CN113166162A - Preparation and application of imidazo aromatic ring compounds - Google Patents

Preparation and application of imidazo aromatic ring compounds Download PDF

Info

Publication number
CN113166162A
CN113166162A CN201980081131.8A CN201980081131A CN113166162A CN 113166162 A CN113166162 A CN 113166162A CN 201980081131 A CN201980081131 A CN 201980081131A CN 113166162 A CN113166162 A CN 113166162A
Authority
CN
China
Prior art keywords
group
substituted
compound
unsubstituted
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201980081131.8A
Other languages
Chinese (zh)
Inventor
江磊
冯志勇
金贤
乔智
尚珂
寿建勇
吴淡宜
许灵灵
徐圆
张淑芸
张毅
张宇星
郑明伟
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Ennovabio Pharmaceuticals Co Ltd
Original Assignee
Shanghai Ennovabio Pharmaceuticals Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Ennovabio Pharmaceuticals Co Ltd filed Critical Shanghai Ennovabio Pharmaceuticals Co Ltd
Publication of CN113166162A publication Critical patent/CN113166162A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/537Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines spiro-condensed or forming part of bridged 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provides preparation and application of a class of aromatic amide compounds, and particularly provides a compound shown as a formula I, wherein the definition of each group is described in the specification. The compound has TRK kinase inhibitory activity and can be used as a pharmaceutical composition for treating TRK dysfunction related diseases.

