WO2015081257A2 - Aminopyridine derivatives as tam family kinase inhibitors - Google Patents

Aminopyridine derivatives as tam family kinase inhibitors Download PDF

Info

Publication number
WO2015081257A2
WO2015081257A2 PCT/US2014/067709 US2014067709W WO2015081257A2 WO 2015081257 A2 WO2015081257 A2 WO 2015081257A2 US 2014067709 W US2014067709 W US 2014067709W WO 2015081257 A2 WO2015081257 A2 WO 2015081257A2
Authority
WO
WIPO (PCT)
Prior art keywords
pyridin
pyrazol
compound
oxadiazol
amine
Prior art date
Application number
PCT/US2014/067709
Other languages
French (fr)
Other versions
WO2015081257A3 (en
Inventor
Zaihui Zhang
Original Assignee
Signalchem Lifesciences Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR112016012146-5A priority Critical patent/BR112016012146B1/en
Priority to EP14819178.6A priority patent/EP3074390B1/en
Application filed by Signalchem Lifesciences Corporation filed Critical Signalchem Lifesciences Corporation
Priority to CA2930324A priority patent/CA2930324C/en
Priority to AU2014354711A priority patent/AU2014354711B2/en
Priority to KR1020167016444A priority patent/KR102398473B1/en
Priority to CN201910548630.5A priority patent/CN110156770B/en
Priority to JP2016534735A priority patent/JP6496731B2/en
Priority to US15/039,609 priority patent/US10233176B2/en
Priority to CN201480065032.8A priority patent/CN105764514B/en
Priority to MX2016006815A priority patent/MX2016006815A/en
Publication of WO2015081257A2 publication Critical patent/WO2015081257A2/en
Publication of WO2015081257A3 publication Critical patent/WO2015081257A3/en
Priority to IL245660A priority patent/IL245660B/en
Priority to US16/254,208 priority patent/US10723725B2/en
Priority to US16/899,489 priority patent/US11034685B2/en
Priority to US16/899,494 priority patent/US11034686B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/443Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • the present disclosure provides compounds that inhibit protein kinases Tyro3, Axl and Mer (TAM family kinases), prodrugs of the compounds, intermediates and methods of synthesizing the compounds and/or prodrugs, pharmaceutical compositions comprising the compounds and/or prodrugs and methods of using the compounds and/or prodrugs in a variety of contexts, including, for example, in the treatment and/or prevention of various diseases that are responsive to TAM family kinase inhibition and/or that are mediated, at least in part, by abnormal TAM family kinase activity.
  • TAM family kinases protein kinases Tyro3, Axl and Mer
  • RTKs receptor tyrosine kinases
  • the receptor tyrosine kinases are transmembrane proteins and function as sensors for extracellular ligands, which transduce signals from extracellular medium to the cytoplasm. Their activation leads to the recruitment, phosphorylation, and activation of the downstream signaling pathways, which ultimately regulate cellular functions such as proliferation, growth, differentiation or motility.
  • Abnormal overexpression levels and/or enhanced activities of RTKs have been associated to a variety of human cancers, leading to a strong interest in the development of inhibitors against these human cancers.
  • Tyro-3, Axl, and Mer constitute the TAM family of RTKs characterized by a conserved sequence within the kinase domain and adhesion molecule like extracellular domains.
  • TAM kinases can be activated by the vitamin K-dependent ligand Gas6 and/or Protein S. Strong evidence supports their association with both cancer (gain-in-function) and autoimmunity (loss-of-function). TAM kinase signaling has been implicated in a myriad of cellular responses, many of which are the hallmarks of cancer, including proliferation, survival, migration, invasion and angiogenesis. In addition, TAM plays pivotal roles innate immunity through inhibiting inflammation in macrophages and dendritic cells and promoting the phagocytosis of apoptotic cells.
  • TAM kinases While the oncogenic activity of TAM kinases appears to be mediated via PI3K/AKT pathway, the JAK- STAT pathway is critical for their roles in immune responses. Overexpression of TAM kinases has been observed in over 20 human cancers. The level of their expression was shown to correlate with shorter progression- free and overall survival and their up- regulation has been linked to cancer resistance to cytotoxic drugs and targeted therapies.
  • Tyro3, Axl, and Mer While broadly expressed in various human tumor cell lines, Tyro3, Axl, and Mer exhibit their respective tissue-specific expression patterns. Tyro-3 is highly expressed in the nervous system whereas, Axl is expressed ubiquitously. Higher level of Mer is often found in hematopoietic lineages such as monocytes/macrophages, dendritic cells, NK cells, NKT cells, megakaryocytes, and platelets.
  • Tyro3 is the least studied kinase of the TAM family. Implication of Tyro3 in tumorigenesis was only recently substantiated by recent studies, which revealed Tyro3 is a potential oncogene in melanoma that is linked to poorer outcome of patients suffering from melanoma regardless the BRAF or NRAS status by conferring survival advantage to melanoma cells. It was also identified as one of kinases significantly up-regulated in lung cancer by a phosphoproteomic screen. High level of Tyro3 expression has also been correlated with thyroid cancer.
  • Axl was discovered as a transforming gene in chronic myelogenous leukemia (CML).
  • CML chronic myelogenous leukemia
  • Axl overexpression has since been reported in a wide range of human malignancies and is associated with invasiveness and metastasis in lung, prostate, breast and pancreatic cancer.
  • Axl is also an important regulator of breast cancer metastasis and EMT.
  • Activation of the Axl kinase confers resistance to EGFR targeted therapy in lung cancer. Upregulation of Axl has been implicated as a mechanism of resistance to imatinib in CML and
  • Axl expression has also been associated with chemoresistance in AML, NSCLC and ovarian cancer.
  • Mer is overexpressed/over-activated in a wide variety of cancers and has been established as a therapeutic target in hematopoietic malignancies and solid tumors including leukemia, non-small cell lung cancer, glioblastoma, melanoma, prostate cancer, breast cancer, colon cancer, gastric cancer, pituitary adenomas, and rhabdomyosarcomas.
  • Oncogenic potential of Mer is mediated through the activation of several canonical oncogenic signaling pathways including the mitogen-activated protein kinase and phosphoinositide 3 -kinase pathways, as well as regulation of signal transducer and activator of transcription family members, migration-associated proteins including the focal adhesion kinase and myosin light chain 2, and prosurvival proteins such as survivin and Bcl-2.
  • these signaling events result in functional phenotypes such as decreased apoptosis, increased migration,
  • the present disclosure provides 2-aminopyridine derivatives that are capable of inhibiting the activity of TAM family kinases. Methods of using such derivatives to inhibit the activity of TAM family kinases and pharmaceutical compositions comprising such derivatives are also disclosed.
  • One embodiment provides compounds of formula (I):
  • L is heteroaryl, heterocyclyl, -N(R 6 )-, -0-, -C(O)-, -C(0)0-, -S(O),- where t is 0, 1, or 2, -CON(R 6 )-, -N(R 6 )CO-, -S0 2 N(R 6 )-, or -N(R 6 )CON(R 6 )-, provided that when L is heteroaryl, L is not pyridinyl, pyrazinyl or thienyl;
  • R 1 is alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or heteroarylalkyl;
  • R 2 is hydrogen, heterocyclyl, heterocyclylalkyl, cycloalkyl, alkyl, aralkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, or aryl;
  • each R 3 and R 4 is the same or different and independently hydrogen, alkyl, aralkyl, alkenyl, aralkenyl, alkynyl, aralkynyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl;
  • R 5 at each occurrence, is hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl;
  • R 6 is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, or haloalkyl, a stereoisomer, enantiomer or tautomer thereof, an isotopically enriched derivative thereof, a pharmaceutically acceptable salt thereof, a pharmaceutical composition thereof or a prodrug thereof.
  • A, A 1 , A 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • A, A 1 , A 2 , R 3 , R 4 , and R 5 are as defined above,
  • L is heteroaryl or heterocycle
  • R 7 is hydrogen, alkyl, alkoxy, or halo
  • X is -0-, -NR 8 -, or -C(R 9 ) 2 -;
  • R 8 is hydrogen, alkyl, or -C(0)R 10 -;
  • each R 9 is independently hydrogen, alkyl, alkoxy, or halo
  • R 10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
  • A, A 1 , A 2 , R 1 , R 3 , R 4 , R 7 , and R 8 are as defined above.
  • A, A 1 , A 2 , R 3 , R 4 , and R 7 are as defined above.
  • A, A 1 , A 2 , R 1 , R 3 , R 4 , R 7 , and R 9 are as defined above.
  • Yet another embodiment provides a pharmaceutical composition of a compound of any one of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3), or any one the specific compounds disclosed herein.
  • Yet another embodiment provides a method of treating leukemia and lymphoma comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3)or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating endometriosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating restenosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Atherosclerosis/thrombosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating psoriasis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating age-related macular degeneration or diabetic retinopathy comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating retinopathy of prematurity comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating renal transplant rejection comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating rheumatoid arthritis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating osteoarthritis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating osteoporosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of treating cataracts comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
  • Yet another embodiment provides a method of preventing/treating tumor metastasis or treating metastasized tumors comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a
  • composition comprising the same.
  • Amino refers to the -NH 2 radical.
  • Metal refers to the -OCH 3 radical.
  • Niro refers to the -N0 2 radical.
  • Trifluoromethyl refers to the -CF 3 radical.
  • Acyl refers to -C(0)R radical, wherein R is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl;
  • Alkyl refers to a straight or branched hydrocarbon chain radical, when unsubstituted, consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms, preferably one to eight carbon atoms or one to six carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (z ' so-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted by one or more
  • alkenyl refers to a straight or branched hydrocarbon chain radical group, when unsubstituted, consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g. , ethenyl, prop- 1 -enyl, but- 1 -enyl, pent- 1 -enyl, penta- 1 ,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group may be optionally substituted by one of or more substituents, as defined herein.
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group, when unsubstituted, consisting solely of carbon and hydrogen atoms, containing at least one triple bond, optionally containing at least one double bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group may be optionally substituted by one or more substituents, as defined herein.
  • Alkylene and “alkylene chain” refer to a straight or branched divalent hydrocarbon chain, linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, preferably having from one to eight carbons, e.g., methylene, ethylene, propylene, n-butylene, and the like.
  • the alkylene chain may be attached to the rest of the molecule and to the radical group through one carbon within the chain or through any two carbons within the chain.
  • alkenylene and alkenylene chain refer to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one double bond and having from two to twelve carbon atoms, e.g. , ethenylene, propenylene, n-butenylene, and the like.
  • the alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond.
  • the points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • Alkoxy refers to a radical of the formula -OR a where R a is an alkyl radical as defined above.
  • R a is an alkyl radical as defined above.
  • the alkyl part of the alkoxy radical may be optionally substituted as defined above for an alkyl radical.
  • Aryl refers to aromatic monocyclic or multi-cyclic hydrocarbon ring system, when unsubstituted, consisting only of hydrogen and carbon and containing from 6 to 19 carbon atoms, preferably 6 to 10 carbon atoms, where the ring system may be partially or fully saturated.
  • Aryl groups include, but are not limited to groups such as fluorenyl, phenyl and naphthyl. Unless stated otherwise specifically in the specification, the term “aryl” or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl radicals optionally substituted by one or more substituents, as defined herein.
  • Alkyl refers to a radical of the formula -R a R b where Ra is an alkyl radical as defined above and R b is one or more aryl radicals as defined above, e.g., benzyl, diphenylmethyl and the like.
  • the aryl part of the aralkyl radical may be optionally substituted as described above for an aryl group.
  • the alkyl part of the aralkyl radical may be optionally substituted as defined above for an alkyl group.
  • Alkenyl refers to a radical of the formula -R c R b where R c is an alkenyl radical as defined above and R b is one or more aryl radicals as defined above, which may be optionally substituted as described above.
  • the aryl part of the aralkenyl radical may be optionally substituted as described above for an aryl group.
  • the alkenyl part of the aralkenyl radical may be optionally substituted as defined above for an alkenyl group.
  • Alkynyl refers to a radical of the formula -R R b where R d is an alkynyl radical as defined above and R b is one or more aryl radicals as defined above.
  • the aryl part of the aralkynyl radical may be optionally substituted as described above for an aryl group.
  • the alkynyl part of the aralkynyl radical may be optionally substituted as defined above for an alkynyl group.
  • Cycloalkyl refers to a stable non-aromatic monocyclic or bicyclic hydrocarbon radical, when unsubstituted, consisting solely of carbon and hydrogen atoms, having from three to fifteen carbon atoms, preferably having from three to twelve carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond, e.g. , cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, decalinyl and the like.
  • cycloalkyl is meant to include cycloalkyl radicals which are optionally substituted by one or more substituents, as defined herein.
  • Cycloalkylalkyl refers to a radical of the formula -R a R d where R a is an alkyl radical as defined above and Rj is a cycloalkyl radical as defined above.
  • the cycloalkyl part of the cycloalkyl radical may be optionally substituted as defined above for a cycloalkyl radical.
  • the alkyl part of the cycloalkyl radical may be optionally substituted as defined above for an alkyl radical.
  • Halo refers to bromo, chloro, fluoro or iodo.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above. One or more carbons of the alkyl radical may be substituted by the one or more halo radicals.
  • haloalkyl include, trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1 -fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoro-propyl, 1 -bromomethyl-2-bromoethyl, and the like.
  • the alkyl part of the haloalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Haloalkenyl refers to an alkenyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., 2-bromoethenyl, 3-bromoprop-l-enyl, and the like.
  • the alkenyl part of the haloalkenyl radical may be optionally substituted as defined above for an alkyl group.
  • Heterocyclyl refers to a stable 3- to 18-membered non-aromatic ring radical including, as ring atoms, at least one carbon atom and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; and the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated.
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl,
  • heterocyclyl is meant to include heterocyclyl radicals as defined above which are optionally substituted by one or more substituents, as defined herein.
  • the heterocyclyl When the heterocyclyl is a substituent of another moiety, the heterocyclyl is monovalent, which means that the heterocyclyl is connected to the other moiety by a single ring atom.
  • An example of a monovalent heterocyclyl can be found in the radical of heterocyclylalkyl, in which a heterocyclyl group is a substituent of an alkyl group.
  • the heterocyclyl is a linker moiety (L) of Formula (I)
  • the heterocyclyl is a divalent radical.
  • the heterocyclyl (as L) is linked, by two of its ring atoms, to the 2-aminopyridine moiety and R 2 of Formula (I).
  • An example of a divalent heterocycl -piperidinyl shown below:
  • Heterocyclylalkyl refers to a radical of the formula -R a Rg where R a is an alkyl radical as defined above and R e is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom.
  • R a is an alkyl radical as defined above
  • R e is a heterocyclyl radical as defined above
  • heterocyclylalkyl radical may be optionally substituted as defined above for an alkyl group.
  • the heterocyclyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for a heterocyclyl group.
  • Heteroaryl refers to a 5- to 18-membered aromatic ring radical including, as ring atoms, at least one carbon atom and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; and the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzothiadiazolyl,
  • benzonaphthofuranyl benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl,
  • heteroaryl is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents, as defined herein.
  • Heteroaryl, as defined herein may be monovalent or divalent.
  • the heteroaryl is monovalent, which means that the heteroaryl is connected to the other moiety by a single ring atom.
  • An example of a monovalent heteroaryl can be found in the radical of heteroarylalkyl, in which an alkyl group is substituted with a heteroaryl group.
  • heteroaryl is a linker moiety (L) of Formula (I)
  • the heteroaryl is a divalent radical. In this occurrence, the heteroaryl (as L) is linked, by two of its ring atoms, to the
  • 2-aminopyridine moiety and R 2 of Formula (I).
  • R 2 of Formula (I) An example of a divalent heteroaryl is lH-pyrazol-4-yl, shown below:
  • Heteroarylalkyl refers to a radical of the formula -R a R f where R a is an alkyl radical as defined above and R f is a heteroaryl radical as defined above.
  • the heteroaryl part of the heteroarylalkyl radical may be optionally substituted as defined above for a heteroaryl group.
  • the alkyl part of the heteroarylalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Heteroarylalkenyl refers to a radical of the formula -R c R g where R c is an alkenyl radical as defined above and R g is a heteroaryl radical as defined above.
  • the heteroaryl part of the heteroarylalkenyl radical may be optionally substituted as defined above for a heteroaryl group.
  • the alkenyl part of the heteroarylalkenyl radical may be optionally substituted as defined above for an alkenyl group.
  • N-heteroaryl is a subset of heteroaryl, and refers to a heteroaryl having at least one nitrogen ring atom. Heteroaryl is otherwise as defined as herein. Examples of N-heteroaryls include, without limitation, benzimidazolyl, benzindolyl,
  • benzotriazolyl benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, imidazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, thi
  • N-heterocyclyl is a subset of heterocyclyl, and refers to a heterocyclyl having at least one nitrogen ring atom. Heterocyclyl is otherwise as defined as herein. Examples of N- heterocyclyls include, without limitation, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl,
  • Substituent refers to a radical (a single non-hydrogen atom or a functional group) that is or can be bonded to another molecule.
  • An substituent is therefore any one of the following radicals: alkyl, alkenyl, amino, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR 14 , -OC(0)R 14 , - N(R 14 ) 2 , -C(0)R 14 , -C(0)OR 14 , -C(0)N(R 14 ) 2 , -N(R 14 )C(0)OR 16 , -N(R 14 )C(0)R 16 , -N(R 14 )C(0)R 16 , -N(R 14
  • each R 16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, and where each of the above substituents is unsubstituted unless otherwise indicated.
  • Prodrugs refers to a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of Formula (I) or any one of the substructures.
  • prodrug refers to a metabolic precursor of a compound of Formula (I) or any one of the substructures, that is pharmaceutically acceptable; the latter is also referred to as a "parent compound.”
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound, i.e., the parent compound.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24, Elsevier, Amsterdam).
  • prodrugs are provided in Higuchi, ⁇ ., ⁇ ⁇ , "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible
  • prodrug is also meant to include any covalently bonded carriers which release the active compound of the disclosure in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound of the disclosure may be prepared by modifying functional groups present in the compound of Formula (I) or any one of the substructures in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • Prodrugs include compounds of the disclosure wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug disclosure is administered to a mammalian subject, cleaves to restore the free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate, and phosphate derivatives of alcohol or amine functional groups in the compounds of Formula (I), (IA), (IB) or any one of the substructures.
  • Solid compound and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Mammal” or “mammalian subject” includes humans and domestic animals, such as cats, dogs, swine, cattle, sheep, goats, horses, rabbits, and the like.
  • Optional or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid,
  • benzenesulfonic acid benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid,
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine,
  • diethanolamine diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine,
  • N-ethylpiperidine polyamine resins and the like.
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • solvate refers to an aggregate that comprises one or more molecules of a compound of the disclosure with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • the compounds of the present disclosure may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms.
  • the compound of the disclosure may be true solvates, while in other cases, the compound of the disclosure may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
  • a “pharmaceutical composition” refers to a formulation of a compound of the disclosure and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans.
  • a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
  • “Therapeutically effective amount” refers to that amount of a compound of the disclosure which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a disease or condition in the mammal, preferably a human.
  • the amount of a compound of the disclosure which constitutes a “therapeutically effective amount” will vary depending on the compound, the condition and its severity, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
  • “Treating” or “treatment” as used herein covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or disorder of interest, and includes:
  • the terms “disease” and “condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
  • the compounds of the disclosure, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to
  • Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as HPLC using a chiral column.
  • a “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are non-superimposeable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any said compounds.
  • isotopically enriched derivative refers to a compound wherein one or more atoms are replaced by atoms having the same atomic number but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes suitable for inclusion in the compounds of the disclosure comprises isotopes of hydrogen, such as 2 H and 3 H, carbon, such as U C, 13 C and 14 C,
  • chlorine such as CI
  • fluorine such as F
  • iodine such as I and I
  • nitrogen such as 13 N and 15 N
  • oxygen such as 15 0, 17 0 and 18
  • phosphorus such as 31 P, 32 P and 33 P
  • sulphur such as 35 S.
  • Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances.
  • Isotopically-enriched compounds of the disclosure can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations Sections using an appropriate isotopically-enriched reagent in place of the non-enriched reagent previously employed.
  • each dash bond may, but not necessarily, indicate the presence of a double bond.
  • A is defined as -O -, A is connected to the two adjacent carbon atoms by single bonds, respectively.
  • L is lH-pyrazol-4-yl
  • R 1 is 4-chlorophenyl
  • R 2 is l-(piperidin-4-yl)
  • R 3 , R 4 and R 5 are each hydrogen, i.e., a compound of the following formula: is named herein as: 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine.
  • linker L which is a divalent radical
  • L may also have two possible ways to connect the 2-aminopyridine and the R 2 group.
  • L is an amido linker, i.e., -CON(R 6 )-
  • a compound of Formula (I) may have the carbonyl portion directly connected to the 2-aminopyridine moiety.
  • Another compound of Formula (I) may have the carbonyl portion directly connected to the R 2 group.
  • L is lH-pyrazol-4-yl
  • R 1 is 2,6-dichlorophenyl
  • R 2 is l-(piperidin-4-yl)
  • R 3 and R 4 are each hydrogen, i.e., a compound of the following formula:
  • aminopyridine derivatives useful as TAM family kinase inhibitors are provided herein.
  • One embodiment provides a compound of formula (I):
  • L is a heteroaryl, heterocyclyl, -N(R 6 )-, -0-, -C(O)-, -C(0)0-, -S(O),- where t is 0, 1 , or 2, -CON(R 6 )-, -N(R 6 )CO-, -S0 2 N(R 6 )-, or -N(R 6 )CON(R 6 )-; provided that when L is heteroaryl, L is not pyridinyl, pyrazinyl or thienyl;
  • R 1 is alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or heteroarylalkyl;
  • R 2 is hydrogen, heterocyclyl, heterocyclylalkyl, cycloalkyl, alkyl, aralkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, or aryl; each R 3 and R 4 is the same or different and independently selected from hydrogen, alkyl, aralkyl, alkenyl, aralkenyl, alkynyl, aralkynyl, cycloalkyl,
  • R 5 at each occurrence, is hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl;
  • R 6 is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, or haloalkyl, a stereoisomer, enantiomer or tautomer thereof, an isotopically enriched derivative thereof, a pharmaceutically acceptable salt thereof, a pharmaceutical composition thereof or a prodrug thereof.
  • L is heteroaryl
  • L is N-heteroaryl
  • L is divalent pyrrozolyl moiety.
  • L is lH-pyrazol-4-yl, and the compound of Formula (I) is represented by Formula (IA):
  • A, A 1 , A 2 , R 3 , and R 4 are as defined above.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl. In other embodiments of Formula (IA1), R 2 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
  • R 2 is heterocyclyl
  • R 2 is N-heterocyclyl. More specifically, R 2 is piperidinyl or substituted piperidinyl.
  • R 2 is
  • R 2 is cyclohexyl
  • each of R 3 and R 4 is hydrogen, R 1 is aryl, R 2 is heterocyclyl.
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl or substituted phenyl, R 2 is heterocyclyl.
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl or substituted phenyl, R 2 is piperidinyl or substituted piperidinyl.
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl (including phenyl substituted with one or more halo), and R 2 is heterocyclyl.
  • R 2 is a l-piperidin-4-yl (including l-piperidin-4-yl substituted at the 1 -position with alkyl or acyl).
  • the compound of Formula (IA1) is: tert-butyl-4-(4-(6-amino-5-(5-phenyloxazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine- 1 -carboxylate;
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl or substituted phenyl, R 2 is tetrahydropyranyl.
  • the compound of Formula (IA1) is: 3-(5-phenyloxazol-2-yl)-5-(l-(tetrahydro-2pyran-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine; or
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl or substituted phenyl, R 2 is cyclohexyl.
  • the compound of Formula (IA1) is: 3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-cyclohexyl-lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-cyclohexyl-lH-pyrazol-4-yl)pyridin-2-amine; or
  • each of R 3 and R 4 is hydrogen, R 1 is heteroaryl
  • R 2 is heterocyclyl
  • the compound is 5-(l-(piperidin-4-yl)- lH-pyrazol-4-yl)-3-(5-(pyridin-3-yl)oxazol-2-yl)pyridin-2-amine.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 2 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R 2 is heterocyclyl. More specifically, R 2 is N-heterocyclyl.
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl or substituted phenyl, R 2 is piperidinyl or substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
  • the compound of Formula (IA2) is: 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenylisoxazol-3-yl)pyridin-2-amine;
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 2 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R 2 is heterocyclyl. More specifically, R 2 is N-heterocyclyl.
  • each of R 3 and R 4 is hydrogen, R 1 is phenyl or substituted phenyl, R 2 is piperidinyl or substituted
  • the compound of Formula (IA3) is: 3-(5-phenylfuran-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine;
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 is aryl, heteroaryl, cycloalkyl, heterocyclyl, aralkyl, or cycloalkylalkyl.
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl. In another more specific embodiment of Formula (IA4), R 1 is heteroaryl. More specifically, R 1 is a pyridinyl, substituted pyridinyl, pyrazinyl, substituted pyrazinyl, thiazolyl, substituted thiazolyl.
  • R 1 is aralkyl. More specifically, R 1 is benzyl.
  • R 1 is cycloalkylalkyl. More specifically, R 1 is cycloalkylalkyl.
  • R 2 is hydrogen, aryl, heteroaryl, cycloalkyl or heterocyclyl.
  • R 2 is heterocyclyl
  • R 2 is N-heterocyclyl.
  • R 2 is piperidinyl or substituted piperidinyl.
  • R 2 is tetr ahy dropyr any 1.
  • R 2 is hydrogen.
  • each of R 3 and R 4 is hydrogen, R 1 is aryl, R 2 is heterocyclyl, including N-heterocyclyl.
  • each of R 3 and R 4 is hydrogen
  • R 1 is phenyl or phenyl substituted with one or more halo or alkyl
  • R 2 is piperidinyl or substituted piperidinyl.
  • a compound of Formula (IA4) is:
  • tert-butyl 4-(4-(6-amino-5-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate; 3-(5-(2,5-difluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
  • each of R 3 and R 4 is hydrogen, R 1 is heteroaryl or substituted heteroaryl, R 2 is piperidinyl or substituted piperidinyl.
  • the compound of Formula (IA4) is: tert-butyl 4-(4-(6-amino-5-(5-(pyrazin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate; tert-butyl 4-(4-(6-amino-5-(5-(pyridin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
  • each of R 3 and R 4 is hydrogen, R 1 is cycloalkylalkyl or aralkyl, R 2 is piperidinyl or substituted piperidinyl.
  • the compound of Formula (IA4) is: tert-butyl 4-(4-(6-amino-5-(5-(cyclopropylmethyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate;
  • each of R 3 and R 4 is hydrogen, R 1 is aryl or more specifically phenyl or substituted phenyl, R 2 is hydrogen.
  • the compound of Formula (IA4) is: 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine;
  • each of R 3 and R 4 is hydrogen, R 1 is aryl or more specifically, phenyl or substituted phenyl, R 2 is cycloalkyl.
  • the compound of Formula (IA4) is:
  • each of R 3 and R 4 is hydrogen, R 1 is aryl or more specifically, phenyl or substituted phenyl, R 2 is tetrahydropyranyl.
  • the compound of Formula (IA4) is:
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 2 is heterocyclyl. More specifically, R 2 is N-heterocyclyl.
  • R 2 is piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
  • R 2 is hydrogen.
  • each of R 3 and R 4 is hydrogen, R 1 is aryl, R 2 is heterocyclyl, including N-heterocyclyl.
  • each of R 3 and R 4 is hydrogen
  • R 1 is aryl or more specifically, phenyl or substituted phenyl
  • R 2 is hydrogen, piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
  • each of R 3 and R 4 is hydrogen
  • R 1 is aryl or more specifically, phenyl or substituted phenyl
  • R 2 is piperidinyl, substituted piperidinyl.
  • the compound of Formula (IA5) is:
  • each of R 3 and R 4 is hydrogen
  • R 1 is aryl or more specifically, phenyl or substituted phenyl
  • R 2 is hydrogen, tetrahydropyranyl or cyclohexyl.
  • the compound of Formula (IA5) is:
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 2 is heterocyclyl
  • R 2 is N-heterocyclyl.
  • R 2 is piperidinyl or substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
  • R 2 is hydrogen.
  • each of R 3 and R 4 is hydrogen, R 1 is aryl, R 2 is heterocyclyl, including N-heterocyclyl.
  • each of R 3 and R 4 is hydrogen
  • R 1 is aryl or more specifically, phenyl or substituted phenyl
  • R 2 is hydrogen, piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
  • the compound of Formula (IA6) is:
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 2 is heterocyclyl. More specifically, R 2 is piperidinyl or substituted piperidinyl.
  • the compound of Formula (IA7) is:
  • A, A 1 , A 2 , R 3 , R 4 , and R 5 are as defined above,
  • L is heteroaryl or heterocycle
  • R is hydrogen, alkyl, alkoxy, or halo
  • X is -0-, -NR 8 -, or -C(R 9 ) 2 -;
  • R 8 is hydrogen, alkyl, or -C(0)R 10 -;
  • each R 9 is independently hydrogen, alkyl, alkoxy, or halo
  • R 10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
  • R 1 is aryl, heteroaryl, cycloalkyl, heterocyclyl, aralkyl, or cycloalkylalkyl;
  • each of R 3 , R 4 and R 7 is hydrogen
  • R 8 is hydrogen, alkyl, or -C(0)R 10 -;
  • R 10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 1 is heteroaryl. More specifically, R 1 is a pyridinyl, substituted pyridinyl, pyrazinyl, substituted pyrazinyl, thiazolyl, substituted thiazolyl.
  • R 1 is aralkyl. More specifically, R 1 is benzyl.
  • R 1 is cycloalkylalkyl. More specifically, R 1 is cyclopropylalkyl.
  • R 1 is aryl, heteroaryl, cycloalkyl, or heterocyclyl
  • each of R 3 , R 4 and R 7 is hydrogen.
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • R 1 is aryl, heteroaryl, cycloalkyl, or heterocyclyl
  • each of R 3 , R 4 , R 7 and R 9 is hydrogen
  • R 1 is aryl. More specifically, R 1 is a phenyl or substituted phenyl.
  • the present disclosure relates to compounds, pharmaceutical compositions and methods of using the compounds and pharmaceutical compositions for the treatment and/or prevention of diseases and conditions mediated by the kinase activity of Tyro3, Axl or Mer individually or by any combination of them, preferably diseases and conditions related to characterized by angiogenesis and/or cell proliferation and migration, and especially a disease and condition related to cancer, inflammatory diseases, autoimmune diseases, neurodisorders, and the like, by administering an effective amount of a compound of the disclosure.
  • the compounds of the disclosure modulate, preferably inhibit, the activity of human Tyro3, Axl or Mer individually or by any combination of them.
  • the general value of the compounds of the disclosure in modulating, especially inhibiting, the activity of Tyro3, Axl or Mer individually or by any combination of them can be determined using the assay described below in Example 21.
  • the compounds of the instant disclosure are inhibitors of Tyro3, Axl or Mer individually or inhibitors of any combination of them and are useful for treating diseases and disorders in humans and other organisms, including all those human diseases and disorders which are the result of abnormal kinase activity of Tyro3, Axl or Mer individually or any combination of them or which may be ameliorated by modulation of the kinase activity of Tyro3, Axl or Mer individually or any combination of them.
  • a disease or condition mediated by the abnormal kinase activity of Tyro3, Axl or Mer individually or any combination of them is defined as any disease or condition in which the activity of Tyro3, Axl or Mer individually or any combination of them is elevated and/or where inhibition of the activity of Tyro3, Axl or Mer individually or any combination of them can be demonstrated to bring about symptomatic improvements for the individual so treated.
  • a disease or condition mediated by the abnormal activity of Tyro3, Axl or Mer individually or any combination of them includes, but is not limited to, a disease or condition which is, or is related to cancer, inflammatory diseases, autoimmune diseases, and neurodisorders.
