US20180099961A1 - Protein kinase inhibitors - Google Patents

Protein kinase inhibitors Download PDF

Info

Publication number
US20180099961A1
US20180099961A1 US15/839,534 US201715839534A US2018099961A1 US 20180099961 A1 US20180099961 A1 US 20180099961A1 US 201715839534 A US201715839534 A US 201715839534A US 2018099961 A1 US2018099961 A1 US 2018099961A1
Authority
US
United States
Prior art keywords
compound
salts
prodrugs
hydrogen
lower alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/839,534
Inventor
Kan He
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Princeton Drug Discovery Inc
Original Assignee
Princeton Drug Discovery Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Princeton Drug Discovery Inc filed Critical Princeton Drug Discovery Inc
Priority to US15/839,534 priority Critical patent/US20180099961A1/en
Assigned to PRINCETON DRUG DISCOVERY INC reassignment PRINCETON DRUG DISCOVERY INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HE, KAN
Publication of US20180099961A1 publication Critical patent/US20180099961A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention relates to novel deuterated and non-deuterated cyclic chemical compounds and salts thereof, to methods of using such compounds in treating protein kinase-mediated diseases or conditions such as autoimmune and cancer diseases or conditions, to pharmaceutical compositions of said compounds, and to combination treatments of said compounds with co-administered therapeutic agents.
  • PTKs protein tyrosine kinases
  • Protein kinases are regulators of cellular signaling, and their functional dysregulation is common in carcinogenesis, autoimmune reactions, and many other disease states or conditions (Lahiry id.; Vargas L et al. Inhibitors of BTK and ITK: state of the new drugs for cancer, autoimmunity and inflammatory diseases. Scand J Immunol. 2013; 78(2):130-9; Nobel M E et al. Protein kinase inhibitors: insights into drug design from structure. Science. 2004; 303:1800-1805).
  • the human genome encodes over 500 protein kinases that share a catalytic domain conserved in sequence and structure but which are notably different in how their catalysis is regulated (Manning G et al. The protein kinase complement of the human genome. Science. 2002; 298:1912-1934; Nobel id.).
  • a partial, non-limiting list of kinases includes: ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN (isoform a), FYN (isoform b), HCK, KIT, LCK, LYNa, PDGFR ⁇ , PDGFR ⁇ , SRC, SRM, YES, PIK3CA/PIK3R1 (Manning id).
  • Aberrant kinase activity has been observed in many disease states including benign and malignant proliferative conditions as well as diseases resulting from inappropriate activation of the immune and nervous systems.
  • novel compounds of this invention inhibit the activity of one or more protein kinases and are expected to be useful in treating kinase-related diseases or conditions.
  • the present invention concerns novel deuterated and non-deuterated cyclic chemical compounds and salts thereof active on protein kinases in general, and in particular as inhibitors of protein kinases. Additionally, methods of treating mammals with protein kinase-mediated diseases or conditions by administering a therapeutically effective amount of the novel deuterated or non-deuterated cyclic chemical compound and/or salts thereof to such mammals in need thereof.
  • the present invention provides compounds having formula I:
  • the present invention provides compounds having formula II:
  • the present invention provides compounds having formula III:
  • the present invention provides compounds having formula IV:
  • the present invention provides compounds having formula V:
  • Exemplary compounds include the following deuterated and non-deuterated cyclic chemical compounds.
  • the present invention provides a compound having the structure of compound I:
  • the present invention provides a compound having the structure of compound II:
  • the present invention provides a compound having the structure of compound III:
  • the present invention provides a compound having the structure of compound IV:
  • the present invention provides a compound having the structure of compound V:
  • the present invention provides a compound having the structure of compound VI:
  • the present invention provides a compound having the structure of compound VII:
  • the present invention provides a compound having the structure of compound VIII:
  • the present invention provides a compound having the structure of compound IX:
  • the present invention provides a compound having the structure of compound X:
  • the present invention provides a compound having the structure of compound XI:
  • the present invention provides a compound having the structure of compound XII:
  • the present invention provides a compound having the structure of compound XIII:
  • the present invention provides a compound having the structure of compound XIV:
  • the invention provides a method for treating a protein kinase-mediated disease or condition in an animal or human subject wherein the method involves administering to the subject an effective amount of one or more of a compound selected from formulas I, II, III, IV, and/or V, and preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X and/or XI.
  • a compound selected from formulas I, II, III, IV, and/or V and preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X and/or XI.
  • the protein kinase mediated disease or condition is an autoimmune disease or a cancer.
  • the autoimmune disease may be at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis.
  • the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
  • CML Philadelphia chromosome-positive chronic myeloid leukemia
  • Ph+ ALL Philadelphia chromosome-positive acute lymphoblastic leukemia
  • DLBCL diffuse large B-cell lymphoma
  • CLL chronic lymphocytic leukemia
  • follicular lymphoma marginal zone lymphomas
  • MCL mantle cell lymphoma
  • WM Waldenstrom's macroglobulinemia
  • T-cell lymphomas and multiple myel
  • the invention provides a method of treating a subject suffering from a protein kinase-mediated disease or condition, comprising administering to the subject suffering from a protein kinase-mediated disease or condition in combination with at least one additional therapeutic agent one or more of a compound selected from formulas I, II, III, IV, and/or V, and preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X and/or XI.
  • a compound selected from formulas I, II, III, IV, and/or V and preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X and/or XI.
  • treat or “therapy” and like terms refer to the administration of compounds in an amount effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or condition, i.e., indication, and/or to prolong the survival of the subject being treated.
  • protein kinase-mediated disease or condition refers to a disease or condition in which the biological function of a protein kinase affects the development, course, and/or symptoms of the disease or condition.
  • a protein kinase-mediated disease or condition includes a disease or condition for which modulation of protein kinase activity provides a positive effect, i.e., one in which treatment with protein kinase inhibitors, including compounds described herein, provides a therapeutic benefit to the subject with or at risk of the disease or condition.
  • the invention provides for pharmaceutical compositions that include a therapeutically effective amount of one or more of a compound selected from formulas I, II, III, IV, and/or V, and preferably compounds I, II, II, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X, and/or XI in free form or in a pharmaceutically acceptable salt form and at least one pharmaceutically acceptable carrier, excipient, and/or diluent.
  • a compound selected from formulas I, II, III, IV, and/or V and preferably compounds I, II, II, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X, and/or XI in free form or in a pharmaceutically acceptable salt form and at least
  • a compound or group of compounds includes pharmaceutically acceptable salts of such compound(s) unless clearly indicated to the contrary, prodrug(s), and all stereoisomers and mixtures thereof.
  • FIG. 1 Metabolic stability of compounds in human liver microsomal incubations. Metabolic stability was determined for compounds I-XIII in incubations with human liver microsomal preparations. Incubations of individual compounds I-XIII at 1 ⁇ M concentrations were carried out for up to 1 hour with human liver microsomes (0.5 mg/mL) in 0.1 M phosphate buffer containing 10 mM MgCl 2 , 1 mM NADPH and 2 mM UDPGA at 37° C. Concentrations at specified times were determined by LC-MS/MS.
  • FIG. 2 Plasma concentration versus time profiles for compound IV and dasatinib in Sprague-Dawley rats following a single oral gavage dose of compound IV and dasatinib administered together and each dosed at 2.5 mg/kg; (Lower panel) plasma concentration versus time profiles for compounds III and V in Sprague-Dawley rats following a single oral gavage dose of compounds III and V administered together and each dosed at 2.5 mg/kg together.
  • FIG. 3 Plasma concentration versus time profiles for compounds X and III in Sprague-Dawley rats following a single oral gavage dose of compounds X and III administered together and each dosed at 5 mg/kg; (Lower panel) plasma concentration versus time profiles for compound XI and dasatinib in Sprague-Dawley rats following a single oral gavage dose of compound XI and dasatinib administered together and each dosed at 5 mg/kg together.
  • chemical structure or “chemical substructure” is meant any definable atom or group of atoms that constitute an individually identifiable molecule, portion of a molecule, such as a substituent moiety, a core which is optionally substituted, and the like.
  • chemical substructures of a ligand can have a role in binding of the ligand to a target molecule, or can influence the three-dimensional shape, electrostatic charge, and/or conformational properties of the ligand.
  • prodrug is a compound that, upon in vivo administration, is metabolized by one or more steps or processes or otherwise converted to the biologically, pharmaceutically, or therapeutically active form of the compound.
  • the pharmaceutically active compound is modified such that the active compound will be regenerated by metabolic or hydrolytic processes.
  • binding in connection with the interaction between a target and a potential binding compound, indicates that the potential binding compound associates with the target to a statistically significant degree as compared to association with proteins generally (i.e., non-specific binding).
  • the term “modulating” or “modulate” refers to an effect of altering a biological activity, especially a biological activity associated with a particular biomolecule such as a protein kinase.
  • a biological activity associated with a particular biomolecule
  • an agonist or antagonist of a particular biomolecule modulates the activity of that biomolecule, e.g., an enzyme, by either increasing (e.g., agonist, activator), or decreasing (e.g., antagonist, inhibitor) its activity.
  • This type of activity is typically indicated in terms of an half maximal effective concentration (EC 50 ) or half maximal inhibitory concentration (IC 50 ) for an activator or inhibitor, respectively.
  • inhibition activity can be expressed in percent inhibition and/or Ki.
  • the term “synthesizing” and like terms means chemical synthesis from one or more precursor materials. Further, by “assaying” is meant the creation of experimental conditions and the gathering of data regarding a particular result of the experimental conditions. For example, enzymes can be assayed based on their ability to act upon a detectable substrate. A compound or ligand can be assayed based on its ability to bind to a particular target molecule or molecules.
  • “D,” “d,” and “ 2 H” refer to a deuterium atom, a stable isotope of hydrogen with a mass twice that of hydrogen (atomic weight of 2.0144). Hydrogen naturally occurs as a mixture of the isotopes hydrogen ( 1 H), deuterium ( 2 H or D), and tritium ( 3 H or T). The natural abundance of deuterium is about 0.015%. A person skilled in the art would recognize that all chemical compounds with a hydrogen atom actually are present as mixtures of the H and D isotopes, with about 0.015% being the deuterium isotope.
  • lower alkyl is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups and branched-chain alkyl groups.
  • a straight chain or branched chain alkyl has about 6 or fewer carbon atoms in its backbone (e.g., C 1 -C 6 for straight chain, C 3 -C 6 for branched chain).
  • lower alkenyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as straight or branched group of 2-6 carbon atoms, referred to herein as C 2 -C 6 alkenyl.
  • cycloalkyl refers to a 3-7 membered moncyclic ring of aliphatic groups, including C 3 -C 7 , that is optionally substituted with alkyl, alkenyl, alkoxyl, optionally substituted amino, halogens, cyano (—CN), or nitro (—NO 2 ).
  • alkoxyl or “alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy, and the like.
  • heterocyclo refers to fully saturated or unsaturated, including non-aromatic (i.e., “heterocycloalkyl”) and aromatic (i.e., “heteroaryl”) cyclic groups having from 5 to 10 atoms with at least one heteroatom (e.g. oxygen (“O”), sulfur (“S”), or nitrogen (“N”)) in at least one carbon atom-containing ring.
  • heterocycloalkyl i.e., “heterocycloalkyl”
  • heteroaryl aromatic i.e., “heteroaryl” cyclic groups having from 5 to 10 atoms with at least one heteroatom (e.g. oxygen (“O”), sulfur (“S”), or nitrogen (“N”)) in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group may have 1, 2, 3, or 4 heteroatoms.
  • the heteroatoms nitrogen and sulfur may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • heterocyclo may be optionally substituted with amino (—NR 5 R 6 ), wherein R 5 and R 6 are independently hydrogen and/or lower alkyl, hydroxyl (—OH), alkoxyl, lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl may be optionally substituted with —OH or alkoxyl groups.
  • Halogen refers to chloro (“Cl”), fluoro (“F”), bromo (“Br”), or iodo (“I”).
  • the compounds provided herein may contain chiral centers. Such chiral centers may be of either the (R) or (S) configuration, or may be a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, or stereoisomeric or diastereomeric mixtures thereof, including racemic mixtures (about 50:50 ratio of enantiomers).
  • pharmaceutically acceptable means that the indicated material does not have properties that would cause a reasonably prudent medical practitioner to avoid administration of the material to a patient, taking into consideration the disease or condition to be treated and the respective route of administration. For example, it is commonly required that such a material be essentially sterile, e.g., for injectable.
  • salts refers to salts that are non-toxic in the amounts and concentrations at which they are administered.
  • the preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of a compound without preventing it from exerting its physiological effect.
  • composition refers to a pharmaceutically active compound and one or more pharmaceutically acceptable carriers, excipients, and/or diluents.
  • terapéuticaally effective or “effective amount” is an amount of a preparation that alone, or together with further doses, and/or in combination with other therapeutic agents produces the desired response. This may involve halting the progression of the disease or delaying the onset of or preventing the disease or condition from occurring, although it may also imply only slowing of the disease or condition temporarily.
  • protein kinase-mediated disease or condition refers to a disease or condition in which the biological function of a protein kinase affects the development, course, and/or symptoms of the disease or condition.
  • mutants refers to single or multiple amino acid changes in a protein as compared to the wild-type protein amino acid sequence.
  • the present invention provides compounds having formula I:
  • W 2 is deuterium or hydrogen, more preferably hydrogen, and W 1 and W 3 are hydrogen.
  • Y is preferably nitrogen.
  • R 2 is preferably lower alkyl, more preferably methyl.
  • R 3 is preferably lower alkyl or hydrogen, more preferably hydrogen.
  • R 1 is preferably a substituted or unsubstituted saturated five or six membered nitrogen containing heterocyclo ring.
  • the substituted or unsubstituted saturated five membered nitrogen containing heterocyclo ring can be substituted or unsubstituted pyrrolidin-1-yl, preferably 3-hydroxy- or 3-amino-pyrrolidin-1-yl, more preferably 3-hydroxy pyrrolidin-1-yl.
  • the present invention provides compounds having formula II:
  • W 1 , W 2 , and W 3 are independently hydrogen or deuterium
  • Y is carbon or nitrogen
  • R 2 , R 3 , and R 4 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR 5 R 6 , wherein R 5 and R 6 are independently hydrogen, lower alkyl, or lower alkenyl; and
  • X is independently hydrogen, —(CH 2 ) n —OR 5 wherein n is an integer number from 0 to 4 and R 5 is hydrogen, lower alkyl, or lower alkenyl, or —NR 5 R 6 .
  • W 2 is deuterium or hydrogen, more preferably hydrogen, and W 1 and W 3 are hydrogen.
  • Y is preferably nitrogen.
  • R 2 is preferably lower alkyl, more preferably methyl.
  • R 5 is preferably lower alkyl or hydrogen, more preferably hydrogen.
  • R 4 is preferably lower alkyl or hydrogen, more preferably hydrogen.
  • X is preferably hydrogen, hydroxyl or amine, more preferably hydroxyl.
  • the present invention provides compounds having formula III:
  • W 2 is deuterium or hydrogen, more preferably hydrogen, and W 1 and W 3 are hydrogen.
  • Y is preferably nitrogen.
  • R 2 is preferably lower alkyl, more preferably methyl.
  • R 3 is preferably lower alkyl or hydrogen, more preferably hydrogen.
  • R 4 is preferably lower alkyl or hydrogen, more preferably hydrogen.
  • the present invention provides compounds having formula IV:
  • the present invention provides compounds having formula V:
  • the present invention provides a compound having the structure of compound I:
  • the present invention provides a compound having the structure of compound II:
  • the present invention provides a compound having the structure of compound III:
  • the present invention provides a compound having the structure of compound IV:
  • the present invention provides a compound having the structure of compound V:
  • the present invention provides a compound having the structure of compound VI:
  • the present invention provides a compound having the structure of compound VII:
  • the present invention provides a compound having the structure of compound VIII:
  • the present invention provides a compound having the structure of compound IX:
  • the present invention provides a compound having the structure of compound X:
  • the present invention provides a compound having the structure of compound XI:
  • the present invention provides a compound having the structure of compound XII:
  • the present invention provides a compound having the structure of compound XIII:
  • the present invention provides a compound having the structure of compound XIV:
  • Protein kinases play key roles in propagating biochemical signals in diverse biological pathways. As such, kinases represent important control points for small molecule therapeutic intervention. More than 500 kinases have been described, and specific kinases have been implicated in a wide range of diseases or conditions.
  • the invention provides methods for treating a protein kinase-mediated disease or condition in an animal or human subject, (i.e., indications), such as without limitation, autoimmune disease, hyperproliferative disease, cancer, cardiovascular disease, inflammatory disease, neurological disease, and other diseases.
  • the protein kinase-mediated disease or condition is an autoimmune disease or cancer.
  • the autoimmune disease is at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis; and the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
  • SLE systemic lupus erythematosus
  • the invention provides a method for modulating the activity of a protein kinase selected from the group consisting of ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN, HCK, KIT, LCK, LYN, PDGFR ⁇ , PDGFR ⁇ , SRC, SRM, YES, PIK3CA/PIK3R1 by administering an effective dose amount of one or more compounds having formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI.
  • Upper and lower case for letters used in kinase nomenclature are used interchange
  • the invention provides methods for treating a protein kinase-mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of a composition including one or more compounds having formulas I, II, III, IV and/or V, preferably one or more of compounds I, II, III, IV, VI, V, VII, VIII, IX, X, XI, XII, XIII, and XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and XI.
  • a composition including one or more compounds having formulas I, II, III, IV and/or V, preferably one or more of compounds I, II, III, IV, VI, V, VII, VIII, IX, X, XI, XII, XIII, and XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and XI.
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN (isoform a), FYN (isoform b), HCK, KIT, LCK, LYNa, PDGFR ⁇ , PDGFR ⁇ , SRC, SRM, YES, PIK3CA/PIK3R1 by administering an effective amount of one or more compounds having formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI.
  • a number of different assays for kinase activity can be utilized for testing to determine active modulators and/or determining specificity of a modulator for a particular kinase or group of kinases.
  • the person of ordinary skill in the art will know and understand that other assays that can be utilized or can be modified for a particular application.
  • compounds of the invention were assayed for inhibition of cell growth using diffuse large B-cell lymphoma cell line SU-DHL-4 and chronic myelogenous leukemia cell line K-562 (see Example 16).
  • the IC 50 values for compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV were all less than 20 nM.
  • Protein kinase targets for compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV include, but are not limited to the following: ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN, HCK, KIT, LCK, LYN, PDGFR ⁇ , PDGFR ⁇ , SRC, SRM, YES, and PIK3CA/PIK3R1.
  • the Tec family of kinases forms the second largest class of cytoplasmic protein tyrosine kinases after the Src family kinases (SFKs) and consists of five mammalian members: Btk, Bmx (bone marrow kinase on the X-chromosome, also known as Etk), Itk (IL-2 inducible T-cell kinase), RIk (resting lymphocyte kinase, also known as Txk), and Tec (Hartkamp et al. Bruton's tyrosine kinase in chronic inflammation: from pathophysiology to therapy. Int J Interferon Cytokine Mediat Res. 2015; 7: 27-34).
  • Tec family of kinases are primarily expressed in the hematopoietic system, although both Tec and Bmx are also expressed in stromal tissues such as liver and endothelial cells, respectively.
  • Activation of Tec family kinases upon cell-surface receptor triggering requires relocalization of the protein to the plasma membrane, which is mediated by the interaction of the PH domain with the lipid phosphatidylinositol (3,4,5) P3, formed by activated phosphatidylinositol-3 kinase.
  • Subsequent phosphorylation by SFKs and autophosphorylation of tyrosine 223 result in the complete activation of Tec family of kinases.
  • BTK is the best-known member of the Tec family of kinases with BTK mutations leading to X-linked agammagiobulinemia in men and X-linked immunodeficiency in mice.
  • BTK is a key regulator of B-cell development, activation, signaling, and survival (Hartkamp id).
  • BTK plays an important role in a number of other hematopoietic cell-signaling pathways, e.g., toll-like receptor (TLR) and cytokine receptor-mediated TNF-alpha production in macrophages, IgE receptor (FcepsilonRI) signaling in mast cells, inhibition of Fas/APO-1 apoptotic signaling in B-lineage lymphoid cells, and collagen-stimulated platelet aggregation.
  • TLR toll-like receptor
  • FcepsilonRI IgE receptor
  • BTK and other members of Tec family kinases can play a critical role in autoimmune diseases, such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), type I diabetes (T1D), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), and rheumatoid arthritis (RA).
  • SLE systemic lupus erythematosus
  • MS multiple sclerosis
  • T1D type I diabetes
  • SSc systemic sclerosis
  • pSS primary Sjögren's syndrome
  • RA rheumatoid arthritis
  • the BTK inhibitor ibrutinib demonstrated high clinical activity in B-cell malignancies, especially in patients with chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), and Waldenstrom's macroglobulinemia (WM).
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • tyrosine kinase BMX regulates inflammation induced by TNF and other mediators appear to do so by regulating the shared TAK1-TAB complex (Gottar-Guillier M et al.
  • the tyrosine kinase BMX is an essential mediator of inflammatory arthritis in a kinase-independent manner. J Immunology. 2011; 186(10):6014).
  • BMX kinase may play a role in the pathogenesis of glioblastoma, prostate, breast, and lung cancers.
  • BMX has also shown potential as an anti-vascular therapy in combination with radiation or as a sensitizer to chemotherapeutic agents. (Jarboe J S et al. Mini-review: bmx kinase inhibitors for cancer therapy. Recent Pat Anticancer Drug Discov. 2013; 8(3):228-38).
  • Src family kinases consist of 11 nonreceptor tyrosine kinases, including Src, Fyn, Yes, Blk, Yrk, Frk (also known as Rak), Fgr, Hck, Lck, Srm, and Lyn (Sen B, Johnson F M. Regulation of SRC family kinases in human cancers. J Signal Transduct. 2011:ID865819). Src is found in keratinocytes, whereas Blk, Fgr, Hck, Lck, and Lyn are found primarily in hematopoietic cells. Frk occurs chiefly in bladder, breast, brain, colon, and lymphoid cells. Src family kinases are involved in proliferation and migration responses in many cell types.
  • Src is a non-receptor protein tyrosine kinase that plays a multitude of roles in cell signaling. Src is involved in the control of many functions, including cell adhesion, growth, movement, and differentiation. Src is widely expressed in many cell types, and can have different locations within a cell. Numerous human malignancies display increased SRC expression and activity, suggesting that SRC may be intimately involved in oncogenesis. SRC inhibitor bosutinib has been used or the treatment of Philadelphia chromosome-positive (Ph+) chronic myelogenous leukemia (CML), and saracatinib has been studied for potential treatment of Alzheimer's disease and schizophrenia.
