US20210244735A1 - Compositions and methods for treating myelofibrosis - Google Patents

Compositions and methods for treating myelofibrosis Download PDF

Info

Publication number
US20210244735A1
US20210244735A1 US17/220,073 US202117220073A US2021244735A1 US 20210244735 A1 US20210244735 A1 US 20210244735A1 US 202117220073 A US202117220073 A US 202117220073A US 2021244735 A1 US2021244735 A1 US 2021244735A1
Authority
US
United States
Prior art keywords
compound
amino
formulation
subject
capsule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/220,073
Inventor
Arvind JAYAN
Janice Cacace
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Impact Biomedicines Inc
Original Assignee
Impact Biomedicines Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46024747&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20210244735(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Impact Biomedicines Inc filed Critical Impact Biomedicines Inc
Priority to US17/220,073 priority Critical patent/US20210244735A1/en
Publication of US20210244735A1 publication Critical patent/US20210244735A1/en
Assigned to IMPACT BIOMEDICINES, INC. reassignment IMPACT BIOMEDICINES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TARGEGEN, INC.
Assigned to TARGEGEN, INC. reassignment TARGEGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JAYAN, ARVIND, CACACE, JANICE
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P35/00Antineoplastic agents
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy

Definitions

  • compositions and methods for treating myelofibrosis relate to treatment of myelofibrosis with compounds that inhibit JAK2 or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • MF Myelofibrosis
  • MPN BCR-ABL1-negative myeloproliferative neoplasm
  • PV polycythemia vera
  • ET essential thrombocythemia
  • Clinical features include progressive anemia, marked splenomegaly, constitutional symptoms (e.g. fatigue, night sweats, bone pain, pruritus, and cough) and weight loss (Tefferi A, N Engl J Med 342:1255-1265, 2000).
  • Some mutations occur at high frequency in MF (e.g. JAK2 mutations in ⁇ 50% patients), and either directly (e.g. JAK2 or MPL mutations) or indirectly (e.g. LNK or CBL mutations) induce JAK-STAT hyperactivation.
  • the currently available treatments are not effective in reversing the process of MF, be it primary or secondary disease.
  • the only potential for cure of the disease to date is bone marrow transplantation.
  • most patients are not suitable bone marrow transplant candidates because of the older median age at diagnosis, in which transplant-related morbidity and mortality tends to be high.
  • management options of MF are currently inadequate to meet the needs of all patients.
  • the main options for active intervention include cyto-reductive therapy, e.g. with hydroxyurea, treatment of anemia with androgens, erythropoietin and splenectomy.
  • the capsule comprises an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate, wherein the admixture is contained in the capsule.
  • the capsule contains about 10 mg to about 680 mg of the compound, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 10 mg to about 500 mg of the compound. In some embodiments, the capsule contains about any of 10 mg, 40 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg of the compound. In some embodiments, the weight ratio of the compound to microcrystalline cellulose in the capsule is between about 1:1.5 to 1:15, wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound.
  • the weight ratio of the compound to sodium stearyl fumarate in the capsule is between about 5:1 to about 50:1, and wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound.
  • the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • unit dosage forms comprising an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate.
  • the unit dosage forms are for treatment of myelofibrosis such as treatment of myelofibrosis according to a method described herein.
  • the unit dosage form comprises an admixture of (i) about 10 mg to about 680 mg (or about 10 mg to about 500 mg) of a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the specified weight is the free base moiety weight of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate.
  • the unit dosage form is in the form of a capsule, and the admixture is contained in the capsule.
  • the compound in the admixture is about 10 mg to about 500 mg, wherein the specified weight is the free base moiety weight of the compound.
  • the admixture comprises (i) about 10 mg (or about any of 40 mg, 100 mg, 200 mg, 300 mg, 400 mg, or 500 mg) of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate, wherein the specified weight is the free base moiety weight of the compound.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the weight ratio of the compound to microcrystalline cellulose in the capsule is between about 1:1.5 to 1:15, wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound.
  • the weight ratio of the compound to sodium stearyl fumarate in the capsule is between about 5:1 to about 50:1, and wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound.
  • the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • sodium stearyl fumarate is about 1% w/w of capsule fill weight.
  • the weight ratio of the compound to microcrystalline cellulose such as silicified microcrystalline cellulose is about 40:60 to about 10:90 (e.g., about 40:60 or about 1:1.5, or about 10:90 or about 1:9).
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the unit dosage form or capsule contains an admixture of about 12 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 122 mg of silicified microcrystalline cellulose, and about 1 mg of sodium stearyl fumarate.
  • the unit dosage form or capsule contains an admixture of about 47 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 448 mg of silicified microcrystalline cellulose, and about 5 mg of sodium stearyl fumarate.
  • the unit dosage form or capsule contains an admixture of about 117 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the unit dosage form or capsule contains an admixture of about 235 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 357 mg of silicified microcrystalline cellulose, and about 6.00 mg of sodium stearyl fumarate.
  • the capsule is a hard gelatin capsule.
  • Also provided herein are methods of preparing a capsule drug product comprising a) blending a lubricant with a compound that is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to generate granules and b) mixing the granules of a) with an excipient.
  • the lubricant is sodium stearyl fumarate.
  • the excipient is microcrystalline cellulose such as silicified microcrystalline cellulose.
  • sodium stearyl fumarate is about 1% w/w of capsule fill weight.
  • the weight ratio of the compound to silicified microcrystalline cellulose is about 1:1.5 to about 1:9. In some embodiments, the weight ratio of the compound to silicified microcrystalline cellulose is about 1:1.5. In some embodiments, the weight ratio of the compound to silicified microcrystalline cellulose is about 1:9.
  • the capsule is a hard gelatin capsule.
  • a method of treating myelofibrosis in a subject comprising orally administering a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and wherein the compound is in a capsule containing an admixture of (i) the compound, (ii) a microcrystalline cellulose (e.g., silicified microcrystalline cellulose), and (iii) sodium stearyl fumarate.
  • a microcrystalline cellulose e.g., silicified microcrystalline cellulose
  • a compound which which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate
  • the previous therapy is a treatment with a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the previous therapy comprises administration of INCB018424 (ruxolitinib).
  • the subject is unresponsive to the previous therapy.
  • the previous therapy is a treatment with N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the previous therapy has been discontinued upon indication of elevated levels of amylase, lipase, aspartate aminotransferase (“AST”), alanine aminotransferase (“ALT”), and/or creatinine.
  • the previous therapy has been discontinued upon indication of a hematologic condition selected from the group consisting of anemia, thrombocytopenia, and neutropenia.
  • Also provided herein are methods of monitoring treatment of myelofibrosis in a subject comprising (a) administering to a subject an effective amount of a compound which is N-tert-butyl-3-[(5 -methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof; (b) monitoring a non-hematologic parameter selected from the group consisting of amylase level, lipase level, aspartate aminotransferase (AST) level, alanine aminotransferase (ALT) level, and creatinine level in the subject; and (c) determining if the subject should continue or discontinue with the treatment.
  • the one or more of the elevated levels are Grade 4 events.
  • Also provided herein are methods of monitoring a treatment of myelofibrosis to a subject comprising (a) administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof; (b) monitoring a hematologic parameter selected from the group consisting of anemia, thrombocytopenia, and neutropenia in the serum of the subject; and (c) determining if the subject should continue or discontinue with the treatment.
  • the one or more hematologic conditions are grade 4 events.
  • the methods further comprise administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof after the subject has been discontinued with the treatment for at least 2 weeks.
  • the subject has been discontinued with the treatment for at least 3 weeks.
  • the subject has been discontinued with the treatment for at least 4 weeks.
  • the treatment has been discontinued without prior dose reduction.
  • the compound is administered to the human subject at a dose of about 240 mg per day to about 680 mg per day, and wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is administered at a dose of about 300 mg per day to about 500 mg per day (e.g., about 300 mg per day to about 400 mg per day, or about 400 mg per day to about 500 mg per day), and wherein the specified weight is the free base moiety weight of the compound.
  • the compound is administered at a dose of about any of 240 mg per day, 250 mg per day, 300 mg per day, 350 mg per day, 400 mg per day, 450 mg per day, 500 mg per day, 550 mg per day, 600 mg per day, 650 mg per day, or 680 mg per day, and wherein the specified weight is the free base moiety weight of the compound.
  • the compound is administered daily and/or orally.
  • the compound is administered over a period of at least 1 cycle, at least 2 cycles, at least 3 cycles, at least 4 cycles, at least 5 cycles, or at least 6 cycles (e.g., at least 7 cycles, at least 8 cycles, at least 9 cycles, at least 10 cycles, at least 11 cycles, at least 12 cycles, at least 15 cycles, at least 18 cycles, or at least 24 cycles) of a 28-day treatment cycle.
  • the compound is in a capsule and administered orally.
  • the compound is in a unit dosage form. Any of the capsules or unit dosage forms described herein may be administered.
  • the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate.
  • the weight ratio of the compound to microcrystalline cellulose in the admixture is between about 1:1.5 to 1:15, and wherein the weight for the compound is the free base moiety weight of the compound.
  • the weight ratio of the compound to sodium stearyl fumarate in the admixture is between about 5:1 to about 50:1, and wherein the weight for the compound is the free base moiety weight of the compound.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the microcrystalline cellulose is silicified microcrystalline cellulose.
  • the subject is a human.
  • the subject has primary myelofibrosis. In some embodiments of the compositions and methods provided herein, the subject has post polycythemia vera myelofibrosis. In some embodiments of the compositions and methods provided herein, the subject has post essential thrombocythemia myelofibrosis. In some embodiments, the subject has high risk myelofibrosis. In some embodiments, the subject has intermediate risk myelofibrosis (such as intermediate risk level 2).
  • the subject is positive for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or positive for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • the subject has palpable splenomegaly.
  • the subject with myelofibrosis has spleen of at least 5cm below costal margin as measured by palpation.
  • the subject is transfusion dependent. In some embodiments of the compositions and methods provided herein, the subject is not transfusion dependent.
  • the Cmax of the compound upon administration of the compound to a human subject, is achieved within about 2 to about 4 hours post-dose. In some embodiments, upon administration of the compound to a human subject, the elimination half life of the compound is about 16 to about 34 hours. In some embodiments, the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the accumulation of the compound is about 1.25 to about 4.0 fold at steady state when the compound is dosed once daily.
  • the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) microcrystalline cellulose, and (iii) sodium stearyl fumarate.
  • the weight ratio of the compound to microcrystalline cellulose in the admixture is between about 1:1.5 to 1:15, and wherein the weight for the compound is the free base moiety weight of the compound.
  • the weight ratio of the compound to sodium stearyl fumarate in the admixture is between about 5:1 to about 50:1, and wherein the weight for the compound is the free base moiety weight of the compound.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the microcrystalline cellulose is silicified microcrystalline cellulose.
  • articles of manufacture or kits comprising (a) any one of the capsules provided herein, and (b) a package insert or a label indicating that the capsule is useful for treating myelofibrosis in a subject. Also provided herein are articles of manufacture or kits comprising (a) any one of the unit dosage forms provided herein, and (b) a package insert or a label indicating that the capsule is useful for treating myelofibrosis in a subject.
  • an article of manufacture or kit comprising (a) an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) microcrystalline cellulose, and (iii) sodium stearyl fumarate, and (b) a package insert or a label indicating that the admixture is useful for treating myelofibrosis in a subject.
  • articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • JAK2 Janus Kinase 2
  • articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, wherein the subject has previously received another myelofibrosis therapy.
  • the previous therapy is a treatment with a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for ameliorating bone marrow cellularity and/or bone marrow fibrosis.
  • articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for improving pruritus associated with myelofibrosis.
  • articles of manufacture or kits comprising a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, and that subject should discontinue the treatment upon indication of elevated levels of one or more enzymes or molecules selected from the group consisting of: amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatinine in the serum of the subject, and/or upon indication of one or more hematologic condition selected from the group consisting of anemia, thrombocytopenia, and neutropenia.
  • the package insert or the label further indicates that the compound can be discontinued without prior dose reduction.
  • the one or more of the elevated levels of the enzymes or molecules are Grade 4 events. In some embodiments, the one or more of the hematologic conditions are Grade 4 events.
  • the package insert or the label is in a position which is visible to prospective purchasers.
  • the compound is in a unit dosage form or capsule form.
  • the package insert or the label indicates that, upon administration of the admixture to a human subject, the Cmax of the compound is achieved within about 2 to about 4 hours post-dose. In some embodiments, the package insert or the label indicates that, upon administration of the compound to a human subject, the elimination half life of the compound is about 16 to about 34 hours. In some embodiments, the package insert or the label indicates that the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the package insert or the label indicates that the accumulation of the compound is about 1.25 to about 4.0 fold at steady state when the compound is dosed once daily.
  • the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate.
  • the weight ratio of the compound to microcrystalline cellulose in the admixture is between about 1:1.5 to 1:15, and wherein the weight for the compound is the free base moiety weight of the compound.
  • the weight ratio of the compound to sodium stearyl fumarate in the admixture is between about 5:1 to about 50:1, and wherein the weight for the compound is the free base moiety weight of the compound.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the microcrystalline cellulose is silicified microcrystalline cellulose.
  • a compound in the manufacture of a medicament for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate).
  • the compound is administered orally.
  • the use is according to a method described herein.
  • a compound in the manufacture of a medicament for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • JAK2 Janus Kinase 2
  • a compound in the manufacture of a medicament for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy.
  • the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof
  • the use is according to a method described herein.
  • a compound for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof
  • the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate).
  • the compound is administered orally.
  • the treatment is according to a method described herein.
  • a compound for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • JAK2 Janus Kinase 2
  • a compound for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy.
  • the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the treatment is according to a method described herein.
  • FIG. 2 shows WBC count in subjects treated with TG101348.
  • the baseline WBC count was >11 ⁇ 10 9 /L.
  • the doses at follow-up ranged from 360 to 680 mg/day.
  • Last follow-up visit ranged from 8 to 24 weeks (median 24 weeks). “ULN” means upper limit of normal.
  • FIG. 3 shows platelet count in subjects treated with TG101348.
  • the doses at follow-up ranged from 360 to 680 mg/day.
  • Last follow-up visit ranged from 12 to 24 weeks (median 24 weeks). “ULN” means upper limit of normal.
  • FIG. 4 shows the percentages of subjects with worsened, unchanged, improved or resolved constitutional symptoms (fatigue, early satiety, cough, night sweats, and pruritus) in subjects treated with TG101348.
  • Last visit ranged from 4 to 24 weeks (median 20 weeks).
  • FIG. 5 shows the cytokine levels (IL-6, IL-8, IL-2 and TNF- ⁇ ) in subjects treated with TG101348. The values shown are median values.
  • the doses at follow-up were 360 to 680 mg/day.
  • Last follow-up visit ranged from 20 to 72 weeks (median 24 weeks).
  • FIG. 7 shows the bone marrow cellularity at baseline (60% cellularity) and after 18 cycles of TG101348 treatment (5-10% cellularity) in a 76-year-old male subject with V617F negative PMF.
  • the starting dose was 30 mg/day and the dose at follow-up was 520 mg/day.
  • FIG. 8 shows the bone marrow fibrosis at baseline (3+) and after 18 cycles of TG101348 treatment (0) in a 56-year-old male subject with V617F negative PMF.
  • the starting dose was 240 mg/day and the dose at follow-up was 440 mg/day.
  • FIG. 9 shows various measurements of a subject with JAK2 V617F-positive PMF treated with TG101348 (starting dose at 680 mg/day).
  • FIG. 12A shows plot of mean plasma TG101348 concentrations versus time on a semi-log scale (Cycle 1, Day 1).
  • FIG. 12B shows plot of mean plasma TG101348 concentrations versus time on a semi-log scale (Cycle 1, Day 28).
  • FIGS. 14A-14C show effects of TG101348 on symptoms of myelofibrosis.
  • FIG. 15 shows response of leukocytosis to TG101348 therapy.
  • WBC white blood cell
  • FIGS. 16A-16D show effect of TG101348 therapy on JAK2V617F allele burden.
  • the y-axis represents the JAK2V617F allele burden from 1.0 (100%) to 0.0 (0%).
  • the change in JAK2V617F allele burden per cycle of treatment up to end of cycle 12; i.e. C13D1 as compared to pre-study baseline is shown for the 2 groups ( FIGS.
  • FIGS. 16B and 16D the change at the end of cycle 6 (i.e. C7D1) and cycle 12 is shown in FIGS. 16B and 16D .
  • the Wilcoxon matched-pair signed-rank test was used to compare the median JAK2V617F allele burden for the comparisons.
  • FIGS. 17A-D shows absolute changes in pro-inflammatory cytokine levels from baseline at cycle 6: IL-6 ( FIG. 17A ), TNF- ⁇ ( FIG. 17B ), IL-8 ( FIG. 17C ), and IL-2 ( FIG. 17D ). Absolute differences in IL-6 ( ⁇ 4719 pg/mL) and IL-2 ( ⁇ 1827 pg/mL) are omitted from FIGS. 17A and 17D , respectively, for 1 subject (101-039) because they skewed presentation of data for other subjects.
  • FIG. 18 shows a plot of mean plasma TG101348 concentrations versus time on a linear plot after once daily oral doses (Cycle 1; Day 28).
  • treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired clinical results can include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals.
  • beneficial clinical results include one or more of reduction of splenomegaly, improvement in constitutional symptoms (such as early satiety, fatigue, night sweats, cough, and pruritus), reduction of leukocytosis, reduction of thrombocytosis, decrease of JAK2V617F allele burden, reduction of bone marrow fibrosis, and/or reduction of bone marrow cellularity.
  • delay development of a disease means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as myelofibrosis) or symptoms of the disease, and can include “progression free survival”. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease.
  • an “effective dosage” or “effective amount” of drug, compound, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results.
  • beneficial or desired results can include, for example, one or more results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results can include, include, for example one or more clinical results such as decreasing one or more symptoms and pathological conditions resulting from or associated with the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival.
  • an effective amount of a drug may have the effect in reducing one or more of splenomegaly, improving constitutional symptoms (such as early satiety, fatigue, night sweats, cough, and pruritus), reducing leukocytosis, reducing thrombocytosis, decreasing JAK2V617F allele burden, reducing bone marrow fibrosis, and/or reducing bone marrow cellularity.
  • An effective dosage can be administered in one or more administrations.
  • An effective dosage of drug, compound, or pharmaceutical composition can be, for example, an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
  • an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • “ameliorating” bone marrow cellularity or bone marrow fibrosis refers to reducing the level of bone marrow cellularity or bone marrow fibrosis in a subject compared to the level of bone marrow cellularity or bone marrow fibrosis prior to commencing treatment with the compound provided herein.
  • the reduction of bone marrow cellularity or bone marrow fibrosis can be at least by 5, 10, 20, 30, 40, 50, 60, 70, 80, or 90%.
  • conjunction with refers to administration of one treatment modality in addition to another treatment modality.
  • in conjunction with can refer to administration of one treatment modality before, during or after administration of the other treatment modality to the individual.
  • a “patient” or a “subject” refers to a mammal including a human, a dog, a horse, a cow or a cat, etc.
  • pharmaceutically acceptable refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and can be administered to a subject.
  • salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof
  • references to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X.”
  • compositions and methods provided herein can include “consisting” and/or “consisting essentially of” aspects and variations.
  • pharmaceutical compositions comprising N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and a pharmaceutically acceptable excipient or carrier.
  • N-tert-Butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide has the following chemical structure:
  • compositions may be formulated into therapeutic compositions as natural or salt forms.
  • Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, or ferric hydroxide, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino-ethanol, histidine, procaine, and the like.
  • Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric acid, sulfuric acid, or phosphoric acid, or organic acids such as acetic acid, citric acid, p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, tartaric acid, mandelic acid, and the like.
  • inorganic acids such as, for example, hydrochloric acid, sulfuric acid, or phosphoric acid
  • organic acids such as acetic acid, citric acid, p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, tartaric acid, mandelic acid, and the like.
  • Salts of the compounds provided herein can include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the compounds provided herein can also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, methanesulfonic acid and the like. Additional excipients which are contemplated for use in the practice of the compositions and methods provided herein are those available to those of ordinary skills in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, Md. (1989), the relevant contents of which are incorporated herein by reference.
  • the compounds provided herein can include polymorphs.
  • the compound described herein may be in alternative forms.
  • the compound described herein may include a hydrate form.
  • “hydrate” refers to a compound provided herein which is associated with water in the molecular form, i.e., in which the H—OH bond is not split, and may be represented, for example, by the formula R.H 2 O, where R is a compound provided herein.
  • a given compound may form more than one hydrate including, for example, monohydrates (R.H 2 O) or polyhydrates (R.nH 2 O wherein n is an integer greater than 1) including, for example, dihydrates (R.2H 2 O), trihydrates (R.3H 2 O), and the like, or fractional hydrates, such as, for example, R.n/2H 2 O, R.n/3H 2 O, R.n/4H 2 O and the like wherein n is an integer.
  • the compounds described herein may also include acid salt hydrate forms.
  • acid salt hydrate refers to a complex that may be formed through association of a compound having one or more base moieties with at least one compound having one or more acid moieties or through association of a compound having one or more acid moieties with at least one compound having one or more base moieties, said complex being further associated with water molecules so as to form a hydrate, wherein said hydrate is as previously defined and R represents the complex herein described above.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate and has the following chemical structure:
  • compositions for the administration of the compound described herein, either alone or in combination with other therapeutic agents may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy and methods described in Examples 4, 5 and 6. Such methods can include bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • the pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as hard or soft capsules.
  • the suitable capsule shell may be hard gelatin or hydroxypropylmethyl cellulose (“HPMC”).
  • formulations comprising (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) one or more excipients, and (iii) one or more lubricants.
  • the formulations may be in capsule form and administered orally.
  • the formulations may be in unit dosage form.
  • the excipient is lactose (such as Fast-Flo), mannitol (such as Parteck M200), microcrystalline cellulose (“MCC”) (such as Avicel PH102), MCC (such as ProSolv 90 HD).
  • the lubricant is magnesium stearate, sodium stearyl fumarate (such as Pruv), or sodium laurel fumarate.
  • the microcrystalline cellulose is silicified microcrystalline cellulose.
  • the capsule is hard gelatin capsule.
  • a capsule suitable for oral administration comprising an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate), wherein the admixture is contained in the capsule.
  • an excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • a lubricant e.g., sodium stearyl fumarate
  • Microcrystalline cellulose may be used as a filler and/or diluent in the capsules provided herein.
  • Sodium stearyl fumarate may be used as a lubricant in the capsules provided herein.
  • the microcrystalline cellulose is silicified microcrystalline cellulose.
  • silicified microcrystalline cellulose may be composed of microcrystalline cellulose and colloidal silicon dioxide particles.
  • the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • the capsule contains about 10 mg to about 680 mg of the compound, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 10 mg to about 650 mg (or about 10 mg to about 550 mg or about 10 mg to about 500 mg), wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 100 mg to about 600 mg (or about 200 mg to about 550 mg or about 300 mg to about 500 mg), wherein the specified weight is the free base moiety weight of the compound.
  • the capsule contains about 10 mg, about 20 mg, about 40 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, or about 650 mg of the compound, wherein the specified weight is the free base moiety weight of the compound.
  • the capsule is a hard gelatin capsule.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the weight ratio of the compound to excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in the capsule is between about 1:1.5 to about 1:15 (e.g., between about 1:5 to about 1:10, between about 1:5 to about 1:12, or between about 1:10 to about 1:15), wherein the weight of the compound is the free base moiety weight of the compound.
  • excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • the weight ratio of the compound to lubricant (e.g., sodium stearyl fumarate) in the capsule is between about 5:1 to about 50:1 (e.g., between about 5:1 to about 10:1, between about 5:1 to about 25:1, between about 5:1 to about 40:1, between about 7:1 to about 34:1, or between about 8:1 to about 34:1), wherein the weight of the compound is the free base moiety weight of the compound.
  • the capsule contains about 5% to about 50% (e.g., about 5% to about 10% or about 5% to about 35%) compound of the total fill weight of the capsule, wherein the weight of the compound is the free base moiety weight of the compound. In some embodiments, the capsule contains about 40% to about 95% (e.g., about 50% to about 90% or about 60% to about 90%) excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) of the total fill weight of the capsule.
  • excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • the capsule contains about 0.2% to about 5% (e.g., about 0.2% to about 2% or about 0.5% to about 1.5%, or about 0.5%, about 1%, or about 1.5%) lubricant (e.g., sodium stearyl fumarate) of the total fill weight of the capsule.
  • lubricant e.g., sodium stearyl fumarate
  • unit dosage forms comprising an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) an excipient (such as microcrystalline cellulose), and (iii) a lubricant (such as sodium stearyl fumarate). Any one of the capsules described herein may be used in a unit dosage form.
  • the unit dosage form is for treating myelofibrosis.
  • the treatment is according to a method described herein.
  • the unit dosage form comprises an admixture of (i) about 10 mg to about 680 mg (or about 10 mg to about 500 mg) of a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the specified weight is the free base moiety weight of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate.
  • the compound in the admixture is about 10 mg to about 500 mg, wherein the specified weight is the free base moiety weight of the compound.
  • the unit dosage form is in the form of a capsule, and the admixture is contained in the capsule.
  • the unit dosage form comprises about 10 mg, about 20 mg, about 40 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, or about 650 mg of the compound, wherein the specified weight is the free base moiety weight of the compound.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the admixture comprises (i) about 10 mg (or about any of 40 mg, 100 mg, 200 mg, 300 mg, 400 mg, or 500 mg) of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate, wherein the specified weight is the free base moiety weight of the compound.
  • the compound is N-tert-butyl-3-[(5 -methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the weight ratio of the compound to excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in the unit dosage form is between about 1:1.5 to about 1:15 (e.g., between about 1:5 to about 1:10, between about 1:5 to about 1:12, or between about 1:10 to about 1:15), wherein the weight of the compound is the free base moiety weight of the compound.
  • excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • the weight ratio of the compound to lubricant (e.g., sodium stearyl fumarate) in the unit dosage form is between about 5:1 to about 50:1 (e.g., between about 5:1 to about 10:1, between about 5:1 to about 25:1, between about 5:1 to about 40:1, between about 7:1 to about 34:1, or between about 8:1 to about 34:1), wherein the weight of the compound is the free base moiety weight of the compound.
  • the microcrystalline cellulose is silicified microcrystalline cellulose.
  • the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • the lubricant e.g., sodium stearyl fumarate
  • the lubricant is about 0.1% to about 10%, about 0.5% to about 5%, about 0.5% to about 3%, about 0.5% to about 2%, about 0.75% to about 1.5% of the capsule fill weight.
  • the lubricant e.g., sodium stearyl fumarate
  • the lubricant is at least about any one of 0.1%, 0.25%, 0.5%, 0.75%, 1%, 1.25%, 1.5%, 1.75%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, or 5% of the capsule fill weight.
  • the lubricant e.g., sodium stearyl fumarate
  • the lubricant is about any one of 0.1%, 0.25%, 0.5%, 0.75%, 1%, 1.25%, 1.5%, 1.75%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, or 5% of the capsule fill weight.
  • the weight ratio of a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in a capsule or unit dosage form is about 40:60 to about 10:90.
  • the weight ratio of the compound to an excipient is about any one of 95:5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, or 5:95.
  • an excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • the weight ratio of the compound to an excipient is about 1:1.5 to about 1:9.5, such as about any of 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5, 1:5, 1:5.5, 1:6, 1:6.5, 1:7, 1:7.5, 1:8, 1:8.5, 1:9, or 1:9.5.
  • the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the capsule contains about 5% to about 50% (e.g., about 5% to about 10% or about 5% to about 35%) compound of the total weight of the admixture, wherein the weight of the compound is the free base moiety weight of the compound.
  • the capsule contains about 40% to about 95% (e.g., about 50% to about 90% or about 60% to about 90%) microcrystalline cellulose (such as silicified microcrystalline cellulose) of the total weight of the admixture.
  • the capsule contains about 0.2% to about 5% (e.g., about 0.2% to about 2% or about 0.5% to about 1.5%, or about 0.5%, about 1%, or about 1.5%) sodium stearyl fumarate of the total weight of the admixture.
  • the capsule or unit dosage form contains an admixture of about 12 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 122 mg of silicified microcrystalline cellulose, and about 1 mg of sodium stearyl fumarate.
  • the capsule or unit dosage form contains an admixture of about 47 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 448 mg of silicified microcrystalline cellulose, and about 5 mg of sodium stearyl fumarate.
  • the capsule or unit dosage form contains an admixture of about 117 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the capsule or unit dosage form contains an admixture of about 235 mg of N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 357 mg of silicified microcrystalline cellulose, and about 6.00 mg of sodium stearyl fumarate.
  • oral solution formulations comprising a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the oral solution formulation further comprises methylcellulose.
  • the oral solution formulation further comprises methylcellulose and Tween 80.
  • the oral solution formulation comprises the compound at about 1 mg/ml to about 25 mg/ml, about 2 mg/ml to about 20 mg/ml, about 3 mg/ml to about 15 mg/ml, about 5 mg/ml to about 10 mg/ml. In some embodiments, the oral solution formulation comprises the compound at about any one of 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 6 mg/ml, 6.25 mg/ml, 6.5 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, or 12.5 mg/ml, or 15 mg/ml.
  • the oral solution formulation comprises about 0.1% to about 5%, 0.2% to about 3%, about 0.25% to about 2%, about 0.25% to about 1%, or about 0.5% by weight of methylcellulose. In some embodiments, the oral solution formulation comprises about 0.01% to about 0.5%, 0.02% to about 0.3%, about 0.025% to about 0.2%, about 0.025% to about 0.1%, or about 0.05% by weight of Tween 80.
  • the capsule does not comprise an absorption enhancer.
  • the capsule comprises an absorption enhancer (e.g., Vitamin E TPGS, Gelucire 44/14, Pluronic F127, or glyceryl monostearate).
  • an absorption enhancer e.g., Vitamin E TPGS, Gelucire 44/14, Pluronic F127, or glyceryl monostearate.
  • a capsule or unit dosage form provided may comprise one or more of the following properties: (1) upon administration to a subject such as human subject, the Cmax of the compound is achieved within about 2 to about 4 hours post-dose; (2) upon administration to a human subject, the elimination half life of the compound is about 16 to about 34 hours; (3) the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day; (4) the accumulation of the compound is about 1.25 to about 4.0 fold at steady state when the compound is dosed once daily.
  • Also provided are methods of preparing a capsule drug product comprising a) blending a lubricant with a compound that is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to generate granules and b) mixing the granules of a) with an excipient.
  • the lubricant is sodium stearyl fumarate.
  • the excipient is microcrystalline cellulose such as silicified microcrystalline cellulose.
  • Such method may be used to prepare a capsule or unit dosage form described herein.
  • the weight (such as weight ratio or weight percentage) and components regarding the compound, excipient, and/or lubricant may be according to any described herein.
  • a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof e.g., N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate).
  • the subject has myelofibrosis. In some embodiments, the subject is at risk of developing myelofibrosis. In some embodiments, the subject is a human subject. Any one of the formulations described herein such as capsule or unit dosage forms described herein may be used to treat a subject with myelofibrosis.
  • the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) an excipient (such as microcrystalline cellulose), and (iii) a lubricant (such as sodium stearyl fumarate).
  • Myelofibrosis that may be treated by the compounds described herein includes primary myelofibrosis (MF) and secondary myelofibrosis (e.g., myelofibrosis arising from antecedent polycythemia vera (post-PV MF) or essential thrombocythemia (post-ET MF)).
  • Myelofibrosis that may be treated by the compounds described herein also includes myelofibrosis of high risk, intermediate risk such as intermediate risk level 2.
  • Methods for diagnosing various types of myelofibrosis are known in the art. See, e.g., Cervantes et al., Blood 2009. In some embodiments, the subject with myelofibrosis has spleen of at least 5cm below costal margin as measured by palpation.
  • the subject has a point mutation from valine 617 to phenylalanie in the Janus kinase 2 (JAK2 kinase) (JAK2V617F) if the subject is a human, or a point mutation corresponding to the valine 617 to phenylalanie in the Janus kinase 2 (JAK2 kinase) if the subject is not a human.
  • the subject is negative for the valine 617 to phenylalanine mutation of JAK2 if the subject is a human, or negative for a mutation corresponding to the valine 617 to phenylalanie in the Janus kinase 2 (JAK2 kinase) if the subject is not a human
  • a subject is positive or negative for JAK2V617F can be determined by a polymerase chain reaction (“PCR”) analysis using genomic DNA from bone marrow cells or blood cells (e.g., whole blood leukocytes).
  • the PCR analysis can be an allele-specific PCR (e.g., allele-specific quantitative PCR) or PCR sequencing. See Kittur J et al., Cancer 2007, 109(11):2279-84 and McLornan D et al., Ulster Med J. 2006, 75(2):112-9, each of which is expressly incorporated herein by reference.
  • the subject treated with the methods described herein has previously received another myelofibrosis therapy or treatment.