Description

Preparation and application of imidazo aromatic ring compounds Technical Field
The invention relates to the field of small molecule drugs, in particular to a TRK kinase inhibitor and preparation and application thereof.
Background
Tropomyosin-receptor kinase (TRK) is a nerve growth factor receptor, belongs to a receptor tyrosine kinase family, mainly comprises three highly homologous members of TRKA, TRKB and TRKC, and is respectively encoded by three genes of NTRK1, NTRK2 and NTRK 3. These receptor tyrosine kinases are mainly expressed in nervous tissues and play an important role in development and physiological functions of the nervous system through activation of neurotrophic factors nts (neurotropins). TRK function as tyrosine kinase receptors, each TRK having a signaling pathway that binds to and activates its downstream ligand. NGF (nerve growth factor) specifically binds to and activates TRKA; the TRKB ligand comprises BDGF (broad-derived growth factor) and NT-4/5 (neurotropin-4/5); NT-3 specifically binds to and activates TRKC. All three TRK receptors contain an extracellular domain for ligand binding, a transmembrane domain, and an intracellular domain with kinase activity.
When a specific ligand is combined with the extracellular domain of a corresponding receptor, oligomerization of the receptor and phosphorylation of specific tyrosine residues in the cytoplasmic kinase domain are initiated, thereby causing activation of downstream signaling pathways such as Ras/MAPK, PLC γ/PKC and PI3K/AKT signaling pathways, and further regulating a series of physiological processes such as proliferation, differentiation and survival of nerve cells (Bergman, et al 1999). The TRK signaling pathway is usually precisely regulated, while its aberrant activation is closely associated with tumorigenesis (Amatu, et al.2016). Research results show that there are many mechanisms causing abnormal activation of TRK pathway, including gene fusion, protein overexpression and single nucleotide mutation, and these abnormalities are closely related to the pathogenesis of tumor, especially NTRK gene fusion has been proved to be an important factor causing multiple tumorigenesis, and is independent of the tissue source and type of tumor. Under the rapid development of current second-generation sequencing technologies and precision medicine, more and more NTRK fusion genes are found, such as ETV6-NTRK3, MPRIP-NTRK 1, CD 74-NTRK 1 and the like. The results of recent clinical trials show that these fusion genes are very effective anticancer targets, and the tumors containing the NTRK fusion gene have very significant response rate to TRK inhibitors (Drilon, et al.2018). Therefore, more and more TRK target inhibitors are reported, e.g. (WO2010048314, WO2011146336, WO 2017004342). Meanwhile, in the clinical trial stage, it has been found that some patients receiving treatment have drug resistance phenomenon and proved to be caused by partial base mutation of enzyme activity region, such as NTRK1G595R or G667C mutation, G623R or G696A mutation of NTRK3, and the development of new generation TRK kinase inhibitor is expected to solve these problems.
In view of the above, there is an urgent need in the art to develop a new generation of TRK kinase inhibitors.
Disclosure of Invention
The invention aims to provide a novel TRK kinase inhibitor.
In a first aspect of the present invention, there is provided a compound of formula I:
Figure PCTCN2019123407-APPB-000001
wherein the content of the first and second substances,
r has the formula-L1-R AThe structure of (1);
L 1selected from the group consisting of: substituted or unsubstituted 5-to 10-membered heterocyclylene having 1 to 3 heteroatoms selected from N, S and O, or substituted or unsubstituted- (X)y-wherein each of said xs is independently selected from the group consisting of: substituted or unsubstituted C1-C 8Alkylene, -O-, -C (═ O) -, -CONH-, -NHCO-, -S (═ O)2-、-NH-;
y is selected from the group consisting of: 1.2 or 3;
R Aselected from the group consisting of: substituted or unsubstituted C6-C 10Aryl, substituted or unsubstituted 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7each independently selected from the group consisting of: CR1Or N; z1、Z 2Each independently selected from the group consisting of: c or N;
and is
Figure PCTCN2019123407-APPB-000002
Is an aromatic ring;
R 1each independently selected from the group consisting of: H. d, OH, Cl, F and NH2
R 12Selected from the group consisting of: h or C1-C4 alkyl;
ring a is selected from the group consisting of: substituted or unsubstituted C6-C 10Aryl, substituted or unsubstituted 5-10 membered heteroaryl (including monocyclic, fused cyclic) having 1-3 heteroatoms selected from N, S and O;
l is selected from the group consisting of: none, -NR7-,-O-;
R 7Selected from: h, substituted C1-C6 alkyl; wherein said substitution means that one or more H atoms on the group are substituted with a substituent selected from the group consisting of: halogen, hydroxy;
ring C is selected from the group consisting of: substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted 3-12 membered cycloalkyl (including monocyclic, fused, spiro or bridged), substituted or unsubstituted 4-12 membered heterocyclyl (including monocyclic, fused, spiro or bridged) having 1-3 heteroatoms selected from N, S and O;
unless otherwise specified, "substituted" means substituted with one or more (e.g., 2,3, 4, etc.) substituents selected from the group consisting of: halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH 2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 amido),
Figure PCTCN2019123407-APPB-000003
Or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-12 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O, - (CH) 6 alkyl2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2OH, -OH, C6-C10 aryl, C1-C6 amino, C1-C6 amido, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
Figure PCTCN2019123407-APPB-000004
is the attachment site of the group;
with the proviso that the compounds of formula I are chemically stable structures.
In another preferred embodiment, L is1Selected from the group consisting of:
Figure PCTCN2019123407-APPB-000005
n is selected from the group consisting of: 0.1, 2 or 3;
R 2、R 2aand R2bEach independently selected from the group consisting of: H. OH, halogen, substituted or unsubstituted C1-C 8An alkyl group;
X 5selected from the group consisting of: NH, O, -CONH-, -NHCO-, S, -S (═ O)2-、-NHS(=O)-、-NHS(=O) 2-;
R AIs composed of
Figure PCTCN2019123407-APPB-000006
Wherein, the
Figure PCTCN2019123407-APPB-000007
Means RAAnd L1The attachment site of (a); x4Is CH or N;
R 3selected from the group consisting of: H. halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy.
In another preferred embodiment, the ring C has the following structure:
Figure PCTCN2019123407-APPB-000008
wherein k1, k2, k3 are each independently selected from the group consisting of: 0.1, 2 or 3;
X 1、X 2each independently is C (R)4)R 5、NR 6Or O;
wherein R is4,R 5,R 11Each independently selected from: h, OH, Me, halogen, oxo, CF3,CN,NH 2,-OMe,COOH,CONH 2;R 6Selected from: none, H, Me, CONH2
In another preferred embodiment, the ring C has the following structure:
Figure PCTCN2019123407-APPB-000009
wherein m, p are each independently selected from the group consisting of: 0.1, 2 or 3;
X 3is C (R)9)R 10
Wherein R is8,R 9,R 10Each independently selected from: h, OH, Me, halogen, CF3,CN,NH 2,-OMe,COOH,CONH 2
In another preferred embodiment, said ring C is linked to L via an N atom.
In another preferred embodiment, the compound has the structure shown in formula II below:
Figure PCTCN2019123407-APPB-000010
in another preferred embodiment, the compound has the structure shown in formula III below:
Figure PCTCN2019123407-APPB-000011
in another preferred embodiment, the compound has a structure selected from the group consisting of:
Figure PCTCN2019123407-APPB-000012
Figure PCTCN2019123407-APPB-000013
Figure PCTCN2019123407-APPB-000014
in a second aspect of the invention, there is provided a pharmaceutical composition comprising (1) a compound according to the first aspect of the invention or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof; (2) a pharmaceutically acceptable carrier.
In another preferred embodiment, the disease is selected from the group consisting of: cancer, proliferative diseases, pain, skin diseases or conditions, metabolic diseases, muscular diseases, neurological diseases, autoimmune diseases, dermatitis-induced pruritus, inflammation-related diseases, bone-related diseases.
In a third aspect of the present invention, there is provided a use of the compound according to the first aspect of the present invention or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, or the pharmaceutical composition according to the second aspect of the present invention, for the preparation of a pharmaceutical composition for the prevention and/or treatment of a disease associated with abnormal TRK function (abnormal TRK activation due to amplification, or overexpression, or mutation, or gene fusion).
In another preferred embodiment, the disease is selected from the group consisting of: the disease is selected from the group consisting of: cancer, proliferative diseases, pain, skin diseases or conditions, metabolic diseases, muscular diseases, neurological diseases, autoimmune diseases, dermatitis-induced pruritus.
In a fourth aspect of the invention, there is provided a TRK inhibitor comprising a compound of the first aspect of the invention, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Detailed Description
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of …," or "consisting of ….
Definition of
As used herein, the term "alkyl" includes straight or branched chain alkyl groups. E.g. C1-C 8Alkyl represents a straight or branched chain alkyl group having 1 to 8 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, and the like.
As used herein, the term "alkenyl" includes straight or branched chain alkenyl groups. E.g. C2-C 6Alkenyl means a straight or branched alkenyl group having 2 to 6 carbon atoms, such as vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, or the like.
As used herein, the term "alkynyl" includes straight or branched chain alkynyl groups. E.g. C2-C 6Alkynyl means straight or branched chain alkynyl having 2 to 6 carbon atoms, such as ethynyl, propynyl, butynyl, or the like.
As used herein, the term "C3-C 8Cycloalkyl "refers to cycloalkyl groups having 3 to 8 carbon atoms. It may be a single ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or the like. It may also be in the form of a double ring, for example a bridged or spiro ring.
As used herein, the term "C1-C 8Alkoxy "means a straight or branched chain alkoxy group having 1 to 8 carbon atoms; for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tert-butoxy and the like.
As used herein, the term "3-10 membered heterocycloalkyl having 1-3 heteroatoms selected from the group consisting of N, S and O" refers to a saturated or partially saturated cyclic group having 3-10 atoms and wherein 1-3 atoms are heteroatoms selected from the group consisting of N, S and O. It may be monocyclic or may be in the form of a double ring, for example a bridged or spiro ring. Specific examples may be oxetane, azetidine, tetrahydro-2H-pyranyl, piperidinyl, tetrahydrofuranyl, morpholinyl, pyrrolidinyl, and the like.
As used herein, the term "C6-C 10Aryl "means an aryl group having 6 to 10 carbon atoms, for example, phenyl or naphthyl and the like.
As used herein, the term "5-10 membered heteroaryl having 1-3 heteroatoms selected from the group consisting of N, S and O" refers to a cyclic aromatic group having 5-10 atoms and wherein 1-3 atoms are heteroatoms selected from the group consisting of N, S and O. It may be a single ring or a condensed ring form. Specific examples may be pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3) -triazolyl and (1,2,4) -triazolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl and the like.
