CN115772170A - Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof - Google Patents

Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof Download PDF

Info

Publication number
CN115772170A
CN115772170A CN202211500443.8A CN202211500443A CN115772170A CN 115772170 A CN115772170 A CN 115772170A CN 202211500443 A CN202211500443 A CN 202211500443A CN 115772170 A CN115772170 A CN 115772170A
Authority
CN
China
Prior art keywords
group
compound
alkyl
cancer
pyrazolo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202211500443.8A
Other languages
Chinese (zh)
Inventor
余聂芳
董金付
夏伟
吴文涛
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Xunuo Pharmaceutical Nanjing Co ltd
Original Assignee
Xunuo Pharmaceutical Nanjing Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Xunuo Pharmaceutical Nanjing Co ltd filed Critical Xunuo Pharmaceutical Nanjing Co ltd
Publication of CN115772170A publication Critical patent/CN115772170A/en
Pending legal-status Critical Current

Links

Images

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a novel pyrazolo [1,5-a ]]Pyridine derivatives, and preparation methods and medical applications thereof. More particularly, the present invention relates to pyrazolo-containing [1,5-a ]]The pyridine derivatives can be used as medicaments for treating proliferative diseases and other diseases related to TRK or abnormal expression of various kinases such as RET, FGHR, PDGFR, VEGFR and the like. The structure is shown in general formula (I), wherein L 1 、L 2 、L 3 、R 1 、R 2 And R 3 The definition is shown in the specification:

