WO2016109215A1 - Btk inhibitors - Google Patents

Btk inhibitors Download PDF

Info

Publication number
WO2016109215A1
WO2016109215A1 PCT/US2015/066218 US2015066218W WO2016109215A1 WO 2016109215 A1 WO2016109215 A1 WO 2016109215A1 US 2015066218 W US2015066218 W US 2015066218W WO 2016109215 A1 WO2016109215 A1 WO 2016109215A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrazin
amino
benzamide
imidazo
trifluoromethyl
Prior art date
Application number
PCT/US2015/066218
Other languages
French (fr)
Inventor
Jian Liu
Joseph A. Kozlowski
Abdul-Basit Alhassan
Rajan Anand
Sobhana Babu Boga
Deodialsingh Guiadeen
Wensheng Yu
Younong Yu
Hao Wu
Chundao YANG
Original Assignee
Merck Sharp & Dohme Corp.
Liu, Shilan
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 Merck Sharp & Dohme Corp., Liu, Shilan filed Critical Merck Sharp & Dohme Corp.
Priority to EP15875967.0A priority Critical patent/EP3240542A4/en
Priority to US15/538,976 priority patent/US10130630B2/en
Publication of WO2016109215A1 publication Critical patent/WO2016109215A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • B lymphocyte activation is key in the generation of adaptive immune responses. Derailed B lymphocyte activation is a hallmark of many autoimmune diseases and modulation of this immune response is therefore of therapeutic interest. Recently the success of B cell therapies in autoimmune diseases has been established. Treatment of rheumatoid arthritis (RA) patients with Rituximab (anti-CD20 therapy) is an accepted clinical therapy by now. More recent clinical trial studies show that treatment with Rituximab also ameliorates disease symptoms in relapsing remitting multiple sclerosis (RRMS) and systemic lupus erythematosus (SLE) patients. This success supports the potential for future therapies in autoimmune diseases targeting B cell immunity.
  • RRMS multiple sclerosis
  • SLE systemic lupus erythematosus
  • Btk Bruton tyrosine kinase
  • Btk in the regulation of the production of auto-antibodies in autoimmune diseases.
  • regulation of Btk may affect BCR-induced production of pro-inflammatory cytokines and chemokines by B cells, indicating a broad potential for Btk in the treatment of autoimmune diseases.
  • B cell malignancies Other diseases with an important role for dysfunctional B cells are B cell malignancies. Indeed anti-CD20 therapy is used effectively in the clinic for the treatment of follicular lymphoma, diffuse large B-cell lymphoma and chronic lymphocytic leukemia [Lim et al, Haematologica, 95 (2010) ppl35-143].
  • the reported role for Btk in the regulation of proliferation and apoptosis of B cells indicates there is potential for Btk inhibitors in the treatment of B cell lymphomas as well. Inhibition of Btk seems to be relevant in particular for B cell lymphomas due to chronic active BCR signaling [Davis et al, Nature, 463 (2010) pp88-94].
  • Btk inhibitor compounds Some classes of Btk inhibitor compounds have been described as kinase inhibitors, e.g. Imidazo[l,5-f][l,2,4]triazine compounds have been described in WO2005097800 and WO2007064993. Imidazo[l,5-a]pyrazine compounds have been described in
  • WO2005037836 and WO2001019828 as IGF-1R enzyme inhibitors.
  • Lyn-deficient mice exhibit autoimmunity mimicking the phenotype of human lupus nephritis.
  • Fyn-deficient mice also show pronounced neurological defects.
  • Lyn knockout mice also show an allergic-like phenotype, indicating Lyn as a broad negative regulator of the IgE-mediated allergic response by controlling mast cell responsiveness and allergy-associated traits [Odom et al, J. Exp. Med., 199 (2004) ppl491- 1502].
  • aged Lyn knock-out mice develop severe splenomegaly (myeloid expansion) and disseminated monocyte/macrophage tumors [Harder et al, Immunity, 15 (2001) pp603-615].
  • mice Female Src knockout mice are infertile due to reduced follicle development and ovulation [Roby et al, Endocrine, 26 (2005) ppl69-176].
  • the double knockouts Src ⁇ Tyn "7" and Src ⁇ Yes "7” show a severe phenotype with effects on movement and breathing.
  • the triple knockouts Src ⁇ Tyn ⁇ es "7" die at day 9.5
  • an effective amount refers to an amount of the compound of Formula I and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventative effect when administered to a subject suffering from a BTK-mediated disease or disorder.
  • an effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount.
  • cyclic moiety refers to cyclopropyl, cyclobutyl and cyclopentyl and also refers to cyclic structures formed as a result of variables coming together to form a carbon bridge (for example, variables X and T forming a carbon bridge as highlighted in example compound 16).
  • halogen refers to fluorine, chlorine, bromine or iodine. Fluorine, chlorine or bromine being preferred halogens; fluorine being more preferred.
  • purified refers to the physical state of a compound after the compound has been isolated through a synthetic process (e.g., from a reaction mixture), from a natural source, or a combination thereof.
  • purified also refers to the physical state of a compound after the compound has been obtained from a purification process or processes described herein or well-known to the skilled artisan (e.g., chromatography, recrystallization, and the like), in sufficient purity to be characterizable by standard analytical techniques described herein or well-known to the skilled artisan.
  • a “subject” is a human or non-human mammal.
  • a subject is a human.
  • the subject is a chimpanzee.
  • the present invention provides Btk inhibitor compounds according to Formula I or pharmaceutically acceptable salts thereof
  • Ring A is selected from the group consisting of
  • Z is C or N
  • X and Y are independently H or X and Y can come together to form cyclopropyl, cyclobutyl or cyclopentyl,
  • M and L are independently H or M and L can come together to form cyclopropyl
  • Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
  • R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
  • the present invention provides Btk inhibitor compounds according to Formula la or pharmaceutically acceptable salts thereof
  • Ring A is selected from the group consisting of
  • n 0, 1 or 2;
  • Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
  • R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
  • the present invention provides Btk inhibitor compounds according to Formula lb or pharmaceutically acceptable salts thereof
  • n 0, 1 or 2;
  • Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
  • R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
  • Non-limiting examples of the compounds of the present invention include:
  • halogen is F or CI.
  • R 3 is H or F.
  • the compounds of this invention include the salts, solvates, hydrates or prodrugs of the compounds.
  • the use of the terms “salt”, “solvate”, “hydrate”, “prodrug” and the like, is intended to equally apply to the salt, solvate, hydrate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, or racemates of the inventive compounds.
  • the Btk inhibitor compounds of the present invention which can be in the form of a free base, may be isolated from the reaction mixture in the form of a pharmaceutically acceptable salt.
  • the compounds of Formula I can form salts which are also within the scope of this invention.
  • Reference to a compound of Formula I herein is understood to include reference to pharmaceutically acceptable salts thereof, unless otherwise indicated.
  • salt(s) denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases.
  • a compound of Formula I contains both a basic moiety, such as, but not limited to a pyridine or imidazole, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term “salt(s)" as used herein.
  • Such acidic and basic salts used within the scope of the invention are pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts.
  • Salts of the compounds of Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.
  • alkali metal salts such as sodium, lithium, and potassium salts
  • alkaline earth metal salts such as calcium and magnesium salts
  • salts with organic bases for example, organic amines
  • organic amines such as dicyclohexylamines, t-butyl amines
  • salts with amino acids such as arginine, lysine and the like.
  • Basic nitrogen- containing groups may be quarternized with agents such as lower alkyl halides (e.g., methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g., decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenethyl bromides), and others.
  • lower alkyl halides e.g., methyl, ethyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates e.g., dimethyl, diethyl, and dibutyl sulfates
  • long chain halides e.g., decyl, lauryl, and
  • the Btk inhibitor compounds of the present invention may exist as amorphous forms or crystalline forms.
  • the compounds of Formula I may have the ability to crystallize in more than one form, a characteristic known as polymorphism, and it is understood that such polymorphic forms (“polymorphs”) are within the scope of Formula I.
  • Polymorphism generally can occur as a response to changes in temperature or pressure or both and can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility and melting point.
  • the compounds having Formula I or the pharmaceutically acceptable salts may form hydrates or solvates. It is known to those of skill in the art that charged compounds form hydrated species when lyophilized with water, or form solvated species when concentrated in a solution with an appropriate organic solvent.
  • the compounds of this invention include the hydrates or solvates of the compounds listed.
  • NSAIDs include, but are not limited to, ibuprofen, flurbiprofen, naproxen and naproxen sodium, diclofenac, combinations of diclofenac sodium and misoprostol, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, sodium nabumetone, sulfasalazine, tolmetin sodium, and hydroxychloroquine.
  • the anti-inflammatory agent is a salicylate.
  • Salicylates include by are not limited to acetylsalicylic acid or aspirin, sodium salicylate, and choline and magnesium salicylates.
  • B-cells and B-cell precursors expressing BTK have been implicated in the pathology of B-cell malignancies, including, but not limited to, B-cell lymphoma, lymphoma (including Hodgkin's and non-Hodgkin's lymphoma), hairy cell lymphoma, multiple myeloma, chronic and acute myelogenous leukemia and chronic and acute lymphocytic leukemia.
  • camptothecin or topotecan topoisomerase II inhibitors (e.g. daunomycin and etoposide), alkylating agents (e.g. cyclophosphamide, melphalan and BCNU), tubulin directed agents (e.g. taxol and vinblastine), and biological agents (e.g. antibodies such as anti CD20 antibody, IDEC 8, immunotoxins, and cytokines).
  • topoisomerase II inhibitors e.g. daunomycin and etoposide
  • alkylating agents e.g. cyclophosphamide, melphalan and BCNU
  • tubulin directed agents e.g. taxol and vinblastine
  • biological agents e.g. antibodies such as anti CD20 antibody, IDEC 8, immunotoxins, and cytokines.
  • Btk activity has also been associated with some leukemias expressing the bcr-abl fusion gene resulting from translocation of parts of chromosome 9 and 22. This abnormality is commonly observed in chronic myelogenous leukemia. Btk is constitutively phosphorylated by the bcr-abl kinase which initiates downstream survival signals which circumvents apoptosis in bcr-abl cells. (N. Feldhahn et al. J. Exp. Med. 2005 201(11): 1837-1852).
  • a compound of Formula I may be combined with one or more other active agents such as: (1) TNF-a inhibitors such as infliximab (Remicade®), etanercept (Enbrel®), adalimumab (Humira®), certolizumab pegol (Cimzia®), and golimumab (Simponi®); (2) non-selective COX-I/COX-2 inhibitors (such as piroxicam, diclofenac, propionic acids such as naproxen, flubiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, etodolac, azapropazone, pyrazolones such as phen
  • metaproterenol isoproterenol, isoprenaline, albuterol, formoterol (particularly the fumarate salt), salmeterol (particularly the xinafoate salt), terbutaline, orciprenaline, bitolterol mesylate, fenoterol, and pirbuterol, or methylxanthanines including theophylline and aminophylline, sodium cromoglycate; (12) insulin-like growth factor type I (IGF-1) mimetic; (13)
  • the present invention also provides for "triple combination" therapy, comprising a compound of Formula I or a pharmaceutically acceptable salt thereof together with beta2- adrenoreceptor agonist and an anti-inflammatory corticosteroid.
  • this combination is for treatment and/or prophylaxis of asthma, COPD or allergic rhinitis.
  • the beta2-adrenoreceptor agonist and/or the anti-inflammatory corticosteroid can be as described above and/or as described in WO 03/030939 Al .
  • Representative examples of such a "triple” combination are a compound of Formula I or a pharmaceutically acceptable salt thereof in combination with the components of Advair® (salmeterol xinafoate and fluticasone propionate), Symbicort®
  • a compound of Formula I may be combined with one or more of an anticancer agents.
  • an anticancer agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6 th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.
  • sargramostim pentosan polysulfate, cyclooxygenase inhibitors, steroidal anti-inflammatories, carboxyamidotriazole, combretastatin A-4, squalamine, 6-0-chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin- 1, angiotensin II antagonists, heparin, carboxypeptidase U inhibitors, and antibodies to VEGF, endostatin, ukrain, ranpirnase, IM862, acetyl dinanaline, 5- amino-l-[[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]methyl]-lH-l,2,3-triazole-4- carboxamide,CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate
  • rituximab inhibitors of cytokine receptors, inhibitors of MET, inhibitors of PI3K family kinase (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in (WO 03/086404, WO 03/086403, WO 03/086394, WO 03/086279, WO 02/083675, WO 02/083139, WO 02/083140 and WO 02/083138), inhibitors of Raf kinase (for example BAY-43-9006 ), inhibitors of MEK (for example CI-1040 and PD-098059) and inhibitors of mTOR (for example Wyeth CCI-779 and Ariad AP23573); (11) a bisphosphonate such as etidronate, pamidronate, alendronate, risedronate, zoledronate, ibandronate, incadronate or cimadronate
  • MK4827 (16) ERK inhibitors; (17) mTOR inhibitors such as sirolimus, ridaforolimus, temsirolimus, everolimus; (18) cytotoxic/cytostatic agents.
  • proteasome inhibitors include but are not limited to lactacystin and bortezomib.
  • topoisomerase inhibitors are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3',4'-0-exo-benzylidene-chartreusin, lurtotecan, 7-[2- (N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2'-dimethylamino-2'-deoxy-etoposide, GL331, N- [2-(dimethylamino)ethyl]-9-hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-l-carboxamide, asulacrine, 2,3-(methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c]-phenanth
  • inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLPl, inhibitors of CENP-E, inhibitors of MCAK, inhibitors of Kifl4, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
  • histone deacetylase inhibitors include, but are not limited to, vorinostat, trichostatin A, oxamflatin, PXD101, MG98, valproic acid and scriptaid.
  • “Inhibitors of kinases involved in mitotic progression” include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK; in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-Rl .
  • PLK Polo-like kinases
  • An example of an "aurora kinase inhibitor” is VX-680.
  • Antiproliferative agents includes antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methylidenecytidine, 2'- fluoromethylene-2'-deoxycytidine, N6-[4-deoxy-4- N2-[2,4-tetradecadienoyl]glycylamino]-L- glycero-
  • Non-limiting examples of suitable agents used in cancer therapy include, but are not limited to, abarelix; aldesleukin; alemtuzumab; alitretinoin; allopurinol; altretamine; amifostine; anastrozole; arsenic trioxide; asparaginase; azacitidine; bendamustine; bevacuzimab; bexarotene; bleomycin; bortezomib; busulfan; calusterone; capecitabine; carboplatin; carmustine; cetuximab; chlorambucil; cisplatin; cladribine; clofarabine; cyclophosphamide; cytarabine; dacarbazine; dactinomycin, actinomycin
  • hydroxyurea ibritumomab tiuxetan; idarubicin; ifosfamide; imatinib mesylate; interferon alfa 2a; interferon alfa-2b; irinotecan; ixabepilone; lapatinib; lenalidomide; letrozole; leucovorin;
  • leuprolide acetate levamisole; lomustine; meclorethamine, nitrogen mustard; megestrol acetate; melphalan, L-PAM; mercaptopurine; mesna; methotrexate; methoxsalen; mitomycin C;
  • mitotane mitoxantrone; nandrolone phenpropionate; nelarabine; nilotinib; Nofetumomab;
  • sargramostim sargramostim
  • sargramostim satraplatin
  • sorafenib streptozocin
  • sunitinib maleate sunitinib maleate
  • tamoxifen temozolomide
  • temsirolimus teniposide
  • testolactone thioguanine
  • thiotepa topotecan;
  • toremifene tositumomab; trastuzumab; tretinoin; uracil mustard; valrubicin; vinblastine;
  • the other therapeutic ingredient(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent, carrier or excipient represent a further aspect of the invention.
  • These combinations are of particular interest in respiratory diseases and are conveniently adapted for inhaled or intranasal delivery.
  • a process for the preparation of a pharmaceutical composition including admixing a compound of the Formula I, or salts, solvates and physiological functional derivatives thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • compositions of the present invention may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • a unit may contain, for example, 5 ⁇ g to 1 g, preferably 1 mg to 700 mg, more preferably 5 mg to 100 mg of a compound of the Formula I, depending on the condition being treated, the route of administration and the age, weight and condition of the patient.
  • Such unit doses may therefore be administered more than once a day.
  • Preferred unit dosage compositions are those containing a daily dose or sub-dose (for administration more than once a day), as herein above recited, or an appropriate fraction thereof, of an active ingredient.
  • such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
  • compositions of the present invention may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, topical, inhaled, nasal, ocular, sublingual, subcutaneous, local or parenteral (including intravenous and intramuscular) route, and the like, all in unit dosage forms for administration.
  • Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like.
  • the present invention provides a pharmaceutical composition adapted for administration by the oral route, for treating, for example, rheumatoid arthritis.
  • the present invention provides a pharmaceutical composition adapted for administration by the nasal route, for treating, for example, allergic rhinitis.
  • the present invention provides a pharmaceutical composition adapted for administration by the inhaled route, for treating, for example, asthma, Chronic Obstructive Pulmonary disease (COPD) or Acute Respiratory Distress Syndrome (ARDS).
  • COPD Chronic Obstructive Pulmonary disease
  • ARDS Acute Respiratory Distress Syndrome
  • the present invention provides a pharmaceutical composition adapted for administration by the ocular route, for treating, diseases of the eye, for example, conjunctivitis.
  • the active agent may be compressed into solid dosage units, such as pills, tablets, or be processed into capsules or suppositories.
  • the active agent can be applied as a fluid composition, e.g. as an injection preparation, in the form of a solution, suspension, emulsion, or as a spray, e.g. a nasal spray.
  • compositions of the present invention which are adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
  • suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone
  • a solution retardant such as paraffin
  • a resorption accelerator such as a quaternary salt
  • an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit compositions for oral administration can be microencapsulated.
  • the formulation can also be prepared to prolong or sustain the release, for example, by coating or embedding particulate material in polymers, wax or the like.
  • the compounds of Formula I, and salts, solvates and physiological functional derivatives thereof, can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or
  • polyvinylpyrrolidone polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • Dosage forms for inhaled administration may conveniently be formulated as aerosols or dry powders.
  • the compound or salt of Formula I is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronisation.
  • the preferable particle size of the size-reduced (e.g. micronised) compound or salt or solvate is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
  • Aerosol formulations can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or non-aqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
  • a metering valve metered dose inhaler
  • the dosage form comprises an aerosol dispenser
  • it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC).
  • suitable HFC propellants include 1 ,1 , 1,2,3,3,3- heptafluoropropane and 1 ,1 , 1,2-tetrafiuoroethane.
  • the aerosol dosage forms can also take the form of a pump-atomiser.
  • the pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol.
  • Other excipient modifiers may also be incorporated to improve, for example, the stability and/or taste and/or fine particle mass characteristics (amount and/or profile) of the formulation.
  • the pharmaceutical composition is a dry powder inhalable composition.
  • a dry powder inhalable composition can comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, the compound of Formula I or salt or solvate thereof (preferably in particle- size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L- leucine or another amino acid, and/or metals salts of stearic acid such as magnesium or calcium stearate.
  • the dry powder inhalable composition comprises a dry powder blend of lactose and the compound of Formula I or salt thereof.
  • the lactose is preferably lactose hydrate e.g. lactose monohydrate and/or is preferably inhalation-grade and/or fine-grade lactose.
  • the particle size of the lactose is defined by 90% or more of the lactose particles being less than 100-200 microns in diameter, and/or 50% or more of the lactose particles being less than 40-70 microns in diameter. It is preferable that about 3 to about 30% (e.g. about 10%) (by weight or by volume) of the particles are less than 50 microns or less than 20 microns in diameter.
  • a suitable inhalation-grade lactose is E9334 lactose (10% fines) (Borculo Domo Ingredients, Hanzeplein 25, 8017 J D Zwolle, Netherlands).
  • a pharmaceutical composition for inhaled administration can be incorporated into a plurality of sealed dose containers (e.g. containing the dry powder composition) mounted longitudinally in a strip or ribbon inside a suitable inhalation device.
  • the container is rupturable or peel-openable on demand and the dose of e.g. the dry powder composition can be administered by inhalation via the device such as the DISKUS® device(GlaxoSmithKline).
  • Dosage forms for ocular administration may be formulated as solutions or suspensions with excipients suitable for ophthalmic use.
  • Dosage forms for nasal administration may conveniently be formulated as aerosols, solutions, drops, gels or dry powders.
  • compositions adapted for administration by inhalation include fine particle dusts or mists, which may be generated by means of various types of metered, dose pressurized aerosols, nebulizers or insufflators.
  • the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof may be formulated as a fluid formulation for delivery from a fluid dispenser.
  • a fluid dispenser may have, for example, a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
  • Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid formulation, the doses being dispensable upon sequential pump actuations.
  • the dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid formulation into the nasal cavity.
  • a fluid dispenser of the aforementioned type is described and illustrated in WO-A-2005/044354, the entire content of which is hereby incorporated herein by reference.
  • the dispenser has a housing which houses a fluid discharge device having a compression pump mounted on a container for containing a fluid formulation.
  • the housing has at least one finger-operable side lever which is movable inwardly with respect to the housing to cam the container upwardly in the housing to cause the pump to compress and pump a metered dose of the formulation out of a pump stem through a nasal nozzle of the housing.
  • a particularly preferred fluid dispenser is of the general type illustrated in FIGS. 30-40 of WO-A-2005/044354.
  • the invention further includes a pharmaceutical composition of a compound of
  • Step 4 3-ethoxy-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin- 2-yl)benzamide
  • N-(4-methylpyridin-2-yl)-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzamide was prepared in a similar method as intermediate 1
  • Methyl l,3-dioxooctahydroindolizine-6-carboxylate (6.82 g, 32.3 mmol) was dissolved in DMF (23.92 ml) and it was added into the reaction solution over a period of 24 h via a syringe pump at 70 °C. The reaction mixture was concentrated to almost dry. TheDMF was removed as much as possible. Then 200 mL water and 200 mL DCM was added. The aqueous layer was separated and extracted with DCM (100 ml) once.
  • Step 1 Preparation of (fra» , )-6'-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazo[1.5- alpyrazin-3-yl)-r-hvdroxytetrahvdro-rH-spiro[cvclopropane-1.2'-indolizinl-3'(5'H)-one
  • Step 7 (3R,6S)- ⁇ -benzyl 3-methyl 6-(((ter/-butyldiphenylsilyl)oxy)methyl)piperidine-1.3- dicarboxylate
  • Step 8 (3i?.6 ⁇ )-l-((benzyloxy)carbonyl)-6-(((ter/-butyldiphenylsilyl)oxy)methyl)piperidine-3- carboxylic acid
  • Step 10 (2ff.5i?)-benzyl 2-(((ter -butyldiphenylsilyl)oxy)methyl)-5-(8-chloroimidazo[1.5- al pyrazin-3-yl)piperidine- 1 -carboxylate
  • Step 12 (2S,5R)-benzy ⁇ 5-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazori.5-alpyrazin-3- yl)-2-(((ter ⁇ -butyldiphenylsilyl)oxy)methyl)piperidine-l -carboxylate
  • Step 3 (2R.5R)-tert-butyl 2-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)-5-(hvdroxymethyl)morpholine-4-carboxylate
  • Step 5 (2R.5R)-tert-butyl 2-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl -5- 2- methoxycarbonyl pyrrolidin-l-yl methyl mo ⁇ holine-4-carboxylate
  • Step 6 (3R.6aR.l laR)-3-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)octahvdropyrrolor .2':4.51pyrazinor2.1-ci ri.41oxazin-6(lH)-one
  • Step 7 (3R.6aR.l laR)-3-(8-amino-l-bromoimidazori.5-alpyrazin-3- yl)octahvdropyrrolor .2':4.51pyrazinor2.1-ciri.41oxazin-6(lH)-one
  • Step 2 (3R.6aS .1 laR)-3-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)octahvdropyrrolo[ .2':4.51pyrazino[2.1-cl [1.41oxazin-6(lH)-one
  • Step 2 l-(5-bromopyridin-2-yl)cvclopropanecarboxylic acid
  • Step 3 l-(5-bromopyridin-2-yl)-N-methoxy-N-methylcvclopropanecarboxamide
  • Step 7 methyl 2-(l-(5-bromopyridin-2-yl)cvclopropyl)acetate
  • Step 8 methyl 6-(l-(2-methoxy-2-oxoethyl)cvclopropyl)nicotinate
  • Step 9 methyl 6-(l-(2-methoxy-2-oxoethyl)cvclopropyl)piperidine-3-carboxylate
  • Step 13 6'-(8-chloroimidazo[1.5-alpyrazin-3-yl)tetrahvdro-2'H-spiro[cvclopropane-l. - indolizinl-3'(5'H)-one (trans)
  • Stepl4 6'-(l-bromo-8-chloroimidazori.5-alpyrazin-3-yl)tetrahvdro-2'H-spirorcvclopropane- 1.1 '-indolizinl-3'(5'H)-one (trans)
  • Step 3 methyl 6-(2-(ethoxycarbonyl)cvclopropyl)piperidine-3-carboxylate
  • Step 8 5-(l-bromo-8-chloroirrudazo[1.5-alpyrazin-3-yl)hexahvdro-lH-cvclopropa[alindolizin- 2(laH)-one
  • Step 2 (7'R.9a'S)-7'-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)hexahvdrospiro[cvclopropane-1.3'-pyrido[1.2-alpyrazinl-4'(2'H)-one
  • Step 2 (3'R.9a'R)-3'-(l-bromo-8-((2 ⁇ -dimethoxybenzyl)amino)imidazori.5-alpyrazin-3- yl)tetrahvdro- H-spirorcvclopropane-1.7'-pyrazinor2.1-ciri.41oxazinl-6'(8'H)-one
  • Step 3 methyl 6-(l-(((benzyloxy)carbonyl)amino)cvclopropyl)piperidine-3-carboxylate
  • Step 1 4-(8-ariiino-3-(2-oxooctahvdro-lH-cvcloproparalindolizin-5-yl)imidazori.5-alpyrazin-l-yl)-3- ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide
  • Step 1 4-(8-((2 ⁇ -dimethoxybenzyl)amino)-3-(2-oxooctahydro-lH-cvclopropa[alindolizin-5- yl)imidazo[1.5-alpyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (isomer 1A)
  • Step 2 4-(8-andno-3-(2-oxooctahvdro-lH-cvclopropa[alindolizin-5-yl)imidazo[1.5-alpyrazin-l- yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (isomer 1A)
  • the Btk inhibitor compounds of the invention having Formula I inhibit the Btk kinase activity. All compounds of the invention have an IC50 of 10 ⁇ or lower. In another aspect the invention relates to compounds of Formula I which have an IC50 of less than 100 nM. In yet another aspect the invention relates to compounds of Formula I which have an IC50 of less than 10 nM.
  • IC50 means the concentration of the test compound that is required for 50% inhibition of its maximum effect in vitro.
  • each reaction was initiated by the addition of 2.5 lx kinase buffer containing 8 ⁇ biotinylated "A5" peptide (Biotin-EQEDEPEGDYFEWLE-NH2) (SEQ.ID.NO.: 1), and 100 ⁇ ATP.
  • the final reaction in each well of 10 ⁇ consists of 50 pM ABTK, 2 ⁇ biotin-A5- peptide, and 25 ⁇ ATP. Phosphorylation reactions were allowed to proceed for 120 minutes.
  • reaction plates were read on a PerkinElmer EnVision plate reader using standard TR-FRET protocol.
  • Table 2 provides specific IC50 values for all the examples.
  • the IC50 values set forth below were determined according to Assay method described above.
  • Adenosine uptake activity was determined by monitoring the accumulation of tritiated adenosine into HeLa cells (ATCC catalog # CCL-2) using a PMT-based radiometric detection instrument.
  • the potency (IC 50 ) of each compound was determined from a ten point (1 :3 serial dilution; final compound concentration range in assay from 10 ⁇ to 0.032 nM) titration curve using the following outlined procedure.

Abstract

The present invention provides Bruton's Tyrosine Kinase (Btk) inhibitor compounds according to Formula (I), or pharmaceutically acceptable salts or stereoisomers thereof, or to pharmaceutical compositions comprising these compounds and to their use in therapy. In particular, the present invention relates to the use of Btk inhibitor compounds of Formula (I) in the treatment of Btk mediated disorders.

