WO2020114499A1 - Tyrosine kinase inhibitors, compositions and methods there of - Google Patents

Tyrosine kinase inhibitors, compositions and methods there of Download PDF

Info

Publication number
WO2020114499A1
WO2020114499A1 PCT/CN2019/123719 CN2019123719W WO2020114499A1 WO 2020114499 A1 WO2020114499 A1 WO 2020114499A1 CN 2019123719 W CN2019123719 W CN 2019123719W WO 2020114499 A1 WO2020114499 A1 WO 2020114499A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrrolidin
pyrazolo
difluorophenyl
pyrimidin
benzo
Prior art date
Application number
PCT/CN2019/123719
Other languages
French (fr)
Inventor
Bang Fu
Yinlong LI
Wei Ren
Jie Chen
Xiangyong LIU
Jiabing Wang
Lieming Ding
Original Assignee
Betta 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
Priority to MX2021006619A priority Critical patent/MX2021006619A/en
Priority to JP2021531534A priority patent/JP2022510380A/en
Priority to CA3122136A priority patent/CA3122136A1/en
Priority to EP19892359.1A priority patent/EP3891152A4/en
Priority to SG11202105881RA priority patent/SG11202105881RA/en
Priority to AU2019394520A priority patent/AU2019394520A1/en
Priority to EA202191318A priority patent/EA202191318A1/en
Priority to CN201980077490.6A priority patent/CN113166156B/en
Application filed by Betta Pharmaceuticals Co., Ltd filed Critical Betta Pharmaceuticals Co., Ltd
Priority to KR1020217018370A priority patent/KR20210124961A/en
Priority to US17/311,105 priority patent/US20210395256A1/en
Priority to BR112021010930-7A priority patent/BR112021010930A2/en
Publication of WO2020114499A1 publication Critical patent/WO2020114499A1/en
Priority to IL283599A priority patent/IL283599A/en
Priority to ZA2021/03858A priority patent/ZA202103858B/en

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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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

Definitions

  • the present application is concerned with pharmaceutically active compounds.
  • the disclosure provides compounds as well as their compositions and methods of use.
  • the compounds inhibit tropomyosin-related kinases (Trks) and are useful in the treatment of various diseases including infectious diseases and cancer.
  • Trks tropomyosin-related kinases
  • Tropomyosin-related kinases are a group of receptor tyrosine kinases which are regulated by neurotrophins, including 3 members TrkA, TrkB and TrkC, encoded by the genes NTRK1, NTRK2 and NTRK3 respectively. Many cellular functions, for example, cell proliferation, cell differentiation, metabolism and apoptosis are mediated by Trks through phosphorylation and regulation of their downstream signal pathway members. Gene fusions involving NTRK genes result in continuous activation or overexpression of these kinases, which increase the risk of tumor genesis.
  • Trk plays an important physiological role in the development of nerves, including the growth and function maintenance of neuronal axons, the development of memory and the protection of neurons from injury, etc. Also, it is showed that Trk uncommonly expresses in normal tissues or cancer, while fusion drives abnormally high expression and activation of Trk kinase domain. Trk fusions are found in diverse cancer histologies with low fusion frequency, such as thyroid cancer, lung cancer, colon cancer, and melanoma. It is estimated that 1,500-5,000 patients harbor Trk fusion-positive cancers in the United States annually.
  • Trk fusion protein is becoming a valid cancer target
  • the small molecule inhibitor for Trk with most rapid development is Loxo Oncology's larotrectinib which is highly potent against Trk in clinical development.
  • WO2010048314, WO2011006074, WO2016097869, and WO2018077246 disclosed a series of Trk inhibitors. Accordingly, there is still a great demand for Trk inhibitors which have more potent activity, and better liver microsomes metabolic stability.
  • Trk inhibitors which can inhibit not only Trk A, B, and C but also mutated forms of Trk A, B and C (for example the G595R, G667C, A608D, F589L, G623R) which are reported in patients receiving first generation Trk kinase inhibitors.
  • Trk inhibitors which can inhibit not only Trk A, B, and C but also mutated forms of Trk A, B and C (for example the G595R, G667C, A608D, F589L, G623R) which are reported in patients receiving first generation Trk kinase inhibitors.
  • potent small molecules that can have activity as Trk inhibitors, and thus may be useful for therapeutic administration to fight against cancer and/or infectious diseases. These small molecules are expected to be useful as pharmaceuticals with desirable stability, solubility, bioavailability, therapeutic index and toxicity values that are crucial to become efficient medicines to promote human health.
  • the present invention relates to compounds that are used as Trk inhibitors. Trk inhibitors are useful in the treatment of cancers and infectious diseases.
  • the compounds of the invention have the general structures as Formula I.
  • ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • ring B is 5-membered aromatic heterocycle
  • X and Z are each independently selected from C, N, O, or S;
  • Y is C or N
  • R 1 is absent, H, or -C 1-8 alkyl
  • R 2 is H, -C 0-4 alkyl-COOR 10 , -C 0-4 alkyl-NH-COOR 10 , -C 0-4 alkyl-O (CO) R 10 , -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10 , -C 1-4 alkyl-NH 2 , -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, or -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring, wherein the -C 0-4 alkyl-COOR 10 , -C 0-4 alkyl-NH-COOR 10 , -C 0-4 alkyl-O (CO) R 10 , -C 0-4 alkyl-O (CO
  • R 3 is absent, C 3-10 heterocyclic ring
  • R 2 and R 3 together with the atoms to which they are attached to form a 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring, wherein the 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring is optionally substituted with halogen, OH, CN, NH 2 , -CONHOH, -CONH 2 , -C 0-4 alkyl-COOR 10 , -C 0-4 alkyl-O (CO) OR 10 , -C 1-8 alkoxy, -C 1-8 haloalkoxy, -C 1-8 alkoxy-C 1-8 alkoxy, -C 1-8 alkylthio, -C 1-8 haloalkylthio, -C 1-8 alkyl, -C 0-4 alkyl-OH, -O-CH 2
  • R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
  • R 10 is H, or -C 1-8 alkyl
  • heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, O or B.
  • ring A is
  • X is independently selected from O, S or N.
  • Y is C.
  • Z is N.
  • R 4 is
  • the compound is of Formula II or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
  • ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • R 1 is H, or -C 1-8 alkyl
  • R 2 is H, -C 0-4 alkyl-COOR 10 , -C 0-4 alkyl-NH-COOR 10 , -C 0-4 alkyl-O (CO) R 10 , -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10 , -C 1-4 alkyl-NH 2 , -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, or -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring, wherein the -C 0-4 alkyl-COOR 10 , -C 0-4 alkyl-NH-COOR 10 , -C 0-4 alkyl-O (CO) R 10 , -C 0-4 alkyl-O (CO
  • R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
  • R 10 is H, or -C 1-8 alkyl
  • heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, O or B.
  • ring A is
  • R 1 is independently selected from H or CH 3 .
  • R 4 is
  • R 2 is independently selected from
  • R 2 is
  • the compound is of Formula III or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof:
  • ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • ring C is a 5-to 6-membered carbocyclic ring, or heterocyclic ring, aryl ring, or heteroaryl ring;
  • X and Z are each independently selected from C, N, O, or S;
  • Y is C or N
  • R 1 is absent, H, or -C 1-8 alkyl
  • R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
  • R 5 and R 6 are each independently selected from H, OH, NH 2 , CN, -COOH, -CONHOH, -CONH 2 , halogen, -C 1-8 alkyl, -C 0-4 alkyl-COOR 10 , -C 0-4 alkyl-O (CO) OR 10 , -C 1-8 alkoxy, -C 1-8 haloalkoxy, -C 1-8 alkoxy-C 1-8 alkoxy, -C 1-8 alkylthio, -C 1-8 haloalkylthio, -C 1-8 alkyl, -C 1-8 haloalkyl, -C 0-4 alkyl-OH, -O-CH 2 -CN, -C 0-4 alkyl-O-C 3-10 heterocyclic ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring;
  • R 5 and R 6 together with the atoms to which they are attached to form a 5 to 12-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring, wherein the 5 to 12-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring is optionally substituted with halogen;
  • R 10 is H, or -C 1-8 alkyl
  • heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, or O.
  • ring A is
  • ring C is 6-membered aromatic ring.
  • ring C is phenyl, pyridyl, pyridazinyl, or pyrimidinyl.
  • ring C is phenyl
  • X is selected from O, S, or N.
  • X is N.
  • Y is C.
  • Z is N.
  • R 1 is absent, H, or CH 3 .
  • R 4 is
  • R 5 and R 6 are each independently selected from H, OH , NH 2 , F, Cl, Br -CN, -CF 3 , -OCF 3 , CH 3 , -O-CH 3 , -S-CH 3 , -CH 2 OH , -COOH ,
  • R 5 and R 6 are both -O-CH 3 .
  • R 5 and R 6 together with the atoms to which they are attached from
  • the compound is of Formula IV or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
  • Ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, -C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
  • R’ is H, NH 2 , or -C 1-4 alkyl
  • Ring B’ is a 5-membered aromatic heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • Ring C’ is a phenyl, 6-membered heterocyclic ring, or 6-membered heteroaryl ring;
  • X’ and Z’ are each independently selected from C, N, O, or S;
  • Y’ is C or N
  • R” is -C (O) -C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2 -C 1-4 alkyl, -NR 7 (CH 2 ) m NR 8 R 9 , - (CH 2 ) m C 4-10 heterocyclyl; or NH 2 , -C (O) OH, -C (O) NH 2 , -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alkyl, -C (O) O-C 1-4 alkyl, -OC (O) O-C 1-4 alkyl, -S-C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2 -C 1-4 alkyl, -OC 4-6 heterocyclyl, -NR 7 (CH 2 ) m NR 8 R 9 , - (CH 2 ) m C 4-10 heterocyclyl optionally substituted with one or more
  • R 7 , R 8 and R 9 are each independently selected from H, or -C 1-4 alkyl
  • n are each independently selected from 0, 1, 2, 3 or 4.
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R’ is selected from H.
  • R 4 is
  • Ring B’ is selected from imidazole, oxazole, thiazole, triazole or pyrrole.
  • Ring B’ is selected from
  • Ring C’ is selected from phenyl, pyridine, pyrazine, pyrimidine, pyridazine, piperidine, or tetrahydropyran.
  • Ring C’ is selected from
  • R is selected from
  • Ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, -C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
  • R 11 is H, NH 2 , or -C 1-4 alkyl
  • Ring B is a 5-membered aromatic heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
  • Ring C is a phenyl, 6-membered heterocyclic ring, or 6-membered heteroaryl ring;
  • X and Z are each independently selected from C, N, O, or S;
  • Y is C or N
  • R 12 is selected from H, OH, CN, NH 2 , -C (O) OH, -C (O) NH 2 , halogen, -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alkyl, -C (O) O-C 1-4 alkyl, -OC (O) O-C 1-4 alkyl, -S-C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2 -C 1-4 alkyl, -OC 4-6 heterocyclyl, -NR 7 ’ (CH 2 ) m NR 8 ’R 9 ’, - (CH 2 ) m C 4-10 heterocyclyl; wherein NH 2 , -C (O) OH, -C (O) NH 2 , halogen, -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alky
  • R 7 ’, R 8 ’ and R 9 ’ are each independently selected from H, or -C 1-4 alkyl;
  • n are each independently selected from 0, 1, 2, 3 or 4.
  • R 11 is selected from H, NH 2 or CH 3 .
  • Ring B is selected from imidazole, oxazole, thiazole, triazole or pyrrole.
  • Ring B is selected from
  • Ring C is selected from phenyl, pyridine, pyrazine, pyrimidine, pyridazine, piperidine or tetrahydropyran.
  • Ring C is selected from
  • R 12 is selected from H, -OH, F, Cl, Br, -CH 3 , -NH 2 , -COOH, -CN,
  • the present invention further provides some preferred technical solutions with regard to compound of Formula I, Formula II, Formula III, Formula IV, or Formula V, or an isomeride, pharmaceutically acceptable salt or solvate thereof, wherein the compound is:
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of any one of the present invention, or a pharmaceutically acceptable salt or a stereoisomer thereof, and at least one pharmaceutically acceptable carrier or excipient.
  • the present invention additionally provided a method of inhibiting Trk, including wildtype TrkA, TrkB and TrkC, the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, and the TrkC G623R, said method comprising administering to a patient a compound of any one of the present invention or a pharmaceutically acceptable salt or an isomeride thereof.
  • the present invention further provides a method of treating a disease associated with inhibition of Trk, including wildtype TrkA, TrkB and TrkC, the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, and the TrkC G623R, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound in any one in the present invention, or a pharmaceutically acceptable salt or an isomeride thereof.
  • the disease is mammary analogue secretory carcinoma (MASC) of the salivary glands, infantile fibrosarcoma, spitz tumors, colon cancer, gastric cancer, thyroid cancer (such as papillary thyroid cancer) , lung cancer, leukemia, pancreatic cancer, melanoma (sunch as multiple melanoma) , brain cancer (such pontine glioma) , renal cancer (such as congenital mesoblastic nephroma) , prostate cancer, ovarian cancer or breast cancer (such as secretory breast carcinoma) .
  • MSC mammary analogue secretory carcinoma
  • the present invention provided a method of inhibiting Trk in a patient, said method comprising administering to the patient in need thereof a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or an isomeride thereof.
  • the present invention also provides a use of the present compound or its pharmaceutical composition for the preparation of a medicament.
  • the medicament is used for the treatment or prevention of cancer.
  • the disease is mammary analogue secretory carcinoma (MASC) of the salivary glands, infantile fibrosarcoma, spitz tumors, colon cancer, gastric cancer, thyroid cancer (such as papillary thyroid cancer) , lung cancer, leukemia, pancreatic cancer, melanoma (such as multiple melanoma) , brain cancer (such pontine glioma) , renal cancer (such as congenital mesoblastic nephroma) , prostate cancer, ovarian cancer or breast cancer (such as secretory breast carcinoma) .
  • MSC mammary analogue secretory carcinoma
  • Trk is wildtype TrkA, TrkB, TrkC, or the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, or the TrkC G623R.
  • the present invention also provides a method of enhancing, stimulating and/or increasing the immune response in a patient, said method comprising administering to the patient in need thereof a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or an isomeride thereof.
  • halogen as used herein, unless otherwise indicated, means fluoro, chloro, bromo or iodo.
  • halogen groups include F, Cl and Br.
  • alkyl includes saturated monovalent hydrocarbon radicals having straight, branched or cyclic moieties.
  • alkyl radicals include methyl, ethyl, propyl, isopropyl, cyclcopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, cyclcobutyl, n-pentyl, 3- (2-methyl) butyl, 2-pentyl, 2-methylbutyl, neopentyl, cyclcopentyl, n-hexyl, 2-hexyl, 2-methylpentyl and cyclohexyl.
  • C 1-8 as in C 1-8 alkyl is defined to identify the group as having 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms in a linear or branched arrangement.
  • Alkenyl and alkynyl groups include straight, branched chain or cyclic alkenes and alkynes.
  • C 2-8 alkenyl and “C 2-8 alkynyl” means an alkenyl or alkynyl radicals having 2, 3, 4, 5, 6, 7 or 8 carbon atoms in a linear or brached arrangement.
  • Alkoxy radicals are oxygen ethers formed from the previously described straight, branched chain or cyclic alkyl groups.
  • aryl refers to an unsubstituted or substituted mono-or polycyclic ring system containing carbon ring atoms.
  • the preferred aryls are mono cyclic or bicyclic 6-10 membered aromatic ring systems. Phenyl and naphthyl are preferred aryls. The most preferred aryl is phenyl.
  • heterocyclyl represents an unsubstituted or substituted stable three to eight membered monocyclic saturated ring system which consists of carbon atoms and from one to three heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heterocyclyl group may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclyl groups include, but are not limited to azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxoazepinyl, azepinyl, tetrahydrofuranyl, dioxolanyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone and oxadiazolyl.
  • heteroaryl represents an unsubstituted or substituted stable five or six membered monocyclic aromatic ring system or an unsubstituted or substituted nine or ten membered benzo-fused heteroaromatic ring system or bicyclic heteroaromatic ring system which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heteroaryl group may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heteroaryl groups include, but are not limited to thienyl, furanyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, pyridyl, pyridazinyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, benzofuranyl, benzothienyl, benzisoxazolyl, benzoxazolyl, benzopyrazolyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl adeninyl, quinolinyl or isoquinolinyl.
  • alkenyloxy refers to the group -O-alkenyl, where alkenyl is defined as above.
  • alknyloxy refers to the group -O-alknyl, where alknyl is defined as above.
  • cycloalkyl to a cyclic saturated alkyl chain having from 3 to 12 carbon atoms, for example, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • substituted refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent (s) .
  • the substituent (s) is independently selected from the group consisting of -F, -Cl, -Br, -I, -OH, trifluromethoxy, ethoxy, propyloxy, iso-propyloxy, n-butyloxy, isobutyloxy, t-butyloxy, -SCH 3 , -SC 2 H 5 , formaldehyde group, -C (OCH 3 ) , cyano, nitro, CF 3 , -OCF 3 , amino, dimethylamino, methyl thio, sulfonyl and acetyl.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts. Accordingly, pharmaceutical compositions containing the compounds of the present invention as the active ingredient as well as methods of preparing the instant compounds are also part of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents and such solvates are also intended to be encompassed within the scope of this invention.
  • substituted alkyl group examples include, but not limited to, 2-aminoethyl, 2-hydroxyethyl, pentachloroethyl, trifluoromethyl, methoxymethyl, pentafluoroethyl and piperazinylmethyl.
  • substituted alkoxy groups include, but not limited to, aminomethoxy, thrifluoromethoxy, 2-diethylaminoethoxy, 2-ethoxycarbonylethoxy, 3-hydroxypropoxy.
  • the compounds of the present invention may also be present in the form of pharmaceutically acceptable salts.
  • the salts of the compounds of this invention refer to non-toxic “pharmaceutically acceptable salts” .
  • the pharmaceutically acceptable salt forms include pharmaceutically acceptable acidic/anionic or basic/cationic salts.
  • the pharmaceutically acceptable acidic/anionic salt generally takes a form in which the basic nitrogen is protonated with an inorganic or organic acid.
  • organic or inorganic acids include hydrochloric, hydrobromic, hydriodic, perchloric, sulfuric, nitric, phosphoric, acetic, propionic, glycolic, lactic, succinic, maleic, fumaric, malic, tartaric, citric, benzoic, mandelic, methanesulfonic, hydroxyethanesulfonic, benzenesulfonic, oxalic, pamoic, 2-naphthalenesulfonic, p-toluenesulfonic, cyclohexanesulfamic, salicylic, saccharinic or trifluoroacetic.
  • Pharmaceutically acceptable basic/cationic salts include, and are not limited to aluminum, calcium, chloroprocaine, choline, diethanolamine, ethylenediamine, lithium, magnesium, potassium, sodium and zinc.
  • the present invention includes within its scope the prodrugs of the compounds of this invention.
  • such prodrugs will be functional derivatives of the compounds that are readily converted in vivo into the required compound.
  • the term “administering” shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the subject.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs” , ed. H. Bundgaard, Elsevier, 1985.
  • the present invention includes compounds described herein can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers.
  • the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof.
  • the above Formula I are shown without a definitive stereochemistry at certain positions.
  • the present invention includes all stereoisomers of Formula I and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
  • the present invention includes any possible tautomers and pharmaceutically acceptable salts thereof, and mixtures thereof, except where specifically stated otherwise.
  • the present invention includes all isomerides of Formula III and Formula IV and pharmaceutically acceptable salts thereof. Further, mixtures of isomerides as well as isolated specific isomerides are also included. During the course of the synthetic procedures known to those skilled in the art used to prepare the present invention, two isomerides may be obtained by ring-closure reactions. For example, the carboxyl of Compound 7-4 reacts with one of two amino groups of Compound 163-3, then it may result two isomerides which are Compound 163 and its isomeride, and their mixture also may be available. Wherein, isomeride may be stereoisomer, or tautomer.
  • the present invention includes any possible solvates and polymorphic forms.
  • a type of a solvent that forms the solvate is not particularly limited so long as the solvent is pharmacologically acceptable.
  • water, ethanol, propanol, acetone or the like can be used.
  • salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids.
  • the compound of the present invention is acidic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases.
  • Salts derived from such inorganic bases include aluminum, ammonium, calcium, copper (ic and ous) , ferric, ferrous, lithium, magnesium, manganese (ic and ous) , potassium, sodium, zinc and the like salts. Particularly preferred are the ammonium, calcium, magnesium, potassium and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, as well as cyclic amines and substituted amines such as naturally occurring and synthesized substituted amines.
  • Other pharmaceutically acceptable organic non-toxic bases from which salts can be formed include ion exchange resins such as, for example, arginine, betaine, caffeine, choline, N', N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine
  • the compound of the present invention When the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, formic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like.
  • citric, hydrobromic, formic, hydrochloric, maleic, phosphoric, sulfuric and tartaric acids particularly preferred are formic and hydrochloric acid.
  • the compounds of Formula I are intended for pharmaceutical use they are preferably provided in substantially pure form, for example at least 60%pure, more suitably at least 75%pure, especially at least 98%pure (%are on a weight for weight basis) .
  • compositions of the present invention comprise a compound represented by Formula I (or a pharmaceutically acceptable salt thereof) as an active ingredient, a pharmaceutically acceptable carrier and optionally other therapeutic ingredients or adjuvants.
  • the compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered.
  • the pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • the compounds represented by Formula I, or a prodrug, or a metabolite, or pharmaceutically acceptable salts thereof, of this invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous) .
  • the pharmaceutical compositions of the present invention can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient.
  • compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil-in-water emulsion, or as a water-in-oil liquid emulsion.
  • the compound represented by Formula I, or a pharmaceutically acceptable salt thereof may also be administered by controlled release means and/or delivery devices.
  • the compositions may be prepared by any of the methods of pharmacy. In general, such methods include a step of bringing into association the active ingredient with the carrier that constitutes one or more necessary ingredients.
  • the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.
  • compositions of this invention may include a pharmaceutically acceptable carrier and a compound, or a pharmaceutically acceptable salt, of Formula I.
  • the compounds of Formula I, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.
  • the pharmaceutical carrier employed can be, for example, a solid, liquid, or gas.
  • solid carriers include such as lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid.
  • liquid carriers include such as sugar syrup, peanut oil, olive oil, and water.
  • gaseous carriers include such as carbon dioxide and nitrogen.
  • oral liquid preparations such as suspensions, elixirs and solutions
  • carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used to form oral solid preparations such as powders, capsules and tablets.
  • oral solid preparations such as powders, capsules and tablets.
  • tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed.
  • tablets may be coated by standard aqueous or nonaqueous techniques.
  • a tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
  • Each tablet preferably contains from about 0.05mg to about 5g of the active ingredient and each cachet or capsule preferably containing from about 0.05mg to about 5g of the active ingredient.
  • a formulation intended for the oral administration to humans may contain from about 0.5mg to about 5g of active agent, compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition.
  • Unit dosage forms will generally contain between from about l mg to about 2g of the active ingredient, typically 25mg, 50mg, l00mg, 200mg, 300mg, 400mg, 500mg, 600mg, 800mg, or l000mg.
  • compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of the active compounds in water.
  • a suitable surfactant can be included such as, for example, hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
  • compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions.
  • the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions.
  • the final injectable form must be sterile and must be effectively fluid for easy syringability.
  • the pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol) , vegetable oils, and suitable mixtures thereof.
  • compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations may be prepared, utilizing a compound represented by Formula I of this invention, or a pharmaceutically acceptable salt thereof, via conventional processing methods. As an example, a cream or ointment is prepared by admixing hydrophilic material and water, together with about 5wt%to about 10wt%of the compound, to produce a cream or ointment having a desired consistency.
  • compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by first admixing the composition with the softened or melted carrier (s) followed by chilling and shaping in molds.
  • the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.
  • additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.
  • additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.
  • additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.
  • other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient.
  • dosage levels on the order of from about 0.01mg/kg to about 150mg/kg of body weight per day are useful in the treatment of the above-indicated conditions, or alternatively about 0.5mg to about 7g per patient per day.
  • colon cancer, rectal cancer, mantle cell lymphoma, multiple myeloma, breast cancer, prostate cancer, glioblastoma, squamous cell esophageal cancer, liposarcoma, T-cell lymphoma melanoma, pancreatic cancer, or lung cancer may be effectively treated by the administration of from about 0.01 to 50mg of the compound per kilogram of body weight per day, or alternatively about 0.5mg to about 3.5g per patient per day.
  • DIBAL-H Diisobutylaluminium hydride
  • DIEA N, N-Diisopropylethylamine
  • DMSO Dimethyl sulfoxide
  • EA Ethyl acetate
  • EDTA Ethylenediaminetetraacetic acid
  • HATU Hexafluorophosphate Azabenzotriazole Tetramethyl Uronium
  • HEPES 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid
  • LCMS Liquid chromatography–mass spectrometry
  • PE Petroleum ether
  • NCS N-Chlorosuccinimide; rt or R. T: room temperature;
  • TFA Trifluoroacetic acid
  • THF Tetrahydrofuran
  • Step 1 Preparation of ethyl (R) -5- (2- (2-chloro-5-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
  • Step 2 Preparation of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
  • Step 3 Preparation of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamide
  • reaction mixture was poured into water (3 L) and extracted with EA (1 L*5) , combined the organic layers and washed with brine (1 L*3) , dried over Na 2 SO 4 . Concentrated under reduced pressure to afford desired product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamide (105 g, 96%) as a yellow solid
  • Step 4 Preparation of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbothioamide
  • Step 5 Preparation of methyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate
  • Step 6 Preparation of (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-2-ol (Compound 7)
  • Step 2 Preparation of (S) - (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) (phenyl) methanol
  • Step 1 Preparation of tert-butyl 2- (1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbonyl) hydrazine-1-carboxylate
  • Step 2 Preparation of 1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbohydrazide hydrochloride
  • Step 3 Preparation of (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzo [c] [1, 2] oxaborol-1 (3H) -ol (Compound 45)
  • Example 45 Prepare the following examples (shown in Table 1) essentially as described for Example 45 using the corresponding starting materials.
  • Example 1 shown in Table 1
  • Example 45 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • Step 1 Preparation of (R) -methyl 2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazole-5-carboxylate
  • Step 2 Preparation of (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) methanol
  • Example 94 Prepare the following examples (shown in Table 2) essentially as described for Example 94 using the corresponding starting materials.
  • Step 2 Preparation of tert-butyl (4, 5-bis (2-methoxyethoxy) -2-nitrophenyl) carbamate
  • Step 4 Preparation of 4, 5-bis (2-methoxyethoxy) benzene-1, 2-diamine
  • Step 5 Preparation of (R) -3- (5, 6-bis (2-methoxyethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine (Compound 101)
  • Example 3 Prepare the following examples (shown in Table 3) essentially as described for Example 101 using the corresponding starting materials.
  • Example 62 shown in Table 3 essentially as described for Example 101 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • the present invention also includes its isomeride, such as tautomer, and also includes their mixture.
  • Step 3 synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile and/or isomeride thereof
  • Step 2 Synthesis of tert-butyl (4-cyano-5-hydroxy-2-nitrophenyl) carbamate
  • Step 3 Synthesis of tert-butyl (4-cyano-5- (difluoromethoxy) -2-nitrophenyl) carba-mate
  • Step 4 tert-butyl (2-amino-4-cyano-5- (difluoromethoxy) phenyl) carbamate
  • Step 5 4, 5-diamino-2- (difluoromethoxy) benzonitrile
  • Step 6 Synthesis of (R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
  • Step 1 synthesis of 4-amino-2- (4-methylpiperazin-1-yl) -5-nitrobenzonitrile
  • Step 2 synthesis of 4, 5-diamino-2- (4-methylpiperazin-1-yl) benzonitrile
  • Step 3 synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (4-methylpiperazin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
  • Example 163 Prepare the following examples (shown in Table 4) essentially as described for Example 163 using the corresponding starting materials.
  • Example 164 shown in Table 4 essentially as described for Example 163 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • the present invention also includes its isomeride and also includes their mixture.
  • Example 171 (shown in Table 5) essentially as described for Example 170 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • the present invention also includes its isomeride and also includes their mixture.
  • Step 1 synthesis of (R) -5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) -2-nitrobenzenethiol
  • Step 2 synthesis of (R) -2-amino-5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzenethiol
  • Step 3 synthesis of 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzo [d] thiazole
  • Example 75 (shown in Table 6) essentially as described for Example 63 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • Step 3 Preparation of 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-6-ol
  • Example 187 (shown in Table 7) essentially as described for Example 132 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • Step 1 Preparation of (R) -methyl 2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazole-5-carboxylate
  • Step 2 Preparation of (R) -tert-butyl 5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-1-carboxylate
  • Step 3 Preparation of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carbonitrile (Compound 133)
  • Example 133 Prepares the following examples (shown in Table 8) essentially as described for Example 133 using the corresponding starting materials. For example, prepare the following Example 183 (shown in Table 8) essentially as described for Example 133 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • Step 1 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbonitrile
  • Step 2 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -N-hydroxypyrazolo [1, 5-a] pyrimidine-3-carboximidamide
  • Step 3 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboximidamide
  • Step 4 synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3, 4, 6, 7-tetrahydropyrano [3, 4-d] imidazole
  • Step 1 synthesis of tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-hydroxypiperidine-1-carboxylate
  • Step 2 synthesis of tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-oxopiperidine-1-carboxylate
  • Step 3 synthesis of tert-butyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydrothiazolo [4, 5-c] pyridine-5 (4H) -carboxylate
  • Step 4 synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [4, 5-c] pyridine hydrochloride
  • Step 1 synthesis of ethyl (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
  • Step 2 synthesis of (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
  • Step 3 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidin-2-amine
  • Example 194 (shown in Table 10) essentially as described for Example 193 using instead of and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
  • the present invention also includes its isomeride and also includes their mixture.
  • Step 1 Synthesis of (R) -2- (5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
  • Step 1 synthesis of ethyl (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
  • Step 2 synthesis of (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
  • Step 3 synthesis of (S) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
  • Step 1 synthesis of (R) -ethyl 5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
  • Step 2 synthesis of (R) -5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
  • Step 3 synthesis of (R) -2- (5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
  • Step 1 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbohydrazide
  • Step 2 synthesis of (R) -2-chloro-1- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) ethanone
  • Step 3 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6, 7-dimethoxyimidazo [1, 2-a] pyridin-2-yl) pyrazolo [1, 5-a] pyrimidine
  • Step 1 synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbohydrazide
  • Step 2 synthesis of (R) -3- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dihydro-8H- [1, 2, 4] triazolo [3, 4-c] [1, 4] oxazine
  • Mobility shift assay was used to determine the inhibitory activity of compounds against TrkA kinase. Assay procedures are as follows:
  • Stop buffer 100 mM HEPES, pH 7.5; 0.015%Brij-35; 0.2%Coating Reagent #3; 50 mM EDTA
  • IC 50 shown as Table 11, Compounds of the present disclosure, as exemplified in the Examples, showed IC 50 values in the following ranges: “*” stands for “IC 50 ⁇ 10nM” ; “**” stands for “10nM ⁇ IC 50 ⁇ 50nM” ; “***” stands for “IC 50 >50nM.
  • test compound (20 mM stock solution) was diluted to 60 ⁇ M with 100%DMSO as starting concentration then 3-fold serial diluted with "9+0" concentrations. in a 96-well dilution plate (Thermo, Cat. No. 249944) ;
  • X The log of the concentration of the compound
  • Y Luminescence value
  • test compounds solutions or control compound solution (verapamil) at the final concentration of 2 ⁇ M and carried out at 37°C.
  • the slope value, k was determined by linear regression of the natural logarithm of the remaining percentage of the parent drug vs. incubation time curve.
  • in vitro half-life (in vitro t 1/2 ) was determined from the slope value:
  • control compound (verapamil) was included in the assay. Any value of the compounds that was not within the specified limits will be rejected and the experiment will be repeated.
  • the plasma protein binding was measured as the following procedure.
  • a basic solution was prepared by dissolving 14.2 g/L Na 2 HPO 4 and 8.77 g/L NaCl in deionized water and the solution could be stored at 4°C for up to 7 days.
  • An acidic solution was prepared by dissolving 12.0 g/L NaH 2 PO 4 and 8.77 g/L NaCl in deionized water and the solution could be stored at 4°C for up to 7 days.
  • the basic solution was titrated with the acidic solution to pH 7.4 and store at 4°C for up to 7 days. pH was checked on the day of experiment and was adjusted if outside specification of 7.4 ⁇ 0.1.
  • the plasma was centrifuged at 3,220 g for 10 minutes to remove clots and supernatant was collected into a fresh tube. The pH of the plasma was checked and recorded.
  • test compounds and control compound ketoconazole were prepared in DMSO at the concentration of 200 ⁇ M. And then 3 ⁇ L of working solution was removed to mix with 597 ⁇ L of human, rat or mouse plasma to achieve a final concentration of 1 ⁇ M (0.5%DMSO) . Plasma samples were vortexed thoroughly.
  • the dialysis membranes were soaked in ultrapure water for 60 minutes to separate strips, then in 20%ethanol for 20 minutes, finally in dialysis buffer for 20 minutes.
  • the dialysis apparatus was assembled according to the manufacturer’s instruction. Each cell was filled with 120 ⁇ L of plasma sample and dialyzed against equal volume of dialysis buffer (PBS) . The assay was performed in duplicate. The dialysis plate was sealed and incubated in an incubator at 37°C with 5%CO 2 at 100 rpm for 6 hours. At the end of incubation, seal was removed and 50 ⁇ L of samples from both buffer and plasma chambers were transferred to wells of a 96-well plate.
  • PBS dialysis buffer
  • %Recovery (Peak Area Ratio buffer chamber + Peak Area Ratio plasma chamber ) /Peak Area Ratio total sample *100
  • Peak Area Ratio buffer chamber means the conc for free fraction
  • Peak Area Ratio plasma chamber means the conc for both free and bound fraction
  • Peak Area Ratio total sample means the conc for starting sample before incubation
  • Plasma protein binding results of control compound and test Compounds in different species are shown in Table 16.
  • the degree of binding to plasma proteins significantly influences the pharmacokinetic and pharmacodynamics properties of a drug.
  • the Comparative compound A reflected a high degree of plasma protein binding, therefore the efficacy of the drug might be reduced.
  • the exemplified compounds of the present invention have lower degree of plasma protein binding compared with the Comparative compound A. It indicated the present invention had superior pharmacokinetic and pharmacodynamics properties in human.
  • the cytochrome P450 was measured as the following procedure:
  • a master solution containing phosphate buffer, ultra-pure H 2 O, MgCl 2 solution and human liver microsomes was made according to Table 17, and then 1 ⁇ L of 2 mM of compound solution or 1 ⁇ L of DMSO (as without inhibitor control) was added to the above master solution. The final concentration of test compounds or control compounds was 10 ⁇ M.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention relates to compounds of Formula (I), methods of using the compounds as Trk inhibitors, and pharmaceutical compositions comprising such compounds. The compounds are useful in treating, preventing or ameliorating diseases or disorders such as cancer or infections. (I)