  • Ph+ Philadelphia chromosome-positive
  • CML chronic myelogenous leukemia
  • saracatinib has been studied for potential treatment of Alzheimer's disease and schizophrenia.
  • ABL is a cytoplasmic and nuclear protein tyrosine kinase that has been implicated in processes of cell differentiation, cell division, cell adhesion, and stress response (Hantschel O. Structure, regulation, signaling, and targeting of abl kinases in cancer. Genes Cancer. 2012; 3:436-46).
  • ABL mutations are associated with cancers such as chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), and acute myelogenous leukemia (AML).
  • CML chronic myeloid leukemia
  • ALL acute lymphoblastic leukemia
  • AML acute myelogenous leukemia
  • Several ABL inhibitors such as imatinib, dasatinib, and nilotinib have been used for treatment of CML, ALL, and AML.
  • Dasatinib a potent inhibitor of BCR-ABL is used for treatment of newly diagnosed Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML) in chronic phase, chronic, accelerated, or myeloid or lymphoid blast phase Ph+ CML with resistance or intolerance to prior therapy including imatinib, and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) with resistance or intolerance to prior therapy.
  • dasatinib is associated with severe respiratory toxicity such as pleural effusion and pulmonary hypertension, which could be a result of dasatinib accumulation in lung tissue.
  • LCK is a 57.9 kDa membrane-associated non-receptor tyrosine kinase encoded by chromosome Ip34.3.
  • the protein structure comprises an SH3 and SH2 domain.
  • LCK inhibitors may be useful in treating acute lymphoblastic leukemia, T-cell lymphoma, lymphopenia, renal carcinoma, colon carcinoma, severe combined immunodeficiency, multiple sclerosis, inflammatory bowel, and type I diabetes.
  • Frk is a 58.5 kDa tyrosine kinase encoded by chromosome 6q21-q22.3.
  • the structure comprises an SH2, an SH3, and a tyrosine kinase domain.
  • Inhibition of Frk could provide means to suppress beta-cell destruction in type I diabetes.
  • Frk inhibitors may be useful in treating acute myeloid leukemia and type I diabetes.
  • Fyn is a 60.6 kDa non-receptor tyrosine kinase encoded by chromosome 6q21. Fyn is involved in regulation of mast cell degranulation in a synergistic confluence of Fyn and Lyn pathways at the level of protein kinase C and calcium regulation. Fyn inhibitors may be useful in treating Alzheimer's disease, schizophrenia, and in prevention of metastases, e.g., in melanoma and squamous cell carcinoma.
  • HCK is a 59.5 kDa tyrosine kinase encoded by chromosome 20ql 1.21.
  • the protein structure comprises an SH3, an SH2, and a bipartite kinase domain.
  • HCK inhibitors may be useful in treating chronic myelogenous leukemia and acute lymphocytic leukemia.
  • Kit is a 109.9 kDa transmembrane tyrosine kinase encoded by chromosome 4q12. Kit plays an important role in the development of melanocytes, mast, germ, and hematopoietic cells. Aberrant expression and/or activation of Kit has been implicated in a variety of pathologic states.
  • Kit inhibitors may be useful in treating malignancies, including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis, inflammatory bowel syndrome, transplant rejection, and hypereosinophilia.
  • malignancies including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastom
  • LCK is a 57.9 kDa membrane associated non-receptor tyrosine kinase encoded by chromosome Ip34.3.
  • the protein structure comprises an SH3 and SH2 domain.
  • LCK inhibitors may be useful in treating acute lymphoblastic leukemia, T-cell lymphoma, lymphopenia, renal carcinoma, colon carcinoma, severe combined immunodeficiency, multiple sclerosis, inflammatory bowel, and type I diabetes.
  • Platelet-derived growth factor receptors are cell surface tyrosine kinase receptors for members of the platelet-derived growth factor (PDGF) family.
  • PDGF subunits-A and -B are important factors regulating cell proliferation, cellular differentiation, cell growth and development, and many diseases including cancer.
  • PDGFR ⁇ is a 122.7 kDa transmembrane tyrosine kinase encoded by chromosome 4ql2 (symbol: PDGFRA).
  • PDGFR ⁇ is a 124.0 kDa transmembrane tyrosine kinase encoded by chromosome 5q31-q32 (symbol: PDGFRB).
  • PDGFR inhibitors may be useful in treating various diseases such as idiopathic hypereosinophilic syndrome, chronic eosinophilic leukemia, glioma, gastrointestinal stromal tumors (GISTs), juvenile myelomonocytic leukemia, metastatic medulloblastoma, atherogenesis, and restenosis.
  • Yes is a 60.8 kDa tyrosine kinase encoded by chromosome 18pl 1.31-pl 1.21 (symbol: YESI).
  • the structure of Yes comprises SH3 and SH2 domains followed by a TK domain.
  • the YES oncogene is homologous to the Yamaguchi sarcoma virus gene, and the amino acid sequence of Yes shows a high degree of homology with that of the SRC gene product of Roussarcoma virus.
  • the Yes kinase is highly expressed in multiple mammalian cell types, including neurons, spermatozoa, platelets, and epithelial cells.
  • the target kinase Yes is amplified and overexpressed in various cancers including esophageal squamous cell carcinoma.
  • Yes inhibitors may be useful in treating cancers including esophageal squamous cell carcinoma.
  • compounds of formulas I, II, III, IV, and V preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV (compounds I-XIV), including salts, prodrugs, and/or isomers thereof, can be used in preparation of medicaments for the treatment of a kinase-mediated disease or condition, in particular when the disease or condition is an autoimmune disease or cancer.
  • the amounts of compounds of formulas I, II, III, IV, and V, compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII and XIV (compounds I-XIV), including salts, prodrugs, and/or isomers thereof, to be administered can be determined by standard procedures taking into account factors such as the compound's IC 50 ; the biological half-life of the compound; the age, size, and weight of the subject; and the condition associated with the subject. In general, routine experimentation in clinical trials will determine specific ranges for optimal therapeutic effect for each therapeutic agent and each administrative protocol and administration to specific patients will be adjusted to within effective and safe ranges depending on the patient's condition and responsiveness to initial administration.
  • doses of active compounds may range from about 0.01 mg/kg per day to about 1000 mg/kg per day.
  • Compounds described herein can be administered in single or multiple doses.
  • these compounds are expected to have a more favorable pharmacokinetic profile compared to dasatinib, specifically longer t 1/2 , longer duration of action, less first pass effect, and higher oral bioavailability.
  • the intrinsic clearances for compounds I, IV, V, VI, VII, VIII, X, XI, XII, and XIII were less than 24 ⁇ L/min/mg compared to 88 ⁇ L/min/mg for dastatinib.
  • compounds IV, V, X, and XI showed surprisingly significantly higher Cmax values of 82 ⁇ 13, 35 ⁇ 19, 93 ⁇ 101, and 102 ⁇ 86 ng/mL, respectively, as compared to dasatinib where Cmax was 15 ⁇ 5 and 7 ⁇ 7 ng/mL and the deuterium analog of dasatinib, compound III, where Cmax was 9 ⁇ 6 and 11 ⁇ 11 ng/mL.
  • the oral bioavailability of compounds IV, V, X, and XI was 18.9 ⁇ 6.7, 11.1 ⁇ 4.3, 18.9 ⁇ 2, and 24 ⁇ 33.1 percent bioavailability (F) compared to dasatinib oral bioavailability of 3.9 ⁇ 1 and 9.8 ⁇ 3.6 percent bioavailability, and compound III where oral bioavailability was 6 ⁇ 0.9 and 7.2 ⁇ 4.2 percent.
  • the results from this study show compounds IV, V, X, and XI have surprisingly much lower intravenous plasma clearance than dasatinib and compound III. This data is consistent with the in vitro metabolic stability data which showed compounds IV, V, X, and XI to be significantly more stable than either dasatinib or compound III, and with significantly lower intrinsic clearance values.
  • compounds IV, V, X, and XI were found to have a surprisingly significantly lower intravenous volume of distribution values than dasatinib and compound III, suggesting these compounds are not as widely distributed to tissues as compared to dasatinib and compound III. Further, this suggests these compounds have lower potential than dasatinib for drug-induced organ toxicity.
  • Dasatinib is associated with severe respiratory toxicity such as pleural effusion and pulmonary hypertension, which have been attributed to the accumulation of dasatinib in lung tissue.
  • the composition to be administered can include a plurality of different pharmacologically active compounds which can include a plurality of compounds of the invention including one or more compounds of formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI, including salts, prodrugs, and/or isomers thereof, and other therapeutically effective agents for the same disease or condition, wherein the compounds have an additive or a synergistic effect on the disease indication.
  • compounds of formulas I, II, III, IV, and/or V preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-
  • the invention provides methods for treating a kinase dysfunction-mediated disease or condition in an animal or human subject, wherein the method involves administering to the subject an effective amount of one or more compounds having formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI, salts, prodrugs, and/or isomers thereof, in combination with one or more other therapies for treating the same disease or condition.
  • compounds having formulas I, II, III, IV, and/or V preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of
  • Other therapies may include medical procedures (such as surgeries), therapeutic agents, and/or radiation.
  • Therapeutic agents include chemotherapeutic agents, biologics, and immunotherapeutics.
  • Combination therapy can include administration of one or more of compounds described herein with one or more other therapeutics at different times or simultaneous administration.
  • dosages may be modified for one or more of the compounds of the invention or other therapeutics used in combination, such modifications being a reduction in the dose amounts relative to a compound or therapy used alone.
  • use in combination includes use with other medical procedures, therapeutics, and therapies where the other therapy or drug may be administered at different times, within a short time period, such as within 1, 2, 3, or 4-24 hours, or within a longer time period, such a 1-2 days, 2-4 days, 4-7 days, or 1-4 weeks.
  • Use of the compounds of the invention can be in combination with a medical procedure such as surgery, performed on the subject once or infrequently, where the compounds are administered within a short time or longer time before or after the medical procedure.
  • the methods and compounds will typically be used in therapy for human subjects with a kinase-mediated disease or condition. However, they may also be used to treat similar or identical indications in other animal subjects.
  • the terms “subject” and “animal subject” and the like refer to human and non-human vertebrates, i.e., mammals, such as non-human primates, sports and commercial animals, e.g., equines, bovines, porcines, ovines, rodents, and pets, e.g., canines and felines.
  • Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, 2-hydroxyethanesulfanates, lactates, maleates, methanesulfonates, nicotinates, nitrates, oxalates, pectinates, phosphates, picrates, salicylates, propionates, tartrates, thiocyantes, toluenesul
  • Exemplary basic salts include ammonium salts, alkali metal salts such sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, and salts with organic bases (for example organic amines), and the like.
  • Compounds of formulas I, II, III, IV, and V including compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), salts, prodrugs, and/or isomers thereof, can be administered intravenously, intramuscularly, subcutaneously, orally, transdermally, transmucosally, rectally, or by inhalation. In the case of intravenous administration, the dose may be administered as a bolus or infusion.
  • compositions for oral use can be obtained, for example, by combining one or more compounds of formula I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV, and more preferably one or more of compounds IV, V, X, and/or XI, salts, prodrugs, and/or isomers thereof, with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone).
  • disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid, or a salt thereof such as sodium alginate.
  • the compounds of formula I, II, III, IV, and V preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII and XIV, and more preferably compounds IV, V, X, and XI, salts, prodrugs, and/or isomers thereof, are formulated in sterile liquid solutions, preferably in physically compatible buffers or solutions, such as saline solution, Hank's solution, or Ringer's solution.
  • the compounds may be formulated in solid form and re-dissolved or suspended immediately prior to use. Lyophilized forms can also be produced.
  • administration of the compounds described herein can occur simultaneously or sequentially with chemotherapy or radiation. It is understood that administration of other therapeutics or drugs to treat a medical disease or condition can be by a different route of administration or by the same route of administration.
  • the use in combination therapy for any route of administration includes delivery of compounds of the invention and one or more other drug therapeutics delivered by the same route of administration together in any formulation, or administered together, within 1 hour, 2 hours, 3 hours, up to 24 hours, in separate formulations, or by different routes of administration.
  • the invention also provides for a pharmaceutical combination, e.g., a kit, comprising (a) a first agent which is a compound of the invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and (b) at least one co-agent.
  • a pharmaceutical combination e.g., a kit, comprising (a) a first agent which is a compound of the invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and (b) at least one co-agent.
  • the kit can include instructions for administration.
  • the present invention also includes processes for the preparation of the novel deuterium-enriched and non-enriched compounds of the invention.
  • reactive functional groups for example hydroxy, amino, imino, thio, or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions.
  • Conventional protecting groups can be used in accordance with standard practice, for example, see TW Greene and PGM Wutsin Protective Groups in Organic Chemistry , John Wiley and Sons, 1991.
  • the initial step of the chemical process involves reacting 4-bromo-2-chloro-6-methylaniline (1) with allyl bromide to form intermediate 2.
  • intermediate 2 in dry tetrahydrofuran under nitrogen gas at about ⁇ 70° C. is added dropwise n-butyl lithium (about 1.5 moles n-butyl lithium to 1 mole intermediate 2).
  • n-butyl lithium about 1.5 moles n-butyl lithium to 1 mole intermediate 2
  • the intermediate lithium complex is quenched with d 1 -methanol (CH 3 OD; 99% deuterium, #550574; Lot # MKBW0355V, Aldrich, St Louis, Mo.) to selectively incorporate deuterium at the 4 position and give intermediate 3.
  • d 1 -methanol CH 3 OD; 99% deuterium, #550574; Lot # MKBW0355V, Aldrich, St Louis, Mo.
  • Intermediate 4 is reacted with 3-ethoxyacryloyl chloride to form the acrylamide intermediate 5 which is then treated with N-bromosuccinimide and thiourea to form the thiazole intermediate 6.
  • the thiazole intermediate 6 is treated with the base sodium hydride followed by addition of 4,6-dichloro-2-methylpyrimidine to form the 2-[(6-chloro-2-methylpyrimidin-4-yl)amino]-N-(2-chloro-4-deutero-6-methylphenyl)thiazole-5-carboxamide (intermediate 7).
  • Intermediate 7 is reacted with piperidine and N,N-diisopropylethylamine to form the desired product compound I (8).
  • Compound V can be synthesized by the method shown in Scheme 5.
  • mass spectrometry results indicated that a compound may have more than one value due to the isotope distribution of an atom in the molecule, such as a compound having a bromo or chloro substituent.
  • reaction mixture was then stirred from ⁇ 70° C. to ⁇ 20° C. over 40 min when thin layer chromatograph (TLC) analysis (hexanes only) showed the reaction was complete.
  • TLC thin layer chromatograph
  • the mixture was poured into cold water (100 mL) followed by extraction with EtOAc (2 ⁇ 100 mL). The combined organic layers were washed with brine, dried (Na 2 SO 4 ), and concentrated under reduced pressure. The residue was purified by column chromatography (hexanes only, then EtOAc:hexanes; 1:20) to give compound 3 (1.38 g, 93%) as a pale yellow liquid.
  • the mixture was not a clear solution.
  • the mixture was cooled to room temperature and concentrated to dryness under reduced pressure, and the resultant residue was suspended in 50 mL acetonitrile, and centrifuged at 4000 rpm for 15 min.
  • the pellet was then suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min.
  • the pellet was re-suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min.
  • the supernatants were combined and concentrated to dryness to afford the target compound XI(H-30) ( ⁇ 75 mg) as an off-white solid.
  • MSA Off-chip Mobility Shift Assay
  • Compound I showed >50% inhibition at 1 nM on the following recombinant kinases: ABL, ABL(E255K), ACK, ARG, BLK, BMX, BTK, DDR2, EPHA1, FGR, FMS, FRK, FYN (isoform a), HCK, LCK, LYNa, PDGFR ⁇ , PDGFR ⁇ , SRC, YES.
  • Compound I showed concentration-dependent inhibition of recombinant BTK, ACK and PDGFR ⁇ activity with an IC 50 of approximately 0.2, 0.5 and 1.4 nM, respectively.
  • Compound I inhibited recombinant PIK3CA/PIK3R1 activity with an IC50 of approximately 12 nM.
  • Compound I showed >50% inhibition at 10 nM on the following recombinant kinases: YES, FRK, SRC, LYNa, FMS, BMX, ABL, FYN (isoform b), FGR, HCK, FYN (isoform a), LCK, DDR2, ARG, ABL(E255K), BTK, EPHA1, BLK, ACK, SRM, PDGFR ⁇ , PIK3CA/PIK3R1, PDGFR ⁇ , CSK, KIT(D816V), KIT(D816Y), BRK.
  • Compound I showed ⁇ 50% inhibition at 10 nM on the following recombinant kinases: PDGFR ⁇ (V561D), DDR1, KIT, KIT(V560G), HER4, KIT(D816E), EGFR(L858R), KIT(V654A), PDGFR ⁇ (D842V), EGFR, EGFR(L861Q), EGFR(d746-750), JAK1, RET, RET(S891A), FGFR3, ALK, WNK3, RET(Y791F), BRAF(V600E), RAF1, ROCK1, AurA, MAP2K2, RET(M918T), KDR, FGFR2, Erk1, EGFR(T790M), RET(G691 S), PDGFR ⁇ (T6741), p38 ⁇ , HER2, JAK3, MAP2K1, p38 ⁇ , EGFR(T790M/L858R), FLT1, BRAF, KIT(T
  • Compound II showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa. FRK, FYN (isoform a), FYN (isoform b), TEC, BMX, LYNb, ABL, FGR, FMS, BLK, EPHA1, ABL(E255K).
  • Compound II showed ⁇ 50% inhibition at 1 nM on the following recombinant kinases: PDGFR ⁇ , PDGFR ⁇ , PIK3CA/PIK3R1, KIT, KIT(V560G), EGFR, HER2, YES(T3481), ITK, p38 ⁇ , ABL(T3151), RAF1, p38 ⁇ , BRAF.
  • Compound II showed concentration-dependent inhibition of recombinant BTK activity with an IC 50 of ⁇ 1 nM.
  • Compound II showed >50% inhibition at 10 nM on the following recombinant kinases: BTK, PDGFR ⁇ , KIT(V560G), PDGFR ⁇ , KIT. Compound II showed ⁇ 50% inhibition at 10 nM on the following recombinant kinases: PIK3CA/PIK3R1, EGFR, p38 ⁇ , RAF1, HER2, p38 ⁇ , BRAF.
  • Compound II showed >50% inhibition at 100 nM on the following recombinant kinases: PDGFR ⁇ , KIT(V560G), KIT, PDGFR ⁇ , EGFR. Compound II showed ⁇ 50% inhibition at 100 nM on the following recombinant kinases: p38 ⁇ , RAF1, PIK3CA/PIK3R1, HER2, p38 ⁇ , BRAF.
  • Compound IV showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound V showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound VI showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound VII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound VIII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound IX showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound X showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound X showed concentration-dependent inhibition of recombinant ABL, ABL (E255K), BTK and BTK (C481S) activity with IC 50 values of ⁇ 1 nM, respectively.
  • Compound XI showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound XI showed concentration-dependent inhibition of recombinant ABL, ABL (E255K), BTK and BTK (C481S) activity with ICsovalues of ⁇ 1 nM, respectively.
  • Compound XII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound XIII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • SU-DHL-4 ATCCC CRL-2957TM
  • K-562 ATCC® CCL-243TM
  • Mino ATCC® CRL-3000TM
  • ATCC American Type Culture Collection
  • SU-DHL-4 cells and Mino cells grew in ATCC-formulated RPMI-1640 medium (ATCC) supplemented with 10% fetal bovine serum (FBS)(Gibco, Life Technologies) (complete medium) a T-75 flask at 37° C. under 5% CO 2 with saturated humidity.
  • FBS fetal bovine serum
  • K-562 cells grew in ATCC-formulated Iscove's Modified Dulbecco's Medium (IMDM) supplemented with 10% FBS (Gibco, Life Technologies) (complete medium) a T-75 flask at 37° C. under 5% CO 2 with saturated humidity.
  • IMDM Iscove's Modified Dulbecco's Medium
  • FBS Gibco, Life Technologies
  • complete medium a T-75 flask at 37° C. under 5% CO 2 with saturated humidity.
  • FBS Gibco, Life Technologies
  • a 200 ⁇ L aliquot of the cell suspension was added to the well of a 96-well plate which was pre-added with 1 ⁇ L of the test compounds at various concentrations, and the plate was incubated at 37° C. under 5% CO 2 with saturated humidity for 48 h.
  • PrestoBlue® Cell Viability reagent (ThermoFisher Scientific) was added into the well, and the plate was incubated at 37° C. for approximately 60 min.
  • the absorptions at 570 and 600 nm were measured with a SpectraMaxMicroplate reader (Molecular Devices).
  • the absorbance at 570 nm was normalized to that at 600 nm.
  • the normalized absorbance at 570 nm was used for IC 50 calculation following the median-effect plot method (TC Chou. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006; 58:621-681).
  • Metabolic stability in liver microsomes was incubated at a concentration of 1 ⁇ M in human liver microsomes (0.5 mg/mL) (Corning Inc., Tewksbury, Mass.) in 0.1 M phosphate buffer containing 10 mM MgCl 2 , 1 mM NADPH and 2 mM UDPGA for time points ranging from zero to 60 min at 37° C. (see FIG. 1 ).
  • Dasatinib HY10181/CS0100, 302962-49-8, Batch No: 13044 was purchased from MedChemExpress USA (Monmouth Junction, N.J.).
  • LC/MS/MS analysis was carried out on a AB Sciex 4000 Q Trap LC/MS/MS system coupled with Shimadzu Prominence UFLCXR 20 series (including a CBM-20A controller, two LC-20ADXR solvent delivery units, SIL-20AC HT autosampler, CTO-20A column oven, and a SPD-20A UV detector).
  • Compound V and compound III (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 20% propylene glycol 300, and 8% dextrose solution (50%) for intravenous dosing, and at a concentration of 0.25 mg/mL (each compound) in water containing 5% DMSO, 20% propylene glycol 300, and 10% dextrose solution (50%) for oral dosing.
  • Compound X and dasatinib (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 30% propylene glycol 300 and 10% dextrose solution (50%) for intravenous dosing, and at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, 50% propylene glycol 300, and 10% Solutol® HS 15 (Sigma-Aldrich) for oral dosing.
  • Compound XI and compound III (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 30% propylene glycol 300, and 10% dextrose solution (50%) for intravenous dosing and at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, 50% propylene glycol, and 10% Solutol® HS 15 for oral dosing.
  • Blood samples were collected into tubes containing EDTA as the anticoagulant at 0, 0.25 (intravenous only), 0.5, 1, 2, 4, 8, 10, and 24 h post-dosing, and plasma samples were prepared by centrifugation. The plasma samples were mixed with 3 ⁇ volumes of acetonitrile containing 100 nM reserpine as the internal standard, and centrifuged at 4000 RPM for 15 min.
  • the supernatants were injected for LC/MS analysis which were carried out on a AB Sciex 4000 Q Trap LC/MS/MS system coupled with Shimadzu Prominence UFLCXR 20 series (including a CBM-20A controller, two LC-20ADXR solvent delivery units, SIL-20AC HT autosampler, CTO-20A column oven, and a SPD-20A UV detector).
  • the samples were separated on a Phenomenex Columbus column (C18, 4 ⁇ m, 50 ⁇ 2 mm) eluted with two solvent systems: 2 mM ammonium acetate in water containing 0.1% formic acid (A) and methanol (B) at a linear gradient starting with 25% B.
  • Electrospray ionization in positive mode was used to acquire LC/MS/MS data. Plasma concentrations of compounds III, IV, V, X, XI and dasatinib were quantitated using standard curves, respectively.
  • Lung Tissue and Plasma Concentration Ratios Compound X and dasatinib (2-in-1) were both dissolved at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, and 50% propylene glycol 300, and 9% Solutol® HS 15. Compound XI and dasatinib (2-in-1) were both dissolved at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, 20% propylene glycol 300.
  • Blood samples were collected into tubes containing EDTA as the anticoagulant at 1, 2, 4, 10 h post-dosing for Compound X, and predose, 1, 2, 3, 8, 10 and 24 h post-dosing for Compound XI.
  • the plasma samples were prepared by centrifugation. Lung samples were also collected at each of the above time points.
  • the lung samples were homogenated in 4 ⁇ distilled water (v/w).
  • the plasma samples and the homogenated lung tissue samples were mixed with 3 ⁇ volumes of acetonitrile containing 100 nM reserpine as the internal standard, and centrifuged at 4000 RPM for 15 min.
  • the supernatants were injected for LC/MS analysis which were carried out on a AB Sciex 4000 Q Trap LC/MS/MS system coupled with Shimadzu Prominence UFLCXR 20 series (including a CBM-20A controller, two LC-20ADXR solvent delivery units, SIL-20AC HT autosampler, CTO-20A column oven, and a SPD-20A UV detector).
  • the samples were separated on a Phenomenex Columbus column (C18, 4 ⁇ m, 50 ⁇ 2 mm) eluted with two solvent systems: 2 mM ammonium acetate in water containing 0.1% formic acid (A) and methanol (B) at a linear gradient starting at 25%.
  • Electrospray ionization in positive mode was used to acquire LC/MS/MS data.
  • the plasma and tissue concentrations of Compounds X, XI and dasatinib were determined using standard curves, respectively.