Description

Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof
Technical Field
The invention relates to a novel pyrazolo [1,5-a ] pyridine derivative, a preparation method and application thereof. In particular to a novel pyrazolo [1,5-a ] pyridine derivative which can inhibit the growth of tumor strains with high expression of kinases or tumor cell strains with corresponding kinase mutation and a preparation method thereof. In particular, these compounds are useful as agents for treating proliferative disorders, and other diseases associated with abnormal expression of any one or more of TRK, RET, FGHR, PDGFR, VEGFR, and the like. Furthermore, the invention relates to medicaments containing these compounds and to the use of these compounds for the preparation of medicaments.
Background
Tumors are one of the major diseases threatening human health, and society has a great need for the treatment of tumors. With the development of tumor molecular biology and tumor pharmacology, chemotherapy and immunotherapy of tumors have been developed. In various treatments of tumors, small molecule targeted drug therapy of tumors is playing an increasingly important role.
[ NTRK/TRK (Tropomosin receptor kinase) ] is a neurotrophic factor tyrosine kinase receptor. The TRK family mainly comprises 3 members, NTRK1/TRKa, NTRK2/TRKb and NTRK3/TRKc. The complete TRK kinase comprises three parts, namely an extracellular region, a transmembrane region and an intracellular region. The extracellular region of TRK kinase can cause the change of kinase configuration after being combined with corresponding ligand to form dimer. The intracellular region of TRK kinase is autophosphorylated to activate the kinase activity of the TRK kinase, and further activate downstream signal transduction pathways (such as MAPK, AKT, PKC and the like) to generate corresponding biological functions; wherein NGF (nerve growth factor) binds TRKa, BDNF (derived neurotrophic factor) binds TRKb, and NT3 (neurotrophic factor 3) binds TRKc.
A large number of researches show that activation of TRK signal transduction pathway is strongly related to generation and development of tumors, and activated TRK signal protein is found in neurocytoma, prostatic cancer, breast cancer and the like. The discovery of various TRK fusion proteins in recent years has further shown their biological function of promoting tumorigenesis (Sharan K Bangal, mark Andrews, bruce M. Bechle, jianwei Bian, james Billand, david C Blakemore, john Bragza, peter J. Bungeay, matthew S. Corbett, cirar N Cronin, jingong Jean Cui, recbecca Dias, neil J Flanagan, samantha E Greasley, rachel Grimley, kim James, eric Johnson, linda kitchen, michelle L aus, inragan Mcipine, asao Nagagata, man Ninkouvic, steyoni, sagnat, saw, wallace L, walsh L lake L, moisture content J. Pat. J.8. And J.J.J.P.J.J.P.J.P.J.J.P.J.J.P.J.J.P.J.J..
In recent years, TRK fusion proteins have become an effective anti-cancer target and research hotspot, for example, as described in larotectornib (1) LISA jarvis. Bayer, loxo to develoop TRK inhibitors.c & EN,20November 2017,11; 2) TRK kinase inhibitors represented by (WO 2010048314), entrectinib (WO 2009013126A 1) and the like have already entered clinical applications. More TRK kinase inhibitors are already in various stages of the clinic. WO2012116217, WO2010033941, JP 2018044010A, MX 2017007748A, US2017057948A1 and the like disclose TRK kinase inhibitors having different mother nuclei. In view of the importance of the physiological functions of TRK, the TRK inhibitor with stronger activity and wider applicable gene fusion range attracts wide interest. In particular, there is still no social need for TRK inhibitors that inhibit not only TRK a, b and C, but also their mutated forms (e.g., G595R, G667C, a608D, F589L, G623R).
Figure BDA0003967308590000021
In another aspect, the transfection Rearrangement (RET) is a nerve growth factor receptor tyrosine kinase; abnormal RET kinase activity is associated with numerous tumors. Thus, RET is also an anti-tumor target that is highly valued.
RET is a neuronal growth factor receptor tyrosine kinase. RET kinase knockout mice lack enteric neurons and have other nervous system abnormalities, suggesting that functional RET kinase protein products are required for development. The study of the patient population with congenital megacolon disease shows that the mutation frequency of the functional RET is higher with familial and sporadic mutation. Abnormal RET kinase activity is associated with diverse endocrine adenomas (MEN 2A and 2B), familial medullary thyroid tumors (FMTC), papillary Thyroid Carcinomas (PTC) and congenital megacolon disorders (HSCR) (Maria Grazia Borrello, elena Ardini, laura D Locati, angela Greco, lisa lictra & Marco a pieroti (2013), RET inhibition: indications in cancer therapy. Expert Opinion on Therapeutic Targets,17, 4, 403-419). MEN 2A is a cancer syndrome that results from the mutation of the extracellular cysteine-rich region of RET cells leading to the dimerization of disulfide bonds, thereby rendering the activity of the tyrosine kinase under continuous activation (Samuel A. Wells, jr, furio Pacini, bruce G. Robinson, and Massimo Santoro. Multiple Endocrine Neoplasty Type 2and Familial Medulary Thyroid Carcinoma.J. Clin Endocrinol Metab 98. Individuals with such mutations may develop medullary thyroid tumors (MTC), thyroid hyperplasia, and pheochromocytoma. MEN 2B and MEN 2A are similar, but without thyroid hyperplasia, also cause various mucosal ganglions of the lips, tongue and intestinal tract. RET is thought to intervene in tumor initiation by PTC during chromosome recapture. PTC comprises 80% of thyroid tumors (viglititto, g.et al, oncogene,1995, 11.
These facts indicate an ideal therapeutic approach for treating tumors associated with sustained RET activation. The research on RET inhibitors is receiving wide attention and has also gained a rapid development. Among them, RET inhibitors represented by Pralsetinib (WO 2017011776A1Array Loxo 292) and Selpercatinib (WO 2017079140A1 Blu 667) were approved by FDA and marketed in order to treat various related fusion tumors regardless of tumor and succeeded. Thereby encouraging various other development efforts. ((1) Lucille Lopez-Delisle, C é circle Pierre-Eug ne, caroline Louis-Brennetot, didier Surdez, virgine Raynal, sylvain Baulande, valentina Boeva, sandrine Groset E-Lalami, valerie Combaret, michel Peuchmaur6, olivier Deltate tre, isabelle Janoux-Lerosey.activated ALK signals through the ERK-ETV5-RET pathway to drive neuro Kinase. Oncogene (2018) 37
Figure BDA0003967308590000031
As described above, research and development of kinase inhibitors represented by TRK and RET have been successful in treating fused tumors, respectively, and have brought good news to patients. However, the proportion of tumors, which have TRK and RET as the core and are fused with other genes, respectively, is still small (Alexander drug, zishuo I.Hu, gilliane G.Y.Lai & Daniel S.W.Tan Targeting RET-driving receptors: organs from developing a differentiating and cloning patent and Clinical landscapes. Nature Reviews Clinical Oncology volume15,2018, 151-167). Clearly, it would be an effective option if one or more of the kinases, TRK, RET, FGHR, PDGFR, VEGFR, etc., could be effectively inhibited. On the other hand, resistance remains a major problem for almost all kinase targets. Therefore, there remains a great social need in the art for more multi-target kinase inhibitors.
Disclosure of Invention
The invention aims to disclose a novel pyrazolo [1,5-a ] pyridine derivative and a preparation method and application thereof. The compounds can be used for tumors, endocrine disorders, immune system diseases, genetic diseases and neurodegenerative diseases.
In order to achieve the above objects, the inventors have conducted extensive studies and found that a compound represented by the following general formula (I) or a pharmaceutically acceptable salt thereof has an excellent effect of inhibiting kinase activity, particularly inhibiting the activity of any one or more of TRK, RET, FGHR, PDGFR, VEGFR and the like, and is useful as a drug for treating proliferative disorders and other diseases associated with abnormal expression of various kinases.
In order to achieve the purpose, the technical scheme of the invention is as follows:
a pyrazolo [1,5-a ] pyridine derivative which is a compound represented by the general formula (I), and an optical isomer or a pharmaceutically acceptable salt thereof:
Figure BDA0003967308590000032
in the above-mentioned general formula (I),
R 1 selected from: a hydrogen atom, carboxyl, nitro, amino, cyano, acyl, alkyl, alkoxy, alkenyl, alkynyl, halogen, isotope, haloalkenyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl; any of the above groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, heteroalkyl, alkenyl, alkynyl, hydroxyl, hydroxyalkyl, alkoxy, and alkoxyalkyl;
L 1 selected from: covalent bond, -CH 2 -CH 2 -、-CH=CH-、-C≡C-、-NH-、
Figure BDA0003967308590000041
Figure BDA0003967308590000042
Any of the foregoing groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, = O, -CF 3 Alkyl, haloalkyl, hydroxy, alkoxy, and alkoxyalkyl;
L 2 selected from: a covalent bond, alkylene, heteroalkylene, carbonyl, -C (O) -NH-, -alkyl-NH-C (O) -, -alkyl-O-; any of the above groups may each independently be substituted with one or more substituents including hydrogen, isotopes, alkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, heteroaryl;
L 3 selected from: covalent bonds, alkylene, heteroalkylene, carbonyl, -O-, -NH-, -C (O) -NH-, -NH-C (O) -, -alkyl-C (O) -NH-, -alkyl-NH-C (O) -, -alkyl-NH-, -alkyl-O-; any of the foregoing groups, independently of each other, may be substituted with one or more substituents including hydrogen, isotopes, alkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, heteroaryl;
R 2 selected from: a hydrogen atom, an alkyl group, a heteroalkyl group, an alkenyl group, an alkynyl group, an arylalkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, a heteroarylalkyl group, a heterocycloalkyl group, an alkylether group, a heteroalkylether group, an arylalkylether group, a cycloalkether group, an aryl ether group, a heteroaryletheryl group, a heteroarylalkylether group, a heterocycloalkyl group, a carboxyl group, a carboxyalkylaminocarbonyl group, a cycloalkylaminocarbonyl group, a heteroarylaminocarbonyl group; any of the above groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl.
R 3 Selected from: a hydrogen atom, an alkyl group, a heteroalkyl group, an alkenyl group, an alkynyl group, an arylalkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, a heteroarylalkyl group, a heterocycloalkyl group, an alkyl ether group, a heteroalkyl ether group, an arylalkyl ether group, a cycloalkyl ether group, an aryl ether group, a heteroarylether group, a heteroarylalkyl ether group, a heterocycloalkyl ether group, an alkylamino group, a heteroalkylamino group, an arylalkylamino group, a cycloalkylamino group, an arylamino group, a heteroarylamino group, a heteroarylalkylamino group, a heterocycloalkylamino group, a carboxyl group, a carboxyalkylaminocarbonyl group, a cycloalkylaminocarbonyl group, a heteroarylaminocarbonyl group; any of the foregoing groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, and the like,Benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxyl, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl.
In certain embodiments, in formula (I), R 1 Is hydrogen atom, carboxyl, nitro, amino, cyano, acyl, alkyl, alkoxy, alkenyl, alkynyl, halogen, isotope, haloalkenyl, heteroalkyl, aryl, heteroaryl; in the above groups, each may be unsubstituted or substituted with one or more substituents including: halogen, isotope, = O, -CF 3 Alkyl, alkenyl, alkynyl, carboxyl, hydroxyl, hydroxyalkyl, alkoxy, alkoxyalkyl, -C.ident.N.
In still other embodiments, in formula (I), L 3 Is selected from
Figure BDA0003967308590000051
O-CH 2 -R 4 (ii) a Wherein R is 4 Is selected from
Figure BDA0003967308590000052
In certain embodiments, in formula (I), R 3 Selected from the group consisting of C1-C5 alkyl, heterocyclyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl; any of the above groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, alkyl.
In certain other embodiments, in formula (I), L 1 Selected from: -CH 2 -CH 2 -、-CH=CH-、-C≡C-。
In certain embodiments, in formula (I), L 2 Selected from the group consisting of: alkylene, heteroalkylene, carbonyl, -C (O) -NH-, -alkyl-NH-C(O)-。
In certain other embodiments, of formula (I), L 2 Selected from: pyridine, substituted pyridine, pyrimidine, substituted benzene ring, -CO-NH-, -C (O) -, -CH 2 -NH-、
Figure BDA0003967308590000053
Figure BDA0003967308590000054
-C(CH 2 ) -NH-, -CO-N (OH) -; the alkyl is C1-C5 straight chain or branched chain alkyl; the substituent of the substituted pyridine is C1-C5 straight chain or branched chain alkyl; the substituent of the substituted pyrimidine is C1-C5 straight-chain or branched alkyl or amino.
In certain embodiments, in formula (I), R 2 Selected from: C1-C8 alkyl,
Figure BDA0003967308590000055
Amino group,
Figure BDA0003967308590000056
Figure BDA0003967308590000061
Benzene ring, substituted benzene ring, picoline,
Figure BDA0003967308590000062
CO-NH-R 7
Figure BDA0003967308590000063
Pyrimidine, substituted pyrimidine,
Figure BDA0003967308590000064
Pyridine, substituted pyridine,
Figure BDA0003967308590000065
Figure BDA0003967308590000066
Figure BDA0003967308590000067
-CH 2 -CN、
Figure BDA0003967308590000068
Figure BDA0003967308590000069
Figure BDA00039673085900000610
-NH-OH、
Figure BDA0003967308590000071
R 5 Selected from hydrogen,
Figure BDA0003967308590000072
R 6 Selected from hydrogen,
Figure BDA0003967308590000073
Benzene ring, substituted benzene ring,
Figure BDA0003967308590000074
A pyrimidine;
the substituted benzene ring contains 1-3 substituents, and the substituents of the substituted benzene ring are selected from amino, C1-C5 alkyl, halogen, trifluoromethyl, trichloromethyl, tribromomethyl, trifluoroethyl, trifluoropropyl, trifluoromethyl, and the like,
Figure BDA0003967308590000075
Methyl ether group, ethyl ether group, propyl ether group,
Figure BDA0003967308590000076
Trifluoromethyl ether group, trifluoroethyl ether group, trifluoropropyl ether group,
Figure BDA0003967308590000077
-C≡CH、-CN;
R 7 Selected from C1-C8 alkyl;
the substituted pyrimidine contains 1-3 substituents, and the substituents of the substituted pyrimidine are selected from amino, C1-C5 alkyl and halogen;
the substituted pyridine contains 1-3 substituents, and the substituents of the substituted pyridine are selected from
Figure BDA0003967308590000078
C1-C8 alkyl,
Figure BDA0003967308590000081
Difluorophenyl rings, methyl ether groups;
R 8 selected from hydrogen, substituted pyridines, C1-C8 alkyls.
In certain embodiments, the substituted phenyl ring is selected from toluene, ethylbenzene, para-fluorotoluene.
In certain embodiments, the pyrazolo [1,5-a ] pyridine derivative may also be a compound represented by the general formula (I):
13.
Figure BDA0003967308590000082
in the above-mentioned general formula (I),
R 1 selected from: a hydrogen atom, carboxyl, nitro, amino, cyano, acyl, alkyl, alkoxy, alkenyl, alkynyl, halogen, isotope, haloalkenyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl; any of the above groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Alkyl halide, alkyl halideAlkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, and alkoxyalkyl;
L 3 selected from: covalent bonds, alkylene, heteroalkylene, carbonyl, O, NH, CO-NH, NH-CO, alkyl-CO-NH, alkyl-NH-CO, alkyl-NH, alkyl-O; any of the above groups may each independently be substituted with one or more substituents including hydrogen atom, alkyl group;
R 3 selected from: a hydrogen atom, an alkyl group, a heteroalkyl group, an alkenyl group, an alkynyl group, an arylalkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, a heteroarylalkyl group, a heterocycloalkyl group, an alkyl ether group, a heteroalkyl ether group, an arylalkylether group, a cycloalkylether group, an aryl ether group, a heteroarylether group, a heteroarylalkyl ether group, an alkylamino group, a heteroalkylamino group, an arylalkylamino group, a cycloalkylamino group, an arylamino group, a heteroarylamino group, a heteroarylalkylamino group, a heterocycloalkylamino group, a carboxyl group, a carboxyalkylaminocarbonyl group, a cycloalkylaminocarbonyl group, a heteroarylaminocarbonyl group; any of the above groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl;
wherein L is 1 -L 2 -R 2 Selected from:
Figure BDA0003967308590000091
wherein R is 9 ,R 10 ,R 11 And R 12 Independently selected from: hydrogen atom, alkyl, heteroalkyl, alkenyl, alkynyl, arylalkylA group, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, alkyl ether group, heteroalkyl ether group, arylalkyl ether group, cycloalkyl ether group, aryl ether group, heteroarylether group, heteroarylalkyl ether group, heterocycloalkyl ether group, alkylamino group, heteroalkyl amino group, arylalkylamino group, cycloalkyl amino group, arylamino group, heteroarylamino group, heteroarylalkylamino group, heterocycloalkylamino group, carboxyl group, carboxyalkylaminocarbonyl group, cycloalkylaminocarbonyl group, heteroarylaminocarbonyl group; any of the foregoing groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxyl, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl.
In certain embodiments, in general formula (I), the structure may be selected from one of the following structures, or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof:
Figure BDA0003967308590000092
Figure BDA0003967308590000101
Figure BDA0003967308590000111
Figure BDA0003967308590000121
Figure BDA0003967308590000131
Figure BDA0003967308590000141
Figure BDA0003967308590000151
Figure BDA0003967308590000161
the invention also discloses the pyrazolo [1,5-a ] as described above]Process for the preparation of pyridine derivatives, L 2 = CO-NH, the preparation method comprises:
s1, selecting I as a raw material, and removing methyl under the action of a catalyst to obtain a compound II;
s2, reacting the compound II with a boron compound III under catalysis to obtain IV;
s3, converting the compound IV into a compound V under the action of N-phenyl bis (trifluoromethanesulfonimide);
s4, condensing the compound VI and the compound VII to obtain a compound VIII;
s5, reacting the compound VIII with the compound V to obtain a compound IX, namely a target compound shown as the general formula (I);
Figure BDA0003967308590000171
in certain embodiments, compound I-1 is:
Figure BDA0003967308590000172
the synthesis method of the compound I-1 comprises the following steps:
A. taking X as a raw material, and carrying out acylation and ammoniation to obtain XIV;
B. under the action of acid, XIV cyclizes to obtain a compound XVI;
C. introducing formyl on the compound XVI, converting the formyl into a nitrile group through hydroxylamination, and removing methyl under the action of a catalyst to obtain a compound I-1;
Figure BDA0003967308590000173
in certain embodiments, L 1 = CH or-C ≡ C-, L 2 -R 2 For substituted phenyl radicals, said pyrazolo [1, 5-a)]The preparation method of the pyridine derivative comprises the following steps:
1) Taking an iodo compound XIX as a raw material, and reacting with XX under the action of a catalyst to obtain a compound XXI;
2) Reacting a compound XXI in an alkaline methanol solution to obtain XXII;
3) XXII reacts with compound V to give compound XXIII;
Figure BDA0003967308590000181
the invention also includes a pharmaceutical composition containing the compound shown in the general formula (I) or a stereoisomer, a geometric isomer, a tautomer, an N-oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof:
the invention also comprises the combination of the compound shown in the general formula (I) or the pharmaceutically acceptable salt thereof and other one or more medicaments.
The invention also includes the use of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition caused by, associated with or associated with disruption of cell proliferation and/or angiogenesis.
The invention also comprises the application of the compound shown in the general formula (I) or the pharmaceutically acceptable salt thereof or the pharmaceutical composition for inhibiting the activity of the kinase.
In certain embodiments, the kinase-inhibiting activity is inhibition of the activity of TRK or one or more of the various kinases RET, RAF, FGHR, PDGFR, VEGFR.
In certain embodiments, the kinase inhibitory activity is inhibition of TRK activity.
In certain embodiments, the kinase inhibitory activity is inhibition of RET activity.
The invention also includes a method for treating a condition in a patient caused by, associated with or accompanied by disruption of cell proliferation and/or angiogenesis, the method comprising administering to the patient a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as described above.