Description

TITLE OF THE INVENTION
BTK INHIBITORS
FIELD OF THE INVENTION
The present invention relates to Btk inhibitor compounds, to pharmaceutical compositions comprising these compounds and to their use in therapy. In particular, the present invention relates to the use of Btk inhibitor compounds in the treatment of Bruton's Tyrosine Kinase (Btk) mediated disorders. BACKGROUND OF THE INVENTION
B lymphocyte activation is key in the generation of adaptive immune responses. Derailed B lymphocyte activation is a hallmark of many autoimmune diseases and modulation of this immune response is therefore of therapeutic interest. Recently the success of B cell therapies in autoimmune diseases has been established. Treatment of rheumatoid arthritis (RA) patients with Rituximab (anti-CD20 therapy) is an accepted clinical therapy by now. More recent clinical trial studies show that treatment with Rituximab also ameliorates disease symptoms in relapsing remitting multiple sclerosis (RRMS) and systemic lupus erythematosus (SLE) patients. This success supports the potential for future therapies in autoimmune diseases targeting B cell immunity.
Bruton tyrosine kinase (Btk) is a Tec family non-receptor protein kinase, expressed in B cells and myeloid cells. The function of Btk in signaling pathways activated by the engagement of the B cell receptor (BCR) and FcsRl on mast cells is well established. In addition, a function for Btk as a downstream target in Toll-like receptor signaling was suggested. Functional mutations in Btk in human results in the primary immunodeficiency disease called XLA which is characterized by a defect in B cell development with a block between pro- and pre-B cell stage. This results in an almost complete absence of B lymphocytes in human causing a pronounced reduction of serum immunoglobulin of all classes. These finding support the key role for Btk in the regulation of the production of auto-antibodies in autoimmune diseases. In addition, regulation of Btk may affect BCR-induced production of pro-inflammatory cytokines and chemokines by B cells, indicating a broad potential for Btk in the treatment of autoimmune diseases.
With the regulatory role reported for Btk in FcsR-mediated mast cell activation, Btk inhibitors may also show potential in the treatment of allergic responses [Gilfillan et al, Immunological Reviews 288 (2009) ppl49-169]. Furthermore, Btk is also reported to be implicated in RANKL-induced osteoclast differentiation [Shinohara et al, Cell 132 (2008) pp794-806] and therefore may also be of interest for the treatment of bone resorption disorders.
Other diseases with an important role for dysfunctional B cells are B cell malignancies. Indeed anti-CD20 therapy is used effectively in the clinic for the treatment of follicular lymphoma, diffuse large B-cell lymphoma and chronic lymphocytic leukemia [Lim et al, Haematologica, 95 (2010) ppl35-143]. The reported role for Btk in the regulation of proliferation and apoptosis of B cells indicates there is potential for Btk inhibitors in the treatment of B cell lymphomas as well. Inhibition of Btk seems to be relevant in particular for B cell lymphomas due to chronic active BCR signaling [Davis et al, Nature, 463 (2010) pp88-94].
Some classes of Btk inhibitor compounds have been described as kinase inhibitors, e.g. Imidazo[l,5-f][l,2,4]triazine compounds have been described in WO2005097800 and WO2007064993. Imidazo[l,5-a]pyrazine compounds have been described in
WO2005037836 and WO2001019828 as IGF-1R enzyme inhibitors.
Some of the Btk inhibitors reported are not selective over Src-family kinases.
With dramatic adverse effects reported for knockouts of Src-family kinases, especially for double and triple knockouts, this is seen as prohibitive for the development of Btk inhibitors that are not selective over the Src-family kinases.
Both Lyn-deficient and Fyn-deficient mice exhibit autoimmunity mimicking the phenotype of human lupus nephritis. In addition, Fyn-deficient mice also show pronounced neurological defects. Lyn knockout mice also show an allergic-like phenotype, indicating Lyn as a broad negative regulator of the IgE-mediated allergic response by controlling mast cell responsiveness and allergy-associated traits [Odom et al, J. Exp. Med., 199 (2004) ppl491- 1502]. Furthermore, aged Lyn knock-out mice develop severe splenomegaly (myeloid expansion) and disseminated monocyte/macrophage tumors [Harder et al, Immunity, 15 (2001) pp603-615]. These observations are in line with hyperresponsive B cells, mast cells and myeloid cells, and increased Ig levels observed in Lyn-deficient mice. Female Src knockout mice are infertile due to reduced follicle development and ovulation [Roby et al, Endocrine, 26 (2005) ppl69-176]. The double knockouts Src^Tyn"7" and Src^Yes"7" show a severe phenotype with effects on movement and breathing. The triple knockouts Src^Tyn^ es"7" die at day 9.5
[Klinghoffer et al, EMBO I, 18 (1999) pp2459-2471]. For the double knockout Src^Hck"7", two thirds of the mice die at birth, with surviving mice developing osteopetrosis, extramedullary hematopoiseis, anemia, leukopenia [Lowell et al, Blood, 87 (1996) ppl780-1792]. Hence, an inhibitor that inhibits multiple or all kinases of the Src-family kinases simultaneously may cause serious adverse effects.
SUMMARY OF THE INVENTION
The present invention provides compounds which inhibit Btk activity, their use for treatment of Btk mediated diseases and disorders, in particular autoimmune diseases and inflammatory diseases, as well as pharmaceutical compositions comprising such compounds and pharmaceutical carriers. DETAILED DESCRIPTION
Definitions
The terms used herein have their ordinary meaning.
The term "amount effective" or "effective amount" as used herein, refers to an amount of the compound of Formula I and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventative effect when administered to a subject suffering from a BTK-mediated disease or disorder. In the combination therapies of the present invention, an effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount.
The term "cyclic moiety" refers to cyclopropyl, cyclobutyl and cyclopentyl and also refers to cyclic structures formed as a result of variables coming together to form a carbon bridge (for example, variables X and T forming a carbon bridge as highlighted in example compound 16).
The term "halogen", as used herein, refers to fluorine, chlorine, bromine or iodine. Fluorine, chlorine or bromine being preferred halogens; fluorine being more preferred.
When any variable occurs more than one time in any constituent or in any formula depicting and describing compounds of the invention, its definition on each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The term "purified" as used herein, refers to the physical state of a compound after the compound has been isolated through a synthetic process (e.g., from a reaction mixture), from a natural source, or a combination thereof. The term "purified" also refers to the physical state of a compound after the compound has been obtained from a purification process or processes described herein or well-known to the skilled artisan (e.g., chromatography, recrystallization, and the like), in sufficient purity to be characterizable by standard analytical techniques described herein or well-known to the skilled artisan.
A "subject" is a human or non-human mammal. In one embodiment, a subject is a human. In another embodiment, the subject is a chimpanzee.
It should be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
Compounds of the Invention
The present invention provides Btk inhibitor compounds according to Formula I or pharmaceutically acceptable salts thereof
Figure imgf000006_0001
Formula I
wherein:
Ring A is selected from the group consisting of
Figure imgf000006_0002
n is 0, 1 or 2; g is 0 or 1 ; W is CH2 or O;
Z is C or N;
X and Y are independently H or X and Y can come together to form cyclopropyl, cyclobutyl or cyclopentyl,
S and T are independently H, F, OH, OCH3, methyl, ethyl or S and T can come together to form cyclopropyl, provided that when Z is C then S and T can come together to form =0,
M and L are independently H or M and L can come together to form cyclopropyl,
X and T can come together to form a one to three carbon bridge,
wherein one of X and Y, S and T, M and L, and X and T must be a cyclic moiety;
Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
The present invention provides Btk inhibitor compounds according to Formula la or pharmaceutically acceptable salts thereof
Ring A
Figure imgf000007_0001
Formula la
wherein:
Ring A is selected from the group consisting of
Figure imgf000008_0001
n is 0, 1 or 2;
S and T are independently H, F, OH, OCH3, methyl, ethyl or S and T can come together to form =0;
Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
The present invention provides Btk inhibitor compounds according to Formula lb or pharmaceutically acceptable salts thereof
Figure imgf000008_0002
Formula lb
wherein:
Ring A is selected from the group consisting of
Figure imgf000008_0003
n is 0, 1 or 2; Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and
R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
Non-limiting examples of the compounds of the present invention include:
4-(8-amino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-^
(4-(tafluoromethyl)pyridin-2-yl)benzamide;
4-(8-amino-3-((6'S,8a'R)-l^ i'-difluoro-3'-oxota
yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-(8-amino-3-((6'R,8a'S)-l^ i'-difluoro-3'-oxota
yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-(8-amino-3-((6¾,8a'S)-3'-oxohexahydro-l^spiro[cy ^
a]pyrazin-l-yl)-3-ethoxy-5-fluoro-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]iinidazo[l,5-a]pyrazin-l-yl}-3-fluoro-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-N-(4-methylpyridin-2-yl)benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-N-(4-cyclopropylpyridin-2-yl)benzamide;
4-{8-ainino-3-[( S,6'R,8a'S)- -hydroxy-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[( S,6'R,8a'S)- -hydroxy-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-(8-amino-3-(( S,6'R,8a'S)- -hydroxy-3'-oxohexahydro- H-spiro[cyclopropane-l,2'- indolizin]-6'-yl)imidazo[l,5-a]pyrazin-l-yl)-3-fluoro-N-(4-(trifluoromethyl)pyridin-2- yl)benzamide;
4-(8-amino-3-(( S,6'R,8a'S)- -hydroxy-3'-oxohexahydro- H-spiro[cyclopropane-l,2'- indolizin]-6'-yl)imidazo[l,5-a]pyrazin-l-yl)-N-(4-cyclopropylpyridin-2-yl)benzamide; 4-(8-amino-3-(( S,6'R,8a'S)- -methoxy-3'-oxohexahydro- H-spiro[cyclopropane-l,2'- indolizin]-6'-yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N- (4-(trifluoromethyl)pyridin-2- yl)benzamide;
4-{8-amino-3-[(5aS,8R,l laR)-l l-oxodecahydro-lH-pyrido[l,2-a]pyrrolo[l,2-d]pyrazin-8- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(5aS,8R,l laS)-l l-oxodecahydro-lH-pyrido[l,2-a]pyrrolo[l,2-d]pyrazin-8- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(7'R,9a'S)-4'-oxohexahydro-2'H-spiro[cyclobutane-l,3'-pyrido[l,2-a]pyrazin]-7'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4- { 8-amino-3 -[(6'R, 8a' S)-3 '-oxohexahy dro-5 Ή-spiro [cyclopropane- 1 , 1 '-indolizin] -6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'S,8a'R)-3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4- { 8-amino-3 -[(6'R, 8a' S)-3 '-oxohexahy dro-5 Ή-spiro [cyclopropane- 1 , 1 '-indolizin] -6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-5-fluoro-N-[4-(trifluoromethyl)pyridin-2- yljbenzamide;
4- { 8-amino-3 -[(6'R, 8a' S)-3 '-oxohexahy dro-5 Ή-spiro [cyclopropane- 1 , 1 '-indolizin] -6'- yl]iinidazo[l,5-a]pyrazin-l-yl}-N-(4-cyclopropylpyridin-2-yl)-3-ethoxy-5-fluorobenzamide; 4-[8-amino-5-fluoro-3-(3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-5-fluoro-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3- yl]benzamide;
4- [8-amino-5-fluoro-3-(3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-5-fluoro-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3- yl]benzamide;
4- [8-amino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl]-3- ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-[8-ainino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl]-3- ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-[8-ainino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl]-3- ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,l'-difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]iinidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-amino-3-[(6'R,8a'S)-l^ -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-5-fluoro-N-[4-(trifluoromethyl)pyridin-2- yl]benzamide;
4-{8-amino-3-[(6'S,8a'R)-l -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- y 1] imidazo [ 1 ,5 -a] py razin- 1 -y 1 } -3 -methoxy-N- [4-(trifluoromethy l)pyridin-2-y 1] benzamide; 4-{8-amino-3-[(6'R,8a'S)-l^ -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- y 1] imidazo [ 1 ,5 -a] py razin- 1 -y 1 } -3 -fluoro-5-methoxy-N- [4-(trifluoromethy l)py ridin-2- yl]benzamide;
4-{8-amino-3-[(6'S,8a'R)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-3- yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-5-ethoxy-2-fluoro-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3 - yl] benzamide;
4- {8-amino-3-[(6'R,8a'S)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-2-chloro-5-ethoxy-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3 - yl] benzamide;
4- {8-amino-3-[(3R,6aR,l laR)-6-oxooctahydro-lH,6H-pyrrolo[r,2':4,5]pyrazino[2,l- c][l,4]oxazin-3-yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2- yljbenzamide;
4-{8-amino-3-[(3R,6aS,l laR)-6-oxooctahydro-lH,6H-pyrrolo[r,2':4,5]pyrazino[2,l- c][l,4]oxazin-3-yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2- yljbenzamide;
4-[8-amino-5-chloro-3-(3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl)imidazo[l,5-a]pyrazin-l-yl]-N-(4-cyanopyridin-2-yl)-3-ethoxy-5-fluorobenzamide;
4-[8-amino-3-(3'-oxohexahydro-5H-spiro[cyclopropane-lJ'-indolizin]-6'-yl)imidazo[l,5- a]pyrazin-l-yl]-N-(4-cyanopyridin-2-yl)-3-ethoxy-5-fluorobenzamide;
4-[8-amino-3-(4'-oxohexahydro-2'H,6'H-spiro[cyclopropane-l, -pyrido[l,2-a]pyrazin]-7'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(7'R,9a'S)-4'-oxohexahydro-2'H-spiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-
7'-yl]imidazo[l,5-a]pyrazin-l-yl}-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-amino-3-[(7'R,9a'S)-4'-oxohexahydro-2'H-spiro[cyclopropane-l ,3'-pyrido[l,2-a]pyrazin]-
7'-yl]imidazo[l ,5-a]pyrazin-l -yl} -3-methoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzam
4-[8-amino-3-(4'-oxohexahydro-2'H,6'H-spiro[cyclopropane-l, -pyrido[l ,2-a]pyrazin]-7'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-[8-amino-3-(4'-oxohexahydro-2'H,6'H-spiro[cyclopropane-l, -pyrido[l ,2-a]pyrazin]-7'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(7'R,9a'S)-2'-ethyl-4'-oxohexahydro-2'H-spiro[cyclopropane-l,3'-pyrido[l ,2- a]pyrazin]-7'-yl]imidazo[l ,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2- yl]benzamide;
4-{8-amino-3-[(3'R,9a'R)-6'-oxohexahydrospiro[cyclopropane-l ,7'-pyrazino[2, l-c] [l,4]oxazin]- 3'-yl]imidazo[l ,5-a]pyrazin-l -yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-amino-3-[(3'R,9a'R)-6'-oxohexahydrospiro[cyclopentane-l,7'-pyrazino[2,l -c] [l,4]oxazin]- 3'-yl]imidazo[l ,5-a]pyrazin-l -yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-amino-3-[(6'S,8a'R)-3'-oxohexahydrospiro[cyclopropane-l ,2'-indolizin]-6'-yl]imidazo[l,5- a]pyrazin-l-yl}-3-methoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; and
4-{8-amino-3-[(6'S,8a'R)-3'-oxohexahydrospiro[cyclopropane-l ,2'-indolizin]-6'-yl]imidazo[l,5- a] py razin- 1 -y 1 } -3 -ethoxy-N- [4-(trifluoromethy l)py ridin-2-yl] benzamide;
or a pharmaceutically acceptable salt thereof.
In a preferred embodiment halogen is F or CI.
In a preferred embodiment R3 is H or F.
The compounds of this invention include the salts, solvates, hydrates or prodrugs of the compounds. The use of the terms "salt", "solvate", "hydrate", "prodrug" and the like, is intended to equally apply to the salt, solvate, hydrate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, or racemates of the inventive compounds.
Salts
The Btk inhibitor compounds of the present invention, which can be in the form of a free base, may be isolated from the reaction mixture in the form of a pharmaceutically acceptable salt.
The compounds of Formula I can form salts which are also within the scope of this invention. Reference to a compound of Formula I herein is understood to include reference to pharmaceutically acceptable salts thereof, unless otherwise indicated. The term
"pharmaceutically acceptable salt(s)" or "salt", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when a compound of Formula I contains both a basic moiety, such as, but not limited to a pyridine or imidazole, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term "salt(s)" as used herein. Such acidic and basic salts used within the scope of the invention are pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts. Salts of the compounds of Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
Exemplary acid addition salts include acetates, ascorbates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates, methanesulfonates, naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates,) and the like.
Additionally, acids which are generally considered suitable for the formation of
pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33 201-217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference.
Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen- containing groups may be quarternized with agents such as lower alkyl halides (e.g., methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g., decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenethyl bromides), and others.
Crystals
The Btk inhibitor compounds of the present invention may exist as amorphous forms or crystalline forms. The compounds of Formula I may have the ability to crystallize in more than one form, a characteristic known as polymorphism, and it is understood that such polymorphic forms ("polymorphs") are within the scope of Formula I. Polymorphism generally can occur as a response to changes in temperature or pressure or both and can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility and melting point.
Solvates
The compounds having Formula I or the pharmaceutically acceptable salts may form hydrates or solvates. It is known to those of skill in the art that charged compounds form hydrated species when lyophilized with water, or form solvated species when concentrated in a solution with an appropriate organic solvent. The compounds of this invention include the hydrates or solvates of the compounds listed.
One or more compounds of the invention having Formula I or the pharmaceutically acceptable salts or solvates thereof may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. "Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H20.
Preparation of solvates is generally known. Thus, for example, M. Caira et al, J. Pharmaceutical Sci. , 93(3). 601-611 (2004) describe the preparation of the solvates of the antifungal fluconazole in ethyl acetate as well as from water. Similar preparations of solvates, hemisolvate, hydrates and the like are described by E. C. van Tonder et al, AAPS
PharmSciTech., 5(1). article 12 (2004); and A. L. Bingham et al, Chem. Commun. 603-604 (2001). A typical, non-limiting, process involves dissolving the inventive compound in desired amounts of the desired solvent (organic or water or mixtures thereof) at a higher than ambient temperature, and cooling the solution at a rate sufficient to form crystals which are then isolated by standard methods. Analytical techniques such as, for example IR spectroscopy, show the presence of the solvent (or water) in the crystals as a solvate (or hydrate). Optical Isomers
The compounds of Formula I may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of Formula I, as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention embraces all geometric and positional isomers. For example, if a compound of Formula I incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the invention. Such stereoisomeric forms also include enantiomers and diastereoisomers, etc.
For chiral compounds, methods for asymmetric synthesis whereby the pure stereoisomers are obtained are well known in the art, e.g. synthesis with chiral induction, synthesis starting from chiral intermediates, enantioselective enzymatic conversions, separation of stereoisomers using chromatography on chiral media. Such methods are described in Chirality in Industry (edited by A.N. Collins, G.N. Sheldrake and J. Crosby, 1992; John Wiley). Likewise methods for synthesis of geometrical isomers are also well known in the art.
Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g. chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g. hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some of the compounds of Formula I may be atropisomers (e.g. substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of chiral HPLC column.
It is also possible that the compounds of Formula I may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. Also, for example, all keto-enol and imine-enamine forms of the compounds are included in the invention.
All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those of the salts, solvates, esters and prodrugs of the compounds as well as the salts, solvates and esters of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The use of the terms "salt", "solvate", "ester", "prodrug" and the like, is intended to equally apply to the salt, solvate, ester and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
Prodrugs
A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press. The term "prodrug" means a compound (e.g, a drug precursor) that is transformed in vivo to yield a compound of Formula I or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms (e.g. by metabolic or chemical processes), such as, for example, through hydrolysis in blood. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
Isotopes
In the compounds of Formula I, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of generic Formula I. For example, different isotopic forms of hydrogen (H) include protium (TT) and deuterium (2H). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
Isotopically-enriched compounds within generic Formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates. Certain isotopically -labelled compounds of Formula I (e.g. those labeled with H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., H) and carbon-14 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labelled compounds of Formula I can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herinbelow, by substituting an appropriate isotopically labeled reagent for a non-isotopically labeled reagent.
Utilities
The compounds having Formula I and pharmaceutical compositions thereof can be used to treat or prevent a variety of conditions, diseases or disorders mediated by Bruton's Tyrosine kinase (Btk). Such Btk-mediated conditions, diseases or disorders include, but are not limited to: (1) arthritis, including rheumatoid arthritis, juvenile arthritis, psoriatic arthritis and osteoarthritis; (2) asthma and other obstructive airways diseases, including chronic asthma, late asthma, airway hyper-responsiveness, bronchitis, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, adult respiratory distress syndrome, recurrent airway obstruction, and chronic obstruction pulmonary disease including emphysema; (3) autoimmune diseases or disorders, including those designated as single organ or single cell-type autoimmune disorders, for example Hashimoto's thyroiditis, autoimmune hemolytic anemia, autoimmune atrophic gastritis of pernicious anemia, autoimmune encephalomyelitis, autoimmune orchitis, Goodpasture's disease, autoimmune thrombocytopenia including idiopathic thrombopenic purpura, sympathetic ophthalmia, myasthenia gravis, Graves' disease, primary biliary cirrhosis, chronic aggressive hepatitis, ulcerative colitis and membranous glomerulopathy, those designated as involving systemic autoimmune disorder, for example systemic lupus
erythematosis, immune thrombocytopenic purpura, rheumatoid arthritis, Sjogren's syndrome, Reiter's syndrome, polymyositis-dermatomyositis, systemic sclerosis, polyarteritis nodosa, multiple sclerosis and bullous pemphigoid, and additional autoimmune diseases, which can be B- cell (humoral) based or T-cell based, including Cogan's syndrome, ankylosing spondylitis, Wegener's granulomatosis, autoimmune alopecia, Type I or juvenile onset diabetes, and thyroiditis; (4) cancers or tumors, including alimentary /gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer including mast cell tumor and squamous cell carcinoma, breast and mammary cancer, ovarian cancer, prostate cancer, lymphoma and leukemia (including but not limited to acute myelogenous leukemia, chronic myelogenous leukemia, mantle cell lymphoma, NHL B cell lymphomas (e.g. precursor B-ALL, marginal zone B cell lymphoma, chronic lymphocytic leukemia, diffuse large B cell lymphoma, Burkitt lymphoma, mediastinal large B-cell lymphoma), Hodgkin lymphoma, NK and T cell lymphomas; TEL-Syk and ITK-Syk fusion driven tumors, myelomas including multiple myeloma, myeloproliferative disorders kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer, brain cancer, melanoma including oral and metastatic melanoma, Kaposi's sarcoma, proliferative diabetic retinopathy, and angiogenic-associated disorders including solid tumors, and pancreatic cancer; (5) diabetes, including Type I diabetes and complications from diabetes; (6) eye diseases, disorders or conditions including autoimmune diseases of the eye, keratoconjunctivitis, vernal conjunctivitis, uveitis including uveitis associated with Behcet's disease and lens-induced uveitis, keratitis, herpetic keratitis, conical keratitis, corneal epithelial dystrophy, keratoleukoma, ocular premphigus, Mooren's ulcer, scleritis, Grave's ophthalmopathy, Vogt-Koyanagi-Harada syndrome, keratoconjunctivitis sicca (dry eye), phlyctenule, iridocyclitis, sarcoidosis, endocrine ophthalmopathy, sympathetic ophthalmitis, allergic conjunctivitis, and ocular
neovascularization; (7) intestinal inflammations, allergies or conditions including Crohn's disease and/or ulcerative colitis, inflammatory bowel disease, coeliac diseases, proctitis, eosinophilic gastroenteritis, and mastocytosis; (8) neurodegenerative diseases including motor neuron disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis,
Huntington's disease, cerebral ischemia, or neurodegenerative disease caused by traumatic injury, strike, glutamate neurotoxicity or hypoxia; ischemic/ reperfusion injury in stroke, myocardial ischemica, renal ischemia, heart attacks, cardiac hypertrophy, atherosclerosis and arteriosclerosis, organ hypoxia; (9) platelet aggregation and diseases associated with or caused by platelet activation, such as arteriosclerosis, thrombosis, intimal hyperplasia and restenosis following vascular injury; (10) conditions associated with cardiovascular diseases, including restenosis, acute coronary syndrome, myocardial infarction, unstable angina, refractory angina, occlusive coronary thrombus occurring post-thrombolytic therapy or post-coronary angioplasty, a thrombotically mediated cerebrovascular syndrome, embolic stroke, thrombotic stroke, transient ischemic attacks, venous thrombosis, deep venous thrombosis, pulmonary embolus, coagulopathy, disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, thromboangiitis obliterans, thrombotic disease associated with heparin-induced
thrombocytopenia, thrombotic complications associated with extracorporeal circulation, thrombotic complications associated with instrumentation such as cardiac or other intravascular catheterization, intra-aortic balloon pump, coronary stent or cardiac valve, conditions requiring the fitting of prosthetic devices, and the like; (1 1) skin diseases, conditions or disorders including atopic dermatitis, eczema, psoriasis, scleroderma, pruritus and other pruritic conditions; (12) allergic reactions including anaphylaxis, allergic rhinitis, allergic dermatitis, allergic urticaria, angioedema, allergic asthma, or allergic reaction to insect bites, food, drugs, or pollen; (13) transplant rejection, including pancreas islet transplant rejection, bone marrow transplant rejection, graft- versus-host disease, organ and cell transplant rejection such as bone marrow, cartilage, cornea, heart, intervertebral disc, islet, kidney, limb, liver, lung, muscle, myoblast, nerve, pancreas, skin, small intestine, or trachea, and xeno transplantation; and (14) low grade scarring including scleroderma, increased fibrosis, keloids, post-surgical scars, pulmonary fibrosis, vascular spasms, migraine, reperfusion injury, and post-myocardial infarction.
The invention thus provides compounds of Formula I and salts thereof for use in therapy, and particularly in the treatment of disorders, diseases and conditions mediated by inappropriate Btk activity.
The inappropriate Btk activity referred to herein is any Btk activity that deviates from the normal Btk activity expected in a particular mammalian subj ect. Inappropriate Btk activity may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and or control of Btk activity. Such inappropriate activity may result then, for example, from overexpression or mutation of the protein kinase leading to inappropriate or uncontrolled activation.
In one embodiment, the present invention provides for the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a Btk-mediated disorder.
In another embodiment, the present invention provides methods of regulating, modulating, or inhibiting Btk for the prevention and/or treatment of disorders related to unregulated or inappropriate Btk activity.
In a further embodiment, the present invention provides a method for treating a subject suffering from a disorder mediated by Btk, which comprises administering to said subject a compound of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat the Btk-mediated disorder. A further aspect of the invention resides in the use of a compound of Formula I or a pharmaceutically acceptable salt thereof for the manufacture of a medicament to be used for the treatment of chronic B cell disorders in which T cells play a prominent role.
Thus, the compounds according to the invention may be used in therapies to treat or prevent Bruton's Tyrosine Kinase (Btk) mediated diseases, conditions and disorders. Btk mediated diseases, conditions and disorders as used herein, mean any disease, condition or disorder in which B cells, mast cells, myeloid cells or osteoclasts play a central role. These diseases include but are not limited to, immune, autoimmune and inflammatory diseases, allergies, infectious diseases, bone resorption disorders and proliferative diseases.
Immune, autoimmune and inflammatory diseases that may be treated or prevented with the compounds of the present invention include rheumatic diseases (e.g. rheumatoid arthritis, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, deforming arthritis, osteoarthritis, traumatic arthritis, gouty arthritis, Reiter's syndrome, polychondritis, acute synovitis and spondylitis), glomerulonephritis (with or without nephrotic syndrome),
Goodpasture's syndrome, (and associated glomerulonephritis and pulmonary hemorrhage), atherosclerosis, autoimmune hematologic disorders (e.g. hemolytic anemia, aplasic anemia, idiopathic thrombocytopenia, chronic idiopathic thrombocytopenic purpura (ITP), and neutropenia), autoimmune gastritis, and autoimmune inflammatory bowel diseases (e.g.
ulcerative colitis and Crohn's disease), irritable bowel syndrome, host versus graft disease, allograft rejection, chronic thyroiditis, Graves' disease, Sjorgren's disease, scleroderma, diabetes (type I and type II), active hepatitis (acute and chronic), pancreatitis, primary billiary cirrhosis, myasthenia gravis, multiple sclerosis, systemic lupus erythematosis, psoriasis, atopic dermatitis, dermatomyositis, contact dermatitis, eczema, skin sunburns, vasculitis (e.g. Behcet's disease), ANCA-associated and other vasculitudes, chronic renal insufficiency, Stevens-Johnson syndrome, inflammatory pain, idiopathic sprue, cachexia, sarcoidosis, Guillain-Barre syndrome, uveitis, conjunctivitis, kerato conjunctivitis, otitis media, periodontal disease, Addison's disease, Parkinson's disease, Alzheimer's disease, diabetes, septic shock, myasthenia gravis, pulmonary interstitial fibrosis, asthma, bronchitis, rhinitis, sinusitis, pneumoconiosis, pulmonary insufficiency syndrome, pulmonary emphysema, pulmonary fibrosis, silicosis, chronic inflammatory pulmonary disease (e.g. chronic obstructive pulmonary disease) and other inflammatory or obstructive disease on airways. Allergies that may be treated or prevented include, among others, allergies to foods, food additives, insect poisons, dust mites, pollen, animal materials and contact allergans, type I hypersensitivity allergic asthma, allergic rhinitis, allergic conjunctivitis.