  • the subject is a non-responder to the other myelofibrosis therapy or has a relapse after receiving the other myelofibrosis therapy.
  • the previous therapy may be a JAK2 inhibitor (e.g. INCB018424 (also known as ruxolitinib, available from Incyte), CEP-701 (lestaurtinib, available from Cephalon), or XL019 (available from Exelixis)) (See Verstovsek S., Hematology Am Soc Hematol Educ Program.
  • the subject has received ruxolitinib treatment for primary myelofibrosis, post-polycythemia vera myelofibrosis (Post-PV MF), post-essential thrombocythemia myelofibrosis (Post-ET MF), polycythemia vera, or essential thrombocythemia for at least 14 days and discontinued the treatment for at least 30 days.
  • Post-PV MF post-polycythemia vera myelofibrosis
  • Post-ET MF post-essential thrombocythemia myelofibrosis
  • polycythemia vera post-essential thrombocythemia vera
  • essential thrombocythemia for at least 14 days and discontinued the treatment for at least 30 days.
  • the previous therapy is a treatment with a compound described herein and the previous therapy has been discontinued upon indication of one or more elevated levels of amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and/or creatinine in the serum from the subject, and/or upon indication of a hematologic condition selected from the group consisting of anemia, thrombocytopenia, and neutropenia.
  • the dose of the compound in the second treatment is the same or lower than the dose in the previous therapy.
  • the subject may be treated orally and/or daily.
  • the subject (such as a human) may be treated by administering at a dose of about 240 mg per day to about 680 mg per day (or about 300 mg per day to about 500 mg per day), wherein the specified weight is the free base moiety weight of the compound.
  • the compound is administered at a dose of about any of 240 mg/day, 250 mg/day, 300 mg/day, 350 my/day, 400 mg/day, 450 mg/day, 500 mg/day, 550 mg/day, 600 mg/day, 650 mg/day, or 680 mg/day.
  • the compound may be in a capsule and/or a unit dosage form described herein.
  • the compound administered is in an admixture with a microcrystalline cellulose and sodium stearyl fumarate, and the admixture is in a capsule.
  • the compound is administered orally.
  • the treatment using the compound described herein is effective in reducing spleen size, ameliorating constitutional symptoms (such as early satiety, fatigue, night sweats, cough, and pruritus), reducing leukocytosis, reducing thrombocytosis, decreasing JAK2V617F allele burden, reducing bone marrow fibrosis, improving pruritus, improving cachexia, and/or reducing bone marrow cellularity.
  • the reduction, decrease, amelioration, or improvement can be at least by 5, 10, 20, 30, 40, 50, 60, 70, 80, or 90% compared to the level prior to commencing treatment with the compound provided herein.
  • the spleen becomes non-palpable in the subject after treatment.
  • the subject has complete resolution of leukocytosis and/or thrombocytosis after treatment. In some embodiments, the subject has complete resolution of pruritus after treatment.
  • the compound is administered to the subject daily for at least 1 cycle, at least 2 cycles, at least 3 cycles, at least 4 cycles, at least 5 cycles, or at least 6 cycles of a 28-day cycle. In some embodiments, the compound is administered to the subject daily for at least 6 cycles of a 28-day cycle, at least 8 cycles of a 28-day cycle, at least 10 cycles of a 28-day cycle, at least 12 cycles of a 28-day cycle, at least 15 cycles of a 28-day cycle, at least 18 cycles of a 28-day cycle, or at least 24 cycles of a 28-day cycle.
  • the compound is administered to the subject daily for at least one month, at least two month, at least three month, at least four month, at least five month, at least six month, at least eight month, or at least one year. In some embodiments, the compound is administered once a day.
  • the Cmax of the compound is achieved within about 1 to about 5 hours, about 1.5 to about 4.5 hours, about 2 to about 4 hours, or about 2.5 to about 3.5 hours post-dose.
  • the elimination half life of the compound is about 12 to about 40 hours, about 16 to about 34 hours, or about 20 to about 30 hours.
  • the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day.
  • the accumulation of the compound is about 1.1 to about 5 fold, about 1.25 to about 4.0 fold, about 1.5 to about 3.5 fold, about 2 to about 3 fold at steady state when the compound is dosed once daily.
  • the method comprises instructing the subject to ingest the effective amount of the compound on an empty stomach. In some embodiments, the methods further comprise instructing the subject to avoid ingesting agents that are at least moderate inducers or inhibitors of CYP3A4. In some embodiments, the subject does not receive concomitant treatment with or use of drugs to herbal agents known to be at least moderate inhibitors or inducers of CYP3A4. Based on in vitro evaluations, N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide is metabolized by human CYP3A4.
  • Agents that may increase N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide plasma concentrations i.e., CYP3A4 inhibitors
  • decrease N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino-]benzenesulfonamide plasma concentrations i.e., CYP3A4 inducers
  • herbal agents and foods e.g.
  • a list of clinically relevant substrates of CYP3A4 include alfentanil, Cyclosporine, Diergotamine, ethinyl estradiol, ergotamine, fentanyl, pimozide, quinidine, sirolimus, tacrolimus, clarithromycin erythromycin, telithromycin, alprazolam, diazepam, midazolam, triazolam, indinavir, ritonavir, saquinavir, prokinetic, cisapride, astemizole, chlorpheniramine, amlodipine, diltiazem, felodipine, nifedipine, verapamil, atorvastatin, cerivastatin, lovastatin, simvastatin, aripiprazole, gleevec, halopericol, sildenafil, tamoxifen, taxanes, trazodone, and Vincri
  • a list of clinically relevant inducers of CYP3A4 include carbamazepine, phenobarbital, phenytoin, pioglitazone, rifabutin, rifampin, St. John's wort, and troglitazone.
  • a list of clinically relevant inhibitors of CYP3A4 include indinavir, nelfinavir, ritonavir, clarithromycin, itraconazole, ketoconazole, nefazodone, erythromycin, grapefruit juice, verapamil, diltiazem, cimetidine, amiodarone, fluvoxamine, mibefradil, and Troleandomycin. See reference Flockhart et al., http://medicine.iupui.edu/clinpharm/ddis/clinicaltable.aspx., 2009.
  • Also provided herein are methods of monitoring treatment of myelofibrosis to a subject comprising (a) administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof; (b) monitoring a hematologic parameter and/or a non-hematologic parameter in the subject; and (c) determining if the subject should continue or discontinue with the treatment.
  • the hematologic parameter is selected from the group consisting of anemia, thrombocytopenia, and neutropenia.
  • the non-hematologic parameter is an enzyme or molecule in the blood or serum wherein an elevated level of the enzyme or molecule is indicative of tissue or organ damage.
  • the serum enzyme or molecule can be, for example, amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, alkaline phosphatase, and calcium. Methods of monitoring these parameters are known in the art and are described herein. See Examples 1-3.
  • the method further comprises administering to the subject an effective amount of the compound described herein after the subject has been discontinued with the treatment for at least 2 week, at least 3 weeks, or at least 4 weeks. In some embodiments, the previous treatment has been discontinued without prior dose reduction.
  • the treatment is discontinued when one or more of the parameters (including hematologic and non-hematologic parameters) are grade 3 or 4 events.
  • RESPONSE hematologic and non-hematologic Definition Grade 3 Grade 4 Hyperlipasemia A finding based on >2.0-5.0 ⁇ ULN* >5.0 ⁇ ULN laboratory test results that indicate an increase in the level of lipase in a biological specimen.
  • Alanine A finding based on >5.0-20.0 ⁇ ULN >20.0 ⁇ ULN aminotransferase laboratory test results that increased indicate an increase in the level of alanine aminotransferase (ALT or SGPT) in the blood specimen.
  • hypocalcemia A disorder characterized Corrected serum Corrected serum by laboratory test results calcium of calcium of that indicate a low ⁇ 7.0-6.0 mg/dL; ⁇ 6.0 mg/dL; concentration of calcium ⁇ 1.75-1.5 mmol/L; ⁇ 1.5 mmol/L; (corrected for albumin) in Ionized calcium Ionized calcium the blood. ⁇ 0.9-0.8 mmol/L; ⁇ 0.8 mmol/L; hospitalization life-threatening indicated consequences Anemia A disorder characterized Hgb ⁇ 8.0 g/dL; Life-threatening by a reduction in the ⁇ 4.9 mmol/L; consequences; amount of hemoglobin in ⁇ 80 g/L; transfusion urgent intervention 100 ml of blood.
  • Signs indicated indicated and symptoms of anemia may include pallor of the skin and mucous membranes, shortness of breath, palpitations of the heart, soft systolic murmurs, lethargy, and fatigability.
  • Thrombocytopenia a platelet count below the 25,000 to ⁇ 50,000/ ⁇ L below 25,000/ ⁇ L normal range for the population ([+ or ⁇ ] 2 standard deviations). In most laboratories, a normal platelet count is between 150,000 to 450,000/ ⁇ L Neutropenia A finding based on ⁇ 1000-500/mm 3 ; ⁇ 500/mm 3 ; laboratory test results that ⁇ 1.0-0.5 ⁇ 10 9 /L ⁇ 0.5 ⁇ 10 9 /L indicate a decrease in number of neutrophils in a blood specimen. *“ULN” refers to upper limit of normal.
  • articles of manufacture or kits containing a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the article of manufacture or the kit further includes instructions for using the compounds described herein in the methods provided herein.
  • the article of manufacture or the kit further comprises a label or a package insert providing the instructions.
  • the compound is in a capsule and/or a unit dosage form described herein.
  • the article of manufacture or kit may further comprise a container.
  • Suitable containers include, for example, bottles, vials (e.g., dual chamber vials), syringes (such as single or dual chamber syringes) and test tubes.
  • the container may be formed from a variety of materials such as glass or plastic, and the container may hold the compound, for example in the formulation to be administered.
  • the article of manufacture or the kit may further comprise a label or a package insert, which is on or associated with the container, may indicate directions for reconstitution and/or use of the compound.
  • the package insert or the label is in a position which is visible to prospective purchasers.
  • the label or package insert may further indicate that the compound is useful or intended for treating or preventing myelofibrosis in a subject.
  • the package insert or the label indicates that the compound can be used for ameliorating bone marrow cellularity and/or bone marrow fibrosis.
  • the package insert or the label indicates that the compound can be used for treating myelofibrosis in a subject, wherein the subject is negative for the valine 617 to phenylalanine mutation of human JAK2 (JAK2V617F) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • the package insert or the label indicates that the compound can be used for treating myelofibrosis in a subject, and that subject should discontinue the treatment upon indication of elevated levels of one or more of amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, and/or alkaline phosphatase and/or decreased level of calcium in the serum of the subject, and/or upon indication of one or more of anemia, thrombocytopenia, and/or neutropenia.
  • the package insert or the label further indicates that the compound can be discontinued without prior dose reduction.
  • kits or article of manufacture comprising (a) an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) lubricant (e.g., sodium stearyl fumarate), and (b) a package insert or a label indicating that the admixture is useful for treating myelofibrosis in a subject.
  • excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • lubricant e.g., sodium stearyl fumarate
  • kits or article of manufacture comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, wherein the subject has previously received another myelofibrosis therapy with a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the package insert or a label indicates that, upon administration of the compound to a human subject, the Cmax of the compound is achieved within about 1 to about 5 hours, about 1.5 to about 4.5 hours, about 2 to about 4 hours, or about 2.5 to about 3.5 hours post-dose. In some embodiments, the package insert or a label indicates that, upon administration of the compound to a human subject, the elimination half life of the compound is about 12 to about 40 hours, about 16 to about 34 hours, or about 20 to about 30 hours. In some embodiments, the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the accumulation of the compound is about 1.1 to about 5 fold, about 1.25 to about 4.0 fold, about 1.5 to about 3.5 fold, about 2 to about 3 fold at steady state when the compound is dosed once daily.
  • the package insert or the label instructs the subject to ingest the effective amount of the compound on an empty stomach. In some embodiments, the package insert or the label instructs the subject to avoid ingesting agents that are at least moderate inducers or inhibitors of CYP3A4. In some embodiments, the inducer or inhibitor of CYP3A4 is any one of the inducers or inhibitors of CYP3A4 described herein.
  • the use is according to a method described herein.
  • the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate).
  • an excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • a lubricant e.g., sodium stearyl fumarate.
  • the compound is administered orally.
  • the use is according to a method described herein.
  • a compound in the manufacture of a medicament for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • JAK2 Janus Kinase 2
  • a compound in the manufacture of a medicament for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy.
  • the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • a compound for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • the treatment is according to a method described herein.
  • the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate).
  • an excipient e.g., microcrystalline cellulose such as silicified microcrystalline cellulose
  • a lubricant e.g., sodium stearyl fumarate
  • the compound is administered orally.
  • the treatment is according to a method described herein.
  • a compound for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • JAK2 Janus Kinase 2
  • a compound for treating myelofibrosis in a subject wherein the compound is N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy.
  • the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • TG101348 refers to N-tert-butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • the subjects in this study were administered with capsule form of TG101348 as described in Example 5.
  • TG101348 was evaluated in a Phase I study for the treatment of myelofibrosis. This study was ongoing at the time the data were collected.
  • TG101348 is a potent, orally bioavailable, JAK2-selective small molecule inhibitor, that was evaluated in a Phase I study for the treatment of myelofibrosis.
  • the dose-limiting toxicity was asymptomatic grade 3 or 4 amylasemia/lipasemia that was reversible, and the maximum tolerated dose (“MTD”) was 680 mg.
  • MTD maximum tolerated dose
  • the most frequent non-hematological toxicities were mild nausea, vomiting, and/or diarrhea that were easily controlled or resolved spontaneously.
  • Grade 3/4 neutropenia and thrombocytopenia were observed in 14% and 25% of patients, respectively.
  • TG101348 had activity in reducing spleen size, leukocyte count, and JAK2V617F (“VF”) allele burden.
  • VF JAK2V617F
  • the cumulative drug exposure to the time when the data in this example were collected was 362 patient-months; exposure at or above MTD ( ⁇ 680 mg) was 154 patient-months.
  • Gr1/2 Grade 1/2
  • TG101348 was well tolerated in patients with myelofibrosis. Spleen and leukocyte responses were frequent, observed early, and produced substantial clinical benefit for patients. These responses were associated with significant decrease in VF allele burden and pointed to activity of TG101348 against the malignant clone in myelofibrosis.
  • TG101348 was evaluated in a Phase I study for the treatment of myelofibrosis. This study is also described in Example 1. This example describes data available at the time of data collection.
  • This study was an open-label, multicenter, and dose-escalation study with expanded cohort dose confirmation at MTD.
  • the primary objective of this study was to determine safety/tolerability, DLT, MTD, and pharmacokinetics of TG101348 in subjects with MF.
  • the secondary objective of this study was to evaluate preliminary clinical and pharmacodynamic activity.
  • the demographic and baseline characteristics for the subjects are included in Table 2.
  • This study was a dose-escalation study with expanded cohort dose confirmation at MTD. Below describes the data with a focus on the dose confirmation cohort who initiated treatment at a dose of 680 mg/day.
  • the effect of TG101348 on leukocytosis is shown in FIG. 2 .
  • the baseline WBC count was >11 ⁇ 10 9 /L. 73% of subjects had normal WBC counts at their follow-up visit.
  • the effect of TG101348 on thrombocytosis is shown in FIG. 3 (baseline platelet count >450 ⁇ 10 9 /L).
  • TG101348 was able to reduce platelet counts.
  • the effects of TG101348 on constitutional symptoms (baseline versus last visit) are shown in FIG. 4 .
  • TG101348 was able to improve the MF-associated constitutional symptoms.
  • TG101348 had no significant changes on cytokine levels (see FIG. 5 , all values shown are medians).
  • FIG. 6 shows that TG101348 was able to decrease JAK2 V617F allele burden in 59% of the subjects with baseline >20%.
  • FIG. 7 shows the effects of TG101348 on bone marrow cellularity in a 76-year-old male with V617F negative PMF.
  • the starting dose was 30 mg/day and the dose at follow-up was 520 mg/day.
  • FIG. 7 shows that TG101348 was able to reduce bone marrow cellularity in this subject from 60% bone marrow cellularity at baseline to 5-10% bone marrow cellularity after 18 cycles.
  • FIG. 8 shows the effect of TG101348 on bone marrow fibrosis in a 56-year-old male with V617F negative PMF.
  • the starting dose was 240 mg/day and the dose at follow-up was 440 mg/day.
  • FIG. 8 shows that TG101348 was able to reduce bone marrow fibrosis in this subject from 3+ at baseline to 0 after 18 cycles.
  • FIG. 9 shows various measurements in a subject with JAK2V617F-positive PMF that started at TG101348 680 mg/day.
  • TG101348 was able to reduce the palpable spleen size from 9 cm to 0 cm and led to complete resolution of pruritus in this subject.
  • TG101348 was generally well tolerated, with manageable, grade 1 gastrointestinal effects, especially at higher doses. The data indicated no long-term toxicities. The expected on-target myelosuppressive effect appeared to be mostly limited to erythropoiesis, which may be attenuated at lower, but still effective, doses. TG101348 had remarkable activity in MF-associated splenomegaly: ⁇ two-thirds achieved ⁇ 50% reduction in palpable splenomegaly; ⁇ 30% had complete response. TG101348 had significant anti-myeloproliferation activity with virtually all treated subjects experiencing complete resolution of leukocytosis and thrombocytosis.
  • TG101348 had remarkable activity against MF-associated constitutional symptoms, pruritus and cachexia. TG101348 induced a significant decrease in JAK2V617F allele burden in a substantial proportion of treated subjects. TG101348 had minimal effect on serum levels of proinflammatory cytokines; this was consistent with the absence of immediate adverse cytokine-rebound phenomenon upon study drug discontinuation. Without wishing to be bound by any theory, the activity of TG101348 appeared to be a direct consequence of its JAK2 inhibitory activity and not an indirect effect from non-specific anti-cytokine activity. Furthermore, the preliminary observations showed reduction in BM cellularity and reticulinfibrosis with extended treatment.
  • Study Design The study constituted a Phase 1, dose-escalation trial (MF-TG101348-001). This study is also described in Examples 1 and 2. Study eligible patients were ⁇ 18 years of age with high- or intermediate-risk primary myelofibrosis (PMF), post-PV MF, or post-ET MF (Tefferi A et al., Leukemia 22:14-22, 2008). Additional eligibility criteria and participating centers are listed in Table 6. All patients provided written informed consent. The primary endpoints were determination of safety and tolerability, dose-limiting toxicity (“DLT”), maximum tolerated dose (“MTD”) and pharmacokinetic (“PK”) behavior of TG101348. The secondary endpoint was assessment of therapeutic activity.
  • DLT dose-limiting toxicity
  • MTD maximum tolerated dose
  • PK pharmacokinetic
  • MF-TG101348-001 Inclusion Criteria Diagnosis of MF (PMF, post-PV MF, or 1. Any chemotherapy, immunomodulatory post-ET MF) according to the revised WHO drug therapy, immunosuppressive therapy, criteria.
  • corticosteroids > 10 mg/day prednisone or equivalent, or growth factor treatment within 14 days (28 days in the case of darbepoetin) prior to initiation of TG101348.
  • High-risk MF (defined by Mayo PSS), or 2. Major surgery or radiation therapy within Mayo PSS intermediate-risk MF** 28 days prior to initiation of TG101348. accompanied by symptomatic splenomegaly and/or unresponsive to available therapy.
  • Women of childbearing potential unless MF, with the exception of adequately treated surgically sterile for at least 3 months (i.e., basal cell carcinoma and squamous cell hysterectomy), postmenopausal for at least 12 carcinoma of the skin. months (FSH > 30 U/mL), unless they agree to use effective, dual contraceptive methods (i.e., oral, injectable, or barrier method with male partner using a condom) while on study drug.
  • 10. Provide written informed consent to 10. Men who partner with a woman of participate. childbearing potential, unless they agree to use effective, dual contraceptive methods (i.e., a condom, with female partner using oral, injectable, or barrier method) while on study drug. 11.
  • AIDS acquired immunodeficiency syndrome
  • ALT alanine aminotransferase
  • ANC absolute neutrophil count
  • AST aspartate aminotransferase
  • CHF congestive heart failure
  • CVA cerebrovascular accident
  • ECG electrocardiogram
  • ECOG Eastern Cooperative Oncology Group
  • EMH extramedullary hematopoiesis
  • FSH follicle stimulating hormone
  • HIV human immunodeficiency virus
  • MF myelofibrosis
  • MI myocardial infarction
  • NYHA New York Heart Association
  • PSS prognostic scoring system
  • TIA transient ischemic attack
  • WBC white blood cell. *Tefferi and Vardiman.
  • TG101348 was administered orally once daily, with a treatment plan for continuous daily therapy for 24 weeks (6 ⁇ 28-day cycles). Intra-subject dose escalation was permitted after completion of at least 3 cycles of treatment at the starting dose. Once DLT was identified, a dose-confirmation cohort initiated treatment at the MTD. Treatment beyond 6 cycles was allowed on an extension study (MF-TG101348-002; NCT00724334) if deemed beneficial to the patient and if well tolerated.
  • Cytokine Assessment Samples for cytokine measurement were collected at baseline and every 4 weeks thereafter. Cytokine levels were measured using multiplexed sandwich ELISAs (Millipore, St. Charles, Mo.).
  • the starting dose of TG101348 was 30 mg/day and subsequent dose levels were 60, 120, 240, 360, 520, 680 and 800 mg/day (Table 7).
  • 800 mg/day 2 of 6 patients experienced DLT; consequently, the MTD was declared at 680 mg/day.
  • TG101348 doses at the end of each cycle per dose cohort are illustrated in FIGS. 10 and 11 .
  • MTD MTD
  • the median cycle at dose-reduction for the MTD cohort was cycle 3 (range 1-7); the median (range) dose at the end of cycle 3 was 680 mg/day (360-680 mg/day); and 520 mg/day (360-680 mg/day) at the end of cycle 6.
  • FIG. 18 shows a plot of mean plasma TG101348 concentrations versus time on a linear plot after once daily oral doses (Cycle 1; Day 28).
  • the figure shows the IC50, IC90, and 3 times IC90 (3 ⁇ IC90) values for TG101348 in relation to the plasma concentration of TG101348 over time.
  • a dose of 520 mg/day exhibited a plasma concentration TG101348 that was above 3 ⁇ IC90 over the course of at least 24 hours after the dose was administered.
  • a dose of 360 mg/day exhibited a Cmax above 3 ⁇ IC90 and a plasma concentration of TG101348 that was above IC90 over the course of at least 24 hours after the dose was administered.
  • the DLT in 2 of 6 patients treated at 800 mg/day was asymptomatic grade 3 or 4 hyperamylasemia (with or without hyperlipasemia) that was reversible.
  • the most common non-hematologic adverse events at least possibly related to TG101348 included predominantly grade 1 nausea, diarrhea and vomiting; grade 3 events were reported overall/in the MTD cohort for 3%/5%, 10%/13%, and 3%/3% of subjects, respectively, and there were no Grade 4 events (Table 9). These adverse events were dose-dependent, with grade 3 occurrences observed almost exclusively with a TG101348 starting dose of ⁇ 680 mg/day.
  • the gastrointestinal symptoms were largely self-limited or controlled by symptomatic treatment and/or dose reduction.
  • Grade 3/4 hematological adverse events considered related to TG101348 included anemia (35% of 37 subjects who were not transfusion dependent at baseline), thrombocytopenia (24%) and/or neutropenia (10%) (Table 10). The majority of treatment-emergent cytopenias were noted in the first three cycles of treatment. Of the 13 subjects who developed grade 3/4 anemia (all in the MTD cohort), 67% entered the study with grade 2 anemia. Emergence of transfusion requirement was significantly lower for subjects who initiated treatment at 240-520 mg/day (33%) as opposed to 680 mg/day (72%). Of the 14 subjects with grade 3/4 thrombocytopenia, 4 and 5 subjects entered the study with grade 1 and 2 thrombocytopenia, respectively.
  • Serious adverse events considered at least possibly related to TG101348 occurred in 8 subjects and included asymptomatic hyperlipasemia, thrombocytopenia/neutropenia, depression, tumor lysis syndrome, cerebrovascular accident, and dehydration (Table 11).
  • One subject discontinued treatment due to Grade 4 thrombocytopenia; all other events were reversible and subjects were able to resume treatment at a lower dose after resolution of the adverse event.
  • Splenomegaly The onset of spleen response was rapid, and generally seen within the first 2 cycles. By cycle 6, 36 subjects (61%) had experienced a minimum 25% decrease in palpable spleen size, including 65% in the MTD cohort (intent-to-treat analysis). By this time-point, a ⁇ 50% decrease in palpable spleen size persistent for at least 8 weeks (i.e. Clinical Improvement (“CI”) per IWG-MRT criteria) had been observed in 39% and 45% of subjects overall and in the MTD cohort, respectively. Spleen responses per treatment cycle for the MTD cohort are shown in FIG. 13 . Three of 4 subjects (75%) with JAK2V617F-negative MF who completed 6 cycles of treatment achieved CI.
  • CI Clinical Improvement
  • the lowest starting dose at which CI was observed was 240 mg/day.
  • the median time (range) to CI across doses was 141 days (41 to 171), and 113 days (41-170) for the MTD cohort.
  • spleen responses (CI) were observed in 48% and 50% of subjects, for the overall and MTD cohorts, respectively.
  • the mean (standard deviation) duration of spleen response per IWG-MRT criteria was 315 ( ⁇ 129) days and 288 ( ⁇ 76) days for the overall and MTD cohorts, respectively.
  • Body weight At the end of 6 and 12 cycles, the median body weight was stable relative to baseline for the overall and MTD cohorts (Table 13).
  • Leukocytosis and Thrombocytosis Leukocytosis (WBC count >11 ⁇ 10 9 /L) was present at baseline in 33 subjects (56%), 28 of whom completed 6 cycles of treatment; of these, 18 were in the MTD cohort. Following 6 cycles, 16 subjects across doses (57%) and 13 subjects in the MTD cohort (72%) achieved a normal WBC count ( FIG. 15 ); following 12 cycles, 14 of 25 (56%) across doses and 10 of 17 (59%) in the MTD cohort had normal WBC counts.
  • JAK2V617F allele burden Fifty-one subjects (86%) were JAK2V617F-positive, with a median (range) allele burden of 20% (3%-100%); of these, 23 (45%) had a “significant” allele burden (defined as >20% at baseline) with a median (range) of 60% (23%-100%).
  • JAK2V617F allele burden Fifty-one subjects (86%) were JAK2V617F-positive, with a median (range) allele burden of 20% (3%-100%); of these, 23 (45%) had a “significant” allele burden (defined as >20% at baseline) with a median (range) of 60% (23%-100%).
  • the median (range) allele burden was 17% (0%-100%) and 19% (0%-100%), respectively.
  • TG101348 therapy can be discontinued without prior dose reduction or tapering.
  • Subjects that were discontinued did not experience “cytokine rebound”. This indicates that the treatment may be discontinued without prior dose reduction.
  • Cytokine rebound in the context of myelofibrosis is a phenomenon that has occurred in patients receiving therapy other than TG101348 therapy and were discontinued for any reason.
  • the discontinued patients experienced severe symptoms including acute spleen size enlargement and relapse of constitutional symptoms.
  • the discontinued patients experienced life-threatening hemodynamic disturbances (Wadleigh and Tefferi, Clinical Advances in Hematology & Oncology, 8:557-563, 2010).
  • TG101348 appeared to be unique in its ability to induce a significant and sustained decrease in JAK2V617F mutant allele burden. Without wishing to be bound by any theory, it appeared that the effect of JAK2 inhibition on disease burden was the basis for evidence of clinical efficacy in myelofibrosis with TG101348, as opposed to an indirect anti-cytokine effect that may play a major role in responses to JAK family antagonists that have off-target activity for JAK1 as well as for JAK2.
  • the DLT (asymptomatic hyperamylasemia, sometimes with hyperlipasemia) for TG101348 was observed with other small molecule inhibitors including nilotinib (Kantarjian HM et al., Blood 110:3540-3546, 2007). Gastrointestinal adverse events were frequent in this study but accounted for treatment discontinuation in only one subject. These symptoms occurred as early as after the first administered dose, and demonstrated a clear dose-dependent relationship. The myelosuppressive effects of TG101348 were also dose-dependent.
  • a lower starting dose e.g. 400-500 mg/day
  • a dynamic dosing schedule may maximize the opportunity for identifying a patient-specific optimal dose.
  • the Intermediate was synthesized from 2,4-dichloro-5-methylpyrimidine (SM1) and N-t-butyl-3-aminobenzenesulfonamide (SM2) in the following steps: (1) Mix MeOH (6.7UOa) and SM1 (Combi Blocks) (UOa); (2) Add SM2 (1.15UOa, 082eq) and H 2 O (8.5UOa); (3) Heat 45° C., 20 h, N 2 , IPC CPL SM2 ⁇ 2%; (4) Cool 20° C.; (5) Centrifuge, N 2 ; (6) Wash H 2 O (2.1UOa)+MeOH (1.7UOa); (7) Mix solid in H 2 O (4.3UOa) +MeOH (3.4UOa); (8) Centrifuge, N 2 ; (9) Wash H 2 O (2.1UOa)+MeOH (1.7UOa); and (10) Dry 45° C., vacuum, 15 h. Obtained Intermediate, mass 49.6kg (UOb);
  • N-tert-Butyl-3-[(5-methyl-2- ⁇ [4-(2-pyrrolidin-1-ylethoxy)phenyl]amino ⁇ pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate was prepared from 4-p-(1-pyrrolidinyl)ethoxylaniline dihydrochloride (SM3) and Intermediate following steps (A) and (B).
  • Step (A) preparation of free base of SM3 (3) from SM3, comprised steps (1)-(9): (1) Solubilize NaOH (0.42UOb) in H 2 O (9UOb); (2) Cool ⁇ 20° C., N2; (3) Add TBME (6UOb) then SM3 (Malladi Drugs) (1.06UOb); (4) Mix >20 mn then stop; (5) Drain Aq Ph then extract by TBME (3UOb); (6) Combine Or Ph; (7) Concentrate, vacuum, T ⁇ 40° C., to an Oil; (8) Solubilize in IPA (2.5UOb); and (9) Calculate dry extract 23%.
  • Step (B) comprised the steps (1)-(6): (1) Mix IPA (10.5UOb) and Intermediate (UOb); (2) Add free base of SM3 (0.75UOb, 1.33eq/interm); (3) add HCl conc (0.413UOb); (4) Heat 70° C., 20 h, N 2 , IPC CPL Interm ⁇ 2%; (5) Cool ⁇ 20° C.; (2) Centrifuge, N 2 ; (3) Wash IPA (3UOb); (4) Dry 50° C., vacuum, 26 h; (5) De-lump in Fitzmill; and (6) polybag (x2)/poly drum. Obtained TG101348 dihydrochloride monohydrate, mass 83.8kg; Yield 98%; OP: 99.5%.
  • TG101348 drug products were provided as 10 mg, 40 mg, and 200 mg capsule strengths, where weights are specified for the amount of active (i.e., free base) moiety of TG101348.
  • the quantitative composition of each strength of TG101348 drug product capsule is shown in Table 14.
  • A. Dry granulation of intragranular components (implemented for all three drug product strengths): 1. TG101348 and intragranular sodium stearyl fumarate were blended within a V-blender for 5 minutes. 2. The blend was passed through a conical mill equipped with a round 18-mesh screen and round impeller. The blend was recharged into the V-blender. 3. Intragranular silicified microcrystalline cellulose was sifted through a 20-mesh screen and added to the blender. The mixture was blended for 15 minutes. 4. The blend was passed through a roller compactor. 5. The roller compacted ribbons were passed through a conical mill equipped with a round 16-mesh screen and round impeller. 6. The milled material was blended within the V-blender for 5 minutes. 7. In-process check (IPC) samples were withdrawn from the V-blender using a sample thief. Samples were subjected to potency analysis.
  • IPC In-process check
  • Capsule-filling (implemented for all three drug product strengths): 1. If potency (from Step 7 in A for the 200 mg capsules, or Step 6 in B for the 10 mg and 40 mg capsules) was outside 98-102% (w/w) nominal, the capsule fill weight was adjusted accordingly. 2. The prepared material was encapsulated using automatic capsule filling machine. The prepared capsules were bottled and stores at 20-28° F. (68-82° C.) and ambient humidity.
  • HPLC method validation was performed using a one-analyst, one-run-per-analyst design, and satisfied all required criteria for specificity, sensitivity, precision, accuracy, linearity, and sample stability. Specificity was evaluated and confirmed by comparing peak resolution between TG101348 and all of its related compounds, intermediates, and degradants (established from forced degradation studies). The limit of quantitation and limit of detection was established at 0.10 ⁇ g/mL and 0.03 ⁇ g/mL TG101348, respectively. Precision for content uniformity was evaluated via six injections of the 10 mg and 200 mg strength capsules, prepared at the target assay concentration. RSD results were 3.7% and 5.8% for the 10 mg and 200 mg strength capsules, respectively.
  • Hard gelatin and hydroxypropylmethyl cellulose (HPMC) capsule shells (size #00250) were filled with 250 mg of TG101348 di-HCl monohydrate.
  • the filled capsules were placed on accelerated stability (40° C./75% relative humidity (RH) and 25° C./60% RH).
  • the capsules were packaged into 30 mL (1 oz) amber high density polyethylene (HDPE) bottles.
  • a summary of the formulations and accelerated stability protocol for the stability study is shown in Table 16.
  • Drug Substance Blend Compatibility with Fillers and Lubricants
  • Blends were prepared at a scale of 2.5 g each by prescreening all components through a 500 ⁇ m sieve, blending all components except the lubricant using aTurbula T2B blender for 10 min at 22 rpm, screening the blend through a 500 ⁇ m sieve, blending for 10 min, adding lubricant (weight adjusted) and blending for 5 min.
  • the blends were manufactured and stored in 30 mL amber HDPE bottles under primary (60° C./ambient humidity) and backup conditions (40° C./75% RH, 25° C./60% RH, and 5° C.).
  • a summary of the accelerated stability protocol is shown in Table 18. No appreciable changes in attributes (appearance, assay, impurities) were observed over the course of the study.
  • Excipient compatibility testing of dry blends of TG101348 di-HCl monohydrate with four fillers lactose, mannitol, microcrystalline cellulose (MCC) Avicel PH102, and MCC ProSolv 90 HD) and two lubricants (magnesium stearate and sodium stearyl fumarate (Pruv) (Table 17) indicated no incompatibilities.
  • Prosolv SMCC 90HD i.e., silicified microcrystalline cellulose
  • Lactose Fast-Flo i.e., spray dried lactose monohydrate
  • Magnesium stearate (vegetable grade) and Pruv were selected as lubricants for further testing. All excipients are global regulatory approved for use in solid oral dosage forms (USA, European Union, Japan).
  • a “matrix” of prototype capsule formulations for stability evaluation was designed, and is summarized in Table 19. Two dosage strengths were selected, 10 and 125 mg.
  • prototype formulations filled into gelatin capsule shells demonstrated >85% drug release within 15 minutes.
  • TG101348 is on the borderline between “low” and “high” permeability based on caco-2 permeability data.
  • bioavailability in multiple species was typically 20-25%. Therefore, it was not known whether an “absorption enhancer” would be required in the formulation to achieve adequate bioavailability.
  • silicified microcrystalline cellulose (Prosolv SMCC 90HD) was used as the primary filler/carrier excipient for the absorption enhancement formulation.
  • Four absorption enhancement excipient candidates were selected for further testing (Table 21).
  • Table 22 summarizes the absorption enhancement formulations tested. Melt granulation, as opposed to a direct blend manufacturing process, was selected to produce the formulation.
  • a crossover beagle dog PK study was performed testing five formulations: an oral solution as described below, two capsule formulations without absorption enhancer and two capsule formulations with absorption enhancer.
  • capsule formulations displayed immediate release characteristics with a demonstration of bioequivalence to the reference solution dose. Therefore, despite the boarderline permeability in human caco-2 cells and the 20-25% bioavailability in various animal species, capsule formulations without an absorption enhancer formulation demonstrated immediate release characteristics.
  • the initial formulation developed with the drug substance having rounded, granular particles with a mean particle size of 25 ⁇ m was 50:50 ratio by weight of TG101348 drug substance and filler, with 0.5% w/w lubricant.
  • a blend of drug, filler, and lubricant was prepared prior to roller compaction.
  • the drug substance was passed through a co-mil to de-agglomerate prior to blending with formulation excipients.
  • the drug substance having small needles demonstrated a high tendency to agglomerate on storage. After deagglomerating the drug substance having small needles, significant reagglomeration or ‘clumping’ would occur almost instantaneously. This reagglomeration was significantly reduced through blending the drug with lubricant prior to milling.
  • TG101348 di-HCL monohydrate comprised about a 50:50 ratio by weight of TG101348 drug substance and filler, with 0.5% w/w lubricant. This formulation exhibited a poor flow and significant sticking to the metal rolls within the roller compactor.
  • the amount of magnesium stearate lubricant could be increased in the formulation, however increasing the concentration within the formulation could adversely affect drug release kinetics.
  • the lubricant sodium laurel fumarate was also shown to be compatible with TG101348 di-HCl monodyrate and is less hygroscopic than magnesium stearate and was added (at a weight ratio of 2.0% w/w) instead of magnesium stearate which minimized sticking of the formulation to the metal rolls of the roller compactor. However, powder flow remained poor.
  • the ratio of TG101348 drug substance to filler was reduced from about 50:50 to about 40:60.
  • the lubricant (Prov) content was also reduced to 1% w/w which provided acceptable flow and minimal sticking within the roller compactor.
  • the dry granulation process developed as described herein to produce a particles having intragranular TG101348 di-HCL monohydrate and sodium laurel fumarate allowed the preparation of a range of capsule dosages using dry blending processes.
  • the mean granule size was approximately 300 ⁇ m and the silicified microcrystalline cellulose mean particle size was approximately 100 ⁇ m. Therefore, 40 mg and 10 mg capsule strength formulations were produced through dilution of the granules with extragranular silicified microcrystalline cellulose.
  • 40 mg strength capsules were produced using a formulation comprising comparable fill volume to the 200 mg formulation, within the same capsule shell (size #00 hard gelatin capsule). For the 10 mg strength capsules, a common blend with the 40 mg capsule strength formulation was used by filing a smaller capsule size.
  • An oral solution formulation was developed that contained the drug substance, 0.5% methylcellulose (MC) and 0.05% Tween 80.
  • a pH-stability study was conducted at 60° C. on formulations passed through a 0.22 ⁇ m polyethersulfone (PES) filter. No appreciable changes in attributes (appearance, assay, impurities) were observed over the course of the study (14 days).
  • a second oral solution formulation was developed that contained the drug substance and 0.5% MC. The second oral solution formulation was used in the dog PK study.