  • Diseases and conditions which are alleviated by the modulation of the activity of Tyro3, Axl or Mer individually or any combination of them include, but are not limited to, solid tumors, including, but not limited to, breast, renal, endometrial, ovarian, thyroid, and non-small cell lung carcinoma, melanoma, prostate carcinoma, sarcoma, gastric cancer and uveal melanoma; liquid tumors, including but not limited to, leukemias particularly myeloid leukemias and lymphomas; endometriosis, vascular disease/injury including, but not limited to, restenosis, atherosclerosis and thrombosis, psoriasis; visual impairment due to macular
  • kidney disease including, but not limited to, glomerulonephritis, diabetic nephropathy and renal transplant rejection, rheumatoid arthritis; osteoarthritis, osteoporosis and cataracts.
  • the compounds of the disclosure are useful in treating diseases and conditions which are affected by the following biological processes: Invasion, migration, metastasis, or drug resistance as manifested in cancer; stem cell biology as manifested in cancer; invasion, migration, adhesion, or
  • angiogenesis as manifested in endometriosis
  • vascular remodeling as manifested in cardiovascular disease, hypertension or vascular injury
  • bone homeostatasis as manifested in osteoporosis or osteoarthritis
  • viral infection as manifested, for example, in ebola virus infection; or differentiation as manifested in obesity.
  • the compounds of the disclosure may also be used to modulate inflammatory processes by treating sepsis, acting as vaccine adjuvants, and/or potentiating the immune response in immunocompromised patients.
  • the compounds of the disclosure may be used or tested for their use in treating leukemias and lymphomas by, respectively, administering a pharmaceutically effective amount or testing the compounds in the xenograft in SCID mouse model using human cancer cell lines which express Tyro3 or Axl or Mer or co-expressing any combination of these three kinases including, but not limited to, A549, K562, HeLa, MDA-MB-231, SK-OV-3, OVCAR-8, DU145, H1299, ACHN, A498 and Caki-1.
  • the compounds of the disclosure may be tested for their use in treating leukemias in the xenograft in SCID or nu/nu mouse model using human AML and CML leukemia cell lines.
  • the compounds of the disclosure may be used or tested for their use in treating endometriosis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the syngenic mouse model of
  • endometriosis see Somigliana, E. et al., "Endometrial ability to implant in ectopic sites can be prevented by interleukin-12 in a murine model of endometriosis", Hum. Reprod. 1999, 14(12), 2944-2950).
  • the compounds may also be tested for their use in treating endometriosis by using the rat model of endometriosis (see Lebovic, D.I. et al, "Peroxisome proliferator-activated receptor-gamma induces regression of endometrial explants in a rat model of endometriosis", Fertil. SteriL, 2004, 82 Suppl 3, 1008-1013).
  • the compounds of the disclosure may be used or tested for their use in treating restenosis by, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or using the balloon-injured rate carotid artery model (see Kim, D.W. et al., "Novel oral formulation of paclitaxel inhibits neointimal hyperplasia in a rat carotid artery injury model", Circulation, 2004, 109(12), 1558- 1563).
  • the compounds of the disclosure may also be tested for their use in treating restenosis by using the percutaneous transluminal coronary angioplasty in apoE deficient mouse model (see von der Thusen, J. H.
  • the compounds of the disclosure may be used or tested for their use in treating atherosclerosis/thrombosis, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or in the ApoE deficient mouse model (see Nakashima, Y. et al., "ApoE-deficient mice develop lesions of all phases of
  • the compounds of the disclosure may also be used or tested for their use in treating thrombosis, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or using the collagen-epinephrine-induced pulmonary thromboembolism model and the stasis induced venous thrombosis model (see Angelillo-Scherrer A. et al, "Role of Gas6 receptors in platelet signaling during thrombus stabilization and implications for antithrombotic therapy", J. Clin. Invest., 2005,115, 237-246).
  • the compounds of the disclosure may be used or tested for their use in treating psoriasis by, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or using the SCID mouse model or the human skin model of psoriasis (see Nickoloff, B.J. et al, "Severe combined immunodeficiency mouse and human psoriatic skin chimeras. Validation of a new animal model", Am. J. Pathol, 1995, 146(3), 580-588).
  • the compounds of the disclosure may be used or tested for their use in treating age- related macular degeneration or diabetic retinopathy by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the rat corneal angiogenesis model (see Sarayba MA, Li L, Tungsiripat T, Liu NH, Sweet PM, Patel AJ, Osann KE, Chittiboyina A, Benson SC, Pershadsingh HA, Chuck RS. Inhibition of corneal neovascularization by a peroxisome proliferator-activated receptor-gamma ligand. Exp. Eye.
  • the compounds of the disclosure may be used or tested for their use in treating retinopathy of prematurity, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or in the mouse retinopathy of prematurity model (see Smith, L.E. et al, "Oxygen-induced retinopathy in the mouse", Invest. Ophthalmol. Vis. Sci., 1994, 35(1), 101-111).
  • the compounds of the disclosure may be tested for their use in treating glomerulonephritis or diabetic nephropathy in the rat anti- Thyl .1 -induced experimental mesengial proliferative glomerulonephritis model (see Smith, L. E. et al. cited above).
  • the compounds of the disclosure may be used or tested for their use in treating renal transplant rejection, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or by using a rat model of chronic renal transplant rejection (see Yin, J. L. et al, "Expression of growth arrest-specific gene 6 and its receptors in a rat model of chronic renal transplant rejection", Transplantation, 2002, 73(4), 657-660).
  • the compounds of the disclosure may be used or tested for their use in treating rheumatoid arthritis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the CAIA mouse model (see Phadke, K. et al., "Evaluation of the effects of various anti-arthritic drugs on type II collagen-induced mouse arthritis model", lmmunopharmacology, 1985, 10(1), 51-60).
  • the compounds of the disclosure may be used or tested for their use in treating osteoarthritis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the STR/ORT mouse model (see Brewster, M. et al., "Ro 32-3555, an orally active collagenase selective inhibitor, prevents structural damage in the STR/ORT mouse model of osteoarthritis", Arthritis. Rheum., 1998, 41(9), 1639-1644).
  • the compounds of the disclosure may be used or tested for their use in treating osteoporosis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the ovariectomized rat model (see Wronski, TJ.
  • the compounds of the disclosure may be used or tested for their use in treating cataracts by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the H 2 0 2 -induced model (see Kadoya, K. et al, "Role of calpain in hydrogen peroxide induced cataract", Curr. Eye Res., 1993, 12(4), 341- 346) or the Emory mouse model (see Sheets, N. L. et al, "Cataract- and lens-specific up-regulation of ARK receptor tyrosine kinase in Emory mouse cataract", Invest.
  • H 2 0 2 -induced model see Kadoya, K. et al, "Role of calpain in hydrogen peroxide induced cataract", Curr. Eye Res., 1993, 12(4), 341- 346
  • Emory mouse model see Sheets, N. L. et al, "Cataract- and lens-specific up-regulation of ARK receptor ty
  • a successful inhibitory therapeutic agent of the activity of Tyro3, Axl or Mer individually or any combination of them will meet some or all of the following criteria.
  • Oral availability should be at or above 20%
  • Animal model efficacy is less than about 20 mg/Kg, 2 mg/Kg, 1 mg/Kg, or 0.5 mg/Kg and the target human dose is between 10 and 250 mg/70 Kg, although doses outside of this range may be acceptable.
  • mg/Kg means milligrams of compound per kilogram of body mass of the subject to whom it is being administered).
  • the required dosage should preferably be no more than about once or twice a day or at meal times.
  • the therapeutic index (or ratio of toxic dose to therapeutic dose) should be greater than 10.
  • the IC 50 (“Inhibitory
  • Concentration - 50% is a measure of the amount of compound required to achieve 50% inhibition of the kinase activity, over a specific time period, in a kinase activity assay.
  • Any process for measuring the kinase activity of Tyro3, Axl or Mer, preferably human Tyro3, Axl or Mer, may be utilized to assay the activity of the compounds useful in the methods of the disclosure in inhibiting said Tyro3, Axl or Mer activity.
  • Compounds of the disclosure demonstrate an IC 50 in a 15 to 60 minute recombinant human kinase assay of preferably less than 10 mM, less than 5 ⁇ , less than 2.5 ⁇ , less than 1 ⁇ .
  • Compounds of the disclosure may show reversible inhibition (i.e. competitive inhibition) or irreversible inhibition and preferably do not inhibit other protein kinases.
  • compounds of the disclosure had greater than 50% inhibitory activity at 10 ⁇ concentration of the test compound, preferably greater than 60% inhibitory activity at 10 ⁇ concentration of the test compound, more preferably greater than 70% inhibitory activity at 10 ⁇ concentration of the test compound, and even more preferably greater than 80% inhibitory activity at 10 ⁇ concentration of the test compound, and the most preferably greater than 90% inhibitory activity at 10 ⁇ concentration of the test compound, thereby demonstrating that the compounds of the disclosure are potent inhibitors of the kinase activity of Tyro3, Axl and Mer.
  • SAR structure-activity relationship
  • the determination of the ability of a compound to inhibit Tyro3, Axl and Mer activity may be accomplished in vivo. In one such embodiment this is accomplished by administering said chemical agent to an animal afflicted with a certain tumor graft model and subsequently detecting a change in tumor growth rate in said animal thereby identifying a therapeutic agent useful in treating the said tumors.
  • the animal may be a human, such as a human patient afflicted with such a disorder and in need of treatment of said disorder.
  • said change in Tyro3, Axl or Mer activity in said animal is a decrease in activity, preferably wherein said
  • Tyro3, Axl or Mer inhibiting agent does not substantially inhibit the biological activity of other kinases.
  • the compounds of the disclosure can be used in combination with other therapeutic agents.
  • alkylating agents that can be carried out in combination with include, but not limited to, fluorouracil (5-FU) alone or in further combination with leukovorin; other pyrimidine analogs such as UFT, capecitabine, gemcitabine and cytarabine, the alkyl sulfonates, e.g., busulfan (used in the treatment of chronic granulocytic leukemia), improsulfan and piposulfan; aziridines, e.g., benzodepa, carboquone, meturedepa and uredepa; ethyleneimines and methylmelamines, e.g., altretamine, triethylenemelamine, triethylenephosphoramide,
  • triethylenethiophosphoramide and trimethylolmelamine and the nitrogen mustards, e.g., chlorambucil (used in the treatment of chronic lymphocytic leukemia, primary macroglobulinemia and non-Hodgkin's lymphoma), cyclophosphamide (used in the treatment of Hodgkin's disease, multiple myeloma, neuroblastoma, breast cancer, ovarian cancer, lung cancer, Wilm' s tumor and rhabdomyosarcoma), estramustine, ifosfamide, novembrichin, prednimustine and uracil mustard (used in the treatment of primary thrombocytosis, non-Hodgkin's lymphoma, Hodgkin's disease and ovarian cancer); and triazines, e.g., dacarbazine (used in the treatment of soft tissue sarcoma).
  • nitrogen mustards e.g., chlorambucil (used in the
  • antimetabolite chemotherapeutic agents examples include, but not limited to, folic acid analogs, e.g.,
  • methotrexate used in the treatment of acute lymphocytic leukemia, choriocarcinoma, mycosis fungiodes, breast cancer, head and neck cancer and osteogenic sarcoma
  • pteropterin used in the treatment of acute lymphocytic leukemia, choriocarcinoma, mycosis fungiodes, breast cancer, head and neck cancer and osteogenic sarcoma
  • purine analogs such as mercaptopurine and thioguanine which find use in the treatment of acute granulocytic, acute lymphocytic and chronic granulocytic leukemias.
  • Examples of natural product-based chemotherapeutic agents that can be carried out in combination with include, but not limited to, the vinca alkaloids, e.g., vinblastine (used in the treatment of breast and testicular cancer), vincristine and vindesine; the epipodophyllotoxins, e.g., etoposide and teniposide, both of which are useful in the treatment of testicular cancer and Kaposi's sarcoma; the antibiotic chemotherapeutic agents, e.g., daunorubicin, doxorubicin, epirubicin, mitomycin (used to treat stomach, cervix, colon, breast, bladder and pancreatic cancer), dactinomycin, temozolomide, plicamycin, bleomycin (used in the treatment of skin, esophagus and genitourinary tract cancer); and the enzymatic chemotherapeutic agents such as L- asparaginase.
  • the vinca alkaloids e
  • Examples of other signal transduction inhibiting agents that can be carried out in combination with include, but not limited to, gefitinib, erlotinib, sorafenib, herceptin, imatinib, dasatinib, sunitinib, nilotinib, lapatinib, pazopanib, vandetanib, vemurafenib, crizotinib, ruxolitinib, axitinib, bosutinib, regorafenib, tofacitinib, cabozantinib, ponatinib, dabrafenib, trametinib, and afatinib.
  • COX-II inhibitors such as, but not limited to, Vioxx, Celebrex (celecoxib), valdecoxib, paracoxib, rofecoxib; matrix
  • metalloproteinase inhibitors such as, but not limited to, AG-3340, RO 32-3555, and RS 13-0830.
  • the present disclosure also relates to pharmaceutical composition containing the compounds of Formula (I), (IA), (IB) or substructures thereof disclosed herein.
  • the present disclosure relates to a composition comprising compounds of Formula (I), (IA), (IB) or substructures thereof in a pharmaceutically acceptable carrier and in an amount effective to modulate the activity of Tyro3, Axl and Mer individually or in any combination of them or to treat diseases related to angiogenesis and/or cell proliferation and migration, and especially cancer,
  • the patient has hyperproliferative disease, and especially cancer, inflammatory diseases, autoimmune diseases, neurodisorders and the like, before administration of said compound of the disclosure and the compound of the disclosure is present in an amount effective to reduce said lipid level.
  • compositions useful herein also contain a pharmaceutically acceptable carrier, including any suitable diluent or excipient, which includes any pharmaceutical agent that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Pharmaceutically acceptable carriers include, but are not limited to, liquids, such as water, saline, glycerol and ethanol, and the like.
  • a thorough discussion of pharmaceutically acceptable carriers, diluents, and other excipients is presented in REMINGTON'S PHARMACEUTICAL SCIENCES (Mack Pub. Co., N.J. current edition). Those skilled in the art are also familiar with determining administration methods (oral, intravenous, inhalation, sub-cutaneous, etc.), dosage forms, suitable pharmaceutical excipients and other matters relevant to the delivery of the compounds to a subject in need thereof.
  • the compounds of the disclosure can be used in in vitro or in vivo studies as exemplary agents for comparative purposes to find other compounds also useful in treatment of, or protection from, the various diseases disclosed herein.
  • compositions of the disclosure can be prepared by combining a compound of the disclosure with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • compositions of the disclosure are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient.
  • Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the disclosure in aerosol form may hold a plurality of dosage units.
  • compositions to be administered will, in any event, contain a therapeutically effective amount of a compound of the disclosure, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this disclosure.
  • a pharmaceutical composition of the disclosure may be in the form of a solid or liquid.
  • the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form.
  • the carrier(s) may be liquid, with the
  • compositions being, for example, an oral oil, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.
  • the pharmaceutical composition When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
  • the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form.
  • a solid composition will typically contain one or more inert diluents or edible carriers.
  • binders such as carboxymethylcellulose, ethyl cellulose,
  • microcrystalline cellulose, gum tragacanth or gelatin excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
  • excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like
  • lubricants such as magnesium stearate or Sterotex
  • glidants such as colloidal silicon dioxide
  • sweetening agents such as sucrose or saccharin
  • a flavoring agent such as peppermint, methyl salicylate or orange flavoring
  • a coloring agent e.g., pepper
  • the pharmaceutical composition when in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.
  • a liquid carrier such as polyethylene glycol or oil.
  • the pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension.
  • the liquid may be for oral administration or for delivery by injection, as two examples.
  • preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer.
  • a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
  • the liquid pharmaceutical compositions of the disclosure may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride
  • fixed oils such as synthetic mono or diglycerides which may
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Physiological saline is a preferred adjuvant.
  • An injectable pharmaceutical composition is preferably sterile.
  • a liquid pharmaceutical composition of the disclosure intended for either parenteral or oral administration should contain an amount of a compound of the disclosure such that a suitable dosage will be obtained. Typically, this amount is at least 0.01% of a compound of the disclosure in the composition. When intended for oral administration, this amount may be varied to be between 0.1 and about 70% of the weight of the composition.
  • Preferred oral pharmaceutical compositions contain between about 4% and about 75% of the compound of the disclosure.
  • Preferred pharmaceutical compositions and preparations according to the present disclosure are prepared so that a parenteral dosage unit contains between 0.01 to 10% by weight of the compound prior to dilution of the disclosure.
  • the pharmaceutical composition of the disclosure may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base.
  • the base for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers.
  • Thickening agents may be present in a pharmaceutical composition for topical administration.
  • the composition may include a transdermal patch or iontophoresis device.
  • Topical formulations may contain a concentration of the compound of the disclosure from about 0.1 to about 10%> w/v (weight per unit volume).
  • the pharmaceutical composition of the disclosure may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug.
  • the composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient.
  • bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.
  • the pharmaceutical composition of the disclosure may include various materials, which modify the physical form of a solid or liquid dosage unit.
  • the composition may include materials that form a coating shell around the active ingredients.
  • the materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents.
  • the active ingredients may be encased in a gelatin capsule.
  • the pharmaceutical composition of the disclosure in solid or liquid form may include an agent that binds to the compound of the disclosure and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.
  • the pharmaceutical composition of the disclosure may consist of dosage units that can be administered as an aerosol.
  • aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the disclosure may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s).
  • the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit.
  • the pharmaceutical compositions of the disclosure may be prepared by methodology well known in the pharmaceutical art.
  • a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the disclosure with sterile, distilled water so as to form a solution.
  • a surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalently interact with the compound of the disclosure so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
  • the compounds of the disclosure, or their pharmaceutically acceptable salts are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
  • a therapeutically effective daily dose is (for a 70 kg mammal) from about 0.001 mg/kg (i.e., 0.7 mg) to about 100 mg/kg (i.e., 7.0 gm); preferably a therapeutically effective dose is (for a 70 kg mammal) from about 0.01 mg/kg (i.e., 7 mg) to about 50 mg/kg (i.e., 3.5 g); more preferably a therapeutically effective dose is (for a 70 kg mammal) from about 1 mg/kg (i.e., 70 mg) to about 25 mg/kg (i.e., 1.75 g).
  • Such combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of the disclosure and one or more additional active agents, as well as administration of the compound of the disclosure and each active agent in its own separate pharmaceutical dosage formulation.
  • a compound of the disclosure and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations.
  • the compounds of the disclosure and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
  • Isotopic enrichment is a process by which the relative abundance of the isotopes of a given element are altered, thus producing a form of the element that has been enriched in one particular isotope and depleted in its other isotopic forms.
  • Isotopic enrichment of a drug are used for the following applications: reducing or eliminating unwanted metabolites; increasing the half-life of the parent drug; decreasing the number of doses needed to achieve a desired effect; decreasing the amount of a dose necessary to achieve a desired effect; increasing the formation of active metabolites, if any are formed; and/or decreasing the production of deleterious metabolites in specific tissues and/or create a more effective drug and/or a safer drug for combination therapy, whether the combination therapy is intentional or not.
  • Suitable protecting groups include hydroxy, amino, mercapto and carboxylic acid.
  • Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like.
  • Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Suitable protecting groups for mercapto include -C(0)-R" (where R" is alkyl, aryl or arylalkyl), /?-methoxybenzyl, trityl and the like.
  • Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
  • the protecting group may also be a polymer resin such as a Wang resin or a 2-chlorotrityl- chloride resin.
  • the starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein.
  • the compounds of the Formula (I) or (IA1) are prepared in the above reaction scheme as follows: The starting a-bromo ketone 101 is treated with hexamethylenetetramine under Delepine reaction conditions to afford the a -amino ketone 102 as a
  • R 5 is hydrogen
  • the starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein.
  • the compounds of the Formula (IA2) are prepared in the above reaction scheme as follows:
  • the starting material, an appropriately substituted 2-amino-5- bromonicotinic aldehyde 201 reacts with hydroxylamine hydrochloride salt under reflux to afford the nicotinic aldoxime intermediate 202.
  • This oxime compound is then treated with N-chlorosuccinimide to generate the chlorinated oxime compound 203 which is cyclized with acetylene compound 204 to afford the isoxazole compound 205.
  • This isoxazole compound is coupled with the boron reagent 206 under the Suzuki coupling reaction conditions to afford the compound of formula (IA2).
  • the starting a— ketoalkene 301 reacts with aldehyde compound 302 (e.g., an appropriately substituted 2-amino-5-bromonicotinic aldehyde) under the Stetter Reaction conditions in the presence of a thiazolium catalyst, such as, but not limited to, 3-benzyl-5-(2-hydroxyethyl)-4-methylthiazolium chloride, to afford 1,4-dione compound303, which is cyclized under the Paal-Knorr furan synthesis conditions to afford the furan compound 304.
  • This furan compound is further coupled with the boron reagent 306 under the Suzuki coupling reaction conditions to afford the compound of formula (IA3).
  • the starting carboxylic acid 401 reacts with hydrazide 402 in the presence of a coupling reagent, such as, but not limited to, HATU and a base, such as, but not limited to, triethylamine to generate the 1 ,2-diacylhydrazine compound 403.
  • a coupling reagent such as, but not limited to, HATU and a base, such as, but not limited to, triethylamine
  • Cyclodehydration of compound 403 in the presence of a dehydrating reagent such as, but not limited to, triphenylphosphine/carbon tetrabromide to generate the 1,3,4- oxadiazole 404.
  • This oxadiazole compound is coupled with the boron reagent 406 under the Suzuki coupling reaction conditions to afford the compound of Formula (IA4).
  • Nitrile compound 501 is treated with hydroxylamine hydrochloride in the presence of a base, such as, but not limited to, triethylamine under reflux to generate amidoxime compound 502.
  • a base such as, but not limited to, triethylamine under reflux
  • a condensation reaction of the amidoxime compound 602 with an appropriately substituted nicotinic acid 503 in the presence of a coupling agent, such as, but not limited to, EDCI generates compound 504.
  • compound 504 Under reflux in the presence of pyridine, compound 504 is cyclized to afford 1,2,4-oxadiazole compound 505.
  • This oxadiazole compound is coupled with a boron reagent 506 under the Suzuki coupling reaction conditions to afford the compound of formula (IA5).
  • Nitrile compound 601 is treated with hydroxylamine hydrochloride in the presence of a base, such as, but not limited to, triethylamine under reflux to generate amidoxime compound 602.
  • a base such as, but not limited to, triethylamine under reflux
  • the condensation of amidoxime 602 with carboxlic acid 303 in the presence of a coupling agent, such as, but not limited to, EDCI generates compound 604.
  • compound 604 Under reflux in the presence of pyridine, compound 604 is cyclized to afford 1,2,4-oxadiazole compound 605.
  • This oxadiazole compound is coupled with the boron reagent 606 under the Suzuki coupling reaction conditions to afford the compound of formula (I) of the disclosure.
  • CD 3 OD CD 3 OD: ⁇ 8.28 (s, 1H), 8.18 (s, 1H), 7.74-7.70 (m, 2H), 7.54-7.34 (m, 4H), 6.85 (br s, 2H).
  • 1,4-dione 300 mg, 0.900 mmol
  • /?-toluenesulfonic acid 0.860 g, 4.50 mmol
  • 10 mL of toluene 10 mL
  • the residue was neutralized with saturated sodium bicarbonate, then extracted with dichloromethane (3 x 20 mL).
  • the organic solution was dried over anhydrous sodium sulfate and filtered.
  • hexafluorophosphate (576 mg, 1.50 mmol) were dissolved in N,N-dimethylformamide (3.0 mL), followed by the addition of triethylamine (0.282 mL, 2.00 mmol). The resulting mixture was stirred at ambient temperature overnight, and then diluted with ethyl acetate (50 mL), washed with brine (2 x 50 mL), and dried over sodium sulfate.
  • dichloromethane was added l-ethyl-3-(3-dimethylaminopropyl)carbodiimide (1.45 g, 7.36 mmol). The resulting mixture was stirred at ambient temperature overnight. After removal of solvent in vacuo, the residue was purified by column chromatography eluted with 5% methanol in dichloromethane to afford a yellow oil which was treated with dichloromethane and ethyl ether to afford N'-((2-amino-5-bromonicotinoyl)oxy)- benzimidamide as a yellow solid in 32% yield (0.25 g).
  • the mixture was degassed for 10 min, and then [l, -bis(diphenylphosphino)ferrocene]dichloro-palladium(II) (0.922 g, 1.26 mmol) was added.
  • the mixture was degassed for another 10 minutes and heated at 90 °C overnight, diluted with ethyl acetate (80 mL), washed with water (2 x 20 mL), brine (20 mL), dried and filtered.
  • dichloromethane (3 mL) was added acetic anhydride (0.013 mL, 0.136 mmol), followed by the addition of triethylamine (0.098 mL, 0.700 mmol) at 0 °C.
  • the mixture was stirred at ambient temperature for 3 h, diluted with dichloromethane (20 mL), washed with saturated sodium bicarbonate (10 mL) and brine (10 mL). The volatiles were removed in vacuo and the residue was purified by flash chromatography eluted with 5% methanol in dichloromethane to afford the title compound as a pale brown solid in 81% yield (55 mg).
  • dioxane:water was degassed with nitrogen for 10 minutes, followed by the addition of tetrakis(triphenylphosphine)-palladium(0) (24 mg, 0.021 mmol). The mixture was degassed for another 10 minutes and then sealed and heated at 110 °C overnight.
  • Recombinant human Tyro3 (455 -end, the gene accession number NM_006293), human Axl (473-end, the gene accession number NM_021913) and human Mer (578-872, the gene accession number NM_006343) were independently expressed by baculovirus in Sf9 insect cells using an N-terminal GST tag.
  • the recombinant proteins were stored at -70 °C in a medium containing 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 10 mM glutathione, 0.1 mM EDTA, 0.25 mM DTT, 0.1 mM PMSF and 25% glycerol.
  • the recombinant proteins were aliquoted into smaller quantities after centrifugation to avoid repeated handling and multiple freeze/thaw cycles for the most favorable performance.
  • This buffer solution consisted of the following components: 25 mM MOPS, pH 7.2, 12.5 mM ⁇ -glycerol-phosphate, 25 mM MgCl 2 , 5 mM EGTA, 2 mM EDTA and DTT 0.25 mM which was added prior to use.
  • Kinase Dilution Buffer Kinase Assay Buffer was diluted at a 1 :4 ratio (5x dilution) with distilled water.
  • [ 33 P]-ATP assay cocktail In a designated radioactive working area, a 250 ⁇ [ 33 P]-ATP assay cocktail was prepared by the addition of the following components: 150 ⁇ of 10 mM ATP stock solution, 100 ⁇ [ 33 P]-ATP (1 mci/100 iL), 5.75 mL of kinase assay buffer. This solution was stored in 1 mL aliquots at -20 °C.
  • ATP stock solution (10 mM): The ATP stock solution was prepared by dissolving 55 mg of ATP in 10 mL of kinase assay buffer. It was stored in 200 aliquots at -20 °C.
  • Active Kinase Stock Solution The active recombinant kinase protein (0.1 ⁇ g/ ⁇ L) was diluted with Kinase Dilution Buffer and the activity was assayed using a serial dilution method. The specific activity is expressed in nmoL/min/mg.
  • Substrates poly(4:l glu:tyr) is the substrate used for each kinase Tyro3, Axl and Mer: The peptide substrate was dissolved in water to give the final concentration of 1 mg/mL.
  • Test compound solution Test compound was dissolved in DMSO to obtain a 10 mM solution.
  • the assay solution was prepared by adding 5 ⁇ of this solution to 955 ⁇ , of 10% DMSO/water to obtain the final concentration of 50 ⁇ .
  • the enzymatic activity of all three kinases was determined as follows. Using a 96-well plate, the wells were divided into three categories: Blank wells, Background wells and Test wells. In Test wells, 5 ⁇ of the test compound solution and 5 ⁇ _, of the substrate solution were added. In Control wells, 5 ⁇ _, of 10%
  • DMSO/water and 5 ⁇ _, of the substrate solution were added.
  • 10 of 10% DMSO/water was added.
  • 10 ⁇ of Active Kinase Stock Solution was added to make up the volume in each well to 20 ⁇ . All test samples, controls and blanks were run in duplicate.
  • the reaction was initiated by the addition of 5 ⁇ of [ 33 P]-ATP assay cocktail, bringing the final volume up to 25 [iL in every well. The mixture was incubated at room temperature for 30 minutes. The reaction was terminated by transferring 10 ⁇ of the reaction mixture into a Millipore MultiScreen filter plate (cat. number MSPHN6B50).
  • the filter plate was washed in a 1% phosphoric acid solution with constant gentle shaking for 15 minutes and this step was repeated once. After the plate was dried in air, scintillation fluid was added to each well and the radioactivity in each well in CPM was counted by a Microbeta TriLux. The corrected CPM in each test well was determined by subtracting the average value of Blank well values. Percentage of inhibition of the kinase enzymatic activity by the test compound was determined using the following formula:
  • the IC 50 values were determined in a similar way following a serial dilution of the test compound. The percentage of inhibition at each concentration was calculated following the above formula. The IC 50 was estimated from the curve of % Inhibition against Concentration in log unit using Prism 5, version 5.01.
  • A549 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM, HyClone) containing 10% fetal bovine serum (FBS, Life technologies) and maintained in a humidified incubator at 37 °C with 5% C0 2 .
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • Cell viability assay protocol 5 x 10 3 A549 cells in 180 of DMEM containing 0.5%> FBS were seeded in 96-well flat bottom plates (Costar) and incubated in a humidified incubator at 37 °C with 5% C0 2 for 24 hours.
  • the test compound (20 ⁇ ) in different concentrations after a serial dilution was added to the wells to a final volume of 200 per well.
  • the wells with zero compound concentration were used as the Control wells and the Test wells contained different concentrations of the test compound.
  • To the Blank wells were added medium only. After 48 hours incubation, 40 of MTS (Promega) was added to each well and the plates were incubated for 10 minutes to 1 hour at 37 °C.
  • the cell viability was estimated by measurement of optical density at 490 nm using a microplate spectrophotometer (Molecular Devices).
  • A549 cells were treated with the test compound at different concentrations (5, 1 and 0.2 ⁇ ) in DMEM containing 0.5% FBS at 37 °C for 1 hour. After treatment, cells were seeded in the agar containing the test compound at different concentrations as outlined above in 6-well plates and the plates were incubated at 37 °C for 2 weeks. DMEM containing the various concentrations of the test compound was added on the top of agar and changed every 2-3 days. The viable colonies were stained with crystal violet and counted. This example is to demonstrate the capability of test compounds to prevent the formation of colonies.
  • A549 cells were seeded in agar in each well of 6-well plates and cultured in DMEM containing 10% FBS at 37 °C for six days and the colonies were formed.
  • the test compound dissolved in DMEM with different concentrations was added to the test wells of the plates and DMEM with no test compound or with diluted DMSO was added to the control wells.
  • the plates were incubated at 37 °C for 18-20 days during which period the DMEM containing 10% FBS and the various test compound dissolved in DMEM with different concentrations was added to the test wells of the plates and DMEM with no test compound or with diluted DMSO was added to the control wells.
  • the plates were incubated at 37 °C for 18-20 days during which period the DMEM containing 10% FBS and the various
  • concentrations of test compounds was changed every 2 to 3 days. The viable colonies were stained with crystal violet and counted. This example is to demonstrate the capability of teat compounds to inhibit the colony growth after they are established and/or to eliminate the established colonies.
  • 5 x l0 5 A549 cells were starved in DMEM contains no FBS in 6-well plate overnight, and treated with different concentrations of test compounds in serum- free medium for 30 minutes at 37 °C. Cells were subsequently stimulated with or without Gas6 (400 ng/mL) for 30 min at 37 °C.
  • Total proteins were extracted from the test compound treated cells using ice-cold RIPA buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 % (v/v) Triton-X 100, 0.1 (w/v) SDS) supplemented with protease inhibitor cocktail and phosphatase inhibitors. Protein concentrations were determined using Bradford assay.
  • Membranes were blocked at ambient temperature with 5% non-fat dry milk in Tris- buffered saline (TBS) containing 0.1% Tween-20 (TBST) for 1 hour and incubated with primary antibodies against Axl, Tyro3, phospho-AKT (Ser 473 ), total AKT or ⁇ -actin at 4 °C. After overnight incubation, membranes were washed with TBST and incubated with a secondary horseradish peroxidase (HRP)-labeled antibody (Jackson ImmunoResearch Laboratories, Inc.) for 1 hour at ambient temperature. Membranes were washed in TBST following incubation with secondary antibodies. Bound antibody complexes were detected and visualized using Luminata Classico Western HRP Substrate