The invention also includes a method of treating a condition treatable by inhibiting a kinase in a subject, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as described above.
In certain embodiments, the disorder is selected from: proliferative diseases such as non-small cell lung cancer, renal cell carcinoma, gastric cancer, hepatocellular carcinoma, colorectal cancer, medullary thyroid carcinoma, follicular thyroid carcinoma, undifferentiated thyroid carcinoma, papillary thyroid carcinoma, brain tumor, peritoneal cavity cancer, solid tumors, other lung cancers, renal cell carcinoma, gastric cancer, head and neck cancer, glioma, neuroblastoma, von Hipple-Lindau syndrome and renal tumors, breast cancer, fallopian tube cancer, ovarian cancer, transitional cell cancer, prostate cancer, cancer at the junction of esophagus and stomach, biliary tract cancer and adenocarcinoma, and any malignancy with increased kinase activity of any one or more of TRK, RET, FGHR, PDGFR, VEGFR, and the like; neurodegenerative diseases, including: huntington's disease, polyglutamine disease, parkinson's disease, alzheimer's disease, seizures, striatonigral degeneration, progressive supranuclear palsy, torsion dystonia, spastic torticollis and dyskinesia, familial tremor, gilles de la tourette's syndrome, diffuse lewy body disease, pick's disease, intracranial hemorrhage, primary lateral sclerosis, spinal muscular atrophy, amyotrophic lateral sclerosis, hypertrophic interstitial polyneuropathy, retinitis pigmentosa, hereditary optic atrophy, hereditary spastic paraplegia, progressive ataxia and Shy-Drager syndrome; a metabolic disease comprising: type 2 diabetes; an ocular degenerative disease comprising: glaucoma, age-related macular degeneration, rubeosis iridis glaucoma: diseases involving angiogenesis, including: cancer, psoriasis; a psychological disorder, comprising: bipolar disorder, schizophrenia, mania, depression and dementia; cardiovascular diseases include: heart failure, restenosis and arteriosclerosis; fibrotic diseases, including: liver fibrosis, cystic fibrosis and vascular fibromyalgia: infectious diseases, including: and (4) fungal infection. For example: candida albicans, bacterial infection, viral infection. For example: herpes simplex, protozoal infections, such as: malaria, leishmania infection, trypanosoma brucei infection, toxoplasmosis and coccidiosis, and hematopoietic disorders including: marine anemia, and sickle cell anemia.
In certain embodiments, in the above-described methods, the patient is undergoing surgery or radiation therapy and the compound is administered to the patient concomitantly with, or prior to, or after the surgery or radiation therapy.
Drawings
FIG. 1 is a graph of the in vivo antitumor activity of compounds of the present invention in a TT model.
Detailed Description
The groups of the compound represented by the general formula (I) of the present invention are defined as follows.
"halogen" refers to fluorine, chlorine, bromine and iodine.
"= O" means oxo.
“CF 3 "refers to trifluoromethyl.
"CO-NH" is "-amide-".
"NH-CO" is an "aminoacyl".
"carbonyl" means
Figure BDA0003967308590000191
"alkyl" when taken as a group or part of a group refers to a straight or branched chain aliphatic hydrocarbon radical in which none or one or more (preferably 1,2 or 3) of the carbon atoms are replaced by oxygen, nitrogen, phosphorus, boron, selenium, silicon or sulfur atoms (preferably oxygen, sulfur or nitrogen).
"alkyl" is preferably: C1-C14 alkyl, more preferably C1-C10 alkyl; most preferred is C1-C6 alkyl, unless otherwise indicated. Examples of C1-C6 alkyl groups include, but are not limited to: methyl, ethyl, n-propyl, 2-propyl, n-butyl, isobutyl, tert-butyl, hexyl and the like.
"alkyl" is most preferably selected from C1-C6 unless otherwise indicated. Examples of linear or and branched C1-C6 alkyl groups include, but are not limited to: methyl, ethyl, n-propyl, 2-propyl, n-butyl, isobutyl, tert-butyl, hexyl and the like.
"cycloalkyl" refers to a saturated or partially saturated monocyclic, fused or spiro carbocyclic ring. Preferably a ring consisting of 3 to 9 carbon atoms. Examples include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
"heteroalkyl" refers to a group formed by replacing one or more (preferably 1,2 or 3) carbon atoms in a straight or branched chain alkyl group with an oxygen, nitrogen, phosphorus, boron, selenium, silicon or sulfur atom (preferably oxygen, sulfur or nitrogen). The heteroalkyl group having 2 to 14 atoms is preferable, the number of atoms is more preferable from 2 to 8, and the number of atoms is particularly preferable from 2 to 6. Heteroalkyl groups include, but are not limited to: ethers, thioethers, alkyl esters, secondary alkyl amines, tertiary alkyl amines, alkylsulfonic acids, nitriles, isonitriles, cyanates, thiocyanates, isocyanates, isothiocyanates, alkylnitriles and the like, and specific examples thereof include: methoxy, trifluoromethoxy, ethoxy, N-propoxy, isopropoxy, tert-butoxy, methoxymethyl, ethoxymethyl, methoxyethyl, methylamino, ethylamino, dimethylamino, diethylamino, isopropylethylamino, methyl-aminomethyl, ethylaminomethyl, di-isopropylaminoethyl, enol ether, dimethylaminomethyl, dimethylaminoethyl, acetyl, propionyl, butyryloxy, acetoxy, methoxycarbonyl, ethoxy-carbonyl, N-ethyl-N-methylcarbamoyl or N-methylcarbamoyl.
"heterocycloalkyl" means a group formed by replacement of one or more (preferably 1,2 or 3) carbon atoms in a "cycloalkyl" group as defined above by an oxygen, nitrogen, phosphorus, boron, selenium, silicon or sulfur atom (preferably oxygen, sulfur or nitrogen). Wherein heterocycloalkyl and alkyl moieties are defined herein. Preferably containing 1-3 heteroatoms. Preferred rings are 3-14 membered rings (i.e., 3-14 membered heterocycloalkyl groups), and more preferred rings are 4-7 membered rings (i.e., 4-7 membered heterocycloalkyl groups). Heterocycloalkyl groups include, but are not limited to: pyrrolidinyl, dihydropyrrolyl, tetrahydropyrrolyl, dihydropyrazolyl, piperidinyl, morpholinyl, tetrahydrofuryl, tetrahydrothiofuranyl, tetrahydropyranyl, oxetanyl, aziridinyl, or 2-pyrazolinyl, as well as lactams, lactones, cyclic imines, and cyclic anhydrides, and the like. The heterocycloalkyl group may be substituted with one or more substituents.
"Heterocycloalkylalkyl" means: (heterocycloalkyl-alkyl) -group. Wherein heterocycloalkyl and alkyl moieties are as defined herein. Heterocycloalkyl alkyl groups include, but are not limited to: (2-tetrahydrofuryl) methyl, (2-tetrahydrothiofuranyl) methyl, and the like.
"alkylamino" includes both monoalkylamino and dialkylamino groups unless otherwise indicated. "monoalkylamino" refers to: (alkyl-NH) -; "dialkylamino" refers to: ((alkyl) 2 N) -. Wherein alkyl is as defined herein. The alkylamino group is preferably a C1-C6 alkylamino group. "C1-C6 alkylamino" refers to an amino group substituted with "C1-C6 alkyl", and examples thereof include, but are not limited to: methylamino, ethylamino, isopropylamino, N- (diethyl) amino, and the like.
"Heteroalkylamino" means: both mono-heteroalkylamino and di-heteroalkylamino unless otherwise indicated. Mono-heteroalkylamino refers to: (heteroalkyl-) NH-; di-heteroalkylamino means (heteroalkyl) 2 The radical of N-. Wherein the "heteroalkyl" moiety is defined in the relevant section hereinAnd (4) dividing.
"aminoalkyl" means: (amino-alkyl) -group. Wherein the "alkyl" moiety is as defined herein. The aminoalkyl radical is preferably an aminoC 1-C6 alkyl radical. "amino-C1-C6 alkyl" refers to C1-C6 alkyl substituted with "amino", and examples thereof include, but are not limited to: aminoethyl, 1-aminopropyl, 2-aminopropyl and the like.
"arylamino" includes both mono-arylamino and di-arylamino unless otherwise indicated. Mono-arylamino means: (aryl-) NH-; by di-arylamino is meant (aryl) 2 The radical of N-. Wherein the definition of the "aryl" moiety is found in the relevant section herein.
"acyl" includes both (alkyl-CO) -and (aryl-CO) -groups unless otherwise indicated. Wherein the "alkyl" or "aryl" moiety is as defined herein. Examples of acyl groups include, but are not limited to: acetyl, propionyl, isobutyryl, benzoyl, and the like.
"amido" includes both (alkyl-CONH) -and (aryl-CONH) -groups unless otherwise indicated. Wherein the "alkyl" or "aryl" moiety is defined herein. Examples of amide groups include, but are not limited to: acetylamino, propionylamino, butyrylamino, isobutyrylamino, benzoylamino and the like.
"alkenyl" as a group or part of a group means an aliphatic hydrocarbon group containing at least one carbon-carbon double bond, and can be straight or branched. Preferably C2-C14 alkenyl, more preferably C2-C12 alkenyl, most preferably C2-C6 alkenyl. The group may contain multiple double bonds in its backbone and may each be in its conformation E or Z. Examples of alkenyl groups include, but are not limited to: vinyl, propenyl, and the like. The "alkenyl group" in the present invention refers to a group in the case where the "alkenyl group" defined above is a chain.
"alkynyl" as a group or part of a group refers to an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond, and can be straight or branched. Preferably C2-C14 alkynyl, more preferably C2-C12 alkynyl, most preferably C2-C6 alkynyl. Examples of such alkynyl groups include, but are not limited to: ethynyl, prop-1-yn-1-yl, prop-2-yn-1-yl, but-1-yn-1-yl, but-3-yn-1-yl, 1-methylprop-2-yn-1-yl, pent-1-yn-1-yl, pent-4-yn-1-yl, hex-1-yn-1-yl, hex-5-yn-1-yl and the like.
"alkoxy" refers to the radical of (alkyl-O) -. Wherein the "alkyl" moiety is defined herein. The alkoxy group is preferably a C1-C8 alkoxy group, more preferably a C1-C6 alkoxy group. Examples of such alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, 1-methylethoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, n-pentoxy, isopentoxy, neopentoxy, 1-methylbutoxy, 1-ethylpropoxy, n-hexoxy, isohexoxy, 3-methylpentoxy, 2-methylpentoxy, 1-methylpentoxy, 3-dimethylbutoxy, 2-dimethylbutoxy, 1-dimethylbutoxy, 1, 2-dimethylbutoxy, 1, 3-dimethylbutoxy, 2, 3-dimethylbutoxy, 1-ethylbutoxy, 2-ethylbutoxy and the like. The "alkoxycarbonyl group" represents a group in which the "alkoxy group" defined above is bonded to a carbonyl group, and examples thereof include a methoxycarbonyl group and an ethoxycarbonyl group.
"alkenyloxy" refers to the radical of (alkenyl-O) -. Wherein the "alkenyl" moiety is defined herein. Preferably C2-C6 alkenyloxy.
"alkynyloxy" refers to the radical of (alkynyl-O) -. Wherein the "alkynyl" moiety is as defined herein. Preferably C2-C6 alkynyloxy.
"alkoxycarbonyl" refers to the radical of (alkyl-O-C (O)) -. Wherein alkyl is as defined herein. Preferred alkyl groups are C1-C6 alkyl groups. Examples include, but are not limited to: methoxycarbonyl, ethoxycarbonyl, and the like.
"Alkylsulfinyl" refers to the group of (alkyl-S (O)) -. Wherein the "alkyl" moiety is defined herein. Preferably C1-C6 alkylsulfinyl. Examples of alkylsulfinyl groups include, but are not limited to: methylsulfinyl, ethylsulfinyl, and the like.
"alkylsulfonyl" means (alkyl-S (O) 2 -O) -. Wherein the "alkyl" moiety is as defined herein. C1-C6 alkylsulfonyl is preferred. The "C1-C6 alkylsulfonyl" refers to a sulfonyl group substituted with a "C1-C6 alkyl", and examples thereof include methylsulfonyl, ethylsulfonyl, n-propylsulfonyl, isopropylsulfonyl, n-butylsulfonyl, isobutylsulfonyl, sec-butylsulfonyl, tert-butylsulfonyl, n-pentylsulfonyl, isopentylsulfonyl, neopentylsulfonyl, and tert-pentylsulfonyl groups.
"alkylaminocarbonyl" refers to alkylamino-carbonyl groups. Wherein the "alkylamino" moiety is as defined herein.
"cycloalkylalkyl" refers to a cycloalkyl-alkyl group. Wherein cycloalkyl and alkyl moieties are defined herein. Monocycloalkyl groups include, but are not limited to: cyclopropylmethyl, cyclopentylmethyl, cyclohexylmethyl, cycloheptylmethyl and the like.
"Heterocycloalkenyl" refers to a group containing at least one double bond in the "heterocycloalkyl" group as defined above.
"aryl" as a group or part of a group means: (1) aromatic monocyclic or fused aromatic hydrocarbon ring groups; preferably 6-12 membered aryl (also denoted C6-C12 aryl), more preferably 6-10 membered aryl (also denoted C6-C10 aryl), examples of which include but are not limited to: phenyl, naphthyl, anthryl and phenanthryl; or (2) may link partially saturated carbocycles, for example: the phenyl and C5-7 cycloalkyl or C5-7 cycloalkenyl groups are fused to each other to form a cyclic structure. Examples include, but are not limited to: tetrahydronaphthyl, indenyl or hydroindenyl and the like. The aryl group may be substituted with one or more substituents.
"arylalkenyl" means: (aryl-alkenyl) -. Wherein the "aryl" and "alkenyl" moieties are as defined herein. Exemplary arylalkenyl groups include, but are not limited to: phenylpropenyl and the like.
"aralkyl" means: (aryl-alkyl) -. Wherein the aryl and alkyl moieties are defined herein. Exemplary aralkyl groups include, but are not limited to: benzyl, phenethyl, 1-naphthylmethyl, and the like.
"cycloalkenyl" means a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond and preferably having 5 to 10 carbon atoms per ring. Exemplary monocyclic cycloalkenyl rings include, but are not limited to: cyclopentene, cyclohexene or cycloheptene. The cycloalkene group may be substituted with one or more substituents.
"heteroaryl" means a monocyclic or fused polycyclic aromatic heterocyclic group, which is preferably an aromatic group having one or more (preferably 3 to 14, more preferably 5 to 10, particularly preferably 5 or 6) carbon atoms, and one or more (preferably 1,2, 3 or 4) oxygen, nitrogen, phosphorus or sulfur ring atoms (preferably O, S or N) as ring-forming atoms, preferably the aromatic group is a 4-15-membered heteroaryl group, more preferably a 5-7-membered heteroaryl group. Examples of said heteroaryl group may be, for example: furyl, thienyl, pyrrolyl, pyrazolyl, triazolyl, thiazolyl, pyridyl, pyrimidinyl, pyrazinyl, indolyl, benzimidazolyl, pyridyl, imidazolyl, 3-phenylpyrrolyl, thiazolyl-oxazolyl, tetrazolyl, isoxazolyl, indazolyl, pyridazinyl, quinolyl, purinyl, carbazolyl, acridinyl, pyrimidinyl, 2,3' -bifuryl, and isoquinolyl.
"heteroarylalkyl" means: (heteroaryl-alkyl) -. Wherein the heteroaryl and alkyl moieties are as defined herein. Exemplary heteroarylalkyl groups include, but are not limited to: 2-furylmethyl, 3-furylmethyl, 2-pyridylmethyl and the like.
"alkyl ether" means: (alkyl) -O-. Wherein the "alkyl" moiety is as defined herein.
"Cycloalkaneether group" means: (cycloalkyl) -O-. Wherein the "cycloalkyl" moiety is defined herein.
"Heteroalkylether" means: (heteroalkyl) -O-. Wherein the "heteroalkyl" moiety is as defined herein.
"aryl ether" means: (aryl) -O-. Wherein the "aryl" moiety is as defined herein.
"arylalkylether group" means: (aryl-alkyl) -O-. Wherein the "aryl" and "alkyl" moieties are as defined herein.
"heteroaryl aryl ether" means: (heteroaryl) -O-. Wherein the "heteroaryl" moiety is as defined herein.
"heteroarylalkylether group" means: (heteroaryl-alkyl) -O-. Wherein the "heteroaryl" and "alkyl" moieties are as defined herein.
"Heterocycloalkylether group" means: (heterocycloalkyl) -O-. Wherein the "heterocycloalkyl" moiety is as defined herein.
"Heterocycloalkylamino" means: both mono-and di-heterocycloalkylamino, unless otherwise indicated. Mono-heterocycloalkylamino means: (heterocycloalkyl-) NH-; di-heterocycloalkylamino means (heterocycloalkyl) 2 The radical of N-. Wherein the definition of "heterocycloalkyl" moiety is found in the relevant sections herein.
"arylalkylamino" refers to: both mono-arylalkylamino and di-arylalkylamino groups, unless otherwise indicated. Mono-arylalkylamino refers to: (aryl-alkyl) -NH-; di-arylalkylamino refers to (aryl-alkyl) 2 -a group of N-. Wherein the definition of "aryl" and "alkyl" moieties is found in the relevant section herein.
"cycloalkylamino" refers to: both mono-cycloalkylamino and di-cycloalkylamino, unless otherwise indicated. Mono-cycloalkylamino means: (cycloalkyl) -NH-; di-arylalkylamino refers to (cycloalkyl) 2 -a group of-N-. Wherein the definition of "cycloalkyl" moiety is found in the relevant section herein.
"arylamino" refers to: both mono-arylamino and di-arylamino groups unless otherwise indicated. Mono-arylamino means: (aryl) -NH-; by di-arylamino is meant (aryl) 2 -a group of-N-. Wherein the definition of the "aryl" moiety is found in the relevant section herein.
"heteroarylamino" means: both mono-heteroarylamino and di-heteroarylamino unless otherwise indicated. Mono-heteroarylamino means: (heteroaryl) -NH-; di-heteroarylamino means (heteroaryl) 2 -a group of-N-. Wherein the "heteroaryl" moiety is defined hereinA relevant portion.
"heteroarylalkylamino" refers to: both mono-heteroarylalkylamino and di-heteroarylalkylamino, unless otherwise indicated. Mono-heteroarylalkylamino means: (heteroaryl-alkyl) -NH-; di-heteroarylalkylamino means (heteroaryl-alkyl) 2 -a group of-N-. Wherein the "heteroaryl" and "alkyl" moieties are defined in the relevant sections herein.
Unless otherwise specified, a subunit of the invention refers to a divalent group, i.e., refers to a group in which one hydrogen atom in a monovalent group is replaced by a valence. For example, "heteroalkylene" refers to a heteroalkyl group in which one hydrogen atom is replaced by a valence; "heterocyclylene" refers to a heterocyclic group in which one hydrogen atom is replaced by a valence; "arylene" refers to an aryl group in which one hydrogen atom is replaced by a valence; "alkylene" refers to an alkyl group in which one hydrogen atom is replaced by a valence; "alkenylene" refers to an alkenyl group in which one hydrogen atom is replaced by a valence; "cycloalkylene" refers to a cycloalkyl group in which one hydrogen atom is replaced by a valence; "heteroarylene" refers to a heteroaryl group in which one hydrogen atom is replaced by a valence; "heterocycloalkylene" means a heterocycloalkyl group in which one hydrogen atom is replaced by a valence; "heterocycloalkenylene" refers to a heterocycloalkenyl group in which one hydrogen atom is replaced by a valence; "Alkyleneoxy" refers to an alkoxy group in which one hydrogen atom is replaced by a valence; "alkenylene" refers to an alkenylene in which one hydrogen atom is replaced by a valence; "alkynyloxy" refers to alkynyloxy in which one hydrogen atom is replaced by a valence, and the like. Wherein the above heterocyclic, aryl, alkyl, alkenyl, cycloalkyl, heteroaryl, heterocycloalkyl, heterocycloalkenyl, alkoxy, alkenyloxy, alkynyloxy, and the like are as defined herein.
The invention includes compounds represented by the general formula (I) and the various isomeric forms thereof which are possible. The method comprises the following steps: non-mirror image isomers, tautomers, and geometric isomers of "E" or "Z" configurational isomers, and the like. Any chemist with a certain basis can isolate the above optically or stereoisomerically pure compounds.
Otherwise, the groups not defined here follow the usual definitions.
Preferred embodiments of the present invention include the following embodiments.
In the compounds of the invention, R 1 Preferably a hydrogen atom, carboxyl, nitro, amino, cyano, acyl, alkyl, alkoxy, alkenyl, alkynyl, halogen, isotope, haloalkenyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl; any of the foregoing groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, amino, cyano, carboxy, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, and alkoxyalkyl.
R1 is more preferably amino, cyano, alkenyl, alkynyl, halogen, isotope, C1-C6 alkyl, C6-C12 aryl, 5-12 membered arylC 1-C6 alkyl, C3-C9 cycloalkyl, 4-15 membered heteroaryl, 4-15 membered heteroarylC 1-C6 alkyl, 3-14 membered heterocycloalkyl, or C1-C6 alkoxy; any of the above groups may each independently be substituted with one or more substituents selected from substituent group a above, including but not limited to halogen, isotope, amino, cyano, carboxy, = O, -CF 3 Alkyl, hydroxyl, hydroxyalkyl, alkoxy.
R 1 More preferably a cyano group, an alkynyl group, a halogen, an isotope, or a C1-C6 alkyl group, a pyrazolyl group, a furyl group, a pyrimidinyl group, an aminopyrazolyl group, a morpholinyl group, or a phenyl group; any of the above groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, amino, cyano, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF3, haloalkyl, alkyl, hydroxyl, hydroxyalkyl, alkoxy.
In the compounds of the present invention, L 1 preferably-CH 2 CH 2 -, -CH = CH-, or-C ≡ C-.
L 1 More preferably-CH = CH-, or-C ≡ C-. .
In the compounds of the present invention, L 2 Preferably alkylene, carbonyl, -CO-NH-, -alkyl-CO-NH-, -alkyl-NH-, or-alkyl-O-. Any of the above groups may be each independently substituted with one or more substituents including a hydrogen atom, an alkyl group.