Infectious diseases that may be treated or prevented include, among others, sepsis, septic shock, endotoxic shock, sepsis by Gram-negative bacteria, shigellosis, meningitis, cerebral malaria, pneumonia, tuberculosis, viral myocarditis, viral hepatitis (hepatitis A, hepatitis B and hepatitis C), HIV infection, retinitis caused by cytomegalovirus, influenza, herpes, treatment of infections associated with severe burns, myalgias caused by infections, cachexia secondary to infections, and veterinary viral infections such as lentivirus, caprine arthritic virus, visna-maedi virus, feline immunodeficiency virus, bovine immunodeficiency virus or canine
immunodeficiency virus.
Bone resorption disorders that may be treated or prevented include, among others, osteoporosis, osteoarthritis, traumatic arthritis, gouty arthritis and bone disorders related with multiple myeloma.
Proliferative diseases that may be treated or prevented include, among others, non-Hodgkin lymphoma (in particular the subtypes diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL)), B cell chronic lymphocytic leukemia and acute lymphoblastic leukemia (ALL) with mature B cell, ALL in particular.
In particular the compounds of Formula I or pharmaceutically acceptable salts may be used for the treatment of B cell lymphomas resulting from chronic active B cell receptor signaling.
Yet another aspect of the present invention provides a method for treating diseases caused by or associated with Fc receptor signaling cascades, including FceRI and/or FcgRI-mediated degranulation as a therapeutic approach towards the treatment or prevention of diseases characterized by, caused by and/or associated with the release or synthesis of chemical mediators of such Fc receptor signaling cascades or degranulation. In addition, Btk is known to play a critical role in immunotyrosine-based activation motif (IT AM) singaling, B cell receptor signaling, T cell receptor signaling and is an essential component of integrin beta (1), beta (2), and beta (3) signaling in neutrophils. Thus, compounds of the present invention can be used to regulate Fc receptor, ITAM, B cell receptor and integrin signaling cascades, as well as the cellular responses elicited through these signaling cascades. Non-limiting examples of cellular responses that may be regulated or inhibited include respiratory burst, cellular adhesion, cellular degranulation, cell spreading, cell migration, phagocytosis, calcium ion flux, platelet aggregation and cell maturation.
Combination Therapy
Included herein are methods of treatment and/ or pharmaceutical compositions in which at least one compound of Formula I or a pharmaceutically acceptable salt thereof is administered in combination with at least one other active agent. The other active agent is an anti-inflammatory agent, an immunosuppressant agent, or a chemotherapeutic agent. Antiinflammatory agents include but are not limited to NSAIDs, non-specific and COX-2 specific cyclooxgenase enzyme inhibitors, gold compounds, corticosteroids, methotrexate, tumor necrosis factor receptor (TNF) receptors antagonists, immunosuppressants and methotrexate.
Examples of NSAIDs include, but are not limited to, ibuprofen, flurbiprofen, naproxen and naproxen sodium, diclofenac, combinations of diclofenac sodium and misoprostol, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, sodium nabumetone, sulfasalazine, tolmetin sodium, and hydroxychloroquine.
Examples of NSAIDs also include COX-2 specific inhibitors such as celecoxib, valdecoxib, lumiracoxib and/or etoricoxib.
In some embodiments, the anti-inflammatory agent is a salicylate. Salicylates include by are not limited to acetylsalicylic acid or aspirin, sodium salicylate, and choline and magnesium salicylates.
The anti-inflammatory agent may also be a corticosteroid. For example, the corticosteroid may be cortisone, dexamethasone, methylprednisolone, prednisolone,
prednisolone sodium phosphate, or prednisone.
In additional embodiments the anti-inflammatory agent is a gold compound such as gold sodium thiomalate or auranofin.
The invention also includes embodiments in which the anti-inflammatory agent is a metabolic inhibitor such as a dihydrofolate reductase inhibitor, such as methotrexate or a dihydroorotate dehydrogenase inhibitor, such as leflunomide.
Other embodiments of the invention pertain to combinations in which at least one anti-inflammatory agent is an anti-C5 monoclonal antibody (such as eculizumab or
pexelizumab), a TNF antagonist, such as entanercept, or infliximab, which is an anti-TNF alpha monoclonal antibody.
Still other embodiments of the invention pertain to combinations in which at least one active agent is an immunosuppressant agent, such as an immunosuppressant compound chosen from methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine, and
mycophenolate mofetil.
B-cells and B-cell precursors expressing BTK have been implicated in the pathology of B-cell malignancies, including, but not limited to, B-cell lymphoma, lymphoma (including Hodgkin's and non-Hodgkin's lymphoma), hairy cell lymphoma, multiple myeloma, chronic and acute myelogenous leukemia and chronic and acute lymphocytic leukemia.
BTK has been shown to be an inhibitor of the Fas/APO-1 (CD-95) death inducing signaling complex (DISC) in B-lineage lymphoid cells. The fate of leukemia/lymphoma cells may reside in the balance between the opposing proapoptotic effects of caspases activated by DISC and an upstream anti-apoptotic regulatory mechanism involving BTK and/or its substrates (Vassilev et al, J. Biol. Chem. 1998, 274, 1646-1656).
It has also been discovered that BTK inhibitors are useful as chemosensitizing agents, and, thus, are useful in combination with other chemotherapeutic agents, in particular, drugs that induce apoptosis. Examples of other chemotherapeutic agents that can be used in combination with chemosensitizing BTK inhibitors include topoisomerase I inhibitors
(camptothecin or topotecan), topoisomerase II inhibitors (e.g. daunomycin and etoposide), alkylating agents (e.g. cyclophosphamide, melphalan and BCNU), tubulin directed agents (e.g. taxol and vinblastine), and biological agents (e.g. antibodies such as anti CD20 antibody, IDEC 8, immunotoxins, and cytokines).
Btk activity has also been associated with some leukemias expressing the bcr-abl fusion gene resulting from translocation of parts of chromosome 9 and 22. This abnormality is commonly observed in chronic myelogenous leukemia. Btk is constitutively phosphorylated by the bcr-abl kinase which initiates downstream survival signals which circumvents apoptosis in bcr-abl cells. (N. Feldhahn et al. J. Exp. Med. 2005 201(11): 1837-1852).
The compound(s) of Formula I and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately this may occur simultaneously or sequentially in any order. The amounts of the compound(s) of Formula I and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
For the treatment of the inflammatory diseases, rheumatoid arthritis, psoriasis, inflammatory bowel disease, COPD, asthma and allergic rhinitis a compound of Formula I may be combined with one or more other active agents such as: (1) TNF-a inhibitors such as infliximab (Remicade®), etanercept (Enbrel®), adalimumab (Humira®), certolizumab pegol (Cimzia®), and golimumab (Simponi®); (2) non-selective COX-I/COX-2 inhibitors (such as piroxicam, diclofenac, propionic acids such as naproxen, flubiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, etodolac, azapropazone, pyrazolones such as phenylbutazone, salicylates such as aspirin); (3) COX-2 inhibitors (such as meloxicam, celecoxib, rofecoxib, valdecoxib and etoricoxib); (4) other agents for treatment of rheumatoid arthritis including methotrexate, leflunomide, sulfasalazine, azathioprine, cyclosporin, tacrolimus, penicillamine, bucillamine, actarit, mizoribine, lobenzarit, ciclesonide, hydroxychloroquine, d-penicillamine, aurothiomalate, auranofin or parenteral or oral gold, cyclophosphamide, Lymphostat-B, BAFF/APRIL inhibitors and CTLA-4-Ig or mimetics thereof; (5) leukotriene biosynthesis inhibitor, 5 -lipoxygenase (5-LO) inhibitor or 5-lipoxygenase activating protein (FLAP) antagonist such as zileuton; (6) LTD4 receptor antagonist such as zafirlukast, montelukast and pranlukast; (7) PDE4 inhibitor such as roflumilast, cilomilast, AWD-12-281 (Elbion), and PD-168787 (Pfizer); (8) antihistaminic HI receptor antagonists such as cetirizine, levocetirizine, loratadine, desloratadine, fexofenadine, astemizole, azelastine, levocabastine, olopatidine, methapyrilene and chlorpheniramine; (9) al- and a2-adrenoceptor agonist vasoconstrictor sympathomimetic agent, such as propylhexedrine, phenylephrine, phenylpropanolamine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline
hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride, and
ethylnorepinephrine hydrochloride; (10) anticholinergic agents such as ipratropium bromide, tiotropium bromide, oxitropium bromide, aclindinium bromide, glycopyrrolate, (R,R)- glycopyrrolate, pirenzepine, and telenzepine; (11) β-adrenoceptor agonists such as
metaproterenol, isoproterenol, isoprenaline, albuterol, formoterol (particularly the fumarate salt), salmeterol (particularly the xinafoate salt), terbutaline, orciprenaline, bitolterol mesylate, fenoterol, and pirbuterol, or methylxanthanines including theophylline and aminophylline, sodium cromoglycate; (12) insulin-like growth factor type I (IGF-1) mimetic; (13)
glucocorticosteroids, especially inhaled glucocorticoid with reduced systemic side effects, such as prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, ciclesonide and mometasone furoate; (14) kinase inhibitors such as inhibitors of the Janus Kinases (JAK 1 and/or JAK2 and/or JAK 3 and/or TYK2), p38 MAPK and IKK2; (15) B-cell targeting biologies such as rituximab (Rituxan®); (16) selective costimulation modulators such as abatacept (Orencia); (17) interleukin inhibitors, such as IL-1 inhibitor anakinra (Kineret) and IL-6 inhibitor tocilizumab (Actemra). The present invention also provides for "triple combination" therapy, comprising a compound of Formula I or a pharmaceutically acceptable salt thereof together with beta2- adrenoreceptor agonist and an anti-inflammatory corticosteroid. Preferably this combination is for treatment and/or prophylaxis of asthma, COPD or allergic rhinitis. The beta2-adrenoreceptor agonist and/or the anti-inflammatory corticosteroid can be as described above and/or as described in WO 03/030939 Al . Representative examples of such a "triple" combination are a compound of Formula I or a pharmaceutically acceptable salt thereof in combination with the components of Advair® (salmeterol xinafoate and fluticasone propionate), Symbicort®
(budesonide and formoterol fumarate), or Dulera® (mometasone furoate and formoterol).
For the treatment of cancer a compound of Formula I may be combined with one or more of an anticancer agents. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Such anti-cancer agents include, but are not limited to, the following: (1) estrogen receptor modulator such as diethylstibestral, tamoxifen, raloxifene, idoxifene, LY353381, LY117081, toremifene, fluoxymestero, and SH646; (2) other hormonal agents including aromatase inhibitors (e.g., aminoglutethimide, tetrazole anastrozole, letrozole and exemestane), luteinizing hormone release hormone (LHRH) analogues, ketoconazole, goserelin acetate, leuprolide, megestrol acetate and mifepristone; (3) androgen receptor modulator such as finasteride and other 5a-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate; (4) retinoid receptor modulator such as bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, a-difluoromethylornithine, ILX23- 7553, trans-N-(4'-hydroxyphenyl) retinamide, and N-4-carboxyphenyl retinamide; (5) antiproliferative agent such asantisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methylidenecytidine, 2'- fluoromethylene-2'-deoxycytidine, N6-[4-deoxy-4-[N2-[2(E),4(E)-tetradeca- dienoyl]glycylamino]-L-glycero-B-L-manno-heptopyranosyl]adenine, aplidine, ecteinascidin, troxacitabine, aminopterin, 5-flurouracil, floxuridine, methotrexate, leucovarin, hydroxyurea, thioguanine (6-TG), mercaptopurine (6-MP), cytarabine, pentostatin, fludarabine phosphate, cladribine (2-CDA), asparaginase, gemcitabine, alanosine, swainsonine, lometrexol,
dexrazoxane, methioninase, and 3-aminopyridine-2-carboxaldehyde thiosemicarbazone; (6) prenyl-protein transferase inhibitor including farnesyl-protein transferase (FPTase),
geranylgeranyl-protein transferase type I (GGPTase-I), and geranylgeranyl-protein transferase type-II (GGPTase-II, also called Rab GGPTase); (7) HMG-CoA reductase inhibitor such as lovastatin, simvastatin, pravastatin, atorvastatin, fluvastatin and rosuvastatin; (8) angiogenesis inhibitor such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-l/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon-a, interleukin-12, erythropoietin (epoietin-a), granulocyte-CSF (filgrastin), granulocyte, macrophage-CSF
(sargramostim), pentosan polysulfate, cyclooxygenase inhibitors, steroidal anti-inflammatories, carboxyamidotriazole, combretastatin A-4, squalamine, 6-0-chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin- 1, angiotensin II antagonists, heparin, carboxypeptidase U inhibitors, and antibodies to VEGF, endostatin, ukrain, ranpirnase, IM862, acetyl dinanaline, 5- amino-l-[[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]methyl]-lH-l,2,3-triazole-4- carboxamide,CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, and 3-[(2,4-dimethylpyrrol-5-yl)methylene]-2-indolinone (SU5416); (9) PPAR-γ agonists, PPAR-δ agonists, thiazolidinediones (such as DRF2725, CS-011, troglitazone, rosiglitazone, and pioglitazone), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR- H039242, JTT-501, MCC-555, GW2331, GW409544, N 2344, KRP297, NP0110, DRF4158, N 622, GI262570, PNU182716, DRF552926, 2-[(5,7-dipropyl-3-trifluoromethyl-l,2- benzisoxazol-6-yl)oxy]-2-methylpropionic acid (disclosed in USSN 09/782,856), and (2R)-7-(3- (2-chloro-4-(4-fluorophenoxy)phenoxy)propoxy)-2-ethylchromane-2-carboxylic acid (disclosed in USSN 60/235,708 and 60/244,697); (9) inhibitor of inherent multidrug resistance including inhibitors of p-glycoprotein (P-gp), such as LY335979, XR9576, OC144-093, R101922, VX853 and PSC833 (valspodar); (10) inhibitor of cell proliferation and survival signaling such as inhibitors of EGFR (for example gefitinib and erlotinib), inhibitors of ERB-2 (for example trastuzumab), inhibitors of IGF1R such as MK-0646 (dalotuzumab), inhibitors of CD20
(rituximab), inhibitors of cytokine receptors, inhibitors of MET, inhibitors of PI3K family kinase (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in (WO 03/086404, WO 03/086403, WO 03/086394, WO 03/086279, WO 02/083675, WO 02/083139, WO 02/083140 and WO 02/083138), inhibitors of Raf kinase (for example BAY-43-9006 ), inhibitors of MEK (for example CI-1040 and PD-098059) and inhibitors of mTOR (for example Wyeth CCI-779 and Ariad AP23573); (11) a bisphosphonate such as etidronate, pamidronate, alendronate, risedronate, zoledronate, ibandronate, incadronate or cimadronate, clodronate, EB-1053, minodronate, neridronate, piridronate and tiludronate; (12) γ-secretase inhibitors, (13) agents that interfere with receptor tyrosine kinases (RTKs) including inhibitors of c-Kit, Eph, PDGF, Flt3 and c-Met; (14) agent that interferes with a cell cycle checkpoint including inhibitors of ATR, ATM, the Chkl and Chk2 kinases and cdk and cdc kinase inhibitors and are specifically exemplified by 7-hydroxystaurosporin, flavopiridol, CYC202 (Cyclacel) and BMS-387032; (15) BTK inhibitors such as PCI32765, AVL-292 and AVL-101; (16) PARP inhibitors including iniparib, olaparib, AG014699, ABT888 and
MK4827; (16) ERK inhibitors; (17) mTOR inhibitors such as sirolimus, ridaforolimus, temsirolimus, everolimus; (18) cytotoxic/cytostatic agents.
"Cytotoxic/cytostatic agents" refer to compounds which cause cell death or inhibit cell proliferation primarily by interfering directly with the cell's functioning or inhibit or interfere with cell mytosis, including alkylating agents, tumor necrosis factors, intercalators, hypoxia activatable compounds, microtubule inhibitors/microtubule-stabilizing agents, inhibitors of mitotic kinesins, inhibitors of histone deacetylase, inhibitors of kinases involved in mitotic progression, antimetabolites; biological response modifiers; hormonal/anti -hormonal therapeutic agents, haematopoietic growth factors, monoclonal antibody targeted therapeutic agents, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors.
Examples of cytotoxic agents include, but are not limited to, sertenef, cachectin, chlorambucil, cyclophosphamide, ifosfamide, mechlorethamine, melphalan, uracil mustard, thiotepa, busulfan, carmustine, lomustine, streptozocin, tasonermin, lonidamine, carboplatin, altretamine, dacarbazine, procarbazine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl- pyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans)-bis-mu-(hexane- l,6-diamine)-mu-[diarnine-platinum(II)]bis[diamine(chloro)platinum (II)]tetrachloride, diarizidinylspermine, arsenic trioxide, l-(l l-dodecylamino-10-hydroxyundecyl)-3,7- dimethylxanthine, zorubicin, doxorubicin, daunorubicin, idarubicin, anthracenedione, bleomycin, mitomycin C, dactinomycin, plicatomycin, bisantrene, mitoxantrone, pirarubicin, pinafide, valrubicin, amrubicin, antineoplaston, 3'-deamino-3'-morpholino-13-deoxo-10- hydroxycarminomycin, annamycin, galarubicin, elinafide, MEN10755, and 4-demethoxy-3- deamino-3-aziridinyl-4-methylsulphonyl-daunorubicin.
An example of a hypoxia activatable compound is tirapazamine.
Examples of proteasome inhibitors include but are not limited to lactacystin and bortezomib.
Examples of microtubule inhibitors/microtubule-stabilising agents include vincristine, vinblastine, vindesine, vinzolidine, vinorelbine, vindesine sulfate, 3',4'-didehydro- 4'-deoxy-8'-norvincaleukoblastine, podophyllotoxins (e.g., etoposide (VP- 16) and teniposide (VM-26)), paclitaxel, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS 184476, vinflunine, cryptophycin, anhydrovinblastine, N,N- dimethy 1-L-valy 1-L-valy 1-N-methy 1-L-valy 1-L-proly 1-L-proline-t-buty lamide, TDX258, the epothilones (see for example U.S. Pat. Nos. 6,284,781 and 6,288,237) and BMS 188797.
Some examples of topoisomerase inhibitors are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3',4'-0-exo-benzylidene-chartreusin, lurtotecan, 7-[2- (N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2'-dimethylamino-2'-deoxy-etoposide, GL331, N- [2-(dimethylamino)ethyl]-9-hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-l-carboxamide, asulacrine, 2,3-(methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c]-phenanthridinium, 5- (3-aininopropylamino)-7,10-dihydroxy-2-(2-hydroxyethylaminomethyl)-6H-pyrazolo[4,5,l- de]acridin-6-one, N-[l -[2-(diethylamino)ethylamino]-7-methoxy-9-oxo-9H-thioxanthen-4- ylmethyl]formamide, N-(2-(dimethylamino)ethyl)acridine-4-carboxarnide, 6-[[2- (dimethylamino)ethyl]amino]-3-hydroxy-7H-indeno[2,l-c]quinolin-7-one, and dimesna.
Examples of inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLPl, inhibitors of CENP-E, inhibitors of MCAK, inhibitors of Kifl4, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
Examples of "histone deacetylase inhibitors" include, but are not limited to, vorinostat, trichostatin A, oxamflatin, PXD101, MG98, valproic acid and scriptaid.
"Inhibitors of kinases involved in mitotic progression" include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK; in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-Rl . An example of an "aurora kinase inhibitor" is VX-680.
"Antiproliferative agents" includes antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methylidenecytidine, 2'- fluoromethylene-2'-deoxycytidine, N6-[4-deoxy-4- N2-[2,4-tetradecadienoyl]glycylamino]-L- glycero-B-L-manno-heptopyranosyl] adenine, aplidine, ecteinascidin, troxacitabine, aminopterin,
5-flurouracil, floxuridine, methotrexate, leucovarin, hydroxyurea, thioguanine (6-TG), mercaptopurine (6-MP), cytarabine, pentostatin, fludarabine phosphate, cladribine (2-CDA), asparaginase, gemcitabine, alanosine, swainsonine, lometrexol, dexrazoxane, methioninase, and
3-aminopyridine-2-carboxaldehyde thiosemicarbazone.
Non-limiting examples of suitable agents used in cancer therapy that may be combined with compounds of Formula I include, but are not limited to, abarelix; aldesleukin; alemtuzumab; alitretinoin; allopurinol; altretamine; amifostine; anastrozole; arsenic trioxide; asparaginase; azacitidine; bendamustine; bevacuzimab; bexarotene; bleomycin; bortezomib; busulfan; calusterone; capecitabine; carboplatin; carmustine; cetuximab; chlorambucil; cisplatin; cladribine; clofarabine; cyclophosphamide; cytarabine; dacarbazine; dactinomycin, actinomycin
D; dalteparin; darbepoetin alfa; dasatinib; daunorubicin; degarelix; denileukin diftitox;
dexrazoxane; docetaxel; doxorubicin; dromostanolone propionate; eculizumab; Elliott's B
Solution; eltrombopag; epirubicin; epoetin alfa; erlotinib; estramustine; etoposide phosphate; etoposide; everolimus; exemestane; filgrastim; floxuridine; fludarabine; fluorouracil; fulvestrant; gefitinib; gemcitabine; gemtuzumab ozogamicin; goserelin acetate; histrelin acetate;
hydroxyurea; ibritumomab tiuxetan; idarubicin; ifosfamide; imatinib mesylate; interferon alfa 2a; interferon alfa-2b; irinotecan; ixabepilone; lapatinib; lenalidomide; letrozole; leucovorin;
leuprolide acetate; levamisole; lomustine; meclorethamine, nitrogen mustard; megestrol acetate; melphalan, L-PAM; mercaptopurine; mesna; methotrexate; methoxsalen; mitomycin C;
mitotane; mitoxantrone; nandrolone phenpropionate; nelarabine; nilotinib; Nofetumomab;
ofatumumab; oprelvekin; oxaliplatin; paclitaxel; palifermin; pamidronat; panitumumab;
pazopanib; pegademase; pegaspargase; Pegfilgrastim; pemetrexed disodium; pentostatin;
pipobroman; plerixafor; plicamycin, mithramycin); porfimer sodium; pralatrexate; procarbazine; quinacrine; Rasburicase; raloxifene hydrochloride; Rituximab; romidepsin; romiplostim;
sargramostim; sargramostim; satraplatin; sorafenib; streptozocin; sunitinib maleate; tamoxifen; temozolomide; temsirolimus; teniposide; testolactone; thioguanine; thiotepa; topotecan;
toremifene; tositumomab; trastuzumab; tretinoin; uracil mustard; valrubicin; vinblastine;
vincristine; vinorelbine; vorinostat; and zoledronate. It will be clear to a person skilled in the art that, where appropriate, the other therapeutic ingredient(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.
The combinations referred to above may conveniently be presented for use in the form of a pharmaceutical composition and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent, carrier or excipient represent a further aspect of the invention. These combinations are of particular interest in respiratory diseases and are conveniently adapted for inhaled or intranasal delivery.
The individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical compositions. Preferably, the individual compounds will be administered simultaneously in a combined pharmaceutical composition. Appropriate doses of known therapeutic agents will be readily appreciated by those skilled in the art.
Pharmaceutical Compositions
While it is possible that, for use in therapy, a compound of Formula I, as well as salts, solvates and physiological functional derivatives thereof, may be administered as the raw chemical, it is possible to present the active ingredient as a pharmaceutical composition.
Accordingly, the invention further provides a pharmaceutical composition which comprises a compound of Formula I and salts, solvates and physiological functional derivatives thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients. The compounds of the Formula I and salts, solvates and physiological functional derivatives thereof, are as described above. The carrier(s), diluent(s) or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical composition including admixing a compound of the Formula I, or salts, solvates and physiological functional derivatives thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients.
Routes of Administration Pharmaceutical compositions of the present invention may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Such a unit may contain, for example, 5μg to 1 g, preferably 1 mg to 700 mg, more preferably 5 mg to 100 mg of a compound of the Formula I, depending on the condition being treated, the route of administration and the age, weight and condition of the patient. Such unit doses may therefore be administered more than once a day. Preferred unit dosage compositions are those containing a daily dose or sub-dose (for administration more than once a day), as herein above recited, or an appropriate fraction thereof, of an active ingredient. Furthermore, such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
Pharmaceutical compositions of the present invention may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, topical, inhaled, nasal, ocular, sublingual, subcutaneous, local or parenteral (including intravenous and intramuscular) route, and the like, all in unit dosage forms for administration. Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s). Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like.
In a further embodiment, the present invention provides a pharmaceutical composition adapted for administration by the oral route, for treating, for example, rheumatoid arthritis.
In a further embodiment, the present invention provides a pharmaceutical composition adapted for administration by the nasal route, for treating, for example, allergic rhinitis.
In a further embodiment, the present invention provides a pharmaceutical composition adapted for administration by the inhaled route, for treating, for example, asthma, Chronic Obstructive Pulmonary disease (COPD) or Acute Respiratory Distress Syndrome (ARDS).
In a further embodiment, the present invention provides a pharmaceutical composition adapted for administration by the ocular route, for treating, diseases of the eye, for example, conjunctivitis.
In a further embodiment, the present invention provides a pharmaceutical composition adapted for administration by the parenteral (including intravenous) route, for treating, for example, cancer. For parenteral administration, the pharmaceutical composition of the invention may be presented in unit-dose or multi-dose containers, e.g. injection liquids in predetermined amounts, for example in sealed vials and ampoules, and may also be stored in a freeze dried (lyophilized) condition requiring only the addition of sterile liquid carrier, e.g. water, prior to use.
Mixed with such pharmaceutically acceptable auxiliaries, e.g. as described in the standard reference, Gennaro, A.R. et al, Remington: The Science and Practice of Pharmacy (20th Edition., Lippincott Williams & Wilkins, 2000, see especially Part 5: Pharmaceutical Manufacturing), the active agent may be compressed into solid dosage units, such as pills, tablets, or be processed into capsules or suppositories. By means of pharmaceutically acceptable liquids the active agent can be applied as a fluid composition, e.g. as an injection preparation, in the form of a solution, suspension, emulsion, or as a spray, e.g. a nasal spray.
For making solid dosage units, the use of conventional additives such as fillers, colorants, polymeric binders and the like is contemplated. In general any pharmaceutically acceptable additive which does not interfere with the function of the active compounds can be used. Suitable carriers with which the active agent of the invention can be administered as solid compositions include lactose, starch, cellulose derivatives and the like, or mixtures thereof, used in suitable amounts. For parenteral administration, aqueous suspensions, isotonic saline solutions and sterile injectable solutions may be used, containing pharmaceutically acceptable dispersing agents and/or wetting agents, such as propylene glycol or butylene glycol.
Pharmaceutical compositions of the present invention which are adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of the present invention can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
Where appropriate, dosage unit compositions for oral administration can be microencapsulated. The formulation can also be prepared to prolong or sustain the release, for example, by coating or embedding particulate material in polymers, wax or the like.
The compounds of Formula I, and salts, solvates and physiological functional derivatives thereof, can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or
phosphatidylcholines.
The compounds of Formula I and salts, solvates and physiological functional derivatives thereof may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include
polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
Dosage forms for inhaled administration may conveniently be formulated as aerosols or dry powders.
For compositions suitable and/or adapted for inhaled administration, it is preferred that the compound or salt of Formula I is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronisation. The preferable particle size of the size-reduced (e.g. micronised) compound or salt or solvate is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
Aerosol formulations, e.g. for inhaled administration, can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or non-aqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
Where the dosage form comprises an aerosol dispenser, it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC). Suitable HFC propellants include 1 ,1 , 1,2,3,3,3- heptafluoropropane and 1 ,1 , 1,2-tetrafiuoroethane. The aerosol dosage forms can also take the form of a pump-atomiser. The pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol. Other excipient modifiers may also be incorporated to improve, for example, the stability and/or taste and/or fine particle mass characteristics (amount and/or profile) of the formulation.
For pharmaceutical compositions suitable and/or adapted for inhaled administration, it is preferred that the pharmaceutical composition is a dry powder inhalable composition. Such a composition can comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, the compound of Formula I or salt or solvate thereof (preferably in particle- size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L- leucine or another amino acid, and/or metals salts of stearic acid such as magnesium or calcium stearate. Preferably, the dry powder inhalable composition comprises a dry powder blend of lactose and the compound of Formula I or salt thereof. The lactose is preferably lactose hydrate e.g. lactose monohydrate and/or is preferably inhalation-grade and/or fine-grade lactose.
Preferably, the particle size of the lactose is defined by 90% or more (by weight or by volume) of the lactose particles being less than 1000 microns (micrometres) (e.g. 10-1000 microns e.g. 30- 1000 microns) in diameter, and/or 50% or more of the lactose particles being less than 500 microns (e.g. 10-500 microns) in diameter. More preferably, the particle size of the lactose is defined by 90% or more of the lactose particles being less than 300 microns (e.g. 10-300 microns e.g. 50-300 microns) in diameter, and/or 50% or more of the lactose particles being less than 100 microns in diameter. Optionally, the particle size of the lactose is defined by 90% or more of the lactose particles being less than 100-200 microns in diameter, and/or 50% or more of the lactose particles being less than 40-70 microns in diameter. It is preferable that about 3 to about 30% (e.g. about 10%) (by weight or by volume) of the particles are less than 50 microns or less than 20 microns in diameter. For example, without limitation, a suitable inhalation-grade lactose is E9334 lactose (10% fines) (Borculo Domo Ingredients, Hanzeplein 25, 8017 J D Zwolle, Netherlands). Optionally, in particular for dry powder inhalable compositions, a pharmaceutical composition for inhaled administration can be incorporated into a plurality of sealed dose containers (e.g. containing the dry powder composition) mounted longitudinally in a strip or ribbon inside a suitable inhalation device. The container is rupturable or peel-openable on demand and the dose of e.g. the dry powder composition can be administered by inhalation via the device such as the DISKUS® device(GlaxoSmithKline). Other dry powder inhalers are well known to those of ordinary skill in the art, and many such devices are commercially available, with representative devices including Aerolizer® (Novartis), Airmax™ (IV AX), ClickHaler® (Innovata Biomed), Diskhaler® (GlaxoSmithKline), Accuhaler (GlaxoSmithKline), Easyhaler® (Orion Pharma), Eclipse™ (Aventis), FlowCaps® (Hovione), Handihaler® (Boehringer