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Provided herein are aminopyridine derivatives and pharmaceutical compositions that are useful as TAM family kinase inhibitors.

Description

AMINOPYRIDINE DERIVATIVES AS TAM FAMILY KINASE INHIBITORS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Patent Application No. 61/909,828 filed November 27, 2013, and U.S. Provisional Patent Application No. 61/909,830 filed November 27, 2013, which applications are incorporated herein by reference in thier entirety.
BACKGROUND
Technical Field
The present disclosure provides compounds that inhibit protein kinases Tyro3, Axl and Mer (TAM family kinases), prodrugs of the compounds, intermediates and methods of synthesizing the compounds and/or prodrugs, pharmaceutical compositions comprising the compounds and/or prodrugs and methods of using the compounds and/or prodrugs in a variety of contexts, including, for example, in the treatment and/or prevention of various diseases that are responsive to TAM family kinase inhibition and/or that are mediated, at least in part, by abnormal TAM family kinase activity.
Description of the Related Art
The receptor tyrosine kinases (RTKs) are transmembrane proteins and function as sensors for extracellular ligands, which transduce signals from extracellular medium to the cytoplasm. Their activation leads to the recruitment, phosphorylation, and activation of the downstream signaling pathways, which ultimately regulate cellular functions such as proliferation, growth, differentiation or motility. Abnormal overexpression levels and/or enhanced activities of RTKs have been associated to a variety of human cancers, leading to a strong interest in the development of inhibitors against these human cancers. Tyro-3, Axl, and Mer constitute the TAM family of RTKs characterized by a conserved sequence within the kinase domain and adhesion molecule like extracellular domains. With varying degree of specificity and affinity, TAM kinases can be activated by the vitamin K-dependent ligand Gas6 and/or Protein S. Strong evidence supports their association with both cancer (gain-in-function) and autoimmunity (loss-of-function). TAM kinase signaling has been implicated in a myriad of cellular responses, many of which are the hallmarks of cancer, including proliferation, survival, migration, invasion and angiogenesis. In addition, TAM plays pivotal roles innate immunity through inhibiting inflammation in macrophages and dendritic cells and promoting the phagocytosis of apoptotic cells. While the oncogenic activity of TAM kinases appears to be mediated via PI3K/AKT pathway, the JAK- STAT pathway is critical for their roles in immune responses. Overexpression of TAM kinases has been observed in over 20 human cancers. The level of their expression was shown to correlate with shorter progression- free and overall survival and their up- regulation has been linked to cancer resistance to cytotoxic drugs and targeted therapies.
While broadly expressed in various human tumor cell lines, Tyro3, Axl, and Mer exhibit their respective tissue-specific expression patterns. Tyro-3 is highly expressed in the nervous system whereas, Axl is expressed ubiquitously. Higher level of Mer is often found in hematopoietic lineages such as monocytes/macrophages, dendritic cells, NK cells, NKT cells, megakaryocytes, and platelets.
Compared to Axl and Mer, Tyro3 is the least studied kinase of the TAM family. Implication of Tyro3 in tumorigenesis was only recently substantiated by recent studies, which revealed Tyro3 is a potential oncogene in melanoma that is linked to poorer outcome of patients suffering from melanoma regardless the BRAF or NRAS status by conferring survival advantage to melanoma cells. It was also identified as one of kinases significantly up-regulated in lung cancer by a phosphoproteomic screen. High level of Tyro3 expression has also been correlated with thyroid cancer.
As the founding member of the TAM kinase family, Axl was discovered as a transforming gene in chronic myelogenous leukemia (CML). Axl overexpression has since been reported in a wide range of human malignancies and is associated with invasiveness and metastasis in lung, prostate, breast and pancreatic cancer. Axl is also an important regulator of breast cancer metastasis and EMT. Activation of the Axl kinase confers resistance to EGFR targeted therapy in lung cancer. Upregulation of Axl has been implicated as a mechanism of resistance to imatinib in CML and
gastrointestinal stromal tumors and to lapatinib in breast cancer. Axl expression has also been associated with chemoresistance in AML, NSCLC and ovarian cancer.
Mer is overexpressed/over-activated in a wide variety of cancers and has been established as a therapeutic target in hematopoietic malignancies and solid tumors including leukemia, non-small cell lung cancer, glioblastoma, melanoma, prostate cancer, breast cancer, colon cancer, gastric cancer, pituitary adenomas, and rhabdomyosarcomas. Oncogenic potential of Mer is mediated through the activation of several canonical oncogenic signaling pathways including the mitogen-activated protein kinase and phosphoinositide 3 -kinase pathways, as well as regulation of signal transducer and activator of transcription family members, migration-associated proteins including the focal adhesion kinase and myosin light chain 2, and prosurvival proteins such as survivin and Bcl-2. In neoplastic cells, these signaling events result in functional phenotypes such as decreased apoptosis, increased migration,
chemoresistance, increased colony formation, and increased tumor formation in murine models. Conversely, Mer inhibition by genetic or pharmacologic means can reverse these pro-oncogenic phenotypes.
To date, the following literature reports described small molecule inhibitors of Tyro3, Axl and/or Mer: Zhang et al, J. Med. Chem., 2014, 57, 7031-7041; Rho et al, Cancer Res., 2014, 74, 253-262; Traore et al, Euro. J. Med. Chem., 2013, 70, 789-801; Zhang et al, J. Med. Chem., 2013, 56, 9683-9692; Zhang et al, J. Med. Chem., 2013, 56, 9693-9700; Liu et al, Euro. J. Med. Chem., 2013, 65, 83-93; Powell et al, Bioorg. Med. Chem. Lett., 2013, 23, 1051-1055; Powell et al, Bioorg. Med. Chem. Lett., 2013, 23, 1046-1050; Suarez et al, Euro. J. Med. Chem., 2013, 61, 2-25; M. F. Burbridge et al, Mol Cancer Ther., 2013, 12, 1749-1762; Powell et al, Bioorg. Med. Chem. Lett. 2012, 22, 190-193; Liu et al, ACS Med. Chem. Lett., 2012, 3, 129- 134; Mollard et al, ACS Med. Chem. Lett, 2011, 2, 907-912; Holland et al, Cancer Res., 2010, 70(4), 1544-1554.; Ono et al, poster number MEDI-393, 244th ACS National Meeting & Exposition, Philadelphia, PA, Aug 19-23, 2012; Zhang et al, poster number MEDI-56, 244th ACS National Meeting & Exposition, Philadelphia, PA, Aug 19-23, 2012; Yang et al, poster number MEDI-265, 242nd ACS National Meeting & Exposition, Denver, CO, August 28-September 1, 2011; Zhang et al, poster number MEDI-62, 242nd ACS National Meeting & Exposition, Denver, CO, August 28- September 1, 2011; Wang et al, poster number MEDI-18, 242nd ACS National Meeting & Exposition, Denver, CO, August 28-September 1, 2011; Huang et al, J. Stru. Biol 2009, 165, 88-96. Axl inhibitors have also been disclosed in US2008188455A1;
WO2007030680A2; WO2008045978A1; WO2008080134A2; WO2008083353A1; WO2008083354A1; WO2008083356A1; WO2008083357A1; WO2008083367A2; WO2008128072A2; WO2009007390A2; WO2009024825A1; WO2009047514A1; WO2009053737A2; WO2009054864A1; WO2009127417A1; WO2010005876A2; WO2010005879A1; WO2010083465A1; WO2010090764A1; WO2011045084A1; WO2011138751A2; WO2012028332A1; WO2012135800A1; WO2013074633A1; WO2013115280A1; WO2013162061 Al .
SUMMARY
The present disclosure provides 2-aminopyridine derivatives that are capable of inhibiting the activity of TAM family kinases. Methods of using such derivatives to inhibit the activity of TAM family kinases and pharmaceutical compositions comprising such derivatives are also disclosed.
One embodiment provides compounds of formula (I):
Figure imgf000005_0001
Formula (I)
wherein:
Figure imgf000005_0002
W is
each A, A1 and A2 is the same or different and independently -N=, CR5=, or -0-;
L is heteroaryl, heterocyclyl, -N(R6)-, -0-, -C(O)-, -C(0)0-, -S(O),- where t is 0, 1, or 2, -CON(R6)-, -N(R6)CO-, -S02N(R6)-, or -N(R6)CON(R6)-, provided that when L is heteroaryl, L is not pyridinyl, pyrazinyl or thienyl;
R1 is alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or heteroarylalkyl;
R2 is hydrogen, heterocyclyl, heterocyclylalkyl, cycloalkyl, alkyl, aralkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, or aryl;
each R3 and R4 is the same or different and independently hydrogen, alkyl, aralkyl, alkenyl, aralkenyl, alkynyl, aralkynyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl;
R5, at each occurrence, is hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl;
R6 is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, or haloalkyl, a stereoisomer, enantiomer or tautomer thereof, an isotopically enriched derivative thereof, a pharmaceutically acceptable salt thereof, a pharmaceutical composition thereof or a prodrug thereof.
Of the compounds of Formula (I), an embodiment provides compound of Formula (IA):
Figure imgf000006_0001
Formula (IA)
wherein,
A, A1, A2,
Figure imgf000006_0002
R3, and R4 are as defined above.
Of the compounds of Formula (IA), an embodiment provides compounds of Formula (IA1):
Figure imgf000006_0003
Formula (IA1)
wherein:
R1, R2, R3, and R4 are as defined above.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA2)
Figure imgf000006_0004
Formula (IA2)
wherein, R1, R2, R3, and R4 are as defined above.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA3)
Figure imgf000006_0005
Formula (I A3)
wherein, R1, R2, R3, and R4 are as defined above. Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA4):
Figure imgf000007_0001
Formula (IA4)
wherein, R1, R2, R3, and R4 are as defined above.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA5
Figure imgf000007_0002
wherein, R1, R2, R3, and R4 are as defined above.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA6):
Figure imgf000007_0003
Formula (IA6)
wherein, R1, R2, R3, and R4 are as defined above.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA7):
Figure imgf000007_0004
wherein, R1, R2, R3, and R4 are as defined above.
Of the compounds of Formula (I), an embodiment provides compound of
Formula (IB):
Figure imgf000008_0001
Formula (IB)
wherein,
A, A1, A2,
Figure imgf000008_0002
R3, R4, and R5 are as defined above,
L is heteroaryl or heterocycle;
R7 is hydrogen, alkyl, alkoxy, or halo;
X is -0-, -NR8-, or -C(R9)2-;
R8 is hydrogen, alkyl, or -C(0)R10-;
each R9 is independently hydrogen, alkyl, alkoxy, or halo; and
R10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
Of the compounds of Formula (IB), an embodiment provides compound of Formula (IB1):
Figure imgf000008_0003
Formula (IB1)
wherein,
A, A1, A2, R1, R3, R4, R7, and R8 are as defined above.
Of the compounds of Formula (IB), an embodiment provides compound of Formula (IB2):
Figure imgf000008_0004
Formula (IB2)
wherein,
A, A1, A2,
Figure imgf000008_0005
R3, R4, and R7 are as defined above.
Of the compounds of Formula (IB), an embodiment provides compound of Formula (IB3):
Figure imgf000009_0001
Formula (IB 3)
wherein,
A, A1, A2, R1, R3, R4, R7, and R9 are as defined above.
Yet another embodiment provides a pharmaceutical composition of a compound of any one of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3), or any one the specific compounds disclosed herein.
Yet another embodiment provides a method of treating leukemia and lymphoma comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3)or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating endometriosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating restenosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating
atherosclerosis/thrombosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same. Yet another embodiment provides a method of treating psoriasis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating age-related macular degeneration or diabetic retinopathy comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating retinopathy of prematurity comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating renal transplant rejection comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating rheumatoid arthritis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating osteoarthritis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating osteoporosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of treating cataracts comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a pharmaceutical composition comprising the same.
Yet another embodiment provides a method of preventing/treating tumor metastasis or treating metastasized tumors comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of a compound of Formula (I), (IA), or (IB), or any one of the substructures as represented by Formula (IA1), (IA2), (IA3), (IA4), (IA5), (IA6), (IA7), (IB1), (IB2), or (IB3) or a
pharmaceutical composition comprising the same.
DETAILED DESCRIPTION
Definitions
Certain chemical groups named herein are preceded by a shorthand notation indicating the total number of carbon atoms that are to be found in the indicated chemical group. For example;
Figure imgf000011_0001
describes an alkyl group, as defined below, having a total of 7 to 12 carbon atoms, and C4-Ci2cycloalkylalkyl describes a cycloalkylalkyl group, as defined below, having a total of 4 to 12 carbon atoms. The total number of carbons in the shorthand notation does not include carbons that may exist in substituents of the group described.
Accordingly, as used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated:
"Amino" refers to the -NH2 radical.
"Methoxy" refers to the -OCH3 radical.
"Cyano" refers to the -CN radical.
"Nitro" refers to the -N02 radical.
"Trifluoromethyl" refers to the -CF3 radical.
"Oxo" refers to the =0.
"Thioxo" refers to the =S. "Acyl" refers to -C(0)R radical, wherein R is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl;
"Alkyl" refers to a straight or branched hydrocarbon chain radical, when unsubstituted, consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms, preferably one to eight carbon atoms or one to six carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (z'so-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted by one or more
substituents, as defined herein. "Alkenyl" refers to a straight or branched hydrocarbon chain radical group, when unsubstituted, consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g. , ethenyl, prop- 1 -enyl, but- 1 -enyl, pent- 1 -enyl, penta- 1 ,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group may be optionally substituted by one of or more substituents, as defined herein.
"Alkynyl" refers to a straight or branched hydrocarbon chain radical group, when unsubstituted, consisting solely of carbon and hydrogen atoms, containing at least one triple bond, optionally containing at least one double bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group may be optionally substituted by one or more substituents, as defined herein. "Alkylene" and "alkylene chain" refer to a straight or branched divalent hydrocarbon chain, linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, preferably having from one to eight carbons, e.g., methylene, ethylene, propylene, n-butylene, and the like. The alkylene chain may be attached to the rest of the molecule and to the radical group through one carbon within the chain or through any two carbons within the chain.
"Alkenylene" and "alkenylene chain" refer to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one double bond and having from two to twelve carbon atoms, e.g. , ethenylene, propenylene, n-butenylene, and the like. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
"Alkoxy" refers to a radical of the formula -ORa where Ra is an alkyl radical as defined above. The alkyl part of the alkoxy radical may be optionally substituted as defined above for an alkyl radical.
"Aryl" refers to aromatic monocyclic or multi-cyclic hydrocarbon ring system, when unsubstituted, consisting only of hydrogen and carbon and containing from 6 to 19 carbon atoms, preferably 6 to 10 carbon atoms, where the ring system may be partially or fully saturated. Aryl groups include, but are not limited to groups such as fluorenyl, phenyl and naphthyl. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals optionally substituted by one or more substituents, as defined herein. "Aralkyl" refers to a radical of the formula -RaRb where Ra is an alkyl radical as defined above and Rb is one or more aryl radicals as defined above, e.g., benzyl, diphenylmethyl and the like. The aryl part of the aralkyl radical may be optionally substituted as described above for an aryl group. The alkyl part of the aralkyl radical may be optionally substituted as defined above for an alkyl group.
"Aralkenyl" refers to a radical of the formula -RcRb where Rc is an alkenyl radical as defined above and Rb is one or more aryl radicals as defined above, which may be optionally substituted as described above. The aryl part of the aralkenyl radical may be optionally substituted as described above for an aryl group. The alkenyl part of the aralkenyl radical may be optionally substituted as defined above for an alkenyl group.
"Aralkynyl" refers to a radical of the formula -R Rb where Rd is an alkynyl radical as defined above and Rb is one or more aryl radicals as defined above. The aryl part of the aralkynyl radical may be optionally substituted as described above for an aryl group. The alkynyl part of the aralkynyl radical may be optionally substituted as defined above for an alkynyl group. "Cycloalkyl" refers to a stable non-aromatic monocyclic or bicyclic hydrocarbon radical, when unsubstituted, consisting solely of carbon and hydrogen atoms, having from three to fifteen carbon atoms, preferably having from three to twelve carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond, e.g. , cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, decalinyl and the like. Unless otherwise stated specifically in the specification, the term "cycloalkyl" is meant to include cycloalkyl radicals which are optionally substituted by one or more substituents, as defined herein. "Cycloalkylalkyl" refers to a radical of the formula -RaRd where Ra is an alkyl radical as defined above and Rj is a cycloalkyl radical as defined above. The cycloalkyl part of the cycloalkyl radical may be optionally substituted as defined above for a cycloalkyl radical. The alkyl part of the cycloalkyl radical may be optionally substituted as defined above for an alkyl radical.
"Halo" refers to bromo, chloro, fluoro or iodo.
"Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above. One or more carbons of the alkyl radical may be substituted by the one or more halo radicals. Examples of haloalkyl include, trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1 -fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoro-propyl, 1 -bromomethyl-2-bromoethyl, and the like. The alkyl part of the haloalkyl radical may be optionally substituted as defined above for an alkyl group.
"Haloalkenyl" refers to an alkenyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., 2-bromoethenyl, 3-bromoprop-l-enyl, and the like. The alkenyl part of the haloalkenyl radical may be optionally substituted as defined above for an alkyl group.
"Heterocyclyl" refers to a stable 3- to 18-membered non-aromatic ring radical including, as ring atoms, at least one carbon atom and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For purposes of this disclosure, the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; and the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl,
oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, the term "heterocyclyl" is meant to include heterocyclyl radicals as defined above which are optionally substituted by one or more substituents, as defined herein. When the heterocyclyl is a substituent of another moiety, the heterocyclyl is monovalent, which means that the heterocyclyl is connected to the other moiety by a single ring atom. An example of a monovalent heterocyclyl can be found in the radical of heterocyclylalkyl, in which a heterocyclyl group is a substituent of an alkyl group. When heterocyclyl is a linker moiety (L) of Formula (I), the heterocyclyl is a divalent radical. In this occurrence, the heterocyclyl (as L) is linked, by two of its ring atoms, to the 2-aminopyridine moiety and R2 of Formula (I). An example of a divalent heterocycl -piperidinyl, shown below:
Figure imgf000015_0001
"Heterocyclylalkyl" refers to a radical of the formula -RaRg where Ra is an alkyl radical as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. The alkyl part of the
heterocyclylalkyl radical may be optionally substituted as defined above for an alkyl group. The heterocyclyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for a heterocyclyl group.
"Heteroaryl" refers to a 5- to 18-membered aromatic ring radical including, as ring atoms, at least one carbon atom and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For purposes of this disclosure, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; and the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzothiadiazolyl,
benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl,
dibenzofuranyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl. Unless stated otherwise specifically in the specification, the term "heteroaryl" is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents, as defined herein. Heteroaryl, as defined herein, may be monovalent or divalent. When heteroaryl is a substituent of another moiety, the heteroaryl is monovalent, which means that the heteroaryl is connected to the other moiety by a single ring atom. An example of a monovalent heteroaryl can be found in the radical of heteroarylalkyl, in which an alkyl group is substituted with a heteroaryl group. When heteroaryl is a linker moiety (L) of Formula (I), the heteroaryl is a divalent radical. In this occurrence, the heteroaryl (as L) is linked, by two of its ring atoms, to the
2-aminopyridine moiety and R2 of Formula (I). An example of a divalent heteroaryl is lH-pyrazol-4-yl, shown below:
Figure imgf000016_0001
Heteroarylalkyl" refers to a radical of the formula -RaRf where Ra is an alkyl radical as defined above and Rf is a heteroaryl radical as defined above. The heteroaryl part of the heteroarylalkyl radical may be optionally substituted as defined above for a heteroaryl group. The alkyl part of the heteroarylalkyl radical may be optionally substituted as defined above for an alkyl group.
Heteroarylalkenyl" refers to a radical of the formula -RcRg where Rc is an alkenyl radical as defined above and Rg is a heteroaryl radical as defined above. The heteroaryl part of the heteroarylalkenyl radical may be optionally substituted as defined above for a heteroaryl group. The alkenyl part of the heteroarylalkenyl radical may be optionally substituted as defined above for an alkenyl group. "N-heteroaryl" is a subset of heteroaryl, and refers to a heteroaryl having at least one nitrogen ring atom. Heteroaryl is otherwise as defined as herein. Examples of N-heteroaryls include, without limitation, benzimidazolyl, benzindolyl,
benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, imidazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, and triazinyl.
"N-heterocyclyl" is a subset of heterocyclyl, and refers to a heterocyclyl having at least one nitrogen ring atom. Heterocyclyl is otherwise as defined as herein. Examples of N- heterocyclyls include, without limitation, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl,
octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl,
2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl.
"Substituent" refers to a radical (a single non-hydrogen atom or a functional group) that is or can be bonded to another molecule. An substituent is therefore any one of the following radicals: alkyl, alkenyl, amino, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR14, -OC(0)R14, - N(R14)2, -C(0)R14, -C(0)OR14, -C(0)N(R14)2, -N(R14)C(0)OR16, -N(R14)C(0)R16, - N(R14)(S(0)tR16) (where t is 1 to 2), -S(0),OR16 (where t is 1 to 2), -S(0)tR16 (where t is 0 to 2), and -S(0)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or
heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, and where each of the above substituents is unsubstituted unless otherwise indicated.
"Prodrugs" refers to a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of Formula (I) or any one of the substructures. Thus, the term "prodrug" refers to a metabolic precursor of a compound of Formula (I) or any one of the substructures, that is pharmaceutically acceptable; the latter is also referred to as a "parent compound." A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound, i.e., the parent compound. Prodrugs are typically rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24, Elsevier, Amsterdam).
A discussion of prodrugs is provided in Higuchi, Ί., εΐ αΙ, "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible
Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
The term "prodrug" is also meant to include any covalently bonded carriers which release the active compound of the disclosure in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the disclosure may be prepared by modifying functional groups present in the compound of Formula (I) or any one of the substructures in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the disclosure wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug disclosure is administered to a mammalian subject, cleaves to restore the free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate, and phosphate derivatives of alcohol or amine functional groups in the compounds of Formula (I), (IA), (IB) or any one of the substructures.
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
"Mammal" or "mammalian subject" includes humans and domestic animals, such as cats, dogs, swine, cattle, sheep, goats, horses, rabbits, and the like.
"Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid,
benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene- 1,5-disulfonic acid, naphthalene-2-sulfonic acid, l-hydroxy-2 -naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, / toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine,
diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine,
N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
Often crystallizations produce a solvate of the compound of the disclosure. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the disclosure with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate.
Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present disclosure may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the disclosure may be true solvates, while in other cases, the compound of the disclosure may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
A "pharmaceutical composition" refers to a formulation of a compound of the disclosure and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
"Therapeutically effective amount" refers to that amount of a compound of the disclosure which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a disease or condition in the mammal, preferably a human. The amount of a compound of the disclosure which constitutes a "therapeutically effective amount" will vary depending on the compound, the condition and its severity, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure. "Treating" or "treatment" as used herein covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or disorder of interest, and includes:
(i) preventing the disease or condition from occurring in a mammal, in particular, when such mammal is predisposed to the condition but has not yet been diagnosed as having it;
(ii) inhibiting the disease or condition, i.e., arresting its development; or
(iii) relieving the disease or condition, i.e., causing regression of the disease or condition.
As used herein, the terms "disease" and "condition" may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
The compounds of the disclosure, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to
enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as HPLC using a chiral column. When the compounds described herein contain olefmic double bonds or other centers of geometric
asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are non-superimposeable mirror images of one another. A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any said compounds.
An "isotopically enriched derivative" refers to a compound wherein one or more atoms are replaced by atoms having the same atomic number but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds of the disclosure comprises isotopes of hydrogen, such as 2H and 3H, carbon, such as UC, 13C and 14C,
38 18 123 125 chlorine, such as CI, fluorine, such as F, iodine, such as I and I, nitrogen, such as 13N and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 31P, 32P and 33P, and sulphur, such as 35S. Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopically-enriched compounds of the disclosure can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations Sections using an appropriate isotopically-enriched reagent in place of the non-enriched reagent previously employed.
Where a bond is shown as a dashed line (— ), it is understood that the location allows for the possibility of a double bond. For example, the structure of the linker W is shown as:
Figure imgf000022_0001
in which each dash bond may, but not necessarily, indicate the presence of a double bond. For instance, when A is -NH=, A is connected to the two adjacent carbon atoms by a single bond and a double bond, respectively. On the other hand, if A is defined as -O -, A is connected to the two adjacent carbon atoms by single bonds, respectively. The location and number of the double bonds in a given ring structure of W should satisfy the valence requirement, as would be recognized by a skilled person in the art.
The chemical naming protocol and structure diagrams used herein employ and rely the chemical naming features as utilized by Chemdraw version 12.0.2.1076 (available from Cambridgesoft Corp., Cambridge, MA). For complex chemical names employed herein, a substituent group is named before the group to which it attaches. For example, cyclopropylethyl comprises an ethyl backbone with cyclopropyl substituent. In chemical structure diagrams, all bonds are identified, except for some carbon atoms which are assumed to be bonded to sufficient hydrogen atoms to complete the valency.
For example, a compound of formula (I), as set forth above in the Summary of this disclosure, where W is a divalent oxazolyl moiety connected to the 2-aminopyridine moiety and the R1 group at 2 and 5 positions:
\VV
L is lH-pyrazol-4-yl, R1 is 4-chlorophenyl, R2 is l-(piperidin-4-yl), R3, R4 and R5 are each hydrogen, i.e., a compound of the following formula:
Figure imgf000023_0001
is named herein as: 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine.
It should be note that there are two possible ways for W to connect with the 2-aminopyridine moiety and the R1 group, namely, the oxazolyl may be connected at its 2 position to the 2-aminopyridine moiety, as shown above; or the oxazolyl may be connected at its 5 position to the 2-aminopyridine moiety. Thus, the following compound is also encompassed by Formula (I), when L is lH-pyrazol-4-yl, R1 is 4-chlorophenyl, R2 is l-(piperidin-4-yl), and R3, R4 and R5 are hydrogens:
Figure imgf000023_0002
Similarly, the linker L, which is a divalent radical, may also have two possible ways to connect the 2-aminopyridine and the R2 group. For instance, when L is an amido linker, i.e., -CON(R6)-, a compound of Formula (I) may have the carbonyl portion directly connected to the 2-aminopyridine moiety. Another compound of Formula (I) may have the carbonyl portion directly connected to the R2 group. As another example, a compound of formula (I), as set forth above in the Summary of this disclosure, where W is a divalent 1,3,4 -oxadiazolyl moiety connected to the 2-aminopyridine moiety and the R1 group at 2 and 5 positions:
Figure imgf000024_0001
L is lH-pyrazol-4-yl, R1 is 2,6-dichlorophenyl, R2 is l-(piperidin-4-yl), R3 and R4 are each hydrogen, i.e., a compound of the following formula:
Figure imgf000024_0002
is named herein as: 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4 yl)-lH-pyrazol-4-yl)pyridin-2-amine.
Embodiments
Provided herein are aminopyridine derivatives useful as TAM family kinase inhibitors. One embodiment provides a compound of formula (I):
Figure imgf000024_0003
Formula (I)
Figure imgf000024_0004
each A, A1 and A2 is the same or different and independently -N=,
CR =, or -0-;
L is a heteroaryl, heterocyclyl, -N(R6)-, -0-, -C(O)-, -C(0)0-, -S(O),- where t is 0, 1 , or 2, -CON(R6)-, -N(R6)CO-, -S02N(R6)-, or -N(R6)CON(R6)-; provided that when L is heteroaryl, L is not pyridinyl, pyrazinyl or thienyl;
R1 is alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or heteroarylalkyl;
R2 is hydrogen, heterocyclyl, heterocyclylalkyl, cycloalkyl, alkyl, aralkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, or aryl; each R3 and R4 is the same or different and independently selected from hydrogen, alkyl, aralkyl, alkenyl, aralkenyl, alkynyl, aralkynyl, cycloalkyl,
cycloalkylalkyl, halo, and haloalkyl;
R5, at each occurrence, is hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl;
R6 is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, or haloalkyl, a stereoisomer, enantiomer or tautomer thereof, an isotopically enriched derivative thereof, a pharmaceutically acceptable salt thereof, a pharmaceutical composition thereof or a prodrug thereof.
In a further embodiment, L is heteroaryl.
In yet another embodiment, L is N-heteroaryl.
In yet another embodiment, L is divalent pyrrozolyl moiety. In a specific embodiment, L is lH-pyrazol-4-yl, and the compound of Formula (I) is represented by Formula (IA):
Figure imgf000025_0001
Formula (IA)
wherein,
A, A1, A2,
Figure imgf000025_0002
R3, and R4 are as defined above.
Of the compounds of Formula (IA), an embodiment provides compounds of Formula (IA1):
Figure imgf000025_0003
Formula (IA1)
wherein:
R1, R2, R3, and R4 are as defined above.
In more specific embodiments of Formula (IA1), R1 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
In a more specific embodiment of Formula (IA1), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl. In other embodiments of Formula (IA1), R2 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
In a more specific embodiment of Formula (IA1), R2 is heterocyclyl.
In more specific embodiments of Formula (IA1), R2 is N-heterocyclyl. More specifically, R2 is piperidinyl or substituted piperidinyl.
In other more specific embodiments of Formula (IA1), R2 is
tetr ahy dropyr any 1.
In another more specific embodiment of Formula (IA1), R2 is cyclohexyl.
In certain specific embodiments of Formula (IA1), each of R3 and R4 is hydrogen, R1 is aryl, R2 is heterocyclyl.
In certain specific embodiments of Formula (IA1), each of R3 and R4 is hydrogen, R1 is phenyl or substituted phenyl, R2 is heterocyclyl.
In certain specific embodiments of Formula (IA1), each of R3 and R4 is hydrogen, R1 is phenyl or substituted phenyl, R2 is piperidinyl or substituted piperidinyl.
In certain specific embodiments, each of R3 and R4 is hydrogen, R1 is phenyl (including phenyl substituted with one or more halo), and R2 is heterocyclyl. In more specific embodiments, R2 is a l-piperidin-4-yl (including l-piperidin-4-yl substituted at the 1 -position with alkyl or acyl).