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Provided herein are compositions and methods for treating myelofibrosis in a subject. The methods comprise administering to the subject an effective amount of compound which is which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional application of U.S. patent application Ser. No. 16/549,043, which is a divisional application of U.S. patent application Ser. No. 13/888,096, filed May 6, 2013, which is a continuation of PCT Application No. PCT/US2011/059643, filed Nov. 7, 2011, which claims the priority benefit of U.S. Provisional Application No. 61/410,924, filed Nov. 7, 2010, the disclosure of each of which is incorporated herein by reference in its entirety.
  • TECHNICAL FIELD
  • Provided herein are compositions and methods for treating myelofibrosis. The compositions and methods provided herein relate to treatment of myelofibrosis with compounds that inhibit JAK2 or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • BACKGROUND
  • Myelofibrosis (“MF”) is a rare disease mainly affecting people of older age. MF is a BCR-ABL1-negative myeloproliferative neoplasm (“MPN”) that presents de novo (primary) or may be preceded by polycythemia vera (“PV”) or essential thrombocythemia (“ET”). Clinical features include progressive anemia, marked splenomegaly, constitutional symptoms (e.g. fatigue, night sweats, bone pain, pruritus, and cough) and weight loss (Tefferi A, N Engl J Med 342:1255-1265, 2000). Median survival ranges from less than 2 years to over 15 years based on currently identified prognostic factors (Cervantes F et al., Blood 113:2895-2901, 2009; Hussein K et al. Blood 115:496-499, 2010; Patnaik M M et al., Eur J Haematol 84:105-108, 2010). Mutations involving JAK2 (James C et al., Nature 434:1144-1148, 2005; Scott L M et al., N Engl J Med 356:459-468, 2007), MPL (Pikman Y et al., PLoS Med 3:e270, 2006), TET2 (Delhommeau F et al., N Engl J Med 360:2289-2301, 2009), ASXL1 (Carbuccia N et al., Leukemia 23:2183-2186, 2009), IDH1/IDH2 (Green A et al., N Engl J Med 362:369-370, 2010; Tefferi A et al., Leukemia 24:1302-1309, 2010), CBL (Grand F H et al., Blood 113:6182-6192, 2009), IKZF1 (Jager R et al., Leukemia 24:1290-1298, 2010), LNK (Oh S T et al., Blood 116:988-992, 2010), or EZH2 (Ernst T et al., Nat Genet 42:722-726) have been described in patients with MPN, including those with MF. Some mutations occur at high frequency in MF (e.g. JAK2 mutations in ˜50% patients), and either directly (e.g. JAK2 or MPL mutations) or indirectly (e.g. LNK or CBL mutations) induce JAK-STAT hyperactivation.
  • The currently available treatments are not effective in reversing the process of MF, be it primary or secondary disease. The only potential for cure of the disease to date is bone marrow transplantation. However, most patients are not suitable bone marrow transplant candidates because of the older median age at diagnosis, in which transplant-related morbidity and mortality tends to be high. Thus management options of MF are currently inadequate to meet the needs of all patients. The main options for active intervention include cyto-reductive therapy, e.g. with hydroxyurea, treatment of anemia with androgens, erythropoietin and splenectomy. These options have not been shown to improve survival and are largely seen as palliative (Cervantes F., Myelofibrosis: Biology and treatment options, European Journal of Haematology, 2007, 79 (suppl.68) 13-17). Therefore, there is a need to provide additional therapy options for MF patients.
  • SUMMARY OF THE INVENTION
  • Provided herein are capsules suitable for oral administration. In some embodiments, the capsule comprises an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate, wherein the admixture is contained in the capsule.
  • In some embodiments, the capsule contains about 10 mg to about 680 mg of the compound, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 10 mg to about 500 mg of the compound. In some embodiments, the capsule contains about any of 10 mg, 40 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg of the compound. In some embodiments, the weight ratio of the compound to microcrystalline cellulose in the capsule is between about 1:1.5 to 1:15, wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to sodium stearyl fumarate in the capsule is between about 5:1 to about 50:1, and wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • Also provided herein are unit dosage forms comprising an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate. In some embodiments, the unit dosage forms are for treatment of myelofibrosis such as treatment of myelofibrosis according to a method described herein.
  • In some embodiments, the unit dosage form comprises an admixture of (i) about 10 mg to about 680 mg (or about 10 mg to about 500 mg) of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the specified weight is the free base moiety weight of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate. In some embodiments, the unit dosage form is in the form of a capsule, and the admixture is contained in the capsule. In some embodiments, the compound in the admixture is about 10 mg to about 500 mg, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the admixture comprises (i) about 10 mg (or about any of 40 mg, 100 mg, 200 mg, 300 mg, 400 mg, or 500 mg) of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the weight ratio of the compound to microcrystalline cellulose in the capsule is between about 1:1.5 to 1:15, wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to sodium stearyl fumarate in the capsule is between about 5:1 to about 50:1, and wherein the weight for the compound in the weight ratio is the free base moiety weight of the compound. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • In some embodiments, sodium stearyl fumarate is about 1% w/w of capsule fill weight. In some embodiments, the weight ratio of the compound to microcrystalline cellulose such as silicified microcrystalline cellulose is about 40:60 to about 10:90 (e.g., about 40:60 or about 1:1.5, or about 10:90 or about 1:9).
  • In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the unit dosage form or capsule contains an admixture of about 12 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 122 mg of silicified microcrystalline cellulose, and about 1 mg of sodium stearyl fumarate. In some embodiments, the unit dosage form or capsule contains an admixture of about 47 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 448 mg of silicified microcrystalline cellulose, and about 5 mg of sodium stearyl fumarate. In some embodiments, the unit dosage form or capsule contains an admixture of about 117 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the unit dosage form or capsule contains an admixture of about 235 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 357 mg of silicified microcrystalline cellulose, and about 6.00 mg of sodium stearyl fumarate. In some embodiments, the capsule is a hard gelatin capsule.
  • Also provided herein are methods of preparing a capsule drug product comprising a) blending a lubricant with a compound that is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to generate granules and b) mixing the granules of a) with an excipient. In some embodiments, the lubricant is sodium stearyl fumarate. In some embodiments, the excipient is microcrystalline cellulose such as silicified microcrystalline cellulose. In some embodiments, sodium stearyl fumarate is about 1% w/w of capsule fill weight. In some embodiments, the weight ratio of the compound to silicified microcrystalline cellulose is about 1:1.5 to about 1:9. In some embodiments, the weight ratio of the compound to silicified microcrystalline cellulose is about 1:1.5. In some embodiments, the weight ratio of the compound to silicified microcrystalline cellulose is about 1:9. In some embodiments, the capsule is a hard gelatin capsule.
  • Also provided herein are methods of treating myelofibrosis in a subject, comprising orally administering a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and wherein the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., a microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate). Any of the unit dosage forms or capsules described herein may be used. In some embodiments, there is provided a method of treating myelofibrosis in a subject comprising orally administering a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and wherein the compound is in a capsule containing an admixture of (i) the compound, (ii) a microcrystalline cellulose (e.g., silicified microcrystalline cellulose), and (iii) sodium stearyl fumarate.
  • Also provided herein are methods of treating myelofibrosis in a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • Also provided herein are methods of treating myelofibrosis in a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy. In some embodiments, the previous therapy is a treatment with a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the previous therapy comprises administration of INCB018424 (ruxolitinib). In some embodiments, the subject is unresponsive to the previous therapy. In some embodiments, the previous therapy is a treatment with N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the previous therapy has been discontinued upon indication of elevated levels of amylase, lipase, aspartate aminotransferase (“AST”), alanine aminotransferase (“ALT”), and/or creatinine. In some embodiments, the previous therapy has been discontinued upon indication of a hematologic condition selected from the group consisting of anemia, thrombocytopenia, and neutropenia.
  • Also provided herein are methods of ameliorating bone marrow cellularity or bone marrow fibrosis associated with myelofibrosis in a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • Also provided herein are methods of improving pruritus associated with myelofibrosis in a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl] amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof
  • Also provided herein are methods of monitoring treatment of myelofibrosis in a subject, comprising (a) administering to a subject an effective amount of a compound which is N-tert-butyl-3-[(5 -methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof; (b) monitoring a non-hematologic parameter selected from the group consisting of amylase level, lipase level, aspartate aminotransferase (AST) level, alanine aminotransferase (ALT) level, and creatinine level in the subject; and (c) determining if the subject should continue or discontinue with the treatment. Also provided herein are methods of monitoring treatment of myelofibrosis to a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and discontinuing the treatment upon indication of elevated levels of one or more enzymes or molecules selected from the group consisting of amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatinine in the serum of the subject without prior dose reduction. In some embodiments, the one or more of the elevated levels are Grade 4 events.
  • Also provided herein are methods of monitoring a treatment of myelofibrosis to a subject, comprising (a) administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof; (b) monitoring a hematologic parameter selected from the group consisting of anemia, thrombocytopenia, and neutropenia in the serum of the subject; and (c) determining if the subject should continue or discontinue with the treatment. Also provided herein are methods of monitoring treatment of myelofibrosis to a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and discontinuing the treatment upon indication of one or more hematologic conditions selected from the group consisting of anemia, thrombocytopenia, and neutropenia without prior dose reduction. In some embodiments, the one or more hematologic conditions are grade 4 events.
  • In some embodiments of the methods of monitoring treatment provided herein, the methods further comprise administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof after the subject has been discontinued with the treatment for at least 2 weeks. In some embodiments, the subject has been discontinued with the treatment for at least 3 weeks. In some embodiments, the subject has been discontinued with the treatment for at least 4 weeks. In some embodiments, the treatment has been discontinued without prior dose reduction.
  • In some embodiments, the compound is administered to the human subject at a dose of about 240 mg per day to about 680 mg per day, and wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is administered at a dose of about 300 mg per day to about 500 mg per day (e.g., about 300 mg per day to about 400 mg per day, or about 400 mg per day to about 500 mg per day), and wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is administered at a dose of about any of 240 mg per day, 250 mg per day, 300 mg per day, 350 mg per day, 400 mg per day, 450 mg per day, 500 mg per day, 550 mg per day, 600 mg per day, 650 mg per day, or 680 mg per day, and wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is administered daily and/or orally. In some embodiments, the compound is administered over a period of at least 1 cycle, at least 2 cycles, at least 3 cycles, at least 4 cycles, at least 5 cycles, or at least 6 cycles (e.g., at least 7 cycles, at least 8 cycles, at least 9 cycles, at least 10 cycles, at least 11 cycles, at least 12 cycles, at least 15 cycles, at least 18 cycles, or at least 24 cycles) of a 28-day treatment cycle. In some embodiments, the compound is in a capsule and administered orally. In some embodiments, the compound is in a unit dosage form. Any of the capsules or unit dosage forms described herein may be administered. In some embodiments of the methods provided herein, the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate. In some embodiments, the weight ratio of the compound to microcrystalline cellulose in the admixture is between about 1:1.5 to 1:15, and wherein the weight for the compound is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to sodium stearyl fumarate in the admixture is between about 5:1 to about 50:1, and wherein the weight for the compound is the free base moiety weight of the compound. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the subject is a human.
  • In some embodiments of the compositions and methods provided herein, the subject has primary myelofibrosis. In some embodiments of the compositions and methods provided herein, the subject has post polycythemia vera myelofibrosis. In some embodiments of the compositions and methods provided herein, the subject has post essential thrombocythemia myelofibrosis. In some embodiments, the subject has high risk myelofibrosis. In some embodiments, the subject has intermediate risk myelofibrosis (such as intermediate risk level 2). In some embodiments of the compositions and methods provided herein, the subject is positive for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or positive for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments of the compositions and methods provided herein, the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments of the compositions and methods provided herein, the subject has palpable splenomegaly. In some embodiments, the subject with myelofibrosis has spleen of at least 5cm below costal margin as measured by palpation. In some embodiments of the compositions and methods provided herein, the subject is transfusion dependent. In some embodiments of the compositions and methods provided herein, the subject is not transfusion dependent.
  • In some embodiments of the methods provided herein, upon administration of the compound to a human subject, the Cmax of the compound is achieved within about 2 to about 4 hours post-dose. In some embodiments, upon administration of the compound to a human subject, the elimination half life of the compound is about 16 to about 34 hours. In some embodiments, the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the accumulation of the compound is about 1.25 to about 4.0 fold at steady state when the compound is dosed once daily. In some embodiments, the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) microcrystalline cellulose, and (iii) sodium stearyl fumarate. In some embodiments, the weight ratio of the compound to microcrystalline cellulose in the admixture is between about 1:1.5 to 1:15, and wherein the weight for the compound is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to sodium stearyl fumarate in the admixture is between about 5:1 to about 50:1, and wherein the weight for the compound is the free base moiety weight of the compound. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose.
  • Also provided herein are articles of manufacture or kits comprising (a) any one of the capsules provided herein, and (b) a package insert or a label indicating that the capsule is useful for treating myelofibrosis in a subject. Also provided herein are articles of manufacture or kits comprising (a) any one of the unit dosage forms provided herein, and (b) a package insert or a label indicating that the capsule is useful for treating myelofibrosis in a subject. In some embodiments, there is provided an article of manufacture or kit comprising (a) an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) microcrystalline cellulose, and (iii) sodium stearyl fumarate, and (b) a package insert or a label indicating that the admixture is useful for treating myelofibrosis in a subject.
  • Also provided herein are articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2.
  • Also provided herein are articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, wherein the subject has previously received another myelofibrosis therapy. In some embodiments, the previous therapy is a treatment with a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • Also provided herein are articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for ameliorating bone marrow cellularity and/or bone marrow fibrosis.
  • Also provided herein are articles of manufacture or kits comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for improving pruritus associated with myelofibrosis.
  • Also provided herein are articles of manufacture or kits comprising a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, and that subject should discontinue the treatment upon indication of elevated levels of one or more enzymes or molecules selected from the group consisting of: amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatinine in the serum of the subject, and/or upon indication of one or more hematologic condition selected from the group consisting of anemia, thrombocytopenia, and neutropenia. In some embodiments, the package insert or the label further indicates that the compound can be discontinued without prior dose reduction. In some embodiments, the one or more of the elevated levels of the enzymes or molecules are Grade 4 events. In some embodiments, the one or more of the hematologic conditions are Grade 4 events.
  • In some embodiments, the package insert or the label is in a position which is visible to prospective purchasers. In some embodiments, the compound is in a unit dosage form or capsule form.
  • In some embodiments, the package insert or the label indicates that, upon administration of the admixture to a human subject, the Cmax of the compound is achieved within about 2 to about 4 hours post-dose. In some embodiments, the package insert or the label indicates that, upon administration of the compound to a human subject, the elimination half life of the compound is about 16 to about 34 hours. In some embodiments, the package insert or the label indicates that the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the package insert or the label indicates that the accumulation of the compound is about 1.25 to about 4.0 fold at steady state when the compound is dosed once daily. In some embodiments, the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate. In some embodiments, the weight ratio of the compound to microcrystalline cellulose in the admixture is between about 1:1.5 to 1:15, and wherein the weight for the compound is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to sodium stearyl fumarate in the admixture is between about 5:1 to about 50:1, and wherein the weight for the compound is the free base moiety weight of the compound. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose.
  • In some embodiments, there is provided use of a compound in the manufacture of a medicament for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate). In some embodiments, the compound is administered orally. In some embodiments, the use is according to a method described herein.
  • In some embodiments, there is provided use of a compound in the manufacture of a medicament for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments, there is provided use of a compound in the manufacture of a medicament for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy. In some embodiments, the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof In some embodiments, the use is according to a method described herein.
  • In some embodiments, there is provided a compound for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof In some embodiments, the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate). In some embodiments, the compound is administered orally. In some embodiments, the treatment is according to a method described herein.
  • In some embodiments, there is provided a compound for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments, there is provided a compound for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy. In some embodiments, the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the treatment is according to a method described herein.
  • It is to be understood that one, some, or all of the properties of the various embodiments described herein may be combined to form other embodiments of the compositions and methods provided herein. These and other aspects of the compositions and methods provided herein will become apparent to one of skill in the art.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows decrease in palpable spleen size by cycle for patients treated with TG101348 680 mg/day (starting dose) (N=37). Doses for cycle 1 were 520-800 mg/day and doses for cycles 2-6 were 360-680 mg/day. For cycle 6≥50% subjects, there was 22-47% increase in 3 subjects with drug held for ˜2-3 weeks immediately prior to measurement.
  • FIG. 2 shows WBC count in subjects treated with TG101348. The baseline WBC count was >11×109/L. The doses at follow-up ranged from 360 to 680 mg/day. Last follow-up visit ranged from 8 to 24 weeks (median 24 weeks). “ULN” means upper limit of normal.
  • FIG. 3 shows platelet count in subjects treated with TG101348. The baseline platelet count >450×109/L. The doses at follow-up ranged from 360 to 680 mg/day. Last follow-up visit ranged from 12 to 24 weeks (median 24 weeks). “ULN” means upper limit of normal.
  • FIG. 4 shows the percentages of subjects with worsened, unchanged, improved or resolved constitutional symptoms (fatigue, early satiety, cough, night sweats, and pruritus) in subjects treated with TG101348. Last visit ranged from 4 to 24 weeks (median 20 weeks). The data here reflected changes from symptoms present at baseline. 18 subjects reported new onset of ≥1 symptom during the study; of these, symptoms for 12 subjects were resolved by last follow-up visit. Severity was rated by subjects on a scale of 1-10: 0=absent; 1-3=mild; 4-7=moderate; 8-10=severe. Improved=downgrade to absent or to mild or moderate from more severe rating at baseline.
  • FIG. 5 shows the cytokine levels (IL-6, IL-8, IL-2 and TNF-α) in subjects treated with TG101348. The values shown are median values.
  • FIG. 6 shows the change in V617F allele burden from baseline as a proportion of baseline in subjects with baseline >20% (N=22) treated with TG101348. The figure shows the subset of JAK2V617F positive subjects in the overall population (N=48). The doses at follow-up were 360 to 680 mg/day. Last follow-up visit ranged from 20 to 72 weeks (median 24 weeks).
  • FIG. 7 shows the bone marrow cellularity at baseline (60% cellularity) and after 18 cycles of TG101348 treatment (5-10% cellularity) in a 76-year-old male subject with V617F negative PMF. The starting dose was 30 mg/day and the dose at follow-up was 520 mg/day.
  • FIG. 8 shows the bone marrow fibrosis at baseline (3+) and after 18 cycles of TG101348 treatment (0) in a 56-year-old male subject with V617F negative PMF. The starting dose was 240 mg/day and the dose at follow-up was 440 mg/day.
  • FIG. 9 shows various measurements of a subject with JAK2 V617F-positive PMF treated with TG101348 (starting dose at 680 mg/day).
  • FIG. 10A-10G show distribution of TG101348 doses at the end of each cycle for subjects who initiated dosing at 30 mg/day, 60 mg/day, 120 mg/day, 240 mg/day, 360 mg/day, 520 mg/day, and 800 mg/day, respectively, (n=25).
  • FIG. 11 shows distribution of TG101348 doses at the end of each cycle for subjects who initiated dosing at 680 mg/day (n=34).
  • FIG. 12A shows plot of mean plasma TG101348 concentrations versus time on a semi-log scale (Cycle 1, Day 1). FIG. 12B shows plot of mean plasma TG101348 concentrations versus time on a semi-log scale (Cycle 1, Day 28).
  • FIG. 13 shows splenomegaly response to TG101348 therapy. This figure shows decrease in palpable spleen size from baseline by cycle for subjects in the maximum tolerated dose cohort (n=37). The proportion of subjects with ≥50% and 100% decrease in palpable splenomegaly is shown. For subjects who completed 6 cycles of treatment, 90% had a ≥25% reduction in palpable spleen size, 66% had a ≥50% reduction, and in 31% the spleen became non palpable.
  • FIGS. 14A-14C show effects of TG101348 on symptoms of myelofibrosis. (A): Proportion of subjects in maximum tolerated dose cohort with complete resolution of early satiety by cycle from a baseline symptom score of “mild” (score=1-3), “moderate” (score=4-7), or “severe” (score=8-10). Twenty-seven (79%) and 19 (56%) patients were evaluable for improvement in early satiety at the end of 1 and 6 cycles, respectively. After 2 cycles of treatment, 56% reported complete resolution of this symptom with durable benefit. (B): Proportion of subjects in maximum tolerated dose cohort with complete resolution of fatigue by cycle from a baseline symptom score of “mild” (score=1-3), or improvement in or complete resolution of fatigue from a baseline score of “moderate” (score=4-7) or “severe” (score=8-10). Twenty-four (71%) and 16 (47%) patients were evaluable for improvement in fatigue at the end of 1 and 6 cycles, respectively. After 6 cycles, 63% reported improvement and 25% had complete resolution of this symptom. (C): Proportion of subjects in maximum tolerated dose cohort with complete resolution of night sweats by cycle from a baseline symptom score of “mild” (score=1-3), “moderate” (score=4-7), or “severe” (score=8-10). Fourteen (40%) and 9 (26%) patients were evaluable for improvement in night sweats at the end of 1 and 6 cycles, respectively. After 1 cycle, 64% of subjects had complete resolution of this symptom; after 6 cycles, this proportion had increased to 89%.
  • FIG. 15 shows response of leukocytosis to TG101348 therapy. Changes in white blood cell (WBC) count after 6 cycles for subjects who entered the study with leukocytosis (WBC count >11×109/L). Following 6 cycles, 16 subjects across doses (57%) and 13 subjects in the MTD cohort (72%) achieved a normal WBC count, with durable benefit.
  • FIGS. 16A-16D show effect of TG101348 therapy on JAK2V617F allele burden. Box plot representation of JAK2V617F allele burden data for all mutation-positive subjects (n=51; FIGS. 16A and 16B) and for the subgroup with baseline allele burden >20% (n=23; FIGS. 16C and 16D). The y-axis represents the JAK2V617F allele burden from 1.0 (100%) to 0.0 (0%). The change in JAK2V617F allele burden per cycle of treatment (up to end of cycle 12; i.e. C13D1) as compared to pre-study baseline is shown for the 2 groups (FIGS. 16A and 16C); the change at the end of cycle 6 (i.e. C7D1) and cycle 12 is shown in FIGS. 16B and 16D. A significant decrease in JAK2V617F allele burden as compared to pre-study baseline was observed at the end of cycle 6 for the mutation-positive group (FIG. 16B; p=0.04) and the subgroup with baseline allele burden >20% (FIG. 16D; p=0.002); a similar significant decrease was seen at the end of cycle 12 for the former (FIG. 16B; p=0.01) and latter (FIG. 16D; p=0.002) groups. The Wilcoxon matched-pair signed-rank test was used to compare the median JAK2V617F allele burden for the comparisons.
  • FIGS. 17A-D shows absolute changes in pro-inflammatory cytokine levels from baseline at cycle 6: IL-6 (FIG. 17A), TNF-α (FIG. 17B), IL-8 (FIG. 17C), and IL-2 (FIG. 17D). Absolute differences in IL-6 (−4719 pg/mL) and IL-2 (−1827 pg/mL) are omitted from FIGS. 17A and 17D, respectively, for 1 subject (101-039) because they skewed presentation of data for other subjects.
  • FIG. 18 shows a plot of mean plasma TG101348 concentrations versus time on a linear plot after once daily oral doses (Cycle 1; Day 28).
  • DETAILED DESCRIPTION I. Definitions
  • As used herein, “treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired clinical results can include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals. In some embodiments, for the treatment of myelofibrosis, beneficial clinical results include one or more of reduction of splenomegaly, improvement in constitutional symptoms (such as early satiety, fatigue, night sweats, cough, and pruritus), reduction of leukocytosis, reduction of thrombocytosis, decrease of JAK2V617F allele burden, reduction of bone marrow fibrosis, and/or reduction of bone marrow cellularity.
  • As used herein, “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as myelofibrosis) or symptoms of the disease, and can include “progression free survival”. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease.
  • As used herein, an “effective dosage” or “effective amount” of drug, compound, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results can include, for example, one or more results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results can include, include, for example one or more clinical results such as decreasing one or more symptoms and pathological conditions resulting from or associated with the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival. In the case of myelofibrosis, an effective amount of a drug may have the effect in reducing one or more of splenomegaly, improving constitutional symptoms (such as early satiety, fatigue, night sweats, cough, and pruritus), reducing leukocytosis, reducing thrombocytosis, decreasing JAK2V617F allele burden, reducing bone marrow fibrosis, and/or reducing bone marrow cellularity. An effective dosage can be administered in one or more administrations. An effective dosage of drug, compound, or pharmaceutical composition can be, for example, an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. As is understood in the clinical context, an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • As used herein, “ameliorating” bone marrow cellularity or bone marrow fibrosis refers to reducing the level of bone marrow cellularity or bone marrow fibrosis in a subject compared to the level of bone marrow cellularity or bone marrow fibrosis prior to commencing treatment with the compound provided herein. The reduction of bone marrow cellularity or bone marrow fibrosis can be at least by 5, 10, 20, 30, 40, 50, 60, 70, 80, or 90%.
  • As used herein, “in conjunction with” refers to administration of one treatment modality in addition to another treatment modality. As such, “in conjunction with” can refer to administration of one treatment modality before, during or after administration of the other treatment modality to the individual.
  • As used herein, a “patient” or a “subject” refers to a mammal including a human, a dog, a horse, a cow or a cat, etc.
  • The term “pharmaceutically acceptable” refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and can be administered to a subject.
  • As used herein, “pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof
  • As used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly indicates otherwise.
  • Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X.”
  • It is understood that aspects and variations of the compositions and methods provided herein can include “consisting” and/or “consisting essentially of” aspects and variations.
  • II. Compounds and Pharmaceutical Compositions
  • Provided herein is a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. Also provided herein are pharmaceutical compositions comprising N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and a pharmaceutically acceptable excipient or carrier. The compound and the pharmaceutical compositions described herein can be used for treating or delaying development of myelofibrosis in a subject. N-tert-Butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide has the following chemical structure:
  • Figure US20210244735A1-20210812-C00001
  • The compound provided herein may be formulated into therapeutic compositions as natural or salt forms. Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, or ferric hydroxide, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino-ethanol, histidine, procaine, and the like. Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric acid, sulfuric acid, or phosphoric acid, or organic acids such as acetic acid, citric acid, p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, tartaric acid, mandelic acid, and the like.
  • Salts of the compounds provided herein can include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the compounds provided herein can also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, methanesulfonic acid and the like. Additional excipients which are contemplated for use in the practice of the compositions and methods provided herein are those available to those of ordinary skills in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, Md. (1989), the relevant contents of which are incorporated herein by reference.
  • In addition, the compounds provided herein can include polymorphs. The compound described herein may be in alternative forms. For example, the compound described herein may include a hydrate form. As used herein, “hydrate” refers to a compound provided herein which is associated with water in the molecular form, i.e., in which the H—OH bond is not split, and may be represented, for example, by the formula R.H2O, where R is a compound provided herein. A given compound may form more than one hydrate including, for example, monohydrates (R.H2O) or polyhydrates (R.nH2O wherein n is an integer greater than 1) including, for example, dihydrates (R.2H2O), trihydrates (R.3H2O), and the like, or fractional hydrates, such as, for example, R.n/2H2O, R.n/3H2O, R.n/4H2O and the like wherein n is an integer.
  • The compounds described herein may also include acid salt hydrate forms. As used herein, “acid salt hydrate” refers to a complex that may be formed through association of a compound having one or more base moieties with at least one compound having one or more acid moieties or through association of a compound having one or more acid moieties with at least one compound having one or more base moieties, said complex being further associated with water molecules so as to form a hydrate, wherein said hydrate is as previously defined and R represents the complex herein described above.
  • In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate and has the following chemical structure:
  • Figure US20210244735A1-20210812-C00002
  • The pharmaceutical compositions for the administration of the compound described herein, either alone or in combination with other therapeutic agents, may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy and methods described in Examples 4, 5 and 6. Such methods can include bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as hard or soft capsules. The suitable capsule shell may be hard gelatin or hydroxypropylmethyl cellulose (“HPMC”).
  • Provided herein are formulations comprising (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) one or more excipients, and (iii) one or more lubricants. The formulations may be in capsule form and administered orally. The formulations may be in unit dosage form. In some embodiments, the excipient is lactose (such as Fast-Flo), mannitol (such as Parteck M200), microcrystalline cellulose (“MCC”) (such as Avicel PH102), MCC (such as ProSolv 90 HD). In some embodiments, the lubricant is magnesium stearate, sodium stearyl fumarate (such as Pruv), or sodium laurel fumarate. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the capsule is hard gelatin capsule.
  • In some embodiments, there is provided a capsule suitable for oral administration comprising an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate), wherein the admixture is contained in the capsule. Methods known in the art and described herein may be used for making the capsules. See, e.g., Example 3. Microcrystalline cellulose may be used as a filler and/or diluent in the capsules provided herein. Sodium stearyl fumarate may be used as a lubricant in the capsules provided herein. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose. For example, silicified microcrystalline cellulose may be composed of microcrystalline cellulose and colloidal silicon dioxide particles. In some embodiments, the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • In some embodiments, the capsule contains about 10 mg to about 680 mg of the compound, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 10 mg to about 650 mg (or about 10 mg to about 550 mg or about 10 mg to about 500 mg), wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 100 mg to about 600 mg (or about 200 mg to about 550 mg or about 300 mg to about 500 mg), wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule contains about 10 mg, about 20 mg, about 40 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, or about 650 mg of the compound, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the capsule is a hard gelatin capsule. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • In some embodiments, the weight ratio of the compound to excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in the capsule is between about 1:1.5 to about 1:15 (e.g., between about 1:5 to about 1:10, between about 1:5 to about 1:12, or between about 1:10 to about 1:15), wherein the weight of the compound is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to lubricant (e.g., sodium stearyl fumarate) in the capsule is between about 5:1 to about 50:1 (e.g., between about 5:1 to about 10:1, between about 5:1 to about 25:1, between about 5:1 to about 40:1, between about 7:1 to about 34:1, or between about 8:1 to about 34:1), wherein the weight of the compound is the free base moiety weight of the compound.
  • In some embodiments, the capsule contains about 5% to about 50% (e.g., about 5% to about 10% or about 5% to about 35%) compound of the total fill weight of the capsule, wherein the weight of the compound is the free base moiety weight of the compound. In some embodiments, the capsule contains about 40% to about 95% (e.g., about 50% to about 90% or about 60% to about 90%) excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) of the total fill weight of the capsule. In some embodiments, the capsule contains about 0.2% to about 5% (e.g., about 0.2% to about 2% or about 0.5% to about 1.5%, or about 0.5%, about 1%, or about 1.5%) lubricant (e.g., sodium stearyl fumarate) of the total fill weight of the capsule.
  • Also provided herein are unit dosage forms comprising an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) an excipient (such as microcrystalline cellulose), and (iii) a lubricant (such as sodium stearyl fumarate). Any one of the capsules described herein may be used in a unit dosage form. In some embodiments, the unit dosage form is for treating myelofibrosis. In some embodiments, the treatment is according to a method described herein.