Description

TYROSINE KINASE INHIBITORS, COMPOSITIONS AND METHODS THERE OF FIELD OF THE INVENTION
The present application is concerned with pharmaceutically active compounds. The disclosure provides compounds as well as their compositions and methods of use. The compounds inhibit tropomyosin-related kinases (Trks) and are useful in the treatment of various diseases including infectious diseases and cancer.
BACKGROUND OF THE INVENTION
Tropomyosin-related kinases (Trks) are a group of receptor tyrosine kinases which are regulated by neurotrophins, including 3 members TrkA, TrkB and TrkC, encoded by the genes NTRK1, NTRK2 and NTRK3 respectively. Many cellular functions, for example, cell proliferation, cell differentiation, metabolism and apoptosis are mediated by Trks through phosphorylation and regulation of their downstream signal pathway members. Gene fusions involving NTRK genes result in continuous activation or overexpression of these kinases, which increase the risk of tumor genesis.
Trk plays an important physiological role in the development of nerves, including the growth and function maintenance of neuronal axons, the development of memory and the protection of neurons from injury, etc. Also, it is showed that Trk uncommonly expresses in normal tissues or cancer, while fusion drives abnormally high expression and activation of Trk kinase domain. Trk fusions are found in diverse cancer histologies with low fusion frequency, such as thyroid cancer, lung cancer, colon cancer, and melanoma. It is estimated that 1,500-5,000 patients harbor Trk fusion-positive cancers in the United States annually.
In recent years, Trk fusion protein is becoming a valid cancer target, the small molecule inhibitor for Trk with most rapid development is Loxo Oncology's larotrectinib which is highly potent against Trk in clinical development. Earlier application, WO2010048314, WO2011006074, WO2016097869, and WO2018077246 disclosed a series of Trk inhibitors. Accordingly, there is still a great demand for Trk inhibitors which have more potent activity, and better liver microsomes metabolic stability. Additionally, in view of the importance of Trk’s physiological functions, there is great demand for Trk inhibitors which can inhibit not only Trk A, B, and C but also mutated forms of Trk A, B and C (for example the G595R, G667C, A608D, F589L, G623R) which are reported in patients receiving first generation Trk kinase inhibitors. In this invention, applicant discovered potent small molecules that can have activity as Trk inhibitors, and thus may be useful for therapeutic administration to fight against cancer and/or  infectious diseases. These small molecules are expected to be useful as pharmaceuticals with desirable stability, solubility, bioavailability, therapeutic index and toxicity values that are crucial to become efficient medicines to promote human health.
SUMMARY OF INVENTION
The present invention relates to compounds that are used as Trk inhibitors. Trk inhibitors are useful in the treatment of cancers and infectious diseases.
The compounds of the invention have the general structures as Formula I. A compound of Formula I, or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
Figure PCTCN2019123719-appb-000001
wherein,
ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
ring B is 5-membered aromatic heterocycle;
X and Z are each independently selected from C, N, O, or S;
Y is C or N;
R 1 is absent, H, or -C 1-8 alkyl;
R 2 is H, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, or -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring, wherein the -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 0-4 alkyl-C 3-10 carbocyclic ring, -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring is optionally substituted with -C 1-8 alkyl, -C 2-8 alkynyl, -C 1-8 haloalkyl, -C 1-8 alkyl-OH, halogen, OH, CN, NH 2, -C 0-4 alkyl-COOR 10, -C 6-10 aryl ring, -O-C 6-10 aryl ring, substituted or unsubstituted -C 3-10 carbocyclic ring, or substituted or unsubstituted -C 3-10 heterocyclic ring;
R 3 is absent, C 3-10 heterocyclic ring; or
R 2 and R 3 together with the atoms to which they are attached to form a 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring, wherein the 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring is optionally substituted with  halogen, OH, CN, NH 2, -CONHOH, -CONH 2, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-O (CO) OR 10, -C 1-8 alkoxy, -C 1-8 haloalkoxy, -C 1-8 alkoxy-C 1-8 alkoxy, -C 1-8 alkylthio, -C 1-8 haloalkylthio, -C 1-8 alkyl, -C 1-8 haloalkyl, -C 0-4 alkyl-OH, -O-CH 2-CN, -C 0-4 alkyl-O-C 3-10 heterocyclic ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring, or the 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring forms a ring structure with other substituted or unsubstituted carbocyclic ring, substituted or unsubstituted heterocyclic ring, substituted or unsubstituted aryl ring, or substituted or unsubstituted heteroaryl ring;
R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
R 10 is H, or -C 1-8alkyl;
wherein the heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, O or B.
In some embodiments of Formula I, ring A is
Figure PCTCN2019123719-appb-000002
In some embodiments of Formula I, X is independently selected from O, S or N.
In some embodiments of Formula I, Y is C.
In some embodiments of Formula I, Z is N.
In some embodiments of Formula I, R 4 is
Figure PCTCN2019123719-appb-000003
In some embodiments of Formula I, the compound is of Formula II or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
Figure PCTCN2019123719-appb-000004
wherein,
ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
R 1 is H, or -C 1-8alkyl;
R 2 is H, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, or -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring, wherein the -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring is optionally substituted with -C 1-8alkyl, -C 2-8 alkynyl, -C 1-8 haloalkyl, -C 1-8 alkyl-OH, halogen, OH, CN, NH 2, -C 0-4 alkyl-COOR 10, -C 6-10 aryl ring, -O-C 6-10 aryl ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring;
R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
R 10 is H, or -C 1-8 alkyl;
wherein the heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, O or B.
In some embodiments of Formula II, ring A is
Figure PCTCN2019123719-appb-000005
In some embodiments of Formula II, R 1 is independently selected from H or CH 3.
In some embodiments of Formula II, R 4 is
Figure PCTCN2019123719-appb-000006
In some embodiments of Formula II, R 2 is independently selected from
Figure PCTCN2019123719-appb-000007
Figure PCTCN2019123719-appb-000008
Figure PCTCN2019123719-appb-000009
In some embodiments of Formula II, R 2 is
Figure PCTCN2019123719-appb-000010
In some embodiments of Formula I, the compound is of Formula III or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof:
Figure PCTCN2019123719-appb-000011
wherein,
ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
ring C is a 5-to 6-membered carbocyclic ring, or heterocyclic ring, aryl ring, or heteroaryl ring;
X and Z are each independently selected from C, N, O, or S;
Y is C or N;
R 1 is absent, H, or -C 1-8alkyl;
R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
R 5 and R 6 are each independently selected from H, OH, NH 2, CN, -COOH, -CONHOH, -CONH 2, halogen, -C 1-8alkyl, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-O (CO) OR 10, -C 1-8 alkoxy, -C 1-8  haloalkoxy, -C 1-8 alkoxy-C 1-8 alkoxy, -C 1-8 alkylthio, -C 1-8 haloalkylthio, -C 1-8 alkyl, -C 1-8 haloalkyl, -C 0-4 alkyl-OH, -O-CH 2-CN, -C 0-4 alkyl-O-C 3-10 heterocyclic ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring;
or R 5 and R 6 together with the atoms to which they are attached to form a 5 to 12-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring, wherein the 5 to 12-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring is optionally substituted with halogen;
R 10 is H, or -C 1-8alkyl;
wherein the heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, or O.
In some embodiments of Formula III, ring A is
Figure PCTCN2019123719-appb-000012
In some embodiments of Formula III, ring C is 6-membered aromatic ring.
In some embodiments of Formula III, ring C is phenyl, pyridyl, pyridazinyl, or pyrimidinyl.
In some embodiments of Formula III, ring C is phenyl.
In some embodiments of Formula III, X is selected from O, S, or N.
In some embodiments of Formula III, X is N.
In some embodiments of Formula III, Y is C.
In some embodiments of Formula III, Z is N.
In some embodiments of Formula III, R 1 is absent, H, or CH 3.
In some embodiments of Formula III, R 4 is
Figure PCTCN2019123719-appb-000013
In some embodiments of Formula III, R 5 and R 6 are each independently selected from H, OH , NH 2, F, Cl, Br -CN, -CF 3, -OCF 3, CH 3, -O-CH 3, -S-CH 3, -CH 2OH , -COOH , 
Figure PCTCN2019123719-appb-000014
Figure PCTCN2019123719-appb-000015
In some embodiments of Formula III, R 5 and R 6 are both -O-CH 3.
In some embodiments of Formula III, R 5 and R 6 together with the atoms to which they are attached from
Figure PCTCN2019123719-appb-000016
In some embodiments of Formula I, the compound is of Formula IV or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
Figure PCTCN2019123719-appb-000017
wherein,
Ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, -C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
R’ is H, NH 2, or -C 1-4 alkyl;
Ring B’ is a 5-membered aromatic heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
Ring C’ is a phenyl, 6-membered heterocyclic ring, or 6-membered heteroaryl ring;
X’ and Z’ are each independently selected from C, N, O, or S;
Y’ is C or N;
R” is -C (O) -C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2-C 1-4 alkyl, -NR 7 (CH 2mNR 8R 9, - (CH 2mC 4-10 heterocyclyl; or NH 2, -C (O) OH, -C (O) NH 2, -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alkyl, -C (O) O-C 1-4 alkyl, -OC (O) O-C 1-4 alkyl, -S-C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2-C 1-4 alkyl, -OC 4-6heterocyclyl, -NR 7 (CH 2mNR 8R 9, - (CH 2mC 4-10 heterocyclyl optionally substituted with one or more substituents independently selected from OH, CN, NH 2, -C (O) OH, halogen, -C 1-4 alkyl or -C 1-4 alkoxyl; or
any two R” together with the atoms to which they are attached form a 5-to 12-membered ring;
R 7, R 8 and R 9 are each independently selected from H, or -C 1-4 alkyl;
m and n are each independently selected from 0, 1, 2, 3 or 4.
In some embodiments of Formula IV, Ring A is
Figure PCTCN2019123719-appb-000018
In some embodiments of Formula IV, R’ is selected from H.
In some embodiments of Formula IV, R 4 is
Figure PCTCN2019123719-appb-000019
In some embodiments of Formula IV, Ring B’ is selected from imidazole, oxazole, thiazole, triazole or pyrrole.
In some embodiments of Formula IV, Ring B’ is selected from
Figure PCTCN2019123719-appb-000020
Figure PCTCN2019123719-appb-000021
In some embodiments of Formula IV, Ring C’ is selected from phenyl, pyridine, pyrazine, pyrimidine, pyridazine, piperidine, or tetrahydropyran.
In some embodiments of Formula IV, Ring C’ is selected from
Figure PCTCN2019123719-appb-000022
Figure PCTCN2019123719-appb-000023
In some embodiments of Formula IV, R” is selected from
Figure PCTCN2019123719-appb-000024
Figure PCTCN2019123719-appb-000025
In some embodiments of Formula IV, two R” with the atoms to which they are attached  form
Figure PCTCN2019123719-appb-000026
Figure PCTCN2019123719-appb-000027
In some embodiments of Formula I, wherein the compound is of Formula V or an isomeride, pharmaceutically acceptable salt thereof,
Figure PCTCN2019123719-appb-000028
wherein,
Ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, -C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
R 11 is H, NH 2, or -C 1-4 alkyl;
Ring B” is a 5-membered aromatic heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
Ring C” is a phenyl, 6-membered heterocyclic ring, or 6-membered heteroaryl ring;
X” and Z” are each independently selected from C, N, O, or S;
Y” is C or N;
R 12 is selected from H, OH, CN, NH 2, -C (O) OH, -C (O) NH 2, halogen, -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alkyl, -C (O) O-C 1-4 alkyl, -OC (O) O-C 1-4 alkyl, -S-C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2-C 1-4 alkyl, -OC 4-6heterocyclyl, -NR 7’ (CH 2mNR 8’R 9’, - (CH 2mC 4-10heterocyclyl; wherein NH 2, -C (O) OH, -C (O) NH 2, halogen, -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alkyl, -C (O) O-C 1-4 alkyl, -OC (O) O-C 1-4 alkyl, -S-C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2-C 1-4 alkyl, -OC 4-6heterocyclyl,  -NR 7’ (CH 2mNR 8’R 9’, - (CH 2mC 4-10heterocyclyl optionally substituted with one or more substituents independently selected from OH, CN, NH 2, -C (O) OH, halogen, -C 1-4 alkyl or -C 1-4 alkoxyl; or
any two R 12 together with the atoms to which they are attached form a 5-to 12-membered ring;
R 7’, R 8’ and R 9’ are each independently selected from H, or -C 1-4 alkyl;
m and n are each independently selected from 0, 1, 2, 3 or 4.
In some embodiments of Formula V, wherein Ring A is
Figure PCTCN2019123719-appb-000029
In some embodiments of Formula V, wherein R 11 is selected from H, NH 2 or CH 3.
In some embodiments of Formula V, wherein R 4 is
Figure PCTCN2019123719-appb-000030
In some embodiments of Formula V, wherein Ring B” is selected from imidazole, oxazole, thiazole, triazole or pyrrole.
In some embodiments of Formula V, wherein Ring B” is selected from
Figure PCTCN2019123719-appb-000031
Figure PCTCN2019123719-appb-000032
In some embodiments of Formula V, wherein Ring C” is selected from phenyl, pyridine, pyrazine, pyrimidine, pyridazine, piperidine or tetrahydropyran.
In some embodiments of Formula V, wherein Ring C” is selected from
Figure PCTCN2019123719-appb-000033
Figure PCTCN2019123719-appb-000034
In some embodiments of Formula V, wherein R 12 is selected from H, -OH, F, Cl, Br, -CH 3, -NH 2, -COOH, -CN, 
Figure PCTCN2019123719-appb-000035
Figure PCTCN2019123719-appb-000036
In some embodiments of Formula V, wherein two R 12 with the atoms to which they are attached form
Figure PCTCN2019123719-appb-000037
Figure PCTCN2019123719-appb-000038
The present invention further provides some preferred technical solutions with regard to compound of Formula I, Formula II, Formula III, Formula IV, or Formula V, or an isomeride, pharmaceutically acceptable salt or solvate thereof, wherein the compound is:
1) (R) -4- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) phenyl) morpholine;
2) (R) -1- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) piperidin-4-ol;
3) 5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (tetrahydrofuran-3-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
4) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethan-1-ol;
5) (1S, 4s) -4- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclohexan-1-ol;
6) (R) -4- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) morpholine;
7) (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-2-ol;
8) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (pyridin-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
9) (R) -4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) phenol;
10) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (pyrazin-2-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
11) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethan-1-amine;
12) methyl ( (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethyl) carbamate;
13) (R) -3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzonitrile;
14) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (6- (trifluoromethyl) pyridin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
15) 3- (5- (azetidin-2-yl) -4H-1, 2, 4-triazol-3-yl) -5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
16) ethyl (R) -5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazole-3-carboxylate;
17) (R) -5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazole-3-carboxylic acid;
18) (3S) -3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclohexan-1-ol;
19) (3S) -3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclopentan-1-ol;
20) tert-butyl 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) azetidine-1-carboxylate;
21) (R) -1- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) piperidin-4-ol;
22) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (piperidin-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
23) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclobutan-1-ol;
24) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclobutan-1-amine;
25) (S) -2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluoropropan-2-ol;
26) (R) -2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluoropropan-2-ol;
27) (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1, 3, 3, 3-hexafluoropropan-2-ol;
28) 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluorobutan-2-ol;
29) 3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluoro-2-methylpropan-2-ol;
30) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2-methylpropan-2-ol;
31) (R) -3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclobutan-1-ol;
32) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (tetrahydro-2H-pyran-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
33) (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2-methylpropan-1-ol;
34) (R) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethan-1-ol;
35) 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) piperidin-4-ol;
36) (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 2, 3, 4-tetrahydroisoquinoline;
37) (1R, 3r) -3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) adamantan-1-ol;
38) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (1-methylpiperidin-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
39) (R) -2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-1-ol;
40) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (4- (piperazin-1-yl) phenyl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
41) (R) -3- (5- (4, 4-difluorocyclohexyl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
42) (R) - (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) (phenyl) methanol;
43) (R) - (3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) bicyclo [1.1.1] pentan-1-yl) methanol;
44) (R) -3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) bicyclo [1.1.1] pentan-1-amine;
45) (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzo [c] [1, 2] oxaborol-1 (3H) -ol;
46) 1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1-difluorobutan-2-ol;
47) 1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2, 2, 2-trifluoroethan-1-ol;
48) 1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) prop-2-yn-1-ol;
49) 3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) morpholine;
50) (R) -3- (5- (1H-indol-5-yl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
51) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethyl L-leucinate hydrochloride;
52) 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2-fluoroethan-1-ol;
53) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclopropan-1-ol;
54) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (6- (4-methylpiperazin-1-yl) pyridin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
55) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethyl L-valylvalinate hydrochloride;
56) (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) quinoline;
57) (R) -3- (5- (1H-benzo [d] imidazol-6-yl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
58) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (4-phenoxyphenyl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
59) (R) -3- (5- (1H-indazol-6-yl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
60) (1R, 2S, 3R, 5S) -5- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclohexane-1, 2, 3, 5-tetraol;
61) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (2, 3-dihydrobenzofuran-6-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
62) 5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
63) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzo [d] thiazole;
64) (R) -4- (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-c] pyridin-6-yl) morpholine;
65) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-c] pyridine;
66) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazol-6-yl) ethan-1-ol;
67) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazol-6-yl) methanol;
68) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) benzo [d] oxazol-6-yl) ethan-1-ol;
69) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) benzo [d] oxazol-6-yl) methanol;
70) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3H-imidazo [4, 5-c] pyridin-6-yl) ethan-1-ol;
71) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (trifluoromethoxy) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
72) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethyl) -3H-imidazo [4, 5-c] pyridine;
73) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) oxazolo [4, 5-c] pyridine;
74) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6-fluoro-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
75) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) thiazolo [4, 5-c] pyridine;
76) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-d] pyridazine;
77) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6-methoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
78) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
79) (R) -6- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -2, 2-difluoro-5H- [1, 3] dioxolo [4', 5': 4, 5] benzo [1, 2-d] imidazole;
80) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethoxy) benzo [d] oxazole;
81) (R) -3- (6- (difluoromethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
82) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7, 9, 10, 12, 13-hexahydro-1H- [1, 4, 7, 10] tetraoxacyclododecino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
83) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1-methyl-6, 7-dihydro-1H- [1, 4] dioxino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
84) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-3H-imidazo [4, 5-b] pyridine;
85) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-imidazo [4, 5-c] pyridine;
86) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methyl-1H-imidazo [4, 5-c] pyridine;
87) (R) -8- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7H-purin-6-amine;
88) (R) -8- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7H-purin-6-ol;
89) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -N-hydroxy-5-methoxy-1H-benzo [d] imidazole-6-carboxamide;
90) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carboxylic acid;
91) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carboxamide;
92) (R) -3- (5-chloro-6- (trifluoromethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
93) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) ethan-1-ol;
94) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) methanol;
95) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3H-imidazo [4, 5-c] pyridin-6-yl) methanol;
96) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydro-1H- [1, 4] dioxino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
97) (R) -3- (7-chloro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
98) (R) -3- (7-chloro-5-fluoro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
99) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7-methyl-1H-imidazo [4, 5-c] pyridine;
100) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-methoxybenzo [d] oxazole;
101) (R) -3- (5, 6-bis (2-methoxyethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
102) (R) -6, 7-dichloro-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] pyridine;
103) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-methyl-3H-imidazo [4, 5-c] pyridine;
104) (R) -3- (4, 7-dichloro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
105) (R) -3- (5, 6-dichloro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
106) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methyl-3H-imidazo [4, 5-b] pyridine;
107) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carbonitrile;
108) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-fluoro-3H-imidazo [4, 5-b] pyridine;
109) (R) -3- (5, 6-bis (difluoromethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
110) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethyl) -1H-imidazo [4, 5-b] pyridine;
111) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 7-difluorobenzo [d] oxazole;