Ingelheim), Pulvinal® (Chiesi), Rotahaler® (GlaxoSmithKline), SkyeHaler™ or Certihaler™ (SkyePharma), Twisthaler (Schering-Plough), Turbuhaler® (AstraZeneca), Ultrahaler®
(Aventis), and the like.
Dosage forms for ocular administration may be formulated as solutions or suspensions with excipients suitable for ophthalmic use.
Dosage forms for nasal administration may conveniently be formulated as aerosols, solutions, drops, gels or dry powders.
Pharmaceutical compositions adapted for administration by inhalation include fine particle dusts or mists, which may be generated by means of various types of metered, dose pressurized aerosols, nebulizers or insufflators.
For pharmaceutical compositions suitable and/or adapted for intranasal administration, the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof may be formulated as a fluid formulation for delivery from a fluid dispenser. Such fluid dispensers may have, for example, a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser. Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid formulation, the doses being dispensable upon sequential pump actuations. The dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid formulation into the nasal cavity. A fluid dispenser of the aforementioned type is described and illustrated in WO-A-2005/044354, the entire content of which is hereby incorporated herein by reference. The dispenser has a housing which houses a fluid discharge device having a compression pump mounted on a container for containing a fluid formulation. The housing has at least one finger-operable side lever which is movable inwardly with respect to the housing to cam the container upwardly in the housing to cause the pump to compress and pump a metered dose of the formulation out of a pump stem through a nasal nozzle of the housing. A particularly preferred fluid dispenser is of the general type illustrated in FIGS. 30-40 of WO-A-2005/044354.
The invention further includes a pharmaceutical composition of a compound of
Formula I or pharmaceutically acceptable salts thereof, as hereinbefore described, in combination with packaging material suitable for said composition, said packaging material including instructions for the use of the composition for the use as hereinbefore described.
The following are examples of representative pharmaceutical dosage forms for the compounds of this invention:
Inj ectable Suspension (l.M.) mg/ml
Compound of Formula I 10
Methylcellulose 5.0
Tween 80 0.5
Benzyl alcohol 9.0
Benzalkonium chloride 1.0
Water for injection to a total volume of 1 ml
Tablet mg/tablet
Compound of Formula I 25
Microcrystalline Cellulose 415
Providone 14.0
Pregelatinized Starch 43.5
Magnesium Stearate 2.5
500
Capsule mg/capsule
Compound of Formula I 25
Lactose Powder 573.5
Magnesium Stearate 1.5
600 Aerosol Per canister
Compound of Formula I 24 mg
Lecithin, NF Liquid Concentrate 1.2 mg
Trichlorofluoromethane, NF 4.025 gm
Dichlorodifluoromethane, NF 12.15 gm
It will be appreciated that when the compound of the present invention is administered in combination with other therapeutic agents normally administered by the inhaled, intravenous, oral or intranasal route, that the resultant pharmaceutical composition may be administered by the same routes.
It should be understood that in addition to the ingredients particularly mentioned above, the compositions may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
A therapeutically effective amount of a compound of the present invention will depend upon a number of factors including, for example, the age and weight of the animal, the precise condition requiring treatment and its severity, the particular compound having Formula I, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. However, an effective amount of a compound of Formula I for the treatment of diseases or conditions associated with inappropriate Btk activity, will generally be in the range of 5 μg to 100 mg/kg body weight of recipient (mammal) per day and more usually in the range of 5 μg to 10 mg/kg body weight per day. This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same. An effective amount of a salt or solvate, thereof, may be determined as a proportion of the effective amount of the compound of Formula I per se.
In general parenteral administration requires lower dosages than other methods of administration which are more dependent upon absorption. However, a dosage for humans preferably contains 0.0001-25 mg of a compound of Formula I or pharmaceutically acceptable salts thereof per kg body weight. The desired dose may be presented as one dose or as multiple subdoses administered at appropriate intervals throughout the day, or, in case of female recipients, as doses to be administered at appropriate daily intervals throughout the menstrual cycle. The dosage as well as the regimen of administration may differ between a female and a male recipient.
General Synthesis
The 8-amino-imidazo[l,5-a]pyrazine, 4-amino-imidazo[l ,5-/] [l ,2,4]triazine, 4-amino- pyrazolo[3,4-cf]pyrimidine and 4-amino-pyrrolo[l,2-/] [l,2,4]triazine derivatives of the present invention can be prepared by methods well known in the art of organic chemistry. See, for example, J. March, 'Advanced Organic Chemistry' 4th Edition, John Wiley and Sons. During synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This is achieved by means of conventional protecting groups, such as those described in T.W. Greene and P.G.M. Wutts 'Protective Groups in
Organic Synthesis' 3rd Edition, John Wiley and Sons, 1999. The protective groups are optionally removed at a convenient subsequent stage using methods well known in the art.
The products of the reactions are optionally isolated and purified, if desired, using conventional techniques, but not limited to, filtration, distillation, crystallization,
chromatography and the like. Such materials are optionally characterized using conventional means, including physical constants and spectral data.
8-amino-imidazo[l ,5-a]pyrazine compounds of formula I, wherein Ri-Rx have the previously defined meanings, can be prepared by the general synthetic route shown in scheme I.
Scheme I
Figure imgf000040_0001
Figure imgf000040_0002
Figure imgf000040_0003
Reduction of 3-chloropyrazine-2-carbonitrile (II) can be accomplished by hydrogenation in the presence of a suitable catalyst system and solvent, for example Raney -Nickel to provide (3-chloropyrazin-2-yl)methanamine (III). This can then be reacted either with an appropriately amine protected amino acid where A5 is equivalent to CH and X is equivalent to OH. The reaction of HO(0)CC(R3,P )Rx can be carried out in a solvent such as DMF, THF or DCM in the presence of a base such as DIPEA, N-methylmorpholine, 4-DMAP or triethylamine and in the presence of a coupling reagent such as PyBOP, TBTU, EDCI or HATU to form N-((3- chloropyrazin-2-yl)methyl)amide (IV). Alternatively, if A5 is equivalent to nitrogen,
NH(R3,R4)RX can be activated with trichloromethyl chloroformate or phosgene to introduce COX, where X is equivalent to a leaving group. Subsequent reaction with (3-chloropyrazin-2- yl)methanamine (III) in a suitable solvent like DCM, EtOAc or DMF in the presence of a base such as DiPEA or triethylamine can give compounds of formula IV. Cyclisation chloropyrazine (IV) can be performed using condensation reagents like phosphorousoxy chloride under heating conditions to provide the 8-chloroimidazo[l,5-a]pyrazine derivatives V. Subsequent bromination can be accomplished using bromine or N-bromosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of formula VI. 8-Aminoimidazo[l,5- a]pyrazine derivatives (VII) can be prepared from compounds VI using ammonia(gas) in isopropanol at elevated temperature in a pressure vessel (>4 atm). Compounds of formula IX can be prepared from compounds of formula VII using an appropriate boronic acid or pinacol ester (VIII), in the presence of a suitable palladium catalyst system, for example
bis(diphenylphosphino)ferrocene palladium(II)chloride complex or
tefrate(triphenylphosphine)palladium(0) in the presence of an anorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Finally, cleaving the protective group of compounds with the formula IX give the unprotected amine or carboxylic acid which after functionalisation, using methods well known in the art, provided compounds of formula I.
The compounds like COXA5(R3,R4)RX are either commercially available or they can be readily prepared using methods well known to the skilled organic chemist, to introduce protecting groups like benzyloxycarbonyl or fert-butyloxycarbonyl.
Palladium catalysts and conditions to form either the pinacol esters or to couple the boronic acids or pinacol esters with the l-bromoimidazo[l,5-a]pyrazin-8-amine are well known to the skilled organic chemist - see, for example, Ei-ichi Negishi (Editor), Armin de Meijere (Associate Editor), Handbook of Organopalladium Chemistry for Organic Synthesis, John Wiley and Sons, 2002. Examples
The following examples are illustrative embodiments of the invention, not limiting the scope of the invention in any way. Reagents are commercially available or are prepared according to procedures in the literature.
Mass Spectrometry: Electron Spray spectra were recorded on the Applied
Biosy stems API- 165 single quad mass spectrometer in alternating positive and negative ion mode using Flow Injection. The mass range was 120-2000 Da. and scanned with a step rate of 0.2 Da. and the capillary voltage was set to 5000 V. N2gas was used for nebulisation.
LC-MS spectrometer (Waters) Detector: PDA (200-320 nm), Mass detector: ZQ and Eluent : A: acetonitrile with 0.05% trifluoroacetic acid , B: acetronitrile/water = 1/9 (v/v) with 0.05% trifluoroacetic acid.
Method A: LC-MS
Column Ascentis Express C18, 100x3.0mm, 2.7 μηι
A : H20 (0.1% TF A)
Mobile Phase B: MeCN (0.05% TFA)
Stop Time :5.0 min
Time (min) B%
0.00 10
3.50 99
4.99 99
Gradient 5.00 10
Sample injection 2 μ1
volume
Flow Rate 1.00 ml/min
Wavelength 220 nm
Oven Tern. 50 °C
MS polarity ESI POS
Method B: LC-MS
Column Ascentis Express C18, 50x2. lmm, 5 μηι
A : H20 (0.1% TFA)
Mobile Phase B: MeCN (0.05% TFA) Stop Time :2.0 min
Time (min) B%
0 10
0.8 99
1.99 99
Gradient 2.00 10
Sample injection
volume 2 μ1
Flow Rate 1.25 ml/min
Wavelength 220 nm
Oven Temp. 50 °C
MS polarity ESI POS
Method C:
Sample Info : Easy-Access Method: 'l-Short_TFA_Pos'
Method Info : B222 Column Agilent SBC (3.0x50 mm, 1.8 um); Flow 1.0 mL/min; solvent A: H2O-0.1% TFA;
solvent B: MeCN-0.1% TFA;
GRADIENT TABLE: 0 min: 10% B, 0.3 min: 10%B, 1.5min: 95% B, 2.70min: 95% B, 2.76 min: 10% B
stop time 3.60 min, PostTime 0.70 min.
Method D;
Sample Info : Easy-Access Method: 'l_Fast'
Method Info : A330 Column Agilent Zorbax SB-C18 (2.1x30 mm, 3.5 um); Flow 2.0 mL/min;
solvent A: H2O-0.1% TFA;
solvent B: MeCN-0.1% TFA;
GRADIENT TABLE: 0.01 min: 10% B, 1.01 min:95% B, 1.37 min:95% B, 1.38 min: 10% B, stop time 1.7min, PostTime=OFF The following abbreviations are used throughout the application with respect to chemical terminology: HATU 0-(7-Azabenzotriazol- 1 -yl)- 1 , 1 ,3,3-tetramethyluroniumhexafluoro phosphate
Cbz Benzyloxycarbonyl
D Deuterated hydrogen
DMF N,N-Dimethylformamide
DCM Dichloromethane
EA Ethyl acetate
EtOAc Ethyl acetate
DIPEA N,N-Diisopropylethylamine
THF Tetrahydrofuran
EtOH Ethanol
EDCI.HC1 l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
4-DMAP 4-Dimethylaminopyridine
PyBOP O-Benzotriazole-l-yl-oxy-trispyrrolidinophosphoniurnhexafluorophosphate
TBTU O-Benzotriazol- 1 -y 1-Ν,Ν,Ν* ,Ν' -tetramethy luronium tetrafluoroborate HBr Hydrogen bromide
HC1 Hydrogen chloride
HO Ac Acetic acid
POCl3 Phosphorous oxychloride
HPLC High Pressure Liquid Chromatography
UPLC Ultra Performance Liquid Chromatography
LiHMDS Lithium hexamethyldisilazide
MeOH Methanol
DCM Dichloromethane
n-BuLi n-Butyllithium
CO2 Carbondioxide
NaHCOj Sodium bicarbonate
K3PO4 Potassium phosphate
P(Cy)3 Tricyclohexylphosphine
Pd(OAc)2 Palladium(II) acetate
Na2S04 Sodium sulfate
Na2COs Sodium carbonate
DAST Diethylaminosulfur trifluoride
CS2CO3 Cesium carbonate Et20 Diethylether
Na2S2C>3 Sodium thiosulfate
Na2S204 Sodium hydrosulfite
NaCNBH3 Sodium cyanoborohydride
NH4CI Ammonium chloride
MgSC Magnesium sulfate
LiOH Lithium hydroxide
IPA Isopropylamine
TFA Trifluoroacetic acid
Cbz-Cl Benzylchloroformate
PE Petroleum ether
EA Ethyl acetate
NaHMDS Sodium hexamethyldisilazide
10% Pd/C 10% Palladium on carbon
TEA Triethylamine
CDI l, l'-Carbonyl diimidazole
DMI 1.3-Dimethy 1-2-imidazolidinone
NBS N-Bromosuccinimide
z-PrOH 2-Propanol
K2CO3 Potassium carbonate
Pd(dppf)Cl2 l, l'-Bis(diphenylphosphino)ferrocene palladium (II) chloride, complex withdichloromethane
Et3N Triethylamine
2-BuOH 2-Butanol
LCMS Liquid Chromatography / Mass Spectrometry
MeCN Acetonitrile
NH3 Ammonia
CD3I Trideuteromethyl iodide
CD3OD Tetradeuteromethanol
CH3I Iodomethane
CBr4 Carbon tetrabromide
Tris-HCl Tris(hydroxymethyl)aminomethane hydrochloride
MgCl2 Magnesium chloride NaN3 Sodium azide
DTT Dithiothreitol
DMSO Dimethyl sulfoxide
IMAP Immobilized Metal Ion Affinity-Based Fluorescence Polarization
ATP Adenosine triphosphate
MnCl2 Manganese(II) chloride
DMA Dimethylacetamide
IPA Isopropyl alcohol
TPP triphenylphosphine
DIAD Diisopropyl azodicarboxylate
DMB 2,4-dimethoxybenzyl
DCE Dichloroethane
DEAD Diethyl azodicarboxylate
ACN Acetonitrile
Ret. Time Retention Time
RT (rt) Room Temperature
Aq Aqueous
EtOH Ethanol
MPLC Medium Pressure Liquid Chromoatography
Xantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene
X-phos 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl
Intermediate 1
Figure imgf000046_0001
3-ethoxy-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2- vDbenzamide
Step 1; methyl 4-bromo-3-ethoxybenzoate
A suspension of methyl 4-bromo-3-hydroxybenzoate (1.0 g, 4.33 mmol) and powder potassium carbonate (0.658 g, 4.76 mmol) in DMF (4.33 ml) under N2 was treated with iodoethane (0.675 g, 4.33 mmol) via a syringe and the mixture stirred at rt for 2 h. The reaction was quenched with water and extracted with EtOAc (x2). The combined EtOAc layer was washed with water (x2) and brine, dried (MgSOzi) an& concentrated to afford a white solid. Trituration with ether / hexane followed by filtration afforded the title compound as a solid. XH NMR, 500 MHz, CDC13, δ 7.62 (5, J= 8.2 Hz, 1H), 7.55 (d, J = 1.7 Hz, 1H), 7.52 (dd, J = 8.2, 1.8 Hz, 1H), 4.19 (q, J= 6.9 Hz, 2H), 3.94 (s, 3H), 1.52 (t, J= 7.0 Hz, 3H) ppm.
Step 2: 4-bromo-3-ethoxybenzoic acid
A solution of the title compound from step 1, methyl 4-bromo-3-ethoxybenzoate (900 mg, 3.47 mmol) in THF (9.0 ml) was treated with LiOH (166 mg, 6.95 mmol) dissolved in water ( 4.5 ml) followed by MeOH ( 4.5 ml). The resulting mixture was then stirred at 45 °C for 2 h. The solvent was evaporated and the residue diluted with water. The pH was adjusted to 6 with 2 N HCl and the resulting white suspension washed with EtOAc (x2). The organic layer was dried (MgSOzi) and concentrated to afford the title compound as a solid. Calc'd m/z = 245.0, Found m/z = 247.0 (M+2).
Step 3; 4-bromo-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide
A suspension of the title compound from step 2, 4-bromo-3-ethoxybenzoic acid, (500 mg, 2.040 mmol), in DCM (5982 μΐ) under N2 was treated with DMF (55.3 μΐ, 0.714 mmol) followed by thionyl chloride (1489 μΐ, 20.40 mmol) via a syringe and the mixture stirred at 35 °C for 18 h. The solvent was evaporated and the residue co-evaporated with DCM and toluene (x2). The resulting residue was then diluted with acetonitrile ( 5982μ1) and treated with DMAP (324 mg, 2.65 mmol) and 4-(trifluoromethyl)pyridin-2-amine (364 mg, 2.244 mmol). The mixture was then stirred at rt for 3 h. The solvent was evaporated and the residue diluted with EtOAc and washed with water (x2). The combined organics was washed with brine, dried (MgSOzi & concentrated. Purification on the CombiFlash RF MPLC, on a 40 g column, eluting with 0 to 20 % EtOAc / Hexane (25 CV) affoded the title compound as a solid. Calc'd m/z = 389.1, Found m/z = 391.0 (M+2).
Step 4 : 3-ethoxy-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin- 2-yl)benzamide
A sealed vial containing the title compound from step 3, 4-bromo-3-ethoxy-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide (500 mg, 1.285 mmol), 4,4,4',4',5,5,5',5'-octamethyl- 2,2'-bi(l,3,2-dioxaborolane) (359 mg, 1.413 mmol), PdC12(dppf)- CH2Cl2Adduct (210 mg, 0.257 mmol) and K3PO4 (252 mg, 2.57 mmol) was evacuated and backfilled with N2. Dioxane (6424 μΐ) was then added via a syringe and the suspension evacuated again and backfilled with N2. The mixture was then stirred at rt for 5 min and then at 75 °C for 4.0 h (dark mixture). The mixture was diluted with EtOAc and filtered. The filtrate was concentrated to afford a brown oil. Purification on the CombiFlash RF MPLC, on a 40 g column, eluting with 0 to 20 % EtOAc / Hexane (40 CV) afforded the title compound, Intermediate 1. Calc'd m/z = 436.2, Found m/z = 437.1 (M+l).
Intermediate 2
Figure imgf000048_0001
3-ethoxy-5-fluoro-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide
Stepl; 3-Ethoxy-5-fluorobenzoic acid
SODIUM (6.54 g, 285 mmol) was dissolved in EtOH (150 ml) and concentrated to give a white solid. The solid was dissolved in DMSO (100 ml) and then added 3,5-difluorobenzoic acid (18g, 114 mmol). The mixture was stirred at 80 °C for 12 hours. The mixture was cooled to room temperature and then the mixture was acided to ph=5 with 2M HC1 , extracted with ethyl acetate (50 mLx3). The combined organic layers were washed with brine(20 mL), dried over anhydrous sodium sulfate, concentrated to afford the product 3-ethoxy-5-fluorobenzoic acid as a solid. 1H NMR (400MHz, CDC13) δ = 7.44 - 7.33 (m, 2H), 6.83 (d, J=10.2 Hz, 1H), 4.06 (q, J=7.0 Hz, 2H), 1.43 (t, J=7.0 Hz, 3H)
Step 2; 4-borono-3-ethoxy-5-fluorobenzoic acid
To a solution of 3-ethoxy-5-fluorobenzoic acid (4 g, 21.72 mmol) in THF (30 ml) was added LDA (32.6 ml, 65.2 mmol) dropwise at -78 °C under N2 atmosphere. The resultant solution was stirred for 15 min followed by slow addition of triisopropyl borate (4.90 g, 26.1 mmol). The mixture was stirred for 30 min and then hydrolyzed with 1M HC1. Extracted with EA (20 mL χ 3). The organic layer was washed with water(10 mL), brine(10 mL), dried over Na2S04, concentrated to afford the crude product, then the crude product was pruified by column chromatography on silica gel eluted with (THF: PE = 10% - 100%) to give 4-borono-3-ethoxy-5- fluorobenzoic acid as a solid. The compound structure was confirmed by HMBC. ¾ NMR
(400MHz, DMSO-de) = 8.40 (s, 1H), 7.21 (s, 1H), 7.14 (d, J=7.8 Hz, 1H), 4.03 (q, J=7.0 Hz, 2H), 1.28 (t, J=6.8 Hz, 3H)
Step 3; 3-Ethoxy-5-fluoro-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)benzoic acid
To a solution of 4-borono-3-ethoxy-5-fluorobenzoic acid (2.45g, 10.75 mmol) in PhCH3 (50 ml) was added 2,3-dimethylbutane-2,3-diol (1.397 g, 11.82 mmol) in one portion at room temperature under N2 atmosphere. The resultant solution was hitted to 120 °C and stirred at this temperature for 14h.The mixture was cooled to room temperature and concentrated to afford the crude product, then the crude product was pruified by column chromatography on silica gel eluted with (THF: PE = 10% - 50%) to give 3-ethoxy-5-fluoro-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)benzoic acid as a solid. *H NMR (CDC13) = 7.34 (d, J=8.2 Hz, 1H), 7.29 (s, 1H), 4.07 (q, J=6.7 Hz, 2H), 1.46 - 1.31 (m, 15H)
Step 4; 3-Ethoxy-5-fluoro-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide
To a solution of 3-ethoxy-5-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)benzoic acid (300 mg, 0.967 mmol) in anhydrous DCM (10 ml) was added OXALYL CHLORIDE (614 mg, 4.84 mmol) at 0 °C, then DMF (one drop) was added and the mixture was stirred at 20 °C for 1.5 hrs. The mixture was concentrated in vacuo, which then diluted with THF (6 ml), to the mixture was added 4-(trifluoromethyl)pyridin-2-amine (314 mg, 1.935 mmol) at 0 °C. The mixture was stirred at 80 °C for 16 hrs. After cooling to room temperature, the mixture was concentrated to give the crude product. The crude product was purified by column chromatography on silica gel eluted with (EA: PE = 1% ~ 50%) to give 3-ethoxy-5-fluoro-4-
(4,4,5, 5 etramethyl-l,3,2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide as a solid. The compound structure was confirmed by NOE. 1H NMR (400MHz, CHLOROFORM- d) = 8.71 - 8.66 (m, 2H), 8.47 (d, J=5.0 Hz, 1H), 7.31 (d, J=5.0 Hz, 1H), 7.18 (s, 1H), 7.12 (dd, J=0.9, 8.2 Hz, 1H), 4.10 (q, J=6.9 Hz, 2H), 1.46 - 1.38 (m, 15H)
Intermediate 3
Figure imgf000049_0001
3-fluoro-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2- vDbenzamide
3-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2- yl)benzamide was prepared in a similar manner as intermediate 1
Intermediate 4
Figure imgf000049_0002
3-methoxy-4-(4.4.5.5-tetramethyl-l .3.2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2- vDbenzamide
3-methoxy-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2- yl)benzamide was prepared in a similar procedure as intermediate 1
Intermediate 5
Figure imgf000050_0001
3-fluoro-5-methoxy-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide
3-fluoro-5-methoxy-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide was prepared in the similar method as intermediate 2
Intermediate 6
Figure imgf000050_0002
N-(4-methylpyridin-2-yl)-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)benzamide
N-(4-methylpyridin-2-yl)-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzamide was prepared in a similar method as intermediate 1
Intermediate 7
Figure imgf000050_0003
N-(4-cvclopropylpyridin-2-yl)-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)benzamide N-(4-cyclopropylpyridin-2-yl)-4-(4,4,5,5 etramethyl-l,3,2-dioxaborolan-2-yl)benzamide was prepared in a similar method as intermediate 1
Intermediate 8
Figure imgf000050_0004
N-(4-cvclopropylpyridin-2-yl)-3-ethoxy-5-fluoro-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2- vDbenzamide
N-(4-cyclopropylpyridin-2-yl)-3-ethoxy-5-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)benzamide was prepared in a similar method as intermediate 2
Intermediate 9
Figure imgf000051_0001
N-(4-cvanopyridin-2-yl)-3-ethoxy-5-fluoro-4-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2- vDbenzamide
N-(4-cyanopyridin-2-yl)-3-ethoxy-5-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)benzamide was prepared in a similar method as intermediate 2
Intermediate 10
Figure imgf000051_0002
3-ethoxy-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4.4.5.5-tetramethyl-1.3.2- dioxaborolan-2 -vDbenzamide
Step 1: 4-bromo-3-ethoxy-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)benzamide
A solution of 4-bromo-3-ethoxy benzoic acid (5.0 g, 20.40 mmol) in DMF (102 ml) was treated with HATU (8.53 g, 22.44 mmol) and the mixture stirred at rt for 15 min. l-methyl-5- (trifluoromethyl)-lH-pyrazol-3-amine (3.37 g, 20.40 mmol) was then added followed by DIEA (7.13 ml, 40.8 mmol) and the mixture stirred at rt for 15 h. The mixture was diluted with EtOAc and washed with water (x2). The organic layer was then washed with brine, dried (MgSOzi) m& concentrated to afford an oil. Purification on the CombiFlash RF MPLC on a 24 g column, eluting with 0 to 20 % EtOAc / Hexane afforded the desired product as an oil which later solidified. Calc'd m/z = 392.1, Found m/z = 394.0 (M+2).
Step 2: 3-ethoxy-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4.4.5.5-tetramethyl-1.3.2- dioxaborolan-2-yl)benzamide In a sealed round bottom flask containing the title compound from step 1, 4-bromo-3- ethoxy-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)benzamide, (1000 mg, 2.55 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (971 mg, 3.82 mmol), PdC12(dppf CH2CI2 adduct (208 mg, 0.255 mmol) and potassium acetate (751 mg, 7.65 mmol) was added 1,4-Dioxane (1.27E+04 μΐ) under a N2 atmosphere. The resulting suspension was then degassed (x3) and back filled with N2. The mixture was then stirred at 80 °C under N2 for 8 h h. The mixture was filtered and the filtrate concentrated. Purification on the CombiFlash RF, on a 80 g column, eluting with 0 to 15 % EtOac / Hexane (80 CV) afforded the title compound,
Intermediate 30 as an oil which later solidified. Calc'd m/z = 439.2, Found m/z = 440.2 (M+l).
Intermediate 11
Figure imgf000052_0001
3-ethoxy-5-fluoro-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4.4.5.5-tetramethyl-
1.3.2-dioxaborolan-2-yl)benzamide
3-ethoxy-5-fluoro-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)benzamide was prepared in a similar method as intermediate 2 and 10
Intermediate 12
Figure imgf000052_0002
2-chloro-5-ethoxy-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4.4.5.5-tetramethyl-
1.3.2-dioxaborolan-2-yl)benzamide
Step 1
Added a solution of Iodine (0.75g, 2.95mmol) and potassium iodide (0.55g, 3.31mmol) in water (5ml) dropwise to a solution of 2-Chloro-5 -hydroxy benzoic acid (0.5g, 2.90mmol) in ammonium hydroxide (cone, 4ml, 48mmol) then stirred 2 hours at room temperature. Reaction was diluted with water (5ml) acidified to pH 3 with cone. HC1, then extracted with EtOAc (50ml) and H20 (20ml). Organic layer was separated , dried over Na2SC>4, then filtered and evaporated solvent yielding 2-Chloro-5-hydroxy-4-iodobenzoic acid as a solid LCMS : C7H4C1I03 MH Found [M+H]+: 298.84 RT = 1.26 min.
Step 2
Added Thionyl Chloride (0.5ml, 6.85mmol) dropwise to a solution of 2-Chloro-5-Hydroxy-4- iodobenzoic acid (0.6g, 2.01mmol) in MeOH (10ml) at 0°C , then stirred overnight at room temperature. The solvent was evaporated and residue extracted with EtOAc (100ml), washed with saturated NaHCCb (40ml),dried over Na2SC>4 then filtered and solvent evaporated yielding the title compound Methyl -2-Chloro-5-hydroxy-4-iodobenzoate as a solid.
Step 3
Added Iodoethane (0.3ml, 3.75mmol) to a suspension of Methyl -2-Chloro-5-hydroxy-4- iodobenzoate (0.9g,2.87mmol) and potassium carbonate (0.4g, 2.89mmol) in DMF (4ml) then stirrede 2 hours at room temperature. Extracted with EtOAc (100ml) and H20 (40ml), separated organic layer, dried over Na2S04, then filtered and evaporated solvent. Purified by
chromatography (40g silica gel,20% ethylacetate in Hexanes) yielding title compound Methyl-2- Chloro-5-Ethoxy-4-Iodobenzoate as a solid.
LCMS : C10H10C1IO3, Found [M+H]+ 340.89, RT = 1.56 min.
Step 4
Added Lithium Hydroxide monohydrate (0.2g , 4.77mmol) to a solution of Methyl-2-Chloro-5- Ethoxy-4-Iodobenzoate (0.3g , 0.846mmol) in Methanol :THF:H20 (7ml, 3:3: 1) then stirred at 40°C overnight. Reaction was cooled to room temperature , the solvent was evaporated ,and residue was diluted with H20 (20ml) ,acidified with cone HC1 to pH 3 ,precipitated white solid was filtered and washed with H20 (10ml). Solid was dissolved in THF (20ml) and solvent evaporated yielding title compound 2-Chloro-5-Ethoxy-4-Iodobenzoic acid.
Step 5
Added oxalyl chloride (0.3ml, 3.16mmol) to solution of 2-Chloro-5-Ethoxy-4-Iodobenzoic acid (0.3g , l. lOlmmol) and DMF (0.05ml , 3.16mmol) in MeCl2 ( 10ml) then stirred at room temperature for 1 hour.
Solvent was evaporated, yielding the title compound 2-Chloro-5-Ethoxy-4-Iodobenzoyl chloride as a solid.
Step 6
Added DMAP ( 3mg , 0.025mmol) and N-Ethyl-N-isopropylpropan-2-amine (0.2ml,
1.148mmol) to mixture of 2-Chloro-5-Ethoxy-4-Iodobenzoyl chloride (0.27g, 0.783mmol) and l-Methyl-5-trifluoromethyl-lH-Pyrazol-3-amine (150mg, 0.908mmol) in THF (3ml) then stirred at room temperature for 2 hours. Extracted with EtOAc (50ml) and H20 (20ml), then separated organic layer,dried over Na2SC>4, then filtered and evaporated solvent.Purified residue by chromatography (24g silica gel, 15% ethylacetate in Hexanes) yielding title compound 2-Chloro- 5-Ethoxy-4-Iodo-N-(l-Methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl) benzamide as a solid. LCMS : C14H12C1F3IN302, Found [M+H]+ 473.96, RT = 1.53 min.
Step 7
Added DMSO (1ml) to a mixture of Bis(Pinacolato)Diboron (160mg, 0.630mmol), potassium acetate (80mg , 0.815mmol); [1,1 -Bis(Diphenylphosphino)ferrocene]Dichloropalladium (11) (20mg,0.027mmol) and 2-Chloro-5-Ethoxy-4-Iodo-N-(l-Methyl-5-(trifluoromethyl)-lH- pyrazol-3-yl) benzamide (200mg,0.422mmol) ,degassed resultant solution, then stirred at 80°C for 4 hours. Cooled to room temperature,then extracted with EtOAc (50ml) and H20 (20ml).
Separated organic layer dried over Na2SC>4, then filtered and evaporated solvent.Purified residue by chromatography (24g silica gel,45% ethylacetate in Hexanes) yielding title compound 2- chloro-5-ethoxy-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4,4,5-trimethyl-l,3,2- dioxaborolan-2-yl)benzamide ) as a solid.
LCMS : C14H12C1F3N302, Found [M+H]+ 474.12, RT = 1.55 min.
Intermediate 13
Figure imgf000054_0001
5-ethoxy-2-fluoro-N-(l-methyl-5-(trifluoromethyl)-lH-pyrazol-3-yl)-4-(4.4.5.5-tetramethyl-
1.3.2-dioxaborolan-2-yl)benzamide
The title compound was prepared in a similar method as Intermediate 12
Intermediate 14
Figure imgf000054_0002
Methyl 1.3-dioxooctahvdroindolizine-6-carboxylate
Step 1. Preparation of (c^)-dimethyl piperidine-2.5-dicarboxylate
To dimethyl pyridine-2,5-dicarboxylate (50 g) was added AcOH (500 mL) and catalyst 5%
RI1/AI2O3 (5 g). The reaction mixture was shaked under H2 (100 psi) at 25 oC for 16 h. The catalyst was removed by filtartion. The AcOH was removed under vacuum and the residue was used without furtherpuirification. Step 2. Preparation of (c^)-dimethyl l-(4-methoxy-3.4-dioxobutanoyl)piperidine-2.5- dicarboxylate
(cw)-Dimethyl piperidine-2,5-dicarboxylate acetate (147 g, 563 mmol) was dissolved in CH2CI2 (1407 ml) in a 3L three-neck round bottom flask with a machine stir, an temperature probe and an additional funnel. The flask was cooled by a ice-water bath. Methyl malonyl chloride (91 ml, 844 mmol) was added slowly via an additional funnel in a speed that the internal temperature was maintained at below 10 °C. After the additional was completed, the reaction mixture was stirred in the ice-water bath for 3 hours. Sat NaHCC was added slowly with efficient stirring. The pH value of the reaction mixture was checked by pH paper. After 500 mL sat NaHC03 was added, solid NaHC03 was added into the solution mixture with efficient stirring, until the pH value of the reaction mixture reaches 8 to 9. The DCM layer was separated and the aquous layer was extracted with DCM (300 ml) twice. The organic laayers were combined and concentrated. It was dried under vacuum with stirring to give the title compound as a oil. It was used without further purification.
Step 3. Preparation of sodium 2.6-bis(methoxycarbonyl)-3-oxo-3.5.6.7.8.8a-hexahydroindolizin- 1-olate
To (cw)-dimethyl l-(3-methoxy-3-oxopropanoyl)piperidine-2,5-dicarboxylate (136.75 g, 454 mmol) in a 2 L round bottom flask was added MeOH (648 ml). It was cooled by a ice-water bath. NaOMe (85 ml, 454 mmol) was added dropwise via an additional funnel over 30 minutes. The internal temperature was kept between 6 to 8 °C. White solid appeared during this time period. MTBE (800 mL) was added over 45 min via an additional funnel. The slurry was stirred at 0 °C for 2 h and filtered, washed with MTBE twice, and dried under vacuum for 3 days to give the title compound as a solid.
Step 4. Preparation of Dimethyl 1.3-dioxooctahvdroindolizine-2.6-dicarboxylate
Sodium 2,6-bis(methoxycarbonyl)-3-oxo-3,5,6,7,8,8a-hexahydroindolizin-l-olate (32.8 g, 113 mmol) was suspended in DCM (282 ml). HC1 (282 ml, 563 mmol) was added. The suspensition was stirred at 25 °C for 20 min until all solid dissolved. The mixture was transfered into a separate funnel and the organic layer was separated. The aquous layer was extracted with DCM (150 mL) twice. The organic layers were combined, concentrated, and dried under vacuum for 3 hours to give the title compound as a solid.
Step 5. Preparation of Methyl 1.3-dioxooctahvdroindolizine-6-carboxylate
To a 500 mL round bottom flask was added dimethyl l-hydroxy-3-oxo-3,5, 6,7, 8,8a- hexahydroindolizine-2,6-dicarboxylate (5.26 g, 19.54 mmol), acetic acid (98 ml) and water (0.704 ml, 39.1 mmol). The reaction was stirred at 70 °C for 2.5 hours. The acetic acid was removed at 25 oC under vacuum. It was azotropied with Toluene (100 mL) twice to give the title compound as a oil which was stored at -20 °C. MS: 212.13 [M+H]+
Intermediate 15
Figure imgf000056_0001
(fra» ,)- . -difluoro-3'-oxohexahvdro- H-spiro[cvclopropane-1.2'-indolizinel-6'-carboxylic acid
Step 1. Preparation of Methyl .3'-dioxohexahvdro- H-spirorcvclopropane-1.2'-indolizinel-6'- carboxylate