  • In some embodiments, the unit dosage form comprises an admixture of (i) about 10 mg to about 680 mg (or about 10 mg to about 500 mg) of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the specified weight is the free base moiety weight of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate. In some embodiments, the compound in the admixture is about 10 mg to about 500 mg, wherein the specified weight is the free base moiety weight of the compound.
  • In some embodiments, the unit dosage form is in the form of a capsule, and the admixture is contained in the capsule. In some embodiments, the unit dosage form comprises about 10 mg, about 20 mg, about 40 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, or about 650 mg of the compound, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the admixture comprises (i) about 10 mg (or about any of 40 mg, 100 mg, 200 mg, 300 mg, 400 mg, or 500 mg) of the compound, (ii) a microcrystalline cellulose, and (iii) sodium stearyl fumarate, wherein the specified weight is the free base moiety weight of the compound. In some embodiments, the compound is N-tert-butyl-3-[(5 -methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • In some embodiments, the weight ratio of the compound to excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in the unit dosage form is between about 1:1.5 to about 1:15 (e.g., between about 1:5 to about 1:10, between about 1:5 to about 1:12, or between about 1:10 to about 1:15), wherein the weight of the compound is the free base moiety weight of the compound. In some embodiments, the weight ratio of the compound to lubricant (e.g., sodium stearyl fumarate) in the unit dosage form is between about 5:1 to about 50:1 (e.g., between about 5:1 to about 10:1, between about 5:1 to about 25:1, between about 5:1 to about 40:1, between about 7:1 to about 34:1, or between about 8:1 to about 34:1), wherein the weight of the compound is the free base moiety weight of the compound. In some embodiments, the microcrystalline cellulose is silicified microcrystalline cellulose. In some embodiments, the silicified microcrystalline cellulose is a combination of 98% microcrystalline cellulose and 2% colloidal silicon dioxide.
  • In some embodiments, the lubricant (e.g., sodium stearyl fumarate) is about 0.1% to about 10%, about 0.5% to about 5%, about 0.5% to about 3%, about 0.5% to about 2%, about 0.75% to about 1.5% of the capsule fill weight. In some embodiments, the lubricant (e.g., sodium stearyl fumarate) is at least about any one of 0.1%, 0.25%, 0.5%, 0.75%, 1%, 1.25%, 1.5%, 1.75%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, or 5% of the capsule fill weight. In some embodiments, the lubricant (e.g., sodium stearyl fumarate) is about any one of 0.1%, 0.25%, 0.5%, 0.75%, 1%, 1.25%, 1.5%, 1.75%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, or 5% of the capsule fill weight.
  • In some embodiments, the weight ratio of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in a capsule or unit dosage form is about 40:60 to about 10:90. In some embodiments, the weight ratio of the compound to an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose) in a capsule of unit dosage form is about any one of 95:5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, or 5:95. In some embodiments, the weight ratio of the compound to an excipient (e.g., microcrystalline such as silicified microcrystalline cellulose) is about 1:1.5 to about 1:9.5, such as about any of 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5, 1:5, 1:5.5, 1:6, 1:6.5, 1:7, 1:7.5, 1:8, 1:8.5, 1:9, or 1:9.5. In some embodiments, the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
  • In some embodiments, the capsule contains about 5% to about 50% (e.g., about 5% to about 10% or about 5% to about 35%) compound of the total weight of the admixture, wherein the weight of the compound is the free base moiety weight of the compound. In some embodiments, the capsule contains about 40% to about 95% (e.g., about 50% to about 90% or about 60% to about 90%) microcrystalline cellulose (such as silicified microcrystalline cellulose) of the total weight of the admixture. In some embodiments, the capsule contains about 0.2% to about 5% (e.g., about 0.2% to about 2% or about 0.5% to about 1.5%, or about 0.5%, about 1%, or about 1.5%) sodium stearyl fumarate of the total weight of the admixture.
  • In some embodiments, the capsule or unit dosage form contains an admixture of about 12 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 122 mg of silicified microcrystalline cellulose, and about 1 mg of sodium stearyl fumarate. In some embodiments, the capsule or unit dosage form contains an admixture of about 47 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 448 mg of silicified microcrystalline cellulose, and about 5 mg of sodium stearyl fumarate. In some embodiments, the capsule or unit dosage form contains an admixture of about 117 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. In some embodiments, the capsule or unit dosage form contains an admixture of about 235 mg of N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate, about 357 mg of silicified microcrystalline cellulose, and about 6.00 mg of sodium stearyl fumarate.
  • Also provided herein are oral solution formulations comprising a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the oral solution formulation further comprises methylcellulose. In some embodiments, the oral solution formulation further comprises methylcellulose and Tween 80. In some embodiments, the oral solution formulation comprises the compound at about 1 mg/ml to about 25 mg/ml, about 2 mg/ml to about 20 mg/ml, about 3 mg/ml to about 15 mg/ml, about 5 mg/ml to about 10 mg/ml. In some embodiments, the oral solution formulation comprises the compound at about any one of 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 6 mg/ml, 6.25 mg/ml, 6.5 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, or 12.5 mg/ml, or 15 mg/ml. In some embodiments, the oral solution formulation comprises about 0.1% to about 5%, 0.2% to about 3%, about 0.25% to about 2%, about 0.25% to about 1%, or about 0.5% by weight of methylcellulose. In some embodiments, the oral solution formulation comprises about 0.01% to about 0.5%, 0.02% to about 0.3%, about 0.025% to about 0.2%, about 0.025% to about 0.1%, or about 0.05% by weight of Tween 80.
  • In some embodiments, the capsule does not comprise an absorption enhancer. In some embodiments, the capsule comprises an absorption enhancer (e.g., Vitamin E TPGS, Gelucire 44/14, Pluronic F127, or glyceryl monostearate).
  • A capsule or unit dosage form provided may comprise one or more of the following properties: (1) upon administration to a subject such as human subject, the Cmax of the compound is achieved within about 2 to about 4 hours post-dose; (2) upon administration to a human subject, the elimination half life of the compound is about 16 to about 34 hours; (3) the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day; (4) the accumulation of the compound is about 1.25 to about 4.0 fold at steady state when the compound is dosed once daily.
  • Also provided are methods of preparing a capsule drug product comprising a) blending a lubricant with a compound that is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to generate granules and b) mixing the granules of a) with an excipient. In some embodiments, the lubricant is sodium stearyl fumarate. In some embodiments, the excipient is microcrystalline cellulose such as silicified microcrystalline cellulose. Such method may be used to prepare a capsule or unit dosage form described herein. The weight (such as weight ratio or weight percentage) and components regarding the compound, excipient, and/or lubricant may be according to any described herein.
  • III. Methods of Treatment and Prevention of Myelofibrosis
  • Provided herein are methods for treating, delaying development, and/or preventing myelofibrosis in a subject comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof (e.g., N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate). In some embodiments, the subject has myelofibrosis. In some embodiments, the subject is at risk of developing myelofibrosis. In some embodiments, the subject is a human subject. Any one of the formulations described herein such as capsule or unit dosage forms described herein may be used to treat a subject with myelofibrosis. In some embodiments, the compound is in an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) an excipient (such as microcrystalline cellulose), and (iii) a lubricant (such as sodium stearyl fumarate).
  • Myelofibrosis that may be treated by the compounds described herein includes primary myelofibrosis (MF) and secondary myelofibrosis (e.g., myelofibrosis arising from antecedent polycythemia vera (post-PV MF) or essential thrombocythemia (post-ET MF)). Myelofibrosis that may be treated by the compounds described herein also includes myelofibrosis of high risk, intermediate risk such as intermediate risk level 2. Methods for diagnosing various types of myelofibrosis are known in the art. See, e.g., Cervantes et al., Blood 2009. In some embodiments, the subject with myelofibrosis has spleen of at least 5cm below costal margin as measured by palpation.
  • In some embodiments, the subject has a point mutation from valine 617 to phenylalanie in the Janus kinase 2 (JAK2 kinase) (JAK2V617F) if the subject is a human, or a point mutation corresponding to the valine 617 to phenylalanie in the Janus kinase 2 (JAK2 kinase) if the subject is not a human. In some embodiments, the subject is negative for the valine 617 to phenylalanine mutation of JAK2 if the subject is a human, or negative for a mutation corresponding to the valine 617 to phenylalanie in the Janus kinase 2 (JAK2 kinase) if the subject is not a human Whether a subject is positive or negative for JAK2V617F can be determined by a polymerase chain reaction (“PCR”) analysis using genomic DNA from bone marrow cells or blood cells (e.g., whole blood leukocytes). The PCR analysis can be an allele-specific PCR (e.g., allele-specific quantitative PCR) or PCR sequencing. See Kittur J et al., Cancer 2007, 109(11):2279-84 and McLornan D et al., Ulster Med J. 2006, 75(2):112-9, each of which is expressly incorporated herein by reference.
  • In some embodiments, the subject treated with the methods described herein has previously received another myelofibrosis therapy or treatment. In some embodiments, the subject is a non-responder to the other myelofibrosis therapy or has a relapse after receiving the other myelofibrosis therapy. The previous therapy may be a JAK2 inhibitor (e.g. INCB018424 (also known as ruxolitinib, available from Incyte), CEP-701 (lestaurtinib, available from Cephalon), or XL019 (available from Exelixis)) (See Verstovsek S., Hematology Am Soc Hematol Educ Program. 2009:636-42) or a non-JAK2 inhibitor (such as hydroxyurea). In some embodiments, the subject has received ruxolitinib treatment for primary myelofibrosis, post-polycythemia vera myelofibrosis (Post-PV MF), post-essential thrombocythemia myelofibrosis (Post-ET MF), polycythemia vera, or essential thrombocythemia for at least 14 days and discontinued the treatment for at least 30 days. In some embodiments, the previous therapy is a treatment with a compound described herein and the previous therapy has been discontinued upon indication of one or more elevated levels of amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and/or creatinine in the serum from the subject, and/or upon indication of a hematologic condition selected from the group consisting of anemia, thrombocytopenia, and neutropenia. In some embodiments, the dose of the compound in the second treatment is the same or lower than the dose in the previous therapy.
  • The subject may be treated orally and/or daily. The subject (such as a human) may be treated by administering at a dose of about 240 mg per day to about 680 mg per day (or about 300 mg per day to about 500 mg per day), wherein the specified weight is the free base moiety weight of the compound. In some embodiment, the compound is administered at a dose of about any of 240 mg/day, 250 mg/day, 300 mg/day, 350 my/day, 400 mg/day, 450 mg/day, 500 mg/day, 550 mg/day, 600 mg/day, 650 mg/day, or 680 mg/day. The compound may be in a capsule and/or a unit dosage form described herein. In some embodiments, the compound administered is in an admixture with a microcrystalline cellulose and sodium stearyl fumarate, and the admixture is in a capsule. In some embodiments, the compound is administered orally.
  • Also provided herein are methods for ameliorating one or more symptoms associated with myelofibrosis. For example, the treatment using the compound described herein is effective in reducing spleen size, ameliorating constitutional symptoms (such as early satiety, fatigue, night sweats, cough, and pruritus), reducing leukocytosis, reducing thrombocytosis, decreasing JAK2V617F allele burden, reducing bone marrow fibrosis, improving pruritus, improving cachexia, and/or reducing bone marrow cellularity. The reduction, decrease, amelioration, or improvement can be at least by 5, 10, 20, 30, 40, 50, 60, 70, 80, or 90% compared to the level prior to commencing treatment with the compound provided herein. In some embodiment, the spleen becomes non-palpable in the subject after treatment. In some embodiments, the subject has complete resolution of leukocytosis and/or thrombocytosis after treatment. In some embodiments, the subject has complete resolution of pruritus after treatment.
  • In some embodiments, the compound is administered to the subject daily for at least 1 cycle, at least 2 cycles, at least 3 cycles, at least 4 cycles, at least 5 cycles, or at least 6 cycles of a 28-day cycle. In some embodiments, the compound is administered to the subject daily for at least 6 cycles of a 28-day cycle, at least 8 cycles of a 28-day cycle, at least 10 cycles of a 28-day cycle, at least 12 cycles of a 28-day cycle, at least 15 cycles of a 28-day cycle, at least 18 cycles of a 28-day cycle, or at least 24 cycles of a 28-day cycle. In some embodiments, the compound is administered to the subject daily for at least one month, at least two month, at least three month, at least four month, at least five month, at least six month, at least eight month, or at least one year. In some embodiments, the compound is administered once a day.
  • In some embodiments, upon administration of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof to a subject such as human subject, the Cmax of the compound is achieved within about 1 to about 5 hours, about 1.5 to about 4.5 hours, about 2 to about 4 hours, or about 2.5 to about 3.5 hours post-dose. In some embodiments, upon administration of the compound to a human subject, the elimination half life of the compound is about 12 to about 40 hours, about 16 to about 34 hours, or about 20 to about 30 hours. In some embodiments, the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the accumulation of the compound is about 1.1 to about 5 fold, about 1.25 to about 4.0 fold, about 1.5 to about 3.5 fold, about 2 to about 3 fold at steady state when the compound is dosed once daily.
  • In some embodiments, the method comprises instructing the subject to ingest the effective amount of the compound on an empty stomach. In some embodiments, the methods further comprise instructing the subject to avoid ingesting agents that are at least moderate inducers or inhibitors of CYP3A4. In some embodiments, the subject does not receive concomitant treatment with or use of drugs to herbal agents known to be at least moderate inhibitors or inducers of CYP3A4. Based on in vitro evaluations, N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide is metabolized by human CYP3A4. Agents that may increase N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide plasma concentrations (i.e., CYP3A4 inhibitors) or decrease N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino-]benzenesulfonamide plasma concentrations (i.e., CYP3A4 inducers), including herbal agents and foods (e.g. grapefruit/grapefruit juice), should be avoided in subjects being treated as described herein. In addition, in vitro data have indicated that N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide inhibits CYP3A4 in a time-dependent fashion. Agents that are sensitive substrates for metabolism by CYP3A4 should be used with caution as coadministration with N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide may result in higher plasma concentrations of the coadministered agent. A list of clinically relevant substrates of CYP3A4 include alfentanil, Cyclosporine, Diergotamine, ethinyl estradiol, ergotamine, fentanyl, pimozide, quinidine, sirolimus, tacrolimus, clarithromycin erythromycin, telithromycin, alprazolam, diazepam, midazolam, triazolam, indinavir, ritonavir, saquinavir, prokinetic, cisapride, astemizole, chlorpheniramine, amlodipine, diltiazem, felodipine, nifedipine, verapamil, atorvastatin, cerivastatin, lovastatin, simvastatin, aripiprazole, gleevec, halopericol, sildenafil, tamoxifen, taxanes, trazodone, and Vincristine. A list of clinically relevant inducers of CYP3A4 include carbamazepine, phenobarbital, phenytoin, pioglitazone, rifabutin, rifampin, St. John's wort, and troglitazone. A list of clinically relevant inhibitors of CYP3A4 include indinavir, nelfinavir, ritonavir, clarithromycin, itraconazole, ketoconazole, nefazodone, erythromycin, grapefruit juice, verapamil, diltiazem, cimetidine, amiodarone, fluvoxamine, mibefradil, and Troleandomycin. See reference Flockhart et al., http://medicine.iupui.edu/clinpharm/ddis/clinicaltable.aspx., 2009.
  • Also provided herein are methods of monitoring treatment of myelofibrosis to a subject, comprising (a) administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof; (b) monitoring a hematologic parameter and/or a non-hematologic parameter in the subject; and (c) determining if the subject should continue or discontinue with the treatment. In some embodiments, the hematologic parameter is selected from the group consisting of anemia, thrombocytopenia, and neutropenia. In some embodiments, the non-hematologic parameter is an enzyme or molecule in the blood or serum wherein an elevated level of the enzyme or molecule is indicative of tissue or organ damage. In some embodiments, the serum enzyme or molecule can be, for example, amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, alkaline phosphatase, and calcium. Methods of monitoring these parameters are known in the art and are described herein. See Examples 1-3. In some embodiments, the method further comprises administering to the subject an effective amount of the compound described herein after the subject has been discontinued with the treatment for at least 2 week, at least 3 weeks, or at least 4 weeks. In some embodiments, the previous treatment has been discontinued without prior dose reduction.
  • Also provided herein are methods of monitoring treatment of myelofibrosis to a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and discontinuing the treatment upon indication of elevated levels of one or more enzymes or molecules selected from the group consisting of amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatinine and/or decreased level of calcium in the blood or serum of the subject without prior dose reduction. Also provided herein are methods of monitoring treatment of myelofibrosis to a subject, comprising administering to the subject an effective amount of a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, and discontinuing the treatment upon indication of one or more hematologic conditions selected from the group consisting of anemia, thrombocytopenia, and neutropenia without prior dose reduction. In some embodiments, the treatment is discontinued when one or more of the parameters (including hematologic and non-hematologic parameters) are grade 3 or 4 events.
  • Grade 3 or 4 adverse events for hematologic and non-hematologic parameters are known in the art and shown in the Table below. See, e.g. C Common Terminology Criteria for Adverse Events (CTCAE), Version 4.0, Published: May 28, 2009 (v4.03: Jun. 14, 2010).
  • RESPONSE
    (hematologic and
    non-hematologic) Definition Grade 3 Grade 4
    Hyperlipasemia A finding based on >2.0-5.0 × ULN* >5.0 × ULN
    laboratory test results that
    indicate an increase in the
    level of lipase in a
    biological specimen.
    Serum amylase A finding based on >2.0-5.0 × ULN >5.0 × ULN
    laboratory test results that
    indicate an increase in the
    levels of amylase in a
    serum specimen.
    Alanine A finding based on >5.0-20.0 × ULN >20.0 × ULN
    aminotransferase laboratory test results that
    increased indicate an increase in the
    level of alanine
    aminotransferase (ALT or
    SGPT) in the blood
    specimen.
    Aspartate A finding based on >5.0-20.0 × ULN >20.0 × ULN
    aminotransferase laboratory test results that
    increased indicate an increase in the
    level of aspartate
    aminotransferase (AST or
    SGOT) in a blood
    specimen.
    Blood creatinine A finding based on >3.0 baseline; >3.0 6.0 × ULN
    increased laboratory test results that >6.0 × ULN
    indicate increased levels
    of creatinine in a
    biological specimen.
    Blood alkaline A finding based on >5.0-20.0 × ULN >20.0 × ULN
    phosphatase increased laboratory test results that
    indicate an increase in the
    level of alkaline
    phosphatase in a blood
    specimen.
    Hypocalcemia A disorder characterized Corrected serum Corrected serum
    by laboratory test results calcium of calcium of
    that indicate a low <7.0-6.0 mg/dL; <6.0 mg/dL;
    concentration of calcium <1.75-1.5 mmol/L; <1.5 mmol/L;
    (corrected for albumin) in Ionized calcium Ionized calcium
    the blood. <0.9-0.8 mmol/L; <0.8 mmol/L;
    hospitalization life-threatening
    indicated consequences
    Anemia A disorder characterized Hgb <8.0 g/dL; Life-threatening
    by a reduction in the <4.9 mmol/L; consequences;
    amount of hemoglobin in <80 g/L; transfusion urgent intervention
    100 ml of blood. Signs indicated indicated
    and symptoms of anemia
    may include pallor of the
    skin and mucous
    membranes, shortness of
    breath, palpitations of the
    heart, soft systolic
    murmurs, lethargy, and
    fatigability.
    Thrombocytopenia a platelet count below the 25,000 to <50,000/μL below 25,000/μL
    normal range for the
    population ([+ or −] 2
    standard deviations). In
    most laboratories, a
    normal platelet count is
    between 150,000 to
    450,000/μL
    Neutropenia A finding based on <1000-500/mm3; <500/mm3;
    laboratory test results that <1.0-0.5 × 109/L <0.5 × 109/L
    indicate a decrease in
    number of neutrophils in a
    blood specimen.
    *“ULN” refers to upper limit of normal.
  • IV. Articles of Manufactures and Kits
  • Also provided herein are articles of manufacture or kits containing a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the article of manufacture or the kit further includes instructions for using the compounds described herein in the methods provided herein. In some embodiments, the article of manufacture or the kit further comprises a label or a package insert providing the instructions. In some embodiments, the compound is in a capsule and/or a unit dosage form described herein.
  • In some embodiments, the article of manufacture or kit may further comprise a container. Suitable containers include, for example, bottles, vials (e.g., dual chamber vials), syringes (such as single or dual chamber syringes) and test tubes. The container may be formed from a variety of materials such as glass or plastic, and the container may hold the compound, for example in the formulation to be administered. The article of manufacture or the kit may further comprise a label or a package insert, which is on or associated with the container, may indicate directions for reconstitution and/or use of the compound. In some embodiments, the package insert or the label is in a position which is visible to prospective purchasers.
  • The label or package insert may further indicate that the compound is useful or intended for treating or preventing myelofibrosis in a subject. In some embodiments, the package insert or the label indicates that the compound can be used for ameliorating bone marrow cellularity and/or bone marrow fibrosis. In some embodiments, the package insert or the label indicates that the compound can be used for treating myelofibrosis in a subject, wherein the subject is negative for the valine 617 to phenylalanine mutation of human JAK2 (JAK2V617F) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments, the package insert or the label indicates that the compound can be used for treating myelofibrosis in a subject, and that subject should discontinue the treatment upon indication of elevated levels of one or more of amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, and/or alkaline phosphatase and/or decreased level of calcium in the serum of the subject, and/or upon indication of one or more of anemia, thrombocytopenia, and/or neutropenia. In some embodiments, the package insert or the label further indicates that the compound can be discontinued without prior dose reduction.
  • In some embodiments, there is provided a kit or article of manufacture comprising (a) an admixture of (i) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, (ii) excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) lubricant (e.g., sodium stearyl fumarate), and (b) a package insert or a label indicating that the admixture is useful for treating myelofibrosis in a subject. In some embodiments, there is provided a kit or article of manufacture comprising (a) a compound which is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutical salt thereof or a hydrate thereof, and (b) a package insert or a label indicating that the compound can be used for treating myelofibrosis in a subject, wherein the subject has previously received another myelofibrosis therapy with a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • In some embodiments, the package insert or a label indicates that, upon administration of the compound to a human subject, the Cmax of the compound is achieved within about 1 to about 5 hours, about 1.5 to about 4.5 hours, about 2 to about 4 hours, or about 2.5 to about 3.5 hours post-dose. In some embodiments, the package insert or a label indicates that, upon administration of the compound to a human subject, the elimination half life of the compound is about 12 to about 40 hours, about 16 to about 34 hours, or about 20 to about 30 hours. In some embodiments, the mean AUC of the compound increases more than proportionally with increasing doses ranging from about 30 mg to about 800 mg per day. In some embodiments, the accumulation of the compound is about 1.1 to about 5 fold, about 1.25 to about 4.0 fold, about 1.5 to about 3.5 fold, about 2 to about 3 fold at steady state when the compound is dosed once daily.
  • In some embodiments, the package insert or the label instructs the subject to ingest the effective amount of the compound on an empty stomach. In some embodiments, the package insert or the label instructs the subject to avoid ingesting agents that are at least moderate inducers or inhibitors of CYP3A4. In some embodiments, the inducer or inhibitor of CYP3A4 is any one of the inducers or inhibitors of CYP3A4 described herein.
  • Also provided are uses of a compound in the manufacture of a medicament for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the use is according to a method described herein. In some embodiments, the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate). In some embodiments, the compound is administered orally. In some embodiments, the use is according to a method described herein. In some embodiments, there is provided use of a compound in the manufacture of a medicament for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments, there is provided use of a compound in the manufacture of a medicament for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy. In some embodiments, the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • Also provided is a compound for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof. In some embodiments, the treatment is according to a method described herein. In some embodiments, the compound is in an admixture of (i) the compound, (ii) an excipient (e.g., microcrystalline cellulose such as silicified microcrystalline cellulose), and (iii) a lubricant (e.g., sodium stearyl fumarate). In some embodiments, the compound is administered orally. In some embodiments, the treatment is according to a method described herein. In some embodiments, there is provided a compound for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject is negative for the valine 617 to phenylalanine mutation of human Janus Kinase 2 (JAK2) or negative for the mutation corresponding to the valine 617 to phenylalanine mutation of human JAK2. In some embodiments, there is provided a compound for treating myelofibrosis in a subject, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof, wherein the subject has previously received another myelofibrosis therapy. In some embodiments, the previous therapy comprises a JAK2 inhibitor which is not N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt thereof or a hydrate thereof.
  • The following are examples of the methods and compositions provided herein. It is understood that various other embodiments may be practiced, given the general description provided above.
  • EXAMPLES Example 1 Evaluation of TG101348 in Myelofibrosis
  • As used herein, “TG101348” refers to N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate. The subjects in this study were administered with capsule form of TG101348 as described in Example 5. TG101348 was evaluated in a Phase I study for the treatment of myelofibrosis. This study was ongoing at the time the data were collected.
  • Background: TG101348 is a potent, orally bioavailable, JAK2-selective small molecule inhibitor, that was evaluated in a Phase I study for the treatment of myelofibrosis. The dose-limiting toxicity was asymptomatic grade 3 or 4 amylasemia/lipasemia that was reversible, and the maximum tolerated dose (“MTD”) was 680 mg. The most frequent non-hematological toxicities were mild nausea, vomiting, and/or diarrhea that were easily controlled or resolved spontaneously. Grade 3/4 neutropenia and thrombocytopenia were observed in 14% and 25% of patients, respectively. TG101348 had activity in reducing spleen size, leukocyte count, and JAK2V617F (“VF”) allele burden. This example describes the results with a focus on the data from the dose confirmation cohort who initiated treatment at a dose of 680 mg/day.
  • Results: Fifty nine patients (median age=66 years; range 43-86) were treated −28 in the dose escalation phase, and 31 in the dose confirmation phase. Overall, 44 patients had PMF, 12 post-PV MF, and 3 post-ET MF; 86% were VF-positive. Median palpable spleen size was 18 cm and 22 patients were red blood cell (“RBC”) transfusion-requiring at study enrollment. After a median follow-up of 12 weeks (range <1-76), 18 (31%) patients discontinued treatment due to toxicity (n=7; thrombocytopenia=3, neutropenia=1), comorbidities (n=5), withdrawal of consent (n=4), or non-compliance/lack of response (1 each). The remaining 41 patients were at the following dose levels when the data in this example were collected: 680 mg (n=14), 520-600 mg (n=16), 360-440 mg (n=10), and 240 mg (n=1). The cumulative drug exposure to the time when the data in this example were collected was 362 patient-months; exposure at or above MTD (≥680 mg) was 154 patient-months. Forty patients (68%) started treatment at ≥680 mg.
  • Toxicity: TG101348 was well tolerated. Of the patients who started at ≥680 mg, Gr3/4 neutropenia was observed in 15/0% and Gr3/4 thrombocytopenia in 20/10%. Twenty four (60%) patients did not require RBC transfusions at baseline (median hemoglobin (“Hgb”)=9.6g/dL; range 7.4-13.1); of these, 42% and 8% of patients developed Grade 3 (“Gr3”) and Grade 4 (“Gr4”) anemia, respectively. The majority of patients who started at ≥680 mg developed mild nausea (1 Gr3), vomiting (1 Gr3), and/or diarrhea (3 Gr3) that were self-limited or easily controlled. Other non-hematological toxicities included Grade 1/2 (“Gr1/2”) transaminitis (38%), Gr1/2 serum creatinine elevation (38%), and asymptomatic hyperlipasemia (33%).
  • Efficacy: Thirty three patients who started at >680 mg completed at least 3 cycles of treatment; at 3 months, reduction in palpable spleen size (baseline median=18 cms; range 6-32) was at least 50% in 22 (67%) patients; the spleen became non-palpable in 9 (27%) patients. All 21 patients with leukocytosis at baseline (WBC range 11 to 203×109/L) who started at ≥680 mg experienced a marked reduction in their WBC count (range 4 to 90); 70% had a normal WBC count at their last follow-up visit. Overall, 48 of the 51 VF-positive patients completed at least 1 cycle and were evaluable for response in VF allele burden; at last available follow-up, the median decrease in granulocyte mutant allele burden was 48%; 21 (44%) patients had a ≥50% reduction, and in the group who started treatment at ≥680 mg, 48% have had a ≥50% reduction. Of those evaluable, there was clinically significant benefit or resolution of constitutional symptoms, including early satiety, fatigue, cough, pruritus, and night sweats.
  • Conclusions: TG101348 was well tolerated in patients with myelofibrosis. Spleen and leukocyte responses were frequent, observed early, and produced substantial clinical benefit for patients. These responses were associated with significant decrease in VF allele burden and pointed to activity of TG101348 against the malignant clone in myelofibrosis.
  • Example 2 Evaluation of TG101348 in Myelofibrosis
  • The subjects in this study were administered with capsule form of TG101348. TG101348 was evaluated in a Phase I study for the treatment of myelofibrosis. This study is also described in Example 1. This example describes data available at the time of data collection.
  • This study was an open-label, multicenter, and dose-escalation study with expanded cohort dose confirmation at MTD. The primary objective of this study was to determine safety/tolerability, DLT, MTD, and pharmacokinetics of TG101348 in subjects with MF. The secondary objective of this study was to evaluate preliminary clinical and pharmacodynamic activity.
  • The key eligibility criteria for subjects included: Myelofibrosis (PMF or post-PV/ET MF); High-risk or intermediate-risk with symptomatic splenomegaly/unresponsive to available therapy; ECOG performance status ≤2; ANC≥1×109/L; Platelet count≥50×109/L; Serum creatinine ≤2 mg/dL; Total bilirubin ≤2 mg/dL; AST/ALT≤3X upper limit of normal.
  • The subject disposition for this study is included in Table 1.
  • TABLE 1
    Subject Disposition
    MTD* Overall
    Enrolled 40 59
    Included in safety analysis 40 59
    Included in drug activity analysis 37 55
    Discontinued 11 (28%) 15 (25%)
    Reasons for discontinuation
    Adverse event 5 6
    Subject withdrew consent 4 6
    Investigator discretion 2 3
    Median (range) treatment 24 weeks 24 weeks
    duration (1-24 weeks) (0.3-84 weeks)**
    *Includes all subjects who initiated treatment at 680 or 800 mg/day.
    **Includes continued treatment in extension study.
  • The demographic and baseline characteristics for the subjects are included in Table 2.
  • TABLE 2
    Demographic and Baseline Characteristics
    MTD Overall
    (n = 40) (N = 59)
    Age (median; years) 65 (43-85) 64 (43-85)
    Male 22 (55%) 34 (58%)
    JAK-2V617F positive 35 (88%) 51 (86%)
    PMF 31 (78%) 44 (75%)
    Post-PV MF  6 (15%) 12 (20%)
    Post-ET MF  3 (8%)  3 (5%)
    High risk 20 (50%) 26 (44%)
    Palpable splenomegaly 39 (98%) 58 (98%)
    Transfusion dependent 16 (40%) 22 (37%)
  • This study was a dose-escalation study with expanded cohort dose confirmation at MTD. Below describes the data with a focus on the dose confirmation cohort who initiated treatment at a dose of 680 mg/day.
  • The decrease in palpable spleen size by cycle for subjects treated with TG101348 680 mg/day (starting dose) (N=37) is shown in FIG. 1. The baseline spleen size was: median=18cm; range=6-32 cm. 49% of subjects achieved clinical improvement based on reduction of palpable splenomegaly (IWG response) (56% of subjects by 12 weeks; 100% of subjects by 20 weeks). There was no relapse or disease progression at the time of data collection.
  • The effect of TG101348 on leukocytosis is shown in FIG. 2. The baseline WBC count was >11×109/L. 73% of subjects had normal WBC counts at their follow-up visit. The effect of TG101348 on thrombocytosis is shown in FIG. 3 (baseline platelet count >450×109/L). TG101348 was able to reduce platelet counts. The effects of TG101348 on constitutional symptoms (baseline versus last visit) are shown in FIG. 4. TG101348 was able to improve the MF-associated constitutional symptoms. TG101348 had no significant changes on cytokine levels (see FIG. 5, all values shown are medians). FIG. 6 shows the effect of TG101348 on V617F allele burden in subjects with baseline >20% (N=22). FIG. 6 shows that TG101348 was able to decrease JAK2 V617F allele burden in 59% of the subjects with baseline >20%.
  • FIG. 7 shows the effects of TG101348 on bone marrow cellularity in a 76-year-old male with V617F negative PMF. The starting dose was 30 mg/day and the dose at follow-up was 520 mg/day. FIG. 7 shows that TG101348 was able to reduce bone marrow cellularity in this subject from 60% bone marrow cellularity at baseline to 5-10% bone marrow cellularity after 18 cycles. FIG. 8 shows the effect of TG101348 on bone marrow fibrosis in a 56-year-old male with V617F negative PMF. The starting dose was 240 mg/day and the dose at follow-up was 440 mg/day. FIG. 8 shows that TG101348 was able to reduce bone marrow fibrosis in this subject from 3+ at baseline to 0 after 18 cycles.
  • The treatment-emergent grade 3 & 4 hematologic toxicities in MTD Subjects (N=40) is shown in Table 3. The treatment-emergent non-hematologic adverse events (reported for at least 5 subjects) in MTD Subjects (N=40) is shown in Table 4.
  • TABLE 3
    Treatment-Emergent Grade 3 & 4 Hematologic
    Toxicities in MTD Subjects (N = 40)
    Anemia
    Neutropenia Thrombocytopenia (N = 24)*
    (N = 40) (N = 40) New Transfusion
    Grade
    3 Grade 4 Grade 3 Grade 4 Requirement on Study‡
    6 (15%) 0 8 (20%) 5 (13%) 16 (67%)
    *Subjects who were not transfusion dependent at baseline.
    ‡Transfusion on at least 2 occasions for hemoglobin (“Hb”) <10 g/dL.
  • TABLE 4
    Treatment-Emergent Non-Hematologic Adverse
    Events in MTD Subjects (N = 40)
    Event Grade 1 Grade 2 Grade 3 Grade 4
    Gastrointestinal disorders
    Diarrhea 21 (53%)  4 (10%)  5 (13%) 0
    Nausea 20 (50%)  6 (15%) 2 (5%) 0
    Vomiting 20 (50%)  7 (18%) 1 (3%) 0
    Constipation  6 (15%) 1 (3%) 0 0
    Abdominal pain  5 (13%) 0 0 0
    Other
    Anorexia  7 (18%) 0 1 (3%) 0
    Edema peripheral  7 (18%) 1 (3%) 0 0
    Fatigue 2 (5%) 3 (8%) 1 (3%) 0
    Contusion  5 (13%) 0 0 0
    Headache  4 (10%) 1 (3%) 0 0
    Proteinuria 2 (5%) 3 (8%) 0 0
  • The grade ≥2 treatment-emergent non-hematologic laboratory findings in MTD subjects (N=40) is shown in Table 5.
  • TABLE 5
    Grade ≥ 2 Treatment-Emergent Non-Hematologic Laboratory Findings
    in MTD Subjects (N = 40)
    Finding Grade 2 Grade 3 Grade 4
    Creatinine increased 11 (28%)  0 0
    Hypocalcemia 8 (20%) 3 (8%) 0
    AST increased 5 (13%) 1 (3%) 0
    ALT increased 8 (20%) 2 (5%) 0
    Hyperkalemia 3 (8%)  2 (5%) 1 (3%)
    Hyperlipasemia 4 (10%) 3 (8%) 2 (5%)
    Hyperamylasemia 0 1 (3%) 1 (3%)