In various specific embodiments, the compound of Formula (IA1) is: tert-butyl-4-(4-(6-amino-5-(5-phenyloxazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine- 1 -carboxylate;
3-(5-phenyloxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidine- 1 -carboxylate;
3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
l-(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)ethanone;
(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(cyclopropyl)methanone; 3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
l-(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)ethanone;
3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(cyclopropyl)methanone;
(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(phenyl)methanone;
l-(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)-2-phenylethanone;
(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(phenyl)methanone;
l-(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)-2-phenylethanone;
3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
l-(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)-2,2-dimethylpropan- 1 -one;
(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(4-fluorophenyl)methanone; or
5-(l-(l-ethylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-phenyloxazol-2-yl)pyridin-2-amine.
In other more specific embodiments of Formula (IA1), each of R3 and R4 is hydrogen, R1 is phenyl or substituted phenyl, R2 is tetrahydropyranyl.
In various specific embodiments, the compound of Formula (IA1) is: 3-(5-phenyloxazol-2-yl)-5-(l-(tetrahydro-2pyran-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine; or
3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(tetrahydro-2pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine.
In other more specific embodiments of Formula (IA1), each of R3 and R4 is hydrogen, R1 is phenyl or substituted phenyl, R2 is cyclohexyl.
In various specific embodiments, the compound of Formula (IA1) is: 3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-cyclohexyl-lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-cyclohexyl-lH-pyrazol-4-yl)pyridin-2-amine; or
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenyloxazol-2-yl)pyridin-2-amine.
In other embodiments, each of R3 and R4 is hydrogen, R1 is heteroaryl
(e.g., pyridinyl), and R2 is heterocyclyl.
In more specific embodiments, the compound is 5-(l-(piperidin-4-yl)- lH-pyrazol-4-yl)-3-(5-(pyridin-3-yl)oxazol-2-yl)pyridin-2-amine.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA2):
Figure imgf000028_0001
Formula (IA2)
wherein, R1, R2, R3, and R4 are as defined above.
In more specific embodiments of Formula (IA2), R1 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
In other embodiments of Formula (IA2), R2 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R2 is heterocyclyl. More specifically, R2 is N-heterocyclyl.
In certain specific embodiments of Formula (IA2), each of R3 and R4 is hydrogen, R1 is phenyl or substituted phenyl, R2 is piperidinyl or substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
In various specific embodiments, the compound of Formula (IA2) is: 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenylisoxazol-3-yl)pyridin-2-amine;
3-(5-phenylisoxazol-3-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
3-(5-phenylisoxazol-3-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine; or 5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-phenylisoxazol-3-yl)pyridin-2- amine.
Of the compounds of Formula (I A), another embodiment provides compounds of Formula (IA3):
Figure imgf000029_0001
Formula (I A3)
wherein, R1, R2, R3, and R4 are as defined above.
In more specific embodiments of Formula (IA3), R1 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
In other embodiments of Formula (IA3), R2 is aryl, heteroaryl, cycloalkyl or heterocyclyl. More specifically, R2 is heterocyclyl. More specifically, R2 is N-heterocyclyl.
In certain specific embodiments of Formula (IA3), each of R3 and R4 is hydrogen, R1 is phenyl or substituted phenyl, R2 is piperidinyl or substituted
piperidinyl, tetrahydropyranyl or cyclohexyl.
In various specific embodiments, the compound of Formula (IA3) is: 3-(5-phenylfuran-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine;
5 -( 1 -( 1 -methylpiperidin-4-yl)- lH-pyrazol-4-yl)-3 -(5 -phenylfuran-2-yl)pyridin-2-amine; 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenylfuran-2-yl)pyridin-2-amine; or
3 -(5 -phenylfuran-2-yl)-5 -( 1 -(tetrahydro-2H-pyran-4-yl)- lH-pyrazol-4-yl)pyridin-2- amine.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA4
Figure imgf000029_0002
Formula (IA4)
wherein, R1, R2, R3, and R4 are as defined above.
In more specific embodiments of Formula (IA4), R1 is aryl, heteroaryl, cycloalkyl, heterocyclyl, aralkyl, or cycloalkylalkyl.
In a more specific embodiment of Formula (IA4), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl. In another more specific embodiment of Formula (IA4), R1 is heteroaryl. More specifically, R1 is a pyridinyl, substituted pyridinyl, pyrazinyl, substituted pyrazinyl, thiazolyl, substituted thiazolyl.
In further specific embodiment of Formula (IA4), R1 is aralkyl. More specifically, R1 is benzyl.
In further specific embodiment of Formula (IA4), R1 is cycloalkylalkyl. More specifically, R1 is cycloalkylalkyl.
In any of the above embodiments of Formula (IA4), R2 is hydrogen, aryl, heteroaryl, cycloalkyl or heterocyclyl.
In a more specific embodiment of Formula (IA4), R2 is heterocyclyl.
More specifically, R2 is N-heterocyclyl.
In more specific embodiments of Formula (IA4), R2 is piperidinyl or substituted piperidinyl.
In other more specific embodiments of Formula (IA4), R2 is tetr ahy dropyr any 1.
In other more specific embodiments of Formula (IA4), R2 is hydrogen. In certain specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is aryl, R2 is heterocyclyl, including N-heterocyclyl.
In certain specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is phenyl or phenyl substituted with one or more halo or alkyl, R2 is piperidinyl or substituted piperidinyl.
In various specific embodiments, a compound of Formula (IA4) is:
tert-butyl 4-(4-(6-amino-5 -(5 -phenyl- 1 ,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine- 1 -carboxylate;
3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
3-(5-(2,6-dichloro-3-fluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine;
3-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate; 3-(5-(2,5-difluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate;
3-(5-(4-fluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(4-fluorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
l-(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)ethanone;
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(l-methylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine;
1 -(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)- 1 ,3,4-oxadiazol-2-yl)pyridin-3-yl)- 1H- pyrazol- 1 -yl)piperidin- 1 -yl)-4-methylpentan- 1 -one;
(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)(cyclopropyl)methanone;
(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)(phenyl)methanone;
l-(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)-2-phenylethanone;
3-(5-(2,6-dichlorophenyl)- 1 ,3,4-oxadiazol-2-yl)-5-(l -(1 -ethylpiperidin-4-yl)- 1H- pyrazol-4-yl)pyridin-2-amine; or
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(l-dodecylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine.
In other more specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is heteroaryl or substituted heteroaryl, R2 is piperidinyl or substituted piperidinyl.
In various specific embodiments, the compound of Formula (IA4) is: tert-butyl 4-(4-(6-amino-5-(5-(pyrazin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate; tert-butyl 4-(4-(6-amino-5-(5-(pyridin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(pyrazin-2-yl)-l,3,4-oxadiazol-2- yl)pyridin-2-amine;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(pyridin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin- 2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(thiazol-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(thiazol-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-
2-amine;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(pyridin-3-yl)-l,3,4-oxadiazol-2-yl)pyridin- 2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(pyridin-3-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
tert-butyl 4-(4-(6-amino-5-(5-(4-(trifiuoromethyl)-thiazol-2-yl)-l,3,4-oxadiazol-2- yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate; or
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(4-(trifluoromethyl)thiazol-2-yl)-l,3,4- oxadiazol-2-yl)pyridin-2-amine.
In other more specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is cycloalkylalkyl or aralkyl, R2 is piperidinyl or substituted piperidinyl.
In various specific embodiments, the compound of Formula (IA4) is: tert-butyl 4-(4-(6-amino-5-(5-(cyclopropylmethyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate;
3-(5-(cyclopropylmethyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5 -(5 -benzyl- 1 ,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine-l -carboxylate; or
3-(5-benzyl-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine.
In other more specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is aryl or more specifically phenyl or substituted phenyl, R2 is hydrogen.
In various specific embodiments, the compound of Formula (IA4) is: 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine;
3-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(2,5-difluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; or 3-(5-(4-fluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine.
In other more specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is aryl or more specifically, phenyl or substituted phenyl, R2 is cycloalkyl.
In a specific embodiment, the compound of Formula (IA4) is:
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2- yl)pyridin-2-amine;
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenyl-l,3,4-oxadiazol-2-yl)pyridin-2-amine.
In yet other more specific embodiments of Formula (IA4), each of R3 and R4 is hydrogen, R1 is aryl or more specifically, phenyl or substituted phenyl, R2 is tetrahydropyranyl.
In a specific embodiment, the compound of Formula (IA4) is:
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine; or
3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA5
Figure imgf000033_0001
Formula (IA5)
In a more specific embodiment of Formula (IA5), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
In a more specific embodiment of Formula (IA5), R2 is heterocyclyl. More specifically, R2 is N-heterocyclyl.
In more specific embodiments of Formula (IA5), R2 is piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
In other more specific embodiments of Formula (IA5), R2 is hydrogen. In certain specific embodiments of Formula (IA5), each of R3 and R4 is hydrogen, R1 is aryl, R2 is heterocyclyl, including N-heterocyclyl.
In yet other more specific embodiments of Formula (IA5), each of R3 and R4 is hydrogen, R1 is aryl or more specifically, phenyl or substituted phenyl, R2 is hydrogen, piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
In yet other more specific embodiments of Formula (IA5), each of R3 and R4 is hydrogen, R1 is aryl or more specifically, phenyl or substituted phenyl, R2 is piperidinyl, substituted piperidinyl.
In a specific embodiment, the compound of Formula (IA5) is:
3-(3-phenyl- 1 ,2,4-oxadiazol-5-yl)-5-(l -(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin-2- amine;
1 -(4-(4-(6-amino-5 -(3 -phenyl- 1 ,2,4-oxadiazol-5-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidin- 1 -yl)ethanone;
(4-(4-(6-amino-5-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(cyclopropyl)methanone;
5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(3-phenyl-l,2,4-oxadiazol-5- yl)pyridin-2-amine;
(4-(4-(6-amino-5-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(phenyl)methanone;
1 -(4-(4-(6-amino-5 -(3 -phenyl- 1 ,2,4-oxadiazol-5-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidin- 1 -yl)-2-phenylethanone;
3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(3-(2,6-dichlorophenyl)- 1 ,2,4-oxadiazol-5-yl)-5-(l -(1 -methylpiperidin-4-yl)- 1H- pyrazol-4-yl)pyridin-2-amine;
3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(l-ethylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine;
5-(l-(l-ethylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-
2-amine; or
3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(l-dodecylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine. In yet other more specific embodiments of Formula (IA5), each of R3 and R4 is hydrogen, R1 is aryl or more specifically, phenyl or substituted phenyl, R2 is hydrogen, tetrahydropyranyl or cyclohexyl.
In a specific embodiment, the compound of Formula (IA5) is:
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-2-amine; 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5- yl)pyridin-2-amine; ;
3-(3-phenyl-l,2,4-oxadiazol-5-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(3-(2,6-dichlorophenyl)- 1 ,2,4-oxadiazol-5-yl)-5-(l -(tetrahydro-2H-pyran-4-yl)- 1H- pyrazol-4-yl)pyridin-2-amine; or
3-(3-phenyl- 1 ,2,4-oxadiazol-5-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine. .
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA6)
Figure imgf000035_0001
Formula (IA6)
wherein, R1, R2, R3, and R4 are as defined above.
In a more specific embodiment of Formula (IA6), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
In a more specific embodiment of Formula (IA6), R2 is heterocyclyl.
More specifically, R2 is N-heterocyclyl.
In more specific embodiments of Formula (IA6), R2 is piperidinyl or substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
In other more specific embodiments of Formula (IA6), R2 is hydrogen. In certain specific embodiments of Formula (IA6), each of R3 and R4 is hydrogen, R1 is aryl, R2 is heterocyclyl, including N-heterocyclyl.
In yet other more specific embodiments of Formula (IA6), each of R3 and R4 is hydrogen, R1 is aryl or more specifically, phenyl or substituted phenyl, R2 is hydrogen, piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
In a specific embodiment, the compound of Formula (IA6) is:
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenyl-l,2,4-oxadiazol-3-yl)pyridin-2-amine; 3-(5-phenyl-l,2,4-oxadiazol-3-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(5-phenyl-l,2,4-oxadiazol-3-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-phenyl-l,2,4-oxadiazol-3- yl)pyridin-2-amine;
3-(5-(2,5-dichlorophenyl)-l,2,4-oxadiazol-3-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3 -(5 -phenyl- 1 ,2,4-oxadiazol-3 -yl)-5 -( 1 H-pyrazol-4-yl)pyridin-2-amine.
Of the compounds of Formula (IA), another embodiment provides compounds of Formula (IA7)
Figure imgf000036_0001
Formula (IA7)
wherein, R1, R2, R3, and R4 are as defined above.In a more specific embodiment of Formula (IA7), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
In a more specific embodiment of Formula (IA7), R2 is heterocyclyl. More specifically, R2 is piperidinyl or substituted piperidinyl.
In a specific embodiment, the compound of Formula (IA7) is:
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrrol-3- yl)pyridin-2-amine.
Of the compounds of Formula (I), an embodiment provides compound of
Formula (IB):
Figure imgf000036_0002
Formula (IB)
wherein,
A, A1, A2,
Figure imgf000036_0003
R3, R4, and R5 are as defined above,
L is heteroaryl or heterocycle; R is hydrogen, alkyl, alkoxy, or halo;
X is -0-, -NR8-, or -C(R9)2-;
R8 is hydrogen, alkyl, or -C(0)R10-;
each R9 is independently hydrogen, alkyl, alkoxy, or halo; and
R10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
Of the compounds of Formula (IB), an embodiment provides compound of Formula (IBl):
Figure imgf000037_0001
Formula (IBl)
wherein,
each A, A1 and A2 is the same or different and independently -N=, -
CR5=, or -0-;
R1 is aryl, heteroaryl, cycloalkyl, heterocyclyl, aralkyl, or cycloalkylalkyl;
each of R3, R4 and R7 is hydrogen,
R8 is hydrogen, alkyl, or -C(0)R10-; and
R10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
In a more specific embodiment of Formula (IBl), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
In another more specific embodiment of Formula (IBl), R1 is heteroaryl. More specifically, R1 is a pyridinyl, substituted pyridinyl, pyrazinyl, substituted pyrazinyl, thiazolyl, substituted thiazolyl.
In further specific embodiment of Formula (IBl), R1 is aralkyl. More specifically, R1 is benzyl.
In further specific embodiment of Formula (IBl), R1 is cycloalkylalkyl. More specifically, R1 is cyclopropylalkyl.
Of the compounds of Formula (IB), an embodiment provides compound of Formula (IB2):
Figure imgf000038_0001
Formula (IB2)
wherein,
each A, A1 and A2 is the same or different and independently -N=, - CR5=, or -0-;
R1 is aryl, heteroaryl, cycloalkyl, or heterocyclyl; and
each of R3, R4 and R7 is hydrogen.
In a more specific embodiment of Formula (IB2), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
Of the compounds of Formula (IB), an embodiment provides compound of Formula (IB3):
Figure imgf000038_0002
Formula (IB 3)
wherein,
each A, A1 and A2 is the same or different and independently -N=, -
CR5=, or -0-;
R1 is aryl, heteroaryl, cycloalkyl, or heterocyclyl; and
each of R3, R4, R7 and R9 is hydrogen;
In a more specific embodiment of Formula (IB3), R1 is aryl. More specifically, R1 is a phenyl or substituted phenyl.
Utility and Testing of the Compounds of the Disclosure
The present disclosure relates to compounds, pharmaceutical compositions and methods of using the compounds and pharmaceutical compositions for the treatment and/or prevention of diseases and conditions mediated by the kinase activity of Tyro3, Axl or Mer individually or by any combination of them, preferably diseases and conditions related to characterized by angiogenesis and/or cell proliferation and migration, and especially a disease and condition related to cancer, inflammatory diseases, autoimmune diseases, neurodisorders, and the like, by administering an effective amount of a compound of the disclosure.
The compounds of the disclosure modulate, preferably inhibit, the activity of human Tyro3, Axl or Mer individually or by any combination of them.
The general value of the compounds of the disclosure in modulating, especially inhibiting, the activity of Tyro3, Axl or Mer individually or by any combination of them can be determined using the assay described below in Example 21.
The compounds of the instant disclosure are inhibitors of Tyro3, Axl or Mer individually or inhibitors of any combination of them and are useful for treating diseases and disorders in humans and other organisms, including all those human diseases and disorders which are the result of abnormal kinase activity of Tyro3, Axl or Mer individually or any combination of them or which may be ameliorated by modulation of the kinase activity of Tyro3, Axl or Mer individually or any combination of them.
As defined herein, a disease or condition mediated by the abnormal kinase activity of Tyro3, Axl or Mer individually or any combination of them is defined as any disease or condition in which the activity of Tyro3, Axl or Mer individually or any combination of them is elevated and/or where inhibition of the activity of Tyro3, Axl or Mer individually or any combination of them can be demonstrated to bring about symptomatic improvements for the individual so treated. As defined herein, a disease or condition mediated by the abnormal activity of Tyro3, Axl or Mer individually or any combination of them includes, but is not limited to, a disease or condition which is, or is related to cancer, inflammatory diseases, autoimmune diseases, and neurodisorders. For purposes of this disclosure, Diseases and conditions which are alleviated by the modulation of the activity of Tyro3, Axl or Mer individually or any combination of them include, but are not limited to, solid tumors, including, but not limited to, breast, renal, endometrial, ovarian, thyroid, and non-small cell lung carcinoma, melanoma, prostate carcinoma, sarcoma, gastric cancer and uveal melanoma; liquid tumors, including but not limited to, leukemias particularly myeloid leukemias and lymphomas; endometriosis, vascular disease/injury including, but not limited to, restenosis, atherosclerosis and thrombosis, psoriasis; visual impairment due to macular
degeneration; diabetic retinopathy and retinopathy of prematurity; kidney disease including, but not limited to, glomerulonephritis, diabetic nephropathy and renal transplant rejection, rheumatoid arthritis; osteoarthritis, osteoporosis and cataracts.
In addition to the foregoing, the compounds of the disclosure are useful in treating diseases and conditions which are affected by the following biological processes: Invasion, migration, metastasis, or drug resistance as manifested in cancer; stem cell biology as manifested in cancer; invasion, migration, adhesion, or
angiogenesis as manifested in endometriosis; vascular remodeling as manifested in cardiovascular disease, hypertension or vascular injury; bone homeostatasis as manifested in osteoporosis or osteoarthritis; viral infection as manifested, for example, in ebola virus infection; or differentiation as manifested in obesity. The compounds of the disclosure may also be used to modulate inflammatory processes by treating sepsis, acting as vaccine adjuvants, and/or potentiating the immune response in immunocompromised patients.
The following animal models provide guidance to one of ordinary skill in the art in testing the compounds of the disclosure for their use in treating the disease or condition indicated.
The compounds of the disclosure may be used or tested for their use in treating leukemias and lymphomas by, respectively, administering a pharmaceutically effective amount or testing the compounds in the xenograft in SCID mouse model using human cancer cell lines which express Tyro3 or Axl or Mer or co-expressing any combination of these three kinases including, but not limited to, A549, K562, HeLa, MDA-MB-231, SK-OV-3, OVCAR-8, DU145, H1299, ACHN, A498 and Caki-1.
The compounds of the disclosure may be tested for their use in treating leukemias in the xenograft in SCID or nu/nu mouse model using human AML and CML leukemia cell lines.
The compounds of the disclosure may be used or tested for their use in treating endometriosis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the syngenic mouse model of
endometriosis (see Somigliana, E. et al., "Endometrial ability to implant in ectopic sites can be prevented by interleukin-12 in a murine model of endometriosis", Hum. Reprod. 1999, 14(12), 2944-2950). The compounds may also be tested for their use in treating endometriosis by using the rat model of endometriosis (see Lebovic, D.I. et al, "Peroxisome proliferator-activated receptor-gamma induces regression of endometrial explants in a rat model of endometriosis", Fertil. SteriL, 2004, 82 Suppl 3, 1008-1013).
The compounds of the disclosure may be used or tested for their use in treating restenosis by, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or using the balloon-injured rate carotid artery model (see Kim, D.W. et al., "Novel oral formulation of paclitaxel inhibits neointimal hyperplasia in a rat carotid artery injury model", Circulation, 2004, 109(12), 1558- 1563). The compounds of the disclosure may also be tested for their use in treating restenosis by using the percutaneous transluminal coronary angioplasty in apoE deficient mouse model (see von der Thusen, J. H. et al, "Adenoviral transfer of endothelial nitric oxide synthase attenuates lesion formation in a novel murine model of postangioplasty restenosis", Arterioscler. Thromb. Vase. Biol, 2004, 24(2), 357-362).
The compounds of the disclosure may be used or tested for their use in treating atherosclerosis/thrombosis, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or in the ApoE deficient mouse model (see Nakashima, Y. et al., "ApoE-deficient mice develop lesions of all phases of
atherosclerosis throughout the arterial tree", Arterioscler. Thromb., 1994, 14(1), 133- 140).
The compounds of the disclosure may also be used or tested for their use in treating thrombosis, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or using the collagen-epinephrine-induced pulmonary thromboembolism model and the stasis induced venous thrombosis model (see Angelillo-Scherrer A. et al, "Role of Gas6 receptors in platelet signaling during thrombus stabilization and implications for antithrombotic therapy", J. Clin. Invest., 2005,115, 237-246).
The compounds of the disclosure may be used or tested for their use in treating psoriasis by, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or using the SCID mouse model or the human skin model of psoriasis (see Nickoloff, B.J. et al, "Severe combined immunodeficiency mouse and human psoriatic skin chimeras. Validation of a new animal model", Am. J. Pathol, 1995, 146(3), 580-588).
The compounds of the disclosure may be used or tested for their use in treating age- related macular degeneration or diabetic retinopathy by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the rat corneal angiogenesis model (see Sarayba MA, Li L, Tungsiripat T, Liu NH, Sweet PM, Patel AJ, Osann KE, Chittiboyina A, Benson SC, Pershadsingh HA, Chuck RS. Inhibition of corneal neovascularization by a peroxisome proliferator-activated receptor-gamma ligand. Exp. Eye. Res., 2005, 80(3), 435-442) or the laser-induced mouse choroidal neovasculation model (see Bora, P. S., et al., "Immunotherapy for choroidal neovascularization in a laser-induced mouse model simulating exudative (wet) macular degeneration", Proc. Natl. Acad. Sci. U. S. A., 2003, 100(5), 2679-2684).
The compounds of the disclosure may be used or tested for their use in treating retinopathy of prematurity, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or in the mouse retinopathy of prematurity model (see Smith, L.E. et al, "Oxygen-induced retinopathy in the mouse", Invest. Ophthalmol. Vis. Sci., 1994, 35(1), 101-111). The compounds of the disclosure may be tested for their use in treating glomerulonephritis or diabetic nephropathy in the rat anti- Thyl .1 -induced experimental mesengial proliferative glomerulonephritis model (see Smith, L. E. et al. cited above).
The compounds of the disclosure may be used or tested for their use in treating renal transplant rejection, respectively, by administering a pharmaceutically effective amount to a subject in need thereof or by using a rat model of chronic renal transplant rejection (see Yin, J. L. et al, "Expression of growth arrest-specific gene 6 and its receptors in a rat model of chronic renal transplant rejection", Transplantation, 2002, 73(4), 657-660).
The compounds of the disclosure may be used or tested for their use in treating rheumatoid arthritis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the CAIA mouse model (see Phadke, K. et al., "Evaluation of the effects of various anti-arthritic drugs on type II collagen-induced mouse arthritis model", lmmunopharmacology, 1985, 10(1), 51-60).
The compounds of the disclosure may be used or tested for their use in treating osteoarthritis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the STR/ORT mouse model (see Brewster, M. et al., "Ro 32-3555, an orally active collagenase selective inhibitor, prevents structural damage in the STR/ORT mouse model of osteoarthritis", Arthritis. Rheum., 1998, 41(9), 1639-1644). The compounds of the disclosure may be used or tested for their use in treating osteoporosis by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the ovariectomized rat model (see Wronski, TJ. et al., "Endocrine and pharmacological suppressors of bone turnover protect against osteopenia in ovariectomized rats", Endocrinology, 1989, 125(2), 810-816) or the ovariectomized mouse model (see Alexander, J. M. et al., "Human parathyroid hormone 1-34 reverses bone loss in ovariectomized mice", J Bone Miner Res., 2001, 16(9), 1665-1673; Fujioka, M. et al, "Equol, a metabolite of daidzein, inhibits bone loss in ovariectomized mice", J. Nut., 2004, 134(10), 2623-2627).
The compounds of the disclosure may be used or tested for their use in treating cataracts by, respectively, administering a pharmaceutically effective amount to a subject in need thereof or using the H202-induced model (see Kadoya, K. et al, "Role of calpain in hydrogen peroxide induced cataract", Curr. Eye Res., 1993, 12(4), 341- 346) or the Emory mouse model (see Sheets, N. L. et al, "Cataract- and lens-specific up-regulation of ARK receptor tyrosine kinase in Emory mouse cataract", Invest.
Ophthalmol. Vis. Sci., 2002, 43(6), 1870-1875).
Typically, a successful inhibitory therapeutic agent of the activity of Tyro3, Axl or Mer individually or any combination of them will meet some or all of the following criteria. Oral availability should be at or above 20% Animal model efficacy is less than about 20 mg/Kg, 2 mg/Kg, 1 mg/Kg, or 0.5 mg/Kg and the target human dose is between 10 and 250 mg/70 Kg, although doses outside of this range may be acceptable. ("mg/Kg" means milligrams of compound per kilogram of body mass of the subject to whom it is being administered). The required dosage should preferably be no more than about once or twice a day or at meal times. The therapeutic index (or ratio of toxic dose to therapeutic dose) should be greater than 10. The IC50 ("Inhibitory
Concentration - 50%") is a measure of the amount of compound required to achieve 50% inhibition of the kinase activity, over a specific time period, in a kinase activity assay. Any process for measuring the kinase activity of Tyro3, Axl or Mer, preferably human Tyro3, Axl or Mer, may be utilized to assay the activity of the compounds useful in the methods of the disclosure in inhibiting said Tyro3, Axl or Mer activity. Compounds of the disclosure demonstrate an IC50 in a 15 to 60 minute recombinant human kinase assay of preferably less than 10 mM, less than 5 μΜ, less than 2.5 μΜ, less than 1 μΜ. less than 750 nM, less than 500 nM, less than 250 nM, less than 100 nM, less than 50 nM, and most preferably less than 20 nM. Compounds of the disclosure may show reversible inhibition (i.e. competitive inhibition) or irreversible inhibition and preferably do not inhibit other protein kinases.
The identification of compounds of the disclosure as Tyro3, Axl or Mer inhibitors was readily accomplished using the recombinant human Tyro3, Axl and Mer proteins and employing the 33P-radiolabeled phosphate transfer assay for which the procedure is known to someone skilled in the art or as described in Example 18. When tested in this assay, compounds of the disclosure had greater than 50% inhibitory activity at 10 μΜ concentration of the test compound, preferably greater than 60% inhibitory activity at 10 μΜ concentration of the test compound, more preferably greater than 70% inhibitory activity at 10 μΜ concentration of the test compound, and even more preferably greater than 80% inhibitory activity at 10 μΜ concentration of the test compound, and the most preferably greater than 90% inhibitory activity at 10 μΜ concentration of the test compound, thereby demonstrating that the compounds of the disclosure are potent inhibitors of the kinase activity of Tyro3, Axl and Mer.
These results provide the basis for analysis of the structure-activity relationship (SAR) between test compounds and the kinase activity of Tyro3, Axl and Mer. Certain-groups tend to provide more potent inhibitory compounds. SAR analysis is one of the tools those skilled in the art may employ to identify preferred
embodiments of the compounds of the disclosure for use as therapeutic agents. Other methods of testing the compounds disclosed herein are also readily available to those skilled in the art. Thus, in addition, the determination of the ability of a compound to inhibit Tyro3, Axl and Mer activity may be accomplished in vivo. In one such embodiment this is accomplished by administering said chemical agent to an animal afflicted with a certain tumor graft model and subsequently detecting a change in tumor growth rate in said animal thereby identifying a therapeutic agent useful in treating the said tumors. In such embodiment, the animal may be a human, such as a human patient afflicted with such a disorder and in need of treatment of said disorder.
In specific embodiments of such in vivo processes, said change in Tyro3, Axl or Mer activity in said animal is a decrease in activity, preferably wherein said
Tyro3, Axl or Mer inhibiting agent does not substantially inhibit the biological activity of other kinases. The compounds of the disclosure can be used in combination with other therapeutic agents. Examples of alkylating agents that can be carried out in combination with include, but not limited to, fluorouracil (5-FU) alone or in further combination with leukovorin; other pyrimidine analogs such as UFT, capecitabine, gemcitabine and cytarabine, the alkyl sulfonates, e.g., busulfan (used in the treatment of chronic granulocytic leukemia), improsulfan and piposulfan; aziridines, e.g., benzodepa, carboquone, meturedepa and uredepa; ethyleneimines and methylmelamines, e.g., altretamine, triethylenemelamine, triethylenephosphoramide,
triethylenethiophosphoramide and trimethylolmelamine; and the nitrogen mustards, e.g., chlorambucil (used in the treatment of chronic lymphocytic leukemia, primary macroglobulinemia and non-Hodgkin's lymphoma), cyclophosphamide (used in the treatment of Hodgkin's disease, multiple myeloma, neuroblastoma, breast cancer, ovarian cancer, lung cancer, Wilm' s tumor and rhabdomyosarcoma), estramustine, ifosfamide, novembrichin, prednimustine and uracil mustard (used in the treatment of primary thrombocytosis, non-Hodgkin's lymphoma, Hodgkin's disease and ovarian cancer); and triazines, e.g., dacarbazine (used in the treatment of soft tissue sarcoma).
Examples of antimetabolite chemotherapeutic agents that can be carried out in combination with include, but not limited to, folic acid analogs, e.g.,
methotrexate (used in the treatment of acute lymphocytic leukemia, choriocarcinoma, mycosis fungiodes, breast cancer, head and neck cancer and osteogenic sarcoma) and pteropterin; and the purine analogs such as mercaptopurine and thioguanine which find use in the treatment of acute granulocytic, acute lymphocytic and chronic granulocytic leukemias. Examples of natural product-based chemotherapeutic agents that can be carried out in combination with include, but not limited to, the vinca alkaloids, e.g., vinblastine (used in the treatment of breast and testicular cancer), vincristine and vindesine; the epipodophyllotoxins, e.g., etoposide and teniposide, both of which are useful in the treatment of testicular cancer and Kaposi's sarcoma; the antibiotic chemotherapeutic agents, e.g., daunorubicin, doxorubicin, epirubicin, mitomycin (used to treat stomach, cervix, colon, breast, bladder and pancreatic cancer), dactinomycin, temozolomide, plicamycin, bleomycin (used in the treatment of skin, esophagus and genitourinary tract cancer); and the enzymatic chemotherapeutic agents such as L- asparaginase. Examples of other signal transduction inhibiting agents that can be carried out in combination with include, but not limited to, gefitinib, erlotinib, sorafenib, herceptin, imatinib, dasatinib, sunitinib, nilotinib, lapatinib, pazopanib, vandetanib, vemurafenib, crizotinib, ruxolitinib, axitinib, bosutinib, regorafenib, tofacitinib, cabozantinib, ponatinib, dabrafenib, trametinib, and afatinib.
Other agents can be used in combination with the compound of the disclosure include, but not limited to, COX-II inhibitors, such as, but not limited to, Vioxx, Celebrex (celecoxib), valdecoxib, paracoxib, rofecoxib; matrix