Abstract

The present invention concerns novel deuterated and non-deuterated cyclic chemical compounds and their corresponding salts thereof active on protein kinases in general, and in particular as inhibitors of protein kinases. Additionally, methods of treating mammals with protein kinase-mediated diseases or conditions by administering a therapeutically effective amount of the novel deuterated or non-deuterated cyclic chemical compound and their corresponding salts thereof.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit to U.S. Provisional Application No. 62/539,785 filed on Aug. 1, 2017 and to U.S. Provisional Application No. 62/433,410 filed on Dec. 13, 2016, the entire contents of which are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to novel deuterated and non-deuterated cyclic chemical compounds and salts thereof, to methods of using such compounds in treating protein kinase-mediated diseases or conditions such as autoimmune and cancer diseases or conditions, to pharmaceutical compositions of said compounds, and to combination treatments of said compounds with co-administered therapeutic agents.
  • BACKGROUND OF THE INVENTION
  • The information provided herein is intended solely to assist the understanding of the reader. None of the information provided nor references cited is admitted to be prior art to the present invention.
  • The identification of the molecular events that undertie the development of human diseases presents a major challenge in the design of improved strategies in the prevention, management, and cure of certain diseases (Lahiry P et al. Kinase mutations in human disease: interpreting genotype-phenotype relationships. Nat Rev Genet. 2010; 11(1):60-74).
  • The role of aberrantly regulated protein tyrosine kinases (PTKs) in human diseases is the subject of intense investigation (Lahiry id.). Protein kinases are regulators of cellular signaling, and their functional dysregulation is common in carcinogenesis, autoimmune reactions, and many other disease states or conditions (Lahiry id.; Vargas L et al. Inhibitors of BTK and ITK: state of the new drugs for cancer, autoimmunity and inflammatory diseases. Scand J Immunol. 2013; 78(2):130-9; Nobel M E et al. Protein kinase inhibitors: insights into drug design from structure. Science. 2004; 303:1800-1805). The human genome encodes over 500 protein kinases that share a catalytic domain conserved in sequence and structure but which are notably different in how their catalysis is regulated (Manning G et al. The protein kinase complement of the human genome. Science. 2002; 298:1912-1934; Nobel id.). Protein kinases regulate key signal transduction cascades that control or are involved in the control of physiological functions, including cellular growth and proliferation, cell differentiation, cellular development, cell division, stress response, transcription regulation, aberrant mitogenesis, angiogenesis, abnormal endothelial cell-cell or cell-matrix interactions during vascular development, inflammation, Jun-N-terminal kinase (JNK) signal transduction, and several other cellular processes (Manning id). Protein kinase inhibitors have been established as promising drugs that inhibit overactive protein kinases in cancer cells (Gross S et al. Targeting cancer with kinase inhibitors. J Clin Invest. 2015; 125(5):1780-1789; Vargas id).
  • A partial, non-limiting list of kinases includes: ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN (isoform a), FYN (isoform b), HCK, KIT, LCK, LYNa, PDGFRα, PDGFRβ, SRC, SRM, YES, PIK3CA/PIK3R1 (Manning id). Aberrant kinase activity has been observed in many disease states including benign and malignant proliferative conditions as well as diseases resulting from inappropriate activation of the immune and nervous systems.
  • The novel compounds of this invention inhibit the activity of one or more protein kinases and are expected to be useful in treating kinase-related diseases or conditions.
  • SUMMARY OF INVENTION
  • The present invention concerns novel deuterated and non-deuterated cyclic chemical compounds and salts thereof active on protein kinases in general, and in particular as inhibitors of protein kinases. Additionally, methods of treating mammals with protein kinase-mediated diseases or conditions by administering a therapeutically effective amount of the novel deuterated or non-deuterated cyclic chemical compound and/or salts thereof to such mammals in need thereof.
  • In one aspect, the present invention provides compounds having formula I:
  • Figure US20180099961A1-20180412-C00001
  • all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof;
      • wherein
      • W1, W2, and W3 are independently hydrogen or deuterium;
      • Y is carbon or nitrogen;
      • R1 is

  • -Q-A
        • wherein
        • Q is a single bond directly attaching A to a ring carbon atom, or a methylene or ethylene group connecting A to a ring carbon atom; and
        • A is
  • Figure US20180099961A1-20180412-C00002
        • wherein
        • Y1 and Y2 are independently carbon or nitrogen;
        • Z1 and Z2 are independently hydrogen, —(CH2)n—OR5 where n is an integer number from 0 to 4, and R5 is hydrogen, lower alkyl, or lower alkenyl, with the proviso that when n is 1 and R5 is hydrogen, R1 is not a 1-piperidinyl group, and that when n is 2, R5 is hydrogen, and R1 is a 1-piperazinyl group, W2 is deuterium, and —NR5R6 where R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl;
        • T1 and T2 are independently an integer number from 0 to 4 with the proviso that when T1 or T2 is 0, —(CH2)T1 or —(CH2)T2 is a single bond, and T1 and T2 are not 0 at the same time;
      • R2 and R3 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo including —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl; and wherein the positions of R1, R2 and R3 are exchangeable.
  • In one aspect, the present invention provides compounds having formula II:
  • Figure US20180099961A1-20180412-C00003
  • all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof:
      • wherein W1, W2, and W3 are independently hydrogen or deuterium;
      • wherein Y is carbon or nitrogen;
      • wherein R2, R3, and R4 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl; and
      • wherein X is independently hydrogen, —(CH2)n—OR5 wherein n is an integer number from 0 to 4 and R5 is hydrogen, lower alkyl, or lower alkenyl, or —NR5R6.
  • In one aspect, the present invention provides compounds having formula III:
  • Figure US20180099961A1-20180412-C00004
  • all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
      • wherein W1, W2, and W3 are independently hydrogen or deuterium;
      • wherein R2, R3, and R4 are independently H; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl.
  • In one aspect, the present invention provides compounds having formula IV:
  • Figure US20180099961A1-20180412-C00005
  • all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
      • wherein W2 is hydrogen or deuterium.
  • In one aspect, the present invention provides compounds having formula V:
  • Figure US20180099961A1-20180412-C00006
  • all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
      • wherein W2 is hydrogen or deuterium.
  • Exemplary compounds include the following deuterated and non-deuterated cyclic chemical compounds.
  • In one aspect, the present invention provides a compound having the structure of compound I:
  • Figure US20180099961A1-20180412-C00007
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound II:
  • Figure US20180099961A1-20180412-C00008
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound III:
  • Figure US20180099961A1-20180412-C00009
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound IV:
  • Figure US20180099961A1-20180412-C00010
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound V:
  • Figure US20180099961A1-20180412-C00011
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound VI:
  • Figure US20180099961A1-20180412-C00012
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound VII:
  • Figure US20180099961A1-20180412-C00013
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound VIII:
  • Figure US20180099961A1-20180412-C00014
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound IX:
  • Figure US20180099961A1-20180412-C00015
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound X:
  • Figure US20180099961A1-20180412-C00016
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XI:
  • Figure US20180099961A1-20180412-C00017
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XII:
  • Figure US20180099961A1-20180412-C00018
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XIII:
  • Figure US20180099961A1-20180412-C00019
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XIV:
  • Figure US20180099961A1-20180412-C00020
  • all salts and prodrugs thereof.
  • In one aspect, the invention provides a method for treating a protein kinase-mediated disease or condition in an animal or human subject wherein the method involves administering to the subject an effective amount of one or more of a compound selected from formulas I, II, III, IV, and/or V, and preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X and/or XI.
  • The protein kinase mediated disease or condition is an autoimmune disease or a cancer. Preferably the autoimmune disease may be at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis. Preferably, the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
  • In one aspect, the invention provides a method of treating a subject suffering from a protein kinase-mediated disease or condition, comprising administering to the subject suffering from a protein kinase-mediated disease or condition in combination with at least one additional therapeutic agent one or more of a compound selected from formulas I, II, III, IV, and/or V, and preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X and/or XI.
  • The terms “treat” or “therapy” and like terms refer to the administration of compounds in an amount effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or condition, i.e., indication, and/or to prolong the survival of the subject being treated. The term “protein kinase-mediated disease or condition” refers to a disease or condition in which the biological function of a protein kinase affects the development, course, and/or symptoms of the disease or condition. A protein kinase-mediated disease or condition includes a disease or condition for which modulation of protein kinase activity provides a positive effect, i.e., one in which treatment with protein kinase inhibitors, including compounds described herein, provides a therapeutic benefit to the subject with or at risk of the disease or condition.
  • In one aspect, the invention provides for pharmaceutical compositions that include a therapeutically effective amount of one or more of a compound selected from formulas I, II, III, IV, and/or V, and preferably compounds I, II, II, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably compounds IV, V, X, and/or XI in free form or in a pharmaceutically acceptable salt form and at least one pharmaceutically acceptable carrier, excipient, and/or diluent.
  • In reference to compounds of the invention a compound or group of compounds includes pharmaceutically acceptable salts of such compound(s) unless clearly indicated to the contrary, prodrug(s), and all stereoisomers and mixtures thereof.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. Metabolic stability of compounds in human liver microsomal incubations. Metabolic stability was determined for compounds I-XIII in incubations with human liver microsomal preparations. Incubations of individual compounds I-XIII at 1 μM concentrations were carried out for up to 1 hour with human liver microsomes (0.5 mg/mL) in 0.1 M phosphate buffer containing 10 mM MgCl2, 1 mM NADPH and 2 mM UDPGA at 37° C. Concentrations at specified times were determined by LC-MS/MS.
  • FIG. 2. (Upper panel) Plasma concentration versus time profiles for compound IV and dasatinib in Sprague-Dawley rats following a single oral gavage dose of compound IV and dasatinib administered together and each dosed at 2.5 mg/kg; (Lower panel) plasma concentration versus time profiles for compounds III and V in Sprague-Dawley rats following a single oral gavage dose of compounds III and V administered together and each dosed at 2.5 mg/kg together.
  • FIG. 3. (Upper panel) Plasma concentration versus time profiles for compounds X and III in Sprague-Dawley rats following a single oral gavage dose of compounds X and III administered together and each dosed at 5 mg/kg; (Lower panel) plasma concentration versus time profiles for compound XI and dasatinib in Sprague-Dawley rats following a single oral gavage dose of compound XI and dasatinib administered together and each dosed at 5 mg/kg together.
  • FIG. 4. Mean ratios of lung tissue concentration to plasma concentration versus time for compound X and dasatinib in mice wherein compound X and dasatinib were administered together as a single oral dose and each dosed at 5 mg/kg. (2-in-1 dosing, N=3).
  • FIG. 5. Mean ratios of lung tissue concentration to plasma concentration versus time for compound XI and dasatinib in mice wherein compound XI and dasatinib were administered together as a single oral dose and each dosed at 5 mg/kg. (2-in-1 dosing, N=3).
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • As used herein the following definitions apply unless clearly indicated otherwise. By “chemical structure” or “chemical substructure” is meant any definable atom or group of atoms that constitute an individually identifiable molecule, portion of a molecule, such as a substituent moiety, a core which is optionally substituted, and the like. Normally, chemical substructures of a ligand can have a role in binding of the ligand to a target molecule, or can influence the three-dimensional shape, electrostatic charge, and/or conformational properties of the ligand.
  • The term “prodrug” is a compound that, upon in vivo administration, is metabolized by one or more steps or processes or otherwise converted to the biologically, pharmaceutically, or therapeutically active form of the compound. To produce a prodrug, the pharmaceutically active compound is modified such that the active compound will be regenerated by metabolic or hydrolytic processes.
  • The term “binds,” in connection with the interaction between a target and a potential binding compound, indicates that the potential binding compound associates with the target to a statistically significant degree as compared to association with proteins generally (i.e., non-specific binding).
  • As used herein, the term “modulating” or “modulate” refers to an effect of altering a biological activity, especially a biological activity associated with a particular biomolecule such as a protein kinase. For example, an agonist or antagonist of a particular biomolecule modulates the activity of that biomolecule, e.g., an enzyme, by either increasing (e.g., agonist, activator), or decreasing (e.g., antagonist, inhibitor) its activity. This type of activity is typically indicated in terms of an half maximal effective concentration (EC50) or half maximal inhibitory concentration (IC50) for an activator or inhibitor, respectively. Additionally, inhibition activity can be expressed in percent inhibition and/or Ki.
  • As used herein in connection with compounds of the invention, the term “synthesizing” and like terms means chemical synthesis from one or more precursor materials. Further, by “assaying” is meant the creation of experimental conditions and the gathering of data regarding a particular result of the experimental conditions. For example, enzymes can be assayed based on their ability to act upon a detectable substrate. A compound or ligand can be assayed based on its ability to bind to a particular target molecule or molecules.
  • “D,” “d,” and “2H” refer to a deuterium atom, a stable isotope of hydrogen with a mass twice that of hydrogen (atomic weight of 2.0144). Hydrogen naturally occurs as a mixture of the isotopes hydrogen (1H), deuterium (2H or D), and tritium (3H or T). The natural abundance of deuterium is about 0.015%. A person skilled in the art would recognize that all chemical compounds with a hydrogen atom actually are present as mixtures of the H and D isotopes, with about 0.015% being the deuterium isotope. Compounds with a level of deuterium that has been enriched to be greater than its natural abundance of 0.015% should be considered unnatural, and as a result novel, over their non-enriched counterparts. The D in structural formulas and chemical compounds herein refers to incorporation of D in amounts greater than 0.015%.
  • The term “lower alkyl” is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups and branched-chain alkyl groups. In certain embodiments, a straight chain or branched chain alkyl has about 6 or fewer carbon atoms in its backbone (e.g., C1-C6 for straight chain, C3-C6 for branched chain).
  • The term “lower alkenyl” refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as straight or branched group of 2-6 carbon atoms, referred to herein as C2-C6 alkenyl.
  • The term “cycloalkyl” refers to a 3-7 membered moncyclic ring of aliphatic groups, including C3-C7, that is optionally substituted with alkyl, alkenyl, alkoxyl, optionally substituted amino, halogens, cyano (—CN), or nitro (—NO2).
  • The terms “alkoxyl” or “alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy, and the like.
  • The terms “heterocyclo,” “heterocyclic,” or “heterocycle” refer to fully saturated or unsaturated, including non-aromatic (i.e., “heterocycloalkyl”) and aromatic (i.e., “heteroaryl”) cyclic groups having from 5 to 10 atoms with at least one heteroatom (e.g. oxygen (“O”), sulfur (“S”), or nitrogen (“N”)) in at least one carbon atom-containing ring. Each ring of the heterocyclic group may have 1, 2, 3, or 4 heteroatoms. The heteroatoms nitrogen and sulfur may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. Further, the heterocyclo may be optionally substituted with amino (—NR5R6), wherein R5 and R6 are independently hydrogen and/or lower alkyl, hydroxyl (—OH), alkoxyl, lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl may be optionally substituted with —OH or alkoxyl groups.
  • “Halogen” refers to chloro (“Cl”), fluoro (“F”), bromo (“Br”), or iodo (“I”).
  • It is to be understood that the compounds provided herein may contain chiral centers. Such chiral centers may be of either the (R) or (S) configuration, or may be a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, or stereoisomeric or diastereomeric mixtures thereof, including racemic mixtures (about 50:50 ratio of enantiomers).
  • The term “pharmaceutically acceptable” means that the indicated material does not have properties that would cause a reasonably prudent medical practitioner to avoid administration of the material to a patient, taking into consideration the disease or condition to be treated and the respective route of administration. For example, it is commonly required that such a material be essentially sterile, e.g., for injectable.
  • The term “pharmaceutically acceptable salts” refers to salts that are non-toxic in the amounts and concentrations at which they are administered. The preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of a compound without preventing it from exerting its physiological effect.
  • The term “pharmaceutically acceptable composition” refers to a pharmaceutically active compound and one or more pharmaceutically acceptable carriers, excipients, and/or diluents.
  • The term “therapeutically effective” or “effective amount” is an amount of a preparation that alone, or together with further doses, and/or in combination with other therapeutic agents produces the desired response. This may involve halting the progression of the disease or delaying the onset of or preventing the disease or condition from occurring, although it may also imply only slowing of the disease or condition temporarily.
  • The term “protein kinase-mediated disease or condition” refers to a disease or condition in which the biological function of a protein kinase affects the development, course, and/or symptoms of the disease or condition.
  • The term “mutants” refers to single or multiple amino acid changes in a protein as compared to the wild-type protein amino acid sequence.
  • Throughout the description and claims of this specification, the word “comprise” and variations of the word, such as “comprising” and “comprises,” means “including but not limited to,” and is not intended to exclude, for example, other additives, components, integers, or steps.
  • Compounds of the Invention
  • In one aspect, the present invention provides compounds having formula I:
  • Figure US20180099961A1-20180412-C00021
  • all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof;
      • wherein
      • W1, W2, and W3 are independently hydrogen or deuterium;
      • Y is carbon or nitrogen;
      • R1 is