To a 500 mL round bottom flask was added tert-Butanol (191 ml), K2C03 (22.31 g, 161 mmol) and 1 ,2-dibromomethane (11.13 ml, 129 mmol). The temperature of the reaction mixture was raised to 70 °C (a temperature probe was placed in the reaction mixture, heating mantle temperature is 74 °C). Methyl l,3-dioxooctahydroindolizine-6-carboxylate (6.82 g, 32.3 mmol) was dissolved in DMF (23.92 ml) and it was added into the reaction solution over a period of 24 h via a syringe pump at 70 °C. The reaction mixture was concentrated to almost dry. TheDMF was removed as much as possible. Then 200 mL water and 200 mL DCM was added. The aqueous layer was separated and extracted with DCM (100 ml) once. The organic layers were combined and then washed with water (100 mL) three times, dried over anhydrous Na2SC>4, concentared and purified by S1O2 chromatography (Hexane/EtOAc, 20% to 100%)to give the title compounds as oil. MS: 238.07 [M+H]+
Step 2. Preparation of Methyl . -difluoro-3'-oxohexahvdro- H-spiro[cvclopropane-1.2'- indolizinel -6'-carboxylate
To a 40 mL pressure vial with pressure release cap was added Methyl Γ,3'- dioxohexahydro- H-spiro[cyclopropane-l,2'-indolizine]-6'-carboxylate (350 mg, 1.475 mmol) and bis(2-methoxylaminosulfurtrifluoride(6337 μΐ, 14.75 mmol) . The reaction was stirred at 90 °C for 8h. More DeoxoFluor (2 ml) was added and the reaction was stirred at 90 °C for another 16 hours. The reaction was cooled down and sat NaHCCb was added carefully until no bubble cames out. The product ws extracted with EtOAc three times. The organic layers were combined, washed with brine once, dried over anhydrous Na2S04, filtered, and concentrated. The product was purified by S1O2 chromatography (Hexane/EtOAc, 0% to 100%) to give the title compound. MS: 260.14 [M+H]+
Step 3. Preparation of (fra» ,)-r.r-difluoro-3'-oxohexahvdro-rH-spiro[cvclopropane-1.2l- indolizinel-6'-carboxylic acid
To a 40 mL round bottom flask was added methyl r,l'-difluoro-3'- oxohexahydro-rH-spiro[cyclopropane-l,2'-indolizine]-6'-carboxylate (235 mg, 0.906 mmol), MeOH (4532 μΐ), and NaOMe (196 mg, 3.63 mmol). Then water (32.7 μΐ, 1.813 mmol) was added. The solution was stirred at 70 °C for 24 hours. More NaOMe (196 mg, 3.63 mmol) and water (32.7 μΐ, 1.813 mmol) were added at 70 °C. The reaction was stirred at 70 °C for another 24h. After the reaction was cooled down, AcOH (about 5 equive) was added until the solution is slightly acidic. The solid was collected by filtration and washed with small amount of water twice. This is the first batch of the title compound. The filtrate was concentrate to about 15 mL and applied to the CI 8 column. The product was purified by CI 8 column (CH3CN in water with 0.1% TFA: 0% to 30%) to give the batch of the title compound. MS: 246.11 [M+H]+
I
Figure imgf000057_0001
(fra» ,)-6'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3-yl)tetrahydro-rH- spirorcvclopropane-1.2'-indolizinl-3'(5'H)-one
Step 1. Preparation of (fra» ,)-6'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5- alpyrazin-3-yl)-r-hvdroxytetrahvdro-rH-spiro[cvclopropane-1.2'-indolizinl-3'(5'H)-one
To a 100 mL pressure vial with pressure release cap was added 6'-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)tetrahydro-rH-spiro[cyclopropane-l,2'- indolizine]-r,3'(5'H)-dione (550 mg, 1.018 mmol), EtOH (1.02E+04 μΐ), and sodium
borohydride (77 mg, 2.035 mmol). The reaction was stirred at 25 °C for 2 hours. Sat NaHC03 was added until no bubble comes out. Then the reaction was extracted with DCM (50 mL x3). The organic layers were combined and concentrated. The product was purified by S1O2 chromatography (EtOAc/EtOH (3: 1) in hexane with 1% NH3 H20, 0% to 100%) to give the title compound. MS: 541.84, 543.84 [M+H]+ Step 2. Preparation of 0-(fra».s,)-6'-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5- alpyrazin-3-yl)-3'-oxohexahvdro- H-spiro[cvclopropane-1.2'-indolizinl- -yl) lH-imidazole-1- carbothioate
To a 8 mL pressure vial with pressure release cap was added (ira«s,)-6'-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)- -hydroxytetrahydro- H- spiro[cyclopropane-l,2'-indolizin]-3'(5'H)-one (300 mg, 0.553 mmol), 1,1'- thiocarbonyldiimidazole (246 mg, 1.383 mmol) and THF (5531 μΐ). The reaction was stirred at reflux for 3 hours. LCMS indicated about 40% SM remain. More thiocarbonyldiimidazole(100 mg) was added and the reaction was stirred at reflux for another 2 hours. Saturated NaHCC (50 mL) and DCM (50 mL) were added. The organic layer was separated and concentrated. The product was purified by S1O2 chromatography (EtOAc/EtOH (3: 1) in hexane with 1% NH3 H20,
0% to 100%) to give the title compound. MS: 652.12, 654.12 [M+H]+
Step 3. Preparation of (fra» ,)-6'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5- alpyrazin-3-yl)tetrahvdro- H-spirorcvclopropane-1.2'-indolizinl-3'(5'H)-one
To a 40 mL pressure vial with pressure release cap was added tri-n-butyltinhydride (246 μΐ, 0.919 mmol) and toluene (2043 μΐ). The solution was heated to 115 °C. 0-((frara)-6'-(l- bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-3'-oxohexahydro- H- spiro[cyclopropane-l,2'-indolizin]- -yl) lH-imidazole-l-carbothioate (300 mg, 0.460 mmol) was dissoved in toluene (4086 μΐ) and added into the solution dropwise and slowly. The reaction was stirred at 115 °C for 4 hours. The solvent was concentrated and the residue was purified by C18 column (Gilson, CH3CN in water with 0.1% TFA: 0% to 90%) to give the title compound. MS: 526.07, 528.07 [M+H]+
Intermediate 17
Figure imgf000058_0001
trans-6'-(8-chloroimidazori.5-alpyrazin-3-yl)- -methoxytetrahvdro- H-spirorcvclopropane-
1.2'-indolizinl-3'(5'H)-one
Step 1. trans-N-((3-chloropyrazin-2-yl)methyl)- .3'-dioxohexahvdro- H-spiro[cvclopropane- 1.2'-indolizinel -6'-carboxamide To the heterogeneous potassium superoxide (468 mg, 6.58 mmol) and trans-N-((3- chloropyrazin-2-yl)methyl)-3'-oxo- -(2-tosylhydrazono)hexahydro- H-spiro[cyclopropane-l,2'- indolizine]-6'-carboxamide (340 mg, 0.658 mmol) solution in acetonitrile was added a solution of 2-nitrobenzene-l-sulfonyl chloride (437 mg, 1.973 mmol) in acetonitrile (6 mL) at -42 °C under nitrogen atmosphere with good stirring for 20 hours. The reaction mixture was then filtered, washed with acetonitrile and DCM and concentrated under vacuum to give a crude residue, which was column purified on silica gel eluting with (30-70% EtOAc/Hexanes) solvent system to afford trans-N-((3-chloropyrazin-2-yl)methyl)- ,3'-dioxohexahydro- H- spiro[cyclopropane-l,2'-indolizine]-6'-carboxamide. [M+H]+ = 349.07, Rt = 1.02 mins method B.
Step 2. trans-6'-(8-chloroimidazo[1.5-alpyrazin-3-yl)tetrahvdro- H-spiro[cvclopropane-1.2'- indolizinel- .3'(5'H)-dione
trans-N-((3-chloropyrazin-2-yl)methyl)- ,3'-dioxohexahydro- H-spiro[cyclopropane- l,2'-indolizine]-6'-carboxamide (140 mg, 0.401 mmol) was dissolved in acetonitrile (6 mL) and DMF (0.2 mL)and cooled to 0 °C. Phosphoryl trichloride (0.128 ml, 1.405 mmol) was added slowly at 0 °C. The ice bath was removed and reaction mixture was stirred at 40 °C for 30 mins under a stream of nitrogen. Upon completion, the reaction mixture was cooled to 0 °C and quenched with saturated NaHCC , and extracted with DCM (3X 15 ml). The combined organic phases were concentrated to dryness and the residue was column purified (3% MeOH/DCM) to afford trans-6'-(8-chloroimidazo[l,5-a]pyrazin-3-yl)tetrahydro- H-spiro[cyclopropane-l,2'- indolizine]-r,3'(5'H)-dione. [M+H]+ = 331.04, Rt = 1.16 mins,method B.
Step 3. trans-6'-(8-chloroimidazo[1.5-alpyrazin-3-yl)- -hvdroxytetrahydro- H- spiro[cvclopropane-1.2'-indolizinl-3'(5'H)-one
To a 25 mL round bottom flask was added trans-6'-(8-chloroimidazo[l,5-a]pyrazin-3- yl)tetrahydro- H-spiro[cyclopropane-l,2'-indolizine]- ,3'(5'H)-dione (100 mg, 0.302 mmol), Ethanol (6 ml), and sodium tetrahydroborate (28.6 mg, 0.756 mmol). The reaction was stirred at RT for 1 hour. The mixture was concentrated under vacuum to give a crude residue, which was column purified on silica gel eluting with (30-60% EtOAc/Hexane) to afford trans-6'-(8- chloroinddazo[l,5-a]pyrazin-3-yl)- -hydroxytetrahydro-lH-spiro[cyclopropane-l,2'-indolizin]- 3'(5'H)-one. [M+H]+ = 333.08, Rt = 1.05 mins, method B.
Step 4. trans-6'-(8-chloroimidazo[1.5-alpyrazin-3-yl)- -methoxytetrahydro- H- spiro[cvclopropane-1.2'-indolizinl-3'(5'H)-one To trans-6'-(8-chloroimidazo[l,5-a]pyrazin-3-yl)- -hydroxytetrahydro- H-spiro[cyclopropane- l,2'-indolizin]-3'(5'H)-one (50 mg, 0.150 mmol) dissolved in DMF (2 mL) at 0 °C was added sodium hydride (10.82 mg, 0.451 mmol) and stirred for 30 minutes under constant flow of nitrogen. lodomethane (25.6 mg, 0.180 mmol) was added and continued to stir for overnight. The reaction was quenched with saturated NH4C1 and extracted with EtOAc (3x5 mL). The combined organic phase was washed with saturated NaCl (lx 5 mL) and concentared under vacuum to give a crude product trans-6'-(8-chloroimidazo[l,5-a]pyrazin-3-yl)- - methoxytetrahydro- H-spiro[cyclopropane-l,2'-indolizin]-3'(5'H)-one. [M+H]+ = 347.02, Rt = 1.22 mins, method B.
Figure imgf000060_0001
(5aS.8RJ laS)-8-(8-amino-l-bromoimidazori.5-alpyrazin-3-yl)octahvdro-lH-pyridori.2- al pyrrolo Γ 1.2-dl pyrazin- 11 11 aHVone
Step 1. Dimethyl pyridine-2.5-dicarboxylate
SOCI2 (855 g, 7.2 mol) was added dropwise into the solution of pyridine-2,5-dicarboxylate (500 g, 3.0 mol) in MeOH (5 L) at room temperature. The mixture was stirred at 70 °C overnight.
After cooling, the mixture was evaporated, and the residue was added EA (5 L), followed by
Na2C03 (sat.) until PH>7. The mixture was separated and the aqueous layer was extracted with
EA (1 L*3). The combined organic layer was washed with brine, dried over Na2SC>4, filtered and concentrated to give dimethyl pyridine-2,5-dicarboxylate. The product was used for next step without further purification.
Step 2. Methyl 6-(hydroxymethyl)nicotinate
The mixture of dimethyl pyridine-2,5-dicarboxylate (200 g, 1.03 mol) in MeOH (2400 ml) and THF (2200 ml) was added CaCl2 (455 g, 4.10mol) and NaBH4 (97 g, 2.56 mol) below -10 °C. The mixture was stirred below 0 °C for 3 hours. The mixture was added NH4C1 (15%, 2000 ml), and the temperature maintain below 5 °C. Then, the solution was extracted with EA (500 ml x 3). The combined organic layers were dried over Na2SC>4, filtered and concentrated to give the crude product. The crude product was washed with PE: DCM=5: 1 to give methyl 6-(hydroxymethyl) nicotinate as a solid. 1H NMR: 400 MHz CDC13: δ 8.052 (s, 1H), 8.315-8.290 (m, 1H),
7.381-7.359 (m, 1H), 4.845 (s, 2H), 3.969 (s, 3H) ppm. Step 3. Methyl 6-(acetoxymethyl)nicotinate
The mixture of methyl 6-(hydroxymethyl) nicotinate (620 g, 3.7 mol) in DCM (6000 ml) was added Et3N (937 g, 9.3 mol), DMAP (31 g, 5%) and Ac20 (571 g, 5.5mol). The mixture was stirred at room temperature for 3 hours. The mixture was added H20 (5000 ml), separated and the aqueous layer was extracted with DCM (1500 ml*3). The combined organic layers were dried over Na2SC>4 and concentrated to give the crude product. The crude product was washed with PE: EA=5: 1 to give methyl 6-(acetoxymethyl)piperidine-3-carboxylate as a solid. 1H
NMR: 400 MHz CDC13 δ: 9.209-9.205 (s, 1H), 8.347-8.322 (m, 1H), 7.469-7.449 (d, J=8 Hz, 1H), 5.305 (s, 2H), 3.984 (s, 3H), 2.218 (s, 3H) , 3.565-3.465 (m,2H) , 2.135-1.878 (m,2H) ppm.
Step 4. Methyl 6-(acetoxymethyl)piperidine-3-carboxylate
NaBHsCN (136 g, 2.15 mol) was added in portions to the solution of methyl 6- (acetoxymethyl)piperidine-3-carboxylate (100 g, 0.48 mol) in CH3COOH (500 ml) at 0 °C for 2 hours and the mixture was stirred at 30 °C for 2 hours, quenched with H20 (120 ml), evaporated in vacuum to give methyl 6-(acetoxymethyl)piperidine-3-carboxylate as an oil, which was directly used for next step without further purification. LCMS:(M+1=216.2).
Step 5. trans- 1 -benzyl 3-methyl 6-(acetoxymethyl)piperidine-1.3-dicarboxylate
NaHCCb (67.2 g, 0.8 mol) was added very slowly to the mixture of methyl 6- (acetoxymethyl)piperidine-3-carboxylate (133 g, 0.16 mol) in THF (300 ml) and H20 (300 ml), followed by Cbz-Cl (270 g, 0.16 mol). The mixture was stirred at room temperature for 18 hours. The mixture was filtered, extracted with EA (600 ml*3). The combined organic layers were dried over Na2SC>4, filtered and concentrated to give the crude product. Chromatograph column (EA in PE from 0% to 33%) gave colorless oil. Prep-HPLC gave trans- 1 -benzyl 3-methyl 6- (acetoxymethyl)piperidine-l,3-dicarboxylate. 1H NMR: 400 MHz CDC13 δ: 7.377-7.323 (m, 5H), 5.188-5.080 (m, 2H), 4.627-4.570 (m, 1H), 4.325-4.284 (m, 2H), 4.235-4.137 (s, 1H), 3.700 (s, 3H), 3.066-2.965 (m, 1H), 2.557-2.400 (m, 1H), 2.018-1.917 (m, 4H), 1.772-1.648 (m, 4H) ppm.
Step 6. (3 S.6R)-1 -benzyl 3-methyl 6-(hvdroxymethyl)piperidine-1.3-dicarboxylate and (3R.6S)- 1 -benzyl 3-methyl 6-(hvdroxymethyl)piperidine-1.3-dicarboxylate
The mixture of trans-1 -benzyl 3-methyl 6-(acetoxymethyl)piperidine-l,3-dicarboxylate (25 g, 0.072 mmol), NaHC03 (54 g, 0.64 mol) and K2C03 (4.94 g, 35.8 mmol) in MeOH (100 ml) was was stirred at room temperature for 1 hour, filtered, diluted with H20 (100 ml), extracted with DCM (100 mlx3). The combined organic layers were washed with H20 (100 ml), brine (100 ml), evaporated in vacuo to get crude product. Chromatograph column (EA in PE from 0% to 33%) gave colorless oil trans-1 -benzyl 3-methyl 6-(hydroxymethyl)piperidine-l,3-dicarboxylate. The racemic mixture was further separated by SFC to obtain (3S,6R)-l-benzyl 3-methyl 6- (hydroxymethyl)piperidine-l,3-dicarboxylate ( retention time = 4.254 min) and (3R,6S)-1 -benzyl 3-methyl 6-(hydroxymethyl)piperidine-l,3-dicarboxylate (retention time = 4.607 min). SFC separation condition: Instrument: SFC Thar 200. Column: Chiralpak AD-H 250x50mmI.D., 10um.Mobile phase: A for C02 and B for ETOH (0.1%NH3H2O). Gradient: B 40% Flow rate: 200mL /min. Back pressure: lOObar. Column temperature: 35 °C. Wavelength: 220nm. 1H NMR: 400 MHz CDC13 δ: 7.379-7.311 (m, 5H), 5.227-5.195 (d, J = 12.8 Hz, 1H), 5.121-5.089 (d, J = 12.8 Hz, 1H), 4.248-4.169 (m, 2H), 3.846-3.818 (m, 1H), 3.721-3.679 (m, 1H), 3.625 (s, 3H), 3.450-3.322 (m, 1H), 2.617-2.594 (m, 1H), 1.999-1.899 (m, 1H), 1.855-1.831 (m, 2H), 1.616-1.582 (m, 1H) ppm ppm.
Step 7: (3R,6S)-\ -benzyl 3-methyl 6-(((ter/-butyldiphenylsilyl)oxy)methyl)piperidine-1.3- dicarboxylate
To a solution of (3R,6S)-\ -benzyl 3-methyl 6-(hydroxymethyl)piperidine-l,3-dicarboxylate (2.00 g, 6.51 mmol), imidazole (0.532 g, 7.81 mmol) in DMF (10 ml) was added TBDPS-C1 (2.0 ml, 7.81 mmol). It was stirred at rt for 2 h. Precipitates were found. The reaction mixture was diluted with ethyl acetate, washed with water three times, then brine once. The organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified by ISCO (Gold 120g, 0- 50% ethyl acetate in hexane ) to give the title compound as a solid. LC-MS: C32H39N05Si, calc. = 546.27; found = 546.29 (M+H)+.
Step 8: (3i?.6^)-l-((benzyloxy)carbonyl)-6-(((ter/-butyldiphenylsilyl)oxy)methyl)piperidine-3- carboxylic acid
To a solution of (3R,6S)-\ -benzyl 3-methyl 6-(((/er/-butyldiphenylsilyl)oxy)methyl)piperidine- 1,3-dicarboxylate (3.40 g, 6.23 mmol) in THF (40 ml), MeOH (40.0 ml) and water (40.0 ml) was added LiOH (5 M, 8 ml, 40.0 mmol) slowly. It was stirred at rt for 3 h and acidified by 1 M HCl (about 40 mL) to adjust pH to 5. The mixture was extracted with ethyl acetate (3x100 mL). The combined organic layers were washed with brine (100 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was used without purification. LC-MS: C31H37NO5S1, calc. = 532.25; found = 532.34 (M+H)+.
Step 9: (2£.5i<?)-benzyl 2-(((ter/-butyldiphenylsilyl)oxy)methyl)-5-(((3-chloropyrazin-2- yl)methyl)carbamoyl)piperidine-l-carboxylate To a mixture of (3i?,6»S)-l-((benzyloxy)carbonyl)-6-(((fer/- butyldiphenylsilyl)oxy)methyl)piperidine-3-carboxylic acid (3.3 g, 6.21 mmol), (3- chloropyrazin-2-yl)methanamine bis-hydrocloride salt (1.478 g, 6.83 mmol) and HATU (2.83 g, 7.45 mmol) in DMF (20 ml) was added DIEA (3.25 ml, 18.62 mmol). The mixture was stirred at rt for 1 h. Most solvent was removed under reduced pressure and the residue was diluted with ethyl acetate (100 mL), washed with water (3x50 mL), then brine (100 mL). The organic layer was seperated and dried over sodium sulfate, filtered and concentrated under redueced pressure. The residue was purified by ISCO (Gold 80g, 0-100% EtOAc/EtOH (3: 1) in hexane) to give the title compound as a solid. LC-MS: C36H41CIN4O4S1, calc. = 656.27; found = 656.41 (M+H)+. Step 10: (2ff.5i?)-benzyl 2-(((ter -butyldiphenylsilyl)oxy)methyl)-5-(8-chloroimidazo[1.5- al pyrazin-3-yl)piperidine- 1 -carboxylate
To a mixture of (25',5i?)-benzyl 2-(((teri-butyldiphenylsilyl)oxy)methyl)-5-(((3-chloropyrazin-2- y l)methyl)carbamoyl)piperi dine- 1 -carboxylate (2.2 g, 3.35 mmol) and sodium carbonate (2.84 g,
26.8 mmol) in acetonitrile (12 mL) and DMF (12.00 mL) at 0 °C was added POCl3 (1 mL, 10.73 mmol) dropwise. It was warmed to 45 °C and stirred for 1 h. The mixture was diluted withlOO mL of ethyl acetate and washed with water (3x50 mL). The combined aqueous layers were extracted with ethyl acetate (100 mL). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by ISCO (Gold 40g, 0-100% EtOAc/EtOH (3: 1) in hexane) to give the title compound. LC-MS:
C36H39CIN4O3S1, calc. = 639.26; found = 656.41 (M+H)+. (
Step 11 : (2S,5R)-benzy\ 5-(l-bromo-8-chloroimidazori.5-alpyrazin-3-yl)-2-(((ter^- butyldiphenylsilyl)oxy)methyl)piperidine- 1 -carboxylate
To a solution of (2S,5i<!)-benzyl 2-(((teri-butyldiphenylsilyl)oxy)methyl)-5-(8- chloroimidazo[l,5-a]pyrazin-3-yl)piperidine-l-carboxylate (1.33 g, 2.081 mmol) in acetonitrile (15 ml) was added NBS (0.444 g, 2.497 mmol). The mixture was stirred at rt for 15 min. It was concentrated and purified by ISCO (gold 40g, 0-50% ethyl acetate in hexane) to give the title compound. LC-MS: CseHsgBrClNiOsSi, calc. = 717.17, 719.17; found = 717.35, 719.30 (M+H)+.
Step 12: (2S,5R)-benzy\ 5-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3- yl)-2-(((ter^-butyldiphenylsilyl)oxy)methyl)piperidine-l -carboxylate
To a solution of (25',5i?)-benzyl 5-(l-bromo-8-chloroimidazo[l,5-a]pyrazin-3-yl)-2-(((teri- butyldiphenylsilyl)oxy)methyl)piperidine-l -carboxylate (1.35 g, 1.880 mmol) in DMF (6 ml) was added 2,4-dimethoxybenzylamine (0.40 g, 2.392 mmol) and triethylamine (0.42 ml, 3.01 mmol). The mixture was stirred at 60 °C for 4 h. It was diluted with ethyl acetate, washed with water three times and brine once. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by ISCO (Gold 40g, 0-50% ethyl acetate in hexane) to give the title compound as a solid. LC-MS: C45H50BrN5O5Si, calc. = 848.36, 850.31 ; found = 848.28, 850.29 (M+H)+.
Step 13: (2S,5R)-benzy\ 5-(l-bromo-8-((2^-dimethoxybenzyl)ariiino)imidazori.5-alpyrazin-3- yl)-2-(hvdroxymethyl)piperidine-l-carboxylate
A solution of (25',5i?)-benzyl 5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-2-(((teri-butyldiphenylsilyl)oxy)methyl)piperidine-l-carboxylate (880 mg, 1.037 mmol) in THF (5 ml) in a plastic vial was treated with HF (70 wt% in pyridine, 0.8 ml, 1.037 mmol) at rt for 3 h. It was diluted with ethyl acetate, washed with sodium bicarbonate, brine. The organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified by ISCO (Gold 40g, 0-100% EtOAc/EtOH in hexane) to give the title compound. LC-MS: C29H32BrN505, calc. = 610.17, 612.17; found = 610.21, 612.16 (M+H)+.
Step 14: (2S,5R)-benzy\ 5-(l-bromo-8-((2^-dimethoxybenzyl)ariiino)imidazo[1.5-alpyrazin-3- yl)-2-formylpiperidine- 1 -carboxylate
To a solution of (25',5i?)-benzyl 5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-2-(hydroxymethyl)piperidine-l-carboxylate (550 mg, 0.901 mmol) in DCM (9 mL) was added Dess-Martin periodinane (535 mg, 1.261 mmol). It was stirred at rt for 30 min. The reaction was quenched with aqueous sodium bicarbonate and sodium thiosulfate. The mixture was extracted with ethyl acetate. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound as a solid. It was used without further purification. LC-MS: (M+H)+ 574.1, 576.1. Retention time = 1.33 min, method B.
Step 15: (2S.5R)-tert-butyl 5-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin- 3-yl)-2-(((S)-2-(methoxycarbonyl)pyrrolidin-l-yl)methyl)piperidine-l-carboxylate
To a 20 ml sample vial was charged with (2S,5R)-tert-butyl 5-(l-bromo-8-((2,4- dimethoxybenzyl)ariiino)imidazo[l,5-a]pyrazin-3-yl)-2-formylpiperidine-l -carboxylate (81 mg, 0.141 mmol), (S)-methyl pyrrolidine-2-carboxylate hydrochloride (28.0 mg, 0.169 mmol), along with MS 4A (100 mg) and C1CH2CH2C1 (2 ml). The mixture was stirred and sodium
triacetoxyhydroborate (32.9 mg, 0.155 mmol) was added. The resulting reaction mixture was stirred at room temperature for 30 min. The reaction was then quenched by diluting with methylene chloride (10 mL) and NaHCC (sat, 3 mL). The organic layer was separated and the aqueous layer was extracted by methylene chloride (2x). The combined organic phases were dried by MgS04, filtered and concentrrated to afford (2S,5R)-tert-butyl 5-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-2-(((S)-2-(methoxycarbonyl)pyrrolidin-l- y l)methyl)piperi dine- 1-carboxy late which is used to next step without further purification. LC- MS: (M+H)+ 687.1, 689.1, retention time = 1.22 min.
Step 16: (8RJ laS)-8-(8-amino-l-bromoimidazori.5-alpyrazin-3-yl)octahydro-lH- pyridori.2-alpyrrolori.2-dlpyrazin-l 1(1 laHVone
To a 20 ml sample vial was charged with (2S,5R)-tert-butyl 5-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-2-(((S)-2-(methoxycarbonyl)pyrrolidin-l- yl)methyl)piperidine- 1-carboxy late, 359570-010 (75 mg, 0.109 mmol) along with 2,2,2- trifluoroacetic acid (2 mL, 26.1 mmol). The mixture was then stirred and heated in an oil bath of 100 °C for 7 hrs. The deprotection happened very fast in a few minutes, however the cyclication for the formation of lactam was very slow. After cooled to room temperature, the mixture was concentragted by rotary evaporation. The residue was then partitioned between methylene chloride (5 mL) and NaHCC (sat,2 mL). The organic layer was separated and the aquouse layer was extracted (2x). The combined organic phases were dried over MgSC>4, filtered and concentrated. The crude was purified by MPLC (4 g silica gel, 0 to 10% methanol in methylene chloride) to afford solid product (8R,l laS)-8-(8-amino-l-bromoimidazo[l,5-a]pyrazin-3- yl)octahydro-lH-pyrido[l,2-a]pyrrolo[l,2-d]pyrazin-l l(l laH)-one. LC-MS: (M+H)+ 404.9, 406.9, retention time = 0.46 min. 1HNMR (CDC13, 500 MHz): δ = 7.31 (1H, d, J = 5.0 Hz), 7.00 (1H, d, J = 5.0 Hz), 5.93 (2H, br), 4.84 (1H, dd, J = 13.0, 3.0 hz), 3.48 (1H, dd, J = 5.0, 2.0 Hz), 3.06 (1H, m), 3.96 (1H, m), 2.86 (1H, dd, J = 11.5, 2.5 Hz), 2.77 (1H, m), 2.69 (2H, m), 2.34 (1H, m), 2.15 (2H, m), 1.92 (3H, m), 1.75 (2H, m) ppm.
Figure imgf000065_0001
(5aS.8RJ laR)-8-(8-andno-l-bromoimidazo[1.5-alpyrazin-3-yl)octahvdro-lH-pyrido[1.2- al pyrrolo [ 1.2-dl pyrazin- 11(11 aH)-one
In the same procedure for the preparation of Intermediate 18, at step 15, (R)-methyl pyrrolidine- 2-carboxylate hydrochloride was applied for the reduction animation, the title compound (5aS,8R,l laR)-8-(8-aniino-l-bromoiniidazo[l,5-a]pyrazin-3-yl)octahydro-lH-pyrido[l,2- a]pyrrolo[l,2-d]pyrazin-l l(l laH)-one was prepared. LC-MS: (M+H)+ 405.0, 406.9, retention time = 0.30 min, method B.
Intermediate 20
Figure imgf000066_0001
(7'R.9a'S)-7'-(8-amino-l-bromoimidazo[1.5-alpyrazin-3-yl)hexahvdrospiro[cvclobutane-1.3'- pyrido[1.2-alpyrazinl-4'(2'H)-one
In the same procedure for the preparation of Intermediate 18, at step 15, methyl 1- aminocyclopentanecarboxylate oxalate was applied for the reduction animation, the title compound (7'R,9a'S)-7'-(8-amino-l-bromoimidazo[l,5-a]pyrazin-3-yl)hexahydrospiro
[cyclobutane-l,3'-pyrido[l,2-a]pyrazin]-4'(2'H)-one was prepared. LC-MS: (M+H)+ 404.9, 406.9, retention time = 0.31 min, method B.
Intermediate 21
Figure imgf000066_0002
4-(8-amino-3-((3R.6aR.l laR)-6-oxodecahvdropyrrolo[ .2':4.51pyrazino[2.1-cl[1.41oxazin-3- yl)irnidazo[1.5-alpyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide Step 1 : (2R.5S)-tert-butyl 2-(l-bromo-8-chloroimidazo[1.5-alpyrazin-3-yl)-5-(((tert- butyldiphenylsilyl)oxy)methyl)morpholine-4-carboxylate
To a solution of (2R,5S)-tert-butyl 5-(((tert-bu†yldiphenylsilyl)oxy)methyl)-2-(8- chloroimidazo[l,5-a]pyrazin-3-yl)morpholine-4-carboxylate (6.2 g, 10.21 mmol) in acetonitrile (80 mL) was added l-bromopyrrolidine-2,5-dione (1.999 g, 11.23 mmol) under N2, and the mixture stirred at 20 °C for 1.5 hours. The mixture was poured into an ice-water (200 mL), and 10 mL of sat. aq. NaHCCb was added. The mixture was filtered, and the filter cake was dissolved with 200 mL of EtOAc, the organic layer was washed with H20 (10 mL χ 5), brine (20 mL x2), dried over Na2SC>4, filtered, and evaporated to afford the title compound, which was used in the next step directly. *H NMR (400MHz, CDC13) δ = 8.05 (d, J=5.1 Hz, IH), 7.68 (t, J=5.1 Hz, 4H), 7.49 - 7.36 (m, 6H), 7.34 (d, J=4.7 Hz, IH), 5.06 (d, J=3.5 Hz, IH), 4.74 (d, J=14.1 Hz, IH), 4.15 (d, J=6.7 Hz, IH), 3.95 - 3.87 (m, 2H), 3.81 - 3.73 (m, IH), 3.38 (dd, J=4.3, 14.1 Hz, IH), 3.28 (dd, J=3.3, 11.9 Hz, IH), 1.50 (s, 9H), 1.05 (s, 9H).
Step 2: (2R.5S)-tert-butyl 2-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)-5-(((tert-butyldiphenylsilyl)oxy)methyl)morpholine-4-carboxylate
To a solution of (2R,5S)-tert-butyl 2-(l-bromo-8-chloroimidazo[l,5-a]pyrazin-3-yl)-5-(((tert- butyldiphenylsilyl)oxy)methyl)morpholine-4-carboxylate (6.9 g, 10.06 mmol) in DMF (70 mL) was added K2CO3 (3.47 g, 25.1 mmol) and (2,4-dimethoxyphenyl)methanamine (2.186 g, 13.07 mmol) under N2, and the reaction mixture was stirred at 80 °C for 2 hours. LCMS showed the reaction was complete, and then the brown mixture was cooled to 20 °C and poured into an ice- water (300 mL) slowly, and the mixture was filtered, and the filter cake was dissolved with EtOAc (300 mL). The organic layer was washed with H20 (50 mL χ 5), brine (50 mL), dried over Na2SC>4, evaporated to give the crude product, which was then purified by flash
chromatography (Pet. ether/EtOAc = 70 ~ 60%) to afford the title compound.
1H NMR (400MHz, CDC13) δ = 7.68 (t, J=5.5 Hz, 4H), 7.49 - 7.34 (m, 7H), 7.28 (s, IH), 7.11 (d, J=5.1 Hz, IH), 6.75 (br. s., IH), 6.49 (s, IH), 6.45 (dd, J=1.8, 8.0 Hz, IH), 4.96 (d, J=3.1 Hz, IH), 4.72 - 4.60 (m, 3H), 4.18 - 4.11 (m, IH), 3.93 - 3.83 (m, 5H), 3.82 - 3.74 (m, 4H), 3.41 - 3.28 (m, 2H), 1.49 (s, 9H), 1.04 (s, 9H) ppm.
Step 3: (2R.5R)-tert-butyl 2-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)-5-(hvdroxymethyl)morpholine-4-carboxylate
To a solution of (2R,5S)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-(((tert-butyldiphenylsilyl)oxy)methyl)mo holine-4-carboxylate (6.6 g, 8.08 mmol) in THF (80 mL) was added TBAF (9.70 mL, 9.70 mmol) slowly, and the light brown mixture was stirred at 20 °C for 12 hours. TLC (Pet. ether/THF=l : 1) showed the reaction was complete, and then the mixture was quenched with H20 (50 mL), the mixture was extracted with EtOAc (50 mL x3). The organic layer was washed with brine (50 mL), dried over Na2SC>4, evaporated to get the crude product, which was then purified by flash chromatography (Pet. ether/THF=50 ~ 40 %) to afford the title compound.
1H NMR (400MHz, CDC13) δ = 7.40 (d, J=5.1 Hz, IH), 7.27 (br. s., IH), 7.11 (d, J=4.7 Hz, IH), 6.76 (br. s., IH), 6.49 (s, IH), 6.44 (dd, J=2.0, 8.2 Hz, IH), 4.98 (br. s., IH), 4.67 (d, J=2.7 Hz, 2H), 4.48 (dd, J=2.3, 13.7 Hz, IH), 3.98 - 3.85 (m, 6H), 3.80 (s, 3H), 3.77 - 3.71 (m, 2H), 3.69 (br. s., IH), 3.47 (dd, J=2.9, 11.9 Hz, IH), 1.52 (s, 9H) ppm. Step 4: (2R.5S)-tert-butyl 2-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)-5-formylmorpholine-4-carboxylate
To a solution of (2R,5R)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-(hydroxymethyl)mo holine-4-carboxylate (1.14 g, 1.971 mmol) in DCM (20 mL) was added Dess-Martin Periodinane (1.254 g, 2.96 mmol) in portions at 0 °C, and the mixture was stirred for 3 hours. TLC showed most of the starting material was consumed, and the mixture was poured into 80 mL of sat. aq. NaHCCb in an ice-bathe, and 1.5 g of Na2S2C>3 was added to the mixture, and stirred for 10 min. The mixture was extracted with DCM (30 mL x 3), and the organic layer was washed with brine (20 mL), dried over Na2SC>4, evaporated to get the crude product, which was purified by flash chromatography (Pet. ether/THF= 70 ~ 60%) to afford the title compound.
*H NMR (400MHz, CDC13) δ = 9.63 (s, 1H), 7.33 (d, J=4.7 Hz, 1H), 7.26 (br. s., 1H), 7.12 (d, J=5.1 Hz, 1H), 6.77 (br. s., 1H), 6.48 (d, J=1.6 Hz, 1H), 6.43 (dd, J=2.0, 8.2 Hz, 1H), 4.92 (br. s., 1H), 4.66 (d, J=3.9 Hz, 2H), 4.37 (dd, J=4.3, 13.7 Hz, 1H), 4.28 (br. s., 1H), 4.03 (d, J=9.4 Hz, 1H), 3.87 (s, 3H), 3.79 (s, 3H), 3.76 - 3.71 (m, 1H), 3.61 (d, J=8.2 Hz, 1H), 1.52 (br. s., 9H) ppm.
Step 5: (2R.5R)-tert-butyl 2-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl -5- 2- methoxycarbonyl pyrrolidin-l-yl methyl moφholine-4-carboxylate
To a mixture of (2R,5S)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-formylmo holine-4-carboxylate (200 mg, 0.347 mmol), methyl pyrrolidine-2- carboxylate (53.8 mg, 0.416 mmol) and sodium cyanoborohydride (65.4 mg, 1.041 mmol) in CH2CI2 (5 mL) was added AcOH (1.986 μΐ, 0.035 mmol). The mixture was stirred at 20 °C for 12 hours. The mixture was diluted with water (5 mL) and extracted with ethyl acetate (10 mLx4). The combined organic layers were washed with brine (10 mL), dried over sodium sulfate and concentrated to afford the crude product ,which was purified on silica gel column chromatograph (DCM/THF = 100% ~ 30 %) to afford the title compound. MS: 689.2/691.2 (M+l).
Step 6: (3R.6aR.l laR)-3-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)octahvdropyrrolor .2':4.51pyrazinor2.1-ci ri.41oxazin-6(lH)-one
A mixture of (2R,5R)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-(((R)-2-(methoxycarbonyl)pyrrolidin-l-yl)methyl)morpholine-4-carboxylate (200 mg, 0.290 mmol) in HC1/ Dioxane (4M) (3 mL, 12.00 mmol) was stirred at 20 °C for 2 hours. LCMS showed that the reaction was complete, then the mixture was concentrated, and the residue was dissolved with H20 (2 mL) and basified with sat. aq. NaHCC to pH=8~9, and the mixture was extracted with DCM (10 mL x4). The organic layer was washed with brine (10 mL), dried over Na2SC>4, filtered, and evaporated to give the product (R)-methyl l-(((3R,6R)-6- (l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)morpholin-3- yl)methyl)pyrrolidine-2-carboxylate (170 mg), which was then dissolved in anhydrous MeOH (4 mL) was stirred at 80 °C for 12 hours. Then the mixture was cooled to room temperature and evaporated to afford the title compound, which was used in the next step directly.
1H NMR (400MHz, CDC13) δ = 7.29 (d, J=5.1 Hz, IH), 7.24 (s, IH), 7.12 (d, J=5.1 Hz, IH), 6.78 (t, J=5.3 Hz, IH), 6.48 (s, IH), 6.44 (d, J=8.2 Hz, IH), 4.89 (dd, J=2.3, 13.7 Hz, IH), 4.73 - 4.63 (m, 3H), 3.98 (dd, J=2.7, 11.3 Hz, IH), 3.87 (s, 3H), 3.79 (s, 3H), 3.49 - 3.41 (m, IH), 3.25 (td, J=5.9, 11.7 Hz, 2H), 3.07 - 2.94 (m, 2H), 2.47 - 2.34 (m, 2H), 2.24 - 2.13 (m, IH), 2.05 - 1.95 (m, IH), 1.92 - 1.76 (m, 3H) ppm.
Step 7: (3R.6aR.l laR)-3-(8-amino-l-bromoimidazori.5-alpyrazin-3- yl)octahvdropyrrolor .2':4.51pyrazinor2.1-ciri.41oxazin-6(lH)-one
A mixture of (3R,6aR,l laR)-3-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)octahydropyrrolo[ ,2':4,5]pyrazino[2,l-c] [l,4]oxazin-6(lH)-one (80 mg, 0.144 mmol) in TFA (3 mL) was stirred at 80 °C for 2 hours under N2. Then the mixture was concentrated, the residue was dissolved with sat. aq. NaHCC (20 mL), and the mixture was extracted with DCM (10 mL x3). The organic layer was washed with brine (10 mL), dried over Na2SC>4, evaporated to afford the title compound, which was used in the next step directly.
1H NMR (400MHz, CDC13) δ = 7.43 (d, J=4.7 Hz, IH), 7.06 (d, J=5.1 Hz, IH), 5.74 (br. s., 2H), 4.94 (dd, J=2.2, 13.9 Hz, IH), 4.75 (dd, J=2.2, 10.8 Hz, IH), 4.00 (dd, J=2.7, 11.3 Hz, IH), 3.88 (d, J=7.4 Hz, IH), 3.50 - 3.43 (m, IH), 3.31 - 3.23 (m, 2H), 3.10 - 2.95 (m, 2H), 2.49 - 2.35 (m, 2H), 2.27 - 2.15 (m, IH), 2.07 - 1.95 (m, IH), 1.92 - 1.80 (m, 2H) ppm.
2
Figure imgf000069_0001
(3R.6aS.l laR)-3-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)octahvdropyrrolo[ .2':4.51pyrazino[2.1-cl[1.41oxazin-6(lH)-one Step 1 : (2R.5R)-tert-butyl 2-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl -5-((2-(methoxycarbonyl pyrrolidin-l -yl methyl moφholine-4-carboxylate
To a mixture of (2R,5S)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-formylmo holine-4-carboxylate (90 mg, 0.156 mmol) , methyl pyrrolidine-2- carboxylate (24.20 mg, 0.187 mmol) and NaCNBH3 (29.4 mg, 0.468 mmol) in CH2C12 (2 mL) was added AcOH (0.894 μΐ, 0.016 mmol). The mixture was stirred at 20 °C for 12 hours. The mixture was diluted with water (5 mL) and extracted with ethyl acetate (10 mLx4). The combined organic layers were washed with brine (10 mL), dried over sodium sulfate and concentrated to afford the crude product, which was purified on silica gel column chromatograph (PE/EA = 100% ~ 60 %) to afford the title compound. MS: 689.2/691.2 (M+l).
Step 2: (3R.6aS .1 laR)-3-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)octahvdropyrrolo[ .2':4.51pyrazino[2.1-cl [1.41oxazin-6(lH)-one