L 2 <xnotran> -CO-NH-, - -CO-NH-, - -NH-C (O) -, - -NH-, - -O-. </xnotran> Any of the above groups may be each independently substituted with one or more substituents including a hydrogen atom, an alkyl group.
L 3 Preferably: covalent bonds, alkylene groups, -O-) -NH-, -C (O) -NH-, -NH-C (O) -, -alkyl-C (O) -NH-, -alkyl-NH-C (O) -, -alkyl-NH-, -alkyl-O-. Any of the above groups may be each independently substituted with one or more substituents including a hydrogen atom, an alkyl group.
L 3 Preferably: covalent bonds, -O-, -NH-, -C (O) -NH-, -NH-C (O) -.
In the compounds of the present invention, R 2 Preferably carboxy, amino, acyl, alkyl, alkoxy, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl; any of the above groups independently can be substituted with one or more substituents including, but not limited to, halogen, isotope, amino, cyano, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF3, haloalkyl, alkyl, alkenyl, alkynyl, hydroxyl, hydroxyalkyl, alkoxy, and alkoxyalkyl.
R 2 More preferably C1-C6 alkyl, C6-C12 aryl, 5-12 membered arylC 1-C6 alkyl, C3-C9 cycloalkyl, 4-15 membered heteroaryl, 4-15 membered heteroarylC 1-C6 alkyl3-14 membered heterocycloalkyl, C1-C6 alkoxyC 1-C6 alkyl; any of the above groups may each independently be substituted with one or more substituents selected from the above substituent group a, including but not limited to halogen, isotope, amino, cyano, carboxy, phenyl, = O, -CF3, haloalkyl, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, and alkoxyalkyl.
R 2 Further preferred are pyrazolyl, furyl, pyrimidinyl, bromopyrimidinyl, aminopyrazolyl, morpholinyl, and phenyl; any of the foregoing groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, cyano, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, and alkoxyalkyl.
In the compounds of the invention, R 3 Preferably: a hydrogen atom, an alkyl group, a heteroalkyl group, an arylalkyl group, a cycloalkyl group, an aryl group, a heteroaryl group, a heteroarylalkyl group, a heterocycloalkyl group, an alkyl ether group, a heteroalkyl ether group, an arylalkyl ether group, a cycloalkyl ether group, an aryl ether group, a heteroarylether group, a heteroarylalkyl ether group, an alkylamino group, a heteroalkyl amino group, an arylalkylamino group, a cycloalkylamino group, an arylamino group, a heteroarylamino group, a heteroarylalkylamino group, a heterocycloalkylamino group; any of the foregoing groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, = O, -CF3, haloalkyl, alkyl, alkenyl, alkynyl, hydroxyl, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl.
R 3 More preferably methyl, ethyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, or aminoalkyl; any one of the above groupsEach of the groups independently may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkenyl, alkoxyalkyl.
The invention includes compounds represented by the general formula (I) and the various isomeric forms thereof. The method comprises the following steps: non-mirror image isomers, tautomers, and geometric isomers of "E" or "Z" configurational isomers, and the like. The optically or stereoisomerically pure compounds described above can be isolated by one skilled in the art according to conventional methods in the art.
The invention comprises compounds represented by the general formula (I) and possible racemates or/and enantiomers or/and mixtures of non-enantiomers thereof.
In addition, the application of the compound represented by the general formula (I) also covers the solvated forms and unsolvated forms of the compound. Accordingly, each form includes compounds having the indicated structure, including hydrates and anhydrates thereof.
In addition to the compounds represented by the general formula (I), various embodiments of the kinase inhibitors include: pharmaceutically acceptable salts, prodrugs and active metabolites of these compounds, and pharmaceutically acceptable salts of these metabolites.
The term "pharmaceutically acceptable salts" refers to certain salts of the above compounds which retain their biological activity and which are suitable for pharmaceutical use. There are two forms of pharmaceutically acceptable salts of the compounds represented by the general formula (I): one is a salt with an acid; the other is a salt with an alkali or alkali metal. Acids that form pharmaceutically acceptable salts with the compounds represented by general formula (I) include inorganic and organic acids. Suitable inorganic acids include: hydrochloric acid, sulfuric acid and phosphoric acid. Suitable organic acids may be selected from aliphatic, cycloaliphatic, aromatic, heterocyclic carboxylic and sulfonic organic acids; examples include, but are not limited to: formic acid, acetic acid, propionic acid, succinic acid, glycolic acid, gluconic acid, lactic acid, malic acid, tartaric acid, glycine, arginine, citric acid, fumaric acid, alkylsulfonic acids, arylsulfonic acids, and the like. Alkali metals that form pharmaceutically acceptable salts with compounds represented by the general formula (I) include: lithium, sodium, potassium, magnesium, calcium, aluminum, zinc, and the like; bases which form pharmaceutically acceptable salts with compounds represented by the general formula (I) include: choline, diethanolamine, morpholine, and the like.
A "prodrug" is a derivative of a compound of formula (I) which is converted (e.g., by hydrolysis, reduction or oxidation) in vivo to the compound of formula (I) by means of metabolism in vivo. For example, a compound having a hydroxyl group represented by the general formula (I) can be reacted with an acid to prepare a corresponding ester. The corresponding ester is a prodrug, which can hydrolyze the parent drug in vivo. Suitable acids for preparing "prodrugs" include, but are not limited to: acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, oxalic acid, salicylic acid, succinic acid, fumaric acid, maleic acid, methylene-bis- β -hydroxynaphthoic acid, gentisic acid, isethionic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid and the like.
The kinase inhibitors referred to in the present invention include IC 50 Those having a value of 10. Mu.M or less. The kinase referred to in the present invention includes, but is not limited to, RET, TRK.
The administration mode of the compound represented by the general formula (I) may be either gastrointestinal administration or parenteral administration. Gastrointestinal administration refers to oral or rectal administration. Parenteral administration includes: subcutaneous, intramuscular, intravenous, and intradermal routes. In general, the active compounds represented by the general formula (I) may be administered using a pharmaceutically acceptable carrier or diluent.
"therapeutically effective amount" or "therapeutic amount" refers to an amount sufficient to produce a therapeutic effect. The effective amount may be administered in one or more divided doses. Generally, an effective amount is sufficient to moderate, ameliorate, stabilize, slow or delay further progression of the disease.
The compounds of the present invention may be used alone or in combination with one or more other drugs; or for a patient undergoing surgery or radiotherapy, wherein a compound of the invention is administered to the patient concomitantly with, or prior to, or subsequent to, the surgery or radiotherapy; or making into certain dosage form with pharmaceutically acceptable carrier, diluent or excipient. The specific dosage form depends on the route of administration.
The medicine formula for parenteral injection comprises a pharmaceutically acceptable sterile aqueous solution or non-aqueous solution, a dispersing agent, a suspending agent or an emulsifying agent and a powder injection which is prepared into an injectable sterile aqueous solution before use.
If desired, and for more effective distribution, the compounds of the invention may be incorporated into slow release or targeted delivery systems, such as: polymer matrices, liposomes and microspheres.
Solid dosage forms for oral administration include: capsules, tablets, troches, powders, and granules. In these solid dosage forms, the active compound represented by general formula (I) is contained together with at least one inert and pharmaceutically acceptable excipient or carrier. These excipients or carriers include sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as: starch, lactose, sucrose, glucose, mannitol, and salicylic acid; b) Binding agents, for example: carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) Disintegrants, for example: agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; e) Dissolution retarders, for example: paraffin wax; f) Absorption accelerators, for example: a quaternary ammonium compound; g) Wetting agents, for example: cetyl alcohol and glycerol monostearate; h) Adsorbents, for example: kaolin and bentonite; and i) lubricants, for example: talcum powder, calcium stearate, magnesium stearate and solid polyethylene glycol.
Lozenges, dragees, capsules, tablets and granules in solid dosage form can be prepared with a coating or shell.
The active compounds may also be administered in microencapsulated form. If desired, one or more of the above-mentioned excipients may be present.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsifiers, solutions, suspensions, syrups and the like. In addition to the active compound, the liquid dosage forms may contain inert diluents commonly used in liquid dosage forms, such as: water or other solvents, stabilizers and emulsifiers, for example: ethyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycol, and fatty acid esters of sorbitan, and the like.
In addition to inert diluents, oral compositions may also include: adjuvants, for example: wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
In suspensions, suspending agents may be present in addition to the active compounds, for example: ethoxylated isostearyl alcohols, polyoxyethylene sorbitol, and sorbitan esters.
Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of the present invention with suitable non-irritating excipients or carriers.
Dosage forms for topical administration of the compounds of the present invention include powders, patches, sprays, ointments and inhalants which may be prepared by mixing the active compound under sterile conditions with a pharmaceutically acceptable carrier and any preservatives, buffers or propellants which may be required.
The preferred dosage range for the compounds of the invention is about 0.01 to about 400 mg per kg of body weight per day. More preferably, the dosage range is 0.2-100 mg per kg body weight per day. The appropriate dosage may also be selected for multiple separate administrations per day.
The pyrazolo [1,5-a ] pyridine derivatives of the invention can be used as, but not limited to, kinase inhibitors. Pyrazolo [1,5-a ] pyridine derivatives are disclosed which may be used alone or in combination with other pharmaceutical or pharmaceutically acceptable carriers, diluents or excipients, and are useful in the prevention or treatment of conditions caused by, associated with or associated with the destruction of cell proliferation and/or angiogenesis. One example of such a condition is cancer.
The compounds of the invention may also be useful in therapy involving, or at least in part being modulated by, any one or more of the kinase activities of TRK, RET, FGHR, PDGFR, VEGFR, and the like, wherein TRK, RET activities are known to play a role in promoting disease onset, or the symptoms are known or have been shown to be alleviated by TRK, RET inhibitors. Conditions of this type which are expected to be treated by the compounds of the invention include, but are not limited to, the following: anti-proliferative disorders (e.g., cancer); neurodegenerative diseases, including: huntington's disease, polyglutamine disease, parkinson's disease, alzheimer's disease, seizures, striatal substantia nigra degeneration, progressive supranuclear palsy, torsion dystonia, spastic torticollis and dyskinesia, familial tremor, gilles de la tourette's syndrome, diffuse lewy body disease, progressive supranuclear palsy, pick's disease, intracranial hemorrhage, primary lateral sclerosis, spinal muscular atrophy, amyotrophic lateral sclerosis, hypertrophic interstitial polyneuropathy, retinitis pigmentosa, hereditary optic atrophy, hereditary spastic paraplegia, progressive ataxia and Shy-Drager syndrome; a metabolic disease comprising: type 2 diabetes mellitus; an ocular degenerative disease comprising: glaucoma, age-related macular degeneration, rubeosis iridis glaucoma; inflammatory diseases and/or immune system disorders, including: rheumatoid Arthritis (RA), osteoarthritis, juvenile chronic arthritis, graft-versus-host disease, psoriasis, asthma, spondyloarthropathies, psoriasis, crohn's disease, inflammatory bowel disease, colonic ulcers, alcoholic hepatitis, diabetes, sjoegrens syndrome, multiple sclerosis, ankylosing spondylitis, membranous glomerulopathy, discogenic pain, systemic lupus erythematosus; diseases involving angiogenesis, including: cancer, psoriasis, rheumatoid arthritis; a psychological disorder, comprising: bipolar disorder, schizophrenia, mania, depression and dementia: cardiovascular diseases include; heart failure, restenosis and arteriosclerosis; fibrotic diseases, including: liver fibrosis, cystic fibrosis, and angiofibroma; infectious diseases, including: fungal infections, for example: candida albicans, bacterial infections, viral infections, for example: herpes simplex, protozoal infections, such as: malaria, leishmania infections, trypanosoma brucei infections, toxoplasmosis and coccidiosis and hematopoietic disorders including: marine anemia, and sickle cell anemia.
The term "cancer" as used in this specification generally refers to a wide range of conditions characterized by uncontrolled abnormal growth of cells.
The compounds of the present invention are expected to be useful in the treatment of various cancers, including but not limited to: bone cancers, including: ewing's sarcoma, osteosarcoma, chondrosarcoma, etc.; brain and CNS tumors, including: acoustic neuroma, neuroblastoma, glioma and other brain tumors, spinal cord tumors, breast cancer, colorectal cancer, advanced colorectal adenocarcinoma; endocrine cancers, including: adrenocortical carcinoma, pancreatic carcinoma, pituitary cancer, thyroid cancer, parathyroid cancer, thymus gland cancer, multiple endocrine tumors; gastrointestinal cancers, including: gastric cancer, esophageal cancer, small intestine cancer, renal cell carcinoma, liver cancer, extrahepatic bile duct cancer, gastrointestinal carcinoid tumor, and gallbladder cancer; genitourinary cancers, including: cuiping cancer, penile cancer, prostate cancer; gynecological cancers, including: cervical cancer, ovarian cancer, vaginal cancer, uterine/endometrial cancer, pudendum cancer, gestational trophoblastic tumors, fallopian tube cancer, uterine sarcoma; head and neck tumors, including: oral cancer, lip cancer, salivary gland cancer, larynx cancer, hypopharynx cancer, eupharyngeal cancer, nasal cancer, sinus cancer, and nasopharyngeal cancer; blood cancers, including: childhood leukemia, acute lymphocytic leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, acute promyelocytic leukemia, plasma cell leukemia; a hematological disorder of bone marrow cancer comprising: myelodysplastic syndrome, myeloproliferative disorders, aplastic anemia, fanconi anemia, idiopathic macroglobulinemia; lung cancer species, including: small cell lung cancer, non-small cell lung cancer; lymphoid cancers including: hodgkin's disease, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, peripheral T-cell lymphoma, AIDS-related lymphoma; eye cancers, including: retinoblastoma, uveal melanoma, skin cancers, including: melanoma, non-melanoma skin cancer, merkel cell carcinoma, soft tissue sarcomas, such as: children soft tissue sarcoma, adult soft tissue sarcoma, kaposi sarcoma, urinary system cancer, including: kidney cancer, wilms' tumor, bladder cancer, urinary tract cancer, and metastatic cell cancer.
Cancers that the compounds of the present invention may be used to treat include, but are not limited to: breast cancer, lung cancer, renal cell carcinoma, gastric cancer, ovarian cancer, thyroid cancer, rectal cancer, prostate cancer, head and neck cancer, kidney, stomach and brain cancer.
Preferred cancers that can be treated by the compounds of the present invention are solid tumors and hematological malignancies.
In addition, the compounds of the invention are useful for treating proliferative diseases that are resistant to other chemotherapeutic treatments; and for the treatment of hyperproliferative diseases, such as: leukemia, psoriasis, and the like.
EXAMPLE Synthesis and use of novel pyrazolo [1,5-a ] pyridine derivatives
The compounds of the present invention represented by general formula (I) can be synthesized using the synthetic routes and synthetic methods discussed below. The raw materials are convenient and easy to obtain. However, the synthetic route and the synthetic method used in the invention can be widely applied to the synthesis of analogues, and only the starting raw materials need to be changed. For example, synthesis of a compound not described in detail in the examples herein can be carried out by replacing the starting materials with those of the corresponding target compound, and then, if necessary, slightly changing the reaction conditions in accordance with the common knowledge of the chemistry.
Reagents for each embodiment can be prepared using available starting materials using reaction pathways or synthetic schemes that are described below using means known in the art. The preparation of particular compounds of particular embodiments is described in detail in the examples below, but those skilled in the art will recognize that the chemistry described may be adapted to the preparation of a variety of other compounds in different embodiments. For example: the synthesis of non-exemplified compounds can be successfully performed by modifications apparent to those skilled in the art, such as: by appropriate protection of interfering groups, by changing to other appropriate reagents known in the art, or by performing routine modifications of reaction conditions. Other reactions disclosed herein or known in the art may be deemed to have applicability for the preparation of other compounds of each embodiment.
Reagents for synthesizing compounds can be obtained or prepared according to techniques known in the art.
In the following examples, all temperatures are in degrees celsius unless otherwise indicated.
Various starting materials and reagents are commercially available. Suppliers include, but are not limited to: aldrich Chemical Company, lancaster Synthesis Ltd, and the like. Commercial starting materials and reagents were used without further purification unless otherwise indicated.
The glassware is oven dried and/or heat dried. The reaction was followed on a glass silica-gel-60F 254 plate (0.25 mm) (TLC). Analytical thin layer chromatography and development with appropriate solvent ratio (v/v). The end of the reaction was determined by the time the starting material was consumed on TLC.
In general, the subsequent treatment is to double the volume of the reaction solution with the solvent used for the reaction and then extract three times with 25% of the total volume of the extraction solvent, unless otherwise specified. The product-containing extract was dehydrated over anhydrous sodium sulfate, filtered on a rotary evaporator, the solvent was evaporated under reduced pressure and the removal of the solvent in vacuo was noted. And finally, separating by using flash column chromatography to obtain the target compound.
1 The H NMR spectrum was obtained using a Bruker instrument (400 MHz) and the chemical shifts were expressed in ppm. Chloroform was used as a reference standard (7.25 ppm) or tetramethylsilane internal standard (0.00 ppm). Other solvents commonly used for NMR may also be used as necessary. 1 Method for H NMR expression: s = singlet, d = doublet, t = triplet, m = multiplet, br = broadened, dd = doublet of doublets, dt = doublet of triplets. If a coupling constant is provided, it is in Hz.
The mass spectrum is measured by an LC/MS instrument, and the ionization mode can be ESI or APCI. All melting points are not modified.
The following examples are merely illustrative of the synthesis of specific compounds of the invention. But there is no limitation on the synthesis method. The compounds not listed below can also be prepared by selecting appropriate starting materials and adjusting reaction conditions slightly appropriate to the degree of common knowledge where necessary, by the same synthetic route and synthetic method as those described below.
Synthesis of
