Novel RET inhibitors, pharmaceutical compositions thereof and uses thereof

文档序号:1810418 发布日期:2021-11-09 浏览:14次 中文

阅读说明:本技术 新型的ret抑制剂、其药物组合物及其用途 (Novel RET inhibitors, pharmaceutical compositions thereof and uses thereof ) 是由 谢洪明 罗明 张英俊 胡扬校 王凯 寇玉辉 于 2021-05-07 设计创作,主要内容包括:本发明属于药物领域,涉及一种新型的RET抑制剂、其药物组合物及其用途。具体地,本发明涉及一种式(I)所示的化合物,或式(I)所示化合物的立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或前药,本发明还涉及包含所述这些化合物的药物组合物、以及所述这些化合物及其药物组合物在制备药物中的用途,该药物尤其用于治疗和预防与RET相关的疾病和病症,包括癌症、肠易激综合征和/或与肠易激综合征相关的疼痛。(The invention belongs to the field of medicines, and relates to a novel RET inhibitor, a pharmaceutical composition thereof and application thereof. Specifically, the invention relates to a compound shown in formula (I), or a stereoisomer, a tautomer and a nitrogen oxide of the compound shown in formula (I)Solvates, metabolites, pharmaceutically acceptable salts or prodrugs thereof, as well as to pharmaceutical compositions comprising said compounds and to the use of said compounds and pharmaceutical compositions thereof in the manufacture of a medicament, particularly for the treatment and prevention of diseases and disorders associated with RET, including cancer, irritable bowel syndrome and/or pain associated with irritable bowel syndrome.)

1. A compound that is a compound of formula (I) or a stereoisomer, tautomer, nitroxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof:

wherein the content of the first and second substances,

X1、X2、X3、X4and X5Each independently is CR4Or N;

e is a bond, -NR6-or-O-;

ring A is 3-12 membered carbocyclyl OR 3-12 membered heterocyclyl, wherein ring A is optionally substituted with 1,2,3 OR 4 substituents selected from-F, -Cl, -Br, -OR5、-NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-, oxo, C1-6Aminoalkyl radical, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C1-6Hydroxyalkyl, 3-12 membered carbocyclyl, 3-12 membered heterocyclyl, (3-12 membered heterocyclyl) -C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl and C1-6Alkylthio group substituent;

R1is-H, -D, -CN, -F, -Cl, -Br, -NH2Methyl or cyclopropyl, whereinThe methyl and cyclopropyl groups can be independently and optionally substituted by 1,2,3 or 4 groups selected from-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

R2is-H, -D, C1-6Alkyl, 3-12 membered heterocyclyl, 3-12 membered carbocyclyl, (3-12 membered heterocyclyl) -C1-6Alkyl, (3-12 membered carbocyclyl) -C1-6Alkyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C1-6Aminoalkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkylamino radical C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl radical, R5O(C=O)NR6-(C1-6Alkyl), R5(C=O)NR6-(C1-6Alkyl), NR)5R6(C=O)-(C1-6Alkyl) or R5(C=O)-(C1-6Alkyl) of said C1-6Alkyl, 3-12 membered heterocyclyl, 3-12 membered carbocyclyl, (3-12 membered heterocyclyl) -C1-6Alkyl, (3-12 membered carbocyclyl) -C1-6Alkyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C1-6Aminoalkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkylamino radical C1-6Alkyl and C1-6Alkoxy radical C1-6Alkyl, and R5O(C=O)NR6-(C1-6Alkyl), R5(C=O)NR6-(C1-6Alkyl), NR)5R6(C=O)-(C1-6Alkyl) and R5(C=O)-(C1-6Alkyl) each independently optionally substituted by 1,2,3 or 4 alkyl moieties selected from-F, -Cl, -Br, -OH, oxo, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkoxy radical C1-6Alkyl radical, R5(C ═ O) -and R5O (C ═ O) -;

R3is-H, -D, NR5R6(C=O)-、C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C6-10Aryl radical C1-6Alkyl, (3-10 membered carbocyclyl) -C1-6Alkyl, (3-10 membered heterocycle)Radical) -C1-6Alkyl or C1-6Aminoalkyl radical, wherein said C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C6-10Aryl radical C1-6Alkyl, (3-10 membered carbocyclyl) -C1-6Alkyl, (3-10 membered heterocyclyl) -C1-6Alkyl and C1-6Aminoalkyl is each independently optionally substituted by 1,2,3 or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NR5R6、-OR7、C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, halo C1-6Alkoxy radical, C6-10Aryl radical, C1-6Alkoxy radical C1-6Alkyl, oxo, C1-6Alkyl acyl, 3-10 membered heterocyclic group and 3-10 membered carbocyclyl;

each R4Independently is-H, -D, -F, -Cl, -Br, methyl, ethyl, n-propyl, methoxy or ethoxy, wherein said methyl, ethyl, n-propyl, methoxy and ethoxy may be independently optionally substituted with 1,2,3 or 4 groups selected from-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

each R5Independently is-H, -D, -NH2Methyl, ethyl, n-propyl, isopropyl, n-butyl, cyclopropyl, cyclopentyl, pyrrolidinyl, phenyl, or pyrazolyl; wherein said methyl, ethyl, n-propyl, cyclopropyl, cyclopentyl, pyrrolidinyl, phenyl, and pyrazolyl are each independently optionally substituted with 1,2,3, or 4 substituents selected from-F, -Cl, -Br, -OH, -NH2Methyl, -S (═ O)2CH3Methoxy, ethoxy, phenoxy and phenyl;

each R6independently-H, -D, methyl, ethyl, n-propyl, n-butyl, methoxymethyl, ethoxymethyl or methoxyethyl, wherein said methyl, ethyl, n-propyl, n-butyl, methoxymethyl, ethoxymethyl and methoxyethyl are each independently optionally substituted by 1,2,3 or 4 substituents selected from the group consisting of-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

each R7independently-OH, methyl, ethyl, cyclopropyl or phenyl.

2. The compound of claim 1, wherein,

ring a is of sub-structural formula:

wherein Z is1、Z2、Z4And Z6Each independently is CH or N;

each Z3And Z5Independently is a bond, CH2O, S, NH, C O, S ═ O or S (═ O)2

m is 0, 1 or 2;

t, n and t1 are each independently 0 or 1;

and each sub-formula of ring A is optionally substituted by 1,2,3 OR 4 groups selected from-F, -Cl, -Br, -OR5Oxo, -NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-、C1-4Aminoalkyl radical, C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl and C1-4Alkylthio groups.

3. The compound of claim 1, wherein,

ring a is of sub-structural formula:

wherein each sub-formula of ring A is independently optionally substituted by 1,23 or 4 are selected from-F, -Cl, -Br, -OH, oxo, -NH2、-NHCH3、-N(CH3)2、CH3(C ═ O) NH-, phenyl (C ═ O) NH-, NH2(C ═ O) NH-, methoxyphenyl (C ═ O) -, phenyl S (═ O)2NH-, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CF3Hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclohexyl, pyrrolidinyl, piperidinyl, tetrahydrofuranyl, phenoxyethoxy, methoxyethoxy, chloroethoxy, hydroxyethoxy, and ethylthio.

4. The compound of claim 1, wherein,

R2is-H, -D, C1-4Alkyl, 3-10 membered heterocyclyl, 3-10 membered carbocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C1-4Aminoalkyl radical, C1-4Hydroxyalkyl radical, C1-4Alkylamino radical C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl radical, R5O(C=O)NR6-(C1-4Alkyl), R5(C=O)NR6-(C1-4Alkyl), NR)5R6(C=O)-(C1-4Alkyl) or R5(C=O)-(C1-6Alkyl) of said C1-4Alkyl, 3-10 membered heterocyclyl, 3-10 membered carbocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C1-4Aminoalkyl radical, C1-4Hydroxyalkyl radical, C1-4Alkylamino radical C1-4Alkyl and C1-4Alkoxy radical C1-4Alkyl, and R5O(C=O)NR6-(C1-4Alkyl), R5(C=O)NR6-(C1-4Alkyl), NR)5R6(C=O)-(C1-4Alkyl) and R5(C=O)-(C1-4Alkyl) each independently optionally substituted by 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -OH, oxo、C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkoxy radical C1-4Alkyl radical, R5(C ═ O) -and R5O (C ═ O) -, is substituted.

5. The compound of claim 1, wherein,

R2is-H, -D, methyl, ethyl, n-propyl, isopropyl, isobutyl, tert-butyl, cyclopropyl, cyclopentyl, cyclohexyl, pyrrolidinyl, azetidinyl, piperidinyl, morpholinyl, pyrrolidinylethyl, pyrrolidinylmethyl, piperidinylmethyl, piperazinylethyl, azetidinylmethyl, azetidinylethyl, morpholinylethyl, cyclohexylethyl, pyridylmethyl, pyridylethyl, pyrrolylethyl, oxazolylmethyl, imidazolylethyl, aminomethyl, aminoethyl, dimethylaminoethyl, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, hydroxy-isopropyl, hydroxy-isobutyl, methoxyethyl, methoxymethyl, NH, or2(C=O)CH2-、NH2(C=O)NH(CH2)2-or CH3(C=O)NH(CH2)2-, wherein said methyl, ethyl, n-propyl, isopropyl, isobutyl, tert-butyl, cyclopropyl, cyclopentyl, cyclohexyl, pyrrolidinyl, azetidinyl, piperidinyl, morpholinyl, pyrrolidinylethyl, pyrrolidinylmethyl, piperidinylmethyl, piperazinylethyl, azetidinylmethyl, azetidinylethyl, morpholinylethyl, cyclohexylethyl, pyridylmethyl, pyridylethyl, pyrrolylethyl, oxazolylmethyl, imidazolylethyl, aminomethyl, aminoethyl, dimethylaminoethyl, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, hydroxyisopropyl, hydroxyisobutyl, methoxyethyl, or methoxymethyl groups are each independently optionally substituted by 1,2,3, or 4 groups selected from-F, -Cl, -Br, -OH, oxo, methyl, ethyl, methoxymethyl, CH, and methoxy methyl groups3(C ═ O) -and CH3O (C ═ O) -, is substituted.

6. The compound of claim 1, wherein,

R3is-H, -D, NR5R6(C=O)-、C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C6-10Aryl radical C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (3-10 membered heterocyclyl) -C1-4Alkyl or C1-4Aminoalkyl radical, wherein said C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C6-10Aryl radical C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (3-10 membered heterocyclyl) -C1-4Alkyl and C1-4Aminoalkyl is each independently optionally substituted by 1,2,3 or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NR5R6、-OR7、C1-4Alkyl, halo C1-4Alkyl, hydroxy C1-4Alkyl, halo C1-4Alkoxy radical, C6-10Aryl radical, C1-4Alkoxy radical C1-4Alkyl, oxo, C1-4Alkyl acyl, 3-10 membered heterocyclic group and 3-10 membered carbocyclyl.

7. The compound of claim 1, wherein,

R3is-H, -D, N (CH)3)2(C=O)-、NH(CH3) (C ═ O) -, phenyl NH (C ═ O) -, 3-fluorophenyl NH (C ═ O) -, methyl, ethyl, isopropyl, n-propyl, cyclopropyl, cyclopentyl, cyclopropylmethyl, cyclopentylmethyl, azetidinyl, pyrrolidinyl, spiro [ 4.4.4 ═ O) -, and the like]Nonanylmethyl, bicyclo [3.3.0 ]]Octyl, azetidinylmethyl, phenyl, pyrimidinyl, pyridyl, thienyl, thiazolyl, furyl, pyrimidinylmethyl, pyridylmethyl, methoxymethyl, ethoxymethyl, isobutoxymethyl, methoxyethyl, aminomethyl or aminoethyl; wherein the methyl, ethyl, isopropyl, n-propyl, cyclopropyl, cyclopentyl and cyclopropylmethylAlkyl, cyclopentylmethyl, azetidinyl, pyrrolidinyl, spiro [4.4]]Nonanylmethyl, bicyclo [3.3.0 ]]Octyl, azetidinylmethyl, phenyl, pyrimidinyl, pyridyl, thienyl, thiazolyl, furyl, pyrimidinylmethyl, pyridylmethyl, methoxymethyl, ethoxymethyl, isobutoxymethyl, methoxyethylaminomethyl, and aminoethyl each independently optionally substituted with 1,2,3, or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NH23-fluoroanilino, methoxy, phenoxy, phenyl, methyl, trifluoromethyl, hydroxymethyl and oxo.

8. The compound of claim 1 having one of the following structures, or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt, or prodrug thereof,

9. a pharmaceutical composition comprising a compound according to any one of claims 1 to 8, and pharmaceutically acceptable adjuvants.

10. Use of a compound according to any one of claims 1 to 8 or a pharmaceutical composition according to claim 9 for the manufacture of a medicament for the prevention or treatment of RET-related diseases; optionally, the RET-associated disease comprises cancer, irritable bowel syndrome and/or pain associated with irritable bowel syndrome.

Technical Field

The present invention is in the field of medicine, and in particular, the present invention relates to novel compounds that inhibit reverse transcriptase (RET) kinase, pharmaceutical compositions comprising the compounds, the use of the compounds or pharmaceutical compositions thereof in the manufacture of medicaments, particularly for the treatment and prevention of RET related diseases and disorders, including cancer, irritable bowel syndrome and/or pain associated with irritable bowel syndrome.

Background

The Re-associated recombinant transfection (RET) kinase is one of the receptor type tyrosine kinases belonging to the cadherin superfamily, which activates multiple downstream pathways involved in cell proliferation and survival.

It has been reported that the results of the abnormalities in the RET gene (point mutation, chromosomal translocation, chromosomal inversion, gene amplification) are related to canceration. RET fusion proteins are associated with several cancers, including papillary thyroid carcinoma and non-small cell lung cancer. RET fusion proteins have been identified as a driver of certain cancers, driving the use of multi-kinase inhibitors with RET inhibitory activity to treat patients whose tumors express RET fusion proteins. It has been reported that multi-kinase inhibitors such as Sorafenib (Sorafenib), sunitinib, vandetanib, and ponatinib exhibit cell proliferation inhibitory effects on KIF 5B-RET-expressing cell lines (J Clin Oncol 30,2012, suppl; Abstract no: 7510). In addition, the multi-kinase inhibitor cabozantinib was reported to exhibit partial efficacy in two RET fusion gene positive non-small cell lung Cancer patients (Cancer Discov,3(6), Jun 2013, p.630-5). However, these drugs cannot always be administered at levels sufficient to inhibit RET, due to toxicity resulting from inhibition of targets other than RET. Furthermore, one of the biggest challenges in treating cancer is the ability of tumor cells to develop resistance to treatment. Reactivation of kinases through mutation is a common mechanism of resistance. When resistance occurs, the treatment options for patients are often very limited, and cancer progression is not inhibited in most cases. WO 2017011776 discloses single-target RET kinase inhibitors with good prophylactic or therapeutic effects on RET and its mutation-related cancers. There is still a need to further develop compounds that inhibit RET and its resistant mutants to address cancers associated with RET gene abnormalities.

Disclosure of Invention

The invention provides a novel compound showing inhibition of trans-transfection phase Rearrangement (RET) kinase, which has good inhibition effect on RET wild type and RET gene mutant, and compared with other kinases, the compound has better inhibition selectivity on RET wild type and RET gene mutant.

The excellent characteristics of some parameters of the compounds of the invention, such as half-life, clearance, selectivity, bioavailability, chemical stability, metabolic stability, membrane permeability, solubility, etc., can promote the reduction of side effects, the expansion of therapeutic index or the improvement of tolerance, etc.

In one aspect, the invention provides a compound of formula (I), or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt, or prodrug of a compound of formula (I),

wherein the content of the first and second substances,

X1、X2、X3、X4and X5Each independently isCR4Or N;

e is a bond, -NR6-or-O-;

ring A is carbocyclyl OR heterocyclyl and ring A is optionally substituted with 1,2,3 OR 4 substituents selected from-F, -Cl, -Br, -OR5、-NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-, oxo, aminoalkyl, alkyl, alkoxy, haloalkyl, hydroxyalkyl, carbocyclyl, heterocyclyl, heterocyclylalkyl, alkoxyalkyl, and alkylthio;

R1is-H, -D, -CN, -F, -Cl, -Br, -NH2Alkyl or cycloalkyl, wherein said alkyl and cycloalkyl are each independently optionally substituted with 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

R2is-H, -D, alkyl, heterocyclyl, carbocyclyl, heterocyclylalkyl, carbocyclylalkyl, heteroarylalkyl, aminoalkyl, hydroxyalkyl, alkylaminoalkyl, alkoxyalkyl, R5O(C=O)NR6Alkyl radical, R5(C=O)NR6Alkyl, NR5R6(C ═ O) alkyl or R5(C ═ O) alkyl, where said alkyl, heterocyclyl, carbocyclyl, heterocyclylalkyl, carbocyclylalkyl, heteroarylalkyl, aminoalkyl, hydroxyalkyl, alkylaminoalkyl, and alkoxyalkyl, and R5O(C=O)NR6Alkyl radical, R5(C=O)NR6Alkyl, NR5R6(C ═ O) alkyl and R5Each alkyl moiety in (C ═ O) alkyl is optionally substituted with 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -OH, oxo, alkyl, alkoxy, alkoxyalkyl, R5(C ═ O) -and R5O (C ═ O) -;

R3is-H, -D, NR5R6(C ═ O) -, alkyl, alkoxyalkyl, heteroaryl, aryl, carbocyclyl, heterocyclyl, heteroarylalkyl, arylalkyl, carbocyclylalkyl, heterocyclylalkyl, or aminoalkyl, wherein said alkyl, alkoxyalkyl, heteroaryl, or aminoalkyl areEach of the group, aryl, carbocyclyl, heterocyclyl, heteroarylalkyl, arylalkyl, carbocyclylalkyl, heterocyclylalkyl or aminoalkyl is independently optionally substituted with 1,2,3 or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NR5R6、-OR7Alkyl, haloalkyl, hydroxyalkyl, haloalkoxy, aryl, alkoxyalkyl, oxo, alkanoyl, heterocyclyl and carbocyclyl;

each R4Independently is-H, -D, -F, -Cl, -Br, alkyl or alkoxy, wherein said alkyl and alkoxy are each independently optionally substituted with 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

each R5Independently is-H, -NH2-D, alkyl, carbocyclyl, heterocyclyl, aryl or heteroaryl, wherein said-NH2Alkyl, carbocyclyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -OH, -NH2Alkylamino, alkyl, alkylsulfonyl, alkoxy, aryloxy, heteroaryloxy, aryl and heteroaryl;

each R6Independently is-H, -D, alkyl or alkoxyalkyl, wherein the alkyl and alkoxyalkyl groups are each independently optionally substituted with 1,2,3, or 4 groups selected from-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s); and

each R7independently-OH, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl.

In some embodiments, ring A is 3-12 membered carbocyclyl OR 3-12 membered heterocyclyl, wherein ring A is optionally substituted with 1,2,3, OR 4 substituents selected from-F, -Cl, -Br, -OR5、-NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-, oxo, C1-6Aminoalkyl radical, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C1-6Hydroxyalkyl, 3-12 membered carbocyclyl, 3-12 membered heterocyclyl, (3-12 membered heterocyclyl))-C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl and C1-6Alkylthio group substituent;

wherein each R is5And R6Having the definitions as described in the present invention.

In some embodiments, ring a is a subformula:

wherein Z is1、Z2、Z4And Z6Each independently is CH or N;

each Z3And Z5Independently is a bond, CH2O, S, NH, C O, S ═ O or S (═ O)2

m is 0, 1 or 2;

t, n and t1 are each independently 0 or 1;

and each sub-formula of ring A is optionally substituted by 1,2,3 OR 4 groups selected from-F, -Cl, -Br, -OR5Oxo, -NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-、C1-4Aminoalkyl radical, C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl and C1-4Alkylthio group substituent;

wherein each R is5And R6Having the definitions as described in the present invention.

In some embodiments, ring a is a subformula:

wherein each sub-structure of ring A is independently optionally substituted with 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -OH, oxo, -NH2、-NHCH3、-N(CH3)2、CH3(C ═ O) NH-, phenyl (C ═ O) NH-, NH2(C ═ O) NH-, methoxyphenyl (C ═ O) -, phenyl S (═ O)2NH-, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CF3Hydroxymethyl, 2-hydroxyethyl, cyclopropyl, cyclohexyl, pyrrolidinyl, piperidinyl, tetrahydrofuranyl, phenoxyethoxy, methoxyethoxy, chloroethoxy, hydroxyethoxy, and ethylthio.