Unless otherwise specified, the groups of the present invention may be substituted with a substituent selected from the group consisting of: halogen, nitrile group, nitro group, hydroxyl group, amino group, C1-C 6Alkyl-amino, C1-C 6Alkyl radical, C2-C 6Alkenyl radical, C2-C 6Alkynyl, C1-C 6Alkoxy, halo C1-C 6Alkyl, halo C2-C 6Alkenyl, halo C2-C 6Alkynyl, halo C1-C 6Alkoxy, allyl, benzyl、C 6-C 12Aryl radical, C1-C 6alkoxy-C1-C 6Alkyl radical, C1-C 6Alkoxy-carbonyl, phenoxycarbonyl, C2-C 6Alkynyl-carbonyl, C2-C 6Alkenyl-carbonyl, C3-C 6Cycloalkyl-carbonyl, C1-C 6Alkyl-sulfonyl, and the like.
As used herein, "halogen" or "halogen atom" refers to F, Cl, Br, and I. More preferably, the halogen or halogen atom is selected from F, Cl and Br. "halogenated" means substituted with an atom selected from F, Cl, Br, and I.
Unless otherwise specified, the structural formulae depicted herein are intended to include all isomeric forms (e.g., enantiomers, diastereomers and geometric isomers (or conformational isomers)): for example, R, S configuration containing an asymmetric center, (Z), (E) isomers of double bonds, and the like. Thus, individual stereochemical isomers of the compounds of the present invention or mixtures of enantiomers, diastereomers or geometric isomers (or conformers) thereof are within the scope of the present invention.
As used herein, the term "tautomer" means that structural isomers having different energies may exceed the low energy barrier, thereby converting with each other. For example, proton tautomers (i.e., proton shifts) include interconversion by proton shift, such as 1H-indazoles and 2H-indazoles. Valence tautomers include interconversion by recombination of some of the bonding electrons.
As used herein, the term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio.
A compound of formula I
The invention provides a compound shown as the following formula I:
Figure PCTCN2019123407-APPB-000015
wherein the content of the first and second substances,
r has the formula-L1-R AThe structure of (1);
L 1selected from the group consisting of: substituted or unsubstituted 5-to 10-membered heterocyclylene having 1 to 3 heteroatoms selected from N, S and O, or substituted or unsubstituted- (X)y-wherein each of said xs is independently selected from the group consisting of: substituted or unsubstituted C1-C 8Alkylene, -O-, -C (═ O) -, -CONH-, -NHCO-, -S (═ O)2-、-NH-;
y is selected from the group consisting of: 1.2 or 3;
R Aselected from the group consisting of: substituted or unsubstituted C6-C 10Aryl, substituted or unsubstituted 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7each independently selected from the group consisting of: CR1Or N; z1、Z 2Each independently selected from the group consisting of: c or N; and is
Figure PCTCN2019123407-APPB-000016
Is an aromatic ring;
R 1each independently selected from the group consisting of: H. d, OH, Cl, F and NH2
Ring a is selected from the group consisting of: substituted or unsubstituted C6-C 10Aryl, substituted or unsubstituted 5-10 membered heteroaryl (including monocyclic, fused, spiro or bridged) having 1-3 heteroatoms selected from N, S and O;
l is selected from the group consisting of: none, -NR7-,-O-;
R 7Selected from: H. substituted C1-C6 alkyl; wherein said substitution means that one or more H atoms on the group are substituted with a substituent selected from the group consisting of: halogen, hydroxy;
Ring C is selected from the group consisting of: substituted or unsubstituted 3-12 membered cycloalkyl (including monocyclic, fused, spiro or bridged ring), substituted or unsubstituted 4-12 membered heterocyclyl (including monocyclic, fused, spiro or bridged ring) having 1-3 heteroatoms selected from N, S and O;
unless otherwise specified, "substituted" means substituted with one or more (e.g., 2,3, 4, etc.) substituents selected from the group consisting of: halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH 2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 amido),
Figure PCTCN2019123407-APPB-000017
Or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-12 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O, - (CH) 6 alkyl2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2OH, -OH, C6-C10 aryl, C1-C6 amino, C1-C6 amido, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
Figure PCTCN2019123407-APPB-000018
is the attachment site of the group;
with the proviso that the compounds of formula I are chemically stable structures.
In another preferred embodiment, the compounds of formula I of the present invention are the compounds prepared in the examples.
Preparation of Compounds of formula I
The compounds of formula I of the present invention may be prepared by the following process:
Figure PCTCN2019123407-APPB-000019
pharmaceutical compositions and methods of administration
Since the compound of the present invention has excellent inhibitory activity of TRK kinase, the compound of the present invention and various crystalline forms, pharmaceutically acceptable inorganic or organic salts, hydrates, or solvates thereof, and a pharmaceutical composition containing the compound of the present invention as a main active ingredient may be used for the prevention and/or treatment of diseases (e.g., cancer) associated with TRK kinase activity or expression level.
The pharmaceutical compositions of the present invention comprise a safe and effective amount of a compound of the present invention in combination with a pharmaceutically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 10-200mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate etc.), gelatin, talc, solid lubricants (e.g. stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g. soybean oil, sesame oil, peanut oil, olive oil etc.), polyols (e.g. propylene glycol, glycerol, mannitol, sorbitol etc.), emulsifiers (e.g. tween, etc.)
Figure PCTCN2019123407-APPB-000020
) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, parenteral (intravenous, intramuscular or subcutaneous).
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
When administered in combination, the pharmaceutical composition further comprises one or more other pharmaceutically acceptable compounds. One or more of the other pharmaceutically acceptable compounds may be administered simultaneously, separately or sequentially with a compound of the invention.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 1 to 2000mg, preferably 20 to 500 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are by weight.
Synthesis of intermediate A
Figure PCTCN2019123407-APPB-000021
(R, E) -N- (2, 5-difluorobenzylidene) -2-methylpropane-2-sulfinamide
Figure PCTCN2019123407-APPB-000022
2, 5-difluorobenzaldehyde (5 g, 35.2 mmol) and (R) -2-methylpropane-2-sulfinamide (4.47 g, 36.9 mmol) were dissolved in dichloromethane (50 ml), and cesium carbonate (8.0 g, 24.6 mmol) was added at room temperature, followed by heating to 50 ℃ for 3 hours, TLC showed completion of the reaction, filtration, washing of the filter cake with dichloromethane, washing of the filtrate with brine, drying over anhydrous sodium sulfate, and spin-drying to give a yellow oily liquid (9 g).
(R) -N- ((R) -1- (2, 5-difluorophenyl) -3- (1, 3-dioxan-2-yl) propyl) -2-methylpropane-2-sulfinamide
Figure PCTCN2019123407-APPB-000023
Magnesium turnings (2 g, 83.3 mmol) were dissolved in tetrahydrofuran (72 ml), and a solution of diisobutylaluminum hydride in tetrahydrofuran (0.1 ml, 1.5M,0.15 mmol) was added dropwise to the system at 40 ℃ under nitrogen blanket, reacted at 40 ℃ for 0.5 hour, and then a solution of 2- (2-bromoethyl) -1, 3-dioxane (14.3 g, 73.47 mmol) in tetrahydrofuran (40 ml) was slowly added dropwise to the system while controlling the temperature at 40-50 ℃ and after completion of the addition, the mixture was stirred at 40 ℃ for 1 hour. The heating was removed and the reaction was cooled to-30 ℃ and then (R, E) -N- (2, 5-difluorobenzylidene) -2-methylpropane-2-sulfinamide (9 g, 36.73 mmol) in tetrahydrofuran (40 ml) was added dropwise to the system, the temperature was controlled at-30 ℃ to-20 ℃, after completion of the addition, the mixture was stirred at-30 ℃ for 2 hours, TLC showed the reaction was complete, quenched with 10% aqueous citric acid and controlled at 10 ℃, extracted with dichloromethane, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and spin dried to give a colorless oily liquid (15.8 g).
(R) -2- (2, 5-difluorophenyl) pyrrolidine
Figure PCTCN2019123407-APPB-000024
(R) -N- ((R) -1- (2, 5-difluorophenyl) -3- (1, 3-dioxan-2-yl) propyl) -2-methylpropane-2-sulfinamide (15.8 g, 43.76 mmol) was added to a mixed solution of trifluoroacetic acid (32 ml) and water (8 ml) at room temperature, stirred at room temperature for 1 hour, then trifluoroacetic acid (60 ml) was added to the system, triethylsilane (15.2 g, 131.1 mmol) was added dropwise to the system, reaction was carried out at room temperature overnight, LCMS monitored for completion of the reaction, most of the trifluoroacetic acid was removed, the remainder was dissolved in hydrochloric acid (1N,100 ml) and stirred for 0.5 hour, extracted with methyl t-butyl ether, the organic phase was washed with hydrochloric acid (1N,50 ml), the aqueous phases were combined, the aqueous phase was adjusted to pH 11 with a 40% aqueous sodium hydroxide solution, then, the mixture was extracted with dichloromethane, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and spin-dried to obtain an oily liquid (6.7 g).
Synthesis of intermediate B
Figure PCTCN2019123407-APPB-000025
(S) -N- ((S) -1- (2, 5-difluorophenyl) but-3-en-1-yl) -2-methylpropane-2-sulfinamide
Figure PCTCN2019123407-APPB-000026
(R) -N- (2, 5-difluorobenzylidene) -2-methylpropane-2-sulfinamide (30 g, 122.45 mmol) is added to an aqueous solution of saturated sodium bromide (480 ml) at room temperature, In (42 g, 367.35 mmol) is added, then allylmagnesium bromide (42 ml, 489.8 mmol) is added, the reaction is carried out at room temperature for 6 hours, TLC monitors the reaction to be completed, the reaction is quenched with saturated sodium bicarbonate solution, filtered, the filtrate is extracted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate and dried by spinning to obtain (S) -N- ((S) -1- (2, 5-difluorophenyl) but-3-en-1-yl) -2-methylpropane-2-sulfinamide as a yellow solid (35 g).
(S) -N- ((1S) -1- (2, 5-difluorophenyl) -2- (oxiran-2-yl) ethyl) -2-methylpropane-2-sulfinamide
Figure PCTCN2019123407-APPB-000027
(S) -N- ((S) -1- (2, 5-difluorophenyl) but-3-en-1-yl) -2-methylpropane-2-sulfinamide (35 g, 121.95 mmol) is dissolved in dichloromethane (800 ml), m-chloroperoxybenzoic acid (80 g, 365.85 mmol) is added in portions at room temperature, the mixture is stirred at room temperature overnight, TLC is used for monitoring the completion of the reaction, the reaction solution is washed by saturated sodium bicarbonate and saturated sodium thiosulfate solution, the saturated saline is washed, anhydrous sodium sulfate is dried, and the (S) -N- ((S) -1- (2, 5-difluorophenyl) but-3-en-1-yl) -2-methylpropane-2-sulfinamide is obtained by spin-drying, yellow solid (31 g, 79%).
(3R, 5R) -1- (tert-butylsulfonyl) -5- (2, 5-difluorophenyl) pyrrolidin-3-ol
Figure PCTCN2019123407-APPB-000028
(S) -N- ((S) -1- (2, 5-difluorophenyl) but-3-en-1-yl) -2-methylpropane-2-sulfinamide (31 g, 97.18 mmol) is dissolved in N, N-dimethylformamide (300 ml), potassium carbonate (40 g, 291.53 mmol), potassium iodide (16 g, 97.18 mmol) are added at room temperature, the mixture is heated to 100 ℃ for reaction for 1 hour, TLC monitors the completion of the reaction, the reaction mixture is cooled to room temperature, filtered, the filtrate is poured into water, the organic phase is extracted with ethyl acetate, the combined organic phase is washed with saturated brine, dried over anhydrous sodium sulfate, and spin-dried through-column purification (petroleum ether/ethyl acetate 10/1-5/1) to obtain the compound (3R, 5R) -1- (tert-butylsulfonyl) -5- (2, 5-difluorophenyl) pyrrolidin-3-ol (7.5 g).
(2R,4S) -1- (tert-butylsulfonyl) -2- (2, 5-difluorophenyl) -4-fluoro
Figure PCTCN2019123407-APPB-000029
