US20180162861A1 - Tyrosine kinase inhibitors - Google Patents

Tyrosine kinase inhibitors Download PDF

Info

Publication number
US20180162861A1
US20180162861A1 US15/578,623 US201615578623A US2018162861A1 US 20180162861 A1 US20180162861 A1 US 20180162861A1 US 201615578623 A US201615578623 A US 201615578623A US 2018162861 A1 US2018162861 A1 US 2018162861A1
Authority
US
United States
Prior art keywords
amino
pyridin
oxo
imidazo
phenoxyphenyl
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
US15/578,623
Other languages
English (en)
Inventor
David M. Goldstein
Timothy D. Owens
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.)
Principia Biopharma Inc
Original Assignee
Principia Biopharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Principia Biopharma Inc filed Critical Principia Biopharma Inc
Priority to US15/578,623 priority Critical patent/US20180162861A1/en
Assigned to PRINCIPIA BIOPHARMA INC. reassignment PRINCIPIA BIOPHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GOLDSTEIN, DAVID, OWENS, TIMOTHY
Publication of US20180162861A1 publication Critical patent/US20180162861A1/en
Assigned to PRINCIPIA BIOPHARMA INC. reassignment PRINCIPIA BIOPHARMA INC. CHANGE OF ADDRESS OF ASSIGNEE Assignors: PRINCIPIA BIOPHARMA INC.
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present disclosure provides compounds that are tyrosine kinase inhibitors, in particular Bruton tyrosine kinase (“BTK”) inhibitors, and are therefore useful for the treatment of diseases such as cancer, autoimmune, inflammatory, and thromboembolic diseases. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.
  • BTK Bruton tyrosine kinase
  • BTK a member of the Tec family non-receptor tyrosine kinases, is essential for B cell signaling downstream from the B-cell receptor. It is expressed in B cells and other hematopoietic cells such as monocytes, macrophages and mast cells. It functions in various aspects of B cell function that maintain the B cell repertoire (see Gauld S. B. et al., B cell antigen receptor signaling: roles in cell development and disease. Science, 296:1641-2. 2002.) B cells pay a role in rheumatoid arthritis (see Perosa F., et al., CD20-depleting therapy in autoimmune diseases: from basic research to the clinic. J Intern Med. 267:260-77.
  • BTK inhibitors for treating allergic diseases (see Honigberg, L., et. al., The selective BTK inhibitor PCI-32765 blocks B cell and mast cell activation and prevents mouse collagen indiced arthritis. Clin. Immunol. 127 S1:S111. 2008). It was noted that the irreversible inhibitor suppresses passive cutaneous anaphylaxis (PCA) induced by IgE antigen complex in mice. These findings are in agreement with those noted with BTK-mutant mast cells and knockout mice and suggest that BTK inhibitors may be useful for the treatment of asthma, an IgE-dependent allergic disease of the airway.
  • PCA passive cutaneous anaphylaxis
  • compounds that inhibit BTK would be useful in treatment for diseases such as autoimmune diseases, inflammatory diseases, and cancer.
  • this disclosure is directed to a compound of Formula (I):
  • R 1 and R 2 are independently hydrogen, alkyl, alkoxy, halolakyl, or halo;
  • X is —O—, —CONR—, —NRCO—, or —NR—CO—NR′ where R and R′ are independently hydrogen or alkyl;
  • Ar is heteroaryl or phenyl where heteroaryl and phenyl are optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy;
  • A is —N— or —CR 3 — wherein R 3 is hydrogen, alkyl, cyclopropyl, halo, haloalkyl, haloalkoxy, alkoxy, or cyano;
  • Y is bond or alkylene
  • ring Z is heterocycloamino optionally substituted with one or two substituents independently selected from alkyl, hydroxy, alkoxy, and fluoro;
  • R 5 is a group of formula (i), (ii), (iii) or (iv):
  • R a is hydrogen, fluoro, or cyano; provided that when R a is cyano then R b is hydrogen and R c is not hydrogen;
  • R b is hydrogen or alkyl
  • R c is hydrogen, hydroxyalkyl, alkoxyalkyl, alkyl (optionally substituted with one or two substituents independently selected from hydroxy, hydoxyalkyl, heteroaryl (optionally substituted with one or two substituents independently selected from alkyl and heterocyclyl wherein heterocyclyl is optionally substituted with one or two substituents independently selected from halo and alkyl), and —CONR 9 R 10 (where R 9 and R 10 are independently hydrogen or alkyl, or R 9 and R 10 together with the nitrogen atom to which they are attached form a heterocyclyl optionally substituted with one or two substituents selected from alkyl and heterocyclyl)), cycloalkyl (optionally substituted with one or two substituents independently selected from halo, alkyl, alkoxyalkyl and aryl; or wherein two adjacent substituents of the cycloalkyl together with the carbon atoms to which they are attached form a heterocyclyl group), heterocycly
  • X 1 , X 2 and X 3 are nitrogen and the rest are carbon and the ring is optionally substituted with one or two substituents independently selected from alkyl, haloalkyl, and halo); and/or
  • R a is cyano and R c is heterocycloaminolalkyl wherein the heterocycloamino in heterocycloaminoalkyl is optionally substituted with one or two substituents independently selected from alkyl, alkoxy, hydroxy, halo, amino, and oxo, and the nitrogen atom of heterocycloamino is substituted with heterocyclyl wherein the heterocyclyl is substituted with one or two substituents independently selected from hydrogen, alkyl, halo, hydroxy, and alkoxy.
  • R 5 in the compounds of Formula (I) and/or a pharmaceutically acceptable salt thereof (and any embodiments thereof disclosed herein) is a group of formula (i), (ii), or (iii) wherein R a is cyano
  • the compounds of the disclosure are reversible covalent inhibitors of BTK, i.e., they can form a reversible covalent bond with a thiol group of a cysteine residue, in particular with Cys481 of BTK.
  • R 5 in the compounds of Formula (I) and/or a pharmaceutically acceptable salt thereof (and any embodiments thereof disclosed herein) is a group of formula (i), (ii), or (iii) where R a is hydrogen or fluoro, or R 5 is a group of formula (iv)
  • the compounds of the disclosure are irreversible covalent inhibitors of BTK, i.e., they can form an irreversible covalent bond with a thiol group of a cysteine residue, in particular with Cys481 of BTK.
  • this disclosure is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) (or any of the embodiments thereof described herein), and/or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable excipient.
  • the formulation is a solid oral formulation comprising:
  • the formulation is a solid oral formulation comprising means for release of a therapeutically effective amount of a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof (or any embodiment thereof disclosed herein) from said oral formulation in the intestine.
  • the compound of Formula (I) and/or a pharmaceutically acceptable salt thereof is released in the small intestine.
  • the enteric coating is a polymer.
  • the enteric coating is an anionic polymer selected from polymethacrylates (e.g., methacrylic acid ethacrylate poly, methacrylic acid methyl methacrylate poly); cellulose-based polymers (e.g., cellulose acetate phthalate CAP, cellulose acetate trimellitate CAT, cellulose acetate succinate CAS, hydroxypropylmethylcellulose phthalate HPMCP, hydroxypropylmethylcellulose acetate succinate HPMCAS), and polyvinyl derivatives such as polyvinyl acetate phthalate PVAP.
  • polymethacrylates e.g., methacrylic acid ethacrylate poly, methacrylic acid methyl methacrylate poly
  • cellulose-based polymers e.g., cellulose acetate phthalate CAP, cellulose acetate trimellitate CAT, cellulose acetate succinate CAS, hydroxypropylmethylcellulose phthalate HPMCP, hydroxypropylmethylcellulose acetate succinate HP
  • the enteric coating erodes in the gastrointestinal track having a pH from about 4.5 to about 7 or from about 5 or 5.5 to about 7 to release the compound of Formula (I) and/or a pharmaceutically acceptable salt thereof (or embodiments thereof disclosed herein).
  • the non-enteric time-delayed release dosage forms can be administered in fasted state and the time-delayed release coating can be designed to erode, burst, or become hightly permeable in about 0.3 to about 3 hours or in about 0.5 to about 2 hours after administration to release the compound of Formula (I) (or embodiments thereof disclosed herein) and/or a pharmaceutically acceptable salt thereof.
  • this disclosure is directed to a method of treating a disease treatable by inhibition of BTK in a mammal in need thereof which method comprises administering to the mammal in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the disease is cancer, autoimmune, inflammatory, or thromboembolic diseases.
  • the disease is acute necrotizing hemorrhagic leukoencephalitis, acute disseminated encephalomyelitis, Addison's disease, agammaglobulinemia, alopecia areata, alopecia universalis, amyloidosis, ankylosing spondylitis, anti-GBM/Anti-TBM nephritis, antiphospholipid syndrome (APS), antiphospholipid antibody syndrome, aplastic anemia, arthritis, autoimmune angioedema, autoimmune dysautonomia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease (AIED), autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura (ATP), autoimmune thyroid disease, autoimmune urticaria, autoimmune hemolytic anemia, axonal & neuronal neuropathies, Balo disease, Beh
  • the mammal is suffering from an autoimmune disease, e.g., inflammatory bowel disease, arthritis, lupus including Lupus Nephritis, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, granulomatosis with polyangiitis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, Sjogren's syndrome, dry eye (including Sjogren's dry eye and non-Sjogren's dry eye), multiple sclerosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylitisis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, coeliac disease, Goodpasture's syndrome, idiopathic thrombocytopenic
  • an autoimmune disease
  • the autoimmune disease is lupus, pemphigus vulgaris, myasthenia gravis, Sjogren's syndrome, dry eye, multiple sclerosis, Wegener's granulomatosis, autoimmune hemolytic anemia, idiopathic thrombocytopenic purpura, Granulomatosis with Polyangiitis, or rheumatoid arthritis.
  • the mammal is suffering from a heteroimmune condition or disease, e.g., graft versus host disease, transplantation, transfusion, anaphylaxis, allergy, type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, or atopic dermatitis.
  • a heteroimmune condition or disease e.g., graft versus host disease, transplantation, transfusion, anaphylaxis, allergy, type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, or atopic dermatitis.
  • the disease is atopic dermatitis.
  • the mammal is suffering from an inflammatory disease, e.g., asthma, appendicitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis,
  • an inflammatory disease e
  • the mammal is suffering from inflammatory skin disease which includes, by way of example, dermatitis, contact dermatitis, eczema, urticaria, rosacea, and scarring psoriatic lesions in the skin, joints, or other tissues or organs.
  • inflammatory skin disease includes, by way of example, dermatitis, contact dermatitis, eczema, urticaria, rosacea, and scarring psoriatic lesions in the skin, joints, or other tissues or organs.
  • the inflammatory disease is asthma or dermatitis.
  • the mammal is suffering from inflammatory and/or autoimmune disease, including acute inflammatory and/or autoimmune disease, where corticosteroid therapy is used as the first or second line therapy or first or second line maintenance therapy.
  • corticosteroid therapy is used as the first or second line therapy or first or second line maintenance therapy.
  • the compound of Formula (I) (or any embodiments thereof disclosed herein) is used for the treatment of:
  • Endocrine Disorders Primary or secondary adrenocortical insufficiency (hydrocortisone or cortisone is the first choice: synthetic analogs may be used in conjunction with mineralocorticoids where applicable; in infancy mineralocorticoid supplementation is of particular importance); congenital adrenal hyperplasia; nonsuppurative thyroiditis; hypercalcemia associated with cancer.
  • Rheumatic Disorders As adjunctive therapy for short-term administration (to tide the patient over an acute episode or exacerbation) in: psoriatic arthritis, rheumatoid arthritis, including juvenile rheumatoid arthritis (selected cases may require low-dose maintenance therapy), ankylosing spondylitis, acute and subacute bursitis, acute nonspecific tenosynovitis, gout, acute gouty arthritis, post-traumatic osteoarthritis, synovitis of osteoarthritis, epicondylitis.
  • Collagen Diseases During an exacerbation or as maintenance therapy in selected cases of: systemic lupus erythematosus, systemic dermatomyositis (polymyositis), acute rheumatic carditis.
  • Dermatologic Diseases Pemphigus; bullous dermatitis herpetiformis; severe erythema multiforme (Stevens-Johnson syndrome); exfoliative dermatitis; mycosis fungoides; severe psoriasis; severe seborrheic dermatitis.
  • Allergic States Control of severe or incapacitating allergic conditions intractable to adequate trials of conventional treatment: seasonal or perennial allergic rhinitis; bronchial asthma; contact dermatitis; atopic dermatitis; serum sickness; drug hypersensitivity reactions.
  • Ophthalmic Diseases Severe acute and chronic allergic and inflammatory processes involving the eye and its adnexa such as: allergic corneal marginal ulcers, herpes zoster ophthalmicus, anterior segment inflammation, diffuse posterior uveitis and choroiditis, sympathetic ophthalmia, allergic conjunctivitis, keratitis, chorioretinitis, optic neuritis, ulceris and iridocyclitis.
  • Respiratory Diseases Symptomatic sarcoidosis; Loeffler's syndrome not manageable by other means; berylliosis; aspiration pneumonitis, fulminating or disseminated pulmonary tuberculosis when used concurrently with appropriate antituberculous chemotherapy
  • Hematologic Disorders Idiopathic thrombocytopenic purpura in adults; secondary thrombocytopenia in adults; acquired (autoimmune) hemolytic anemia; erythroblastopenia (RBC anemia); congenital (erythroid) hypoplastic anemia.
  • Neoplastic Diseases For palliative management of: leukemias and lymphomas in adults, acute leukemia of childhood.
  • Edematous States To induce a diuresis or remission of proteinuria in the nephrotic syndrome, without uremia, of the idiopathic type or that due to lupus erythematosus.
  • Gastrointestinal Diseases To tide the patient over a critical period of the disease in: ulcerative colitis, regional enteritis.
  • the compound of Formula (I) and/or a pharmaceutically acceptable salt thereof can be used for the treatment of above listed diseases optionally in combination with a corticosteroid, noncorticosteroidal, immunosupressive, and/or antiinflammatory agents.
  • the immunosuppressive agent is selected from interferon alpha, interferon gamma, cyclophosphamide, tacrolimus, mycophenolate mofetil, methotrexate, dapsone, sulfasalazine, azathioprine, an anti-CD20 agent (such as rituximab, ofatumumab, obinutuzumab, or veltuzumab, or a biosimilar version thereof), anti-TNFalpha agent (such as entanercept, infliximab, golilumab, adalimumab, or certolizumab pegol or a biosimilar version thereof), anti-IL6 agent toward ligand or its receptors (
  • the mammal is suffering from a cancer.
  • the cancer is a B-cell proliferative disorder, e.g., diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma (CLL), chronic lymphocytic leukemia, chromic myleogenous leukemia, B-cell acute lymphoblastic leukemia (B-ALL), Philadelphia chromosome positive B-ALL, B-cell prolymphocytic leukemia, small lymphocytic lymphoma (SLL), multiple myeloma, B-cell non-Hodgkin lymphoma, lymphoplamascytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, mantle cell lymphoma
  • B-cell proliferative disorder e
  • the mammal is suffering from a thromboembolic disorder, e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, or deep venous thrombosis.
  • a thromboembolic disorder e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, or deep venous thrombosis.
  • the disclosure is directed a compound of Formula (I) (and any embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof for use as a medicament.
  • the use of the compound of Formula (I) and/or a pharmaceutically acceptable salt thereof is for treating a disease mediated by BTK, for example, the disease is an inflammatory disease, autoimmune disease, cancer, or thromboembolic diseases described in the third aspect and embodiments therein.
  • a fifth aspect is the use of a compound of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a disease in a mammal in which BTK contributes to the pathology and/or symptoms of the disease.
  • the disease is cancer, autoimmune, inflammatory, or thromboembolic disease described in the third aspect and embodiments therein.
  • any of the aforementioned aspects involving the treatment of cancer are further embodiments comprising administering the compound of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof, in combination with an anticancer agent.
  • the agents can be administered simultaneously (such as a fixed combination drug product) or sequentially.
  • this disclosure is directed to an intermediate of Formula (II):
  • R 1 , R 2 , R 3 , X, Ar, Y, and ring Z are as defined in the first aspect above; or a salt thereof.
  • R 1 , R 2 , R 3 , X, Ar, Y, and ring Z are as defined in the first aspect above; with an aldehyde of formula R c CHO where R c is as defined in the first aspect above; or
  • R 1 , R 2 , R 3 , X, Ar, Y, and Z are as defined in the first aspect above;
  • R 1 , R 2 , R 3 , X, Ar, Y, and ring Z are as defined in the first aspect above; with a compound of formula R c R b C ⁇ CHCOL where L is a leaving group under acylation reaction conditions where R b and R c are as defined in the first aspect above;
  • Alkyl means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
  • Alkylene means a linear saturated divalent hydrocarbon radical of one to six carbon atoms or a branched saturated divalent hydrocarbon radical of three to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1-methylpropylene, 2-methylpropylene, butylene, pentylene, and the like.