In some embodiments, R2is-H, -D, C1-6Alkyl, 3-12 membered heterocyclyl, 3-12 membered carbocyclyl, (3-12 membered heterocyclyl) -C1-6Alkyl, (3-12 membered carbocyclyl) -C1-6Alkyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C1-6Aminoalkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkylamino radical C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl radical, R5O(C=O)NR6-(C1-6Alkyl), R5(C=O)NR6-(C1-6Alkyl), NR)5R6(C=O)-(C1-6Alkyl) or R5(C=O)-(C1-6Alkyl) of said C1-6Alkyl, 3-12 membered heterocyclyl, 3-12 membered carbocyclyl, (3-12 membered heterocyclyl) -C1-6Alkyl, (3-12 membered carbocyclyl) -C1-6Alkyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C1-6Aminoalkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkylamino radical C1-6Alkyl and C1-6Alkoxy radical C1-6Alkyl, and R5O(C=O)NR6-(C1-6Alkyl), R5(C=O)NR6-(C1-6Alkyl), NR)5R6(C=O)-(C1-6Alkyl) and R5(C=O)-(C1-6Alkyl) is optionally substituted by 1,2,3 or 4 alkyl moietiesSelected from-F, -Cl, -Br, -OH, oxo, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkoxy radical C1-6Alkyl radical, R5(C ═ O) -and R5O (C ═ O) -;

wherein each R is5And R6Having the definitions as described in the present invention.

In some embodiments, R2is-H, -D, C1-4Alkyl, 3-10 membered heterocyclyl, 3-10 membered carbocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C1-4Aminoalkyl radical, C1-4Hydroxyalkyl radical, C1-4Alkylamino radical C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl radical, R5O(C=O)NR6-(C1-4Alkyl), R5(C=O)NR6-(C1-4Alkyl), NR)5R6(C=O)-(C1-4Alkyl) or R5(C=O)-(C1-6Alkyl) of said C1-4Alkyl, 3-10 membered heterocyclyl, 3-10 membered carbocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C1-4Aminoalkyl radical, C1-4Hydroxyalkyl radical, C1-4Alkylamino radical C1-4Alkyl, and C1-4Alkoxy radical C1-4Alkyl, and R5O(C=O)NR6-(C1-4Alkyl), R5(C=O)NR6-(C1-4Alkyl), NR)5R6(C=O)-(C1-4Alkyl) and R5(C=O)-(C1-4Alkyl) each independently optionally substituted by 1,2,3 or 4 alkyl moieties selected from-F, -Cl, -Br, -OH, oxo, C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkoxy radical C1-4Alkyl radical, R5(C ═ O) -and R5O (C ═ O) -;

wherein each R is5And R6Having the definitions as described in the present invention.

In some embodiments, R2is-H, -D,Methyl, ethyl, n-propyl, isopropyl, isobutyl, tert-butyl, cyclopropyl, cyclopentyl, cyclohexyl, pyrrolidinyl, azetidinyl, piperidinyl, morpholinyl, pyrrolidinylethyl, pyrrolidinylmethyl, piperidinyl methyl, piperazinylethyl, azetidinylmethyl, azetidinylethyl, morpholinylethyl, cyclohexylethyl, pyridylmethyl, pyridylethyl, pyrrolylethyl, oxazolylmethyl, imidazolylethyl, aminomethyl, aminoethyl, dimethylaminoethyl, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, hydroxyisopropyl, hydroxyisobutyl, methoxyethyl, methoxymethyl, NH, ethyl, piperidinyl, azetidinyl, piperidinyl, morpholinyl, pyrrolidinyl, oxazolylethyl, imidazolyl-ethyl, aminomethyl, aminoethyl, dimethylaminoethyl, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, hydroxyisopropyl, hydroxyisobutyl, methoxyethyl, methoxymethyl, NH, and the like2(C=O)CH2-、NH2(C=O)NH(CH2)2-or CH3(C=O)NH(CH2)2-, wherein said methyl, ethyl, n-propyl, isopropyl, isobutyl, tert-butyl, cyclopropyl, cyclopentyl, cyclohexyl, pyrrolidinyl, azetidinyl, piperidinyl, morpholinyl, pyrrolidinylethyl, pyrrolidinylmethyl, piperidinylmethyl, piperazinylethyl, azetidinylmethyl, azetidinylethyl, morpholinylethyl, cyclohexylethyl, pyridylmethyl, pyridylethyl, pyrrolylethyl, oxazolylmethyl, imidazolylethyl, aminomethyl, aminoethyl, dimethylaminoethyl, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, hydroxyisopropyl, hydroxyisobutyl, methoxyethyl, or methoxymethyl groups are each independently optionally substituted by 1,2,3, or 4 groups selected from-F, -Cl, -Br, -OH, oxo, methyl, ethyl, methoxymethyl, CH, and methoxy methyl groups3(C ═ O) -and CH3O (C ═ O) -, is substituted.

In some embodiments, R3is-H, -D, NR5R6(C=O)-、C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C6-10Aryl radical C1-6Alkyl, (3-10 membered carbocyclyl) -C1-6Alkyl, (3-10 membered heterocyclyl) -C1-6Alkyl or C1-6Aminoalkyl radical, wherein said C1-6Alkyl radical, C1-6Alkoxy radical C1-6Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-6Alkyl radical, C6-10Aryl radical C1-6Alkyl, (3-10 membered carbocyclyl) -C1-6Alkyl, (3-10 membered heterocyclyl) -C1-6Alkyl and C1-6Aminoalkyl is each independently optionally substituted by 1,2,3 or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NR5R6、-OR7、C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, halo C1-6Alkoxy radical, C6-10Aryl radical, C1-6Alkoxy radical C1-6Alkyl, oxo, C1-6Alkyl acyl, 3-10 membered heterocyclic group and 3-10 membered carbocyclyl;

wherein each R is5、R6And R7Having the definitions as described in the present invention.

In some embodiments, R3is-H, -D, NR5R6(C=O)-、C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C6-10Aryl radical C1-4Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (3-10 membered heterocyclyl) -C1-4Alkyl or C1-4Aminoalkyl radical, wherein said C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl, 5-10 membered heteroaryl, C6-10Aryl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (5-10 membered heteroaryl) -C1-4Alkyl radical, C6-10Aryl radical C14Alkyl, (3-10 membered carbocyclyl) -C1-4Alkyl, (3-10 membered heterocyclyl) -C1-4Alkyl and C1-4Aminoalkyl is each independently optionally substituted by 1,2,3 or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NR5R6、-OR7、C1-4Alkyl, halo C1-4Alkyl, hydroxy C1-4Alkyl, halo C1-4Alkoxy radical, C6-10Aryl radical, C1-4Alkoxy radical C1-4Alkyl, oxo, C1-4Alkyl acyl, 3-10 membered heterocyclic group and 3-10 membered carbocyclyl;

wherein each R is5、R6And R7Having the definitions as described in the present invention.

In some embodiments, R3is-H, -D, N (CH)3)2(C=O)-、NH(CH3) (C ═ O) -, phenyl NH (C ═ O) -, 3-fluorophenyl NH (C ═ O) -, methyl, ethyl, isopropyl, n-propyl, cyclopropyl, cyclopentyl, cyclopropylmethyl, cyclopentylmethyl, azetidinyl, pyrrolidinyl, spiro [ 4.4.4 ═ O) -, and the like]Nonanylmethyl, bicyclo [3.3.0 ]]Octyl, azetidinylmethyl, phenyl, pyrimidinyl, pyridyl, thienyl, thiazolyl, furyl, pyrimidinylmethyl, pyridylmethyl, methoxymethyl, ethoxymethyl, isobutoxymethyl, methoxyethyl, aminomethyl or aminoethyl; wherein said methyl, ethyl, isopropyl, n-propyl, cyclopropyl, cyclopentyl, cyclopropylmethyl, cyclopentylmethyl, azetidinyl, pyrrolidinyl, spiro [4.4] methyl]Nonanylmethyl, bicyclo [3.3.0 ]]Octyl, azetidinylmethyl, phenyl, pyrimidinyl, pyridyl, thienyl, thiazolyl, furyl, pyrimidinylmethyl, pyridylmethyl, methoxymethyl, ethoxymethyl, isobutoxymethyl, methoxyethyl, aminomethyl and aminoethyl each independently optionally substituted with 1,2,3 or 4 substituents selected from-D, -F, -Cl, -CN, -NO2、-OH、-NH23-fluoroanilino, methoxy, phenoxy, phenyl, methyl, trifluoromethyl, hydroxymethyl and oxo.

In some embodiments, R1is-H, -D, -CN, -F, -Cl, -Br, -NH2Methyl or cyclopropyl, wherein said methyl and cyclopropyl are independently optionally substituted by 1,2,3 or 4 substituents selected from the group consisting of-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

each R4independently-H, -D, -F, -Cl, -Br, methyl, ethyl, n-propyl, methoxy or ethoxy, wherein said methyl, ethyl, n-propyl, methoxy and ethoxy may beIndependently optionally substituted by 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

each R5Independently is-H, -D, -NH2Methyl, ethyl, n-propyl, isopropyl, n-butyl, cyclopropyl, cyclopentyl, pyrrolidinyl, phenyl, or pyrazolyl; wherein said-NH2Methyl, ethyl, n-propyl, cyclopropyl, cyclopentyl, pyrrolidinyl, phenyl and pyrazolyl each independently optionally substituted by 1,2,3 or 4 substituents selected from-F, -Cl, -Br, -OH, -NH2Methyl, -S (═ O)2CH3Methoxy, ethoxy and phenyl;

each R6independently-H, -D, methyl, ethyl, n-propyl, n-butyl, methoxymethyl, ethoxymethyl or methoxyethyl, wherein said methyl, ethyl, n-propyl, n-butyl, methoxymethyl, ethoxymethyl and methoxyethyl are each independently optionally substituted by 1,2,3 or 4 substituents selected from the group consisting of-F, -Cl, -Br, -CN, -NH2-OH and-NO2Substituted with the substituent(s);

each R7independently-OH, methyl, ethyl, cyclopropyl or phenyl.

In some embodiments, the compounds of the present invention have the structure of formula (I-1), or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug of the structure of formula (I-1),

wherein q is 0, 1,2,3 or 4;

each RaIndependently is-F, -Cl, -Br, -OR5Oxo, -NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-、C1-4Aminoalkyl radical, C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Haloalkyl, C1-4Hydroxy radicalAlkyl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl and C1-4An alkylthio group;

each R1、R2、R3、X1、X2、X3、X4、X5、E、Z1、Z2、Z3、Z5And m has the definition as described in the present invention.

In some embodiments, the compounds of the present invention have the structure of formula (I-2), or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug of the structure of formula (I-2),

wherein the content of the first and second substances,

ring a1 is of the subformula: and the sub-formulae of each ring A1 are optionally substituted by 1,2,3 OR 4 substituents selected from the group consisting of-F, -Cl, -Br, -OR5Oxo, -NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-、C1-4Aminoalkyl radical, C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl and C1-4Alkylthio group substituent;

each R1、R2、R3、X1、X2、X3、X4、X5、E、Z1And Z2Having the definitions as described in the present invention.

In some embodiments, the compounds of the present invention have the structure of formula (I-3) or (I-4), or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug of the structure of formula (I-3) or (I-4),

wherein the content of the first and second substances,each optionally substituted by 1,2,3 OR 4 groups selected from-F, -Cl, -Br, -OR5Oxo, -NR5R6、R5(C=O)NR6-、R5(C=O)-、R5S(=O)2NR6-、C1-4Aminoalkyl radical, C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 3-10 membered carbocyclyl, 3-10 membered heterocyclyl, (3-10 membered heterocyclyl) -C1-4Alkyl radical, C1-4Alkoxy radical C1-4Alkyl and C1-4Alkylthio groups.

Each R1、R2、R3、X1、X2、X3、X4、X5、E、Z1、Z2、Z4、Z6M, n, t and t1 have the definitions as described herein.

In some embodiments, the compounds of the present invention have one of the following structures, or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof,

in another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention, and a pharmaceutically acceptable adjuvant.

In another aspect, the present invention also provides a use of the compound of the present invention or the pharmaceutical composition of the present invention for the preparation of a medicament for preventing or treating a RET-related disease.

In some embodiments, the RET-associated disease includes cancer, irritable bowel syndrome, and/or pain associated with irritable bowel syndrome.

In another aspect, the present invention also provides a compound according to the present invention or a pharmaceutical composition according to the present invention for use in preventing or treating RET-related diseases.

In some embodiments, the RET-associated disease includes cancer, irritable bowel syndrome, and/or pain associated with irritable bowel syndrome.

In another aspect, the present invention also provides a method for preventing or treating a RET-associated disease, the method comprising administering to a patient a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.

In some embodiments, the RET-associated disease includes cancer, irritable bowel syndrome, and/or pain associated with irritable bowel syndrome.

In another aspect, the invention relates to intermediates for the preparation of compounds of formula (I), (I-1), (I-2), (I-3) or (I-4).

In another aspect, the present invention relates to a method for the preparation, isolation and purification of a compound represented by formula (I), (I-1), (I-2), (I-3) or (I-4).

In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention and pharmaceutically acceptable adjuvants thereof. In some embodiments, adjuvants of the present invention include, but are not limited to, carriers, excipients, diluents, vehicles, or combinations thereof. In some embodiments, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.

The invention also provides a method of inhibiting cell proliferation in vitro or in vivo, comprising contacting a cell with an effective amount of a compound of the invention or a pharmaceutical composition thereof.

The present invention also provides a method of treating Irritable Bowel Syndrome (IBS) and/or pain associated with IBS in a patient in need of such treatment, comprising administering to said patient a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.

The invention also provides a compound of the invention or a pharmaceutical composition of the invention for use in the prevention or treatment of Irritable Bowel Syndrome (IBS) and/or pain associated with IBS.

Unless otherwise indicated, all stereoisomers, tautomers, nitroxides, hydrates, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds of the present invention are within the scope of the present invention.

In particular, the salts are pharmaceutically acceptable salts. The term "pharmaceutically acceptable" includes substances or compositions which must be compatible with chemical or toxicological considerations, in connection with the other components which make up the formulation and the mammal being treated.

The salt of the compound of the present invention also includes an intermediate for preparing or purifying the compound represented by formula (I), (I-1), (I-2), (I-3) or (I-4) or an isolated enantiomeric salt of the compound represented by formula (I), (I-1), (I-2), (I-3) or (I-4), but is not necessarily a pharmaceutically acceptable salt.

In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.

Nitroxides of the compounds of the present invention are also included within the scope of the present invention. The nitroxides of the compounds of the present invention can be prepared by oxidation of the corresponding nitrogen-containing basic substances using common oxidants (e.g. hydrogen peroxide) in the presence of acids such as acetic acid at elevated temperature, or by reaction with peracids in suitable solvents, for example with peracetic acid in dichloromethane, ethyl acetate or methyl acetate, or with 3-chloroperoxybenzoic acid in chloroform or dichloromethane.

If the compounds of the invention are basic, the desired salts may be prepared by any suitable method provided in the literature, for example, using inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric and phosphoric acids and the like. Or using organic acids such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid and salicylic acid; pyranonic acids, such as glucuronic acid and galacturonic acid; alpha-hydroxy acids such as citric acid and tartaric acid; amino acids such as aspartic acid and glutamic acid; aromatic acids such as benzoic acid and cinnamic acid; sulfonic acids such as p-toluenesulfonic acid, ethanesulfonic acid, and the like.

If the compounds of the invention are acidic, the desired salts can be prepared by suitable methods, e.g., using inorganic or organic bases, such as ammonia (primary, secondary, tertiary), alkali or alkaline earth metal hydroxides, and the like. Suitable salts include, but are not limited to, organic salts derived from amino acids such as glycine and arginine, ammonia such as primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine, piperazine and the like, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.

Detailed description of the invention

Definitions and general terms

Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.

It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.

The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.

The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.

"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans isomers), atropisomers, and the like. Unless otherwise indicated, all stereoisomers or mixtures of stereoisomers of the formulae depicted herein are within the scope of the present invention. In addition, unless otherwise indicated, the structural formulae of the compounds described herein include isotopically enriched concentrations of one or more different atoms.

The stereochemical definitions and rules used in the present invention generally follow the general definitions of S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E.and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.

Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.

The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (low energy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers (valenctautomers) include interconversion by recombination of some of the bonding electrons. A specific example of keto-enol tautomerism is the tautomerism of the pentan-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the tautomerism of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.

The compounds of the invention may be independently optionally substituted with one or more substituents, as described herein, in the general formula above, or as specified in the examples, subclasses, and classes of compounds encompassed by the invention. It will be appreciated that the term "independently optionally substituted with … …" is used interchangeably with the term "substituted or unsubstituted". In general, the term "substituted" means that one or more hydrogen atoms in a given structure is replaced with a particular substituent. Unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently.

In addition, unless otherwise explicitly indicated, the descriptions of the terms "… independently" and "… independently" and "… independently" used in the present invention are interchangeable and should be understood in a broad sense to mean that the specific items expressed between the same symbols do not affect each other in different groups or that the specific items expressed between the same symbols in the same groups do not affect each other.

In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group.

In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.

The term "alkyl" denotes a saturated, straight or branched chain, monovalent hydrocarbon radical containing 1 to 20 carbon atoms, wherein the alkyl radical may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 3 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH)3) Ethyl group (Et, -CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl group (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) Tert-butyl (t-Bu, -C (CH)3)3) N-pentyl (-CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH(CH3)2) 3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) N-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2) 2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2) 3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)3) N-heptyl, n-octyl, and the like.

When alkyl is the linking group, and "alkyl" is enumerated for this markush group definition, then "alkyl" represents the linked alkylene group. When M is alkyl as defined herein, it means that M is an attached alkylene group. The term "alkylene" refers to a saturated divalent hydrocarbon radical resulting from the removal of two hydrogen atoms from a saturated straight or branched chain hydrocarbon radical. Examples of alkyl groups represented as attached alkylene groups include, but are not limited to: -CH2-、-CH2CH2-、-CH(CH3)CH2-, and the like.

The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "tans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 10 carbon atoms; in one embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) Propenyl (-CHCH)3) Isopropenyl (-C (CH)3)=CH2) And so on.

When alkenyl is attachedWhere groups are recited, and "alkenyl" is recited with respect to the markush group definition, then "alkenyl" represents an attached alkenylene group. When M is alkenyl as defined herein, it means that M is an attached alkenylene group. Examples of alkenyl groups represented as attached alkenylene groups include, but are not limited to: -CH ═ CH2-、-CH2CH=CH-、-CH2CH=CHCH2-, and the like.

The terms "carbocyclyl" and "carbocycle" are used interchangeably to refer to a saturated or partially unsaturated monocyclic, bicyclic, or polycyclic system in which the ring atoms are all carbon atoms, including mono-, bridged-, and fused-and spiro-carbocyclyl. When carbocyclyl and carbocycle are linking groups, and "carbocyclyl" and "carbocycle" are listed for this markush group definition, then "carbocyclyl" and "carbocycle" represent the linked carbocyclylene group. The terms "monocyclic" and "carbocycle" are used interchangeably to denote a saturated or partially unsaturated monocyclic ring system in which the ring atoms are all carbon atoms, -CH2A group-may be optionally substituted by-C (═ O) - (or- (C ═ O) -). In some embodiments, a monocyclic group is a 3-12 membered monocyclic group. In some embodiments, a monocyclic group is a 3-7 membered monocyclic group. In some embodiments, a monocyclic group is a 3-6 membered monocyclic group. Examples of monocyclic groups include, but are not limited to: cyclopropyl, cyclobutyl, cyclohexyl, cyclopentyl, cyclopentadienyl, cycloheptyl, and the like. When a monocyclic group and a monocyclic ring are linking groups, and a monocyclic group and a monocyclic ring are enumerated for this markush group definition, then "monocyclic group" and "monocyclic ring" represent linked submonocarbocyclic group groups.

The terms "fused carbocyclic ring" and "fused carbocyclic ring" are used interchangeably and both represent non-aromatic, saturated or partially unsaturated bicyclic or polycyclic ring systems that share two adjacent ring carbon atoms, with the ring atoms being carbon atoms. In some embodiments, the fused carbocyclyl is a 4-12 membered fused carbocyclyl. In some embodiments, the fused carbocyclic group is a 5-12 membered fused carbocyclic group. Examples of fused carbocyclic groups include, but are not limited to: bicyclo [2.1.0] pentyl, bicyclo [3.1.0] heptyl, bicyclo [4.1.0] heptyl, bicyclo [3.2.0] heptyl, bicyclo [4.2.0] octyl, octahydro-1H-indenyl, octahydro-cyclopentadienyl, decahydronaphthyl, decahydro-1H-benzo [7] annulene, and the like.

The terms "bridged carbocycle" and "bridged carbocyclyl" are used interchangeably and both represent a non-aromatic, saturated or partially unsaturated bicyclic or polycyclic ring system that shares two or more nonadjacent ring carbon atoms, with the ring atoms being carbon atoms. C-CH in bridged carbocycle2-the group may optionally be replaced by-C (═ O) -. In some embodiments, bridged carbocycles contain 6 to 12 ring carbon atoms, i.e., represent 6 to 12 membered bridged carbocycles; in other embodiments, bridged carbocycles contain 6 to 10 ring carbon atoms, i.e., represent 6 to 10 membered bridged carbocycles. Examples of bridged carbocycles include, but are not limited to: bicyclo [3.1.1]Heptane, bicyclo [3.2.1]Octane, bicyclo [2.2.2]Octane, bicyclo [2.2.0]Hexane, octahydro-1H-indene, and the like. When a bridged carbocycle or carbocyclyl group is the linking group, and bridged carbocycle or bridged carbocyclyl is enumerated for this markush group definition, then bridged carbocycle or bridged carbocyclyl represents the linked bridged carbocyclylene group. The term "bridged carbocyclyl" refers to a divalent bridged carbocyclic group formed by the removal of two hydrogen atoms from a ring atom bridging a carbocyclic ring. The bridged carbocycle or carbocyclyl may independently be optionally substituted with one or more substituents described herein.