  • -Q-A
        • wherein
        • Q is a single bond directly attaching A to a ring carbon atom, or a methylene or ethylene group connecting A to a ring carbon atom; and A is
  • Figure US20180099961A1-20180412-C00022
      • wherein
      • Y1 and Y2 are independently carbon or nitrogen;
      • Z1 and Z2 are independently hydrogen, —(CH2)n—OR5 where n is an integer number from 0 to 4, and R5 is hydrogen, lower alkyl, or lower alkenyl, with the proviso that when n is 1 and R5 is hydrogen, R1 is not a 1-piperidinyl group, and that when n is 2, R5 is hydrogen, and R1 is a 1-piperazinyl group, W2 is deuterium, and —NR5R6 where R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl; T1 and T2 are independently an integer number from 0 to 4 with the proviso that when T1 or T2 is 0, —(CH2)T1 or —(CH2)T2 is a single bond, and T1 and T2 are not 0 at the same time;
        R2 and R are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo including —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl; and wherein the positions of R1, R2 and R3 are exchangeable.
  • Preferably W2 is deuterium or hydrogen, more preferably hydrogen, and W1 and W3 are hydrogen. Y is preferably nitrogen. R2 is preferably lower alkyl, more preferably methyl. R3 is preferably lower alkyl or hydrogen, more preferably hydrogen. R1 is preferably a substituted or unsubstituted saturated five or six membered nitrogen containing heterocyclo ring. The substituted or unsubstituted saturated five membered nitrogen containing heterocyclo ring can be substituted or unsubstituted pyrrolidin-1-yl, preferably 3-hydroxy- or 3-amino-pyrrolidin-1-yl, more preferably 3-hydroxy pyrrolidin-1-yl.
  • In one aspect, the present invention provides compounds having formula II:
  • Figure US20180099961A1-20180412-C00023
  • all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof:
  • wherein W1, W2, and W3 are independently hydrogen or deuterium;
  • wherein Y is carbon or nitrogen;
  • wherein R2, R3, and R4 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl; and
  • wherein X is independently hydrogen, —(CH2)n—OR5 wherein n is an integer number from 0 to 4 and R5 is hydrogen, lower alkyl, or lower alkenyl, or —NR5R6.
  • Preferably W2 is deuterium or hydrogen, more preferably hydrogen, and W1 and W3 are hydrogen. Y is preferably nitrogen. R2 is preferably lower alkyl, more preferably methyl. R5 is preferably lower alkyl or hydrogen, more preferably hydrogen. R4 is preferably lower alkyl or hydrogen, more preferably hydrogen. X is preferably hydrogen, hydroxyl or amine, more preferably hydroxyl.
  • In one aspect, the present invention provides compounds having formula III:
  • Figure US20180099961A1-20180412-C00024
  • all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
      • wherein W1, W2, and W3 are independently hydrogen or deuterium;
      • wherein R2, R3, and R4 are independently H; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl.
  • Preferably W2 is deuterium or hydrogen, more preferably hydrogen, and W1 and W3 are hydrogen. Y is preferably nitrogen. R2 is preferably lower alkyl, more preferably methyl. R3 is preferably lower alkyl or hydrogen, more preferably hydrogen. R4 is preferably lower alkyl or hydrogen, more preferably hydrogen.
  • In one aspect, the present invention provides compounds having formula IV:
  • Figure US20180099961A1-20180412-C00025
  • all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
      • wherein W2 is hydrogen or deuterium.
  • In one aspect, the present invention provides compounds having formula V:
  • Figure US20180099961A1-20180412-C00026
  • all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
      • wherein W2 is hydrogen or deuterium.
    Exemplary Compounds
  • In one aspect, the present invention provides a compound having the structure of compound I:
  • Figure US20180099961A1-20180412-C00027
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound II:
  • Figure US20180099961A1-20180412-C00028
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound III:
  • Figure US20180099961A1-20180412-C00029
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound IV:
  • Figure US20180099961A1-20180412-C00030
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound V:
  • Figure US20180099961A1-20180412-C00031
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound VI:
  • Figure US20180099961A1-20180412-C00032
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound VII:
  • Figure US20180099961A1-20180412-C00033
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound VIII:
  • Figure US20180099961A1-20180412-C00034
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound IX:
  • Figure US20180099961A1-20180412-C00035
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound X:
  • Figure US20180099961A1-20180412-C00036
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XI:
  • Figure US20180099961A1-20180412-C00037
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XII:
  • Figure US20180099961A1-20180412-C00038
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XIII:
  • Figure US20180099961A1-20180412-C00039
  • all salts and prodrugs thereof.
  • In one aspect, the present invention provides a compound having the structure of compound XIV:
  • Figure US20180099961A1-20180412-C00040
  • all salts and prodrugs thereof.
  • Protein Kinase Targets and Indications of the Invention
  • Protein kinases play key roles in propagating biochemical signals in diverse biological pathways. As such, kinases represent important control points for small molecule therapeutic intervention. More than 500 kinases have been described, and specific kinases have been implicated in a wide range of diseases or conditions. In one aspect, the invention provides methods for treating a protein kinase-mediated disease or condition in an animal or human subject, (i.e., indications), such as without limitation, autoimmune disease, hyperproliferative disease, cancer, cardiovascular disease, inflammatory disease, neurological disease, and other diseases.
  • Preferably, the protein kinase-mediated disease or condition is an autoimmune disease or cancer. More preferably, the autoimmune disease is at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis; and the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
  • In another aspect, the invention provides a method for modulating the activity of a protein kinase selected from the group consisting of ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN, HCK, KIT, LCK, LYN, PDGFRα, PDGFRβ, SRC, SRM, YES, PIK3CA/PIK3R1 by administering an effective dose amount of one or more compounds having formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI. Upper and lower case for letters used in kinase nomenclature are used interchangeably in the present document.
  • In another aspect, the invention provides methods for treating a protein kinase-mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of a composition including one or more compounds having formulas I, II, III, IV and/or V, preferably one or more of compounds I, II, III, IV, VI, V, VII, VIII, IX, X, XI, XII, XIII, and XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and XI.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN (isoform a), FYN (isoform b), HCK, KIT, LCK, LYNa, PDGFRα, PDGFRβ, SRC, SRM, YES, PIK3CA/PIK3R1 by administering an effective amount of one or more compounds having formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI.
  • A number of different assays for kinase activity can be utilized for testing to determine active modulators and/or determining specificity of a modulator for a particular kinase or group of kinases. In addition to the assay mentioned in the Examples below, the person of ordinary skill in the art will know and understand that other assays that can be utilized or can be modified for a particular application.
  • In a commonly used in vitro screen for measuring inhibition of a battery of selected protein kinases (see Example 15) including ABL, ABL(E255K), ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN, HCK, KIT, LCK, LYN, PDGFRα, PDGFRβ, SRC, SRM, YES, and PIK3CA/PIK3R1, compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, and XIII were found to display potent activity to inhibit BTK, BMX, ABL, ABL(E255K), SRC, ACK, ARG, BLK, DDR2, EPHA, FGR, FMS, FRK, FYN, HCK, LCK, LYN, PDGFRα, PDGFRβ, YES, and PIK3CA/PIK3R1, among others.
  • In the above referenced in vitro screen of the battery of protein kinases (see Example 15), compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, and XIII displayed greater than 50% inhibition at 10 nM of BTK, BMX, ABL, ABL(E255K), SRC, ACK, ARG, BLK, DDR2, EPHA1, FGR, FMS, FRK, FYN (isoform a), HCK, LCK, LYNa, PDGFRα, PDGFRβ, YES, and PIK3CA/PIK3R1.
  • As a further test of biological activity, compounds of the invention were assayed for inhibition of cell growth using diffuse large B-cell lymphoma cell line SU-DHL-4 and chronic myelogenous leukemia cell line K-562 (see Example 16). In this cell-based assay, the IC50 values for compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV were all less than 20 nM.
  • Protein kinase targets for compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV include, but are not limited to the following: ABL, ACK, ARG, BLK, BMX, BRK, BTK, CSK, DDR1, DDR2, EGFR, EPHA1, FGR, FMS, FRK, FYN, HCK, KIT, LCK, LYN, PDGFRα, PDGFRβ, SRC, SRM, YES, and PIK3CA/PIK3R1.
  • The Tec family of kinases forms the second largest class of cytoplasmic protein tyrosine kinases after the Src family kinases (SFKs) and consists of five mammalian members: Btk, Bmx (bone marrow kinase on the X-chromosome, also known as Etk), Itk (IL-2 inducible T-cell kinase), RIk (resting lymphocyte kinase, also known as Txk), and Tec (Hartkamp et al. Bruton's tyrosine kinase in chronic inflammation: from pathophysiology to therapy. Int J Interferon Cytokine Mediat Res. 2015; 7: 27-34). Most of the Tec family of kinases are primarily expressed in the hematopoietic system, although both Tec and Bmx are also expressed in stromal tissues such as liver and endothelial cells, respectively. Activation of Tec family kinases upon cell-surface receptor triggering requires relocalization of the protein to the plasma membrane, which is mediated by the interaction of the PH domain with the lipid phosphatidylinositol (3,4,5) P3, formed by activated phosphatidylinositol-3 kinase. Subsequent phosphorylation by SFKs and autophosphorylation of tyrosine 223 result in the complete activation of Tec family of kinases.
  • BTK is the best-known member of the Tec family of kinases with BTK mutations leading to X-linked agammagiobulinemia in men and X-linked immunodeficiency in mice. BTK is a key regulator of B-cell development, activation, signaling, and survival (Hartkamp id). In addition, BTK plays an important role in a number of other hematopoietic cell-signaling pathways, e.g., toll-like receptor (TLR) and cytokine receptor-mediated TNF-alpha production in macrophages, IgE receptor (FcepsilonRI) signaling in mast cells, inhibition of Fas/APO-1 apoptotic signaling in B-lineage lymphoid cells, and collagen-stimulated platelet aggregation. BTK and other members of Tec family kinases can play a critical role in autoimmune diseases, such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), type I diabetes (T1D), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), and rheumatoid arthritis (RA). The BTK inhibitor ibrutinib demonstrated high clinical activity in B-cell malignancies, especially in patients with chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), and Waldenstrom's macroglobulinemia (WM). However, resistance to ibrutinib has been demonstrated in a subgroup of patients receiving ibrutinib treatment, mainly due to the development of BTK mutant enzyme C481S (Woyach J A et al. Resistance mechanisms for the Bruton's tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014; 370(24):2286-94).
  • The tyrosine kinase BMX regulates inflammation induced by TNF and other mediators appear to do so by regulating the shared TAK1-TAB complex (Gottar-Guillier M et al. The tyrosine kinase BMX is an essential mediator of inflammatory arthritis in a kinase-independent manner. J Immunology. 2011; 186(10):6014). BMX kinase may play a role in the pathogenesis of glioblastoma, prostate, breast, and lung cancers. BMX has also shown potential as an anti-vascular therapy in combination with radiation or as a sensitizer to chemotherapeutic agents. (Jarboe J S et al. Mini-review: bmx kinase inhibitors for cancer therapy. Recent Pat Anticancer Drug Discov. 2013; 8(3):228-38).
  • Src family kinases (SFKs) consist of 11 nonreceptor tyrosine kinases, including Src, Fyn, Yes, Blk, Yrk, Frk (also known as Rak), Fgr, Hck, Lck, Srm, and Lyn (Sen B, Johnson F M. Regulation of SRC family kinases in human cancers. J Signal Transduct. 2011:ID865819). Src is found in keratinocytes, whereas Blk, Fgr, Hck, Lck, and Lyn are found primarily in hematopoietic cells. Frk occurs chiefly in bladder, breast, brain, colon, and lymphoid cells. Src family kinases are involved in proliferation and migration responses in many cell types.
  • Src is a non-receptor protein tyrosine kinase that plays a multitude of roles in cell signaling. Src is involved in the control of many functions, including cell adhesion, growth, movement, and differentiation. Src is widely expressed in many cell types, and can have different locations within a cell. Numerous human malignancies display increased SRC expression and activity, suggesting that SRC may be intimately involved in oncogenesis. SRC inhibitor bosutinib has been used or the treatment of Philadelphia chromosome-positive (Ph+) chronic myelogenous leukemia (CML), and saracatinib has been studied for potential treatment of Alzheimer's disease and schizophrenia.
  • ABL is a cytoplasmic and nuclear protein tyrosine kinase that has been implicated in processes of cell differentiation, cell division, cell adhesion, and stress response (Hantschel O. Structure, regulation, signaling, and targeting of abl kinases in cancer. Genes Cancer. 2012; 3:436-46). ABL mutations are associated with cancers such as chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), and acute myelogenous leukemia (AML). Several ABL inhibitors such as imatinib, dasatinib, and nilotinib have been used for treatment of CML, ALL, and AML. Dasatinib, a potent inhibitor of BCR-ABL is used for treatment of newly diagnosed Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML) in chronic phase, chronic, accelerated, or myeloid or lymphoid blast phase Ph+ CML with resistance or intolerance to prior therapy including imatinib, and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) with resistance or intolerance to prior therapy. However, dasatinib is associated with severe respiratory toxicity such as pleural effusion and pulmonary hypertension, which could be a result of dasatinib accumulation in lung tissue. (Quinths-Cardama A, et al. Pleural effusion in patients with chronic myelogenous leukemia treated with dasatinib after imatinib failure. J Clin Oncol. 2007; 25(25):3908-14; Guignabert C, et al. Dasatinib induces lung vascular toxicity and predisposes to pulmonary hypertension. J Clin Invest. 2016; 126(9):3207-18).
  • LCK is a 57.9 kDa membrane-associated non-receptor tyrosine kinase encoded by chromosome Ip34.3. The protein structure comprises an SH3 and SH2 domain. LCK inhibitors may be useful in treating acute lymphoblastic leukemia, T-cell lymphoma, lymphopenia, renal carcinoma, colon carcinoma, severe combined immunodeficiency, multiple sclerosis, inflammatory bowel, and type I diabetes.
  • Frk is a 58.5 kDa tyrosine kinase encoded by chromosome 6q21-q22.3. The structure comprises an SH2, an SH3, and a tyrosine kinase domain. Inhibition of Frk could provide means to suppress beta-cell destruction in type I diabetes. Frk inhibitors may be useful in treating acute myeloid leukemia and type I diabetes.
  • Fyn is a 60.6 kDa non-receptor tyrosine kinase encoded by chromosome 6q21. Fyn is involved in regulation of mast cell degranulation in a synergistic confluence of Fyn and Lyn pathways at the level of protein kinase C and calcium regulation. Fyn inhibitors may be useful in treating Alzheimer's disease, schizophrenia, and in prevention of metastases, e.g., in melanoma and squamous cell carcinoma.
  • HCK is a 59.5 kDa tyrosine kinase encoded by chromosome 20ql 1.21. The protein structure comprises an SH3, an SH2, and a bipartite kinase domain. HCK inhibitors may be useful in treating chronic myelogenous leukemia and acute lymphocytic leukemia.
  • Kit is a 109.9 kDa transmembrane tyrosine kinase encoded by chromosome 4q12. Kit plays an important role in the development of melanocytes, mast, germ, and hematopoietic cells. Aberrant expression and/or activation of Kit has been implicated in a variety of pathologic states. Kit inhibitors may be useful in treating malignancies, including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis, inflammatory bowel syndrome, transplant rejection, and hypereosinophilia.
  • LCK is a 57.9 kDa membrane associated non-receptor tyrosine kinase encoded by chromosome Ip34.3. The protein structure comprises an SH3 and SH2 domain. LCK inhibitors may be useful in treating acute lymphoblastic leukemia, T-cell lymphoma, lymphopenia, renal carcinoma, colon carcinoma, severe combined immunodeficiency, multiple sclerosis, inflammatory bowel, and type I diabetes.
  • Platelet-derived growth factor receptors (PDGF-R) are cell surface tyrosine kinase receptors for members of the platelet-derived growth factor (PDGF) family. PDGF subunits-A and -B are important factors regulating cell proliferation, cellular differentiation, cell growth and development, and many diseases including cancer. There are two forms of the PDGF-R, alpha and beta, each encoded by a different gene. PDGFRα is a 122.7 kDa transmembrane tyrosine kinase encoded by chromosome 4ql2 (symbol: PDGFRA). PDGFRβ is a 124.0 kDa transmembrane tyrosine kinase encoded by chromosome 5q31-q32 (symbol: PDGFRB). PDGFR inhibitors may be useful in treating various diseases such as idiopathic hypereosinophilic syndrome, chronic eosinophilic leukemia, glioma, gastrointestinal stromal tumors (GISTs), juvenile myelomonocytic leukemia, metastatic medulloblastoma, atherogenesis, and restenosis.
  • Yes is a 60.8 kDa tyrosine kinase encoded by chromosome 18pl 1.31-pl 1.21 (symbol: YESI). The structure of Yes comprises SH3 and SH2 domains followed by a TK domain. The YES oncogene is homologous to the Yamaguchi sarcoma virus gene, and the amino acid sequence of Yes shows a high degree of homology with that of the SRC gene product of Roussarcoma virus. The Yes kinase is highly expressed in multiple mammalian cell types, including neurons, spermatozoa, platelets, and epithelial cells. The target kinase Yes is amplified and overexpressed in various cancers including esophageal squamous cell carcinoma. Yes inhibitors may be useful in treating cancers including esophageal squamous cell carcinoma.
  • In one aspect, compounds of formulas I, II, III, IV, and V, preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV (compounds I-XIV), including salts, prodrugs, and/or isomers thereof, can be used in preparation of medicaments for the treatment of a kinase-mediated disease or condition, in particular when the disease or condition is an autoimmune disease or cancer.
  • The amounts of compounds of formulas I, II, III, IV, and V, compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII and XIV (compounds I-XIV), including salts, prodrugs, and/or isomers thereof, to be administered can be determined by standard procedures taking into account factors such as the compound's IC50; the biological half-life of the compound; the age, size, and weight of the subject; and the condition associated with the subject. In general, routine experimentation in clinical trials will determine specific ranges for optimal therapeutic effect for each therapeutic agent and each administrative protocol and administration to specific patients will be adjusted to within effective and safe ranges depending on the patient's condition and responsiveness to initial administration. However, the ultimate administration protocol will be regulated according to the judgment of the attending clinician considering such factors as age, gender, condition, and size of the patient. Generally, doses of active compounds may range from about 0.01 mg/kg per day to about 1000 mg/kg per day. Compounds described herein can be administered in single or multiple doses.
  • Preclinical Pharmacokinetics
  • In Vitro Metabolic Stability:
  • An in vitro metabolic stability study in human liver microsomes was conducted for compounds I through XIII and dasatinib (see Example 17 for experimental conditions). Results from this study are shown in FIG. 1 and table 1.
  • Compounds I, IV, V, VI, VII, VIII, X, XI, XII and XIII displayed significantly greater stability as compared to dasatinib. The in vitro tin for compounds I, IV, V, VI, VII, VIII, X, XI, XII, and XIII were all >59 min compared to 16 min for dasatinib. This significant increase in metabolic stability for compounds I, IV, V, VI, VII, VIII, X, XI, XII, and XIII was unexpected. Longer in vitro metabolic stability half-life (t1/2) is an indicator of longer in vivo human plasma t1/2 for these compounds compared to dasatinb. Therefore, these compounds are expected to have a more favorable pharmacokinetic profile compared to dasatinib, specifically longer t1/2, longer duration of action, less first pass effect, and higher oral bioavailability. The intrinsic clearances for compounds I, IV, V, VI, VII, VIII, X, XI, XII, and XIII were less than 24 μL/min/mg compared to 88 μL/min/mg for dastatinib.
  • Surprisingly compound III, a deuterium labeled analog of dasatinib, was found to have similar metabolic stability to dasatinib (FIG. 1, table 1). This was an unexpected finding given the reported in vitro metabolism of dasatinib in human liver microsomes where hydroxylation at the 4-position of the 2-chloro-6-methylphenyl ring is a route of oxidative metabolism (Christopher U et al. Biotransformation of [14C]dasatinib: in vitro studies in rat, monkey, and human and disposition after administration to rats and monkey. Drug Metab. Dispos. 2007; 36(7):1341-1356).
  • Compounds I, IV, V, VI, VII, VIII, X, XI, XII, and XIII, showed unexpectedly significantly increased metabolic stability in human liver microsomes compared to dasatinib, and deuterium substitution results in additional unexpected results as compared to dasatinib.
  • TABLE 1
    In vitro t1/2 and intrinsic clearance (CLint) of
    Compounds in human liver microsomal incubations*
    In vitro t1/2 (min) CLint (μL/min/mg)
    Dasatinib 16 88
    Compound I 261 5
    Compound II 32 44
    Compound III 24 58
    Compound IV 88 16
    Compound V 67 21
    Compound VI 403 3
    Compound VII 404 3
    Compound VIII >500 <3
    Compound IX 25 56
    Compound X 87 16
    Compound XI 59 24
    Compound XII 448 3
    Compound XIII 261 5
    *Compound I-XIII (1 μM) were incubated with human liver microsomes (0.5 mg/mL) in 0.1M phosphate buffer containing 10 mM MgCl2, 1 mM NADPH and 2 mM UDPGA at 37° C. for various time points through 60 min. The concentrations of remaining test compounds at the various time points were determined by LC-MS/MS.
  • In Vivo Pharmacokinetics:
  • The in vivo pharmacokinetic profiles of compounds III, IV, V, X, XI, and dasatinib were investigated in Sprague Dawley rats following oral and intravenous administration using the technique of 2-in-1 dosing (see Example 18 for experimental conditions). Compound IV was dosed with dasatinib, compound V was dosed with compound III, compound X was dosed with dasatinib, and compound XI was dosed with compound III. Results are shown in FIGS. 2 and 3 and tables 2 and 3.
  • TABLE 2
    Pharmacokinetic parameters of compounds III, IV, V and dasatinib in Sprague-Dawley
    rats following a single intravenous or oral dose administration (Example 18).
    Compound IV Dasatinib Compound V Compound III
    intravenous PO intravenous PO intravenous PO intravenous PO
    Dose (mg/kg) 1 2.5 1 2.5 1 2.5 1 2.5
    Cmax (ng/ml) N/A 82 ± 23 N/A 15 ± 5  N/A 35 ± 19 N/A 9 ± 6
    Tmax (hr) N/A 5.5 ± 4.3 N/A 5.5 ± 4.3 N/A 5.5 ± 4.3 N/A 5.5 ± 4.3
    AUClast (ng/mL*hr) 1877 ± 152 891 ± 341 783 ± 54 193 ± 86  1960 ± 214 524 ± 191 835 ± 114 139 ± 69 
    AUCinf (ng/mL*hr) 1879 ± 153 892 ± 341 784 ± 54 196 ± 85  1961 ± 214 539 ± 188 835 ± 114 144 ± 68 
    t1/2 (hr)  1.4 ± 0.1 2.3 ± 0.2 1.5 ± 0  3.5 ± 0.5  1.4 ± 0.1 4.4 ± 2.8 1.6 ± 0.1 4.7 ± 2.2
    CL (mL/hr/kg) 535 ± 45 N/A 1279 ± 88  N/A 514 ± 54 N/A 1211 ± 157  N/A
    V (L/kg)  1 ± 0 N/A  3 ± 0 N/A  1 ± 0 N/A 3 ± 0 N/A
    F (%) N/A 18.9 ± 6.7  N/A 9.8 ± 3.6 N/A 11.1 ± 4.3  N/A 7.2 ± 4.2
    Cmax—plasma maximum concentration following oral dosing;
    Tmax—time to maximum plasma concentration following oral dosing;
    AUClast—area under the plasma concentration versus time curve to the last detectable concentration;
    AUCinf—area under the plasma concentration versus time curve extrapolated to time infinity;
    t1/2—plasma concentration half-life;
    CL—plasma clearance;
    V—volume of distribution;
    F (%)—percent oral bioavailability as determined by AUCinf (oral) versus AUCinf (intravenous) dose normalized.
  • TABLE 3
    Pharmacokinetic parameters of compounds III, X, XI, and dasatinib
    in Sprague-Dawley rats following a single intravenous or oral dose.
    Compound X Dasatinib Compound XI Compound III
    intravenous PO intravenous PO intravenous PO intravenous PO
    Dose (mg/kg) 1 5 1 5 1 5 1 5
    Cmax (ng/mL) N/A  93 ± 101 N/A 7 ± 7 N/A 102 ± 86 N/A 11 ± 11
    Tmax (hr) N/A 0.5 ± 0  N/A 0.5 ± 0 N/A 0.5 ± 0  N/A 0.5 ± 0
    AUClast (ng/mL*hr) 2519 ± 446 1122 ± 369 1013 ± 68 93 ± 23 2480 ± 243 1420 ± 343 916 ± 161 124 ± 26 
    AUCinf (ng/mL*hr) 2520 ± 446 1204 ± 349 1014 ± 68 97 ± 22 2481 ± 243 1495 ± 273 917 ± 161 137 ± 20 
    t½ (hr)  1.6 ± 0.2  6.8 ± 4.1  1.7 ± 0.3 5.9 ± 1.5  1.6 ± 0.2   6 ± 3.6 1.7 ± 0.2 8.2 ± 4.1
    CL (mL/hr/kg) 405 ± 66 N/A  989 ± 64 N/A 406 ± 42 N/A 1113 ± 193  N/A
    V (L/kg)  1 ± 0 N/A  2 ± 0 N/A  1 ± 0 N/A 3 ± 0 N/A
    F (%) N/A 18.9 ± 2.2 N/A 3.9 ± 1 N/A 24 ± 3.1 N/A 6 ± 0.9
    Cmax—plasma maximum concentration following oral dosing;
    Tmax—time to maximum plasma concentration following oral dosing;
    AUClast—area under the plasma concentration versus time curve to the !ast detectable concentration;
    AUCinf—area under the plasma concentration versus time curve extrapolated to time infinity;
    t1/2—plasma concentration half-life;
    CL—plasma clearance;
    V—volume of distribution;
    F (%)—percent oral bioavailability as determined by AUCinf (oral) versus AUCinf (intravenous) dose normalized.
  • Following oral administrations, compounds IV, V, X, and XI showed surprisingly significantly higher Cmax values of 82±13, 35±19, 93±101, and 102±86 ng/mL, respectively, as compared to dasatinib where Cmax was 15±5 and 7±7 ng/mL and the deuterium analog of dasatinib, compound III, where Cmax was 9±6 and 11±11 ng/mL. The oral bioavailability of compounds IV, V, X, and XI was 18.9±6.7, 11.1±4.3, 18.9±2, and 24±33.1 percent bioavailability (F) compared to dasatinib oral bioavailability of 3.9±1 and 9.8±3.6 percent bioavailability, and compound III where oral bioavailability was 6±0.9 and 7.2±4.2 percent. The results from this study show compounds IV, V, X, and XI have surprisingly much lower intravenous plasma clearance than dasatinib and compound III. This data is consistent with the in vitro metabolic stability data which showed compounds IV, V, X, and XI to be significantly more stable than either dasatinib or compound III, and with significantly lower intrinsic clearance values.