  • Alkylsulfonyl means a —SO 2 R radical where R is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like.
  • Amino means a —NH 2 .
  • Alkoxy means a —OR radical where R is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
  • Alkoxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with an alkoxy group, (in one embodiment one or two alkoxy groups), as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like.
  • Alkoxycarbonyl means a —C(O)OR radical where R is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
  • “Acyl” means a —COR radical where R is alkyl, haloalkyl, or cycloalkyl, e.g., acetyl, propionyl, cyclopropylcarbonyl, and the like. When R is alkyl, the radical is also referred to herein as alkylcarbonyl.
  • Cycloalkyl means a cyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms wherein one or two carbon atoms may be replaced by an oxo group, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, and the like.
  • Carboxy means —COOH.
  • Halo means fluoro, chloro, bromo, or iodo; in one embodiment fluoro or chloro.
  • Haloalkyl means alkyl radical as defined above, which is substituted with one or one to five halogen atoms (in one embodiment fluorine or chlorine,) including those substituted with different halogens, e.g., —CH 2 Cl, —CF 3 , —CHF 2 , —CH 2 CF 3 , —CF 2 CF 3 , —CF(CH 3 ) 2 , and the like.
  • halogen atoms in one embodiment fluorine or chlorine,
  • fluoroalkyl e.g., —CH 2 Cl, —CF 3 , —CHF 2 , —CH 2 CF 3 , —CF 2 CF 3 , —CF(CH 3 ) 2 , and the like.
  • fluoroalkyl When the alkyl is substituted with only fluoro, it can referred to in this disclosure as fluoroalkyl.
  • Haloalkoxy means a —OR radical where R is haloalkyl as defined above e.g., —OCF 3 , —OCHF 2 , and the like.
  • R is haloalkyl where the alkyl is substituted with only fluoro, it can referred to in this disclosure as fluoroalkoxy.
  • Hydroalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 1-(hydroxymethyl)-2-methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1-(hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl.
  • Further examples include, but are not limited to, 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1-(hydroxymethyl)-2-hydroxyethyl.
  • Heterocyclyl means a saturated or unsaturated monovalent monocyclic or bi-cyclic group (fused bi-cyclic or bridged bi-cyclic) of 4 to 10 ring atoms in which one or two ring atoms are heteroatom selected from N, O, and S(O) n , where n is an integer from 0 to 2, the remaining ring atoms being C. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a —CO— group.
  • heterocyclyl includes, but is not limited to, oxetanyl, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydropyranyl, thiomorpholino, hexahydropyrrolo[1,2-a]pyrazin-6(2H)-one-yl, tetrahydro-1H-oxazolo[3,4-a]pyrazin-3(5H)-one-yl, 5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine-yl, 3-oxa-8-azabicyclo[3.2.1]octane-yl, and the like.
  • the heterocyclyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic
  • Heterocyclylalkyl means a -(alkylene)-R radical where R is heterocyclyl ring as defined above e.g., tetraydrofuranylmethyl, piperazinylmethyl, morpholinylethyl, and the like.
  • Heterocycloamino means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom selected from N, O, or S(O) n , where n is an integer from 0 to 2, the remaining ring atoms being C provided that at least one of the ring atoms is N. Additionally, one or two ring carbon atoms in the heterocycloamino ring can optionally be replaced by a —CO— group. When the heterocycloamino ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.
  • Heterocycloaminoalkyl means a -(alkylene)-R radical where R is heterocycloamino as described above.
  • Heteroaryl means a monovalent monocyclic or bicyclic aromatic radical of 5 to 10 ring atoms where one or more, (in one embodiment one, two, or three), ring atoms are heteroatom selected from N, O, and S, the remaining ring atoms being carbon.
  • Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, and the like.
  • mammal as used herein means domesticated animals (such as dogs, cats, and horses), and humans. In one embodiment, mammal is a human.
  • the present disclosure also includes the prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof.
  • the term prodrug is intended to represent covalently bonded carriers, which are capable of releasing the active ingredient of Formula (I) (or any of the embodiments thereof described herein) when the prodrug is administered to a mammalian subject. Release of the active ingredient occurs in vivo.
  • Prodrugs can be prepared by techniques known to one skilled in the art. These techniques generally modify appropriate functional groups in a given compound. These modified functional groups however regenerate original functional groups in vivo or by routine manipulation.
  • Prodrugs of compounds of Formula (I) include compounds wherein a hydroxy, amino, carboxylic, or a similar group is modified.
  • Examples of prodrugs include, but are not limited to esters (e.g., acetate, formate, and benzoate derivatives), carbamates (e.g., N,N-dimethylaminocarbonyl) of hydroxy or amino functional groups in compounds of Formula (I)), amides (e.g., trifluoroacetylamino, acetylamino, and the like), and the like.
  • Prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof are also within the scope of this disclosure.
  • the present disclosure also includes polymorphic forms (amorphous as well as crystalline) and deuterated forms of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • Such salts include:
  • acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulf
  • a metal ion e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion
  • organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • the compounds of the present disclosure may have asymmetric centers.
  • Compounds of the present disclosure containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of materials. All chiral, diastereomeric, racemic forms, as individual forms and mixtures thereof, are within the scope of this disclosure, unless the specific stereochemistry or isomeric form is specifically indicated.
  • Certain compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof can exist as tautomers and/or geometric isomers. All possible tautomers and cis and trans isomers, as individual forms and mixtures thereof, are within the scope of this disclosure. Additionally, as used herein the term alkyl includes all the possible isomeric forms of said alkyl group albeit only a few examples are set forth. Furthermore, when the cyclic groups such as heteroaryl, heterocyclyl are substituted, they include all the positional isomers albeit only a few examples are set forth. Furthermore, all hydrate forms of a compound of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt thereof are within the scope of this disclosure.
  • heterocyclyl group optionally substituted with an alkyl group means that the alkyl may but need not be present, and the description includes situations where the heterocyclyl group is substituted with an alkyl group and situations where the heterocyclyl group is not substituted with alkyl.
  • a “pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier or an excipient that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable carrier/excipient” as used in the specification and claims includes both one and more than one such excipient.
  • both substituents can be selected from alkyl, alkoxy, hydroxy, halo, and oxo or one of the two substituent is selected from alkyl, alkoxy, hydroxy, halo, and oxo and the other substituent is selected from alkyl, cycloalkyl, hydroxyalkyl, alkoxyalkyl, acyl, haloalkyl, alkylsulfonyl, alkoxycarbonyl, and heterocyclyl.
  • heterocyclyl ring when heterocyclyl ring is substituted with three substituents, then two substituents are selected from alkyl, alkoxy, hydroxy, halo, and oxo and the third substituent is selected from alkyl, cycloalkyl, hydroxyalkyl, alkoxyalkyl, acyl, haloalkyl, alkylsulfonyl, alkoxycarbonyl, and heterocyclyl.
  • Treating” or “treatment” of a disease includes:
  • a “therapeutically effective amount” means the amount of a compound of Formula (I) (or any of the embodiments thereof described herein), that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • a compound of Formula (I) as defined in the first embodiment of the first aspect above including an E or Z isomer thereof, and/or a pharmaceutically acceptable salt thereof.
  • R 1 and R 2 are independently hydrogen, alkyl, alkoxy, halolakyl, or halo;
  • X is —O—, —CONR—, —NRCO—, or —NR—CO—NR′ where R and R′ are independently hydrogen or alkyl;
  • Ar is heteroaryl or phenyl where heteroaryl and phenyl are optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy;
  • A is —N— or —CR 3 — wherein R 3 is hydrogen, alkyl, cyclopropyl, halo, haloalkyl, haloalkoxy, alkoxy, or cyano;
  • Y is bond or alkylene
  • ring Z is heterocycloamino optionally substituted with one or two substituents independently selected from alkyl, hydroxy, alkoxy, and fluoro;
  • R 5 is a group of formula (i), (ii), (iii) or (iv):
  • R a is hydrogen, fluoro, or cyano; provided that when R a is cyano then R b is hydrogen and R c is not hydrogen;
  • R b is hydrogen or alkyl
  • R c is hydrogen, alkyl optionally substituted with one or two substituents independently selected from OH, heteroaryl (optionally substituted with one or two substituents independently selected from alkyl and heterocyclyl wherein heterocyclyl is optionally substituted with one or two substituents independently selected from halo and alkyl), and —CONR 9 R 10 (where R 9 and R 10 are independently hydrogen or alkyl, or R 9 and R 10 together with the nitrogen atom to which they are attached form a heterocyclyl optionally substituted with one or two substituents selected from alkyl and heterocyclyl),
  • cycloalkyl optionally substituted with one or two substituents independently selected from halo, alkyl and aryl,
  • heterocyclylalkyl heterocyclyl (wherein heterocyclyl and heterocyclyl in heterocyclylalkyl are optionally substituted with one, two, or three substituents where two of the optional substituents are independently selected from alkyl, alkoxy, hydroxy, halo, amino, and oxo, and one of the optional substituent is alkyl, hydroxyalkyl, alkoxy, alkoxyalkyl, acyl, haloalkyl, alkylsulfonyl, alkoxycarbonyl, or heterocyclyl wherein the heterocyclyl is substituted with one or two substitutents independently selected from hydrogen, alkyl, halo, hydroxy, and alkoxy), or
  • R 6 and R 7 are independently hydrogen, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, cycloalkyl, or heterocyclyl wherein the heterocyclyl is optionally substituted with one or two substituents independently selected from alkyl, halo, hydroxy, hydroxyalkyl, alkoxyalkyl, acyl, and alkoxycarbonyl; or R 6 and R 7 together with the nitrogen atom to which they are attached form
  • X 1 , X 2 and X 3 are nitrogen and the rest are carbon and the ring is optionally substituted with one or two substituents independently selected from alkyl, haloalkyl, and halo); and/or a pharmaceutically acceptable salt thereof provided that:
  • R a is cyano and R c is heterocycloaminolalkyl wherein the heterocycloamino in heterocycloaminoalkyl is optionally substituted with one or two substituents independently selected from alkyl, alkoxy, hydroxy, halo, amino, and oxo, and the nitrogen atom of heterocycloamino is substituted with heterocyclyl wherein the heterocyclyl is substituted with one or two substituents independently selected from hydrogen, alkyl, halo, hydroxy, and alkoxy.
  • R 3 is hydrogen, methyl, ethyl, isopropyl, fluoro, or chloro. In a second embodiment of embodiment 3, R 3 is hydrogen.
  • Ar is heteroaryl or phenyl where heteroaryl and phenyl are optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy.
  • Ar is pyridinyl, pyrimidinyl, thienyl, or pyrazinyl, optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy.
  • Ar is phenyl where phenyl is optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy, preferably substituted with one or two fluoro.
  • Ar is heteroaryl or phenyl where heteroaryl and phenyl are optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy.
  • Ar is pyridinyl, pyrimidinyl, thienyl, or pyrazinyl, optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy.
  • Ar is phenyl where phenyl is optionally substituted with one, two, or three substituents independently selected from alkyl, halo, haloalkyl, alkoxy, and hydroxy, preferably one or two fluoro.
  • R 1 and R 2 are independently hydrogen or halo, preferably hydrogen or fluoro, more preferably R 1 and R 2 are hydrogen or R 1 is hydrogen and R 2 is fluoro.
  • Y is alkylene and ring Z is pyrrolidinyl, and in one embodiment, pyrrolidin-2-yl or azetidin-3-yl.
  • Y is methylene.
  • the pyrrolidinyl ring ring attached at C2 and the stereochemistry at carbon of the pyrrolidinyl ring attached to Y is (R) or (S).
  • R 5 is a group of formula (i) or (iv).
  • R 5 is a group of formula (i).
  • R 5 is a group of formula (ii) or (iii).
  • R 5 is a group of formula (iv).
  • R b and R c are hydrogen.
  • R b is hydrogen and R c is alkyl or or -(alkylene)-NR 6 R 7 (where R 6 and R 7 are independently hydrogen, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, cycloalkyl, or heterocyclyl wherein the heterocyclyl ring is optionally substituted with one or two substituents independently selected from alkyl, halo, hydroxy, hydroxyalkyl, alkoxyalkyl, acyl, and alkoxycarbonyl), preferably R 6 and R 7 are independently hydrogen or alkyl.
  • R 5 is a group of formula (i).
  • R 5 is a group of formula (ii) or (iii).
  • R c is cycloalkyl, which is optionally substituted with one or two substituents independently selected from halo, alkyl, alkoxyalkyl and aryl; or wherein two adjacent substituents of the cycloalkyl together with the carbon atoms to which they are attached form a heterocyclyl group.
  • R c is cyclopropyl, 1-methylcyclobutyl, 1-phenylcyclopropyl, 1-methylcyclopropyl, 2,2-difluorocyclopropyl,
  • R c is unsubstituted alkyl.
  • R c is isopropyl or tert-butyl.
  • R c is -(alkylene)-NR 6 R 7 (where R 6 and R 7 are independently hydrogen, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, cycloalkyl, or heterocyclyl.
  • R c is-C(CH 3 ) 2 NH 2 , —C(CH 3 ) 2 NHCH 3 , —C(CH 3 ) 2 N(CH 3 ) 2 , —C(CH 3 ) 2 NHCH 2 CH 3 , —C(CH 3 ) 2 NHCH(CH 3 ) 2 , —C(CH 3 ) 2 NHcyclopropyl, —C(CH 3 ) 2 NH(CH 2 ) 2 OCH 3 , —C(CH 3 ) 2 OCH 2 CH 3 , —C(CH 3 ) 2 N(CH 2 CH 3 )(oxetan-3-yl), —C(CH 3 ) 2 N(CH 3 )(oxetan-3-yl), or —C(CH 3 ) 2 NH(oxetan-3-yl).
  • R c is heterocyclylalkyl wherein the heterocyclyl in heterocyclylalkyl is optionally substituted with one, two, or three substituents where two of the optional substituents are independently selected from alkyl, alkoxy, hydroxy, halo, amino, and oxo, and one of the optional substituent is alkyl, hydroxyalkyl, alkoxyalkyl, acyl, haloalkyl, alkylsulfonyl, alkoxycarbonyl, or heterocyclyl wherein the heterocyclyl is optionally substituted with one or two substitutents independently selected from alkyl, halo, hydroxy, and alkoxy.
  • R c is —C(CH 3 ) 2 morpholine-4-yl, —C(CH 3 ) 2 -4-(2,2,2-trifluoroethyl)piperazin-1-yl, —C(CH 3 ) 2 -4-(1-methyl)piperidin-1-yl,
  • R c is heterocyclylalkyl wherein the heterocyclyl in heterocyclylalkyl is substituted with another heterocyclyl wherein the another heterocyclyl is substituted with alkyl on a carbon of the another heterocyclyl.
  • R c is heterocyclyl optionally substituted with one, two, or three substituents where two of the optional substituents are independently selected from alkyl, alkoxy, hydroxy, halo, amino, and oxo, and one of the optional substituent is alkyl, hydroxyalkyl, alkoxyalkyl, acyl, or heterocyclyl.
  • R c is 3-methyloxetan-3-yl, 3-ethyloxetan-3-yl, 3-fluorooxetan-3-yl, 3-aminooxetan-3-yl, 4-methylpiperidin-4-yl, 3-methylazetidin-3-yl, 1-methylazetidin-3-yl, 4-methyl-4-tetrahydropyranyl or 1,3-dimethylazetidin-3-yl.
  • R is 3-methyloxetan-3-yl, 3-ethyloxetan-3-yl, 3-fluorooxetan-3-yl, 3-aminooxetan-3-yl, 4-methylpiperidin-4-yl, 3-methylazetidin-3-yl, 1-methylazetidin-3-yl, 4-methyl-4-tetrahydropyranyl or 1,3-dimethylazetidin-3-yl.
  • R is
  • R c is alkyl which is optionally substituted with one or two substituents independently selected from hydroxy, hydoxyalkyl, and heteroaryl which is substituted with one or two substituents independently selected from alkyl and heterocyclyl wherein heterocyclyl is optionally substituted with one or two substituents independently selected from halo and alkyl.
  • R c is alkyl which is substituted with one or two hydroxy substituents.
  • R c is
  • R c is alkyl which is substituted with a heteroaryl that is optionally substituted with one or two substituents independently selected from alkyl and heterocyclyl wherein heterocyclyl is optionally substituted with one or two substituents independently selected from halo and alkyl.
  • R c is
  • R c is alkyl that is substituted with —CONR 9 R 10 , where R 9 and R 10 are independently hydrogen or alkyl, or R 9 and R 0 together with the nitrogen atom to which they are attached form a heterocyclyl optionally substituted with one or two substituents selected from alkyl and heterocyclyl.
  • R 9 and R 10 are both hydrogen or alkyl.
  • R c is —C(CH 3 ) 2 —CONH 2 or —C(CH 3 ) 2 —CON(CH 3 ) 2 .
  • Re is alkyl that is substituted with —CONR 9 R 10 , wherein R 9 and R 10 together with the nitrogen atom to which they are attached form a heterocyclyl optionally substituted with one or two substituents selected from alkyl and heterocyclyl.
  • the heterocyclyl formed by R 9 and R 10 together with the nitrogen atom to which they are attached is 4-methylpiperazinyl, or 4-(oxetan-3-yl)piperazin-1-yl.
  • A is —N—; —X—Ar is attached to carbon at the 4-position of the phenyl ring, the carbon of the phenyl ring attached to N of the cyclic urea ring being position 1; X is O; Y is a bond; ring Z is pyrrolidinyl or piperidinyl and is attached to the cyclic urea nitrogen at the C-3 carbon, the nitrogen atom of pyrrolidinyl or piperidinyl ring being C-1; the stereochemistry at carbon of the pyrrolidinyl or piperidinyl attached to the cyclic nitrogen being (R)), wherein R 5 is a group of formula (i), R a is cyano, R b is hydrogen and R c is heterocyclylalkyl, wherein heterocyclyl in heterocyclylalkyl is optionally substituted with one, two, or three substituents where two of the optional substitu