A solution of (2R,5R)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-(((S)-2-(methoxycarbonyl)pyrrolidin-l-yl)methyl)morpholine-4-carboxylate (60 mg, 0.087 mmol) in HC1/ Dioxane (2mL, 7.13 mmol) was stirred at 20 °C for 3 hours.
LCMS showed that the reaction was complete, then the mixture was concentrated, and the residue was dissolved with H2O (5 mL), and basified with sat. aq. NaHCCb to pH=8 ~ 9, and the mixture was extracted with DCM (10 mL x4). The organic layer was washed with brine (10 mL), dried over Na2SC>4, filtered, and evaporated to get the compound (S)-methyl l -(((3R,6R)-6- (l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)morpholin-3- yl)methyl)pyrrolidine-2-carboxylate (50 mg, 88 % yield), which was dissolved in anhydrous methanol (3 mL) and was heated at 80 °C for 12 hrs under N2. The mixture was cooled to room temperature, and evaporated to afford the title compound, which was used in the next step directly.
*H NMR (400MHz, CDC13) δ = 7.28 - 7.26 (m, 1H), 7.25 - 7.23 (m, 1H), 7.13 (d, J=5.1 Hz,
1H), 6.79 (br. s., 1H), 6.49 (s, 1H), 6.44 (d, J=8.2 Hz, 1H), 4.94 (d, J=14.1 Hz, 1H), 4.74 - 4.65 (m, 3H), 3.88 (s, 4H), 3.80 (s, 3H), 3.75 (br. s., 1H), 3.39 - 3.29 (m, 1H), 3.06 (t, J=8.0 Hz, 1H), 2.92 (br. s., 1H), 2.84 - 2.71 (m, 2H), 2.42 (br. s., 1H), 2.18 (br. s., 1H), 2.01 - 1.85 (m, 2H), 1.81 (br. s., 1H), 1.65 (br. s., 1H) ppm.
Intermediate 23, 24
6'-(8-andno-l -bromoimidazo[1.5-alpyrazin-3-yl)tetrahvdro-2'H-spiro[cvclopropane-l. - indolizinl-3'(5'H)-one (23). and 6'-(8-amino-l-bromoimidazo[1.5-alpyrazin-3-yl)tetrahydro-2'H- spiro [cyclopropane- 1.1 '-indolizinl -3'(5 'H)-one (24)
Figure imgf000071_0001
Step 1 : 1 -(5-bromopyridin-2-yl)cvclopropanecarbonitrile
To a stirred solution of cyclopropanecarbonitrile (19.06 g, 284 mmol) and 5-bromo-2- fluoropyridine (50 g, 284 mmol) in toluene (400 mL) was added potassium
bis(trimethylsilyl)amide (500 ml, 500 mmol) in THF (1M) dropwise at 0 °C, the reaction mixture was stirred at 10°C for 16 hours. After quenching with a saturated aqueous solution of ammonium chloride (500 mL), the mixture was extracted with EtOAc (200mLx3) .The combined organic layers were washed with brine (500 mL) The extracts were dried over Na2SC>4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (Petr. ether / ethyl acetate=100%~80%) to afford l-(5-bromopyridin-2- y cyclopropanecarbonitrile H NMR (CDC13 400 MHz) δ = 8.48 (d, J=1.6 Hz, 1H), 7.79 (dd, J=2.0, 8.2 Hz, 1H), 7.59 (d, J=8.2 Hz, 1H), 1.80 - 1.75 (m, 2H), 1.74 - 1.69 (m, 2H) ppm.
Step 2: l-(5-bromopyridin-2-yl)cvclopropanecarboxylic acid
To a solution of l-(5-bromopyridin-2-yl)cyclopropanecarbonitrile (58 g, 260 mmol) in ethanol(500mL) was added a solution of NaOH (31.2 g, 780 mmol) in water (lOOmL), then the mixture was heated to 100°C for 24 hours. After cooling to room temperature, the solution was poured into an ice-cold saturated aqueous Na2HPC>4 solution (500 mL) and the resulting mixture was adjusted to pH 4 by the addition of 1 M aqueous hydrochloric acid. The mixture was extracted with EtOAc (300mLx4) .The combined organic layers were washed with brine (500 mL) The extracts were dried over Na2SC>4, filtered and concentrated under reduced pressure to give l-(5-bromopyridin-2-yl)cyclopropanecarboxylic acid. XH NMR (CDC13 400 MHz) δ = 8.50 (d, J=1.6 Hz, 1H), 7.87 (dd, J=2.0, 8.6 Hz, 1H), 6.80 (d, J=8.6 Hz, 1H), 2.09 (q, J=4.0 Hz, 2H), 1.43 - 1.34 (m, 2H) ppm.
Step 3: l-(5-bromopyridin-2-yl)-N-methoxy-N-methylcvclopropanecarboxamide
To a solution of l-(5-bromopyridin-2-yl)cyclopropanecarboxylic acid (60 g, 248 mmol) in anhydrous DMF (600 mL) was added HATU (113 g, 297 mmol) and the mixture was stirred for 30 mins under nitrogen. ΕΪ Ν (104 ml, 744 mmol) was added and followed by Ν,Ο- dimethylhydroxylamine hydrochloride (26.6 g, 273 mmol). The mixture was stirred for 16 hrs at 10 °C. The mixture was diluted with water (1500 mL) and extracted with ethyl acetate (500 mL χ 5). The combined organic layers were washed with water (200 mL χ 2), brine (1000 mL), dried over Na2SC>4 and evaporated to get the crude product, which was purified on silica gel flash chromatography (Pet. ether/EtOAc = 100 ~ 50 %) to givel-(5-bromopyridin-2-yl)-N-methoxy- N-methylcyclopropanecarboxamide. Ti NMR (CDC13400 MHz ) δ = 8.49 (d, J=1.6 Hz, 1H), 7.68 (dd, J=2.2, 8.4 Hz, 1H), 7.06 (d, J=8.6 Hz, 1H), 3.43 (br. s., 3H), 3.19 (s, 3H), 1.49 - 1.38 (m, 4H) ppm.
Step 4: (l-(5-bromopyridin-2-yl)cvclopropyl)methanol
To a solution of l-(5-bromopyridin-2-yl)-N-methoxy-N-methylcyclopropanecarboxamide (60 g, 210 mmol) in EtOH (600 ml) was added NaBH4 (23.88 g, 631 mmol) in portions at 0 °C. The mixture was stirred at 30°C for 20 hours, then poured into a saturated aqueous solution of NH4C1(1000 mL), and extracted with EtOAc (300mLx4). The combined organic layers were washed with brine (500 mL) The extracts were dried over Na2SC>4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (Petr. ether / ethyl acetate=100%~50%) to afford (l-(5-bromopyridin-2-yl)cyclopropyl)methanol . XH NMR (CDC13 400MHz) δ = 8.51 (d, J=2.3 Hz, 1H), 7.71 (dd, J=2.5, 8.5 Hz, 1H), 6.88 (d, J=8.5 Hz, 1H), 3.82 (s, 2H), 3.75 (br. s., 1H), 1.07 (d, J=3.3 Hz, 4H) ppm.
Step 5:2-(l-(5-bromopyridin-2-yl)cvclopropyl)acetonitrile
To a solution of (l-(5-bromopyridin-2-yl)cyclopropyl)methanol (20 g, 88 mmol) and ΕΪ Ν (36.7 ml, 263 mmol) in CH2Cl2(200mL) was added methanesulfonyl chloride (20.07 ml, 258 mmol) dropwise at 0 °C under nitrogen atmosphere. The mixture was stirred at 0 °C for 4 hours, then washed with saturate aqueous NaHCC (50mL), brine (50 mL), dried over Na2SC>4 and concentrated under reduced pressure. The residue was dissolved in DMF (200mL) and then NaCN (13.21 g, 270 mmol) was added. The mixture was stirred at 70 °C fori 8 hours, then diluted with water (600mL) and extracted with EtOAt (200mLx4). The combined organic layers were washed with water (100mLx3), brine (200 mL), dried over Na2SC>4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column
chromatography (Petr. ether / ethyl acetate=100%~60%) to give 2-(l-(5-bromopyridin-2- yl)cyclopropyl)acetonitrile. lH NMR (CDC13 400MHz) δ = 8.56 (d, J=2.0 Hz, 1H), 7.74 (dd,
J=2.3, 8.6 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 2.92 (s, 2H), 1.30 - 1.21 (m, 2H), 1.20 - 1.11 (m, 2H) ppm.
Step 6:2-(l-(5-bromopyridin-2-yl)cvclopropyl)acetic acid To a solution of 2-(l-(5-bromopyridin-2-yl)cyclopropyl)acetonitrile (13.5 g, 56.9 mmol) in ethanol(130mL) was added sodium hydroxide (6.83 g, 171 mmol) in water (30mL). The mixture was heated to 100°C for 24 hours . After cooling to room temperature, the solution was poured into ice-cold saturated aqueous Na2HPC>4 solution (50 mL) and the resulting mixture was adjusted to pH 4 by the addition of 1 M aqueous hydrochloric acid. The mixture was extracted with EtOAc (200mLx4) .The combined organic layers were washed with brine (200 mL) The extracts were dried over Na2SC>4, filtered and concentrated under reduced pressure to give 2-(l- (5-bromopyridin-2-yl)cyclopropyl)acetic acid . XH NMR 0351907-0092-1A (CDC13 400MHz ) δ = 8.54 (d, J=2.0 Hz, 1H), 7.82 (dd, J=2.3, 8.6 Hz, 1H), 6.92 (d, J=8.6 Hz, 1H), 2.81 (s, 2H), 1.17 (s, 4H) ppm.
Step 7: methyl 2-(l-(5-bromopyridin-2-yl)cvclopropyl)acetate
A solution of 2-(l-(5-bromopyridin-2-yl)cyclopropyl)acetic acid (30 g, 117 mmol) was dissolved in HCI/CH3OH (250 mL)(4M), and the mixture was stirred at 15 °C for 2 hours. The TLC showed completion of the reaction .The mixture was concentrated under reduced pressure. The residue was dissolved in EtOAc(100 mL) and neutralized with sat. NaHCC pOOmL) (aq.). The aqueous was extracted with EtOAc (100mLx4). The combined organic layers were washed with brine (200 mL) The extracts were dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (EtOAc: Pet. ether=0%~30%) to give methyl 2-(l-(5-bromopyridin-2-yl)cyclopropyl)acetate.1H NMR (CDC13 400 MHz ) δ = 8.48 (br. s., 1H), 7.65 (d, J=6.7 Hz, 1H), 6.99 (d, J=8.2 Hz, 1H), 3.75 - 3.53 (m, 3H), 2.76 (s, 2H), 1.22 (br. s., 1H), 1.01 (br. s., 1H) ppm.
Step 8: methyl 6-(l-(2-methoxy-2-oxoethyl)cvclopropyl)nicotinate
To a solution of methyl 2-(l-(5-bromopyridin-2-yl)cyclopropyl)acetate (10 g, 37.0 mmol) in MeOH (lOOmL) and DMF(lOOmL) was added Et3N (15.48 ml, 111 mmol) ,DPPF (4.10 g, 7.40 mmol) and diacetoxypalladium (0.831 g, 3.70 mmol). The mixture was stirred at 80 °C for 48 hours under CO (50psi). The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. Then the residue was diluted with water (300 mL) and extracted with EtOAc (100mLx4 ). The combined organic layers were washed with water (50mLx3), brine (100 mL) dried over Na2S04, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (EtOAc: Pet. ether =0%~40%) to afford methyl 6-(l-(2- methoxy-2-oxoethyl)cyclopropyl)nicotinate.1H NMR (CDC13 400MHz) δ = 9.05 (br. s., 1H), 8.14 (d, J=6.7 Hz, 1H), 7.14 (d, J=8.2 Hz, 1H), 4.00 - 3.86 (m, 3H), 3.75 - 3.59 (m, 3H), 2.83 (br. s., 2H), 1.36 (br. s., 2H), 1.10 (br. s., 2H) ppm. Step 9: methyl 6-(l-(2-methoxy-2-oxoethyl)cvclopropyl)piperidine-3-carboxylate
To a solution of methyl 6-(l-(2-methoxy-2-oxoethyl)cyclopropyl)nicotinate (8.5 g, 34.1 mmol) in AcOH (100ml) was added NaCNBH4 (6.43 g, 102 mmol) in portions and the mixture was stirred at 40 °C for 24 hours. Then the mixture was concentrated under reduced pressure. The residue was dissolved in water (50ml) and pH adjusted with a saturated aqueous solution of NaHC03(5 mL) to 9, then extracted with EtOAc (50mLx4). The combined organic layers were washed with brine (100 mL) The extracts were dried over Na2SC>4, filtered and concentrated under reduced pressure to give crude methyl 6-(l-(2-methoxy-2- oxoethyl)cyclopropyl)piperidine-3-carboxylate. MS: 256.0 (M+l).
Step 10: methyl3'-oxohexahvdro-2'H-spiro[cvclopropane-l. -indolizinel- 6'- Carboxylate
A solution of methyl 6-(l-(2-methoxy-2-oxoethyl)cyclopropyl)piperidine-3-carboxylate (10 g, 39.2 mmol) in MeOH (100 ml) was refluxed for 16 hours. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column
chromatography (Pet. ether /THF=100%~20%) to afford methyl3'-oxohexahydro-2'H- spiro[cyclopropane-l, -indolizine]-6'-Carboxylate l,l'-indolizine]-6'-carboxylate (trans isomer). XH NMR (CDC13 400MHz )δ = 4.46 (dd, J=3.3, 13.1 Hz, 1H), 3.74 - 3.61 (m, 3H), 3.18 (dd, J=2.7, 11.7 Hz, 1H), 2.78 (t, J=12.3 Hz, 1H), 2.50 - 2.31 (m, 3H), 2.19 (d, J=12.9 Hz, 1H), 1.64 - 1.51 (m, 2H), 1.28 - 1.15 (m, 1H), 0.72 (dtd, J=5.5, 9.8, 19.2 Hz, 2H), 0.65 - 0.56 (m, 1H), 0.56 - 0.46 (m, 1H) ppm.
Step 11: 3'-oxohexahvdro-2'H-spiro[cvclopropane-l. -indolizinel-6'-carboxylic acid (trans) To a solution of methyl3'-oxohexahydro-2'H-spiro[cyclopropane-l, -indolizine]-6'-carboxylate (trans isomer) (1.5g, 6.72 mmol) in THF (lOmL) was added a solution of LiOH (0.483 g, 20.16 mmol) in water (lOmL) and the mixture was stirred at 10°C for 3 hours . The mixture was adjusted to pH 4 by the addition of aqueous hydrochloric acid (1 M), then extracted with EtOAc (10mLx4) .The combined organic layers were washed with brine (30 mL) The extracts were dried over Na2S04, filtered and concentrated under reduced pressure to give 3'-oxohexahydro- 2'H-spiro[cyclopropane-l, -indolizine]-6'-carboxylic acid (trans). XH NMR (CDC13 400MHz) δ = 4.49 (dd, J=3.3, 13.5 Hz, 1H), 3.22 - 3.14 (m, 1H), 2.79 (t, J=12.3 Hz, 1H), 2.50 - 2.35 (m, 3H), 2.23 (d, J=12.1 Hz, 1H), 1.66 - 1.53 (m, 2H), 1.21 (d, J=3.5 Hz, 1H), 0.72 (ddd, J=4.7, 9.8, 18.0 Hz, 2H), 0.60 (td, J=5.0, 9.9 Hz, 1H), 0.52 (dd, J=4.5, 9.6 Hz, 1H) ppm.
Stepl2: N-((3-chloropyrazin-2-yl)methyl)-3'-oxohexahvdro-2'H-spirorcvclopropane-l. - indolizinel-6'-carboxamide (trans) To a solution of (6'R,8a'S)-3'-oxohexahydro-2'H-spiro[cyclopropane-l, -indolizine]-6'- carboxylic acid (trans) (100 mg, 0.478 mmol) in DMF (2 mL) were added HATU (113 g, 297 mmol) , DIEA (0.250 ml, 1.434 mmol) and (3-chloropyrazin-2-yl)methanamine hydrochloride (86 mg, 0.478 mmol) and the mixture was stirred for 16 hrs at 15 °C. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (5 mL χ 4). The combined organic layers were washed with water (5 mL χ 3), brine (10 mL), dried over Na2SC>4, evaporated to get the crude product, which was purified on flash chromatography (Pet. ether/EtOAc = 100 ~ 60 %) to give N-((3-chloropyrazin-2-yl)methyl)-3'-oxohexahydro-2'H-spiro[cyclopropane-l, - indolizine]-6'-carboxamide (trans). lH NMR (400MHz, CDC13) δ = 8.44 (d, J=2.0 Hz, 1H), 8.33 (s, 1H), 6.87 (br. s., 1H), 4.76 - 4.60 (m, 2H), 4.41 (dd, J=3.1, 12.9 Hz, 1H), 3.21 (dd, J=2.9, 11.5 Hz, 1H), 2.91 (t, J=12.3 Hz, 1H), 2.52 - 2.28 (m, 3H), 2.16 - 2.01 (m, 1H), 1.86 - 1.72 (m, 1H), 1.62 (dd, J=3.1, 13.3 Hz, 1H), 1.31 - 1.19 (m, 1H), 0.79 - 0.65 (m, 2H), 0.62 (td, J=5.0, 9.9 Hz, 1H), 0.56 - 0.47 (m, 1H) ppm.
Step 13 :6'-(8-chloroimidazo[1.5-alpyrazin-3-yl)tetrahvdro-2'H-spiro[cvclopropane-l. - indolizinl-3'(5'H)-one (trans)
To a stirred solution of N-((3-chloropyrazin-2-yl)methyl)-3'-oxohexahydro-2'H- spiro[cyclopropane-l, -indolizine]-6'-carboxamide (1.8g, 5.38 mmol) in acetonitrile (20 ml) was added PCI5 (3.36 g, 16.13 mmol),the reaction mixture was stirred at 10°C for 3 hours. After cooling to 0 °C, the mixture was poured into a saturated aqueous solution of NaHCO3(50 mL) at 0°C and extracted with EtOAc (30mLx4). The combined organic layers were washed with brine (50 mL) The extracts were dried over Na2SC>4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CH2CI2 / MeOH =100%~80%) to afford 6'-(8-chloroimidazo[l,5-a]pyrazin-3-yl)tetrahydro-2'H-spiro[cyclopropane-l, - indolizin]-3'(5'H)-one (trans isomer) . ¾ NMR (CDC13 400MHz) δ = 7.79 (s, 1H), 7.68 (d, J=4.7 Hz, 1H), 7.35 (d, J=4.7 Hz, 1H), 4.51 - 4.42 (m, 1H), 3.32 (dd, J=3.1, 11.3 Hz, 1H), 3.03 (d, J=9.0 Hz, 2H), 2.58 - 2.40 (m, 2H), 2.24 - 2.16 (m, 1H), 2.14 - 2.02 (m, 1H), 1.76 (dd, J=3.1, 12.9 Hz, 1H), 1.48 - 1.35 (m, 1H), 0.85 - 0.63 (m, 3H), 0.62 - 0.53 (m, 1H) ppm.
Stepl4: 6'-(l-bromo-8-chloroimidazori.5-alpyrazin-3-yl)tetrahvdro-2'H-spirorcvclopropane- 1.1 '-indolizinl-3'(5'H)-one (trans)
To a solution of (6'R, 8a'S)-6'-(8-chloroimidazo [1, 5-a]pyrazin-3-yl)tetrahydro-2'H- spiro[cyclopropane-l, -indolizin]-3'(5'H)-one (trans isomer) (1.3g, 4.10 mmol) in acetonitrile (20 ml) was added NBS (0.803 g, 4.51 mmol). The resulting mixture was stirred at 10 °C for 1 hour. LCMS showed that the reaction was complete, the mixture was filtered to remove a white solid and the filtrate was poured into a saturated aqueous solution of NaHCC (30 mL) and extracted with EtOAc (40 mLx3). The combined organic layers were washed with brine (50 mL), dried over Na2SC>4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford 6'-(l-bromo-8-chloroimidazo[l,5- a]pyrazin-3-yl)tetrahydro-2'H-spiro[cyclopropane-l, -indolizin]-3'(5'H)-one (trans). XH NMR (CDC13 400MHz) δ = 7.69 (d, J=5.1 Hz, 1H), 7.36 (d, J=5.1 Hz, 1H), 4.45 (d, J=8.6 Hz, 1H), 3.31 (dd, J=2.9, 11.5 Hz, 1H), 3.07 - 2.95 (m, 2H), 2.61 - 2.42 (m, 2H), 2.23 - 2.08 (m, 2H), 1.77 (dd, J=3.1, 13.3 Hz, 1H), 1.47 - 1.35 (m, 1H), 0.84 - 0.66 (m, 3H), 0.60 (dd, J=4.5, 9.6 Hz, 1H) ppm.
Stepl5: 6'-(8-andno-l-bromoimidazo[1.5-alpyrazin-3-yl)tetrahvdro-2'H-spiro[cvclopropane-l. - indolizinl-3'(5'H)-one (trans)
To a solution of (6'R,8a'S)-6'-(l-bromo-8-chloroimidazo[l,5-a]pyrazin-3
-yl)tetrahydro-2'H-spiro[cyclopropane-l, -indolizin]-3'(5'H)-one (trans isomer) (1.5g, 3.79 mmol) in 2-Propanol (15 ml) was added aq. ammonia (30 ml, 189 mmol). The mixture was stirred at 120 °C for 18 hours in a lOOmL of autoclave. The reaction mixture was concentrated and the residue was purified by silica gel column chromatography (DCM / MeOH= 0%~20%) to afford 6'-(8-andno-l-bromoimidazo[l,5-a]pyrazin-3-yl)tetrahydro-2'H-spiro[cyclopropane-l, - indolizin]-3'(5'H)-one (trans). XH NMR (CDC13 400MHz) δ = 7.25 (d, J=5.1 Hz, 1H), 7.06 (d, J=4.7 Hz, 1H), 5.67 (br. s., 2H), 4.46 (d, J=11.0 Hz, 1H), 3.30 (dd, J=2.9, 11.5 Hz, 1H), 3.06 - 2.91 (m, 2H), 2.60 - 2.42 (m, 2H), 2.22 - 2.13 (m, 1H), 2.11 - 1.99 (m, 1H), 1.74 (dd, J=2.9, 13.1 Hz, 1H), 1.39 (dq, J=3.1, 12.7 Hz, 1H), 0.85 - 0.64 (m, 3H), 0.63 - 0.54 (m, 1H) ppm.
Stepl6: 6'-(8-andno-l-bromoimidazo[1.5-alpyrazin-3-yl)tetrahvdro-2'H-spiro[cvclopropane-l. - indolizinl-3'(5'H)-one (trans isomerl) and 6'-(8-amino-l-bromoimidazori.5-alpyrazin-3- yl)tetrahvdro-2'H-spirorcvclopropane-l. -indolizinl-3'(5'H)-one (trans isomer2)
The trans racemic miture was resolved with chiral SFC (Instrument : Thar 200
Column : OJ 250mmx50mm,10um Mobile phase: A: Supercritical C02 , B: EtOH (0.05% NH3H2O, A:B =70:30 at 200ml/min, Column Temp: 38°C, Nozzle Pressure: lOOBar, Nozzle Temp: 60°C, Evaporator Temp: 20°C, Trimmer Temp: 25°C
Wavelength: 220nm) to give 6'-(8-amino-l-bromoimidazo[l,5-a]pyrazin-3-yl)tetrahydro-2'H- spiro[cyclopropane-l,r-indolizin]-3'(5'H)-one (23). Retention time : 1.203min. ¾ NMR CDC13 400MHz = 7.24 (d, J=5.1 Hz, 1H), 7.06 (d, J=5.1 Hz, 1H), 5.68 (br. s., 2H), 4.45 (d, J=11.0 Hz, 1H), 3.29 (dd, J=2.7, 11.7 Hz, 1H), 2.99 (q, J=12.3 Hz, 2H), 2.58 - 2.42 (m, 2H), 2.21 - 2.12 (m, 1H), 2.10 - 1.98 (m, 1H), 1.78 - 1.72 (m, 1H), 1.45 - 1.33 (m, 1H), 0.82 - 0.64 (m, 3H), 0.58 (dd, J=4.5, 9.6 Hz, 1H) ppm. 6'-(8-amino-l -bromoimidazo[l ,5-a]pyrazin-3-yl)tetrahydro-2'H- spiro[cyclopropane-l, -indolizin]-3'(5'H)-one (24). Retention time : 1.362min. 'H NMR (CDC13 400MHz) = 7.17 (d, J=5.1 Hz, 1H), 6.97 (d, J=5.1 Hz, 1H), 5.85 (br. s., 2H), 4.38 (d, J=1 1.0 Hz, 1H), 3.23 (dd, J=2.9, 1 1.5 Hz, 1H), 3.00 - 2.86 (m, 2H), 2.50 - 2.36 (m, 2H), 2.16 - 2.06 (m, 1H), 2.04 - 1.91 (m, 1H), 1.66 (dd, J=2.9, 13.1 Hz, 1H), 1.38 - 1.26 (m, 1H), 0.78 - 0.58 (m, 3H), 0.51 (dd, J=4.9, 9.2 Hz, 1H) ppm.
Intermediate 25, 26, 27, 28
Figure imgf000077_0001
isomer 1 A isomer 1B isomer 2A isomer 2B
25 26 27 28
Trans -4-(8-anTino-3-(2-oxooctahvdro-lH-cvclopropa[alindolizin-5-yl)imidazo[1.5-alpyrazin-l- yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (Four isomers) Step 1 : methyl 6-vinylnicotinate
To a solution of methyl 6-bromonicotinate (lOg, 46.3 mmol) in Me2CHOH (200 mL) was added C2H3BF4K+ (14.16 g, 93 mmol), Et3N (19.36 mL, 139 mmol), Pd(dppf)C12 (0.2 g). The mixture was stirred at 100 °C for 12 hours under N2 atmosphere. The mixture was concentrated and the residue was purified by column chromatography on silica gel (Pet.ether: EtOAc =15: 1) to give methyl 6-vinylnicotinate. MS : 164.2 (M+l). method DM (Rt =0.824 min).
Step 2: methyl 6-(2-(ethoxycarbonyl)cvclopropyl)nicotinate
Ethyl diazoacetate (381 mg, 3.31 mmol) was added to a solution of methyl 6-vinylnicotinate (450 mg, 2.76 mmol) in dimethylbenzene (20 mL). The mixture was heated to 130 °C to reflux for 2 hours, then cooled to 25°C and stirred for another 12 hours. The reaction solution was concentrated in vacuo and purified with column chromatography on silica gel (CH2CI2 / MeOH = 100% ~ 10%) to give methyl 6-(2-(ethoxycarbonyl)cyclopropyl)nicotinate. MS: 250.2 (M+l). method D (Rt =0.995 min). XH NMR (400MHz, CD3OD): δ= 8.94 (s, 1H), 8.23 (d, J=6.0 Hz, 1H), 7.45 (d, J=8.0 Hz, 1H), 4.13 (q, J=6.8 Hz, 2H), 3.90 (s, 3H), 2.68-2.66 (m, 1H), 2.27-2.24 (m,lH), 1.64-1.59 (m, 2H), 1.23 (t, J=7.2 Hz, 3H) ppm.
Step 3: methyl 6-(2-(ethoxycarbonyl)cvclopropyl)piperidine-3-carboxylate
To a solution of methyl 6-(2-(ethoxycarbonyl)cyclopropyl)nicotinate (250 mg, 1.003 mmol) in acetonitrile (50 mL) was added NaB¾CN (63.0 mg, 1.003 mmol) at 0°C. The mixture was stirred at 15°C for 12 h. The reaction solution was quenched with water (20 mL) and extracted with EtOAc (20 mLx2). The organic layer was concentrated to give crude product and purified with prep-HPLC (TFA) to give methyl 6-(2-(ethoxycarbonyl)cyclopropyl)piperidine-3- carboxylate. MS: 256.2 (M+l). method D (Rt =0.918 min). XH NMR (400MHz, CDC13) = 4.21-4.09 (m, 2H), 3.36-3.39 (m, 4H), 2.91 (s, 2H), 2.31-1.55 (m, 6H), 3.90 (s, 3H), 1.28-0.93 (m, 5H) ppm.
Step 4: methyl 2-oxooctahvdro-lH-cvcloproparalindolizine-5-carboxylate
A solution of methyl 6-(2-(ethoxycarbonyl)cyclopropyl)piperidine-3-carboxylate (200 mg, 0.783 mmol) in toluene (100 mL) was heated to 80°C and stirred for 12 hours. The reaction solution was concentrated and purified with prep-HPLC (TFA) to give methyl 6-(2-
(ethoxycarbonyl)cyclopropyl)piperidine-3-carboxylate. MS: 210.2 (M+l). method D (Rt =0.678 min). XH NMR (400MHz, CDC13) δ = 4.38 (d, J=6.8 Hz, 1H), 3.67 (s, 3H), 3.60 (s, 1H), 2.77-2.73 (m, 1H), 2.64 (s, 1H), 2.25 (d, J=6.8 Hz, 1H), 1.95-1.92 (m, 2H), 1.84-1.80 (m, 5H), 1.63-1.62 (m, 1H), 1.50-1.47 (m, 1H), 0.96-0.92 (m, 1H), 0.59 (d, J=3.6 Hz, 1H) ppm.
Step 5: 2-oxooctahvdro-lH-cvclopropa[alindolizine-5-carboxylic acid
To a solution of methyl 6-(2-(ethoxycarbonyl)cyclopropyl)piperidine-3-carboxylate (5 g, 23.90 mmol) in THF/ H20 (1 : 1, 80 mL) was added lithium hydroxide (1.431 g, 59.7 mmol). The mixture was stirred at 8°C for 13 hours. The reaction was acidified to pH = 3-5 with 2M aq.HCl and concentrated to give the crude product 2-oxooctahydro-lH-cyclopropa[a]indolizine-5- carboxylic acid. MS: 196.2 (M+l). method D (Rt =0.418 min).
Step 6: N-((3-chloropyrazin-2-yl)methyl)-2-oxooctahydro-lH-cvclopropa[alindolizine-5- carboxamide
To the solution of 2-oxooctahydro-lH-cyclopropa[a]indolizine-5-carboxylic acid (3.5 g, 10.76 mmol) in CH2C12 (150 mL) and DMF (20 mL) was added EDC (3.09 g, 16.14 mmol) and DMAP (1.971 g, 16.14 mmol) under N2, followed by (3-chloropyrazin-2-yl)methanamine hydrochloride (2.324 g, 12.91 mmol). The resulting mixture was stirred at 15 °C for 3 h. TLC (DCM: MeOH = 10: 1) and LCMS showed the starting material was consumed completely. The mixture was diluted with H20 (350mL), extracted with DCM (80mLx2). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under vacuum to give crude product which was purified by combi flash (DCM: THF= 100-20%) to give N-((3-chloropyrazin- 2-yl)methyl)-2-oxooctahydro-lH-cyclopropa[a]indolizine-5-carboxarnide. MS (ESI): 321.0 (M+l). method D (Rt =1.078 min). lH NMR (400MHz, CDC13) = 8.45 (br. s., 1H), 8.37 - 8.30 (m, 1H), 7.72 (br. s., 1H), 4.75 - 4.60 (m, 2H), 4.24 - 4.13 (m, 1H), 3.79 - 3.64 (m, 1H), 2.89 - 2.78 (m, 1H), 2.15 - 2.03 (m, 3H), 1.99 - 1.87 (m, 1H), 1.85 - 1.72 (m, 1H), 1.36 - 1.22 (m, 2H), 1.19 - 1.01 (m, 1H), 0.79 - 0.61 (m, 1H) ppm.
Step 7: 5-(8-chloroimidazo[1.5-alpyrazin-3-yl)hexahvdro-lH-cvclopropa[alindolizin-2(laH)-one To a solution of N-((3-chloropyrazin-2-yl)methyl)-2-oxooctahydro-lH-cyclopropa[a]indolizine- 5-carboxamide (2.0 g, 6.23 mmol) in acetonitrile (80 mL) was added PCI5 (3.90 g, 18.70 mmol) in portions at 0°C. After addition, the mixture was allowed to warm to room temperature (20 °C) and stirred for 12 hrs under N2 atmosphere. Then the mixture was poured into ice aq.NaHCC (100 mL) slowly, and stirred for 20 min. The mixture was then extracted with DCM (20mLx3). The organic layer was washed with brine (20 mL), dried over Na2SC>4, and concentrated in vacuo to give the residue, which was then purified with prep HPLC to give 5-(8-chloroimidazo[l,5- a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin-2(laH)-one. MS (ESI):303.0 (M+l). Acq Method 0-60AB_2min_220&254.1cm (Rt =1.178 min). lH NMR (400MHz, CD3OD)5= 8.27 - 8.22 (m, 1H), 7.96 (s, 1H), 7.47 - 7.40 (m, 1H), 4.18 - 4.07 (m, 1H), 3.89 - 3.79 (m, 1H), 3.25 - 3.14 (m, 1H), 3.07 - 2.95 (m, 1H), 2.15 (d, J=9.0 Hz, 2H), 2.04 - 1.85 (m, 3H), 1.63 - 1.47 (m, 1H), 1.13 - 1.05 (m, 1H), 0.82 (d, J=3.5 Hz, 1H).
Step 8: 5-(l-bromo-8-chloroirrudazo[1.5-alpyrazin-3-yl)hexahvdro-lH-cvclopropa[alindolizin- 2(laH)-one
NBS was added in one portion to the solution of 5-(8-chloroimidazo[l,5-a]pyrazin-3- yl)hexahydro-lH-cyclopropa[a]indolizin-2(laH)-one (900 mg, 2.97 mmol) in DMF (12 mL) and the mixture was stirred at room temperature (20 °C) for 2h. The mixture was then pouted into aq. NaHCCb (30 mL). The mixture was extracted with EtOAc (35 mLx3), washed with water (10 mLx2) and dried over Na2SC>4. The organic layer was concentrated in vacuo, and purified with silica gel (methol/dichloromethane 0%~ 15%) to give the crude product of 5-(l-bromo-8- chloroimidazo[l,5-a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin-2(laH)-one. XH NMR (400MHz, CD3OD) = 8.20 (dt, J=5.1, 16.3 Hz, 1H), 7.39 - 7.32 (m, 1H), 4.15 - 4.06 (m, 2H), 3.88 - 3.79 (m, 1H), 3.63 (d, J=13.3 Hz, 1H), 3.17 - 3.08 (m, 1H), 2.19 - 2.11 (m, 3H), 2.00 - 1.88 (m, 3H), 1.58 - 1.47 (m, 1H). MS (ESI): 381/383 (M+l). Acq Method 10- 80AB_2min_220&254.1cm (Rt =1.014 min). Step 9: 5-(l-bromo-8-chloroimidazo[1.5-alpyrazin-3-yl)hexahvdro-lH-cvclopropa[alindolizin- 2(laH)-one
To the solution of 5-(l-bromo-8-chloroimidazo[l,5-a]pyrazin-3-yl)hexahydro-lH- cyclopropa[a]indolizin-2(laH)-one (850 mg, 2.227 mmol)) in DMF (15 niL) was added K2C03 (923 mg, 6.68 mmol) and (2,4-dimethoxyphenyl)methanamine (447 mg, 2.67 mmol) and the mixture was stirred at 80°C for 2 hours under N2. The mixture was partitioned between water (50 mL) and ethyl acetate (150 mL), the organic layer was concentrated in vacuo. The residue was purified with preparative HPLC on Gilson 281 instrument fitted with a Waters XSELECT C18 150*30mm*5um using water and acetonitrile as the eluents. Mobile phase A: water (containing 0.1%TFA, v/v), mobile phase B: acetonitrile. Gradient: 19-35% B, 0-13.0 min; 100% B, 13.2- 15.2min; 10% B, 15.4-17minFlowRate:25ml/min to give 5-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin- 2(laH)-one (isomer 1) (the faster eluting) XH NMR (400MHz, CDC13) δ = 7.11 - 7.06 (m, 1H), 7.05 - 7.01 (m, 1H), 6.74 (br. s., 1H), 6.47 (d, J=2.0 Hz, 1H), 6.42 (dd, J=2.2, 8.0 Hz, 1H), 4.65 (d, J=5.5 Hz, 2H), 4.13 (dd, J=3.7, 13.5 Hz, 1H), 3.86 (s, 3H), 3.78 (s, 3H), 3.71 - 3.64 (m, 1H), 2.87 (t, J=12.3 Hz, 1H), 2.71 - 2.61 (m, 1H), 2.12 - 2.02 (m, 2H), 1.97 (d, J=7.8 Hz, 2H), 1.40 - 1.30 (m, 1H), 1.28 - 1.20 (m, 1H), 1.04 - 0.96 (m, 1H), 0.71 (d, J=3.9 Hz, 1H). HPLC
METHOD: 0-60AB_l .2ml.met (Rt = 3.11 min). And 5-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin- 2(laH)-one (isomer 2) (the slower eluting). XH NMR (400MHz, CDC13) 5 = 7.14 - 7.07 (m, 2H), 6.72 (br. s., 1H), 6.47 (d, J=2.0 Hz, 1H), 6.42 (dd, J=2.3, 8.2 Hz, 1H), 4.65 (d, J=5.5 Hz, 2H), 4.19 (dd, J=3.5, 13.3 Hz, 1H), 3.86 (s, 3H), 3.78 (s, 3H), 3.74 - 3.67 (m, 1H), 3.46 (d, J=10.6 Hz, 1H), 2.98 - 2.89 (m, 1H), 2.84 - 2.75 (m, 1H), 2.10 (d, J=6.3 Hz, 2H), 1.97 (dd, J=3.1, 12.9 Hz, 1H), 1.90 (br. s., 1H), 1.74 - 1.68 (m, 1H), 1.51 - 1.40 (m, 1H), 1.12 - 1.05 (m, 1H). HPLC METHOD: 0-60AB_l.2ml.met (Rt = 3.21 min).
Step 10: 5-(l-bromo-8-chloroimidazori.5-alpyrazin-3-yl)hexahvdro-lH-cvcloproparalindolizin- 2(laH)-one (isomer 1A). 5-(l-bromo-8-chloroimidazori.5-alpyrazin-3-yl)hexahydro-lH- cvclopropa[alindolizin-2(laH)-one (isomer IB). 5-(l-bromo-8-chloroimidazo[1.5-alpyrazin-3- yl)hexahvdro-lH-cvclopropa[alindolizin-2(laH)-one (isomer 2A). and 5-(l-bromo-8- chloroimidazo[1.5-alpyrazin-3-yl)hexahvdro-lH-cvclopropa[alindolizin-2(laH)-one (isomer 2B)
5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydro-lH- cyclopropa[a]indolizin-2(laH)-one (isomer 1) (300 mg, 0.585 mmol) was purified with chiral HPLC [SFC condition: Instrument: SFC-80; Column: AS 250x30mm I.D.,20um; Mobile phase: A: Supercritical C02 , B: MeOH ( 0.1% NH3H20 ) , A:B =60:40 at 70ml/min; Column Temp: 38°C; Nozzle Pressure: l OOBar; Nozzle Temp: 60°C; Evaporator Temp: 20°C; Trimmer Temp: 25°C; Wavelength: 220nm.] to get trans-5-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin- 2(laH)-one (isomer 1 A)(faster eluting): lH NMR (400MHz, CDC13) δ = 7.24 (d, J=8.6 Hz, IH), 7.10 - 7.06 (m, IH), 7.06 - 7.01 (m, IH), 6.73 (t, J=5.3 Hz, IH), 6.47 (s, IH), 6.42 (dd, J=2.0, 8.2 Hz, IH), 4.65 (d, J=5.5 Hz, 2H), 4.13 (dd, J=3.3, 13.1 Hz, IH), 3.86 (s, 3H), 3.78 (s, 3H), 3.67 (d, J=9.4 Hz, IH), 2.87 (t, J=12.3 Hz, IH), 2.72 - 2.62 (m, IH), 2.12 - 2.03 (m, 2H), 2.00 - 1.93 (m, 3H), 1.40 - 1.27 (m, IH), 1.03 - 0.97 (m, IH), 0.71 (d, J=3.9 Hz, IH). Acq Method AS- H_S_3_5_40_3ML_8MIN_15CM.M (Rt = 4.848min, 100%Area). 5-(l -bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin- 2(l aH)-one (isomer IB) (slower eluting). XH NMR (400MHz, CDC13) δ = 7.25 (d, J=8.2 Hz, IH), 7.10 - 7.07 (m, IH), 7.06 - 7.02 (m, IH), 6.74 (t, J=5.3 Hz, IH), 6.48 (s, IH), 6.43 (dd, 3=1.6, 8.2 Hz, IH), 4.65 (d, J=5.5 Hz, 2H), 4.14 (dd, J=3.7, 13.1 Hz, IH), 3.87 (s, 3H), 3.78 (s, 3H), 3.68 (d, J=12.1 Hz, IH), 2.87 (t, J=12.3 Hz, IH), 2.71 - 2.63 (m, IH), 2.12 - 2.04 (m, 2H), 2.01 - 1.92 (m, 3H), 1.39 - 1.30 (m, IH), 1.04 - 0.96 (m, IH), 0.71 (d, J=3.9 Hz, IH). Acq Method AS-H_S_3_5_40_3ML_8MIN_15CM.M (Rt = 4.977min, 99.6%Area).
5-(l -bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l ,5-a]pyrazin-3-yl)hexahydro-lH- cyclopropa[a]indolizin-2(laH)-one (isomer 2) (150 mg, 0.293 mmol) was purified with chiral HPLC [SFC condition: Instrument: SFC-80; Column: AS 250x30mm I.D., 10um; Mobile phase: A: Supercritical C02 , B: MeOH ( 0.1% NH3H20 ) , A:B =50:50 at 80ml/min; Column Temp: 38°C; Nozzle Pressure: l OOBar; Nozzle Temp: 60°C; Evaporator Temp: 20°C; Trimmer Temp: 25°C; Wavelength: 220nm.] to get Peak 1 5-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydro-lH-cyclopropa[a]indolizin- 2(l aH)-one (isomer 2A): XH NMR (400MHz, CDC13) δ = 7.29 - 7.22 (m, IH), 7.15 - 7.06 (m, 2H), 6.73 (t, J=5.1 Hz, IH), 6.48 (s, IH), 6.43 (dd, J=2.0, 8.2 Hz, IH), 4.66 (d, J=5.5 Hz, 2H), 4.19 (dd, J=3.7, 13.1 Hz, IH), 3.87 (s, 3H), 3.79 (s, 3H), 3.47 (d, J=10.6 Hz, IH), 3.00 - 2.89 (m, IH), 2.84 - 2.75 (m, IH), 2.10 (d, J=6.3 Hz, IH), 1.98 (dd, 3=2.7, 12.9 Hz, IH), 1.91 (br. s., IH), 1.76 - 1.67 (m, IH), 1.51 - 1.41 (m, IH), 1.27 - 1.20 (m, IH), 1.13 - 1.06 (m, IH), 0.72 (d, J=3.5 Hz, IH). Acq Method AS-H_S_3_5_40_3ML_8MIN_15CM.M (Rt = 4.030min, 90. l %Area). 5- (l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l ,5-a]pyrazin-3-yl)hexahydro-lH- cyclopropa[a]indolizin-2(laH)-one (isomer 2B): lH NMR (400MHz, CDC13) δ = 7.29 - 7.22 (m, IH), 7.16 - 7.06 (m, 2H), 6.73 (t, J=5.1 Hz, IH), 6.48 (s, IH), 6.43 (dd, J=2.0, 8.2 Hz, IH), 4.66 (d, J=5.5 Hz, 2H), 4.19 (dd, J=3.5, 12.9 Hz, 1H), 3.87 (s, 3H), 3.79 (s, 3H), 3.71 (q, J=6.8 Hz, 1H), 3.47 (d, J=9.4 Hz, 1H), 2.99 - 2.90 (m, 1H), 2.85 - 2.76 (m, 1H), 2.10 (d, J=6.3 Hz, 1H), 1.98 (dd, J=2.7, 12.9 Hz, 1H), 1.90 (br. s., 1H), 1.75 - 1.68 (m, 1H), 1.46 (dq, J=5.3, 12.1 Hz, 1H), 1.26 - 1.20 (m, 1H), 1.13 - 1.05 (m, 1H). Acq Method AS- H_S_3_5_40_3ML_8MIN_15CM.M (Rt = 4.597min, 95.97%Area).
Intermediate 29
Figure imgf000082_0001
(7'R.9a'S)-7'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3- yl)hexahvdrospirorcvclopropane-1.3'-pyridori.2-alpyrazinl-4'(2'H)-one Step 1 : (2S.5R)-tert-butyl 5-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3- yl)-2-(((l-(methoxycarbonyl)cvclopropyl)amino)methyl)piperidine-l-carboxylate
To a solution of (2S,5R)-tert-butyl 5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-2-formylpiperi dine- 1-carboxy late (400 mg, 0.696 mmol) in MeOH (3 mL) was added methyl 1-aminocyclopropanecarboxylate (321 mg, 2.79 mmol), then followed by
NaCNBH3 (131 mg, 2.089 mmol) and acetic Acid (0.1 mL). The mixture was stirred at 18 °C for 2 h. Then to the reaction mixture was added to aq.NaHCC (20 mL), extracted with DCM (25 mLx3). The organic layer was washed with brine, dried over Na2SC>4, concentrated in vacuo and purified by silica gel chromatography (12g, DCM:THF=100%~30%) to give (2S,5R)-tert-butyl 5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-2-(((l- (methoxycarbonyl)cyclopropyl)amino)methyl)piperidine-l-carboxylate. MS (ESI) m/z: 673/675 (M+l). Acq Method 0-60AB_2min_220&254.1cm (Rt =1.233 min). XH NMR (400MHz, CD3OD) δ = 7.40 (d, J=5.1 Hz, 1H), 7.21 (d, J=8.2 Hz, 1H), 7.02 (d, J=5.1 Hz, 1H), 6.57 (s, 1H), 6.44 (dd, J=2.0, 8.2 Hz, 1H), 4.69 - 4.61 (m, 1H), 4.59 - 4.50 (m, 1H), 4.23 (br. s., 1H), 4.10 - 4.03 (m, 1H), 3.89 (s, 3H), 3.77 (s, 3H), 3.69 - 3.65 (m, 3H), 3.49 - 3.40 (m, 1H), 3.34 (br. s., 1H), 3.20 - 3.10 (m, 1H), 2.75 (dd, J=5.9, 11.7 Hz, 1H), 2.59 (br. s., 1H), 2.17 - 2.08 (m, 2H), 1.57 - 1.47 (m, 1H), 1.29 - 1.14 (m, 4H).
Step 2: (7'R.9a'S)-7'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl)hexahvdrospiro[cvclopropane-1.3'-pyrido[1.2-alpyrazinl-4'(2'H)-one A solution of (2S,5R)-tert-butyl 5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-2-(((l-(methoxycarbonyl)cyclopropyl)amino)methyl)piperidine-l-carboxylate (350 mg, 0.520 mmol) in 4M HCl/Dioxane (2.60 mL, 10.39 mmol) was stirred at room temperature (15 °C) for 2 h. The mixture was concentrated in vacuo. The residue was dissolved into MeOH (4 mL) and K2C03 (215 mg, 1.55 mmol) added. The mixture was stirred at 90 °C for 2h, then filtered. The filtrate was concentrated in vacuo, and purified with silica gel column chromatography (THF/dichloromethane 0%~ 80%) to give the (7'R,9a'S)-7'-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)hexahydrospiro[cyclopropane-l,3'- pyrido[l,2-a]pyrazin]-4'(2'H)-one. MS (ESI) m/z: 541/543(M+1). Acq Method 0- 60AB_2min_220&254.1cm (Rt =1.001 min). lH NMR (400MHz, CDC13) δ = 7.50 (d, J=5.1 Hz, 1H), 7.21 (d, J=8.2 Hz, 1H), 7.04 (d, J=5.1 Hz, 1H), 6.58 (s, 1H), 6.47 (d, J=6.3 Hz, 1H), 4.80 (d, J=13.7 Hz, 1H), 4.60 (s, 2H), 3.90 (s, 3H), 3.78 (s, 3H), 3.65 (d, J=11.3 Hz, 1H), 3.18 (t, J=11.7 Hz, 1H), 2.92 - 2.76 (m, 2H), 2.14 (d, J=l 1.3 Hz, 1H), 1.92 (d, J=12.5 Hz, 2H), 1.69 - 1.57 (m, 1H), 1.41 - 1.27 (m, 2H), 1.22 - 1.13 (m, 1H), 0.87 (d, J=2.3 Hz, 2H) ppm.