112) (R) -3- (5-chloro-6-methoxy-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
113) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (7- (trifluoromethoxy) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
114) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (trifluoromethyl) -3H-imidazo [4, 5-b] pyridine;
115) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-diyl dimethyl bis (carbonate) ;
116) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- ( (trifluoromethyl) thio) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
117) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-diol;
118) 5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- ( ( (R) -tetrahydrofuran-3-yl) oxy) -6- ( ( (S) -tetrahydrofuran-3-yl) oxy) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
119) (R) -2, 2'- ( (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-diyl) bis (oxy) ) diacetonitrile;
120) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dimethoxybenzo [d] oxazole;
121) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] quinoxaline;
122) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7-methyl-3H-imidazo [4, 5-b] pyridine;
123) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-fluoro-1H-benzo [d] imidazole-6-carbonitrile;
124) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1-methyl-1H-imidazo [4, 5-c] pyridine;
125) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile;
126) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylthio) -1H-benzo [d] imidazole-6-carbonitrile;
127) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (7-fluoro-6-methoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
128) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] pyrazine;
129) (R) -6-bromo-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] pyrazine;
130) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] phenazine;
131) (R) -6- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) - [1 ,3] dioxolo [4', 5': 4, 5] benzo [1, 2-d] oxazole;
132) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-6-ol;
133) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carbonitrile;
134) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7, 8-dihydro-1H, 6H- [1, 4] dioxepino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
135) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3H-imidazo [4, 5-c] pyridin-6-yl) methanol;
136) (R) -3- (5, 6-difluoro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
137) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5-difluoro-1H-benzo [d] imidazole-6-carboxylate;
138) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5-difluoro-1H-benzo [d] imidazole-6-carboxylic acid;
139) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5-fluoro-6- (trifluoromethyl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
140) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-ethoxy-1H-benzo [d] imidazole-5-carbonitrile;
141) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluoro-1H-benzo [d] imidazole-5-carboxylic acid;
142) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (methylamino) -1H-benzo [d] imidazole-5-carbonitrile;
143) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-morpholino-1H-benzo [d] imidazole-5-carbonitrile;
144) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (dimethylamino) -1H-benzo [d] imidazole-5-carbonitrile;
145) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (3-hydroxyazetidin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile;
146) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydroimidazo [4', 5': 4, 5] benzo [1, 2-e] [1, 4] diazepin-9 (3H) -one;
147) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7, 8-dihydro-3H-imidazo [4', 5': 4, 5] benzo [1, 2-f] [1, 4] oxazepin-9 (6H) -one;
148) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-dicarbonitrile;
149) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-hydroxy-1H-benzo [d] imidazole-5-carbonitrile;
150) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (2-hydroxyethoxy) -1H-benzo [d] imidazole-5-carbonitrile;
151) (R) -6-bromo-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile;
152) methyl (R) -5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carboxylate;
153) (R) -5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carboxylic acid;
154) (R) -5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carboxamide;
155) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carboxylate;
156) (R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile;
157) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (trifluoromethyl) -1H-benzo [d] imidazole-6-carbonitrile;
158) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluoro-1H-benzo [d] imidazole-7-carboxylate;
159) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methyl-1H-benzo [d] imidazole-5-carbonitrile;
160) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-N-methyl-1H-benzo [d] imidazole-5-carboxamide;
161) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-N, N-dimethyl-1H-benzo [d] imidazole-5-carboxamide;
162) (R) -4- ( (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazol-5-yl) methyl) morpholine;
163) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (4-methylpiperazin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile;
164) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (S) -3-hydroxypyrrolidin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile;
165) 6- ( (S) -2-cyanopyrrolidin-1-yl) -2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile;
166) methyl (5-cyano-2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazol-6-yl) -L-prolinate;
167) (5-cyano-2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazol-6-yl) -L-proline;
168) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (2- (dimethylamino) ethyl) (methyl) amino) -1H-benzo [d] imidazole-5-carbonitrile;
169) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (2-methoxyethoxy) -1H-benzo [d] imidazole-5-carbonitrile;
170) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- (methylsulfonyl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
171) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylsulfinyl) -1H-benzo [d] imidazole-6-carbonitrile;
172) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylsulfonyl) -1H-benzo [d] imidazole-6-carbonitrile;
173) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylsulfonyl) -1H-benzo [d] imidazole-6-carboxamide;
174) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxybenzo [d] oxazole-5-carbonitrile;
175) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-fluorobenzo [d] oxazole-7-carboxylate;
176) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethoxy) benzo [d] oxazole-5-carbonitrile;
177) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-hydroxybenzo [d] oxazole-5-carbonitrile;
178) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxybenzo [d] oxazole-6-carboxylate;
179) (R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methylbenzo [d] oxazole;
180) ( (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxybenzo [d] oxazol-5-yl) methyl) -L-proline;
181) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 8-dimethoxy- [1, 2, 4] triazolo [1, 5-c] pyrimidine;
182) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dimethoxy- [1, 2, 4] triazolo [1, 5-a] pyridine;
183) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6-fluoro-1H-indol-2-yl) pyrazolo [1, 5-a] pyrimidine;
184) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carboxylate;
185) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carboxylic acid;
186) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indol-6-ol;
187) (S) -2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-7-ol;
188) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3, 4, 6, 7-tetrahydropyrano [3, 4-d] imidazole;
189) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [4, 5-c] pyridine;
190) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [5, 4-c] pyridine;
191) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydrothiazolo [5, 4-c] pyridine-5 (4H) -carboxamide;
192) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydro-4H-pyrano [4, 3-d] thiazole;
193) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidin-2-amine;
194) (R) -2- (2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile;
195) (R) -2- (5- (2- (2-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
196) (R) -2- (5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
197) (S) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
198) (R) -2- (5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
199) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6, 7-dimethoxyimidazo [1, 2-a] pyridin-2-yl) pyrazolo [1, 5-a] pyrimidine; or
200) (R) -3- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dihydro-8H- [1, 2, 4] triazolo [3, 4-c] [1, 4] oxazine.
The present invention also provides a pharmaceutical composition comprising a compound of any one of the present invention, or a pharmaceutically acceptable salt or a stereoisomer thereof, and at least one pharmaceutically acceptable carrier or excipient.
The present invention additionally provided a method of inhibiting Trk, including wildtype TrkA, TrkB and TrkC, the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, and the TrkC G623R, said method comprising administering to a patient a compound of any one of the present invention or a pharmaceutically acceptable salt or an isomeride thereof.
The present invention further provides a method of treating a disease associated with inhibition of Trk, including wildtype TrkA, TrkB and TrkC, the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, and the TrkC G623R, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound in any one in the present invention, or a pharmaceutically acceptable salt or an isomeride thereof. Wherein the disease is mammary analogue secretory carcinoma (MASC) of the salivary glands, infantile fibrosarcoma, spitz tumors, colon cancer, gastric cancer, thyroid cancer (such as papillary thyroid cancer) , lung cancer, leukemia, pancreatic cancer, melanoma (sunch as multiple melanoma) , brain cancer (such pontine glioma) , renal cancer (such as congenital mesoblastic nephroma) , prostate cancer, ovarian cancer or breast cancer (such as secretory breast carcinoma) .
The present invention provided a method of inhibiting Trk in a patient, said method comprising administering to the patient in need thereof a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or an isomeride thereof.
The present invention also provides a use of the present compound or its pharmaceutical composition for the preparation of a medicament.
In some embodiments, wherein the medicament is used for the treatment or prevention of cancer.
In some embodiments, wherein the disease is mammary analogue secretory carcinoma (MASC) of the salivary glands, infantile fibrosarcoma, spitz tumors, colon cancer, gastric cancer, thyroid cancer (such as papillary thyroid cancer) , lung cancer, leukemia, pancreatic cancer, melanoma (such as multiple melanoma) , brain cancer (such pontine glioma) , renal cancer (such as congenital mesoblastic nephroma) , prostate cancer, ovarian cancer or breast cancer (such as secretory breast carcinoma) .
In some embodiments, wherein the medicament is used as an inhibitor of Trk.
In some embodiments, wherein the Trk is wildtype TrkA, TrkB, TrkC, or the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, or the TrkC G623R.
The present invention also provides a method of enhancing, stimulating and/or increasing the immune response in a patient, said method comprising administering to the patient in need thereof a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or an isomeride thereof.
The general chemical terms used in the formula above have their usual meanings. For example, the term “halogen” , as used herein, unless otherwise indicated, means fluoro, chloro, bromo or iodo. The preferred halogen groups include F, Cl and Br.
As used herein, unless otherwise indicated, alkyl includes saturated monovalent hydrocarbon radicals having straight, branched or cyclic moieties. For example, alkyl radicals include methyl, ethyl, propyl, isopropyl, cyclcopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, cyclcobutyl, n-pentyl, 3- (2-methyl) butyl, 2-pentyl, 2-methylbutyl, neopentyl, cyclcopentyl, n-hexyl, 2-hexyl, 2-methylpentyl and cyclohexyl. Similary, C 1-8, as in C 1-8 alkyl is defined to identify the group as having 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms in a linear or branched arrangement.
Alkenyl and alkynyl groups include straight, branched chain or cyclic alkenes and alkynes. Likewise, “C 2-8 alkenyl” and “C 2-8 alkynyl” means an alkenyl or alkynyl radicals having 2, 3, 4, 5, 6, 7 or 8 carbon atoms in a linear or brached arrangement.
Alkoxy radicals are oxygen ethers formed from the previously described straight, branched chain or cyclic alkyl groups.
The term “aryl” , as used herein, unless otherwise indicated, refers to an unsubstituted or substituted mono-or polycyclic ring system containing carbon ring atoms. The preferred aryls are mono cyclic or bicyclic 6-10 membered aromatic ring systems. Phenyl and naphthyl are preferred aryls. The most preferred aryl is phenyl.
The term “heterocyclyl” , as used herein, unless otherwise indicated, represents an unsubstituted or substituted stable three to eight membered monocyclic saturated ring system which consists of carbon atoms and from one to three heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heterocyclyl group may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclyl groups include, but are not limited to azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxoazepinyl, azepinyl, tetrahydrofuranyl, dioxolanyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone and oxadiazolyl.
The term “heteroaryl” , as used herein, unless otherwise indicated, represents an unsubstituted or substituted stable five or six membered monocyclic aromatic ring system or an unsubstituted or substituted nine or ten membered benzo-fused heteroaromatic ring system or bicyclic heteroaromatic ring system which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heteroaryl group may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of heteroaryl groups include, but are not limited to thienyl, furanyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, pyridyl, pyridazinyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, benzofuranyl, benzothienyl, benzisoxazolyl, benzoxazolyl, benzopyrazolyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl adeninyl, quinolinyl or isoquinolinyl. The term “alkenyloxy” refers to the group -O-alkenyl, where alkenyl is defined as above.
The term “alknyloxy” refers to the group -O-alknyl, where alknyl is defined as above.
The term “cycloalkyl” to a cyclic saturated alkyl chain having from 3 to 12 carbon atoms, for example, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
The term “substituted” refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent (s) . Typical substituents include, but are not limited to, halogen (F, Cl, Br or I) , C 1-8 alkyl, C 3-12 cycloalkyl, -OR 1, SR 1, =O, =S, -C (O) R 1, -C (S) R 1, =NR 1, -C (O) OR 1, -C (S) OR 1, -NR 1R 2, -C (O) NR 1R 2, cyano, nitro, -S (O)  2R 1, -OS (O 2) OR 1, -OS (O)  2R 1, -OP (O) (OR 1) (OR 2) ; wherein R 1 and R 2 is independently selected from -H, lower alkyl, lower haloalkyl. In some embodiments, the substituent (s) is independently selected from the group consisting of -F, -Cl, -Br, -I, -OH, trifluromethoxy, ethoxy, propyloxy, iso-propyloxy, n-butyloxy, isobutyloxy, t-butyloxy, -SCH 3, -SC 2H 5, formaldehyde group,  -C (OCH 3) , cyano, nitro, CF 3, -OCF 3, amino, dimethylamino, methyl thio, sulfonyl and acetyl.
The term “composition” , as used herein, is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts. Accordingly, pharmaceutical compositions containing the compounds of the present invention as the active ingredient as well as methods of preparing the instant compounds are also part of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents and such solvates are also intended to be encompassed within the scope of this invention.
Examples of substituted alkyl group include, but not limited to, 2-aminoethyl, 2-hydroxyethyl, pentachloroethyl, trifluoromethyl, methoxymethyl, pentafluoroethyl and piperazinylmethyl.
Examples of substituted alkoxy groups include, but not limited to, aminomethoxy, thrifluoromethoxy, 2-diethylaminoethoxy, 2-ethoxycarbonylethoxy, 3-hydroxypropoxy.
The compounds of the present invention may also be present in the form of pharmaceutically acceptable salts. For use in medicine, the salts of the compounds of this invention refer to non-toxic “pharmaceutically acceptable salts” . The pharmaceutically acceptable salt forms include pharmaceutically acceptable acidic/anionic or basic/cationic salts. The pharmaceutically acceptable acidic/anionic salt generally takes a form in which the basic nitrogen is protonated with an inorganic or organic acid. Representative organic or inorganic acids include hydrochloric, hydrobromic, hydriodic, perchloric, sulfuric, nitric, phosphoric, acetic, propionic, glycolic, lactic, succinic, maleic, fumaric, malic, tartaric, citric, benzoic, mandelic, methanesulfonic, hydroxyethanesulfonic, benzenesulfonic, oxalic, pamoic, 2-naphthalenesulfonic, p-toluenesulfonic, cyclohexanesulfamic, salicylic, saccharinic or trifluoroacetic. Pharmaceutically acceptable basic/cationic salts include, and are not limited to aluminum, calcium, chloroprocaine, choline, diethanolamine, ethylenediamine, lithium, magnesium, potassium, sodium and zinc.
The present invention includes within its scope the prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds that are readily converted in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term “administering” shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after  administration to the subject. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs” , ed. H. Bundgaard, Elsevier, 1985.
It is intended that the definition of any substituent or variable at a particular location in a molecule be independent of its definitions elsewhere in that molecule. It is understood that substituents and substitution patterns on the compounds of this invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques know in the art as well as those methods set forth herein.
The present invention includes compounds described herein can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof.
The above Formula I are shown without a definitive stereochemistry at certain positions. The present invention includes all stereoisomers of Formula I and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
When a tautomer of the compound of Formula I exists, the present invention includes any possible tautomers and pharmaceutically acceptable salts thereof, and mixtures thereof, except where specifically stated otherwise.
The present invention includes all isomerides of Formula III and Formula IV and pharmaceutically acceptable salts thereof. Further, mixtures of isomerides as well as isolated specific isomerides are also included. During the course of the synthetic procedures known to those skilled in the art used to prepare the present invention, two isomerides may be obtained by ring-closure reactions. For example, the carboxyl of Compound 7-4 reacts with one of two amino groups of Compound 163-3, then it may result two isomerides which are Compound 163 and its isomeride, and their mixture also may be available. Wherein, isomeride may be stereoisomer, or tautomer.
When the compound of Formula I and pharmaceutically acceptable salts thereof exist in the form of solvates or polymorphic forms, the present invention includes any possible solvates and polymorphic forms. A type of a solvent that forms the solvate is not particularly limited so long  as the solvent is pharmacologically acceptable. For example, water, ethanol, propanol, acetone or the like can be used.
The term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids. When the compound of the present invention is acidic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases. Salts derived from such inorganic bases include aluminum, ammonium, calcium, copper (ic and ous) , ferric, ferrous, lithium, magnesium, manganese (ic and ous) , potassium, sodium, zinc and the like salts. Particularly preferred are the ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, as well as cyclic amines and substituted amines such as naturally occurring and synthesized substituted amines. Other pharmaceutically acceptable organic non-toxic bases from which salts can be formed include ion exchange resins such as, for example, arginine, betaine, caffeine, choline, N', N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
When the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, formic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like. Preferred are citric, hydrobromic, formic, hydrochloric, maleic, phosphoric, sulfuric and tartaric acids, particularly preferred are formic and hydrochloric acid. Since the compounds of Formula I are intended for pharmaceutical use they are preferably provided in substantially pure form, for example at least 60%pure, more suitably at least 75%pure, especially at least 98%pure (%are on a weight for weight basis) .
The pharmaceutical compositions of the present invention comprise a compound represented by Formula I (or a pharmaceutically acceptable salt thereof) as an active ingredient, a pharmaceutically acceptable carrier and optionally other therapeutic ingredients or adjuvants. The compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most  suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
In practice, the compounds represented by Formula I, or a prodrug, or a metabolite, or pharmaceutically acceptable salts thereof, of this invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous) . Thus, the pharmaceutical compositions of the present invention can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil-in-water emulsion, or as a water-in-oil liquid emulsion. In addition to the common dosage forms set out above, the compound represented by Formula I, or a pharmaceutically acceptable salt thereof, may also be administered by controlled release means and/or delivery devices. The compositions may be prepared by any of the methods of pharmacy. In general, such methods include a step of bringing into association the active ingredient with the carrier that constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.
Thus, the pharmaceutical compositions of this invention may include a pharmaceutically acceptable carrier and a compound, or a pharmaceutically acceptable salt, of Formula I. The compounds of Formula I, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.
The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas. Examples of solid carriers include such as lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers include such as sugar syrup, peanut oil, olive oil, and water. Examples of gaseous carriers include such as carbon dioxide and nitrogen. In preparing the compositions for oral dosage form, any convenient pharmaceutical media may be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like may be used to form oral liquid preparations such as  suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed. Optionally, tablets may be coated by standard aqueous or nonaqueous techniques.
A tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Each tablet preferably contains from about 0.05mg to about 5g of the active ingredient and each cachet or capsule preferably containing from about 0.05mg to about 5g of the active ingredient. For example, a formulation intended for the oral administration to humans may contain from about 0.5mg to about 5g of active agent, compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition. Unit dosage forms will generally contain between from about l mg to about 2g of the active ingredient, typically 25mg, 50mg, l00mg, 200mg, 300mg, 400mg, 500mg, 600mg, 800mg, or l000mg.
Pharmaceutical compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol) , vegetable oils, and suitable mixtures thereof.
Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations may be prepared, utilizing a compound represented by Formula I of this invention, or a pharmaceutically acceptable salt thereof, via conventional processing methods. As an example, a cream or ointment is prepared by admixing hydrophilic material and water, together with about 5wt%to about 10wt%of the compound, to produce a cream or ointment having a desired consistency.
Pharmaceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by first admixing the composition with the softened or melted carrier (s) followed by chilling and shaping in molds.
In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including antioxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing a compound described by Formula I, or pharmaceutically acceptable salts thereof, may also be prepared in powder or liquid concentrate form.
Generally, dosage levels on the order of from about 0.01mg/kg to about 150mg/kg of body weight per day are useful in the treatment of the above-indicated conditions, or alternatively about 0.5mg to about 7g per patient per day. For example, colon cancer, rectal cancer, mantle cell lymphoma, multiple myeloma, breast cancer, prostate cancer, glioblastoma, squamous cell esophageal cancer, liposarcoma, T-cell lymphoma melanoma, pancreatic cancer, or lung cancer, may be effectively treated by the administration of from about 0.01 to 50mg of the compound per kilogram of body weight per day, or alternatively about 0.5mg to about 3.5g per patient per day.
It is understood, however, that lower or higher doses than those recited above may be required. Specific dose level and treatment regimens for any particular subject will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, the severity and course of the particular disease undergoing therapy, the subject disposition to the disease, and the judgment of the treating physician.
These and other aspects will become apparent from the following written description of the invention.
The following Examples are provided to better illustrate the present invention. All parts and percentages are by weight and all temperatures are degrees Celsius, unless explicitly stated otherwise.
The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results. The compounds of the Examples have been found to inhibit Trk according to at least one assay described herein.
EXAMPLES
Experimental procedures for compounds of the invention are provided below.
The following abbreviations have been used in the examples:
AcOH: Acetic acid;
DCM: Dichloromethane;
DIBAL-H: Diisobutylaluminium hydride;
DIEA: N, N-Diisopropylethylamine;
DMF: Dimethylformamide;
DMAP: 4-Dimethylaminopyridine;
DMSO: Dimethyl sulfoxide;
EA: Ethyl acetate;
EDTA: Ethylenediaminetetraacetic acid;
HATU: Hexafluorophosphate Azabenzotriazole Tetramethyl Uronium;
HEPES: 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid;
LCMS: Liquid chromatography–mass spectrometry;
h or hrs: hour or hours;
PE: Petroleum ether;
MeOH: Methanol;
min: minute;
NCS: N-Chlorosuccinimide; rt or R. T: room temperature;
TFA: Trifluoroacetic acid;
THF: Tetrahydrofuran;
TLC: Preparative thin layer chromatography;
1N: 1mol. L -1, (2N: 2mol. L -1, etc. ) .
Example 7 Synthesis of Compound 7
(R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-2-ol
Step 1: Preparation of ethyl (R) -5- (2- (2-chloro-5-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
Figure PCTCN2019123719-appb-000039
To a solution of (R) -2- (2, 5-difluorophenyl) pyrrolidine hydrochloride (76 g) in 1-BuOH (1 L) was added ethyl 5-chloropyrazolo [1, 5-a] pyrimidine-3-carboxylate (78 g) and DIEA (89 g) . The mixture was heated to 120℃ for 14 h. Monitored by LCMS until the reaction was completed.
The mixture was concentrated under reduced pressure to remove 1-BuOH, the residue was poured into ice-water and extracted with EA (300 mL*3) , combined the organic layers, washed with brine and dried over Na 2SO 4. Concentrated in vacuo and the residue was washed with Hexane (500 mL) to afford the desired product ethyl (R) -5- (2- (2-chloro-5-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (122 g, 95%) as a white solid.
Step 2: Preparation of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
Figure PCTCN2019123719-appb-000040
To a solution of ethyl (R) -5- (2- (2-chloro-5-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (122 g) in EtOH (1 L) was added LiOH aqueous solution (1M, 1 L) . The reaction mixture was heated to 80℃ for 8 h. Monitored by LCMS until the reaction was completed.
The mixture was concentrated in vacuo to remove EtOH , the residue was added water (1 L) and acidified with HCl (1M) to pH=4~5, filtered and the solid was washed with water, dried in vacuo to afford desired product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (110 g, 98%) as a white solid.
Step 3: Preparation of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamide
Figure PCTCN2019123719-appb-000041
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (110 g) in DMF (1 L) was added HATU (146 g) , DIEA (82 g) and NH 4Cl (85 g) . the mixture was stirred at room temperature for 8 h. Monitored by LCMS until the reaction was completed.
The reaction mixture was poured into water (3 L) and extracted with EA (1 L*5) , combined the organic layers and washed with brine (1 L*3) , dried over Na 2SO 4. Concentrated under reduced pressure to afford desired product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamide (105 g, 96%) as a yellow solid
Step 4: Preparation of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbothioamide
Figure PCTCN2019123719-appb-000042
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamide (105 g) in dioxane (1 L) was added Lawesson’s Reagent (210 g) , the mixture was heated to 100℃ for 3 h. Monitored by LCMS until the reaction was completed.
The reaction mixture was cooled down to room temperature and filtered , the solid was washed with Dioxane, the filtrate was concentrated and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 95%: 5%) to afford desired product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbothioamide (85 g, 78%) as a yellow solid.
Step 5: Preparation of methyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate
Figure PCTCN2019123719-appb-000043
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbothioamide (78 g) in MeOH (800 mL) was added CH 3I (46 g) , the mixture was heated to 80℃ for 2 h. Monitored by LCMS until the reaction was completed.
The reaction mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 90%: 10%) to afford desired product methyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate (90 g, 83%) as a yellow solid
Step 6: Preparation of (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-2-ol (Compound 7)
Figure PCTCN2019123719-appb-000044
To a solution of methyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate hydroiodide (5 g) in pyridine (50 mL) was added 2-hydroxy-2-methylpropanehydrazide (2.37 g) , the mixture was heated to 110℃ overnight. Monitored by LCMS until the reaction was completed.
The reaction mixture was concentrated under reduced pressure to remove pyridine. The residue was purified by combi flash (DCM: MeOH = 100%: 0%to 90%: 10%) to yield 3.48g (61%yield) of the title compound. MS (ES +) : m/z=426.2 (M+H)  +
1H NMR (500 MHz, CD 3OD) δ 8.62-8.30 (m, 2H) , 7.19 (s, 1H) , 7.13-6.88 (m, 2H) , 6.65 and 6.11 (1H, s+s) , 5.70 and 5.35 (1H, s+s) , 4.27-3.71 (m, 2H) , 2.57 (s, 1H) , 2.31-1.95 (m, 3H) , 1.63 (s, 6H) .
Example 42 Synthesis of Compound 42
(S) - (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) (phenyl) methanol
Step 1: Preparation of (S) -2-hydroxy-2-phenylacetohydrazide
Figure PCTCN2019123719-appb-000045
To a solution of methyl (S) -2-hydroxy-2-phenylacetate (166 mg) in MeOH (10 mL) was added hydrazine hydrate (200 mg) , the mixture was heated to 80℃ and stirred overnight. Monitored by LCMS until the reaction was completed.
The reaction mixture was concentrated in vacuo to afford desired product (S) -2-hydroxy-2-phenylacetohydrazide (100 mg, 60%) as a yellow oil.
Step 2: Preparation of (S) - (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) (phenyl) methanol
Figure PCTCN2019123719-appb-000046
To a solution of (S) -2-hydroxy-2-phenylacetohydrazide (100 mg) in pyridine (10 mL) was added methyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate (100 mg) , the mixture was heated to 110℃ overnight. Monitored by LCMS until the reaction was completed.
The reaction mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 90%: 10%) to yield 60 mg (44.7%, yield) of the title compound. MS (ES +) : m/z=474.5 (M+H)  +
1H NMR (500 MHz, CD 3OD) δ 8.65-8.32 (m, 2H) , 7.38-7.18 (m, 6H) , 7.12-6.83 (m, 2H) , 6.63 and 6.11 (1H, s+s) , 5.86 (s, 1H) , 5.71 and 5.33 (1H, s+s) , 4.27-3.71 (m, 2H) , 2.57 (s, 1H) , 2.30-1.93 (m, 3H) .
Example 45 Synthesis of Compound 45
(R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzo [c] [1, 2] oxaborol-1 (3H) -ol
Step 1: Preparation of tert-butyl 2- (1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbonyl) hydrazine-1-carboxylate
Figure PCTCN2019123719-appb-000047
To a solution of 1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carboxylic acid (178 g) in DMF (10 mL) was added HATU (572 mg) , DIEA (259 mg) and tert-butyl hydrazinecarboxylate (158 mg) , the mixture was stirred at room temperature overnight. Monitored by LCMS until the reaction was completed.
The reaction mixture was poured into water (50 mL) and extracted with EA (30 mL*3) , combined the organic layers, washed with brine, dried over Na 2SO 4. Concentrated in vacuo, the residue was purified by combi flash (PE: EA = 100%: 0%to 50%: 50%) to afford desired product  tert-butyl 2- (1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbonyl) hydrazine-1-carboxylate (120 mg, 41%) as a white solid
Step 2: Preparation of 1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbohydrazide hydrochloride
Figure PCTCN2019123719-appb-000048
To tert-butyl 2- (1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbonyl) hydrazine-1-carboxylate (120 mg) was added HCl in dioxane (4M) , the mixture was stirred for 2 h. Monitored by LCMS until the reaction was completed.
The reaction mixture was concentrated in vacuo to afford desired product 1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbohydrazide hydrochloride (90 mg, 97%) as a yellow solid
Step 3: Preparation of (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzo [c] [1, 2] oxaborol-1 (3H) -ol (Compound 45)
Figure PCTCN2019123719-appb-000049
To a solution of 1-hydroxy-1, 3-dihydrobenzo [c] [1, 2] oxaborole-6-carbohydrazide hydrochloride (90 mg) in pyridine (10 mL) was added methyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate (100 mg) , the mixture was heated to 110℃ overnight. Monitored by LCMS until the reaction was completed.
The reaction mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0% to 90%: 10%) to yield 40 mg (2%, yield) of the title compound. MS (ES +) : m/z=500.3 (M+H)  +.
1H NMR (500 MHz, CD 3OD) δ 8.71-8.42 (m, 3H) , 7.70 (s, 1H) , 7.47 (d, J=8.1Hz, 1H) , 7.20 (s, 1H) , 7.12-6.85 (m, 2H) , 6.61and 6.10 (1H, s+s) , 5.71 and 5.37 (1H, s+s) , 5.12 (s, 2H) , 4.29-3.74 (m, 2H) , 2.56 (s, 1H) , 2.33-1.92 (m, 3H) .
Prepare the following examples (shown in Table 1) essentially as described for Example 45 using the corresponding starting materials. For example, prepare the following Example 1 (shown in Table 1) essentially as described for Example 45 using
Figure PCTCN2019123719-appb-000050
instead of
Figure PCTCN2019123719-appb-000051
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 1
Figure PCTCN2019123719-appb-000052
Figure PCTCN2019123719-appb-000053
Figure PCTCN2019123719-appb-000054
Figure PCTCN2019123719-appb-000055
Figure PCTCN2019123719-appb-000056
Figure PCTCN2019123719-appb-000057
Figure PCTCN2019123719-appb-000058
Figure PCTCN2019123719-appb-000059
Example 94 Synthesis of Compound 94
(R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) methanol
Figure PCTCN2019123719-appb-000060
Step 1: Preparation of (R) -methyl 2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazole-5-carboxylate
Figure PCTCN2019123719-appb-000061
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (365.8 mg) in POCl 3 (5 mL) was added methyl 5-amino-2-fluoro-4-hydroxybenzoate (203.6 mg) at 100℃ for 3h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was adjusted pH=8, the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 93%: 7%) to afford crude product (R) -methyl 2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazole-5-carboxylate (193.6 mg, 37%) as a yellow solid.
Step 2: Preparation of (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) methanol
Figure PCTCN2019123719-appb-000062
To a solution of (R) -methyl 2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazole-5-carboxylate (193.6 mg) in THF (3 mL) was added DIBAL-H (1 mL) at 0℃ for 1h. The reaction was detected by TLC and LCMS. The mixture was added saturated NH 4Cl solution (3 mL) and acetic ether. The mixure was extracted by acetic ether (3*15 mL) , the organic layer was dried by Na 2SO 4, then the mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 95%: 5%) to yield 56.3 mg (31%, yield) of the title compound. MS (ES +) : m/z=466.4 (M+H)  +
1H NMR (500 MHz, CD 3OD) δ 8.61-8.28 (m, 2H) , 7.74 (s, 1H) , 7.19 (s, 1H) , 7.16-6.90 (m, 3H) , 6.60 and 6.12 (1H, s+s) , 5.73 and 5.36 (1H, s+s) , 4.61 (s, 2H) , 4.30-3.68 (m, 2H) , 2.57 (s, 1H) , 2.31-1.95 (m, 3H) .
Prepare the following examples (shown in Table 2) essentially as described for Example 94 using the corresponding starting materials.
Table 2
Figure PCTCN2019123719-appb-000063
Figure PCTCN2019123719-appb-000064
Figure PCTCN2019123719-appb-000065
Example 101 Synthesis of Compound 101
(R) -3- (5, 6-bis (2-methoxyethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine
Step 1: Preparation of 4, 5-bis (2-methoxyethoxy) -2-nitrobenzoic acid
Figure PCTCN2019123719-appb-000066
To a solution of methyl 4, 5-bis (2-methoxyethoxy) -2-nitrobenzoate (986.5 mg) in MeOH (15 mL) at R. T was added H 2O (3 mL) and KOH (526.7 mg) for 6h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was adjusted pH=6, the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 93%: 7%) to afford crude product 4, 5-bis (2-methoxyethoxy) -2-nitrobenzoic acid (726.5 mg, 77%) as a yellow solid.
Step 2: Preparation of tert-butyl (4, 5-bis (2-methoxyethoxy) -2-nitrophenyl) carbamate
Figure PCTCN2019123719-appb-000067
To a solution of 4, 5-bis (2-methoxyethoxy) -2-nitrobenzoic acid (722.8 mg) in THF (15 mL) was added Et 3N (687.3 mg) and DPPA (628.1 mg) at R. T for 12h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was added t-BuOH (10 mL) at 80℃ for 6h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was purified by combi flash (PE: EA = 100%: 0%to 66%: 34%) to afford crude product 4, 5-bis (2-methoxyethoxy) -2-nitrophenyl) carbamate (586.2 mg, 73%) as a yellow solid.
Step 3: Preparation of 4, 5-bis (2-methoxyethoxy) -2-nitroaniline
Figure PCTCN2019123719-appb-000068
To a solution of 4, 5-bis (2-methoxyethoxy) -2-nitrophenyl) carbamate (580.2 mg) in dioxane (2 mL) was added HCl . dioxane (8 mL) , the reaction was stirred at R. T for 13h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was adjusted pH=8, and the crude product 4, 5-bis (2-methoxyethoxy) -2-nitroaniline (381.7 mg, 89%) was a yellow solid.
Step 4: Preparation of 4, 5-bis (2-methoxyethoxy) benzene-1, 2-diamine
Figure PCTCN2019123719-appb-000069
To a solution of 4, 5-bis (2-methoxyethoxy) -2-nitroaniline (380.2 mg) in MeOH (6 mL) was added Zn powder (418.7 mg) , NH 4Cl (406.2 mg) , H 2O (2 mL) , DCM (4 mL) at R. T for 6h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 93%: 7%) to afford crude product 4, 5-bis (2-methoxyethoxy) benzene-1, 2-diamine (257.6 mg, 76%) as a yellow solid.
Step 5: Preparation of (R) -3- (5, 6-bis (2-methoxyethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine (Compound 101)
Figure PCTCN2019123719-appb-000070
To a solution of 4, 5-bis (2-methoxyethoxy) benzene-1, 2-diamine (85.9 mg) in POCl 3 (3 mL) was added (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (112.6 mg) , the mixture was stirred at 100℃ for 6h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was adjusted pH=8, the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 93%: 7%) to yield 22.6 mg (12%, yield) of the title compound. MS (ES +) : m/z=565.6 (M+H)  + .
1H NMR (500 MHz, CD 3OD) δ 8.56-8.24 (m, 2H) , 7.15 (s, 1H) , 7.12-6.87 (m, 4H) , 6.63 and 6.12 (1H, s+s) , 5.62 and 5.27 (1H, s+s) , 4.27-3.71 (m, 6H) , 4.87-3.81 (m, 4H) , 3.30 (s, 6H) , 2.56 (s, 1H) , 2.30-1.94 (m, 3H) .
Prepare the following examples (shown in Table 3) essentially as described for Example 101 using the corresponding starting materials. For example, prepare the following Example 62 (shown in Table 3) essentially as described for Example 101 using
Figure PCTCN2019123719-appb-000071
instead of 
Figure PCTCN2019123719-appb-000072
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 3
Figure PCTCN2019123719-appb-000073
Figure PCTCN2019123719-appb-000074
Figure PCTCN2019123719-appb-000075
Figure PCTCN2019123719-appb-000076
Figure PCTCN2019123719-appb-000077
Figure PCTCN2019123719-appb-000078
Figure PCTCN2019123719-appb-000079
Figure PCTCN2019123719-appb-000080
Figure PCTCN2019123719-appb-000081
Figure PCTCN2019123719-appb-000082
Figure PCTCN2019123719-appb-000083
Figure PCTCN2019123719-appb-000084
*Remark: If there is an isomeride, such as a tautomer in the above compounds, the present invention also includes its isomeride, such as tautomer, and also includes their mixture.
Example 125 Synthesis of Compound 125 and/or its isomeride
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000085
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000086
Step 1: Preparation of 4-amino-2-methoxy-5-nitrobenzonitrile
Figure PCTCN2019123719-appb-000087
To a solution of CH 3ONa (14.6 g) in MeOH (300 mL) was added 4-amino-2-fluoro-5-nitrobenzonitrile (9.8 g) below 15 ℃. Then the solution was warmed to room temperature and stirred for 8h. LCMS showed the reaction was completed, concentrated under reduced pressure to remove MeOH, the residue was added 1L water and adjust pH to 4-5 with 2N HCl aqueous solution. Filtered and the solid was washed with water, dried under reduced pressure at 50 ℃ for 10 h to afford the product 4-amino-2-methoxy-5-nitrobenzonitrile (9.6 g) as a yellow solid.
Step 2: Preparation of 4, 5-diamino-2-methoxybenzonitrile
Figure PCTCN2019123719-appb-000088
To a solution of 4-amino-2-methoxy-5-nitrobenzonitrile (9.6 g) in DCM/MeOH (1: 1, 60 mL) was added saturated NH 4Cl (aq) (60 mL) . Zinc powder (32.5 g) was added to the mixture,  then the mixture was stirred at room temperature for 2h. LCMS showed the reaction was completed. The reaction mixture was filtered and the filtrate was extracted with DCM (3*100mL) , combined the organic layers, washed with brine, concentrated under reduced pressure and the residue was purified by combiflash (PE: EA=50%: 50%) to afford the product 4, 5-diamino-2-methoxybenzonitrile (7.3 g) as a red solid.
Step 3: synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile and/or isomeride thereof
Figure PCTCN2019123719-appb-000089
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (3.44 g) in POCl 3 (30 mL) was added 4, 5-diamino-2-methoxybenzonitrile (1.96 g) . The mixture was heater to 90 ℃ and stirred for 3h. LCMS showed the reaction was completed. Cooled to room temperature and concentrated under reduced pressure to remove POCl 3, the residue was poured into water (300 mL) and precipitated, the precipitation was filtered, then the solid was added to 1N NaOH aqueous solution and stirred overnight, then the solid was filtered and washed with water, and the filter cake dried under vacuum at 60 ℃ for 10h to afford final product (Compound 125 and/or the isomeride of Compound 125) as a yellow solid (3.98 g) . MS: [M+H]  +: 472.16.
1H NMR (500 MHz, DMSO-d6) δ 10.53-11.44 (m, 1H) , 8.63 and 8.78 (br+br, 1H) , 8.37 and 8.46 (s+s, 1H) , 7.56 and 7.87 and 7.90 (s+s+s, 1H) , 6.97 and 7.21-7.36 (m+m, 4H) , 6.09 and 6.63 (br+br, 1H) , 5.37 and 5.66 (br+br, 1H) , 3.72 and 4.25 (br+br, 1H) , 3.94-4.00 (m, 4H) , 2.57 (br, 1H) , 1.98-2.15 (m, 3H) .
Example 156 Synthesis of Compound 156 and/or its isomeride
(R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000090
(R) -5- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000091
Step 1: Synthesis of 4-amino-2-hydroxy-5-nitrobenzonitrile
Figure PCTCN2019123719-appb-000092
To a solution of NaOH (8.8 g) in water (100 mL) was added 4-amino-2-fluoro-5-nitrobenzonitrile (10 g) below 15 ℃, and the mixture was stirred for 8h at 80 ℃. The reaction was monitored by LC-MS. After the 4-amino-2-fluoro-5-nitrobenzonitrile was consumed completely, the reaction mixture was adjusted to PH 6-7 using 6 N HCl below 20 ℃. The mixture was filtered and the filter cake was washed by water, dried under reduced pressure at 50 ℃ for 10 h to afford 4-amino-2-hydroxy-5-nitrobenzonitrile (9.0 g) as a yellow solid.
Step 2: Synthesis of tert-butyl (4-cyano-5-hydroxy-2-nitrophenyl) carbamate
Figure PCTCN2019123719-appb-000093
To a solution of 4-amino-2-hydroxy-5-nitrobenzonitrile (500 mg) in THF (15 mL) was added Boc 2O (670 mg) and DMAP (34 mg) . The mixture was stirred for 4 h at room temperature,  and TLC showed 4-amino-2-hydroxy-5-nitrobenzonitrile was completely. The mixture was evaporated under in vacuo, and the residue was diluted by EA. The organic phase was washed with 0.5 N HCl, water, brine and dried over Na 2SO 4. The solvent was evaporated in vacuo, and the residue was purified by silica gel column chromatography (EA/PE: 0~20%in 30 min) to afford the desired product (646 mg) as a yellow solid.
Step 3: Synthesis of tert-butyl (4-cyano-5- (difluoromethoxy) -2-nitrophenyl) carba-mate
Figure PCTCN2019123719-appb-000094
To a mixture of tert-butyl (4-cyano-5-hydroxy-2-nitrophenyl) carbamate (100 mg) , ClCF 2COONa (109 mg) and Cs 2CO 3 (140 mg) was added DMF (3 mL) and water (0.3 mL) . The mixture was stirred for 2 h at 90 ℃. After TLC showed tert-butyl (4-cyano-5- (difluoromethoxy) -2-nitrophenyl) carbamate was consumed completely, the reaction mixture diluted by EA. The organic phase was washed with water, brine and dried over Na 2SO 4. The solvent was evaporated in vacuo, and the residue was purified by silica gel column chromatography (EA/PE: 0~20%in 30 min) to afford the desired product (77 mg) as a yellow solid.
Step 4: tert-butyl (2-amino-4-cyano-5- (difluoromethoxy) phenyl) carbamate
Figure PCTCN2019123719-appb-000095
To a mixture of tert-butyl (4-cyano-5- (difluoromethoxy) -2-nitrophenyl) carbamate (77 mg) , zinc powder (91 mg) and NH 4Cl (126 mg) was added EtOH (3 mL) and water (1 mL) . The mixture was stirred for 12 h at 80 ℃. After TLC and LC-MS showed tert-butyl (4-cyano-5- (difluoromethoxy) -2-nitrophenyl) carbamate was consumed completely, the reaction mixture filtered. The filtrated was concentrated in vacuo, and the residue was diluted by water. The aqueous phase was extracted with DCM, and the combined organic phases were washed with brine, dried over Na 2SO 4 and concentrated in vacuo to afford crude product, which was purified by silica gel column chromatography (MeOH/DCM: 0~5%in 30 min) to afford the desired product (56 mg) as a yellow solid.
Step 5: 4, 5-diamino-2- (difluoromethoxy) benzonitrile
Figure PCTCN2019123719-appb-000096
To tert-butyl (2-amino-4-cyano-5- (difluoromethoxy) phenyl) carbamate (56 mg) was added 4M HCl in dioxane (4 mL) . The mixture was stirred for 4 h at room temperature. After LC-MS showed 4, 5-diamino-2- (difluoromethoxy) benzonitrile was consumed completely, the reaction mixture was concentrated in vacuo, and the residue was diluted by aq. NaHCO 3. The aqueous phase was extracted with DCM, and the combined organic phases were washed with brine, dried over Na 2SO 4 and concentrated in vacuo to afford desired product (37 mg) , which was used directly in next step.
Step 6: Synthesis of (R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
Figure PCTCN2019123719-appb-000097
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (64 mg) in CH 3CN (2 mL) was added POCl 3 (54 μL, 0.561 mmol) and 4, 5-diamino-2- (difluoromethoxy) benzonitrile (37 mg) . The mixture was stirred for 3h at 90 ℃. After LC-MS showed 4, 5-diamino-2- (difluoromethoxy) benzonitrile was consumed completely, the reaction mixture was concentrated in vacuo, and the residue was diluted by EA. The organic phase was washed with water, brine and dried over Na 2SO 4. The solvent was evaporated in vacuo, and the residue was purified by silica gel column chromatography (MeOH/DCM: 0~8%in 30 min) to afford the desired product (Compound 156 and/or the isomeride of Compound 156) as yellow solid (18 mg) . MS: [M+H]  + 508.18.