  • Additionally, compounds IV, V, X, and XI were found to have a surprisingly significantly lower intravenous volume of distribution values than dasatinib and compound III, suggesting these compounds are not as widely distributed to tissues as compared to dasatinib and compound III. Further, this suggests these compounds have lower potential than dasatinib for drug-induced organ toxicity.
  • These results indicate that chemical substitution on the pyrimidin-4-yl group to produce compounds IV, V, X, and XI results in unexpected and significant changes in the pharmacokinetic profile of these novel compounds as compared to dasatinib.
  • In Vivo Tissue Distribution: A study was conducted in mice to determine the ratio of parent compound concentrations in lung tissue versus plasma for compounds X, XI, and dasatinib dosed by oral gavage (see Example 19 for experimental conditions). Results are shown in FIGS. 4 and 5.
  • Compounds X and XI unexpectedly showed significantly lower ratios for parent compound concentration levels in lung tissue compared to their plasma concentrations as compared to dasatinib at all time points tested (FIGS. 4 and 5). These results show that compounds X and XI distribute less than dasatinib into lung tissue.
  • Dasatinib is associated with severe respiratory toxicity such as pleural effusion and pulmonary hypertension, which have been attributed to the accumulation of dasatinib in lung tissue. (Quintes-Cardama A et al. Pleural effusion in patients with chronic myelogenous leukemia treated with dasatinib after imatinib failure. J Clin Oncol 2007; 25(25):3908-14; Wang X et al. Differential effects of dosing regimen on the safety and efficacy of dasatinib: retrospective exposure-response analysis of a Phase III study. Clin Pharmacol, 2013; 5: 85-97; Guignabert C et al. Dasatinib induces lung vascular toxicity and predisposes to pulmonary hypertension. J Clin Invest 2016; 126(9):3207-18; lurlo A, et al. Pleural effusion and molecular response in dasatinib-treated chronic myeloid leukemia patients in a real-life Italian multicenter series. Ann Hematol. 2017; Oct. 2. Doi: 10.1007/soo277-017-3144-1).
  • Therefore, the unexpected significantly lower distribution of compounds X and XI into lung tissue from plasma is suggestive of lower potential of these compounds to accumulate in lung tissue, and therefore, a lower potential for drug-induced lung toxicity as compared to dasatinib.
  • Combination Therapy
  • In one aspect, the composition to be administered can include a plurality of different pharmacologically active compounds which can include a plurality of compounds of the invention including one or more compounds of formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI, including salts, prodrugs, and/or isomers thereof, and other therapeutically effective agents for the same disease or condition, wherein the compounds have an additive or a synergistic effect on the disease indication.
  • In one preferred aspect, the invention provides methods for treating a kinase dysfunction-mediated disease or condition in an animal or human subject, wherein the method involves administering to the subject an effective amount of one or more compounds having formulas I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), and more preferably one or more of compounds IV, V, X, and/or XI, salts, prodrugs, and/or isomers thereof, in combination with one or more other therapies for treating the same disease or condition. Other therapies, may include medical procedures (such as surgeries), therapeutic agents, and/or radiation. Therapeutic agents include chemotherapeutic agents, biologics, and immunotherapeutics. Combination therapy can include administration of one or more of compounds described herein with one or more other therapeutics at different times or simultaneous administration. In some embodiments, dosages may be modified for one or more of the compounds of the invention or other therapeutics used in combination, such modifications being a reduction in the dose amounts relative to a compound or therapy used alone.
  • It is understood that use in combination includes use with other medical procedures, therapeutics, and therapies where the other therapy or drug may be administered at different times, within a short time period, such as within 1, 2, 3, or 4-24 hours, or within a longer time period, such a 1-2 days, 2-4 days, 4-7 days, or 1-4 weeks.
  • Use of the compounds of the invention can be in combination with a medical procedure such as surgery, performed on the subject once or infrequently, where the compounds are administered within a short time or longer time before or after the medical procedure.
  • Administration
  • The methods and compounds will typically be used in therapy for human subjects with a kinase-mediated disease or condition. However, they may also be used to treat similar or identical indications in other animal subjects. In this context, the terms “subject” and “animal subject” and the like refer to human and non-human vertebrates, i.e., mammals, such as non-human primates, sports and commercial animals, e.g., equines, bovines, porcines, ovines, rodents, and pets, e.g., canines and felines.
  • Compounds of formulas I, II, III, IV, and V, preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV, and more preferably compounds IV, V. X, and XI may in some cases form salts which are also within the scope of this invention. The term “salts(s)”, as employed herein, denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases.
  • Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, 2-hydroxyethanesulfanates, lactates, maleates, methanesulfonates, nicotinates, nitrates, oxalates, pectinates, phosphates, picrates, salicylates, propionates, tartrates, thiocyantes, toluenesulfonates, and the like.
  • Exemplary basic salts include ammonium salts, alkali metal salts such sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, and salts with organic bases (for example organic amines), and the like.
  • Compounds of formulas I, II, III, IV, and V, including compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV (compounds I-XIV), salts, prodrugs, and/or isomers thereof, can be administered intravenously, intramuscularly, subcutaneously, orally, transdermally, transmucosally, rectally, or by inhalation. In the case of intravenous administration, the dose may be administered as a bolus or infusion.
  • Pharmaceutical compositions for oral use can be obtained, for example, by combining one or more compounds of formula I, II, III, IV, and/or V, preferably one or more of compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and/or XIV, and more preferably one or more of compounds IV, V, X, and/or XI, salts, prodrugs, and/or isomers thereof, with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone). If desired, disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid, or a salt thereof such as sodium alginate.
  • For injection, the compounds of formula I, II, III, IV, and V, preferably compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII and XIV, and more preferably compounds IV, V, X, and XI, salts, prodrugs, and/or isomers thereof, are formulated in sterile liquid solutions, preferably in physically compatible buffers or solutions, such as saline solution, Hank's solution, or Ringer's solution. In addition, the compounds may be formulated in solid form and re-dissolved or suspended immediately prior to use. Lyophilized forms can also be produced.
  • The administration of the compounds described herein can occur simultaneously or sequentially with chemotherapy or radiation. It is understood that administration of other therapeutics or drugs to treat a medical disease or condition can be by a different route of administration or by the same route of administration.
  • In another aspect, the use in combination therapy for any route of administration includes delivery of compounds of the invention and one or more other drug therapeutics delivered by the same route of administration together in any formulation, or administered together, within 1 hour, 2 hours, 3 hours, up to 24 hours, in separate formulations, or by different routes of administration.
  • The invention also provides for a pharmaceutical combination, e.g., a kit, comprising (a) a first agent which is a compound of the invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and (b) at least one co-agent. The kit can include instructions for administration.
  • General Synthetic Methods
  • The present invention also includes processes for the preparation of the novel deuterium-enriched and non-enriched compounds of the invention. In the reactions described, it can be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio, or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups can be used in accordance with standard practice, for example, see TW Greene and PGM Wutsin Protective Groups in Organic Chemistry, John Wiley and Sons, 1991.
  • Compounds of formulas I, II, III, IV, and V, including the exemplary compounds, can generally be synthesized by making appropriate modifications to reagents of scheme 1 (and other applicable schemes) below as would be understood by the person of ordinary skill in the art. It is noted that non-deuterated intermediates are generally commercially available and so the synthesis could be started, for example, at compound 7 (see e.g. compound 7H in schemes 8-14) using the appropriate commercially available intermediate.
  • Compound I can be synthesized by the method shown in Scheme 1.
  • Figure US20180099961A1-20180412-C00041
    Figure US20180099961A1-20180412-C00042
  • The initial step of the chemical process involves reacting 4-bromo-2-chloro-6-methylaniline (1) with allyl bromide to form intermediate 2. To intermediate 2 in dry tetrahydrofuran under nitrogen gas at about ˜70° C. is added dropwise n-butyl lithium (about 1.5 moles n-butyl lithium to 1 mole intermediate 2). After about 40 minutes, the intermediate lithium complex is quenched with d1-methanol (CH3OD; 99% deuterium, #550574; Lot # MKBW0355V, Aldrich, St Louis, Mo.) to selectively incorporate deuterium at the 4 position and give intermediate 3. The allyl protecting groups are removed by standard procedure to give the aniline intermediate 4. Intermediate 4 is reacted with 3-ethoxyacryloyl chloride to form the acrylamide intermediate 5 which is then treated with N-bromosuccinimide and thiourea to form the thiazole intermediate 6. The thiazole intermediate 6 is treated with the base sodium hydride followed by addition of 4,6-dichloro-2-methylpyrimidine to form the 2-[(6-chloro-2-methylpyrimidin-4-yl)amino]-N-(2-chloro-4-deutero-6-methylphenyl)thiazole-5-carboxamide (intermediate 7). Intermediate 7 is reacted with piperidine and N,N-diisopropylethylamine to form the desired product compound I (8).
  • Compound II can be synthesized by the method shown in Scheme 2.
  • Figure US20180099961A1-20180412-C00043
  • In the synthesis of compound II, the first 6 steps in the method are identical to those used in the synthesis of compound I to produce intermediate 7. The last step in the synthesis uses 4-hydroxypiperidine and N,N-diisopropylethylamine to react with intermediate 7 to form the desired product compound II.
  • Compound III can be synthesized by the method shown in Scheme 3.
  • Figure US20180099961A1-20180412-C00044
  • In the synthesis of compound III, the first 6 steps in the method are identical to those used in the synthesis of compound I to produce intermediate 7. The last step in the synthesis uses 1-(2-hydroxyethyl)piperazine (Sigma-Aldrich; St Louis, Mo.) and N,N-diisopropylethylamine to react with intermediate 7 to form the desired addition product compound III.
  • Compound IV can be synthesized by the method shown in Scheme 4.
  • Figure US20180099961A1-20180412-C00045
  • In the synthesis of compound IV, the first 6 steps in the method are identical to those used in the synthesis of compound I to produce intermediate 7. Intermediate 7 is react with(S)-3-hydroxypyrrolidine and N,N-diisopropylethylamine to form the desired product compound IV.
  • Compound V can be synthesized by the method shown in Scheme 5.
  • Figure US20180099961A1-20180412-C00046
  • In the synthesis of compound V, the first 6 steps in the method are identical to those used in the synthesis of compound I to produce intermediate 7. Intermediate 7 is reacted with (R)-3-hydroxypyrrolidine and N,N-diisopropylethylamine to form the desired product compound V.
  • Compound VI can be synthesized by the method shown in Scheme 6.
  • Figure US20180099961A1-20180412-C00047
  • In the synthesis of compound VI, the first 6 steps in the method are identical to those used in the synthesis of compound I to produce intermediate 7. Intermediate 7 is reacted with tert-butyl (S)-pyrrolidin-3-ylcarbamate and N,N-diisopropylethylamine in dioxane to form compound 300-98. Compound 300-98 is deprotected to produce the product compound VI.
  • Compound VII can be synthesized by the method shown in Scheme 7.
  • Figure US20180099961A1-20180412-C00048
  • In the synthesis of compound VII, the first 6 steps in the method are identical to those used in the synthesis of compound I to produce intermediate 7. Intermediate 7 is reacted with tert-butyl (R)-pyrrolidin-3-ylcarbamate and N,N-diisopropylethylamine in dioxane to form compound 300-100. Compound 300-100 is deprotected to produce the product compound VII.
  • Compound VIII can be synthesized by the method shown in Scheme 8.
  • Figure US20180099961A1-20180412-C00049
  • In the synthesis of compound VIII, commercially available intermediate 7H (Combi-Blocks, Inc., San Diego, Calif.) is reacted with piperidine and N,N-diisopropylethylamine in dioxane to form the desired product compound VIII.
  • Compound IX can be synthesized by the method shown in Scheme 9.
  • Figure US20180099961A1-20180412-C00050
  • In the synthesis of compound IX, commercially available intermediate 7H is reacted with 4-hydoxypiperidine and N,N-diisopropylethylamine in dioxane to form the desired product compound IX.
  • Compound X can be synthesized by the method shown in Scheme 10.
  • Figure US20180099961A1-20180412-C00051
  • In the synthesis of compound X, commercially available intermediate 7H is reacted with (S)-pyrrolidin-3-ol hydrochloride and N,N-diisopropylethylamine in dioxane to form the desired product compound X.
  • Compound XI can be synthesized by the method shown in Scheme 11.
  • Figure US20180099961A1-20180412-C00052
  • In the synthesis of compound X, commercially available intermediate 7H is reacted with (R)-pyrrolidin-3-olhydrochloride and N,N-diisopropylethylamine in dioxane to form the desired product compound XI.
  • Compound XII can be synthesized by the method shown in Scheme 12.
  • Figure US20180099961A1-20180412-C00053
  • In the synthesis of compound XII, commercially available intermediate 7H is reacted with tert-butyl (S)-pyrrolidin-3-ylcarbamate and N,N-diisopropylethylamine in dioxane to form intermediate 300-92. Intermediate 300-92 is deprotected to produce compound XII.
  • Compound XIII can be synthesized by the method shown in Scheme 13.
  • Figure US20180099961A1-20180412-C00054
  • In the synthesis of compound XIII, commercially available intermediate 7H is reacted with tert-butyl (R)-pyrrolidin-3-ylcarbamate and N,N-diisopropylethylamine in dioxane to form intermediate 300-92. Intermediate 300-92 is deprotected to produce compound XII.
  • Compound XIV can be synthesized by the method shown in Scheme 14.
  • Figure US20180099961A1-20180412-C00055
  • In the synthesis of compound XIV, commercially available intermediate 7H is reacted with pyrrolidine and N,N-diisopropylethylamine in dimethyl sulfoxide (DMSO) to form compound XIV.
  • EXAMPLES
  • Examples related to the present invention are described below. In more cases, alternative techniques can be used. The examples are intended to be illustrative and are not limiting or restrictive to the scope of the invention. In some examples, the mass spectrometry results indicated that a compound may have more than one value due to the isotope distribution of an atom in the molecule, such as a compound having a bromo or chloro substituent.
  • Example 1 Synthesis of N-(2-chloro-4-deuterio-6-methyl-phenyl)-2-[[2-methyl-6-(1-piperidyl)pyrimidin-4-yl]amino]thiazole-5-carboxamide(Compound I)
  • Preparation of N,N-diallyl-4-bromo-2-chloro-6-methylaniline (2): To a 250 mL flask were added 4-bromo-2-chloro-6-methylaniline (3 g, 13.61 mmol), dimethyl formamide (DMF) (50 mL), and sodium carbonate (6.37 g, 60.09 mmol, 4.4 eq) at 0° C. With stirring, allyl bromide (9.4 mL, 108.62 mmol, 8 eq) was added dropwise at 0-5° C. under nitrogen. After addition, the mixture was stirred at room temperature for 20 min and heated at 120° C. under nitrogen for 3 h when TLC analysis showed no presence of the starting material. The mixture was then cooled to room temperature and poured into cold water followed by extraction with ethyl acetate (EtOAc) (2×150 mL). The combined organic layers were washed with brine (3×100 mL), dried (sodium sulfate, Na2SO4), and concentrated under reduced pressure to get a black-brown liquid residue, which was purified by column chromatography (hexanes only) to give compound 2 (3.9 g, 95%) as a pale brown liquid.
  • Preparation of N,N-diallyl-2-chloro-4-deutero-6-methylaniline (3): To a solution of compound 2 (2 g, 6.65 mmol) in tetrahydrofuran (THF) (40 mL) at −70° C. under nitrogen was added 2.5 M solution of n-butyl lithium (4 ml, 10 mmol) dropwise. After addition, the mixture was stirred at −70° C. for 40 min and then quenched by addition of deuterated methanol (MeOD) (2 mL, 49.2 mmol, 99% deuterium, #550574; Lot # MKBW0355V, Aldrich, St Louis, Mo.) dropwise. The reaction mixture was then stirred from −70° C. to −20° C. over 40 min when thin layer chromatograph (TLC) analysis (hexanes only) showed the reaction was complete. The mixture was poured into cold water (100 mL) followed by extraction with EtOAc (2×100 mL). The combined organic layers were washed with brine, dried (Na2SO4), and concentrated under reduced pressure. The residue was purified by column chromatography (hexanes only, then EtOAc:hexanes; 1:20) to give compound 3 (1.38 g, 93%) as a pale yellow liquid. 1H Nuclear magnetic resonance (NMR) (CDCl3): 7.20 (s, 1H), 7.09 (s, 1H), 5.95 (m, 2H), 5.21-5.20 (m, 4H), 3.79 (d, 4H), 2.41 (s, 3H). 1H NMR showed absence of the proton signal at the 4 position of compound 3.
  • Preparation of 2-chloro-4-deutero-6-methylaniline (4): To a stirring solution of the aniline 3 (3 g, 13.31 mmol) in DCM (130 mL) were added N,N.dimethylbarbituric acid (8.3 g, 53.16 mmol, 4 eq) and Pd(PPh3)4 (0.5 g, 0.43 mmol, 0.032 eq). The mixture was heated at reflux under nitrogen for 4 h. TLC analysis (EtOAc:hexanes; 1:9) showed the reaction was complete. After the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was dissolved in EtOAc (120 mL) and the organic layer was washed with 10% sodium bicarbonate (NaHCO3) solution (4×60 mL), dried (Na2SO4), and concentrated. The crude product was purified by column chromatography (EtOAc:hexanes; 1:9) to give the desired aniline 4 (1.67 g, 88%) as a pale brown liquid.
  • Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-3-ethoxyacrylamide (5): To a mixture of compound 4 (460 mg, 3.23 mmol), pyridine (0.4 mL, 4.95 mmol, 1.5 eq), and THF (25 mL) at 0-5° C. was added 3-ethoxyacryloyl chloride (666 mg, 4.95 mmol, 1.5 eq) dropwise. After addition, the mixture was stirred at room temperature under nitrogen overnight. TLC analysis (EtOAc:hexanes; 1:2) showed absence of starting material. EtOAc (80 mL) and water (80 mL) were added to the mixture and the organic layer separated and washed with 1N hydrochloric acid (HCl) solution, water, and 5% NaHCO3 solution, dried (Na2SO4), and concentrated to give the compound 5 as a white solid, which was used in the next step without purification.
  • Preparation of 2-amino-N-(2-chloro-4-deutero-6-methylphenyl)thiazole-5-carboxamide (6): To a mixture of the acrylamide 5 (900 mg, 3.74 mmol), 1,4-dioxane(7 mL), and water (7 mL) was added N-bromosuccinimide (730 mg, 4.10 mmol, 1.1 eq) at 0° C. The slurry was stirred at room temperature for 3 h. Thiourea (285 mg, 3.75 mmol, 1 eq) was added and the mixture heated to 80° C. After 3 h, the mixture was cooled to room temperature followed by addition of concentrated ammonium hydroxide solution (1 mL). After stirring, EtOAc (80 mL) and water (80 mL) were added to the mixture. The organic layer was separated and the aqueous layer was extracted with EtOAc (80 mL). The combined organic layers were washed with water, dried (Na2SO4), and concentrated. The residue was subjected to column chromatography (EtOAc-hexanes, 1:4 to 1:2) to give compound 6 (0.82 g, 82%) as a pale brown solid. Liquid chromatography-mass spectrometry (LC-MS) analysis showed a protonated parent ion at 269.14 (M+H).
  • Preparation of 2-[(6-chloro-2-methylpyrimidin-4-yl)amino]-N-(2-chloro-4-deutero-6-methylphenyl)thiazole-5-carboxamide (7): To a mixture of sodium hydride (60%, 67 mg, 1.67 mmol) and THF (15 mL) at 0° C. was added compound 6 (150 mg, 0.56 mmol) in portions. The mixture was stirred at 0° C. for 30 min and a solution of 4,6-dichloro-2-methylpyrimidine (109 mg, 0.67 mmol, 1.2 eq) was added dropwise. The resulting mixture was stirred at room temperature for 3 h when LC-MS analysis showing absence of starting material. The mixture was concentrated to provide the crude compound 7, which was used in next step without purification.
  • Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-(piperdin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (8, compound I): To a mixture of the crude compound 7 (about 0.56 mmol) in dioxane (10 mL) was added piperidine (143 mg, 1.68 mmol, 3 eq) and N,N-diisopropylethylamine (DIEA) (217 mg, 1.68 mmol, 3 eq) at room temperature. The mixture was stirred at 85-90° C. under nitrogen for 20 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was concentrated to dryness and suspended in 50 mL acetonitrile containing 20% HPLC grade water. The mixture was then centrifuged at 4000 rpm for 15 minutes. The pellet was re-suspended in acetonitrile and centrifuged again at 4000 rpm for 15 minutes. The final pellet was dried under nitrogen, and recovered as an off-white solid of the target Compound I (compound 8) (12 mg). The purity of the final product was determined to be greater than 95% by liquid chromatograph-ultraviolet-mass spectrometry (LC-UV-MS). LC-MS: 444.14 (M+H); 1H NMR (DMSO-d6): 11.52 (s, 1H. NH), 9.82 (s, 1H, NH), 8.18 (s, 1H), 7.39 (s, 1H), 7.21 (s, 1H), 6.00 (s, 1H), 3.55 (m, 4H), 3.22 (s, 3H), 2.43 (s, 3H), 1.65 (m, 6H). 1H NMR showed absence of a proton signal at the 4-position of compound I.
  • Example 2 Preparation of N-(2-chloro-4-deuterio-6-methyl-phenyl)-2-[[6-(4-hydroxy-1-piperidyl)-2-methyl-pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound II)
  • Compound II was synthesized starting from 2-[(6-chloro-2-methylpyrimidin-4-yl)amino]-N-(2-chloro-4-deutero-6-methylphenyl)thiazole-5-carboxamide (7) by the synthetic procedure shown in Scheme 2.
  • 2-[(6-chloro-2-methylpyrimidin-4-yl)amino]-N-(2-chloro-4-deutero-6methylphenyl)thiazole-5-carboxamide (7) was prepared in six steps starting from 4-bromo-2-chloro-6-methylaniline as described in Example 1.
  • Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-(4-hydroxypiperidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (compound II): To a mixture of the crude compound 7 (about 0.56 mmol) in dioxane (10 mL) was added 4-hydroxypiperidine (170 mg, 1.68 mmol, 3 eq) and DIEA (217 mg, 1.68 mmol, 3 eq) at room temperature. The mixture was stirred at 85-90° C. under nitrogen for 20 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was concentrated to dryness followed by suspending in 50 mL acetonitrile. The mixture was centrifuged at 4000 rpm for 15 min. The pellet was dissolved in methanol, and purified with column chromatography (methanol-methylene chloride, 1:9). The column chromatograph fractions containing only compound II were combined and dried under a nitrogen flow and recovered as an off-white solid of the target compound II (17 mg). The purity of the final product was determined to be greater than 95% by LC-UV-MS. LC-MS: 460.14 (M+H); 1H NMR (DMSO-d6): 11.42 (s, 1H. NH), 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (s, 1H), 7.24 (s, 1H), 6.05 (s, 1H), 4.78 (s, 1H), 3.95 (m, 2H), 3.75 (m, 1H), 3.22 (s, 3H), 3.18 (m, 2H), 2.43 (s, 3H), 1.85 (m, 2H), 1.35 (m, 2H). 1H NMR showed absence of a proton signal at the 4-position of compound II.
  • Example 3 Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-(4-(2-hydroxyethyl)piperidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound III)
  • To a mixture of the crude compound 7 (about 0.56 mmol) and dioxane (10 mL) were added 4-(2-hydroxyethyl)piperidine (219 mg, 1.68 mmol, 3 eq) and DIEA (217 mg, 1.68 mmol, 3 eq) at room temperature. The mixture was then stirred at 85-90° C. under nitrogen for 20 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was cooled to room temperature, concentrated under reduced pressure to dryness, and suspended in 50 mL acetonitrile containing 20% HPLC grade water. The mixture was then centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in acetonitrile and centrifuged again at 4000 rpm for 15 min. The final pellet was dried under nitrogen, and recovered as an off-white solid of the target compound III (about 40 mg) as an off-white solid. LC-MS: 489.16 (M+H); 1H NMR (DMSO-d6): 11.42 (s, 1H. NH), 9.83 (s, 1H, NH), 8.20 (s, 1H), 7.40 (s, 1H), 7.24 (s, 1H), 6.05 (s, 1H), 4.42 (m, 1H), 3.35 (m, 2H), 2.48 (m, 8H), 2.40 (s, 3H), 2.15 (m, 2H).
  • Example 4 Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-((S)-3-hydroxypyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound IV)
  • To a mixture of the crude compound 7 (about 0.56 mmol) and dioxane (10 mL) were added (S)-pyrrolidin-3-ol hydrochloride (208 mg, 1.68 mmol, 3 eq) and DIEA (400 mg, 3.1 mmol, 5.5 eq) at room temperature. The mixture was then stirred at 85-90° C. under nitrogen for 6 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was cooled to room temperature and concentrated to dryness under reduced pressure, and the resultant residue was suspended in 50 mL acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was then suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatants were combined and concentrated to dryness to afford the target compound IV (about 60 mg) as an off-white solid. LC-MS: 446 (M+H); 1H NMR (DMSO-d6): 11.40 (s, 1H. NH), 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (s, 1H), 7.24 (s, 1H), 5.80 (s, 1H), 4.98 (s, 1H), 4.35 (s, 1H), 2.53 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 5 Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-((R)-3-hydroxypyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound V)