(3R, 5R) -1- (tert-butylsulfonyl) -5- (2, 5-difluorophenyl) pyrrolidin-3-ol (2.0 g, 6.27 mmol) was dissolved in dichloromethane (50 ml), cooled to-60 ℃, DAST (2 ml) was added dropwise to the system, then naturally warmed to room temperature and stirred overnight, lcms monitored for completion of the reaction, the reaction was diluted with dichloromethane, slowly poured into ice water, separated, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and purified by spin-drying column chromatography (petroleum ether/ethyl acetate ═ 10/1) to give (2R,4S) -1- (tert-butylsulfonyl) -2- (2, 5-difluorophenyl) -4-fluoro as a yellow solid (1.2 g, 60%).
(2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidine
Figure PCTCN2019123407-APPB-000030
To a solution of (2R,4S) -1- (tert-butylsulfonyl) -2- (2, 5-difluorophenyl) -4-fluoro (500 mg, 1.55 mmol) in dichloromethane (20 ml) was added trifluoromethanesulfonic acid (0.7 ml), reacted for 2 hours, the solvent was dried, washed with 2 mol sodium hydroxide solution, extracted with ethyl acetate, separated, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and purified by spin-column chromatography (petroleum ether/ethyl acetate ═ 4/1) to give (2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidine as a yellow solid (305 mg, 99%).
Synthesis of intermediate C
Figure PCTCN2019123407-APPB-000031
(R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3-nitroimidazo [1,2-b ] pyridazine
Figure PCTCN2019123407-APPB-000032
Dimethyl sulfoxide (30 ml) was added to a three-necked flask (100 ml) at room temperature, the compounds 6-chloro-3-nitroimidazo [1,2-b ] pyridazine (1.0 g, 5.0 mmol), (R) -2- (2, 5-difluorophenyl) pyrrolidine (1.0 g, 5.5 mmol) and N, N-diisopropylethylamine (2.0 g, 15.1 mmol) were added, stirred overnight at 110 ℃, poured into ice water, extracted with ethyl acetate (100 ml _ 3), the organic phase was spin dried, and purified with a silica gel column (petroleum ether: ethyl acetate ═ 2:1) to give a yellow solid (650 mg, 37.6%).
(R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-amine
Figure PCTCN2019123407-APPB-000033
Ethanol (30 ml) was added to a three-necked flask (100 ml) at room temperature, the compound (R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3-nitroimidazo [1,2-b ] pyridazine (650 mg, 1.9 mmol), iron powder (516.4 mg, 9.2 mmol) and ammonium chloride (985.5 mg, 18.5 mmol) were added, stirred at 80 ℃ for 3 hours, cooled to room temperature, filtered, the filtrate was concentrated and extracted with ethyl acetate (50 ml × 3), the organic phase was spin-dried and purified with a silica gel column (dichloromethane: methanol: 50:1) to give a green solid (200 mg, 33.7%).
1HNMR(400MHz,CDCl 3):δ(ppm)7.52-7.50(m,1H),7.08-7.00(m,1H),6.98(s,1H),6.92-6.87(m,1H),6.83-6.78(m,1H),6.27(s,1H),5.30-5.27(m,1H),3.92-3.87(m,1H),3.70-3.63(m,1H),2.49-2.44(m,1H),2.10-2.01(m,5H).
Synthesis of intermediate D
Figure PCTCN2019123407-APPB-000034
6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) -3-nitroimidazo [1,2-b ] pyridazine
Figure PCTCN2019123407-APPB-000035
To a solution of (2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidine (150 mg, 0.75 mmol), 6-chloro-3-nitroimidazo [1,2-b ] pyridazine (222 mg, 1.12 mmol) in dimethyl sulfoxide (2 ml) was added triethylamine (151 mg, 1.5 mmol), and the reaction was stirred at 100 ℃ for 16 hours. LCMS showed most of the starting material disappeared. The reaction was purified by preparative high performance liquid chromatography to give a yellow solid (150 mg, 55%) as a yellow solid.
MS(ESI):m/z=363.9[M+H] +.
6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-amine
Figure PCTCN2019123407-APPB-000036
To a solution of 6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) -3-nitroimidazo [1,2-b ] pyridazine (60 mg, 0.16 mmol) in methanol (20 ml) was added palladium on charcoal (15 mg). The reaction solution was replaced three times with a hydrogen balloon by a water pump and then stirred at room temperature for 8 hours. LCMS showed most of the starting material disappeared. The reaction was filtered through celite and rinsed with dichloromethane (20 ml). The filtrate was concentrated and purified by preparative high performance liquid chromatography to give a yellow solid (30 mg, 57%).
MS(ESI):m/z=333.9[M+H] +.
Synthesis of intermediate E
Figure PCTCN2019123407-APPB-000037
6- (3-hydroxypyrrolidin-1-yl) nicotinic acid
Figure PCTCN2019123407-APPB-000038
Anhydrous N-butanol (10 mL) was added to a microwave tube (20 mL), compound 6-chloronicotinic acid (1.576 g, 10.0 mmol), compound pyrrolidin-3-ol (1.05 g, 12.0 mmol) and N, N-diisopropylethylamine (3.87 g, 30.0 mmol) were added. The reaction was stirred at 150 ℃ for 16 hours. The reaction was concentrated on a silica gel column and purified (dichloromethane: methanol ═ 4:1) to give a white solid (1.1 g, 52%).
1HNMR(400MHz,CDCl 3):δ(ppm)12.29(s,1H),8.61(d,J=2.0Hz,1H),7.90-7.87(m,1H),6.46(d,J=8.8Hz,1H),5.0(s,1H),4.39(s,1H),3.53-2.43(m,3H),2.0-1.2(m,2H),1.19(s,1H).
Example 1:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6- (3-hydroxypyrrolidine-1- Yl) Nicotinamide
Figure PCTCN2019123407-APPB-000039
To a solution of (R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-amine (25 mg, 0.08 mmol), N-diisopropylethylamine (30 mg, 0.24 mmol), 6- (3-hydroxypyrrolidin-1-yl) nicotinic acid (50 mg, 0.24 mmol) in dichloromethane (6 ml) was added 2- (7-oxabenzotriazol-oxide) -N, N' -tetramethyluronium hexafluorophosphate (130 mg, 0.24 mmol). The reaction solution was stirred at room temperature for 3 days. Concentrating, and purifying the residue by preparative high performance liquid chromatography to obtain white solid.
MS(ESI):m/z=506.2[M+H] +.
1HNMR(400MHz,CD 3OD)δ(ppm)8.63(s,1H),7.95(dd,J=9.0,2.3Hz,1H),7.65(d,J=9.9Hz,1H),7.51(s,1H),6.97-6.82(m,3H),6.79(d,J=10.0Hz,1H),6.58(d,J=9.0Hz,1H),5.32(d,J=5.6Hz,1H),4.61–4.52(m,1H),3.98–3.87(m,1H),3.72–3.49(m,5H),2.51-2.42(m,1H),2.26–2.14(m,1H),2.11-2.08(m,3H),1.99–1.87(m,1H).
Example 2:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6- ((S) -3-hydroxypyrroles Alk-1-yl) nicotinamide
Figure PCTCN2019123407-APPB-000040
(R) -N- (6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6-fluoroniacinamide
Figure PCTCN2019123407-APPB-000041
To a solution of (R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-amine (150 mg, 0.476 mmol), N-diisopropylethylamine (307 mg, 2.18 mmol), 6-fluoronicotinic acid (134 mg, 0.951 mmol) in dichloromethane (12 ml) was added 2- (7-oxabenzotriazol) -N, N' -tetramethylurea hexafluorophosphate (362 mg, 0.951 mmol). The reaction solution was stirred at room temperature for 3 days. Liquid quality monitoring showed product formation. Concentration and purification on column (ethyl acetate/dichloromethane ═ 5/1) gave a brown oil (197 mg, 94.4%).
MS(ESI):m/z=439.1[M+H] +.
N- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6- ((S) -3-hydroxypyrroles Alk-1-yl) nicotinamide
Figure PCTCN2019123407-APPB-000042
To (R) -N- (6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6-fluoronicotinamide (55 mg, 0.125 mmol l), N-diisopropylethylamine (129 mg, 1.004 mmol) in N, N-dimethylformamide (2 ml) was added (S) -3-hydroxypyrrolidine hydrochloride (47 mg, 0.374 mmol). The temperature was raised to 90 ℃ and stirred for 5 hours. Liquid quality monitoring showed product formation. Concentrating, and purifying the residue by preparative high performance liquid chromatography to obtain yellow solid. MS (ESI) 506.2[ M + H ]] +.
1HNMR(400MHz,CD 3OD)δ(ppm)8.61(d,J=1.7Hz,1H),7.94(dd,J=8.8,2.1Hz,1H),7.64(d,J=9.8Hz,1H),7.51(s,1H),6.98–6.81(m,3H),6.78(d,J=9.7Hz,1H),6.57(d,J=9.0Hz,1H),5.31(dd,J=8.2,2.7Hz,1H),4.56(s,3H),3.97–3.87(m,1H),3.72–3.50(m,4H),2.54–2.38(m,1H),2.23-2.13(m,1H),2.11–2.03(m,2H),1.98–1.86(m,1H).
Example 3:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6- ((R) -3-hydroxypyrroles Alk-1-yl) nicotinamide
Figure PCTCN2019123407-APPB-000043
The title compound was prepared from intermediate C and (R) -3-hydroxypyrrolidine hydrochloride under conditions analogous to example 2.
MS(ESI):m/z=506.5[M+H] +.
1HNMR(400MHz,CD 3OD)δ(ppm)8.62(s,1H),7.94(d,J=7.2Hz,1H),7.64(d,J=9.9Hz,1H),7.52(s,1H),6.95-6.85(m,3H),6.77(d,J=9.7Hz,1H),6.56(d,J=9.0Hz,1H),5.32(d,J=5.9Hz,1H),4.57(s,2H),3.97–3.88(m,1H),3.73–3.51(m,5H),2.52-2.42(m,1H),2.20-2.16(m,1H),2.13-2.02(m,3H),1.98-1.90(m,1H).
Example 4:
n- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -6- ((S) -3- Hydroxypyrrolidin-1-yl) nicotinamide
Figure PCTCN2019123407-APPB-000044
The title compound was prepared from intermediate D and (S) -3-hydroxypyrrolidine hydrochloride under conditions analogous to example 2.
MS(ESI):m/z=523.9[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.62(s,1H),8.66(s,1H),8.00–7.92(m,1H),7.78(d,J=9.8Hz,1H),7.46(s,1H),7.16–7.09(m,1H),7.06–6.89(m,2H),6.78–6.69(m,1H),6.52(d,J=9.0Hz,1H),5.53(s,0.5H),5.40(s,0.5H),5.30(m,1H),5.03–4.98(m,1H),4.40(s,1H),4.13–3.98(m,2H),3.60–3.49(m,2H),3.05–2.86(m,1H),2.79–2.67(m,1H),2.24–1.86(m,4H).
Example 5:
(R) -N- (6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- ((2-hydroxyethyl) amino) nicotinamide
Figure PCTCN2019123407-APPB-000045
The title compound was prepared from intermediate C and 2-aminoethan-1-ol under conditions analogous to example 2.
MS(ESI):m/z=480.0[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.53(s,1H),8.51(s,1H),7.76(d,J=9.8Hz,2H),7.43(s,1H),7.26-7.19(m,1H),7.02(m,2H),6.92–6.84(m,1H),6.68(d,J=9.8Hz,1H),6.54(d,J=8.9Hz,1H),5.24(d,J=6.0Hz,1H),4.75(m,1H),3.93–3.83(m,1H),3.61–3.48(m,3H),3.39(dd,J=11.6,5.8Hz,2H),2.43–2.30(m,1H),2.03-1.88(m,2H),1.85-1.72(m,1H).
Example 6:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- ((3-hydroxycyclopentyl) amino) nicotinamide
Figure PCTCN2019123407-APPB-000046
The title compound was prepared from intermediate C and 3-aminocyclopentane-1-ol hydrochloride under conditions analogous to example 2.
MS(ESI):m/z=519.9[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.51(s,1H),8.50(s,1H),7.77-7.74(m,2H),7.43(s,1H),7.21(d,J=7.4Hz,1H),7.08–6.96(m,2H),6.91–6.85(m,1H),6.68(d,J=9.6Hz,1H),6.50(d,J=8.9Hz,1H),5.25(d,J=5.9Hz,1H),4.64(d,J=3.9Hz,1H),4.26–4.07(m,2H),3.92-3.83(m,1H),3.55(dd,J=17.3,8.1Hz,1H),2.42–2.19(m,2H),2.05–1.88(m,3H),1.86-1.76(m,1H),1.75–1.52(m,3H),1.45–1.33(m,1H).
Example 7:
(R) -N- (6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- (dimethylamino) nicotinamide
Figure PCTCN2019123407-APPB-000047
The title compound was prepared from intermediate C and 3-aminocyclopentane-1-ol hydrochloride under conditions analogous to example 2.
MS(ESI):m/z=464.0[M+H]+.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.61(s,1H),8.61(s,1H),7.90(d,J=8.0Hz,1H),7.76(d,J=9.8Hz,1H),7.44(s,1H),7.08-6.98(m,2H),6.92-6.86(m,1H),6.69(d,J=8.8Hz,2H),5.24(d,J=6.5Hz,1H),3.95-3.85(m,1H),3.55(dd,J=17.4,7.8Hz,1H),3.27(s,1H),3.11(s,5H),2.42-2.29(m,1H),2.03-1.90(m,2H),1.85-1.76(m,1H).
Example 8:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -2- ((S) -3-hydroxypyrrolidin-1-yl) pyrimidine-5-carboxamide
Figure PCTCN2019123407-APPB-000048
The title compound was prepared from intermediate C and 2-chloropyrimidine-5-carboxylic acid under conditions analogous to example 2.
MS(ESI):m/z=506.9[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.81(s,1H),8.75(s,1H),7.77(d,J=10.0Hz,2H),7.43(s,1H),7.01–6.83(m,3H),6.73–6.49(m,1H),5.21–5.19(m,1H),5.01(d,J=3.2Hz,1H),4.39(s,1H),3.88–3.86(m,2H),3.69-3.52(m,5H),2.39–2.29(m,2H),2.04–1.78(m,6H).
Example 9:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -2- ((S) -3-hydroxypyrrolidin-1-yl) thiazole-5-carboxamide
Figure PCTCN2019123407-APPB-000049
Methyl (S) -2- (3-hydroxypyrrolidin-1-yl) thiazole-5-carboxylic acid ester
Figure PCTCN2019123407-APPB-000050
To a solution of methyl 2-bromothiazole-5-carboxylate (500 mg, 2.27 mmol) in 1, 4-dioxane (10 ml) was added (S) -3-hydroxypyrrolidine hydrochloride (234 mg, 2.72 mmol) and 1, 8-diazabicycloundec-7-ene (690 mg, 4.54 mmol). The reaction was stirred at 85 ℃ for 16 hours. The reaction was concentrated on a silica gel column (ethyl acetate as eluent) to give a white solid (465 mg, 90%).
MS(ESI):m/z=228.8[M+H] +.
(S) -2- (3-hydroxypyrrolidin-1-yl) thiazole-5-carboxylic acid
Figure PCTCN2019123407-APPB-000051