    Laboratory findings were transient and reversible, and resolved spontaneously or following dose interruption and/or reduction.
  • FIG. 9 shows various measurements in a subject with JAK2V617F-positive PMF that started at TG101348 680 mg/day. TG101348 was able to reduce the palpable spleen size from 9 cm to 0 cm and led to complete resolution of pruritus in this subject.
  • Conclusions: TG101348 was generally well tolerated, with manageable, grade 1 gastrointestinal effects, especially at higher doses. The data indicated no long-term toxicities. The expected on-target myelosuppressive effect appeared to be mostly limited to erythropoiesis, which may be attenuated at lower, but still effective, doses. TG101348 had remarkable activity in MF-associated splenomegaly: ˜two-thirds achieved ≥50% reduction in palpable splenomegaly; ˜30% had complete response. TG101348 had significant anti-myeloproliferation activity with virtually all treated subjects experiencing complete resolution of leukocytosis and thrombocytosis. TG101348 had remarkable activity against MF-associated constitutional symptoms, pruritus and cachexia. TG101348 induced a significant decrease in JAK2V617F allele burden in a substantial proportion of treated subjects. TG101348 had minimal effect on serum levels of proinflammatory cytokines; this was consistent with the absence of immediate adverse cytokine-rebound phenomenon upon study drug discontinuation. Without wishing to be bound by any theory, the activity of TG101348 appeared to be a direct consequence of its JAK2 inhibitory activity and not an indirect effect from non-specific anti-cytokine activity. Furthermore, the preliminary observations showed reduction in BM cellularity and reticulinfibrosis with extended treatment.
  • Example 3 Evaluation of TG101348 in Myelofibrosis
  • The subjects in this study were administered with capsule form of TG101348.
  • Study Design: The study constituted a Phase 1, dose-escalation trial (MF-TG101348-001). This study is also described in Examples 1 and 2. Study eligible patients were ≥18 years of age with high- or intermediate-risk primary myelofibrosis (PMF), post-PV MF, or post-ET MF (Tefferi A et al., Leukemia 22:14-22, 2008). Additional eligibility criteria and participating centers are listed in Table 6. All patients provided written informed consent. The primary endpoints were determination of safety and tolerability, dose-limiting toxicity (“DLT”), maximum tolerated dose (“MTD”) and pharmacokinetic (“PK”) behavior of TG101348. The secondary endpoint was assessment of therapeutic activity.
  • TABLE 6
    Detailed enrollment criteria for MF-TG101348-001
    Inclusion Criteria Exclusion Criteria
    1. Diagnosis of MF (PMF, post-PV MF, or 1. Any chemotherapy, immunomodulatory
    post-ET MF) according to the revised WHO drug therapy, immunosuppressive therapy,
    criteria.* corticosteroids > 10 mg/day prednisone or
    equivalent, or growth factor treatment within 14
    days (28 days in the case of darbepoetin) prior
    to initiation of TG101348.
    2. High-risk MF (defined by Mayo PSS), or 2. Major surgery or radiation therapy within
    Mayo PSS intermediate-risk MF** 28 days prior to initiation of TG101348.
    accompanied by symptomatic splenomegaly
    and/or unresponsive to available therapy.
    3. At least 18 years of age. 3. Concomitant treatment with agents known
    to inhibit or induce CYP3A4, unless approved
    by the sponsor.
    4. Body weight ≥ 50 kg. 4. Known hypersensitivity to any ingredients
    in the study drug formulation.
    5. ECOG performance status ≤ 2. 5. Active infection requiring antibiotics.
    6. Within 4 days prior to initiation of 6. Uncontrolled CHF (NYHA Classification 3
    TG101348: or 4), angina, MI, CVA, coronary/peripheral
    ANC ≥ 1 × 109/L artery bypass graft surgery, TIA, or pulmonary
    Platelet count ≥ 50 × 109/L embolism within 3 months prior to initiation of
    Serum creatinine ≤ 2.0 mg/dL study drug.
    Total bilirubin ≤ 2.0 mg/dL
    AST or ALT ≤ 3 times the ULN (unless
    clinically compatible with hepatic EMH)
    7. Life expectancy ≥ 12 weeks. 7. Cardiac dysrhythmias requiring ongoing
    treatment, bundle branch block on ECG or QRS
    duration > 120 ms, or prolongation of the QTc
    (Fridericia) interval to > 450 ms for males or > 470 ms
    for females.
    8. Negative serum pregnancy test result for 8. Pregnant or lactating females.
    women of childbearing potential.
    9. Absence of active malignancy other than 9. Women of childbearing potential, unless
    MF, with the exception of adequately treated surgically sterile for at least 3 months (i.e.,
    basal cell carcinoma and squamous cell hysterectomy), postmenopausal for at least 12
    carcinoma of the skin. months (FSH > 30 U/mL), unless they agree to
    use effective, dual contraceptive methods (i.e.,
    oral, injectable, or barrier method with male
    partner using a condom) while on study drug.
    10. Provide written informed consent to 10. Men who partner with a woman of
    participate. childbearing potential, unless they agree to use
    effective, dual contraceptive methods (i.e., a
    condom, with female partner using oral,
    injectable, or barrier method) while on study
    drug.
    11. Willing to comply with scheduled visits, 11. Known HIV- or AIDS-related illness.
    treatment plans, laboratory assessments, and 12. Clinically active hepatitis B or C.
    other study-related procedures. 13. Any severe, acute or chronic medical,
    neurological, or psychiatric condition or
    laboratory abnormality that may increase the
    risk associated with study participation or
    study drug administration, may interfere with
    the informed consent process and/or with
    compliance with the requirements of the study,
    or may interfere with the interpretation of
    study results and, in the investigator's opinion,
    would make the patient inappropriate for entry
    into this study.
    Abbreviations:
    AIDS = acquired immunodeficiency syndrome;
    ALT = alanine aminotransferase;
    ANC = absolute neutrophil count;
    AST = aspartate aminotransferase;
    CHF = congestive heart failure;
    CVA = cerebrovascular accident;
    ECG = electrocardiogram;
    ECOG = Eastern Cooperative Oncology Group;
    EMH = extramedullary hematopoiesis;
    FSH = follicle stimulating hormone;
    HIV = human immunodeficiency virus;
    MF = myelofibrosis;
    MI = myocardial infarction;
    NYHA = New York Heart Association;
    PSS = prognostic scoring system;
    TIA = transient ischemic attack;
    WBC = white blood cell.
    *Tefferi and Vardiman. Leukemia. 2008 Jan; 22(1): 14-22
    **High-risk disease requires two and intermediate-risk disease requires one of the following prognostic factors: hemoglobin < 10 g/dL, WBC count <4 or> 30 × 109/L, platelet count < 100 × 109/L, absolute monocyte count ≥ 1 × 109/L.
  • Patients were assigned to one of 8 dose cohorts, ranging from 30 to 800 mg per day, using standard 3+3 cohort design. TG101348 was administered orally once daily, with a treatment plan for continuous daily therapy for 24 weeks (6×28-day cycles). Intra-subject dose escalation was permitted after completion of at least 3 cycles of treatment at the starting dose. Once DLT was identified, a dose-confirmation cohort initiated treatment at the MTD. Treatment beyond 6 cycles was allowed on an extension study (MF-TG101348-002; NCT00724334) if deemed beneficial to the patient and if well tolerated.
  • Assessment of Toxicity and Response: Safety assessments were performed weekly during cycle 1, every other week during cycles 2 and 3, and every 4 weeks thereafter. Toxicity was graded in accordance with the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 3.0.
  • Responses were measured every 4 weeks per International Working Group for MPN Research and Treatment (IWG-MRT) criteria (Tefferi A et al., Blood 108:1497-1503, 2006). Assessment of bone marrow histology was performed at baseline and every 24 weeks of therapy. Changes in JAK2V 617F allele burden in the granulocyte fraction of peripheral blood were measured as previously described (Kittur J et al., Cancer 109:2279-2284, 2007); the assessments were at baseline and every 4 weeks during the first 6 cycles, and every 6th cycle of therapy in the extension study.
  • Pharmacokinetics: The concentration—time curves of TG101348 in plasma were evaluated by a non-compartmental analysis (with the use of WinNonlin® software, version 5.2).
  • Cytokine Assessment: Samples for cytokine measurement were collected at baseline and every 4 weeks thereafter. Cytokine levels were measured using multiplexed sandwich ELISAs (Millipore, St. Charles, Mo.).
  • Results
  • Enrollment of patients: A total of 59 subjects were enrolled; 28 in the dose-escalation phase and 31 in the dose-confirmation phase (Table 7). Forty-four subjects had PMF, 12 post-PV MF, and 3 post-ET MF; 86% were JAK2V617F-positive. The median duration of disease was 3.4 years (range 0.06 to 25.8). At study enrollment, the median palpable spleen size was 18 cm below the left costal margin (83% had a palpable spleen size >10 cm), median hemoglobin level was 9.2 g/dL (range 6.6 to 15.2) and 21 (36%) subjects were red cell transfusion-dependent by IWG-MRT criteria.
  • TABLE 7
    Demographic and Baseline Subject Characteristics
    TG101348 Starting Dose (mg/day) MTD All
    30 60 120 240 360 520 680 800 Cohort Doses
    Characteristic n = 4 n = 3 n = 3 n = 3 n = 3 n = 3 n = 34 n = 6 n = 40 n = 59
    Age-years 63.5 64.0 63.0 68.0 66.0 57.0 63.5 69.0 65.1 (10.47)† 64.5 (9.70)†
    Range 55-76 56-66 53-71 55-79 61-71 50-66 43-83 50-85 43-85 43-85
    Gender
    Male 2 3 1 2 2 2 18 4   22 (55.0%)   34 (57.6%)
    Female 2 0 2 1 1 1 16 2   18 (45.0%)   25 (42.4%)
    Race
    White 3 2 3 3 3 2 29 6   35 (87.5%)   51 (86.4%)
    Black, African 0 0 0 0 0 0 1 0   1 (2.5%)   1 (1.7%)
    American
    Asian 1 1 0 0 0 0 3 0   3 (7.5%)   5 (8.5%)
    Other 0 0 0 0 0 1 1 0   1 (2.5%)   2 (3.4%)
    Diagnosis
    PMF 3 2 1 3 2 2 27 4   31 (77.5%)   44 (74.6%)
    Post-PV MF 1 1 2 0 1 1 6 0   6 (15.0%)   12 (20.3%)
    Post-ET MF 0 0 0 0 0 0 1 2   3 (7.5%)   3 (5.1%)
    Risk Category
    (Mayo PSS)
    High 0 0 1 2 0 3 14 6   20 (50.0%)   26 (44.1%)
    Not high* 4 3 2 1 3 0 20 0   20 (50.0%)   33 (55.9%)
    JAK2V617F 3 3 3 2 3 2 29 6   35 (87.5%)   51 (86.4%)
    Positive
    Transfusion 1 1 0 1 0 2 13 3   16 (40.0%)   21 (35.6%)
    Dependent
    Spleen Size 3 3 3 2 3 2 28 5   33 (82.5%)   49 (83.1%)
    >10 cm
    Abbreviations:
    ET, essential thrombocythemia;
    JAK, Janus kinase;
    MF, myelofibrosis;
    PMF, primary myelofibrosis;
    PV, polycythemia vera;
    PSS, prognostic scoring system.
    *Equivalent to symptomatic/treatment refractory intermediate-risk disease.
    †Mean (standard deviation)
  • In the dose-escalation phase, the starting dose of TG101348 was 30 mg/day and subsequent dose levels were 60, 120, 240, 360, 520, 680 and 800 mg/day (Table 7). At 800 mg/day, 2 of 6 patients experienced DLT; consequently, the MTD was declared at 680 mg/day. In the dose-confirmation phase, all patients started treatment at the MTD. The “MTD cohort” (n=40; Table 7) included patients who received 680 mg/day as their starting dose (dose-escalation cohort, n=3; dose-confirmation cohort, n=31) and those whose drug dose was decreased from 800 mg/day (n=6) to 680 mg/day after MTD was declared.
  • The median (range) exposure to TG101348 for the overall (n=59) and MTD (n=40) cohorts was 155 (2-172) and 147 (8-171) days, respectively. TG101348 doses at the end of each cycle per dose cohort are illustrated in FIGS. 10 and 11. In the MTD cohort, 28 subjects (70%) required dose-reduction during the first 6 cycles; the primary reasons were: cytopenia(s) (20%), gastrointestinal adverse events (12.5%), amylase/lipase elevation (10%), ALT elevation (7.5%), investigator discretion (7.5%), or other adverse events (12.5%). The median cycle at dose-reduction for the MTD cohort was cycle 3 (range 1-7); the median (range) dose at the end of cycle 3 was 680 mg/day (360-680 mg/day); and 520 mg/day (360-680 mg/day) at the end of cycle 6.
  • Forty three (73%) subjects, including 28 (70%) from the MTD cohort, continued treatment on the extension study; at entry into the extension study, 31 (72%) subjects were receiving <680 mg/day of the drug (median 520 mg/day; range 120-680 mg/day). At data cutoff, the median (range) cumulative exposure to TG101348 for the 43 subjects was 380 days (170-767). The number of treatment cycles completed ranged from 7-29; 39 subjects (66%), including 27 (68%) from the MTD cohort completed 12 treatment cycles. At data cutoff, 28%, and 14% of subjects who entered the extension study had completed 18 and 24 treatment cycles, respectively. The median (range) treatment dose during the extension phase was 440 mg/day (120-680 mg/day).
  • Pharmacokinetics: Peak plasma concentration of TG101348 was achieved 1-4 hours after dosing. TG101348 showed greater than dose-proportional increases in plasma PK parameters (Cmax and AUC0-t) (Table 8 and FIG. 12). Mean steady-state C. and AUC0-t values increased approximately 54- and 88-fold, respectively, over a 27-fold increase in dose. The terminal phase half-life at steady state remained similar across all doses (16 to 34 hours), consistent with linear drug elimination. FIG. 18 shows a plot of mean plasma TG101348 concentrations versus time on a linear plot after once daily oral doses (Cycle 1; Day 28). The figure shows the IC50, IC90, and 3 times IC90 (3×IC90) values for TG101348 in relation to the plasma concentration of TG101348 over time. A dose of 520 mg/day exhibited a plasma concentration TG101348 that was above 3×IC90 over the course of at least 24 hours after the dose was administered. A dose of 360 mg/day exhibited a Cmax above 3×IC90 and a plasma concentration of TG101348 that was above IC90 over the course of at least 24 hours after the dose was administered.
  • TABLE 8
    Mean (SD) plasma pharmacokinetic parameters following multiple daily doses of
    TG101348 (Cycle 1, Day 28) in MF-TG101348-001
    Dose/Day
    30 mg 60 mg 120 mg 240 mg 360 mg 520 mg 680 mg 800 mg
    Parameter (n = 3) (n = 3) (n = 3) (n = 3) (n = 3) (n = 3) (n = 27) (n = 5)
    Cmax 81.85 257.33 556.67 1796.67 1717.33 3886.67 3064.07 4380.00
    (ng/mL) (95.630) (121.138) (135.500) (648.254) (1558.705) (3560.707) (1129.671) (1764.809)
    Tmax* (hr) 2.00 1.00 2.00 2.00 2.00 4.00 4.00 2.25
    (0.5, 4.0) (1.0, 4.0) (0.5, 4.0) (2.0, 2.1) (2.0, 4.0) (4.0, 4.0) (0.0, 8.3) (2.0, 4.0)
    AUC(0-t) 806.76 2426.53 7645.69 26193.40 23879.05 61749.22 55111.68 70840.97
    (hr*ng/mL) (806.973) (1048.264) (2810.740) (11767.460) (16898.162) (57240.295) (25702.038) (32668.886)
    T1/2 (hr) 20.94 15.68 24.42 20.77 21.39 20.94 33.71 23.99
    (7.039) (3.464) (8.434) (6.238) (7.090) (5.006) (33.674) (9.674)
    λz (1/hr) 0.0354 0.0456 0.0305 0.0352 0.0353 0.0343 0.0301 0.0331
    (0.01016) (0.00918) (0.00932) (0.00903) (0.01309) (0.00723) (0.01421) (0.01321)
    *Tmax is presented as median (min, max)
    SD indicates standard deviation; Cmax, peak plasma concentration; Tmax, the time to the maximal concentration; AUC(0-t), area under the concentration-time curve from time zero to the last measurable concentration; T1/2, terminal half-life; and λz, the elimination rate constant.
  • Safety profile: The DLT in 2 of 6 patients treated at 800 mg/day was asymptomatic grade 3 or 4 hyperamylasemia (with or without hyperlipasemia) that was reversible. The most common non-hematologic adverse events at least possibly related to TG101348 included predominantly grade 1 nausea, diarrhea and vomiting; grade 3 events were reported overall/in the MTD cohort for 3%/5%, 10%/13%, and 3%/3% of subjects, respectively, and there were no Grade 4 events (Table 9). These adverse events were dose-dependent, with grade 3 occurrences observed almost exclusively with a TG101348 starting dose of ≥680 mg/day. The gastrointestinal symptoms were largely self-limited or controlled by symptomatic treatment and/or dose reduction. Other adverse events (Grades 3/4; overall/MTD cohort) included asymptomatic increases in serum lipase (10%/15%), AST (2%/3%), ALT (7%/8%), creatinine (0%/0%) and alkaline phosphatase (0%/0%) (Table 9).
  • TABLE 9
    Treatment-Emergent Non-Hematologic Adverse Events Considered at
    Least Possibly Related to TG101348 and Reported for ≥10% of Subjects
    MTD Cohort All Subjects
    (n = 40) (n = 59)
    Severity Severity Severity Severity
    Adverse Events Grade 1-2 Grade 3- Grade 1- Grade 3-4
    Gastrointestinal disorders
    Nausea 31 2 (5.0%) 39 2 (3.4%)
    Diarrhea 25 5 32 6 (10.2%)
    Vomiting 27 1 (2.5%) 32 2 (3.4%)
    Abdominal pain 4 (10.0%) 0 6 (10.2%) 0
    General disorders
    Anorexia 6 (15.0%) 0 8 (13.6%) 0
    Edema peripheral 4 (10.0%) 0 6 (10.2%) 0
    Abnormal laboratory
    values
    Hyperlipasemia 9 (22.5%) 6 10 6 (10.2%)
    Alanine 9 (22.5%) 3 (7.5%) 11 4 (6.8%)
    aminotransferase
    increased
    Aspartate 13 1 (2.5%) 15 1 (1.7%)
    aminotransferase
    increased
    Blood creatinine 11 0 14 0
    increased
    Blood alkaline 9 (22.5%) 0 10 0
    phosphatase increased
    Hypocalcemia 6 (15.0%) 1 (2.5%) 7 (11.9%) 1 (1.7%)
    Skin and subcutaneous
    tissue disorders
    Skin exfoliation 8 (20.0%) 0 8 (13.6%) 0
    Dry skin 6 (15%) 0 6 (10.2%) 0
  • Grade 3/4 hematological adverse events considered related to TG101348 included anemia (35% of 37 subjects who were not transfusion dependent at baseline), thrombocytopenia (24%) and/or neutropenia (10%) (Table 10). The majority of treatment-emergent cytopenias were noted in the first three cycles of treatment. Of the 13 subjects who developed grade 3/4 anemia (all in the MTD cohort), 67% entered the study with grade 2 anemia. Emergence of transfusion requirement was significantly lower for subjects who initiated treatment at 240-520 mg/day (33%) as opposed to 680 mg/day (72%). Of the 14 subjects with grade 3/4 thrombocytopenia, 4 and 5 subjects entered the study with grade 1 and 2 thrombocytopenia, respectively.
  • TABLE 10
    Treatment-Emergent Hematologic Adverse Events Considered at Least
    Possibly Related to TG101348 and Reported for ≥10% of Subjects
    MTD Cohort All Subjects
    (n = 40) (n = 59)
    Severity Severity Severity Severity
    Grade 1-2 Grade 3-4 Grade 1-2 Grade 3-4
    Anemia* 2 (8.3%) 13 (54.2%) 3 (8.1%) 13 (35.1%)
    Throm-  8 (20.0%) 11 (27.5%) 10 (17.0%) 14 (23.7%)
    bocytopenia
    Neutropenia 2 (5.0%)  4 (10.0%) 2 (3.4%)  6 (10.2%)
    *Events reported only for subjects who were not transfusion dependent at study entry (MTD Cohort, n = 24; All Subjects, n = 37) are presented.
  • At data cutoff, no unique safety findings have emerged with continued dosing of TG101348 beyond 6 cycles of therapy.
  • Serious adverse events considered at least possibly related to TG101348 occurred in 8 subjects and included asymptomatic hyperlipasemia, thrombocytopenia/neutropenia, depression, tumor lysis syndrome, cerebrovascular accident, and dehydration (Table 11). One subject discontinued treatment due to Grade 4 thrombocytopenia; all other events were reversible and subjects were able to resume treatment at a lower dose after resolution of the adverse event.
  • TABLE 11
    Serious Adverse Events Assessed by Investigators as at Least Possibly Related to Therapy
    (MF-TG101348-001 and MF-TG101348-002)
    Onset
    Starting From
    Dose/Dose Start of CTCAE Action Taken
    at Event Dosing Severity With Study
    Subject # Event (mg/day) (days) Grade Drug Outcome
    105-013 Thrombocytopenia 240/360 215 4 None Recovered/resolved
    Thrombocytopenia 240/360 247 4 Permanently Not recovered/not
    discontinued resolved
    Hyperlipasemia 240/0 356 4 None Recovered/resolved
    104-015 Depression 360/520 256 3* Permanently Not recovered/not
    discontinued resolved
    106-024 Nausea 800/680  87 2 Stopped Recovered/resolved
    temporarily
    Vomiting 800/680  87 2 Stopped Recovered/resolved
    temporarily
    Diarrhea 800/680  87 3 Stopped Recovered/resolved
    temporarily
    Dehydration 800/680  87 2 Stopped Recovered/resolved
    temporarily
    Tumor lysis 800/440 366 3 Stopped Recovered/resolved
    syndrome temporarily
    Dehydration 800/400 474 3 None Recovered/resolved
    106-033 Pleuritic pain 680/680  8 2 Stopped Recovered/resolved
    temporarily
    106-045 Dehydration 680/440 170 3 Stopped Recovered/resolved
    temporarily
    101-047 Neutropenia 680/680  52 2 Stopped Recovered/resolved
    temporarily
    105-056 Cerebrovascular 680/680  22 4 Stopped Recovered/resolved
    accident temporarily
    Gallbladder pain 680/520  95 3 Stopped Recovered/resolved
    temporarily with sequelae
    105-059 Hyperlipasemia 680/680  8 3 Stopped Recovered/resolved
    temporarily
    Hyperlipasemia 680/520  28 3 Stopped Recovered/resolved
    temporarily
    Cardiac arrest 680/360  42 5 Permanently Fatal
    discontinued
    *Subject died (suicide) approximately 12 weeks after discontinuation of study drug.