  • R c is
  • the present invention is also directed to the following compounds:
  • the starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
  • the reactions described herein take place at atmospheric pressure over a temperature range from about ⁇ 78° C. to about 150° C., or from about 0° C. to about 125° C. or at about room (or ambient) temperature, e.g., about 20° C.
  • Reaction of a dihaloheteroaryl compound such as 4,6-dichloro-5-nitropyrimidine with an amine of formula NH(PG) 2 where PG is a suitable amino protecting group such as benzyl provides a compound of formula 1.
  • the reaction is carried out in a suitable organic solvent such as dioxane, dichloromethane, and the like.
  • the reaction is carried out in dichloromethane, dioxane, tetrahydrofuran, and the like with additional base such as triethylamine.
  • Compounds of formula 4 can be cyclized to form the benzimidazolones of formula 5 by heating 4 in an organic solvent such as dichloroethane and the like, with carbonyl diimidazole, phosgene or a phosgene equivalent (e.g., diphosgene or triphosgene), in the presence of a base such as triethyl amine, diisopropylethyl amine, and the like. Removal of the amino protecting group PG provides compound of formula 6. The reaction conditions utilized are based on the nature of the amino protecting group.
  • PG when PG is a benzyl group it can be removed via hydrogenation using a Pd/C catalyst and the like with an additive such as acetic acid to afford a compound of formula 6.
  • compound 8 can be prepared by reacting compound 5 first with boronic acid 7, followed by removal of the amino protecting group under conditions described above. Removal of the amino protecting group PG 1 in compound 8 provides compound of formula 9.
  • the reaction conditions utilized are based on the nature of the amino protecting group. For example, when PG 1 is Boc it can be removed under acid hydrolysis reaction conditions such as treatment with an acid such TFA, HCl, and the like.
  • Compound 9 can be then converted to a compound of Formula (I) by methods well known in the art.
  • compounds of Formula (I) can be prepared by coupling compound 9 with an acid of formula 10 or an acid derivative of compound 10 such as acid chloride, where R a , R b , and R c are as described in the Summary to give a compound of Formula (I).
  • the reaction is carried out under standard amide coupling conditions such as in the presence of HATU, DCC, carbon diimidazole (CDI) and the like.
  • Compounds of formula 10 or acid chloride derivatives thereof are commercially available (e.g. acryloyl chloride) or they can be prepared by methods well known in the art, such as the condensation product of cyanoacetic acid and an aldehyde such as isobutyraldeyde or pivaldehyde.
  • Compound of Formula (I) where R a is cyano also be prepared by first condensing compound 9 with 2-cyanoacetic acid under standard amide coupling conditions such as carbon diimidazole (CDI) and the like to give a compound of formula 11.
  • Condensation of a compound of formula 11 with an aldehyde of formula R c CHO where R c is as defined in the Summary under standard condensation reaction conditions such as using a base such as piperidine and the like, in the presence or absence of acetic acid and the like, in solvents such as ethanol and the like at temperatures ranging from room temperature to reflux then provides a compound of Formula (I).
  • R c CHO Compounds of formula R c CHO are commercially available or they can be prepared by methods well known in the art, e.g., acetaldehyde, cyclopropylaldehyde, isobutyraldehyde, 3-methyloxetane-3-carbaldehyde, 2-(dimethylamino)-2-methylpropanal, 2-methyl-2-(1-piperidyl)propanal, tert-butyl (2S)-2-formylpyrrolidine-1-carboxylate and 2-methyl-2-(morpholin-4-yl)propanal are commercially available.
  • Ethoxy-2-methylpropanal was prepared from isobutyraldehyde as described in PCT Int.
  • R c CHO where R c is -(alkylene)-NR 6 R 7 can be prepared by treatment of isobutryaldhyde with bromine to form bromoisobutryaldehyde followed displacement of the bromide by addition of HNR 6 R 7 .
  • compound 11 can also be condensed with a precursor group of R c CHO and then converted to a compound of Formula (I).
  • compound 11 can be condensed with tert-butyl 2-methyl-1-oxopropan-2-ylcarbamate followed by removal of the amino protecting group to give a compound of Formula (I) where R c is 2-aminopropan-2-yl.
  • the condensation reaction can be also be carried out by adding the desired aldehyde R c CHO with a base such as pyrrolidine or piperidine with or without chlorotrimethylsilane in dichloromethane or other suitable solvent (e.g. dioxane and ethanol).
  • compounds of formula 11 can be reacted with cyanomethanesulfonyl chloride, available commercially, to afford a cyanomethylsulfonamide which can be condensed with aldehydes of formula 12 with TMSCl and pyrrolidine to afford structures of formula (I).
  • compound 17 can be prepared by treating first with an acid such as TFA to remove both protecting groups and subsequently installing the PG 1 (e.g. Boc) group. Subsequent treatment with dimethylformamide dimethyl acetal affords a compound of formula 18. Reacting under Chan-Lam conditions as described above then affords compound 19. Subsequent deprotection by treatment of 19 with an acid such as HCl or TFA in solvents such as Dichloromethane, dioxane, MeOH, or EtOH affords a compound of formula 20. Preparation of compounds of formula (I) are then prepared in analogous fashion to the methods described in Schemes 1 and 2.
  • an acid such as TFA
  • solvents such as Dichloromethane, dioxane, MeOH, or EtOH affords a compound of formula 20.
  • scheme 4 shows the preparation of the compounds of formula 21, 22, 24, and 25.
  • Coupling of a commercially available acid such as 3-hydroxypropanoic acid or 2-hydroxypropanoic acid (as a racemate or as the (S) or (R) isomer) with compound 20 from scheme 2 using a reagent such as HATU in a solvent such as DMF affords compounds of formula 21 and 22 respectively.
  • Acryloyl chloride may be added to compound 20 in a solvent such as DMF with a base such as trimethylamine or diisopropylethylamine to afford a compound of formula 23.
  • Compound 25 can be prepared from a compound of formula 23 by oxidation with an oxidant such as mCPBA in a solvent such as toluene or dichloromethane or by tert-butyl hydrogen peroxide (TBHP) and a cinchona alkaloid catalyst (Sharpless epoxidation).
  • the BTK inhibitory activity, residence time of the inhibitor BTK bound complex, and the ability of the of the compounds of the present disclosure to form an irreversible covalent bond or a reversible covalent bond with Cys 481 (UniprotKB Sequence ID Q06187) of BTK can be tested using the in vitro and/or in vivo assays described in Biological Examples below.
  • the BTK inhibitory activity of the compound of Formula (I) and/or a pharmaceutically acceptable salt thereof of the present disclosure can be tested using the in vitro and/or in vivo assays described in Biological Examples 1, 3, 4, and 5 below. A determination of kinase inhibitory activity by any of those assays is considered to be kinase inhibitory activity within the scope of this disclosure even if any or all of the other assays do not result in a determination of kinase inhibitory activity.
  • the cysteine sulfhydryl group and a carbon atom forming part of the carbon-carbon double bond in the R 5 group in a compound of Formula (I) where R 5 is a group of formula (i), (ii) or (iii) where R a is cyano, can form a reversible, i.e., labile, covalent bond, such as wherein Cys 481 of BTK attacks an electron deficient carbon atom of the carbon-carbon double bond in the above listed R 5 groups in the compound of present disclosure to form a thiol adduct.
  • the electron deficient carbon atom of the olefin is distal to the carbon attached to the R a group (where R a is cyano) i.e., the carbon atom attached to the R b and R c group (see Formula (I) in the compounds of the present disclosure). Therefore, the combination of the R a group (where R a is cyano) and the “—N—CO—, —NSO 2 or —N—SO—” moieties and the olefinic moiety to which they are bonded in the compounds of the present disclosure can increase the reactivity of the olefin to form a thiol adduct with the active site cysteine residue in BTK.
  • the compounds of the present disclosure which are reversible covalent inhibitors can bind with BTK in two different manners.
  • they are believed to also form non-covalent binding (e.g., via van der Waals binding, hydrogen binding, hydrophobic binding, hydrophilic binding, and/or electrostatic charge binding) with BTK, the non-covalent binding sufficient to at least partially inhibit the kinase activity of the BTK.
  • the labile covalent binding occurs between the olefin in the inhibitor and the cysteine 481 residue thiol side chain at or near the site where the inhibitor has the aforementioned non-covalent binding with the BTK.
  • the compounds of the present disclosure which are reversible covalent inhibitors have both a cysteine-mediated covalent binding and a non-covalent binding with the BTK. This is in contrast with non-covalent reversible inhibitors which inhibit the BTK only via non-covalent binding and lack the cysteine-mediated covalent binding.
  • the binding of the compounds of the present disclosure with BTK in the two different manners mentioned above provides a reversible covalent inhibitor having a slow off-rate and a protracted duration of action, in some instances comparable to an irreversible covalent inhibitor without forming permanent irreversible protein adducts.
  • the difference between irreversible and reversible covalent inhibitors, particularly the compounds disclosed herein, can be ascertained utilizing assays disclosed herein.
  • the binding involved an inhibitor that forms a reversible covalent bond with BTK which is stable when the BTK is in certain configurations and susceptible to being broken when the BTK is in different configurations (in both cases under physiologic conditions), whereas the interaction between an inhibitor that forms an irreversible covalent bond with BTK is stable under physiologic conditions even when the BTK is in different configurations.
  • a reversible covalent bond often imparts unique properties related to the residence time of the compound within the cysteine-containing binding site.
  • residence time refers to the temporal duration of the compound-target complex under different conditions (see Copeland R A, Pompliano D L, Meek T D. Drug-target residence time and its implications for lead optimization. Nat. Rev. Drug Discov. 5(9), 730-739 (2006).
  • a reversible covalent bond in a reversible covalent inhibitor as disclosed herein can lead to an extended residence time when compared to a compound that does not form a covalent bond with BTK.
  • the compounds of the present disclosure that are reversible covalent inhibitors have a residence time of at least about 1 h. Residence time may be measured using an occupancy assay in a biochemical or cellular environment (see Biological Example 2 and 9 below). Additionally, residence time may be measured using a functional assay following a defined wash-out period.
  • therapeutically effective amounts of compounds of Formula (I) may range from about 0.01 to about 500 mg per kg patient body weight per day, which can be administered in single or multiple doses.
  • the dosage level will be about 0.1 to about 250 mg/kg per day.
  • the dosage level will be about 0.5 to about 100 mg/kg per day.
  • a suitable dosage level may be about 0.01 to about 250 mg/kg per day, about 0.05 to about 100 mg/kg per day, or about 0.1 to about 50 mg/kg per day. Within this range the dosage can be about 0.05 to about 0.5, about 0.5 to about 5 or about 5 to about 50 mg/kg per day.
  • compositions may be provided in the form of tablets containing about 1.0 to about 1000 milligrams of the active ingredient, particularly about 1.0, 5.0, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient.
  • the actual amount of the compound of this disclosure, i.e., the active ingredient will depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound being utilized, the route and form of administration, and other factors.
  • compositions will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration.
  • routes e.g., oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration.
  • parenteral e.g., intramuscular, intravenous or subcutaneous
  • compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.
  • formulation depends on various factors such as the mode of drug administration (e.g., for oral administration, formulations in the form of tablets, pills or capsules are preferred) and the bioavailability of the drug substance.
  • pharmaceutical formulations have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size.
  • U.S. Pat. No. 4,107,288 describes a pharmaceutical formulation having particles in the size range from 10 to 1,000 nm in which the active material is supported on a crosslinked matrix of macromolecules.
  • 5,145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanoparticles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.
  • Bioavailablity of drugs that decompose at stomach pH can be increased by administration of such drugs in a formulation that releases the drug intraduodenally.
  • compositions are comprised of in general, a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof in combination with a pharmaceutically acceptable excipient such as binders, surfactants, diluents, buffering agents, antiadherents, glidants, hydrophilic or hydrophobic polymers, retardants, stabilizing agents or stabilizers, disintegrants or superdisintegrants, antioxidants, antifoaming agents, fillers, flavors, colors, lubricants, sorbents, preservatives, plasticizers, or sweeteners, or mixtures thereof, which facilitate processing of the compound of Formula (I) (or embodiments thereof disclosed herein) and/or a pharmaceutically acceptable salt thereof into preparations which can be used pharmaceutically.
  • a pharmaceutically acceptable excipient such as binders, surfactants, diluents, buffering agents, antiadherents, glidants, hydrophilic or hydrophobic polymers, retardants, stabilizing agents or stabilizers, disintegr
  • the formulations may include one or more pH adjusting agents or buffering agents, for example, acids such as acetic, boric, citric, fumaric, maleic, tartaric, malic, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate, ammonium chloride, and the like.
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium bicarbonate, ammonium chloride, and the like.
  • Such buffers used as bases may have other counterions than sodium, for example, potassium, magnesium, calcium, ammonium, or other counterions.
  • Such acids, bases and buffers are included in an amount required to maintain pH of the composition in
  • the formulations may also include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • the formulations may also include one or more antifoaming agents to reduce foaming during processing which can result in coagulation of aqueous dispersions, bubbles in the finished film, or generally impair processing.
  • anti-foaming agents include silicon emulsions or sorbitan sesquoleate.
  • the formulations may also include one or more antioxidants, such as non-thiol antioxidants, for example, butylated hydroxytoluene (BHT), sodium ascorbate, ascorbic acid or its derivative, and tocopherol or its derivatives.
  • antioxidants enhance chemical stability where required.
  • Other agents such as citric acid or citrate salts or EDTA may also be added to slow oxidation.
  • the formulations may also include one or more preservatives to inhibit microbial activity.
  • Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide, and cetylpyridinium chloride.
  • the formulations may also include one or more binders.
  • Binders impart cohesive qualities and include, e.g., alginic acid and salts thereof; cellulose derivatives such as carboxymethylcellulose, methylcellulose (e.g., Methocel®), hydroxypropylmethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose (e.g., Klucel®), ethylcellulose (e.g., Ethocel®), and microcrystalline cellulose (e.g., Avicel®); microcrystalline dextrose; amylose; magnesium aluminum silicate; polysaccharide acids; bentonites; gelatin; polyvinyl-pyrrolidone/vinyl acetate copolymer; crosspovidone; povidone; starch; pregelatinized starch; tragacanth, dextrin, a sugar, such as sucrose (e.g., Dipac®), glucose, dextrose, molasses, mannitol, sorb
  • the formulations may also include dispersing agents and/or viscosity modulating agents.
  • Dispersing agents and/or viscosity modulating agents include materials that control the diffusion and homogeneity of a drug through liquid media or a granulation method or blend method. In some embodiments, these agents also facilitate the effectiveness of a coating or eroding matrix.
  • Exemplary diffusion facilitators/dispersing agents include, e.g., hydrophilic polymers, electrolytes, Tween®60 or 80, PEG, polyvinylpyrrolidone (PVP; commercially known as Plasdone®), and the carbohydrate-based dispersing agents such as, for example, hydroxypropyl celluloses (e.g., HPC, H-PC-SL, and HPC-L), hydroxypropyl methylcelluloses (e.g., HPMC K100, RPMC K4M, HPMC K15M, and HPMC K100M), carboxymethylcellulose sodium, methylcellulose, hydroxyethyl-cellulose, hydroxypropyl-cellulose, hydroxypropylmethylcellulose phthalate, hydroxypropyl-methylcellulose acetate stearate (HPMCAS), noncrystalline cellulose, polyethylene oxides, magnesium aluminum silicate, triethanolamine, polyvinyl alcohol (PVA), vinyl pyrrolidone/vinyl acetate copolymer (S
  • Plasticizcers such as cellulose or triethyl cellulose can also be used as dispersing agents.
  • Dispersing agents particularly useful in liposomal dispersions and self-emulsifying dispersions are dimyristoyl phosphatidyl choline, natural phosphatidyl choline from eggs, natural phosphatidyl glycerol from eggs, cholesterol and isopropyl myristate.