Methyl (S) -2- (3-hydroxypyrrolidin-1-yl) thiazole-5-carboxylate (465 mg, 2.04 mmol), lithium hydroxide monohydrate (257 mg, 6.11 mmol) were sequentially added to a mixed solution of tetrahydrofuran/water (10 ml/4 ml). Stirred at room temperature for 3 hours. LCMS showed reaction complete. The reaction solution was evaporated to remove tetrahydrofuran, 3 ml of water was added, and the solution was adjusted to pH 4 with 1N hydrochloric acid. The precipitated solid was filtered and washed four times with water. A solid was obtained as the title compound (400 mg, 91%).
N- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -2- ((S) -3-hydroxypyrrolidin-1-yl) thiazole-5-carboxamide
Figure PCTCN2019123407-APPB-000052
The title compound was prepared from intermediate C and (S) -2- (3-hydroxypyrrolidin-1-yl) thiazole-5-carboxylic acid under conditions analogous to example 1.
MS(ESI):m/z=511.9[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.61(s,1H),7.92(s,1H),7.74(d,J=9.6Hz,2H),7.38(s,1H),7.03–7.02(m,2H),6.89–6.86(m,1H),6.68–6.66(m,1H),5.25–5.22(m,1H),5.12(d,J=3.6Hz,1H),4.40(s,1H),3.88–3.84(m,2H),3.55-3.47(m,4H),2.39–2.28(m,2H),2.12–1.79(m,6H).
Example 10:
3-cyano-N- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -4- ((S) -3-hydroxypyrrolidin-1-yl) benzamide
Figure PCTCN2019123407-APPB-000053
The title compound was prepared from intermediate C and 3-cyano-4-fluorobenzoic acid under conditions analogous to example 2.
MS(ESI):m/z=529.9[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.76(s,1H),8.02(s,1H),7.84(d,J=8.0Hz,1H),7.77(d,J=9.2Hz,1H),7.41(s,1H),7.02–6.98(m,2H),6.89–6.85(m,1H),6.83(d,J=9.2Hz,1H),6.73–6.71(m,1H),5.22–5.21(m,1H),5.10(d,J=3.6Hz,1H),4.40(s,1H),3.89–3.85(m,1H),3.81-3.64(m,3H),3.56-3.48(m,2H),2.38–2.33(m,1H),2.03–1.90(m,5H),1.81–1.78(m,1H).
Example 11:
(R) -N- (6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- (pyrrolidin-1-yl) nicotinamide
Figure PCTCN2019123407-APPB-000054
The title compound was prepared from intermediate C and pyrrolidine under conditions analogous to example 2.
MS(ESI):m/z=489.9[M+H]+.
1HNMR(400MHz,CD 3OD)δ(ppm)8.61(s,1H),7.92(d,J=7.0Hz,1H),7.64(d,J=9.9Hz,1H),7.52(s,1H),6.95-6.85(m,3H),6.77(d,J=9.7Hz,1H),6.55(d,J=9.0Hz,1H),5.32(d,J=5.4Hz,1H),3.96–3.88(m,1H),3.65(dd,J=17.5,8.0Hz,1H),3.60-3.50(m,4H),2.49-2.44(m,1H),2.12–2.00(m,6H),2.00–1.90(m,1H).
Example 12:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- ((S) -3-hydroxypyrrolidin-1-yl) pyridazine-3-carboxamide
Figure PCTCN2019123407-APPB-000055
Methyl (S) -6- (3-hydroxypyrrolidin-1-yl) pyridazine-3-carboxylic acid ester
Figure PCTCN2019123407-APPB-000056
To a solution of methyl 6-chloropyridazine-3-carboxylate (1.0 g, 5.8 mmol) in tetrahydrofuran (20 ml) were added (S) -3-hydroxypyrrolidine hydrochloride (610 mg, 7.0 mmol), potassium carbonate (1.2 g, 0.58 mmol), and tetrabutylammonium iodide (210 mg, 0.58 mmol). The reaction was stirred at 70 ℃ for 16 hours. The reaction was quenched with water, extracted with dichloromethane and isopropanol (3:1), and the organic phase was concentrated to a silica gel column (dichloromethane/methanol-95/5) to give a white solid (1 g, 76%).
MS(ESI):m/z=224.3[M+H] +.
(S) -6- (3-hydroxypyrrolidin-1-yl) pyridazine-3-carboxylic acid
Figure PCTCN2019123407-APPB-000057
Methyl (S) -6- (3-hydroxypyrrolidin-1-yl) pyridazine-3-carboxylate (300 mg, 1.3 mmol), lithium hydroxide monohydrate (170 mg, 3.0 mmol) were sequentially added to a mixed solution of tetrahydrofuran/water (13 ml/5 ml). Stirred at room temperature for 2 hours. LCMS showed reaction complete. The reaction solution was evaporated to remove tetrahydrofuran, 3 ml of water was added, and the solution was adjusted to pH 4 with 1N hydrochloric acid. The precipitated solid was filtered and washed four times with water. A solid was obtained as the title compound (470 mg, 97%).
MS(ESI):m/z=209.9[M+H] +.
N- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- ((S) -3-hydroxypyrrolidin-1-yl) pyridazine-3-carboxamide
Figure PCTCN2019123407-APPB-000058
The title compound was prepared from intermediate C and (S) -6- (3-hydroxypyrrolidin-1-yl) pyridazine-3-carboxylic acid under conditions analogous to example 1.
MS(ESI):m/z=507.4[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)10.22(s,1H),7.88(d,J=9.4Hz,1H),7.76(d,J=9.9Hz,1H),7.64(s,1H),7.31–7.22(m,1H),7.08–6.93(m,3H),6.67–6.57(m,1H),5.25(d,J=5.5Hz,1H),5.09(s,1H),4.46(s,1H),3.93(s,1H),3.84–3.44(m,5H),2.45–2.36(m,1H),2.14–1.93(m,4H),1.91–1.81(m,1H).
Example 13:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-hydroxypyrrolidin-1-yl) pyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000059
The title compound was prepared from intermediate C and methyl 5-chloropyrazine-2-carboxylate under conditions analogous to example 12.
MS(ESI):m/z=507.3[M+H] +.
1HNMR(400MHz,DMSO-d6)δ(ppm)9.84(s,1H),8.68(d,J=1.1Hz,1H),7.95(s,1H),7.77(d,J=9.8Hz,1H),7.64(s,1H),7.24(td,J=9.3,4.5Hz,1H),7.07–6.96(m,2H),6.73–6.64(m,1H),5.29(d,J=5.6Hz,1H),5.09(s,1H),4.44(s,1H),3.99–3.89(m,1H),3.72–3.43(m,5H),2.45–2.35(m,1H),2.16–1.81(m,5H).
Example 14:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-hydroxypyrrolidin-1-yl) pyrimidine-2-carboxamide
Figure PCTCN2019123407-APPB-000060
The title compound was prepared from intermediate C and 5-bromopyrimidine-2-carboxylic acid under conditions analogous to example 12.
MS(ESI):m/z=501.8[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)10.17(s,1H),8.19(s,2H),7.77(d,J=9.8Hz,1H),7.67(s,1H),7.28–7.19(m,1H),7.07–6.97(m,2H),6.74–6.61(m,1H),5.34–5.28(m,1H),5.11(d,J=3.8Hz,1H),4.46(s,1H),3.99–3.91(m,1H),3.65–3.43(m,4H),2.14–1.81(m,5H).
Example 15:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b]Pyridazin-3-yl) -1- (3-hydroxycyclopentyl) - 1H-pyrazole-3-carboxamides (cis or tran)
Figure PCTCN2019123407-APPB-000061
Methyl 1- (3-carbonyl cyclopentyl) -1H-pyrazole-3-carboxylate
Figure PCTCN2019123407-APPB-000062
To a mixture of methyl 1H-pyrazole-3-carboxylate (1 g, 7.9 mmol) and cyclopent-2-en-1-one (1.3 g, 16.0 mmol) was added p-toluenesulfonic acid monohydrate (100 mg, 0.53 mmol). The reaction solution was stirred at 60 ℃ for 16 hours and then purified by flash column (dichloromethane to dichloromethane: methanol-9: 1). The crude product was purified by high performance liquid chromatography to afford a light yellow oil (1.1 g, 67%).
MS(ESI):m/z=208.9[M+H] +.
Methyl 1- (3-Hydroxycyclopentyl) -1H-pyrazole-3-carboxylate and example 492C methyl 1- (3-Hydroxycyclopentyl) -1H-pyrazole-3-carboxylate
Figure PCTCN2019123407-APPB-000063
Will be provided withMethyl 1- (3-carbonyl cyclopentyl) -1H-pyrazole-3-carboxylateA solution of (500 mg, 2.4 mmol) in methanol (20 ml) was cooled to 0 ℃. To the reaction was added sodium borohydride (500 mg, 13.0 mmol), the reaction was stirred at 0 ℃ for 1.5 hours and poured into a mixture of ice and water, extracted three times with ethyl acetate, dichloromethane: isopropanol-3: 1 was extracted three times. The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by flash separation (dichloromethane/methanol ═ 9/1) to give the desired compounds as colorless oils 15A-2-1(300 mg, 59%) and 15A-2-2(100 mg, 22%).
MS(ESI):m/z=210.9[M+H] +
N- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -1- (3-hydroxycyclopentyl) -1H-pyrazole-3-carboxamide
Figure PCTCN2019123407-APPB-000064
To a solution of 15A-2-1(84 mg, 0.4 mmol) in tetrahydrofuran (3 ml) and water (3 ml) was added lithium hydroxide (50 mg, 1.2 mmol). after stirring the reaction at room temperature for 3 hours, it was acidified with concentrated hydrochloric acid, concentrated and lyophilized. The resulting oil was dissolved in isopropanol and concentrated to give a white solid. A mixture of the resulting white solid and (R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-amine (50 mg, 0.16 mmol), 2- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (91 mg, 0.24 mmol), N, N-diisopropylethylamine (62 mg, 0.48 mmol) and anhydrous N, N-dimethylformamide (1 ml) was stirred at 40 ℃ for 16 hours. The reaction solution was purified by preparative high performance liquid chromatography to obtain compound 15A (33 mg, 42%) as a white solid.
MS(ESI):m/z=494.4[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.36(s,1H),8.00(d,J=2.3Hz,1H),7.77(d,J=9.7Hz,1H),7.56(s,1H),7.21–7.09(m,1H),7.09–7.01(m,1H),7.01–6.91(m,1H),6.75(d,J=2.2Hz,1H),6.66(d,J=9.2Hz,1H),5.30(d,J=6.7Hz,1H),4.93–4.75(m,2H),4.24(s,1H),3.97–3.89(m,1H),3.63–3.54(m,1H),2.46–2.36(m,2H),2.27–1.68(m,8H).
Figure PCTCN2019123407-APPB-000065
Lithium hydroxide (50 mg, 1.2 mmol) was added to a solution of 15A-2-2(84 mg, 0.4 mmol) in tetrahydrofuran (3 ml) and water (3 ml) and the reaction was stirred at room temperature for 3 hours, acidified with concentrated hydrochloric acid, concentrated and lyophilized. The resulting oil was dissolved in isopropanol and concentrated to give a white solid. A mixture of the resulting white solid and (R) -6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-amine (50 mg, 0.16 mmol), 2- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (91 mg, 0.24 mmol), N, N-diisopropylethylamine (62 mg, 0.48 mmol) and anhydrous N, N-dimethylformamide (1 ml) was stirred at 40 ℃ for 16 hours. The reaction was purified by preparative high performance liquid chromatography to give 15B (34 mg, 43%) as a white solid.
MS(ESI):m/z=494.4[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.56(s,1H),8.00(s,1H),7.94(d,J=10.0Hz,1H),7.87(s,1H),7.20–6.89(m,4H),6.75(d,J=1.4Hz,1H),5.33(d,J=7.8Hz,1H),5.08–4.94(m,1H),4.73(brs,1H),4.39(s,1H),4.01–3.91(m,1H),3.66–3.55(m,1H),2.46–2.37(m,1H),2.36–2.25(m,1H),2.22–1.79(m,7H),1.69–1.56(m,1H).
Example 16:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -1- (3-hydroxycyclopentyl) -1H-imidazole-4-carboxamide
Figure PCTCN2019123407-APPB-000066
The title compound was prepared from intermediate C and methyl 1H-imidazole-4-carboxylate under conditions analogous to example 15.
16A:MS(ESI):m/z=494.3[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.50(s,1H),7.96(d,J=3.8Hz,2H),7.76(d,J=9.9Hz,1H),7.60(s,1H),7.30–7.20(m,1H),7.08–7.01(m,1H),7.00–6.92(m,1H),6.71–6.61(m,1H),5.31–5.24(m,1H),4.87–4.77(m,1H),4.76–4.70(m,1H),4.35–4.27(m,1H),3.97–3.88(m,1H),3.63–3.54(m,1H),2.44–2.38(m,1H),2.34–2.23(m,1H),2.14–1.93(m,5H),1.91–1.82(m,1H),1.82–1.69(m,1H),1.57(s,1H).
16B:MS(ESI):m/z=494.3[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.69(s,1H),8.07–7.89(m,4H),7.29–7.17(m,1H),7.17–6.92(m,3H),5.31(d,J=6.5Hz,1H),4.76–4.64(m,1H),4.27–4.17(m,1H),4.03–3.94(m,1H),3.68–3.59(m,1H),2.46–2.35(m,2H),2.26–2.16(m,1H),2.08–1.83(m,4H),1.78–1.69(m,3H).
Example 17:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-hydroxypyrrolidin-1-yl) picolinamide
Figure PCTCN2019123407-APPB-000067
The title compound was prepared from intermediate C and 5-fluoro-o-picolinic acid under conditions analogous to example 2.
MS(ESI):m/z=505.9[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)10.16(s,1H),7.95(d,J=2.7Hz,1H),7.88(d,J=8.8Hz,1H),7.77(d,J=9.8Hz,1H),7.65(s,1H),7.28–7.20(m,1H),7.08–6.97(m,3H),6.75–6.65(m,1H),5.35–5.29(m,1H),5.07(d,J=3.7Hz,1H),4.45(s,1H),4.00–3.91(m,1H),3.65–3.44(m,4H),3.28–3.22(m,1H),2.17–1.83(m,6H).
Example 18:
(R) -N- (6- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- (oxetan-3-ylamino) nicotinamide
Figure PCTCN2019123407-APPB-000068
The target compound was prepared from intermediate C and oxetan-3-amine under conditions similar to example 2.
MS(ESI):m/z=492.4[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.59(s,1H),8.32(s,1H),8.24(d,J=12.0Hz,1H),7.76(d,J=9.5Hz,1H),7.52(t,J=9.0Hz,1H),7.38(s,1H),7.13-7.00(m,2H),6.93–6.82(m,1H),6.75-6.67(m,1H),6.46(dd,J=9.5,4.0Hz,1H),5.22(d,J=8.1Hz,1H),4.29(t,J=10.5Hz,1H),4.25–4.15(m,1H),4.13-4.05(m,1H),3.92-3.83(m,1H),3.57-3.48(m,2H),3.45-3.40(m,1H),2.45-2.32(m,1H),2.00-1.90(m,2H),1.85-1.75(m,1H).
Example 19:
n- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -6- ((S) -3-methoxypyrrolidin-1-yl) nicotinamide
Figure PCTCN2019123407-APPB-000069
The title compound was prepared from intermediate C and (S) -3-methoxypyrrolidine under conditions analogous to example 2.
MS(ESI):m/z=520.0[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.61(s,1H),8.61(s,1H),7.90(d,J=8.1Hz,1H),7.76(d,J=9.8Hz,1H),7.44(s,1H),7.10-7.00(m,2H),6.93-6.85(m,1H),6.68(d,J=9.0Hz,1H),6.52(d,J=8.9Hz,1H),5.24(d,J=6.2Hz,1H),4.12-4.06(m,1H),3.93-3.85(m,1H),3.65–3.48(m,4H),3.46-3.38(m,1H),3.26(s,3H),2.43–2.30(m,1H),2.15–2.02(m,2H),2.00-1.90(m,2H),1.85-1.77(m,1H).
Example 20:
n- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-hydroxypyrrolidin-1-yl) pyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000070