  • To a mixture of the crude compound 7 (about 0.56 mmol) and dioxane (10 mL) were added (R)-pyrrolidin-3-ol hydrochloride (208 mg, 1.68 mmol, 3 eq) and DIEA (400 mg, 3.1 mmol, 5.5 eq) at room temperature. The mixture was then stirred at 85-90° C. under nitrogen for 6 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was cooled to room temperature and concentrated to dryness under reduced pressure, and the resultant residue was suspended in 50 mL acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was then suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatants were combined and concentrated to dryness to afford the target compound V (about 103 mg) as an off-white solid. LC-MS: 446 (M+H); 1H NMR (DMSO-d6): 11.40 (s, 1H. NH), 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (s, 1H), 7.24 (s, 1H), 5.80 (s, 1H), 4.98 (s, 1H), 4.35 (s, 1H), 2.53 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 6 Preparation of N-(2-chloro-4-dutero-6-methylphenyl)-2-[[2-methyl-6-((S)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound VI, 300-101)
  • Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-((S)-3 aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound VII, 300-101). To a mixture of 300-97 (200 mg, 0.51 mmol) and dioxane (8 mL) were added tert-butyl (S)-pyrrolidin-3-ylcarbamate (186 mg, 1 mmol, 2 eq) and DIEA (147 mg, 1.14 mmol, 2 eq) at room temperature. The mixture was then stirred at 90-91° C. under nitrogen for 12 h. LC-MS analysis showed the product peak. The mixture was cooled to room temperature and concentrated under reduced pressure, after addition of methanol (4 mL), the mixture was re-concentrated to give the intermediate 300-100 as a grey solid. LC-MS: 545.12 (M+H).
  • To the crude sample (300-100) was added DCM (4 mL). The mixture was cooled to 5° C., and then a mixture of TFA-DCM (1:1, 5 mL) was added dropwise. After addition, the mixture was stirred at room temperature for 3 h and concentrated under reduced pressure. The residue was mixed with methanol (4 mL), followed by addition of triethylamine (2 mL) and stirring a while. The mixture was concentrated to dryness, then suspended in 50 mL distilled water, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended with 50 mL distilled water, and then centrifuged at 4000 rpm for 15 min. The pellet was further suspended with 100 mL 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatant was evaporated to dryness to afford the target compound VI (300-101) (approximately 108 mg) as an off-white solid. LC-MS: 445 (M+H); 1H NMR (DMSO-d6): 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.30 (s, 1H), 7.19 (s, 1H), 5.80 (s, 1H), 3.60-3.35 (m, 2H), 2.43 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 7 Preparation of N-(2-chloro-4-deutero-6-methylphenyl)-2-[[2-methyl-6-((R)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound VII, 300-99)
  • Preparation of 2-[(6-chloro-2-methylpyrimidin-4-yl)amino]-N-(2-chloro-4-deutero-6-methylphenyl)thiazole-5-carboxamide (300-97). To a stirred mixture of the starting material 300-77-2 (200 mg, 0.74 mmol), 4,6-dichloro-2-methylpyrimidine (147 mg, 0.90 mmol, 1.2 eq), and THF (4 mL) was added a solution of sodium tertiary-butoxide in THF (2M, 1.31 mL, 2.62 mmol, 3.5 eq) dropwise at 0-5° C. After addition, the mixture was stirred at room temperature for 1.5 h and re-cooled to 0-5° C. 2N HCl solution (1 mL) was added dropwise. The mixture was stirred for 15 min and concentrated under reduced pressure. The residue was mixed with EtOAc-hexane (1:1) and stirred for 5 min. The solid was filtered and washed with EtOA-hexane (1:1) and dried to give a yellow solid (210 mg), which was used in the next step without purification. LC-MS: 395.03 (M+H).
  • Preparation of N-(2-chloro-4-dutero-6-methylphenyl)-2-[[2-methyl-6-((R)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound VI, 300-99). To a mixture of 300-97 (200 mg, 0.51 mmol) and dioxane (8 mL) were added tert-butyl (R)-pyrrolidin-3-ylcarbamate (186 mg, 1 mmol, 2 eq) and DIEA (147 mg, 1.14 mmol, 2 eq) at room temperature. The mixture was then stirred at 90-91° C. under nitrogen for 12 h. LC-MS analysis showed the product peak. The mixture was cooled to room temperature and concentrated under reduced pressure. After addition of methanol (4 mL), the mixture was re-concentrated to give the intermediate 300-98 as a grey solid. LC-MS: 545.12 (M+H).
  • To the crude sample (300-98) was added dichloromethane (DCM) (4 mL). The mixture was cooled to 5° C., and then a mixture of trifluoroacetic acid (TFA)-DCM (1:1, 5 mL) was added dropwise. After addition, the mixture was stirred at room temperature for 3 h and concentrated under reduced pressure. The residue was mixed with methanol (4 mL), followed by addition of triethylamine (2 mL) and stirred. The mixture was concentrated to dryness, then suspended in 50 mL distilled water, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended with 50 mL distilled water, and then centrifuged at 4000 rpm for 15 min. The pellet was further suspended with 100 mL 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatant was evaporated to dryness to afford the target compound VII (300-99) as an off-white solid (˜105 mg). LC-MS: 445 (M+H); 1H NMR (DMSO-d6): 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.30 (s, 1H), 7.19 (s, 1H), 5.80 (s, 1H), 3.60-3.35 (m, 2H), 2.43 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 8 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-(piperidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound VIII, H-20)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-(piperidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound VIII, H-20). To a mixture of the starting material 7H (150 mg, 0.38 mmol), dioxane (8 mL) were added piperidine (97 mg, 1.14 mmol, 3 eq) and DIEA (147 mg, 1.14 mmol, 3 eq) at room temperature. The mixture was stirred at 90-91° C. under nitrogen for 15 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was concentrated to dryness and suspended in 50 mL acetonitrile containing 20% HPLC grade water. The mixture was then centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in acetonitrile and centrifuged again at 4000 rpm for 15 min. The final pellet was dried under nitrogen to afford the target compound VIII (H-20) (˜129 mg) as an off-white solid. LC-MS: 443.14 (M+H); 1H NMR (DMSO-d6): 11.52 (s, 1H. NH), 9.82 (s, 1H, NH), 8.18 (s, 1H), 7.39 (m, 1H), 7.21 (m, 2H), 6.00 (s, 1H), 3.55 (m, 4H), 3.22 (s, 3H), 2.43 (s, 3H), 1.65 (m, 6H).
  • Example 9 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-(4-hydroxypiperidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound IX, H-21)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-(4-hydroxypiperdin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (H-21). To a mixture of the starting material 7H (150 mg, 0.38 mmol) and dioxane (8 mL) were added 4-hydroxypiperidine (115 mg, 1.14 mmol, 3 eq) and DIEA (147 mg, 1.14 mmol, 3 eq) at room temperature. The mixture was then stirred at 90-92° C. under nitrogen for 10 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was concentrated to dryness and suspended in 50 mL acetonitrile containing 20% HPLC grade water. The mixture was then centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in acetonitrile and centrifuged again at 4000 rpm for 15 min. The final pellet was dried under nitrogen to afford the target compound IX (H-21) (130 mg) as an off-white solid. LC-MS: 459.14 (M+H); 1H NMR (DMSO-d6): 11.42 (s, 1H. NH), 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (m, 1H), 7.24 (m, 2H), 6.05 (s, 1H), 4.78 (s, 1H), 3.95 (m, 2H), 3.75 (m, 1H), 3.22 (s, 3H), 3.18 (m, 2H), 2.43 (s, 3H), 1.85 (m, 2H), 1.35 (m, 2H).
  • Example 10 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((S)-3-hydroxypyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound X, H-31, 300-89)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((S)-3-hydroxypyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound XI, H-31, 300-89). To a mixture of the starting material 7H (150 mg, 0.38 mmol) and dioxane (8 mL) were added (S)-pyrrolidin-3-ol hydrochloride (141 mg, 1.14 mmol, 3 eq) and DIEA (245 mg, 1.90 mmol, 5 eq) at room temperature. The mixture was then stirred at 90-92° C. under nitrogen for 12 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was cooled to room temperature and concentrated to dryness under reduced pressure, and the resultant residue was suspended in 50 mL acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was then suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatants were combined and concentrated to dryness to afford the target compound X (H-31) (105 mg) as an off-white solid. LC-MS: 445 (M+H); 1H NMR (DMSO-d6): 11.40 (s, 1H. NH), 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (m, 1H), 7.24 (m, 2H), 5.80 (s, 1H), 4.98 (s, 1H), 4.35 (s, 1H), 2.53 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 11 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((R)-3-hydroxypyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound XI, H-30, 300-87)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((R)-3-hydroxypyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound X, H-30, 300-87). To a mixture of the starting material 7H (150 mg, 0.38 mmol) and dioxane (8 mL) were added (R)-pyrrolidin-3-ol (100 mg, 1.1 mmol, 3 eq) and DIEA (147 mg, 1.14 mmol, 3 eq) at room temperature. The mixture was then stirred at 90-92° C. under nitrogen for 12 h. LC-MS analysis showed the product peak. The mixture was not a clear solution. The mixture was cooled to room temperature and concentrated to dryness under reduced pressure, and the resultant residue was suspended in 50 mL acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was then suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended in cooled 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatants were combined and concentrated to dryness to afford the target compound XI(H-30) (˜75 mg) as an off-white solid. LC-MS: 445 (M+H); 1H NMR (DMSO-d6): 11.40 (s, 1H. NH), 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (m, 1H), 7.24 (m, 2H), 5.80 (s, 1H), 4.98 (s, 1H), 4.35 (s, 1H), 2.53 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 12 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((S)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound XII, H-41, 300-93)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((S)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (H-41, 300-93). To a mixture of the starting material 7H (150 mg, 0.38 mmol) and dioxane (8 mL) were added tert-butyl (S)-pyrrolidin-3-ylcarbamate (142 mg, 0.76 mmol, 2 eq) and DIEA (147 mg, 1.14 mmol, 3 eq) at room temperature. The mixture was then stirred at 90-91° C. under nitrogen for 10 h. LC-MS analysis showed the product peak. The mixture was cooled to room temperature and concentrated under reduced pressure. After addition of methanol (4 mL), the mixture was re-concentrated to give the intermediate 300-92 as a grey solid. LC-MS: 544.12 (M+H).
  • To the crude sample (300-92) was added DCM (4 mL). The mixture was cooled to 5° C., and then a mixture of TFA-DCM (1:1, 5 mL) was added dropwise. After addition, the mixture was stirred at room temperature for 3 h and concentrated under reduced pressure. The residue was mixed with methanol (4 mL), followed by addition of TEA (2 mL) and stirring a while. The mixture was concentrated to dryness, then suspended in 50 mL distilled water, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended with 50 mL distilled water, and then centrifuged at 4000 rpm for 15 min. The pellet was further suspended with 100 mL 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatant was evaporated to dryness to afford the target compound XII (H-41) as an off-white solid (approximately 88 mg). LC-MS: 444 (M+H); 1H NMR (DMSO-d6): 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (m, 1H), 7.24 (m, 2H), 5.80 (s, 1H), 3.60-3.35 (m, 2H), 2.53 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 13 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((R)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound XIII, H-40, 300-91)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-((R)-3-aminopyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound XII, H-40, 300-91). To a mixture of the starting material 7H (150 mg, 0.38 mmol) and dioxane (8 mL) were added tert-butyl (R)-pyrrolidin-3-ylcarbamate (142 mg, 0.76 mmol, 2 eq) and DIEA (147 mg, 1.14 mmol, 3 eq) at room temperature. The mixture was then stirred at 90-91° C. under nitrogen for 12 h. LC-MS analysis showed the product peak. The mixture was cooled to room temperature and concentrated under reduced pressure. After addition of methanol (4 mL), the mixture was re-concentrated to give the intermediate 300-90 as a grey solid. LC-MS: 544.12 (M+H).
  • To the crude sample (300-90) was added DCM (4 mL). The mixture was cooled to 5° C., and then a mixture of TFA-DCM (1:1, 5 mL) was added dropwise. After addition, the mixture was stirred at room temperature for 3 h and concentrated under reduced pressure. The residue was mixed with methanol (4 mL), followed by addition of trimethylamine (TEA) (2 mL) and stirring a while. The mixture was concentrated to dryness, then suspended in 50 mL distilled water, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended with 50 mL distilled water, and then centrifuged at 4000 rpm for 15 min. The pellet was further suspended with 100 mL 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatant was evaporated to dryness to afford the target compound XIII(H-40) as an off-white solid (approximately 96 mg). LC-MS: 444 (M+H); 1H NMR (DMSO-d6): 9.83 (s, 1H, NH), 8.19 (s, 1H), 7.40 (m, 1H), 7.24 (m, 2H), 5.80 (s, 1H), 3.60-3.35 (m, 2H), 2.53 (s, 3H), 2.20 (s, 2H), 2.12 (s, 2H), 1.85 (m, 2H).
  • Example 14 Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-(pyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (Compound XIV, H-50)
  • Preparation of N-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-(pyrrolidin-1-yl)pyrimidin-4-yl]amino]thiazole-5-carboxamide (H-50): To a mixture of the starting material 7H (300 mg, 0.76 mmol) and DMSO (10 mL) was added pyrrolidine (162 mg, 2.28 mmol, 3 eq) and DIEA (294 mg, 2.28 mmol, 3 eq) and DMAP (3 mg) at room temperature. The mixture was then heated to 110° C. under nitrogen and stirred at 110° C. for 15 h. LC-MS analysis showed the complete reaction. The mixture was concentrated to dryness, then suspended in 50 mL distilled water, and centrifuged at 4000 rpm for 15 min. The pellet was re-suspended with 50 mL distilled water, and then centrifuged at 4000 rpm for 15 min. The pellet was further suspended with 100 mL 80% acetonitrile, and centrifuged at 4000 rpm for 15 min. The supernatant was evaporated to dryness to afford the target compound XIV (H-50) as an off-white solid (approximately 314 mg). LC-MS: 429 (M+H); 1H NMR (DMSO-d6): 11.52 (s, 1H. NH), 9.82 (s, 1H, NH), 8.18 (s, 1H), 7.39 (m, 1H), 7.21 (m, 2H), 5.79 (s, 1H), 3.35 (m, 4H), 2.39 (s, 3H), 2.23 (s, 3H), 1.90 (m, 4H).
  • Example 15 Protein Kinase Inhibition Studies
  • Off-chip Mobility Shift Assay (MSA) by Cama Biosciences, Inc (Natick, Mass.) was used for measuring the kinase activity and inhibition.
      • 1) The 5 μL of ×4 compound solution, 5 μL of ×4 Substrate/ATP/Metal solution, and 10 μL of ×2 kinase solution were prepared with assay buffer (20 mM HEPES, 0.01% Triton X100, 2 mM DTT, pH 7.5) and mixed and incubated in a well of polypropylene 384 well microplate for 1 or 5 h (depending on kinase) at room temperature.
      • 2) 70 μL of Termination Buffer (QuickScout Screening Assist MSA; Cama Biosciences) was added to the well.
      • 3) The reaction mixture was applied to a LabChip system (Perkin Elmer), and the product and substrate peptide peaks were separated and quantitated.
      • 4) The kinase reaction was evaluated by the product ratio calculated from peak heights of product (P) and substrate (S) peptides (P/(P+S)).
      • 5) The reaction conditions were followed according to assay protocols of Carna Biosciences, Inc (BMA 3F, 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan; www.camabio.com).
      • 6) Data analysis: The readout value of reaction control (complete reaction mixture) was set as a 0% inhibition, and the readout value of background (Enzyme (−)) was set as a 100% inhibition, then the percent inhibition of each test solution was calculated.
  • Compound I showed >50% inhibition at 1 nM on the following recombinant kinases: ABL, ABL(E255K), ACK, ARG, BLK, BMX, BTK, DDR2, EPHA1, FGR, FMS, FRK, FYN (isoform a), HCK, LCK, LYNa, PDGFRα, PDGFRβ, SRC, YES. Compound I showed concentration-dependent inhibition of recombinant BTK, ACK and PDGFRα activity with an IC50 of approximately 0.2, 0.5 and 1.4 nM, respectively. Compound I inhibited recombinant PIK3CA/PIK3R1 activity with an IC50 of approximately 12 nM.
  • Compound I showed >50% inhibition at 10 nM on the following recombinant kinases: YES, FRK, SRC, LYNa, FMS, BMX, ABL, FYN (isoform b), FGR, HCK, FYN (isoform a), LCK, DDR2, ARG, ABL(E255K), BTK, EPHA1, BLK, ACK, SRM, PDGFRβ, PIK3CA/PIK3R1, PDGFRα, CSK, KIT(D816V), KIT(D816Y), BRK.
  • Compound I showed <50% inhibition at 10 nM on the following recombinant kinases: PDGFRα(V561D), DDR1, KIT, KIT(V560G), HER4, KIT(D816E), EGFR(L858R), KIT(V654A), PDGFRα(D842V), EGFR, EGFR(L861Q), EGFR(d746-750), JAK1, RET, RET(S891A), FGFR3, ALK, WNK3, RET(Y791F), BRAF(V600E), RAF1, ROCK1, AurA, MAP2K2, RET(M918T), KDR, FGFR2, Erk1, EGFR(T790M), RET(G691 S), PDGFRα(T6741), p38α, HER2, JAK3, MAP2K1, p38β, EGFR(T790M/L858R), FLT1, BRAF, KIT(T670I), MET, skMLCK, MNK1, MST1, PKD1, JAK2, YES(T3481), FGFR1, EGFR(d746-750/T790M).
  • Compound II showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa. FRK, FYN (isoform a), FYN (isoform b), TEC, BMX, LYNb, ABL, FGR, FMS, BLK, EPHA1, ABL(E255K). Compound II showed <50% inhibition at 1 nM on the following recombinant kinases: PDGFRβ, PDGFRα, PIK3CA/PIK3R1, KIT, KIT(V560G), EGFR, HER2, YES(T3481), ITK, p38α, ABL(T3151), RAF1, p38β, BRAF. Compound II showed concentration-dependent inhibition of recombinant BTK activity with an IC50 of <1 nM.
  • Compound II showed >50% inhibition at 10 nM on the following recombinant kinases: BTK, PDGFRβ, KIT(V560G), PDGFRα, KIT. Compound II showed <50% inhibition at 10 nM on the following recombinant kinases: PIK3CA/PIK3R1, EGFR, p38α, RAF1, HER2, p38β, BRAF.
  • Compound II showed >50% inhibition at 100 nM on the following recombinant kinases: PDGFRα, KIT(V560G), KIT, PDGFRβ, EGFR. Compound II showed <50% inhibition at 100 nM on the following recombinant kinases: p38α, RAF1, PIK3CA/PIK3R1, HER2, p38β, BRAF.
  • Compound IV showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound V showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound VI showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound VII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound VIII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound IX showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound X showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K). Compound X showed concentration-dependent inhibition of recombinant ABL, ABL (E255K), BTK and BTK (C481S) activity with IC50 values of <1 nM, respectively.
  • Compound XI showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K). Compound XI showed concentration-dependent inhibition of recombinant ABL, ABL (E255K), BTK and BTK (C481S) activity with ICsovalues of <1 nM, respectively.
  • Compound XII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Compound XIII showed >50% inhibition at 1 nM on the following recombinant kinases: SRC, YES, LCK, HCK, BTK, LYNa, TEC, BMX, ABL, ABL (E255K).
  • Example 16 Cell Inhibition Assay
  • Cell lines of SU-DHL-4 (ATCCC CRL-2957™), K-562 (ATCC® CCL-243™), and Mino (ATCC® CRL-3000™) were purchased from American Type Culture Collection (ATCC, Manassas, Va.). SU-DHL-4 cells and Mino cells grew in ATCC-formulated RPMI-1640 medium (ATCC) supplemented with 10% fetal bovine serum (FBS)(Gibco, Life Technologies) (complete medium) a T-75 flask at 37° C. under 5% CO2 with saturated humidity. K-562 cells grew in ATCC-formulated Iscove's Modified Dulbecco's Medium (IMDM) supplemented with 10% FBS (Gibco, Life Technologies) (complete medium) a T-75 flask at 37° C. under 5% CO2 with saturated humidity. When the cells grew to a concentration of approximately 1×106 cells/mL, they were diluted to 2.5-5×104 cells/mL with the corresponding complete medium for each cell line. A 200 μL aliquot of the cell suspension was added to the well of a 96-well plate which was pre-added with 1 μL of the test compounds at various concentrations, and the plate was incubated at 37° C. under 5% CO2 with saturated humidity for 48 h. At end of the cell culture, a 10 μL aliquot of PrestoBlue® Cell Viability reagent (ThermoFisher Scientific) was added into the well, and the plate was incubated at 37° C. for approximately 60 min. The absorptions at 570 and 600 nm were measured with a SpectraMaxMicroplate reader (Molecular Devices). The absorbance at 570 nm was normalized to that at 600 nm. The normalized absorbance at 570 nm was used for IC50 calculation following the median-effect plot method (TC Chou. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006; 58:621-681).
  • Compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV showed concentration-dependent inhibition of growth of SU-DHL-4 with IC50 values of <15 nM, respectively.
  • Compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, and XIV showed concentration-dependent inhibition of growth of K562 cells with IC50 values of <2 nM, respectively.
  • Compounds I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII and XIV showed concentration-dependent inhibition of growth of Mino with IC50 values of <50 nM, respectively.
  • Example 17 Metabolic Stability
  • Metabolic stability in liver microsomes:test compounds and dasatinib were incubated at a concentration of 1 μM in human liver microsomes (0.5 mg/mL) (Corning Inc., Tewksbury, Mass.) in 0.1 M phosphate buffer containing 10 mM MgCl2, 1 mM NADPH and 2 mM UDPGA for time points ranging from zero to 60 min at 37° C. (see FIG. 1). Dasatinib (HY10181/CS0100, 302962-49-8, Batch No: 13044) was purchased from MedChemExpress USA (Monmouth Junction, N.J.). The incubation reactions at various time points were quenched by adding 2× volumes of acetonitrile containing 100 nM reserpine. The quenched incubation samples were centrifuged at 4000 RPM for 10 min, and the supernatants injected for LC/MS/MS analysis. LC/MS/MS analysis was carried out on a AB Sciex 4000 Q Trap LC/MS/MS system coupled with Shimadzu Prominence UFLCXR 20 series (including a CBM-20A controller, two LC-20ADXR solvent delivery units, SIL-20AC HT autosampler, CTO-20A column oven, and a SPD-20A UV detector). The samples were separated on a Phenomenex Columbus column (C18, 4 μm, 50×2 mm) eluted with two solvent systems: 2 mM ammonium acetate in water containing 0.1% formic acid (A) and methanol (B) at a linear gradient starting with 25% B. Electrospray ionization in positive mode was used to acquire LC/MS/MS data. The in vitro t1/2 and intrinsic clearance were calculated using method previous reported (Obatch S. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos. 1999; 27(11):1350-9).
  • Example 18 Pharmacokinetics
  • Pharmacokinetics: Compound IV and dasatinib (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 20% propylene glycol 300, and 8% dextrose solution (50%) for intravenous dosing, and at a concentration of 0.25 mg/mL (each compound) in water containing 5% DMSO, 20% propylene glycol 300, and 10% dextrose solution (50%) for oral dosing. Compound V and compound III (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 20% propylene glycol 300, and 8% dextrose solution (50%) for intravenous dosing, and at a concentration of 0.25 mg/mL (each compound) in water containing 5% DMSO, 20% propylene glycol 300, and 10% dextrose solution (50%) for oral dosing. Compound X and dasatinib (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 30% propylene glycol 300 and 10% dextrose solution (50%) for intravenous dosing, and at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, 50% propylene glycol 300, and 10% Solutol® HS 15 (Sigma-Aldrich) for oral dosing. Compound XI and compound III (2-in-1) were both dissolved at a concentration of 0.2 mg/mL (each compound) in water containing 2.5% DMSO, 30% propylene glycol 300, and 10% dextrose solution (50%) for intravenous dosing and at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, 50% propylene glycol, and 10% Solutol® HS 15 for oral dosing.
  • Sprague-Dawley rats (approximate weight 275-300 g, N=3) were dosed intravenously at 5 mL/kg and orally at 10 mL/kg with the above dosing solutions. Blood samples were collected into tubes containing EDTA as the anticoagulant at 0, 0.25 (intravenous only), 0.5, 1, 2, 4, 8, 10, and 24 h post-dosing, and plasma samples were prepared by centrifugation. The plasma samples were mixed with 3× volumes of acetonitrile containing 100 nM reserpine as the internal standard, and centrifuged at 4000 RPM for 15 min. The supernatants were injected for LC/MS analysis which were carried out on a AB Sciex 4000 Q Trap LC/MS/MS system coupled with Shimadzu Prominence UFLCXR 20 series (including a CBM-20A controller, two LC-20ADXR solvent delivery units, SIL-20AC HT autosampler, CTO-20A column oven, and a SPD-20A UV detector). The samples were separated on a Phenomenex Columbus column (C18, 4 μm, 50×2 mm) eluted with two solvent systems: 2 mM ammonium acetate in water containing 0.1% formic acid (A) and methanol (B) at a linear gradient starting with 25% B. Electrospray ionization in positive mode was used to acquire LC/MS/MS data. Plasma concentrations of compounds III, IV, V, X, XI and dasatinib were quantitated using standard curves, respectively.
  • Example 19 Tissue Distribution
  • Lung Tissue and Plasma Concentration Ratios: Compound X and dasatinib (2-in-1) were both dissolved at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, and 50% propylene glycol 300, and 9% Solutol® HS 15. Compound XI and dasatinib (2-in-1) were both dissolved at a concentration of 0.5 mg/mL (each compound) in water containing 5% DMSO, 20% propylene glycol 300. CD-1 mouse (approximate weight 30 g, N=3) were dosed by oral gavage at 10 mL/kg. Blood samples were collected into tubes containing EDTA as the anticoagulant at 1, 2, 4, 10 h post-dosing for Compound X, and predose, 1, 2, 3, 8, 10 and 24 h post-dosing for Compound XI. The plasma samples were prepared by centrifugation. Lung samples were also collected at each of the above time points. The lung samples were homogenated in 4× distilled water (v/w). The plasma samples and the homogenated lung tissue samples were mixed with 3× volumes of acetonitrile containing 100 nM reserpine as the internal standard, and centrifuged at 4000 RPM for 15 min. The supernatants were injected for LC/MS analysis which were carried out on a AB Sciex 4000 Q Trap LC/MS/MS system coupled with Shimadzu Prominence UFLCXR 20 series (including a CBM-20A controller, two LC-20ADXR solvent delivery units, SIL-20AC HT autosampler, CTO-20A column oven, and a SPD-20A UV detector). The samples were separated on a Phenomenex Columbus column (C18, 4 μm, 50×2 mm) eluted with two solvent systems: 2 mM ammonium acetate in water containing 0.1% formic acid (A) and methanol (B) at a linear gradient starting at 25%. Electrospray ionization in positive mode was used to acquire LC/MS/MS data. The plasma and tissue concentrations of Compounds X, XI and dasatinib were determined using standard curves, respectively.
  • It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of appended claim. All publications, patents, and patent applications cited herein are hereby incorporated by reference.