  • binder levels of about 10 to about 70% are used in powder-filled gelatin capsule formulations.
  • Binder usage level in tablet formulations varies whether direct compression, wet granulation, roller compaction, or usage of other excipients such as fillers which itself can act as moderate binder. Formulators skilled in art can determine the binder level for the formulations, but binder usage level of up to 90% and more typically up to 70% in tablet formulations is common.
  • the formulations may also include one or more diluents which refer to chemical compounds that are used to dilute the compound of interest prior to delivery. Diluents can also be used to stabilize compounds because they can provide a more stable environment Salts dissolved in buffered solutions (which also can provide pH control or maintenance) are utilized as diluents in the art, including, but not limited to a phosphate buffered saline solution. In certain embodiments, diluents increase bulk of the composition to facilitate compression or create sufficient bulk for homogenous blend for capsule filling.
  • Such compounds include e.g., lactose, starch, mannitol, sorbitol, dextrose, microcrystalline cellulose such as Avicel®; dibasic calcium phosphate, dicalcium phosphate dihydrate; tricalcium phosphate, calcium phosphate; anhydrous lactose, spray-dried lactose; pregelatinized starch, compressible sugar, such as Di-Pac® (Amstar); hydroxypropyl-methylcellulose, hydroxypropylmethylcellulose acetate stearate, sucrose-based diluents, confectioner's sugar; monobasic calcium sulfate monohydrate, calcium sulfate dihydrate; calcium lactate trihydrate, dextrates; hydrolyzed cereal solids, amylose; powdered cellulose, calcium carbonate; glycine, kaolin; mannitol, sodium chloride; inositol, bentonite, and the like.
  • Avicel® dibasic calcium phosphat
  • the formulations may also include one or more disintegrant which includes both the dissolution and dispersion of the dosage form when contacted with gastrointestinal fluid.
  • Disintegration agents or disintegrants facilitate the breakup or disintegration of a substance.
  • disintegration agents include a starch, e.g., a natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or sodium starch glycolate such as Promogel® or Explotab®, a cellulose such as a wood product, methylcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PH 102, Avicel® PH105, Elceme® P100, Emcocel®, Vivacel®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethyl-cellulose (Ac-Di-Sol®), cross-linked carboxymethylcellulose, or cross-linked croscar
  • the formulations may also include erosion facilitators.
  • Erosion facilitators include materials that control the erosion of a particular material in gastrointestinal fluid. Erosion facilitators are generally known to those of ordinary skill in the art. Exemplary erosion facilitators include, e.g., hydrophilic polymers, electrolytes, proteins, peptides, and amino acids.
  • the formulations may also include one or more filling agents which include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • filling agents include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • the formulations may also include one or more flavoring agentsand/or sweeteners e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate, maltol, mannitol, maple, marshmallow, menthol, mint cream, mixed berry, neohesperidine DC
  • the formulations may also include one or more lubricants and glidants which are compounds that prevent, reduce or inhibit adhesion or friction of materials.
  • lubricants include, e.g., stearic acid, calcium hydroxide, talc, sodium stearyl lumerate, a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil, higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol (e.g., PEG4000) or a methoxypolyethylene glycol such as Carbowax®, sodium oleate, sodium benzoate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silic
  • the formulations may also include one or more plasticizers which are compounds used to soften the enteric or delayed release coatings to make them less brittle.
  • plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, triethyl citrate, dibutyl sebacate, triethyl cellulose and triacetin.
  • plasticizers can also function as dispersing agents or wetting agents.
  • the formulations may also include one or more solubilizers which include compounds such as triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, sodium lauryl sulfate, sodium doccusate, vitamin E TPGS, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropylmethyl cellulose, hydroxypropyl cyclodextrins for example Captisol®, ethanol, n-butanol, isopropyl alcohol, cholesterol, bile salts, polyethylene glycol 200-600, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide and the like.
  • the solubilizer is vitamin E TPGS and/or Captisol® or B-hydroxypropylcyclodextrin.
  • the formulations may also include one or more suspending agents which include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K112, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, vinyl pyrrolidone/vinyl acetate copolymer (S630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, hydroxymethylcellulose acetate stearate, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gun, sugars, cell
  • the formulations may also include one or more surfactants which include compounds such as sodium lauryl sulfate, sodium docusate, Tween 20, 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like.
  • surfactants include compounds such as sodium lauryl sulfate, sodium docusate, Tween 20, 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of
  • surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g. octoxynol 10, octoxynol 40. In some embodiments, surfactants may be included to enhance physical stability or for other purposes.
  • the formulations may also include one or more viscosity enhancing agents which include, e.g., methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose acetate stearate, hydroxypropylmethyl cellulose phthalate, carbomer, polyvinyl alcohol alginates, acacia, chitosans and combinations thereof.
  • viscosity enhancing agents include, e.g., methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose acetate stearate, hydroxypropylmethyl cellulose phthalate, carbomer, polyvinyl alcohol alginates, acacia, chitosans and combinations thereof.
  • the formulations may also include one or more wetting agents which include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium docusate, sodium oleate, sodium lauryl sulfate, sodium doccusate, triacetin, Tween 80, vitamin E TPGS, ammonium salts and the like.
  • wetting agents include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium docusate, sodium oleate, sodium lauryl sulfate, sodium doccusate, triacetin, Tween 80, vitamin E
  • compositions disclosed herein can be obtained by mixing one or more solid excipient such as carrier, binder, filling agent, suspending agent, flavoring agent, sweetening agent, disintegrating agent, dispersing agent, surfactant, lubricant, colorant diluent, solubilizer, moistening agent, plasticizer, stabilizer, penetration enhancer, wetting agent, anti-foaming agent, antioxidant, preservative, or one or more combination thereof with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable excipients, if desired, to obtain tablets.
  • solid excipient such as carrier, binder, filling agent, suspending agent, flavoring agent, sweetening agent, disintegrating agent, dispersing agent, surfactant, lubricant, colorant diluent, solubilizer, moistening agent, plasticizer, stabilizer, penetration enhancer, wetting agent, anti-foaming agent, antioxidant, preservative, or one or
  • compositions disclosed herein also include capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Capsules may also be made of polymers such as hypromellose.
  • the capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, lipids, solubilizers, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • formulations can be manufactured by conventional pharmacological techniques.
  • Conventional pharmacological techniques include, e.g., one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, (6) fusion, or (7) extrusion. See, e.g., Lachman et al., The Theory and Practice of Industrial Pharmacy, 3 rd ed. (1986).
  • Other methods include, e.g., spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., wurster coating), tangential coating, top spraying, tableting, extruding, extrusion/spheronization, and the like.
  • the solid dosage forms described herein are enteric coated oral dosage forms, i.e., as an oral dosage form of a pharmaceutical composition as described herein which utilizes an enteric coating to effect the release of the compound in the intestine of the gastrointestinal tract.
  • An “enterically coated” drug and/or tablet refers to a drug and/or tablet that is coated with a substance that remains intact in the stomach but dissolves and releases the drug once the intestine (in one embodiment small intestine) is reached.
  • enteric coating is a material, such as a polymer material or materials which encase the therapeutically active agent core either as a dosage form or as particles.
  • enteric coating material typically, a substantial amount or all of the enteric coating material is dissolved before the therapeutically active agent is released from the dosage form, so as to achieve delayed dissolution of the therapeutically active agent core or particles in the small and/or large intestine.
  • Enteric coatings are discussed, for example, Loyd, V. Allen, Remington: The Science and Practice of Pharmacy, Twenty-first Ed., (Pharmaceutical Press, 2005; and P. J. Tarcha, Polymers for Controlled Drug Delivery, Chapter 3, CRC Press, 1991.
  • Methods for applying enteric coatings to pharmaceutical compositions are well known in the art, and include for example, U.S. Patent Publication No. 2006/0045822.
  • the enteric coated dosage form may be a compressed or molded or extruded tablet (coated or uncoated) containing granules, powder, pellets, beads or particles of the compound of Formula (I) (or any embodiments thereof) and/or a pharmaceutically acceptable salt thereof and/or other excipients, which are themselves coated or uncoated provided at least the tablet or the compound of Formula (I) is coated.
  • the enteric coated oral dosage form may also be a capsule (coated or uncoated) containing pellets, beads or granules of the compound of Formula (I) (or any embodiments thereof) and/or a pharmaceutically acceptable salt thereof and/or other excipients, which are themselves coated or uncoated provided at least one of them is coated.
  • coatings that were originally used as enteric coatings are beeswax and glyceryl monostearate; beeswax, shellac and cellulose; and cetyl alcohol, mastic and shellac as well as shellac and stearic acid (U.S. Pat. No. 2,809,918); polyvinylacetate and ethyl cellulose (U.S. Pat. No. 3,835,221). More recently, the coatings used are neutral copolymers of polymethacrylic acid esters (Eudragit L30D). (F. W. Goodhart et al, Pharm. Tech ., p.
  • any anionic polymer exhibiting a pH-dependent solubility profile can be used as an enteric coating in the methods and compositions described herein to achieve delivery to the intestine. In one embodiment, delivery to the small intestine. In another embodiment, delivery to the duodenum.
  • the polymers described herein are anionic carboxylic polymers. In other embodiments, the polymers and compatible mixtures thereof, and some of their properties, include, but are not limited to:
  • purified lac it is a refined product obtained from the resinous secretion of an insect. This coating dissolves in media of pH>7;
  • acrylic polymers primarily their solubility in biological fluids
  • suitable acrylic polymers include methacrylic acid copolymers and ammonium methacrylate copolymers.
  • the Eudragit series L, S, and RS (manufactured Rohm Pharma and known as Evonik®) are available as solubilized in organic solvent, aqueous dispersion, or dry powders.
  • the Eudragit series RL, NE, and RS are insoluble in the gastrointestinal tract but are permeable and are used primarily for colonic targeting.
  • the Eudragit series L, L-30D and S are insoluble in stomach and dissolve in the intestine and may be selected and formulated to dissolve at a value of pH greater than 5.5 or as low as greater than 5 or as high as greater than 7.
  • Suitable cellulose derivatives are: ethyl cellulose; reaction mixtures of partial acetate esters of cellulose with phthalic anhydride. The performance can vary based on the degree and type of substitution.
  • Cellulose acetate phthalate (CAP) dissolves in pH>6.
  • Aquateric (FMC) is an aqueous based system and is a spray dried CAP pseudolatex with particles ⁇ 1 ⁇ m.
  • Other components in Aquateric can include pluronics, Tweens, and acetylated monoglycerides.
  • Suitable cellulose derivatives include; cellulose acetate tritnellitate (Eastman); methylcellulose (Pharmacoat, Methocel); hydroxypropylmethyl cellulose phthalate (HPMCP); hydroxypropylmethyl cellulose succinate (HPMCS); and hydroxypropylmethylcellulose acetate succinate (HPMCAS e.g., AQOAT (Shin Etsu)).
  • HPMCP such as, HP-50, HP-55, HP-55S, HP-55F grades are suitable.
  • the performance can vary based on the degree and type of substitution.
  • suitable grades of hydroxypropylmethylcellulose acetate succinate include, but are not limited to, AS-LG (LF), which dissolves at pH 5, AS-MG (MF), which dissolves at pH 5.5, and AS-HG (HF), which dissolves at higher pH.
  • AS-LG LF
  • AS-MG MF
  • AS-HG HF
  • polymers are offered as granules, or as fine powders for aqueous dispersions;
  • PVAP Poly Vinyl Acetate Phthalate
  • PVAP dissolves in pH>5, and it is much less permeable to water vapor and gastric fluids.
  • the coating can, and usually does, contain a plasticizer and possibly other coating excipients such as colorants, talc, and/or magnesium stearate, which are well known in the art.
  • Suitable plasticizers include triethyl citrate (Citroflex 2), triacetin (glyceryl triacetate), acetyl triethyl citrate (Citroflec A2), Carbowax 400 (polyethylene glycol 400), diethyl phthalate, tributyl citrate, acetylated monoglycerides, glycerol, fatty acid esters, propylene glycol, and dibutyl phthalate.
  • anionic carboxylic acrylic polymers usually will contain 10-25% by weight of a plasticizer, especially dibutyl phthalate, polyethylene glycol, triethyl citrate and triacetin.
  • Conventional coating techniques such as fluid bed or Wurster coaters, or spray or pan coating are employed to apply coatings. The coating thickness must be sufficient to ensure that the oral dosage form remains intact until the desired site of topical delivery in the intestinal tract is reached.
  • Colorants may be added to the coatings besides plasticizers to solubilize or disperse the coating material, and to improve coating performance and the coated product.
  • a half-thickness, double coat of enteric polymer for instance, Eudragit L30 D-55
  • the inner enteric coat may have a buffer up to pH 6.0 in the presence of 10% citric acid, followed by a final layer of standard Eudragit L 30 D-55.
  • the intactness of the enteric coating may be measured, for example, by the degradation of the drug within the micropellets.
  • the enteric coated dosage forms or pellets may be tested in dissolution testing first in gastric fluid and separately in intestinal fluid as described in USP to determine its function.
  • enteric coated tablets and capsules formulation containing the disclosed compounds can be made by methods well known in the art.
  • tablets containing a compound disclosed herein can be enterically coated with a coating solution containing Eudragit®, diethylphthlate, isopropyl alcohol, talc, and water using a side vented coating pan (Freund Hi-Coater).
  • a multi-unit dosage form comprising enteric-coated pellets that can be incorporated into a tablet or into a capsule can be prepared as follows.
  • the core material for the individually enteric coating layered pellets can be constituted according to different principles. Seeds layered with the active agent (i.e., the compound of Formula (I) (including embodiments disclosed herein) and/or a pharmaceutically acceptable sale thereof), optionally mixed with alkaline substances or buffer, can be used as the core material for the further processing.
  • the seeds which are to be layered with the active agent can be water insoluble seeds comprising different oxides, celluloses, organic polymers and other materials, alone or in mixtures or water-soluble seeds comprising different inorganic salts, sugars, non-pareils and other materials, alone or in mixtures. Further, the seeds may comprise the active agent in the form of crystals, agglomerates, compacts etc.
  • the size of the seeds is not essential for the present invention but may vary between approximately 0.1 and 2 mm.
  • the seeds layered with the active agent are produced either by powder or solution/suspension layering using for instance granulation or spray coating layering equipment.
  • active agent Before the seeds are layered, active agent may be mixed with further components.
  • Such components can be binders, surfactants, fillers, disintegrating agents, alkaline additives or other and/or pharmaceutically acceptable ingredients alone or in mixtures.
  • the binders are for example polymers such as hydroxypropyl methylcellulose (HPMC), hydroxypropyl-cellulose (HPC), carboxymethylcellulose sodium, polyvinyl pyrrolidone (PVP), or sugars, starches or other pharmaceutically acceptable substances with cohesive properties.
  • Suitable surfactants are found in the groups of pharmaceutically acceptable non-ionic or ionic surfactants such as for instance sodium lauryl sulfate.
  • the active agent optionally mixed with suitable constituents can be formulated into a core material.
  • Said core material may be produced by extrusion/spheronization, balling or compression utilizing conventional process equipment.
  • the size of the formulated core material is approximately between 0.1 and 4 mm and for example, between 0.1 and 2 mm.