    One subject presented with severe pulmonary hypertension and right heart failure during cycle 4 (at 240 mg/day); the event was considered unrelated to TG101348 per the investigator.
  • Fifteen (25%) subjects discontinued treatment during the first 6 cycles of therapy (Table 12). Reasons for discontinuation included treatment-related adverse events (n=6); investigator decision/intercurrent illness (n=3) or withdrawal of consent (n=6). Eight of 43 subjects (19%) discontinued treatment during the extension study, including 3 because of adverse events following a total of 24 to 46 weeks on therapy (Table 12).
  • TABLE 12
    Subjects discontinuing study due to death, toxicity,
    withdrawal of consent, or intercurrent illness
    MF-TG101348-001 Reasons for Discontinuation (Table 12 A)
    Starting Dose at Duration of
    Dose Termination Treatment
    Subject (mg/day) (mg/day) (days) Reason
    102-002 30 30 2 Investigator discretion - previously
    undiagnosed cardiac condition with long
    QTc interval
    106-009 120 240 109 Patient withdrew consent
    101-011 240 240 100 Patient withdrew consent
    102-019 520 520 42 Adverse event - neutropenia (grade 3;
    probably related)
    102-023 800 680 70 Investigator discretion - recurrent
    Waldenstrom's macroglobulinemia
    104-027 800 680 77 Adverse event - thrombocytopenia (grade
    4; possibly related)
    106-028 800 520 44 Adverse event - thrombocytopenia (grade
    4; possibly related)
    104-029 680 680 44 Adverse event - endocarditis (grade 3;
    not related), embolic stroke (grade 3;
    not related)
    101-032 680 680 8 Investigator discretion - Acquired factor
    VIII inhibitor
    101-040 680 520 24 Adverse events - diarrhea (grade 3;
    possibly related)
    103-043 680 360 68 Patient withdrew consent
    103-046 680 680 26 Patient withdrew consent
    102-051 680 600 108 Patient withdrew consent
    102-054 680 680 75 Patient withdrew consent
    105-059 680 360 27 Adverse event - cardiac arrest (grade 5;
    possibly related)
    MF-TG101348-002 Reasons for Discontinuation (Table 12B)
    Cumulative
    Starting Dose at Duration of
    Dose Termination Treatment
    Subject (mg/day) (mg/day) (days) Reason
    101-005 60 360 196 Investigator discretion - lack of
    response to treatment
    106-010 120 520 185 Investigator discretion
    105-013 240 360 321 Adverse event - thrombocytopenia
    (grade 4; probably related)
    104-015 360 520 257 Adverse event - depression (grade 3;
    possibly related)
    106-016 360 680 527 Investigator discretion - lack of
    response to treatment
    104-017 520 200 309 Investigator discretion - disease
    progression
    105-021 680 520 357 Patient withdrew consent
    101-047 680 320 233 Adverse event - elevated creatinine
    (grade 2; possibly related)
  • Three subjects had disease progression (doses at study start and discontinuation are indicated): one each with progressive hepatosplenomegaly and ascites with superimposed endocarditis (cycle 2; 680 and 520 mg/day), accelerated myelofibrosis (cycle 13; 520 and 200 mg/day), and leukemic transformation (cycle 2; 520 and 520 mg/day).
  • Responses are shown below.
  • Splenomegaly: The onset of spleen response was rapid, and generally seen within the first 2 cycles. By cycle 6, 36 subjects (61%) had experienced a minimum 25% decrease in palpable spleen size, including 65% in the MTD cohort (intent-to-treat analysis). By this time-point, a ≥50% decrease in palpable spleen size persistent for at least 8 weeks (i.e. Clinical Improvement (“CI”) per IWG-MRT criteria) had been observed in 39% and 45% of subjects overall and in the MTD cohort, respectively. Spleen responses per treatment cycle for the MTD cohort are shown in FIG. 13. Three of 4 subjects (75%) with JAK2V617F-negative MF who completed 6 cycles of treatment achieved CI. The lowest starting dose at which CI was observed was 240 mg/day. The median time (range) to CI across doses was 141 days (41 to 171), and 113 days (41-170) for the MTD cohort. By cycle 12, spleen responses (CI) were observed in 48% and 50% of subjects, for the overall and MTD cohorts, respectively. The mean (standard deviation) duration of spleen response per IWG-MRT criteria was 315 (±129) days and 288 (±76) days for the overall and MTD cohorts, respectively.
  • Constitutional symptoms: Thirty five subjects in the MTD cohort endorsed the presence and severity of early satiety, fatigue, night sweats, cough, and pruritus on an 11-point scale (0=absence of symptoms to 10=worst imaginable symptoms) at baseline and at the end of at least one cycle. Symptoms were categorized as “absent” (score=0), “mild” (score=1-3), “moderate” (score=4-7), or “severe” (score=8-10).
  • Early satiety was reported by 29 (85%) subjects at baseline. After 2 cycles of treatment (n=27), 56% reported complete resolution of this symptom (FIG. 14A). Fatigue was reported at baseline by 26 (76%) subjects. After 6 cycles (n=16), 63% reported improvement and 25% complete resolution of this symptom (FIG. 14B). Night sweats were reported at baseline by 14 (40%) subjects. After 1 cycle, 64% of subjects had complete resolution of this symptom; after 6 cycles, this proportion had increased to 89% (n=9) (FIG. 14C). Cough was reported at baseline by 13 (37%) subjects. After 1 cycle (n=12), 75% reported improvement and 67% complete resolution of this symptom. Pruritus was reported by 8 (23%) subjects at baseline. After 1 cycle, 75% had improvement, with 50% reporting complete resolution. Responses in constitutional symptoms were durable in most instances.
  • Body weight: At the end of 6 and 12 cycles, the median body weight was stable relative to baseline for the overall and MTD cohorts (Table 13).
  • TABLE 13
    Change in weight during study treatment
    Baseline
    6 Cycles 12 Cycles
    MTD MTD MTD
    Overall Cohort Overall Cohort Overall Cohort
    Weight (kg) (n = 57) (n = 38) (n = 43) (n = 28) (n = 36) (n = 26)
    Median (range) 75.6 77.7 76.9 77.7 76.1 76.5
    (48.2-105.2) (48.2-96.1) (51.4-105.8) (51.4-97.6) (49.8-106.8) (49.8-99.5)
    Change from n/a n/a 0.4 0.6 0.7 0.35
    baseline Median (−11.7-8.9) (−9.2-8.9) (−10.7-13.7) (−10.7-13.7)
    (range)
    kg indicates kilograms, n, number, and MTD, maximum tolerated dose
  • Leukocytosis and Thrombocytosis: Leukocytosis (WBC count >11×109/L) was present at baseline in 33 subjects (56%), 28 of whom completed 6 cycles of treatment; of these, 18 were in the MTD cohort. Following 6 cycles, 16 subjects across doses (57%) and 13 subjects in the MTD cohort (72%) achieved a normal WBC count (FIG. 15); following 12 cycles, 14 of 25 (56%) across doses and 10 of 17 (59%) in the MTD cohort had normal WBC counts.
  • Thrombocytosis (platelet count >450×109/L) was noted at baseline for 10 (17%) subjects across doses and 7 (19%) in the MTD cohort (n=37), all of whom completed 6 cycles of therapy. At this time point, 90% and 100% of subjects across doses and in the MTD cohort, respectively, achieved a normal platelet count; following 12 cycles, 7 of 8 subjects (88%) across doses and all 6 subjects in the MTD cohort had a normal platelet count.
  • JAK2V617F allele burden: Fifty-one subjects (86%) were JAK2V617F-positive, with a median (range) allele burden of 20% (3%-100%); of these, 23 (45%) had a “significant” allele burden (defined as >20% at baseline) with a median (range) of 60% (23%-100%). For the overall mutation-positive subjects, there was a significant decrease in the JAK2V617F allele burden after 6 cycles (p=0.04) and 12 cycles of treatment (p=0.01) (FIGS. 16A and 16B). After 6 and 12 cycles of treatment, the median (range) allele burden was 17% (0%-100%) and 19% (0%-100%), respectively. Similarly, for the 23 subjects with baseline JAK2V617F allele burden of >20%, there was a significant and even more pronounced decrease in the JAK2V617F allele burden after 6 cycles (p=0.002) and 12 cycles of treatment (p=0.002) (FIGS. 16C and 16D). After 6 and 12 cycles of treatment, the median (range) allele burden was 31% (4%-100%) and 32% (7%-100%), respectively. After 6 cycles, 16 of 20 subjects (80%) with baseline allele burden >20% who reached this time-point exhibited a median 61% (range 6% to 96%) decrease, and 9 subjects (45%) had a ≥50% decrease in JAK2V617F allele burden. In contrast, 4 subjects (20%) exhibited an increase (18%, 21%, 30%, and 58%). Eighteen subjects (78%) of the group with allele burden >20% completed 12 cycles of treatment with a median 50% (range 29% to 82%) decrease, and 7 (39%) subjects had a ≥50% decrease in JAK2V617F. Three (17%) subjects exhibited an increase in allele burden (7%, 18%, and 22%), and 2 others with 100% allele burden at baseline exhibited no change.
  • Discussion: A significant proportion of patients treated in this study experienced rapid, substantial, and durable control of symptomatic splenomegaly, leukocytosis, thrombocytosis, and constitutional symptoms. In addition, there was also evidence for a significant reduction in genomic disease burden that indicates potential for disease modifying activity. There were responses in MF patients who were JAK2V617F negative. It is unknown whether the subjects in this study have other mutations in the JAK-STAT signal transduction pathway such as MPL, LNK or as yet unknown alleles (Pardanani A D et al., Blood 108:3472-3476, 2006; Oh S T et al., Blood First Edition Paper, prepublished online Apr. 19, 2010; DOI 101182/blood-2010-02-270108 2010; Pardanani A et al., Leukemia In press:2010).
  • The clinical study results show that TG101348 therapy can be discontinued without prior dose reduction or tapering. Subjects that were discontinued (whether or not recontinued at a later date) did not experience “cytokine rebound”. This indicates that the treatment may be discontinued without prior dose reduction.
  • Cytokine rebound in the context of myelofibrosis is a phenomenon that has occurred in patients receiving therapy other than TG101348 therapy and were discontinued for any reason. In some cases, the discontinued patients experienced severe symptoms including acute spleen size enlargement and relapse of constitutional symptoms. In some cases, the discontinued patients experienced life-threatening hemodynamic disturbances (Wadleigh and Tefferi, Clinical Advances in Hematology & Oncology, 8:557-563, 2010).
  • Of note, among small molecule inhibitors of the JAK-STAT pathway in MF, TG101348 appeared to be unique in its ability to induce a significant and sustained decrease in JAK2V617F mutant allele burden. Without wishing to be bound by any theory, it appeared that the effect of JAK2 inhibition on disease burden was the basis for evidence of clinical efficacy in myelofibrosis with TG101348, as opposed to an indirect anti-cytokine effect that may play a major role in responses to JAK family antagonists that have off-target activity for JAK1 as well as for JAK2. In support of this, there were no consistent changes in levels of pro-inflammatory cytokines (interleukin (“IL”)-6, IL-2, IL-8, and TNF-α) relative to baseline during the course of TG101348 treatment (FIG. 17). In contrast, and consistent with the on-target activity of TG101348 for JAK2, increases in serum EPO and to a lesser extent TPO levels relative to baseline were observed after treatment initiation (data not shown).
  • The DLT (asymptomatic hyperamylasemia, sometimes with hyperlipasemia) for TG101348 was observed with other small molecule inhibitors including nilotinib (Kantarjian HM et al., Blood 110:3540-3546, 2007). Gastrointestinal adverse events were frequent in this study but accounted for treatment discontinuation in only one subject. These symptoms occurred as early as after the first administered dose, and demonstrated a clear dose-dependent relationship. The myelosuppressive effects of TG101348 were also dose-dependent.
  • While the MTD (680 mg/day) of TG101348 was the most efficacious dose, it was also associated with the highest incidence of adverse events. Therefore, a lower starting dose (e.g. 400-500 mg/day) may provide an optimal risk/benefit balance. Furthermore, because myelofibrosis is a heterogeneous disease, a dynamic dosing schedule may maximize the opportunity for identifying a patient-specific optimal dose.
  • These observations suggest that, in addition to MF, TG101348 may also have a potential role for the treatment of PV and ET.
  • Example 4 Synthesis of TG101348 Example 4.1 N-tert-Butyl-3-(2-chloro-5-methyl-pyrimidin-4-ylamino)-benzenesulfonamide (Intermediate) Example 4.1(a)
  • Figure US20210244735A1-20210812-C00003
  • A mixture of 2-chloro-5-methyl-pyrimidin-4-ylamine (1) (0.4 g, 2.8 mmol), 3-bromo-N-tert-butyl-benzenesulfonamide (2) (1.0 g, 3.4 mmol), Pd2(dba)3 (0.17 g, 0.19 mmol), Xantphos (0.2 g, 3.5 mmol) and cesium carbonate (2.0 g, 6.1 mmol) was suspended in dioxane (25 mL) and heated at reflux under the argon atmosphere for 3 h. The reaction mixture was cooled to room temperature and diluted with DCM (30 mL). The mixture was filtered and the filtrate concentrated in vacuo. The residue was dissolved in EtOAc and hexanes added until solid precipitated. After filtration, the title compound (1.2 g, 98%) was obtained as a light brown solid. It was used in the next step without purification. MS (ES+): m/z 355 (M+H)+.
  • Example 4.1(b)
  • Figure US20210244735A1-20210812-C00004
  • The Intermediate was synthesized from 2,4-dichloro-5-methylpyrimidine (SM1) and N-t-butyl-3-aminobenzenesulfonamide (SM2) in the following steps: (1) Mix MeOH (6.7UOa) and SM1 (Combi Blocks) (UOa); (2) Add SM2 (1.15UOa, 082eq) and H2O (8.5UOa); (3) Heat 45° C., 20 h, N2, IPC CPL SM2<2%; (4) Cool 20° C.; (5) Centrifuge, N2; (6) Wash H2O (2.1UOa)+MeOH (1.7UOa); (7) Mix solid in H2O (4.3UOa) +MeOH (3.4UOa); (8) Centrifuge, N2; (9) Wash H2O (2.1UOa)+MeOH (1.7UOa); and (10) Dry 45° C., vacuum, 15 h. Obtained Intermediate, mass 49.6kg (UOb); Yield 79%; OP: 99.6%.
  • Example 4.2 N-tert-Butyl-3-[)5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide
  • Figure US20210244735A1-20210812-C00005
  • Example 4.2(a)
  • A mixture of N-tert-Butyl-3-(2-chloro-5-methyl-pyrimidin-4-ylamino)-benzenesulfonamide (Intermediate) (0.10 g, 0.28 mmol) and 4-(2-pyrrolidin-1-yl-ethoxy)-phenylamine (3) (0.10 g, 0.49 mmol) in acetic acid (3 mL) was sealed in a microwave reaction tube and irradiated with microwave at 150° C. for 20 min After cooling to room temperature, the cap was removed and the mixture concentrated. The residue was purified by HPLC and the corrected fractions combined and poured into saturated NaHCO3 solution (30 mL). The combined aqueous layers were extracted with EtOAc (2×30 mL) and the combined organic layers washed with brine, dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated and the resulting solid dissolved in minimum amount of EtOAc and hexanes added until solid precipitated. After filtration, the title compound was obtained as a white solid (40 mg, 27%). 1H NMR (500 MHz, DMSO-d6): δ 1.12 (s, 9H), 1.65-1.70 (m, 4H), 2.12 (s, 3H), 2.45-2.55 (m, 4H), 2.76 (t, J=5.8 Hz, 2H), 3.99 (t, J=6.0 Hz, 2H), 6.79 (d, J=9.0 Hz, 2H), 7.46-7.53 (m, 4H), 7.56 (s, 1H), 7.90 (s, 1H), 8.10-8.15 (m, 2H), 8.53 (s, 1H), 8.77 (s, 1H). MS (ES+): m/z 525 (M+H)+.
  • Example 4.2(b)
  • N-tert-Butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate was prepared from 4-p-(1-pyrrolidinyl)ethoxylaniline dihydrochloride (SM3) and Intermediate following steps (A) and (B).
  • Step (A), preparation of free base of SM3 (3) from SM3, comprised steps (1)-(9): (1) Solubilize NaOH (0.42UOb) in H2O (9UOb); (2) Cool <20° C., N2; (3) Add TBME (6UOb) then SM3 (Malladi Drugs) (1.06UOb); (4) Mix >20 mn then stop; (5) Drain Aq Ph then extract by TBME (3UOb); (6) Combine Or Ph; (7) Concentrate, vacuum, T<40° C., to an Oil; (8) Solubilize in IPA (2.5UOb); and (9) Calculate dry extract 23%.
  • Step (B) comprised the steps (1)-(6): (1) Mix IPA (10.5UOb) and Intermediate (UOb); (2) Add free base of SM3 (0.75UOb, 1.33eq/interm); (3) add HCl conc (0.413UOb); (4) Heat 70° C., 20 h, N2, IPC CPL Interm<2%; (5) Cool <20° C.; (2) Centrifuge, N2; (3) Wash IPA (3UOb); (4) Dry 50° C., vacuum, 26 h; (5) De-lump in Fitzmill; and (6) polybag (x2)/poly drum. Obtained TG101348 dihydrochloride monohydrate, mass 83.8kg; Yield 98%; OP: 99.5%.
  • Example 5 Capsule Form of TG101348 and Process of Making TG101348
  • TG101348 drug products were provided as 10 mg, 40 mg, and 200 mg capsule strengths, where weights are specified for the amount of active (i.e., free base) moiety of TG101348. The quantitative composition of each strength of TG101348 drug product capsule is shown in Table 14.
  • TABLE 14
    List of all components and unit formula for 10 mg, 40 mg, and 200 mg
    strengths of TG101348 drug product capsules
    Component
    and Quality Unit Formula
    Standard (and TG101348 TG101348 TG101348
    Grade, if 10 mg 40 mg 200 mg
    Applicable) Function Capsule Capsule Capsule
    TG101348 (drug Active  11.73 mg  46.90 mg 234.80 mg
    substance)* ingredient
    Silicified Filler/ 121.92 mg 448.10 mg 356.70 mg
    Microcrystalline Diluent
    Cellulose
    (Prosolv SMCC
    90HD ®)†
    Sodium Stearyl Lubricant  1.35 mg  5.00 mg  6.00 mg
    Fumarate
    (Pruv ®)
    Total Capsule NA 135.00 mg 500.00 mg 597.50 mg
    Fill Weight
    Hard Gelatin Container 1 capsule 1 capsule 1 capsule
    Capsule (white (white (Swedish
    opaque, size opaque, size orange,
    3, each 00, each opaque,
    capsule is capsule is size 00, each
    48 ± 3 mg) 118 ± 7 mg) capsule is
    118 ± 7 mg)
    *Adjusted to obtain full potency based on the purity of the TG101348 drug substance lot used.
    †Adjusted to accommodate all component weights so as to ensure the total capsule fill weight is constant.
    USP = United States Pharmacopoeia;
    NF = National Formulary;
    EP = European Pharmacopoeia;
    JP = Japanese Pharmacopoeia;
    NA = not applicable.
  • The components that were used in the manufacturing process for each capsule strength, on a per batch basis, are shown in Table 15.
  • TABLE 15
    List of all components for manufacturing of the dosage forms
    TG101348
    TG101348 TG101348
    200 mg
    Strength (Label Claim) 10 mg Capsule 40 mg Capsule Capsule
    Batch Size 1,620.000 g 6,000.000 g 5,975.000 g
    Component and Quality
    Standard (and Grade if Quantity per Quantity per Quantity per
    Applicable) Batch (g) Batch (g) Batch (g)
    INTRAGRANULAR COMPONENTS
    TG101348* 140.780 562.800 2,348.175
    Silicified 214.160 856.800 3,567.075
    Microcrystalline
    Cellulose (Prosolv
    SMCC 90HD)†
    Sodium Stearyl Fumarate 3.560 14.400 59.750
    (Pruv)
    EXTRAGRANULAR COMPONENTS
    Silicified 1248.860 4,520.400 Not included
    Microcrystalline
    Cellulose (Prosolv
    SMCC 90HD)
    Sodium Stearyl Fumarate 12.640 45.600 Not included
    (Pruv)
    TOTAL OF INTRAGRANULAR COMPONENTS +
    EXTRAGRANULAR COMPONENTS
    Total Batch Weight 1,620.000 g 6,000.000 g 5,975.000 g
    CAPSULE SHELLS
    Capsule Shell Type Hard gelatin Hard gelatin Hard gelatin
    Capsule Size Size 3 Size 00 Size 00
    Capsule Color White, opaque White, opaque Swedish
    orange,
    opaque
    Total Batch Scale 12,000 12,000 10,000
    (Capsules)
    *Adjusted to obtain full potency based on the purity of the TG101348 drug substance lot used.
    †Adjusted to accommodate all component weights so as to ensure the total batch weight is constant.
  • The process for making TG101348 capsules is described below:
  • A. Dry granulation of intragranular components (implemented for all three drug product strengths): 1. TG101348 and intragranular sodium stearyl fumarate were blended within a V-blender for 5 minutes. 2. The blend was passed through a conical mill equipped with a round 18-mesh screen and round impeller. The blend was recharged into the V-blender. 3. Intragranular silicified microcrystalline cellulose was sifted through a 20-mesh screen and added to the blender. The mixture was blended for 15 minutes. 4. The blend was passed through a roller compactor. 5. The roller compacted ribbons were passed through a conical mill equipped with a round 16-mesh screen and round impeller. 6. The milled material was blended within the V-blender for 5 minutes. 7. In-process check (IPC) samples were withdrawn from the V-blender using a sample thief. Samples were subjected to potency analysis.
  • B. Addition of extragranular components (implemented for 10 mg and 40 mg capsules): 1. Where potency of granules (from Step 7 in A) was outside 98-102% (w/w) nominal, extragranular silicified microcrystalline cellulose was adjusted accordingly. 2. The V-blender was charged with TG101348 di-HCl monohydrate/silicified microcrystalline cellulose/sodium stearyl fumarate granules (from A). 3. The extragranular silicified microcrystalline cellulose was sifted through a 20-mesh screen and added to the V-blender. 4. The Extragranular sodium stearyl fumarate was added to the V-blender. 5. The intragranular and extragranular components were blended for 15 minutes. 6. IPC samples were withdrawn from the V-blender using a sample thief and analyzed for potency.
  • C. Capsule-filling (implemented for all three drug product strengths): 1. If potency (from Step 7 in A for the 200 mg capsules, or Step 6 in B for the 10 mg and 40 mg capsules) was outside 98-102% (w/w) nominal, the capsule fill weight was adjusted accordingly. 2. The prepared material was encapsulated using automatic capsule filling machine. The prepared capsules were bottled and stores at 20-28° F. (68-82° C.) and ambient humidity.
  • Content uniformity and dissolution were examined HPLC method validation was performed using a one-analyst, one-run-per-analyst design, and satisfied all required criteria for specificity, sensitivity, precision, accuracy, linearity, and sample stability. Specificity was evaluated and confirmed by comparing peak resolution between TG101348 and all of its related compounds, intermediates, and degradants (established from forced degradation studies). The limit of quantitation and limit of detection was established at 0.10 μg/mL and 0.03 μg/mL TG101348, respectively. Precision for content uniformity was evaluated via six injections of the 10 mg and 200 mg strength capsules, prepared at the target assay concentration. RSD results were 3.7% and 5.8% for the 10 mg and 200 mg strength capsules, respectively. Precision for dissolution was evaluated via six injections at each dissolution timepoint of the 10 mg and 200 mg strength capsules. Relative standard deviation (“RSD”) results for all strengths and corresponding time points were well within the acceptance criteria (±10%) specified in the validation protocol. Accuracy (defined by the recovery of the analyte spiked into a placebo solution for the 10 mg and 200 mg strength capsules) was evaluated at 70%, 100%, and 130% of the target assay standard concentration. Recovery values for all measurements were within the acceptance criteria (93%-105%) specified in the validation protocol. Linearity was demonstrated over the range of 50% to 120% of the target assay standard concentration, and exhibited an r2 of 1.00. Sample stability and method robustness were also demonstrated during method validation.
  • Example 6 Formulation Study for TG101348
  • The formulation study for N-tert-Butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide di-HCl monohydrate salt was conducted.
  • N-tert-Butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide di-HCl monohydrate salt compatibility with capsule shells
  • It was unknown whether the caustic/acidic nature of N-tert-Butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide di-HCl monohydrate salt (TG101348 di-HCl monohydrate) would be incompatible with capsules due to the potential caustic/acidic nature of a di-HCl salt.
  • Hard gelatin and hydroxypropylmethyl cellulose (HPMC) capsule shells (size #00250) were filled with 250 mg of TG101348 di-HCl monohydrate. The filled capsules were placed on accelerated stability (40° C./75% relative humidity (RH) and 25° C./60% RH). The capsules were packaged into 30 mL (1 oz) amber high density polyethylene (HDPE) bottles. A summary of the formulations and accelerated stability protocol for the stability study is shown in Table 16.
  • TABLE 16
    Formulations (i) Size #00 Hard Gelatin (White Opaque)
    Capsule Filled with 250 mg(1)
    (ii) Size #00 Hard HPMC (White Opaque)
    Capsule Filled with 250 mg(1)
    Lot Numbers 191-031A: Gelatin capsule fills stored at
    25° C./60% RH
    191-031B: Gelatin capsule fills stored at
    40° C./75% RH
    191-031C: HPMC capsule fills stored at
    25° C./60% RH
    191-031D: HPMC capsule fills stored at
    40° C./75% RH
    Storage Timepoint (weeks)
    Condition t = 0 t = 1 weeks t = 2 weeks t = 3 weeks
    40° C./75% RH 1 capsule 1 capsule 1 capsule 1 capsule
    25° C./60% RH 1 capsule 1 capsule 1 capsule
    Comments
    Storage Nalgene
    30 mL (1 oz) Amber Wide Mouth, HDPE
    Store capsules for each formulation at each condition
    in the same bottle
    Appearance/ 1 capsules per pull
    Assay/
    Impurities
    (1)a free base assay content of 83.78%. 250 mg of TG101348 is equivalent to 209.45 mg of free base.
  • It was found that TG101348 di-HCL monohydrate was compatible with hard gelatine capsules. No appreciable changes in attributes (appearance, assay, impurities) were observed over the timepoints of the study (t=1, 2, and 3 weeks).
  • Drug Substance Blend Compatibility with Fillers and Lubricants
  • A matrix of formulation blends was designed to study the compatibility of TG101348 di-HCl monohydrate with four fillers and two lubricants (Table 17). Blends were prepared at a scale of 2.5 g each by prescreening all components through a 500 μm sieve, blending all components except the lubricant using aTurbula T2B blender for 10 min at 22 rpm, screening the blend through a 500 μm sieve, blending for 10 min, adding lubricant (weight adjusted) and blending for 5 min The blends were manufactured and stored in 30 mL amber HDPE bottles under primary (60° C./ambient humidity) and backup conditions (40° C./75% RH, 25° C./60% RH, and 5° C.). A summary of the accelerated stability protocol is shown in Table 18. No appreciable changes in attributes (appearance, assay, impurities) were observed over the course of the study.
  • TABLE 17
    Formulation (% w/w)
    Ingredients 1 2 3 4 5 6 7 8 Control
    TG101348 50.46% 50.46% 50.46% 50.46% 50.46% 50.46% 50.46% 50.46% 100%
    di-HCl
    monohydrate (1)
    Lactose 48.54% 48.54%
    (Fast-Flo)
    Mannitol 48.54% 48.54%
    (Parteck M200)
    MCC (Avicel 49.04% 49.04%
    PH102)
    MCC (ProSolv 49.04% 49.04%
    90 HD)
    Magnesium  1.0%  1.0%  0.5%  0.5%
    Stearate
    Sodium Stearyl  1.0%  1.0%  0.5%  0.5%
    Fumarate
    (Pruv)
    (1) Based on TG101348 with a free-base assay content of 81.26% (equivalent to 41.00% w/w free base content in the formulation).
  • TABLE 18
    Timepoint (weeks)
    Storage t = t =
    Condition t = 0 t = 1 weeks t = 2 weeks 4 weeks 12 weeks
    60° C./ X X X X X
    ambient
    humidity
    40° C./75%
    RH
    25° C./60%
    RH
    5° C.
    Comments
    Storage Nalgene
    30 mL (1 oz) Amber Wide Mouth, HDPE
    Store blend for each formulation at each condition in the
    separate bottles
    Testing Visual appearance
    Assay
    Impurities
    ◯ = optional.
  • Powder-In-Capsule Development Excipient Selection
  • Excipient compatibility testing of dry blends of TG101348 di-HCl monohydrate with four fillers (lactose, mannitol, microcrystalline cellulose (MCC) Avicel PH102, and MCC ProSolv 90 HD) and two lubricants (magnesium stearate and sodium stearyl fumarate (Pruv) (Table 17) indicated no incompatibilities. Prosolv SMCC 90HD (i.e., silicified microcrystalline cellulose) and Lactose Fast-Flo (i.e., spray dried lactose monohydrate) were selected as fillers for further testing based on properties amenable to direct dry blending processes. Magnesium stearate (vegetable grade) and Pruv were selected as lubricants for further testing. All excipients are global regulatory approved for use in solid oral dosage forms (USA, European Union, Japan).
  • Suitability for Dry Powder Process Development
  • True densities of TG101348 di-HCl monohydrate having particles of rounded/granular appearance, silicified microcrystalline cellulose (Prosolv SMCC 90HD), and sodium stearyl fumarate (Pruv) were measured using a helium pycnometer (Micromeritics Accupyc 1340). The true density of the drug substance and excipients (fillers/diluents) appeared well matched.
  • Capsule Formulations
  • A “matrix” of prototype capsule formulations for stability evaluation was designed, and is summarized in Table 19. Two dosage strengths were selected, 10 and 125 mg.
  • TABLE 19
    Prototype Formulation
    P1 P2 P3 P4 P5 P6 P7 P8
    Dry Blend
    Component % w/w composition
    TG101348 49.37 49.37 49.37 49.37 49.37 49.37 49.37 49.37
    di-HCl
    monohydrate (1)
    Prosolv SMCC 50.13 50.13 50.13 50.13
    90HD
    Lactose 49.63 49.63 49.63 49.63
    Fast-Flo
    Magnesium 0.5 0.5 0.5 0.5 1.0 1.0 1.0 1.0
    Stearate
    Total
    100% 100% 100% 100% 100% 100% 100% 100%
    Capsule Fill
    Capsule Type Gelatin Gelatin HPMC HPMC Gelatin Gelatin HPMC HPMC
    Size Size Size Size Size Size Size Size
    #00 #00 #00 #00 #00 #00 #00 #00
    Capsule Color White White White White White White White White
    Opaque Opaque Opaque Opaque Opaque Opaque Opaque Opaque
    Capsule Fill 24.39 mg 304.86 mg 24.39 mg 304.86 mg 24.39 mg 304.86 mg 24.39 mg 304.86 mg
    blend fill blend fill blend fill blend fill blend fill blend fill blend fill blend fill
    Dose
    Dose
      10 mg   125 mg   10 mg   125 mg   10 mg   125 mg   10 mg   125 mg
    (mg, i.e.,
    free base)
    (1) Based on a free-base assay content of 83.05% (equivalent to 41.00% w/w free base content in the formulation).
  • Accelerated Stability Testing Protocol
  • Table 20 summarizes the accelerated stability protocol applied for the capsule prototypes. No appreciable changes in attributes (appearance, assay, impurities, in vitro dissolution) were observed over the timepoints of the study (t=1, 2, 4, and 8 weeks at 40° C./75% RH and 25° C./60% RH). Based on these results, Prototypes P2 and P6 were selected for further evaluation.
  • TABLE 20
    Storage Timepoint (weeks)
    Condition t = 0 2 weeks 4 weeks 8 weeks Contingency
    40° C./75% RH 16 capsules 5 capsules 5 capsules 5 capsules 7 capsules
    25° C./60% RH 5 capsules 5 capsules 5 capsules 7 capsules
    Comments
    Storage Store each pull separately into individual bottles
    Nalgene
    30 mL (1 oz) Amber Wide Mouth, HDPE
    Appearance/ 2 capsules per pull
    Assay/
    Impurities
    In vitro 3 capsules per pull
    Dissolution
    Notes
    16 capsules for t = 0 to include support for in vitro dissolution
    methodology development
  • In Vitro Dissolution Performance Considerations
  • On in vitro dissolution testing, prototype formulations filled into gelatin capsule shells (P1, P2, P5, P6) demonstrated >85% drug release within 15 minutes. Prototypes formulations filled into HPMC capsule shells (e.g., P3, P4, P7, P8) typically demonstrated <60% drug release after 60 minutes. Prototypes in HPMC capsules were therefore not progressed beyond t=0 testing.
  • Absorption Enhancer Capsule Development
  • TG101348 is on the borderline between “low” and “high” permeability based on caco-2 permeability data. In addition, bioavailability in multiple species was typically 20-25%. Therefore, it was not known whether an “absorption enhancer” would be required in the formulation to achieve adequate bioavailability.
  • Excipient Selection
  • Based on excipient compatibility described above, silicified microcrystalline cellulose (Prosolv SMCC 90HD) was used as the primary filler/carrier excipient for the absorption enhancement formulation. Four absorption enhancement excipient candidates were selected for further testing (Table 21).
  • TABLE 21
    Absorption Enhancement Absorption Enhancement
    Excipient Excipient Manufacturer Mechanism
    Vitamin E D-α-tocopheryl Eastman Chemical Lipid excipient (surfactant),
    polyethylene glycol succinate Company P-glycoprotein (PGP)
    (Vitamin E TPGS) inhibitor
    Gelucire 44/14 Gattefossé S. A. Lipid excipient (amphiphile)
    (PEG-32 glyceryl laurate)
    Pluronic F127 BASF Chemical Company Polymeric amphiphilic
    surfactant/
    micellar solubilization/
    P-glycoprotein (PGP)
    inhibitor
    Glyceryl monostearate 40-55 Gattefossé S.A. Lipid excipient (emulsifier)
    P-glycoprotein (PGP)
    inhibitor
  • Formulations and Manufacturing Processes
  • Table 22 summarizes the absorption enhancement formulations tested. Melt granulation, as opposed to a direct blend manufacturing process, was selected to produce the formulation.
  • TABLE 22
    Absorption Enhancer Formulations
    A B C D
    Component % w/w composition
    Granules
    TG101348 di-HCl 50.17% 50.17% 50.17% 50.17%
    monohydrate(1)
    Vitamin E TPGS 25.00%
    Gelucire 44/14 25.00%
    Pluronic F127  3.33%
    Glyceryl  3.33%
    monostearate
    ProSolv SMCC 24.33% 24.33% 46.00% 46.00%
    90HD
    Magnesium Stearate  0.50%  0.50%  0.50%  0.50%
    Capsule Fill
    Capsule Type Gelatin Size Gelatin Size Gelatin Size Gelatin Size
    #00 #00 #00 #00
    Capsule Color White Opaque White Opaque White Opaque White Opaque
    Capsule Fill 300 mg 300 mg 300 mg 300 mg
    of granules of granules of granules of granules
    Dose
    Dose 125 mg 125 mg 125 mg 125 mg
    (mg, i.e. free base)
    (1)Based on TG101348 with a free-base assay content of 83.05% (equivalent to 41.67% w/w free base content in the formulation).
  • Crossover Beagle Dog PK Study
  • A crossover beagle dog PK study was performed testing five formulations: an oral solution as described below, two capsule formulations without absorption enhancer and two capsule formulations with absorption enhancer.
  • Five beagle dogs were dosed with each formulation at a TG101348 dose of 125 mg, or approximately 11 mg/kg based on mean body weights, with a ‘washout’ of one week in between doses. The formulations administered are summarized in Table 23.
  • TABLE 23
    Dose Dose Matrix
    Phase Dose Formulation Strength Amount Collected
    1 Oral solution Aqueous, 125 mg 20 mL Plasma
    0.5% MC,
    6.25 mg/mL
    2 Prototype Capsule fill 125 mg One Plasma
    Capsule P2 containing capsule
    dry blend of:
    49.37% TG101348
    Lot K-18
    50.13% Prosolv
    SMCC 90HD
    0.5% Magnesium
    Stearate
    3 Prototype Capsule fill 125 mg One Plasma
    Capsule P6 containing capsule
    dry blend of:
    49.37% TG101348
    Lot K-18
    49.63% Lactose
    Fast-Flo
    1.0% Magnesium
    Stearate
    4 Prototype Capsule fill 125 mg One Plasma
    Capsule A containing capsule
    granules composed
    of:
    50.17% TG101348
    Lot K-18
    3.33% Glyceryl
    Monostearate
    46.00% Prosolv
    SMCC 90HD
    0.5% Magnesium
    Stearate
    5 Prototype Capsule fill 125 mg One Plasma
    Capsule D containing capsule
    granules composed
    of:
    50.17% TG101348
    Lot K-18
    25.00% Vitamin E
    TPGS
    24.33% Prosolv
    SMCC 90HD
    0.5% Magnesium
    Stearate
  • All four capsule formulations displayed immediate release characteristics with a demonstration of bioequivalence to the reference solution dose. Therefore, despite the boarderline permeability in human caco-2 cells and the 20-25% bioavailability in various animal species, capsule formulations without an absorption enhancer formulation demonstrated immediate release characteristics.
  • Process Development Drug Substance Particle Morphology
  • Different particle morphology, from rounded, granular particles (mean particle size 25 μm), to that of small needles (mean particle size≈7-10 μm) was found between different lots of the drug substance. The needle form was found to be highly static, which could negatively affect drug product manufacturing and also negatively affect drug product content uniformity.
  • Dry Granulation Process
  • The initial formulation, developed with the drug substance having rounded, granular particles with a mean particle size of 25 μm was 50:50 ratio by weight of TG101348 drug substance and filler, with 0.5% w/w lubricant. Prior to roller compaction, a blend of drug, filler, and lubricant was prepared. As described herein, the drug substance was passed through a co-mil to de-agglomerate prior to blending with formulation excipients. The drug substance having small needles demonstrated a high tendency to agglomerate on storage. After deagglomerating the drug substance having small needles, significant reagglomeration or ‘clumping’ would occur almost instantaneously. This reagglomeration was significantly reduced through blending the drug with lubricant prior to milling.
  • The initial formulation of TG101348 di-HCL monohydrate comprised about a 50:50 ratio by weight of TG101348 drug substance and filler, with 0.5% w/w lubricant. This formulation exhibited a poor flow and significant sticking to the metal rolls within the roller compactor.
  • The amount of magnesium stearate lubricant could be increased in the formulation, however increasing the concentration within the formulation could adversely affect drug release kinetics. The lubricant sodium laurel fumarate was also shown to be compatible with TG101348 di-HCl monodyrate and is less hygroscopic than magnesium stearate and was added (at a weight ratio of 2.0% w/w) instead of magnesium stearate which minimized sticking of the formulation to the metal rolls of the roller compactor. However, powder flow remained poor.
  • The ratio of TG101348 drug substance to filler was reduced from about 50:50 to about 40:60. The lubricant (Prov) content was also reduced to 1% w/w which provided acceptable flow and minimal sticking within the roller compactor.
  • Development of Multi-Dose Formulations
  • On capsule hand-filling trials, with aggressive tamping, into size #00 gelatin capsule shells, approximately 600 mg of granules appeared to be the maximum achievable fill. With a drug substance loading of 40% w/w in the formulation, and with the TG101348 di-HCL monohydrate batch comprising 83.78% free base content, it therefore appeared that an upper capsule strength of 200 mg strength was feasible.
  • The dry granulation process developed as described herein to produce a particles having intragranular TG101348 di-HCL monohydrate and sodium laurel fumarate allowed the preparation of a range of capsule dosages using dry blending processes.
  • The mean granule size was approximately 300 μm and the silicified microcrystalline cellulose mean particle size was approximately 100 μm. Therefore, 40 mg and 10 mg capsule strength formulations were produced through dilution of the granules with extragranular silicified microcrystalline cellulose. The general, particles sizes within a The size of the granules and the extragranular silicified microcrystalline cellulose are similar enough to allow homogeneous mixing.
  • 40 mg strength capsules were produced using a formulation comprising comparable fill volume to the 200 mg formulation, within the same capsule shell (size #00 hard gelatin capsule). For the 10 mg strength capsules, a common blend with the 40 mg capsule strength formulation was used by filing a smaller capsule size.
  • Oral Solution Formulation
  • An oral solution formulation was developed that contained the drug substance, 0.5% methylcellulose (MC) and 0.05% Tween 80. A pH-stability study was conducted at 60° C. on formulations passed through a 0.22 μm polyethersulfone (PES) filter. No appreciable changes in attributes (appearance, assay, impurities) were observed over the course of the study (14 days). A second oral solution formulation was developed that contained the drug substance and 0.5% MC. The second oral solution formulation was used in the dog PK study.
  • Although the foregoing examples have been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention.