The terms "spirocarbocycle" and "spirocarbocyclyl" are used interchangeably to refer to a non-aromatic, saturated or partially unsaturated bicyclic or polycyclic system formed by two carbocycles sharing a single carbon atom. Spiro carbocyclic ring-CH2-the group may optionally be replaced by-C (═ O) -. In some embodiments, a spiro carbocyclic ring contains 7 to 12 ring carbon atoms, i.e., represents a 7 to 12 membered spiro carbocyclic ring; in other embodiments, spiro carbocyclic rings contain 7 to 10 ring carbon atoms, i.e., represent 7 to 10 membered spiro carbocyclic rings. Examples of spiro carbocycles include, but are not limited to: spiro [4.4]]Nonane, spiro [3.4 ]]Octane, spiro [4.5 ]]Decane, etc. When a spiro carbocyclic or spiro carbocyclic group is the linking group and spiro carbocyclic or spiro carbocyclic groups are listed for the markush group definition, the spiro carbocyclic or spiro carbocyclic group represents the linked spirocyclic carbocyclylene group. The term "spirorylene carbocyclyl" refers to a divalent spirocarbocyclic group formed by removing two hydrogen atoms from a ring atom of a spirocarbocyclic ring. The above-mentionedThe spiro carbocycle or spiro carbocyclyl may independently be optionally substituted with one or more substituents described herein.

The terms "heterocycle" or "heterocyclyl" are used interchangeably and all refer to a monovalent non-aromatic saturated or partially unsaturated monocyclic, bicyclic, or polycyclic system of 3 to 12 ring atoms containing at least 1 carbon atom and containing 1,2, or 3 heteroatoms selected from O, N, S including mono-, bridged, and heterocyclic and spiroheterocyclic groups. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide and the nitrogen atom of the ring may optionally be oxidized to the N-oxygen compound. The heterocycle may be monocyclic or bicyclic; in particular, the bicyclic ring system can be a hetero-bicyclic, a spiro-hetero-bicyclic, or a bridged hetero-bicyclic ring. In some embodiments, heterocyclyl contains 4-7 ring atoms, i.e., represents a 4-7 membered heterocyclyl; examples of heterocyclyl groups include, but are not limited to: oxirane, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, homopiperazinyl, homopiperidinyl, 1-dioxo-1, 3-thiomorpholinyl, and the like. In heterocyclic radicals of-CH2Examples of-groups substituted by-C (═ O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl. Examples of heterocyclic groups in which the nitrogen atom is oxidized to the N-oxide compound include, but are not limited to, 1-dioxo-1, 3-thiomorpholine. When a heterocycle or heterocyclyl is the linking group and a heterocycle or heterocyclyl is listed for this markush group definition, then a heterocycle or heterocyclyl represents the linked heterocyclylene group. The term "heterocyclylene" refers to a divalent heterocyclic group formed by removing two hydrogen atoms from a ring atom of a heterocycle. Said heterocycle orThe heterocyclyl group may be independently optionally substituted with one or more substituents described herein.

The terms "fused heterocycle" and "fused heterocyclyl" are used interchangeably and both refer to a non-aromatic, saturated or partially unsaturated bicyclic or polycyclic ring system which shares two adjacent ring atoms and which has at least one ring atom which is a heteroatom selected from O, N, S. In some embodiments, the heterocyclo is a 4-12 membered heterocyclo. In some embodiments, the heterocyclo is a 5-12 membered heterocyclo. Examples of heterocyclyls include, but are not limited to: 3-azabicyclo [3.1.0] hexanyl, 2-oxa-5-azabicyclo [2.2.0] hexanyl, 2, 5-diazabicyclo [2.2.0] hexanyl, 2-azabicyclo [2.1.0] pentanyl, 2-azabicyclo [3.1.0] hexanyl, 3-oxabicyclo [3.1.0] hexanyl, octahydrocyclopenta [ c ] pyrrole, octahydropyrrolo [3,4-c ] pyrrole, hexahydrofuro [3,2-b ] furan, hexahydrofuro [2,3-b ] furan, octahydropyrrolo [3,4-b ] pyrrole, hexahydro-1H-thieno [3,4-c ] pyrrole, hexahydro-1H-furo [3,4-c ] pyrrole, hexahydro-2H- [1,4] dioxino [2,3-c ] pyrrole, octahydro- [1,4] dioxino [2,3-c ] pyridine, etc.

The terms "bridged heterocyclic ring" or "bridged heterocyclic group" are used interchangeably and both refer to a non-aromatic, saturated or partially unsaturated bicyclic or polycyclic ring system that shares two or more non-adjacent ring atoms and that contains at least 1 carbon atom and contains 1,2, or 3 heteroatoms selected from O, N, S. Bridged heterocycle-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide and the nitrogen atom of the ring may optionally be oxidized to the N-oxygen compound. In some embodiments, bridged heterocycles contain 6 to 12 ring atoms, i.e., represent 6-12 membered bridged heterocycles; in other embodiments, bridged heterocycles contain 6 to 10 ring atoms, i.e., represent 6 to 10 membered bridged heterocycles. Examples of bridged heterocycles include, but are not limited to: 3, 6-diazabicyclo [3.1.1]Heptane, 3, 8-diazabicyclo [3.2.1]Octane, 2-azabicyclo [2.2.1]Heptane, 6-azabicyclo [3.1.1]Heptane, 3-azabicyclo [3.1.1]Heptane, 8-azabicyclo [3.2.1]Octane, 3-azabicyclo [3.2.1]Octane, 2-dinitrogenHetero-bicyclo [2.2.2]Octane, etc. When a bridged heterocyclic or bridged heterocyclic group is a linking group, and bridged heterocyclic or bridged heterocyclic group is enumerated for this markush group definition, then bridged heterocyclic or bridged heterocyclic group represents an attached bridged heterocyclylene group. The term "bridged heterocyclic group" means a divalent bridged heterocyclic group formed by removing two hydrogen atoms from a ring atom of the bridged heterocyclic ring. The bridged heterocyclic or bridged heterocyclic group may be independently optionally substituted with one or more substituents described herein.

The terms "spiroheterocycle" or "spiroheterocyclyl" are used interchangeably and both refer to a non-aromatic, saturated or partially unsaturated ring system of two rings sharing a single carbon atom and containing 1,2 or 3 heteroatoms selected from O, N, S. Spiro-heterocyclic ring-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide and the nitrogen atom of the ring may optionally be oxidized to the N-oxygen compound. In some embodiments, the spiroheterocycle contains 7-12 ring atoms, i.e., represents a 7-12 membered spiroheterocycle; in other embodiments, the spiroheterocycle contains 7-10 ring atoms, i.e., represents a 7-10 membered spiroheterocycle. Examples of spiroheterocycles include, but are not limited to: 4, 7-diazaspiro [2.5 ]]Octane, 2, 8-diazaspiro [4.5 ]]Decane, 2, 7-diazaspiro [4.5 ]]Decane, 2, 7-diazaspiro [3.5 ]]Decane, 2, 6-diazaspiro [3.3]Heptane, 2, 7-diazaspiro [4.4]]Nonane, 3-azaspiro [5.5 ]]Undecane, 2, 7-diazaspiro [4.4]]Nonan-1-one, and the like. When a spiroheterocycle or spiroheterocyclyl is a linking group and a spiroheterocycle or spiroheterocyclyl is listed for this markush group definition, then a spiroheterocycle or spiroheterocyclyl represents the linked spiroheterocyclylene. The term "spiroheterocyclylene" denotes a divalent spiroheterocyclylene group formed by removing two hydrogen atoms from the ring atoms of a spiroheterocycle. The spiroheterocycle or spiroheterocyclyl may be independently optionally substituted with one or more substituents described herein.

The term "carbocyclylalkyl" denotes an alkyl group substituted with one or more carbocyclyl groups, wherein carbocyclyl and alkyl have the definitions as described herein. In some embodiments, carbocyclylalkyl represents alkyl substituted with one carbocyclyl. Examples of carbocyclylalkyl groups include, but are not limited to: cyclopropylmethyl, cyclopentylmethyl, cyclobutylmethyl, cyclohexylmethyl, and the like.

The term "hydroxyalkyl" denotes an alkyl group substituted with one or more hydroxyl groups. In some embodiments, hydroxyalkyl represents alkyl substituted with 1,2,3, or 4 hydroxy groups. In some embodiments, hydroxyalkyl represents an alkyl group substituted with one or two hydroxy groups. In some embodiments, hydroxyalkyl represents hydroxy C1-6Alkyl radicals, i.e. C1-6Alkyl is substituted with one or more hydroxy groups; preferably, a hydroxyl group C1-6Alkyl radicals being represented by, i.e. C1-6The alkyl group is substituted with one hydroxyl group. In some embodiments, hydroxyalkyl represents hydroxy C1-4An alkyl group. In some embodiments, hydroxyalkyl represents hydroxy C1-3An alkyl group. Examples of hydroxyalkyl include, but are not limited to, CH2(OH)-、CH2(OH)CH2CH2CH2-、CH2(OH)CH2-、CH2(OH)CH2CH(OH)CH2-、CH(CH3)(OH)CH2CH(OH)CH2-, etc.

The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein. Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-PrO, n-propoxy, -OCH)2CH2CH3) 2-propoxy (i-PrO, i-propoxy, -OCH (CH)3)2) 1-butoxy (n-BuO, n-butoxy, -OCH)2CH2CH2CH3) 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH)2CH(CH3)2) 2-butoxy (s-BuO, s-butoxy, -OCH (CH)3)CH2CH3) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH)3)3) 1-pentyloxy (n-pentyloxy, -OCH)2CH2CH2CH2CH3) 2-pentyloxy (-OCH (CH)3)CH2CH2CH3) 3-pentyloxy (-OCH (CH))2CH3)2) 2-methyl-2-butoxy (-OC (CH))3)2CH2CH3) 3-methyl-2-butoxy (-OCH (CH)3)CH(CH3)2) 3-methyl-l-butoxy (-OCH)2CH2CH(CH3)2) 2-methyl-l-butoxy (-OCH)2CH(CH3)CH2CH3) And so on.

The term "alkoxyalkyl" denotes an alkyl group substituted with one alkoxy group, wherein alkoxy and alkyl have the definitions as described herein. In some embodiments, alkoxyalkyl represents C1-6Alkoxy radical C1-6An alkyl group; in other embodiments, alkoxyalkyl represents C1-4Alkoxy radical C1-4An alkyl group; in other embodiments, alkoxyalkyl represents C1-4Alkoxy radical C1-3An alkyl group; in some embodiments, alkoxyalkyl represents C1-3Alkoxy radical C1-3An alkyl group. Examples of alkoxy groups include, but are not limited to, methoxymethyl, ethoxymethyl, n-propoxymethyl, isopropoxymethyl, methoxyethyl, methoxy-n-propyl, methoxyisopropyl, ethoxyethyl, ethoxy-n-propyl, ethoxyisopropyl, n-propoxyethyl, isopropoxyethyl, n-propoxy-n-propyl, n-propoxy-isopropyl, isopropoxy-n-propyl, isopropoxy-isopropyl, and the like.

The term "halogen" denotes F (fluorine), Cl (chlorine), Br (bromine) or I (iodine).

The term "oxo" denotes ═ O.

The term "haloalkyl" denotes an alkyl group substituted with one or more halogen atoms, examples of which include, but are not limited to, monofluoromethyl, difluoromethyl, trifluoromethyl, 1-fluoroethyl, 2-fluoroethyl, 1, 2-difluoroethyl, 1-difluoroethyl, 2-difluoroethyl, monochloromethyl, dichloromethyl, trichloromethyl, 2-chloroethyl, 1, 2-dichloroethyl, 1-dichloroethyl, 2-dichloroethyl, 1-dibromoethyl, and the like.

The term "haloalkoxy" denotes an alkoxy group substituted with one or more halogen atoms, examples of which include, but are not limited to, monofluoromethoxy, difluoromethoxy, trifluoromethoxy, 1-fluoroethoxy, 2-fluoroethoxy, 1, 2-difluoroethoxy, 1-difluoroethoxy, 2-difluoroethoxy, monochloromethoxy, dichloromethoxy, trichloromethoxy, 2-chloroethoxy, 1, 2-dichloroethoxy, 1-dichloroethoxy, 2-dichloroethoxy, 1-dibromoethoxy, and the like.

The term "alkanoyl" means an alkyl group attached to the remainder of the molecule through a carbonyl group, wherein alkyl has the meaning described herein, and carbonyl represents-C (═ O) -. In some embodiments, alkanoyl represents C1-6An alkyl acyl group; in some other embodiments, alkanoyl represents C1-4An alkyl acyl group. Examples of alkanoyl groups include, but are not limited to: formyl, acetyl, and the like.

The terms "cycloalkyl" or "cycloalkane" are used interchangeably and both represent monovalent, saturated, monocyclic carbocyclic ring systems of 3 to 7 carbon atoms. In the carbocyclic ring-CH2A group-may be optionally substituted by-C (═ O) - (or- (C ═ O) -). In one embodiment, the cycloalkyl group contains 3 to 6 carbon atoms, i.e., C3-6A cycloalkyl group; in another embodiment, the cycloalkyl group contains 3 to 5 carbon atoms, i.e., C3-5A cycloalkyl group. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. In the carbocyclic ring-CH2Examples of — groups that may be replaced by-C (═ O) -include, but are not limited to: cyclopentanone, cyclobutanone, and the like. When cycloalkyl is the linking group, and "cycloalkyl" is enumerated for this markush group definition, then "cycloalkyl" represents the linked cycloalkylene group. The term "cycloalkylene" denotes a divalent cycloalkane group formed from a cycloalkyl group by the removal of two hydrogen atoms from a ring carbon atom. Said cycloalkyl radicalOr the cycloalkane may independently be optionally substituted with one or more substituents described herein.

The term "aryloxyalkyloxy" or "aryloxyalkoxy" denotes an alkoxy group substituted with one or more aryloxy groups, wherein the aryloxy and alkoxy groups have the definitions as described herein. In some embodiments, aryloxyalkyloxy represents an alkoxy group substituted with one aryloxy group. Examples of aryloxyalkyloxy groups include, but are not limited to: phenoxyethyloxy, phenoxymethyloxy, and the like.

The term "alkylheterocyclyl" denotes a heterocyclyl group substituted with one or more alkyl groups, wherein alkyl and heterocyclyl have the definitions as described herein. In some embodiments, alkyl heterocyclyl represents a heterocyclyl group substituted with one alkyl group. Examples of alkyl heterocyclic groups include, but are not limited to: isopropyl azetidinyl, methyl pyrrolidinyl, and the like.

The term "aromatic ring" or "arene" means monocyclic, bicyclic and tricyclic carbon ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring system is aromatic, wherein each ring system comprises a ring of 3 to 7 atoms. Examples of the aromatic ring may include benzene, naphthalene, and anthracene.

The term "aryl" refers to a monovalent aromatic ring radical formed by the removal of one hydrogen atom from a ring carbon atom of an aromatic ring. Examples of the aryl group may include phenyl, naphthyl and anthracenyl. When aryl is the linking group, and "aryl" is enumerated for this markush group definition, then "aryl" represents the linked arylene group. When M is aryl as defined herein, it means that M is an attached arylene group. The term "arylene" refers to a divalent aromatic ring radical formed by the removal of two hydrogen atoms from a ring carbon atom of an aromatic ring. Examples of arylene groups represented as attached arylene groups may include phenylene, naphthylene, and anthracenylene. The aryl group may independently be optionally substituted with one or more substituents described herein.

The term "aryloxy" denotes aryl-O-, i.e. the aryl group in aryloxy is attached to the rest of the molecule via an oxygen atom, wherein aryl has the meaning as described in the present invention. Examples of aryloxy groups include, but are not limited to, phenoxy, naphthoxy.

The term "heteroaromatic ring" denotes monocyclic, bicyclic and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring system is aromatic and at least one ring system contains one or more heteroatoms, wherein each ring system contains a ring of 5 to 7 atoms.

The term "heteroaryl" refers to a monovalent aromatic ring radical formed by the removal of one hydrogen atom from a ring atom of a heteroaromatic ring. The heteroaryl group is optionally substituted with one or more substituents described herein. In one embodiment, a 5-10 atom composed heteroaryl or 5-10 membered heteroaryl group comprises 1,2,3, or 4 heteroatoms independently selected from O, S, and N. In some embodiments, the term "heteroaryl" denotes a heteroaryl ring group or 5-6 membered heteroaryl group containing 5-6 ring atoms, which contains 1,2,3 or 4 heteroatoms independently selected from O, S and N. In some embodiments, the term "heteroaryl" denotes a heteroaryl ring group or 5-membered heteroaryl group containing 5 ring atoms, wherein 1,2,3 or 4 heteroatoms independently selected from O, S and N are contained. Examples of heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), and the like, 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, pyrazinyl, 1,3, 5-triazinyl; the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridyl, and the like. When heteroaryl is a linking group, and heteroaryl is enumerated for this markush group definition, heteroaryl represents a linked heteroarylene group. When M is heteroaryl as defined herein, it means M is a linked heteroarylene group. The term "heteroarylene" refers to a divalent heteroaromatic ring group formed by removing two hydrogen atoms from a ring atom of a heteroaryl group. The heteroaryl group can be independently optionally substituted with one or more substituents described herein.

The term "aryloxyalkyl" denotes an aryloxy-substituted alkyl group wherein the aryloxy and alkyl groups have the definitions as described herein. In some embodiments, aryloxyalkyl represents C6-10Aryloxy radical C1-6An alkyl group; in other embodiments, aryloxyalkyl represents phenoxy C1-6An alkyl group; in other embodiments, aryloxyalkyl represents phenoxy C1-4An alkyl group. Specific examples of aryloxyalkyl groups include, but are not limited to, phenoxymethyl, phenoxyethyl, phenoxy-n-propyl, phenoxyisopropyl, phenoxy-n-butyl, phenoxyisobutyl, phenoxy-tert-butyl, and the like.

The term "arylalkyl" denotes aryl substituted alkyl groups, wherein the aryl and alkyl groups have the definitions as described herein. In some embodiments, arylalkyl represents C6-10Aryl radical C1-6An alkyl group; in other embodiments, arylalkyl represents phenyl C1-6An alkyl group; in other embodiments, arylalkyl represents phenyl C1-4An alkyl group. Specific examples of arylalkyl groups include, but are not limited to, phenylmethyl, phenylethyl, phenyl-n-propyl, phenylisopropyl, phenyl-n-butyl, phenylisobutyl, phenyl-tert-butyl, and the like.

The term "heteroarylalkyl" denotes an alkyl substituted by a heteroaryl groupSubstituted alkyl groups, wherein the heteroaryl and alkyl groups have the definitions as described herein. In some embodiments, heteroarylalkyl represents (5-10 membered heteroaryl) -C1-6An alkyl group; in other embodiments, heteroarylalkyl represents (5-10 membered heteroaryl) -C1-4An alkyl group; in other embodiments, heteroarylalkyl represents (5-6 membered heteroaryl) -C1-4An alkyl group. Examples of heteroarylalkyl include, but are not limited to, imidazolylmethyl, imidazolylethyl, pyrazolylmethyl, pyrazolylethyl, oxazolylmethyl, oxazolylethyl, imidazolylpropyl, pyridylpropyl, pyridylmethyl, pyridylethyl, pyrimidylmethyl, furylmethyl, furylethyl, indolylmethyl, pyrazolo [1,5-a ]]Pyrimidylmethyl and the like.

The term "heterocyclylalkyl" denotes an alkyl group substituted with one or more heterocyclyl groups, wherein heterocyclyl and alkyl have the definitions set forth herein. In some embodiments, heterocyclylalkyl represents an alkyl group substituted with one heterocyclyl. Examples of heterocyclylalkyl groups include, but are not limited to: spiro [4.4] nonanylmethyl, azetidinylmethyl, pyrrolidinylmethyl, and the like.