Example 163 Synthesis of Compound 163 and/or its isomeride
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (4-methylpiperazin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000098
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (4-methylpiperazin-1-yl) -1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000099
Step 1: synthesis of 4-amino-2- (4-methylpiperazin-1-yl) -5-nitrobenzonitrile
Figure PCTCN2019123719-appb-000100
To a solution of 4-amino-2-fluoro-5-nitrobenzonitrile (18.1g) in THF (300 mL) was added 1-methylpiperazine (12.1 g) and DIEA (25.8 g) below 15 ℃. Then the solution was warmed to room temperature and stirred for 3h. LCMS showed the reaction was completed. The reaction mixture was poured into ice-water and extracted with EA (3*100 mL) , combined the organic layers and washer with brine , dried over Na 2SO 4. Concentreted to afford the desired product 4-amino-2- (4-methylpiperazin-1-yl) -5-nitrobenzonitrile (21.5 g) as a brown solid.
Step 2: synthesis of 4, 5-diamino-2- (4-methylpiperazin-1-yl) benzonitrile
Figure PCTCN2019123719-appb-000101
To a solution of 4-amino-2- (4-methylpiperazin-1-yl) -5-nitrobenzonitrile (13.1 g) in DCM/MeOH (1: 1, 60 mL) was added NH 4Cl/H 2O (60 mL) . The mixture was stirred, Zn (32.8 g) was added, then the solution was stirred at room temperature for 2h. LCMS showed the reaction was completed. The reaction mixture was filtered and the filtrate was extracted with DCM (3*100mL) , combined the organic layers, washed with brine , Concentrated under reduced pressure and the residue was purified by combiflash (PE: EA=50%: 50%) to afford the desired product 4, 5-diamino-2- (4-methylpiperazin-1-yl) benzonitrile (8.7 g) as a brown solid.
Step 3: synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (4-methylpiperazin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
Figure PCTCN2019123719-appb-000102
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (3.44 g) in POCl 3 (30 mL) was added 4, 5-diamino-2- (4-methylpiperazin-1-yl) benzonitrile (2.77 g) . The mixture was heater to 90 ℃ and stirred for 3h. LCMS showed the reaction was completed. Cooled to room temperature and concentrated under reduced pressure to remove POCl 3, the residue was poured into water (300 mL) and filtered, the solid was washed with NaHCO 3 saturated solution and water, dried under reduce pressure at 60 ℃ for 10h to afford the desired product (Compound 163 and/or the isomeride of Compound 163) (3.58 g) as a yellow solid. MS : [M+H]  + 540.81
Prepare the following examples (shown in Table 4) essentially as described for Example 163 using the corresponding starting materials. For example, prepare the following Example 164  (shown in Table 4) essentially as described for Example 163 using
Figure PCTCN2019123719-appb-000103
instead of 
Figure PCTCN2019123719-appb-000104
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 4
Figure PCTCN2019123719-appb-000105
Figure PCTCN2019123719-appb-000106
*Remark: If there is an isomeride in the above compounds, the present invention also includes its isomeride and also includes their mixture.
Example 170 Synthesis of Compound 170 and/or its isomeride
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- (methylsulfonyl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine
Figure PCTCN2019123719-appb-000107
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (methylsulfonyl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine
Figure PCTCN2019123719-appb-000108
Figure PCTCN2019123719-appb-000109
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (344 mg) in POCl 3 (5 mL) was added 4- (methylsulfonyl) benzene-1, 2-diamine (223 mg) . The mixture was heater to 90 ℃ and stirred for 3h. LCMS showed the reaction was completed. Cooled to room temperature and concentrated under reduced pressure to remove POCl 3, the residue was poured into water (300 mL) and filtered, the solid was washed with NaHCO 3 saturated solution and water, dried under reduce pressure at 60 ℃ for 10 h to afford the desired product (Compound 170 and/or the isomeride of Compound 170) as a yellow solid (397 mg) . LC-MS : [M+H]  + 495.66.
Prepare the following examples (shown in Table 5) essentially as described for Example 170 using the corresponding starting materials. For example, prepare the following Example 171 (shown in Table 5) essentially as described for Example 170 using
Figure PCTCN2019123719-appb-000110
instead of 
Figure PCTCN2019123719-appb-000111
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 5
Figure PCTCN2019123719-appb-000112
Figure PCTCN2019123719-appb-000113
*Remark: If there is an isomeride in the above compounds, the present invention also includes its isomeride and also includes their mixture.
Example 63 Synthesis of Compound 63
2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzo [d] thiazole
Figure PCTCN2019123719-appb-000114
Step 1: synthesis of (R) -5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) -2-nitrobenzenethiol
Figure PCTCN2019123719-appb-000115
To a solution of 5-fluoro-2-nitrobenzenethiol (1.73g) in THF (300 mL) was added (R) -octahydropyrrolo [1, 2-a] pyrazine (1.51 g) and DIEA (2.58 g) below 15 ℃. Then the solution was  warmed to room temperature and stirred for 3h. LCMS showed the reaction was completed. The reaction mixture was poured into ice-water and extracted with EA (3*100 mL) , combined the organic layers and washer with brine , dried over Na 2SO 4. Concentreted to afford the desired product (R) -5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) -2-nitrobenzenethiol (2.13 g) as a brown solid.
Step 2: synthesis of (R) -2-amino-5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzenethiol
Figure PCTCN2019123719-appb-000116
To a solution of (R) -5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) -2-nitrobenzenethiol (2.13 g) in DCM/MeOH (1: 1, 30 mL) was added NH 4Cl/H 2O (30 mL) . The mixture was stirred, Zn (4.9 g) was added, then the solution was stirred at room temperature for 2h. LCMS showed the reaction was completed. The reaction mixture was filtered and the filtrate was extracted with DCM (3*100mL) , combined the organic layers, washed with brine , Concentrated under reduced pressure and the residue was purified by combiflash (PE: EA=50%: 50%) to afford the desired product (R) -2-amino-5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzenethiol (1.37 g) as a brown solid.
Step 3: synthesis of 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzo [d] thiazole
Figure PCTCN2019123719-appb-000117
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (3.44 g) was added to toluene, then with SOCl 2 (2.38 g) was added, stirred at 80℃ for 2 h, excess SOCl 2 was distilled off, the residue was dissolve in toluene (30 mL) and then (R) -2-amino-5- (hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzenethiol (2.49 g) was added at 0 ℃, followed by stirring at room temperature for 1 h.
LCMS showed the reaction was completed. The mixture was diluted with EtOAc (10 mL) and sat. aq NaHCO 3 (5 mL) . The organic layer was separated and the aqueous layer extracted with EA (3 *5 mL) . The combined EtOAc extracts were washed with H 2O (3 *5 mL) , dried over  Na 2SO 4 and concentrated under reduced pressure, the residue was purified by combiflash (DCM: MeOH=95%: 5%) to afford the desired product 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzo [d] thiazole (2.43 g) as a yellow solid. MS : [M+H]  + 558.81.
Prepare the following Example 75 (shown in Table 6) essentially as described for Example 63 using
Figure PCTCN2019123719-appb-000118
instead of
Figure PCTCN2019123719-appb-000119
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 6
Figure PCTCN2019123719-appb-000120
Example 132 Synthesis of Compound 132
2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-6-ol
Figure PCTCN2019123719-appb-000121
Step 1: Preparation of (5-oxotetrahydrofuran-3-yl) methyl methanesulfonate
Figure PCTCN2019123719-appb-000122
To a solution of 4- (hydroxymethyl) dihydrofuran-2 (3H) -one (236.5 mg) in DCM (5 mL) was added Et 3N (658.7 mg) and MsCl (392.6 mg) at 0℃ for 1h. The reaction was detected by TLC and LCMS. The mixture was added saturated NH 4Cl solution (3 mL) and DCM. The mixure was extracted by DCM (3*15 mL) , the organic layer was dried by Na 2SO 4, then the mixture was concentrated in vacuo and the residue was purified by combi flash  (DCM: MeOH = 100%: 0%to 95%: 5%) to afford product (5-oxotetrahydrofuran-3-yl) methyl methanesulfonate (228.7 mg, 58%) as a yellow liquid.
Step 2: Preparation of 1-amino-5-hydroxypiperidin-2-one
Figure PCTCN2019123719-appb-000123
To a solution of (5-oxotetrahydrofuran-3-yl) methyl methanesulfonate (226.7 mg) in EtOH (5 mL) was added N 2H 4 . H 2O (52.8 mg) at 80℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 95%: 5%) to afford product 1-amino-5-hydroxypiperidin-2-one (56.3 mg, 90%) as a yellow solid.
Step 3: Preparation of 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-6-ol
Figure PCTCN2019123719-appb-000124
To a solution of 1-amino-5-hydroxypiperidin-2-one (56.3 mg) in pyridine was added (R) -methyl 5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbimidothioate (148.7 mg) at 110℃ for 12h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 95%: 5%) to yield 37.5mg (20%, yield) of the title compound. MS (ES +) : m/z=438.5 (M+H)  +
1H NMR (500 MHz, CD 3OD) δ 8.52-8.20 (m, 2H) , 7.16 (s, 1H) , 7.11-6.86 (m, 2H) , 6.62 and 6.08 (1H, s+s) , 5.65 and 5.30 (1H, s+s) , 4.25-3.69 (m, 4H) , 3.16 (s, 1H) , 2.67-2.62 (m, 2H) , 2.28-1.92 (m, 4H) , 1.63-1.59 (m, 2H) .
Prepare the following Example 187 (shown in Table 7) essentially as described for Example 132 using
Figure PCTCN2019123719-appb-000125
instead of
Figure PCTCN2019123719-appb-000126
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 7
Figure PCTCN2019123719-appb-000127
Figure PCTCN2019123719-appb-000128
Example 133 Synthesis of Compound 133
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carbonitrile
Figure PCTCN2019123719-appb-000129
Step 1: Preparation of (R) -methyl 2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazole-5-carboxylate
Figure PCTCN2019123719-appb-000130
To a solution of (R) -2- (2, 5-difluorophenyl) pyrrolidine hydrochloride (2.2 g) in n-BuOH (30 mL) was added DIEA (4.82 g) and 3-bromo-5-chloropyrazolo [1, 5-a] pyrimidine (2.61 g) at 100℃ for 3h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the mixure was extracted by acetic ether (3×100 mL) , the organic layer was dried by Na 2SO 4, then the mixture was concentrated in vacuo and the residue to afford product ( (R) -3-bromo-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine (3.5 g, 92%) as a yellow solid.
Step 2: Preparation of (R) -tert-butyl 5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-1-carboxylate
Figure PCTCN2019123719-appb-000131
To a solution of ( (R) -3-bromo-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine (162.8 mg) in dioxane (5 mL) was added Cs 2CO 3 (418.3 mg) , H 2O (1 mL) , Pd (dppf) Cl 2 (62.5 mg) and (1- (tert-butoxycarbonyl) -5-cyano-1H-indol-2-yl) boronic acid (192.7 mg) at 80℃ for 6h with N 2. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the mixure was extracted by acetic ether (3×50 mL) , the organic layer was dried by Na 2SO 4, then the mixture was concentrated in vacuo and the residue was purified by combi flash (PE: EA = 100%: 0%to 50%: 50%) to afford crude product ( (R) -tert-butyl 5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-1-carboxylate (106.3 mg, 46 %) as a yellow solid.
Step 3: Preparation of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carbonitrile (Compound 133)
Figure PCTCN2019123719-appb-000132
To a solution of ( (R) -tert-butyl 5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-1-carboxylate (102.8 mg) in DCM (2 mL) was added TFA (2 mL) at R.T for 12h. The reaction was detected by TLC and LCMS. Then the mixture was concentrated in vacuo and the residue was adjusted pH=8, the residue was purified by combi flash (DCM: MeOH = 100%: 0%to 93%: 7%) to yield 36.9 mg (45%, yield) of the title compound. MS (ES +) : m/z=441.5 (M+H)  + .
1H NMR (500 MHz, CD 3OD) δ 8.60-8.78 (m, 2H) , 7.52-7.77 (m, 3H) , 7.18 (s, 1H) , 7.10-6.85 (m, 3H) , 6.61 and 6.07 (1H, s+s) , 5.66 and 5.31 (1H, s+s) , 4.22-3.66 (m, 2H) , 2.53 (s, 1H) , 2.29-1.93 (m, 3H) .
Prepare the following examples (shown in Table 8) essentially as described for Example 133 using the corresponding starting materials. For example, prepare the following Example 183 (shown in Table 8) essentially as described for Example 133 using
Figure PCTCN2019123719-appb-000133
instead of 
Figure PCTCN2019123719-appb-000134
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 8
Figure PCTCN2019123719-appb-000135
Example 188 Synthesis of Compound 188
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3, 4, 6, 7-tetrahydropyrano [3, 4-d] imidazole
Figure PCTCN2019123719-appb-000136
Step 1: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbonitrile
Figure PCTCN2019123719-appb-000137
To a solution of ethyl 5-chloropyrazolo [1, 5-a] pyrimidine-3-carbonitrile (17.8 g) in EtOH (400 mL) was added (R) -2- (2, 5-difluorophenyl) pyrrolidine hydrochloride (26.2 g) and DIEA (25.8 g) . The mixture was heated to 90℃ for 2 h.
TLC showed the reaction was completed, Concentrated under reduced pressure to remove EtOH and the residue was poured into cooled water and filtered, the solid was washed with 1N HCl and water, dried under reduce pressure at 60 ℃ for 10h to afford the desired product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbonitrile (29.9 g, 92%) as a white solid.
Step 2: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -N-hydroxypyrazolo [1, 5-a] pyrimidine-3-carboximidamide
Figure PCTCN2019123719-appb-000138
A mixture of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbonitrile (16.2 g) , NH 2OH·HCl (4.17 g) , and DIEA (19.3 g) in THF (200 mL) was stirred overnight at 70 ℃. After cooling to room temperature, the mixture was concentrated under reduced pressure. The residue was taken up in water and the pH was adjusted to 2-3 with HCl  (aqueous, 1 M) . After washing the mixture with 3*40 mL EA, the pH of the aqueous layer was adjusted to 8-9 with NaOH (aqueous, 2 M) followed by extraction with 3*30 mL EA. The combined organic layers were dried over Na 2SO 4 and concentrated under reduced pressure to afford product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -N-hydroxypyrazolo [1, 5-a] pyrimidine-3-carboximidamide (5.1 g) as a white solid.
Step 3: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboximidamide
Figure PCTCN2019123719-appb-000139
A round-bottom flask, containing a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -N-hydroxypyrazolo [1, 5-a] pyrimidine-3-carboximidamide (5.0 g) in methanol (150 mL) was purged with nitrogen gas. To the solution was added Pd/C (1 g, 10%, 60%water) and the flask was then further purged with nitrogen gas. The atmosphere was then changed to hydrogen and the mixture was stirred overnight at 25 ℃ under a balloon. After purging the system with nitrogen, the solids were removed by filtration and the filtrate was concentrated under reduced pressure to afford desired product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboximidamide (2.9 g) as a brown solid.
Step 4: synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3, 4, 6, 7-tetrahydropyrano [3, 4-d] imidazole
Figure PCTCN2019123719-appb-000140
A mixture of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboximidamide (685 mg) , 3-bromodihydro-2H-pyran-4 (3H) -one (358 mg) , and potassium carbonate (552 mg) in CH3CN (15 mL) was stirred overnight at 80 ℃ under nitrogen. After cooling to ambient temperature, the reaction mixture was concentrated under reduced pressure. The residue was dissolved in EA (50 mL) and washed with 2*10 mL of H 2O. The organic phase was dried over (Na 2SO 4) and concentrated under reduced pressure. The residue was purified  using silica gel column chromatography, eluting with EA/PE (1/3) to afford desired product (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3, 4, 6, 7-tetrahydropyrano [3, 4-d] imidazole (295 mg) as a white solid. LC-MS : [M+H]  + 423.72.
Example 189 Synthesis of Compound 189
(R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [4, 5-c] pyridine
Figure PCTCN2019123719-appb-000141
Step 1: synthesis of tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-hydroxypiperidine-1-carboxylate
Figure PCTCN2019123719-appb-000142
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (3.44 g) was treated with SOCl 2 (2.38 g) at 80℃ for 2 h, till an aliquot portion on quenching with a few drops of MeOH revealed the complete consumption of the acid and appearance of a new spot in the TLC. Excess SOCl 2 was distilled off, the residue was dissolve in DCM (30 mL) and then tert-butyl 3-amino-4-hydroxypiperidine-1-carboxylate (2.16 g) , Et 3N (2.02 g) was added at 0 ℃, followed by stirring for 1 h.
100 mL ethylacetoacetate was added to the mixture and then washing with water. An organic layer dried over anhydrous magnesium sulfate. Filtered off and distilled off under reduced pressure, then the residue was purified by combiflash to afford desired product tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-hydroxypiperidine-1-carboxylate (2.77 g, 51%) as a yellow solid.
Step 2: synthesis of tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-oxopiperidine-1-carboxylate
Figure PCTCN2019123719-appb-000143
T-butyl 4-hydroxy-3- { [4- (trifluoromethyl) benzoyl] amino} piperidin-1-carboxylate (2.17 g) was dissolved in DCM (30 mL) , and Dess-Martin periodinane (2.5 g) was dropwise added thereto. After stirring for 5 hours, ethylacetoacetate (50 mL) was dropwise added thereto, and the resulting solution was washed with water. An organic layer dried over anhydrous magnesium sulfate. The reaction solution was filtered off and distilled off under reduced pressure, then the residue was purified by combiflash (DCM: MeOH=95%: 5%) to afford the desired product tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-oxopiperidine-1-carboxylate (1.6 g, 76%) as a yellow solid.
Step 3: synthesis of tert-butyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydrothiazolo [4, 5-c] pyridine-5 (4H) -carboxylate
Figure PCTCN2019123719-appb-000144
To a solution of tert-butyl 3- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxamido) -4-oxopiperidine-1-carboxylate (540 mg) in toluene was added Lawesson's reagent (485 mg) , and the resulting solution was stirred under refluxing for 4 hours. LCMS show the reaction was completed, distilled off under reduced pressure to remove toluene. The residue was purified by combiflash (DCM: MeOH=95%: 5%) to afford the product tert-butyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydrothiazolo [4, 5-c] pyridine-5 (4H) -carboxylate (242 mg) as a brown solid.
Step 4: synthesis of (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [4, 5-c] pyridine hydrochloride
Figure PCTCN2019123719-appb-000145
To a solution of tert-butyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydrothiazolo [4, 5-c] pyridine-5 (4H) -carboxylate (240 mg) in DCM (10 mL) was added 4N HCl/Dioxane (4 mL) . The mixture was stirred for 3h. LCMS showed the reaction was completed. Concentrated under reduced pressure to remove DCM and Dioxane to afford the product (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [4, 5-c] pyridine hydrochloride (148 mg) as a brown solid. MS: [M+H]  + 439.78.
Prepare the following examples (shown in Table 9) essentially as described for Example 189 using the corresponding starting materials.
Table 9
Figure PCTCN2019123719-appb-000146
Example 193 Synthesis of Compound 193
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidin-2-amine
Figure PCTCN2019123719-appb-000147
Step 1: synthesis of ethyl (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
Figure PCTCN2019123719-appb-000148
To a solution of (R) -2- (2, 5-difluorophenyl) pyrrolidine hydrochloride (1.00 g) in EtOH (150 mL) was added DIEA (1.93 g) and ethyl 2-amino-5-chloropyrazolo [1, 5-a] pyrimidine-3-carboxylate (1.13 g) at R. T and then heated to 80 ℃ for 3h. The reaction was detected by TLC and LCMS. The reaction was cooled to room temperature, then the mixture was concentrated in vacuo. The reaction mixture was added into cooled water and stirring. The mixure was filtrated and the residue solid was stirring in 1N HCl solution, then the mixture was filtrated to afford crude product and drying at 50 ℃ for 16h to afford ethyl (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (1.51 g) as a light yellow solid.
Step 2: synthesis of (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
Figure PCTCN2019123719-appb-000149
To a solution of ethyl (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (1.50 g) in EtOH (150 mL) and H 2O (150mL) was added NaOH (467.9 mg) at 80 ℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was pour into 1N HCl solution. The mixture was filtrated and dried at 50 ℃ for 16h to afford crude product (R) -2-amino-5- (2- (2, 5- difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (1.30 g, 93%) as off-white solid.
Step 3: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidin-2-amine
Figure PCTCN2019123719-appb-000150
To a solution of (R) -2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (1.30 g) in MeCN (150 mL) was added 4, 5-dimethoxybenzene-1, 2-diamine (669 mg) and POCl 3 (1.66 g) at 100 ℃ for 16h. The reaction was detected by TLC and LCMS. The mixture was added MeCN (150 mL) and then filtered , the filter cake was adjusted PH=8 by 0.5N NaOH solution, then the mixture was filtrated to afford crude product, the filter cake was dried at 50 ℃ for 16h to afford product (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidin-2-amine (1.2 g) as off-white solid. MS : [M+H]  + 492.81.
Prepare the following Example 194 (shown in Table 10) essentially as described for Example 193 using
Figure PCTCN2019123719-appb-000151
instead of
Figure PCTCN2019123719-appb-000152
and other starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.
Table 10
Figure PCTCN2019123719-appb-000153
*Remark: If there is an isomeride in the above compounds, the present invention also includes its isomeride and also includes their mixture.
Example 195 Synthesis of Compound 195 and/or its isomeride
(R) -2- (5- (2- (2-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000154
(R) -2- (5- (2- (2-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000155
To a solution of (R) -5- (2- (2-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (13.0 g) in MeCN (150 mL) was added 4, 5-diamino-2-methoxybenzonitrile (7.15 g) and POCl 3 (18.34 g) at 100 ℃ for 16h. The reaction was detected by TLC and LCMS. The  mixture was added MeCN (150 mL) and then filtrated, the filter cake was adjusted PH=8 by 0.5N NaOH solution, then the mixture was filtrated to afford crude product, the filter cake was dried to afford product (Compound 195 and/or the isomeride of Compound 195) as off-white solid (13.9g) . LC-MS : [M+H]  + 454.78.
Example 196 Synthesis of Compound 196 and/or its isomeride
(R) -2- (5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000156
(R) -2- (5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000157
Step 1: Synthesis of (R) -2- (5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
Figure PCTCN2019123719-appb-000158
To a solution of (R) -5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (13.0 g) in MeCN (150 mL) was added 4, 5-diamino-2-methoxybenzonitrile (7.15 g) and POCl 3 (18.34 g) at 100 ℃ for 16h. The reaction was detected by TLC and LCMS. The mixture was added MeCN (150 mL) and then filtrated, the filter cake was dried to afford the desired product (Compound 196 and/or the isomeride of Compound 196) as off-white solid (13.6 g) . MS: [M+H]  + 454.78.
Example 197 Synthesis of Compound 197
(S) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000159
(S) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000160
Figure PCTCN2019123719-appb-000161
Step 1: synthesis of ethyl (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
Figure PCTCN2019123719-appb-000162