The title compound was prepared from intermediate D and methyl 5-chloropyrazine-2-carboxylate under conditions analogous to example 13.
MS(ESI):m/z=524.9[M+H] +
1HNMR(400MHz,DMSO-d 6)δ(ppm)9.81(s,1H),8.69(s,1H),8.04(s,1H),7.81(d,J=9.8Hz,1H),7.65(s,1H),7.29–7.23(m,1H),7.21–7.13(m,1H),7.02(mf,1H),6.77(d,J=9.6Hz,1H),5.52(d,J=52.3Hz,1H),5.38–5.32(m,1H),5.09(s,1H),4.45(s,1H),4.23–3.98(m,2H),3.73–3.46(m,4H),2.84–2.71(m,1H),2.31–1.91(m,3H).
Example 21:
n- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-hydroxypyrrolidin-1-yl) pyrimidine-2-carboxamide
Figure PCTCN2019123407-APPB-000071
The title compound was prepared from intermediate D and 5-bromopyrimidine-2-carboxylic acid under conditions analogous to example 14.
MS(ESI):m/z=524.9[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ(ppm)10.13(s,1H),8.31–8.18(m,2H),7.82(d,J=9.8Hz,1H),7.67(s,1H),7.36–7.25(m,1H),7.21–7.11(m,1H),7.06–6.96(m,1H),6.77(d,J=9.8Hz,1H),5.52(d,J=52.5Hz,1H),5.42–5.30(m,1H),5.12(s,1H),4.46(s,1H),4.23–3.98(m,2H),3.60–3.48(m,3H),3.31(d,J=10.3Hz,1H),2.87–2.73(m,1H),2.31–2.12(m,1H),2.11–2.01(m,1H),2.00–1.92(m,1H).
Example 22:
n- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) pyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000072
Tert-butyl (S) -3-methoxypyrrolidine-1-carboxylic acid ester
Figure PCTCN2019123407-APPB-000073
To a solution of (S) -1-N-t-butoxycarbonyl-3-hydroxypyrrolidine (1.0 g, 5.34 mmol) in N, N-dimethylformamide (10 ml) was added 60% sodium hydride (320 mg, 6.01 mmol) in portions at 0 ℃ and then reacted at 0 ℃ for half an hour. Methyl iodide (850 mg, 6.01 mmol) was added to the reaction solution, and the reaction was continued at room temperature for 2 hours. Water (50 ml) was added, extracted with ethyl acetate (100 ml _ 1), washed with water (100 ml _ 1) and saturated brine (100 ml _ 1) the organic phase was dried, filtered and concentrated to give the title compound 22A tert-butyl (S) -3-methoxypyrrolidine-1-carboxylate (1.05 g, 97.7%) as a colorless oil.
MS(ESI):m/z=223.9[M+Na] +.
(S) -3-methoxypyrrolidine hydrochloride
Figure PCTCN2019123407-APPB-000074
A solution of tert-butyl (S) -3-methoxypyrrolidine-1-carboxylate (1.05 g, 5.22 mmol) in 3N methanol hydrochloride (10 ml) was stirred at room temperature for 4 hours. The reaction was concentrated to give (S) -3-methoxypyrrolidine hydrochloride (710 mg, 98.8%) as a colorless oil.
MS(ESI):m/z=101.9[M+H] +.
N- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) pyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000075
To 6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrroleAlk-1-yl) imidazo [1,2-b]To a solution of pyridazin-3-amine (60 mg, 0.18 mmol), 5-chloropyrazine-2-carboxylic acid (57 mg, 0.36 mmol) and N, N-diisopropylethylamine (116 mg, 0.90 mmol) in N, N-dimethylformamide (1 ml) was added O- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate (110 mg, 0.29 mmol). The reaction was stirred overnight at room temperature. N, N-diisopropylethylamine (234 mg, 1.8 mmol) and 22B (S) -3-methoxypyrrolidine hydrochloride (124 mg, 0.90 mmol) were added to the reaction solution, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated and the reverse phase was followed to afford the title compound 22N- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1, 2-b)]Pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) pyrazine-2-carboxamide (55 mg, 56.7%) as a yellow solid. MS (ESI) M/z 539.0[ M + H ]] +.
1HNMR(400MHz,DMSO-d 6)δ9.81(s,1H),8.69(s,1H),8.05(s,1H),7.81(d,J=9.8Hz,1H),7.65(s,1H),7.28-7.22(m,1H),7.21-7.14(m,1H),7.05-6.97(m,1H),6.76(d,J=9.8Hz,1H),5.52(d,J=52.2Hz,1H),5.39–5.30(m,1H),4.24–3.97(m,3H),3.74-3.48(m,4H),3.27(s,3H),2.85–2.70(m,1H),2.31–2.19(m,1H),2.19-2.04(m,2H).
Example 23:
n- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) -N-methylpyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000076
N- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) pyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000077
According to the procedure for the synthesis of example 22, the title compound 23A N- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) pyrazine-2-carboxamide (140 mg, 85.8%) was obtained as a yellow solid.
MS(ESI):m/z=538.9[M+H] +.
Example 23N- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) -N-methylpyrazine-2-carboxamide
Figure PCTCN2019123407-APPB-000078
Sodium hydride (6 mg, 0.148 mmol) was added to a solution of N, N-dimethylformamide (2 ml) under ice-cooling, and after 1 hour at room temperature, iodomethane (21 mg, 0.148 mmol) was added and the reaction was continued at room temperature for 4 hours. Quench with water, add ethyl acetate, wash the organic phase with water and brine, dry over sodium sulfate, filter, and concentrate the filtrate. The residue was purified by reverse phase column to give the title compound 23N- (6- ((2R,4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) -5- ((S) -3-methoxypyrrolidin-1-yl) -N-methylpyrazine-2-carboxamide (9 mg, 19.5%) as a white solid.
MS(ESI):m/z=553.0[M+H] +.
1HNMR(400MHz,DMSO-d 6)δ7.77(d,J=9.8Hz,1H),7.36(s,1H),7.22–7.03(m,3H),6.86(t,J=23.6Hz,3H),5.49(d,J=52.9Hz,1H),5.28–5.18(m,1H),4.18-3.92(m,3H),3.39(s,3H),3.26(d,J=11.7Hz,1H),3.18(s,3H),3.09-2.84(m,2H),2.78–2.68(m,2H),2.19-2.08(m,1H),1.98-1.93(m,2H).
The following compounds were obtained by a method similar to that of example 2, substituting the corresponding starting materials.
Figure PCTCN2019123407-APPB-000079
Figure PCTCN2019123407-APPB-000080
Figure PCTCN2019123407-APPB-000081
Figure PCTCN2019123407-APPB-000082
Figure PCTCN2019123407-APPB-000083
Figure PCTCN2019123407-APPB-000084
Biological test example 1 TRKA, TRKB, TRKC kinase in vitro Activity test
Experimental Material
Recombinant human TRKA, TRKB, TRKC proteins were purchased from Carna Biosciences. HTRF kinEASE TKkit was purchased from CisbioBioassays. The BioTek microplate reader Synergy Neo 2 plate was used.
Experimental methods
Test compounds were diluted in 3-fold concentration gradient to a final concentration of 1 μ M to 0.05nM 10 concentrations, two replicate wells per concentration; the content of DMSO in the assay reaction was 1%.
TRKA enzyme reaction:
0.2 ng/. mu.l TRKA protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 14.68. mu.M ATP, 1 × enzymetic buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), and the reaction is carried out at room temperatureThe reaction was carried out for 40 minutes in a volume of 10. mu.l.
TRKB enzyme reaction:
0.037 ng/. mu.l TRKB protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 4.77. mu.M ATP, 1 × enzymetic buffer, 5mM MgCl2,1mMMnCl 21mM DTT. The assay plate was a White Proxiplate384-Plus plate (PerkinElmer) and reacted at room temperature for 50 minutes in a 10. mu.l reaction system.
TRKC enzyme reaction:
0.037 ng/. mu.l TRKC protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 25.64. mu.M ATP, 1 × enzymatic buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 40 minutes at room temperature, and the reaction system is 10 mu l.
Reaction detection:
add 10. mu.l of assay reagent to the reaction plate, containing SA-XL665 at a final concentration of 0.125. mu.M and 1 XTK-Antibody 5. mu.l, incubated overnight at room temperature, Synergy Neo 2 read plate.
Data analysis
The 665/620Ratio value was converted into an inhibition Ratio (%) (1-Ratio) by the following equationtest/Ratio max)×100%。Ratio maxRatio as a positive control without test CompoundtestThe values were measured for each concentration of different compounds. IC50(nM) data were obtained by 4-parameter curve fitting, as shown in Table 1.
TABLE 1
Compound TRKA(nm) TRKB(nm) TRKC(nm)
Example 1 <10 <10 <10
Example 2 <10 <10 <10
Example 3 <10 <10 <10
Example 4 <10 <10 <10
Example 5 <10 <10 <10
Example 6 <10 <10 <10
Example 7 <10 <10 <10
Example 8 <10 <10 <10
Example 9 <10 <10 <10
Example 10 <10 <10 <10
Example 11 <10 <10 <10
Example 12 <10 <10 <10
Example 13 <10 <10 <10
Example 14 <10 <10 <10
Example 15A <10 <10 <10
Example 15B <10 <10 <10
Example 16A <10 <10 <10
Example 16B <10 <10 <10
Example 17 <10 <10 <10
Example 18 <10 <10 <10
Example 19 <10 <10 <10
Example 20 <10 <10 <10
Example 21 <10 <10 <10
Example 22 <10 <10 <10
Example 23 <10 <10 <10
Example 24 <10 <10 <10
Example 25 <10 <10 <10
Example 26 <10 <10 <10
Example 27 <10 <10 <10
Example 28 <10 <10 <10
Example 29 <10 <10 <10
Example 30 <10 <10 <10
Example 31 <10 <10 <10
Example 32 <10 <10 <10
Example 33 <10 <10 <10
Example 34 <10 <10 <10
Example 35 <10 <10 <10
Example 36 <10 <10 <10
Example 37 <10 <10 <10
Example 38 <10 <10 <10
Example 39 <10 <10 <10
Example 40 <10 <10 <10
EXAMPLE 41 <10 <10 <10
Example 42 <10 <10 <10
Example 43 <10 <10 <10
Example 44 <10 <10 <10
Example 45 <10 <10 <10
Bioassay example 2 KM12-LUC cell proliferation assay
A human colon cancer cell line KM12-LUC (LUC, stably expressing Luciferace) containing TPM3-NTRK1 fusion gene is used as a model for evaluating the pharmacodynamics of a test compound at a cytological level. The TRK fusion gene in KM12-LUC cells makes the cells independent of stimulation of extracellular growth factors, and can continuously and spontaneously activate downstream signaling pathways MAPK-ERK, PI3K-AKT and other signaling pathways closely related to cell proliferation. Therefore, inhibition of TRK activity in KM12-LUC cells significantly inhibited cell proliferation. The method comprises the following steps: the first day, cells were seeded in 384-well plates, 2000 cells/well; adding the compounds to be detected with different concentrations on the next day; on the fifth day, cell activity was measured by adding CellTiter-glo (Promega), and the cell proliferation inhibition rate was calculated for 72 hours. Statistical analysis with prism5 and IC of test compound50The values are shown in Table 2.
The results show that the compound of the invention can effectively inhibit the proliferation of KM12-LUC cells.
TABLE 2
Compound KM12-LUC IC 50(nm)
Example 4 <10
Example 10 <10
Example 13 <10
Example 27 <10
EXAMPLE 41 <10
Bioassay 3 mutant TRKA (G595R), TRKA (G667C) and TRKC (G623R) kinase in vitro Activity assay
Experimental Material
Recombinant human TRKA (G595R), TRKA (G667C), and TRKC (G623R) proteins were purchased from SignalChem. HTRF kinEASE TKkit was purchased from CisbioBioassays. The BioTek microplate reader Synergy Neo 2 plate was used.
Experimental methods
Test compounds were diluted in 4-fold concentration gradient to a final concentration of 10 concentrations from 1 μ M to 0.004nM, two duplicate wells per concentration; the content of DMSO in the assay reaction was 1%.
TRKA (G595R) enzyme reaction:
0.12 ng/. mu.l TRKA (G595R) protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 4.5. mu.M ATP, 1 × enzymic buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 30 minutes at room temperature, and the reaction system is 10 mu l.
TRKA (G667C) enzyme reaction:
0.026 ng/. mu.l TRKA (G667C) protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 5.5. mu.M ATP, 1 × enzymic buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 30 minutes at room temperature, and the reaction system is 10 mu l.
TRKC (G623R) enzymatic reaction:
1.0 ng/. mu.l TRKC (G623R) protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 62.9. mu.M ATP, 1 × enzymic buffer, 5mM MgCl21mM DTT. The assay plate was a White Proxiplate384-Plus plate (PerkinElmer) and reacted at room temperature for 50 minutes in a 10. mu.l reaction system.
Reaction detection:
add 10. mu.l of assay reagent to the reaction plate, containing SA-XL665 at a final concentration of 0.125. mu.M and 1 XTK-Antibody 5. mu.l, incubated overnight at room temperature, Synergy Neo 2 read plate.
Data analysis
The value of 665/620Ratio minus the value of the enzyme-free negative control well was converted to the percent inhibition (%) (1-Ratio) by the following equationtest/Ratio max)×100%。Ratio maxRatio as a positive control without test CompoundtestThe values were measured for each concentration of different compounds. IC50(nM) data were obtained by 4-parameter curve fitting, as shown in Table 3.
TABLE 3
Figure PCTCN2019123407-APPB-000085
Figure PCTCN2019123407-APPB-000086
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