The term "aminoalkyl" denotes an alkyl group substituted with one or more amino groups. In some embodiments, the term "aminoalkyl" denotes an alkyl group substituted with one amino group. In some embodiments, the term "aminoalkyl" denotes amino C1-6An alkyl group. In other embodiments, the term "aminoalkyl" denotes amino C1-4An alkyl group. In other embodiments, the term "aminoalkyl" denotes amino C1-3An alkyl group. Examples of aminoalkyl groups include, but are not limited to, aminomethyl, aminoethyl, amino-n-propyl, amino-isopropyl, amino-isobutyl, amino-tert-butyl, 1, 2-diaminoethyl, and the like. The term "alkylamino" or "alkylamino" denotes an amino group substituted by one or two alkyl groups. In some embodiments, the term "alkylamino" denotes an amino group substituted with one alkyl group. In some embodiments, the term "alkylamino" denotes C1-6An alkylamino group. In other embodiments, the term "alkylamino" denotes C1-4An alkylamino group. In other embodiments, the term "alkylamino" denotes C1-3An alkylamino group. Examples of alkylamino include, but are not limited to, methylamino, ethylamino, n-propylamino, isopropylamino, isobutylamino, tert-butylamino, dimethylamino, diethylamino, di-n-propylamino, diisopropylamino, diisobutylamino, di-tert-butylamino, and the like.

The term "alkylaminoalkyl" denotes an alkyl group substituted with an alkylamino group, wherein alkylamino and alkyl have the meaning as defined herein. In some embodiments, the term "alkylaminoalkyl" denotes C1-6Alkylamino radical C1-6An alkyl group. In other embodiments, the term "alkylaminoalkyl" denotes C1-4Alkylamino radical C1-4An alkyl group. In other embodiments, the term "alkylamino" denotes C1-3Alkylamino radical C1-4An alkyl group. Examples of alkylaminoalkyl radicals include, but are not limited to, dimethylaminomethyl (N (CH)3)2CH2-), dimethylaminoethyl (N (CH)3)2CH2CH2-) methylaminomethyl (CH)3NHCH2-, etc.

The term "alkylsulfonyl" denotes alkyl-S (═ O)2-, i.e. alkyl by-S (═ O)2-to the rest of the molecule. In some embodiments, alkylsulfonyl represents C1-6An alkylsulfonyl group; in other embodiments, alkylsulfonyl represents C1-4An alkylsulfonyl group; in other embodiments, alkylsulfonyl represents C1-4An alkylsulfonyl group. Examples of alkylsulfonyl include, but are not limited to, methylsulfonyl, ethylmethylsulfonyl, n-propylmethylsulfonyl, isopropylmethylsulfonyl, n-butylmethylsulfonyl, and the like.

In the general formula of the compound, the left end of a ring A is connected with E, and the right end of the ring A is connected withFor example, when A isWhen it is, then it represents

As described herein, unless otherwise specified, a ring substituent may be attached to the rest of the molecule through any available position on the ring. For example, piperidinyl includes piperidin-1-yl, piperidin-2-yl, piperidin-3-yl, and piperidin-4-yl.

The term "protecting group" or "PG" refers to a substituent that, when reacted with other functional groups, is generally used to block or protect a particular functionality. For example, "amino protecting group" means a substituent attached to an amino group to block or protect the functionality of the amino group in a compound, and suitable amino protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC ), benzyloxycarbonyl (CBZ ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). Similarly, "hydroxyl protecting group" refers to the functionality of a substituent of a hydroxyl group to block or protect the hydroxyl group, and suitable protecting groups include acetyl and silyl groups. "carboxy protecting group" refers to the functionality of a substituent of a carboxy group to block or protect the carboxy group, and typical carboxy protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General descriptions of protecting groups can be found in the literature: greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.

The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-24) Esters, acyloxy groupsMethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel Delivery Systems, Vol.14of the A.C.S.Symphosis Series, Edward B.Roche, ed., Bioreversible Carriers in Drug designs, American Pharmaceutical Association and Pergamon Press,1987, J.Rautio et al, Prodrugs in Design and Clinical Applications, Nature Review Delivery, 2008,7,255 and 270, S.J.Herer et al, Prodrugs of pharmaceuticals and pharmaceuticals, Journal of chemical Chemistry,2008,51,2328 and 5.

"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.

As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, description of the scientific acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, inorganic acid salts formed by reaction with amino groups such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, and organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonateOr by other methods described in the literature, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.

The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of Pharmaceutical Salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable Salts.

In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms.

"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.

"nitroxide" in the context of the present invention means that when a compound contains several amine functional groups, 1 or more than 1 nitrogen atom can be oxidized to form an N-oxide. Specific examples of N-oxides are N-oxides of tertiary amines or N-oxides of nitrogen-containing heterocyclic nitrogen atoms. The corresponding amines can be treated with an oxidizing agent such as hydrogen peroxide or a peracid (e.g., peroxycarboxylic acid) to form the N-oxide (see Advanced Organic Chemistry, Wiley Interscience, 4 th edition, Jerry March, pages). In particular, the N-oxide may be prepared by the method of L.W.Deady (Syn.Comm.1977,7,509-514) in which an amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.

The term "treating" or "treatment" as used herein refers, in some embodiments, to ameliorating a disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.

The term "RET-associated cancer" as used herein refers to a cancer associated with dysregulated expression or activity or levels of the RET gene, RET kinase (also referred to herein as RET kinase protein or RET kinase), or any one thereof. The present invention describes non-limiting examples of RET-associated cancers. The deregulation of the expression or activity or level of either of the RET gene, RET kinase or any of these is one or more point mutations in the RET gene.

Dysregulation of the expression or activity or level of RET kinase, RET gene, or any (e.g., one or more) thereof, may contribute to tumorigenesis. For example, a disorder of RET kinase, RET gene, or a disorder of expression or activity or level of any of these may be translocation, overexpression, activation, amplification, or mutation of the RET kinase, RET gene, or RET kinase domain. Translocation may include translocation involving the RET kinase domain, mutation may include mutation involving the RET ligand binding site, and amplification may be of the RET gene. Other disorders may include RET mRNA splice variants and RET autocrine/paracrine signaling, which may also contribute to tumorigenesis.

In some embodiments, the deregulation of the expression or activity or level of a RET gene, RET kinase, or any of these includes one or more deletions (e.g., deletion of amino acid 4), insertions, or point mutations in RET kinase. In some embodiments, deregulation of the expression or activity or level of a RET gene, RET kinase, or any of these, includes deletion of one or more residues of RET kinase, resulting in constitutive activity of the RET kinase domain.

The term "irritable bowel syndrome" includes diarrhea predominant, constipation predominant or alternating pattern of bowel movement, functional bloating, functional constipation, functional diarrhea, non-specific functional bowel disease, functional abdominal pain syndrome, chronic idiopathic constipation, functional esophageal disease, functional gastroduodenal disease, functional anorectal pain, inflammatory bowel disease, and the like.

Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H,3H,11C,13C,14C,15N,17O,18O,18F,31P,32P,35S,36Cl and125I。

in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present3H,14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13C. the isotopically enriched compounds can be used for metabolic studies (use)14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.

In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stabilityIn (1). For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is to be considered as a substituent of the compound of formula (I), (I-1), (I-2), (I-3) or (I-4). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D2O, acetone-d6、DMSO-d6Those solvates of (a).

Compounds of the invention and pharmaceutical compositions, formulations and administrations thereof

The present invention provides compounds of the invention or pharmaceutical compositions thereof that inhibit wild-type RET and RET mutants. In addition, the compounds of the invention or pharmaceutical compositions thereof are selective for inhibition of both wild-type RET and RET gene mutants relative to other kinases, resulting in reduced toxicity associated with inhibition of other kinases.

The pharmaceutical composition of the present invention comprises a compound represented by formula (I), (I-1), (I-2), (I-3) or (I-4), a compound listed in the present invention, or a compound of the examples. The amount of the compound in the compositions of the invention is effective to treat or alleviate RET-associated diseases or disorders in a patient, including RET-associated cancer, irritable bowel syndrome and/or pain associated with irritable bowel syndrome.

As described herein, the pharmaceutically acceptable compositions of the present invention further comprise pharmaceutically acceptable adjuvants, as used herein, including any solvents, diluents, or other liquid excipients, dispersing or suspending agents, surfactants, isotonic agents, thickening agents, emulsifiers, preservatives, solid binders or lubricants, and the like, as appropriate for the particular target dosage form. As described in the following documents: in Remington, The Science and Practice of Pharmacy,21st edition,2005, ed.D.B.Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds.J.Swarbrick and J.C.Boylan, 1988. Annu 1999, Marcel Dekker, New York, taken together with The disclosure of this document, indicates that different adjuvants can be used In The preparation of pharmaceutically acceptable compositions and their well-known methods of preparation. Except insofar as any conventional adjuvant is incompatible with the compounds of the present invention, e.g., any adverse biological effect produced or interaction in a deleterious manner with any other component of a pharmaceutically acceptable composition, their use is contemplated by the present invention.

In preparing the compositions provided herein, the active ingredient is typically mixed with an excipient, diluted by an excipient or encapsulated in such a carrier, for example, in the form of a capsule, sachet, paper, or other container. If the excipient serves as a diluent, it may be a solid, semi-solid, or liquid material that serves as a vehicle, carrier, or medium for the active ingredient. Suitable carriers include, but are not limited to, magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low melting wax, cocoa butter, and the like. Thus, the compositions may be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (in solid form or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders. In one embodiment, the composition is formulated for oral administration. In one embodiment, the composition is formulated as a tablet or capsule.

When useful in therapy, a therapeutically effective amount of a compound of the present invention, particularly a compound of formula (I), (I-1), (I-2), (I-3) or (I-4) and pharmaceutically acceptable salts thereof, may be administered as the raw chemical and may also be provided as the active ingredient of a pharmaceutical composition. Accordingly, the present disclosure also provides pharmaceutical compositions comprising a therapeutically effective amount of a compound of the present invention, particularly a compound of formula (I), (I-1), (I-2), (I-3), or (I-4), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable adjuvants, including but not limited to carriers, diluents, or excipients, and the like. The term "therapeutically effective amount" as used herein refers to the total amount of each active component sufficient to show meaningful patient benefit (e.g., cancer cell reduction). When the active ingredient alone is used for separate administration, the term refers only to that ingredient. When used in combination, the term refers to the combined amounts of the active ingredients that, when combined, administered sequentially or simultaneously, result in a therapeutic effect. The compounds of the present invention, particularly the compounds represented by formula (I), (I-1), (I-2), (I-3) or (I-4), and pharmaceutically acceptable salts thereof are as described above. The carrier, diluent or excipient must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. According to a further aspect of the present disclosure there is also provided a process for the preparation of a pharmaceutical formulation which comprises mixing a compound of the present invention, especially a compound of formula (I), (I-1), (I-2), (I-3) or (I-4) or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients. The term "pharmaceutically acceptable" as used herein refers to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and which are effective for their intended use.

The amount of active ingredient that is combined with one or more adjuvants to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration. The amount of the active ingredient to be mixed with a carrier material to prepare a single dosage form of a compound represented by the formula (I), (I-1), (I-2), (I-3) or (I-4) will vary depending on the disease to be treated, the severity of the disease, the administration time, the administration route, the excretion rate of the compound used, the treatment time, and the age, sex, body weight and condition of the patient. Preferred unit dosage forms are those containing a daily or divided dose or suitable fraction thereof of the active ingredient described herein above. Treatment can be initiated with small doses, which are clearly below the optimal dose of the compound. Thereafter, the dosage is increased in smaller increments until the optimum effect is achieved in this case. In general, the compounds are most desirably administered at concentration levels that generally provide effective results in antitumor terms without causing any harmful or toxic side effects.

Compositions containing the compounds of the present invention may be formulated in unit dosage forms, each dosage containing from about 5 to about 1,000mg (1g), more typically from about 100mg to about 500mg, of the active ingredient. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects or other patients, each unit containing a predetermined quantity of active material (i.e. a compound of formula I as herein provided) calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.

The pharmaceutical compositions are suitable for administration by any suitable route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intradermal, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional, intravenous or subdermal injection or infusion) route. Such formulations may be prepared by any method known in the art of pharmacy, for example by mixing the active ingredient with a carrier or excipient. Oral administration or injection administration is preferred.

The invention also provides methods of treating an individual having a RET-associated cancer comprising administering a compound of the invention before, during, or after administering another anti-cancer agent (e.g., other than a compound of the invention).

The present invention provides a method for treating cancer in a patient in need thereof, the method comprising: (a) determining whether the cancer in the patient is a RET-associated cancer (e.g., including a RET-associated cancer having one or more RET inhibitor resistance mutations) (e.g., using a regulatory agency-approved, e.g., FDA-approved, kit to identify a deregulation of expression or activity or levels of a RET gene, RET kinase, or any of these in a patient or in a biopsy sample of a patient, or by performing any of the non-limiting examples of assays described herein); and (b) administering a therapeutically effective amount of a compound represented by formula (I), (I-1), (I-2), (I-3) or (I-4) or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof to the patient if the cancer is determined to be RET-associated cancer. Some embodiments of these methods further comprise administering to the subject another anti-cancer agent (e.g., another RET inhibitor, e.g., a RET inhibitor that is not a compound of the present invention). In some embodiments, the subject has been previously treated with a RET inhibitor that is not a compound of formula (I), (I-1), (I-2), (I-3), or (I-4), or a pharmaceutically acceptable salt or solvate thereof, or has been previously treated (e.g., after tumor resection or radiotherapy) with another anti-cancer agent.

In some embodiments of any of the methods described herein, a compound of formula (I), (I-1), (I-2), (I-3), or (I-4) (or a pharmaceutically acceptable salt or solvate thereof) is combined with a therapeutically effective amount of at least one additional therapeutic agent selected from one or more additional therapeutic or therapeutic (e.g., chemotherapeutic) agents.

Non-limiting examples of other therapeutic agents include: other RET-targeted therapeutics (i.e., other RET kinase inhibitors: RET inhibitors that are not compounds described herein), receptor tyrosine kinase-targeted therapeutics, signal transduction pathway inhibitors, checkpoint inhibitors, apoptosis pathway modulators (e.g., obataclx); cytotoxic chemotherapeutic agents, angiogenesis targeted therapeutic agents, immune targeted agents and radiotherapy.

In some embodiments, the other RET-targeted therapeutic agent is a multi-kinase inhibitor that exhibits RET inhibitory activity.

Non-limiting examples of RET targeted therapeutics include alatinib, apatinib, cabozantinib (XL-184), multidimensional, lenvatinib, motaxanib, nintedanib, ponatinib, ragrafenib, statinib (sitravatinib) (MGCD516), sunitinib, sorafenib, vatanib, vandetanib, AUY-922(5- (2, 4-dihydroxy-5-isopropyl-phenyl) -N-ethyl-4- [4- (morpholinomethyl) phenyl ] isoxazole-3-carboxamide), BLU6864, BLU-667, DCC-2157, NVP-AST487(1- [4- [ (4-ethylpiperazin-1-yl) methyl ] -3- (trifluoromethyl) phenyl ] -3- [4- [6- (methylamino) pyrimidin-4-yl ] oxyphenyl ] urea), PZ-1, RPI-1(1, 3-dihydro-5, 6-dimethoxy-3- [ (4-hydroxyphenyl) methylene ] -1H-indol-2-one), RXDX-105(1- (3- (6, 7-dimethoxyquinazolin-4-yl) oxy) phenyl) -3- (5- (1,1, 1-trifluoro-2-methylpropan-2-yl) isoxazol-3-yl) urea), SPP86 (1-isopropyl-3- (phenylethynyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine) and TG101209(N- (1, 1-dimethylethyl) -3- [ [ 5-methyl-2- [ [4- (4-methyl-1-methyl-4-amine) -piperazinyl) phenyl ] amino ] -4-pyrimidinyl ] amino ] benzenesulfonamide).

Other therapeutic agents include RET inhibitors such as those described, for example, in the following: U.S. patent nos. 7,504,509; 8,299,057, respectively; 8,399,442, respectively; 8,067,434, respectively; 8,937,071, respectively; 9,006,256, respectively; and 9,035,063; U.S. publication No. 2014/0121239; 20160176865, respectively; 2011/0053934, respectively; 2011/0301157, respectively; 2010/0324065, respectively; 2009/0227556, respectively; 2009/0130229, respectively; 2009/0099167, respectively; 2005/0209195, respectively; international publication nos. WO 2014/184069; WO 2014/072220; WO 2012/053606; WO 2009/017838; WO 2008/031551; WO 2007/136103; WO 2007/087245; WO 2007/057399; WO 2005/051366; WO 2005/062795; and WO 2005/044835; and j.med.chem.2012,55(10), 4872-.

Also provided herein are methods of treating cancer comprising administering to a patient in need thereof a pharmaceutical combination for treating cancer comprising (a) a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, (b) an additional therapeutic agent, and (c) optionally at least one pharmaceutically acceptable carrier, for simultaneous, separate or sequential use in treating cancer, wherein the amount of the compound of formula I or a pharmaceutically acceptable salt or solvate thereof and the amount of the additional therapeutic agent are jointly effective in treating cancer.

The compounds and compositions described herein can be administered alone or in combination with other compounds (including other RET modulating compounds) or other therapeutic agents. In some embodiments, a compound or composition of the invention may be administered in combination with one or more compounds selected from the group consisting of: cabozantinib (COMETRIQ), vandetanib (CALPRESA), sorafenib (NEXAVAR), Sunitinib (SUTENT), ragrafenib (STAVARGA), ponatinib (icluti), bevacizumab (avastin), crizotinib (XALKORI) or gefitinib (IRESSA). The compounds or compositions of the invention may be administered simultaneously or sequentially with other therapeutic agents by the same or different routes of administration. The compounds of the present invention may be included in a single formulation with other therapeutic agents or in separate formulations.

In some embodiments, the compounds of the invention may be used to treat Irritable Bowel Syndrome (IBS) in combination with one or more other therapeutic agents or therapies that are effective in the treatment of irritable bowel syndrome by acting through the same or different mechanisms of action. The at least one additional therapeutic agent may be administered as part of the same or separate dosage forms, via the same or different routes of administration, and according to the same or different schedules of administration, with the compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, according to standard pharmaceutical practice known to those skilled in the art. Non-limiting examples of other therapeutic agents for treating Irritable Bowel Syndrome (IBS) include probiotics, fibrous supplements (e.g., psyllium, methylcellulose), antidiarrheals (e.g., loperamide), bile acid binders (e.g., cholestyramine, colestipol, colesevelam), anticholinergics and antispasmodics (e.g., hyoscyamine, dicyclomine), antidepressants (e.g., tricyclic antidepressants such as imipramine or nortriptyline or selective 5-hydroxytryptamine reuptake inhibitors (SSRIs) such as fluoxetine or paroxetine), antibiotics (e.g., rifaximin), alosetron and rubiprostone.

Use of the Compounds and pharmaceutical compositions of the invention

The present invention also provides use of a compound of the present invention or a pharmaceutical composition of the present invention in the manufacture of a medicament for preventing or treating a RET-associated disease or disorder, wherein the RET-associated disease or disorder comprises RET-associated cancer, irritable bowel syndrome and/or pain associated with irritable bowel syndrome.

The present invention provides compounds of the invention or pharmaceutical compositions thereof that inhibit wild-type RET and RET mutants, e.g., RET mutants that are resistant to current standard of care therapy ("RET resistant mutants"). In addition, the compounds of the invention or pharmaceutical compositions thereof may be selective for wild-type RET over other kinases, resulting in reduced toxicity associated with inhibition of other kinases.

The invention provides application of the compound for inhibiting wild type RET and RET mutants or a pharmaceutical composition thereof in preparing a medicament for preventing or treating diseases or symptoms related to the wild type RET and RET mutants.