To a solution of (S) -2- (2, 4-difluorophenyl) pyrrolidine hydrochloride (10.00 g) in EtOH (150 mL) was added DIEA (17.62 g) and ethyl 5-chloropyrazolo [1, 5-a] pyrimidine-3-carboxylate (9.76 g) at R. T and then heated to 80 ℃ for 3h. The reaction was detected by LCMS. The reaction was cooled to room temperature, then the mixture was concentrated in vacuo. The reaction mixture was added into cooled water and stirring. The mixure was filtrated and the residue solid was stirring in 1N HCl solution, then the mixture was filtrated to afford crude product and drying at 50 ℃ for 16h to afford ethyl (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (15.20 g) as a light yellow solid.
Step 2: synthesis of (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
Figure PCTCN2019123719-appb-000163
To a solution of ethyl (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (15.2 g) in EtOH (150 mL) and H 2O (150mL) was added NaOH (4.90 g) at 80 ℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was pour into 1N HCl solution. The mixture was filtrated and dried to afford product (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (13.0 g) as off-white solid.
Step 3: synthesis of (S) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
Figure PCTCN2019123719-appb-000164
To a solution of (S) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (13.0 g) in MeCN (150 mL) was added 4, 5-diamino-2-methoxybenzonitrile (6.77 g) and POCl 3 (17.37 g ) at 100℃ for 16h. The reaction was detected by TLC and LCMS. The mixture was added MeCN (150 mL) and then filtrated , the filter cake was adjusted PH=8 by 0.5N NaOH solution, then the mixture was filtrated to afford crude product, the filter cake was dried to afford the desired product (Compound 197 and/or the isomeride of Compound 197) as off-white solid (13.5 g) . MS: [M+H]  + 472.81.
Example 198 Synthesis of Compound 198 and/or its isomeride
(R) -2- (5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile
Figure PCTCN2019123719-appb-000165
(R) -2- (5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile
Figure PCTCN2019123719-appb-000166
Step 1: synthesis of (R) -ethyl 5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate
Figure PCTCN2019123719-appb-000167
To a solution of (R) -2- (4-fluorophenyl) pyrrolidine hydrochloride (10.00 g) in EtOH (150 mL) was added DIEA (19.25 g) and ethyl 5-chloropyrazolo [1, 5-a] pyrimidine-3-carboxylate (10.62 g) at R. T and then heated to 80 ℃ for 3h. The reaction was detected by TLC and LCMS. The reaction was cooled to room temperature, then the mixture was concentrated in vacuo. The reaction mixture was added into cooled water and stirring. The mixture was filtrated and the residue solid was stirring in 1N HCl solution, then the mixture was filtrated to afford crude product and drying at 50 ℃ for 16h to afford (R) -ethyl 5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] Pyrimidine-3-carboxylate (15.20 g) as a light yellow solid.
Step 2: synthesis of (R) -5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid
Figure PCTCN2019123719-appb-000168
To a solution of (R) -ethyl 5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (15.2 g) in EtOH (150 mL) and H 2O (150mL) was added NaOH (5.15 g) at 80℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was pour into 1N HCl solution. The mixture was filtrated and heated at 50 ℃ for 16h to afford crude product (R) -5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (13.0 g) as off-white solid.
Step 3: synthesis of (R) -2- (5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile and/or isomeride thereof
Figure PCTCN2019123719-appb-000169
To a solution of (R) -5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylic acid (13.0 g) in MeCN (150 mL) was added 4, 5-diamino-2-methoxybenzonitrile (7.15 g) and POCl 3 (18.34 g) at 100 ℃ for 16h. The reaction was detected by TLC and LCMS. The mixture was added MeCN (150 mL) and then filtrated , the filter cake was adjusted PH=8 by 0.5N NaOH solution, then the mixture was filtrated to afford crude product, the filter cake was heated at 50 ℃ for 16h to afford the desired product (Compound 198 and/or the isomeride of Compound 198) as off-white solid (13 g) . MS : [M+H]  + 454.81.
Example 199 Synthesis of Compound 199
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6, 7-dimethoxyimidazo [1, 2-a] pyridin-2-yl) pyrazolo [1, 5-a] pyrimidine
Figure PCTCN2019123719-appb-000170
Step 1: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbohydrazide
Figure PCTCN2019123719-appb-000171
To a solution of (R) -2- (4-fluorophenyl) pyrrolidine hydrochloride (1.00 g) in EtOH (15 mL) was added DIEA (1.93 g) and 1- (5-chloropyrazolo [1, 5-a] pyrimidin-3-yl) ethan-1-one (923.5 mg) at R.T and then heated to 80 ℃ for 3h. The reaction was detected by TLC and LCMS. The reaction was cooled to room temperature, then the mixture was concentrated in vacuo. The reaction mixture was added into cooled water and stirring. The mixure was filtrated and the residue solid was stirring in 1N HCl solution, then the mixture was filtrated to afford crude product and drying at 50 ℃ for 16h to afford (R) -1- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) ethanone (1.56 g) as a light yellow solid.
Step 2: synthesis of (R) -2-chloro-1- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) ethanone
Figure PCTCN2019123719-appb-000172
To a solution of (R) -1- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) ethanone (1.50 g) in MeCN (15 mL) was added NCS (1.17 g) and p-TsOH (151.5 mg) at 90 ℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo the residue was purified by combiflash (PE: EA=50%: 50%) to afford (R) -2-chloro-1- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) ethanone (906 mg) as a light yellow solid.
Step 3: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6, 7-dimethoxyimidazo [1, 2-a] pyridin-2-yl) pyrazolo [1, 5-a] pyrimidine
Figure PCTCN2019123719-appb-000173
To a solution of (R) -2-chloro-1- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) ethanone (900.00 mg) in n-BuOH (10 mL) was added 4, 5-dimethoxypyridin-2-amine (1.11 g) at 130 ℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was purified by combiflash (DCM: MeOH=95%: 5%) to afford (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6, 7-dimethoxyimidazo [1, 2-a] pyridin-2-yl) pyrazolo [1, 5-a] pyrimidine (906 mg) as a light yellow solid. MS: [M+H]  + 477.53.
Example 200 Synthesis of Compound 200
(R) -3- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dihydro-8H- [1, 2, 4] triazolo [3, 4-c] [1, 4] oxazine
Figure PCTCN2019123719-appb-000174
Step 1: synthesis of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbohydrazide
Figure PCTCN2019123719-appb-000175
To a solution of ethyl (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carboxylate (1.00 g) in EtOH (10 mL) was added N 2H 4 . H 2O (437 mg) at R.T and then heated to 80 ℃ for 3h. The reaction was detected by LCMS. The reaction was cooled to room temperature, then the mixture was concentrated in vacuo, the residue was purified by combiflash (PE: EA=50%: 50%) to afford (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbohydrazide (1.01 g) as a light yellow solid.
Step 2: synthesis of (R) -3- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dihydro-8H- [1, 2, 4] triazolo [3, 4-c] [1, 4] oxazine
Figure PCTCN2019123719-appb-000176
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine-3-carbohydrazide (1.00 g) in THF (10 mL) was added Lawesson’s reagent (3.39 g) and morpholin-3-one (459.8 mg) at 70 ℃ for 6h. The reaction was detected by TLC and LCMS. The mixture was concentrated in vacuo and the residue was pour into 1N HCl solution. The mixture was added t-BuOH (10 mL) at 130 ℃ for 6h. The reaction was cooled to room temperature, then the mixture was concentrated in vacuo the residue was purified by combiflash (DCM: MeOH=5%: 95%) to afford (R) -3- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dihydro-8H- [1, 2, 4] triazolo [3, 4-c] [1, 4] oxazine (906 mg) as a light yellow solid. MS: [M+H]  + 424.48.
Comparative compound A 5- [2- (2, 5-Difluorophenyl) pyrrolidin-1-yl] -3- (5-methyl-1H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine
Prepare the following Comparative compound A as described for Example 43 in WO2016097869.
Figure PCTCN2019123719-appb-000177
Example 201 TrkA Kinase assay
Mobility shift assay was used to determine the inhibitory activity of compounds against TrkA kinase. Assay procedures are as follows:
1. Reaction buffer:
1x Kinase base buffer (50 mM HEPES, pH 7.5; 0.0015%Brij-35)
Stop buffer (100 mM HEPES, pH 7.5; 0.015%Brij-35; 0.2%Coating Reagent #3; 50 mM EDTA)
2. Prepare compounds:
1) Dilute the compound to 50X of the final desired highest inhibitor concentration in reaction by 100%DMSO. Transfer 100 μl of this compound dilution to a well in a 96-well plate.
2) For all compounds , transfer the compound in tubes to one well on 96-well storage plate and serially dilute the compound by transferring 30 μl to 60 μl of 100%DMSO in the next well and so forth for a total of 10 concentrations.
3) Add 100 μl of 100%DMSO to two empty wells for no compound control and no enzyme control in the same 96-well plate. Mark the plate as source plate.
4) Prepare intermediate plate
Transfer 10 μl of compound from source plate to a new 96-well plate as the intermediate plate
Add 90 μl of 1x kinase buffer to each well of the intermediate plate.
Mix the compounds in intermediate plate for 10 min on shaker.
3. Prepare assay plate
1) Transfer 5 μl of each well from the 96-well intermediate plate to a 384-well plate in duplicates. For example, A1 of the 96-well plate is transferred to A1 and A2 of the 384-well plate. A2 of the 96-well plate is transferred to A3 and A4 of the 384-well plate, and so on.
4. Kinase reaction
1) Prepare 2.5x enzyme solution
Add kinase in 1x kinase base buffer.
2) Prepare 2.5x peptide solution
Add FAM-labeled peptide and ATP in the 1x kinase base buffer.
3) Assay plate already contains 5 μl of compound in 10%DMSO.
4) Transfer 2.5x enzyme solution to the assay plate
Add 10 μl of 2.5x enzyme solution to each well of the 384-well assay plate.
5) Incubate at room temperature for 10 min.
6) Transfer 2.5x peptide solution to the assay plate
Add 10 μl of 2.5x peptide solution to each well of the 384-well assay plate.
Kinase reaction conditions are shown as Table 11:
Table 11
Figure PCTCN2019123719-appb-000178
7) Kinase reaction and stop
Incubate at 28℃ for specified period of time.
Add 25 μl of stop buffer to stop reaction.
5. Caliper reading
Collect data on Caliper.
6. Curve fitting
1) Copy conversion data from Caliper program.
2) Convert conversion values to inhibition values.
Percent inhibition = (max-conversion) / (max-min) *100.
“max” stands for DMSO control; “min” stands for low control.
3) Fit the data in XLFit excel add-in version 5.4.0.8 to obtain IC 50 values.
Equation used is: Y=Bottom + (Top-Bottom) / (1+ (IC 50/X) ^HillSlope)
The result is expressed with IC 50, shown as Table 11, Compounds of the present disclosure, as exemplified in the Examples, showed IC 50 values in the following ranges: “*” stands for “IC 50 ≤10nM” ; “**” stands for “10nM<IC 50≤50nM” ; “***” stands for “IC 50>50nM.
Table 12
Figure PCTCN2019123719-appb-000179
Figure PCTCN2019123719-appb-000180
Example 202 Ba/F3-TPM3-NTRK1 and Ba/F3-ETV6-NTRK3 cells proliferation assay
1. Cell culture
Cell line: Ba/F3 cells with TPM3-NTRK1 or ETV6-NTRK3 fusion mutation gene stably overexpressed named Ba/F3-TPM3-NTRK1 and Ba/F3-ETV6-NTRK3.
A. Culture medium
RPMI 1640 and 10%FBS and 1%PS and 2 ug/mL puromycin.
B. Cell recovery
a) The medium was preheated in a 37℃ water bath in advance.
b) Remove the cryogenic vials from the liquid nitrogen tank, quickly put it into a 37℃ water bath, and completely melt it in 1 min.
c) Transfer the cell suspension to a 15 mL centrifuge tube containing 8 mL medium, centrifuge 1000 rpm, 5 min.
d) Discard the supernatant, resuspend the cells in 1 mL medium, and transfer to a 75 cm 2 flask containing 15 mL medium, culture the cells in an incubator at 37℃, 5%CO 2.
C. Cell passage
a) The medium was preheated in a 37℃ water bath in advance.
b) Collect cell to a 15 mL centrifuged tube, centrifuge at 1000 rpm for 5 min. Discard the supernatant, count, and make the cell density at 1.49x10 4 cells/mL, then place it in a 37℃, 5%CO 2 incubator.
2. Compound preparation
a) The test compound (20 mM stock solution) was diluted to 60 μM with 100%DMSO as starting concentration then 3-fold serial diluted with "9+0" concentrations. in a 96-well dilution plate (Thermo, Cat. No. 249944) ;
b) The above compound solution was diluted 1: 20 times with culture medium to prepare a 10 fold working solution;
3. Cell plating
a) Take cells in log phase growth, centrifuge at 1000 rpm for 5 min, then resuspend the cells with culture medium, then count cells;
b) Cells were seeded to 96-well cell culture plate with density at 2000 cells/well;
4. Compound treatment
a) Compounds prepared at step 2 were added to cell plate with 15 μL per well, the final concentrations were 300, 100, 33.33, 11.11, 3.70, 1.23, 0.41, 0.14, 0.05 and 0 nM, and the final concentration of DMSO was 0.5%. The blank control well was a culture medium (0.5%DMSO) ;
c) The cells were incubated for an additional 72 h in the incubator.
5. Detection
a) Remove the 96-well cell culture plate and add 50 μl of CTG reagent (CellTiter Glo kit, promega, Cat #G7573) .
b) Plate was shaked for 2 min and then let it cool for 30 min at room temperature.
c) The Luminescence signal value was read using a PerkinElmer reader.
Experimental data analysis
Data were analyzed using GraphPad Prism 6.0 software to obtain a fitted curve of compound activity.
Fit the Compound IC50 from non-linear regression equation:
Y=Bottom + (Top-Bottom) / (1+10^ ( (LogIC50-X) ×HillSlope) ) ;
X: The log of the concentration of the compound; Y: Luminescence value.
Table 13
EX No. TPM3-NTRK1 IC 50 (nM) ETV6-NTRK3 IC 50 (nM) NTRK1-G595R IC 50 (nM)
LOXO-101 - - 3073.1
Compound 4 3.55 2.78 -
Compound 11 13.64 5.785 -
Compound 7 8.17 5.68 107
Compound 8 2.209 1.548 -
Compound 9 2.05 2.11 -
Compound 10 1.16 1.2 -
Compound 11 13.64 5.785 -
Compound 12 7.713 2.716 -
Compound 13 0.98 1.83 -
Compound 14 12.52 10.03 -
Compound 16 9.182 4.212 -
Compound 17 4.1 3.56 -
Compound 18 3.813 2.833 -
Compound 22 2.929 2.454 -
Compound 23 20.87 - -
Compound 30 13.78 - -
Compound 31 3.88 - -
Compound 34 3.33 - -
Compound 39 7.42 - -
Compound 94 1.38 - 62.9
Compound 101 0.28 - 164.6
Compound 125 1.38 2.21 46.6
Compound 123 1.9 - 116.8
Compound 126 1.04 - 105.9
Compound 141 30.08 20.77 -
Compound 144 3.39 - 221.5
Compound 148 2.19 1.56 81.8
Compound 155 1.2 0.53 18.2
Compound 156 3.92 5.65 182.9
Compound 160 1.06 2.05 15.6
Compound 162 1.33 - 118.8
Compound 166 4.67 7.73 -
Compound 167 13.24 19.6 -
Compound 169 2.91 - 102.7
Compound 171 3.41 2.63 215.7
Compound 172 2.08 2.49 256.9
Compound 173 2.19 - 89.8
Note: “-” stands for “not tested” .
Example 203 Liver microsomes metabolic stability assay
Pooled human liver microsomes (Cat. 452117) were purchased from Corning. Pooled male rat liver microsomes (Cat. R1000) and pooled male mouse liver microsomes (Cat. M1000) were purchased from XENOTECH. Microsomes were stored at -80℃.
1) Make a master solution containing phosphate buffer, ultra-pure H 2O and MgCl 2 solution according to Table 14.
Table 14 Preparation of master solution
Buffer Stock Concentration Volume Final Concentration
Phosphate buffer 200 mM 200 μL 100 mM
Ultra-pure H 2O - 106 μL -
MgCl 2 solution 50 mM 40 μL 5 mM
2) Two separated experiments were performed as follows.
a) With NADPH: 10 μL of 20 mg/mL liver microsomes and 40 μL of 10 mM NADPH were added to the incubations. The final concentrations of microsomes and NADPH were 0.5 mg/mL and 1 mM, respectively.
b) Without NADPH: 10 μL of 20 mg/mL liver microsomes and 40 μL of ultra-pure H 2O were added to the incubations. The final concentration of microsomes was 0.5 mg/mL.
3) The reaction was started with the addition of 4 μL test compounds solutions or control compound solution (verapamil) at the final concentration of 2 μM and carried out at 37℃.
4) Aliquots of 50 μL were taken from the reaction solution at 0, 15, 30, 45 and 60 min. The reaction solutions were stopped by the addition of 4 volumes of cold acetonitrile with IS (100 nM alprazolam, 200 nM caffeine, 200 nM labetalol and 2 μM ketoprofen) . Samples were centrifuged at 3,220g for 40 minutes. Aliquot of 100 μL of the supernatant was mixed with 100 μL of ultra-pure H 2O and then used for LC-MS/MS analysis. All experiments were performed in duplicate.
The slope value, k, was determined by linear regression of the natural logarithm of the remaining percentage of the parent drug vs. incubation time curve.
The in vitro half-life (in vitro t 1/2) was determined from the slope value:
in vitro t 1/2=- (0.693/k)
Conversion of the in vitro t 1/2 (in min) into the in vitro intrinsic clearance (in vitro CL int, in μL/min/mg proteins) was done using the following equation (mean of duplicate determinations) :
Figure PCTCN2019123719-appb-000181
The control compound (verapamil) was included in the assay. Any value of the compounds that was not within the specified limits will be rejected and the experiment will be repeated.
The results of metabolic stability in different species of liver microsomes are shown in Table 15.
Table 15
Figure PCTCN2019123719-appb-000182
Figure PCTCN2019123719-appb-000183
Note: “-” stands for “not tested” .
As a result, it has been confirmed that the exemplified compounds of the present invention have drastically improved metabolic stability in Human/Rat/Mouse liver microsomes compared with the Comparative compound A. This improved stability indicated superior pharmacokinetic properties and better clinical output in human.
Example 204 Plasma protein binding measurements
The plasma protein binding was measured as the following procedure.
1) Preparation of 100 mM sodium phosphate and 150 mM NaCl buffer (PBS)
A basic solution was prepared by dissolving 14.2 g/L Na 2HPO 4 and 8.77 g/L NaCl in deionized water and the solution could be stored at 4℃ for up to 7 days. An acidic solution was prepared by dissolving 12.0 g/L NaH 2PO 4 and 8.77 g/L NaCl in deionized water and the solution could be stored at 4℃ for up to 7 days. The basic solution was titrated with the acidic solution to pH 7.4 and store at 4℃ for up to 7 days. pH was checked on the day of experiment and was adjusted if outside specification of 7.4 ± 0.1.
2) Preparation of plasma
Frozen plasma was thawed immediately at room temperature.
The plasma was centrifuged at 3,220 g for 10 minutes to remove clots and supernatant was collected into a fresh tube. The pH of the plasma was checked and recorded.
Note: a) . Only use plasma that has been thawed no more than twice since arrival. b) . Only use plasma within the range of pH 7 to pH 8.
3) Preparation of working solutions
The working solutions of test compounds and control compound ketoconazole were prepared in DMSO at the concentration of 200 μM. And then 3 μL of working solution was removed to mix with 597 μL of human, rat or mouse plasma to achieve a final concentration of 1 μM (0.5%DMSO) . Plasma samples were vortexed thoroughly.
4) Preparation of dialysis membranes
The dialysis membranes were soaked in ultrapure water for 60 minutes to separate strips, then in 20%ethanol for 20 minutes, finally in dialysis buffer for 20 minutes.
5) Procedure for equilibrium dialysis
The dialysis apparatus was assembled according to the manufacturer’s instruction. Each cell was filled with 120 μL of plasma sample and dialyzed against equal volume of dialysis buffer (PBS) . The assay was performed in duplicate. The dialysis plate was sealed and incubated in an incubator at 37℃ with 5%CO 2 at 100 rpm for 6 hours. At the end of incubation, seal was removed and 50 μL of samples from both buffer and plasma chambers were transferred to wells of a 96-well plate.
6) Procedure for sample analysis
50 μL of blank plasma was added to each buffer samples and an equal volume of PBS was supplemented to the collected plasma sample. 300 μL of room temperature quench solution (acetonitrile containing internal standards (IS, 100 nM Alprazolam, 500 nM Labetalol and 2 μM Ketoprofen) ) was added to precipitate protein. Samples in plate were vortexed for 5 minutes and centrifuged at 3,220 g for 30 minutes at 4℃. And then 100 μL of the supernatant was transferred to a new 96-well plate with 100 μL or 200 μL water (depends on the LC-MS signal response and peak shape) for LC-MS/MS analysis.
Calculate the percentages of test compound and control compound bound as follows:
%Free = (Peak Area Ratio buffer chamber /Peak Area Ratio plasma chamber) *100
%Bound = 100 -%Free
%Recovery = (Peak Area Ratio buffer chamber + Peak Area Ratio plasma chamber) /Peak Area Ratio total sample*100
Peak Area Ratio buffer chamber means the conc for free fraction
Peak Area Ratio plasma chamber means the conc for both free and bound fraction
Peak Area Ratio total sample means the conc for starting sample before incubation
Plasma protein binding results of control compound and test Compounds in different species are shown in Table 16.
Table 16
Figure PCTCN2019123719-appb-000184
Note: “-” stands for “not tested” .
Generally, only the unbound fraction can have a biological effect or be metabolized. Therefore, the degree of binding to plasma proteins significantly influences the pharmacokinetic and pharmacodynamics properties of a drug.
As shown in Table 16, the Comparative compound A reflected a high degree of plasma protein binding, therefore the efficacy of the drug might be reduced. Unexpectedly, the exemplified compounds of the present invention have lower degree of plasma protein binding compared with the Comparative compound A. It indicated the present invention had superior pharmacokinetic and pharmacodynamics properties in human.
Example 205 Cytochrome P450 measurements
The cytochrome P450 was measured as the following procedure:
1) A master solution containing phosphate buffer, ultra-pure H 2O, MgCl 2 solution and human liver microsomes was made according to Table 17, and then 1 μL of 2 mM of compound solution or 1 μL of DMSO (as without inhibitor control) was added to the above master solution. The final concentration of test compounds or control compounds was 10 μM.
Table 17
Reagent Stock Concentration Volume Final Concentration
MgCl 2 solution 50 mM 20 μL 5 mM
Phosphate buffer 200 mM 100 μL 100 mM
Ultra-pure H 2O - 56 μL -
Human liver microsomes 20 mg/mL 2 μL 0.2 mg/mL
2) For CYP1A2 inhibition, 1 μL of specific drug substrate (Phenacetin: 8 mM) was added at the final concentration of 40 μM to the above solution.
3) For CYP2C8 inhibition, 1 μL of specific drug substrate (Paclitaxel: 1 mM) was added at the final concentration of 5 μM to the above solution.
4) For CYP2C9 inhibition, 1 μL of specific drug substrate (Tolbutamide: 40 mM) was added at the final concentration of 200 μM to the above solution.
5) For CYP2C19 inhibition, 1 μL of specific drug substrate ( (s) -Mephenytoin: 10 mM) was added at the final concentration of 50 μM to the above solution.
6) For CYP3A4 inhibition, 1 μL of specific drug substrate (Midazolam: 1 mM) was added at the final concentration of 5 μM to the above solution.
7) For CYP3A4 inhibition, 1 μL of specific drug substrate (Testosterone: 10 mM) was added at the final concentration of 50 μM to the above solution.
8) The mixture was pre-warmed at 37℃ for 5 min. The reaction was started by the addition of 20 μL of 10 mM NADPH solution at the final concentration of 1 mM and carried out at 37℃.
9) The reaction was stopped by addition of 300 μL of cold quench solution (methanol containing internal standards (IS, 500 nM Labetalol, 100 nM Alprazolam and 2 μM Ketoprofen) at the designated time points (Phenacetin: 20 min; Paclitaxel: 10 min; Tolbutamide: 20 min; (s) -Mephenytoin: 20 min; Midazolam: 5 min; Testosterone: 10 min) . Samples were vortexed for 5 minutes and centrifuged at 3220 g for 40 minutes at 4℃. And then 100 μL of the supernatant was transferred to a new 96-well plate with 100 μL or 200 μL water (depends on the LC-MS signal response and peak shape) for LC-MS/MS analysis.
All experiments were performed in duplicates.
Inhibition percentages for compounds against CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 shown in Table 18 (the unit is %) .
Table 18
Figure PCTCN2019123719-appb-000185
As a result, it has been confirmed that the exemplified compounds of the present invention have low inhibition against CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4. Especially for CYP3A4, which is a main isoform for drug metabolism, compounds in the present invention have much less inhibition compared with the Comparative compound A.