  1. A compound of formula I:
    Figure PCTCN2019123407-APPB-100001
    wherein the content of the first and second substances,
    r has the formula-L1-R AThe structure of (1);
    L 1selected from the group consisting of: substituted or unsubstituted 5-to 10-membered heterocyclylene having 1 to 3 heteroatoms selected from N, S and O, or substituted or unsubstituted- (X)y-wherein each of said xs is independently selected from the group consisting of: substituted or unsubstituted C1-C 8Alkylene, -O-, -C (═ O) -, -CONH-, -NHCO-, -S (═ O)2-、-NH-;
    y is selected from the group consisting of: 1.2 or 3;
    R Aselected from the group consisting of: substituted or unsubstituted C6-C 10Aryl, substituted or unsubstituted 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
    Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7each independently selected from the group consisting of: CR1Or N; z1、Z 2Each independently selected from the group consisting of: c or N;
    and is
    Figure PCTCN2019123407-APPB-100002
    Is an aromatic ring;
    R 1each independently selected from the group consisting of: H. d, OH, Cl, F and NH2
    R 12Selected from the group consisting of: h or C1-C4 alkyl;
    ring a is selected from the group consisting of: substituted or unsubstituted C6-C 10Aryl, substituted or unsubstituted 5-10 membered heteroaryl (including monocyclic, fused cyclic) having 1-3 heteroatoms selected from N, S and O;
    l is selected from the group consisting of: none, -NR7-,-O-;
    R 7Selected from: h, substituted C1-C6 alkyl; wherein said substitution means that one or more H atoms on the group are substituted with a substituent selected from the group consisting of: halogen, hydroxy;
    ring C is selected from the group consisting of: substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted 3-12 membered cycloalkyl (including monocyclic, fused, spiro or bridged), substituted or unsubstituted 4-12 membered heterocyclyl (including monocyclic, fused, spiro or bridged) having 1-3 heteroatoms selected from N, S and O;
    unless otherwise specified, "substituted" means substituted with one or more (e.g., 2,3, 4, etc.) substituents selected from the group consisting of: halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH 2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 carboxamido)、
    Figure PCTCN2019123407-APPB-100003
    Or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-12 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O, - (CH) 6 alkyl2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2OH, -OH, C6-C10 aryl, C1-C6 amino, C1-C6 amido, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
    Figure PCTCN2019123407-APPB-100004
    is the attachment site of the group;
    with the proviso that the compounds of formula I are chemically stable structures.
  2. The compound of claim 1, wherein L is1Selected from the group consisting of:
    Figure PCTCN2019123407-APPB-100005
    n is selected from the group consisting of: 0.1, 2 or 3;
    R 2、R 2aand R2bEach independently selected from the group consisting of: H. OH, halogen, substituted or unsubstituted C1-C 8An alkyl group;
    X 5selected from the group consisting of: NH, O, -CONH-, -NHCO-, S, -S (═ O)2-、-NHS(=O)-、-NHS(=O) 2-;
    R AIs composed of
    Figure PCTCN2019123407-APPB-100006
    Wherein, the
    Figure PCTCN2019123407-APPB-100007
    Means RAAnd L1The attachment site of (a); x4Is CH or N;
    R 3selected from the group consisting of: H. halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy.
  3. The compound of claim 1, wherein said compound has the structure shown in formula II:
    Figure PCTCN2019123407-APPB-100008
  4. the compound of claim 1, wherein the compound has the structure shown in formula III below:
    Figure PCTCN2019123407-APPB-100009
  5. the compound of claim 1, wherein said compound has a structure selected from the group consisting of:
    Figure PCTCN2019123407-APPB-100010
    Figure PCTCN2019123407-APPB-100011
    Figure PCTCN2019123407-APPB-100012
    Figure PCTCN2019123407-APPB-100013
    Figure PCTCN2019123407-APPB-100014
    Figure PCTCN2019123407-APPB-100015
  6. a pharmaceutical composition comprising (1) a compound of claim 1 or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof; (2) a pharmaceutically acceptable carrier.
  7. The use according to claim 6, wherein the disease is selected from the group consisting of: cancer, proliferative diseases, pain, skin diseases or conditions, metabolic diseases, muscular diseases, neurological diseases, autoimmune diseases, dermatitis-induced pruritus, inflammation-related diseases, bone-related diseases.
  8. Use of a compound according to claim 1 or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition according to claim 6, for the preparation of a pharmaceutical composition for the prophylaxis and/or treatment of a disease associated with an abnormal TRK function (abnormal TRK activation due to amplification, or overexpression, or mutation, or gene fusion).
  9. The use according to claim 8, wherein the disease is selected from the group consisting of: the disease is selected from the group consisting of: cancer, proliferative diseases, pain, skin diseases or conditions, metabolic diseases, muscular diseases, neurological diseases, autoimmune diseases, dermatitis-induced pruritus.
  10. A TRK inhibitor, wherein said inhibitor comprises a compound of claim 1, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
CN201980081131.8A 2018-12-05 2019-12-05 Preparation and application of imidazo aromatic ring compounds Pending CN113166162A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2018114810619 2018-12-05
CN201811481061.9A CN111269233A (en) 2018-12-05 2018-12-05 Preparation and application of imidazo aromatic ring compounds
PCT/CN2019/123407 WO2020114465A1 (en) 2018-12-05 2019-12-05 Preparation and application of heteroaromatic iminazole compound