Intermediate 30
(7'R.9a'S)-7'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3-yl)-2'- methylhexahvdrospiro[cvclopropane-1.3'-pyrido[1.2-alpyrazinl-4'(2'H)-one
Figure imgf000083_0001
To a solution of compound (7'R,9a'S)-7'-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)hexahydrospiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-4'(2'H)-one (120 mg, 0.222 mmol) in MeOH (5 mL) was added formaldehyde (0.165 mL, 2.216 mmol), NaCNBH3 (69.6 mg, 1.108 mmol) and follow by acetic acid (0.2 mL). The mixture was stirred at 25 °C for 2 h, then pH was adjusted to 9 with aq. NaHC03, extracted with DCM (20 mLx3). The organic layer was washed with brine (20 mL), dried over Na2S04, concentrated in vacuo to give the residue, which was purified with silica gel column chromatography (THF/dichloromethane 0%~ 80%) to give (7'R,9a'S)-7'-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3- yl)-2'-methylhexahydrospiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-4'(2'H)-one. MS (ESI) m/z: 555/557(M+l). XH NMR (400MHz, CDC13) δ = 7.58 - 7.55 (m, IH), 7.48 - 7.43 (m, IH), 7.39 (d, J=5.0 Hz, IH), 7.03 (t, J=5.4 Hz, IH), 6.78 (d, J=2.3 Hz, IH), 6.73 (dd, J=2.5, 8.3 Hz, IH), 5.28 - 5.22 (m, IH), 4.96 (d, J=5.5 Hz, 2H), 4.17 (s, 3H), 4.14 (dd, J=2.8, 5.5 Hz, IH), 4.09 (s, 3H), 3.48 - 3.33 (m, 2H), 3.27 (tt, J=3.9, 11.5 Hz, IH), 3.00 (dd, J=11.7, 13.2 Hz, IH), 2.78 (s, 3H), 2.48 - 2.34 (m, 2H), 2.25 - 2.12 (m, 2H), 1.89 - 1.84 (m, IH), 1.43 - 1.36 (m, IH), 1.21 - 1.16 (m, 2H) ppm.
Intermediate 31
(7'R.9a'S)-7'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3-yl)-2'- ethylhexahvdrospirorcvclopropane-1.3'-pyridori.2-alpyrazinl-4'(2'H)-one
Figure imgf000084_0001
To a solution of (7'R,9a'S)-7'-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin- 3-yl)hexahydrospiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-4'(2'H)-one (120 mg, 0.213 mmol) in MeOH (5 mL) was added acetaldehyde (94 mg, 2.128 mmol), NaCNBH3 (66.9 mg, 1.064 mmol) and follow by Acetic Acid (0.2 mL). The mixture was stirred at 25 °C for 2 h, then pH was adjusted to 9 with aq. NaHCC , extracted with DCM (20 mLx3). The organic layer was washed with brine, dried over Na2SC>4, concentrated in vacuo to give the residue, which was purified with silica gel column chromatography (THF/dichloromethane 0%~ 80%) to give (7'R,9a'S)-7'-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-2'- ethylhexahydrospiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-4'(2'H)-one. MS (ESI) m/z:
569/571(M+l). method D (Rt =1.110 min). XH NMR (400MHz, CDCI3) δ = 7.27 (br. s., IH), 7.17 (d, J=5.1 Hz, IH), 7.10 (d, J=4.7 Hz, IH), 6.75 (t, J=5.3 Hz, IH), 6.49 (s, IH), 6.44 (dd, J=2.0, 8.2 Hz, IH), 5.30 (s, 2H), 4.91 (d, J=13.3 Hz, IH), 4.66 (d, J=5.5 Hz, 2H), 4.59 (q, J=6.7 Hz, IH), 3.88 (s, 3H), 3.80 (s, 3H), 3.75 (br. s., IH), 3.30 (dd, J=5.3, 14.3 Hz, IH), 3.04 - 2.93 (m, 2H), 2.75 (d, J=7.4 Hz, IH), 2.71 - 2.63 (m, IH), 2.19 - 2.06 (m, 2H), 1.91 (d, J=13.3 Hz, IH), 1.60 (d, J=6.7 Hz, 3H), 1.46 - 1.38 (m, IH), 1.29 - 1.20 (m, IH), 0.91 (br. s., IH) ppm.
Intermediate 32
Figure imgf000085_0001
4-(8-airdno-3-((3'R.9a'R)-6'-oxohexahydro
cl[l^loxazinl-3'-yl)imidazo[1.5-alpyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridm^ vDbenzamide
Step 1: (2R.5R)-tert-butyl 2-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl -5-(((l-(methoxycarbonyl cvclopropyl amino methyl moφholine-4-carboxylate
To a solution of (2R,5S)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-formylmo holine-4-carboxylate (200 mg, 0.347 mmol) in MeOH (3 mL) was added methyl 1-aminocyclopropanecarboxylate (160 mg, 1.388 mmol), then followed by NaCNBH3 (65.4 mg, 1.041 mmol) and acetic Acid (0.5 mL). The mixture was stirred at 25 °C for 2 h, then pH was adjusted to 9 with aq. NaHC03, extracted with DCM (20 mLx3). The organic layer was washed with brine, dried over Na2SC>4, concentrated in vacuo and purified by silica gel chromatography (4g, DCM:THF=100%~30%) to give (2R,5R)-tert-butyl 2-(l-bromo- 8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-5-(((l- (methoxycarbonyl)cyclopropyl)amino)methyl)morpholine-4-carboxylate. MS (ESI) m/z:
675/677(M+l). method D (Rt =1.225 min). XH NMR (400MHz, CDC13) = 7.44 (d, J=4.7 Hz, 1H), 7.09 (d, J=4.7 Hz, 1H), 6.75 (br. s., 1H), 6.49 (s, 1H), 6.44 (d, J=8.2 Hz, 1H), 5.00 (br. s., 1H), 4.67 (d, J=5.1 Hz, 2H), 3.88 (s, 4H), 3.80 (s, 3H), 3.69 (s, 3H), 3.64 (d, J=11.7 Hz, 1H), 3.50 (dd, J=4.1, 13.9 Hz, 1H), 3.26 (d, J=9.0 Hz, 1H), 3.19 - 3.10 (m, 1H), 3.02 - 2.92 (m, 1H), 1.57 - 1.50 (m, 9H), 1.50 - 1.41 (m, 2H), 1.32 - 1.22 (m, 2H), 0.98 (d, J=3.5 Hz, 1H).
Step 2: (3'R.9a'R)-3'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3- yl)tetrahvdro- H-spirorcvclopropane-1.7'-pyrazinor2.1-ciri.41oxazinl-6'(8'H)-one
A solution of (2R,5R)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-(((l-(methoxycarbonyl)cyclopropyl)amino)methyl)morpholine-4-carboxylate (80 mg, 0.118 mmol) in 4M HCl/Dioxane (0.030 mL, 0.118 mmol) was stirred at room temperature (15 °C) for 2 h. The mixture was concentrated in vacuo. The residue was dissolved into MeOH (2 mL) and treated with K2C03 (49.1 mg, 0.355 mmol) at 90 °C for 2 hours. The mixture was filtered and filtrate concentrated in vacuo. The residue was purified with silica gel chromatography (THF/dichloromethane 0%~ 80%) to give (3'R,9a'R)-3'-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)tetrahydro-rH-spiro[cyclopropane-l,7'- pyrazino[2,l-c] [l,4]oxazin]-6'(8'H)-one. MS (ESI) m/z: 543/545(M+l). method D (Rt =1.091 min). XH NMR (400MHz, CDC13) d = 7.29 (br. s., 1H), 7.15 (d, J=4.7 Hz, 1H), 6.80 (br. s., 1H), 6.51 (s, 1H), 6.45 (d, J=8.2 Hz, 1H), 4.97 (dd, J=2.3, 13.7 Hz, 1H), 4.73 (dd, J=2.5, 10.8 Hz, 1H), 4.68 (d, J=5.5 Hz, 2H), 4.01 (dd, J=3.1, 11.3 Hz, 1H), 3.89 (s, 3H), 3.81 (s, 3H), 3.53 - 3.46 (m, 2H), 3.36 - 3.30 (m, 1H), 3.25 (dd, J=4.7, 13.3 Hz, 1H), 3.17 - 3.06 (m, 1H), 2.85 (dd, J=9.6, 13.9 Hz, 1H), 1.24 - 1.19 (m, 1H), 0.98 - 0.90 (m, 1H), 0.86 (dd, J=6.5, 10.0 Hz, 1H) ppm.
Figure imgf000086_0001
Trans, isomer 1
Trans, isomer 2
33
34
7'-(8-ammo- 1 -bromoimidazof .1.3-a]pyrazin-3-yl)hexahy drospiro cy clopropane- 1. Γ-pyridof 1.2- a]pyrazin]-4'(6'H)-one (trans isomer 1), and 7'-{8-aii¾no-l--bromoiimdazoj l,5-a^
y i)hexah drospirof cy clopropane-1 , 1 '-pyridol'l ^-ajpyrazin j-4'(6'H)-one (trans isomer 2) Step 1 : benzyl benzyl (l-(5-bromopyridin-2-yl)cvclopropyl)carbamate
To a solution of l-(5-bromopyridin-2-yl)cyclopropanecarboxylic acid (15 g, 62.0 mmol) in Toluene (150 mL) was added TEA (12.96 mL, 93 mmol) and DPPA (34.1 g, 124 mmol) and stirred at 15°C for 30 mins, then heated to 90°C for 2 hours. After cooling to room temperature, then to the reaction mixture was added benzylalcohol (33.5 g, 310 mmol) and mixture stirred at 90°C for 3 hours. After cooling to room temperature, the reaction mixture washed with a saturated NaHCC aqueous solution (50mLx4). The organic layer was concentrated under reduced pressure, and the residue was purified by flash chromatography (ethyl acetate/Pet.
ether= 0%~50%) to give crude product. The residue was suspended in Pet. Ether and filtered to give benzyl benzyl (l-(5-bromopyridin-2-yl)cyclopropyl)carbamate.
XH NMR (400MHz, CDC13) δ = 8.47 (br. s., 1H), 7.71 - 7.56 (m, 1H), 7.37 (br. s., 4H), 7.24 - 7.02 (m, 2H), 5.50 (br. s., 1H), 5.12 (s, 2H), 1.61 (br. s., 2H), 1.35 - 1.21 (m, 2H) ppm.
Step 2: methyl 6-(l-(((benzyloxy)carbonyl)amino)cvclopropyl)nicotinate To a solution of benzyl (l-(5-bromopyridin-2-yl)cyclopropyl)carbamate (6.0 g, 17.28 mmol) in MeOH (100 mL) and DMF (100 mL) was added TEA (7.23 mL, 51.8 mmol), DPPF (3.83 g, 6.91 mmol) and Pd(OAc)2 (0.776 g, 3.46 mmol). The mixture was stirred at 80°C for 48 hours under CO (50psi), then filtered, and the filtrate was concentrated in vacuo. The residue was partitioned between water (100 mL) and EA (300 mL). The organic layer was washed with water (50mLx3), brine (100 mL) dried over Na2SC>4, and concentrated in vacuo. The residue was purified by silica gel column chromatography (EtOAc: Pet. Ether =0%~40%) to afford methyl 6- (l-(((benzyloxy)carbonyl)amino)cyclopropyl)nicotinate.1H NMR (400MHz, CDC13) δ = 9.05 (d, J=1.5 Hz, 1H), 8.18 (d, J=7.5 Hz, 1H), 7.48 - 7.30 (m, 5H), 7.26 - 7.14 (m, 1H), 5.56 (br. s., 1H), 5.19 - 5.13 (m, 2H), 3.94 (s, 3H), 1.81 - 1.72 (m, 2H), 1.39 (br. s., 2H) ppm.
Step 3: methyl 6-(l-(((benzyloxy)carbonyl)amino)cvclopropyl)piperidine-3-carboxylate
To a solution of methyl 6-(l-(((benzyloxy)carbonyl)amino)cyclopropyl)nicotinate (700 mg, 2.145 mmol) in AcOH (10 mL) was added sodium cyanoborohydride (472mg, 7.51 mmol) in portions. The mixture was stirred at 30°C for 24 hours, then poured into a saturated aqueous NaHCO3(50 mL). The mixture was extracted with EtOAc (30mLx4). The combined organic layers were washed with brine (50 mL) The extracts were dried over Na2S04, filtered and concentrated under reduced pressure to give crude methyl 6-(l-
(((benzyloxy)carbonyl)amino)cyclopropyl)piperidine-3-carboxylate. MS: 333.3 (M+l).
Step 4: methyl 6-(l-(((benzyloxy)carbonyl)amino)cvclopropyl)-l-(2-chloroacetyl)
piperidine-3-carboxylate
To a solution of methyl 6-(l-(((benzyloxy)carbonyl)amino)cyclopropyl)piperidine-3-carboxylate (600 mg, 1.805 mmol) in THF (6 mL) was added TEA (0.277 mL, 1.986 mmol) and 2- chloroacetyl chloride (224 mg, 1.986 mmol). The mixture was stirred at 0°C for 3h. Then the reaction mixture was diluted with EtOAc (lOmL) and washed with water (5mLx3), brine(10 mL) dried over Na2S04, and concentrated in vacuo. The residue was purified by silica gel column chromatography (12g, THF: Pet. ether=0%~40%) to afford methyl 6-(l- (((benzyloxy)carbonyl)amino)cyclopropyl)-l-(2-chloroacetyl)piperidine-3-carboxylate. 'H NMR (400MHz, CDC13) δ = 7.34 (br. s., 5H), 5.29 - 5.14 (m, 1H), 5.03 (br. s., 2H), 4.41 (d, J=12.9 Hz, 1H), 4.19 (br. s., 1H), 4.04 - 3.90 (m, 1H), 3.72 - 3.65 (m, 3H), 3.61 (d, J=11.7 Hz, 1H), 2.72 - 2.50 (m, 1H), 1.98 (br. s., 1H), 1.84 - 1.62 (m, 3H), 1.14 - 0.69 (m, 5H) ppm.
Step 5: 2'-benzyl 7'-methyl 4'-oxohexahvdrospirorcvclopropane-l. r-pyrido|T.2- alpyrazinel-2'.7'(6'H)-dicarboxylate (trans) To a solution of methyl 6-(l-(((benzyloxy)carbonyl)amino)cyclopropyl)-l-(2- chloroacetyl)piperidine-3-carboxylate (100 mg, 0.245 mmol) in DMF (2 mL) was added CS2CO3 (239 mg, 0.734 mmol). The mixture was stirred at 70 °C for 2 hours, then poured into water (10 mL) and extracted with EtOAc (10mLx4). The combined organic layers were washed with water (5mLx3), brine (10 mL) dried over Na2SC>4, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (4g, EtOAc: Pet. ether=0%~80%) to afford trans-2'-benzyl 7'-methyl 4'-oxohexahydrospiro[cyclopropane-l,l'-pyrido[l,2-a]pyrazine]- 2',7'(6'H)-dicarboxylate. XH NMR (400MHz, CD3OD) δ = 7.36 (br. s., 5H), 5.12 (br. s., 2H), 4.78 (dd, J=2.0, 12.9 Hz, 1H), 4.51 (d, J=17.2 Hz, 1H), 3.86 (d, J=8.6 Hz, 1H), 3.67 (s, 3H), 2.76 (br. s., 1H), 2.62 (t, J=12.5 Hz, 1H), 2.38 (ddd, J=4.1, 8.2, 11.9 Hz, 1H), 2.10 (d, J=11.0 Hz, 1H), 1.82 (br. s., 1H), 1.68 - 1.54 (m, 1H), 1.53 - 1.32 (m, 2H), 1.18 - 1.05 (m, 1H), 0.94 - 0.77 (m, 2H) ppm.
Step 6: 2'-((benzyloxy)carbonyl)-4'-oxooctahvdrospirorcvclopropane-l. -pyridori.2- alpyrazinel-7'-carboxylic acid (trans)
To a solution of 2'-benzyl 7'-methyl 4'-oxohexahydrospiro[cyclopropane-l, -pyrido[l,2- a]pyrazine]-2',7'(6'H)-dicarboxylate (120 mg, 0.322 mmol) in THF (3 mL) was added lithium hydroxide in H2O (0.483 mL, 0.483 mmol). The mixture was stirred at 15°C for 1.5h. The reaction mixture was diluted with water (lOmL). The pH of the reaction mixture was adjusted to 3 with aq. HC1 (1M). The mixture was extracted with EtOAc (10mLx4). Then the organic layer was washed with brine (30 mL), dried over Na2SC>4, and concentrated under reduced pressure to afford 2'-((benzyloxy)carbonyl)-4'-oxooctahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]- 7'-carboxylic acid. ¾ NMR (400MHz, CDC13) δ = 7.35 (br. s., 5H), 5.12 (br. s., 2H), 4.95 (d, J=12.5 Hz, 1H), 4.74 - 4.48 (m, 1H), 3.85 (br. s., 1H), 2.58 (t, J=12.5 Hz, 2H), 2.52 - 2.40 (m, 1H), 2.28 - 2.13 (m, 1H), 1.87 (br. s., 1H), 1.70 - 1.44 (m, 3H), 1.04 (br. s., 1H), 0.96 - 0.72 (m, 2H) ppm.
Step 7: benzyl 7'-(((3-chloropyrazin-2-yl)methyl)carbamoyl)-4'-oxohexahydrospiro[
cvclopropane-l. -pyrido[1.2-alpyrazinel-2'(6'H)-carboxylate (trans)
To a solution of 2'-((benzyloxy)carbonyl)-4'-oxooctahydrospiro[cyclopropane-l, -pyrido[l,2- a]pyrazine]-7'-carboxylic acid (100 mg, 0.279 mmol)in DMF (3 mL) was added HATU (117 mg, 0.307 mmol), DIEA (0.146 mL, 0.837 mmol) and (3-chloropyrazin-2-yl)methanamine hydrochloride (55.3 mg, 0.307 mmol) and the mixture was stirred for 16 hrs at 15°C. The mixture was diluted with water (15 mL) and extracted with ethyl acetate (10 mL χ 4). The combined organic layers were washed with water (5 mL χ 3), brine (10 mL), dried over Na2SC>4, and evaporated to give the crude product which was purified on flash chromatography (Pet.
ether/EtOAc = 100% ~ 60 %) to give benzyl 7'-(((3-chloropyrazin-2-yl)methyl)carbamoyl)-4'- oxohexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate. XH NMR (400MHz, CDC13) δ = 8.45 (d, J=2.0 Hz, 1H), 8.34 (s, 1H), 7.35 (s, 5H), 6.92 (br. s., 1H), 5.13 (br. s., 2H), 4.90 - 4.80 (m, 1H), 4.73 - 4.53 (m, 3H), 3.84 (br. s., 1H), 3.22 - 2.98 (m, 1H), 2.75 - 2.51 (m, 2H), 2.39 (t, J=11.5 Hz, 1H), 2.04 (s, 1H), 1.92 - 1.86 (m, 1H), 1.57 - 1.44 (m, 2H), 1.06 (br. s., 1H), 0.91 - 0.75 (m, 2H) ppm.
Step 8: benzyl 7'-(8-chloroimidazo[1.5-alpyrazin-3-yl)-4'-oxohexahvdrospiro[
cvclopropane-l. -pyridori.2-alpyrazinel-2'(6'H)-carboxylate (trans)
To a stirred solution of benzyl 7'-(((3-chloropyrazin-2-yl)methyl)carbamoyl)-4'- oxohexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate (1.8g, 3.72 mmol) in Acetonitrile (18 mL) was added PCI5 (0.775 g, 3.72 mmol), and the reaction mixture was stirred at 15°Cfor 3 hours. The reaction mixture was poured slowly to a saturated aq.
NaHC03 (100 mL) at 0°C. The mixture was extracted with ethyl acetate (50 mL χ 4). The combined organic layers were washed with brine (10 mL), dried over Na2SC>4, evaporated to give a product which was purified on flash chromatography (20g Pet. ether/EtOAc = 100% ~ 20 %) to give benzyl 7'-(8-chloroirnidazo[l,5-a]pyrazin-3-yl)-4'-oxohexahydrospiro[cyclopropane- l,r-pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate. lH NMR (400MHz, CD3OD) δ = 8.18 (d, J=4.7 Hz, 1H), 7.86 (s, 1H), 7.46 - 7.27 (m, 6H), 5.16 (br. s., 2H), 4.80 (d, J=11.3 Hz, 1H), 4.60 (d, J=15.7 Hz, 1H), 3.94 (br. s., 1H), 2.96 (d, J=12.5 Hz, 1H), 2.81 (s, 2H), 2.16 (br. s., 1H), 1.98 (d, J=11.0 Hz, 2H), 1.56 (br. s., 2H), 1.20 (d, J=8.2 Hz, 1H), 0.98 - 0.87 (m, 2H) ppm.
Step 9: benzyl 7'-(l-bromo-8-chloroimidazori.5-alpyrazin-3-yl)-4'- oxohexahydrospiro [cyclopropane- 1.1 '-pyrido Γ 1.2-al pyrazinel -2'(6'H)-carboxylate (trans)
To a solution of benzyl 7'-(8-chloroimidazo[l,5-a]pyrazin-3-yl)-4'- oxohexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate (1.4g, 3.00 mmol) in acetonitrile (14 mL) was added NBS (0.588 g, 3.31 mmol). The resulting mixture was stirred at 15°C for 1 hour, then poured into a saturated aqueous NaHCCb (10 mL) and extracted with EtOAc (10 mLx3). The combined organic layers were washed with brine (10 mL), dried over Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography(4g Pet. ether/EtOAc=l 00%~0%) to afford benzyl 7'-(l -bromo-8- chloroimidazo[l,5-a]pyrazin-3-yl)-4'-oxohexahydrospiro[cyclopropane-l, -pyrido[l,2- a]pyrazine]-2'(6'H)-carboxylate. XH NMR (400MHz, CD3OD) δ = 8.15 (d, J=4.7 Hz, 1H), 7.50 - 7.22 (m, 6H), 5.15 (br. s., 2H), 4.76 (d, J=12.9 Hz, 1H), 4.58 (d, J=17.6 Hz, 1H), 3.93 (br. s., 1H), 2.96 - 2.89 (m, 1H), 2.84 (s, 2H), 2.15 (br. s., 1H), 1.95 (d, J=11.0 Hz, 2H), 1.73 - 1.50 (m, 2H), 1.25 - 1.14 (m, 1H), 0.92 (dd, J=3.3, 6.1 Hz, 2H) ppm.
Step 10: benzyl 7'-(l-bromo-8-((2.4-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3-yl)-4'- oxohexahvdrospiro[cvclopropane-l. -pyrido[1.2-alpyrazinel-2'(6'H)-carboxylate (trans isomer 1} and benzyl 7'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3-yl)-4'- oxohexahvdrospirorcvclopropane-l. -pyridori.2-alpyrazinel-2'(6'H)-carboxylate (trans isomer }
To a solution of benzyl 7'-(l-bromo-8-chloroimidazo[l,5-a]pyrazin-3-yl)-4'- oxohexahy drospiro [cyclopropane- 1 , 1 '-pyrido [ 1 ,2-a] py razine] -2'(6'H)-carboxy late (900 mg, 1.652 mmol) in DMF (9 mL) was added K2C03 (685 mg, 4.96 mmol) and (2,4- dimethoxyphenyl)methanamine (414 mg, 2.478 mmol). The mixture was stirred at 90°C for 2 hours. The reaction mixture was diluted with EtOAc (lOOmL) then washed with water (10 mL χ 4) and brine (40mL), dried over Na2SC>4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (4g Pet. ether/THF=100%~30%) and SFC (Instrument : Thar SFC 200; Column : AS 300mmx50mm, lOum; Mobile phase: A:
Supercritical C02 , B: MeOH(0.1% NH3H20), A:B =55:45 at 200ml/min; Column Temp: 38°C; Nozzle Pressure: lOOBar; Nozzle Temp: 60°C; Evaporator Temp: 20°C; Trimmer Temp: 25°C; Wavelength: 220nm) to give two trans isomers. Benzyl 7'-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-4'-oxohexahydrospiro[cyclopropane-l,r- pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate(trans enantiomer 1): Rt=4.820 min H NMR
(400MHz, CD3OD) δ = 7.44 (d, J=5.1 Hz, 1H), 7.41 - 7.26 (m, 5H), 7.19 (d, J=8.2 Hz, 1H), 7.02 (d, J=5.1 Hz, 1H), 6.56 (d, J=2.0 Hz, 1H), 6.45 (dd, J=2.3, 8.2 Hz, 1H), 5.12 (br. s., 2H), 4.71 (d, J=13.3 Hz, 1H), 4.57 (s, 3H), 3.87 (s, 3H), 3.76 (s, 3H), 3.19 - 3.11 (m, 1H), 2.93 - 2.77 (m, 2H), 2.07 (br. s., 1H), 1.88 (d, J=11.0 Hz, 2H), 1.54 (d, J=10.2 Hz, 3H), 1.17 - 1.13 (m, 1H), 0.89 (d, J=7.4 Hz, 2H) ppm. MS: 677.2(M+1). Benzyl 7'-(l-bromo-8-((2,4- dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)-4'-oxohexahydrospiro[cyclopropane-l,r- pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate (trans enantiomer 2). Rt=5.643 min. 'H NMR (400MHz, CD3OD) δ = 7.50 - 7.27 (m, 6H), 7.21 (d, J=8.2 Hz, 1H), 7.03 (d, J=5.1 Hz, 1H), 6.57 (d, J=2.0 Hz, 1H), 6.46 (dd, J=2.3, 8.2 Hz, 1H), 5.14 (br. s., 2H), 4.73 (d, J=11.3 Hz, 1H), 4.59 (s, 3H), 3.89 (s, 3H), 3.78 (s, 3H), 3.20 - 3.12 (m, 1H), 2.95 - 2.79 (m, 2H), 2.09 (br. s., 1H), 1.96 - 1.83 (m, 2H), 1.72 - 1.42 (m, 3H), 1.17 (d, J=7.0 Hz, 1H), 0.90 (d, J=7.4 Hz, 2H) ppm. MS: 677.2(M+1).
Step 11 : benzyl7'-(8-amino-l-bromoimidazo[1.5-alpyrazin-3-yl)-4'-oxohexahydrospiro[ cvclopropane-l. -pyrido[1.2-alpyrazinel-2'(6'H)-carboxylate (trans isomer 1) and benzyl7'-(8- arnino-l-bromoimidazo[1.5-alpyrazin-3-yl)-4'-oxohexahvdrospiro[cvclopropane-l. -pyrido[1.2- alpyrazinel-2'(6'H)-carboxylate (trans isomer 2)
A solution of benzyl 7'-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)- 4'-oxohexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate (trans isomer 1) (100 mg, 0.148 mmol) in TFA (3 mL) was stirred at 90 °Cfor 2 hours under N2 atmosphere. The mixture was concentrated in vacuo to give 7'-(8-amino-l-bromoimidazo[l,5- a]pyrazin-3-yl)hexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazin]-4'(6'H)-one as TFA salt. MS : 391.3/393.3 (M+l).
Starting from benzyl 7'-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3-yl)- 4'-oxohexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate (trans isomer 2), benzyl7'-(8-amino-l-bromoimidazo[l,5-a]pyrazin-3-yl)-4'- oxohexahydrospiro[cyclopropane-l, -pyrido[l,2-a]pyrazine]-2'(6'H)-carboxylate (trans isomer 2) was obtained using the same procedure. MS : 391.3/393.3 (M+l).
Intermediate 35
Figure imgf000091_0001
(3'R.9a'R)-3'-(l-bromo-8-((2^-dimethoxybenzyl)amino)imidazori.5-alpyrazin-3-yl)tetrahvdro- H-spirorcvclopentane-1.7'-pyrazinor2.1-ci ri.41oxazinl-6'(8'H)-one
Step l :(2R.5R)-tert-butyl 2-(l¾omo-8-((2^-dimethoxybenzyl)amino)imidazo[1.5-alpyrazin-3- yl -5-(((l-(methoxycarbonyl cvclopentyl amino methyl moφholine-4-carboxylate
To a solution of (2R,5S)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-formylmo holine-4-carboxylate (120 mg, 0.208 mmol), methyl 1- aminocyclopentanecarboxylate (44.7 mg, 0.312 mmol), and sodium cyanoborohydride (39.2 mg, 0.625 mmol) in DCM (3 mL) was added acetic acid (0.1 mL). The mixture was stirred at 20 °C for 12 hrs under N2, then poured into 20 mL of sat. aq. NaHCC , and extracted with DCM (10 mL x 3). The organic layer was washed with brine (10 mL), dried over Na2SC>4, evaporated to get the crude product, which was purified by flash chromatography (Pet. ether/THF= 65 ~ 50 %) to afford the title compound. lH NMR (400MHz, CDC13) δ = 7.42 (d, J=5.1 Hz, 1H), 7.26 - 7.23 (m, IH), 7.07 (d, J=5.1 Hz, IH), 6.73 (t, J=5.3 Hz, IH), 6.47 (s, IH), 6.42 (dd, J=2.0, 8.2 Hz, IH), 4.98 (d, J=3.5 Hz, IH), 4.71 - 4.61 (m, 3H), 3.85 (s, 4H), 3.80 - 3.75 (m, 3H), 3.70 (s, 3H), 3.67 (br. s., IH), 3.44 (dd, J=4.5, 13.9 Hz, IH), 3.25 (dd, J=2.9, 11.5 Hz, IH), 2.76 (dd, J=3.1, 7.0 Hz, 2H), 2.11 - 2.02 (m, 2H), 1.84 - 1.81 (m, 2H), 1.77 - 1.71 (m, 2H), 1.67 (br. s., 9H), 1.60 - 1.55 (m, 2H) ppm.
Step 2: (3'R.9a'R)-3'-(l-bromo-8-((2^-dimethoxybenzvnariiino)imidazori.5-alpyrazin-3- yl)tetrahvdro- H-spirorcvclopentane-1.7'-pyrazinor2.1-ciri.41oxazinl-6'(8'H)-one
A mixture of (2R,5R)-tert-butyl 2-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5- a]pyrazin-3-yl)-5-(((l-(methoxycarbonyl)cyclopenfyl)ariiino)methyl)morpholine-4-carboxylate (50 mg, 0.047 mmol) in 4M HCl/Dioxane (2 mL, 8.00 mmol) was stirred at 20 °C for 2 hours. After removal of all solvent, the residue was dissolved with sat. aq. NaHCC (10 mL) and extracted with DCM/i-PrOH (10 mL χ 3). The organic layer was washed with brine (10 mL), dried over Na2S04, evaporated. The residue was dissolved in MeOH (3 mL) and heated at 80 °C for 12 hours under N2. The mixture was concentrated to afford the title compound, which was used in the next step directly. XH NMR (400MHz, CDCI3) δ = 7.22 (br. s., IH), 7.18 - 7.16 (m, IH), 7.06 (d, J=5.1 Hz, IH), 6.72 (br. s., IH), 6.43 (br. s., IH), 6.38 (d, J=8.2 Hz, IH), 4.86 (d, J=16.0 Hz, IH), 4.67 - 4.58 (m, 3H), 3.92 - 3.87 (m, IH), 3.81 (s, 3H), 3.74 (s, 3H), 3.69 - 3.61 (m, 2H), 3.38 (t, J=11.0 Hz, IH), 3.18 (dd, J=11.0, 13.7 Hz, IH), 3.08 (dd, J=4.9, 13.5 Hz, IH), 2.71 (dd, J=9.0, 13.3 Hz, IH), 2.31 (d, J=5.9 Hz, IH), 1.98 (br. s., 2H), 1.74 (d, J=12.1 Hz, 2H), 1.69 (d, J=7.4 Hz, 2H).ppm
Example 1
Figure imgf000092_0001
1
4-(8-ariiino-3-(2-oxooctahvdro-lH-cvcloproparalindolizin-5-yl)imidazori.5-alpyrazin-l-yl)-3- ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (1) Step 1 : 4-(8-((2^-dimethoxybenzyl)amino)-3-(2-oxooctahydro-lH-cvclopropa[alindolizin-5- yl)imidazo[1.5-alpyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (isomer 1A)
To a solution of 5-(l-bromo-8-((2,4-dimethoxybenzyl)amino)imidazo[l,5-a]pyrazin-3- yl)hexahydro-lH-cyclopropa[a]indolizin-2(laH)-one (isomer 1A) (30 mg, 0.059 mmol) in 2.5 mL of DMF/dioxane/ H20 (2:2: 1) was added 3-ethoxy-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-N-(5-(trifluoromethyl)pyridin-2-yl)benzamide (30.6 mg, 0.070 mmol), K2C03 (24.28 mg, 0.176 mmol) and PdCl2(dppf) (4.28 mg, 5.85 μηιοΐ) under nitrogen protection. Then the mixture was heated to 80 °C for 4 hour, then poured into H20 (10 mL), extracted with DCM (15 mLx3). The organic layer was washed with brine, dried over Na2SC>4, concentrated in vacuo to give the crude product 4-(8-((2,4-dimethoxybenzyl)amino)-3-(2- oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(5- (trifluoromethyl)pyridin-2-yl)benzamide used to the next step without further purification. MS (ESI): 742.2 (M+l). method D (Rt =1.310 min).
Step 2: 4-(8-andno-3-(2-oxooctahvdro-lH-cvclopropa[alindolizin-5-yl)imidazo[1.5-alpyrazin-l- yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (isomer 1A)
A solution of 4-(8-((2,4-dimethoxybenzyl)amino)-3-(2-oxooctahydro-lH- cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide (55 mg, 0.052 mmol) in TFA (3 mL) was stirred at 90 °C for 2 h. TLC showed the starting materials was consumed and concentrated in vacuo. The residue was purified with preparative HPLC with Waters XSELECT C18 150x30mmx5um. Gradient: 22-52 % 15 min; FlowRate:25 ml/min to get the product 4-(8-amino-3-(2- oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(4- (trifluoromethyl)pyridin-2-yl)benzamide (isomer 1 A). l NMR(400MHz, CD3OD) δ = 8.67 - 8.63 (m, 2H), 7.86 (d, J=6.0 Hz, 1H), 7.81 - 7.77 (m, 2H), 7.73 - 7.69 (m, 1H), 7.49 - 7.46 (m, 1H), 7.01 (d, J=6.0 Hz, 1H), 4.28 (q, J=6.9 Hz, 2H), 4.16 (d, J=10.3 Hz, 1H), 3.91 - 3.83 (m, 1H), 3.20 - 3.08 (m, 2H), 2.23 - 2.13 (m, 3H), 2.04 - 1.94 (m, 2H), 1.62 - 1.51 (m, 1H), 1.36 (t, J=6.9 Hz, 3H), 1.12 (dt, J=4.8, 8.0 Hz, 1H), 0.87 - 0.81 (m, 1H) ppm. MS (ESI): 592.1 (M+l). method C (Rt =2.567 min). Chiral SFC: Acq Method OD-H_3_5_40_2,35ML.M (Rt =
9.417min, 99.65%Area).
Example 2
4-(8-alτ^no-3-((6'S■8a'R -l -difluoro-3'-oxohexahvdro-lΉ-spiro[cvclopropane-l■2'-indolizinl- 6'-yl)inddazo[1.5-alpyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide (2) and 4-(8-amino-3-((6'R.8a'S)- . -difluoro-3'-oxohexahvdro- H-spiro[cvclopropane-1.2'- indolizinl-6'-yl)inTidazo[1.5-alpyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2- vDbenzamide (3)
Figure imgf000094_0001
2 3
To a 40 mL vial was added (6'R,8a'S)-6'-(8-amino-l-bromoimidazo[l,5-a]pyrazin-3-yl)- , - difluorotetrahydro- H-spiro[cyclopropane-l,2'-indolizin]-3'(5'H)-one (50 mg, 0.121 mmol) , 3- ethoxy-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridin-2- yl)benzamide (79 mg, 0.182 mmol), and PdCl2(dppf)- CH2Cl2Adduct (9.91 mg, 0.012 mmol). The flask was degassed by vacuum and re-filled with N2. Then K2CO3 (2M, 243 μΐ, 0.485 mmol) and Dioxane (2426 μΐ) were added. The reaction was stirred at 80 °C for 2 hours.
After cooling down to room temperature, the solution was diluted with EtOAc and dried over anhydrous Na2SC>4, filtered, and concentrated. The product was purified by S1O2
chromatography (12 g, EtOAc in Hexane 0% to 100%) to give a mixture of the title compounds. The two isomers were separated by chiral SFC separation to give the two title compounds.
SFC conditions: Column: AS-H (2 x 25 cm); Mobile pahse: 30% methanol(0.1% DEA)/C02, 100 bar, 60 mL/min, 220 nm ; inj vol.: 0.75 mL , 7 mg/mL methanol.
Example 3
4-(8-amino-3-((6'R.8a'S)-3'-oxohexahvdro-rH-spiro[cvclopropane-1.2'-indolizinl-6'- yl)inddazo[1.5-alpyrazin-l-yl)-3-ethoxy-5-fluoro-N-(4-(trifluoromethyl)pyridin-2-yl)benzarm
Figure imgf000094_0002
4 To a 8 niL vial was added 3-ethoxy-5-fluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-N- (4-(trifluoromethyl)pyridin-2-yl)benzamide (24.14 mg, 0.053 mmol), (6'R,8a'S)-6'-(8-amino-l- bromoiniidazo[l,5-a]pyrazin-3-yl)tetrahydro-lH-spiro[cyclopropane-l,2'-indolizin]-3 5'H)-one (10 mg, 0.027 mmol), and PALLADIUM(II) ACETATE/1, l'-BIS(DI-T- BUTYLPHOSPHINO)FERROCENE/POTASSIUM PHOSPHATE ADMIXTURE (4.84 mg, 5.32 μηιοΐ). The flask was degassed by vacuum and re-filled with N2 . Then K2CO3 (2M, 39.9 μΐ, 0.080 mmol) and Dioxane (532 μΐ) were added. The reaction was stirred at 90 °C for 16 hours. The solvent was removed by blowing N2 for 30 min. The residue was dissolved in 2 mL DMF and purified by C18 column (Gilson, CH3CN in water with 0.1% TFA: 0% to 90%) to the title compound.
The following compounds were prepared based on the same procedures as described in examples 1, 2, and 3 using different bromide and boronic ester intermediates for the Suzuki coupling step.
Table 1
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Biological Activity
The Btk inhibitor compounds of the invention having Formula I inhibit the Btk kinase activity. All compounds of the invention have an IC50 of 10 μΜ or lower. In another aspect the invention relates to compounds of Formula I which have an IC50 of less than 100 nM. In yet another aspect the invention relates to compounds of Formula I which have an IC50 of less than 10 nM.
The term IC50 means the concentration of the test compound that is required for 50% inhibition of its maximum effect in vitro.
Btk enzyme activity Assay Methods
BTK enzymatic activity was determined with the LANCE (Lanthanide Chelate Excite) TR-FRET (Time-resolved fluorescence resonance energy transfer) assay. In this assay, the potency (IC50) of each compound was determined from an eleven point (1 :3 serial dilution; final compound concentration range in assay from 1 μΜ to 0.017 nM) titration curve using the following outlined procedure. To each well of a black non-binding surface Corning 384-well microplate (Corning Catalog #3820), 5 nL of compound (2000 fold dilution in final assay volume of 10 μί) was dispensed, followed by the addition of 7.5 of lx kinase buffer (50 mM Hepes 7.5, 10 mM MgCl2, 0.01% Brij-35, 1 mM EGTA, 0.05% BSA & 1 mM DTT) containing 5.09 ^/μΐ^ (66.67 pM) of BTK enzyme (recombinant protein from baculovirus-transfected Sf9 cells: full-length BTK, 6HIS-tag cleaved). Following a 60 minute compound and enzyme incubation, each reaction was initiated by the addition of 2.5 lx kinase buffer containing 8 μΜ biotinylated "A5" peptide (Biotin-EQEDEPEGDYFEWLE-NH2) (SEQ.ID.NO.: 1), and 100 μΜ ATP. The final reaction in each well of 10 μί consists of 50 pM ABTK, 2 μΜ biotin-A5- peptide, and 25 μΜ ATP. Phosphorylation reactions were allowed to proceed for 120 minutes. Reactions were immediately quenched by the addition of 20 uL of lx quench buffer (15 mM EDTA, 25 mM Hepes 7.3, and 0.1% Triton X-100) containing detection reagents (0.626 nM of LANCE-Eu-W1024-anti-phosphoTyrosine antibody, PerkinElmer and 86.8 nM of Streptavidin- conjugated Dylight 650, Dyomics/ThermoFisher Scientific). After 60 minutes incubation with detection reagents, reaction plates were read on a PerkinElmer EnVision plate reader using standard TR-FRET protocol. Briefly, excitation of donor molecules (Eu-chelate:anti-phospho- antibody) with a laser light source at 337 nm produces energy that can be transferred to Dylight- 650 acceptor molecules if this donor: acceptor pair is within close proximity. Fluorescence intensity at both 665 nm (acceptor) and 615 nm (donor) are measured and a TR-FRET ratio calculated for each well (acceptor intensity/donor intensity). IC50 values were determined by 4 parameter robust fit of TR-FRET ratio values vs. (Logio) compound concentrations.
The following Table 2 provides specific IC50 values for all the examples. The IC50 values set forth below were determined according to Assay method described above.
Table 2. BTK binding potency
Figure imgf000109_0001
7 0.4252 27 1 .598 47 0.5283
8 2.066 28 1 .539 48 1 .188
9 1 .621 29 1 .512
10 0.9515 30 1 .61 1
11 0.7013 31 0.4102
12 0.3845 32 6.225
13 1 .223 33 9.041
14 1 .281 34 6.952
15 0.4622 35 1 .255
16 0.489 36 1 .004
17 0.5006 37 0.1856
18 0.1271 38 0.2221
19 3.809 39 0.3217
20 0.1708 40 0.1728
Compounds were also screened in an adenosine uptake functional cellular assay using the protocol described below.
[3HlAdenosine Uptake Assay Methods
Adenosine uptake activity was determined by monitoring the accumulation of tritiated adenosine into HeLa cells (ATCC catalog # CCL-2) using a PMT-based radiometric detection instrument. In this assay, the potency (IC50) of each compound was determined from a ten point (1 :3 serial dilution; final compound concentration range in assay from 10 μΜ to 0.032 nM) titration curve using the following outlined procedure. To each well of a 96-well CytoStar-T scintillating microplate (Perkin Elmer Catalog
# RPNQ0163), 25 000 HeLa cells in 100 μί, of growth medium comprising: Minimum Essential Media (Life Technologies Catalog # 11095-080) + 10% (v/v) foetal bovine serum (FBS; Sigma Aldrich Catalog
# F2442) was added. These cells were incubated overnight at 37 °C in a humidified atmosphere with 5 % (v/v) C02. After this time the growth medium was removed and replaced with 40 μΐ. assay medium comprising: Hanks balanced salts solution (HBSS; Thermo Fisher Catalog # SH30268.01) + 5% (v/v) FBS. Compound stock solutions in DMSO were diluted in assay medium to 2.5x final compound concentration maintaining a constant DMSO concentration of 0.25% (v/v). 40 μΐ. of compound in assay medium was dispensed into individual wells of the Cytostar-T plates and the plates were incubated for 30 minutes under ambient laboratory conditions. Following this incubation, 20 μΐ. of 500 nM [3H] adenosine (American Radiolabeled Chemicals Inc. Catalog # ART0287) in assay medium was added and incubated for a further 60 minutes under ambient laboratory conditions. The amount of radiolabel accumulation was then determined using a Perkin Elmer Topcount NXT microplate reader. In brief, HeLa cells adhere to the bottom of the Cytostar-T plate, uptake of [3H]adenosine into these cells brings the radiolabel into sufficient proximity to excite the scintillant in the base of the plates. These events are captured by single PMT, time-resolved coincidence counting. IC50 values were determined by 4 parameter robust fit of counts per second values vs. (Logio) compound concentrations.
Table 3. Adenosine uptake inhibition potency
Figure imgf000111_0001