metalloproteinase inhibitors, such as, but not limited to, AG-3340, RO 32-3555, and RS 13-0830.
Pharmaceutical Compositions of the Disclosure and Administration
The present disclosure also relates to pharmaceutical composition containing the compounds of Formula (I), (IA), (IB) or substructures thereof disclosed herein. In one embodiment, the present disclosure relates to a composition comprising compounds of Formula (I), (IA), (IB) or substructures thereof in a pharmaceutically acceptable carrier and in an amount effective to modulate the activity of Tyro3, Axl and Mer individually or in any combination of them or to treat diseases related to angiogenesis and/or cell proliferation and migration, and especially cancer,
inflammatory diseases, autoimmune diseases, neurodisorders and the like when administered to an animal, preferably a mammal, most preferably a human patient. In an embodiment of such composition, the patient has hyperproliferative disease, and especially cancer, inflammatory diseases, autoimmune diseases, neurodisorders and the like, before administration of said compound of the disclosure and the compound of the disclosure is present in an amount effective to reduce said lipid level.
The pharmaceutical compositions useful herein also contain a pharmaceutically acceptable carrier, including any suitable diluent or excipient, which includes any pharmaceutical agent that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity. Pharmaceutically acceptable carriers include, but are not limited to, liquids, such as water, saline, glycerol and ethanol, and the like. A thorough discussion of pharmaceutically acceptable carriers, diluents, and other excipients is presented in REMINGTON'S PHARMACEUTICAL SCIENCES (Mack Pub. Co., N.J. current edition). Those skilled in the art are also familiar with determining administration methods (oral, intravenous, inhalation, sub-cutaneous, etc.), dosage forms, suitable pharmaceutical excipients and other matters relevant to the delivery of the compounds to a subject in need thereof.
In an alternative use of the disclosure, the compounds of the disclosure can be used in in vitro or in vivo studies as exemplary agents for comparative purposes to find other compounds also useful in treatment of, or protection from, the various diseases disclosed herein.
Administration of the compounds of the disclosure, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the disclosure can be prepared by combining a compound of the disclosure with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques. Pharmaceutical compositions of the disclosure are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the disclosure in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the disclosure, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this disclosure. A pharmaceutical composition of the disclosure may be in the form of a solid or liquid. In one aspect, the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form. The carrier(s) may be liquid, with the
compositions being, for example, an oral oil, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.
When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
As a solid composition for oral administration, the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition will typically contain one or more inert diluents or edible carriers. In addition, one or more of the following may be present: binders such as carboxymethylcellulose, ethyl cellulose,
microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
When the pharmaceutical composition is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.
The pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension. The liquid may be for oral administration or for delivery by injection, as two examples. When intended for oral administration, preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer. In a composition intended to be administered by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
The liquid pharmaceutical compositions of the disclosure, whether they be solutions, suspensions or other like form, may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Physiological saline is a preferred adjuvant. An injectable pharmaceutical composition is preferably sterile. A liquid pharmaceutical composition of the disclosure intended for either parenteral or oral administration should contain an amount of a compound of the disclosure such that a suitable dosage will be obtained. Typically, this amount is at least 0.01% of a compound of the disclosure in the composition. When intended for oral administration, this amount may be varied to be between 0.1 and about 70% of the weight of the composition. Preferred oral pharmaceutical compositions contain between about 4% and about 75% of the compound of the disclosure. Preferred pharmaceutical compositions and preparations according to the present disclosure are prepared so that a parenteral dosage unit contains between 0.01 to 10% by weight of the compound prior to dilution of the disclosure.
The pharmaceutical composition of the disclosure may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base. The base, for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers. Thickening agents may be present in a pharmaceutical composition for topical administration. If intended for transdermal administration, the composition may include a transdermal patch or iontophoresis device. Topical formulations may contain a concentration of the compound of the disclosure from about 0.1 to about 10%> w/v (weight per unit volume). The pharmaceutical composition of the disclosure may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug. The composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient. Such bases include, without limitation, lanolin, cocoa butter and polyethylene glycol. The pharmaceutical composition of the disclosure may include various materials, which modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating shell around the active ingredients. The materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents. Alternatively, the active ingredients may be encased in a gelatin capsule.
The pharmaceutical composition of the disclosure in solid or liquid form may include an agent that binds to the compound of the disclosure and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome. The pharmaceutical composition of the disclosure may consist of dosage units that can be administered as an aerosol. The term aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the disclosure may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One of ordinary skill in the art, without undue experimentation may determine preferred aerosols. The pharmaceutical compositions of the disclosure may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the disclosure with sterile, distilled water so as to form a solution. A surfactant may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with the compound of the disclosure so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
The compounds of the disclosure, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy. Generally, a therapeutically effective daily dose is (for a 70 kg mammal) from about 0.001 mg/kg (i.e., 0.7 mg) to about 100 mg/kg (i.e., 7.0 gm); preferably a therapeutically effective dose is (for a 70 kg mammal) from about 0.01 mg/kg (i.e., 7 mg) to about 50 mg/kg (i.e., 3.5 g); more preferably a therapeutically effective dose is (for a 70 kg mammal) from about 1 mg/kg (i.e., 70 mg) to about 25 mg/kg (i.e., 1.75 g).
Compounds of Formula (I), (IA), (IB) or substructures thereof, or pharmaceutically acceptable derivatives thereof, may also be administered
simultaneously with, prior to, or after administration of one or more other therapeutic agents. Such combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of the disclosure and one or more additional active agents, as well as administration of the compound of the disclosure and each active agent in its own separate pharmaceutical dosage formulation. For example, a compound of the disclosure and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations. Where separate dosage formulations are used, the compounds of the disclosure and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
Isotopic Enrichment of Compounds
Isotopic enrichment is a process by which the relative abundance of the isotopes of a given element are altered, thus producing a form of the element that has been enriched in one particular isotope and depleted in its other isotopic forms. Isotopic enrichment of a drug are used for the following applications: reducing or eliminating unwanted metabolites; increasing the half-life of the parent drug; decreasing the number of doses needed to achieve a desired effect; decreasing the amount of a dose necessary to achieve a desired effect; increasing the formation of active metabolites, if any are formed; and/or decreasing the production of deleterious metabolites in specific tissues and/or create a more effective drug and/or a safer drug for combination therapy, whether the combination therapy is intentional or not.
Replacement of an atom for one of its isotopes often will result in a change in the reaction rate of a chemical reaction. This phenomenon is known as the Kinetic Isotope Effect. For example, if a C-H bond is broken during a rate-determining step in a chemical reaction (i.e. the step with the highest transition state energy), substitution of a deuterium for that hydrogen will cause a decrease in the reaction rate and the process will slow down. This phenomenon is known as the Deuterium Kinetic Isotope Effect (Foster et. al., Adv. Drug Res., 1985, 14, 1-36; Kushner et. al, Can. J. Physiol. Pharmacol, 1999, 77, 79-88).
Improvement of metabolism, pharmacokinetics, pharmacodynamics, and toxicity profiles of pharmaceuticals by isotopic enrichment such as deuteration has been demonstrated by the following examples: Lijinsky et. al., J. Nat. Cancer Inst., 1982, 69, 1127-1133; Gately et. al, J. Nucl Med., 1986, 27, 388-394; Gordon et. al, Drug Metab. Dispos., 1987, 15, 589-594; Mangold et. al, Mutation Res., 1994, 308, 33-42; Zello et. al, Metabolism, 1994, 43, 487-491; Wade D., Chem. Biol. Interact., 1999, 117, 191-637.
Preparation of the Compounds of Formula (I), (IA) or (IB)
It is understood that in the following description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.
It will also be appreciated by those skilled in the art that in the process described below the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(0)-R" (where R" is alkyl, aryl or arylalkyl), /?-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
Protecting groups may be added or removed in accordance with standard techniques, which are well-known to those skilled in the art and as described herein.
The use of protecting groups is described in detail in Green, T.W. and P.G.M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley. The protecting group may also be a polymer resin such as a Wang resin or a 2-chlorotrityl- chloride resin.
It will also be appreciated by those skilled in the art, although such protected derivatives of compounds of this disclosure may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the disclosure which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". All prodrugs of compounds of this disclosure are included within the scope of the disclosure.
The following Reaction Schemes illustrate methods to make compounds of this disclosure. It is understood that one of those skilled in the art would be able to make these compounds by similar methods or by methods known to one skilled in the art. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, e.g., Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described in this disclosure.
As representative examples, compounds of formula (IA1), in which W is
Figure imgf000053_0001
(i.e., A is -0-, A1 is -C(R5)=; A2 is -N=) and R5 is hydrogen can be synthesized following the general procedure as described in Reaction Scheme 1.
REACTION SCHEME 1
Figure imgf000053_0002
105 Formula (IA1)
The starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein. In general, the compounds of the Formula (I) or (IA1) are prepared in the above reaction scheme as follows: The starting a-bromo ketone 101 is treated with hexamethylenetetramine under Delepine reaction conditions to afford the a -amino ketone 102 as a
hydrochloride salt. Under amide formation conditions, 2-amino-5-bromonicotinic acid 103 reacts with compound 102 in the presence of a coupling reagent, such as, but not limited to, EDCI to form compound 104. The oxazole compound 105 is obtained under acid cyclization conditions. This oxazole compound is coupled with the boron reagent 106 under the Suzuki coupling reaction conditions to afford the compound of formula (IA1) where W is
Figure imgf000054_0001
and R5 is hydrogen.
Compounds of formula (IA2), in which W is
Figure imgf000054_0002
(i.e., A is -C(R5)=; A1 is -0-, A2 is -N=) and R5 is hydrogen can be synthesized following the general procedure as described in Reaction Scheme 2.
Figure imgf000054_0003
205 Formula (IA2)
The starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein. In general, the compounds of the Formula (IA2) are prepared in the above reaction scheme as follows: The starting material, an appropriately substituted 2-amino-5- bromonicotinic aldehyde 201 reacts with hydroxylamine hydrochloride salt under reflux to afford the nicotinic aldoxime intermediate 202. This oxime compound is then treated with N-chlorosuccinimide to generate the chlorinated oxime compound 203 which is cyclized with acetylene compound 204 to afford the isoxazole compound 205. This isoxazole compound is coupled with the boron reagent 206 under the Suzuki coupling reaction conditions to afford the compound of formula (IA2).
Alternatively, compounds of formula (IA3), in which W is
Figure imgf000055_0001
(i.e., A is -0-, each of A1 and A2 is -C(R5)=) and each R5 is hydrogen, can be synthesized following the general procedure as described in Reaction Scheme 3.
Figure imgf000055_0002
304 Formula (IA3)
The starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein. In general, the compounds of the Formula (IA3) are prepared in the above reaction scheme as follows:
The starting a— ketoalkene 301 reacts with aldehyde compound 302 (e.g., an appropriately substituted 2-amino-5-bromonicotinic aldehyde) under the Stetter Reaction conditions in the presence of a thiazolium catalyst, such as, but not limited to, 3-benzyl-5-(2-hydroxyethyl)-4-methylthiazolium chloride, to afford 1,4-dione compound303, which is cyclized under the Paal-Knorr furan synthesis conditions to afford the furan compound 304. This furan compound is further coupled with the boron reagent 306 under the Suzuki coupling reaction conditions to afford the compound of formula (IA3).
In general, compounds of formula (IA4), in which W is
Figure imgf000056_0001
(i.e., A is -0-, each A1 and A2 is -N=) can be synthesized following the general procedure as described in Reaction Scheme 4.
REACTION SCHEME 4
Figure imgf000056_0002
404 Formula (IA4) The starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein. In general, the compounds of the disclosure are prepared in the above reaction scheme as follows:
The starting carboxylic acid 401 reacts with hydrazide 402 in the presence of a coupling reagent, such as, but not limited to, HATU and a base, such as, but not limited to, triethylamine to generate the 1 ,2-diacylhydrazine compound 403. Cyclodehydration of compound 403 in the presence of a dehydrating reagent, such as, but not limited to, triphenylphosphine/carbon tetrabromide to generate the 1,3,4- oxadiazole 404. This oxadiazole compound is coupled with the boron reagent 406 under the Suzuki coupling reaction conditions to afford the compound of Formula (IA4).
Compounds of formula (IA5) of this disclosure in which W is
N-O (i.e., A is -N=, A1 is -N=, A2 is -0-) can be synthesized following the general procedure as described in Reaction Scheme 5.
REACTION SCHEME 5
Figure imgf000057_0001
505 Formula (IA5) The starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein. In general, the compounds of the disclosure are prepared in the above reaction scheme as follows:
Nitrile compound 501 is treated with hydroxylamine hydrochloride in the presence of a base, such as, but not limited to, triethylamine under reflux to generate amidoxime compound 502. A condensation reaction of the amidoxime compound 602 with an appropriately substituted nicotinic acid 503 in the presence of a coupling agent, such as, but not limited to, EDCI generates compound 504. Under reflux in the presence of pyridine, compound 504 is cyclized to afford 1,2,4-oxadiazole compound 505. This oxadiazole compound is coupled with a boron reagent 506 under the Suzuki coupling reaction conditions to afford the compound of formula (IA5).
Compounds of Formula (IA6) of this disclosure in which W is
Figure imgf000057_0002
(i.e., A is -N=, A1 is -0-, A2 is -N=) can be synthesized following the general procedure as described in Reaction Scheme 6. REACTION SCHEME 6
Figure imgf000058_0001
605 Formula (IA6)
The starting materials for the above reaction scheme are commercially available or can be prepared according to methods known to one skilled in the art or by methods disclosed herein. In general, the compounds of the disclosure are prepared in the above reaction scheme as follows:
Nitrile compound 601 is treated with hydroxylamine hydrochloride in the presence of a base, such as, but not limited to, triethylamine under reflux to generate amidoxime compound 602. The condensation of amidoxime 602 with carboxlic acid 303 in the presence of a coupling agent, such as, but not limited to, EDCI generates compound 604. Under reflux in the presence of pyridine, compound 604 is cyclized to afford 1,2,4-oxadiazole compound 605. This oxadiazole compound is coupled with the boron reagent 606 under the Suzuki coupling reaction conditions to afford the compound of formula (I) of the disclosure.
More specific details on synthetic techniques for compounds of Formula (I), (IA) and (IB) and their substructures are provided herein. Unless otherwise provided, all reagents and reaction conditions employed in synthesis are known to those skilled in the art and are available from ordinary commercial sources. PREPARATIONS
PREPARATION 1
PREPARATION OF 2-AMINO-1-PHENYLETHANONE HYDROCHLORIDE
Figure imgf000059_0001
To a solution of 2-bromo-l-phenylethanone (5.00 g, 25.0 mmol) in 25 mL of chloroform was added a solution of hexamethylenetetramme (3.50 g, 25.0 mmol) in 15 mL of chloroform dropwise. The resulting mixture was stirred at ambient temperature for 1 hour to yield a white solid. The white solid collected by filtration was dissolved in 50 mL of methanol and 10.0 mL of concentrated hydrochloroic acid was added. The mixture was refluxed for 3 hours to afford white precipitates. The precipitates were collected by filtration and washed with tetrahydrofuran to afford 2- amino-l-phenylethanone hydrochloride as a white solid in 66% yield (2.80 g) which was used directly for the next step reaction without further purification. 1H NMR (400 MHz, DMSO- 6): δ 8.49 (br s, 3H), 8.00-7.96 (m, 2H), 7.72-7.66 (m, 1H), 7.56-7.50 (m, 2H), 4.54 (s, 2H).
PREPARATION 1.1
PREPARATION OF 2-AMINO-1-(4-CHLOROPHENYL)ETHANONE HYDROCHLORIDE
Figure imgf000059_0002
Following the procedure as described in Preparation 1 , making variations using 2-bromo-l-(4-chlorophenyl)ethanone to replace 2-bromo-l-phenylethanone, 2- amino-l-(4-chlorophenyl)ethanone hydrochloride was obtained as a white solid in a quantitative yield. 1H NMR (400 MHz, DMSO- 6): δ 8.52 (br s, 3H), 8.02-7.97 (m, 2H), 7.65-7.60 (m, 2H), 4.53 (s, 2H). PREPARATION 1.2
PREPARATION OF 2-AMINO-1-(3-CHLOROPHENYL)ETHANONE HYDROCHLORIDE
Figure imgf000060_0001
Following the procedure as described in Preparation 1 , making variations using 2-bromo-l-(3-chlorophenyl)ethanone to replace 2-bromo-l-phenylethanone, 2- amino-l-(3-chlorophenyl)ethanone hydrochloride was obtained as a white solid in 96% yield. 1H NMR (400 MHz, DMSO-d6): δ 8.54 (br s, 3H), 8.02-7.98 (m, 1H), 7.96-7.92 (m, 1H), 7.78-7.74 (m, 1H), 7.62-7.56 (m, 1H), 4.57 (s, 2H).
PREPARATION 2
PREPARATION OF 2-AMiNO-5-BROMO-N-(2-oxo-2-PHENYLETHYL)NicoTiNAMiDE
Figure imgf000060_0002
To a suspension of 2-amino-5-bromopyridine-3-carboxylic acid (0.43 g,
2.00 mmol) and 2-amino-l-phenylethanone hydrochloride (0.51 g, 3.00 mmol) in 40 mL of dichloromethane was added l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide (1.19 g, 6.00 mmol). The mixture was stirred at ambient temperature for 3 hours. After removal of the solvent in vacuo, the residue was purified by column chromatography eluted with 1 : 1 ethyl acetate :hexane to afford 2-amino-5-bromo-N-(2-oxo-2- phenylethyl)nicotinamide as a yellow solid in 72% yield (0.48 g). 1H NMR (400 MHz, CDCI3): δ 8.21 (d, J= 2.0 Hz, 1H), 8.13-8.02 (m, 2H), 8.03 (d, J= 2.0 Hz, 1H), 7.70- 7.65 (m, 1H), 7.58-7.52 (m, 2H), 7.24 (t, J= 4.4 Hz, 1H), 6.57 (br s, 2H), 4.91 (d, J = 4.4 Hz, 2H). PREPARATION 2.1
PREPARATION OF 2-AMiNO-5-BROMO-N-(2-(4-CHLOROPHENYL)-2- OXOETHYL)NICOTINAMIDE
Figure imgf000061_0001
Following the procedure as described in Preparation 2, making variations using 2-amino-l-(4-chlorophenyl)ethanone hydrochloride to replace 2-amino-l-phenyl- ethanone hydrochloride, 2-amino-5-bromo-N-(2-(4-chlorophenyl)-2-oxoethyl)- nicotinamide was obtained as a yellow solid in 55% yield. 1H NMR (400 MHz, CDC13): 5 8.12 (s, 1H), 8.11 (s, 1H), 7.90 (d, J= 6.8 Hz, 2H), 7.80 (d, J= 4.8 Hz, 1H), 7.44 (d, J = 6.8 Hz, 2H), 6.58 (br s, 2H), 4.77 (d, J= 4.8 Hz, 2H).
PREPARATION 3
PREPARATION OF 5-BR 2-YL)PYRIDIN-2-AMINE
Figure imgf000061_0002
A mixture of 2-amino-5-bromo-N-(2-oxo-2-phenylethyl)nicotinamide (450 mg, 1.35 mmol) in 10 mL of concentrated sulfuric acid was stirred at ambient temperature for 18 hours. The mixture was mixed with 20 mL of ice cold water and neutralized with aqueous ammonia to yield yellow precipitates. After suction filtration, the yellow precipitates were washed with water to afford 5-bromo-3-(5-phenyloxazol-2- yl)pyridin-2-amine as a yellowish solid in 88% yield (375 mg). 1H NMR (400 MHz,
CD3OD): δ 8.28 (s, 1H), 8.18 (s, 1H), 7.74-7.70 (m, 2H), 7.54-7.34 (m, 4H), 6.85 (br s, 2H).
PREPARATION 3.1
PREPARATION OF 5 -BROMO OL-2-YL)PYRIDIN-2-AMINE
Figure imgf000061_0003
Following the procedure as described in Preparation 3, making variations using 2-amino-5-bromo-N-(2-(4-chlorophenyl)-2-oxoethyl)nicotinamide to replace 2- amino-5-bromo-N-(2-oxo-2-phenylethyl)nicotinamide, 5-bromo-3-(5-(4-chlorophenyl)- oxazol-2-yl)pyridin-2-amine was obtained as a yellow solid in 86%. 1H NMR (400 MHz, CDCls): δ 8.24 (s, 1H), 8.19 (s, 1H), 7.65 (d, J= 8.0 Hz, 2H), 7.50-7.38 (m, 3H), 6.75 (br s, 2H).
PREPARATION 4
PREPARATION OF 5 -BROMO-3 ZOL-2-YL)PYRIDIN-2-AMINE
Figure imgf000062_0001
To a suspension of 2-amino-5-bromopyridine-3-carboxylic acid (1.75 g,
8.09 mmol) and 2-amino-l-(3-chlorophenyl)ethanone hydrochloride (2.50 g, 12.1 mmol) in 100 mL of dichloromethane was added l-ethyl-3-(3- dimethylaminopropyl)carbodiimide (3.19 g, 16.2 mmol). The mixture was stirred at ambient temperature overnight to yield yellow precipitates. After filtration, the yellow precipitates were washed with dichloromethane to afford a yellow solid which was mixed with 10 mL of concentrated sulfuric acid and was stirred at ambient temperature for 20 hours. The reaction mixture was mixed with 40 mL of ice cold water and neutralized with ammonia solution to yield yellow precipitates. After suction filtration, the yellow precipitates were washed with water to afford 5 -bromo-3 -(5 -(3- chlorophenyl)oxazol-2-yl)pyridin-2-amine as an orange solid in 77% (2.00 g). 1H NMR (400 MHz, CDCI3): δ 8.27 (d, J = 2.4 Hz, 1H), 8.19 (d, J= 2.4 Hz, 1H), 7.71 (s, 1H), 7.65-7.57 (m, 1H), 7.55-7.45 (m, 1H), 7.45-7.33 (m, 2H), 6.78 (br s, 2H).
PREPARATION 5
PREPARATION OF 5-BRO -3-YL)PYRIDIN-2 -AMINE
Figure imgf000062_0002
A. To a solution of 2-aminopyridine-3-carbaldehyde (10.1 g, 81.0 mmol) in acetonitrile (150 mL) was added N-bromosuccinimide (15.1 g, 84.0 mmol). The resulting mixture was stirred at reflux for 2 hours. After cooled to room temperature, the reaction mixture was filtered. The collected solid was washed with methanol, and dried in vacuo to afford 2-amino-5-bromopyridine-3-carbaldehyde as a brown solid in 79% yield (12.9 g). 1H NMR (400 MHz, CDC13): δ 9.81 (s, 1H), 8.30 (d, J= 2.4 Hz, 1H), 7.89 (d, J= 2.4 Hz, 1H), 6.78 (br s, 2H).
B. A mixture of 2-amino-5-bromopyridine-3-carbaldehyde (8.90 g,
44.3 mmol), hydroxylamine hydrochloride (3.72 g, 53.1 mmol) and sodium acetate (4.36 g, 53.1 mmol) in 95% ethanol (150 mL) was stirred under reflux for 3 hours. After cooled to room temperature, the reaction mixture was concentrated and the residue was re-dissolved in ethyl acetate (500 mL), washed with brine (2 x 300 mL). The aqueous phase was back- washed with ethyl acetate (2 x 250 mL). The combined organic layer was washed with brine (2 x 250 mL), dried and filtered. The solvent of the filtrate was removed in vacuo, and the residue was dried in vacuo to afford 2-amino-5- bromopyridine-3-carbaldehyde oxime as a yellow solid in 90% yield (8.60 g). 1H NMR (400 MHz, CDC13): δ 11.4 (s, 1H), 8.19 (s, 1H), 7.98 (d, J= 2.4 Hz, 1H), 7.77 (d, J = 2.4 Hz, 1H), 7.10 (br s, 2H).
C. To a suspension of 2-amino-5-bromopyridine-3-carbaldehyde oxime (4.32 g, 20.0 mmol) was added N-chlorosuccinimide (2.99 g, 22.0 mmol). The mixture was stirred at 50 °C overnight and then cooled to room temperature, and followed by the addition of dichloromethane (100 mL), phenylacetylene (2.24 mL, 20.0 mmol) and triethylamine (4.20 mL, 30.0 mmol). The resulting mixture was stirred at room temperature overnight, then diluted with ethyl acetate (500 mL), washed with brine (4 x 300 mL). The aqueous phase was back-washed with ethyl acetate (1 x 300 mL). The combined organic layer was washed with brine (3 x 150 mL), dried and filtered. The solvent of the filtrate was removed in vacuo and the residue was purified by column chromatography eluted with hexanes: ethyl acetate (from 10: 1 to 5: 1) to afford 5-bromo-3-(5-phenylisoxazol-3-yl)pyridin-2-amine as a yellow solid in 6% yield (366 mg). 1H NMR (400 MHz, CDC13): δ 8.19 (d, J= 2.4 Hz, 1H), 7.89 (d, J= 2.4 Hz, 1H), 7.87-7.82 (m, 2H), 7.55-7.48 (m, 3H), 6.87 (s, 1H), 6.40-6.25 (br s, 2H). PREPARATION 6
PREPARATION OF 5-BR -YL)PYRIDIN-2-AMINE
Figure imgf000064_0001
A. A mixture of 3-chloro-l-phenylpropan-l-one (2.00 g, 11.9 mmol) and triethylamine (1.44 g, 14.2 mmol) in chloroform was stirred at room temperature overnight. The reaction mixture was diluted with dichloromethane, washed with 0.1 M hydrochloric acid solution (2 x 20 mL) and brine (20 mL). The organic solution was dried over anhydrous sodium sulfate and filtered. The filtrate was dried in vacuo to afford l-phenylprop-2-en-l-one as a yellow liquid in 91% yield (1.42 g). 1H NMR (400
MHz, CDCls): δ 7.97-7.92 (m, 2H), 7.61-7.54 (m, 1H), 7.51-7.44 (m, 2H), 7.16 (dd, J 17.2, 10.8 Hz, 1H), 6.44 (dd, J= 17.2, 1.6 Hz, 1H), 5.93 (dd, J= 10.8, 1.6 Hz, 1H).
B. A mixture of l-phenylprop-2-en-l-one (1.40 g, 10.6 mmol), 2- amino-5-bromopyridine-3-carbaldehyde (1.70 g, 8.48 mmol) and triethylamine in 5 mL of dioxane was heated at 100 °C for 10 minutes, followed by the addition of 3-benzyl-5- (2-hydroxyethyl)-4-methylthiazolium chloride (0.49 g, 1.59 mmol) in 20 mL of dioxane dropwise in 0.5 hour. The resulting mixture was heated at 100 °C overnight, then concentrated. The residue was purified by column chromatography eluted with 10% ethyl acetate in hexane to afford l-(2-amino-5-bromopyridin-3-yl)-4-phenylbutane-l,4- dione as a yellow solid in 13% yield (300 mg). 1H NMR (400 MHz, CDC13): δ 8.25 (d,
J= 2.4 Hz, 1H), 8.21 (d, J= 2.4 Hz, 1H), 8.08-8.02 (m, 2H), 7.62-7.56 (m, 1H), 7.52- 7.46 (m, 2H), 3.47-3.36 (m, 4H).
C. A mixture of l-(2-amino-5-bromopyridin-3-yl)-4-phenylbutane-
1,4-dione (300 mg, 0.900 mmol) and /?-toluenesulfonic acid (0.860 g, 4.50 mmol) in 10 mL of toluene was heated at 100 °C for 2 hours, then concentrated. The residue was neutralized with saturated sodium bicarbonate, then extracted with dichloromethane (3 x 20 mL). The organic solution was dried over anhydrous sodium sulfate and filtered. The solvent of the filtrate was removed in vacuo and the residue was purified by column chromatography eluted with 20% ethyl acetate in hexane to afford 5-bromo-3- (5-phenylfuran-2-yl)pyridin-2-amine as a yellow solid in 48% yield (136 mg). 1H NMR
(400 MHz, CDCI3): δ 8.08 (d, J= 2.4 Hz, 1H), 7.89 (d, J= 2.4 Hz, 1H), 7.73-7.68 (1 2H), 7.47-7.41 (m, 2H), 7.35-7.28 (m, 1H), 6.80 (d, J= 3.6 Hz, 1H), 6.76 (d, J= 3.6 Hz, 1H), 5.23 (br s, 2H).
PREPARATION 7
PREPARATION OF 5-BROMO OL-2-YL)PYRIDIN-2-AMINE
Figure imgf000065_0001
A. To a solution of l-(pyridin-3-yl)ethanone (10.0 g, 82.4 mmol) in 100 mL of acetic acid was added 48% (w/v) hydrobromic acid in acetic acid (21.8 mL, 123.6 mmol), followed by the addition of bromine dropwise. The resulting mixture was heated at 60 °C for 2 hours to yield white precipitates. After filtration, the solid was washed with ether to afford a white solid which was mixed with 200 mL of acetonitrile and sodium diformylamide (16.9 g, 178 mmol) was added. The mixture was heated at 60 °C for 3 hours. After removal of solvent, the residue was purified by column chromatography to afford N-formyl-N-(2-oxo-2-(pyridin-3-yl)ethyl)formamide as a yellowish wax in 23% yield (1.6 g). 1H NMR (400 MHz, CDC13): δ 9.23 (m, 1H), 9.05 (s, 2H), 8.89-8.83 (m, 1H), 8.27-8.22 (m, 1H), 7.52-7.45 (m, 1H), 5.10 (s, 2H).
B. To a solution of of N-formyl-N-(2-oxo-2-(pyridin-3- yl)ethyl)formamide (1.60 g, 8.33 mmol) in 150 mL of ethanol was added 10.0 mL of concentrated hydrochloric acid. The mixture was heated at 50 °C overnight to yield white precipitates which were collected by filtration and washed with ice cold ethanol to afford 2-amino-l-(pyridin-3-yl)ethanone hydrochloride as a white solid in 81% yield (1.17 g). 1H NMR (400 MHz, DMSO-d6): δ 9.25-9.22 (m, 1H), 8.93-8.90 (m, 1H), 8.62- 8.53 (br s, 3H), 8.63-8.49 (m, 1H), 7.78-7.75 (m, 1H), 4.65-4.58 (m, 2H).
C. To a suspension of 2-amino-5-bromopyridine-3-carboxylic acid (0.566 g, 2.60 mmol) and 2-amino-l-(pyridin-3-yl)ethanone hydrochloride (0.500 g, 2.90 mmol) in 40 mL of dichloromethane was added HATU (1.97 g, 5.20 mmol), followed by the addition of N,N-diisopropylethylamine (1.00 mL, 5.80 mmol). The mixture was stirred at room temperature overnight to yield white precipitates. After filtration, the white precipitates were washed with water to afford 2-amino-5-bromo-N- (2-oxo-2-(pyridin-3-yl)ethyl)pyridine-3-carboxamide as a white solid in 69%> yield (0.60 g). 1H NMR (400 MHz, DMSO-d6): 5 9.17-9.14 (m, 1H), 9.00 (t, J = 5.6 Hz, 1H), 8.82-8.78 (m, 1H), 8.35-8.28 (m, 1H), 8.18-8.13 (m, 2H), 7.60-7.53 (m, 1H), 7.21 (br s, 2H), 4.74 (d, J = 5.6 Hz, 2H).
D. To a cold (0 °C) suspension of 2-amino-5-bromo-N-(2-oxo-2- (pyridin-3-yl)ethyl)pyridine-3-carboxamide (0.56 g, 1.67 mmol) in 25 mL of dichloromethane was added triethylamine (1.14 mL, 8.35 mmol), followed by the addition of triphenylphosphine (0.55 g, 2.09 mmol) and tetrabromomethane (0.69 g, 2.09 mmol). The resulting mixture was stirred at room temperature for 5 hours. After removal of solvent, the residue was purified by column chromatography eluted with 3:2 ethyl acetate: dichloromethane to afford a yellow solid which was further purified by recrystallization in methanol to afford 5-bromo-3-(5-(pyridin-3-yl)oxazol-2-yl)pyridin- 2-amine as a light yellow solid in 35% yield (0.185 g). 1H NMR (400 MHz, CDC13): δ 9.02-8.98 (m, 1H), 8.64-8.60 (m, 1H), 8.27 (d, J = 2.4 Hz, 1H), 8.21 (d, J = 2.4 Hz, 1H), 8.04-7.99 (m, 1H), 7.56 (s, 1H), 7.44-7.40 (m, 1H), 6.75 (br s, 2H).
PREPARATION 8
PREPARATION OF 2-AMINO-5 -BROMONICOTINOHYDRAZIDE
Figure imgf000066_0001
1. To a suspension of 2-aminonicotinic acid 1 (13.8 g, 100 mmol) in 100 mL of acetic acid was added bromine (5.70 mL, 110 mmol) dropwise over 20 min. The mixture was stirred at ambient temperature for 1 h, evaporated, triturated with ether (50 mL). The solid was collected by filtration, washed with diethyl ether (2 x 20 mL) and dried in air. The product, 2-amino-5-bromonicotinic acid hydrobromide, was obtained as a brown solid in 93 % yield (27.8 g). 1H NMR (400 MHz, OMSO-d6): δ 8.28 (d, J= 2.4 Hz, 1H), 8.15 (d, J= 2.4 Hz, 1H), 5.00 (br s, 4H).
2. To a suspension of 2-amino-5-bromonicotinic acid hydrobromide (18.8 g, 60.0 mmol) in 150 mL of methanol was added cone, sulfuric acid (6 mL). The mixture was heated at 75 °C for 2 days, evaporated, diluted with water (100 mL), basified with solid sodium bicarbonate to pH 7-8, and extracted with ethyl acetate (3 x 100 mL). The extracts were washed with brine (50 mL), dried over sodium sulfate and filtered. The filtrate was dried in vacuo and the residue was purified by flash chromatography eluted with ethyl acetate/hexanes (1/4) to afford methyl 2-amino-5- bromonicotinate as a white solid in 36% yield (5.0 g). 1H NMR (400 MHz, CDC13): δ 8.26 (d, J= 2.8 Hz, 1H), 8.23 (d, J= 2.8 Hz, 1H), 3.92 (s, 3H).
3. To a solution of methyl 2-amino-5 -bromonicotinate (4.95 g, 21.4 mmol) in tetrahydrofuran/methanol (30 mL/30 mL) was added 5.2 mL of hydrazine hydrate (NH2NH H20). The mixture was stirred at 75 °C for 18 h, and cooled. The solid was collected by filtration, washed with methanol (2 x 10 mL) and dried in air to afford 2-amino-5-bromonicotinohydrazide as a white solid (3.6 g). The filtrate was evaporated and purified by column chromatography eluted with 5% methanol in dichloromethane to afford another 1.1 g of 2-amino-5-bromonicotinohydrazide as a white solid (total 4.7 g, 95%). 1H NMR (400 MHz, DMSO-<¾): δ 9.74 (s, 1H), 8.09 (d, J= 2.4 Hz, 1H), 7.95 (d, J= 2.4 Hz, 1H), 7.13 (s, 2H), 4.42 (s, 2H).
PREPARATION 9
PREPARATION OF 2-AMiNO-5-BROMO-N-(2,6-DiCHLORO-3- FLUOROBENZOYL)NICOTINOHYDRAZIDE
Figure imgf000067_0001
To a cooled (0 °C) solution of 2-amino-5-bromonicotinohydrazide (0.71 g, 3.08 mmol) in 18 mL of pyridine was added a solution of 2,6-dichloro-3- fluorobenzoyl chloride (crude, 3.24 mmol, prepared from 2,6-dichloro-3-fluorobenzoic acid (0.68 g, 3.24 mmol) with oxalyl chloride) in dichloromethane (5 mL) dropwise. The mixture was warmed to ambient temperature overnight and evaporated. The residue was purified by flash chromatography eluted with 3% methanol in dichloromethane to afford 2-amino-5-bromo-N'-(2,6-dichloro-3-f uorobenzoyl)nicotino-hydrazide as a white solid in 26% yield (0.34 g). 1H NMR (400 MHz, DMSO-<¾): δ 10.8 (s, 1H), 10.7 (s, 1H), 8.24-8.18 (m, 2H), 7 '.62-7 '.52 (m, 2H), 7.24 (br s, 2H). PREPARATION 9.1
PREPARATION OF 2 -AMiN0-N'-BENZ0YL-5 -BROMONICOTINOHYDRAZIDE
Figure imgf000068_0001
Following the procedure as described in Preparation 9, making variations using benzoyl chloride to replace 2,6-dichloro-3-fluorobenzoyl chloride to react with 2- amino-5 -bromonicotinohydrazide, 2-amino-N'-benzoyl-5 -bromonicotinohydrazide was obtained as a white solid in 72% yield. 1H NMR (400 MHz, OMSO-d6): δ 10.5 (s, 2H), 8.20-8.15 (m, 2H), 7.89-7.85 (m, 2H), 7.59-7.46 (m, 3H), 7.22 (br s, 2H).
PREPARATION 9.2
PREPARATION OF SYNTHESIS OF 2-AMINO-5-BROMO-N '-(4-(TERT-
BU DE
Figure imgf000068_0002
Following the procedure as described in Preparation 9, making variations using 4-tert-butylbenzoyl chloride to replace 2,6-dichloro-3-fluorobenzoyl chloride to react with 2-amino-5 -bromonicotinohydrazide, 2-amino-5-bromo-N'-(4-(fert- butyl)benzoyl)-nicotinohydrazide was obtained as a white solid in a quantitative yield. 1H NMR (400 MHz, DMSO-<¾): δ 10.5 (s, 1H), 10.4 (s, 1H), 8.20-8.15 (m, 2H), 7.84- 7.78 (m, 2H), 7.52-7.46 (m, 2H), 7.22 (br s, 2H), 1.23 (s, 9H).