Claims (44)

We claim:
1. A method of treating a subject suffering from a protein kinase-mediated disease or condition, comprising administering to the subject suffering from a protein kinase-mediated disease or condition a therapeutically effective amount of a compound having the structure of formula I:
Figure US20180099961A1-20180412-C00056
all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof;
wherein
W1, W2, and W3 are independently hydrogen or deuterium;
Y is carbon or nitrogen;
R1 is

-Q-A
wherein
Q is a single bond directly attaching A to a ring carbon atom, or a methylene or ethylene group connecting A to a ring carbon atom; and
A is
Figure US20180099961A1-20180412-C00057
wherein
Y1 and Y2 are independently carbon or nitrogen;
Z1 and Z2 are independently hydrogen, —(CH2)n—OR5 where n is an integer number from 0 to 4, and R5 is hydrogen, lower alkyl, or lower alkenyl, with the proviso that when n is 1 and R5 is hydrogen, R1 is not a 1-piperidinyl group, and that when n is 2, R5 is hydrogen, and R1 is a 1-piperazinyl group, W2 is deuterium, and —NR5R6 where R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl;
T1 and T2 are independently an integer number from 0 to 4 with the proviso that when T1 or T2 is 0, —(CH2)T1 or —(CH2)T2 is a single bond, and T1 and T2 are not 0 at the same time;
R2 and R3 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo including —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl;
and wherein the positions of R1, R2 and R3 are exchangeable.
2. A method of treating a subject suffering from a protein kinase-mediated disease or condition comprising administering to the subject suffering from a protein kinase-mediated disease or condition a therapeutically effective amount of a compound having the structure of formula II:
Figure US20180099961A1-20180412-C00058
all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof:
wherein W1, W2, and W3 are independently hydrogen or deuterium;
wherein Y is carbon or nitrogen;
wherein R2, R3, and R4 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl; and
wherein X is independently hydrogen, —(CH2)n—OR5 wherein n is an integer number from 0 to 4 and R5 is hydrogen, lower alkyl, or lower alkenyl, or —NR5R6.
3. A method of treating a subject suffering from a protein kinase-mediated disease or condition, comprising administering to the subject suffering from a protein kinase-mediated disease or condition a therapeutically effective amount of a compound having the structure of formula III:
Figure US20180099961A1-20180412-C00059
all salts, prodrugs, enantiomers and enantiomeric mixtures thereof:
wherein W1, W2, and W3 are independently hydrogen or deuterium;
wherein R2, R3, and R4 are independently H; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo, wherein optionally substituted includes —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl.
4. The method of claim 1, wherein the protein kinase-mediated disease or condition is an autoimmune disease or a cancer.
5. The method of claim 4, wherein the autoimmune disease is at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis.
6. The method of claim 4, wherein the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
7. The method of claim 1, wherein W2 is deuterium, and W1 and W3 are hydrogen.
8. The method of claim 7, wherein said compound is compound I:
Figure US20180099961A1-20180412-C00060
all salts and prodrugs thereof.
9. The method of claim 7, wherein said compound is compound II:
Figure US20180099961A1-20180412-C00061
all salts and prodrugs thereof.
10. The method of claim 7, wherein said compound is compound III:
Figure US20180099961A1-20180412-C00062
all salts and prodrugs thereof.
11. The method of claim 7, wherein said compound is compound IV:
Figure US20180099961A1-20180412-C00063
all salts and prodrugs thereof.
12. The method of claim 11, wherein the protein kinase-mediated disease or condition is an autoimmune disease or a cancer.
13. The method of claim 12, wherein the autoimmune disease is at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis.
14. The method of claim 12, wherein the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
15. The method of claim 7, wherein said compound is compound V:
Figure US20180099961A1-20180412-C00064
all salts and prodrugs thereof.
16. The method of claim 15, wherein the protein kinase-mediated disease or condition is an autoimmune disease or a cancer.
17. The method of claim 16, wherein the autoimmune disease is at least one of systemic lupus erythematosus (SLE), transplant rejection, multiple sclerosis (MS), systemic sclerosis (SSc), primary Sjögren's syndrome (pSS), rheumatoid arthritis (RA), and psoriasis.
18. The method of claim 16, wherein the cancer is at least one of Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML), Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma, marginal zone lymphomas, mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), T-cell lymphomas, and multiple myeloma.
19. The method of claim 7, wherein said compound is compound VI:
Figure US20180099961A1-20180412-C00065
all salts and prodrugs thereof.
20. The method of claim 7, wherein said compound is compound VII:
Figure US20180099961A1-20180412-C00066
all salts and prodrugs thereof.
21. The method of claim 1, wherein W1, W2, and W3 are each hydrogen.
22. The method of claim 21, wherein said compound is compound VIII:
Figure US20180099961A1-20180412-C00067
all salts and prodrugs thereof.
23. The method of claim 21, wherein said compound is compound IX:
Figure US20180099961A1-20180412-C00068
all salts and prodrugs thereof.
24. The method of claim 21, wherein said compound is compound XII:
Figure US20180099961A1-20180412-C00069
all salts and prodrugs thereof.
25. The method of claim 21, wherein said compound is compound XIII:
Figure US20180099961A1-20180412-C00070
all salts and prodrugs thereof.
26. The method of claim 21, wherein said compound is compound XIV:
Figure US20180099961A1-20180412-C00071
all salts and prodrugs thereof.
27. A method of treating a subject suffering from a protein kinase-mediated disease or condition, comprising administering to the subject suffering from a protein kinase-mediated disease or condition in combination with at least one additional therapeutic agent a compound having the structure of formula I:
Figure US20180099961A1-20180412-C00072
all salts, prodrugs, enantiomers, and enantiomeric mixtures thereof;
wherein
W1, W2, and W3 are independently hydrogen or deuterium;
Y is carbon or nitrogen;
R1 is