Claims

WHAT IS CLAIMED IS :
1. A compound according to Formula I or a pharmaceutically acceptable salt thereof
Ring A
Figure imgf000112_0001
Formula I
wherein:
Ring A is selected from the group consisting of
Figure imgf000112_0002
n is 0, 1 or 2; g is 0 or 1 ;
W is CH2 or O;
Z is C or N;
X and Y are independently H or X and Y can come together to form cyclopropyl, cyclobutyl or cyclopentyl,
S and T are independently H, F, OH, OCH3, methyl, ethyl or S and T can come together to form cyclopropyl, provided that when Z is C then S and T can come together to form =0, M and L are independently H or M and L can come together to form cyclopropyl,
X and T can come together to form a one to three carbon bridge,
wherein one of X and Y, S and T, M and L, and X and T must be a cyclic moiety;
Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
2. The compound according to claim 1 of Formula la or a pharmaceutically acceptable salt thereof
Ring A
Figure imgf000113_0001
Formula la
wherein:
Ring A is selected from the group consisting of
Figure imgf000113_0002
n is 0, 1 or 2;
S and T are independently H, F, OH, OCH3, methyl, ethyl or S and T can come together to form =0;
- I l l - Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
3. The compound according to claim 1 of Formula lb or a pharmaceutically acceptable salt thereof
Ring A
Figure imgf000114_0001
Formula lb
wherein:
Ring A is selected from the group consisting of
Figure imgf000114_0002
n is 0, 1 or 2;
Ri is selected from the group consisting of hydrogen, triflouromethyl, cyclopropyl, methyl, and cyano;
R2 is independently selected from the group consisting of methoxy, ethoxy and halogen; and R3 is hydrogen, halogen, methyl, ethyl, propyl or isopropyl.
4. The compound of claim 1 selected from the group consisting of:
4-(8-amino-3-(2-oxooctahydro-lH-cyclopropa[a]m^
(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-(8-amino-3-((6'S,8a'R)-l^ i'-difluoro-3'-oxo
yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-(8-amino-3-((6¾,8a'S) ',r-difluoro-3'-oxo
yl)imidazo[l,5-a]pyrazin-l-yl)-3-ethoxy-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-(8-amino-3-((6¾,8a'S)-3'-oxohexahydro-l^spiro[^^
a]pyrazin-l-yl)-3-ethoxy-5-fluoro-N-(4-(trifluoromethyl)pyridin-2-yl)benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]irrddazo[l,5-a]pyrazin-l -yl} -N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-fluoro-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l -yl} -N-(4-methylpyridin-2-yl)benzamide;
4-{8-arnino-3-[(6'R,8a'S)- ,3'-dioxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l -yl} -N-(4-cyclopropylpyridin-2-yl)benzamide;
4-{8-amino-3-[( S,6'R,8a'S)- -hydroxy-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l -yl} -N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[( S,6'R,8a'S)- -hydroxy-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-(8-amino-3-(( S,6'R,8a'S)- -hydroxy-3'-oxohexahydro- H-spiro[cyclopropane-l,2'- indolizin]-6'-yl)imidazo[l,5-a]pyrazin-l -yl)-3-fluoro-N-(4-(trifluoromethyl)pyridin-2- yl)benzamide;
4-(8-amino-3-(( S,6'R,8a'S)- -hydroxy-3'-oxohexahydro- H-spiro[cyclopropane-l,2'- indolizin]-6'-yl)imidazo[l,5-a]pyrazin-l -yl)-N-(4-cyclopropylpyridin-2-yl)benzamide;
4-(8-amino-3-(( S,6'R,8a'S)- -methoxy-3'-oxohexahydro- H-spiro[cyclopropane-l,2'- indolizin]-6'-yl)imidazo[l,5-a]pyrazin-l -yl)-3-ethoxy-N- (4-(trifluoromethyl)pyridin-2- yl)benzamide;
4-{8-amino-3-[(5aS,8R, l laR)-l l -oxodecahydro-lH-pyrido[l ,2-a]pyrrolo[l ,2-d]pyrazin-8- yl]imidazo[l,5-a]pyrazin-l-yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-amino-3-[(5aS,8R,l laS)-l l-oxodecahydro-lH-pyrido[l,2-a]pyrrolo[l,2-d]pyrazin-8- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(7'R,9a'S)-4'-oxohexahydro-2'H-spiro[cyclobutane-l,3'-pyrido[l,2-a]pyrazin]-7'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4- { 8-amino-3 -[(6'R, 8a' S)-3 '-oxohexahy dro-5 Ή-spiro [cyclopropane- 1 , 1 '-indolizin] -6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'S,8a'R)-3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4- { 8-amino-3 -[(6'R, 8a' S)-3 '-oxohexahy dro-5 Ή-spiro [cyclopropane- 1 , 1 '-indolizin] -6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-5-fluoro-N-[4-(trifluoromethyl)pyridin-2- yljbenzamide;
4- { 8-amino-3 -[(6'R, 8a' S)-3 '-oxohexahy dro-5 Ή-spiro [cyclopropane- 1 , 1 '-indolizin] -6'- yl]iinidazo[l,5-a]pyrazin-l-yl}-N-(4-cyclopropylpyridin-2-yl)-3-ethoxy-5-fluorobenzamide; 4-[8-amino-5-fluoro-3-(3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-5-fluoro-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3- yl]benzamide;
4- [8-amino-5-fluoro-3-(3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl)iinidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-5-fluoro-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3- yl]benzamide;
4- [8-amino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl]-3- ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-[8-ainino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl]-3- ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-[8-ainino-3-(2-oxooctahydro-lH-cyclopropa[a]indolizin-5-yl)imidazo[l,5-a]pyrazin-l-yl]-3- ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- ,l'-difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-5-fluoro-N-[4-(trifluoromethyl)pyridin-2- yl]benzamide;
4-{8-amino-3-[(6'S,8a'R)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- y 1] imidazo [ 1 ,5 -a] py razin- 1 -y 1 } -3 -methoxy-N- [4-(trifluoromethy l)pyridin-2-y 1] benzamide; 4-{8-amino-3-[(6'R,8a'S)-l^ -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- y 1] imidazo [ 1 ,5 -a] py razin- 1 -y 1 } -3 -fluoro-5-methoxy-N- [4-(trifluoromethy l)py ridin-2- yl]benzamide;
4-{8-amino-3-[(6'S,8a'R)-l -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)-l^ -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-3- yl]benzamide;
4-{8-amino-3-[(6'R,8a'S)-l^ -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-5-ethoxy-2-fluoro-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol^
3- yl]benzamide;
4- {8-amino-3-[(6'R,8a'S)- , -difluoro-3'-oxohexahydrospiro[cyclopropane-l,2'-indolizin]-6'- yl]imidazo[l,5-a]pyrazin-l-yl}-2-chloro-5-ethoxy-N-[l-methyl-5-(trifluoromethyl)-lH-pyrazol-
3- yl]benzamide;
4- {8-amino-3-[(3R,6aR,l laR)-6-oxooctahydro-lH,6H-pyrrolo[r,2':4,5]pyrazino[2,l- c][l,4]oxazin-3-yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2- yl]benzamide;
4-{8-amino-3-[(3R,6aS,l laR)-6-oxooctahydro-lH,6H-pyrrolo[r,2':4,5]pyrazino[2,l- c][l,4]oxazin-3-yl]imidazo[l,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2- yl]benzamide;
4-[8-amino-5-chloro-3-(3'-oxohexahydro-5'H-spiro[cyclopropane-l, -indolizin]-6'- yl)imidazo[l,5-a]pyrazin-l-yl]-N-(4-cyanopyridin-2-yl)-3-ethoxy-5-fluorobenzamide;
4-[8-amino-3-(3'-oxohexahydro-5H-spiro[cyclopropane-lJ'-indolizin]-6'-yl)imidazo[l,5- a]pyrazin-l-yl]-N-(4-cyanopyridin-2-yl)-3-ethoxy-5-fluorobenzamide;
4-[8-amino-3-(4'-oxohexahydro-2'H,6'H-spiro[cyclopropane-l, -pyrido[l,2-a]pyrazin]-7'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(7'R,9a'S)-4'-oxohexahydro-2'H-spiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-
7'-yl]imidazo[l,5-a]pyrazin-l-yl}-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-amino-3-[(7'R,9a'S)-4'-oxohexahydro-2'H-spiro[cyclopropane-l,3'-pyrido[l,2-a]pyrazin]-
7'-yl]imidazo[l,5-a]pyrazin-l-yl}-3-methoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-[8-amino-3-(4'-oxohexahydro-2'H,6'H-spiro[cyclopropane-l, -pyrido[l,2-a]pyrazin]-7'- yl)imidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-[8-aniino-3-(4'-oxohexahydro-2'H,6'H-spiro[cyclopropane-l, -pyrido[l ,2-a]pyrazin]-7'- yl)iniidazo[l,5-a]pyrazin-l-yl]-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide;
4-{8-aniino-3-[(7'R,9a'S)-2'-ethyl-4'-oxohexahydro-2'H-spiro[cyclopropane-l,3'-pyrido[l ,2- a]pyrazin]-7'-yl]iniidazo[l ,5-a]pyrazin-l-yl}-3-ethoxy-N-[4-(trifluoromethyl)pyridin-2- yl]benzamide;
4-{8-amino-3-[(3'R,9a'R)-6'-oxohexahydrospiro[cyclopropane-l ,7'-pyrazino[2, l-c] [l,4]oxazin]- 3'-yl]imidazo[l ,5-a]pyrazin-l -yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-amino-3-[(3'R,9a'R)-6'-oxohexahydrospiro[cyclopentane-l,7'-pyrazino[2,l -c] [l,4]oxazin]- 3'-yl]imidazo[l ,5-a]pyrazin-l -yl} -3-ethoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; 4-{8-aniino-3-[(6'S,8a'R)-3'-oxohexahydrospiro[cyclopropane-l ,2'-indolizin]-6'-yl]iniidazo[l,5- a]pyrazin-l-yl}-3-methoxy-N-[4-(trifluoromethyl)pyridin-2-yl]benzamide; and
4-{8-aniino-3-[(6'S,8a'R)-3'-oxohexahydrospiro[cyclopropane-l ,2'-indolizin]-6'-yl]iniidazo[l,5- a] py razin- 1 -y 1 } -3 -ethoxy-N- [4-(trifluoromethy l)py ridin-2-yl] benzamide; or a pharmaceutically acceptable salt thereof.
5. A pharmaceutical composition which comprises the compound of claim 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers.
6. The pharmaceutical composition of claim 5, which further comprises at least one additional therapeutically active agent.
7. The compound of claim 1 or a pharmaceutically acceptable salt thereof for use in therapy.
8. The compound of claim 1 or a pharmaceutically acceptable salt thereof for use in the treatment of Bruton's Tyrosine Kinase (Btk) mediated disorders.
9. Use of the compound of Formula I according to claim 1 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of Bruton's Tyrosine Kinase (Btk) mediated disorders.
10. A method for treating a subject suffering with a Bruton's Tyrosine Kinase (Btk) mediated disorder comprising administering to the subject the compound of claim 1 in an amount effective to treat the Btk mediated disorder, thereby treating the subject.
1 1. The method of claim 10, wherein the Btk mediated disorder is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, deforming arthritis, osteoarthritis, traumatic arthritis, gouty arthritis, Reiter's syndrome, polychondritis, acute synovitis and spondylitis, glomerulonephritis (with or without nephrotic syndrome), autoimmune hematologic disorders, hemolytic anemia, aplasic anemia, idiopathic thrombocytopenia, and neutropenia, autoimmune gastritis, and autoimmune inflammatory bowel diseases, ulcerative colitis, Crohn's disease, host versus graft disease, allograft rejection, chronic thyroiditis, Graves' disease, schleroderma, diabetes (type I and type II), active hepatitis (acute and chronic), pancreatitis, primary billiary cirrhosis, myasthenia gravis, multiple sclerosis, systemic lupus erythematosis, psoriasis, atopic dermatitis, contact dermatitis, eczema, skin sunburns, vasculitis (e.g. Behcet's disease) chronic renal insufficiency, Stevens-Johnson syndrome, inflammatory pain, idiopathic sprue, cachexia, sarcoidosis, Guillain-Barre syndrome, uveitis, conjunctivitis, kerato conjunctivitis, otitis media, periodontal disease, pulmonary interstitial fibrosis, asthma, bronchitis, rhinitis, sinusitis, pneumoconiosis, pulmonary insufficiency syndrome, pulmonary emphysema, pulmonary fibrosis, silicosis, chronic inflammatory pulmonary disease, and chronic obstructive pulmonary disease.
12. The method of claim 1 1, wherein the Btk mediated disorder is rheumatoid arthritis, psoriatic arthritis, or osteoarthritis.
PCT/US2015/066218 2014-12-31 2015-12-17 Btk inhibitors WO2016109215A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP15875967.0A EP3240542A4 (en) 2014-12-31 2015-12-17 Btk inhibitors
US15/538,976 US10130630B2 (en) 2014-12-31 2015-12-17 BTK inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/CN2014/095770 WO2016106629A1 (en) 2014-12-31 2014-12-31 Btk inhibitors
CNPCT/CN2014/095770 2014-12-31

Publications (1)

Publication Number Publication Date
WO2016109215A1 true WO2016109215A1 (en) 2016-07-07

Family

ID=56283903

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2014/095770 WO2016106629A1 (en) 2014-12-31 2014-12-31 Btk inhibitors
PCT/US2015/066218 WO2016109215A1 (en) 2014-12-31 2015-12-17 Btk inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/CN2014/095770 WO2016106629A1 (en) 2014-12-31 2014-12-31 Btk inhibitors

Country Status (3)

Country Link
US (1) US10130630B2 (en)
EP (1) EP3240542A4 (en)
WO (2) WO2016106629A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2022140246A1 (en) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Methods and compounds for targeted autophagy

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016106629A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Btk inhibitors

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001019828A2 (en) 1999-09-17 2001-03-22 Basf Aktiengesellschaft Kinase inhibitors as therapeutic agents
US6284781B1 (en) 1996-12-03 2001-09-04 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US6288237B1 (en) 1995-11-17 2001-09-11 Gesellschaft Fur Biotechnologische Forschung Mbh (Gbf) Epothilons C and D, preparation and compositions
WO2002083138A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2002083140A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2002083675A2 (en) 2001-04-10 2002-10-24 Merck Sharp & Dohme Limited Inhibitors of akt activity
WO2002083139A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2003030939A1 (en) 2001-10-05 2003-04-17 Glaxo Group Limited Therapies for treating respiratory diseases
WO2003086279A2 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2003086404A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Fused quinoxaline derivatives as inhibitors of akt activity
WO2003086403A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2003086394A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2005037836A2 (en) 2003-10-15 2005-04-28 Osi Pharmaceuticals, Inc. Imidazo ‘1, 5 - a ! pyrazine tyrosine kinase inhibitors
WO2005044354A1 (en) 2003-11-03 2005-05-19 Glaxo Group Limited A fluid dispensing device
WO2005097800A1 (en) 2004-04-02 2005-10-20 Osi Pharmaceuticals, Inc. 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
WO2007064993A2 (en) 2005-12-02 2007-06-07 Osi Pharmaceuticals, Inc. Bicyclic protein kinase inhibitors
WO2013010868A1 (en) * 2011-07-19 2013-01-24 Msd Oss B.V. 4 - imidazopyridazin- 1 -yl-benzamides and 4 - imidazotriazin- 1 - yl - benzamides as btk- inhibitors
US8673925B1 (en) * 2013-04-09 2014-03-18 Principia Biopharma Inc. Tyrosine kinase inhibitors
US20140206681A1 (en) * 2013-01-23 2014-07-24 Ronald M. Kim Btk inhibitors
WO2014116504A1 (en) * 2013-01-23 2014-07-31 Merck Sharp & Dohme Corp. Btk inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005014599A1 (en) * 2003-06-04 2005-02-17 Cellular Genomics, Inc. Imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of bruton’s tyrosine kinase by such compounds
BRPI0622054B8 (en) * 2006-09-22 2021-05-25 Oxford Amherst Llc compound and pharmaceutical composition
MX2012009208A (en) * 2010-02-08 2012-09-07 Msd Oss Bv 8-methyl-1-phenyl-imidazol[1,5-a]pyrazine compounds.
JP5985658B2 (en) * 2012-01-31 2016-09-06 ナンジン アルゲン ファルマ カンパニー リミテッドNanjing Allgen Pharma Co. Ltd. Cyclic molecules as breton tyrosine kinase inhibitors
WO2014113932A1 (en) 2013-01-23 2014-07-31 Merck Sharp & Dohme Corp. Btk inhibitors
WO2016106629A1 (en) 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Btk inhibitors

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6288237B1 (en) 1995-11-17 2001-09-11 Gesellschaft Fur Biotechnologische Forschung Mbh (Gbf) Epothilons C and D, preparation and compositions
US6284781B1 (en) 1996-12-03 2001-09-04 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
WO2001019828A2 (en) 1999-09-17 2001-03-22 Basf Aktiengesellschaft Kinase inhibitors as therapeutic agents
WO2002083138A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2002083140A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2002083675A2 (en) 2001-04-10 2002-10-24 Merck Sharp & Dohme Limited Inhibitors of akt activity
WO2002083139A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2003030939A1 (en) 2001-10-05 2003-04-17 Glaxo Group Limited Therapies for treating respiratory diseases
WO2003086403A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2003086404A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Fused quinoxaline derivatives as inhibitors of akt activity
WO2003086279A2 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2003086394A1 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
WO2005037836A2 (en) 2003-10-15 2005-04-28 Osi Pharmaceuticals, Inc. Imidazo ‘1, 5 - a ! pyrazine tyrosine kinase inhibitors
WO2005044354A1 (en) 2003-11-03 2005-05-19 Glaxo Group Limited A fluid dispensing device
WO2005097800A1 (en) 2004-04-02 2005-10-20 Osi Pharmaceuticals, Inc. 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
WO2007064993A2 (en) 2005-12-02 2007-06-07 Osi Pharmaceuticals, Inc. Bicyclic protein kinase inhibitors
WO2013010868A1 (en) * 2011-07-19 2013-01-24 Msd Oss B.V. 4 - imidazopyridazin- 1 -yl-benzamides and 4 - imidazotriazin- 1 - yl - benzamides as btk- inhibitors
US20140206681A1 (en) * 2013-01-23 2014-07-24 Ronald M. Kim Btk inhibitors
WO2014116504A1 (en) * 2013-01-23 2014-07-31 Merck Sharp & Dohme Corp. Btk inhibitors
US8673925B1 (en) * 2013-04-09 2014-03-18 Principia Biopharma Inc. Tyrosine kinase inhibitors

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Cancer Principles and Practice of Oncology", 15 February 2001, LIPPINCOTT WILLIAMS & WILKINS PUBLISHERS
"Chirality in Industry", 1992, JOHN WILEY
"Handbook of Pharmaceutical Salts. Properties, Selection and Use", 2002, JOHN WILEY AND SONS
A. L. BINGHAM ET AL., CHEM. COMMUN., 2001, pages 603 - 604
ANDERSON ET AL.: "The Practice of Medicinal Chemistry", 1996, ACADEMIC PRESS
DAVIS ET AL., NATURE, vol. 463, 2010, pages 88 - 94
E. C. VAN TONDER ET AL., AAPS PHARMSCITECH., vol. 5, no. 1, 2004
GENNARO, A.R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS
GILFILLAN ET AL., IMMUNOLOGICAL REVIEWS, vol. 288, 2009, pages 149 - 169
HARDER ET AL., IMMUNITY, vol. 15, 2001, pages 603 - 615
KLINGHOFFER ET AL., EMBO J., vol. 18, 1999, pages 2459 - 2471
LIM ET AL., HAEMATOLOGICA, vol. 95, 2010, pages 135 - 143
LOWELL ET AL., BLOOD, vol. 87, 1996, pages 1780 - 1792
M. CAIRA ET AL., J. PHARMACEUTICAL SCI., vol. 93, no. 3, 2004, pages 601 - 611
N. FELDHAHN ET AL., J. EXP. MED., vol. 201, no. 11, 2005, pages 1837 - 1852
ODOM ET AL., J. EXP. MED., vol. 199, 2004, pages 1491 - 1502
P. GOULD, INTERNATIONAL J. OF PHARMACEUTICS, vol. 33, 1986, pages 201 - 217
ROBY ET AL., ENDOCRINE, vol. 26, 2005, pages 169 - 176
S. BERGE ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, no. 1, 1977, pages 1 - 19
See also references of EP3240542A4
SHINOHARA ET AL., CELL, vol. 132, 2008, pages 794 - 806
T. HIGUCHIV. STELLA: "Bioreversible Carriers in Drug Design", vol. 14, 1987, AMERICAN PHARMACEUTICAL ASSOCIATION, article "Bioreversible Carriers"
T.W. GREENEP.G.M. WUTTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS
VASSILEV ET AL., J. BIOL. CHEM., vol. 274, 1998, pages 1646 - 1656

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2022140246A1 (en) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Methods and compounds for targeted autophagy

Also Published As

Publication number Publication date
EP3240542A4 (en) 2018-07-04
US20170340631A1 (en) 2017-11-30
WO2016106629A1 (en) 2016-07-07
US10130630B2 (en) 2018-11-20
EP3240542A1 (en) 2017-11-08

Similar Documents

Publication Publication Date Title
EP2948458B1 (en) Btk inhibitors
EP3082809B1 (en) Btk inhibitors
US9446130B2 (en) BTK inhibitors
EP3240546B1 (en) Tertiary alcohol imidazopyrazine btk inhibitors
EP3240536B1 (en) Imidazopyrazine analogs with 3-tertiary carbon substitutions as btk inhibitors
EP3082811B1 (en) Btk inhibitors
US10214546B2 (en) Btk inhibitors
EP3240545A1 (en) Biarylether imidazopyrazine btk inhibitors
WO2016109217A2 (en) Btk inhibitors
WO2014113932A1 (en) Btk inhibitors
WO2016196418A1 (en) Btk inhibitors
WO2016109221A1 (en) Btk inhibitors
US10130630B2 (en) BTK inhibitors

Legal Events

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

Ref document number: 15875967

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15538976

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015875967

Country of ref document: EP