A compound of the formula (I) when L 2 When the catalyst is = -C (O) -NH-, the synthesis method is as follows: the methyl group is removed by selecting a proper I as a raw material and under a proper catalyst and a proper condition (for example, under the action of 1-dodecanethiol), and a compound II is obtained. Under catalysis, the compound II reacts with the boron compound III to obtain IV. Compound IV is converted to compound V by the action of N-phenylbis (trifluoromethanesulfonimide) (1, 1-trifluorooro-N-phenyl-N- ((trifluoromethenyl) sulfonyl) methanesulfonamide). On the other hand, compound VIII obtained by condensing compound VI and compound VII reacts with compound V to obtain compound IX, namely the target compound shown in the same general formula (I) (synthesis scheme 1),
synthesis scheme 1
Figure BDA0003967308590000281
When R in the formula (I) 1 And (= -CN), it can be synthesized according to scheme 2. In this case, the synthesis method is: proper X is selected as a raw material, and is acylated and aminated to obtain XIV. Under the action of acid, cyclizing to obtain the compound XVI. A formyl group is introduced into the compound XVI, which is converted into a nitrile group by hydroxylamination. Compound I-1 is obtained by removal of the methyl group in the presence of a suitable catalyst and under suitable conditions, for example, under the action of 1-dodecanethiol.
Synthesis scheme 2
Figure BDA0003967308590000282
A compound of the formula (I) when L 1 = CH or-C ≡ C-, L 2 -R 2 In order to substitute an aryl group, the corresponding object compound, compound XXIII, can be synthesized by the method shown in scheme 3. The method comprises the following steps: the appropriate iodo compound XIX is selected as starting material and reacted with XX in the presence of an appropriate catalyst and under appropriate conditions to give compound XXI. The silicon protecting group is then removed in basic methanol solution to afford XXII. The latter is reacted with compound V to give compound XXIII.
Synthetic scheme 3
Figure BDA0003967308590000291
The present invention will be further clarified by the following examples. The purpose of the present invention is to make the detailed contents of the present invention more clearly understood and practiced by those having basic knowledge in the field. However, the scope of the present invention is not limited to these examples only.
EXAMPLE 1 Synthesis of (S) -3- (3-cyano-6- (1-methyl-1H-pyrazol-4-yl) pyrazolo [1,5-a ] pyridin-4-yl) -N- (1- (1-methyl-1H-imidazol-4-yl) methyl) pyrrolidin-3-yl) propionamide (IX-1)
Step-16-bromo-4-hydroxypyrazolo [1,5-a ]]Synthesis of pyridine-3-carbonitrile (II-2):
Figure BDA0003967308590000292
the compound 6-bromo-4-methoxy pyrazolo [1,5-a ]]Pyridine-3-carbonitrile (I-1) (100.0g, 396.7mmol and 1.0eq) is added into N, N-dimethylformamide (1000.0mL, 10.0Vol), the system is heated to 40-50 ℃ under the condition of stirring, a mixed solution of sodium hydroxide (31.7g, 793.4mmol, 2.0eq) and water (64.9mL and 0.65Vol) which are prepared in advance and cooled to room temperature is added, the temperature is controlled to be 40-50 ℃, N-dodecyl mercaptan (160.6g, 793.4mmol and 2.0eq) is slowly dripped into the reaction system, and after the feeding is finished, the temperature is kept at 45-50 ℃ for reaction for 10-12 hours. After the reaction is monitored by HPLC, the system is loweredHeating to 10-20 deg.C and controlling the temperature in this range, slowly adding water (3000.0mL, 30.0Vol) to quench, adding methyl tert-butyl ether (1000.0mL x2, 10.0Vol 2) to extract, separating water phase, cooling to 0-10 deg.C and controlling in this temperature range, slowly adding 10% citric acid solution (about 1200 g) prepared in advance into water phase, regulating pH = 5-6, separating out a large quantity of solid, stirring at 0-10 deg.C for 10-20 min, suction-filtering, washing filter cake with water (300.0mL x2, 3.0Vol 2), suction-drying, transferring filter cake into oven and drying at 50-60 deg.C to obtain white-like solid II-2 (88.0g, 93.2%). 1H MNR (400MHz, DMSO-d) 6 )δ:8.87-8.75(m,1H),8.62-8.51(m,1H),8.03-7.89(m,1H),6.95-6.84(m,1H).ESI-MS(m/z):239.8[M+H]+.
Step-24-hydroxy-6- (1-methyl-1H-pyrazol-4-yl) pyrazolo [1,5-a]Synthesis of pyridine-3-carbonitrile IV-1:
Figure BDA0003967308590000293
the compound 6-bromo-4-hydroxypyrazolo [1,5-a ]]Pyridine-3-carbonitrile (II-1) (55.0g, 231.1mmol, 1.0eq) and potassium carbonate (63.8g, 462.1mmol, 2.0eq) were added to a mixed solution of dioxane (1100.0 mL,20.0 Vol) and water (275.0 mL,5.0 Vol), the catalyst tetratriphenylphosphine palladium (13.4g, 11.6mmol, 0.05eq) was added to the reaction system with stirring, nitrogen was substituted for 5 times, and the system was heated to 95-100 ℃ to react for 4-5 hours. After the reaction was monitored by HPLC, the system was cooled to 10-20 ℃ and controlled within this temperature range, water (3300.0mL, 60.0Vol) was slowly added to the system to quench, the mixture was passed through a Celite pad, methyl t-butyl ether (1100.0mL x 3,20.0Vol x 3) was added to the filtrate to extract, the mixture was separated to obtain an aqueous phase, the temperature was controlled to 15-25 ℃, a previously prepared 1M diluted hydrochloric acid solution was slowly added to the aqueous phase to adjust the pH = 5-6, a large amount of solid was precipitated, the mixture was stirred at 15-25 ℃ for 10-20 min, filtered, the filter cake was washed with water (275.0mL x2, 5.0Vol 2), dried, and the filter cake was dried in a oven at 50-60 ℃ to obtain a gray solid IV-1 (51.6g, 93.4%). 1 H NMR(400MHz,DMSO-d 6 )δ:11.36(s,1H),8.74(s,1H),8.50(s,1H),8.21(s,1H),7.94(s,1H),6.96(s,1H),3.88(s,3H).ESI-MS(m/z):240.1[M+H] + .
Step-33-cyano-6- (1-methyl-1H-pyrazol-4-yl) pyrazolo [1,5-a]Synthesis of pyridin-4-yl trifluoromethanesulfonate (V-1):
Figure BDA0003967308590000301
the compound 4-hydroxy-6- (1-methyl-1H-pyrazol-4-yl) pyrazole [1,5-a]Pyridine-3-nitrile (IV-1) (73.6g, 307.6mmol and 1.0eq) is dissolved in N, N-dimethylacetamide (1472.0mL and 20.0Vol), the temperature is controlled to be 10-20 ℃, N-phenyl bis (trifluoromethyl) sulfimide (120.9g, 338.4mmol and 1.1eq) is added into the system in batches, the temperature of the system is reduced to 10-15 ℃ and controlled within the temperature range, N-diisopropylethylamine (79.5g, 615.2mmol and 2.0eq) is slowly dripped into the system, and the temperature is controlled to be 10-15 ℃ to react for 4-5 hours after the dripping is finished. After the reaction is monitored by HPLC, the temperature of the system is reduced to 0-10 ℃ and controlled within the temperature range, water (3200.0mL and 43.5 Vol) is slowly added into the system for quenching, a large amount of solid is separated out, the temperature is kept at 10-20 ℃, stirring is carried out for 10-20 min, suction filtration is carried out, a filter cake is washed by water (441.6mL, 2and 6.0Vol, 2), suction drying is carried out, and the filter cake is transferred into a drying oven to be dried at 50-60 ℃ to obtain an off-white solid V-1 (112.0g and 98.0%). 1 HNMR(40 0MHz,DMSO-d 6 )δ:9.53-9.28(m,1H),8.90-8.64(m,1H),8.58-8.29(m,1H),8.25-7.89(m,2H),4.01-3.68(m,3H).ESI-MS(m/z):372.0[M+H] + .
Step-4Synthesis of tert-butyl 3- (S) -propionylaminopyrrolidine-1-carboxylate (VIII-1):
Figure BDA0003967308590000302
the compound 3- (S) -aminopyrrolidine-1-carboxylic acid tert-butyl ester (VI-1) (1.8g, 9.6mmol, 1.0eq) is added into dichloromethane (100mL, 55.5 Vol), under the condition of nitrogen protection, 2-ethoxy-1-ethoxycarbonyl-1, 2-dihydroquinoline (EEDQ, 2.8g,11.6mmol,1.2 0eq) and propiolic acid (VII.1, 678mg,9.6mmol, 1.0eq) are added, and the temperature is controlled at 20-30 ℃ to stir for 15-20h. After the completion of the TLC monitoring reaction, the system was concentrated, and the residue was purified by silica gel column chromatography with petroleum ether/ethyl acetate =5/1 andpetroleum ether/ethyl acetate =2/1 as eluent, and concentrated to give yellow solid VIII-1 (1.50g, 65%). 1 H NMR(400MHz,CDCl 3 )δ:6.37-6.47(d,J=42Hz,1H),4.47-4.51(dd,J=5.4Hz,1H),3.59-3.63(dd,J=6.0Hz,1H),3.45(s,2H),3.25-3.30(d,J=19.6Hz,1H),2.83(s,1H),2.14-2.19(m,1H),1.83(s,1H),1.47(s,9H).ESI-MS(m/z):237.2[M-H] -
Step-53- (S) - (3-cyano-6- (1-methyl-1H-pyrazol-4-yl) pyrazole [1,5-a]Synthesis of pyridin-4-yl) -N- (pyrrolidin-3-yl) propionamide (IX-1) hydrochloride:
Figure BDA0003967308590000311
under the protection of nitrogen, 3-cyano-6- (1-methyl-1H-pyrazol-4-yl) pyrazole [1,5-a]Pyridin-4-yl trifluoromethanesulfonate (V-1) (0.82g, 2.2mmol, 1.0eq) and 3- (S) -propionylaminopyrrolidine-1-carboxylic acid tert-butyl ester (VIII-1) (0.80g, 3.4mmol, 1.5eq) were dissolved in N, N-dimethylacetamide (50.0mL, 60.0Vol), cuprous iodide (0.042g, 0.22mmol, 0.1eq), N-diisopropylethylamine (0.571g, 4.4mmol, 2.0eq) and a catalyst of bistriphenylphosphine palladium dichloride (0.154g, 0.22mmol, 0.1eq) were added, and the mixture was replaced with nitrogen 6 times, and the system was heated to 95-100 ℃ to react for 1-2 hours. After TLC monitoring of the reaction was complete, the system was quenched by pouring into water (200.0 mL), filtered with suction, and the filter cake was purified by silica gel column chromatography using petroleum ether/ethyl acetate =1/1 and ethyl acetate as eluent and concentrated to afford Key intermedate-1 as a white solid (0.59g, 58%). 1 H NMR(400MHz,CDCl 3 )δ:8.71(s,1H),8.30(s,1H),7.79-7.83(d,J=17.6Hz,2H),7.69(s,1H),7.28(s,1H),4.55(s,1H),4.03(s,3H),3.66-3.70(dd,J=6.4Hz,1H),3.30-3.53(m,3H),2.07(s,1H),1.93-1.98(m,1H),1.50(s,9H).ESI-MS(m/z):404.0[M+H-56] + .
Reacting 3- (S) - (3-cyano-6- (1-methyl-1H-pyrazol-4-yl) pyrazole [1,5-a ]]Pyridin-4-yl) propionamido) pyrrolidine-1-carboxylic acid tert-butyl ester (Key Intermediate-1) (500mg, 1.08mmol, 1.0eq) was dissolved in ethyl acetate (38.5mL, 77.0Vol), 4M ethyl hydrogen chloride-acetate solution (3.85mL, 7.7Vol) was added, and the temperature was controlled at 20-30 ℃ and stirring was carried out for reaction for 15-20h. TLC monitored reverseAfter completion of the reaction, the system was filtered by suction, and the filter cake was washed with ethyl acetate and recrystallized from water and ethanol to give the target compound 1 (IX-1) (305mg, 71%) as a yellow solid. 1 H NMR(400MHz,DMSO-d 6 )δ:9.42(d,J=11.2Hz,1H),9.22(d,J=6.4Hz,2H),8.75(d,J=8.0Hz,1H),8.39(s,1H),8.29(br,1H),8.13(s,1H),4.43-4.48(q,J=6.4Hz,1H),3.89(s,3H),3.42-3.46(m,1H),3.21-3.30(m,3H),3.05-3.09(m,1H),2.14-2.23(m,1H),1.87-1.95(m,1H).ESI-MS(m/z):360.2[M+H] + .
EXAMPLE-Synthesis of 6- (1-methyl-1H-pyrazol-4-yl) -4- (5-methyl-2- (4-methylpiperazin-1-yl) pyridin-4-yl) ethynyl) pyrazolo [1,5-a ] pyridine-3-carbonitrile (2)
Step-1Synthesis of tert-butyl (mesitylenesulfonyl) oxycarbarbamate (XII):
Figure BDA0003967308590000312
a solution of 2,4, 6-trimethylbenzene-1-sulfonyl chloride (X) (50.0g, 0.228mol) and tert-butyl hydroxycarbamate (XI, 30.4g, 0.228mol) in tert-butyl methyl ether (500 mL) was cooled to 0 ℃. Triethylamine (25.4 g, 0.25mol) was added dropwise. After the addition was complete, the mixture was stirred at 0 ℃ for 0.5 h. The mixture was then warmed to room temperature and stirred for 2 hours. The mixture was filtered. The filtrate was washed with water (100 mL) and brine (50 mL), na 2 SO 4 Dried and concentrated to give a residue. The residue was triturated with petroleum ether (50 mL) and filtered. The filter cake was collected and dried to give the title compound XII (60.2 g, 84% yield) as an off-white solid. 1 H NMR(400MHz,CDCl 3 )δ:7.57(br s,1H),6.99(s,2H),2.68(s,6H),2.32(s,3H),1.32(s,9H).
Synthesis of Step-2-1-amino-3-bromo-5-methoxypyridin-1-ium 2,4, 6-trimethylbenzenesulfonate (XIV):
Figure BDA0003967308590000321
trifluoroacetic acid (100 mL) was cooled to 0 ℃ and tert-butyl (mesitylenesulfonyl) oxycarbarbamate (XII) (60.0 g, 0.19mol) was added portionwise. After the addition was complete, the mixture was stirred at 20 ℃ for 2 hours. Ice water (300 mL) was added dropwise and the mixture was stirred for 0.5 h and filtered. The filter cake was washed with water (50 mL × 3) and collected to give the corresponding intermediate (58 g, wet weight) as a white solid. The product was used immediately in the next step without further purification.
Intermediate (58 g, wet weight) was dissolved in dichloromethane (300 mL) and Na was added 2 SO 4 Dried for 5 minutes and filtered. The filtrate was cooled to 0 ℃. 3-bromo-5-methoxypyridine (XIII) (35.8g, 0.19mol) was added in portions. After the addition was complete, the mixture was stirred at room temperature for 2 hours. The mixture was filtered. The filter cake was washed with dichloromethane (50 mL × 2), collected and dried to give the title compound (XIV) (48.2 g, yield 63%) as a white solid. 1 H NMR(400MHz,DMSO-d 6 )δ:8.71(s,1H),8.62(s,1H),8.59(s,2H),8.27(s,1H),6.74(s,2H),3.97(s,3H),2.50(s,6H),2.17(s,3H).
Step-36-bromo-4-methoxypyrazolo [1,5-a ]]Synthesis of pyridine (XVI):
Figure BDA0003967308590000322
a solution of 1-amino-3-bromo-5-methoxypyridin-1-yl 2,4, 6-trimethylbenzenesulfonate (XIV) (48.0 g, 0.12mol) in N, N-dimethylformamide (150 mL) was cooled to 0 ℃. Triethylamine (24.1g, 0.24mol) was added dropwise. After the addition was complete, the mixture was stirred at 0 ℃ for 5 minutes. Ethyl propiolate (XV) (23.3g, 0.24mol) was added dropwise. After the addition was complete, the mixture was stirred at room temperature overnight. The mixture was poured into water (500 mL) and extracted with ethyl acetate (100 mL. Times.4). The combined organic phases were washed with brine (50 mL) and Na 2 SO 4 Dried and concentrated. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate = 5) to give a crude intermediate (28 g) as a yellow solid. The solid was dissolved in hydrobromic acid (48% aqueous, 60 mL) and refluxed for 1.5 h. The mixture was then stirred at room temperature overnight. The mixture was filtered. The filter cake was washed with water (10 mL. Times.5)And dried to give the title compound (3.0 g) as a yellow solid. The filtrates were combined and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with brine (20 mL) and Na 2 SO 4 Dried and concentrated to give a residue. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10) to give the title compound (1.20 g) as a white solid. The two batches were combined to give the title compound (XVI, 4.2g, yield 16%) as a yellow solid. LC-MS [ mobile phase: from 95% water and 5% CH% 3 CN to 5%water and 95% CH 3 CN in 2.5min],Rt=1.58min;Purity:100%;MS Calcd:226.0;MS Found:226.9[M+H] + .1H NMR(300MHz,CDCl 3 )δ:8.28(s,1H),7.85(d,J=1.8Hz,1H),6.63(d,J=1.5Hz,1H),6.46(s,1H),3.96(s,3H).
Step-46-bromo-4-methoxypyrazolo [1,5-a ]]Synthesis of pyridine-3-carbaldehyde (XVII):
Figure BDA0003967308590000331
to 6-bromo-4-methoxypyrazolo [1,5-a ] at 0 deg.C]To a solution of pyridine (XVI) (3.0 g, 13.2mmol) in N, N-dimethylformamide (75 mL) was added dropwise phosphorus oxychloride (6.10 g, 39.2mmol). After the addition was complete, the mixture was stirred at room temperature for 4 hours. The mixture was poured into ice water (100 mL) and stirred for 30 minutes. Aqueous sodium hydroxide (1 mol/L) was added dropwise until pH =9 the mixture was filtered. The filter cake was washed with water (5 mL. Times.2) and tert-butyl methyl ether (5 mL. Times.2). The solid was collected and dried to give the title compound XVII (3.2 g, yield 95%) as a white solid. 1 H NMR(400MHz,DMSO-d 6 )δ:10.21(s,1H),8.94(s,1H),8.47(s,1H),7.31(s,1H),4.08(s,3H).
Step-56-bromo-4-methoxypyrazolo [1,5-a ]]Synthesis of pyridine-3-carbaldehyde oxime (XVIII):
Figure BDA0003967308590000332
reacting 6-bromo-4-methoxyPyrazolo [1,5-a]Pyridine-3-carbaldehyde (XVII) (3.2g, 12.5 mmol) was dissolved in ethanol (80 mL) and stirred for 10 minutes. Water (40 mL) and hydroxylamine hydrochloride (1.3g, 18.8mmol) were added. The mixture was stirred at 55 ℃ for 4 hours. The mixture was concentrated to remove ethanol. The residue was filtered. The filter cake was washed with water (5 mL. Times.3) and tert-butyl methyl ether (5 mL. Times.3). The solid was collected and dried to give the title compound XVIII (2.8 g, yield 83%) as an off-white solid. 1 H NMR(400MHz,DMSO-d 6 )δ:11.42(br s,1H),8.74(s,1H),8.69(s,1H),7.96(s,1H),7.01(s,1H),4.03(s,3H).
Step-66-bromo-4-methoxypyrazolo [1,5-a ]]Synthesis of pyridine-3-carbonitrile (I-1):
Figure BDA0003967308590000333
reacting 6-bromo-4-methoxy pyrazolo [1,5-a ]]A suspension of pyridine-3-carbaldehyde oxime (XVII) (2.8g, 10.4mmol) and acetic anhydride (90 mL) was stirred at 120 ℃ overnight. The mixture was cooled and concentrated to give a residue. The residue was triturated with (petroleum ether: ethyl acetate =1, 5 ml) and filtered. The solids were collected and dried to give the title compound (I-1, 2.38g, 91% yield) as a grey solid. 1 H NMR(400MHz,DMSO-d 6 )δ:8.95(s,1H),8.60(s,1H),7.25(s,1H),4.04(s,3H).
Step-7Synthesis of tert-butyl 4- (5-methyl-4- (trimethylsilyl) ethynyl) pyridin-2-yl) piperazine-1-carboxylate (XXI-1):
Figure BDA0003967308590000334
tert-butyl 4- (4-iodo-5-methylpyridin-2-yl) piperazine-1-carboxylate (XIX-1) (3.3g, 8.19mmol), ethynyltrimethylsilane (0.96g, 9.83mmol) and triethylamine (1.65g, 16, 14mmol) were dissolved in a solution of tetrahydrofuran (30 mL), after which copper (I) iodide (78mg, 0.41mmol) was added and bis (triphenylphosphine) palladium (II) chloride (287mg, 0.41mmol) was added. After stirring at 40 ℃ for 2 days under nitrogen, the mixture was cooled and poured into water (50 mL).The mixture was extracted with ethyl acetate (30 mL. Times.2). The combined organic layers were washed with brine (20 mL) and Na 2 SO 4 Dried and concentrated to give a residue. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate = 20) to give the title compound XXI-1 (2.80 g, yield 92%) as a brown solid. 1 H NMR(300MHz,CDCl 3 )δ:8.03(s,1H),6.69(s,1H),3.53-3.51(m,4H),3.47–3.43(m,4H),2.25(s,3H),1.48(s,9H),0.26(s,9H).
Step-8Synthesis of tert-butyl 4- (4-ethynyl-5-methylpyridin-2-yl) piperazine-1-carboxylate (XXII-1):
Figure BDA0003967308590000341
to a mixture of tert-butyl 4- (5-methyl-4- ((trimethylsilyl) ethynyl) pyridin-2-yl) piperazine-1-carboxylate (XXI-1, 2.6g, 6.95mmol) in methanol (50 mL) was added potassium carbonate (4.8g, 6.95mmol). After stirring overnight at room temperature, the mixture was filtered. The filtrate was concentrated to give a residue. The residue was diluted with water (30 mL) and extracted with ethyl acetate (30 mL. Times.2). The combined organic layers were washed with brine (20 mL) and Na 2 SO 4 Dried and concentrated to give a residue. The residue was triturated with petroleum ether (20 mL) and filtered. The filter cake was dried to give the title compound (XXII-1, 1.42g, 68% yield) as a brown solid. 1 H NMR(400MHz,CDCl 3 )δ:8.05(s,1H),6.73(s,1H),3.53-3.44(m,8H),3.33(s,1H),2.27(s,3H),1.48(s,9H).
Step-96- (1-methyl-1H-pyrazol-4-yl) -4- ((5-methyl-2- (4-methylpiperazin-1-yl) pyridin-4-yl) ethynyl) pyrazolo [1,5-a]Pyridine-3-carbonitrile (2) Synthesis:
Figure BDA0003967308590000342
reacting 3-cyano-6- (1-methyl-1H-pyrazol-4-yl) pyrazolo [1,5-a]Pyridin-4-yl triflate (V-1, 1.11g, 2.99mmol), tert-butyl 4- (4-ethynyl-5-methylpyridin-2-yl) piperazineButyl 1-carboxylate (XVII-1, 0.90g, 2.99mmol), copper iodide (57mg, 0.30mmol) and N, N-diisopropylethylamine (772mg, 5.99mmol) were suspended in N, N-dimethylacetamide (15 mL), and bis (triphenylphosphine) palladium (II) chloride (210mg, 0.30mmol) was added. After stirring at 100 ℃ under nitrogen for 5 hours, the mixture was cooled and poured into water (50 mL). Dichloromethane (50 mL × 3) was added to extract the desired compound. The combined organic layers were washed with water (20 mL), brine (20 mL), and Na 2 SO 4 Dried and concentrated to give a residue. The residue was triturated with petroleum ether (30 mL) and filtered to give the crude product (1.1 g, crude). The crude product was further triturated with methanol (5 mL) and filtered to give Key intermedate-2 (920 mg, 59% yield) as a yellow solid. 1 H NMR(400MHz,CDCl 3 )δ:8.63(d,J=1.2Hz,1H),8.29(s,1H),8.10(s,1H),7.80(s,1H),7.71(d,J=1.2Hz,2H),7.21(s,1H),4.00(s,3H),3.54-3.52(m,8H),2.41(s,3H),1.49(s,9H).
Trifluoroacetic acid (5 mL) was added to a mixture of Key Intermediate-2 (900mg, 1.72mmol) in dichloromethane (20 mL). After stirring at room temperature for 1 hour, the mixture was concentrated to give a residue. The residue was diluted with dichloromethane (100 mL) and washed with saturated aqueous sodium bicarbonate (50 mL). The aqueous layer was extracted with dichloromethane (50 mL. Times.2). The combined organic phases were concentrated to give a residue. This residue was dissolved in (dichloromethane: methanol =2:1,100ml). Aqueous formaldehyde (37%, 0.5 mL) was added. After stirring at room temperature for 30 minutes, sodium cyanoborohydride (325mg, 5.16mmol) was added in portions. The mixture was stirred at room temperature overnight. The mixture was poured into water (30 mL) and extracted with dichloromethane (100 mL. Times.2). The combined organic phases were concentrated to give a residue. The residue was triturated with methanol (5 mL) and filtered to give the title compound (620 mg, 83% yield) as a yellow solid with a purity of 95%. The product was recrystallized from (chloroform: methanol =1, 15ml) to give the title compound 2 (500 mg, yield 67%) as a yellow solid with a purity of 96%. The product was recrystallized from (chloroform: methanol =3:1,15ml) to give a yellow solid of 97.3% purity (230 mg, yield 31%). LC-MS [ mobile phase: from 80% ]water (0.02% NH) 4 OAc)and 20% CH 3 CN to 30%water(0.02% NH 4 OAc)and 70% CH 3 CN in 6.5min],Rt=3.552min;Purity:97.35%(214nm),97.28%(254nm);MS Calcd:436.2;MS Found:437.2[M+H]+. 1 H NMR(400MHz,CDCl 3 )δ:8.63(d,J=1.2Hz,1H),8.28(s,1H),8.10(s,1H),7.80(s,1H),7.71(s,2H),7.20(s,1H),4.00(s,3H),3.56(t,J=5.2Hz,4H),2.53(t,J=5.2Hz,4H),2.40(s,3H),2.34(s,3H).
