EP3030227B1 (en) 2020-04-08
EP3721873A1 (en) 2020-10-14

Similar Documents

Publication Publication Date Title
US11045421B2 (en) Sustained release dosage forms for a JAK1 inhibitor
NZ756083A (en) Sustained release dosage forms for a jak1 inhibitor
NZ717230B2 (en) Sustained release dosage forms for a jak1 inhibitor
NZ756083B2 (en) Sustained release dosage forms for a jak1 inhibitor
NZ756084B2 (en) Sustained release dosage forms for a jak1 inhibitor
BR112016002571B1 (en) SUSTAINED RELEASE TABLETS WITH A JAK1 INHIBITOR
EA043723B1 (en) JAK1 INHIBITOR TIME-RELEASE TABLET

Legal Events

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

Ref document number: 201480052299.3

Country of ref document: CN

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

Ref document number: 14753182

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2920108

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 243920

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 122020001831

Country of ref document: BR

Ref document number: 12016500243

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2016533398

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 000226-2016

Country of ref document: PE

Ref document number: MX/A/2016/001639

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016002571

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 16051256

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2014753182

Country of ref document: EP

Ref document number: CR2016-000102

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 201690357

Country of ref document: EA

Ref document number: A201602100

Country of ref document: UA

Ref document number: IDP00201601463

Country of ref document: ID

ENP Entry into the national phase

Ref document number: 20167006096

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2014305989

Country of ref document: AU

Date of ref document: 20140806

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016002571

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160204