PREPARATION 9.3
PREPARATION OF 2-AMINO- '-(2,5-DIFLUOROBENZOYL)NICOTINOHYDRAZIDE
Figure imgf000068_0003
Following the procedure as described in Preparation 9, making variations using 2,5-difluorobenzoyl chloride to replace 2,6-dichloro-3-fluorobenzoyl chloride to react with 2-amino-5-bromonicotinohydrazide, 2-amino-5-bromo-N'-(2,5- difluorobenzoyl)-nicotinohydrazide was obtained as a white solid in 94% yield. 1H NMR (400 MHz, DMSC ¾): δ 10.6 (s, 1H), 10.4 (s, 1H), 8.19 (d, J= 2.4 Hz, 1H), 8.15 (d, J= 2.4 Hz, 1H), 7.48-7.34 (m, 3H), 7.23 (br s, 2H).
PREPARATION 9.4
PREPARATION OF 2-AMINO-5-BROMO-N^2,6-DICHLOROBENZOYL)NICOTINOHYDRAZIDE
Figure imgf000069_0001
Following the procedure as described in Preparation 9, making variations using 2,6-dichlorobenzoyl chloride to replace 2,6-dichloro-3-fluorobenzoyl chloride to react with 2-amino-5-bromonicotinohydrazide, 2-amino-5-bromo-N'-(2,6- dichlorobenzoyl)nicotinohydrazide was obtained as a white solid in 97% yield. 1H NMR (400 MHz, DMSC ¾): δ 10.7 (s, 1H), 10.6 (s, 1H), 8.25-8.17 (m, 2H), 7.55-7.43 (m, 3H), 7.23 (br s, 2H).
PREPARATION 9.5
PREPARATION OF 2-AMIN YL)NICOTINOHYDRAZIDE
Figure imgf000069_0002
Following the procedure as described in Preparation 9, making variations using 4-fluorobenzoyl chloride to replace 2,6-dichloro-3-fluorobenzoyl chloride to react with 2-amino-5-bromonicotinohydrazide, 2-amino-5-bromo-N'-(4- fluorobenzoyl)nicotinehydrazide was obtained as a white solid in 71% yield. 1H NMR (400 MHz, OMSO-de) δ 10.5 (s, 2H), 8.19 (d, J= 2.4 Hz, 1H), 8.15 (d, J= 2.4 Hz, 1H), 7.98-7.82 (m, 2H), 7.36-7.28 (m, 2H), 7.21 (br s, 2H).
PREPARATION 9.6
PREPARATION OF 2-AMiNO-5-BROMO-N'- (CYCLOHEXANECARBONYL)NICOTINOHYDRAZIDE
Figure imgf000070_0001
Following the procedure as described in Preparation 9, making variations using cyclohexanecarbonyl chloride to replace 2,6-dichloro-3-fluorobenzoyl chloride to react with 2-amino-5-bromonicotinohydrazide, 2-amino-5-bromo-N'- (cyclohexanecarbonyl)nicotinohydrazide was obtained as a white solid in 16% yield. 1H NMR (400 MHz, DMSO-d6): £ 10.26 (br s, 1H), 9.75 (br s, 1H), 8.16 (d, J= 2.4 Hz, 1H), 8.08 (d, J= 2.4 Hz, 1H), 7.18 (br s, 2H), 2.26-2.17 (m, 1H), 1.75-1.66 (m, 4H), 1.64-1.56 (m, 1H), 1.42-1.30 (m, 2H), 1.39-1.11 (m, 3H).
PREPARATION 10
PREPARATION OF 5-BROMO-3-(5-(PYRIDIN-2-YL)-1 ,3,4-OXADIAZOL-2-YL)PYRIDIN-2-
AMINE
Figure imgf000070_0002
Picolinic acid (149 mg, 99 %, 1.20 mmol), 2-amino-5-bromo-N'- nicotinoylnicotinohydrazide (231 mg, 1.00 mmol) and 1- [bis(dimethylamino)methylene]- 1H- 1 ,2,3-triazolo[4, 5-¾]pyridinium 3-oxid
hexafluorophosphate (576 mg, 1.50 mmol) were dissolved in N,N-dimethylformamide (3.0 mL), followed by the addition of triethylamine (0.282 mL, 2.00 mmol). The resulting mixture was stirred at ambient temperature overnight, and then diluted with ethyl acetate (50 mL), washed with brine (2 x 50 mL), and dried over sodium sulfate. After filtration and removal of the solvent, the residue was dried under vacuum to afford a yellow solid N'-(2-amino-5-bromonicotinoyl)picolinohydrazide which was suspended in dichloromethane (20 mL), followed by the addition of triethylamine (0.697 mL, 5.00 mmol), triphenylphosphine (331 mg, 1.25 mmol) and carbon tetrachloride (419 mg, 99 %, 1.25 mmol). The resulting mixture was stirred at ambient temperature for 3 hrs, and then concentrated. The residue was treated with methanol (5.0 mL), and filtered. The solid collected was washed with methanol and dried in vacuo to afford 5-bromo-3-(5-(pyridin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-2-amine as a yellow solid in 59% yield (188 mg). 1H NMR (400 MHz, CDC13): δ 8.84 (d, J= 4.8 Hz, 1H), 8.40 (d, J= 2.4 Hz, 1H), 8.33 (d, J= 8.0 Hz, 1H), 8.28 (d, J= 2.4 Hz, 1H), 7.98- 7.90 (m, 1H), 7.68-7.64 (m, 1H), 6.90-6.70 (br s, 2 H).
PREPARATION 10.1
PREPARATION OF 5-BROMO-3-(5-(PYRIDIN-3-YL)-1 ,3,4-OXADIAZOL-2-YL)PYRIDIN-2-
AMINE
Figure imgf000071_0001
Following the procedure as described in Preparation 10, making variations using nicotinic acid to replace picolinic acid to react with 2-amino-5- bromonicotinohydrazide, 5-bromo-3-(5-(pyridin-3-yl)-l,3,4-oxadiazol-2-yl)pyridin-2- amme was obtained as a yellow solid in 62 % yield. 1H NMR (400 MHz, CDC13): δ 9.38 (s, 1H), 8.84 (d, J= 4.8 Hz, 1H), 8.42 (d, J= 4.8 Hz, 1H), 8.30 (s, 1H), 8.22 (s, 1H), 7.53 (m, 1H), 6.85-6.55 (br s, 2H).
PREPARATION 10.2
PREPARATION OF 5 -BROMO-3 -(5 -(PYRAZIN-2-YL)- 1 ,3 ,4-OXADIAZOL-2-YL)PYRIDIN-2-
Figure imgf000071_0002
Following the procedure as described in Preparation 10, making variations using pyrazine-2-carboxylic acid to replace picolinic acid to react with 2- amino-5-bromonicotinohydrazide, 5-bromo-3-(5-(pyrazin-2-yl)-l,3,4-oxadiazol-2- yl)pyridin-2-amine was obtained as a yellowish solid in 57% yield. 1H NMR (400 MHz, CDC13): δ 9.33 (s, 1H), 8.64-8.60 (m, 2H), 8.18 (d, J= 2.4 Hz, 1H), 8.10 (d, J= 2.4 Hz, 1H), 7.20-7.05 (br s, 2H). PREPARATION 10.3
PREPARATION OF 3 -(5 -BENZ -5 -BROMOP YRIDIN-2-AMINE
Figure imgf000072_0001
Following the procedure as described in Preparation 10, making variations using 2-phenylacetic acid to replace picolinic acid to react with 2-amino-5- bromonicotinohydrazide, 3-(5-benzyl-l,3,4-oxadiazol-2-yl)-5-bromopyridin-2-amine was obtained as a white solid in 49% yield. 1H NMR (400 MHz, CDC13): δ 8.21 (d, J 2.4 Hz, 1H), 8.02 (d, J= 2.4 Hz, 1H), 7.41-7.29 (m, 5H), 6.80-6.55 (br s, 2H), 4.28 (s, 2H).
PREPARATION 10.4
PREPARATION OF 5 -BROMO-3 -(5 -(CYCLOPROPYLMETHYL)- 1 ,3 ,4-OXADIAZOL-2-
Figure imgf000072_0002
Following the procedure as described in Preparation 10, making variations using 2-cyclopropylacetic acid to replace picolinic acid to react with 2- amino-5-bromonicotinohydrazide, 5-bromo-3-(5-(cyclopropylmethyl)-l,3,4-oxadiazol- 2-yl)-pyridin-2-amine was obtained as a yellowish solid in 38% yield. 1H NMR (400 MHz, CDC13): δ 8.24 (d, J= 2.4 Hz, 1H), 8.10 (d, J= 2.4 Hz, 1H), 6.85-6.60 (br s, 2H), 2.85 (d, J= 7.2 Hz, 2H), 1.27-1.16 (m, 1H), 0.71-0.65 (m, 2H), 0.39-0.36 (m, 2H).
PREPARATION 10.5
PREPARATION OF 5 -BROMO-3 -(5 -(THIAZOL-2-YL)- 1 ,3 ,4-OXADIAZOL-2-YL)PYRIDIN-2-
Figure imgf000072_0003
Following the procedure as described in Preparation 10, making variations using thiazole-2-carboxylic acid to replace picolinic acid to react with 2- amino-5-bromonicotinohydrazide, 5-bromo-3-(5-(thiazol-2-yl)-l,3,4-oxadiazol-2- yl)pyridin-2-amine was obtained as a yellow solid in 53% yield. 1H NMR (400 MHz, CDCI3): δ 7.90 (d, J= 2.4 Hz, 1H), 7.87 (d, J= 2.4 Hz, 1H), 7.73 (d, J= 3.2 Hz, 1H), 7.46 (d, J= 3.2 Hz, 1H), 7.00-6.85 (br s, 2H).
PREPARATION 10.6
PREPARATION OF 5 -BROMO-3 -(5 -(4-(TRIFLUOROMETHYL)THIAZOL-2-YL)- 1,3,4- OX NE
Figure imgf000073_0001
Following the procedure as described in Preparation 10, making variations using 4-(trifluoromethyl)thiazole-2-carboxylic acid to replace picolinic acid to react with 2-amino-5-bromonicotinohydrazide, 5-bromo-3-(5-(4-(trifluoromethyl)- thiazol-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-2-amine was obtained as a yellow solid in 35% yield. 1H NMR (400 MHz, CDC13): δ 8.37 (d, J= 2.4 Hz, 1H), 8.32 (d, J= 2.4 Hz, 1H), 8.07 (s, 1H), 6.85-6.55 (br s, 2H).
PREPARATION 11
PREPARATION OF 5-BROMO-3-(5-(2,6-DICHLORO-3-FLUOROPHENYL)-1,3,4-OXADIAZOL-2-
YL)PYRIDIN-2-AMINE
Figure imgf000073_0002
To a suspension of 2-amino-5-bromo-N'-(2,6-dichloro-3-fluorobenzoyl)- nicotinohydrazide (0.34 g, 0.81 mmol) in 20 mL of dichloromethane were added triethylamine (0.59 mL, 4.30 mmol), triphenylphosphine (0.28 g, 1.05 mmol) and tetrabromomethane (CBr4) (0.35 g, 1.05 mmol). The mixture was stirred for 18 h, and evaporated. The residue was purified by flash chromatography eluted with ethyl acetate/dichloromethane (1/20) to afford 5-bromo-3-(5-(2,6-dichloro-3-fluorophenyl)- l,3,4-oxadiazol-2-yl)pyridin-2-amine as a pale brown solid in 38% yield (0.125 g). 1H NMR (400 MHz, DMSO-<¾): δ 8.32 (d, J= 2.4 Hz, 1H), 8.20 (d, J= 2.4 Hz, 1H), 7.88- 7.81 (m, 2H), 7.53 (br s, 2H). PREPARATION 11.1
PREPARATION OF 5-BROMO- ZOL-2-YL)PYRIDIN-2-AMINE
Figure imgf000074_0001
Following the procedure as described in Preparation 11 , making variations using 2-amino-N'-benzoyl-5-bromonicotinohydrazide to replace 2-amino-5- bromo-N'-(2,6-dichloro-3 -fluorobenzoyl)nicotinohydrazide, 5 -bromo-3 -(5 -phenyl- l,3,4-oxadiazol-2-yl)pyridin-2-amine was obtained as a white solid in 89% yield. 1H NMR (400 MHz, OMSO-d6): δ 8.44 (d, J= 2.4 Hz, 1H), 8.28 (d, J= 2.4 Hz, 1H), 8.21 8.16 (m, 2H), 7.65-7.57 (m, 3H), 7.48 (br s, 2H).
PREPARATION 11.2
PREPARATION OF 5-BROMO-3-(5-(4-(7¾^7,-BUTYL)PHENYL)-1,3,4-OXADIAZOL-2-
Figure imgf000074_0002
Following the procedure as described in Preparation 11 , making variations using 2-amino-5-bromo-N'-(4-(tert-butyl)benzoyl)nicotinohydrazide to replace 2-amino-5-bromo-N'-(2,6-dichloro-3-fluorobenzoyl)nicotinohydrazide, 5- bromo-3-(5-(4-(tert-butyl)-phenyl)-l,3,4-oxadiazol-2-yl)pyridin-2-amine was obtained as a white solid in 82% yield. 1H NMR (400 MHz, DMSO-<¾: δ 8.42 (d, J= 2.4 Hz, 1H), 8.27 (d, J= 2.4 Hz, 1H), 8.12-8.08 (m, 2H), 7.64-7.59 (m, 2H), 7.49 (br s, 2H), 1.31 (s, 9H).
PREPARATION 11.3
PREPARATION OF 5 -BROMO-3 -(5 -(2,5 -DIFLUOROPHENYL)- 1 ,3 ,4-OXADIAZOL-2-
Figure imgf000074_0003
Following the procedure as described in Preparation 11 , making variations using 2-amino-5-bromo-N'-(2,5-difluorobenzoyl)nicotinohydrazide to replace 2-amino-5-bromo-N'-(2,6-dichloro-3-fluorobenzoyl)nicotinohydrazide, 5-bromo-3-(5- (2,5-difluoro-phenyl)-l,3,4-oxadiazol-2-yl)pyridin-2-amine was obtained as a pale yellow solid in 87% yield. 1H NMR (400 MHz, OMSO-d6): δ 8.47 (d, J= 2.4 Hz, 1H), 8.29 (d, J= 2.4 Hz, 1H), 8.23-8.17 (m, 1H), 7.59-7.53 (m, 2H), 7.49 (br s, 2H).
PREPARATION 11.4
PREPARATION OF 5-BROMO-3-(5-(2,6-DICHLOROPHENYL)-1 ,3,4-OXADIAZOL-2-
Figure imgf000075_0001
Following the procedure as described in Preparation 11 , making variations using 2-amino-5-bromo-N'-(2,6-dichlorobenzoyl)nicotinohydrazide to replace 2-amino-5-bromo-N'-(2,6-dichloro-3-fluorobenzoyl)nicotinohydrazide, 5- bromo-3-(5-(2,6-dichloro-phenyl)-l,3,4-oxadiazol-2-yl)pyridin-2-amine was obtained as a white solid in 70% yield. 1H NMR (400 MHz, DMSO-<¾: δ 8.31 (d, J= 2.4 Hz, 1H), 8.20 (d, J= 2.4 Hz, 1H), 7.78-7.70 (m, 3H), 7.53 (br s, 2H).
PREPARATION 11.5
PREPARATION OF 5-BROMO-3-(5-(4-FLUOROPHENYL)-1,3,4-OXADIAZOL-2-YL)-PYRIDIN-
Figure imgf000075_0002
Following the procedure as described in Preparation 11 , making variations using 2-amino-5-bromo-N'-(4-fluorobenzoyl)nicotinohydrazide to replace 2- amino-5-bromo-N'-(2,6-dichloro-3-fluorobenzoyl)nicotinohydrazide, 5-bromo-3-(5-(4- fluorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-2-amine was obtained as a pale yellow solid in 83% yield. 1H NMR (400 MHz, DMSO-<¾): δ 8.47 (d, J= 2.4 Hz, 1H), 8.28 (d, J = 2.4 Hz, 1H), 8.27-8.23 (m, 2H), 7.48 (br s, 2H), 7.48-7.42 (m, 2H). PREPARATION 12
PREPARATION OF 1 -CYCLOHEXYL -4-(4,4,5,5-TETRAMETHYL-1,3,2-DIOXABOROLAN-2-
Figure imgf000076_0001
1. To a cooled (0 °C) solution of cyclohexanol (2.50 g, 25.0 mmol) in dichloromethane (50 mL) was added triethylamine (7.00 mL, 50.0 mmol), followed by the addition of methanesulfonyl chloride (2.91 mL, 37.5 mmol). The mixture was stirred at ambient temperature for 20 h, diluted with dichloromethane (50 mL), washed with 1 N hydrochloroic acid solution (20 mL), saturated sodium bicarbonate (30 mL), water (30 mL), brine (30 mL) and dried. The crude cyclohexyl methanesulfonate was obtained as pale yellow oil after removal of the solvent (4.47 g, 100% yield). 1Η NMR (300 MHz, CDC13): δ 4.75-4.67 (m, 1H), 3.00 (s, 3H), 2.04-1.95 (m, 2H), 1.83-1.74 (m, 2H), 1.70-1.59 (m, 2H), 1.57-1.49 (m, 1H), 1.46-1.25 (m, 3H).
2. To a cooled (0 °C) solution of 4-bromo-lH-pyrazole (2.21 g, 15.0 mmol) in dimethylformamide (25 mL) was added 60% sodium hydride (0.900 g, 22.5 mmol) in portions. After 30 min, a solution of cyclohexyl methanesulfonate (2.84 g, 16.0 mmol) in dimethylformamide (4 mL) was added. The mixture was heated at 105 °C for 20 h. The volatiles were removed in vacuo and the residue was diluted with ethyl acetate (100 mL), washed with water (2 x 20 mL), brine (20 mL) and dried. The residue after removal of the solvents was purified by flash column chromatography eluted with 5% ethyl acetate in hexanes to afford 4-bromo-l -cyclohexyl- lH-pyrazole as a white solid in 33% yield (1.14 g). 1H NMR (400 MHz, CDC13): δ 7.45 (s, 1H), 7.44 (s, 1H), 4.13-4.05 (m, 1H), 2.18-2.10 (m, 2H), 1.94-1.86 (m, 2H), 1.75-1.62 (m, 3H), 1.48-1.36 (m, 2H), 1.31-1.19 (m, 1H).
3. To a solution of 4-bromo-l -cyclohexyl- lH-pyrazole (1.13 g, 4.95 mmol) in dimethyl sulfoxide (10 mL) was added potassium acetate (1.47 g, 15 mmol), pinacol diboron (1.52 g, 6.00 mmol). The mixture was degassed for 10 min, followed by the addition of PdCl2(dppf) (0.300 g, 0.400 mmol). The mixture was degassed for another 10 min and heated at 90 °C for 20 h, diluted with ethyl acetate (80 mL), washed with water (2 x 20 mL), brine (20 mL) and dried. The residue after removal of the solvents was purified by flash column chromatography eluted with 10% ethyl acetate in hexanes to afford l-cyclohexyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole as a white solid in 20% yield (0.265 g). 1H NMR (300 MHz, CDC13) δ 7.80 (s, 1H), 7.75 (s, 1H), 4.21-4.11 (m, 1H), 2.21-2.14 (m, 2H), 1.94-1.85 (m, 2H), 1.77-1.65 (m, 3H), 1.48-1.34 (m, 3H), 1.32 (s, 12H).
PREPARATION 13
PREPARATION OF N -HYDROXYBENZAMIDINE
Figure imgf000077_0001
A mixture of benzonitrile (5.00 g, 48.5 mmol), hydroxylamine hydrochloride (8.42 g, 121 mmol) and triethylamine (30 mL) was refluxed in 30 mL of ethanol for 7 hours. After removal of solvent in vacuo, the residue (white suspension) was mixed with water and extracted with ethyl acetate (3 x 50 mL). The collected organic solution was dried over sodium sulfate. N'-hydroxybenzamidine was obtained as a grey gum in 70% yield (4.60 g) after the removal of the solvent in vacuo. 1H NMR (400 MHz, CDC13): £7.65-7.60 (m, 2H), 7.46-7.35 (m, 3H), 4.95 (br s, 2H).
PREPARATION 13.1
PREPARATION OF (Z)-2, HYDROXYBENZAMiDiNE
Figure imgf000077_0002
Following the procedure as described in Preparation 13, making variations using 2,6-dichlorobenzonitrile to replace benzonitrile, (Z)-2,6-dichloro-N- hydroxybenzamidine was obtained as a white solid in 37% yield. 1H NMR (400 MHz, DMSO-de): δ 9.38 (s, 1H), 7.48-7.45 (m, 2H), 7.41-7.35 (m, 1H), 5.88 (br s, 2H).
PREPARATION 14
PREPARATION OF N'-((2- YL)oxY)BENziMiD AMIDE
Figure imgf000078_0001
To a mixture of N'-hydroxybenzamidine (0.50 g, 3.68 mmol) and 2- amino-5-bromopyridine-3-carboxylic acid (0.83 g, 4.05 mmol) in 30 mL of
dichloromethane was added l-ethyl-3-(3-dimethylaminopropyl)carbodiimide (1.45 g, 7.36 mmol). The resulting mixture was stirred at ambient temperature overnight. After removal of solvent in vacuo, the residue was purified by column chromatography eluted with 5% methanol in dichloromethane to afford a yellow oil which was treated with dichloromethane and ethyl ether to afford N'-((2-amino-5-bromonicotinoyl)oxy)- benzimidamide as a yellow solid in 32% yield (0.25 g). 1H NMR (400 MHz, DMSO- d6): £8.61 (d, J= 2.4 Hz, 1H), 8.25 (d, J= 2.4 Hz, 1H), 7.74-7.70 (m, 2H), 7.52-7.42 (m, 3H), 7.34 (br s, 2H), 7.04 (br s, 2H).
PREPARATION 14.1
PREPARATION OF (Z)-N'-((2-AMiNO-5-BROMONicoTiNOYL)oxY)-2,6-
Figure imgf000078_0002
Following the procedure as dedscribed in Preparation 14, making variations using (Z)-2,6-dichloro-N'-hydroxybenzamidine to replace N'- hydroxybenzamidine to react with 2-amino-5-bromopyridine-3-carboxylic acid, (Ζ)-Ν'- ((2-amino-5-bromonicotinoyl)oxy)-2,6-dichlorobenzimidamide was obtained as a yellowish solid in 41% yield. 1H NMR (400 MHz, DMSO-d6): δ 8.67 (d, J= 2.4 Hz, 1H), 8.25 (d, J= 2.4 Hz, 1H), 7.56-7.46 (m, 3H), 7.34 (br s, 4H). PREPARATION 15
PREPARATION OF 5-BROMO-3-(3-PHENYL-1 ,2,4-OXADIAZOL-5-YL)PYRIDIN-2-AMINE
H2N N
A solution of N-((2-amino-5-bromonicotinoyl)oxy)benzimidamide (0.30 g, 0.90 mmol) in 5 mL of pyridine was refluxed for 3 hours. After cooling, precipitates were formed. After removal of pyridine by gravity filtration, the precipitates were washed with water to afford 5-bromo-3-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-2-amine as a yellowish solid in 92% yield (0.25 g). 1H NMR (400 MHz, DMSO-d6): £8.35 (d, J = 2.4 Hz, 1H), 8.32 (d, J= 2.4 Hz, 1H), 8.15-8.10 (m, 2H), 7.66 (br s, 2H), 7.60-7.54 (m, 3H).
PREPARATION 15.1
PREPARATION OF 5-BROMO-3-(3-(2,6-DICHLOROPHENYL)-1 ,2,4-OXADIAZOL-5-
Figure imgf000079_0001
Foloowing the procedure as described in Preparation 15, making variations using N'-((2-amino-5-bromonicotinoyl)oxy)-2,6-dichlorobenzimidamide to replace N'-((2-amino-5-bromonicotinoyl)oxy)benzimidamide, 5-bromo-3-(3-(2,6- dichlorophenyl)-l,2,4-oxadiazol-5-yl)pyridin-2-amine was obtained as a yellowish solid in 67% yield. 1H NMR (400 MHz, CDC13): δ 8.43 (d, J= 2.4 Hz, 1H), 8.33 (d, J= 2.0 Hz, 1H), 7.50-7.42 (m, 3H), 6.80 (br s, 2H).
PREPARATION 16
PREPARATION OF (Z)-2-AMiNO-5-BROMO-N'-HYDROXYNicoTiNiMiD AMIDE
Figure imgf000079_0002
A solution of 2-amino-5-bromopyridine-3-carbonitrile (2.00 g, 10.1 mmol), hydroxylamine hydrochloride (1.06 g, 15.3 mmol) and 24 mL of triethylamine in 40 mL of ethanol was refluxed for 3 h. The resulting mixture was poured into ice cold water to afford a white solid. The solid was separated by filtration and washed with water. The filtrate was extracted with ethyl acetate and the organic layers were combined, dried over anhydrous sodium sulfate and filtered. The filtrate was evaporate to afford a white solid which was combined with the white solid obtained earlier to afford (Z)-2-amino-N'-(benzoyloxy)-5-bromonicotinimidamide in quantitative yield (2.30 g). 1H NMR (400 MHz, OMSO-d6):S9.95 (s, 1H), 7.96 (d, J= 2.4 Hz, 1H), 7.89 (d, J= 2.4 Hz, 1H), 7.20 (br s, 2H), 5.93 (s, 2H).
PREPARATION 17
PREPARATION OF 5-BROMO-3-(5-PHENYL-1,2,4-OXADIAZOL-3-YL)PYRIDIN-2-AMINE
Figure imgf000080_0001
A. A mixture of benzoic acid (319 mg, 2.60 mmol), 2-amino-5- bromo-A/"-hydroxypyridine-3-carboxamidine (400 mg, 1.73 mmol) and l-ethyl-3-(3- dimethylaminopropyl)carbodiimide (627 mg, 3.24 mmol) in dichloromethane (10 mL) was stirred at ambient temperature for 5 hours and a white solid was precipitated. The solid was collected by filtration, washed with dichloromethane, and dried in vacuo to afford (Z)-2-amino-N'-(benzoyloxy)-5-bromonicotinimidamide as a white solid in 73% yield (422 mg). 1H NMR (400 MHz, DMSO-<¾): ^ 8.18 (dd, J= 8.0, 1.2 Hz, 2H), 8.10 (d, J= 2.4 Hz, 1H), 8.00 (d, J= 2.4 Hz, 1H), 7.66 (t, J= 8.0 Hz, 1H), 7.52 (t, J= 8.0 Hz, 2H), 7.29 (br s, 2H), 7.04 (br s, 2H).
B. A suspension of (Z)-2-amino-N'-(benzoyloxy)-5-bromo- nicotinimidamide (405 mg, 1.21 mmol) in pyridine (6.0 mL, 77.0 mmol) was stirred at 110-115 °C overnight. After cooled to ambient temperature, the reaction mixture was concentrated under high vacuum. The residue was azeotroped with toluene (4 X 10 mL), and dried under vacuum for 30 minutes. The dried residue was treated with methanol (10 mL), and a precipitate was obtained. The precipitate was collected by filtration, and washed with methanol (~20 mL), and dried under vacuum to afford 5- bromo-3-(5-phenyl-l,2,4-oxadiazol-3-yl)pyridin-2-amine as a yellow solid (285 mg, 74%). 1H NMR (400 MHz, CDC13): δ 8.56 (d, J= 2.4 Hz, 1H), 8.27 (d, J= 2.4 Hz, 1H), 8.25-8.20 (m, 2H), 7.68-7.63 (m, 1H), 7.62-7.56 (m, 2H), 6.30-6.17 (br s, 2H). PREPARATION 18
PREPARATION OF 5-BROMO-3-(5-(2,5-DICHLOROPHENYL)-1 ,2,4-OXADIAZOL-3-
YL)PYRIDIN-2-AMINE
Figure imgf000081_0001
A. To a solution of 2-amino-5-bromo-N'-hydroxypyridine-3- carboxamidine (74 mg, 0.344 mmol) and 2,5-dichlorobenzoic acid (72 mg, 0.378 mmol) in 5 mL of dichloromethane was added l-ethyl-3-(3-dimethylaminopropyl)- carbodiimide (98 mg, 0.516 mmol) and 1 mL of N,N-dimethylformamide. The mixture was stirred at ambient temperature overnight, followed by the addition of water to quench the reaction. The resulting mixture was extracted with dichloromethane (2 x 10 mL). The combined organic solution was dried over anhydrous sodium sulfate. After filtration and removal of solvent, the residue was purified by flash column
chromatography to afford (Z)-2-amino-5-bromo-N'-((2,5-dichlorobenzoyl)oxy)- nicotinimidamide as a white solid in 72 % yield (96 mg). 1H NMR (400 MHz, DMSO- d6): £8.12 (d, J= 2.4 Hz, 1H), 8.11 (d, J= 2.4 Hz, 1H), 7.97 (d, J= 2.4 Hz, 1H), 7.65 (dd, J= 8.0, 2.4 Hz, 1H), 7.62 (d, J= 8.0 Hz, 1H), 7.19 (br s, 2H), 7.08 (br s, 2H).
B. A solution of (Z)-2-amino-5-bromo-N-((2,5- dichlorobenzoyl)oxy)-nicotinimidamide (170 mg, 0.463 mmol) in 5 mL of pyridine was refluxed for 2 h. The reaction mixture was cooled to room temperature and pyridine was removed in vacuo. The resulting white solid was washed with water and collected by filtration. The collected white solid was washed with a mixture of ethyl
acetate: dichloromethane (1 : 10) to remove the remaining starting material to afford pure 5-bromo-3-(5-(2,5-dichlorophenyl)-l,2,4-oxadiazol-3-yl)pyridin-2-amine as a yellow solid in 34% yield (60 mg). 1H NMR (400 MHz, DMSO-d6): £8.35 (d, J= 2.4 Hz, 1H), 8.31-8.28 (m, 2H), 7.82-7.77 (m, 2H), 7.12 (br s, 2H). PREPARATION 19
PREPARATION OF TERT-EUTYL 4-(4-(4,4,5,5-TETRAMETHYL-1,3,2-DIOXABOROLAN-2-YL)- 1H-PYRA 1 -CARBOXYLATE
Figure imgf000082_0001
A. To a cold solution (0 °C) of tert-butyl 4-hydroxypiperidine- 1- carboxylate (175 g, 0.87 mol) in 750 mL of dichloromethane was added triethylamine (237 mL, 1.74 mol) dropwise, followed by the addition of mesyl chloride (101 mL, 1.31 mol) dropwise. The resulting mixture was stirred at room temperature for 2 hours, then diluted with 400 mL of dichloromethane and washed with saturated sodium bicarbonate (2 x 750 mL), water (1 x 600 mL) and brine (1 x 600 mL). The organic solution was dried over anhydrous sodium sulfate, and filtered. Removal of the solvent afforded tert- butyl 4-((methylsulfonyl)oxy)piperidine-l-carboxylate as a yellow solid in 96.8% yield (235 g). 1H NMR (400 MHz, CDC13) δ 4.90-4.82 (m, 1H), 3.73-3.64 (m, 2H), 3.34-3.25 (m, 2H), 3.03 (s, 3H), 2.00-1.92 (m, 2H), 1.88-1.75 (m, 2H), 1.44 (s, 9H).
B. To a cold solution (0 °C) of 4-bromo-lH-pyrazole (14.9 g, 0.10 mol) in 80 mL of dichloromethane was added sodium hydride (8.13 g, 0.20 mol) portion wise. The mixture was stirred at 0 °C for 1 hour, followed by the addition of a solution of tert-butyl 4-((methylsulfonyl)oxy)piperidine-l-carboxylate (34.0 g, 0.12 mol) in 30 mL of N,N-dimethylformamide. The resulting mixture was heated at 110 °C overnight. Purification by column chromatography afforded tert-butyl 4-(4-bromo-lH- pyrazol-l-yl)piperidine-l-carboxylate as a yellow oil in 58% yield (19.5 g). 1H NMR (400 MHz, CDC13) δ 7.48 (s, 1H), 7.44 (s 1H), 4.35-4.20 (m, 3H), 2.95-2.85 (m, 2H), 2.15-2.05 (m, 2H), 1.92-1.81 (m, 2H), 1.48 (s, 9H).
C. A mixture of tert-butyl 4-(4-bromo- lH-pyrazol- 1 -yl)piperidine- 1-carboxylate (20.0 g, 0.61 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-l,3,2-dioxaborolane (30.8 g, 0.12 mmol) and potassium acetate (17.8 g, 0.18 mol) in 50 mL of dimethyl sulfoxide was purged with nitrogen gas for 10 min. After the addition of [l, -bis(diphenylphosphino)ferrocene]dichloropalladium(II) (3.55 g, 4.85 mmol), the mixture was purged with nitrogen gas for another 10 minutes, heated at 80 °C overnight under nitrogen atmosphere and filtered through celite and washed with ethyl acetate. The filtrate was extracted with ethyl acetate (2 x 200 mL). The organic layer was dried over anhydrous sodium sulfate. After filtration and removal of the solvent, the residue was purified by column chromatograph eluted with hexane to afford an oil which was recrystallized from hexane to afford tert-butyl 4-(4-(4,4,5,5- tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate as a white solid in 44% yield (10 g). 1H NMR (400 MHz, CDC13) δ 7.79 (s, 1H), 7.72 (s, 1H), 4.32-4.13 (m, 3H), 2.95-2.80 (m, 2H), 2.15-2.05 (m, 2H), 1.93-1.82 (m, 2H), 1.47 (s, 9H), 1.31 (s, 12H).
PREPARATION 20
PREPARATION OF 5 -BROMO-3 -(5 -CYCLOHEXYL- 1 ,3 ,4-OXADIAZOL-2-YL)PYRIDIN-2 -AMINE
Figure imgf000083_0001
To a suspension of 2-amino-5-bromo-N'-(cyclohexanecarbonyl)- nicotinohydrazide (0.20 g, 0.59 mmol) in 15 mL of dichloromethane was added triethylamine (0.40 mL, 2.93 mmol) and acetic anhydride (13 xL, 0.14 mmol), followed by the addition of triphenylphosphine (0.19 g, 0.73 mmol) and tetrabromomethane (0.24 g, 0.73 mmol). The resulting mixture was stirred at room temperature for 2 hours. After removal of the solvent in vacuo, the residue was purified by column
chromatography eluted with 1 :1 ethyl acetate:hexane to afford 5-bromo-3-(5- cyclohexyl-l,3,4-oxadiazol-2-yl)pyridin-2-amine as a white solid in 77% yield (0.14 g). 1H NMR (400 MHz, CDC13): £8.23 (d, J= 2.4 Hz, 1H), 8.07 (d, J= 2.4 Hz, 1H), 6.61 (br s, 2H), 3.04-2.95 (m, 1H), 2.20-2.10 (m, 2H), 1.94-1.84 (m, 2H), 1.80-1.60 (m, 3H), 1.50-1.30 (m, 3H).
PREPARATION 21
PREPARATION OF ΓΕΚΓ-BUTYL 4-(3-(4,4,5,5-TETRAMETHYL-1,3,2-DIOXABOROLAN-2-YL)- 1H-PYRR 1 -CARBOXYLATE
Figure imgf000083_0002
A. To an ice-cold solution of pyrrole (611 mg, 9.11 mmol) in 6 mL of A,N-dimethylformamide was slowly added sodium hydride (364 mg, 9.11 mmol, 60% in mineral oil). After stirred at room temperature for 1 hour, the resulting mixture was cooled to 0 °C, followed by the addition of l-(tert-butoxycarbonyl)piperidin-4-yl methanesulfonate slowly. The mixture was then heated up to 110 °C and stirred overnight. After removal of the solvent in vacuo, the residue was purified by column chromatography eluted with 20% ethyl acetate in hexane to afford fert-butyl 4-(lH- pyrrol- l-yl)piperidine-l-carboxylate as a colorless oil in 21% yield (317 mg). 1H NMR (400 MHz, CDCI3): δ 6.75-6.71 (m, 2H), 6.18-6.15 (m, 2H), 4.32-4.20 (m, 2H), 4.02- 3.93 (m, 1H), 2.90-2.76 (m, 2H), 2.07-2.03 (m, 2H), 1.89-1.78 (m, 2H), 1.48 (s, 9H).
B. To a solution of tert-butyl 4-(lH-pyrrol-l-yl)piperidine-l- carboxylate (1.03 g, 3.72 mmol) in 30 mL of anhydrous tetrahydrofuran at -78 °C was added phosphorus tribromide (60 mg, 0.220 mmol) and N-bromosuccinimide (663 mg, 3.72 mmol) in sequence. The mixture was stirred at -78 °C for 1 hour, and it was left in the freezer (-15 °C) overnight. A white solid was precipitated after the addition of triethylamine (0.42 g, 1 equiv) and hexanes (40 mL) to the mixture. The suspension was filtered through neutral aluminium oxide pad and dried over sodium sulfate. The removal of the solvent of the filtrate afforded tert-butyl 4-(3-bromo-lH-pyrrol-l- yl)piperidine-l-carboxylate (1.23 g) as yellowish oil which was used for the next step reaction without further purification. 1H NMR (400 MHz, CDC13): δ 6.71-6.69 (m, 1H), 6.63-6.61 (m, 1H), 6.16-6.13 (m, 1H), 4.35-4.20 (m, 2H), 3.95-3.85 (m, 1H), 2.89-2.76 (m, 2H), 2.06-1.98 (m, 2H), 1.89-1.76 (m, 2H), 1.47 (s, 9H).
C. A solution of tert-butyl 4-(3-bromo-lH-pyrrol-l-yl)piperidine-l- carboxylate (1.23 g, 3.7 mmol), bis(pinacolato)diboron (1.90 g, 7.4 mmol) and potassium acetate (1.09 g, 11.1 mmol) in 20 mL of dioxane in a sealed tube was degassed with nitrogen gas for 10 min, followed by the addition of 1,1 '- bis(diphenylphosphino)ferrocene-palladium(II) di chloride (270 mg, 0.37 mmol). The mixture was degassed for another 10 min. The tube was sealed and heated at 80 °C overnight. The resulting solution was washed with water and extracted with ethyl acetate. Organic layers were combined and evaporated and the residue was purified by column chromatography eluted with 15% ethyl acetate in hexanes to afford tert-butyl 4- (3-(4,4,5 ,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- lH-pyrrol- 1 -yl)piperidine- 1 - carboxylate as colorless oil in 14% yield (70 mg). 1H NMR (400 MHz, CDC13): δ 7.19- 7.17 (m, 1H), 6.76-6.74 (m, 1H), 6.51-6.49 (m, 1H), 4.28-4.19 (m, 2H), 4.02-3.92 (m, 1H), 2.86-2.78 (m, 2H), 2.04-2.01 (m, 2H), 1.88-1.78 (m, 2H), 1.48 (s, 9H), 1.32 (s, 12 H). PREPARATION 22
PREPARATION OF l-(TETRAHYDRO-2H-PYRAN-4-YL)-4-(4,4,5,5-TETRAMETHYL-l,3,2-
DIOXA AZOLE
Figure imgf000085_0001
A. To a cooled (0 °C) solution of tetrahydro-2H-pyran-4-ol (5.00 g,
49.0 mmol) in dichloromethane (85 mL) was added triethylamine (5.95 mL, 58.8 mmol), followed by the addition of methanesulfonyl chloride (6.74 mL, 58.8 mmol). The mixture was stirred at room temperature overnight, diluted with dichloromethane (85 mL), then sequentially washed with 0.5 N hydrochloric acid solution (40 mL), saturated sodium bicarbonate (50 mL), water (50 mL), brine (50 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was evaporated to afford the crude product tetrahydro-2H-pyran-4-yl methanesulfonate (8.6 g, 97.4%) as a pale yellow solid. 1H NMR (400 Hz, CDC13) δ 4.95-4.85 (m, 1H), 3.97-3.90 (m, 2H), 3.57-3.51 (m, 2H), 3.04 (s, 3H), 2.08-2.02 (m, 2H), 1.93-1.84 (m, 2H).
B. To a cooled (0 °C) solution of 4-bromo-lH-pyrazole (3.00 g, 20.4 mmol) in N,N-dimethylformamide (10 mL) was added 60% sodium hydride (0.90 g, 22.5 mmol) in portions, followed by the addition of a solution of tetrahydro-2H-pyran- 4-yl methanesulfonate (5.52 g, 30.6 mmol) in N,N-dimethylformamide (10 mL) 30 minutes later. The mixture was heated at 110 °C overnight, cooled to room temperature and mixed with water (50 mL). The precipitates obtained were collected by filtration, washed with water and dried to afford 4-bromo-l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazole (2.90 g, 62%) as a brown solid. 1H NMR (400 Hz, CDC13) δ 7.47 (s, 1H), 7.46 (s, 1H), 4.35-4.30 (m, 1H), 4.12-4.08 (m, 2H), 3.55-3.48 (m, 2H), 2.15-2.00 (m, 4H).
C. To a solution of 4-bromo-l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazole (2.90 g, 12.6 mmol) in dimethyl sulfoxide (25 mL) was added potassium acetate (4.94 g, 50.4 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-l,3,2-dioxaborolane (6.40 g, 25.2 mmol). The mixture was degassed for 10 min, and then [l, -bis(diphenylphosphino)ferrocene]dichloro-palladium(II) (0.922 g, 1.26 mmol) was added. The mixture was degassed for another 10 minutes and heated at 90 °C overnight, diluted with ethyl acetate (80 mL), washed with water (2 x 20 mL), brine (20 mL), dried and filtered. The solvent of the filtrate was removed and the residue was purified by flash column chromatography eluted with 20% ethyl acetate in hexanes to afford l-(tetrahydro-2H-pyran-4-yl)-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (0.66 g, 19%) as a white solid. 1H NMR (400 Hz, CDCls) δ 7.81 (s, 1H), 7.76 (s, 1H), 4.42-4.32 (m, 1H), 4.14-4.06 (m, 2H), 3.58-3.50 (m, 2H), 2.16-1.98 (m, 4H), 1.32 (s, 12H).
EXAMPLES EXAMPLE 1
SYNTHESIS OF TERT-BUYYL 4-(4-(6-AMINO-5 -(5 -PHENYL)- 1,3, 4-OXADIAZOL-2- YL)PYRIDIN- 1 -CARBOXYLATE
Figure imgf000086_0001
To a solution of 5-bromo-3-(5-phenyl-l,3,4-oxadiazol-2-yl)pyridin-2- amine (0.112 g, 0.353 mmol) in dimethyl sulfoxide (5 mL) were added tert-butyl 4-(4- (4,4,5 ,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- IH-pyrazol- 1 -yl)piperidine- 1 -carboxylate (0.160 g, 0.424 mmol), potassium acetate (0.140 g, 1.40 mmol) and PdCl2(dppf) (26.0 mg, 0.035 mmol). The mixture was degassed with nitrogen gas for 5 min, evaporated, heated at 100 °C for 20 h, cooled, diluted with 50 mL of ethyl acetate, washed with water (10 mL), and brine (10 mL). The organic phase was dried in vacuo and the residue was purified by flash chromatography eluted with 60% ethyl acetate in hexanes to afford the title compound as a pale brown solid in 24% yield (42 mg). 1H NMR (400 MHz, CDCI3): δ 8.40 (d, J= 2.0 Hz, 1H), 8.19-8.15 (m, 3H), 7.78 (d, J= 0.8 Hz, 1H), 7.68 (d, J= 0.8 Hz, 1H), 7.62-7.55 (m, 3H), 6.63 (br s, 2H), 4.38-4.23 (m, 3H), 3.00- 2.87 (m, 2H), 2.23-2.16 (m, 2H), 2.06-1.83 (m, 2H), 1.50 (s, 9H).
The table below shows additional examples of compounds of Formula (IA4) that were prepared according to the above process.
Figure imgf000087_0001
Figure imgf000088_0001
EXAMPLE 2
SYNTHESIS OF 3-(3-PHENYL-l,2,4-OXADIAZOL-5-YL)-5-(l-(TETRAHYDRO-2H-PYRAN-4- E
Figure imgf000088_0002
A mixture of 5-bromo-3-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-2-amine
(100 mg, 0.33 mmol), l-(tetrahydro-2H-pyran-4-yl)-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (0.11 g, 0.40 mmol) and cesium carbonate (0.54 g, 1.65 mmol) in 5 mL of 10: 1 dioxane:water in a sealed tube was degassed for 10 min, followed by the addition of tetrakis(triphenylphosphine)palladium(0) (38 mg, 0.03 mmol). The mixture was degassed for another 10 minutes and then heated at 100 °C overnight. After removal of the solvents in vacuo, the residue was purified by column chromatography eluted with ethyl acetate to afford the title compound as a yellowish solid in 20% yield (25 mg). 1H NMR (400 MHz, DMSO-d6): £8.48 (s, 1H), 8.35 (d, J = 2.4 Hz, 1H), 8.20-8.14 (m, 2H), 7.79 (s, 1H), 7.71 (s, 1H), 7.58-7.50 (m, 3H), 6.66 (br s, 2H), 4.48-4.38 (m, 1H), 4.20-4.10 (m, 2H), 3.62-3.55 (m, 2H), 2.22-2.10 (m, 4H); MS (ES+): m/z 389.3 (M + 1).
The table below shows additional examples of compounds of Formula (IA5) that were prepared according to the above process.
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
456.3 and 457.3 (M + 1). Ex.
Chemical Name Starting Material and Yield Characterization Data No.
fert-butyl 4-(3-(6- 5-bromo-3-(5-(2,6- ¾ NMR (400 MHz, CDC13): δ 8.45 amino-5-(5-(2,6- dichlorophenyl)- 1 ,3 ,4- (d, J= 2.4 Hz, 1H), 8.13 (.7= 2.4 Hz, dichlorophenyl)- oxadiazol-2-yl)pyridin-2- 1H), 7.53-7.48 (m, 3H), 6.99-6.97 l,3,4-oxadiazol-2- amine and feri-butyl 4-(3- (m, 1H), 6.78-6.75 (m, 1H), 6.55 (br
2.19
yl)pyridin- 3 -yl) - 1 H- (4,4,5,5-tetramethyl-l,3,2- s, 2H), 6.43-6.40 (m, 1H), 4.35-4.22 pyrrol-1- dioxaborolan-2-yl)- \H- (m, 2H), 4.02-3.92 (m, 1H), 2.80- yl)piperidine-l- pyrrol- 1 -yl)piperidine- 1 - 2.78 (m, 2H), 2.12-2.05 (m, 2H), carboxylate carboxylate; 46% 1.92-1.80 (m, 2H), 1.48 (s, 9H).
EXAMPLE 3
SYNTHESIS OF 3-(5-(2,6-DICHLORO-3-FLUOROPHENYL)-1,3,4-OXADIAZOL-2-YL)-5-(1- PIPERIDIN-4-YL)-LH-PYRAZOL-4-YL)PYRIDIN-2-AMINE
Figure imgf000092_0001
To a solution of tert-butyl 4-(4-(6-amino-5-(5-(2,6-dichloro-3- fluorophenyl)- 1 ,3 ,4-oxadiazol-2-yl)pyridin-3 -yl)- lH-pyrazol- 1 -yl)piperidine- 1 - carboxylate (30.0 mg, 0.052 mmol) in dichloromethane (3 mL) was added a solution of 2 N hydrochloric acid in diethyl ether (1.00 mL, 2.00 mmol). The mixture was stirred at ambient temperature overnight, diluted with saturated sodium bicarbonate (5 mL), and extracted with dichloromethane (3 x 20 mL). The combined extracts were washed with brine (10 mL), dried and evaporated to afford the title compound as a pale brown solid in 92% yield (23 mg). 1H NMR (400 Hz, CDC13): δ 8.43 (d, J= 2.4 Hz, 1H), 8.11 (d, J = 2.4 Hz, 1H), 7.74 (d, J= 0.8 Hz, 1H), 7.66 (d, J= 0.8 Hz, 1H), 7.53-7.48 (m, 1H), 7.41-7.36 (m, 1H), 6.61 (br s, 2H), 4.30-4.21 (m, 1H), 3.30-3.23 (m, 2H), 2.83-2.75 (m, 2H), 2.23-2.17 (m, 2H), 1.98-1.87 (m, 2H); MS (ES+): m/z 474.2, 475.2 and 476.3 (M + 1).
The table below shows additional examples of compounds of Formula (IA4) that were prepared according to the above process.
Ex.
Chemical Name Starting Material and Yield Characterization Data No.
¾ NMR (400 MHz, CDC13): δ 8.40 (d, J= 2.4 Hz, 1H), 8.19-8.15 (m,
3-(5-phenyl-l,3,4- feri-butyl 4-(4-(6-amino-5- 3H), 7.77 (d, J= 0.8 Hz, 1H), 7.70 (d, oxadiazol-2-yl)-5- (5-phenyl)- 1 ,3 ,4-oxadiazol- J= 0.8 Hz, 1H), 7.61-7.55 (m, 3H),
3.1 ( 1 -(piperidin-4-yl)- 2 -yl)pyridin- 3 -yl) - 1 H- 6.59 (br s, 2H), 4.34-4.25 (m, 1H),
17/-pyrazol-4- pyrazol- 1 -yl)piperidine- 1 - 3.32-3.25 (m, 2H), 2.85-2.77 (m, 2H), yl)pyridin-2 - amine carboxylate; 76%
2.26-2.19 (m, 2H), 2.03-1.91 (m, 2H); MS (ES+): m/z 388.3 (M + 1).
Figure imgf000093_0001
Figure imgf000094_0001
y pyr n- - amne .
Figure imgf000095_0001
EXAMPLE 4
SYNTHESIS OF 1-(4-(4-(6-AMINO-5-(5-(2,6-DICHLOROPHENYL)-1,3,4-OXADIAZOL-2-
YL)PYRIDIN-3 -YL)- 1 H-PYRAZOL- 1 -YL)PIPERIDIN- 1 -YL)ETHANONE
Figure imgf000095_0002
To a solution of 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l- piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine (62.0 mg, 0.136 mmol) in