Examples 3 to 183
A wide variety of derivatives can be synthesized according to the methods of example 1 or example 2, provided that the appropriate starting materials, the appropriate corresponding intermediates, are changed. Examples 3-183 are some representative exemplary compounds thereof (see table 1).
TABLE 1 representative Compounds of formula I
Figure BDA0003967308590000351
Figure BDA0003967308590000361
Figure BDA0003967308590000371
Figure BDA0003967308590000381
Figure BDA0003967308590000391
Figure BDA0003967308590000401
Figure BDA0003967308590000411
Figure BDA0003967308590000421
Figure BDA0003967308590000431
Figure BDA0003967308590000441
Figure BDA0003967308590000451
Figure BDA0003967308590000461
Figure BDA0003967308590000471
Figure BDA0003967308590000481
Figure BDA0003967308590000491
Figure BDA0003967308590000501
Figure BDA0003967308590000511
Figure BDA0003967308590000521
Figure BDA0003967308590000531
Figure BDA0003967308590000541
Figure BDA0003967308590000551
Figure BDA0003967308590000561
Figure BDA0003967308590000571
Figure BDA0003967308590000581
Figure BDA0003967308590000591
Figure BDA0003967308590000601
Figure BDA0003967308590000611
Figure BDA0003967308590000621
Figure BDA0003967308590000631
Figure BDA0003967308590000641
Figure BDA0003967308590000651
Figure BDA0003967308590000661
Figure BDA0003967308590000671
Figure BDA0003967308590000681
Figure BDA0003967308590000691
Figure BDA0003967308590000701
Figure BDA0003967308590000711
Figure BDA0003967308590000721
Figure BDA0003967308590000731
Figure BDA0003967308590000741
Figure BDA0003967308590000751
Figure BDA0003967308590000761
Figure BDA0003967308590000771
In addition, with reference to the methods of example 1 or and example 2, a wider variety of derivatives can also be synthesized, provided that the starting materials are appropriately selected. For example, the compounds listed in Table 2 are some typical exemplary compounds.
TABLE 2 comparative exemplary Compounds of formula I
Figure BDA0003967308590000772
Figure BDA0003967308590000781
Figure BDA0003967308590000791
Figure BDA0003967308590000801
Figure BDA0003967308590000811
Figure BDA0003967308590000821
Figure BDA0003967308590000831
Figure BDA0003967308590000841
Figure BDA0003967308590000851
Figure BDA0003967308590000861
Example 241 biological experiments and pharmacodynamic analyses
1. Detection of kinase Activity
The kinase activity test has more literature reports, and related kinase detection kits are available. Products from Cisbio may be selected but are not limited to: HTRF kinEASE-STK KIT. Taking the detection of the HTRF (homogeneous time-resolved fluorescence) kinase detection kit on the NTRK kinase inhibitory activity as an example, the experimental operation steps are as follows:
1. the experimental method and the steps are as follows:
1.1 test compounds are formulated in 10mM (mmol/L) DMSO solution.
1.2 the compound at 10mM concentration was diluted with Kinase buffer (Kinase buffer) to 2.5. Mu.M (2.5 XCompound) working solution, then diluted in 3-fold gradient with 2.5. Mu.M as the highest concentration, and 2.5 Xworking solution of the test compound was serially diluted to 9 concentrations: 2.5, 0.833333, 0.277778, 0.092593, 0.030864, 0.010288, 0.003429, 0.001143, 0.000381 μ M; 10mM of control compound is diluted into 2.5 mu M of working solution by using kinase buffer, then the working solution is diluted by 3 times of gradient with 2.5 mu M as the highest concentration, and 2.5 times of working solution of the compound to be tested is continuously diluted to 9 concentrations, wherein the concentrations are respectively as follows: 2.5, 0.833333, 0.277778, 0.092593, 0.030864, 0.010288, 0.003429, 0.001143, 0.000381 μ M.
1.3 mu.L of 2.5 Xcompound working solution was added to a 384 well plate (Greiner, cat # 781280), blank wells (no compound and kinase added) and Control wells (no compound added to kinase only) were placed, and 4. Mu.L of kinase buffer was added to the Blank and Control wells.
1.4 preparing the corresponding 5 Xworking solution from the kinase stock solution by using a kinase buffer, adding 2 mu L of 5 Xkinase working solution into each hole containing the compound working solution, adding 2 mu L of kinase buffer into a Blank hole, and adding 2 mu L of 5 Xkinase working solution into a Control hole.
1.5 mu.L of 5 Xsubstrate stock (TK Antibody-Cryptate) was added to each well containing the compound and kinase working solution, 2. Mu.L of kinase buffer was added to the Blank well, and 2. Mu.L of 5 Xsubstrate stock was added to the Control well.
1.6 Add 2. Mu.L of ATP working solution (5X) to each well.
1.7 incubation of 384-well plates with sealed membranes at 37 ℃ for 1 hour, followed by addition of 5. Mu.L (4X) of reaction stop solution (Streptavidin-XL 665) to each well;
1.8 the 384 well plates were further sealed and incubated at 37 ℃ for 1 hour before detecting 665, 620 signal values on a 2104EnVision plate reader.
Preparing a detection system working solution:
Figure BDA0003967308590000862
Figure BDA0003967308590000871
2. and (3) data analysis:
the Inhibition Rate (IR) of the test compound was calculated by the following formula: IR (%) = (RLU CTR (665/620) -RLU compound (665/620))/(RLU CTR (665/620) -RLU BLANK (665/620)) × 100%. At ExInhibition rates were calculated for compounds at different concentrations in cel, and IC was calculated using GraphPad Prism5 software 50
Results of detection of inhibition of TRKa, TRKb, TRKc kinase Activity
Some biological activity test results are shown in Table 3
TABLE 3 kinase inhibitory Activity of Compounds
Compound (I) TRKa IC 50 (μM) TRKb IC 50 (μM) TRKc IC 50 (μM)
1 0.098 0.019 0.155
2 0.049 0.010 0.341
3 0.020 0.014 0.784
Similarly, the activity of other compounds on TRKa, TRKb and TRKc kinase is inhibited, and the inhibition effect is better.
Meanwhile, the compound of the invention has obvious inhibition effect on other kinases, such as TRK, RET, RAF, FGHR, PDGFR, VEGFR and other kinases.
Example 242 tumor cell inhibitory Activity GI 50 Determination of value
CELL activity assay CTG (CELL TITER-GLO) luminescence method was used to assay the activity of target compounds. The principle is as follows: ATP adenosine triphosphate (adenosine triphosphate for short) participates in a plurality of enzymatic reactions in organisms, is an index of living cell metabolism, the content of ATP adenosine triphosphate directly reflects the number and the state of cells, and in the experimental process, equal volume of CellTiter-Glo is added into a cell culture medium TM The reagent is used for measuring a luminous value, in an optical signal and system, the luminous value is in direct proportion to the amount of ATP, the ATP is in positive correlation with the number of living cells and in inverse proportion to the activity of the anti-tumor medicament, and anti-proliferation activity data of the compound on the tumor cells can be obtained by detecting a fluorescence signal of the ATP according to a calculation formula.
The test of tumor cell inhibitory activity was carried out in two parts: using kinase as a target spot, and checking a target compound on human thyroid duct cancer cells (TT) and human colon cancer cells (KM 12); to determine the activity on NTRK Fusion, the growth inhibitory activity of the target compound on NTRK Fusion cells was specifically examined.
Reagents used for the experiment: F-12K basal medium (ATCC, 30-2004), fetal bovine serum (Corning, 35-076-CV), diabody (GIBCO, 15240-062), pancreatin (GIBCO, 25200072), DMSO (SIGMA, D2650), DMEM basal medium (Corning, 10-013-CV), fetal bovine serum (Gibco, 10091-148).
1. Activity measurement of compound on human thyroid duct cancer cell and human colon cancer cell
The following methods were used to determine the effect of compounds on tumor cell proliferation by using CTG luminescence.
The specific experimental operation method and the flow are as follows:
1.1, cell recovery:
immediately placing the frozen cells in a constant-temperature water bath at 37 ℃ from a liquid nitrogen storage tank, shaking for 2min, transferring the cell suspension into a 15mL centrifuge tube after the cell frozen solution is completely melted, slowly adding 4mL culture solution, centrifuging (1000 r/min,5 min), discarding the supernatant, sucking the stock solution, adding 5mL of the culture medium, gently blowing to obtain single cell suspension, transferring the single cell suspension into a culture bottle, and placing the single cell suspension into an incubator for culture.
1.2, cell culture:
cells were run out of whole medium at 37 ℃ in 5% CO 2 The incubator of (2) for cultivation. Cells were plated at regular passage and at logarithmic growth phase.
1.3, cell plating:
cell staining was performed with trypan blue and viable cells were counted, the cell concentration was adjusted to a cell sap of an appropriate plate concentration (TT: 50000cell/mL, KM12:35000 cell/mL), 90. Mu.L of cell suspension was added to each well of a 96-well culture plate (Corning, 3599), and blank control wells and vehicle control wells were set. Cell-containing medium was added to the blank control wells and cell-free medium was added to the vehicle control wells. The plates were then placed at 37 ℃ in 5% CO 2 And overnight in an incubator at 100% relative humidity.
1.4, compound preparation:
compounds were weighed into 10mM stock solutions in DMSO, and test compound stocks were diluted on formulation plates (Beaver, suzhou) in serum-free medium to a final concentration of 100 μ M in 10 × compound working solution (including controls). The solution was diluted with serum-free medium in 3-fold concentration gradient to obtain 9 concentration gradient 10 × compound working solution, the compound concentrations were 100, 33.33, 11.11, 3.70, 1.23, 0.411, 0.137, 0.046 and 0.015 μ M, respectively.
1.5 addition of Compounds
Different concentration gradients of 10 x compound working solution were added to 96 well cell culture plates at 10 μ L/well, 10 μ L DMSO-cell culture solution mixture was added to vehicle control wells and blank control wells at a final DMSO concentration of 0.1%, with 2 replicate wells set at each concentration. The 96-well cell plates were returned to 37 ℃ and cultured in a 5% carbon dioxide incubator for 5 days.
1.6, CTG detection:
the cell culture plate was removed and allowed to equilibrate to room temperature for 30 minutes, 50. Mu.L (equal to half the volume of the cell culture solution in each well) of CellTiter-Glo working solution was added to each well, the plate was wrapped with aluminum foil paper to protect from light, the plate was shaken on an orbital shaker for 2 minutes to induce cell lysis, the plate was allowed to stand at room temperature for 10 minutes to stabilize the luminescence signal, and the luminescence signal was detected on a 2104EnVision plate reader.
1.7, data analysis
The Inhibition Rate (IR) of the test compound was calculated by the following formula: IR (%) = (1- (RLU compound-RLU blank)/(RLU vehicle control-RLU blank)). 100%), and finally the IC of the compound inhibiting cell proliferation was obtained by nonlinear regression analysis using the log-inhibition rate of compound concentration in Graphpad prism5 software 50 The value is obtained.
Results the results are shown in table 4.
Results of measurement of growth inhibitory Activity of the obtained Compounds on TT cells and KM12 cells
Figure BDA0003967308590000881
Figure BDA0003967308590000891
Figure BDA0003967308590000901
Figure BDA0003967308590000911
Figure BDA0003967308590000921
Figure BDA0003967308590000931
EXAMPLE 243 determination of Activity of Compounds on hybridoma cell lines
In vitro anti-tumor inhibition activity the activity of the target compound on NTRK Fusion is specifically detected by using NTRK Fusion engineering cell strains besides related tumor cell strains. The following method was used to determine the effect of compounds on the proliferation of cells engineered for NTRK fusion, and was performed by using CTG luminescence. For NTRK fusion, baF3 ETV6-NTRK3 fusion engineered cell lines (constructed by Precedo Co.) were used. Culturing according to corresponding conditions.
Reagents used for the experiment: RPMI-1640 basic medium (GIBCO, 22400-089), fetal bovine serum (SH 30084.03 ), diabody (GIBCO, 15240-062), pancreatin (GIBCO, 25200072), DMSO (SIGMA, D2650).
The experimental operating method is as follows:
2.1, recovering cells:
immediately placing the frozen cells in a liquid nitrogen storage tank, shaking in a constant-temperature water bath at 37 ℃ for 2min until the frozen cell liquid is completely melted, transferring the cell suspension into a 15mL centrifuge tube, slowly adding 4mL culture solution, centrifuging (1000 r/min,5 min), removing the supernatant, sucking the stock solution, adding 5mL of the culture medium, slightly blowing and beating to obtain a single-cell suspension, transferring the single-cell suspension into a culture bottle, and placing the single-cell suspension into an incubator for culture.
2.2, cell culture:
the Ba/F3-ETV6-NTRK3 engineered cells were subjected to the complete medium (RPMI-1640 +10% FBS +1% P/S) at 37 ℃,5% CO 2 The incubator of (2) for cultivation. Cells in logarithmic growth phase were taken for plating after regular passage.
2.3, cell plating:
cell staining was performed with trypan blue and viable cells were counted, the cell concentration was adjusted to 30000 cells/mL of cell fluid, 90 μ L of cell suspension was added to each well of a 96-well culture plate (Corning, 3599), and blank control wells and vehicle control wells were set. Cell-containing medium was added to the blank control wells and cell-free medium was added to the vehicle control wells. Then will beThe culture plate was placed at 37 ℃ and 5% CO 2 And overnight in an incubator at 100% relative humidity.
2.4, preparing a compound:
compounds were weighed into DMSO as 10mM stock solutions and test compound stocks were diluted to 100 μ M final concentration 10 × compound working solutions (including controls) in serum-free medium on formulation plates (Beaver, suzhou). The solution was diluted with serum-free medium in 3-fold concentration gradient to obtain 9 concentration gradient 10 × compound working solution, the compound concentrations were 100, 33.33, 11.11, 3.70, 1.23, 0.411, 0.137, 0.046 and 0.015 μ M, respectively.
2.5, addition of Compound:
different concentration gradient of 10 x compound working solution is added to 96 hole cell culture plate, 10 u L/hole, in the vehicle control hole and blank control hole to add 10 u L DMSO-cell culture solution mixture, DMSO final concentration is 0.1%, each concentration set up 2 multiple hole. The 96-well cell plates were returned to 37 ℃ and cultured in a 5% carbon dioxide incubator for 5 days.
2.6, CTG detection:
the cell culture plate was removed and allowed to equilibrate to room temperature for 30 minutes, 50. Mu.L (equal to half the volume of the cell culture solution in each well) of CellTiter-Glo working solution was added to each well, the plate was wrapped with aluminum foil paper to protect from light, the plate was shaken on an orbital shaker for 2 minutes to induce cell lysis, the plate was allowed to stand at room temperature for 10 minutes to stabilize the luminescence signal, and the luminescence signal was detected on a 2104En Vision plate reader.
2.7, data analysis:
the Inhibition Rate (IR) of the test compound was calculated by the following formula: IR (%) = (1- (RLU compound-RLU blank)/(RLU vehicle control-RLU blank)). 100%), and finally, nonlinear regression analysis was performed in Graphpad prism5 software using the logarithmic inhibition rate of compound concentration to obtain IC of the compound for inhibiting cell proliferation 50 The value is obtained.
Results the results of the tests are shown in Table 5.
Results of the detection of the growth inhibitory Activity of the Compounds obtained in Table 5 on fused tumor cells
Figure BDA0003967308590000941
Figure BDA0003967308590000951
The results show that the target compound has obvious inhibitory activity on NTRK Fusion. The rest of the compound data are not provided, but all have more remarkable inhibitory effect.
Example 244 in vivo antitumor activity of compounds of the invention:
a fraction of the compound with strong in vitro activity and low toxicity was selected to determine the Maximum Tolerated Dose (MTD) in mice. The in vivo antitumor activity of the compound of the present invention is measured on a model of human cancer nude mouse xenograft tumor, and the administration dose, administration route, administration frequency and period of the test compound for generating the drug effect are explored.
And (3) breeding female BALB/C nude mice with the age of 5-6 weeks, wherein the weight of the female BALB/C nude mice is about 18-20 g.
The subcutaneous allografting tumor of the cell has the drug effect in a BALB/c nude mouse model and the drug effect evaluation is carried out.
Constructing a human cancer nude mouse xenograft tumor model: TT (human thyroid cancer cell) culture, digesting and removing the wall of the tumor cells cultured in a single layer, collecting and suspending the tumor cells in a serum-free culture medium, and adjusting the concentration to 5X 10 6 0.2mL, carried in an ice box to an animal room, and transplanted into the scapular part under the back of the left axilla of the nude mouse by 5 multiplied by 10 6 Measuring the tumor formation volume every 2-3 days, selecting tumor-bearing nude mice with vigorous tumor growth and no rupture, taking out the tumor under aseptic condition, shearing tumor tissue into diameter of about 2-3mm, inoculating to the subcutaneous part of scapula at the back of left axilla of the nude mice, and after three generations, when the tumor volume grows to 100-150mm 3 The nude mice with over-large or under-small tumor masses are randomly divided and administered.
5 groups were randomly divided, including a negative control group (vehicle), a positive control group (Blu-667, 10mg/kg), a high, medium and low treatment groups (5 mg/kg,10mg/kg and 40mg/kg respectively, wherein the high dose is lower than MTD), and 5 nude mice in each group were administered by intraperitoneal injection twice a day for 3 weeks. During this period, animal body weight, tumor volume and animal mortality were measured every 3 days. Animals were sacrificed 24 hours after the last administration, tumor volume size, tumor weight, nude mouse body weight were measured, tumor volume growth curve, nude mouse body weight growth curve and tumor inhibition rate were plotted, animal mortality was calculated, and relative tumor proliferation rate T/C (%) was calculated according to the formula T/C (%) = TRTV/CRTV 100%. (TRTV: treatment RTV; CRTV: negative control RTV, relative tumor volume RTV = Vt/V0, where V0 is the tumor volume at the time of the group administration and Vt is the tumor volume after the administration). The in vivo anti-tumor effect of the compound of the invention has a relative tumor proliferation rate T/C (%) less than or equal to 40% and TGI% more than 80% (see figure 1 for in vivo anti-tumor activity of compound 171 in TT model). And the difference has statistical significance, has obvious pharmacodynamic action and shows good in-vivo tumor inhibition effect. At the same time, the animals with different dose groups of the compound 171 of the present invention showed no significant weight loss and showed good tolerance.
In this experiment. Compound 171 was evaluated for in vivo efficacy in a TT cell xenograft tumor model with Blu-667 as a control. The experimental result shows that the compound 171 is gavaged at the dose of 30mg/kg, the T/C is less than 40 percent, and the compound shows good in-vivo tumor inhibition effect. Similarly, other compounds have similar in vivo tumor-inhibiting effects.
In vivo antitumor activity in addition to the tumor cell lines, engineered lines, such as KIF5B-RET fusion tumor cell lines, are also used.
The details of the particular examples described in this disclosure are not to be interpreted as limitations. Various changes, synonyms and modifications can be made without departing from the spirit and scope of the invention, and it is known that embodiments of such changes, synonyms and modifications are a part of the present invention.