Claims (30)

1-69. (canceled)
70. A pharmaceutical formulation comprising a compound that is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide or a pharmaceutically acceptable salt and/or hydrate thereof, characterized in that the formulation provides:
a steady state patient Cmax of the compound that is achieved within 2 to 4 hours post administration; and/or
a steady state patient Cmax of the compound between 1717.33 ng/mL and 3886.67 ng/mL post administration.
71. The formulation of claim 70, characterized in that the formulation provides a Cmax of the compound that is achieved within 2 to 4 hours post administration.
72. The formulation of claim 70, characterized in that the formulation provides a Cmax of the compound between 1717.33 ng/mL and 3886.67 ng/mL post administration.
73. The formulation of claim 70, wherein the formulation further comprises a filler.
74. The formulation of claim 73, wherein the filler is a microcrystalline cellulose.
75. The formulation of claim 70, wherein the formulation further comprises a lubricant.
76. The formulation of claim 73, wherein the formulation further comprises a lubricant.
77. The formulation of claim 74, wherein the formulation further comprises a lubricant.
78. The formulation of claim 75, wherein the lubricant is sodium stearyl fumarate.
79. The formulation of claim 76, wherein the lubricant is sodium stearyl fumarate.
80. The formulation of claim 77, wherein the lubricant is sodium stearyl fumarate.
81. The formulation of claim 70, wherein the formulation comprises about 400 mg of the compound.
82. The formulation of claim 73, wherein the formulation comprises about 400 mg of the compound.
83. The formulation of claim 74 wherein the formulation comprises about 400 mg of the compound.
84. The formulation of claim 75, wherein the formulation comprises about 400 mg of the compound.
85. The formulation of claim 76, wherein the formulation comprises about 400 mg of the compound.
86. The formulation of claim 77, wherein the formulation comprises about 400 mg of the compound.
87. The formulation of claim 78, wherein the formulation comprises about 400 mg of the compound.
88. The formulation of claim 79, wherein the formulation comprises about 400 mg of the compound.
89. The formulation of claim 80, wherein the formulation comprises about 400 mg of the compound.
90. The formulation of claim 70, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
91. The formulation of claim 73, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonamide dihydrochloride monohydrate.
93. The formulation of claim 74, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]bezenesulfonamide dihydrochloride monohydrate.
94. The formulation of claim 75, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]bezenesulfonamide dihydrochloride monohydrate.
95. The formulation of claim 76, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]bezenesulfonamide dihydrochloride monohydrate.
96. The formulation of claim 77, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]bezenesulfonamide dihydrochloride monohydrate.
97. The formulation of claim 78, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]bezenesulfonamide dihydrochloride monohydrate.
98. The formulation of claim 79, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]benzenesulfonami de dihydrochloride monohydrate.
99. The formulation of claim 80, wherein the compound is N-tert-butyl-3-[(5-methyl-2-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}pyrimidin-4-yl)amino]bezenesulfonamide dihydrochloride monohydrate.
US17/220,073 2010-11-07 2021-04-01 Compositions and methods for treating myelofibrosis Abandoned US20210244735A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/220,073 US20210244735A1 (en) 2010-11-07 2021-04-01 Compositions and methods for treating myelofibrosis

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US41092410P 2010-11-07 2010-11-07
PCT/US2011/059643 WO2012061833A1 (en) 2010-11-07 2011-11-07 Compositions and methods for treating myelofibrosis
US13/888,096 US10391094B2 (en) 2010-11-07 2013-05-06 Compositions and methods for treating myelofibrosis
US16/549,043 US20190381041A1 (en) 2010-11-07 2019-08-23 Compositions and methods for treating myelofibrosis
US17/220,073 US20210244735A1 (en) 2010-11-07 2021-04-01 Compositions and methods for treating myelofibrosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/549,043 Division US20190381041A1 (en) 2010-11-07 2019-08-23 Compositions and methods for treating myelofibrosis

Publications (1)

Publication Number Publication Date
US20210244735A1 true US20210244735A1 (en) 2021-08-12

Family

ID=46024747

Family Applications (4)

Application Number Title Priority Date Filing Date
US13/888,096 Active 2032-05-29 US10391094B2 (en) 2010-11-07 2013-05-06 Compositions and methods for treating myelofibrosis
US13/888,073 Abandoned US20130252988A1 (en) 2010-11-07 2013-05-06 Compositions and methods for treating myelofibrosis
US16/549,043 Pending US20190381041A1 (en) 2010-11-07 2019-08-23 Compositions and methods for treating myelofibrosis
US17/220,073 Abandoned US20210244735A1 (en) 2010-11-07 2021-04-01 Compositions and methods for treating myelofibrosis

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US13/888,096 Active 2032-05-29 US10391094B2 (en) 2010-11-07 2013-05-06 Compositions and methods for treating myelofibrosis
US13/888,073 Abandoned US20130252988A1 (en) 2010-11-07 2013-05-06 Compositions and methods for treating myelofibrosis
US16/549,043 Pending US20190381041A1 (en) 2010-11-07 2019-08-23 Compositions and methods for treating myelofibrosis

Country Status (34)

Country Link
US (4) US10391094B2 (en)
EP (2) EP2635282B1 (en)
JP (1) JP6133211B2 (en)
KR (4) KR20200083676A (en)
CN (2) CN108125923A (en)
AU (2) AU2010363329A1 (en)
BR (1) BR112013011184A2 (en)
CA (2) CA2816957A1 (en)
CL (1) CL2013001252A1 (en)
CO (1) CO6801724A2 (en)
DK (1) DK2635282T3 (en)
DO (1) DOP2013000097A (en)
EC (1) ECSP13012658A (en)
ES (1) ES2930650T3 (en)
HR (1) HRP20221269T1 (en)
HU (1) HUE060254T2 (en)
IL (1) IL226101A (en)
LT (1) LT2635282T (en)
MA (1) MA34723B1 (en)
MX (1) MX360246B (en)
MY (1) MY161164A (en)
NI (1) NI201300038A (en)
NZ (1) NZ611363A (en)
PE (1) PE20140389A1 (en)
PL (1) PL2635282T3 (en)
PT (1) PT2635282T (en)
RS (1) RS63996B1 (en)
RU (1) RU2616262C2 (en)
SG (1) SG190134A1 (en)
SI (1) SI2635282T1 (en)
TW (1) TWI531389B (en)
UA (1) UA114076C2 (en)
WO (2) WO2012060847A1 (en)
ZA (1) ZA201303423B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190381041A1 (en) * 2010-11-07 2019-12-19 Impact Biomedicines, Inc. Compositions and methods for treating myelofibrosis
US11400092B2 (en) 2018-09-25 2022-08-02 Impact Biomedicines, Inc. Methods of treating myeloproliferative disorders

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8604042B2 (en) 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
WO2013059548A1 (en) * 2011-10-19 2013-04-25 Sanofi Compositions and methods for treating cancer using jak2 inhibitor
UA114948C2 (en) * 2013-03-06 2017-08-28 Новартіс Аг Formulations of organic compounds
CN104771761A (en) * 2015-03-19 2015-07-15 深圳国源国药有限公司 Novel pharmaceutical auxiliary material namely silicified microcrystalline cellulose and preparation method thereof
WO2017018385A1 (en) * 2015-07-24 2017-02-02 公立大学法人名古屋市立大学 Method for assisting diagnosis of conditions of myelofibrosis, method for assisting prediction of prognosis, method for monitoring therapeutic effect, and marker and device to be used therein
CN105919955A (en) * 2016-06-13 2016-09-07 佛山市腾瑞医药科技有限公司 Ruxolitinib preparation and application thereof
US11384069B2 (en) 2018-01-16 2022-07-12 Shenzhen Targetrx, Inc. Diphenylaminopyrimidine compound for inhibiting kinase activity
EA202190751A1 (en) * 2018-09-25 2021-06-28 Импакт Биомедисинс, Инк. METHODS FOR TREATMENT OF MYELOPROLIFERATIVE DISORDERS
FR3092581A1 (en) * 2019-02-12 2020-08-14 Impact Biomedicines, Inc CRYSTALLINE FORMS OF A JAK2 INHIBITOR
US20230250068A1 (en) * 2019-02-12 2023-08-10 Inpact Biomedicines, Inc. Crystalline forms of a jak2 inhibitor
US11306062B2 (en) 2019-10-29 2022-04-19 Johnson Matthey Public Limited Forms of fedratinib dihydrochloride
DK4076404T3 (en) * 2019-12-20 2024-01-22 Intervet Int Bv PHARMACEUTICAL PYRAZOLE COMPOSITION
WO2021207051A1 (en) * 2020-04-06 2021-10-14 Bristol-Myers Squibb Company Methods of treating acute respiratory disorders
WO2022132933A1 (en) 2020-12-16 2022-06-23 Impact Biomedicines, Inc. Dosing of fedratinib
WO2022182857A1 (en) 2021-02-25 2022-09-01 Impact Biomedicines, Inc. Use of a bet inhibitor alone or in combination with fedratinib or ruxolitinib for treating a hematological malignancy such as myelofibrosis
WO2023044297A1 (en) 2021-09-14 2023-03-23 Impact Biomedicines, Inc. Fedratinib for treating myeloproliferative disorders

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060210627A1 (en) * 2005-01-18 2006-09-21 Sabine Pfeffer Direct compression formulation and process