Claims (49)

  1. A compound of Formula I, or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
    Figure PCTCN2019123719-appb-100001
    wherein,
    ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
    ring B is 5-membered aromatic heterocycle;
    X and Z are each independently selected from C, N, O, or S;
    Y is C or N;
    R 1 is absent, H, or -C 1-8 alkyl;
    R 2 is H, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, or -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring, wherein the -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 0-4 alkyl-C 3-10 carbocyclic ring, -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring is optionally substituted with -C 1-8 alkyl, -C 2-8 alkynyl, -C 1-8 haloalkyl, -C 1-8 alkyl-OH, halogen, OH, CN, NH 2, -C 0-4 alkyl-COOR 10, -C 6-10 aryl ring, -O-C 6-10 aryl ring, substituted or unsubstituted -C 3-10 carbocyclic ring, or substituted or unsubstituted -C 3-10 heterocyclic ring;
    R 3 is absent, C 3-10 heterocyclic ring; or
    R 2 and R 3 together with the atoms to which they are attached to form a 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring, wherein the 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring is optionally substituted with halogen, OH, CN, NH 2, -CONHOH, -CONH 2, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-O (CO) OR 10, -C 1-8 alkoxy, -C 1-8 haloalkoxy, -C 1-8 alkoxy-C 1-8 alkoxy, -C 1-8 alkylthio, -C 1-8 haloalkylthio, -C 1-8 alkyl, -C 1-8 haloalkyl, -C 0-4 alkyl-OH, -O-CH 2-CN, -C 0-4 alkyl-O-C 3-10 heterocyclic ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring, or the 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring forms a ring structure with other substituted or unsubstituted carbocyclic ring, substituted or  unsubstituted heterocyclic ring, substituted or unsubstituted aryl ring, or substituted or unsubstituted heteroaryl ring;
    R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
    R 10 is H, or -C 1-8 alkyl;
    wherein the heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, O or B.
  2. The compound of claim 1, wherein ring A is
    Figure PCTCN2019123719-appb-100002
  3. The compound of claim 1 or 2, wherein X is independently selected from O, S or N.
  4. The compound of anyone of claims 1-3, wherein Y is C.
  5. The compound of anyone of claims 1-4, wherein Z is N.
  6. The compound of anyone of claims 1-5, wherein R 4 is
    Figure PCTCN2019123719-appb-100003
  7. The compound of claim 1, wherein the compound is formula II or a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
    Figure PCTCN2019123719-appb-100004
    wherein,
    ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
    R 1 is H, or -C 1-8alkyl;
    R 2 is H, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring, or -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring, wherein the -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-NH-COOR 10, -C 0-4 alkyl-O (CO) R 10, -C 0-4 alkyl-O (CO) -C 1-4 alkyl-NHCO-R 10, -C 1-4 alkyl-NH 2, -C 0-4 alkyl-OH, -C 1-4 alkyl-C 3-10 carbocyclic ring,  -C 0-4 alkyl-C 3-10 heterocyclic ring, -C 0-4 alkyl-C 6-10 aryl ring, or -C 0-4 alkyl-C 5-10 heteroaryl ring is optionally substituted with -C 1-8alkyl, -C 2-8 alkynyl, -C 1-8 haloalkyl, -C 1-8 alkyl-OH, halogen, OH, CN, NH 2, -C 0-4 alkyl-COOR 10, -C 6-10 aryl ring, -O-C 6-10 aryl ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring;
    R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
    R 10 is H, or -C 1-8 alkyl;
    wherein the heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, O or B.
  8. The compound of claim 7, wherein ring A is
    Figure PCTCN2019123719-appb-100005
  9. The compound of claim 7 or 8, wherein R 1 is independently selected from H or CH 3.
  10. The compound of anyone of claims 7-9, wherein R 4 is
    Figure PCTCN2019123719-appb-100006
  11. The compound of anyone of claims 7-10, wherein R 2 is independently selected from
    Figure PCTCN2019123719-appb-100007
    Figure PCTCN2019123719-appb-100008
    Figure PCTCN2019123719-appb-100009
  12. The compound of anyone of claims 7-11, wherein R 2 is
    Figure PCTCN2019123719-appb-100010
  13. The compound of claim 1, wherein the compound is of formula III or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof:
    Figure PCTCN2019123719-appb-100011
    wherein,
    ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
    ring C is a 5-to 6-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring;
    X and Z are each independently selected from C, N, O, or S;
    Y is C or N;
    R 1 is absent, H, or -C 1-8 alkyl;
    R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
    R 5 and R 6 are each independently selected from H, OH, NH 2, CN, -COOH, -CONHOH, -CONH 2, halogen, -C 1-8 alkyl, -C 0-4 alkyl-COOR 10, -C 0-4 alkyl-O (CO) OR 10, -C 1-8 alkoxy, -C 1-8 haloalkoxy, -C 1-8 alkoxy-C 1-8 alkoxy, -C 1-8 alkylthio, -C 1-8 haloalkylthio, -C 1-8 alkyl, -C 1-8 haloalkyl, -C 0-4 alkyl-OH, -O-CH 2-CN, -C 0-4 alkyl-O-C 3-10 heterocyclic ring, substituted or unsubstituted -C 3-10 carbocyclic ring or substituted or unsubstituted -C 3-10 heterocyclic ring;
    or R 5 and R 6 together with the atoms to which they are attached to form a 5 to 12-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring, wherein the 5 to 12-membered carbocyclic ring, heterocyclic ring, aryl ring, or heteroaryl ring is optionally substituted with halogen;
    R 10 is H, or -C 1-8 alkyl;
    wherein the heterocyclic ring or the heteroaryl ring optionally has 1, 2 or 3 heteroatoms independently selected from N, S, or O.
  14. The compound of claim 13, wherein ring A is
    Figure PCTCN2019123719-appb-100012
  15. The compound of claim 13 or 14, wherein ring C is 6-membered aromatic ring.
  16. The compound of anyone of claims 13-15 wherein ring C is phenyl, pyridyl, pyridazinyl, or pyrimidinyl.
  17. The compound of anyone of claims 13-16, wherein ring C is phenyl.
  18. The compound of anyone of claims 13-17, wherein X is selected from O, S, or N.
  19. The compound of anyone of claims 13-18, wherein X is N.
  20. The compound of anyone of claims 13-19, wherein Y is C.
  21. The compound of anyone of claims 13-20, wherein Z is N.
  22. The compound of anyone of claims 13-21, wherein R 1 is absent, H, or CH 3.
  23. The compound of anyone of claims 13-22, wherein R 4 is
    Figure PCTCN2019123719-appb-100013
  24. The compound of anyone of claims 13-23, wherein R 5 and R 6 are each independently selected from H, OH, NH 2, F, Cl, Br, -CN, -CF 3, -OCF 3, CH 3, -O-CH 3, -S-CH 3, -CH 2OH , -COOH ,
    Figure PCTCN2019123719-appb-100014
    Figure PCTCN2019123719-appb-100015
  25. The compound of anyone of claims 13-24, wherein R 5 and R 6 are both -O-CH 3.
  26. The compound of anyone of claims 13-25, wherein R 5 and R 6 together with the atoms to which they are attached from
    Figure PCTCN2019123719-appb-100016
  27. The compound of claim 1, wherein the compound is Formula IV or an isomeride, a stereoisomer, tautomer, pharmaceutically acceptable salt, prodrug, chelate, non-covalent complex, or solvate thereof,
    Figure PCTCN2019123719-appb-100017
    wherein,
    Ring A is C 5-6 heterocyclic ring, wherein the C 5-6 heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
    R 4 is (i) phenyl optionally substituted with one or more substituents independently selected from halogen, -C 1-4 alkyl, -C 1-4 haloalkyl, -C 1-4 alkoxyl, or (ii) a C 5-6 heteroaryl ring having a ring heteroatom selected from N, S, or O, wherein the C 5-6 heteroaryl ring is optionally substituted with one or more halogen atoms;
    R’ is H, NH 2, or -C 1-4 alkyl;
    Ring B’ is a 5-membered aromatic heterocyclic ring optionally comprising 1, 2 or 3 hetero atoms independently selected from N, S, or O;
    Ring C’ is a phenyl, 6-membered heterocyclic ring, or 6-membered heteroaryl ring;
    X’ and Z’ are each independently selected from C, N, O, or S;
    Y’ is C or N;
    R” is -C (O) -C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2-C 1-4 alkyl, -NR 7 (CH 2mNR 8R 9, - (CH 2mC 4-10 heterocyclyl; or NH 2, -C (O) OH, -C (O) NH 2, -C 1-4 alkyl, -C 1-4 alkoxyl, -C (O) -C 1-4 alkyl, -C (O) O-C 1-4 alkyl, -OC (O) O-C 1-4 alkyl, -S-C 1-4 alkyl, -SO-C 1-4 alkyl, -SO 2-C 1-4 alkyl, -OC 4-6heterocyclyl, -NR 7 (CH 2mNR 8R 9, - (CH 2mC 4-10 heterocyclyl optionally substituted with one or more substituents independently selected from OH, CN, NH 2, -C (O) OH, halogen, -C 1-4 alkyl or -C 1-4 alkoxyl; or
    any two R” together with the atoms to which they are attached form a 5-to 12-membered ring;
    R 7, R 8 and R 9 are each independently selected from H, or -C 1-4 alkyl;
    m and n are each independently selected from 0, 1, 2, 3 or 4.
  28. The compound of claim 27, wherein Ring A is
    Figure PCTCN2019123719-appb-100018
  29. The compound of claim 27 or 28, wherein R’ is selected from H.
  30. The compound of anyone of claims 27-29, wherein R 4 is
    Figure PCTCN2019123719-appb-100019
    Figure PCTCN2019123719-appb-100020
  31. The compound of anyone of claims 27-30, wherein Ring B’ is selected from imidazole, oxazole, thiazole, triazole or pyrrole.
  32. The compound of anyone of claims 27-31, wherein Ring B’ is selected from
    Figure PCTCN2019123719-appb-100021
  33. The compound of anyone of claims 27-32, wherein Ring C’ is selected from phenyl, pyridine, pyrazine, pyrimidine, pyridazine, piperidine, or tetrahydropyran.
  34. The compound of anyone of claims 27-33, wherein Ring C’ is selected from
    Figure PCTCN2019123719-appb-100022
  35. The compound of anyone of claims 27-34, wherein R” is selected from
    Figure PCTCN2019123719-appb-100023
    Figure PCTCN2019123719-appb-100024
  36. The compound of anyone of claims 27-34, wherein two R” with the atoms to  which they are attached form
    Figure PCTCN2019123719-appb-100025
    Figure PCTCN2019123719-appb-100026
  37. The compound of claim 1, or an isomeride, pharmaceutically acceptable salt or solvate thereof, wherein the compound is:
    1) (R) -4- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) phenyl) morpholine;
    2) (R) -1- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) piperidin-4-ol;
    3) 5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (tetrahydrofuran-3-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    4) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethan-1-ol;
    5) (1S, 4s) -4- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclohexan-1-ol;
    6) (R) -4- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) morpholine;
    7) (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-2-ol;
    8) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (pyridin-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    9) (R) -4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) phenol;
    10) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (pyrazin-2-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    11) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethan-1-amine;
    12) methyl ( (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethyl) carbamate;
    13) (R) -3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzonitrile;
    14) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (6- (trifluoromethyl) pyridin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    15) 3- (5- (azetidin-2-yl) -4H-1, 2, 4-triazol-3-yl) -5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    16) ethyl (R) -5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazole-3-carboxylate;
    17) (R) -5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazole-3-carboxylic acid;
    18) (3S) -3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclohexan-1-ol;
    19) (3S) -3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclopentan-1-ol;
    20) tert-butyl 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) azetidine-1-carboxylate;
    21) (R) -1- (4- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) piperidin-4-ol;
    22) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (piperidin-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    23) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclobutan-1-ol;
    24) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclobutan-1-amine;
    25) (S) -2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluoropropan-2-ol;
    26) (R) -2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluoropropan-2-ol;
    27) (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1, 3, 3, 3-hexafluoropropan-2-ol;
    28) 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluorobutan-2-ol;
    29) 3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1, 1-trifluoro-2-methylpropan-2-ol;
    30) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2-methylpropan-2-ol;
    31) (R) -3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclobutan-1-ol;
    32) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (tetrahydro-2H-pyran-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    33) (R) -2- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2-methylpropan-1-ol;
    34) (R) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethan-1-ol;
    35) 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) piperidin-4-ol;
    36) (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 2, 3, 4-tetrahydroisoquinoline;
    37) (1R, 3r) -3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) adamantan-1-ol;
    38) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (1-methylpiperidin-4-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    39) (R) -2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) propan-1-ol;
    40) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (4- (piperazin-1-yl) phenyl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    41) (R) -3- (5- (4, 4-difluorocyclohexyl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    42) (R) - (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) (phenyl) methanol;
    43) (R) - (3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) bicyclo [1.1.1] pentan-1-yl) methanol;
    44) (R) -3- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) bicyclo [1.1.1] pentan-1-amine;
    45) (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) benzo [c] [1, 2] oxaborol-1 (3H) -ol;
    46) 1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -1, 1-difluorobutan-2-ol;
    47) 1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2, 2, 2-trifluoroethan-1-ol;
    48) 1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) prop-2-yn-1-ol;
    49) 3- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) morpholine;
    50) (R) -3- (5- (1H-indol-5-yl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    51) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethyl L-leucinate hydrochloride;
    52) 2- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) -2-fluoroethan-1-ol;
    53) (R) -1- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclopropan-1-ol;
    54) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (6- (4-methylpiperazin-1-yl) pyridin-3-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    55) (S) -1- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) ethyl L-valylvalinate hydrochloride;
    56) (R) -6- (5- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) quinoline;
    57) (R) -3- (5- (1H-benzo [d] imidazol-6-yl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    58) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (4-phenoxyphenyl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    59) (R) -3- (5- (1H-indazol-6-yl) -4H-1, 2, 4-triazol-3-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    60) (1R, 2S, 3R, 5S) -5- (5- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4H-1, 2, 4-triazol-3-yl) cyclohexane-1, 2, 3, 5-tetraol;
    61) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (2, 3-dihydrobenzofuran-6-yl) -4H-1, 2, 4-triazol-3-yl) pyrazolo [1, 5-a] pyrimidine;
    62) 5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    63) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (R) -hexahydropyrrolo [1, 2-a] pyrazin-2 (1H) -yl) benzo [d] thiazole;
    64) (R) -4- (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-c] pyridin-6-yl) morpholine;
    65) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-c] pyridine;
    66) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazol-6-yl) ethan-1-ol;
    67) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazol-6-yl) methanol;
    68) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) benzo [d] oxazol-6-yl) ethan-1-ol;
    69) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) benzo [d] oxazol-6-yl) methanol;
    70) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3H-imidazo [4, 5-c] pyridin-6-yl) ethan-1-ol;
    71) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- (trifluoromethoxy) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    72) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethyl) -3H-imidazo [4, 5-c] pyridine;
    73) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) oxazolo [4, 5-c] pyridine;
    74) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6-fluoro-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    75) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) thiazolo [4, 5-c] pyridine;
    76) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-d] pyridazine;
    77) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6-methoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    78) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    79) (R) -6- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -2, 2-difluoro-5H- [1, 3] dioxolo [4', 5': 4, 5] benzo [1, 2-d] imidazole;
    80) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethoxy) benzo [d] oxazole;
    81) (R) -3- (6- (difluoromethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    82) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7, 9, 10, 12, 13-hexahydro-1H- [1, 4, 7, 10] tetraoxacyclododecino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
    83) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1-methyl-6, 7-dihydro-1H- [1, 4] dioxino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
    84) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-3H-imidazo [4, 5-b] pyridine;
    85) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-imidazo [4, 5-c] pyridine;
    86) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methyl-1H-imidazo [4, 5-c] pyridine;
    87) (R) -8- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7H-purin-6-amine;
    88) (R) -8- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7H-purin-6-ol;
    89) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -N-hydroxy-5-methoxy-1H-benzo [d] imidazole-6-carboxamide;
    90) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carboxylic acid;
    91) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carboxamide;
    92) (R) -3- (5-chloro-6- (trifluoromethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    93) 1- (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) ethan-1-ol;
    94) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluorobenzo [d] oxazol-5-yl) methanol;
    95) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3H-imidazo [4, 5-c] pyridin-6-yl) methanol;
    96) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydro-1H- [1, 4] dioxino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
    97) (R) -3- (7-chloro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    98) (R) -3- (7-chloro-5-fluoro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    99) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7-methyl-1H-imidazo [4, 5-c] pyridine;
    100) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-methoxybenzo [d] oxazole;
    101) (R) -3- (5, 6-bis (2-methoxyethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    102) (R) -6, 7-dichloro-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] pyridine;
    103) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-methyl-3H-imidazo [4, 5-c] pyridine;
    104) (R) -3- (4, 7-dichloro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    105) (R) -3- (5, 6-dichloro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    106) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methyl-3H-imidazo [4, 5-b] pyridine;
    107) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carbonitrile;
    108) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-fluoro-3H-imidazo [4, 5-b] pyridine;
    109) (R) -3- (5, 6-bis (difluoromethoxy) -1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    110) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethyl) -1H-imidazo [4, 5-b] pyridine;
    111) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 7-difluorobenzo [d] oxazole;
    112) (R) -3- (5-chloro-6-methoxy-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    113) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (7- (trifluoromethoxy) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    114) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (trifluoromethyl) -3H-imidazo [4, 5-b] pyridine;
    115) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-diyl dimethyl bis (carbonate) ;
    116) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- ( (trifluoromethyl) thio) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    117) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-diol;
    118) 5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5- ( ( (R) -tetrahydrofuran-3-yl) oxy) -6- ( ( (S) -tetrahydrofuran-3-yl) oxy) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    119) (R) -2, 2'- ( (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-diyl) bis (oxy) ) diacetonitrile;
    120) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dimethoxybenzo [d] oxazole;
    121) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] quinoxaline;
    122) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7-methyl-3H-imidazo [4, 5-b] pyridine;
    123) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-fluoro-1H-benzo [d] imidazole-6-carbonitrile;
    124) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1-methyl-1H-imidazo [4, 5-c] pyridine;
    125) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile;
    126) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylthio) -1H-benzo [d] imidazole-6-carbonitrile;
    127) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (7-fluoro-6-methoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    128) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] pyrazine;
    129) (R) -6-bromo-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] pyrazine;
    130) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-imidazo [4, 5-b] phenazine;
    131) (R) -6- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) - [1, 3] dioxolo [4', 5': 4, 5] benzo [1, 2-d] oxazole ;
    132) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-6-ol ;
    133) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carbonitrile;
    134) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7, 8-dihydro-1H, 6H- [1, 4] dioxepino [2', 3': 4, 5] benzo [1, 2-d] imidazole;
    135) (R) - (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3H-imidazo [4, 5-c] pyridin-6-yl) methanol;
    136) (R) -3- (5, 6-difluoro-1H-benzo [d] imidazol-2-yl) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidine;
    137) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5-difluoro-1H-benzo [d] imidazole-6-carboxylate;
    138) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5-difluoro-1H-benzo [d] imidazole-6-carboxylic acid;
    139) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5-fluoro-6- (trifluoromethyl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    140) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-ethoxy-1H-benzo [d] imidazole-5-carbonitrile;
    141) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluoro-1H-benzo [d] imidazole-5-carboxylic acid;
    142) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (methylamino) -1H-benzo [d] imidazole-5-carbonitrile;
    143) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-morpholino-1H-benzo [d] imidazole-5-carbonitrile;
    144) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (dimethylamino) -1H-benzo [d] imidazole-5-carbonitrile;
    145) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (3-hydroxyazetidin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile;
    146) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydroimidazo [4', 5': 4, 5] benzo [1, 2-e] [1, 4] diazepin-9 (3H) -one;
    147) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -7, 8-dihydro-3H-imidazo [4', 5': 4, 5] benzo [1, 2-f] [1, 4] oxazepin-9 (6H) -one;
    148) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5, 6-dicarbonitrile;
    149) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-hydroxy-1H-benzo [d] imidazole-5-carbonitrile;
    150) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (2-hydroxyethoxy) -1H-benzo [d] imidazole-5-carbonitrile;
    151) (R) -6-bromo-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile;
    152) methyl (R) -5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carboxylate;
    153) (R) -5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carboxylic acid;
    154) (R) -5-cyano-2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-6-carboxamide;
    155) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carboxylate;
    156) (R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile;
    157) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (trifluoromethyl) -1H-benzo [d] imidazole-6-carbonitrile;
    158) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-fluoro-1H-benzo [d] imidazole-7-carboxylate;
    159) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methyl-1H-benzo [d] imidazole-5-carbonitrile;
    160) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-N-methyl-1H-benzo [d] imidazole-5-carboxamide;
    161) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-N, N-dimethyl-1H-benzo [d] imidazole-5-carboxamide;
    162) (R) -4- ( (2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazol-5-yl) methyl) morpholine;
    163) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (4-methylpiperazin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile;
    164) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (S) -3-hydroxypyrrolidin-1-yl) -1H-benzo [d] imidazole-5-carbonitrile;
    165) 6- ( (S) -2-cyanopyrrolidin-1-yl) -2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazole-5-carbonitrile;
    166) methyl (5-cyano-2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazol-6-yl) -L-prolinate;
    167) (5-cyano-2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-benzo [d] imidazol-6-yl) -L-proline;
    168) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- ( (2- (dimethylamino) ethyl) (methyl) amino) -1H-benzo [d] imidazole-5-carbonitrile;
    169) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (2-methoxyethoxy) -1H-benzo [d] imidazole-5-carbonitrile;
    170) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6- (methylsulfonyl) -1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    171) 2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylsulfinyl) -1H-benzo [d] imidazole-6-carbonitrile;
    172) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylsulfonyl) -1H-benzo [d] imidazole-6-carbonitrile;
    173) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5- (methylsulfonyl) -1H-benzo [d] imidazole-6-carboxamide;
    174) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxybenzo [d] oxazole-5-carbonitrile;
    175) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4-fluorobenzo [d] oxazole-7-carboxylate;
    176) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6- (trifluoromethoxy) benzo [d] oxazole-5-carbonitrile;
    177) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-hydroxybenzo [d] oxazole-5-carbonitrile;
    178) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxybenzo [d] oxazole-6-carboxylate;
    179) (R) -6- (difluoromethoxy) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methylbenzo [d] oxazole;
    180) ( (2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxybenzo [d] oxazol-5-yl) methyl) -L-proline;
    181) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 8-dimethoxy- [1, 2, 4] triazolo [1, 5-c] pyrimidine;
    182) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dimethoxy- [1, 2, 4] triazolo [1, 5-a] pyridine;
    183) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6-fluoro-1H-indol-2-yl) pyrazolo [1, 5-a] pyrimidine;
    184) methyl (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carboxylate;
    185) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indole-5-carboxylic acid;
    186) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -1H-indol-6-ol;
    187) (S) -2- (5- ( (R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6, 7, 8-tetrahydro- [1, 2, 4] triazolo [1, 5-a] pyridin-7-ol;
    188) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -3, 4, 6, 7-tetrahydropyrano [3, 4-d] imidazole;
    189) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [4, 5-c] pyridine;
    190) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -4, 5, 6, 7-tetrahydrothiazolo [5, 4-c] pyridine;
    191) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydrothiazolo [5, 4-c] pyridine-5 (4H) -carboxamide;
    192) (R) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6, 7-dihydro-4H-pyrano [4, 3-d] thiazole;
    193) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (5, 6-dimethoxy-1H-benzo [d] imidazol-2-yl) pyrazolo [1, 5-a] pyrimidin-2-amine;
    194) (R) -2- (2-amino-5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5-methoxy-1H-benzo [d] imidazole-6-carbonitrile;
    195) (R) -2- (5- (2- (2-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
    196) (R) -2- (5- (2- (3-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
    197) (S) -2- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
    198) (R) -2- (5- (2- (4-fluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -6-methoxy-1H-benzo [d] imidazole-5-carbonitrile;
    199) (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (6, 7-dimethoxyimidazo [1, 2-a] pyridin-2-yl) pyrazolo [1, 5-a] pyrimidine; or
    200) (R) -3- (5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1, 5-a] pyrimidin-3-yl) -5, 6-dihydro-8H- [1, 2, 4] triazolo [3, 4-c] [1, 4] oxazine.
  38. A pharmaceutical composition comprising a compound of any one of claims 1-37, or an isomeride、 a pharmaceutically acceptable salt or a stereoisomer thereof, and at least one pharmaceutically acceptable carrier or excipient.
  39. A method of inhibiting various different forms of Trk, including wildtype TrkA, TrkB and TrkC, the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, and TrkC G623R, said method comprising administering to a patient a compound of any one of claims 1-37, or a pharmaceutically acceptable salt or an isomeride thereof.
  40. A method of treating a disease associated with inhibition of Trk, including wildtype TrkA, TrkB and TrkC, the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589L, and TrkC G623R, said method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of claims 1-37, or a pharmaceutically acceptable salt or isomeride thereof.
  41. The method of claim 40, wherein the disease is mammary analogue secretory carcinoma (MASC) of the salivary glands, infantile fibrosarcoma, spitz tumors, colon cancer, gastric cancer, thyroid cancer, lung cancer, leukemia, pancreatic cancer, melanoma, brain cancer, renal cancer, prostate cancer, ovarian cancer or breast cancer.
  42. The method of claim 41, wherein the thyroid cancer is papillary thyroid cancer, the brain cancer is pontine glioma, the renal cancer is congenital mesoblastic nephroma, the breast cancer is secretory breast carcinoma.
  43. Use of the pharmaceutical composition of claim 38, or the compound of any one of claims 1-37 for the preparation of a medicament.
  44. The use of claim 43, wherein the medicament is used for the treatment or prevention of cancer.
  45. The use of claim 44, wherein the cancer is mammary analogue secretory carcinoma (MASC) of the salivary glands, infantile fibrosarcoma, spitz tumors, colon cancer, gastric cancer, thyroid cancer, lung cancer, leukemia, pancreatic cancer, melanoma, brain cancer, renal cancer, prostate cancer, ovarian cancer or breast cancer.
  46. The use of claim 45, wherein the thyroid cancer is papillary thyroid cancer, the brain cancer is pontine glioma, the renal cancer is congenital mesoblastic nephroma, the breast cancer is secretory breast carcinoma.
  47. The use of claim 43, wherein the medicament is used as an inhibitor of Trk.
  48. The use of claim 47, wherein the Trk is wildtype TrkA, TrkB, TrkC, or the TrkA G595R, the TrkA G667C, the TrkA A608D, the TrkA F589, or TrkC G623R..
  49. A method of enhancing, stimulating and/or increasing the immune response in a patient, said method comprising administering to the patient in need thereof a therapeutically effective amount of a compound of any one of claims 1-37, or a pharmaceutically acceptable salt or an isomeride thereof.
PCT/CN2019/123719 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of WO2020114499A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
EA202191318A EA202191318A1 (en) 2019-05-09 2019-12-06 TYROSINE KINASE INHIBITORS, COMPOSITIONS AND METHODS OF THEIR APPLICATION
CA3122136A CA3122136A1 (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of
EP19892359.1A EP3891152A4 (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of
SG11202105881RA SG11202105881RA (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of
AU2019394520A AU2019394520A1 (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of
MX2021006619A MX2021006619A (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of.
CN201980077490.6A CN113166156B (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods
JP2021531534A JP2022510380A (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitor, composition and method thereof
KR1020217018370A KR20210124961A (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods
US17/311,105 US20210395256A1 (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of
BR112021010930-7A BR112021010930A2 (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and their procedures
IL283599A IL283599A (en) 2018-12-07 2021-06-01 Tyrosine kinase inhibitors, compositions and methods there of
ZA2021/03858A ZA202103858B (en) 2018-12-07 2021-06-04 Tyrosine kinase inhibitors, compositions and methods there of