-Q-A
wherein
Q is a single bond directly attaching A to a ring carbon atom, or a methylene or ethylene group connecting A to a ring carbon atom; and
A is
Figure US20180099961A1-20180412-C00073
wherein
Y1 and Y2 are independently carbon or nitrogen;
Z1 and Z2 are independently hydrogen, —(CH2)n—OR5 where n is an integer number from 0 to 4, and R5 is hydrogen, lower alkyl, or lower alkenyl, with the proviso that when n is 1 and R5 is hydrogen, R1 is not a 1-piperidinyl group, and that when n is 2, R5 is hydrogen, and R1 is a 1-piperazinyl group, W2 is deuterium, and —NR5R6 where R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl;
T1 and T2 are independently an integer number from 0 to 4 with the proviso that when T1 or T2 is 0, —(CH2)T1 or —(CH2)T2 is a single bond, and T1 and T2 are not 0 at the same time;
R2 and R3 are independently hydrogen; halogen; alkoxyl; lower alkyl or lower alkenyl, wherein the lower alkyl or lower alkenyl is optionally substituted with one or more substituents selected from —OH and alkoxyl, wherein alkoxyl is methoxy, ethoxy, propyloxy, or tert-butoxy; or substituted heterocyclo including —NR5R6, wherein R5 and R6 are independently hydrogen, lower alkyl, or lower alkenyl;
and wherein the positions of R1, R2 and R3 are exchangeable.
28. The method of claim 27, wherein the at least one therapeutic agent is a chemotherapeutic agent, biologic agent, steroid, immune suppressor, and non-steroidal anti-inflammatory agent.
29. The method of claim 27, wherein W2 is deuterium, and W1 and W3 are hydrogen.
30. The method of claim 29, wherein said compound is compound I:
Figure US20180099961A1-20180412-C00074
all salts and prodrugs thereof.
31. The method of claim 29, wherein said compound is compound II:
Figure US20180099961A1-20180412-C00075
all salts and prodrugs thereof.
32. The method of claim 29, wherein said compound is compound III:
Figure US20180099961A1-20180412-C00076
all salts and prodrugs thereof.
33. The method of claim 29, wherein said compound is compound IV:
Figure US20180099961A1-20180412-C00077
all salts and prodrugs thereof.
34. The method of claim 33, wherein the at least one therapeutic agent is a chemotherapeutic agent, biologic agent, steroid, immune suppressor, and non-steroidal anti-inflammatory agent.
35. The method of claim 29, wherein said compound is compound V:
Figure US20180099961A1-20180412-C00078
all salts and prodrugs thereof.
36. The method of claim 35, wherein the at least one therapeutic agent is a chemotherapeutic agent, biologic agent, steroid, immune suppressor, and non-steroidal anti-inflammatory agent.
37. The method of claim 29, wherein said compound is compound VI:
Figure US20180099961A1-20180412-C00079
all salts and prodrugs thereof.
38. The method of claim 29, wherein said compound is compound VII:
Figure US20180099961A1-20180412-C00080
all salts and prodrugs thereof.
39. The method of claim 27, wherein W1, W2, and W3 are each hydrogen.
40. The method of claim 39, wherein said compound is compound VIII:
Figure US20180099961A1-20180412-C00081
all salts and prodrugs thereof.
41. The method of claim 39, wherein said compound is compound IX:
Figure US20180099961A1-20180412-C00082
all salts and prodrugs thereof.
42. The method of claim 39, wherein said compound is compound XII:
Figure US20180099961A1-20180412-C00083
all salts and prodrugs thereof.
43. The method of claim 39, wherein said compound is compound XIII:
Figure US20180099961A1-20180412-C00084
all salts and prodrugs thereof.
44. The method of claim 39, wherein said compound is compound XIV:
Figure US20180099961A1-20180412-C00085
all salts and prodrugs thereof.
US15/839,534 2016-12-13 2017-12-12 Protein kinase inhibitors Abandoned US20180099961A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/839,534 US20180099961A1 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662433410P 2016-12-13 2016-12-13
US201762539785P 2017-08-01 2017-08-01
US15/839,534 US20180099961A1 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors

Publications (1)

Publication Number Publication Date
US20180099961A1 true US20180099961A1 (en) 2018-04-12

Family

ID=61829613

Family Applications (5)

Application Number Title Priority Date Filing Date
US15/839,534 Abandoned US20180099961A1 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors
US15/839,469 Active US10479786B2 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors
US15/839,332 Active US10174018B2 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors
US16/179,174 Active US10556897B2 (en) 2016-12-13 2018-11-02 Protein kinase inhibitors
US16/721,142 Active 2038-04-09 US11236080B2 (en) 2016-12-13 2019-12-19 Protein kinase inhibitors

Family Applications After (4)

Application Number Title Priority Date Filing Date
US15/839,469 Active US10479786B2 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors
US15/839,332 Active US10174018B2 (en) 2016-12-13 2017-12-12 Protein kinase inhibitors
US16/179,174 Active US10556897B2 (en) 2016-12-13 2018-11-02 Protein kinase inhibitors
US16/721,142 Active 2038-04-09 US11236080B2 (en) 2016-12-13 2019-12-19 Protein kinase inhibitors

Country Status (8)

Country Link
US (5) US20180099961A1 (en)
EP (1) EP3555092A4 (en)
JP (1) JP7077335B2 (en)
KR (1) KR102582752B1 (en)
CN (1) CN108473487B (en)
AU (1) AU2017375949B2 (en)
CA (1) CA3044066A1 (en)
WO (1) WO2018111893A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020123277A1 (en) * 2018-12-10 2020-06-18 Princeton Drug Discovery Inc. Protein kinase inhibitor prodrugs
US11236080B2 (en) 2016-12-13 2022-02-01 Princeton Drug Discovery, Inc Protein kinase inhibitors

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102256804B1 (en) * 2019-02-26 2021-05-26 한국화학연구원 N-phenyl-2-(pyrimidin-4-ylamino)thiazol-5-carboxamide derivatives, pharmaceutically acceptable salts thereof and a whitening material composition containing the same as an active ingredient

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040054186A1 (en) * 1999-04-15 2004-03-18 Jagabandhu Das Cyclic protein tyrosine kinase inhibitors
US20090076025A1 (en) * 2007-09-14 2009-03-19 Protia, Llc Deuterium-enriched dasatinib
US20120244116A1 (en) * 2009-10-01 2012-09-27 Csl Limited Method of treatment of philadelphia chromosome positive leukaemia

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TR200102969T2 (en) 1999-04-15 2002-08-21 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors.
HUP0303538A2 (en) 2000-12-21 2005-02-28 Bristol-Myers Squibb Co. Thiazolyl inhibitors of tec family tyrosine kinases and pharmaceutical compositions containing them
US20040209930A1 (en) * 2002-10-02 2004-10-21 Carboni Joan M. Synergistic methods and compositions for treating cancer
TW200501960A (en) 2002-10-02 2005-01-16 Bristol Myers Squibb Co Synergistic kits and compositions for treating cancer
US7491725B2 (en) 2004-02-06 2009-02-17 Bristol-Myers Squibb Company Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
US20060021105A1 (en) * 2004-07-29 2006-02-02 Wilson Nick L Ergonomic hand protection apparatus
PE20061394A1 (en) 2005-03-15 2006-12-15 Bristol Myers Squibb Co METABOLITES OF N- (2-CHLORO-6-METHYLPHENYL) -2 - [[6- [4- (2-HYDROXYETHYL) -1-PIPERAZINYL] -2-METHYL-4-PYRIMIDINYL] AMINO] -5-THIAZOLCARBOXAMIDES
JP5563300B2 (en) * 2006-09-11 2014-07-30 キュリス,インコーポレイテッド Tyrosine kinase inhibitors containing zinc binding moieties
TW200906825A (en) 2007-05-30 2009-02-16 Scripps Research Inst Inhibitors of protein kinases
ES2396366T3 (en) 2007-12-10 2013-02-21 Concert Pharmaceuticals Inc. Heterocyclic Kinase Inhibitors
AP2909A (en) 2009-03-17 2014-05-31 Concert Pharmaceuticals Inc Pyrazinoisoquinoline Compounds
BRPI1010881A2 (en) 2009-06-08 2016-05-31 California Capital Equity Llc triazine derivatives and their therapeutic applications.
CN101891738B (en) 2010-02-08 2011-09-28 南京卡文迪许生物工程技术有限公司 Dasatinib polymorph and preparation method and medical composition thereof
AU2011261185A1 (en) 2010-06-03 2013-01-10 Pharmacyclics, Inc. The use of inhibitors of Bruton's tyrosine kinase (Btk)
EP2872159A2 (en) 2012-07-13 2015-05-20 Concert Pharmaceuticals Inc. Deuterated carfilzomib
EP2970213A4 (en) 2013-03-14 2016-09-07 Concert Pharmaceuticals Inc Deuterated pacritinib
CN104130250B (en) 2013-05-07 2016-06-22 郑州泰基鸿诺药物科技有限公司 Deuterated Dasatinib and its preparation method and application
CN104151321B (en) * 2013-05-15 2016-09-07 复旦大学 N-(2-chloro-6-aminomethyl phenyl)-2 [(2-methylpyrimidine-4-base) amino] thiazole-5-methanamide compound and its production and use
AU2015211021B2 (en) 2014-01-28 2020-07-02 Buck Institute For Research On Aging Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders
US20170224688A1 (en) 2016-02-04 2017-08-10 Acerta Pharma B.V. Methods of Using BTK Inhibitors to Treat Dermatoses
WO2017168454A2 (en) * 2016-04-02 2017-10-05 Sun Pharma Advanced Research Company Limited Novel compounds as btk inhibitors
US20180099961A1 (en) 2016-12-13 2018-04-12 Princeton Drug Discovery Inc Protein kinase inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040054186A1 (en) * 1999-04-15 2004-03-18 Jagabandhu Das Cyclic protein tyrosine kinase inhibitors
US20090076025A1 (en) * 2007-09-14 2009-03-19 Protia, Llc Deuterium-enriched dasatinib
US20120244116A1 (en) * 2009-10-01 2012-09-27 Csl Limited Method of treatment of philadelphia chromosome positive leukaemia

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11236080B2 (en) 2016-12-13 2022-02-01 Princeton Drug Discovery, Inc Protein kinase inhibitors
WO2020123277A1 (en) * 2018-12-10 2020-06-18 Princeton Drug Discovery Inc. Protein kinase inhibitor prodrugs

Also Published As

Publication number Publication date
US20190071437A1 (en) 2019-03-07
EP3555092A4 (en) 2020-05-13
WO2018111893A1 (en) 2018-06-21
KR20190091312A (en) 2019-08-05
JP7077335B2 (en) 2022-05-30
AU2017375949B2 (en) 2021-09-09
US10479786B2 (en) 2019-11-19
US10556897B2 (en) 2020-02-11
US20200123148A1 (en) 2020-04-23
KR102582752B1 (en) 2023-09-22
CN108473487A (en) 2018-08-31
US10174018B2 (en) 2019-01-08
EP3555092A1 (en) 2019-10-23
CN108473487B (en) 2021-08-03
CA3044066A1 (en) 2018-06-21
US11236080B2 (en) 2022-02-01
AU2017375949A1 (en) 2019-05-23
JP2020503376A (en) 2020-01-30
US20180099959A1 (en) 2018-04-12
US20180099960A1 (en) 2018-04-12

Similar Documents

Publication Publication Date Title
US10596174B2 (en) Pyrrolopyrimidine compounds as inhibitors of protein kinases
US11007197B2 (en) EGFR modulators and uses thereof
RU2645672C2 (en) New compounds of pyrrolopyrimidine as inhibitors of protein kinases
US10350210B2 (en) EGFR and ALK dual inhibitor
EP2727918B1 (en) Compounds and Compositions as Protein Kinase Inhibitors
US11236080B2 (en) Protein kinase inhibitors
US10106538B2 (en) Inhibitors of protein kinases
WO2018022911A1 (en) Macrocycle kinase inhibitors
US9586965B2 (en) Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases

Legal Events

Date Code Title Description
AS Assignment

Owner name: PRINCETON DRUG DISCOVERY INC, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HE, KAN;REEL/FRAME:044384/0535

Effective date: 20171208

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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