Family Cites Families (253)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2003199A (en) 1930-05-31 1935-05-28 Johnson Frank James Automatic coal stoker
US2003149A (en) 1931-05-22 1935-05-28 Autographic Register Co Manifolding
US2003065A (en) 1931-06-20 1935-05-28 John R Ditmars Composition for coating sheets, fibrous stocks, and the like
US2004102A (en) 1932-02-24 1935-06-11 Daniel A Dickey Hollow steel propeller construction
US2004138A (en) 1932-11-30 1935-06-11 Byers A M Co Method of making wrought iron pipe
US2002165A (en) 1933-07-08 1935-05-21 Charles A Winslow Air cleaner
US2003187A (en) 1933-10-02 1935-05-28 Frederick H Shaw Automobile radio device
US2003166A (en) 1933-10-26 1935-05-28 Zancan Ottavio Front drive for motor cars
US2001051A (en) 1933-12-01 1935-05-14 Angelina Mariani Tamperproof meter box with cutout control and fuse drawer for electric meters
US2004092A (en) 1933-12-15 1935-06-11 John L Chaney Device for indicating the temperature of liquids
US2003060A (en) 1934-04-02 1935-05-28 Ernest L Heckert Thermostatic controlling device
US2667486A (en) 1951-05-24 1954-01-26 Research Corp 2,4-diamino pteridine and derivatives
IE38778B1 (en) 1973-02-02 1978-05-24 Ciba Geigy New 1,2,4-benzotriazine derivatives
AU535258B2 (en) 1979-08-31 1984-03-08 Ici Australia Limited Benzotriazines
DE3205638A1 (en) 1982-02-17 1983-08-25 Hoechst Ag, 6230 Frankfurt Trisubstituted pyrimidine-5-carboxylic acids and their derivatives, processes for their preparation, and their use as pesticides
US4490289A (en) 1982-09-16 1984-12-25 Hoffmann-La Roche Inc. Homogeneous human interleukin 2
US5616584A (en) 1986-09-25 1997-04-01 Sri International 1,2,4-benzotriazine oxides as radiosensitizers and selective cytotoxic agents
JP2598100B2 (en) 1988-08-31 1997-04-09 キヤノン株式会社 Electrophotographic photoreceptor
US5214059A (en) 1989-07-03 1993-05-25 Hoechst-Roussel Pharmaceuticals Incorporated 2-(aminoaryl) indoles and indolines as topical antiinflammatory agents for the treatment of skin disorders
US5776502A (en) 1989-07-18 1998-07-07 Oncogene Science, Inc. Methods of transcriptionally modulating gene expression
US5665543A (en) 1989-07-18 1997-09-09 Oncogene Science, Inc. Method of discovering chemicals capable of functioning as gene expression modulators
US5041430A (en) * 1989-09-18 1991-08-20 Du Pont Mereck Pharmaceutical Company Oral anticoagulant/platelet inhibitor low dose formulation
JPH03127790A (en) 1989-10-11 1991-05-30 Morishita Pharmaceut Co Ltd N-(1h-tetrazol-5-yl)-2-anilino-5-pyrimidinecarboxamides and synthetic intermediate therefor
GB9003553D0 (en) 1990-02-16 1990-04-11 Ici Plc Herbicidal compositions
JP2839106B2 (en) 1990-02-19 1998-12-16 キヤノン株式会社 Electrophotographic photoreceptor
GB9012592D0 (en) 1990-06-06 1990-07-25 Smithkline Beecham Intercredit Compounds
FR2665159B1 (en) 1990-07-24 1992-11-13 Rhone Poulenc Sante NEW PYRIDINE AND QUINOLEIN DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
MY107955A (en) 1990-07-27 1996-07-15 Ici Plc Fungicides.
GB9016800D0 (en) 1990-07-31 1990-09-12 Shell Int Research Tetrahydropyrimidine derivatives
DE4025891A1 (en) 1990-08-16 1992-02-20 Bayer Ag PYRIMIDYL-SUBSTITUTED ACRYLIC ACID ESTERS
JPH05345780A (en) 1991-12-24 1993-12-27 Kumiai Chem Ind Co Ltd Pyrimidine or triazine derivative and herbicide
HUT63941A (en) 1992-05-15 1993-11-29 Hoechst Ag Process for producing 4-alkyl-substituted pyrimidine-5-carboxanilide derivatives, and fungicidal compositions comprising same
US5438130A (en) 1993-01-15 1995-08-01 Cambridge Neuroscience, Inc. Fucosylated guanosine disulfates as excitatory amino acid antagonists
JPH0741461A (en) 1993-05-27 1995-02-10 Eisai Co Ltd Sulfonic acid ester derivative
JPH0782183A (en) 1993-09-09 1995-03-28 Canon Inc Liquid crystal instermediate compound, liquid crystal compound, high molecular liquid crystal compound, liquid crystal copolymer, composition thereof, liquid crystal element and recorder
DE4338704A1 (en) 1993-11-12 1995-05-18 Hoechst Ag Stabilized oligonucleotides and their use
US5530000A (en) 1993-12-22 1996-06-25 Ortho Pharmaceutical Corporation Substituted pyrimidinylaminothiazole derivatives useful as platelet aggreggation inhibitors
GB9506466D0 (en) 1994-08-26 1995-05-17 Prolifix Ltd Cell cycle regulated repressor and dna element
US5597826A (en) 1994-09-14 1997-01-28 Pfizer Inc. Compositions containing sertraline and a 5-HT1D receptor agonist or antagonist
DE19502912A1 (en) 1995-01-31 1996-08-01 Hoechst Ag G-Cap Stabilized Oligonucleotides
US6326487B1 (en) 1995-06-05 2001-12-04 Aventis Pharma Deutschland Gmbh 3 modified oligonucleotide derivatives
EP0850228A1 (en) 1995-09-01 1998-07-01 Signal Pharmaceuticals, Inc. Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions
GB9523675D0 (en) 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
CA2241724A1 (en) 1995-12-29 1997-07-10 Smithkline Beecham Corporation Vitronectin receptor antagonists
JPH09274290A (en) 1996-02-07 1997-10-21 Fuji Photo Film Co Ltd Developing solution and processing method for silver halide photographic material
JP4212112B2 (en) 1996-02-23 2009-01-21 イーライ・リリー・アンド・カンパニー Non-peptidyl vasopressin V1a antagonist
DE59707681D1 (en) 1996-10-28 2002-08-14 Rolic Ag Zug Crosslinkable, photoactive silane derivatives
KR100503701B1 (en) 1996-11-20 2005-07-26 인트로겐 테라페티스, 인코퍼레이티드 An improved method for the production and purification of adenoviral vectors
JP3734903B2 (en) 1996-11-21 2006-01-11 富士写真フイルム株式会社 Development processing method
JPH10153838A (en) 1996-11-22 1998-06-09 Fuji Photo Film Co Ltd Method for processing silver halide photographic sensitive material
JP3720931B2 (en) 1996-11-26 2005-11-30 富士写真フイルム株式会社 Processing method of silver halide photographic light-sensitive material
CN1240010A (en) 1996-12-04 1999-12-29 金伯利-克拉克环球有限公司 Method for making wet strength paper
US5935383A (en) 1996-12-04 1999-08-10 Kimberly-Clark Worldwide, Inc. Method for improved wet strength paper
WO1998028282A2 (en) 1996-12-23 1998-07-02 Du Pont Pharmaceuticals Company OXYGEN OR SULFUR CONTAINING 5-MEMBERED HETEROAROMATICS AS FACTOR Xa INHIBITORS
JPH10207019A (en) 1997-01-22 1998-08-07 Fuji Photo Film Co Ltd Method for processing silver halide photographic sensitive material
JPH10213820A (en) 1997-01-31 1998-08-11 Canon Inc Liquid crystal element and liquid crystal device
DE59807348D1 (en) 1997-02-05 2003-04-10 Rolic Ag Zug Photocrosslinkable silane derivatives
GB9705361D0 (en) 1997-03-14 1997-04-30 Celltech Therapeutics Ltd Chemical compounds
JPH10260512A (en) 1997-03-19 1998-09-29 Fuji Photo Film Co Ltd Method for processing silver halide photosensitive material
JPH10310583A (en) 1997-05-09 1998-11-24 Mitsubishi Chem Corp Pyridone azo-based compound, coloring matter for heat-sensitive transfer and heat-sensitive transfer material using the same
US6070126A (en) 1997-06-13 2000-05-30 William J. Kokolus Immunobiologically-active linear peptides and method of identification
TW436485B (en) 1997-08-01 2001-05-28 American Cyanamid Co Substituted quinazoline derivatives
US6635626B1 (en) 1997-08-25 2003-10-21 Bristol-Myers Squibb Co. Imidazoquinoxaline protein tyrosine kinase inhibitors
US6022884A (en) 1997-11-07 2000-02-08 Amgen Inc. Substituted pyridine compounds and methods of use
AU1507199A (en) 1997-12-15 1999-07-05 Yamanouchi Pharmaceutical Co., Ltd. Novel pyrimidine-5-carboxamide derivatives
BR9813835A (en) 1997-12-22 2000-10-10 Du Pont Pharm Co Compound, pharmaceutical composition and method of treatment or prevention of a thromboembolic disorder
NZ506417A (en) 1998-02-17 2003-05-30 Tularik Inc Anti-viral pyrimidine derivatives
BRPI9909191B8 (en) 1998-03-27 2021-07-06 Janssen Pharmaceutica Nv hiv inhibitor pyrimidine derivatives, their use and pharmaceutical composition comprising them
SI0945443T1 (en) 1998-03-27 2003-08-31 Janssen Pharmaceutica N.V. HIV inhibiting pyrimidine derivatives
US6685938B1 (en) 1998-05-29 2004-02-03 The Scripps Research Institute Methods and compositions useful for modulation of angiogenesis and vascular permeability using SRC or Yes tyrosine kinases
DE19830181C1 (en) 1998-07-06 1999-09-30 Inter Control Koehler Hermann Multi-pole electrical plug-in coupling for domestic electrical appliance e.g. electric kettle or iron
US6136971A (en) 1998-07-17 2000-10-24 Roche Colorado Corporation Preparation of sulfonamides
US6378526B1 (en) 1998-08-03 2002-04-30 Insite Vision, Incorporated Methods of ophthalmic administration
US6288082B1 (en) 1998-09-29 2001-09-11 American Cyanamid Company Substituted 3-cyanoquinolines
ES2211175T3 (en) 1998-09-29 2004-07-01 Wyeth Holdings Corporation INHIBITORS OF TYPEOSIN KINASE PROTEINS BASED ON REPLACED 3-CYANOQUINOLINS.
CN1144786C (en) 1998-09-29 2004-04-07 美国氰胺公司 Substituted 3-cyanoquinolines as protein tyrosine kinases inhibitors
US6297258B1 (en) 1998-09-29 2001-10-02 American Cyanamid Company Substituted 3-cyanoquinolines
CA2320730A1 (en) 1998-12-23 2000-07-06 Renhua Li Thrombin or factor xa inhibitors
GB9828511D0 (en) 1998-12-24 1999-02-17 Zeneca Ltd Chemical compounds
EP1150955A2 (en) 1999-02-04 2001-11-07 Millennium Pharmaceuticals, Inc. G-protein coupled heptahelical receptor binding compounds and methods of use thereof
MXPA01010292A (en) 1999-04-15 2002-10-23 Squibb Bristol Myers Co Cyclic protein tyrosine kinase inhibitors.
ATE264328T1 (en) 1999-04-21 2004-04-15 Wyeth Corp SUBSTITUTED 3-CYANO-(1.7), (1.5) AND (1.8)NAPHTHHYRIDINE INHIBITORS OF TYROSINE KINASES
AU5020400A (en) 1999-05-20 2000-12-12 E.I. Du Pont De Nemours And Company Heteroaryloxypyrimidine insecticides and acaricides
US6710221B1 (en) 1999-06-15 2004-03-23 Kimberly-Clark Worldwide, Inc. Absorbent articles incorporating color change graphics
BR0012046A (en) 1999-07-01 2002-05-14 Ajinomoto Kk Heterocyclic compound, pharmaceutical composition, inhibitor of activation of ap-1 or an inhibitor of activation of nf-kappab, inhibitor of the production of inflammatory cytokine, and inhibitor of the production of matrix metalloprotease or inhibitor of the expression of inflammatory cell adhesion factor
WO2001007027A2 (en) 1999-07-22 2001-02-01 Vertex Pharmaceuticals Incorporated Pyrimidine derivatives for the treatment of viral diseases
TR200200099T2 (en) 1999-07-23 2002-06-21 Shionogi &Co., Ltd. Th2 differentiation inhibitors
AU6016000A (en) 1999-07-23 2001-02-13 Shionogi & Co., Ltd. Tricyclic compounds and drug compositions containing the same
CZ2002474A3 (en) 1999-08-12 2003-02-12 American Cyanamid Company NSAID and EFGR kinase inhibitor containing composition for the treatment or inhibition of colonic polyps and colorectal cancer
US6093838A (en) 1999-08-16 2000-07-25 Allergan Sales, Inc. Amines substituted with a dihydro-benzofuranyl or with a dihydro-isobenzofuranyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
US6127382A (en) 1999-08-16 2000-10-03 Allergan Sales, Inc. Amines substituted with a tetrahydroquinolinyl group an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
PL355639A1 (en) 1999-09-10 2004-05-04 Merck & Co, Inc. Tyrosine kinase inhibitors
GB9922171D0 (en) 1999-09-21 1999-11-17 Zeneca Ltd Chemical compounds
JP2001089412A (en) 1999-09-22 2001-04-03 Otsuka Pharmaceut Co Ltd Benzene derivative or its pharmaceutically acceptable salt
US6506769B2 (en) 1999-10-06 2003-01-14 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
EP1223170B1 (en) 1999-10-12 2005-12-28 Takeda Pharmaceutical Company Limited Pyrimidine-5-carboxamide compounds, process for producing the same and use thereof
EP1226123A1 (en) 1999-11-03 2002-07-31 Du Pont Pharmaceuticals Company Cyano compounds as factor xa inhibitors
GB9929988D0 (en) 1999-12-17 2000-02-09 Celltech Therapeutics Ltd Chemical compounds
ATE353320T1 (en) 1999-12-29 2007-02-15 Wyeth Corp TRICYCLIC PROTEIN KINASE INHIBITORS
US6638929B2 (en) 1999-12-29 2003-10-28 Wyeth Tricyclic protein kinase inhibitors
EP1294709A2 (en) 2000-01-28 2003-03-26 AstraZeneca AB Quinoline derivatives and their use as aurora 2 kinase inhibitors
US6153752A (en) 2000-01-28 2000-11-28 Creanova, Inc. Process for preparing heterocycles
US20020165244A1 (en) 2000-01-31 2002-11-07 Yuhong Zhou Mucin synthesis inhibitors
CA2400447C (en) 2000-02-17 2008-04-22 Amgen Inc. Kinase inhibitors
EP1261361B1 (en) 2000-02-18 2006-06-14 Yeda Research And Development Company, Ltd. Oral, nasal and pulmonary dosage formulations of copolymer 1 (glatiramer acetate)
EP1261327B1 (en) 2000-02-25 2005-04-27 F.Hoffmann-La Roche Ag Adenosine receptor modulators
GB0004887D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
ES2317889T3 (en) 2000-03-01 2009-05-01 Janssen Pharmaceutica Nv DERIVATIVES OF TIAZOLIL 2,4-DISPOSED.
GB0004890D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
WO2001064646A2 (en) 2000-03-01 2001-09-07 Tularik Inc. Hydrazones and analogs as cholesterol lowering agents
GB0004888D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
JP2001247411A (en) 2000-03-09 2001-09-11 Tomono Agrica Co Ltd Pest-controlling agent
BR0109165A (en) 2000-03-13 2003-04-22 American Cyanamid Co Cholemic polyps treatment method
US6613917B1 (en) 2000-03-23 2003-09-02 Allergan, Inc. Amines substituted with a dihydronaphthalenyl, chromenyl, or thiochromenyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
AR028261A1 (en) 2000-03-28 2003-04-30 Wyeth Corp TRICICLIC INHIBITORS OF PROTEIN QUINASA
US6608048B2 (en) 2000-03-28 2003-08-19 Wyeth Holdings Tricyclic protein kinase inhibitors
CN1431896A (en) 2000-04-04 2003-07-23 盐野义制药株式会社 Oily compsns. contg. highly fat-soluble drugs
AU2001244610A1 (en) 2000-04-05 2001-10-23 Shionogi And Co., Ltd. Oil-in-water microemulsions containing tricyclic compounds or preconcentrates thereof
US6471968B1 (en) 2000-05-12 2002-10-29 Regents Of The University Of Michigan Multifunctional nanodevice platform
DE10024622A1 (en) 2000-05-18 2001-11-22 Piesteritz Stickstoff New N-(2-pyrimidinyl)-(thio)phosphoric acid triamide compounds are urease inhibitors useful for preventing hydrolysis of urea fertilizers or for reducing release of ammonia in animal stalls
EP1170353B1 (en) 2000-07-06 2005-11-02 Fuji Photo Film Co., Ltd. Liquid crystal composition comprising liquid crystal molecules and aligment promoter
JP5000068B2 (en) 2000-08-11 2012-08-15 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 Heterocyclic compounds useful as inhibitors of tyrosine kinases
KR100876069B1 (en) 2000-09-15 2008-12-26 버텍스 파마슈티칼스 인코포레이티드 Pyrazole Compounds Useful as Protein Kinase Inhibitors and Pharmaceutical Compositions Comprising the Same
EP1578341A4 (en) 2000-10-11 2005-09-28 Tularik Inc Modulation of ccr4 function
US7253184B2 (en) 2000-11-02 2007-08-07 Astrazeneca Ab 4-Substituted quinolines as antitumor agents
AU2002210714A1 (en) 2000-11-02 2002-06-11 Astrazeneca Ab Substituted quinolines as antitumor agents
CA2430037A1 (en) 2000-11-20 2002-05-30 Michael S. South Substituted polycyclic aryl and heteroaryl pyridines useful for selective inhibition of the coagulation cascade
US20070026433A1 (en) * 2001-03-09 2007-02-01 Hildebrand William H Epitope testing using soluble HLA
WO2002053160A1 (en) 2000-12-29 2002-07-11 Alteon, Inc. Method for treating glaucoma ivb
WO2002053101A2 (en) 2000-12-29 2002-07-11 Alteon, Inc. Method for treating fibrotic diseases or other indications
JP2002221770A (en) 2001-01-24 2002-08-09 Fuji Photo Film Co Ltd Silver halide photographic sensitive material and method of processing the same
WO2002064096A2 (en) 2001-02-16 2002-08-22 Tularik Inc. Methods of using pyrimidine-based antiviral agents
IL157254A0 (en) 2001-02-23 2004-02-19 Merck & Co Inc N-substituted nonaryl-heterocyclic nmda/mr2b antagonists
AU2002247402A1 (en) 2001-03-23 2002-10-08 Chugai Seiyaku Kabushiki Kaisha Flt-1 ligands and their uses in the treatment of diseases regulatable by angiogenesis
ATE339416T1 (en) 2001-04-13 2006-10-15 Vertex Pharma INHIBITORS OF C-JUN N-TERMINAL KINASES (JNK) AND OTHER PROTEIN KINASES
EP1383760A1 (en) 2001-04-30 2004-01-28 Fujisawa Pharmaceutical Co., Ltd. Biarylcarboxamide compounds as apolipoprotein b inhibitors
CA2446879A1 (en) 2001-05-11 2002-11-21 Vertex Pharmaceuticals Incorporated 2,5-disubstituted pyridine, pyrimidine, pyridazine and 1, 2, 4-triazine derivatives for use as p38 inhibitors
WO2002094766A1 (en) 2001-05-18 2002-11-28 Nihon Nohyaku Co., Ltd. Phthalamide derivative, agricultural or horticultural insecticide, and use thereof
AU2002314252A1 (en) 2001-05-28 2002-12-09 Aventis Pharma S.A. Chemical derivatives and the use thereof as an anti-telomerase agent
JP4291135B2 (en) 2001-05-29 2009-07-08 バイエル・シエーリング・ファーマ アクチエンゲゼルシャフト CDK-inhibiting pyrimidines, their manufacture and use as pharmaceuticals
CA2449054C (en) 2001-05-30 2011-01-04 The Scripps Research Institute Integrin targeting liposome for nucleic acid delivery
DE60214703T2 (en) 2001-06-01 2007-09-13 Vertex Pharmaceuticals Inc., Cambridge THIAZOL COMPOUNDS SUITABLE AS INHIBITORS OF PROTEIN KINASES
CA2452603A1 (en) 2001-07-03 2003-01-16 Vertex Pharmaceuticals Incorporated Isoxazolyl-pyrimidines as inhibitors of src and lck protein kinases
US6861529B2 (en) 2001-07-06 2005-03-01 Pfizer Inc Cycloalkypyrrole-3-carboxylic acid derivatives and heterocycloalkylpyrrole-3-carboxylic acid derivatives
JO3429B1 (en) 2001-08-13 2019-10-20 Janssen Pharmaceutica Nv Hiv inhibiting pyrimidines derivatives
BR0212510A (en) 2001-09-14 2004-08-24 Methylgene Inc Histone Deacetylase Inhibitor, Compound and Composition
WO2003030909A1 (en) 2001-09-25 2003-04-17 Bayer Pharmaceuticals Corporation 2- and 4-aminopyrimidines n-substtituded by a bicyclic ring for use as kinase inhibitors in the treatment of cancer
DE10150615A1 (en) 2001-10-12 2003-04-30 Clariant Gmbh Process for organometallic production of organic intermediates
DE10150614A1 (en) 2001-10-12 2003-04-30 Clariant Gmbh Process for organometallic production of organic intermediates via halogen-metal exchange reactions
DE10150610A1 (en) 2001-10-12 2003-04-30 Clariant Gmbh Process for organometallic production of organic intermediates using amide bases
EP1453516A2 (en) 2001-10-17 2004-09-08 Boehringer Ingelheim Pharma GmbH & Co.KG Novel tri-substituted pyrimidines, method for production and use thereof as medicament
BR0213792A (en) 2001-11-01 2004-12-07 Janssen Pharmaceutica Nv Heteroaryl amines as glycogen synthase kinase 3beta inhibitors (gsk3 inhibitors)
JP2005507419A (en) 2001-11-01 2005-03-17 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Amide derivatives as glycogen synthase kinase 3 beta inhibitors
UA77469C2 (en) 2001-11-27 2006-12-15 White Holdings Corp 3-cyanoquinolines as egf-r and her2 kinase inhibitors
AU2002344567A1 (en) 2001-11-28 2003-06-10 Daiso Co., Ltd. Heterocyclic amide compounds as apolipoprotein b inhibitors
US20030187026A1 (en) 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
US20030166932A1 (en) 2002-01-04 2003-09-04 Beard Richard L. Amines substituted with a dihydronaphthalenyl, chromenyl, or thiochromenyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
WO2003057165A2 (en) 2002-01-04 2003-07-17 The Rockefeller University COMPOSITIONS AND METHODS FOR PREVENTION AND TREATMENT OF AMYLOID-β PEPTIDE-RELATED DISORDERS
TWI329105B (en) 2002-02-01 2010-08-21 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds and their uses
EP1472213B1 (en) 2002-02-05 2010-06-09 Wyeth a Corporation of the State of Delaware Process for the synthesis of n-acyl-2-amino-4-alkoxy-5-nitrobenzoic acids
WO2003066601A1 (en) 2002-02-08 2003-08-14 Smithkline Beecham Corporation Pyrimidine compounds
CA2477505A1 (en) 2002-03-01 2003-09-12 Smithkline Beecham Corporation Diamino-pyrimidines and their use as angiogenesis inhibitors
GB0206215D0 (en) 2002-03-15 2002-05-01 Novartis Ag Organic compounds
WO2003095448A1 (en) 2002-05-06 2003-11-20 Bayer Pharmaceuticals Corporation Pyridinyl amino pyrimidine derivatives useful for treating hyper-proliferative disorders
TW200406374A (en) 2002-05-29 2004-05-01 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
MY141867A (en) 2002-06-20 2010-07-16 Vertex Pharma Substituted pyrimidines useful as protein kinase inhibitors
AU2003245624A1 (en) 2002-07-02 2004-01-23 Kimberly-Clark Worldwide, Inc. Elastomeric adhesive compositions and laminates
US7361665B2 (en) 2002-07-09 2008-04-22 Vertex Pharmaceuticals Incorporated Inhibitors of c-Jun N-terminal kinases (JNK) and other protein kinases
SG176311A1 (en) 2002-07-29 2011-12-29 Rigel Pharmaceuticals Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
DK1525200T3 (en) 2002-08-02 2007-12-03 Ab Science 2- (3-aminoaryl) amino-4-aryl-thiazoles for disease treatment
WO2004016597A2 (en) 2002-08-14 2004-02-26 Vertex Pharmaceuticals Incorporated Protein kinase inhibitors and uses thereof
UY27939A1 (en) 2002-08-21 2004-03-31 Glaxo Group Ltd COMPOUNDS
DE10240262A1 (en) 2002-08-31 2004-03-11 Clariant Gmbh Production of aryllithium-electrophile reaction products of interest for the pharmaceutical and agrochemical industries comprises using an organolithium compound prepared by reacting an aryl halide with lithium
DE10240261A1 (en) 2002-08-31 2004-03-11 Clariant Gmbh Production of aryllithium-electrophile reaction products of interest for the pharmaceutical and agrochemical industries comprises using an organolithium compound prepared by reacting an aryl halide with lithium
JP4560483B2 (en) 2002-10-03 2010-10-13 ターゲジェン インコーポレーティッド Angiogenic substances and methods for their use
US20050282814A1 (en) 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
CA2500368A1 (en) 2002-10-04 2004-04-22 Caritas St. Elizabeth's Medical Center Of Boston, Inc. Inhibition of src for treatment of reperfusion injury related to revascularization
EP1553947A4 (en) 2002-10-21 2006-11-29 Bristol Myers Squibb Co Quinazolinones and derivatives thereof as factor xa inhibitors
US7262200B2 (en) 2002-10-25 2007-08-28 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
US7452911B2 (en) 2002-10-31 2008-11-18 Boehringer Ingelheim Pharma Gmbh & Co. Kg Alkyne compounds with MCH antagonistic activity and medicaments comprising these compounds
DE10250708A1 (en) 2002-10-31 2004-05-19 Boehringer Ingelheim Pharma Gmbh & Co. Kg New alkyne compounds having MCH antagonist activity and medicaments containing these compounds
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
US20050101576A1 (en) * 2003-11-06 2005-05-12 Novacea, Inc. Methods of using vitamin D compounds in the treatment of myelodysplastic syndromes
AU2003280188A1 (en) 2002-12-06 2004-06-30 Warner-Lambert Company Llc Benzoxazin-3-ones and derivatives thereof as inhibitors of pi3k
JP3837670B2 (en) 2002-12-12 2006-10-25 富士通株式会社 Data relay apparatus, associative memory device, and associative memory device utilization information retrieval method
US20050043344A1 (en) 2002-12-20 2005-02-24 Pharmacia Corporation Heteroarylalkanoic acids as integrin receptor antagonists derivatives
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
EP1572210A1 (en) 2002-12-20 2005-09-14 Pharmacia Corporation The r-isomer of beta amino acid compounds as integrin receptor antagonists derivatives
AU2003294142A1 (en) 2002-12-24 2004-07-22 Astrazeneca Ab Therapeutic quinazoline derivatives
US7144911B2 (en) 2002-12-31 2006-12-05 Deciphera Pharmaceuticals Llc Anti-inflammatory medicaments
EA010379B1 (en) 2003-02-07 2008-08-29 Янссен Фармацевтика Н.В. Hiv inhibiting 1,2,4 triazines
CA2513527C (en) 2003-02-07 2012-08-14 Janssen Pharmaceutica N.V. Pyrimidine derivatives for the prevention of hiv infection
WO2004071426A2 (en) 2003-02-11 2004-08-26 Kemia Inc. Compounds for the treatment of viral infection
CL2004000303A1 (en) 2003-02-20 2005-04-08 Tibotec Pharm Ltd COMPOUNDS DERIVED FROM PYRIMIDINS AND TRIAZINES; PREPARATION PROCESS; PHARMACEUTICAL COMPOSITION; AND ITS USE TO INHIBIT HIV REPLICATION.
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
US20050019395A1 (en) 2003-04-14 2005-01-27 Gabor Pragai Formulations of amlodipine maleate
US20040265375A1 (en) 2003-04-16 2004-12-30 Platteeuw Johannes J. Orally disintegrating tablets
EP1623269B2 (en) 2003-04-24 2022-08-31 CooperVision International Limited Hydrogel contact lenses and package systems and production methods for same
CN102358738A (en) 2003-07-30 2012-02-22 里格尔药品股份有限公司 2,4-pyrimidinediamine compounds and uses of treating or preventing autoimmune diseases thereof
JP4607879B2 (en) 2003-08-15 2011-01-05 ノバルティス アーゲー 2,4-Pyrimidinediamine useful for the treatment of neoplastic diseases, inflammation and immune disorders
GB0321710D0 (en) 2003-09-16 2003-10-15 Novartis Ag Organic compounds
AU2004272288B2 (en) 2003-09-18 2008-11-13 Novartis Ag 2,4-di (phenylamino) pyrimidines useful in the treatment of proliferative disorders
NZ588139A (en) 2004-04-08 2012-02-24 Targegen Inc 7-Aryl-3-arylamino-benzo[1,2,4]triazine derivatives
MX2007002208A (en) 2004-08-25 2007-05-08 Targegen Inc Heterocyclic compounds and methods of use.
US7210697B2 (en) 2004-12-16 2007-05-01 Tricam International, Inc. Convertible handle
AU2005322855B2 (en) 2004-12-30 2012-09-20 Exelixis, Inc. Pyrimidine derivatives as kinase modulators and method of use
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
CA2600531A1 (en) 2005-03-16 2006-09-28 Targegen, Inc. Pyrimidine compounds and methods of use
WO2006128172A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating b cell regulated autoimmune disorders
WO2006128129A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating cancer
AU2006254825A1 (en) 2005-06-08 2006-12-14 Targegen, Inc. Methods and compositions for the treatment of ocular disorders
ES2651349T3 (en) 2005-06-08 2018-01-25 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibiting the JAK route
WO2006137658A1 (en) 2005-06-20 2006-12-28 Dongbu Hitek Co., Ltd. New substituted 1,3-thiazole derivatives or pharmaceutically acceptable salts thereof having immunosuppression and inflammation inhibitory acitivity, intermediate compounds or pharmaceutically acceptable salts thereof, a process for the preparation thereof, and pharmaceutical composition comprising the same
RS20070511A (en) 2005-06-29 2009-01-22 Panacea Biotec Ltd., Modified release pharmaceutical compositions and processes thereof
WO2007008541A2 (en) 2005-07-08 2007-01-18 Kalypsys, Inc. Cellular cholesterol absorption modifiers
US7754717B2 (en) 2005-08-15 2010-07-13 Amgen Inc. Bis-aryl amide compounds and methods of use
CA2621261C (en) 2005-09-22 2014-05-20 Incyte Corporation Azepine inhibitors of janus kinases
US20070072682A1 (en) 2005-09-29 2007-03-29 Crawford James T Iii Head to head electronic poker game assembly and method of operation
US8133900B2 (en) * 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
UA109412C2 (en) * 2005-11-01 2015-08-25 5-METHYL-N2- [4- (4-METHYLPIPERAZIN-1-yl) PHENYL] -PYRIMIDINE-2,4-DIAMINE AND ITS APPLICATION IN THE METHOD OF PREPARATION
US8604042B2 (en) 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
PT1951684T (en) * 2005-11-01 2016-10-13 Targegen Inc Bi-aryl meta-pyrimidine inhibitors of kinases
US20070161645A1 (en) 2005-11-02 2007-07-12 Targegen, Inc. Thiazole inhibitors targeting resistant kinase mutations
CA2628474A1 (en) 2005-11-03 2007-05-10 Sgx Pharmaceuticals, Inc. Pyrimidinyl-thiophene kinase modulators
EP1954256A2 (en) 2005-11-09 2008-08-13 Torrent Pharmaceuticals Ltd Pharmaceutical composition comprising a compound having a catechol moiety and an alkalising agent
WO2007058628A1 (en) 2005-11-16 2007-05-24 S*Bio Pte Ltd Heteroalkyl linked pyrimidine derivatives
PT3184526T (en) 2005-12-13 2018-12-19 Incyte Holdings Corp Pyrrolo[2,3-d]pyrimidine derivatives as janus kinase inhibitor
WO2007089768A2 (en) * 2006-01-30 2007-08-09 Exelixis, Inc. 4-aryl-2-amino-pyrimidines or 4-aryl-2-aminoalkyl-pyrimidines as jak-2 modulators and pharmaceutical compositions containing them
JP2009528295A (en) 2006-02-24 2009-08-06 ライジェル ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibition of the JAK pathway
CN101460154B (en) * 2006-04-04 2015-07-01 Kg艾克维泽生公司 Oral dosage forms including an antiplatelet agent and an acid inhibitor
WO2007127366A2 (en) 2006-04-25 2007-11-08 Targegen, Inc. Kinase inhibitors and methods of use thereof
EP2037918A2 (en) * 2006-05-15 2009-03-25 Acadia Pharmaceuticals Inc. Pharmaceutical formulations of pimavanserin
US8030487B2 (en) 2006-07-07 2011-10-04 Targegen, Inc. 2-amino—5-substituted pyrimidine inhibitors
US20080021013A1 (en) 2006-07-21 2008-01-24 Cephalon, Inc. JAK inhibitors for treatment of myeloproliferative disorders
JP2009544592A (en) 2006-07-21 2009-12-17 ノバルティス アクチエンゲゼルシャフト 2,4-Di (arylamino) -pyrimidine-5-carboxamide compounds as JAK kinase inhibitors
WO2008057233A2 (en) * 2006-10-25 2008-05-15 The Regents Of The University Of California Models of erythropoiesis
US20080312259A1 (en) 2007-06-13 2008-12-18 Incyte Corporation SALTS OF THE JANUS KINASE INHIBITOR (R)-3-(4-(7H-PYRROLO[2,3-d]PYRIMIDIN-4-YL)-1H-PYRAZOL-1-YL)-3-CYCLOPENTYLPROPANENITRILE
DE102007034947B4 (en) * 2007-07-26 2009-06-18 Siemens Ag Device for unloading a stack of flat objects standing on their narrow sides
CN101802182A (en) * 2007-08-21 2010-08-11 诺达利蒂公司 Be used to diagnose, the method for prognosis and methods of treatment
WO2009046416A1 (en) 2007-10-05 2009-04-09 Targegen Inc. Anilinopyrimidines as jak kinase inhibitors
US20100310563A1 (en) * 2007-11-30 2010-12-09 Bumm Thomas G P Methods for treating induced cellular proliferative disorders
EP2296632A4 (en) * 2008-07-14 2014-11-12 Otonomy Inc Controlled-release apoptosis modulating compositions and methods for the treatment of otic disorders
RU2011108563A (en) 2008-08-05 2012-09-10 Таргеджен, Инк. (US) THALASSEMIA TREATMENT METHODS
US8987243B2 (en) 2008-12-11 2015-03-24 Cti Biopharma Corp. 11-(2-pyrrolidin-1-yl-ethoxy)-14,19-dioxa-5,7,26-triaza-tetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-maleate salt
ES2382773T3 (en) 2009-03-13 2012-06-13 Sanovel Ilac Sanayi Ve Ticaret A.S. Ezetimibe compositions
US20100278921A1 (en) * 2009-04-30 2010-11-04 Fischer Cristina M Solid oral formulation of abt-263
US20100297194A1 (en) 2009-04-30 2010-11-25 Nathaniel Catron Formulation for oral administration of apoptosis promoter
US20120190806A1 (en) 2009-07-31 2012-07-26 Basf Se Phosphine Borane Compounds Comprising Imidazol Groups And Method For Producing Phosphine Borane Compounds Comprising Imidazol Groups
US20120157500A1 (en) 2009-08-24 2012-06-21 Weikang Tao Jak inhibition blocks rna interference associated toxicities
WO2012060847A1 (en) * 2010-11-07 2012-05-10 Targegen, Inc. Compositions and methods for treating myelofibrosis
WO2013013195A1 (en) 2011-07-21 2013-01-24 Sanofi Compositions and methods for treating polycythemia vera and essential thrombocythemia
WO2013059548A1 (en) 2011-10-19 2013-04-25 Sanofi Compositions and methods for treating cancer using jak2 inhibitor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060210627A1 (en) * 2005-01-18 2006-09-21 Sabine Pfeffer Direct compression formulation and process

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
D.A. Doornbos, Optimisation in pharmaceutical science, 1981, Pharmaceutisch Weekblad Scientific Edition, Vol. 3 (Year: 1981) *
Z.T. Chowhan, Drug-Excipient Interactions Resulting from Powder Mixing IV: Role of Lubricants and Their Effect on In Vitro Dissolution, June 1986, Journal of pharmaceutical sciences, vol. 75, no.6 (Year: 1986) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190381041A1 (en) * 2010-11-07 2019-12-19 Impact Biomedicines, Inc. Compositions and methods for treating myelofibrosis
US11400092B2 (en) 2018-09-25 2022-08-02 Impact Biomedicines, Inc. Methods of treating myeloproliferative disorders

Also Published As

Publication number Publication date
IL226101A0 (en) 2013-06-27
AU2011323108B2 (en) 2015-12-17
CN108125923A (en) 2018-06-08
PT2635282T (en) 2023-01-05
SI2635282T1 (en) 2023-01-31
LT2635282T (en) 2022-12-12
AU2011323108A1 (en) 2013-05-09
HUE060254T2 (en) 2023-02-28
RU2616262C2 (en) 2017-04-13
CA2816957A1 (en) 2012-05-10
KR20130137647A (en) 2013-12-17
BR112013011184A2 (en) 2018-01-30
ZA201303423B (en) 2015-06-24
US20190381041A1 (en) 2019-12-19
WO2012061833A1 (en) 2012-05-10
MA34723B1 (en) 2013-12-03
RS63996B1 (en) 2023-03-31
NI201300038A (en) 2013-09-09
JP6133211B2 (en) 2017-05-24
KR101940979B1 (en) 2019-01-23
EP2635282A4 (en) 2014-04-30
MX360246B (en) 2018-10-26
US10391094B2 (en) 2019-08-27
TWI531389B (en) 2016-05-01
RU2013126121A (en) 2015-01-27
UA114076C2 (en) 2017-04-25
HRP20221269T1 (en) 2022-12-23
CA2816710A1 (en) 2012-05-10
EP2635282A1 (en) 2013-09-11
WO2012060847A1 (en) 2012-05-10
CL2013001252A1 (en) 2014-03-28
SG190134A1 (en) 2013-06-28
EP4159216A1 (en) 2023-04-05
JP2013541595A (en) 2013-11-14
DK2635282T3 (en) 2022-10-17
KR20200083676A (en) 2020-07-08
ECSP13012658A (en) 2013-08-30
NZ611363A (en) 2015-09-25
US20130243853A1 (en) 2013-09-19
CN103282036B (en) 2018-01-26
PE20140389A1 (en) 2014-03-26
US20130252988A1 (en) 2013-09-26
MY161164A (en) 2017-04-14
TW201306882A (en) 2013-02-16
KR102131241B1 (en) 2020-07-07
ES2930650T3 (en) 2022-12-20
EP2635282B1 (en) 2022-09-28
AU2010363329A1 (en) 2013-05-09
KR20180122029A (en) 2018-11-09
DOP2013000097A (en) 2013-11-30
PL2635282T3 (en) 2023-02-06
IL226101A (en) 2017-12-31
KR20240029118A (en) 2024-03-05
MX2013005020A (en) 2013-07-29
CN103282036A (en) 2013-09-04
CO6801724A2 (en) 2013-11-29

Similar Documents

Publication Publication Date Title
US20210244735A1 (en) Compositions and methods for treating myelofibrosis
US20110207660A1 (en) Pharmaceutical formulations of an hcv protease inhibitor in a solid molecular dispersion
JP6404886B2 (en) Method for stabilizing pharmaceutical compositions containing irbesartan and amlodipine or a salt thereof
KR20160013068A (en) Cenicriviroc compositions and methods of making and using the same
TW201204711A (en) Pharmaceutical composition comprising amide derivative or pharmaceutically acceptable salt thereof
US20190358240A1 (en) Hiv treatment formulation of atazanavir and cobicistat
US10603282B2 (en) Pharmaceutical compositions containing doravirine, tenofovir disoproxil fumarate and lamivudine
KR20210041589A (en) Bioavailable oral dosage form
US20120141586A1 (en) Thrombin receptor antagonist and clopidogrel fixed dose tablet
CN113242859B (en) Therapeutic use of pyrrolopyrimidine compounds and solid pharmaceutical compositions thereof
US11648242B2 (en) Pharmaceutical composition comprising pimavanserin, process of preparation and use thereof
OA17692A (en) Compositions and methods for treating myelofibrosis.
CN114259497A (en) Pharmaceutical composition of aminopyrimidine compounds and preparation method thereof
NZ623628B2 (en) Solid oral pharmaceutical formulations comprising amorphous (S)-methyl (1- ((4-(3-(5-chloro-2-fluoro-3-(methylsulfonamido)phenyl)-1-isopropy1-1H-pyrazo1-4-yl)pyrimidin-2-yl)amino)propan-2-yl)carbamate (Compound A)

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: SPECIAL NEW

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: IMPACT BIOMEDICINES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TARGEGEN, INC.;REEL/FRAME:059237/0351

Effective date: 20170816

Owner name: TARGEGEN, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JAYAN, ARVIND;CACACE, JANICE;SIGNING DATES FROM 20111106 TO 20111107;REEL/FRAME:059237/0284

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: SPECIAL NEW

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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