Claims (12)

1. A pyrazolo [1,5-a ] pyridine derivative, which is a compound represented by the general formula (I):
Figure FDA0003967308580000011
in the above-mentioned general formula (I),
R 1 selected from: a hydrogen atom, carboxyl, nitro, amino, cyano, acyl, alkyl, alkoxy, alkenyl, alkynyl, halogen, isotope, haloalkenyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl; any of the foregoing groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, and alkoxyalkyl;
L 1 selected from the group consisting of: covalent bond, -CH 2 -CH 2 -、-CH=CH-、-C≡C-、-NH-、
Figure FDA0003967308580000012
Figure FDA0003967308580000013
Any of the above groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, = O, -CF 3 Alkyl, haloalkyl, hydroxy, alkoxy, and alkoxyalkyl;
L 2 selected from: a covalent bond, alkylene, heteroalkylene, carbonyl, -C (O) -NH-, -alkyl-NH-C (O) -, -alkyl-O-; any of the above groups each independently may be substituted by one or moreSubstituents including hydrogen atoms, isotopes, alkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, heteroaryl;
L 3 selected from: a covalent bond, alkylene, heteroalkylene, carbonyl, -O-, -NH-, -C (O) -NH-, -NH-C (O) -, -alkyl-C (O) -NH-, -alkyl-NH-C (O) -, -alkyl-NH-, -alkyl-O-; any of the above groups may each independently be substituted with one or more substituents including hydrogen, isotopes, alkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, heteroaryl;
R 2 selected from: a hydrogen atom, an alkyl group, a heteroalkyl group, an alkenyl group, an alkynyl group, an arylalkyl group, a cycloalkyl group, an aryl group, an arylalkyl group, a heteroaryl group, a heteroarylalkyl group, a heterocycloalkyl group, an alkylether group, a heteroalkylether group, an arylalkylether group, a cycloalkylether group, an arylene ether group, a heteroarylalkylether group, a carboxyl group, a carboxyalkylaminocarbonyl group, a cycloalkylaminocarbonyl group, a heteroarylaminocarbonyl group; any of the above groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl;
R 3 selected from the group consisting of: <xnotran> , , , , , , , , , , , , , , , , , , , , , , , , , , , , </xnotran>Carboxyalkylaminocarbonyl, cycloalkylaminocarbonyl, heteroarylaminocarbonyl; any of the foregoing groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, carboxyl, phenyl, benzyl, phenyloxy, = O, -CF 3 Haloalkyl, alkyl, alkenyl, alkynyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, heteroalkyl, arylalkyl, cycloalkyl, aryl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkenyl, alkoxyalkyl, alkenyloxy, alkynyloxy, alkylamino, aminoalkyl, alkylaminocarbonyl, sulfonyl, alkylsulfonyl, alkylsulfinyl, or aminosulfonyl.
2. Pyrazolo [1,5-a ] according to claim 1]Pyridine derivatives characterized in that R 1 Is hydrogen atom, carboxyl, nitro, amino, cyano, acyl, alkyl, alkoxy, alkenyl, alkynyl, halogen, isotope, haloalkenyl, heteroalkyl, aryl, heteroaryl; in the above groups, each may be unsubstituted or substituted with one or more substituents including: halogen, isotope, = O, -CF 3 Alkyl, alkenyl, alkynyl, carboxyl, hydroxyl, hydroxyalkyl, alkoxy, alkoxyalkyl, -CN.
3. Pyrazolo [1,5-a ] according to claim 1]Pyridine derivative characterized in that L 3 Is selected from
Figure FDA0003967308580000021
O-CH 2 -R 4 (ii) a Wherein R is 4 Is selected from
Figure FDA0003967308580000031
Any of the foregoing groups independently may be substituted with one or more substituents including, but not limited to, halogen, isotope, = O, -CF 3 Alkyl, haloalkyl, hydroxy, alkoxyAnd alkoxyalkyl groups.
4. Pyrazolo [1,5-a according to claim 1]Pyridine derivatives characterized in that R 3 Selected from the group consisting of C1-C5 alkyl, heterocyclyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl; any of the foregoing groups, independently of each other, may be unsubstituted or substituted with one or more substituents including, but not limited to, halogen, isotope, amino, alkyl.
5. Pyrazolo [1,5-a ] pyridine derivative according to claim 1, having a structure which may be selected from one of the following structures, or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof:
Figure FDA0003967308580000032
Figure FDA0003967308580000041
Figure FDA0003967308580000051
Figure FDA0003967308580000061
Figure FDA0003967308580000071
Figure FDA0003967308580000081
Figure FDA0003967308580000091
Figure FDA0003967308580000101
6. pyrazolo [1,5-a according to any one of claims 1 to 5]A process for the preparation of pyridine derivatives, characterized in that L 2 The preparation method comprises the following steps:
s1, selecting I as a raw material, and removing methyl under the action of a catalyst to obtain a compound II;
s2, reacting the compound II with a boron compound III under catalysis to obtain IV;
s3, converting the compound IV into a compound V under the action of N-phenyl bis (trifluoromethanesulfonimide);
s4, condensing the compound VI and the compound VII to obtain a compound VIII;
s5, reacting the compound VIII with the compound V to obtain a compound IX, namely a target compound shown in the same general formula (I);
Figure FDA0003967308580000102
7. pyrazolo [1,5-a according to claims 1-5]A process for the preparation of pyridine derivatives, characterized in that compound I-1 is:
Figure FDA0003967308580000111
the synthesis method of the compound I-1 comprises the following steps:
A. taking X as a raw material, and carrying out acylation and ammoniation to obtain XIV;
B. under the action of acid, XIV is cyclized to obtain a compound XVI;
C. introducing formyl on the compound XVI, converting the formyl into a nitrile group through hydroxylamination, and removing methyl under the action of a catalyst to obtain a compound I-1;
Figure FDA0003967308580000112
8. pyrazolo [1,5-a according to any one of claims 1 to 5]A process for the preparation of pyridine derivatives, characterized in that L 1 = CH or-C ≡ C-, L 2 -R 2 For substituted phenyl radicals, said pyrazolo [1,5-a ]]The preparation method of the pyridine derivative comprises the following steps:
1) Taking an iodo compound XIX as a raw material, and reacting with XX under the action of a catalyst to obtain a compound XXI;
2) Reacting a compound XXI in an alkaline methanol solution to obtain XXII;
3) XXII reacts with compound V to obtain compound XXIII;
Figure FDA0003967308580000113
9. a pharmaceutical composition comprising a pyrazolo [1,5-a ] pyridine derivative according to any one of claims 1 to 5, an optical isomer thereof, or a pharmaceutically acceptable salt thereof, preferably wherein the pyrazolo [1,5-a ] pyridine derivative, an optical isomer thereof, or a pharmaceutically acceptable salt thereof is in any form together with a pharmaceutically acceptable diluent, excipient, or carrier; further preferably, the pharmaceutical composition further comprises one or more other drugs for combination therapy.
10. Use of a pyrazolo [1,5-a ] pyridine derivative according to any one of claims 1 to 5, an optical isomer thereof, or a pharmaceutically acceptable salt thereof for inhibiting the activity of a kinase; the activity of inhibiting the kinase refers to inhibiting the activity of TRK or RET, FGHR, PDGFR and VEGFR.
11. Use of a pyrazolo [1,5-a ] pyridine derivative according to any one of claims 1 to 5, an optical isomer thereof, or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a disorder caused by, associated with or accompanied by disruption of cell proliferation and/or angiogenesis; preferably, the disorder is a proliferative disease; further preferably, the proliferative disease is cancer.
12. Use according to claim 11, wherein the condition is selected from: proliferative diseases including non-small cell lung cancer, renal cell carcinoma, gastric cancer, hepatocellular carcinoma, colorectal cancer, medullary thyroid carcinoma, follicular thyroid carcinoma, undifferentiated thyroid carcinoma, papillary thyroid carcinoma, brain tumor, peritoneal cavity cancer, solid tumors, other lung cancers, head and neck cancers, gliomas, neuroblastoma, von Hipple-Lindau syndrome and renal tumors, breast cancer, fallopian tube cancer, ovarian cancer, transitional cell carcinoma, prostate cancer, cancers at the junction of the esophagus and stomach, biliary tract cancers and adenocarcinomas, and any malignancy with increased kinase activity of any one or more of TRK, RET, FGHR, PDGFR, VEGFR, and the like; neurodegenerative diseases, including: huntington's disease, polyglutamine disease, parkinson's disease, alzheimer's disease, seizures, striatal substantia nigra degeneration, progressive supranuclear palsy, torsion dystonia, spastic torticollis and dyskinesia, familial tremor, gilles de la tourette's syndrome, diffuse lewy body disease, pick's disease, intracranial hemorrhage, primary lateral sclerosis, spinal muscular atrophy, amyotrophic lateral sclerosis, hypertrophic interstitial polyneuropathy, retinitis pigmentosa, hereditary optic atrophy, hereditary spastic paraplegia, progressive ataxia and Shy-Drager syndrome; a metabolic disease comprising: type 2 diabetes; an ocular degenerative disease comprising: glaucoma, age-related macular degeneration, rubeosis iridis glaucoma: diseases involving angiogenesis, including: cancer, psoriasis; a psychological disorder, including: bipolar disorder, schizophrenia, mania, depression and dementia; cardiovascular diseases include: heart failure, restenosis and arteriosclerosis; fibrotic diseases, including:
liver fibrosis, cystic fibrosis, and vascular fibromyalgia: infectious diseases, including: and (4) fungal infection. For example: candida albicans, bacterial infection, viral infection. For example: herpes simplex, protozoal infections, such as: malaria, leishmania infections, trypanosoma brucei infections, toxoplasmosis and coccidiosis, and hematopoietic disorders including: marine anemia, and sickle cell anemia.
CN202211500443.8A 2021-12-03 2022-11-28 Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof Pending CN115772170A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021114683028 2021-12-03
CN202111468302 2021-12-03