In some embodiments of any of the methods or uses described herein, the cancer (e.g., a RET-associated cancer) is a hematologic cancer. In some embodiments of any of the methods or uses described herein, the cancer (e.g., a RET-associated cancer) is a solid tumor. In some embodiments of any of the methods or uses described herein, the cancer (e.g., a RET-associated cancer) is lung cancer (e.g., small cell lung cancer or non-small cell lung cancer), papillary thyroid cancer, medullary thyroid cancer, differentiated thyroid cancer, recurrent thyroid cancer, refractory differentiated thyroid cancer, lung adenocarcinoma, bronchiolar lung cancer, multiple endocrine tumors of type 2A or 2B (MEN 2A or MEN2B, respectively), pheochromocytoma, parathyroid hyperplasia, breast cancer, colorectal cancer (e.g., metastatic colorectal cancer), papillary renal cell carcinoma, ganglioneuroma disease of the gastrointestinal mucosa, inflammatory myofibroblastic tumor, or cervical cancer. In some embodiments of any of the methods or uses described herein, the cancer (e.g., a RET-associated cancer) is selected from the group consisting of: acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), juvenile cancer, adrenocortical cancer, anal cancer, appendiceal cancer, astrocytoma, atypical teratoma/rhabdoid tumor, basal cell carcinoma, cholangiocarcinoma, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumor, Burkitt's lymphoma, carcinoid tumor, unknown primary cancer, heart tumor, cervical cancer, childhood cancer, chordoma, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), chronic myeloproliferative tumor, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, cholangiocarcinoma, ductal carcinoma in situ, embryonal tumor, endometrial cancer, ependymoma, esophageal cancer, adult neuroblastoma, Ewing's sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, extrahepatic bile duct cancer, ocular cancer, fallopian tube cancer, fibroblastic bone cancer, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), germ cell tumor, gestational trophoblastic disease, glioma, hairy cell tumor, hairy cell leukemia, head and neck cancer, heart cancer, hepatocellular carcinoma, histiocytosis, Hodgkin's lymphoma, hypopharynx cancer, intraocular melanoma, islet cell tumor, pancreatic neuroendocrine tumor, Kaposi's sarcoma, kidney cancer, Langerhans ' cell histiocytosis, laryngeal cancer, leukemia, lip and oral cancer, liver cancer, lung cancer, lymphoma, macroglobulinemia, malignant fibrous histiocytoma of bone, bone cancer, melanoma, Merkel cell cancer, mesothelioma, metastatic neck cancer, midline cancer, squamous cell cancer, oral cancer, multiple endocrine syndrome, multiple myeloma, mycosis fungoides granulomatosis, myelodysplastic syndrome, myelodysplastic/myeloproliferative tumors, myelogenous leukemia, multiple myeloma, myeloproliferative tumors, cancers of the nasal cavity and sinuses, nasopharyngeal carcinoma, neuroblastoma, non-hodgkin's lymphoma, non-small cell lung cancer, oral cancer, lip cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, papillomatosis, paragangliomas, paranasal sinuses and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pituitary cancer, plasmacytoma, pleuropneumonias, pregnancy and breast cancer, primary central nervous system lymphoma, primary peritoneal cancer, prostate cancer, rectal cancer, renal cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland carcinoma, sarcoma, sezary syndrome, skin cancer, small cell lung cancer, small bowel cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer, gastric cancer, T-cell lymphoma, testicular cancer, cancer of the throat, cancer of the thymus and thymus, cancer of the thyroid gland, transitional cell carcinoma of the renal pelvis and ureter, cancer of unknown primary, cancer of the urethra, cancer of the uterus, sarcoma of the uterus, cancer of the vagina, cancer of the vulva and wilm's tumor.

In some embodiments, the RET-associated cancer of the invention is selected from lung cancer, papillary thyroid carcinoma, medullary thyroid carcinoma, differentiated thyroid carcinoma, recurrent thyroid carcinoma, refractory differentiated thyroid carcinoma, multiple endocrine tumors of type 2A or 2B (MEN 2A or MEN2B, respectively), pheochromocytoma, parathyroid hyperplasia, breast cancer, colorectal cancer, papillary renal cell carcinoma, gastrointestinal mucosal ganglionic cell tumors, and cervical cancer. In some embodiments, the RET-associated cancer is RET fusion lung cancer or medullary thyroid cancer.

In some embodiments, compounds of formula (I), (I-1), (I-2), (I-3), or (I-4), and pharmaceutically acceptable salts and solvates thereof, are useful for treating patients with RET inhibitor resistance mutations that result in increased resistance, e.g., a substitution at amino acid position 804, e.g., V804M, V804L, or V804E, to a cancer that is not a compound of formula (I), (I-1), (I-2), (I-3), or (I-4), or a pharmaceutically acceptable salt or solvate thereof, the treatment is by co-administration or subsequent treatment with an existing drug therapy (e.g., other RET kinase inhibitors that are not compounds of formula (I), (I-1), (I-2), (I-3), or (I-4) or a pharmaceutically acceptable salt or solvate thereof). Described herein are exemplary RET kinase inhibitors (e.g., other RET kinase inhibitors that are not compounds of formula (I), (I-1), (I-2), (I-3), or (I-4), or a pharmaceutically acceptable salt or solvate thereof). In some embodiments, the RET kinase inhibitor may be selected from cabozantinib, vandetanib, alatinib, sorafenib, lenvatinib, ponatinib, multidimensional, sunitinib, fortinib (foretinib), BLU667, and BLU 6864.

In some embodiments of any of the methods or uses described herein, the Irritable Bowel Syndrome (IBS) comprises diarrhea predominant, constipation predominant or alternating, functional abdominal bloating, functional constipation, functional diarrhea, unspecified functional bowel disorder, functional abdominal pain syndrome, chronic idiopathic constipation, functional esophageal disease, functional gastroduodenal disease, functional anorectal pain, and inflammatory bowel disease.

The compounds and compositions according to the methods of the present invention can be administered in any amount and by any route effective to treat or reduce the severity of the disease. The exact amount necessary will vary depending on the patient, depending on the race, age, general condition of the patient, severity of infection, particular factors, mode of administration, and the like. The compound or composition may be administered in combination with one or more other therapeutic agents, as discussed herein.

General Synthesis of Compounds of the invention

In general, the compounds of the present invention may be prepared by the methods described herein, wherein the substituents are as defined for formula (I), (I-1), (I-2), (I-3) or (I-4), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.

Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.

The examples described below, unless otherwise indicated, are all temperatures set forth in degrees Celsius. Unless otherwise indicated, reagents are commercially available, for example, from commercial suppliers such as Lingkai medicine, Aldrich Chemical Company, Inc., Arco Chemical Company and Alfa Chemical Company, and are used without further purification. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin HaoLiyu Chemicals Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaseiki chemical plant.

The anhydrous tetrahydrofuran is obtained by refluxing and drying the metallic sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, N, N-dimethylacetamide and petroleum ether were used dried over anhydrous sodium sulfate in advance.

The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. The glassware was dried.

The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants. Nuclear magnetic resonance spectroscopy with CDC13Or DMSO-d6As solvent (reported in ppm) TMS (0ppm) or chloroform (7.25ppm) was used as reference standard. When multiple peaks occur, the following abbreviations will be used: s (singleton), d (doublet), t (triplet ), m (multiplet, multiplet), br (broad ), dd (doublet of doublets), dt (doublet of triplets ). Coupling constants are expressed in hertz (Hz).

Low resolution Mass Spectral (MS) data were measured by an Agilent 6320 series LC-MS spectrometer equipped with a G1312A binary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315B DAD detector were applied for analysis, and an ESI source was applied to the LC-MS spectrometer.

Low resolution Mass Spectral (MS) data were determined by Agilent 6120 series LC-MS spectrometer equipped with a G1311A quaternary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315D DAD detector were used for analysis, and an ESI source was used for the LC-MS spectrometer.

Both spectrometers were equipped with an Agilent Zorbax SB-C18 column, 2.1X 30mm, 5 μm. The injection volume is determined by the sample concentration; the flow rate is 0.6 mL/min; peaks of HPLC were recorded by UV-Vis wavelength at 210nm and 254 nm. The mobile phases were 0.1% formic acid in acetonitrile (phase a) and 0.1% formic acid in ultrapure water (phase B).

Compound purification was assessed by Agilent 1100 series High Performance Liquid Chromatography (HPLC) with UV detection at 210nm and 254nm, a Zorbax SB-C18 column, 2.1X 30mm, 4 μm, 10min, flow rate 0.6mL/min, 5-95% (0.1% formic acid in acetonitrile) in (0.1% formic acid in water), the column temperature was maintained at 40 ℃.

The following acronyms are used throughout the invention:

DCE 1, 2-dichloroethane

THF tetrahydrofuran

DCM dichloromethane

TEA Triethylamine

DMSO dimethyl sulfoxide

DIPEA N, N-diisopropylethylamine

DMF N, N-dimethylformamide

Pd(PPh3)4Tetrakis (triphenylphosphine) palladium

Tf2O-Trifluoromethanesulfonic anhydride

NH4Cl ammonium chloride

AlCl3Aluminium trichloride

NaSO4·10H2Sodium O decahydrate sulfate

EtMgBr Ethyl magnesium bromide

FeBr2Ferrous bromide

NMP N-methylpyrrolidone

K2CO3Potassium carbonate

MeOH methanol

HCl hydrogen chloride

Pd(PPh3)2Cl2Bis (triphenylphosphine) palladium dichloride

CuI cuprous iodide

n-BuLi n-butyllithium

NaH sodium hydride

H2O water

PE Petroleum Ether

EA Ethyl acetate

DEG C

mL of

g

mmol millimole

TLC thin layer chromatography

min for

h hours

mol/L mol/liter

The following synthetic schemes describe the steps for preparing the compounds disclosed herein. Unless otherwise stated, each R1、R2、R3、R6、X1、X2、X3、X4、X5And A has the definition as described in the present invention.

Intermediate I-a synthesis scheme:

the synthesis of intermediate I-A can be obtained by reference to the synthetic steps of the intermediate synthesis scheme above. Wherein, hal1Is F, Cl, Br or I, preferably Cl or Br; hal2Is F, Cl, Br or I, preferably F, Cl or Br. A compound of formula I-A-1 is reacted with a compound of formula I-A-2 under suitable coupling agent conditions (e.g., a palladium coupling agent, preferably Pd (PPh)3)4) The coupling reaction is carried out in a suitable solvent (e.g., dioxane, etc.) to afford the compound of formula I-A.

Synthesis scheme 1:

when E in the general formula (I) is a bond, the compound of the formula (I) is the compound of the formula (Ia) in the synthesis scheme 1. The synthesis of the compounds of formula (Ia) of the present invention can be obtained by reference to the synthetic procedure of FIG. 1. Wherein, hal2Is F, Cl, Br or I, preferably F, Cl or Br. Coupling of a compound of formula I-A with a compound of formula I-B1 or a salt of a compound of formula I-B1 (e.g., hydrochloride, trifluoroacetate, hydrobromide, etc.) in the presence of a suitable reagent (e.g., DIPEA, etc.) affords a compound of formula (Ia).

Synthesis scheme 2:

when E in the general formula (I) of the present invention is-NR6When the compound of formula (I) of the present invention is a compound of formula (Ib) in FIG. 2. The synthesis of the compounds of formula (Ib) according to the invention can be obtained by reference to the synthetic procedure of scheme 2. Wherein, hal2Is F, Cl, Br or I, preferably F, Cl or Br. Coupling of a compound of formula I-A with a compound of formula I-B2 or a salt of a compound of formula I-B2 (e.g., hydrochloride, trifluoroacetate, hydrobromide, etc.) in the presence of a suitable reagent (e.g., DIPEA, etc.) affords a compound of formula (Ib).

Synthesis scheme 3:

when E in the general formula (I) is a bond and the left end of A is connected with E through an N atom, the compound of the formula (I) is the compound of the formula (Ic) in the synthesis scheme 3. The synthesis of the compounds of formula (Ic) according to the invention can be obtained by reference to the synthetic procedure of scheme 3. Wherein, hal2Is F, Cl, Br or I, preferably F, Cl or Br. Coupling of a compound of formula I-A with a compound of formula I-B3 or a salt of a compound of formula I-B3 (e.g., hydrochloride, trifluoroacetate, hydrobromide, etc.) in the presence of a suitable reagent (e.g., DIPEA, etc.) affords a compound of formula (Ic).

Synthesis scheme 4:

when E in the general formula (I) is a bond, the left end of A is connected with E through N atom, and R3When H and D are not present, the compound of formula (I) according to the invention is a compound of formula (Ic') according to FIG. 3, wherein hal3Is F, Cl, Br or I, preferably Cl, Br or I; hal2Is F, Cl, Br or I, preferablyF, Cl or Br. . The synthesis of the compounds of formula (Ic) according to the invention can be obtained by reference to the synthetic procedure of scheme 3. Coupling a compound of formula I-A with a compound of formula I-B4 or a salt of a compound of formula I-B4 (e.g., hydrochloride, trifluoroacetate, hydrobromide, etc.) under suitable reagent conditions (e.g., DIPEA, etc.) to provide a compound of formula (Id-1); a compound of formula (Id-1) and formula hal3-R3The compound is reacted with a suitable reagent (e.g., Pd (PPh)3)2Cl2CuI and triethylamine) in a suitable solvent (such as THF, etc.) to obtain the compound of formula (Ic').

Examples

Intermediate 1: 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a ] pyridine

Step 1: 3-cyano-4-bromo-6-methoxypyrazolo [1,5-a ] pyridines

(E) -4-bromo-6-methoxypyrazolo [1,5-a ] pyridine-3-carbaldehyde oxime (10.0g,37.026mmol) was dissolved in acetic anhydride (250mL) in a 500mL single vial and reacted at 140 ℃ under reflux. The solution gradually changed from yellow to brown. The reaction was completed by TLC detection for 4.5 h. The solvent was distilled off at this temperature under reduced pressure, 100mL of water was added to the residue, stirring was carried out for 5min, then suction filtration was carried out, the filter cake was washed with 20mL of water and dried in a vacuum oven at 50 ℃ for 24h to give 6.83g of a gray solid with a yield of 73.2%.

Step 2: 3-cyano-4-bromo-6-hydroxypyrazolo [1,5-a ] pyridines

In a 1000mL single-neck flask, 3-cyano-4-bromo-6-methoxy pyrazolo [1,5-a]Pyridine (6.83g,27.1mmol) was dissolved in DCE (273mL) and AlCl was added in one portion3(10.8g,81.0mmol) and transferred to an 80 ℃ oil bath for reflux overnight. The reaction was complete by TLC. The reaction solution was cooled to room temperature, and 200mL of NaSO was added thereto with stirring4·10H2And stirring the THF solution of O for 4 hours, then carrying out suction filtration, collecting filtrate, and spin-drying to obtain 5.8g of brown solid. The yield thereof was found to be 90%.

And step 3: 3-cyano-4-bromo-6-ethoxypyrazolo [1,5-a ] pyridine

In a 250mL single vial, 3-cyano-4-bromo-6-hydroxypyrazolo [1,5-a ] pyridine (5g,21.005mmol) and potassium carbonate (8.7g,63mmol) were dissolved with DMA (125mL), iodoethane (5g,32.057mmol) was added with stirring, and after the addition was completed, the reaction mixture was transferred to 60 ℃. The reaction was carried out for 6h and the reaction was complete by TLC. To the reaction mixture was added 40mL of a solution of ammonia water to water (v: v) ═ 1:1, and the mixture was stirred for 1 hour, then filtered under suction, and the filter cake was washed with 50mL of water, and then spin-dried and suction-dried to obtain 4.48g of a gray solid with a yield of 80.2%.

And 4, step 4: 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a ] pyridine

Reacting 3-cyano-4-bromo-6-ethoxypyrazolo [1,5-a ] in a pressure tube]Pyridine (2.5g,9.4mmol), 2-fluoropyridine-5-boronic acid pinacol ester (2.5g,11mmol) were dissolved in 1, 4-dioxane (94mL), and Pd (PPh) was added3)4(1.1g,0.95mmol) and sodium carbonate solution (9.4mL,19mmol,2mol/L) were sealed and heated in a 100 ℃ oil bath. After 9h, heating was stopped and the reaction was complete by TLC. Adding 50mL of saturated ammonium chloride solution to quench reaction, transferring the reaction solution to a separating funnel, adding 300mL of EA and 50mL of saturated saline solution, oscillating, separating flocculent gray solid in the middle layer, performing suction filtration, separating gray solid, separating an organic phase from a filtrate, extracting an aqueous phase twice with 250mL of EA, combining the organic phase with the gray solid, performing spin-drying column chromatography (eluent EA: PE (v: v) ═ 4:1-1:1) to obtain a product, namely 2.7g of light yellow solid, and obtaining the yield of 100%.1H NMR(400MHz,CDCl3):δ8.39(s,1H),8.21(s,1H),8.18(d,1H),8.01(td,J=8.3,2.6Hz,1H),7.16(d,J=1.8Hz,1H),7.12(dd,J=8.4,2.9Hz,1H),4.11(q,J=6.9Hz,2H),1.51(t,J=6.9Hz,3H)。

Intermediate 2: 4- (6-Fluoropyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4-bromo-6-hydroxypyrazolo [1,5-a ] pyridine-3-carbonitrile

4-bromo-6-methoxypyrazolo [1,5-a ] in a 1000mL single-necked flask]Pyridine-3-carbonitrile (6.83g,27.1mmol) Using DCE (2)73mL) and adding AlCl in one time3(10.8g,81.0mmol) and transferred to an 80 ℃ oil bath for reflux overnight. The reaction was complete by TLC. The reaction solution was cooled to room temperature, and 200mL of NaSO was added thereto with stirring4·10H2And stirring the THF solution of O for 4 hours, then carrying out suction filtration, collecting filtrate, and spin-drying to obtain 5.8g of brown solid. The yield thereof was found to be 90%.

Step 2: 4- (6-Fluoropyridin-3-yl) -6-hydroxypyrazolo [1,5-a ] pyridine-3-carbonitrile

4-bromo-6-hydroxy-pyrazolo [1,5-a ] pyridine-3-carbonitrile (1.54g,6.47mmol), 2-fluoro-5- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (1.88g,8.43mmol), 2mol/L sodium carbonate (9.7mL,19mmol), 1, 4-dioxane (30.8mL), tetrakis (triphenylphosphine) palladium (0.75g,0.65mmol) were sequentially added to a 50mL sealed tube, purged with nitrogen for 10min with stirring, sealed and stirred at 100 ℃ for 7 h. Then cooled to room temperature and stirred overnight. The reaction mixture was directly filtered by suction, the cake was washed with a large amount of EA (80mL), 50mL of water was added to the filtrate, the aqueous phase was separated and extracted with ethyl acetate (50mL × 2), the organic phases were combined, washed with water (50mL), washed with saturated brine (50mL), dried over anhydrous sodium sulfate and purified by spin-dry column chromatography (eluent PE: EA (v: v) ═ 4:1-2:1) to give 1.14g of a pale yellow solid, with a yield of 69.3%.

And step 3: 4- (6-Fluoropyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a ] pyridine-3-carbonitrile

4- (6-Fluoropyridin-3-yl) -6-hydroxypyrazolo [1,5-a ] was added in this order to a 20mL pressure tube]Pyridine-3-carbonitrile (0.34g,1.3mmol), potassium carbonate (0.55g,4.0mmol), 2, 2-dimethyloxirane (0.96g,13mmol), DMF (4mL), sealed, reacted at 60 ℃ for 10h with magnetic stirring, and then heated to 80 ℃ for overnight reaction. After completion of the TLC detection reaction, the reaction mixture was added to cold 50mL of water, stirred at room temperature for 1 hour, extracted with EA (80 mL. times.3), dried over anhydrous sodium sulfate for the organic phase, and then subjected to spin-dry silica gel column chromatography (PE: EA (v: v) ═ 9:1-2:1) to collect 183mg of a white solid with a yield of 42%. LC-MS 327.1[ M +1 ]]+1H NMR(400MHz,CDCl3)δ8.40(d,1H),8.24(d,J=1.7Hz,1H),8.22(s,1H),8.05-7.98(m,1H),7.23(d,J=1.8Hz,1H),7.13(dd,J=8.5,2.8Hz,1H),3.88(s,2H),1.40(s,6H)。

Example 1: 6-ethoxy 4- (6- (4-ethynyl-4- (2-phenoxyethoxy) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4-hydroxy-4- ((trimethylsilyl) ethynyl) piperidine-1-carboxylic acid tert-butyl ester

A dry nitrogen-blanketed two-necked round bottom flask was charged with n-BuLi (8.2mL,13mmol,1.6mol/L in n-hexane), 20mL anhydrous THF (40mL), ethynyl (trimethyl) silane (1.84mL,13.0mmol) was added dropwise at-78 deg.C for 15min, the reaction was stirred at-78 deg.C for 30min, tert-butyl 4-oxopiperidine-1-carboxylate (2g,10.04mmol) was dissolved in 20mL anhydrous THF, the reaction was stirred at this temperature for 1h, allowed to warm to room temperature and stirred overnight. To the reaction mixture was added 80mL of water, extracted with ethyl acetate (80mL × 3), the organic phases were combined, washed with saturated sodium chloride and dried over anhydrous sodium sulfate, and subjected to silica gel column chromatography, the eluent was PE: EA (v: v) ═ 20:1-8:1, and 2.23g of a white solid was collected, with a yield of 74.78%.1H-NMR(600MHz,CDCl3):δ3.79(m,2H),3.21(ddd,J=13.2,9.7,3.2Hz,2H),2.22(s,1H),1.86(d,J=12.0Hz,2H),1.68–1.62(m,2H),1.45(s,9H),0.24(s,9H)。

Step 2: 4-ethynyl-4-hydroxypiperidine-1-carboxylic acid tert-butyl ester

Tert-butyl 4-hydroxy-4- ((trimethylsilyl) ethynyl) piperidine-1-carboxylate (853mg,2.87mmol) was dissolved in THF (14mL) at room temperature, to which tetrabutylammonium fluoride (3.4mL,3.4mmol,1mol/L in tetrahydrofuran) was added in one portion with stirring, and the reaction was continued at room temperature. After 5 hours of reaction, TLC was carried out to monitor that no starting material remained, water (10mL) was added and the reaction was quenched, THF was spin-dried under reduced pressure, the residue was extracted with EA (80 mL. times.2), the organic phases were combined, washed once with water (20mL) and saturated brine (20mL), dried over anhydrous sodium sulfate, spin-dried under reduced pressure, and purified by silica gel column chromatography (PE: EA (v: v) ═ 4:1) to give 640mg of a colorless transparent oil which was left to stand at room temperature as a white solid. The yield thereof was found to be 99%. LC-MS M/z-126.05 [ M-Boc +2H]+1H NMR(400MHz,CDCl3):δ3.76(brs,2H),3.36–3.20(m,2H),2.54(s,1H),2.11(s,1H),1.95-1.85(m,2H),1.75-1.67(m,2H),1.47(s,9H)。