  • the manufactured core material can further be layered with additional ingredients comprising the active agent and/or be used for further processing.
  • the active agent is mixed with pharmaceutical constituents to obtain preferred handling and processing properties and a suitable concentration of the active agent in the final preparation.
  • Pharmaceutical constituents such as fillers, binders, lubricants, disintegrating agents, surfactants and other pharmaceutically acceptable additives may be used.
  • the aforementioned core material can be prepared by using spray drying or spray congealing technique.
  • the pellets Before applying the enteric coating layer(s) onto the core material in the form of individual pellets, the pellets may optionally be covered with one or more separating layer(s) comprising pharmaceutical excipients optionally including alkaline compounds such as pH-buffering compounds.
  • This/these separating layer(s) separate(s) the core material from the outer layers being enteric coating layer(s).
  • This/these separating layer(s) protecting the core material of active agent should be water soluble or rapidly disintegrating in water.
  • a separating layer(s) can be optionally applied to the core material by coating or layering procedures in suitable equipments such as coating pan, coating granulatooror in a fluidized bed apparatus using water and/or organic solvents for the coating process.
  • the separating layer(s) can be applied to the core material by using powder coating technique.
  • the materials for the separating layers are pharmaceutically acceptable compounds such as, for instance, sugar, polyethylene glycol, polyvinylpyrrolidone, polyvinyl alcohol, polyvinyl acetate, hydroxypropyl cellulose, methylcellulose, ethylcellulose, hydroxypropyl methyl cellulose, carboxymethylcellulose sodium, water soluble salts of enteric coating polymers and others, used alone or in mixtures.
  • Additives such as plasticizers, colorants, pigments, fillers anti-tacking and anti-static agents, such as for instance magnesium stearate, titanium dioxide, talc and other additives may also be included into the separating layer(s).
  • the optional separating layer When the optional separating layer is applied to the core material it may constitute a variable thickness.
  • the maximum thickness of the separating layer(s) is normally only limited by processing conditions.
  • the separating layer may serve as a diffusion barrier and may act as a pH-buffering zone.
  • the optionally applied separating layer(s) is not essential for the invention. However, the separating layer(s) may improve the chemical stability of the active substance and/or the physical properties of the novel multiple unit tableted dosage form.
  • the separating layer may be formed in situ by a reaction between an enteric coating polymer layer applied on the core material and an alkaline reacting compound in the core material.
  • the separating layer formed comprises a water soluble salt formed between the enteric coating layer polymer(s) and an alkaline reacting compound which is in the position to form a salt
  • enteric coating layers are applied onto the core material or onto the core material covered with separating layer(s) by using a suitable coating technique.
  • the enteric coating layer material may be dispersed or dissolved in either water or in suitable organic solvents.
  • enteric coating layer polymers one or more, separately or in combination, of the following can be used, e.g. solutions or dispersions of methacrylic acid copolymers, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, cellulose acetate trimellitate, carboxymethylethylcellulose, shellac or other suitable enteric coating polymer(s).
  • the enteric coating layers contain pharmaceutically acceptable plasticizers to obtain the desired mechanical properties, such as flexibility and hardness of the enteric coating layers.
  • plasticizers are for instance, but not restricted to triacetin, citric acid esters, phthalic acid esters, dibutyl sebacate, cetyl alcohol, polyethylene glycols, polysorbates or other plasticizers.
  • the amount of plasticizer is optimized for each enteric coating layer formula, in relation to the selected enteric coating layer polymer(s), selected plasticizer(s) and the applied amount of said polymer(s), in such a way that the mechanical properties, i.e. flexibility and hardness of the enteric coating layer(s), for instance exemplified as Vickers hardness, are adjusted so that if a tablet is desired the acid resistance of the pellets covered with enteric coating layer(s) does not decrease significantly during compression of pellets into tablets.
  • the amount of plasticizer is usually above 5% by weight of the enteric coating layer polymer(s), such as 15-50% and further such as 20-50%. Additives such as dispersants, colorants, pigments polymers e.g.
  • poly(ethylacrylate, methylmethacrylate), anti-tacking and anti-foaming agents may also be included into the enteric coating layer(s).
  • Other compounds may be added to increase film thickness and to decrease diffusion of acidic gastric juices into the acid susceptible material.
  • the maximum thickness of the applied enteric coating is normally only limited by processing conditions and the desired dissolution profile.
  • Pellets covered with enteric coating layer(s) may optionally further be covered with one or more over-coating layer(s).
  • the over-coating layer(s) should be water soluble or rapidly disintegrating in water.
  • the over-coating layer(s) can be applied to the enteric coating layered pellets by coating or layering procedures in suitable equipments such as coating pan, coating granulator or in a fluidized bed apparatus using water and/or organic solvents for the coating or layering process.
  • the materials for over-coating layers are chosen among pharmaceutically acceptable compounds such as, for instance sugar, polyethylene glycol, polyvinylpyrrolidone, polyvinyl alcohol, polyvinyl acetate, hydroxypropyl cellulose, methylcellulose, ethylcellulose, hydroxypropyl methyl cellulose, carboxymethylcellulose sodium and others, used alone or in mixtures.
  • pharmaceutically acceptable compounds such as, for instance sugar, polyethylene glycol, polyvinylpyrrolidone, polyvinyl alcohol, polyvinyl acetate, hydroxypropyl cellulose, methylcellulose, ethylcellulose, hydroxypropyl methyl cellulose, carboxymethylcellulose sodium and others, used alone or in mixtures.
  • Additives such as plasticizers, colorants, pigments, fillers, anti-tacking and anti-static agents, such for instance magnesium stearate, titanium dioxide, talc and other additives may also be included into the over-coating layer(s).
  • the over-coating layer may further prevent potential agglomeration of enteric coating layered pellets, further it may protect the enteric coating layer towards cracking during the compaction process and enhance the tableting process.
  • the maximum thickness of the applied over-coating layer(s) is normally limited by processing conditions and the desired dissolution profile.
  • the over-coating layer may also be used as a tablet film coating layer.
  • Enteric coating of soft gelatin capsules may contain an emulsion, oil, microemulsion, self-emulsifying system, lipid, triglycerides, polyethylene glycol, surfactants, other solubilizers and the like, and combinations thereof, to solubilize the active agent.
  • the flexibility of the soft gelatin capsule is maintained by residual water and plasticizer.
  • the gelatin may be dissolved in water so that spraying must be accomplished at a rate with relatively low relative humidity such as can be accomplished in a fluid bed or Wurster. In addition, drying should be accomplished without removing the residual water or plasticizer causing cracking of the capsule shell.
  • enteric coated capsules may be prepared by: a) rotating capsules in a flask or dipping capsules in a solution of the gently heated enteric coating material with plasticizer at the lowest possible temperature or b) in a lab scale sprayer/fluid bed and then drying.
  • water-in-oil emulsion
  • water-in-oil formulations can provide a lipid layer, which can interact favorably with lipids in cells of the body, and can increase the partition of the formulation onto the membranes of cells. Such partition can increase the absorption of drugs in such formulations into the circulation and therefore can increase the bioavailability of the drug.
  • the water-in-oil emulsion contains an oily phase composed of medium or long chain carboxylic acids or esters or alcohols thereof, a surfactant or a surface active agent, and an aqueous phase containing primarily water and the active agent.
  • Medium and long chain carboxylic acids are those ranging from C 8 to C 22 with up to three unsaturated bonds (also branching).
  • saturated straight chain acids are n-dodecanoic acid, n-tetradecanoic acid, n-hexadecanoic acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, montanic acid and melissic acid.
  • unsaturated monoolefinic straight chain monocarboxylic acids are also useful.
  • these are oleic acid, gadoleic acid and erucic acid.
  • unsaturated (polyolefinic) straight chain monocarboxylic acids examples of these are linoleic acid, ricinoleic acid, linolenic acid, arachidonic acid and behenolic acid.
  • Useful branched acids include, for example, diacetyl tartaric acid.
  • Unsaturated olefinic chains may also be hydroxylated or ethoxylated to prevent oxidation or to alter the surface properties.
  • long chain carboxylic acid esters include, but are not limited to, those from the group of: glyceryl monostearates; glyceryl monopalmitates; mixtures of glyceryl monostearate and glyceryl monopalmitate; glyceryl monolinoleate; glyceryl monooleate; mixtures of glyceryl monopalmitate, glyceryl monostearate, glyceryl monooleate and glyceryl monolinoleate; glyceryl monolinolenate; glyceryl monogadoleate; mixtures of glyceryl monopalmitate, glyceryl monostearate, glyceryl monooleate, glyceryl monolinoleate, glyceryl monolinolenate and glyceryl monogadoleate; acetylated glycerides such as distilled acetylated monoglycerides; mixtures of
  • the self-emulsifying long chain carboxylic acid esters include those from the groups of stearates, pamitates, ricinoleates, oleates, behenates, ricinolenates, myristates, laurates, caprylates, and caproates.
  • the oily phase may comprise a combination of 2 or more of the long chain carboxylic acids or esters or alcohols thereof.
  • medium chain surfactants may be used and the oil phase may comprise a mixture of caprylic/capric triglyceride and C 8 /C 10 mono-/di-glycerides of caprylic acid, glyceryl caprylate or propylene glycol monocaprylate or their mixtures.
  • the alcohols that can be used are exemplified by the hydroxyl forms of the carboxylic acids exemplified above and also strearyl alcohol.
  • the surfactant may comprise: Tween@ (polyoxyethylene sorbate) family of surfactants, Span® (sorbitan long chain carboxylic acid esters) family of surfactants, Pluronic® (ethylene or propylene oxide block copolymers) family of surfactants, Labrasol®, Labrafil® and Labrafac® (each polyglycolyzed glycerides) families of surfactants, sorbitan esters of oleate, stearate, laurate or other long chain carboxylic acids, poloxamers (polyethylene-polypropylene glycol block copolymers or Pluronic®), other sorbitan or sucrose long chain carboxylic acid esters, mono and diglycerides, PEG derivative
  • the aqueous phase may optionally comprise the active agent suspended in water and a buffer.
  • such emulsions are coarse emulsions, microemulsions and liquid crystal emulsions.
  • such emulsion may optionally comprise a permeation enhancer.
  • spray-dried dispersions or microparticles or nanoparticles containing encapsulated microemulsion, coarse emulsion or liquid crystal can be used.
  • the solid dosage forms described herein are non-enteric time-delayed release dosage forms.
  • non-enteric time-delayed release refers to the delivery so that the release of the drug can be accomplished at some generally predictable location in the intestinal tract more distal to that which would have been accomplished if there had been no delayed release alterations.
  • the method for delay of release is a coating that becomes permeable, dissolves, ruptures, and/or is no longer intact after a designed duration.
  • the coating in the time-delayed release dosage forms can have a fixed time to erode after which the drug is released (suitable coating include polymeric coating such as HPMC, PEO, and the like) or has a core comprised of a superdisinegrant(s) or osmotic agent(s) or water attractant such as a salt, hydrophilic polymer, typically polyethylene oxide or an alkylcellulose, salts such as sodium chloride, magnesium chloride, sodium acetate, sodium citrate, sugar, such as glucose, lactose, or sucrose, or the like, which draw water through a semi-permeable membrane or a gas generating agent such as citric acid and sodium bicarbonate with or without an acid such as citric acid or any of the aforementioned acids incorporated in dosage forms.
  • a superdisinegrant(s) or osmotic agent(s) or water attractant such as a salt, hydrophilic polymer, typically polyethylene oxide or an alkylcellulose, salts such as sodium chloride,
  • the semi-permeable membrane while mostly not permeable to the drug nor the osmotic agent, is permeable to water that permeates at a near constant rate to enter the dosage form to increase the pressure and ruptures after the swelling pressure exceeds a certain threshold over a desired delay time.
  • the permeability through this membrane of the drug should be less than 1/10 than water and in one embodiment less than 1/100 the water permeability.
  • a membrane could become porous by leaching an aqueous extractable over a desired delay time.
  • Osmotic dosage forms have been described in Theeuwes U.S. Pat. No. 3,760,984, and an osmotic bursting dosage form is described in Baker U.S. Pat. No. 3,952,741.
  • This osmotic bursting dosage form can provide a single pulse of release or multiple pulses if different devices with different timings are employed.
  • the timing of the osmotic burst may be controlled by the choice of polymer and the thickness or the area of the semipermeable membrane surrounding the core that contains both the drug and the osmotic agent or attractant. As the pressure in the dosage form increase with additional permeated water, the membrane elongates until its breaking point, and then the drug is released.
  • specific areas of rupture can be created in the membrane by having a thinner, weaker area in the membrane or by adding a weaker material to an area of the coating membrane.
  • Some preferred polymers with high water permeabilities that may be used as semipermeable membranes are cellulose acetate, cellulose acetate butyrate, cellulose nitrate, crosslinked polyvinyl, alcohol, polyurethanes, nylon 6, nylon 6.6, and aromatic nylon. Cellulose acetate is an especially preferred polymer.
  • the time-delayed coating that begins its delay to releasing drug after the enteric coating is at least partially dissolved is comprised of hydrophilic, erodible polymers that upon contact with water begin to gradually erode over time.
  • hydrophilic, erodible polymers include cellulose polymers and their derivatives including, but not limited to, hydroxyalkyl celluloses, hydroxymethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, carboxymethylcellulose, microcrystalline cellulose; polysaccharides and their derivatives; polyalkylene oxides, such as polyethylene oxide or polyethylene glycols, particularly high molecular weight polyethylene glycols; chitosan; poly(vinyl alcohol); xanthan gum; maleic anhydride copolymers; poly(vinyl pyrrolidone); starch and starch-based polymers; maltodextrins; poly (2-ethyl-2-oxazoline); poly(ethyleneimine); polyure
  • Some preferred erodible hydrophilic polymers suitable for forming the erodible coating are poly(ethylene oxide), hydroxypropyl methyl cellulose, and combinations of poly(ethylene oxide) and hydroxypropyl methyl cellulose.
  • Poly(ethylene oxide) is used herein to refer to a linear polymer of unsubstituted ethylene oxide.
  • the molecular weight of the poly(ethylene oxide) polymers can range from about 10 5 Daltons to about 10 7 . Daltons.
  • a preferred molecular weight range of poly(ethylene oxide) polymers is from about 2 times 10 5 to 2 times 10 6 Daltons and is commercially available from The Dow Chemical Company (Midland, Mich.) referred to as SENTRY® POLYOXTM water-soluble resins, NF (National Formulary) grade.
  • SENTRY® POLYOXTM water-soluble resins NF (National Formulary) grade.
  • other hydrophilic agents such as salts or sugars, like glucose, sucrose, or lactose, that promote erosion or
  • the time-delayed dosage form can be a mechanical pill such as an Enterion® capsule or pH sensitive capsule which can release the drug after a pre-programmed time or when it receives a signal which can be transmitted or once it leaves the stomach.
  • the amount of the compound of the disclosure in a formulation can vary within the full range employed by those skilled in the art.
  • the formulation will contain, on a weight percent (wt %) basis, from about 0.01-99.99 wt % of a compound of Formula (I) based on the total formulation, with the balance being one or more suitable pharmaceutical excipients.
  • the compound is present at a level of about 1-80 wt %.
  • the compounds of the present disclosure may be used in combination with one or more other drugs in the treatment of diseases or conditions for which compounds of the present disclosure or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone.
  • Such other drug(s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of the present disclosure.
  • a pharmaceutical composition in unit dosage form containing such other drugs and the compound of the present disclosure is preferred.
  • the combination therapy may also include therapies in which the compound of the present disclosure and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of the present disclosure and the other active ingredients may be used in lower doses than when each is used singly.