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2018119895 2018-12-07
CNPCT/CN2018/119895 2018-12-07
CNPCT/CN2019/086204 2019-05-09
CN2019086204 2019-05-09

Publications (1)

Publication Number Publication Date
WO2020114499A1 true WO2020114499A1 (en) 2020-06-11

Family

ID=70973421

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/123719 WO2020114499A1 (en) 2018-12-07 2019-12-06 Tyrosine kinase inhibitors, compositions and methods there of

Country Status (13)

Country Link
US (1) US20210395256A1 (en)
EP (1) EP3891152A4 (en)
JP (1) JP2022510380A (en)
KR (1) KR20210124961A (en)
CN (1) CN113166156B (en)
AU (1) AU2019394520A1 (en)
BR (1) BR112021010930A2 (en)
CA (1) CA3122136A1 (en)
IL (1) IL283599A (en)
MX (1) MX2021006619A (en)
SG (1) SG11202105881RA (en)
TW (1) TW202033526A (en)
WO (1) WO2020114499A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020224626A1 (en) * 2019-05-08 2020-11-12 浙江同源康医药股份有限公司 Compound used as kinase inhibitor and application thereof
WO2021249450A1 (en) * 2020-06-11 2021-12-16 贝达药业股份有限公司 Salt form, crystal form, pharmaceutical composition and use of tyrosine kinase inhibitor
WO2022060421A1 (en) * 2020-09-17 2022-03-24 Arog Pharmaceuticals, Inc. Crenolanib for treating trk kinase associated proliferative disorders
CN114315899A (en) * 2020-09-30 2022-04-12 上海美迪西生物医药股份有限公司 3- (aromatic benzimidazolyl) pyrazolopyrimidine derivative and application thereof
WO2022095909A1 (en) * 2020-11-03 2022-05-12 上海瑶琪生物科技有限公司 Compound used as ntrk inhibitor and application thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112979654B (en) * 2019-12-16 2024-03-19 赛诺哈勃药业(成都)有限公司 Heteroaryl fused ring compounds, preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2767648A1 (en) * 2009-07-09 2011-01-13 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
WO2016097869A1 (en) * 2014-12-15 2016-06-23 Cmg Pharmaceutical Co., Ltd. Fused ring heteroaryl compounds and their use as trk inhibitors
WO2018077246A1 (en) * 2016-10-28 2018-05-03 正大天晴药业集团股份有限公司 Amino pyrazolopyrimidine compound used as neurotrophic factor tyrosine kinase receptor inhibitor

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ567151A (en) * 2005-10-06 2012-03-30 Schering Corp Pyrazolo [1,5 -A] pyrimidine derivatives and their use for inhibiting Checkpoint kinases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2767648A1 (en) * 2009-07-09 2011-01-13 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
WO2016097869A1 (en) * 2014-12-15 2016-06-23 Cmg Pharmaceutical Co., Ltd. Fused ring heteroaryl compounds and their use as trk inhibitors
WO2018077246A1 (en) * 2016-10-28 2018-05-03 正大天晴药业集团股份有限公司 Amino pyrazolopyrimidine compound used as neurotrophic factor tyrosine kinase receptor inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3891152A4 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020224626A1 (en) * 2019-05-08 2020-11-12 浙江同源康医药股份有限公司 Compound used as kinase inhibitor and application thereof
CN112867717A (en) * 2019-05-08 2021-05-28 浙江同源康医药股份有限公司 Compounds useful as kinase inhibitors and uses thereof
CN112867717B (en) * 2019-05-08 2023-01-10 浙江同源康医药股份有限公司 Compounds useful as kinase inhibitors and uses thereof
WO2021249450A1 (en) * 2020-06-11 2021-12-16 贝达药业股份有限公司 Salt form, crystal form, pharmaceutical composition and use of tyrosine kinase inhibitor
WO2022060421A1 (en) * 2020-09-17 2022-03-24 Arog Pharmaceuticals, Inc. Crenolanib for treating trk kinase associated proliferative disorders
US11524006B2 (en) 2020-09-17 2022-12-13 Arog Pharmaceuticals, Inc. Crenolanib for treating TRK kinase associated proliferative disorders
CN114315899A (en) * 2020-09-30 2022-04-12 上海美迪西生物医药股份有限公司 3- (aromatic benzimidazolyl) pyrazolopyrimidine derivative and application thereof
WO2022095909A1 (en) * 2020-11-03 2022-05-12 上海瑶琪生物科技有限公司 Compound used as ntrk inhibitor and application thereof

Also Published As

Publication number Publication date
TW202033526A (en) 2020-09-16
EP3891152A1 (en) 2021-10-13
AU2019394520A1 (en) 2021-07-01
KR20210124961A (en) 2021-10-15
MX2021006619A (en) 2021-07-07
US20210395256A1 (en) 2021-12-23
CN113166156B (en) 2024-02-27
EP3891152A4 (en) 2022-09-07
CA3122136A1 (en) 2021-06-11
SG11202105881RA (en) 2021-07-29
CN113166156A (en) 2021-07-23
JP2022510380A (en) 2022-01-26
BR112021010930A2 (en) 2021-08-24
IL283599A (en) 2021-07-29

Similar Documents

Publication Publication Date Title
WO2020114499A1 (en) Tyrosine kinase inhibitors, compositions and methods there of
CN114269735B (en) Dihydro-or tetrahydroquinazoline compound, intermediate thereof, preparation method and application
AU2012299899B2 (en) Bicyclic heteroaromatic compounds
US20210040118A1 (en) Immunomodulators, compositions and methods thereof
CN111499634B (en) Quinazoline compound and application thereof in medicine
CA2960275A1 (en) Smyd inhibitors
EP2086979A2 (en) Imidazoý1,2-b¨pyridazine and pyrazoloý1,5-a¨pyrimidine derivatives and their use as protein kinase inhibitors
JP2020504747A (en) Benzimidazole derivatives, methods of preparation and their use
US20170267696A1 (en) Certain protein kinase inhibitors
US20220119411A1 (en) Immunomodulators, compositions and methods thereof
WO2022037568A1 (en) Bicyclic compounds, compositions and use thereof
CN114423751B (en) Novel heterocyclic compounds useful as selective AURORA a inhibitors
US20170137427A1 (en) Imidazo-pyridazine derivatives as casein kinase 1 delta/epsilon inhibitors
CN115362149B (en) SHP2 inhibitor, composition and application thereof
AU2019280356B2 (en) ERK inhibitor and use thereof
JP7201800B2 (en) 3,9-diazaspiro[5,5]undecane-based compounds as inhibitors of FLT3 and AXL
WO2023067546A1 (en) Novel bicyclic heteroaryl derivatives as sos1:kras proteinprotein interaction inhibitors
US20220017520A1 (en) Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine
CA3156625A1 (en) Imidazolidinone compound, preparation method therefor and use thereof
CN118139855A (en) 1, 4-Oxaazepane derivatives and their use
CN113164481A (en) Cycloalkane-1, 3-diamine derivatives

Legal Events

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

Ref document number: 19892359

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021531534

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3122136

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021010930

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2019394520

Country of ref document: AU

Date of ref document: 20191206

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019892359

Country of ref document: EP

Effective date: 20210707

ENP Entry into the national phase

Ref document number: 112021010930

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210604