And step 3: 4-ethynyl-4- (2-phenoxyethoxy) piperidine-1-carboxylic acid tert-butyl ester

4-ethynyl-4-hydroxypiperidine-1-carboxylic acid tert-butyl ester (400mg,1.78mmol) is dissolved with anhydrous DMF (9mL) at 0 ℃ under nitrogen protection, NaH (107mg,2.68mmol), NaI (533mg,3.55mmol) are added, a large number of bubbles are formed, after stirring for 15min 2-bromoethoxybenzene (714mg,3.55mmol) is added in one portion, and the mixture is transferred to room temperature and stirred overnight. 30mL of EA and 10mL of water were added to the reaction mixture, the mixture was shaken, the upper organic phase was separated, the aqueous phase was extracted twice with 30mL of EA, the organic phases were combined, washed with water (10 mL. times.3), dried over anhydrous sodium sulfate and then spin-dried, and the residue was purified by silica gel column chromatography (eluent EA: PE (v: v) ═ 1:8-1:5) to give 205mg of a colorless transparent oil in a yield of 33.4%. LC-MS (LC-MS) with M/z of 290.2[ M-t-Bu +2H [)]+1H NMR(400MHz,CDCl3):δ7.27(dd,J=10.5,5.0Hz,2H),6.98–6.88(m,3H),4.18–4.12(m,2H),3.93(t,J=5.1Hz,2H),3.68(d,J=4.2Hz,2H),3.42–3.25(m,2H),2.54(s,1H),1.90(d,J=3.9Hz,2H),1.83–1.73(m,2H),1.46(s,9H)。

And 4, step 4: 4-ethynyl-4- (2-phenoxyethoxy) piperidine 2,2, 2-trifluoroacetate salt

Tert-butyl 4-ethynyl-4- (2-phenoxyethoxy) piperidine-1-carboxylate (200mg,0.58mmol) was dissolved in DCM (4mL) in a single vial, and TFA (0.86mL,12mmol) was added dropwise with stirring at room temperature, after which stirring at this temperature was continued. The reaction was completed by TLC after 5 h. The reaction solution was directly spin-dried to give 240mg of a pale yellow oil, which was subjected to the next reaction according to theoretical yield without further purification, the yield being 100%. And 5: 6-ethoxy-4- (6- (4-ethynyl-4- (2-phenoxyethoxy) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a ] was added to a 10mL microwave tube]Pyridine (80mg,0.28mmol) (intermediate 1), 4-ethynyl-4- (2-phenoxyethoxy) piperidine trifluoroacetate (208mg,0.58mmol), DMSO (1.42mL) was added to dissolve it, DIPEA (0.25mL,1.5mmol) was added, and the reaction was carried out for 5h using microwave (temperature 150 ℃ C., pressure 10 bar). TLC detectionThe reaction was complete. The reaction mixture was diluted with 10mL of water, and EA was added thereto for extraction (30mL × 3), the organic phases were combined, washed once with 30mL of saturated brine, dried over anhydrous sodium sulfate and then spin-dried, and the residue was purified by silica gel column chromatography (eluent EA: PE (v: v) ═ 1:8-1:4) to give 100mg of a pale yellow solid. The yield thereof was found to be 69.52%. LC-MS M/z 508.30[ M + H ]]+1H-NMR(600MHz,CDCl3)δ8.33(d,J=2.3Hz,1H),8.19(s,1H),8.10(d,J=1.6Hz,1H),7.71(s,1H),7.28(d,J=8.7Hz,2H),7.09(s,1H),6.94(t,J=8.1Hz,3H),6.80(d,J=7.8Hz,1H),4.20–4.15(m,2H),4.08(q,J=7.0Hz,2H),3.99(dd,J=10.0,4.9Hz,4H),3.63–3.55(m,2H),2.57(s,1H),2.05(d,J=9.5Hz,2H),1.98–1.91(m,2H),1.50(t,J=7.0Hz,3H)。

Example 2: 6-ethoxy-4- (6- (4- (3- (2-phenoxyethoxy) prop-1-yn-1-yl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4- (3- (2-Phenoxyethoxy) prop-1-yn-1-yl) piperidine-1-carboxylic acid tert-butyl ester

Tert-butyl 4- (3-hydroxyprop-1-yn-1-yl) piperidine-1-carboxylate (177mg,0.74mmol) was dissolved in DMF (3.7mL) at room temperature, and potassium hydroxide (83mg,1.48mmol), (2-bromoethoxy) benzene (297mg, 1.48mmol) and water (0.5mL) were added in that order and heated to 60 ℃ for 24 h. Saturated NH for reaction4The reaction was diluted with EA (100mL), and the organic phase was washed with water (20mL), brine (20 mL). Dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 10:1-4:1) to obtain 75mg of the objective product as a colorless oil, yield: 28 percent. LC-MS (sodium chloride-sodium chloride) with M/z being 304.2[ M-t-Bu +2H]+,260.1[M-Boc+2H]+1H NMR(400MHz,CDCl3):δ7.30–7.25(m,2H),6.93(t,J=7.2Hz,3H),4.27(d,J=1.7Hz,2H),4.15(dd,J=9.2,4.3Hz,2H),3.91–3.84(m,2H),3.74–3.63(m,2H),3.23-3.12(m,2H),2.61(s,1H),1.81–1.72(m,2H),1.64(s,2H),1.46(s,9H)。

Step 2: 4- (3- (2-Phenoxyethoxy) prop-1-yn-1-yl) piperidin-1-ium 2,2, 2-trifluoroacetate salt

Tert-butyl 4- (3- (2-phenoxyethoxy) prop-1-yn-1-yl) piperidine-1-carboxylate (79mg,0.23mmol) was dissolved in dichloromethane (4.5mL) at room temperature, to which trifluoroacetic acid (0.34mL,4.6mmol) was added and stirring was continued at room temperature overnight. TLC monitored that no starting material remained. The reaction solution was spin-dried under reduced pressure to obtain a pale yellow solid 80mg (97% yield), which was a crude product, which was directly fed without purification for the next reaction. LC-MS M/z 246.35[ M-CF ]3COO]+1H-NMR(400MHz,CDCl3):δ8.79(brs,2H),7.28(t,J=8.0Hz,2H),6.96(t,J=7.3Hz,1H),6.90(d,J=8.2Hz,2H),4.14(d,J=4.9Hz,2H),3.92(d,J=4.9Hz,2H),3.33-3.24(m,2H),3.28(d,J=28.5Hz,4H),2.63(s,1H),2.17(t,J=5.4Hz,4H)。

And step 3: 6-ethoxy-4- (6- (4- (3- (2-phenoxyethoxy) prop-1-yn-1-yl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

4- (3- (2-Phenoxyethoxy) prop-1-yn-1-yl) piperidin-1-ium 2,2, 2-trifluoroacetate (74mg,0.20mmol), 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyridine (intermediate 1, 51mg, 0.18mmol) and potassium carbonate (74mg,0.54mmol) were weighed into a 10mL flask in sequence, the flask was replaced with nitrogen, DMSO (2mL) was added for dissolution, and the reaction was heated to 120 ℃ in an oil bath for 3 days. The heating was stopped. The reaction mixture was extracted with EA (30mL × 3), the organic phases were combined, washed with water (20mL), and saturated brine (20mL), dried over anhydrous sodium sulfate, and dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 4:1-2:1) to obtain 20.1mg of a white solid, with a yield of 21.3%. LC-MS M/z 522.20[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.32(s,1H),8.19(s,1H),8.10(s,1H),7.70(d,J=8.6Hz,1H),7.29-7.25(m,2H),7.08(s,1H),6.97-6.88(m,3H),6.77(d,J=8.8Hz,1H),4.29(s,2H),4.19-4.14(m,2H),4.08(dd,J=13.7,6.9Hz,2H),4.00-3.92(m,2H),3.92-3.86(m,2H),3.48-3.37(m,2H),2.73(s,1H),1.97-1.90(m,2H),1.76-1.70(m,2H),1.49(t,J=6.6Hz,3H)。

Example 3: 6-ethoxy-4- (6- (4- ((3-fluorophenyl) ethynyl) -4-methoxypiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4- ((3-fluorophenyl) ethynyl) -4-hydroxypiperidine-1-carboxylic acid tert-butyl ester

1-ethynyl-3-fluoro-benzene (0.25g,2.1mmol), anhydrous THF (3mL) were added sequentially to a 25mL nitrogen-blanketed, magnetic stir bar in a two-necked flask with syringe, stirred at-78 ℃ for 5min, n-BuLi in tetrahydrofuran (1.4mL,2.2mmol,1.6mol/L) was injected over 10min and stirred for 30min, then a mixed solution of tert-butyl 4-oxopiperidine-1-carboxylate (400mg,2.01mmol) and anhydrous THF (1mL) was added over 15min, reacted at-78 ℃ and monitored by TLC. After 2h, the reaction was allowed to warm up naturally, 15mL of water and 30mL of ethyl acetate were slowly added to the reaction, the aqueous phase was separated and extracted with 30mL of ethyl acetate, the organic phase was combined and washed with 15mL of saturated brine, the organic phase was dried over anhydrous sodium sulfate and then spin-dried, and purified by silica gel column chromatography (eluent PE: EA (v: v) ═ 8:1-4:1) to obtain 0.425g of a colorless viscous liquid with a yield of 66.3%. LC-MS M/z 342.1[ M + Na ]]+1H NMR(400MHz,CDCl3):δ7.27(t,J=6.9Hz,1H),7.20(d,J=7.7Hz,1H),7.12(d,J=9.3Hz,1H),7.04(dd,J=11.5,5.3Hz,1H),3.88–3.75(m,2H),3.37–3.28(m,2H),2.35(s,1H),2.02–1.94(m,2H),1.83–1.75(m,2H),1.47(s,9H)。

Step 2: 4- ((3-fluorophenyl) ethynyl) -4-methoxypiperidine-1-carboxylic acid tert-butyl ester

Under nitrogen, a 50mL two-necked flask was charged with tert-butyl 4- ((3-fluorophenyl) ethynyl) -4-hydroxypiperidine-1-carboxylate (420mg,1.31mmol) and DMF (8.5mL), NaH (0.08g,2mmol, 60% content) was added portionwise at 0 deg.C, the mixture was stirred at room temperature for 1h, iodomethane (0.17mL,2.7mmol) was added, and the reaction was stirred overnight. The reaction solution was poured into 25mL of ice water, extracted with ethyl acetate (30mL × 3), the organic phase was washed with saturated sodium chloride (20mL), dried over anhydrous sodium sulfate, and purified by silica gel column chromatography (PE: EA (v: v) ═ 40:1-7:1) to give 390mg of a colorless transparent liquid, with a yield of 89%. LC-MS M/z 356.1[ M + Na ]]+

And step 3: 4- ((3-fluorophenyl) ethynyl) -4-methoxypiperidine 2,2, 2-trifluoroacetate

After adding DCM (4mL) and TFA (1.2mL) to a 50mL single vial containing tert-butyl 4- ((3-fluorophenyl) ethynyl) -4-methoxypiperidine-1-carboxylate (0.39g,1.2mmol), the reaction turned yellow-brown, stirred at room temperature and checked for completion by TLC after 4 h. The reaction solution is directly spun to dry the solvent to obtain a colorless transparent oily liquid product, and the yield is calculated by 100%.1H NMR(400MHz,CDCl3):δ8.18(s,2H),7.34–7.27(m,1H),7.23(d,J=7.7Hz,1H),7.15(d,J=9.2Hz,1H),7.08(td,J=8.3,2.0Hz,1H),3.45(s,3H),3.43–3.31(m,4H),2.30–2.16(m,4H)。

And 4, step 4: 6-ethoxy-4- (6- (4- ((3-fluorophenyl) ethynyl) -4-methoxypiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

To a microwave tube were added 4- ((3-fluorophenyl) ethynyl) -4-methoxypiperidin-1-ium 2,2, 2-trifluoroacetate (0.31g,0.89mmol), 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyridine (100mg,0.3542mmol) and DMSO (3mL), N-diisopropylethylamine (0.25mL,1.5mmol) were reacted at 135 ℃ for 5h with a microwave. EA (30mL) and water (20mL) were added, the aqueous phase was extracted with EA (30mL × 2), the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate and filtered, the filtrate was spin-dried, and silica gel column chromatography was performed on the residue (PE: EA (v: v) ═ 10:1-3:1) to give 99mg of the product as a pale yellow solid with a yield of 56.4%. LC-MS (LC-MS) M/z is 496.1[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.34(s,1H),8.19(s,1H),8.10(s,1H),7.72(d,J=7.4Hz,1H),7.26(s,2H),7.16(d,J=9.0Hz,1H),7.06(d,J=18.7Hz,2H),6.82(d,J=8.5Hz,1H),4.16–3.97(m,4H),3.64–3.53(m,2H),3.49(s,3H),2.21–2.07(m,2H),2.00–1.89(m,2H),1.55–1.44(m,3H)。

Example 4: 6-ethoxy-4- (6- (4-methoxy-4- (pyrimidin-5-ylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4-hydroxy-4- (pyrimidin-5-ylethynyl) piperidine-1-carboxylic acid tert-butyl ester

5-ethynylpyrimidine (250mg,2.40mmol) was dissolved in anhydrous THF (6mL) under nitrogen, cooled to-78 deg.C, and a solution of n-butyllithium in tetrahydrofuran (1.5mL,2.4mmol,1.60mol/L) was added dropwise slowly, with the temperature being maintained over, and the reaction was continued for 20 min. A solution of tert-butyl 4-oxopiperidine-1-carboxylate (400mg,2.01mmol) in dry THF (4mL) was added dropwise to the reaction flask over 3min, and the reaction was continued for 1.5h while maintaining the temperature. The reaction was quenched with saturated ammonium chloride (8 mL). The aqueous phase was extracted with DCM (30mL × 2), the organic phases were combined, washed with saturated brine (20mL), dried over anhydrous sodium sulfate, dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 2:1-1.5:1 (with 1% triethylamine)) to give 406mg of a white solid, which was the target product in 66.7% yield. LC-MS M/z 248.25[ M-t-Bu + 2H%]+1HNMR(400MHz,CDCl3):δ9.15(s,1H),8.77(s,2H),3.77(s,2H),3.42–3.32(m,2H),2.01(d,J=13.9Hz,2H),1.88–1.78(m,2H),1.47(s,9H)。

Step 2: 4-methoxy-4- (pyrimidin-5-ylethynyl) piperidine-1-carboxylic acid tert-butyl ester

Tert-butyl 4-hydroxy-4- (pyrimidin-5-ylethynyl) piperidine-1-carboxylate (400mg,1.32mmol) was dissolved in anhydrous DMF (6.5mL) under nitrogen and sodium hydride (94mg, 2.35mmol, 60% content) was added in one portion and stirred at room temperature for 30 min. Methyl iodide (0.2mL,3.00mmol) was then added and stirred overnight. The reaction was quenched with saturated ammonium chloride solution (5mL) and diluted with EA (50mL) and water (10 mL). The aqueous phase was extracted with EA (25mL × 2), the organic phases were combined, washed with water (20mL × 2), saturated brine (20mL), dried over anhydrous sodium sulfate, and spin-dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 2:1) to give 217mg of a colorless transparent oil, which was the target product in a yield of 51.9%. LC-MS M/z 262.05[ M-t-Bu + 2H%]+1H NMR(400MHz,CDCl3):δ9.15(s,1H),8.78(s,2H),3.75–3.65(m,2H),3.44(s,3H),3.43–3.34(m,2H),2.03-1.95(m,2H),1.88-1.79(m,2H),1.47(s,9H)。

And step 3: 4-methoxy-4- (pyrimidin-5-ylethynyl) piperidin-1-ium 2,2, 2-trifluoroacetate salt

4-methoxy-4- (pyrimidine-Tert-butyl 5-ylethynyl piperidine-1-carboxylate (207mg,0.65mmol) was dissolved in DCM (6.5mL), and trifluoroacetic acid (1.0mL,13mmol) was added thereto with stirring for reaction overnight. And (3) decompressing and spin-drying the reaction solvent to obtain a crude product, and directly putting the crude product into the next step of reaction without purification according to 100 percent of yield. LC-MS M/z 218.10[ M-CF ]3COO]+1H NMR(400MHz,CDCl3):δ9.25(s,1H),8.96(s,2H),8.24(d,J=38.4Hz,2H),3.46(s,3H),3.39(s,4H),2.30(d,J=4.9Hz,4H)。

And 4, step 4: 6-ethoxy-4- (6- (4-methoxy-4- (pyrimidin-5-ylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

4-methoxy-4- (pyrimidin-5-ylethynyl) piperidin-1-ium 2,2, 2-trifluoroacetate (120mg,0.36mmol) and 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a ]]Pyridine (intermediate 1, 50mg,0.18mmol) was dissolved in DMSO (2mL), DIPEA (0.22mL,1.3mmol) was added thereto, and the reaction was microwaved for 3h (150 ℃,10 bar, pre-stirred for 30 s). After the reaction mixture was cooled to room temperature, EA (100mL) was added to dilute the reaction mixture, the organic phase was washed successively with water (20mL) and saturated brine (20mL), dried over anhydrous sodium sulfate, filtered, the filtrate was spin-dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 2:1-1:1) to obtain 18.6mg of a pale yellow solid, with a yield of 21.9%, which was the target product. LC-MS M/z 480.30[ M + H ]]+1H NMR(400MHz,CDCl3):δ9.15(s,1H),8.80(s,2H),8.33(d,J=2.3Hz,1H),8.19(s,1H),8.10(d,J=1.9Hz,1H),7.72(dd,J=8.8,2.5Hz,1H),7.08(d,J=1.9Hz,1H),6.82(d,J=8.9Hz,1H),4.08(q,J=6.9Hz,2H),4.02-3.94(m,2H),3.68-3.56(m,2H),3.49(s,3H),2.20-2.08(m,2H),2.05-1.93(m,2H),1.49(t,J=7.0Hz,3H)。

Example 5: 6-ethoxy-4- (6- (4- (3-hydroxypropan-1-yn-1-yl) -4-methoxypiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4- (3- (tert-butoxy) prop-1-yn-1-yl) -4-hydroxypiperidine-1-carboxylic acid tert-butyl ester

Under the protection of nitrogen, 3- (tert-butoxy) prop-1-yne (0.32mL,2.4mmol) is dissolved in anhydrous THF (6mL), the temperature is reduced to-78 ℃, n-hexane solution of n-butyllithium (1.5mL,2.4mmol,1.60mol/L) is slowly added dropwise, and the reaction is continued for 20min after the temperature is kept. A solution of tert-butyl 4-oxopiperidine-1-carboxylate (400mg,2.01mmol) in dry THF (4mL) was added dropwise to the reaction flask over 3min, and the reaction was continued for 1.5h while maintaining the temperature. The reaction was quenched with saturated ammonium chloride (8mL), the aqueous phase was extracted with DCM (30mL × 2), the organic phases were combined, washed with saturated brine (20mL), dried over anhydrous sodium sulfate, and spin-dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 4:1-2:1) to obtain 595mg (yield 95.2%) of a colorless transparent oil, which was the target product. LC-MS M/z 334.20[ M + Na ]]+1H NMR(400MHz,CDCl3):δ4.12(s,2H),3.80-3.63(m,2H),3.35-3.20(m,2H),1.87(d,J=12.9Hz,2H),1.74-1.63(m,2H),1.48(s,1H),1.44(s,9H),1.23(s,9H)。

Step 2: 4- (3- (tert-butoxy) prop-1-yn-1-yl) -4-methoxypiperidine-1-carboxylic acid tert-butyl ester

Tert-butyl 4- (3- (tert-butoxy) prop-1-yn-1-yl) -4-hydroxypiperidine-1-carboxylate (590mg,1.90mmol) was dissolved in anhydrous DMF (9.5mL) under nitrogen protection, sodium hydride (136mg,3.40mmol, 60% content) was added in one portion, and the mixture was stirred at room temperature for 30 min. Methyl iodide (0.30mL,5.0mmol) was then added and stirred overnight. The reaction was quenched with saturated ammonium chloride solution (5mL), diluted with EA (50mL) and water (10mL), the aqueous phase was extracted with EA (25 mL. times.2), the organic phases were combined, washed sequentially with water (20 mL. times.2), saturated brine (20mL), dried over anhydrous sodium sulfate, and spun dry under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 10: -6:1) to give 498mg (yield 80.8%) of a colorless clear oil, which was the target product. LC-MS M/z 348.15[ M + Na ]]+1H NMR(400MHz,CDCl3):δ4.15(s,2H),3.71-3.59(m,2H),3.35(s,3H),3.34-3.24(m,2H),1.92-1.81(m,2H),1.74-1.66(m,2H),1.44(s,9H),1.24(s,9H)。

And step 3: 4- (3-hydroxy-1-yn-1-yl) -4-methoxypiperidin-1-ium 2,2, 2-trifluoroacetate salt

Tert-butyl 4- (3- (tert-butoxy) prop-1-yn-1-yl) -4-methoxypiperidine-1-carboxylate (498 m) at room temperatureg,1.53) was dissolved in dichloromethane (15mL), and trifluoroacetic acid (2.2mL,30mmol) was added thereto with stirring and reacted overnight. And (4) decompressing and spin-drying the reaction solvent to obtain a crude product, and directly putting the crude product into the next step of reaction without purification. LC-MS M/z 170.10[ M-CF ]3COO]+1H NMR(400MHz,CDCl3):δ8.10(s,2H),4.99(s,2H),3.37(s,3H),3.35-3.26(m,4H),2.15(t,J=5.3Hz,4H)。

And 4, step 4: 6-ethoxy-4- (6- (4- (3-hydroxypropan-1-yn-1-yl) -4-methoxypiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

4- (3-hydroxy-1-yn-1-yl) -4-methoxypiperidin-1-ium 2,2, 2-trifluoroacetate (125mg,0.44mmol) and 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyridine (intermediate 1, 50mg,0.18mmol) was dissolved in DMSO (2mL), DIPEA (0.2mL,1mmol) was added thereto, and the reaction was microwaved for 4h (150 ℃,10 bar, pre-stirred for 30 s). After the reaction mixture was cooled to room temperature, EA (100mL) was added to dilute the reaction mixture, the organic phase was washed with water (20mL), saturated brine (20mL) in this order, dried over anhydrous sodium sulfate, filtered, and spin-dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 1.5:1-1:1) to obtain 47.8mg (yield 62.5%) of a pale yellow solid, which was the target product. LC-MS M/z 432.20[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.32(d,J=2.2Hz,1H),8.18(s,1H),8.10(s,1H),7.70(dd,J=8.8,2.4Hz,1H),7.08(s,1H),6.79(d,J=8.9Hz,1H),4.36(s,2H),4.08(dd,J=13.9,7.0Hz,2H),4.00-3.89(m,2H),3.58-3.48(m,2H),3.41(s,3H),2.07-1.99(m,2H),1.90-1.82(m,2H),1.49(t,J=6.9Hz,3H)。

Example 6: 6-ethoxy-4- (6- (4- (4-methoxybenzoyl) -3- (prop-1-yn-1-yl) piperazin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 3-Oxopiperazine-1-carboxylic acid benzyl ester

A100 mL reaction flask was charged with piperazin-2-one (2.5g,25mmol) and sodium carbonate (7.95g,75.0mmol), followed by dioxane (41mL) and water (10.3mL), and benzyl chloroformate (3.5mL,25mmol) was added slowly dropwise at room temperature over about 5min, with stirring continued at room temperature for 2.5 h. The reaction was diluted with water (90mL), EA extracted (100 mL. times.3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was rotary dried under reduced pressure to give 5.8g of a white solid, which was the crude product, which was directly fed for the next reaction without purification.