dichloromethane (3 mL) was added acetic anhydride (0.013 mL, 0.136 mmol), followed by the addition of triethylamine (0.098 mL, 0.700 mmol) at 0 °C. The mixture was stirred at ambient temperature for 3 h, diluted with dichloromethane (20 mL), washed with saturated sodium bicarbonate (10 mL) and brine (10 mL). The volatiles were removed in vacuo and the residue was purified by flash chromatography eluted with 5% methanol in dichloromethane to afford the title compound as a pale brown solid in 81% yield (55 mg). 1H NMR (400 Hz, CDC13): δ 8.35 (d, J= 2.4 Hz, 1H), 8.19 (d, J= 2.4 Hz, 1H), 7.41 (s, 1H), 7.65 (s, 1H), 7.53-7.50 (m, 3H), 7.10 (br s, 2H), 4.79-4.72 (m, 1H), 4.43-4.33 (m, 1H), 4.02-3.94 (m, 1H), 3.30-3.21 (m, 1H), 2.82-2.73 (m, 1H), 2.30- 2.16 (m, 2H), 2.14 (s, 3H), 2.04-1.90 (m, 2H); MS (ES+): m/z 498.3, 499.3 and 500.3 (M + 1).
EXAMPLE 4.1
SYNTHESIS OF 1-(4-(4-(6-AMINO-5-(3-PHENYL-1,2,4-OXADIAZOL-5-YL)PY IDIN-3-YL)- l E
Figure imgf000096_0001
Following the procedure as described in Example 4, making variations using 3-(3-phenyl- 1 ,2,4-oxadiazol-5-yl)-5-(l -(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin- 2-amine to replace 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-piperidin-4- yl)-lH-pyrazol-4-yl)pyridin-2-amine to react with acetic anhydride, the title compound was obtained as a pale yellow solid in 60% yield. 1H NMR (400 MHz, CDC13): £8.46 (d, J = 2.4 Hz, 1H), 8.33 (d, J = 2.4 Hz, 1H), 8.18-8.13 (m, 2H), 7.78 (s, 1H), 7.68 (s, 1H), 7.57-7.50 (m, 3H), 6.67 (br s, 2H), 4.82-4.74 (m, 1H), 4.44-4.35 (m, 1H), 4.04- 3.96 (m, 1H), 3.32-3.22 (m, 1H), 2.85-2.76 (m, 1H), 2.32-2.19 (m, 2H), 2.16 (s, 3H), 1.90-1.74 (m, 2H); MS (ES+): m/z 430.5 (M + 1).
EXAMPLE 5
SYNTHESIS OF 3-(5-(2,6-DICHLOROPHENYL)-1,3,4-OXADIAZOL-2-YL)-5-(1-(1- METHYLPIPERIDIN-4-YL)-LH-PYRAZOL-4-YL)PYRIDINE-2-AMINE
Figure imgf000096_0002
To a cooled (0 °C) solution of 3-(5-(2,6-dichlorophenyl)-l,3,4- oxadiazol-2-yl)-5-(l-piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine (62.0 mg, 0.136 mmol) in dichloromethane/methanol (2 mL/2 mL) was added 37% formaldehyde in water (0.0110 mL, 0.148 mmol). Sodium cyanoborohydride (11.0 mg, 0.148 mmol) was added 10 minutes later. The mixture was stirred at 0 °C for 1 h, then at ambient temperature for 1 h, and evaporated. The residue was purified by flash chromatography eluted with 3-5% methanol in dichloromethane containing 0.1% ammonium hydroxide to afford the title compound as a yellow solid in 81% yield (52 mg). 1H NMR (400 Hz, CDCls): (5 8.41 (d, J= 2.4 Hz, 1H), 8.11 (d, J= 2.4 Hz, 1H), 7.22 (s, 1H), 7.66 (s, 1H), 7.53-7.48 (m, 3H), 6.62 (br s, 2H), 4.25-4.15 (m, 1H), 3.22-3.03 (m, 2H), 2.40 (s, 3H), 2.34-2.20 (m, 4H), 2.20-2.08 (m, 2H); MS (ES+): m/z 470.3, 471.3 and 472.4 (M + 1).
The table below shows additional examples of compounds of Formula
(IA4) that were prepared according to the above process.
Figure imgf000097_0001
Figure imgf000098_0001
an . + .
EXAMPLE 6
SYNTHESIS OF 1-(4-(4-(6-AMINO-5-(5-(2,6-DICHLOROPHENYL)-1,3,4-OXADIAZOL-2-
YL)PYRIDIN-3 -YL)- lH-PYRAZOL- 1 -YL)PIPERIDIN- 1 -YL)-4-METHYLPENTAN- 1 -ONE
Figure imgf000098_0002
To a mixture of 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l- (piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine (63.0 mg, 0.138 mmol), 4- methylpentanoic acid (0.021 mL, 0.166 mmol) and l-[bis(dimethylamino)methylene]- lH-l,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (78.7 mg, 99%, 0.20 mmol) dissolved in N,N-dimethylformamide (1.0 mL) was added triethylamine (0.039 mL, 0.276 mmol). The resulting mixture was stirred at ambient temperature overnight, and diluted with ethyl acetate (30 mL), washed with brine (2 x 30 mL), and dried over sodium sulfate. After filtration and removal of the solvent, the residue was purified by column chromatography eluted with dichloromethaneimethanohNlHLOH (from 200:6: 1 to 200:8:1) to afford the title compound as a yellow solid in 39% yield (30.0 mg). 1H NMR (400 MHz, CDC13): δ 8.38 (d, J= 2.4 Hz, 1H), 8.12 (d, J= 2.4 Hz, 1H), 7.73 (s, 1H), 7.64 (s, 1H), 7.51-7.49 (m, 3H), 6.86-6.60 (br s, 2H), 4.80-4073 (m, 1H), 4.40- 4.35 (m, 1H), 4.04-3.98 (m, 1H), 3.25-3.18 (m, 1H), 2.80-2.71 (m, 1H), 2.38-2.32 (m, 2H), 2.30-2.14 (m, 2H), 2.03-1.88 (m, 2H), 1.67-1.49 (m, 3H), 0.91 (d, J= 6.4 Hz, 6H). MS: (ES+): m/z 554.4, 555.3, and 556.3 (M + 1), 576.3, 577.3 and 578.3 (M + Na).
EXAMPLE 7
SYNTHESIS OF (4-(4-(6-AMiNO-5-(3-PHENYL-l,2,4-oxADiAZOL-5-YL)PYMDiN-3-YL)-lH- PYRAZ ANONE
Figure imgf000099_0001
To a solution of 3-(3-phenyl-l,2,4-oxadiazol-5-yl)-5-(l-(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin-2-amine (60 mg, 0.15 mmol) in 1 mL of pyridine was added cyclopropanecarbonyl chloride (28 xL, 0.30 mmol). The resulting mixture was stirred at ambient temperature for 2 hours, followed by the addition of 20 mL of water to yield yellow precipitates. The precipitates were collected by filtration, washed with water and purified by column chromatography eluted with 5% methanol in dichloromethane to afford the title compound as a yellow solid in 66% yield (45 mg). 1H NMR (400 MHz, CDC13): 8.46 (d, J= 2.4 Hz, 1H), 8.33 (d, J= 2.4 Hz, 1H), 8.18-8.13 (m, 2H), 7.79 (s, 1H), 7.70 (s, 1H), 7.57-7.50 (m, 3H), 6.69 (br s, 2H), 4.85-4.73 (m, 1H), 4.48-4.35 (m, 2H), 3.39-3.25 (m, 1H), 2.90-2.76 (m, 1H), 2.38-2.20 (m, 2H), 2.15-1.95 (m, 2H), 1.84- 1.76 (m, 1H), 1.05-0.98 (m, 2H), 0.84-0.78 (m, 2H); MS (ES+): m/z 456.5 (M + 1).
EXAMPLE 7.1
SYNTHESIS OF (4-(4-(6-AMINO-5-(5-(2,6-DICHLOROPHENYL)-1,3,4-OXADIAZOL-2- YL)P YRIDIN-3 - YL)- 1 - YL)(CYCLOPROP YL)METHANONE
Figure imgf000099_0002
Following the procedure as described in Example 7, making variations using 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol- 4-yl)-pyridin-2-amine to replace 3-(3-phenyl-l,2,4-oxadiazol-5-yl)-5-(l-(piperidin-4- yl)-lH-pyrazol-4-yl)pyridin-2-amine to react with cyclopropanecarbonyl chloride, the title compound was obtained as a yellowish solid in 41% yield. 1H NMR (400 MHz, CDCI3): 8.40 (d, J = 2.0 Hz, 1H), 8.14 (d, J = 2.0 Hz, 1H), 7.74 (s, 1H), 7.65 (s, 1H), 7.54-7.47 (m, 3H), 6.82 (br s, 2H), 4.82-4.69 (m, 1H), 4.46-4.32 (m, 2H), 3.38-3.22 (m, 1H), 2.88-2.74 (m, 1H), 2.36-2.14 (m, 2H), 2.10-1.90 (m, 2H), 1.82-1.74 (m, 1H), 1.04- 0.96 (m, 2H), 0.82-0.75 (m, 2H); MS (ES+): m/z 524.3, 525.3 and 526.3 (M + 1).
EXAMPLE 7.2
SYNTHESIS OF (4-(4-(6-AMINO-5-(5-(2,6-DICHLO OPHENYL)-1,3,4-OXADIAZOL-2- YL)P YRIDIN-3 - YL)- 1 H-P YRAZOL- 1 - YL)PIPERIDIN- 1 - YL)(PHENYL)METHANONE
Figure imgf000100_0001
Following the procedure as described in Example 7, making variations using 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol- 4-yl)-pyridin-2-amine to replace 3-(3-phenyl-l,2,4-oxadiazol-5-yl)-5-(l-(piperidin-4- yl)-lH-pyrazol-4-yl)pyridin-2-amine to react with benzoyl chloride, the title compound was obtained as a yellowish solid in 83% yield. 1H NMR (400 MHz, CDC13): £8.42 (s, 1H), 8.12 (s, 1H), 7.75 (s, 1H), 7.67 (s, 1H), 7.53-7.49 (m, 3H), 7.46-7.39 (m, 5H), 6.74 (br s, 2H), 4.95-4.75 (m, 1H), 4.47-4.37 (m, 1H), 4.05-3.78 (m, 1H), 3.25-1.95 (m, 2H), 2.35-2.18 (m, 2H), 2.18-1.98 (m, 2H); MS (ES+): m/z 560.3, 561.3 and 562.3 (M + 1).
EXAMPLE 8
SYNTHESIS OF 5-(l-CYCL0HEXYL-lH-PYRAZ0L-4-YL)-3-(5-PHENYL-l,2,4-0XADiAZ0L-3-
Figure imgf000100_0002
In a sealed tube, a mixture of 5-bromo-3-(5-phenyl-l,2,4-oxadiazol-3- yl)pyridin-2-amine (70.0 mg, 0.221 mmol), l-cyclohexyl-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (61.0 mg, 0.221 mmol) and cesium carbonate (145 mg, 0.442 mmol) in 1 ,4-dioxane/water (2.0 mL, 4:1) was degassed with nitrogen gas for 10 minutes, followed by the addition of tetrakis(triphenylphosphine)palladium (26.0 mg, 22.1 μηιοΐ). The mixture was degassed for another 10 minutes. The resulting mixture was sealed and heated at 100 °C overnight. After cooled to ambient temperature, the reaction mixture was filtered through a celite cake, and washed with dichloromethane and methanol. The filtrate was concentrated and the residue was treated with dichloromethane/methanol, and filtered. The filtrate was concentrated and the residue was purified by column chromatography eluted with dichloromethane :methanol from 100:1 to 50:1 to afford the title compound as a white solid in 41% yield (35 mg). 1H
NMR (400 MHz, CDC13): δ 8.52 (d, J= 2.4 Hz, 1H), 8.40 (d, J= 2.4 Hz, 1H), 8.28- 8.23 (m, 2H), 7.79 (s, 1H), 7.70 (s, 1H), 7.68-7.63 (m, 1H), 7.62-7.56 (m, 2H), 6.25- 6.10 (br s, 2H), 4.21-4.12 (m, 1H), 2.27-2.20 (m, 2H), 2.00-1.90 (m, 2H), 1.85-1.74 (m, 3H), 1.54-1.40 (m, 2H), 1.37-1.25 (m, 1H). MS (ES+): m/z 387.3 (M + 1), 409.4 (M + Na).
EXAMPLE 9
SYNTHESIS OF 3 -(5 -(2,5 -DICHLO OPHENYL)- 1 ,2,4-OXADIAZOL-3 -YL)-5 -( 1 -(PIPERIDIN-4-
Figure imgf000101_0001
A. A mixture of 5-bromo-3-(5-cyclohexyl-l,3,4-oxadiazol-2- yl)pyridin-2-amine (80 mg, 0.208 mmol), tert-butyl 4-(4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazol-l-yl)piperidine-l-carboxylate (94 mg, 0.249 mmol) and cesium carbonate (338 mg, 1.04 mmol) in a sealed tube with 5 mL of 10: 1
dioxane:water was degassed with nitrogen for 10 minutes, followed by the addition of tetrakis(triphenylphosphine)-palladium(0) (24 mg, 0.021 mmol). The mixture was degassed for another 10 minutes and then sealed and heated at 110 °C overnight. After removal of the solvents, the residue was purified by column chromatography and preparative thin layer chromatography eluted with 40% ethyl acetate in hexane to afford tert- vXy\ 4-(4-(6-amino-5-(5-(2,5-dichlorophenyl)- 1 ,2,4-oxadiazol-3-yl)pyridin-3-yl)- lH-pyrazol-l-yl)piperidine-l-carboxylate as a yellowish solid in 30% yield (33 mg). 1H NMR (400 MHz, CDC13): £8.48 (d, J= 2.4 Hz, 1H), 8.40 (d, J= 2.4 Hz, 1H), 8.19 (d, J = 2.4 Hz, 1H), 7.80 (s, 1H), 7.69 (s, 1H), 7.57 (d, J= 8.8 Hz, 1H), 7.53 (dd, J= 8.8, 2.4 Hz, 1H), 6.19 (br s, 2H), 4.38-4.20 (m, 1H), 3.30-3.27 (m, 2H), 2.95-2.80 (m, 2H), 2.23-2.18 (m, 2H), 2.05-1.95 (m, 2H), 1.48 (s, 9H).
B. To a suspension of tert-butyl 4-(4-(6-amino-5-(5-(2,5- dichlorophenyl)- 1 ,2,4-oxadiazol-3-yl)pyridin-3-yl)- lH-pyrazol-1 -yl)piperidine- 1 - carboxylate (20 mg, 0.15 mmol) in 5 mL of dichloromethane was added 3 mL of 2M HC1 in ether. The resulting mixture was stirred at room temperature overnight. After removal of the solvents in vacuo, the residue was purified by preparative thin layer chromatography eluted with 1 : 10:89 ammonium hydroxide :methanol: dichloromethane to afford the title compound as a white solid in 68% yield (11 mg). 1H NMR (400 MHz, CDCls): 8.49 (d, J= 2.4 Hz, 1H), 8.41 (d, J= 2.4 Hz, 1H), 8.19 (d, J= 2.4 Hz, 1H), 7.79 (s, 1H), 7.71 (s, 1H), 7.57 (d, J= 8.8 Hz, 1H), 7.52 (dd, J= 8.8, 2.4 Hz, 1H), 6.19 (br s, 2H), 4.28 (m, 1H), 3.30-3.27 (m, 2H), 2.85-2.75 (m, 2H), 2.26-2.20 (m, 2H), 2.02-1.91 (m, 2H); MS (ES+): m/z 456.3 and 458.3 (M + 1).
EXAMPLE 10
SYNTHESIS OF 3-(5-PHENYL-l,2,4-oxADiAZOL-3-YL)-5-(l-(TETRAHYD O-2H-PYRAN-4-
Y E
Figure imgf000102_0001
A mixture of 5-bromo-3-(5-phenyl-l,2,4-oxadiazol-3-yl)pyridin-2-amine (50 mg, 0.16 mmol), l-(tetrahydro-2H-pyran-4-yl)-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (55 mg, 0.20 mmol), and cesium carbonate (0.22 g, 0.82 mmol) in 3 mL of 10: 1 dioxane: water in a sealed tube was purged with nitrogen gas for 10 minutes, followed by the addition of tetrakis(triphenylphosphine)- palladium(O) (10 mg, 0.008 mmol). The mixture was purged for another 10 minutes and heated at 110 °C overnight. After removal of the solvents, the residue was purified by column chromatography and preparative thin layer chromatography eluted with 2% methanol in dichloromethane to afford the title compound as a white solid in 39% yield (25 mg). 1H NMR (400 MHz, CDC13): £8.52 (d, J= 2.4 Hz, 1H), 8.40 (d, J= 2.0 Hz, 1H), 8.28-8.23 (m, 2H), 7.81 (s, 1H), 7.72 (s, 1H), 7.68-7.63 (m, 1H), 7.62-7.56 (m, 2H), 6.20 (br s, 2H), 4.47-4.37 (m, 1H), 4.20-4.10 (m, 2H), 3.63-3.54 (m, 2H), 2.23- 2.10 (m, 4H); MS (ES+): m/z 389.2 (M + 1). EXAMPLE 11
SYNTHESIS OF 3-(5-PHENYL-l,2,4-oxADiAZOL-3-YL)-5-(l-(piPERiDiN-4-YL)-lH-
Figure imgf000103_0001
A. A mixture of 5-bromo-3-(5-phenyl-l,2,4-oxadiazol-3-yl)pyridin-
2-amine (100 mg, 0.33 mmol), tert-butyl 4-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)-lH-pyrazol-l-yl)piperidine-l-carboxylate (0.15 mg, 0.40 mmol), and cesium carbonate (0.54 g, 1.65 mmol) in 5 mL of 10: 1 dioxane:water in a sealed tube was purged with nitrogen gas for 10 min, followed by the addition of tetrakis(triphenyl- phosphine)palladium(O) (19 mg, 0.05 mmol). The mixture was purged for another 10 minutes and then heated at 110 °C overnight. After removal of the solvents, the residue was purified by column chromatography eluted with 2% methanol in dichloromethane to afford tert-butyl 4-(4-(6-amino-5-(5-phenyl-l,2,4-oxadiazol-3-yl)pyridin-3-yl)-lH- pyrazol-l-yl)piperidine-l-carboxylate as a yellowish solid in 63% yield (100 mg). 1H NMR (400 MHz, CDCls): £8.51 (d, J= 2.4 Hz, 1H), 8.39 (d, J= 2.4 Hz, 1H), 8.28- 8.23 (m, 2H), 7.81 (s, 1H), 7.70 (s, 1H), 7.68-7.63 (m, 1H), 7.62-7.57 (m, 2H), 6.19 (br s, 2H), 4.47-4.20 (m, 3H), 3.00-2.85 (m, 2H), 2.24-2.10 (m, 2H), 2.05-1.92 (m, 2H), 1.48 (s, 9H).
B. To a suspension of tert-butyl 4-(4-(6-amino-5 -(5 -phenyl- 1,2,4- oxadiazol-3-yl)pyridin-3-yl)-lH-pyrazol-l-yl)piperidine-l-carboxylate (100 mg, 0.20 mmol) in 5 mL of dichloromethane was added 3 mL of 2M HC1 in ether. The resulting mixture was stirred at room temperature for 2 hours, then diluted with 20 mL of dichloromethane, and washed with saturated sodium bicarbonate solution. The organic solution was collected and dried over anhydrous sodium sulfate. After filtration and removal of the solvent in vacuo, the residue was purified by column chromatography eluted with 1 : 10:89 ammonium hydroxide :methanol: dichloromethane to afford the title compound as a yellow solid in 50% yield (40 mg). 1H NMR (400 MHz, CDC13): £8.52 (d, J= 2.4 Hz, 1H), 8.39 (d, J= 2.4 Hz, 1H), 8.28-8.23 (m, 2H), 7.80 (s, 1H), 7.72 (s, 1H), 7.68-7.63 (m, 1H), 7.62-7.57 (m, 2H), 6.19 (br s, 2H), 4.33-4.24 (m, 1H), 3.33- 3.24 (m, 2H), 2.86-2.76 (m, 2H), 2.26-2.18 (m, 2H), 2.02-1.90 (m, 2H); MS (ES+): m/z 388.4 (M + 1). EXAMPLE 12
SYNTHESIS OF 1-(4-(4-(6-AMINO-5-(5-(2,6-DICHLOROPHENYL)-1,3,4-OXADIAZOL-2- YL)PYRIDIN-3 -YL)- 1H-PYRAZOL- 1 -YL)PIPERIDIN- 1 -YL)-2-PHENYLETHANONE
Figure imgf000104_0001
To a suspension of 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-
(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine (50 mg, 0.11 mmol) in 5 mL of dichloromethane at room temperature was added triethylamine (77 μί, 0.55 mmol), followed by the addition of phenylacetyl chloride (18 μΐ,, 0.13 mmol). The resulting mixture was stirred at room temperature for 1 hour. After removal of the solvent in vacuo, the residue was purified by column chromatography and preparative thin layer chromatography eluted with neat ethyl acetate to afford the title compound as a yellow solid in 34% yield (21 mg). 1H NMR (400 MHz, CDC13): δ 8.40 (d, J= 2.4 Hz, 1H), 8.10 (d, J= 2.4 Hz, 1H), 7.72 (s, 1H), 7.56 (s, 1H), 7.53-7.49 (m, 3H), 7.36-7.32 (m, 2H), 7.29-7.24 (m, 3H), 6.64 (br s, 2H), 4.83-4.73 (m, 1H), 4.48-4.38 (m, 1H), 4.07- 3.97 (m, 1H), 3.78 (s, 2H), 3.22-3.11 (m, 1H), 2.86-2.75 (m, 1H), 2.25-2.15 (m, 1H), 2.15-2.05 (m, 1H), 2.00-1.86 (m, 1H), 1.76-1.60 (m, 1H); MS (ES+): m/z 596.2, 597.2 and 598.2 (M + Na).
EXAMPLE 12.1
SYNTHESIS OF (4-(4-(6-AMiNO-5-(3-PHENYL-l,2,4-oxADiAZOL-5-YL)PYRiDiN-3-YL)-lH- PYRAZOL- 1 -YL)PIPERIDIN- 1 -YL)(PHENYL)METHANONE
Figure imgf000104_0002
Following the procedure as described in Example 12, making variations using 3-(3-phenyl- 1 ,2,4-oxadiazol-5-yl)-5-(l -(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin- 2-amine to replace 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-piperidin-4- yl)-lH-pyrazol-4-yl)pyridin-2-amine to react with benzoyl chloride, the title compound was obtained as a pale yellowish solid in 45% yield. 1H NMR (400 MHz, CDC13): δ 8.47 (d, J= 2.4 Hz, 1H), 8.35 (d, J= 2.4 Hz, 1H), 8.19-8.15 (m, 2H), 7.80 (s, 1H), 7.71 (s, 1H), 7.57-7.53 (m, 3H), 7.47-7.43 (m, 5H), 6.68 (br s, 2H), 4.98-4.82 (m, 1H), 4.50- 4.41 (m, IH), 4.10-3.95 (m, IH), 3.25-3.00 (m, 2H), 2.38-2.20 (m, 2H), 2.20-2.03 (m, 2H); MS (ES+): m/z 492.4 (M + 1).
EXAMPLE 12.2
SYNTHESIS OF 1-(4-(4-(6-AMINO-5-(3-PHENYL-1,2,4-OXADIAZOL-5-YL)PY IDIN-3-YL)- lH-PYRAZOL- 1 -YL)PIPERIDIN- 1 -YL)-2-PHENYLETHANONE
Figure imgf000105_0001
Following the procedure as described in Example 12, making variations using 3-(3-phenyl- 1 ,2,4-oxadiazol-5-yl)-5-(l -(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin- 2-amine to replace 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-piperidin-4- yl)-lH-pyrazol-4-yl)pyridin-2-amine to react with phenylacetyl chloride, the title compound was obtained as a pale yellowish solid in 49% yield. 1H NMR (400 MHz, CDC13): δ 8.43 (d, J= 2.4 Hz, IH), 8.31 (d, J= 2.4 Hz, IH), 8.19-8.13 (m, 2H), 7.75 (s, IH), 7.59 (s, IH), 7.56-7.49 (m, 3H), 7.39-7.24 (m, 5H), 6.68 (br s, 2H), 4.85-4.75 (m, IH), 4.40-4.28 (m, IH), 4.08-3.98 (m, IH), 3.80 (s, 2H), 3.25-3.12 (m, IH), 2.88-2.78 (m, IH), 2.28-2.17 (m, IH), 2.17-2.06 (m, IH), 2.02-1.89 (m, IH), 1.78-1.65 (m, IH); MS (ES+): m/z 506.4 (M + 1).
EXAMPLE 13
SYNTHESIS OF 3-(5-PHENYLOXAZOL-2-YL)-5-(1 -(TETRAHYDRO-2H-PYRAN-4-YL)- 1H-
Figure imgf000105_0002
A mixture of 5-bromo-3-(5-phenyloxazol-2-yl)pyridin-2-amine (60 mg, 0.20 mmol), l-(tetrahydro-2H-pyran-4-yl)-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)-lH-pyrazole (56 mg, 0.20 mmol) and cesium carbonate (0.32 g, 1.00 mmol) in a sealed tube with 5 mL of 10: 1 dioxane: water was degassed for 10 min. After the addition of tetrakis(triphenylphosphine)palladium(0) (23 mg, 0.02 mmol), the mixture was degassed for another 10 minutes. The resulting mixture was sealed and heated at 100 °C overnight. After removal of the solvent in vacuo, the residue was purified by column chromatography eluted with ethyl acetate to afford the title compound as a yellow solid in 18% yield (14 mg). 1H NMR (400 MHz, CDC13): δ 8.32 (d, J= 2.4 Hz, 1H), 8.24 (d, J= 2.4 Hz, 1H), 7.79 (s, 1H), 7.77-7.72 (m, 2H), 7.69 (s, 1H), 7.50-7.44 (m, 3H), 7.42-7.36 (m, 1H), 6.68 (br s, 2H), 4.50-4.38 (m, 1H), 4.20-4.13 (m, 2H), 3.63-3.53 (m, 2H), 2.22-2.15 (m, 4H); MS (ES+): m/z 388.3 (M + 1).
The table below shows additional examples of compounds of Formula
(IA1) that were prepared according to the above process.
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
EXAMPLE 14
SYNTHESIS OF 3-(5-PHENYLOXAZOL-2-YL)-5-(1 -(PIPERIDIN-4-YL)- lH-PYRAZ0L-4-
Figure imgf000108_0002
To a solution of tert-butyl 4-(4-(6-amino-5-(5-phenyloxazol-2- yl)pyridin-3-yl)-lH-pyrazol-l-yl)piperidine-l-carboxylate (60 mg, 0.12 mmol) in 4 mL of dichloromethane was added 1.0 mL of 2 M hydrochloric acid in ethyl ether. The resulting mixture was stirred at ambient temperature overnight, washed with saturated sodium bicarbonate (3 x 20 mL) and the organic layer was separated from the aqueous layer. The aqueous solution was extracted with dichloromethane (2 x 20 mL). The combined organic solution was dried over sodium sulfate and filtered. After removal of the solvent of the organic layer in vacuo, the residue was purified by preparative thin layer chromatography eluted with 1 : 10:89 ammonium hydroxide :methanol: dichloro- methane to afford the title compound as a yellow solid in 56% yield (26 mg). 1H NMR (400 MHz, CD3OD): δ 8.38 (d, J= 2.4 Hz, 1H), 8.27 (d, J= 2.4 Hz, 1H), 8.10 (s, 1H), 7.86 (s, 1H), 7.85-7.80 (m, 2H), 7.63 (s, 1H), 7.49-7.43 (m, 2H), 7.39-7.33 (m, 1H), 4.42-4.32 (m, 1H), 3.30-3.25 (m, 2H), 2.90-2.80 (m, 2H), 2.22-2.14 (m, 2H), 2.10-1.95 (m, 2H); MS (ES+): m/z 387.4 (M + 1).
The table below shows additional examples of compounds of Formula (IA1) that were prepared according to the above process.
Figure imgf000109_0001
Figure imgf000110_0001
EXAMPLE 15
SYNTHESIS OF 1-(4-(4-(6-AMINO-5-(5-(4-CHLOROPHENYL)OXAZOL-2-YL)PYRIDIN-3-YL)-
Figure imgf000110_0002
To a solution of 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin-2-amine (60 mg, 0.14 mmol) in 5 mL of dichloromethane at 0 °C was added acetic anhydride (13 \L, 0.14 mmol), followed by the addition of triethylamine (99 xL, 0.71 mmol). The resulting mixture was stirred at 0 °C for 30 min, then warmed up to ambient temperature and stirred for 3 hours. The solvent was removed in vacuo, and the residue was purified by column chromatography eluted with 5% methanol in dichloromethane followed by recrystallization from methanol to afford the title compound as a yellowish solid in 51% yield (33 mg). lU NMR (400 MHz, DMSO-de): δ 8.40 (d, J= 2.4 Hz, 1H), 8.37 (d, J= 2.4 Hz, 1H), 8.27 (s, 1H), 7.96-7.92 (m, 4H), 7.57-7.54 (m, 2H), 7.34 (br s, 2H), 4.49-4.37 (m, 2H), 3.95-3.87 (m, 1H), 3.25-3.16 (m, 1H), 2.75-2.67 (m, 1H), 2.12-2.03 (m, 2H), 2.02 (s, 3H), 1.94-1.85 (m, 1H), 1.81-1.70 (m, 1H); MS (ES+): m/z 463.4 and 465.4 (M + 1).
EXAMPLE 15.1
SYNTHESIS OF 1 -(4-(4-(6-AMINO-5 -(5 -(3 -CHLOROPHENYL)OXAZOL-2-YL)PYRIDIN-3 -YL)- 1H-PYRAZOL- 1 -YL)PIPERIDIN- 1 -YL)ETHANONE
Figure imgf000110_0003
Following the procedure as described in Example 15, making variations using 3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin- 2-amine to replace 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol- 4-yl)pyridin-2-amine to react with acetic anhydride, the title compound was obtained as a yellow solid in 65% yield. 1H NMR (400 MHz, CDC13): δ 8.32 (d, J= 2.4 Hz, 1H), 8.22 (d, J= 2.4 Hz, 1H), 7.80 (s, 1H), 7.74-7.71 (m, 1H), 7.69 (s, 1H), 7.64-7.60 (m, 1H), 7.50 (s, 1H), 7.45-7.39 (m, 1H), 7.37-7.33 (m, 1H), 6.70 (br s, 2H), 4.84-4.76 (m, 1H), 4.47-4.37 (m, 1H), 4.06-3.97 (m, 1H), 3.32-3.22 (m, 1H), 2.85-2.76 (m, 1H), 2.35- 2.20 (m, 2H), 2.17 (s, 3H), 2.10-1.98 (m, 2H); MS (ES+): m/z 485.3 and 487.3 (M + Na).
EXAMPLE 16
SYNTHESIS OF (4-(4-(6-AMINO-5-(5-(4-CHLO OPHENYL)OXAZOL-2-YL)PY IDIN-3-YL)- lH-PY ANONE
Figure imgf000111_0001
To a solution of 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin-2-amine (60 mg, 0.14 mmol) in 1 mL of pyridine was added cyclopropanecarbonyl chloride (15 xL, 0.17 mmol). The resulting mixture was stirred at ambient temperature for 2 hours. The solvent was removed in vacuo, and the residue was purified by column chromatography eluted with 5% methanol in dichloromethane followed by recrystallization from a mixed solvent of methanol and water to afford the title compound as an orange solid in 63% yield (43 mg). 1H NMR (400 MHz, CDC13): δ 8.31 (s, 1H), 8.21 (s, 1H), 7.78 (s, 1H), 7.70-7.63 (m, 3H), 7.48-7.40 (m, 3H), 6.72 (br s, 2H), 4.85-4.70 (m, 1H), 4.50-4.35 (m, 2H), 3.40-3.22 (m, 1H), 2.91-2.75 (m, 1H), 2.40-2.20 (m, 2H), 2.15-1.90 (m, 2H), 1.85-1.75 (m, 1H), 1.05-0.95 (m, 2H), 0.85-0.74 (m, 2H); MS (ES+): m/z 511.4 and 513.4 (M + Na).
The table below shows additional examples of compounds of Formula (IA1) that were prepared according to the above process.
Figure imgf000112_0001
Ill Ex. Starting Material and
Chemical Name Characterization Data No. Yield
l-(4-(4-(6-
¾ NMR (400 MHz, CDC13): δ 8.32 (d, amino-5-(5-(3- J = pivaloyl chloride and 3- 2.4 Hz, 1H), 8.22 (d, J= 2.4 Hz, 1H), 7.79 chlorophenyl)ox
(5-(3- (s, 1H), 7.74-7.72 (m, 1H), 7.69 (s, 1H), azol-2- chlorophenyl)oxazol-2- 7.64- 7.60 (m, 1H), 7.50 (s, 1H), 7.44-7.40 yl)pyridin-3-yl)-
16.6 yl) - 5 - ( 1 - (piperidin-4 - (m, 1H), 7.37-7.33 (m, 1H), 6.67 (br s, 2H), l /-pyrazol-l- yl)- li/-pyrazol-4- 4.65- 4.55 (m, 2H), 4.48-4.35 (m, 1H), 3.08- yl)piperidin-l- yl)pyridin-2 - amine ; 2.95 (m, 2H), 2.30-2.23 (m, 2H), 2.10-1.97 yl)-2,2- 10% (m, 2H), 1.33 (s, 9H); MS (ES+): m/z 505.5 dimethylpropan- and 507.5 (M + 1).
1-one
(4-(4-(6-amino-
¾ NMR (400 MHz, CDC13): δ 8.32 (d, J =
5-(5-(3-
4-fluorobenzoyl 2.4 Hz, 1H), 8.22 (d, J= 2.4 Hz, 1H), 7.80 chlorophenyl)ox
chloride and 3-(5-(3- (s, 1H), 7.74-7.72 (m, 1H), 7.70 (s, 1H), azol-2- chlorophenyl)oxazol-2- 7.64-7.60 (m, 1H), 7.51-7.46 (m, 3H), 7.44- yl)pyridin-3-yl)-
16.7 yl) - 5 - ( 1 - (piperidin-4 - 7.39 (m, 1H), 7.37-7.33 (m, 1H), 7.16-7.09 l /-pyrazol-l- yl)- li/-pyrazol-4- (m, 2H), 6.70 (br s, 2H), 4.48-4.35 (m, 2H), yl)piperidin-l- yl)pyridin-2 - amine ; 4.51-4.42 (m, 1H), 3.22-3.04 (m, 2H), 2.34- yl)(4- 12% 2.20 (m, 2H), 2.19-2.05 (m, 2H); MS (ES+): iluorophenyl)met
m/z 543.3 and 545.3 (M + 1).
hanone
EXAMPLE 17
SYNTHESIS OF 3 -(5 -(3 -CHLOROPHENYL)OXAZOL-2-YL)-5 -( 1 -( 1 -METHYLPIPERIDIN-4-YL)-
L E
Figure imgf000113_0001
To a solution of 3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)- lH-pyrazol-4-yl)pyridin-2-amine (50 mg, 0.12 mmol) in 4 mL of dichloromethane and methanol (1 : 1) at 0 °C was added formaldehyde (10 \L, 0.14 mmol), followed by the addition of sodium cyanoborohydride (9 mg, 0.14 mmol). The resulting mixture was warmed up to ambient temperature and stirred for another hour. The solvent was removed in vacuo and the residue was purified by column chromatography eluted with 1 : 10:89 ammonium hydroxide :methanol : dichloromethane to afford the title compound as a yellow solid in 83% yield (43.2 mg). 1H NMR (400 MHz, CDC13): £8.32 (d, J = 2.4 Hz, 1H), 8.21 (d, J= 2.4 Hz, 1H), 7.78 (s, 1H), 7.73-7.68 (m, 2H), 7.63-7.58 (m, 1H), 7.49 (s, 1H), 7.43-7.38 (m, 1H), 7.38-7.32 (m, 1H), 6.67 (br s, 2H), 4.26-4.16 (m, 1H), 3.09-3.00 (m, 2H), 2.38 (s, 3H), 2.30-2.10 (m, 6H); MS (ES+): m/z 435.3 and 437.3 (M + 1).
The table below shows additional examples of compounds of Formula (IA4) that were prepared according to the above process.
Figure imgf000114_0001
BIOLOGICAL EXAMPLES
EXAMPLE 18
KINASE ENZYMATIC ACTIVITY ASSAYS
Preparation of active recombinant kinase proteins:
Recombinant human Tyro3 (455 -end, the gene accession number NM_006293), human Axl (473-end, the gene accession number NM_021913) and human Mer (578-872, the gene accession number NM_006343) were independently expressed by baculovirus in Sf9 insect cells using an N-terminal GST tag. The recombinant proteins were stored at -70 °C in a medium containing 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 10 mM glutathione, 0.1 mM EDTA, 0.25 mM DTT, 0.1 mM PMSF and 25% glycerol. The recombinant proteins were aliquoted into smaller quantities after centrifugation to avoid repeated handling and multiple freeze/thaw cycles for the most favorable performance.
Preparation of assay reagents:
Kinase Assay Buffer: This buffer solution consisted of the following components: 25 mM MOPS, pH 7.2, 12.5 mM β-glycerol-phosphate, 25 mM MgCl2, 5 mM EGTA, 2 mM EDTA and DTT 0.25 mM which was added prior to use.
Kinase Dilution Buffer: Kinase Assay Buffer was diluted at a 1 :4 ratio (5x dilution) with distilled water.
[33P]-ATP assay cocktail: In a designated radioactive working area, a 250 μΜ [33P]-ATP assay cocktail was prepared by the addition of the following components: 150 μΕ of 10 mM ATP stock solution, 100 μΕ [33P]-ATP (1 mci/100 iL), 5.75 mL of kinase assay buffer. This solution was stored in 1 mL aliquots at -20 °C.
ATP stock solution (10 mM): The ATP stock solution was prepared by dissolving 55 mg of ATP in 10 mL of kinase assay buffer. It was stored in 200 aliquots at -20 °C.
Active Kinase Stock Solution: The active recombinant kinase protein (0.1 μg/μL) was diluted with Kinase Dilution Buffer and the activity was assayed using a serial dilution method. The specific activity is expressed in nmoL/min/mg.
Substrates: poly(4:l glu:tyr) is the substrate used for each kinase Tyro3, Axl and Mer: The peptide substrate was dissolved in water to give the final concentration of 1 mg/mL.
Test compound solution: Test compound was dissolved in DMSO to obtain a 10 mM solution. The assay solution was prepared by adding 5 μί of this solution to 955 μΐ, of 10% DMSO/water to obtain the final concentration of 50 μΜ. Assay procedure:
The enzymatic activity of all three kinases was determined as follows. Using a 96-well plate, the wells were divided into three categories: Blank wells, Background wells and Test wells. In Test wells, 5 μί of the test compound solution and 5 μΙ_, of the substrate solution were added. In Control wells, 5 μΙ_, of 10%
DMSO/water and 5 μΙ_, of the substrate solution were added. In Blank wells, 10 of 10%) DMSO/water was added. To each well was added 10 μί of Active Kinase Stock Solution to make up the volume in each well to 20 μί. All test samples, controls and blanks were run in duplicate. The reaction was initiated by the addition of 5 μί of [33P]-ATP assay cocktail, bringing the final volume up to 25 [iL in every well. The mixture was incubated at room temperature for 30 minutes. The reaction was terminated by transferring 10 μί of the reaction mixture into a Millipore MultiScreen filter plate (cat. number MSPHN6B50). The filter plate was washed in a 1% phosphoric acid solution with constant gentle shaking for 15 minutes and this step was repeated once. After the plate was dried in air, scintillation fluid was added to each well and the radioactivity in each well in CPM was counted by a Microbeta TriLux. The corrected CPM in each test well was determined by subtracting the average value of Blank well values. Percentage of inhibition of the kinase enzymatic activity by the test compound was determined using the following formula:
Average of corrected CPM in Test wells
% Inhibition = 1 - x 100
Average of corrected CPM in Control wells.
The IC50 values were determined in a similar way following a serial dilution of the test compound. The percentage of inhibition at each concentration was calculated following the above formula. The IC50 was estimated from the curve of % Inhibition against Concentration in log unit using Prism 5, version 5.01.
The following table summarizes the inhibitory activity on Tyro3, Axl and Mer of the compounds of the disclosure. "+" represents the IC50 is > 10 μΜ; "++" represents the 1 μΜ < IC50 < 10 μΜ; "+++" represents the 0.5 μΜ < IC50 < 1 μΜ and "++++" represents the IC50 < 0.5 μΜ.
Figure imgf000116_0001
Figure imgf000117_0001
tetra y ro- -pyran- -y - -pyrazo- -y py n- -amne
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
EXAMPLE 19
CELL VIABILITY ASSAY
Cell lines and reagents: A549 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM, HyClone) containing 10% fetal bovine serum (FBS, Life technologies) and maintained in a humidified incubator at 37 °C with 5% C02.
Cell viability assay protocol: 5 x 103 A549 cells in 180 of DMEM containing 0.5%> FBS were seeded in 96-well flat bottom plates (Costar) and incubated in a humidified incubator at 37 °C with 5% C02 for 24 hours. The test compound (20 μί) in different concentrations after a serial dilution was added to the wells to a final volume of 200 per well. The wells with zero compound concentration were used as the Control wells and the Test wells contained different concentrations of the test compound. To the Blank wells were added medium only. After 48 hours incubation, 40 of MTS (Promega) was added to each well and the plates were incubated for 10 minutes to 1 hour at 37 °C. The cell viability was estimated by measurement of optical density at 490 nm using a microplate spectrophotometer (Molecular Devices).
EXAMPLE 20
THYMIDINE INCORPORATION ASSAY
1 x 104 A549 cells in 180 μί of DMEM containing 0.5% FBS were seeded in a 96-well white Isoplate (PerkinElmer) and incubated in a humidified incubator at 37 °C with 5% C02 for 24 hours. The test compound in different concentrations after a serial dilution was added to the wells to a final volume of 200 [iL per well. The wells with zero compound concentration were used as the Control wells and the wells containing different concentrations of the test compound were used as the Test wells. To the Blank wells were added water only. After 24-hour incubation, each well was labeled with 1 μθ of [3H]thymidine (specific activity, 26.8 Ci/mmol,
PerkinElmer), and plates were again incubated at 37 °C overnight. After incubation, 50 of cold trichloroacetic acid was added into each well and the plates were incubated at 4 °C for 1-2 hours. Plates were subsequently washed with distilled water 5 times and air-dried at room temperature. Scintillation liquid was added to each well and radioactivity in CPM was counted using a MicroBeta TriLux (PerkinElmer) counter. The corrected CPM in each well was determined by subtracting the average value of the Blank well values. Percentage of inhibition of the thymidine incorporation at tested concentrations by the test compound was determined using the following formula:
( Average of corrected CPM in Test wells \
1 - x 100
Average of corrected CPM in Control wells/ The IC50 was then estimated from the curve of % Inhibition against concentration in log unit using Prism 5, version 5.01.
EXAMPLE 21
COLONY FORMATION ASSAY (METHOD #1)
A549 cells were treated with the test compound at different concentrations (5, 1 and 0.2 μΜ) in DMEM containing 0.5% FBS at 37 °C for 1 hour. After treatment, cells were seeded in the agar containing the test compound at different concentrations as outlined above in 6-well plates and the plates were incubated at 37 °C for 2 weeks. DMEM containing the various concentrations of the test compound was added on the top of agar and changed every 2-3 days. The viable colonies were stained with crystal violet and counted. This example is to demonstrate the capability of test compounds to prevent the formation of colonies.
EXAMPLE 22
COLONY FORMATION ASSAY (METHOD #2)
17,500 A549 cells were seeded in agar in each well of 6-well plates and cultured in DMEM containing 10% FBS at 37 °C for six days and the colonies were formed. The test compound dissolved in DMEM with different concentrations was added to the test wells of the plates and DMEM with no test compound or with diluted DMSO was added to the control wells. The plates were incubated at 37 °C for 18-20 days during which period the DMEM containing 10% FBS and the various
concentrations of test compounds was changed every 2 to 3 days. The viable colonies were stained with crystal violet and counted. This example is to demonstrate the capability of teat compounds to inhibit the colony growth after they are established and/or to eliminate the established colonies.
EXAMPLE 23
WESTERN BLOT ASSAY
5 x l05 A549 cells were starved in DMEM contains no FBS in 6-well plate overnight, and treated with different concentrations of test compounds in serum- free medium for 30 minutes at 37 °C. Cells were subsequently stimulated with or without Gas6 (400 ng/mL) for 30 min at 37 °C. Total proteins were extracted from the test compound treated cells using ice-cold RIPA buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 % (v/v) Triton-X 100, 0.1 (w/v) SDS) supplemented with protease inhibitor cocktail and phosphatase inhibitors. Protein concentrations were determined using Bradford assay. Twenty micrograms of total proteins were fractionated on 10% SDS-PAGE gels and transferred onto nitrocellulose membrane (Bio-rad). Transfer efficiency and loading were confirmed by reversible staining of the membrane with Ponseau S solution (MP Biomedicals) following protein transfer.
Membranes were blocked at ambient temperature with 5% non-fat dry milk in Tris- buffered saline (TBS) containing 0.1% Tween-20 (TBST) for 1 hour and incubated with primary antibodies against Axl, Tyro3, phospho-AKT (Ser473), total AKT or β-actin at 4 °C. After overnight incubation, membranes were washed with TBST and incubated with a secondary horseradish peroxidase (HRP)-labeled antibody (Jackson ImmunoResearch Laboratories, Inc.) for 1 hour at ambient temperature. Membranes were washed in TBST following incubation with secondary antibodies. Bound antibody complexes were detected and visualized using Luminata Classico Western HRP Substrate
(Millipore). All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety.
From the foregoing it will be appreciated that, although specific embodiments of the disclosure have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the disclosure. Accordingly, the disclosure is not limited except as by the appended claims.