  • compositions of the present disclosure also include those that contain one or more other active ingredients, in addition to a compound of the present disclosure.
  • the above combinations include combinations of a compound of the present disclosure not only with one other active compound, but also with two or more other active compounds.
  • compounds of the present disclosure may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which compounds of the present disclosure are useful.
  • Such other drugs may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of the present disclosure.
  • a pharmaceutical composition such as a fixed-combination drug product, containing such other drugs in addition to the compound of the present disclosure is preferred.
  • compositions of the present disclosure also include those that also contain one or more other active ingredients, in addition to a compound of the present disclosure.
  • the weight ratio of the compound of the present disclosure to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
  • a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof can be used with one or more of the following therapeutic agents in any combination: immunosuppressants (e.g., tacrolimus, cyclosporin, rapamicin, methotrexate, cyclophosphamide, azathioprine, mercaptopurine, mycophenolate, or FTY720), glucocorticoids (e.g., prednisone, cortisone acetate, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone), non-steroidal anti-inflammatory drugs (e.g., salicylates, arylalkanoic acids, 2-aryl
  • binding proteins e.g., infliximab, etanercept, or adalimumab
  • abatacept anakinra
  • interferon-.beta. interferon-.gamma.
  • interleukin-2 allergy vaccines
  • antihistamines antileukotrienes
  • beta-agonists theophylline, or anticholinergics.
  • the subject can be treated with a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof in any combination with one or more other anti-cancer agents.
  • one or more of the anti-cancer agents are proapoptotic agents.
  • anti-cancer agents include, but are not limited to, any of the following: gossyphol, genasense, polyphenol E, Chlorofusin, all trans-retinoic acid (ATRA), bryostatin, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), 5-aza-2′-deoxycytidine, all trans retinoic acid, doxorubicin, vincristine, etoposide, gemcitabine, imatinib (GleevecTM), geldanamycin, 17-N-Allylamino-17-Demethoxygeldanamycin (17-AAG), flavopiridol, LY294002, bortezomib, trastuzumab, BAY 11-7082, PKC412, or PD184352, TaxolTM, also referred to as “paclitaxel,” which is a well-known anti-cancer drug which acts by enhancing and stabilizing microtubule formation
  • anti-cancer agents for use in combination with a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include inhibitors of mitogen-activated protein kinase signaling, e.g., U0126, PD98059, PD184352, PD0325901, ARRY-142886, SB239063, SP600125, BAY 43-9006, wortmannin, or LY294002; Syk inhibitors; mTOR inhibitors; and antibodies (e.g., rituxan).
  • mitogen-activated protein kinase signaling e.g., U0126, PD98059, PD184352, PD0325901, ARRY-142886, SB239063, SP600125, BAY 43-9006, wortmannin, or LY294002
  • Syk inhibitors e.g., mTOR inhibitors
  • mTOR inhibitors e.g., rituxan
  • anti-cancer agents that can be employed in combination with a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include Adriamycin, Dactinomycin, Bleomycin, Vinblastine, Cisplatin, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide;
  • anti-cancer agents that can be employed in combination with a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include: 20-epi-1, 25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators
  • anticancer agents that can be employed in combination with a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include alkylating agents, antimetabolites, natural products, or hormones, e.g., nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc.), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, etc.), or triazenes (decarbazine, etc.).
  • nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc.
  • alkyl sulfonates e.g., busulfan
  • nitrosoureas e.g., carmustine, lomusitne, etc.
  • triazenes decarbazine, etc.
  • antimetabolites include but are not limited to folic acid analog (e.g., methotrexate), or pyrimidine analogs (e.g., Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin).
  • folic acid analog e.g., methotrexate
  • pyrimidine analogs e.g., Cytarabine
  • purine analogs e.g., mercaptopurine, thioguanine, pentostatin.
  • Examples of natural products useful in combination with a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include but are not limited to vinca alkaloids (e.g., vinblastin, vincristine), epipodophyllotoxins (e.g., etoposide), antibiotics (e.g., daunorubicin, doxorubicin, bleomycin), enzymes (e.g., L-asparaginase), or biological response modifiers (e.g., interferon alpha).
  • vinca alkaloids e.g., vinblastin, vincristine
  • epipodophyllotoxins e.g., etoposide
  • antibiotics e.g., daunorubicin, doxorubicin, bleomycin
  • enzymes e.g., L-asparaginase
  • biological response modifiers e.g., interferon alpha
  • alkylating agents that can be employed in combination a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include, but are not limited to, nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, melphalan, etc.), ethylenimine and methylmelamines (e.g., hexamethlymelamine, thiotepa), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, semustine, streptozocin, etc.), or triazenes (decarbazine, etc.).
  • nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, melphalan, etc.
  • ethylenimine and methylmelamines e.g., hexamethlymelamine
  • antimetabolites include, but are not limited to folic acid analog (e.g., methotrexate), or pyrimidine analogs (e.g., fluorouracil, floxuridine, Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin.
  • folic acid analog e.g., methotrexate
  • pyrimidine analogs e.g., fluorouracil, floxuridine, Cytarabine
  • purine analogs e.g., mercaptopurine, thioguanine, pentostatin.
  • hormones and antagonists useful in combination a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof include, but are not limited to, adrenocorticosteroids (e.g., prednisone), progestins (e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate), estrogens (e.g., diethlystilbestrol, ethinyl estradiol), antiestrogen (e.g., tamoxifen), androgens (e.g., testosterone propionate, fluoxymesterone), antiandrogen (e.g., flutamide), gonadotropin releasing hormone analog (e.g., leuprolide).
  • adrenocorticosteroids e.g., prednisone
  • progestins e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyproge
  • platinum coordination complexes e.g., cisplatin, carboblatin
  • anthracenedione e.g., mitoxantrone
  • substituted urea e.g., hydroxyurea
  • methyl hydrazine derivative e.g., procarbazine
  • adrenocortical suppressant e.g., mitotane, aminoglutethimide
  • anti-cancer agents which act by arresting cells in the G2-M phases due to stabilized microtubules and which can be used in combination with an BTK inhibitor compound of the disclosure include without limitation the following marketed drugs and drugs in development: Erbulozole (also known as R-55104), Dolastatin 10 (also known as DLS-10 and NSC-376128), Mivobulin isethionate (also known as CI-980), Vincristine, NSC-639829, Discodermolide (also known as NVP-XX-A-296), ABT-751 (Abbott, also known as E-7010), Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C), Spongistatins (such as Spongistatin 1, Spongistatin 2, Spongistatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, Spongistatin 7, Spongistatin 8, and Spongistatin 9), Cemadotin
  • anti-thromboembolic agents include, but are not limited any of the following: thrombolytic agents (e.g., alteplase anistreplase, streptokinase, urokinase, or tissue plasminogen activator), heparin, tinzaparin, warfarin, dabigatran (e.g., dabigatran etexilate), factor Xa inhibitors (e.g., fondaparinux, draparinux, rivaroxaban, DX-9065a, otamixaban, LY517717, or YM150), ticlopidine, clopidogrel, CS-747 (prasugrel, LY640315), ximelag
  • tert-butyl (R,E)-3-(4-(((dimethylamino)-methylene)amino)-2-oxo-2,3-dihydro-1H-imidazo[4,5-c]pyridin-1-yl)piperidine-1-carboxylate 200 mg, 0.51 mmol, 1.00 equiv
  • dichloromethane (20 mL
  • TEA 208 mg, 2.06 mmol, 4.00 equiv
  • TEMPO 88.5 mg, 0.57 mmol, 1.10 equiv
  • Cu(OAc) 2 (46.7 mg, 0.26 mmol, 0.50 equiv).
  • the reaction was then quenched by the addition of 50 mL of water.
  • the resulting solution was extracted with 3 ⁇ 50 mL of dichloromethane and the organic layers combined.
  • the resulting mixture was washed with 50 mL of brine.
  • the mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (30:1).
  • the resulting solution was stirred for 16 h at rt.
  • the crude product was purified by Prep-HPLC with the following conditions (Column, Gemini-NX C18 AXAI Packed, 21.2 ⁇ 150 mm Sum 11 nm; mobile phase, WATER WITH 0.05% TFA and ACN (20.0% ACN up to 50.0% in 8 min); Detector, 254 nm.
  • HMPA 6.0 mL
  • LDA 16.8 mmol
  • THF dry THF (20 mL)
  • acetonitrile 690 mg, 16.8 mmol
  • the solution was stirred for 30 min, and (2-bromoethoxy)(tert-butyl)dimethylsilane (3.4 g, 14.3 mmol) in THF (15 mL) was added dropwise. Stirring was continued for 2 h, after which a second portion of LDA (16.8 mmol in 20 mL THF) was added.
  • a Caliper-based kinase assay (Caliper Life Sciences, Hopkinton, Mass.) was used to measure inhibition of BTK kinase activity of a compound of the present disclosure.
  • Serial dilutions of test compounds were incubated with human recombinant BTK (0.5 nM), ATP (16 M) and a phosphoacceptor peptide substrate FAM-GEEPLYWSFPAKKK-NH 2 (1 ⁇ M) at room temperature for 3 h.
  • the reaction was then terminated with EDTA, final concentration 20 mM and the phosphorylated reaction product was quantified on a Caliper Desktop Profiler (Caliper LabChip 3000). Percent inhibition was calculated for each compound dilution and the concentration that produced 50% inhibition was calculated. This value is presented as the IC 50 .
  • the ICso for certain compounds of the disclosure are provided below.
  • the potency of compounds for inhibition of BTK activity can be assessed by binding of compounds to the target in human peripheral blood mononuclear cells (PBMC) that contain BTK.
  • PBMC peripheral blood mononuclear cells
  • the extent of BTK occupancy is measured after treating the cells with compounds and detecting unoccupied BTK through binding of occupancy of (R,E)-N-(2-(4-(4-(3-(4-amino-3-(2-fluoro-4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)piperidin-1-yl)-4-oxobut-2-en-1-yl)piperazin-1-yl)ethyl)-3-(5,5-difluoro-7,9-dimethyl-5H-4l4,5l4-dipyrrolo[1,2-c:2′,1′-f][1,3,2]diazaborinin-2-yl)propanamide as the probe.
  • This assay was also modified to measure the durability of BTK binding in PBMCs.
  • a concentration of 2 uM compound was added to human whole blood for 1 h.
  • PBMCs were isolated using Ficoll, washed, and resuspended in media for either 4 h or 18 h at 37° C.
  • the occupancy probe was added to a concentration of 1 uM for each sample for 1 h, and BTK occupancy was then determined in the same manner as described above.
  • BTK inhibitors have been shown to block B cell activation as measured by CD69 expression (see Karp, R., et. al., Inhibition of BTK with AVL-292 Translates to Protective Activity in Animal Models of Rheumatoid Arthritis. Inflammation Research Association Meeting, September, 2010).
  • CD69 was expressed following B cell activation as a measure of BTK activity in whole blood.
  • mice are injected with an emulsion of Type II collagen in Complete Freund's Adjuvant. Mice are boosted 21 days later to synchronize development of disease. After development of mild disease, animals are enrolled in the study and randomized. Dosing is oral, Q.D. typically for 11 days with test compound or dexamethasone (0.2 mg/kg) as control. One group receives vehicle alone.
  • Clinical scoring (0-4) is based on the extent of swelling and severity of arthritis. Scores for all four paws are added for maximum score of 16. Anti-collagen antibodies and total Ig are measured for each animal by Elisa at the end of the study (Bolder BioPath, Boulder, Colo.).
  • a compound and/or pharmaceutically acceptable salt of the present disclosure at a concentration 10 times greater than its IC 50 value is added to a solution of protein kinase (5 nM) in a buffer containing 20 mM Hepes [pH 7.5], 5 mM MgCl 2 , 0.01% Triton X-100, and 1 mM dithiothreitol.
  • the reactions are transferred to a dialysis cassette (0.1-0.5 mL Slide-A-Lyzer, MWCO 10 kDa, Pierce) and dialyzed against 1 L of buffer (20 mM Hepes [pH 7.5], 5 mM MgCl 2 , 0.01% Triton X-100, and 1 mM dithiothreitol.) at 22° C.
  • the dialysis buffer is exchanged twice per day until the end of the experiment. Aliquots are removed from the dialysis cassettes every 24 h and analyzed for protein kinase activity. Kinase activity for each sample is normalized to the DMSO control for that time point and expressed as the mean ⁇ SD. It will be observed that the kinase activity will return on dialysis for a compound of the present disclosure where R a is cyano and will not return for for a compound of the present disclosure where R a is hydrogen or fluoro.
  • a protein kinase that is inhibited by compound and/or a pharmaceutically acceptable salt of the present disclosure may be subjected to mass spectral analysis to assess the formation of permanent, irreversible covalent adducts.
  • Suitable analytical methods to examine intact full protein or peptide fragments generated upon tryptic cleavage of the protein kinase are generally known in the art (see Lipton, Mary S., Ljiljana, Pasa-Tolic, Eds. Mass Spectrometry of Proteins and Peptides, Methods and Protocols, Second Edition. Humana Press. 2009).
  • Such methods identify permanent, irreversible covalent protein adducts by observing a mass peak that corresponds to the mass of a control sample plus the mass of an irreversible adduct. Two such methods are described below.
  • a protein kinase (5 ⁇ M) (such as BTK) is incubated with a compound of the present disclosure (25 ⁇ M, 5 equiv) for 1 h at room temperature in buffer (20 mM Hepes [pH 8.0], 100 mM NaCl, 10 mM MgCl 2 ).
  • a control sample is also prepared which does not have a compound of the present disclosure.
  • the reaction is stopped by adding an equal volume of 0.4% formic acid, and the samples are analyzed by liquid chromatography (Microtrap C18 Protein column [Michrom Bioresources], 5% MeCN, 0.2% formic acid, 0.25 mL/min; eluted with 95% MeCN, 0.2% formic acid) and in-line ESI mass spectrometry (LCT Premier, Waters).
  • Molecular masses of the protein kinase and any adducts may be determined with MassLynx deconvolution software (see patent application WO2014 011900, and PCT/US2010/048916).
  • High-resolution intact mass spectrometry analysis of protein kinase, such as BTK, that is inhibited by a compound of the present disclosure where R a is cyano will reveal a spectrum similar to the kinase in the absence of inhibitor (e.g. control sample). There will be no formation of a new peak in the mass spectrum corresponding to the molecular mass of the kinase plus the molecular mass of the compound.
  • high-resolution intact mass spectrometry analysis of a protein kinase that is inhibited by a a compound of the present disclosure where R a is hydrogen or fluoro will reveal formation of a new peak (e.g.
  • a protein (10-100 pmols) is incubated with a compound and/or a pharmaceutically acceptable salt of the present disclosure (100-1000 pmols, 10 equiv) for 3 h prior to tryptic digestion.
  • Iodoacetamide may be used as the alkylating agent after compound incubation.
  • a control sample is also prepared which does not utilize the compound and/or a pharmaceutically acceptable salt of the present disclosure.
  • the following is a protocol that can be used to distinguish whether a compound displays a slow or non-existent dissociation rate from BTK, such as typically would occur if a covalent bond is formed between the compound and the target.
  • the read-out for slow dissociation is the ability of the compound of interest to block binding of a high affinity fluorescent tracer molecule to the kinase active site, as detected using time-resolved fluorescence resonance energy transfer (TR-FRET).
  • TR-FRET time-resolved fluorescence resonance energy transfer
  • the first step of the procedure was incubation of 500 nM BTK (Invitrogen Cat. #PV3587) with 1.5 ⁇ M of a compound of the present disclosure for 30 min in a volume of 10 ⁇ L.
  • the mixture was then diluted 5-fold by addition of 40 ⁇ L of buffer.
  • a 10 ⁇ L volume of the diluted kinase/compound solution was then added to a well of a small volume 384 well plate (such as Greiner Cat. #784076).
  • a competition solution containing both a high affinity fluorescent tracer and an antibody coupled to Europium was prepared.
  • the competition solution contained 1.5 ⁇ M Tracer 178 (Invitrogen Cat.
  • the competition solution also contained 80 nM of an Anti-polyhistidine antibody coupled to Europium (Invitrogen Cat. #PV5596) which is designed to bind the polyhistidine purification tag in BTK.
  • Binding of the tracer to BTK is detected using TR-FRET between the Europium moiety of the Anti-histidine antibody and the AlexaFluor 647 group of Tracer 178. Binding was evaluated using a Perkin Elmer Envision instrument (Model 2101) equipped with filters and mirrors compatible with LANCE-type TR-FRET experiments. Data were plotted at percentage of signal obtained in the absence of competitor compound.
  • the background signal was obtained by omission of BTK from the reaction. If the compound is an irreversible covalent inhibitor, tracer will be completely blocked from binding to the target throughout the entire course of the experiment. If the compound is a reversible covalent inhibitor, the tracer will bind the target as the compound dissociates from the target. For the durability measurements, the range of occupancy for the compounds disclosed herein at 1, 6, and 24 h of washout is shown below.
  • Compound of the disclosure e.g., compound 1 in 2% HPMC, 1% Tween 80 in DI water, pH 2.2 with MSA, q.s. to at least 20 mg/mL