Step 2: 2-oxopiperazine-1, 4-dicarboxylic acid 1-tert-butyl 4-benzyl ester

Benzyl 3-oxopiperazine-1-carboxylate (5.8g,25mmol) was dissolved in dichloromethane (35mL) at room temperature, triethylamine (8.8mL) and DMAP (305mg,2.50mmol) were added in this order, di-tert-butyl dicarbonate (8.6mL,37mmol) was added dropwise over about 5min, and stirring at room temperature was continued for 2 h. The reaction mixture was quenched with water (30mL), the mixture was poured into a separatory funnel, the aqueous phase was extracted with DCM (40mL × 2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was spin-dried under reduced pressure to give a brown oil, which was then purified by silica gel column chromatography (PE: EA (v: v) ═ 4:1) to give 8.3g (yield: 100%) of a pale yellow oil, which was the target product. LC-MS M/z 357.15[ M + Na ]]+1H NMR(400MHz,CDCl3):δ7.35(s,5H),5.15(s,2H),4.24(s,2H),3.87–3.75(m,2H),3.75–3.63(m,2H),1.53(s,9H).。

And step 3: (2- ((tert-Butoxycarbonyl) amino) ethyl) (2-oxopent-3-yn-1-yl) carbamic acid benzyl ester

A50 mL reaction flask was charged with 4-benzyl 2-oxopiperazine-1, 4-dicarboxylic acid 1-tert-butyl ester (2.29g,6.85mmol) and the nitrogen was replaced three times, the mixture was dissolved in anhydrous THF (8.1mL), the temperature was reduced to 0 ℃ and a solution of 1-propynylmagnesium bromide (32.5mL,16.3mmol,0.50mol/L) in THF was slowly added dropwise over about 30min, and the reaction was continued at 0 ℃ for 2 h. Adding saturated NH4The reaction was quenched with Cl solution (30mL), the aqueous phase extracted with DCM (40 mL. times.2), the organic phases combined, dried over anhydrous sodium sulfate and spin dried under reduced pressure. The residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 4:1-2:1) to give 2.23g (yield 87.1%) of a pale yellow viscous oil, which was the objective product LC-MS: M/z ═ 275.2[ M-Boc + 2H:]+1H NMR(400MHz,CDCl3):δ7.40–7.28(m,5H),5.13(d,J=19.6Hz,2H),5.01(s,1H),4.15(d,J=24.6Hz,2H),3.51–3.38(m,2H),3.32–3.15(m,2H),2.00(d,J=12.9Hz,3H),1.75(s,1H),1.42(d,J=4.2Hz,9H)。

and 4, step 4: 3- (prop-1-yn-1-yl) piperazine-1-carboxylic acid benzyl ester

Benzyl (2- ((tert-butoxycarbonyl) amino) ethyl) (2-oxopent-3-yn-1-yl) carbamate (1.28g,3.42mmol) was dissolved in dichloromethane (4.8mL), cooled in an ice-water bath, trifluoroacetic acid (2.5mL,34mmol) was added, the cold bath was removed, warmed to room temperature, and stirring was continued for 15 min. Sodium triacetoxyborohydride (2.9g,14mmol) was added portionwise over about 10 min. Stirring was continued at room temperature for 24 h. The solvent was dried under reduced pressure, and the residue was diluted with EA (100mL), washed with water (20mL) and saturated brine (20mL) in this order, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (DCM: MeOH (v: v) ═ 50:1-20:1) to give 300mg of a yellowish brown oil (yield 34%), which was the target product. LC-MS M/z 259.20[ M + H ]]+1HNMR(400MHz,CDCl3):δ7.54-7.29(m,5H),5.15(d,J=14.9Hz,2H),4.06(s,1H),3.99-3.59(m,4H),3.40(s,1H),3.06(s,1H),1.76(s,3H)。

And 5: 4- (4-Methoxybenzoyl) -3- (prop-1-yn-1-yl) piperazine-1-carboxylic acid benzyl ester

Benzyl 3- (prop-1-yn-1-yl) piperazine-1-carboxylate (300mg,1.16mmol) was dissolved in DCM (11mL) under nitrogen at room temperature, to which was added 4-methoxybenzoyl chloride (297mg,1.74mmol) and triethylamine (0.4mL,3mmol) in that order, and reacted overnight. The reaction mixture was diluted with DCM (50mL), washed with water (20mL) and saturated brine (20mL) in this order, dried over anhydrous sodium sulfate, filtered, dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 2:1) to give 262mg of yellow oil (yield 57.5%) which was the target product. LC-MS (sodium-magnesium chloride) with M/z 415.1[ M + Na ]]+,393.2[M+H]+1H NMR(400MHz,CDCl3):δ7.47(d,J=8.0Hz,2H),7.43–7.30(m,5H),6.95(d,J=8.6Hz,2H),5.40–4.96(m,3H),4.36–4.05(m,3H),3.86(s,3H),3.47(s,1H),3.16–2.83(m,2H),1.78(s,3H)。

Step 6: (4-methoxyphenyl) (2- (prop-1-yn-1-yl) piperazin-1-yl) methanone

4- (4-methoxybenzoyl) -3- (propane-1-carboxylic acid) at room temperatureBenzyl alkynyl-1-yl) piperazine-1-carboxylate (260mg,0.66mmol) in trifluoroacetic acid (6.6mL) was added trifluoromethanesulfonic acid (0.3mL,3mmol) and stirred at room temperature for 4 h. The pH of the reaction solution was adjusted to 12 with a saturated sodium carbonate solution. Extraction with DCM (100 mL. times.2), combining the organic phases, washing with saturated brine (50mL), drying over anhydrous sodium sulfate, filtering, and spin-drying the filtrate under reduced pressure to obtain the crude product. The reaction mixture was used in the next reaction without further purification. LC-MS M/z 259.20[ M + H ]]+1H NMR(400MHz,CDCl3):δ7.44(d,J=7.8Hz,2H),6.92(d,J=8.5Hz,2H),4.01-3.70(m,5H),3.40-3.24(m,1H),3.10–2.67(m,4H),1.89(d,J=1.9Hz,3H)。

And 7: 6-ethoxy-4- (6- (4- (4-methoxybenzoyl) -3- (prop-1-yn-1-yl) piperazin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

(4-methoxyphenyl) (2- (prop-1-yn-1-yl) piperazin-1-yl) methanone (68mg,0.26mmol) and 3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyridine (intermediate 1, 25mg,0.089mmol) was dissolved in DMSO (1mL), DIPEA (0.10mL,0.60mmol) was added and the reaction was microwaved for 6h (150 ℃,10 bar, pre-stirred for 30 s). After the reaction was completed, the reaction mixture was cooled to room temperature, EA (100mL) was added to dilute the reaction mixture, the organic phase was washed with water (20mL), saturated brine (20mL), dried over anhydrous sodium sulfate, filtered, and spin-dried under reduced pressure, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 2:1-1:1) to obtain 8.7mg (yield 20%) of a pale yellow solid, which was the target product. LC-MS M/z 521.30[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.34(d,J=2.1Hz,1H),8.19(s,1H),8.11(s,1H),7.74(dd,J=8.8,2.3Hz,1H),7.51(d,J=8.2Hz,2H),7.09(d,J=1.6Hz,1H),6.95(d,J=8.6Hz,2H),6.83(d,J=8.9Hz,1H),4.44(t,J=14.4Hz,2H),4.08(q,J=6.9Hz,2H),3.93–3.78(m,4H),3.61(s,1H),3.19(d,J=11.3Hz,1H),3.06–2.94(m,2H),1.78(d,J=1.7Hz,3H),1.49(t,J=6.9Hz,3H)。

Example 7: 4- (6- (4-ethynyl-4-hydroxypiperidin-1-yl) pyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4-ethynyl-4-hydroxypiperidine-1-carboxylic acid tert-butyl ester

Magnesium bromo (ethynyl) (10mL,5mmol,0.5mol/L in THF) was added to a two-necked flask under nitrogen protection at 0 deg.C, tert-butyl 4-oxopiperidine-1-carboxylate (1g,5.0188mmol) dissolved in THF (5mL) was slowly injected into the reaction mixture, and after 30min at 0 deg.C, the mixture was allowed to slowly warm to room temperature for 4.5 h. Saturated ammonium chloride solution (30mL) is added into the reaction solution, EA (50mL) is added after stirring for 5min, the water phase is separated and extracted by EA (50mL), the organic phase is combined, the mixture is washed by saturated saline solution (30mL), dried by anhydrous sodium sulfate, filtered, the filtrate is concentrated, and the residue is purified by silica gel column chromatography (eluent PE: EA (v: v) ═ 5:1-3:1) to obtain 460mg of colorless oily matter (yield is 40.68 percent), namely the target product.1HNMR(400MHz,CDCl3):δ3.83-3.72(m,2H),3.32-3.22(m,2H),2.53(s,1H),2.13(s,1H),1.93-1.85(m,2H),1.76-1.66(m,2H),1.46(s,9H)。

Step 2: 4-ethynylpiperidin-4-ol hydrochloride

In a 100mL single-necked flask, tert-butyl 4-ethynyl-4-hydroxypiperidine-1-carboxylate (450mg,2.00mmol), HCl/MeOH (30mL,120mmol,4mol/L) were added in this order and stirred at room temperature overnight. Directly spin-drying the reaction liquid to obtain a light yellow solid, namely the target product, and putting the light yellow solid into the next reaction without further purification.1H NMR(400MHz,CDCl3):δ3.10-3.01(m,2H),2.91-2.81(m,2H),2.52(s,1H),1.98-1.91(m,2H),1.73-1.65(m,2H)。

And step 3: 4- (6- (4-ethynyl-4-hydroxypiperidin-1-yl) pyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Weighing 4- (6-fluoropyridin-3-yl) -6- (2-hydroxy-2-methylpropyloxy) pyrazolo [1,5-a in 10mL microwave tube]Pyridine-3-carbonitrile (intermediate 2, 25mg,0.077mmol), 4-ethynylpiperidine-4-ol hydrochloride (20mg,0.16mmol), dissolved in DMSO (1.5mL), followed by the addition of DIPEA (0.1mL,0.6mmol) and microwave (135 deg.C, 10bar pressure) for 5 h. Pouring the reaction solution into water (20mL), adding EA for extraction (40mL × 3), combining organic phases, washing with saturated saline (30mL), drying the organic phase with anhydrous sodium sulfate, filtering, and filtering the filtrateConcentration and purification of the residue on silica gel column chromatography (eluent DCM: MeOH (v: v) ═ 100:1-30:1) gave 4mg (yield 12.10%) of a pale yellow solid, the expected product. LC-MS M/z 432.2[ M + H ]];1H NMR(400MHz,CDCl3):δ8.33(d,J=2.0Hz,1H),8.19(s,1H),8.15(d,J=1.7Hz,1H),7.70(dd,J=8.8,2.4Hz,1H),7.14(d,J=1.7Hz,1H),6.80(d,J=8.8Hz,1H),4.11-4.04(m,2H),3.86(s,2H),3.54-3.48(m,2H),2.57(s,1H),2.06-2.03(m,2H),1.88-1.85(m,2H),1.39(s,6H)。

Example 8: 6- (2-hydroxy-2-methylpropoxy) -4- (6- (4-hydroxy-4- (phenylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4-hydroxy-4- (phenylethynyl) piperidine-1-carboxylic acid tert-butyl ester

N2Under protection, THF (5mL), phenylacetylene (0.33mL,3.0mmol) and n-BuLi (2.5mol/L n-hexane solution, 1.25mL) are added sequentially in a 50mL double-neck flask at a low temperature of 78 ℃. Tert-butyl 4-oxopiperidine-1-carboxylate (498mg,2.50mmol) was dissolved in 3mL THF and added slowly to the reaction over 15 min. The reaction was maintained at-78 ℃ for 3 h. The reaction mixture was added with 15mL of a saturated ammonium chloride solution, EA extraction (15mL × 3) was performed, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 4:1 as an eluent) to obtain 580mg of a solid. The yield is 76 percent, namely the target product. LC-MS (sodium-magnesium chloride) with M/z of 324.3[ M + Na ]]+1HNMR(400MHz,CDCl3):δ7.45-7.37(m,2H),7.34-7.28(m,3H),3.81(m,2H),3.36-3.27(m,2H),2.41(s,1H),1.98(m,2H),1.83-1.74(m,2H),1.46(s,9H)。

Step 2: 4- (Phenylethynyl) piperidin-4-ol trifluoroacetate salt

A50 mL single vial was charged with tert-butyl 4-hydroxy-4- (phenylethynyl) piperidine-1-carboxylate (92mg,0.31mmol), DCM (4mL), TFA (0.50mL) slowly and stirred at rt for 25 min. The reaction solution was dried and evacuated to give 310mg of pale yellow oil, which was directly used for the next reaction. LC-MS (LC-MS) with m/z 202.2[M+H]+

And step 3: 6- (2-hydroxy-2-methylpropoxy) -4- (6- (4-hydroxy-4- (phenylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

4- (6-fluoropyridin-3-yl) -6- (2-hydroxy-2-methylpropyloxy) pyrazolo [1, 5-a) is sequentially added into a 5mL microwave tube]Pyridine-3-carbonitrile (intermediate 2, 39mg,0.12mmol), 4- (phenylethynyl) piperidin-4-ol trifluoroacetate (0.31mmol), DMSO (3mL), N, N-diisopropylethylamine (0.13mL,0.79mmol), reacted at 110 ℃ for 5 h. EA (20mL) was added to the reaction mixture, washed with water (80 mL. times.3), the aqueous phases combined, EA extracted (15mL), the organic phases combined, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated. The residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 2:3) to obtain 35mg (yield 58%) of a pale yellow solid, which was the objective product. LC-MS (LC-MS) with M/z being 508.1[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.34(d,J=2.3Hz,1H),8.20(s,1H),8.15(d,J=2.0Hz,1H),7.71(dd,J=8.8,2.5Hz,1H),7.45(dd,J=6.5,3.1Hz,2H),7.31(m,3H),7.14(d,J=2.0Hz,1H),6.82(d,J=8.9Hz,1H),4.18–4.09(m,2H),3.86(s,2H),3.56(m,2H),2.36(s,1H),2.14(m,3H),1.99–1.90(m,2H),1.39(s,6H)。

Example 9: 6-ethoxy-4- (6- (4-hydroxy-4- (phenylethynylpiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4-hydroxy-4- (phenylethynyl) piperidine-1-carboxylic acid tert-butyl ester

N2Under protection, THF (5mL), phenylacetylene (0.33mL,3.0mmol) and n-BuLi (2.5mol/L n-hexane solution, 1.25mL) are sequentially added into a 50mL double-neck flask at the low temperature of 78 ℃. Tert-butyl 4-oxopiperidine-1-carboxylate (498mg,2.50mmol) was dissolved in 3mL THF and added slowly to the reaction mixture over 15 min. The reaction was maintained at-78 ℃ for 3 h. Adding 15mL of saturated ammonium chloride solution into the reaction solution, extracting with EA (15 mL. times.3), combining organic phases, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, and separating the residue by silica gel column chromatography (PE: EA (v: v) ═ 4:1 as eluent)Conversion gave 580mg of solid. The yield thereof was found to be 76%. LC-MS (sodium-magnesium chloride) with M/z of 324.3[ M + Na ]]+1H NMR(400MHz,CDCl3):δ7.45-7.37(m,2H),7.34-7.28(m,3H),3.81(m,2H),3.36-3.27(m,2H),2.41(s,1H),1.98(m,2H),1.83-1.74(m,2H),1.46(s,9H)。

Step 2: 4- (Phenylethynyl) piperidin-4-ol trifluoroacetate salt

In a 50mL single vial was added tert-butyl 4-hydroxy-4- (phenylethynyl) piperidine-1-carboxylate (92mg,0.31mmol), DCM (4mL) followed by TFA (0.50mL) slowly and stirred at rt for 25 min. The reaction solution was dried and evacuated to give 310mg of pale yellow oil, which was directly used for the next reaction. LC-MS with M/z 202.2[ M + H ]]+

And step 3: 6-ethoxy-4- (6- (4-hydroxy-4- (phenylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

3-cyano-6-ethoxy-4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a ] is added into a 5mL microwave tube in sequence]Pyridine-3-carbonitrile (40mg,0.14mmol), 4- (phenylethynyl) piperidin-4-ol trifluoroacetate (0.34mmol), DMSO (3mL), N, N-diisopropylethylamine (0.15mL,0.91mmol), microwave reacted at 105 ℃ for 4 h. The reaction mixture was added with EA 20mL, washed with water (8 mL. times.3), the aqueous phases combined, EA 15mL extracted, the organic phases combined, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated. The residue was purified by silica gel column chromatography (PE: EA (v: v) ═ 1:1 as eluent) to obtain 26mg of a pale yellow solid, which was the objective product (yield 39%). LC-MS M/z 464.3[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.33(d,J=2.2Hz,1H),8.18(s,1H),8.09(d,J=1.9Hz,1H),7.70(dd,J=8.8,2.5Hz,1H),7.46–7.39(m,2H),7.33–7.27(m,3H),7.08(d,J=2.0Hz,1H),6.81(d,J=8.9Hz,1H),4.13(m,2H),4.07(m,2H),3.62–3.48(m,2H),2.13(m,2H),2.04(s,1H),1.98–1.90(m,2H),1.48(t,J=7.0Hz,3H)。

Example 10: 4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 4- ((trimethylsilyl) ethynyl) piperidine-1-carboxylic acid tert-butyl ester