Claims

WHAT IS CLAIMED IS
1. A compound of Formula (I):
Figure imgf000125_0001
each A, A1 and A2 is the same or different and independently -N=, -
CR5=, or -0-;
L is a heteroaryl, heterocyclyl, -N(R6)-, -0-, -C(O)-, -C(0)0-, -S(O),- where t is 0, 1, or 2, -CON(R6)-, -N(R6)CO-, -S02N(R6)-, or -N(R6)CON(R6)-; provided that when L is heteroaryl, L is not pyridinyl, pyrazinyl or thienyl;
R1 is alkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or heteroarylalkyl;
R2 is hydrogen, heterocyclyl, heterocyclylalkyl, cycloalkyl, alkyl, aralkyl, cycloalkylalkyl, heteroaryl, heteroarylalkyl, or aryl;
each R3 and R4 is the same or different and independently selected from hydrogen, alkyl, aralkyl, alkenyl, aralkenyl, alkynyl, aralkynyl, cycloalkyl,
cycloalkylalkyl, halo, or haloalkyl;
R5, at each occurrence, is hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, halo, or haloalkyl; and
R6 is hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, or haloalkyl, a stereoisomer, enantiomer or tautomer thereof, an isotopically enriched derivative thereof, a pharmaceutically acceptable salt thereof, a pharmaceutical composition thereof or a prodrug thereof.
2. The compound of claim 1 wherein L is heteroaryl.
3. The compound of claim 1 or claim 2 wherein L is N-heteroaryl.
4. The compound of any one of claims 1-3 wherein L is divalent pyrrozolyl moiety and the com ound is represented by Formula (IA):
Figure imgf000126_0001
Formula (IA)
The compound of any one of claims 1-4, represented by Formula
Figure imgf000126_0002
Formula (IA1)
6. The compound of claim 5 wherein R1 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
7. The compound of claim 6 wherein R1 is a phenyl or substituted phenyl.
8. The compound of claim 6 or claim 7 wherein R2 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
9. The compound of any one of claims 6-8, wherein R2 is piperidinyl, substituted piperidinyl, tetrahydropyranyl, or cyclohexyl.
10. The compound of any one of claims 6-9, wherein each of R3 and
R4 is hydrogen.
11. The compound of any one of claims 6-9 wherein each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is l-piperidin-4-yl or substituted l-piperidin-4-yl.
12. The compound of claim 11 being:
tert-butyl-4-(4-(6-amino-5-(5-phenyloxazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine- 1 -carboxylate;
3-(5-phenyloxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidine- 1 -carboxylate;
3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
l-(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)ethanone;
(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(cyclopropyl)methanone;
3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
l-(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)ethanone;
3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(cyclopropyl)methanone;
(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(phenyl)methanone;
l-(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)-2-phenylethanone;
(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(phenyl)methanone;
l-(4-(4-(6-amino-5-(5-(4-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)-2-phenylethanone;
3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
l-(4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)-2,2-dimethylpropan- 1 -one; (4-(4-(6-amino-5-(5-(3-chlorophenyl)oxazol-2-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(4-fluorophenyl)methanone; or
5-(l-(l-ethylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-phenyloxazol-2-yl)pyridin-2-amine.
13. The compound of any one of claims 6-9 wherein each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is tetrahydropyranyl.
14. The compound of claim 13 being:
3-(5-phenyloxazol-2-yl)-5-(l-(tetrahydro-2pyran-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine; or
3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l-(tetrahydro-2pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine.
15. The compound of any one of claims 6-9 wherein each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is cyclohexyl.
16. The compound of claim 15 being:
3-(5-(3-chlorophenyl)oxazol-2-yl)-5-(l -cyclohexyl- lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(4-chlorophenyl)oxazol-2-yl)-5-(l -cyclohexyl- lH-pyrazol-4-yl)pyridin-2-amine; or
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenyloxazol-2-yl)pyridin-2-amine.
17. The compound of any one of claims 6-9, wherein each of R3 and R4 is hydrogen;
R1 is heteroaryl; and
R2 is heterocyclyl.
18. The compound of claim 17 being 5-(l-(piperidin-4-yl)-lH- pyrazol-4-yl)-3-(5-(pyridin-3-yl)oxazol-2-yl)pyridin-2-amine. The compound of any one of claims 1-4, represented by Formula
(IA2):
Figure imgf000129_0001
Formula (IA2)
The compound of claim 19 wherein R is aryl, heteroaryl, cycloalkyl or heterocyclyl.
21. The compound of claim 19 or 20 wherein R2 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
22. The compounds of any one of claims 19-21 wherein:
each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
The compound of claim 22 being
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenylisoxazol-3-yl)pyridin-2-amine;
3 -(5 -phenylisoxazol-3 -yl)-5 -( 1 -(tetrahydro-2H-pyran-4-yl)- lH-pyrazol-4-yl)pyridin-2- amine;
3-(5-phenylisoxazol-3-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine; or 5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-phenylisoxazol-3-yl)pyridin-2- amine.
The compound of any one of claims 1-4, represented by Formula
(IA3):
Figure imgf000129_0002
Formula (IA3)
25. The compound of claim 24 wherein R1 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
26. The compound of claim 24 or 25, wherein R2 is aryl, heteroaryl, cycloalkyl or heterocyclyl.
27. The compound of any one of claims 24-26, wherein each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
28. The compound of claim 27 being:
3-(5-phenylfuran-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2-amine;
5 -( 1 -( 1 -methylpiperidin-4-yl)- lH-pyrazol-4-yl)-3 -(5 -phenylfuran-2-yl)pyridin-2-amine; 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenylfuran-2-yl)pyridin-2-amine; or
3 -(5 -phenylfuran-2-yl)-5 -( 1 -(tetrahydro-2H-pyran-4-yl)- lH-pyrazol-4-yl)pyridin-2- amine.
29. The compound of any one of claims 1-4, represented by Formula
Figure imgf000130_0001
Formula (IA4)
30. The compound of claim 29, wherein R1 is aryl, heteroaryl, cycloalkyl, heterocyclyl, aralkyl, or cycloalkylalkyl.
31. The compound of claim 30 wherein R1 is a phenyl, substituted phenyl, benzyl or cyclopropylalkyl.
32. The compound of claim 30 wherein R1 is a pyridinyl, substituted pyridinyl, pyrazinyl, substituted pyrazinyl, thiazolyl, or substituted thiazolyl.
33. The compound of any one of claims 29-32, wherein R2 is hydrogen, aryl, heteroaryl, cycloalkyl or heterocyclyl.
34. The compound of claim 33 wherein R2 is piperidinyl or substituted piperidinyl, tetrahydropyranyl or hydrogen.
35. The compound of claim 30 wherein:
each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is piperidinyl or substituted piperidinyl.
36. The compound of claim 35 being:
tert-butyl 4-(4-(6-amino-5 -(5 -phenyl- 1 ,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine- 1 -carboxylate;
3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
3-(5-(2,6-dichloro-3-fluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine;
3-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate;
3-(5-(2,5-difluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate;
3-(5-(4-fluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(4-fluorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate; l-(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)ethanone;
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(l-methylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine;
l-(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)-4-methylpentan- 1 -one;
(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)(cyclopropyl)methanone;
(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)(phenyl)methanone;
l-(4-(4-(6-amino-5-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidin- 1 -yl)-2-phenylethanone;
3-(5-(2,6-dichlorophenyl)- 1 ,3,4-oxadiazol-2-yl)-5-(l -(1 -ethylpiperidin-4-yl)- 1H- pyrazol-4-yl)pyridin-2-amine; or
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(l-dodecylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine.
37. The compound of claim 30 wherein:
each of R3 and R4 is hydrogen;
R1 is heteroaryl or substituted heteroaryl; and
R2 is piperidinyl or substituted piperidinyl.
38. The compound of claim 37 being:
tert-butyl 4-(4-(6-amino-5-(5-(pyrazin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
tert-butyl 4-(4-(6-amino-5-(5-(pyridin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(pyrazin-2-yl)-l,3,4-oxadiazol-2- yl)pyridin-2-amine;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(pyridin-2-yl)-l,3,4-oxadiazol-2-yl)pyridin- 2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(thiazol-2-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate; 5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(thiazol-2-yl)-l,3,4-oxadiazol-2-yl)pyridin- 2-amine;
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(pyridin-3-yl)-l,3,4-oxadiazol-2-yl)pyridin- 2-amine;
tert-butyl 4-(4-(6-amino-5-(5-(pyridin-3-yl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)-lH- pyrazol- 1 -yl)piperidine- 1 -carboxylate;
tert-butyl 4-(4-(6-amino-5-(5-(4-(trifluoromethyl)-thiazol-2-yl)-l,3,4-oxadiazol-2- yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate; or
5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-(4-(trifluoromethyl)thiazol-2-yl)-l,3,4- oxadiazol-2-yl)pyridin-2-amine.
39. The compound of claim 30 wherein:
each of R3 and R4 is hydrogen;
R1 is cycloalkylalkyl or aralkyl; and
R2 is piperidinyl or substituted piperidinyl.
40. The compound of claim 39 being:
tert-butyl 4-(4-(6-amino-5-(5-(cyclopropylmethyl)-l,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 -yl)piperidine- 1 -carboxylate;
3-(5-(cyclopropylmethyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
tert-butyl 4-(4-(6-amino-5 -(5 -benzyl- 1 ,3,4-oxadiazol-2-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidine-l -carboxylate; or
3-(5-benzyl-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine.
41. The compound of claim 30 wherein:
each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is hydrogen, cycloalkyl or tetrahydropyranyl
42. The compound of claim 41 being:
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2- 3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(4-(tert-butyl)phenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(2,5-difluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 3-(5-(4-fluorophenyl)-l,3,4-oxadiazol-2-yl)-5-(lH-pyrazol-4-yl)pyridin-2-amine; 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2- yl)pyridin-2-amine;
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenyl-l,3,4-oxadiazol-2-yl)pyridin-2-amine; 3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine; or
3-(5-phenyl-l,3,4-oxadiazol-2-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine.
43. The compound of any one of claims 1-4, represented by Formula
Figure imgf000134_0001
Formula (IA5)
44. The compound of claim 43 wherein R1 is aryl; and R2 is heterocyclyl.
45. The compound of claim 43 or claim 44 wherein R1 is phenyl or substituted phenyl; and R2 is hydrogen, piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
46. The compound of any one of claims 43-45 wherein: each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl;
R2 is piperidinyl or substituted piperidinyl.
47. The compound of claim 46 being:
3-(3-phenyl-l,2,4-oxadiazol-5-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine; 1 -(4-(4-(6-amino-5 -(3 -phenyl- 1 ,2,4-oxadiazol-5-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidin- 1 -yl)ethanone;
(4-(4-(6-amino-5-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(cyclopropyl)methanone;
5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(3-phenyl-l,2,4-oxadiazol-5- yl)pyridin-2-amine;
(4-(4-(6-amino-5-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-3-yl)-lH-pyrazol-l- yl)piperidin- 1 -yl)(phenyl)methanone;
1 -(4-(4-(6-amino-5 -(3 -phenyl- 1 ,2,4-oxadiazol-5-yl)pyridin-3-yl)- lH-pyrazol- 1 - yl)piperidin- 1 -yl)-2-phenylethanone;
3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(3-(2,6-dichlorophenyl)- 1 ,2,4-oxadiazol-5-yl)-5-(l -(1 -methylpiperidin-4-yl)- 1H- pyrazol-4-yl)pyridin-2-amine;
3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(l-ethylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine;
5-(l-(l-ethylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-
2-amine; or
3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(l-dodecylpiperidin-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine.
48. The compound of any one of claims 43-45 wherein: each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl;
R2 is hydrogen, tetrahydropyranyl or cyclohexyl.
49. The compound of claim 48 being:
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(3-phenyl-l,2,4-oxadiazol-5-yl)pyridin-2-amine; 5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5- yl)pyridin-2-amine;
3-(3-phenyl-l,2,4-oxadiazol-5-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine; 3-(3-(2,6-dichlorophenyl)-l,2,4-oxadiazol-5-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazol-4-yl)pyridin-2-amine; or
3 -(3 -phenyl- 1 ,2,4-oxadiazol-5 -yl)-5 -( 1 H-pyrazol-4-yl)pyridin-2-amine.
50. The compound of any one of claims 1-4, represented by Formula
(IA6):
Figure imgf000136_0001
Formula (IA6)
51. The compound of claim 50 wherein R1 is aryl, R2 is heterocyclyl.
52. The compound of claim 50 or claim 51 wherein R1 is phenyl or substituted phenyl; and R2 is hydrogen, piperidinyl or substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
53. The compound of any one of claims 50-52 wherein: each of R3 and R4 is hydrogen;
R1 is phenyl or substituted phenyl; and
R2 is hydrogen, piperidinyl, substituted piperidinyl, tetrahydropyranyl or cyclohexyl.
54. The compound of claim 53 being:
5-(l-cyclohexyl-lH-pyrazol-4-yl)-3-(5-phenyl-l,2,4-oxadiazol-3-yl)pyridin-2-amine; 3-(5-phenyl-l,2,4-oxadiazol-3-yl)-5-(l-(tetrahydro-2H-pyran-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3-(5-phenyl-l,2,4-oxadiazol-3-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4-yl)pyridin-2- amine;
5-(l-(l-methylpiperidin-4-yl)-lH-pyrazol-4-yl)-3-(5-phenyl-l,2,4-oxadiazol-3- yl)pyridin-2-amine;
3-(5-(2,5-dichlorophenyl)-l,2,4-oxadiazol-3-yl)-5-(l-(piperidin-4-yl)-lH-pyrazol-4- yl)pyridin-2-amine;
3 -(5 -phenyl- 1 ,2,4-oxadiazol-3 -yl)-5 -( 1 H-pyrazol-4-yl)pyridin-2-amine. The compound of any one of claims 1-4, represented by Formula
(IA7):
Figure imgf000137_0001
Formula (IA7)
56. The compound of claim 55 wherein R1 is aryl and R2 is heterocyclyl.
57. The compound of claim 55 or claim 56 being:
3-(5-(2,6-dichlorophenyl)-l,3,4-oxadiazol-2-yl)-5-(l-(piperidin-4-yl)-lH-pyrrol-3- yl)pyridin-2-amine.
The compound of any one of claims 1-4, represented by Formula
(IB):
Figure imgf000137_0002
CR5=, or -0-;
each R3, R4 and R5 is independently hydrogen or alkyl;
L is heteroaryl or heterocycle;
R7 is hydrogen, alkyl, alkoxy, or halo;
X is -0-, -NR8-, or -C(R9)2-;
R8 is hydrogen, alkyl, or -C(0)R10-;
each R9 is independently hydrogen, alkyl, alkoxy, or halo
R10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
59. The compound of claim 58, represented by Formula (IB 1):
Figure imgf000138_0001
Formula (IB
wherein,
each A, A1 and A2 is the same or different and independently -N=, -
CR5=, or -0-;
R1 is aryl, heteroaryl, cycloalkyl, heterocyclyl, aralkyl, or cycloalkylalkyl;
each of R3, R4 and R7 is hydrogen,
R8 is hydrogen, alkyl, or -C(0)R10-; and
R10 is alkyl, cycloalkyl, aryl, alkoxy, or aralkyl.
60. The compound of claim 59 wherein R1 is aryl, aralkyl, heteroaryl or cycloalkylalkyl.
61. The compound of claim 59 wherein R1 is pyridinyl, substituted pyridinyl, pyrazinyl, substituted pyrazinyl, thiazolyl, substituted thiazolyl, phenyl, substituted phenyl, benzyl or cyclopropylalkyl.
62. The com ound of claim 58, represented by Formula (IB2):
Figure imgf000138_0002
Formula (IB2)
wherein,
each A, A1 and A2 is the same or different and independently -N=
CR5=, or -0-;
R1 is aryl, heteroaryl, cycloalkyl, or heterocyclyl; and
each of R3, R4 and R7 is hydrogen.
63. The compound of claim 58, represented by Formula (IB3):
Figure imgf000139_0001
Formula (IB3)
wherein,
each A, A1 and A2 is the same or different and independently -N=,
CR5=, or -0-;
R is aryl, heteroaryl, cycloalkyl, or heterocyclyl; and
each of R3, R4, R7 and R9 is hydrogen;
64. A pharmaceutical composition comprising a compound of any one of claims 1-63 and a pharmaceutically acceptable excipient.
65. A method of treating leukemia and lymphomas comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
66. A method of treating endometriosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
67. A method of treating restenosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
68. A method of treating atherosclerosis/thrombosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
69. A method of treating psoriasis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
70. A method of treating age- related macular degeneration or diabetic retinopathy comprising administering to a subject in need thereof a
pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
71. A method of treating retinopathy of prematurity comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
72. A method of treating renal transplant rejection comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
73. A method of treating rheumatoid arthritis comprising
administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
74. A method of treating osteoarthritis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
75. A method of treating osteoporosis comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
76. A method of treating cataracts comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
77. A method of treating metastasized tumors comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of any one of claims 1-63 or a pharmaceutical composition of claim 64.
PCT/US2014/067709 2013-11-27 2014-11-26 Aminopyridine derivatives as tam family kinase inhibitors WO2015081257A2 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
CN201480065032.8A CN105764514B (en) 2013-11-27 2014-11-26 Aminopyrazole derivatives as TAM race kinase inhibitor
US15/039,609 US10233176B2 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as TAM family kinase inhibitors
CA2930324A CA2930324C (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as tam family kinase inhibitors
EP14819178.6A EP3074390B1 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as tam family kinase inhibitors
KR1020167016444A KR102398473B1 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as tam family kinase inhibitors
CN201910548630.5A CN110156770B (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as inhibitors of TAM family kinases
MX2016006815A MX2016006815A (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as tam family kinase inhibitors.
BR112016012146-5A BR112016012146B1 (en) 2013-11-27 2014-11-26 AMINOPYRIDINE DERIVATIVE COMPOUNDS AS KINASE INHIBITORS OF THE TAM FAMILY, PHARMACEUTICAL COMPOSITION COMPRISING SAID COMPOUNDS AND THERAPEUTIC USE THEREOF
AU2014354711A AU2014354711B2 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as TAM family kinase inhibitors
JP2016534735A JP6496731B2 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as TAM family kinase inhibitors
IL245660A IL245660B (en) 2013-11-27 2016-05-16 Aminopyridine derivatives as tam family kinase inhibitors
US16/254,208 US10723725B2 (en) 2013-11-27 2019-01-22 Aminopyridine derivatives as TAM family kinase inhibitors
US16/899,489 US11034685B2 (en) 2013-11-27 2020-06-11 Aminopyridine derivatives as TAM family kinase inhibitors
US16/899,494 US11034686B2 (en) 2013-11-27 2020-06-11 Aminopyridine derivatives as TAM family kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361909828P 2013-11-27 2013-11-27
US201361909830P 2013-11-27 2013-11-27
US61/909,830 2013-11-27
US61/909,828 2013-11-27

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/039,609 A-371-Of-International US10233176B2 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as TAM family kinase inhibitors
US16/254,208 Division US10723725B2 (en) 2013-11-27 2019-01-22 Aminopyridine derivatives as TAM family kinase inhibitors

Publications (2)

Publication Number Publication Date
WO2015081257A2 true WO2015081257A2 (en) 2015-06-04
WO2015081257A3 WO2015081257A3 (en) 2015-07-02

Family

ID=52146717

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/067709 WO2015081257A2 (en) 2013-11-27 2014-11-26 Aminopyridine derivatives as tam family kinase inhibitors

Country Status (11)

Country Link
US (4) US10233176B2 (en)
EP (1) EP3074390B1 (en)
JP (1) JP6496731B2 (en)
KR (1) KR102398473B1 (en)
CN (2) CN110156770B (en)
AU (1) AU2014354711B2 (en)
BR (1) BR112016012146B1 (en)
CA (1) CA2930324C (en)
IL (1) IL245660B (en)
MX (2) MX2016006815A (en)
WO (1) WO2015081257A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018224455A1 (en) 2017-06-07 2018-12-13 Basf Se Substituted cyclopropyl derivatives
WO2020061216A1 (en) 2018-09-18 2020-03-26 Signalchem Lifesciences Corporation Combination therapy for treating blood cancer
WO2022165436A1 (en) 2021-02-01 2022-08-04 Signalchem Lifesciences Corporation Use of the axl inhibitor slc-391 as antiviral therapeutic agent

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019060693A1 (en) 2017-09-22 2019-03-28 Kymera Therapeutics, Inc. Crbn ligands and uses thereof
JP7366031B2 (en) 2017-09-22 2023-10-20 カイメラ セラピューティクス, インコーポレイテッド Proteolytic agents and their use
IL315310A (en) 2017-12-26 2024-10-01 Kymera Therapeutics Inc Irak degraders and uses thereof
US11512080B2 (en) 2018-01-12 2022-11-29 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
WO2019140380A1 (en) 2018-01-12 2019-07-18 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US20220356185A1 (en) * 2018-07-06 2022-11-10 Kymera Therapeutics, Inc. Mertk degraders and uses thereof
EP3817748A4 (en) 2018-07-06 2022-08-24 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
MX2021006154A (en) 2018-11-30 2021-08-24 Kymera Therapeutics Inc Irak degraders and uses thereof.
US11591332B2 (en) 2019-12-17 2023-02-28 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
KR20220145325A (en) 2019-12-17 2022-10-28 카이메라 쎄라퓨틱스 인코포레이티드 IRAK disintegrants and uses thereof
CN115297931A (en) 2019-12-23 2022-11-04 凯麦拉医疗公司 SMARCA degrading agents and uses thereof
JP2023518423A (en) 2020-03-19 2023-05-01 カイメラ セラピューティクス, インコーポレイテッド MDM2 degrading agents and their uses
TW202210483A (en) 2020-06-03 2022-03-16 美商凱麥拉醫療公司 Crystalline forms of irak degraders
CN112707898A (en) * 2020-12-21 2021-04-27 徐新杰 Heteroaryl-substituted pyrazine derivative and application thereof
TW202309030A (en) 2021-05-07 2023-03-01 美商凱麥拉醫療公司 Cdk2 degraders and uses thereof
AU2023214044A1 (en) 2022-01-31 2024-08-08 Kymera Therapeutics, Inc. Irak degraders and uses thereof

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007030680A2 (en) 2005-09-07 2007-03-15 Rigel Pharmaceuticals, Inc. Triazole derivatives useful as axl inhibitors
WO2008045978A1 (en) 2006-10-10 2008-04-17 Rigel Pharmaceuticals, Inc. Pinane-substituted pyrimidinediamine derivatives useful as axl inhibitors
WO2008080134A2 (en) 2006-12-22 2008-07-03 Rigel Pharmaceuticals, Inc. 4-amin0-2- (hetero) arylamino-5- (hetero) arylthiazoles useful as axl inhibitors
WO2008083356A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Substituted triazoles useful as axl inhibitors
WO2008083353A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Bicyclic aryl and bicyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2008083367A2 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2008083354A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. N3-heteroaryl substituted triazoles and n5-heteroaryl substituted triazoles useful as axl inhibitors
WO2008083357A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Bridged bicyclic aryl and bridged bicyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2008128072A2 (en) 2007-04-13 2008-10-23 Supergen, Inc. Axl kinase inhibitors useful for the treatment of cancer or hyperproliferative disorders
WO2009007390A2 (en) 2007-07-09 2009-01-15 Astrazeneca Ab 2-pyraz inylbenz imidazole derivatives as receptor tyrosine kinase inhibitors
WO2009024825A1 (en) 2007-08-21 2009-02-26 Astrazeneca Ab 2-pyrazinylbenzimidazole derivatives as receptor tyrosine kinase inhibitors
WO2009047514A1 (en) 2007-10-10 2009-04-16 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use
WO2009053737A2 (en) 2007-10-25 2009-04-30 Astrazeneca Ab Pyridine and pyrazine derivatives useful in the treatment of cell proliferative disorders
WO2009054864A1 (en) 2007-10-26 2009-04-30 Rigel Pharmaceuticals, Inc. Polycyclic aryl substituted triazoles and polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2009127417A1 (en) 2008-04-16 2009-10-22 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinoline derivatives as axl kinase inhibitors
WO2010005879A1 (en) 2008-07-09 2010-01-14 Rigel Pharmaceuticals, Inc. Bridged bicyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2010005876A2 (en) 2008-07-09 2010-01-14 Rigel Pharmaceuticals, Inc. Polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2010083465A1 (en) 2009-01-16 2010-07-22 Rigel Pharmaceuticals, Inc. Axl inhibitors for use in combination therapy for preventing, treating or managing metastatic cancer
WO2010090764A1 (en) 2009-02-09 2010-08-12 Supergen, Inc. Pyrrolopyrimidinyl axl kinase inhibitors
WO2011045084A1 (en) 2009-10-16 2011-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinolinyloxyphenylsulfonamides
WO2011138751A2 (en) 2010-05-04 2011-11-10 Pfizer Inc. Heterocyclic derivatives as alk inhibitors
WO2012028332A1 (en) 2010-08-28 2012-03-08 Lead Discovery Center Gmbh Pharmaceutically active compounds as axl inhibitors
WO2012135800A1 (en) 2011-04-01 2012-10-04 University Of Utah Research Foundation Substituted n-phenylpyrimidin-2-amine analogs as inhibitors of the axl kinase
WO2013074633A1 (en) 2011-11-14 2013-05-23 Cephalon, Inc. Uracil derivatives as axl and c-met kinase inhibitors
WO2013115280A1 (en) 2012-01-31 2013-08-08 第一三共株式会社 Pyridone derivative
WO2013162061A1 (en) 2012-04-26 2013-10-31 第一三共株式会社 Bicyclic pyrimidine compound

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20070978A1 (en) 2006-02-14 2007-11-15 Novartis Ag HETEROCICLIC COMPOUNDS AS INHIBITORS OF PHOSPHATIDYLINOSITOL 3-KINASES (PI3Ks)
WO2007111904A2 (en) * 2006-03-22 2007-10-04 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors for the treatment of proliferative disorders
EP1900727A1 (en) 2006-08-30 2008-03-19 Cellzome Ag Aminopyridine derivatives as kinase inhibitors
PE20090717A1 (en) * 2007-05-18 2009-07-18 Smithkline Beecham Corp QUINOLINE DERIVATIVES AS PI3 KINASE INHIBITORS
EP2265270A1 (en) 2008-02-04 2010-12-29 OSI Pharmaceuticals, Inc. 2-aminopyridine kinase inhibitors
CA2716949A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
AU2010344973B2 (en) 2010-02-05 2016-06-16 Merck Patent Gmbh Hetaryl-[1,8]naphthyridine derivatives
MX2012013082A (en) * 2010-05-12 2013-05-09 Vertex Pharma 2 -aminopyridine derivatives useful as inhibitors of atr kinase.
WO2012087938A1 (en) * 2010-12-20 2012-06-28 Glaxosmithkline Llc Quinazolinone derivatives as antiviral agents
EP2941425B1 (en) 2013-01-02 2017-07-12 H. Lundbeck A/S Aminopyridine derived compounds as lrrk2 inhibitors

Patent Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007030680A2 (en) 2005-09-07 2007-03-15 Rigel Pharmaceuticals, Inc. Triazole derivatives useful as axl inhibitors
WO2008045978A1 (en) 2006-10-10 2008-04-17 Rigel Pharmaceuticals, Inc. Pinane-substituted pyrimidinediamine derivatives useful as axl inhibitors
WO2008080134A2 (en) 2006-12-22 2008-07-03 Rigel Pharmaceuticals, Inc. 4-amin0-2- (hetero) arylamino-5- (hetero) arylthiazoles useful as axl inhibitors
WO2008083356A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Substituted triazoles useful as axl inhibitors
WO2008083353A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Bicyclic aryl and bicyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2008083367A2 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2008083354A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. N3-heteroaryl substituted triazoles and n5-heteroaryl substituted triazoles useful as axl inhibitors
WO2008083357A1 (en) 2006-12-29 2008-07-10 Rigel Pharmaceuticals, Inc. Bridged bicyclic aryl and bridged bicyclic heteroaryl substituted triazoles useful as axl inhibitors
US20080188455A1 (en) 2006-12-29 2008-08-07 Rigel Pharmaceuticals, Inc. Polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2008128072A2 (en) 2007-04-13 2008-10-23 Supergen, Inc. Axl kinase inhibitors useful for the treatment of cancer or hyperproliferative disorders
WO2009007390A2 (en) 2007-07-09 2009-01-15 Astrazeneca Ab 2-pyraz inylbenz imidazole derivatives as receptor tyrosine kinase inhibitors
WO2009024825A1 (en) 2007-08-21 2009-02-26 Astrazeneca Ab 2-pyrazinylbenzimidazole derivatives as receptor tyrosine kinase inhibitors
WO2009047514A1 (en) 2007-10-10 2009-04-16 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use
WO2009053737A2 (en) 2007-10-25 2009-04-30 Astrazeneca Ab Pyridine and pyrazine derivatives useful in the treatment of cell proliferative disorders
WO2009054864A1 (en) 2007-10-26 2009-04-30 Rigel Pharmaceuticals, Inc. Polycyclic aryl substituted triazoles and polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2009127417A1 (en) 2008-04-16 2009-10-22 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinoline derivatives as axl kinase inhibitors
WO2010005879A1 (en) 2008-07-09 2010-01-14 Rigel Pharmaceuticals, Inc. Bridged bicyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2010005876A2 (en) 2008-07-09 2010-01-14 Rigel Pharmaceuticals, Inc. Polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2010083465A1 (en) 2009-01-16 2010-07-22 Rigel Pharmaceuticals, Inc. Axl inhibitors for use in combination therapy for preventing, treating or managing metastatic cancer
WO2010090764A1 (en) 2009-02-09 2010-08-12 Supergen, Inc. Pyrrolopyrimidinyl axl kinase inhibitors
WO2011045084A1 (en) 2009-10-16 2011-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinolinyloxyphenylsulfonamides
WO2011138751A2 (en) 2010-05-04 2011-11-10 Pfizer Inc. Heterocyclic derivatives as alk inhibitors
WO2012028332A1 (en) 2010-08-28 2012-03-08 Lead Discovery Center Gmbh Pharmaceutically active compounds as axl inhibitors
WO2012135800A1 (en) 2011-04-01 2012-10-04 University Of Utah Research Foundation Substituted n-phenylpyrimidin-2-amine analogs as inhibitors of the axl kinase
WO2013074633A1 (en) 2011-11-14 2013-05-23 Cephalon, Inc. Uracil derivatives as axl and c-met kinase inhibitors
WO2013115280A1 (en) 2012-01-31 2013-08-08 第一三共株式会社 Pyridone derivative
WO2013162061A1 (en) 2012-04-26 2013-10-31 第一三共株式会社 Bicyclic pyrimidine compound

Non-Patent Citations (52)

* Cited by examiner, † Cited by third party
Title
"Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", December 2000, WILEY
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"Remington: The Science and Practice of Pharmacy", 2000, PHILADELPHIA COLLEGE OF PHARMACY AND SCIENCE
ALEXANDER, J. M. ET AL.: "Human parathyroid hormone 1-34 reverses bone loss in ovariectomized mice", J BONE MINER RES., vol. 16, no. 9, 2001, pages 1665 - 1673
ANGELILLO-SCHERRER A. ET AL.: "Role of Gas6 receptors in platelet signaling during thrombus stabilization and implications for antithrombotic therapy", J. CLIN. INVEST., vol. 115, 2005, pages 237 - 246
BORA, P. S. ET AL.: "Immunotherapy for choroidal neovascularization in a laser-induced mouse model simulating exudative (wet) macular degeneration", PROC. NATL. ACAD. SCI. U. S. A., vol. 100, no. 5, 2003, pages 2679 - 2684
BREWSTER, M. ET AL.: "Ro 32-3555, an orally active collagenase selective inhibitor, prevents structural damage in the STR/ORT mouse model of osteoarthritis", ARTHRITIS. RHEUM., vol. 41, no. 9, 1998, pages 1639 - 1644
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
FOSTER, ADV. DRUG RES., vol. 14, 1985, pages 1 - 36
FUJIOKA, M. ET AL.: "Equol, a metabolite of daidzein, inhibits bone loss in ovariectomized mice", J. NUT., vol. 134, no. 10, 2004, pages 2623 - 2627
GATELY, J. NUCL. MED., vol. 27, 1986, pages 388 - 394
GORDON, DRUG METAB. DISPOS., vol. 15, 1987, pages 589 - 594
GREEN, T.W.; P.G.M. WUTZ: "Protective Groups in Organic Synthesis", 1999, WILEY
HIGUCHI, T. ET AL.: "A.C.S. Symposium Series", vol. 14, article "Pro-drugs as Novel Delivery Systems"
HOLLAND ET AL., CANCER RES., vol. 70, no. 4, 2010, pages 1544 - 1554
HUANG ET AL., J. STRU. BIOL., vol. 165, 2009, pages 88 - 96
KADOYA, K. ET AL.: "Role of calpain in hydrogen peroxide induced cataract", CURR. EYE RES., vol. 12, no. 4, 1993, pages 341 - 346
KIM, D.W. ET AL.: "Novel oral formulation of paclitaxel inhibits neointimal hyperplasia in a rat carotid artery injury model", CIRCULATION, vol. 109, no. 12, 2004, pages 1558 - 1563
KUSHNER, CAN. J. PHYSIOL. PHARMACOL., vol. 77, 1999, pages 79 - 88
LEBOVIC, D.I. ET AL.: "Peroxisome proliferator-activated receptor-gamma induces regression of endometrial explants in a rat model of endometriosis", FERTIL. STERIL., vol. 82, no. 3, 2004, pages 1008 - 1013
LIJINSKY, J. NAT. CANCER INST., vol. 69, 1982, pages 1127 - 1133
LIU ET AL., ACS MED. CHEM. LETT., vol. 3, 2012, pages 129 - 134
LIU ET AL., EURO. J. MED. CHEM., vol. 65, 2013, pages 83 - 93
M. F. BURBRIDGE ET AL., MOL. CANCER THER., vol. 12, 2013, pages 1749 - 1762
MANGOLD, MUTATION RES., vol. 308, 1994, pages 33 - 42
MOLLARD ET AL., ACS MED. CHEM. LETT., vol. 2, 2011, pages 907 - 912
NAKASHIMA, Y. ET AL.: "ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree", ARTERIOSCLER. THROMB., vol. 14, no. 1, 1994, pages 133 - 140
NICKOLOFF, B.J. ET AL.: "Severe combined immunodeficiency mouse and human psoriatic skin chimeras. Validation of a new animal model", AM. J. PATHOL., vol. 146, no. 3, 1995, pages 580 - 588
ONO ET AL.: "poster number MEDI-393", 244TH ACS NATIONAL MEETING & EXPOSITION, 19 August 2012 (2012-08-19)
PHADKE, K. ET AL.: "Evaluation of the effects of various anti-arthritic drugs on type II collagen-induced mouse arthritis model", LMMUNOPHARMACOLOGY, vol. 10, no. 1, 1985, pages 51 - 60
POWELL ET AL., BIOORG. MED. CHEM. LETT., vol. 22, 2012, pages 190 - 193
POWELL ET AL., BIOORG. MED. CHEM. LETT., vol. 23, 2013, pages 1046 - 1050
POWELL ET AL., BIOORG. MED. CHEM. LETT., vol. 23, 2013, pages 1051 - 1055
RHO ET AL., CANCER RES., vol. 74, 2014, pages 253 - 262
SARAYBA MA; LI L; TUNGSIRIPAT T; LIU NH; SWEET PM; PATEL AJ; OSANN KE; CHITTIBOYINA A; BENSON SC; PERSHADSINGH HA: "Inhibition of corneal neovascularization by a peroxisome proliferator-activated receptor-gamma ligand", EXP. EYE. RES., vol. 80, no. 3, 2005, pages 435 - 442
SHEETS, N. L. ET AL.: "Cataract- and lens-specific up-regulation of ARK receptor tyrosine kinase in Emory mouse cataract", INVEST. OPHTHALMOL. VIS. SCI., vol. 43, no. 6, 2002, pages 1870 - 1875
SMITH, L.E. ET AL.: "Oxygen-induced retinopathy in the mouse", INVEST. OPHTHALMOL. VIS. SCI., vol. 35, no. 1, 1994, pages 101 - 111
SOMIGLIANA, E. ET AL.: "Endometrial ability to implant in ectopic sites can be prevented by interleukin-12 in a murine model of endometriosis", HUM. REPROD., vol. 14, no. 12, 1999, pages 2944 - 2950
SUAREZ ET AL., EURO. J. MED. CHEM., vol. 61, 2013, pages 2 - 25
TRAORE ET AL., EURO. J. MED. CHEM., vol. 70, 2013, pages 789 - 801
VON DER THUSEN, J. H. ET AL.: "Adenoviral transfer of endothelial nitric oxide synthase attenuates lesion formation in a novel murine model of postangioplasty restenosis", ARTERIOSCLER. THROMB. VASE. BIOL., vol. 24, no. 2, 2004, pages 357 - 362
WADE D., CHEM. BIOL. INTERACT., vol. 117, 1999, pages 191 - 637
WANG ET AL.: "poster number MEDI-18", 242ND ACS NATIONAL MEETING & EXPOSITION, 28 August 2011 (2011-08-28)
WRONSKI, TJ. ET AL.: "Endocrine and pharmacological suppressors of bone turnover protect against osteopenia in ovariectomized rats", ENDOCRINOLOGY, vol. 125, no. 2, 1989, pages 810 - 816
YANG ET AL.: "poster number MEDI-265", 242ND ACS NATIONAL MEETING & EXPOSITION, 28 August 2011 (2011-08-28)
YIN, J. L. ET AL.: "Expression of growth arrest-specific gene 6 and its receptors in a rat model of chronic renal transplant rejection", TRANSPLANTATION, vol. 73, no. 4, 2002, pages 657 - 660
ZELLO, METABOLISM, vol. 43, 1994, pages 487 - 491
ZHANG ET AL., J. MED. CHEM., vol. 56, 2013, pages 9683 - 9692
ZHANG ET AL., J. MED. CHEM., vol. 56, 2013, pages 9693 - 9700
ZHANG ET AL., J. MED. CHEM., vol. 57, 2014, pages 7031 - 7041
ZHANG ET AL.: "poster number MEDI-56", 244TH ACS NATIONAL MEETING & EXPOSITION, 19 August 2012 (2012-08-19)
ZHANG ET AL.: "poster number MEDI-62", 242ND ACS NATIONAL MEETING & EXPOSITION, 28 August 2011 (2011-08-28)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018224455A1 (en) 2017-06-07 2018-12-13 Basf Se Substituted cyclopropyl derivatives
WO2020061216A1 (en) 2018-09-18 2020-03-26 Signalchem Lifesciences Corporation Combination therapy for treating blood cancer
US20220031685A1 (en) * 2018-09-18 2022-02-03 Signalchem Lifesciences Corporation Combination therapy for treating blood cancer
US12097194B2 (en) * 2018-09-18 2024-09-24 Signalchem Lifesciences Corporation Combination therapy for treating blood cancer
WO2022165436A1 (en) 2021-02-01 2022-08-04 Signalchem Lifesciences Corporation Use of the axl inhibitor slc-391 as antiviral therapeutic agent

Also Published As

Publication number Publication date
EP3074390A2 (en) 2016-10-05
US10233176B2 (en) 2019-03-19
US20190152965A1 (en) 2019-05-23
EP3074390B1 (en) 2020-07-08
CN105764514A (en) 2016-07-13
WO2015081257A3 (en) 2015-07-02
CN105764514B (en) 2019-07-12
IL245660B (en) 2020-01-30
US11034685B2 (en) 2021-06-15
CA2930324C (en) 2022-01-18
BR112016012146A8 (en) 2018-01-30
BR112016012146B1 (en) 2022-11-01
MX2020005181A (en) 2020-08-17
KR102398473B1 (en) 2022-05-16
AU2014354711A1 (en) 2016-06-02
JP2017501981A (en) 2017-01-19
US20200299288A1 (en) 2020-09-24
IL245660A0 (en) 2016-06-30
US11034686B2 (en) 2021-06-15
JP6496731B2 (en) 2019-04-03
CA2930324A1 (en) 2015-06-04
US20170022189A1 (en) 2017-01-26
MX2016006815A (en) 2016-12-02
US20200299287A1 (en) 2020-09-24
CN110156770A (en) 2019-08-23
AU2014354711B2 (en) 2018-10-25
BR112016012146A2 (en) 2017-08-08
CN110156770B (en) 2022-10-04
US10723725B2 (en) 2020-07-28
KR20160091358A (en) 2016-08-02

Similar Documents

Publication Publication Date Title
US11034686B2 (en) Aminopyridine derivatives as TAM family kinase inhibitors
US9382241B2 (en) Aminoquinazoline and pyridopyrimidine derivatives
CN111386118A (en) Combination pharmaceutical agents as RSV inhibitors
JP7476216B2 (en) Substituted Bicyclic Compounds as Farnesoid X Receptor Modulators - Patent application
JPWO2006004027A1 (en) Pyrazole derivative
JP2013525489A (en) Bicyclic heteroaryl analogues as GPR119 modifiers
JP2022130404A (en) Substituted Nitrogen-Containing Compounds
CA3232128A1 (en) Pyridine derivative and use thereof
AU2014353006B2 (en) Quinazoline derivatives as TAM family kinase inhibitors
CN115697991A (en) Piperazine cyclic ureas
US11440922B2 (en) Heterocyclylmethylidene derivatives and their use as modulators of mGluR5 receptors
TW202409011A (en) Inhibitors of human respiratory syncytial virus and metapneumovirus
WO2022129915A1 (en) Pyrazole derivatives as c-abl inhibitors
IL291418B2 (en) C-myc mrna translation modulators and uses thereof in the treatment of cancer
EA047337B1 (en) SUBSTITUTED BICYCLIC COMPOUNDS AS MODULATORS OF THE FARNEZOID X RECEPTOR

Legal Events

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

Ref document number: 14819178

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2930324

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 245660

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/006815

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2016534735

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15039609

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2014354711

Country of ref document: AU

Date of ref document: 20141126

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2014819178

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014819178

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016012146

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20167016444

Country of ref document: KR

Kind code of ref document: A

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

Ref document number: 14819178

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 112016012146

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160527