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Pain & Pain Management (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Transplantation (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
US15/578,623 2015-06-03 2016-06-02 Tyrosine kinase inhibitors Abandoned US20180162861A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/578,623 US20180162861A1 (en) 2015-06-03 2016-06-02 Tyrosine kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562170547P 2015-06-03 2015-06-03
US201562271689P 2015-12-28 2015-12-28
PCT/US2016/035588 WO2016196840A1 (en) 2015-06-03 2016-06-02 Tyrosine kinase inhibitors
US15/578,623 US20180162861A1 (en) 2015-06-03 2016-06-02 Tyrosine kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/035588 A-371-Of-International WO2016196840A1 (en) 2015-06-03 2016-06-02 Tyrosine kinase inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/029,184 Continuation US20210171526A1 (en) 2015-06-03 2020-09-23 Tyrosine Kinase Inhibitors

Publications (1)

Publication Number Publication Date
US20180162861A1 true US20180162861A1 (en) 2018-06-14

Family

ID=56118090

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/578,623 Abandoned US20180162861A1 (en) 2015-06-03 2016-06-02 Tyrosine kinase inhibitors
US15/410,716 Active US9688676B2 (en) 2015-06-03 2017-01-19 Tyrosine kinase inhibitors
US17/029,184 Abandoned US20210171526A1 (en) 2015-06-03 2020-09-23 Tyrosine Kinase Inhibitors
US17/974,613 Pending US20230151012A1 (en) 2015-06-03 2022-10-27 Tyrosine kinase inhibitors

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/410,716 Active US9688676B2 (en) 2015-06-03 2017-01-19 Tyrosine kinase inhibitors
US17/029,184 Abandoned US20210171526A1 (en) 2015-06-03 2020-09-23 Tyrosine Kinase Inhibitors
US17/974,613 Pending US20230151012A1 (en) 2015-06-03 2022-10-27 Tyrosine kinase inhibitors

Country Status (32)