The flask was connected to a reflux condenser, and after nitrogen substitution, THF (3mL) was added, ethynyl (trimethyl) silane (1.0mL,7.1mmol) was added, nitrogen substitution was again performed, and then EtMgBr (7.5mL,7.5mmol,1mol/L) was added dropwise, reacted at room temperature for 10min, and then heated to 50 ℃ for 2 h. Another two-necked bottle was charged with 4-iodopiperidine-1-carboxylic acid tert-butyl ester (1.5g,4.8mmol), FeBr2(102mg,0.47299mmol), nitrogen was replaced, NMP (14mL) was added and dissolved, the Grignard reagent prepared in the above step was added, the solution was blackened, and the reaction was carried out at room temperature for 16.5 hours. After the reaction was terminated, 20mL of water was added to the reaction mixture, EA (100mL × 2) was extracted, the organic phase was washed with water (15mL × 2), washed with saturated brine (20mL), dried over anhydrous sodium sulfate and purified by silica gel column chromatography, and 480mg of a pale yellow liquid was obtained as a product with a yield of 35.0% in which EA (v: v) was 20: 1. LC-MS M/z 226.20[ M-t-Bu + 2H%]+

Step 2: 4-ethynylpiperidine-1-carboxylic acid tert-butyl ester

In a single-necked flask, tert-butyl 4- ((trimethylsilyl) ethynyl) piperidine-1-carboxylate (480mg,1.70mmol) was added, MeOH (5mL) was added and dissolved, and K was added at room temperature2CO3(240mg,1.74mmol), and reacted at room temperature for 16 h. After the reaction is stopped, the reaction solution is directly concentrated by silica gel column chromatography for purification, and the eluent is PE: EA (v: v) ═ 20:1, so that the product is obtained as colorless liquid 280mg, and the yield is 78.46%. LC-MS M/z 210.10[ M + H ]]+

And step 3: 4-ethynylpiperidine hydrochloride

In a single-neck flask, tert-butyl 4-ethynylpiperidine-1-carboxylate (280mg,1.34mmol) was added, EA (2mL) was added and dissolved, HCl/EA (3.0mL,12mmol,4mol/L) was added with stirring, and the reaction was carried out at room temperature for 2.5 h. After the reaction is finished, the reaction solution is directly concentrated, and the obtained white solid is dried for 2 hours in a vacuum drying oven at the temperature of 60 ℃ to obtain 180mg of solid, namely the product, wherein the yield is 92.38%. LC-MS M/z 110.30[ M-HCl + H ]]+. And 4, step 4: 4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a]Pyridine-3-carbonitriles

In a single-necked flask was added 4-ethynylpiperidine hydrochloride (140mg, 0.9)6mmol), 4- (6-Fluoropyridin-3-yl) -6- (2-hydroxy-2-Methylpropoxy) pyrazolo [1,5-a]Pyridine-3-carbonitrile (200mg,0.62mmol, intermediate 2), dissolved by addition of DMSO (3mL), and K was added2CO3(260mg,1.88mmol) and then reacted at 90 ℃ in an oil bath for 18 h. After the reaction was terminated, water (10mL) was added to the reaction mixture, EA (20mL × 2) was extracted, the organic phase was washed with water (10mL × 5), washed with saturated brine (10mL × 2), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated and purified by silica gel column chromatography, and the eluent was DCM: MeOH (v: v) ═ 30:1, and 250mg of a yellow solid was obtained as a product with a yield of 98.16%. LC-MS M/z 416.70[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.33(d,J=2.3Hz,1H),8.20(s,1H),8.14(d,J=2.0Hz,1H),7.70(dd,J=8.8,2.5Hz,1H),7.14(d,J=2.0Hz,1H),6.78(d,J=8.9Hz,1H),4.03–3.93(m,2H),3.86(s,2H),3.49–3.41(m,2H),2.74–2.67(m,1H),2.13(d,J=2.3Hz,1H),2.09(s,1H),1.99–1.90(m,2H),1.81–1.71(m,2H),1.39(s,6H).

Example 11: 6- (2-hydroxy-2-methylpropoxy) -4- (6- (4- (phenylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Adding Pd (PPh) into a double-mouth bottle3)2Cl2(10mg,0.014mmol), CuI (2.5mg,0.013mmol), 4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a]Pyridine-3-carbonitrile (60mg,0.14mmol, example 10) was replaced with nitrogen, THF (1.5mL) was added, and triethylamine (0.1mL,0.7mmol) and iodobenzene (0.1mL,0.9mmol) were added again to replace nitrogen, and the mixture was reacted at room temperature for 18 h. After the reaction is stopped, the reaction liquid is directly concentrated on a silica gel column for chromatography, the eluent is DCM: MeOH (v: v) ═ 20:1, the obtained compound is collected, DCM is dissolved, and then TLC purification is carried out, the developing agent is DCM: EA (v: v) ═ 40:1, the target product is eluted by the eluent, the eluent is DCM: MeOH (v: v) ═ 20:1, 15.0mg of the obtained solid is the product, and the yield is 21.13%. LC-MS M/z 492.10[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.34(d,J=2.2Hz,1H),8.20(s,1H),8.15(d,J=1.8Hz,1H),7.71(dd,J=8.8,2.4Hz,1H),7.42(dd,J=6.5,2.9Hz,2H),7.31–7.27(m,3H),7.14(d,J=1.8Hz,1H),6.80(d,J=8.9Hz,1H),4.06–3.97(m,2H),3.86(s,2H),3.56–3.47(m,2H),2.96–2.89(m,1H),2.06–1.98(m,2H),1.89–1.81(m,2H),1.39(s,6H).

Example 12: 6- (2-hydroxy-2-methylpropoxy) -4- (6- (4- ((6-methoxypyridin-3-yl) ethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Adding Pd (PPh) into a double-mouth bottle3)2Cl2(10mg,0.014mmol), CuI (2.5mg,0.013mmol), 4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) -6- (2-hydroxy-2-methylpropoxy) pyrazolo [1,5-a]Pyridine-3-carbonitrile (60mg,0.14mmol, example 10) was replaced with nitrogen, DMF (1.5mL) was added, and triethylamine (0.1mL,0.7mmol) and 5-bromo-2-methoxypyridine (0.1mL,0.8mmol) were added again to replace nitrogen, and the reaction was carried out at room temperature for 6 h. After the reaction is stopped, 5mL of water is added into the reaction liquid, EA (20mL × 2) is extracted, an organic phase is washed with water (10mL × 5), the organic phase is washed with saturated saline water, dried by anhydrous sodium sulfate and purified by silica gel column chromatography, an eluent is DCM: MeOH (v: v) ═ 50:1, a collected compound is dissolved in DCM, then TLC purification is carried out, a developing solvent is DCM: MeOH (v: v) ═ 40:1, a target product is eluted by the eluent, DCM: MeOH (v: v) ═ 20:1 is used as the eluent, and 12.0mg of the obtained solid is the product, and the yield is 15.90%. LC-MS M/z 523.10[ M + H ]]+1HNMR(400MHz,CDCl3):δ8.34(d,J=2.1Hz,1H),8.23(d,J=1.5Hz,1H),8.20(s,1H),8.15(d,J=1.7Hz,1H),7.71(dd,J=8.8,2.3Hz,1H),7.58(dd,J=8.6,2.1Hz,1H),7.15(d,J=1.7Hz,1H),6.80(d,J=8.9Hz,1H),6.68(d,J=8.6Hz,1H),4.07–3.99(m,2H),3.93(s,3H),3.86(s,2H),3.53–3.44(m,2H),2.96–2.87(m,1H),2.04–1.97(m,2H),1.86–1.79(m,2H),1.39(s,6H)。

Example 13: 6- (2- (1H-imidazol-1-yl) ethoxy) -4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Step 1: 1- (2-chloroethyl) -1H-imidazole

Imidazole (500mg,7.344mmol) was added to a 25mL single-neck flask, acetonitrile (8mL) was added thereto and dissolved, and potassium carbonate (5.2g,36mmol), 1-bromo-2-chloroethane (1.8mL,22mmol) were added with stirring and reacted at 50 ℃ for 15 hours. After the reaction was stopped, the reaction was directly concentrated and purified by silica gel column chromatography using DCM: MeOH (v: v) ═ 20:1 as eluent to give 340mg of colorless liquid as the product in 52.14% yield. LC-MS M/z 131.30[ M + H ]]+

Step 2: 6- (2- (1H-imidazol-1-yl) ethoxy) -4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Adding 4- (6-fluoro-pyridine-3-yl) -6-hydroxypyrazolo [1,5-a ] into a single-mouth bottle]Pyridine-3-carbonitrile (206mg,0.8104mmol), potassium carbonate (500mg,3.50931mmol), 1- (2-chloroethyl) imidazole (253mg,1.9375mmol), DMF (3mL), oil bath at 90 ℃ for 2.5 h. After the reaction was stopped, 5mL of water was added to the reaction mixture, EA (15mL × 2) was extracted, the organic phase was washed with water 6 times, saturated brine was washed once, and the mixture was concentrated and purified by silica gel column chromatography with DCM: MeOH (v: v) ═ 20:1 to obtain 130mg of a yellow solid, which was the target product, with a yield of 46.06%. LC-MS M/z 349.10[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.38(d,J=2.3Hz,1H),8.24(d,J=2.1Hz,2H),8.05–7.98(m,1H),7.64(s,1H),7.18(d,J=2.0Hz,1H),7.16(dd,J=8.5,2.8Hz,1H),7.08(d,J=3.6Hz,2H),4.46(t,J=4.9Hz,2H),4.33(t,J=4.9Hz,2H)。

And step 3: 6- (2- (1H-imidazol-1-yl) ethoxy) -4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

In a single-necked flask was added 4-ethynylpiperidine hydrochloride (82mg,0.56mmol, example 10, step 3), 6- (2- (1H-imidazol-1-yl) ethoxy) -4- (6-fluoropyridin-3-yl) pyrazolo [1,5-a]Pyridine-3-carbonitrile (130mg,0.37mmol), dissolved by addition of DMSO (2mL), and K was added2CO3(154mg,1.11mmol), then reacted at 90 ℃ in an oil bath for 23 h. After the reaction was stopped, water (10mL) was added to the reaction mixture, EA (20 mL. times.2) was extracted, and the organic phase was washed with waterWashing (10mL × 5), washing with saturated brine (10mL × 2), drying over anhydrous sodium sulfate, filtering, concentrating the filtrate, and purifying by silica gel column chromatography with DCM: MeOH (v: v) ═ 10:1 to obtain 140mg of a yellow solid, which is the product, with a yield of 85.74%. LC-MS M/z 438.70[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.31(d,J=2.4Hz,1H),8.19(s,1H),8.08(d,J=2.0Hz,1H),7.67(dd,J=8.9,2.5Hz,1H),7.61(s,1H),7.10(s,1H),7.08–7.01(m,2H),6.77(d,J=8.9Hz,1H),4.42(t,J=4.9Hz,2H),4.27(t,J=4.9Hz,2H),4.02–3.93(m,2H),3.52–3.38(m,2H),2.76–2.66(m,1H),2.14(d,J=2.3Hz,1H),1.98–1.88(m,2H),1.81–1.70(m,2H).

Example 14: 6- (2- (1H-imidazol-1-yl) ethoxy) -4- (6- (4- (phenylethynyl) piperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a ] pyridine-3-carbonitrile

Adding Pd (PPh) into a double-mouth bottle3)2Cl2(7mg,0.001mmol), CuI (2mg,0.01mmol), 6- (2- (1H-imidazol-1-yl) ethoxy) -4- (6- (4-ethynylpiperidin-1-yl) pyridin-3-yl) pyrazolo [1,5-a]Pyridine-3-carbonitrile (40mg,0.09mmol, example 13) was replaced with nitrogen, THF (1.5mL) was added, and triethylamine (0.1mL,0.7mmol) and iodobenzene (0.1mL,0.9mmol) were added again to replace nitrogen, and the mixture was reacted at room temperature for 18 h. After the reaction is stopped, the reaction liquid is directly concentrated by a silica gel column for chromatography and purification, the eluent is DCM: MeOH (v: v) ═ 15:1, the obtained compound is collected, DCM is dissolved, and then TLC purification is carried out, the developing agent is DCM: MeOH (v: v) ═ 30:1, the target product is eluted by the eluent, the eluent is DCM: MeOH (v: v) ═ 20:1, 14.0mg of the obtained solid is the product, and the yield is 29.81%. LC-MS M/z 514.20[ M + H ]]+1H NMR(400MHz,CDCl3):δ8.32(d,J=2.0Hz,1H),8.20(s,1H),8.09(s,1H),7.75(s,1H),7.68(dd,J=8.8,2.2Hz,1H),7.42(d,J=3.6Hz,2H),7.32–7.27(m,3H),7.10(d,J=22.1Hz,2H),7.05(s,1H),6.79(d,J=8.9Hz,1H),4.44(t,J=4.5Hz,2H),4.28(t,J=4.6Hz,2H),4.08–3.95(m,2H),3.57–3.46(m,2H),2.96–2.89(m,1H),2.05–1.96(m,2H),1.88–1.78(m,2H).

Biological activity test example:

test example 1: testing of Compounds of the invention for Ret wt and Ret V804M kinase inhibitory Activity

1. Purpose of the experiment:

HTRF method was used to test the inhibitory activity of the series of compounds on 6 kinases, Ret wt, VEGFR2, CCDC6-RET, Ret M918T, Ret V804L and Ret V804M, and to determine IC50The value is obtained.

2. The experimental reagents and consumables used were as follows:

1)HTRF KinEASE-TK kit(Cisbio,62TK0PEC)

2)Ret wt(Invitrogen,PV3082)

3)CCDC6-RET(Signalchem,R02-19BG-10)

4)Ret M918T(Signalchem,R02-12JG-10)

5)Ret V804L(Signalchem,R02-12BG-10)

6)Ret V804M(Signalchem,R02-12GG-10)

7)MgCl2(Sigma,M1028)

8)ATP(Promega,V910B)

9)DTT(Invitrogen,P2325)

10)DMSO(Sigma,D8418)

11)384-well plate,white,low volume,round-bottom(Greiner,784075)

12)384-Well Polypropylene microplate,Clear,Flatt Bottom,Bar Code(Labcyte,P-05525-BC)

13)96-well polypropylene plate(Nunc,249944)

14)Plate shaker(Thermo,4625-1CECN/THZ Q)

15)Centrifuge(Eppendorf,5810R)

16)Envision 2104multi-label Reader(PerkinElmer,2104-10-1)

17)Echo(Labcyte,550)

3. experimental procedure

3.1 preparation of 1 × kinase reaction buffer:

1 volume of 5x kinase reaction buffer and 4 volumes of water; 5mM MgCl2;1mM DTT;1mM MnCl2

3.2 transfer of 10nl of the diluted compound per well with an Echo 550 reaction plate (784075, Greiner);

3.3 seal the reaction plate with sealing plate and centrifuge at 1000g for 1 min.

3.4 prepare 2 Xkinase using 1 Xenzyme reaction buffer.

3.5 Add 5. mu.l kinase per well to the reaction plate (prepared in step 3.4). Sealing the plate with sealing plate membrane, centrifuging at 1000g for 30s, standing at room temperature for 10 s

And (3) minutes.

3.6 prepare 4 XTK-substrate-biotin and 4 XTATP with 1 Xenzyme reaction buffer, mix well, add 5 u l K-substrate-biotin/ATP mixture to the reaction plate.

3.7 plates were centrifuged at 1000g for 30 seconds with a sealing plate membrane and reacted at room temperature for 40 minutes.

3.8 prepare 4 × Sa-XL 665(250nM) in HTRF detection buffer.

3.9 mu.l of Sa-XL 665 and 5. mu.l of TK-antisense-Cryptate were added to each well, centrifuged at 1000g for 30 seconds, and reacted at room temperature for 1 hour.

3.10 reading the fluorescence signals at 615nm (Cryptote) and 665nm (XL665) with Envision 2104.

4. Data analysis

4.1 calculate the Ratio per well (Ratio _665/615nm)

4.2 inhibition was calculated as follows:

average of CEP-32496 readings for all positive control wells

Average of DMSO well readings for all negative control wells

Wherein, the chemical name of CEP-32496 is as follows: n- [3- [ (6, 7-dimethoxy-4-quinazolinyl) oxy ] phenyl ] -N' - [5- (2,2, 2-trifluoro-1, 1-dimethylethyl) -3-isoxazolyl ] urea.

4.3 calculating IC50And the inhibition curves of the compounds were plotted:

IC of the compound was obtained using the following non-linear fit equation50(median inhibitory concentration): data analysis was performed using Graphpad 6.0 software.

Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*Hill Slope))

X is the log value of the concentration of the compound Y is the inhibition ratio (% inhibition)

5. The results are shown in Table A-1 to Table A-3:

TABLE A-1 kinase inhibitory Activity of Compounds of the invention

TABLE A-2 kinase inhibitory Activity of Compounds of the invention

Examples IC50(nM),Ret wt IC50(nM),Ret V804M
Example 8 0.395 13.26

TABLE A-3 kinase inhibitory Activity of Compounds of the invention

Examples IC50(nM),Ret wt
Example 1 10.6
Example 2 50.1
Example 3 12.3
Example 4 29.6
Example 6 50.0
Example 7 6.151

As can be seen from tables A-1, A-2 and A-3, the compounds of the examples of the invention have good inhibition effect on Ret wt, and in addition, the compounds of the examples of the invention have good inhibition effect on Ret CCDC-6, Ret M918T, Ret V804L and Ret V804M.

In addition to the compounds of the examples in tables A-1 to A-3, the other compounds of the present invention also have a good inhibitory effect on Ret wt, IC50The value is 0 to 50nM, preferably IC50The value is 0-10 nM; in addition, other compounds of the invention also have good inhibition effects on Ret CCDC6, Ret M918T, Ret V804L and Ret V804M, and IC50A value of 0 to 100nM, preferably IC50The value is 0-50 nM; more preferably, the IC50The value is 0-10 nM.

Test example 2: cell proliferation inhibitory Activity of Compounds of the invention on BAF3-KIF5B-RET-WT cells

1. Purpose of the experiment:

the cell proliferation inhibitory activity of the compounds in tumor cells was tested by the CTG method, and the median inhibitory rate (IC) was calculated50)。

2. The experimental reagents and test articles used were as follows:

1)CTG:CellTiter-Glo(Promega)

2) RPMI-1640 medium (Gibco)

3) FBS fetal bovine serum (Gibco)

4)DMSO(Sigma)

5) Double resistance: penicillin and streptomycin (Hyclone)

6) 96-well cell culture plate, white wall impermeable bottom (Corning)

7) BAF3 cells (purchased from Shanghai Mingjin biol.)

8) BAF3-KIF5B-RET-WT cell (Steady cell line, constructed by Pharmacology division of Guangdong Dongyang pharmaceutical Co., Ltd.)

3. The experimental steps are as follows:

1) cell seeding

Cells in exponential growth phase, BAF3 and BAF3-KIF5B-RET-WT, were collected and viable Cell counts were performed using a Vi-Cell XR cytometer. The cell suspension was adjusted to the corresponding concentration with RPMI-1640 complete medium (89% RPMI-1640+ 10% FBS + 1% double antibody). Add 90. mu.L of cell suspension to each well in 96-well cell culture plates, at cell concentrations of 2000 cells/well and 10000 cells/well for BAF3 and BAF3-KIF5B-RET-WT, respectively.

2) Adding chemicals for treatment

a, preparing a working solution: each test compound was dissolved in DMSO to give a final concentration of 10mM stock solution. Stock solutions were diluted 100-fold in RPMI-1640 complete medium and further diluted 3-fold in gradient 9 times to give 10 working solutions each with a final DMSO concentration of 0.1%.

b, adding medicine into cells: after the cells were incubated overnight, 10. mu.l of the above 10 concentrations of working solution were added in sequence and placed at 37 ℃ in 5% CO2Incubating for 72 hours in an incubator; simultaneously setting up no-added compoundNegative control (3).

3) Read plate detection

After 72 hours of drug treatment, according to the CTG operation instruction, 50 μ l (1/2 culture volume) of CTG solution which is pre-melted and balanced to room temperature is added into each well, mixed evenly for 2 minutes by a microplate shaker, and placed at room temperature for 10 minutes, and then the fluorescence signal value is measured by a multifunctional microplate reader.

4) Data analysis

Cell survival%: Vsample/Vvehicle control x 100%. Where Vsample is the reading for the drug treated group and Vvehicle control is the mean value for the solvent control group. Sigmoidal dose-survival curves were plotted using a non-linear regression model using GraphPad Prism 5.0 software and IC calculated50The values, the experimental results are shown in table B.

TABLE B results of experiments on the cell proliferation inhibitory Activity of the Compounds of the present invention on BAF3-KIF5B-RET-WT

As is clear from Table B, the compounds of the present invention also have a good inhibitory effect on cell proliferation of BAF3-KIF5B-RET-WT cells.

In the description herein, reference to the description of the terms "one embodiment," "some embodiments," "an example," "a specific example" or "some examples" or the like means that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above do not necessarily refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, the various examples, embodiments, or examples described in this specification, as well as features of various examples, embodiments, or examples, may be combined and combined by one skilled in the art without contradiction.

Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that those skilled in the art can make changes, modifications, substitutions and alterations to the above embodiments without departing from the principle and spirit of the present invention, the scope of which is defined by the appended claims and their equivalents.

69页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:RET抑制剂、其药物组合物及其在药物中的应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!