Publications (1)

Publication Number Publication Date
CN115772170A true CN115772170A (en) 2023-03-10

Family

ID=85390588

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211500443.8A Pending CN115772170A (en) 2021-12-03 2022-11-28 Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN115772170A (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104114553A (en) * 2011-12-12 2014-10-22 雷迪博士实验室有限公司 Substituted pyrazolo[1,5-a] pyridine as tropomyosin receptor kinase (Trk) inhibitors
WO2016007722A1 (en) * 2014-07-10 2016-01-14 Incyte Corporation Triazolopyridines and triazolopyrazines as lsd1 inhibitors
CN108349969A (en) * 2015-07-16 2018-07-31 阵列生物制药公司 Substituted pyrazolo [1,5-a] pyridine compounds as RET kinase inhibitors
WO2019157879A1 (en) * 2018-02-13 2019-08-22 北京诺诚健华医药科技有限公司 Heterocyclic compound which acts as trk inhibitor
CN113490666A (en) * 2018-12-19 2021-10-08 奥瑞生物药品公司 Substituted pyrazolo [1,5-A ] pyridine compounds as inhibitors of FGFR tyrosine kinases

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104114553A (en) * 2011-12-12 2014-10-22 雷迪博士实验室有限公司 Substituted pyrazolo[1,5-a] pyridine as tropomyosin receptor kinase (Trk) inhibitors
WO2016007722A1 (en) * 2014-07-10 2016-01-14 Incyte Corporation Triazolopyridines and triazolopyrazines as lsd1 inhibitors
CN108349969A (en) * 2015-07-16 2018-07-31 阵列生物制药公司 Substituted pyrazolo [1,5-a] pyridine compounds as RET kinase inhibitors
WO2019157879A1 (en) * 2018-02-13 2019-08-22 北京诺诚健华医药科技有限公司 Heterocyclic compound which acts as trk inhibitor
CN113490666A (en) * 2018-12-19 2021-10-08 奥瑞生物药品公司 Substituted pyrazolo [1,5-A ] pyridine compounds as inhibitors of FGFR tyrosine kinases

Similar Documents

Publication Publication Date Title
US11827644B2 (en) Pyrazine derivative and application thereof in inhibiting SHP2
EP2513114B1 (en) Pyrrolo[2,3-d]pyrimidine compounds
WO2021169990A1 (en) Kras inhibitors for treating cancers
TW202120488A (en) Compound as shp2 inhibitor and use thereof
JP6986032B2 (en) Crystals of pyrrolopyrimidine compounds as JAK inhibitors
US9604935B2 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing the same
KR100818060B1 (en) Amide compounds and use thereof
WO2017035753A1 (en) 2-arylamino pyridine, pyridine or triazine derivative, preparation method and use thereof
US11267815B2 (en) Class of amino-substituted nitrogen-containing fused ring compounds, preparation method therefor, and use thereof
CN103204825B (en) Benzothiazole compounds as protein kinase inhibitors, and preparation method and application thereof
TW479058B (en) 2,7-substituted octahydro-pyrrolo[1,2-a]pyrazine derivatives
JP2019510055A (en) Compounds for the inhibition of cyclophilin and their use
CN116096372A (en) EGFR inhibitor, preparation method and pharmaceutical application thereof
CN115772170A (en) Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof
CN110283174B (en) PI3K delta inhibitor and application thereof
CN106565657A (en) Hesperetin cinnamate compound with anti-tumor activity and synthetic method thereof
TW201240666A (en) Piperazinedione compounds
CN110950848B (en) Synthesis and application of novel aminopyrazole derivative
CN115925684A (en) Pyrimidine derivative and preparation method and application thereof
CN115701429B (en) 4- (1H-indol-1-yl) pyrimidine-2-amino derivative, and preparation method and application thereof
CN117624179A (en) 3- (amino) methylene indoline derivative and preparation method and application thereof
CN117700397A (en) 2-oxo indoline derivative and preparation method and application thereof
CN117624014A (en) 3- (indolylamino) methylene indoline derivative and preparation method and application thereof
WO2022199045A1 (en) Bicyclic heterocyclic fgfr4 inhibitor, pharmaceutical composition and preparation comprising same, and application thereof
CA3240780A1 (en) Heterocyclic compound having anti-tumor activity and use thereof

Legal Events

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