Country Link
US (4) US20180162861A1 (hu)
EP (3) EP4112618A1 (hu)
JP (2) JP6646072B2 (hu)
KR (1) KR102552653B1 (hu)
CN (2) CN106459049B (hu)
AU (2) AU2016270973B2 (hu)
CA (1) CA2987335A1 (hu)
CL (2) CL2017003073A1 (hu)
CO (1) CO2017012947A2 (hu)
CR (1) CR20170593A (hu)
DK (1) DK3303334T3 (hu)
DO (1) DOP2017000279A (hu)
EA (1) EA039006B1 (hu)
ES (1) ES2878030T3 (hu)
HK (1) HK1252378A1 (hu)
HR (1) HRP20211249T1 (hu)
HU (1) HUE055419T2 (hu)
IL (2) IL255946A (hu)
LT (1) LT3303334T (hu)
MX (2) MX2021006571A (hu)
MY (1) MY190548A (hu)
PE (1) PE20180521A1 (hu)
PH (1) PH12017502203A1 (hu)
PL (1) PL3303334T3 (hu)
PT (1) PT3303334T (hu)
RS (1) RS62290B1 (hu)
SG (1) SG10201912443XA (hu)
SI (1) SI3303334T1 (hu)
TN (1) TN2017000501A1 (hu)
TW (2) TWI732765B (hu)
UA (1) UA124090C2 (hu)
WO (1) WO2016196840A1 (hu)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10456403B2 (en) 2014-02-21 2019-10-29 Principia Biopharma Inc. Salts and solid form of a BTK inhibitor
US10485797B2 (en) 2014-12-18 2019-11-26 Principia Biopharma Inc. Treatment of pemphigus
US10533013B2 (en) 2012-09-10 2020-01-14 Principia Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as kinase inhibitors
WO2020028258A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
US11155544B2 (en) 2015-06-24 2021-10-26 Principia Biopharma Inc. Heterocycle comprising tyrosine kinase inhibitors
US11872229B2 (en) 2016-06-29 2024-01-16 Principia Biopharma Inc. Modified release formulations of 2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104159635B (zh) 2012-02-27 2018-09-25 奥雷制药有限公司 用于耳蜗内递送治疗剂以治疗耳部病症的固体药物植入物
WO2015080252A1 (ja) 2013-11-29 2015-06-04 株式会社ダイゾー 内容物収容容器、それを用いた内容物収容製品、吐出製品および吐出装置
CN106459049B (zh) * 2015-06-03 2020-11-27 普林斯匹亚生物制药公司 酪氨酸激酶抑制剂
CN105753863B (zh) * 2015-09-11 2018-07-31 东莞市真兴贝特医药技术有限公司 氧代二氢咪唑并吡啶类化合物及其应用
CN108349940B (zh) * 2015-10-14 2021-08-13 淄博百极常生制药有限公司 布鲁顿酪氨酸激酶抑制剂
CN109790164B (zh) * 2016-07-25 2022-01-25 内尔维亚诺医疗科学公司 作为胆碱激酶抑制剂的嘌呤和3-去氮杂嘌呤类似物
US10314844B2 (en) * 2017-02-24 2019-06-11 Gilead Sciences, Inc. Inhibitors of Bruton's tyrosine kinase
EP3789040A4 (en) * 2018-04-27 2022-03-09 ONO Pharmaceutical Co., Ltd. PREVENTIVE AND/OR THERAPEUTIC AGENT FOR AUTOIMMUNE DISEASES WITH A COMPOUND WITH BTK INHIBITING ACTIVITY AS AN INGREDIENT
WO2020021447A1 (en) 2018-07-25 2020-01-30 Novartis Ag Nlrp3 inflammasome inhibitors
KR20210057014A (ko) * 2018-09-13 2021-05-20 깃세이 야쿠힌 고교 가부시키가이샤 이미다조피리디논 화합물
EP3942045A1 (en) 2019-03-21 2022-01-26 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
AR119731A1 (es) 2019-05-17 2022-01-05 Novartis Ag Inhibidores del inflamasoma nlrp3
EP4054579A1 (en) 2019-11-08 2022-09-14 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
CN113045484B (zh) * 2019-12-27 2024-01-26 南京药石科技股份有限公司 一种2-氨基-2-(1-甲基-4-哌啶基)丙烷-1-醇的制备方法
CN113149891B (zh) * 2020-01-07 2024-02-02 南京药石科技股份有限公司 一种2-氨基-2-(1-甲基-4-哌啶基)乙醇的制备方法
WO2021150476A1 (en) 2020-01-20 2021-07-29 Genzyme Corporation Therapeutic tyrosine kinase inhibitors for relapsing multiple sclerosis (rms)
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
EP4125913A4 (en) * 2020-03-30 2023-08-23 IGIA Pharmaceuticals, Inc. PEDIATRIC TYROSINE KINASE INHIBITOR FORMULATION
AU2021325431B2 (en) 2020-08-14 2024-01-18 Novartis Ag Heteroaryl substituted spiropiperidinyl derivatives and pharmaceutical uses thereof
JP2023550591A (ja) 2020-11-02 2023-12-04 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング EGFR阻害薬としての置換1H-ピラゾロ[4,3-c]及び誘導体
EP4261212A1 (en) 2020-12-10 2023-10-18 Genzyme Corporation Crystal form of tolebrutinib, preparation method therefor and use thereof
WO2022140511A1 (en) * 2020-12-23 2022-06-30 Genzyme Corporation 4-amino-3-(4-phenoxyphenyl)-1,3- dihydro-2h-imidazo[4,5-c]pyridin-2-one derivatives and salts thereof
EP4313023A1 (en) 2021-04-02 2024-02-07 Biogen MA Inc. Combination treatment methods of multiple sclerosis
WO2022218430A1 (zh) * 2021-04-16 2022-10-20 南京明德新药研发有限公司 咪唑并吡啶类化合物及其应用
WO2022223027A1 (zh) 2021-04-23 2022-10-27 杭州领业医药科技有限公司 Tolebrutinib晶型、无定型及其制备方法和用途
EP4342468A1 (en) 2021-05-21 2024-03-27 Hangzhou Solipharma Co., Ltd. Tolebrutinib salt and crystal form thereof, preparation method therefor, pharmaceutical composition thereof, and use thereof
IL309263A (en) 2021-06-11 2024-02-01 Genzyme Corp Crystalline form of tolbrotinib, method of preparation and use thereof
WO2023280132A1 (zh) * 2021-07-06 2023-01-12 苏州晶云药物科技股份有限公司 氧代二氢咪唑并吡啶类化合物的晶型及其制备方法
US11793820B2 (en) * 2021-09-24 2023-10-24 Chadwick Donaldson Dry powder foamable formulations for delivery of medicaments through the mucosa
WO2023122072A1 (en) 2021-12-21 2023-06-29 Genzyme Corporation Crystalline forms of (r)-1-(1-acryloylpiperidin-3-yl)-4-amino-3-(4-phenoxyphenyl)-1h-imidazo[4,5-c]pyridin-2(3h)-one and salts thereof
WO2023172663A1 (en) * 2022-03-09 2023-09-14 Teva Pharmaceuticals International Gmbh Solid state forms of tolebrutinib and of tolebrutinib salts
US20230399313A1 (en) * 2022-06-10 2023-12-14 Advenchen Pharmaceuticals, LLC Biological activities of 5-(2-(4-(4-fluoro-2-methyl-1h-indol-5-yloxy)-6-methoxyquinolin-7-yloxy)ethyl)-5-azaspiro[2.4]-heptan-7-ol crystalline, phosphoric acid salt and its enantiomers
WO2023244587A1 (en) * 2022-06-14 2023-12-21 Genzyme Corporation Methods of making tolebrutinib
WO2023249980A1 (en) * 2022-06-22 2023-12-28 Genzyme Corporation Methods of making modified btk inhibitors
WO2024006406A1 (en) 2022-06-30 2024-01-04 Genzyme Corporation Therapeutic tyrosine kinase inhibitors for multiple sclerosis and myasthenia gravis
TW202406550A (zh) 2022-08-03 2024-02-16 瑞士商諾華公司 Nlrp3炎性小體抑制劑
US20240173313A1 (en) 2022-10-11 2024-05-30 Genzyme Corporation Therapeutic tyrosine kinase inhibitors for multiple sclerosis
WO2024099242A1 (zh) * 2022-11-07 2024-05-16 天津征程医药科技有限公司 氘代的氨基吡啶衍生物以及包含该化合物的药物组合物

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9688676B2 (en) * 2015-06-03 2017-06-27 Principia Biopharma Inc. Tyrosine kinase inhibitors

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2809918A (en) 1955-10-17 1957-10-15 Victor M Hermelin Sustained release pharmaceutical preparations
DE2010416B2 (de) 1970-03-05 1979-03-29 Hoechst Ag, 6000 Frankfurt Oral anwendbare Arzneiform mit Retardwirkung
US3760984A (en) 1971-09-29 1973-09-25 Alza Corp Osmotically powered agent dispensing device with filling means
US4107288A (en) 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
US3952741A (en) 1975-01-09 1976-04-27 Bend Research Inc. Controlled release delivery system by an osmotic bursting mechanism
US4728512A (en) 1985-05-06 1988-03-01 American Home Products Corporation Formulations providing three distinct releases
US4794001A (en) 1986-03-04 1988-12-27 American Home Products Corporation Formulations providing three distinct releases
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
CA2465328C (en) 2001-11-01 2011-06-14 Michael Francis Gross Piperidines
US20060045822A1 (en) 2004-09-01 2006-03-02 Board Of Regents, The University Of Texas System Plasma polymerization for encapsulating particles
CN101056877B (zh) * 2004-09-14 2010-06-09 诺华疫苗和诊断公司 咪唑并喹啉化合物
AU2006213746A1 (en) * 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted imidazo(4,5-c) ring compounds and methods
EP2029597A4 (en) 2006-05-31 2011-11-23 Univ California purine analogs
TW200804383A (en) 2006-06-05 2008-01-16 Astrazeneca Ab New compounds
EP2526933B1 (en) 2006-09-22 2015-02-25 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
NZ620174A (en) 2009-09-16 2016-08-26 Celgene Avilomics Res Inc Protein kinase conjugates and inhibitors
US7741330B1 (en) * 2009-10-12 2010-06-22 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of Bruton's tyrosine kinase
AU2011260961B9 (en) * 2010-05-31 2015-02-26 Ono Pharmaceutical Co., Ltd. Purinone derivative
PL2710007T3 (pl) * 2011-05-17 2020-06-01 The Regents Of The University Of California Inhibitory kinazy
EP2710006A1 (en) * 2011-05-17 2014-03-26 Principia Biopharma Inc. Azaindole derivatives as tyrosine kinase inhibitors
EP2710005B1 (en) * 2011-05-17 2016-10-05 Principia Biopharma Inc. Tyrosine kinase inhibitors
PT2786996T (pt) * 2011-11-29 2016-11-07 Ono Pharmaceutical Co Cloridrato de derivado de purinona
US8501724B1 (en) 2012-01-31 2013-08-06 Pharmacyclics, Inc. Purinone compounds as kinase inhibitors
KR102150432B1 (ko) 2012-06-18 2020-09-01 프린시피아 바이오파마, 인코퍼레이티드 암 및 자가면역 질환 치료에 유용한 가역적인 공유 피롤로- 또는 피라졸로피리미딘
ES2916220T3 (es) 2012-07-11 2022-06-29 Blueprint Medicines Corp Inhibidores del receptor de factor de crecimiento de fibroblasto
PT2892900T (pt) * 2012-09-10 2017-11-06 Principia Biopharma Inc Compostos de pirazolopirimidina como inibidores de cinase
US20140142099A1 (en) * 2012-11-20 2014-05-22 Principia Biopharma Inc. Purinone Derivatives as Tyrosine Kinase Inhibitors
US8957080B2 (en) * 2013-04-09 2015-02-17 Principia Biopharma Inc. Tyrosine kinase inhibitors
CN105753863B (zh) 2015-09-11 2018-07-31 东莞市真兴贝特医药技术有限公司 氧代二氢咪唑并吡啶类化合物及其应用
CN108349940B (zh) 2015-10-14 2021-08-13 淄博百极常生制药有限公司 布鲁顿酪氨酸激酶抑制剂

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9688676B2 (en) * 2015-06-03 2017-06-27 Principia Biopharma Inc. Tyrosine kinase inhibitors

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10533013B2 (en) 2012-09-10 2020-01-14 Principia Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as kinase inhibitors
US11040980B2 (en) 2012-09-10 2021-06-22 Principia Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as kinase inhibitors
US10456403B2 (en) 2014-02-21 2019-10-29 Principia Biopharma Inc. Salts and solid form of a BTK inhibitor
US10828307B2 (en) 2014-02-21 2020-11-10 Principia Biopharma Inc. Salts and solid form of a BTK inhibitor
US11369613B2 (en) 2014-02-21 2022-06-28 Principia Biopharma Inc. Salts and solid form of a BTK inhibitor
US10485797B2 (en) 2014-12-18 2019-11-26 Principia Biopharma Inc. Treatment of pemphigus
US10946008B2 (en) 2014-12-18 2021-03-16 Principia Biopharma Inc. Treatment of pemphigus
US11155544B2 (en) 2015-06-24 2021-10-26 Principia Biopharma Inc. Heterocycle comprising tyrosine kinase inhibitors
US11872229B2 (en) 2016-06-29 2024-01-16 Principia Biopharma Inc. Modified release formulations of 2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile
WO2020028258A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide

Also Published As

Publication number Publication date
LT3303334T (lt) 2021-07-12
IL279258A (en) 2021-01-31
AU2016270973A1 (en) 2018-01-18
PE20180521A1 (es) 2018-03-14
DOP2017000279A (es) 2018-04-15
TW202210478A (zh) 2022-03-16
MY190548A (en) 2022-04-27
CN106459049A (zh) 2017-02-22
SI3303334T1 (sl) 2021-09-30
CN106459049B (zh) 2020-11-27
SG10201912443XA (en) 2020-02-27
CN113149983A (zh) 2021-07-23
PL3303334T3 (pl) 2021-11-08
EA201792529A1 (ru) 2018-05-31
HK1252378A1 (zh) 2019-05-24
TN2017000501A1 (en) 2019-04-12
EP4112618A1 (en) 2023-01-04
EP3303334B1 (en) 2021-06-02
CO2017012947A2 (es) 2018-03-28
ES2878030T3 (es) 2021-11-18
CN113149983B (zh) 2024-03-29
WO2016196840A1 (en) 2016-12-08
DK3303334T3 (da) 2021-06-28
EA039006B1 (ru) 2021-11-19
BR112017025850A2 (pt) 2018-08-14
MX2021006571A (es) 2022-08-25
TW201713654A (zh) 2017-04-16
RS62290B1 (sr) 2021-09-30
KR102552653B1 (ko) 2023-07-07
US20170129890A1 (en) 2017-05-11
JP6985433B2 (ja) 2021-12-22
EP3912979A1 (en) 2021-11-24
UA124090C2 (uk) 2021-07-21
IL279258B (en) 2022-04-01
JP2018516935A (ja) 2018-06-28
TWI810582B (zh) 2023-08-01
US20230151012A1 (en) 2023-05-18
PH12017502203A1 (en) 2018-06-11
AU2016270973B2 (en) 2020-05-14
MX2017015470A (es) 2018-08-15
AU2020203447B2 (en) 2021-04-01
PT3303334T (pt) 2021-07-02
JP6646072B2 (ja) 2020-02-14
CL2017003073A1 (es) 2018-06-08
KR20220146657A (ko) 2022-11-01
TWI732765B (zh) 2021-07-11
AU2020203447A1 (en) 2020-06-11
EP3303334A1 (en) 2018-04-11
CL2019002150A1 (es) 2019-11-29
CA2987335A1 (en) 2016-12-08
US9688676B2 (en) 2017-06-27
JP2020073561A (ja) 2020-05-14
CR20170593A (es) 2018-05-14
IL255946A (en) 2018-01-31
HRP20211249T1 (hr) 2021-11-12
KR20180021730A (ko) 2018-03-05
HUE055419T2 (hu) 2021-11-29
US20210171526A1 (en) 2021-06-10

Similar Documents

Publication Publication Date Title
AU2020203447B2 (en) Tyrosine kinase inhibitors
US11155544B2 (en) Heterocycle comprising tyrosine kinase inhibitors
US11369613B2 (en) Salts and solid form of a BTK inhibitor
EP2861599B1 (en) Reversible covalent pyrrolo- or pyrazolopyrimidines useful for the treatment of cancer and autoimmune diseases
KR102666352B1 (ko) 티로신 키나제 억제제
BR112017025850B1 (pt) Compostos inibidores de tirosina quinase, seus usos e composição farmacêutica
KR20240070721A (ko) 티로신 키나제 억제제

Legal Events

Date Code Title Description
AS Assignment

Owner name: PRINCIPIA BIOPHARMA INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GOLDSTEIN, DAVID;OWENS, TIMOTHY;REEL/FRAME:044267/0552

Effective date: 20160602

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

AS Assignment

Owner name: PRINCIPIA BIOPHARMA INC., CALIFORNIA

Free format text: CHANGE OF ADDRESS OF ASSIGNEE;ASSIGNOR:PRINCIPIA BIOPHARMA INC.;REEL/FRAME:049473/0795

Effective date: 20190614

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: ADVISORY ACTION 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: 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