Publications (1)

Publication Number Publication Date
CN113166162A true CN113166162A (en) 2021-07-23

Family

ID=70974936

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201811481061.9A Pending CN111269233A (en) 2018-12-05 2018-12-05 Preparation and application of imidazo aromatic ring compounds
CN201980081131.8A Pending CN113166162A (en) 2018-12-05 2019-12-05 Preparation and application of imidazo aromatic ring compounds

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN201811481061.9A Pending CN111269233A (en) 2018-12-05 2018-12-05 Preparation and application of imidazo aromatic ring compounds

Country Status (2)

Country Link
CN (2) CN111269233A (en)
WO (1) WO2020114465A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102224153A (en) * 2008-09-22 2011-10-19 阵列生物制药公司 Substituted imidazo[1,2b]pyridazine compounds as trk kinase inhibitors
CN102264736A (en) * 2008-10-22 2011-11-30 阵列生物制药公司 Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
CN102596957A (en) * 2009-07-09 2012-07-18 阵列生物制药公司 Substituted pyrazolo[1,5- a]pyrimidine compounds as TRK kinase inhibitors
CN106456581A (en) * 2014-03-26 2017-02-22 默沙东公司 Trka kinase inhibitors, compositions and methods thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102224153A (en) * 2008-09-22 2011-10-19 阵列生物制药公司 Substituted imidazo[1,2b]pyridazine compounds as trk kinase inhibitors
CN102264736A (en) * 2008-10-22 2011-11-30 阵列生物制药公司 Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
CN102596957A (en) * 2009-07-09 2012-07-18 阵列生物制药公司 Substituted pyrazolo[1,5- a]pyrimidine compounds as TRK kinase inhibitors
CN106456581A (en) * 2014-03-26 2017-02-22 默沙东公司 Trka kinase inhibitors, compositions and methods thereof

Also Published As

Publication number Publication date
WO2020114465A1 (en) 2020-06-11
CN111269233A (en) 2020-06-12

Similar Documents

Publication Publication Date Title
CN113286794B (en) KRAS mutein inhibitors
CN113242857A (en) Preparation and application of imidazo aromatic ring compounds
DE60315615T2 (en) TRICYCLIC COMPOUNDS BASED ON THIOPHES AND MEDICAMENTS COMPRISING THEM
JP2022523274A (en) JAK inhibitor compounds and their use
CN113166142B (en) Five-membered and six-membered heterocyclic compounds and application thereof as protein receptor kinase inhibitors
KR20150028999A (en) 5-azaindazole compounds and methods of use
AU2014307437A1 (en) Novel fused pyrimidine compound or salt thereof
AU2017323112B2 (en) Pyrido five-element aromatic ring compound, preparation method therefor and use thereof
KR20230043955A (en) Compounds with kinase inhibitory activity
CN112654605B (en) Bridged heterocyclic group substituted pyrimidine compound and preparation method and medical application thereof
CA3120430C (en) Six-membered fused with six-membered heterocyclic compound and uses thereof serving as protein receptor kinase inhibitor
CN113637013A (en) Preparation and application of biaryl ring linked aromatic heterocyclic derivative as immunomodulator
KR20210093870A (en) MDM2 inhibitor, preparation method thereof, pharmaceutical composition thereof and use thereof
CN113166162A (en) Preparation and application of imidazo aromatic ring compounds
CN114641476B (en) Preparation and application of protein receptor kinase inhibitors
TW202237596A (en) Fused ring compound as wee-1 inhibitor
EP2986605B1 (en) Rohitukine analogs as cyclin-dependent kinase inhibitors and a process for the preparation thereof
CN111978325B (en) Imidazopyridazine MNK1/MNK2 kinase inhibitor, and preparation method and application thereof
CN117186097A (en) Carboxamide derivative with RSK inhibition effect, pharmaceutical composition containing carboxamide derivative and application of carboxamide derivative
WO2023236960A1 (en) Carboxamide derivative having rsk inhibitory effect, pharmaceutical composition comprising same, and use thereof
CN117402161A (en) Sulfoxide imine compound with FGFR inhibition effect, pharmaceutical composition containing sulfoxide imine compound and application of sulfoxide imine compound
CN111978318A (en) Imidazopyridine MNK1/MNK2 kinase inhibitor and preparation method and application thereof
CN117069719A (en) Diazaspiro-pyridazine compounds and uses thereof
CN116655636A (en) Five-membered and six-membered heterocyclic compounds and application thereof as protein kinase inhibitors
CN113372364A (en) JAK kinase inhibitor and preparation and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination