Free base crystal form of heteroaromatic derivative and preparation method thereof

文档序号:1900186 发布日期:2021-11-30 浏览:25次 中文

阅读说明:本技术 杂芳类衍生物的自由碱晶型及其制备方法 (Free base crystal form of heteroaromatic derivative and preparation method thereof ) 是由 李媛媛 呙临松 于 2021-05-25 设计创作,主要内容包括:本发明涉及一种含杂芳类衍生物的自由碱晶型及其制备方法。具体涉及一种具有通式(I)化合物自由碱的晶型、制备方法和含有治疗有效量的该晶型的药物组合物,及其作为JAK激酶抑制剂,在治疗类风湿性关节炎、皮炎、银屑病、炎症性肠病等炎症性疾病及结肠癌、非小细胞肺癌等肿瘤疾病的用途。(The invention relates to a free base crystal form containing heteroaromatic derivatives and a preparation method thereof. In particular to a crystal form of free alkali of a compound with a general formula (I), a preparation method, a pharmaceutical composition containing a therapeutically effective amount of the crystal form, and application of the crystal form as a JAK kinase inhibitor in treating inflammatory diseases such as rheumatoid arthritis, dermatitis, psoriasis and inflammatory bowel diseases, and tumor diseases such as colon cancer and non-small cell lung cancer.)

1. The crystal form of the compound shown in the general formula (I) has the following structure:

wherein:

L1selected from the group consisting of a bond, - (CH)2)r-、-(CH2)rS(O)2-、-S(O)2(CH2)r-、-(CH2)rS(O)2NRa-、-(CH2)rNRa-、-C(O)(CH2)r-、-C(O)(CH2)rNRa-、-C(O)(CH2)rNRa(CH2)s-、-(CH2)rC (O) -or- (CH)2)rC(O)NRa-;

RaSelected from hydrogen, C1-6Alkyl radical, C1-6Hydroxyalkyl or C1-6An alkoxy group;

R1selected from hydrogen, cyano, halogen, C1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl, 3-12 membered heterocyclyl, C6-12Aryl or 5-10 membered heteroaryl, optionally further substituted by cyano, halogen, C1-6Alkyl radical, C1-6Alkoxy or C1-6Substituted by one or more substituents of hydroxyalkyl, preferably hydrogen, cyano, halogen, C1-3Alkyl radical, C3-6Cycloalkyl, 3-8 membered heterocyclyl, phenyl or 5-6 membered nitrogen containing heteroaryl, optionally further substituted by cyano, halogen, C1-3Alkyl radical, C1-3Alkoxy or C1-3Substituted with one or more substituents in hydroxyalkyl;

R2selected from hydrogen or C1-6An alkyl group;

R3selected from hydrogen, hydroxy, halogen, amino, C1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkoxy radical, C1-6Alkyl monosubstituted amino, C1-6Alkyl disubstituted amino, C3-8Cycloalkyl, 3-10 membered heterocyclyl, C6-12Aryl or 5-10 membered heteroaryl, optionally further substituted by halogen, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Hydroxyalkyl radical, C1-6Alkyl monosubstituted amino, C1-6Alkyl disubstituted amino, C3-8Cycloalkyl, 3-10 membered heterocyclyl, C6-12Aryl or 5-to 10-membered heteroaryl, preferably hydrogen, hydroxy, halogen, amino, C1-3Alkyl radical, C3-6Cycloalkyl, 4-7 membered heterocyclyl containing 1-2 heteroatoms selected from N, O or S, phenyl or 5-6 membered nitrogen containing heteroaryl, optionally further substituted by halogen, amino, cyano, C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Hydroxyalkyl radical, C1-3Alkyl monosubstituted amino, C1-3Alkyl disubstituted amino, C3-6Cycloalkyl, 4-7 membered heterocyclyl, phenyl or 5-6 membered nitrogen containing heteroaryl;

R4selected from hydrogen, C1-6Alkyl radical, C1-6Hydroxyalkyl or C1-6An alkoxy group;

ring A is selected from aryl, five-membered sulfur-containing heterocyclic group, preferably phenyl,

Ring B is selected from 5-10 membered nitrogen-containing heterocyclic group, preferably

m is 1,2 or 3;

r is 0, 1,2 or 3;

s is 1,2 or 3.

2. A crystalline form according to claim 1, characterized in that the general formula (I) is further represented by general formula (II):

wherein n is 1 or 2.

3. A crystalline form according to claim 1 or 2, characterized in that R is1Selected from hydrogen, cyano, halogen, C1-3Alkyl, phenyl, Optionally further substituted by cyano, halogen, C1-3Alkyl radical, C1-3Substituted with one or more substituents in the alkoxy group.

4. A crystalline form according to claim 1, characterized in that R is3Selected from the group consisting of hydrogen, hydroxy, fluoro, chloro, bromo, amino, methylamino, ethylamino, dimethylamino, methyl, ethyl, hydroxymethyl, methoxy, ethoxy, phenyl, methyl, ethyl, propyl, butyl, or butyl, or a,

Optionally further substituted by halogen, amino, C1-3Alkyl monosubstituted amino, C1-3Alkyl disubstituted amino, C1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Alkoxy, phenyl, or a salt thereof, Is substituted with one or more substituents.

5. A crystalline form according to claim 1, characterized in that the general formula (I) is selected from the following compounds:

crystalline forms of 1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (methylamino) -ethan-1-one, including crystalline forms a through Q, wherein:

the X-ray powder diffraction pattern of the crystal form A has a characteristic peak at a 2 theta of 19.7 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 2 theta of 7.0 +/-0.2 degrees and 16.1 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 13.9 ± 0.2 °, 16.6 ± 0.2 °, 20.9 ± 0.2 ° and 22.8 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 10.5 +/-0.2 degrees, 15.1 +/-0.2 degrees, 19.3 +/-0.2 degrees, 19.7 +/-0.2 degrees, 20.4 +/-0.2 degrees, 24.0 +/-0.2 degrees, 24.5 +/-0.2 degrees and 29.2 +/-0.2 degrees; most preferably, the X-ray powder diffraction pattern is substantially as shown in FIG. 1;

the X-ray powder diffraction pattern of the crystal form B has a characteristic peak at a 2 theta of 17.4 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 20.9 +/-0.2 degrees and 26.5 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 14.4 ± 0.2 °, 16.7 ± 0.2 °, 19.2 ± 0.2 ° and 20.3 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 15.2 +/-0.2 degrees, 15.9 +/-0.2 degrees, 19.6 +/-0.2 degrees, 21.6 +/-0.2 degrees, 23.0 +/-0.2 degrees, 23.6 +/-0.2 degrees, 26.1 +/-0.2 degrees and 28.3 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 6.8 + -0.2 deg., 10.8 + -0.2 deg. and 29.5 + -0.2 deg. 2 theta, most preferably, the X-ray powder diffraction pattern thereof is substantially as shown in FIG. 2;

the X-ray powder diffraction pattern of the crystal form C has a characteristic peak at a 2 theta of 20.2 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 14.3 +/-0.2 degrees and 15.5 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 19.2 ± 0.2 °, 21.5 ± 0.2 °, 25.3 ± 0.2 ° and 27.5 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 16.9 +/-0.2 degrees, 18.3 +/-0.2 degrees, 23.0 +/-0.2 degrees and 24.5 +/-0.2 degrees of 2 theta, and most preferably, the X-ray powder diffraction pattern is basically as shown in figure 3;

the X-ray powder diffraction pattern of the crystal form D has a characteristic peak at a 2 theta of 23.3 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 16.1 +/-0.2 degrees and 17.6 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 7.4 ± 0.2 °, 20.4 ± 0.2 °, 22.0 ± 0.2 ° and 22.4 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 11.6 +/-0.2 degrees, 11.8 +/-0.2 degrees, 14.4 +/-0.2 degrees, 16.6 +/-0.2 degrees, 18.8 +/-0.2 degrees, 19.6 +/-0.2 degrees, 23.8 +/-0.2 degrees and 28.9 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 2 θ of 8.7 ± 0.2 °, 9.2 ± 0.2 °, 10.9 ± 0.2 °, 13.6 ± 0.2 °, 15.0 ± 0.2 °, 15.3 ± 0.2 °, 18.0 ± 0.2 °, 18.4 ± 0.2 °, 20.0 ± 0.2 °, 20.8 ± 0.2 °, 24.9 ± 0.2 °, 27.6 ± 0.2 ° and 28.1 ± 0.2 °, most preferably, its X-ray powder diffraction pattern is substantially as shown in fig. 6;

the X-ray powder diffraction pattern of the crystal form E has a characteristic peak at a 2 theta of 7.3 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 20.5 +/-0.2 degrees and 21.7 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 2 θ of 8.5 ± 0.2 °, 17.2 ± 0.2 °, 22.1 ± 0.2 ° and 22.8 ± 0.2 °; further preferably, the compound also comprises characteristic peaks at 2 theta of 10.8 +/-0.2 degrees, 11.6 +/-0.2 degrees, 14.4 +/-0.2 degrees, 16.2 +/-0.2 degrees, 16.5 +/-0.2 degrees, 17.7 +/-0.2 degrees, 23.8 +/-0.2 degrees and 28.3 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 27.7 ± 0.2 ° and 29.4 ± 0.2 ° 2 θ, most preferably, the X-ray powder diffraction pattern thereof is substantially as shown in fig. 9;

the X-ray powder diffraction pattern of the crystal form F has a characteristic peak at a 2 theta of 22.9 +/-0.2 degrees; preferably, the composition also comprises characteristic peaks at 2 theta of 12.3 +/-0.2 degrees and 20.2 +/-0.2 degrees; more preferably, the compound also comprises characteristic peaks at 2 theta of 12.6 +/-0.2 degrees, 14.6 +/-0.2 degrees, 18.4 +/-0.2 degrees and 24.6 +/-0.2 degrees; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.1 +/-0.2 degrees, 16.1 +/-0.2 degrees, 16.6 +/-0.2 degrees, 17.8 +/-0.2 degrees, 22.0 +/-0.2 degrees, 23.1 +/-0.2 degrees, 29.5 +/-0.2 degrees and 30.2 +/-0.2 degrees, and most preferably, the X-ray powder diffraction pattern is basically as shown in figure 12;

the X-ray powder diffraction pattern of the crystal form G has a characteristic peak at a 2 theta of 19.6 +/-0.2 degrees; preferably, the composition also comprises characteristic peaks at 15.4 +/-0.2 degrees and 19.0 +/-0.2 degrees of 2 theta; more preferably, further comprising having characteristic peaks at 6.9 ± 0.2 °, 7.7 ± 0.2 °, 11.0 ± 0.2 ° and 22.7 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 13.6 +/-0.2 degrees, 13.8 +/-0.2 degrees, 16.0 +/-0.2 degrees, 17.6 +/-0.2 degrees, 18.1 +/-0.2 degrees, 20.7 +/-0.2 degrees, 21.3 +/-0.2 degrees and 27.3 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 2 θ of 8.7 ± 0.2 °, 10.4 ± 0.2 °, 13.1 ± 0.2 °, 14.9 ± 0.2 °, 20.3 ± 0.2 °, 22.4 ± 0.2 °, 23.5 ± 0.2 °, 25.5 ± 0.2 ° and 27.8 ± 0.2 °, most preferably, the X-ray powder diffraction pattern thereof is substantially as shown in FIG. 15;

the X-ray powder diffraction pattern of the crystal form H has a characteristic peak at a 2 theta of 7.2 +/-0.2 degrees; preferably, the composition also comprises characteristic peaks at 16.0 +/-0.2 degrees and 20.8 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 16.4 ± 0.2 °, 17.6 ± 0.2 °, 19.7 ± 0.2 ° and 21.9 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.3 +/-0.2 degrees, 11.7 +/-0.2 degrees, 14.6 +/-0.2 degrees, 15.2 +/-0.2 degrees, 19.1 +/-0.2 degrees, 20.3 +/-0.2 degrees, 23.6 +/-0.2 degrees and 27.3 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 9.2 + -0.2 deg., 13.3 + -0.2 deg., 18.4 + -0.2 deg. and 22.7 + -0.2 deg. 2 theta, most preferably, its X-ray powder diffraction pattern is substantially as shown in FIG. 16;

the X-ray powder diffraction pattern of the crystal form I has a characteristic peak at a 2 theta of 6.8 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 2 theta of 13.7 +/-0.2 degrees and 18.6 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 15.1 ± 0.2 °, 15.5 ± 0.2 °, 20.5 ± 0.2 ° and 20.7 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.2 +/-0.2 degrees, 11.8 +/-0.2 degrees, 17.7 +/-0.2 degrees, 20.2 +/-0.2 degrees, 22.4 +/-0.2 degrees, 23.1 +/-0.2 degrees, 23.5 +/-0.2 degrees and 27.0 +/-0.2 degrees, and most preferably, the X-ray powder diffraction pattern is basically as shown in figure 19;

the X-ray powder diffraction pattern of the crystal form J has a characteristic peak at a 2 theta of 20.5 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at the 2 theta of 18.7 +/-0.2 degrees and 20.2 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 6.8 ± 0.2 °, 12.2 ± 0.2 °, 12.6 ± 0.2 ° and 22.8 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 14.5 +/-0.2 degrees, 15.5 +/-0.2 degrees, 16.0 +/-0.2 degrees, 16.5 +/-0.2 degrees, 18.4 +/-0.2 degrees, 21.9 +/-0.2 degrees, 23.1 +/-0.2 degrees and 24.6 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 13.7 + -0.2 deg., 15.1 + -0.2 deg., 22.5 + -0.2 deg. and 30.1 + -0.2 deg. 2 theta in 2 theta, most preferably, it has an X-ray powder diffraction pattern substantially as shown in FIG. 22;

the X-ray powder diffraction pattern of the crystal form K has a characteristic peak at a 2 theta of 19.5 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 2 theta of 6.7 +/-0.2 degrees and 15.9 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 10.3 ± 0.2 °, 13.7 ± 0.2 °, 16.3 ± 0.2 ° and 22.5 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 10.0 +/-0.2 degrees, 12.2 +/-0.2 degrees, 14.9 +/-0.2 degrees, 19.1 +/-0.2 degrees, 20.2 +/-0.2 degrees, 20.7 +/-0.2 degrees, 23.7 +/-0.2 degrees, 26.2 +/-0.2 degrees and 29.0 +/-0.2 degrees of 2 theta, and most preferably, the X-ray powder diffraction pattern is basically as shown in figure 25;

the X-ray powder diffraction pattern of the crystal form L has a characteristic peak at a 2 theta of 7.2 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 21.3 +/-0.2 degrees and 22.5 +/-0.2 degrees of 2 theta; more preferably, it further comprises characteristic peaks at 16.0 + -0.2 °, 21.9 + -0.2 °, 22.2 + -0.2 ° and 27.3 + -0.2 ° 2 θ, further preferably, it further comprises characteristic peaks at 8.4 + -0.2 °, 16.9 + -0.2 °, 17.5 + -0.2 °, 19.5 + -0.2 °, 20.3 + -0.2 °, 23.6 + -0.2 °, 24.1 + -0.2 ° and 27.9 + -0.2 ° 2 θ, further preferably, it further comprises characteristic peaks at 11.6 + -0.2 °, 14.5 + -0.2 °, 15.6 + -0.2 °, 16.4 + -0.2 °, 20.1 + -0.2 ° and 30.4 + -0.2 ° 2 θ, most preferably, its X-ray powder diffraction pattern is substantially as shown in fig. 26;

the X-ray powder diffraction pattern of the crystal form M has a characteristic peak at a 2 theta of 18.6 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 15.7 +/-0.2 degrees and 25.1 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 21.7 ± 0.2 °, 23.4 ± 0.2 °, 24.0 ± 0.2 ° and 28.1 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 10.5 +/-0.2 degrees, 12.9 +/-0.2 degrees, 14.2 +/-0.2 degrees, 16.0 +/-0.2 degrees, 16.8 +/-0.2 degrees, 17.4 +/-0.2 degrees, 19.6 +/-0.2 degrees and 22.3 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 9.3 + -0.2 deg., 21.2 + -0.2 deg., 22.8 + -0.2 deg., 29.4 + -0.2 deg., and 32.2 + -0.2 deg. in 2 theta, most preferably, its X-ray powder diffraction pattern is substantially as shown in FIG. 29;

the X-ray powder diffraction pattern of the crystal form N has a characteristic peak at a 2 theta of 19.1 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 15.4 +/-0.2 degrees and 19.7 +/-0.2 degrees of 2 theta; more preferably, further comprising having characteristic peaks at 2 θ of 7.6 ± 0.2 °, 10.9 ± 0.2 °, 21.0 ± 0.2 ° and 21.3 ± 0.2 °; further preferably, the compound also comprises characteristic peaks at 13.5 +/-0.2 degrees, 18.0 +/-0.2 degrees, 18.8 +/-0.2 degrees, 22.6 +/-0.2 degrees, 25.4 +/-0.2 degrees, 26.2 +/-0.2 degrees and 27.3 +/-0.2 degrees of 2 theta, and most preferably, the X-ray powder diffraction pattern is basically as shown in figure 32;

the X-ray powder diffraction pattern of the crystal form O has a characteristic peak at a 2 theta of 23.8 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 2 theta of 12.7 +/-0.2 degrees and 17.9 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 14.4 ± 0.2 °, 17.6 ± 0.2 °, 21.0 ± 0.2 ° and 24.7 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.4 +/-0.2 degrees, 13.0 +/-0.2 degrees, 19.6 +/-0.2 degrees, 20.7 +/-0.2 degrees, 22.7 +/-0.2 degrees, 26.6 +/-0.2 degrees, 28.7 +/-0.2 degrees and 30.9 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 6.7 + -0.2 deg., 12.0 + -0.2 deg., 14.8 + -0.2 deg., 15.8 + -0.2 deg., 16.5 + -0.2 deg., 19.0 + -0.2 deg., and 23.0 + -0.2 deg. in 2 theta, most preferably, its X-ray powder diffraction pattern is substantially as shown in FIG. 33;

the X-ray powder diffraction pattern of the crystal form P has a characteristic peak at the 2 theta of 23.2 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 14.9 +/-0.2 degrees and 16.5 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 8.3 ± 0.2 °, 13.4 ± 0.2 °, 19.2 ± 0.2 ° and 20.2 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 7.8 +/-0.2 degrees, 9.3 +/-0.2 degrees, 11.6 +/-0.2 degrees, 12.6 +/-0.2 degrees, 16.0 +/-0.2 degrees, 18.5 +/-0.2 degrees, 19.6 +/-0.2 degrees and 27.4 +/-0.2 degrees; still more preferably, it further comprises characteristic peaks at 15.3 ± 0.2 ° and 22.1 ± 0.2 ° 2 θ, most preferably, the X-ray powder diffraction pattern thereof is substantially as shown in fig. 36;

the X-ray powder diffraction pattern of the crystal form Q has a characteristic peak at a 2 theta of 16.4 +/-0.2 degrees; preferably, the compound also comprises characteristic peaks at 2 theta of 13.5 +/-0.2 degrees and 20.0 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 9.1 ± 0.2 °, 14.7 ± 0.2 °, 15.9 ± 0.2 ° and 27.9 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 7.7 +/-0.2 degrees, 11.7 +/-0.2 degrees, 11.9 +/-0.2 degrees, 18.4 +/-0.2 degrees, 19.5 +/-0.2 degrees, 22.3 +/-0.2 degrees, 26.4 +/-0.2 degrees and 27.3 +/-0.2 degrees, and most preferably, the X-ray powder diffraction pattern is basically as shown in figure 39.

Form A of 3- ((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile having an X-ray powder diffraction pattern with a diffraction peak at 8.2 ± 0.2 ° 2 θ (± 0.2 °); preferably, further comprising diffraction peaks at 9.7 ± 0.2 ° and 20.4 ± 0.2 ° in 2 θ (± 0.2 °); more preferably, further comprising diffraction peaks at 13.5 ± 0.2 °, 17.4 ± 0.2 °, 21.1 ± 0.2 ° and 26.6 ± 0.2 ° in 2 θ (± 0.2 °); further preferably, it further comprises diffraction peaks at 16.5 ± 0.2 °, 16.8 ± 0.2 °, 14.1 ± 0.2 °, 19.1 ± 0.2 °, 19.4 ± 0.2 °, 22.8 ± 0.2 °, 27.1 ± 0.2 ° and 28.5 ± 0.2 ° in 2 θ (± 0.2 °); still further preferably, further comprising diffraction peaks at 9.4 ± 0.2 °, 19.8 ± 0.2 ° and 23.6 ± 0.2 ° in 2 θ (± 0.2 °);

further preferably, the X-ray powder diffraction pattern is substantially as shown in figure 42, the DSC pattern is substantially as shown in figure 43, and the TGA pattern is substantially as shown in figure 44.

8. A crystalline form according to any one of claims 1 to 7, which is solvent-containing or solvent-free, wherein the solvent is selected from water, methanol, acetone, ethyl acetate, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, tert-butanol, 2-butanone, 3-pentanone, N-heptane, ethyl formate, isopropyl acetate, cyclohexane, methyl tert-butyl ether or isopropyl ether.

9. A crystalline form according to claim 8, characterized in that the number of solvents is from 0.2 to 3, preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3, more preferably 0.5, 1,2 or 3.

10. A process for preparing a crystalline form of the compound of any one of claims 1-9, comprising the steps of:

1) weighing a proper amount of compound, and suspending with a poor solvent, wherein the suspension density is preferably 50-200 mg/mL;

2) shaking the suspension obtained above at a certain temperature for a certain time, wherein the temperature is preferably 0-50 ℃, and the time is preferably 1-10 days;

3) quickly centrifuging the suspension, removing supernatant, and drying to constant weight to obtain a target product;

wherein:

the poor solvent is selected from one or more of acetone, ethyl formate, ethyl acetate, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, chlorobenzene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, ethyl formate, isopropyl acetate, tert-butanol, 2-butanone or 3-pentanone; preferably one or more of ethyl formate, ethyl acetate, tetrahydrofuran, acetone, acetonitrile, methanol, ethanol, dichloromethane, isopropyl acetate or isopropanol;

alternatively, the first and second electrodes may be,

1) weighing a proper amount of compound, and dissolving the compound by using a good solvent;

2) adding an anti-solvent into the obtained solution at a certain temperature, and stirring until a solid is separated out, wherein the preferable temperature is 0-50 ℃;

3) quickly centrifuging the suspension, removing supernatant, and drying the residual solid to constant weight to obtain a target product;

wherein:

the benign solvent is selected from one or more of methanol, acetone, ethyl acetate, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, tert-butanol, 2-butanone or 3-pentanone; preferably one or two of methanol, dimethyl sulfoxide or isobutanol;

the poor solvent is selected from one or more of dichloromethane, toluene, ethyl formate, ethyl acetate, 2-methyl-tetrahydrofuran, ethanol, n-heptane, 1, 4-dioxane, water, isopropyl acetate, cyclohexane, methyl tert-butyl ether, toluene or isopropyl ether; one or more of water, n-heptane, cyclohexane or methyl tert-butyl ether are preferred.

11. A pharmaceutical composition comprising a therapeutically effective amount of a crystalline form of a compound of any one of claims 1-9, and one or more pharmaceutically acceptable carriers, diluents, or excipients.

12. Use of the crystalline form according to any one of claims 1-9 or the pharmaceutical composition according to claim 11 for the preparation of a medicament for the prevention and/or treatment of a JAK kinase-associated disease, preferably an inflammatory and/or neoplastic disease;

preferably, the inflammatory disease is selected from rheumatoid arthritis, dermatitis, psoriasis, inflammatory bowel disease; wherein the inflammatory bowel disease is preferably chronic intestinal inflammatory disease, further preferably ulcerative colitis and Crohn's disease;

the tumor disease is selected from myelofibrosis, polycythemia vera, essential thrombocythemia, myeloid leukemia, acute lymphocytic leukemia, ductal carcinoma of breast and non-small cell lung cancer.

Technical Field

The invention belongs to the field of drug synthesis, and particularly relates to a crystal form of a heteroaromatic derivative, and a preparation method and application thereof.

Background

Janus kinase (JAK) is an intracellular non-receptor tyrosine kinase that mediates the signaling and activation of various cytokines. The JAK kinase family contains four subfamily members of JAK1, JAK2, JAK3 and TYK2, each of which mediates different types of cytokine signaling pathways, JAK1, JAK2 and TYK2 are expressed in each tissue cell of human body, and JAK3 is mainly expressed in each hematopoietic tissue cell. A common feature of cytokine receptors is that the receptor itself has no kinase activity, but the intracellular segment of the receptor has a binding site for the tyrosine kinase JAK. After the cell factor receptor is combined with a ligand thereof, JAKs coupled with the receptor are activated, so that the receptor is phosphorylated, a phosphorylated tyrosine site can be combined with STAT protein containing an SH2 structural domain, STAT is recruited to the receptor and is phosphorylated through JAKs, then phosphotyrosine mediates STAT dimerization, and the activated STAT dimer is transferred to a cell nucleus and activates target gene transcription of the cell nucleus, so that multiple functions of growth, activation, differentiation and the like of multiple cells are regulated.

The JAK/STAT signal pathway mediates the signal transduction of most of intracellular cytokines and plays a key role in the biological processes of immune regulation, immune cell proliferation and the like. The JAK/STAT signal channel has wide functions, participates in a plurality of important biological processes such as proliferation, differentiation, apoptosis, immunoregulation and the like of cells, and is closely related to a plurality of inflammatory diseases such as rheumatoid arthritis, dermatitis, psoriasis, inflammatory bowel diseases (ulcerative colitis and Crohn's disease) and the like; meanwhile, the JAK/STAT signal pathway is closely related to neoplastic diseases such as myelofibrosis, polycythemia vera and essential thrombocythemia, and the mutation of JAK molecules can also cause Acute Myelocytic Leukemia (AML), Acute Lymphocytic Leukemia (ALL), ductal breast cancer, non-small cell lung cancer (NSCLC) and other neoplastic diseases.

Inflammatory bowel disease is a chronic inflammatory disease of the intestinal tract, including Ulcerative Colitis (UC) and Crohn's Disease (CD). The existing medicines for treating inflammatory bowel diseases mainly comprise aminosalicylic acid preparations, glucocorticoids, immunosuppressive agents, antibiotics and the like. The treatment of UC mainly comprises the regulation of immune response and the inhibition of inflammation. Currently, sulfasalazine is mainly used for treating mild to moderate UC in clinic. While the drugs commonly used for the treatment of moderate to severe UC include glucocorticoids, they are not considered as long-term treatments because of the greater risk than benefit. The monoclonal antibody has the problems of high cost, influence on the safety and effectiveness of the drug due to the generation of the drug antibody, inconvenience in intravenous administration mode and the like, and still has a far-unmet medical requirement in the field. Many patients receiving treatment have not been alleviated, and up to 80% of patients with crohn's disease and 30% of patients with UC eventually require surgical treatment.

Tofacitinib (Xeljanz) is the first oral JAK inhibitor for treating moderate-to-severe active adult patients with UC, has obvious inhibitory activity on JAK1, subtypes 2 and 3, increases the curative effect of the tofacitinib, and brings more serious side effects. Adverse reactions include infection, tuberculosis, tumors, anemia, liver damage, increased cholesterol, and the like. The specification for Tofacitinib is indicated by a number of black boxes: severe infections (tuberculosis, bacteria, fungi, viruses) and malignancies (lymphomas, etc.). Due to the wide range of functions mediated by each JAK, these side effects are caused by the simultaneous inhibition of multiple JAKs by the drug. Since JAKs are widely involved in the regulation of immune cells, JAK inhibitors inevitably cause side effects associated with immunosuppression, such as severe infection, even tumorigenesis, and the like. Even with the numerous highly selective inhibitors currently under investigation, such side effects caused by the inhibitory target are unavoidable.

In view of the good curative effect and the relevant serious side effect of multiple targets of the JAK inhibitor, the problem to be solved urgently at present is to develop a JAK inhibitor medicine with higher safety. Since inflammatory bowel disease occurs on the luminal surface of the gastrointestinal tract and acts without the need for drugs to enter the blood system, the development of a drug that reduces systemic exposure of the drug in the blood circulation and increases the local exposure of the drug at the site of inflammation is a good strategy to increase safety. International application WO2016191524a1 reports that Theravance corporation synthesizes a series of compounds that have very low systemic exposure and form enrichment at intestinal inflammatory sites, and can effectively treat intestinal inflammation without causing serious side effects, indicating that the strategy has great feasibility and may generate great clinical application value.

The invention discloses a series of structures of heteroaromatic derivative inhibitors in PCT patent (application number: PCT/CN2019/121944), and comprehensively researches free base crystal forms of the compounds in subsequent development for the purposes of easy product treatment, filtration and drying, and searching for suitable crystals which are convenient to store, stable for a long time and high in bioavailability.

Disclosure of Invention

All that is referred to in patent PCT/CN2019/121944 is added to the present invention by way of reference.

The invention aims to provide a crystal form of a compound shown in a general formula (I), which has a structure shown in the formula (I):

wherein:

L1selected from the group consisting of a bond, - (CH)2)r-、-(CH2)rS(O)2-、-S(O)2(CH2)r-、-(CH2)rS(O)2NRa-、- (CH2)rNRa-、-C(O)(CH2)r-、-C(O)(CH2)rNRa-、-C(O)(CH2)rNRa(CH2)s-、-(CH2)rC (O) -or- (CH)2)rC(O)NRa-;

RaSelected from hydrogen, C1-6Alkyl radical, C1-6Hydroxyalkyl or C1-6An alkoxy group;

R1selected from hydrogen, cyano, halogen, C1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl, 3-12 membered heterocyclyl, C6-12Aryl or 5-10 membered heteroaryl, optionally further substituted by cyano, halogen, C1-6Alkyl radical, C1-6Alkoxy or C1-6Substituted by one or more substituents of hydroxyalkyl, preferably hydrogen, cyano, halogen, C1-3Alkyl radical, C3-6Cycloalkyl, 3-8 membered heterocyclyl, phenyl or 5-6 membered nitrogen containing heteroaryl, optionally further substituted by cyano, halogen, C1-3Alkyl radical, C1-3Alkoxy or C1-3Substituted with one or more substituents in hydroxyalkyl;

R2selected from hydrogen or C1-6An alkyl group;

R3selected from hydrogen, hydroxy, halogen, amino, C1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Alkoxy radical, C1-6Alkyl monosubstituted amino, C1-6Alkyl disubstituted amino, C3-8Cycloalkyl, 3-10 membered heterocyclyl, C6-12Aryl or 5-10 membered heteroaryl, optionally further substituted by halogen, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Hydroxyalkyl radical, C1-6Alkyl monosubstituted amino, C1-6Alkyl disubstituted amino, C3-8Cycloalkyl, 3-10 membered heterocyclyl, C6-12Aryl or 5-to 10-membered heteroaryl, preferably hydrogen, hydroxy, halogen, amino, C1-3Alkyl radical, C3-6Cycloalkyl, 4-7 membered heterocyclyl containing 1-2 heteroatoms selected from N, O or S, phenyl or 5-6 membered nitrogen containing heteroaryl, optionally further substituted by halogen, amino, cyano, C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Hydroxyalkyl radical, C1-3Alkyl monosubstituted amino, C1-3Alkyl disubstituted amino, C3-6Cycloalkyl, 4-7 membered heterocyclyl, phenyl or 5-6 membered nitrogen containing heteroaryl;

R4selected from hydrogen, C1-6Alkyl radical, C1-6Hydroxyalkyl or C1-6An alkoxy group;

ring A is selected from aryl, five-membered sulfur-containing heterocyclic group, preferably phenyl,

Ring B is selected from 5-10 membered nitrogen-containing heterocyclic group, preferably

m is 1,2 or 3;

r is 0, 1,2 or 3;

s is 1,2 or 3.

In a preferred embodiment of the present invention, there is provided a crystalline form of a compound of formula (II),

wherein n is 1 or 2.

In a preferred embodiment of the present invention, R is1Selected from hydrogen, cyano, halogen, C1-3Alkyl, phenyl,

Optionally further substituted by cyano, halogen, C1-3Alkyl radical, C1-3Substituted with one or more substituents in the alkoxy group.

In a preferred embodiment of the present invention, R is3Selected from the group consisting of hydrogen, hydroxy, fluoro, chloro, bromo, amino, methylamino, ethylamino, dimethylamino, methyl, ethyl, hydroxymethyl, methoxy, ethoxy, phenyl, methyl, ethyl, propyl, butyl, or butyl, or a, Optionally further substituted by halogen, amino, C1-3Alkyl monosubstituted amino, C1-3Alkyl disubstituted amino, C1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Alkoxy, phenyl, or a salt thereof, Is substituted with one or more substituents.

A preferred embodiment of the invention is that the general formula (I) is selected from the following compounds:

in a preferred embodiment of the present invention there is provided a crystalline form of 1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (methylamino) -ethan-1-one (compound III) having the structure:

in a further preferred embodiment of the present invention, said form a of compound III has an X-ray powder diffraction pattern with a characteristic peak at 19.7 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 2 theta of 7.0 +/-0.2 degrees and 16.1 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 13.9 ± 0.2 °, 16.6 ± 0.2 °, 20.9 ± 0.2 ° and 22.8 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 10.5 +/-0.2 degrees, 15.1 +/-0.2 degrees, 19.3 +/-0.2 degrees, 19.7 +/-0.2 degrees, 20.4 +/-0.2 degrees, 24.0 +/-0.2 degrees, 24.5 +/-0.2 degrees and 29.2 +/-0.2 degrees;

characteristic diffraction peaks of X-rays expressed in terms of 2 theta angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 1.

TABLE 1

The X-ray powder diffraction pattern of the crystal form A of the compound III is basically shown as figure 1.

In a further preferred embodiment of the present invention, said form B of compound III has an X-ray powder diffraction pattern having a characteristic peak at 2 θ of 17.4 ± 0.2 °; preferably, the compound also comprises characteristic peaks at 20.9 +/-0.2 degrees and 26.5 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 14.4 ± 0.2 °, 16.7 ± 0.2 °, 19.2 ± 0.2 ° and 20.3 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 15.2 +/-0.2 degrees, 15.9 +/-0.2 degrees, 19.6 +/-0.2 degrees, 21.6 +/-0.2 degrees, 23.0 +/-0.2 degrees, 23.6 +/-0.2 degrees, 26.1 +/-0.2 degrees and 28.3 +/-0.2 degrees; still more preferably, the composition further comprises characteristic peaks at 6.8 + -0.2 deg., 10.8 + -0.2 deg. and 29.5 + -0.2 deg. of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2 theta angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 2.

TABLE 2

The X-ray powder diffraction pattern of the crystal form B of the compound III is basically shown as figure 2.

In a further preferred embodiment of the present invention, said compound III has a form C, X-ray powder diffraction pattern having a characteristic peak at 20.2 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 14.3 +/-0.2 degrees and 15.5 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 19.2 ± 0.2 °, 21.5 ± 0.2 °, 25.3 ± 0.2 ° and 27.5 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 16.9 +/-0.2 degrees, 18.3 +/-0.2 degrees, 23.0 +/-0.2 degrees and 24.5 +/-0.2 degrees of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 3.

TABLE 3

The X-ray powder diffraction pattern of the crystal form C of the compound III is basically shown as figure 3; the DSC pattern is basically shown in figure 4; the TGA profile is substantially as shown in figure 5.

In a further preferred embodiment of the present invention, said compound III in crystalline form D, has an X-ray powder diffraction pattern with a characteristic peak at 23.3 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 16.1 +/-0.2 degrees and 17.6 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 7.4 ± 0.2 °, 20.4 ± 0.2 °, 22.0 ± 0.2 ° and 22.4 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 11.6 +/-0.2 degrees, 11.8 +/-0.2 degrees, 14.4 +/-0.2 degrees, 16.6 +/-0.2 degrees, 18.8 +/-0.2 degrees, 19.6 +/-0.2 degrees, 23.8 +/-0.2 degrees and 28.9 +/-0.2 degrees; still further preferably, the composition further comprises characteristic peaks at 8.7 + -0.2 °, 9.2 + -0.2 °, 10.9 + -0.2 °, 13.6 + -0.2 °, 15.0 + -0.2 °, 15.3 + -0.2 °, 18.0 + -0.2 °, 18.4 + -0.2 °, 20.0 + -0.2 °, 20.8 + -0.2 °, 24.9 + -0.2 °, 27.6 + -0.2 ° and 28.1 + -0.2 ° of 2 θ.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 4.

TABLE 4

The X-ray powder diffraction pattern of the crystal form D of the compound III is basically shown as figure 6; the DSC pattern is basically shown in figure 7; the TGA profile is substantially as shown in figure 8.

In a further preferred embodiment of the present invention, said compound III in crystalline form E, has an X-ray powder diffraction pattern with a characteristic peak at 7.3 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 20.5 +/-0.2 degrees and 21.7 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 2 θ of 8.5 ± 0.2 °, 17.2 ± 0.2 °, 22.1 ± 0.2 ° and 22.8 ± 0.2 °; further preferably, the compound also comprises characteristic peaks at 2 theta of 10.8 +/-0.2 degrees, 11.6 +/-0.2 degrees, 14.4 +/-0.2 degrees, 16.2 +/-0.2 degrees, 16.5 +/-0.2 degrees, 17.7 +/-0.2 degrees, 23.8 +/-0.2 degrees and 28.3 +/-0.2 degrees; still more preferably, the composition further comprises characteristic peaks at 27.7 + -0.2 DEG and 29.4 + -0.2 DEG in terms of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 5.

TABLE 5

The compound III of the invention has a crystal form E, and the X-ray powder diffraction pattern of the compound III is basically as shown in figure 9; the DSC pattern is basically shown in figure 10; the TGA profile is substantially as shown in figure 11.

In a further preferred embodiment of the present invention, said compound III in crystalline form F, has an X-ray powder diffraction pattern with a characteristic peak at 22.9 ± 0.2 ° 2 Θ; preferably, the composition also comprises characteristic peaks at 2 theta of 12.3 +/-0.2 degrees and 20.2 +/-0.2 degrees; more preferably, the compound also comprises characteristic peaks at 2 theta of 12.6 +/-0.2 degrees, 14.6 +/-0.2 degrees, 18.4 +/-0.2 degrees and 24.6 +/-0.2 degrees; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.1 +/-0.2 degrees, 16.1 +/-0.2 degrees, 16.6 +/-0.2 degrees, 17.8 +/-0.2 degrees, 22.0 +/-0.2 degrees, 23.1 +/-0.2 degrees, 29.5 +/-0.2 degrees and 30.2 +/-0.2 degrees;

characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 6.

TABLE 6

The compound III of the invention has a crystal form F, and the X-ray powder diffraction pattern of the crystal form F is basically as shown in figure 12; a DSC profile substantially as shown in figure 13; the TGA profile is substantially as shown in figure 14.

In a further preferred embodiment of the present invention, said compound III in form G, has an X-ray powder diffraction pattern with a characteristic peak at 19.6 ± 0.2 ° 2 Θ; preferably, the composition also comprises characteristic peaks at 15.4 +/-0.2 degrees and 19.0 +/-0.2 degrees of 2 theta; more preferably, further comprising having characteristic peaks at 6.9 ± 0.2 °, 7.7 ± 0.2 °, 11.0 ± 0.2 ° and 22.7 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 13.6 +/-0.2 degrees, 13.8 +/-0.2 degrees, 16.0 +/-0.2 degrees, 17.6 +/-0.2 degrees, 18.1 +/-0.2 degrees, 20.7 +/-0.2 degrees, 21.3 +/-0.2 degrees and 27.3 +/-0.2 degrees; still more preferably, the compound also comprises characteristic peaks at 2 theta of 8.7 +/-0.2 degrees, 10.4 +/-0.2 degrees, 13.1 +/-0.2 degrees, 14.9 +/-0.2 degrees, 20.3 +/-0.2 degrees, 22.4 +/-0.2 degrees, 23.5 +/-0.2 degrees, 25.5 +/-0.2 degrees and 27.8 +/-0.2 degrees.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 7.

TABLE 7

The X-ray powder diffraction pattern of the crystal form G of the compound III is basically shown as figure 15.

In a further preferred embodiment of the present invention, said compound III in crystalline form H, has an X-ray powder diffraction pattern with a characteristic peak at 7.2 ± 0.2 ° 2 Θ; preferably, the composition also comprises characteristic peaks at 16.0 +/-0.2 degrees and 20.8 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 16.4 ± 0.2 °, 17.6 ± 0.2 °, 19.7 ± 0.2 ° and 21.9 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.3 +/-0.2 degrees, 11.7 +/-0.2 degrees, 14.6 +/-0.2 degrees, 15.2 +/-0.2 degrees, 19.1 +/-0.2 degrees, 20.3 +/-0.2 degrees, 23.6 +/-0.2 degrees and 27.3 +/-0.2 degrees; still more preferably, the composition further comprises characteristic peaks at 9.2 + -0.2 deg., 13.3 + -0.2 deg., 18.4 + -0.2 deg. and 22.7 + -0.2 deg. of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 8.

TABLE 8

The X-ray powder diffraction pattern of the crystal form H of the compound III is basically shown in figure 16; a DSC profile substantially as shown in figure 17; the TGA profile is substantially as shown in figure 18.

In a further preferred embodiment of the present invention, said compound III in form I, has an X-ray powder diffraction pattern with a characteristic peak at 6.8 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 2 theta of 13.7 +/-0.2 degrees and 18.6 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 15.1 ± 0.2 °, 15.5 ± 0.2 °, 20.5 ± 0.2 ° and 20.7 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.2 +/-0.2 degrees, 11.8 +/-0.2 degrees, 17.7 +/-0.2 degrees, 20.2 +/-0.2 degrees, 22.4 +/-0.2 degrees, 23.1 +/-0.2 degrees, 23.5 +/-0.2 degrees and 27.0 +/-0.2 degrees;

characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 9.

TABLE 9

The compound III of the invention has the crystal form I, and the X-ray powder diffraction pattern of the compound III is basically as shown in figure 19; a DSC profile substantially as shown in figure 20; the TGA profile is substantially as shown in figure 21.

In a further preferred embodiment of the present invention, form J, the X-ray powder diffraction pattern of compound III has a characteristic peak at 20.5 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at the 2 theta of 18.7 +/-0.2 degrees and 20.2 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 6.8 ± 0.2 °, 12.2 ± 0.2 °, 12.6 ± 0.2 ° and 22.8 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 14.5 +/-0.2 degrees, 15.5 +/-0.2 degrees, 16.0 +/-0.2 degrees, 16.5 +/-0.2 degrees, 18.4 +/-0.2 degrees, 21.9 +/-0.2 degrees, 23.1 +/-0.2 degrees and 24.6 +/-0.2 degrees; still more preferably, the composition further comprises characteristic peaks at 13.7 + -0.2 DEG, 15.1 + -0.2 DEG, 22.5 + -0.2 DEG and 30.1 + -0.2 DEG in terms of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 10.

Watch 10

The X-ray powder diffraction pattern of the crystal form J of the compound III is basically shown in figure 22; a DSC profile substantially as shown in figure 23; the TGA profile is substantially as shown in figure 24.

In a further preferred embodiment of the present invention, said compound III in form K, has an X-ray powder diffraction pattern with a characteristic peak at 19.5 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 2 theta of 6.7 +/-0.2 degrees and 15.9 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 10.3 ± 0.2 °, 13.7 ± 0.2 °, 16.3 ± 0.2 ° and 22.5 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 10.0 +/-0.2 degrees, 12.2 +/-0.2 degrees, 14.9 +/-0.2 degrees, 19.1 +/-0.2 degrees, 20.2 +/-0.2 degrees, 20.7 +/-0.2 degrees, 23.7 +/-0.2 degrees, 26.2 +/-0.2 degrees and 29.0 +/-0.2 degrees of 2 theta;

characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 11.

TABLE 11

The X-ray powder diffraction pattern of the crystal form K of the compound III is basically shown in figure 25.

In a further preferred embodiment of the present invention, said compound III has crystalline form L, an X-ray powder diffraction pattern having a characteristic peak at 7.2 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 21.3 +/-0.2 degrees and 22.5 +/-0.2 degrees of 2 theta; more preferably, it further comprises characteristic peaks at 16.0 ± 0.2 °, 21.9 ± 0.2 °, 22.2 ± 0.2 ° and 27.3 ± 0.2 ° in 2 θ, further preferably, it further comprises characteristic peaks at 8.4 ± 0.2 °, 16.9 ± 0.2 °, 17.5 ± 0.2 °, 19.5 ± 0.2 °, 20.3 ± 0.2 °, 23.6 ± 0.2 °, 24.1 ± 0.2 ° and 27.9 ± 0.2 ° in 2 θ, further preferably, it further comprises characteristic peaks at 11.6 ± 0.2 °, 14.5 ± 0.2 °, 15.6 ± 0.2 °, 16.4 ± 0.2 °, 20.1 ± 0.2 ° and 30.4 ± 0.2 ° in 2 θ.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 12.

TABLE 12

A crystalline form L of compound III of the present invention having an X-ray powder diffraction pattern substantially as shown in figure 26; a DSC profile substantially as shown in figure 27; the TGA profile is substantially as shown in figure 28.

In a further preferred embodiment of the present invention, said compound III in crystalline form M, has an X-ray powder diffraction pattern with a characteristic peak at 18.6 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 15.7 +/-0.2 degrees and 25.1 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 21.7 ± 0.2 °, 23.4 ± 0.2 °, 24.0 ± 0.2 ° and 28.1 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 10.5 +/-0.2 degrees, 12.9 +/-0.2 degrees, 14.2 +/-0.2 degrees, 16.0 +/-0.2 degrees, 16.8 +/-0.2 degrees, 17.4 +/-0.2 degrees, 19.6 +/-0.2 degrees and 22.3 +/-0.2 degrees; still further preferably, the composition further comprises characteristic peaks at 9.3 + -0.2 deg., 21.2 + -0.2 deg., 22.8 + -0.2 deg., 29.4 + -0.2 deg., and 32.2 + -0.2 deg. of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 13.

Watch 13

The X-ray powder diffraction pattern of the crystal form M of the compound III is basically shown in figure 29; a DSC profile substantially as shown in figure 30; the TGA profile is substantially as shown in figure 31.

In a further preferred embodiment of the present invention, said compound III has a crystalline form N, X-ray powder diffraction pattern having a characteristic peak at 19.1 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 15.4 +/-0.2 degrees and 19.7 +/-0.2 degrees of 2 theta; more preferably, further comprising having characteristic peaks at 2 θ of 7.6 ± 0.2 °, 10.9 ± 0.2 °, 21.0 ± 0.2 ° and 21.3 ± 0.2 °; further preferably, the compound also comprises characteristic peaks at 2 theta of 13.5 +/-0.2 degrees, 18.0 +/-0.2 degrees, 18.8 +/-0.2 degrees, 22.6 +/-0.2 degrees, 25.4 +/-0.2 degrees, 26.2 +/-0.2 degrees and 27.3 +/-0.2 degrees;

characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 14.

TABLE 14

The X-ray powder diffraction pattern of the crystal form N of the compound III is basically shown in figure 32.

In a further preferred embodiment of the present invention, said compound III has crystalline form O, having an X-ray powder diffraction pattern with a characteristic peak at 23.8 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 2 theta of 12.7 +/-0.2 degrees and 17.9 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 14.4 ± 0.2 °, 17.6 ± 0.2 °, 21.0 ± 0.2 ° and 24.7 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 8.4 +/-0.2 degrees, 13.0 +/-0.2 degrees, 19.6 +/-0.2 degrees, 20.7 +/-0.2 degrees, 22.7 +/-0.2 degrees, 26.6 +/-0.2 degrees, 28.7 +/-0.2 degrees and 30.9 +/-0.2 degrees; still more preferably, the composition further comprises characteristic peaks at 6.7 + -0.2 deg., 12.0 + -0.2 deg., 14.8 + -0.2 deg., 15.8 + -0.2 deg., 16.5 + -0.2 deg., 19.0 + -0.2 deg., and 23.0 + -0.2 deg. of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 15.

Watch 15

The X-ray powder diffraction pattern of the crystal form O of the compound III is basically shown in figure 33; a DSC profile substantially as shown in figure 34; the TGA profile is substantially as shown in figure 35.

In a further preferred embodiment of the present invention, said compound III in crystalline form P, has an X-ray powder diffraction pattern with a characteristic peak at 23.2 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 14.9 +/-0.2 degrees and 16.5 +/-0.2 degrees of 2 theta; more preferably, further comprising characteristic peaks at 8.3 ± 0.2 °, 13.4 ± 0.2 °, 19.2 ± 0.2 ° and 20.2 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 7.8 +/-0.2 degrees, 9.3 +/-0.2 degrees, 11.6 +/-0.2 degrees, 12.6 +/-0.2 degrees, 16.0 +/-0.2 degrees, 18.5 +/-0.2 degrees, 19.6 +/-0.2 degrees and 27.4 +/-0.2 degrees; still more preferably, the composition further comprises characteristic peaks at 15.3 + -0.2 DEG and 22.1 + -0.2 DEG in terms of 2 theta.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 16.

TABLE 16

The X-ray powder diffraction pattern of the crystal form P of the compound III is basically shown in figure 36; a DSC profile substantially as shown in figure 37; its TGA profile is substantially as shown in figure 38.

In a further preferred embodiment of the present invention, said compound III has crystalline form Q, an X-ray powder diffraction pattern having a characteristic peak at 16.4 ± 0.2 ° 2 Θ; preferably, the compound also comprises characteristic peaks at 2 theta of 13.5 +/-0.2 degrees and 20.0 +/-0.2 degrees; more preferably, further comprising characteristic peaks at 9.1 ± 0.2 °, 14.7 ± 0.2 °, 15.9 ± 0.2 ° and 27.9 ± 0.2 ° in 2 θ; further preferably, the compound also comprises characteristic peaks at 2 theta of 7.7 +/-0.2 degrees, 11.7 +/-0.2 degrees, 11.9 +/-0.2 degrees, 18.4 +/-0.2 degrees, 19.5 +/-0.2 degrees, 22.3 +/-0.2 degrees, 26.4 +/-0.2 degrees and 27.3 +/-0.2 degrees.

Characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 17.

TABLE 17

A crystalline form Q of compound III of the present invention having an X-ray powder diffraction pattern substantially as shown in figure 39; a DSC profile substantially as shown in figure 40; the TGA profile is substantially as shown in figure 41.

The invention provides a crystal form A of a compound 3- ((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-yl) propionitrile (compound IV), which has the following structure:

an X-ray powder diffraction pattern of the compound has a diffraction peak at the 2 theta (+ -0.2 DEG) of 8.2 +/-0.2 DEG; preferably, further comprising diffraction peaks at 9.7 ± 0.2 ° and 20.4 ± 0.2 ° in 2 θ (± 0.2 °); more preferably, further comprising diffraction peaks at 13.5 ± 0.2 °, 17.4 ± 0.2 °, 21.1 ± 0.2 ° and 26.6 ± 0.2 ° in 2 θ (± 0.2 °); further preferably, it further comprises diffraction peaks at 16.5 ± 0.2 °, 16.8 ± 0.2 °, 14.1 ± 0.2 °, 19.1 ± 0.2 °, 19.4 ± 0.2 °, 22.8 ± 0.2 °, 27.1 ± 0.2 ° and 28.5 ± 0.2 ° in 2 θ (± 0.2 °); still further preferably, further comprising diffraction peaks at 9.4 ± 0.2 °, 19.8 ± 0.2 ° and 23.6 ± 0.2 ° in 2 θ (± 0.2 °);

characteristic diffraction peaks of X-rays expressed in terms of 2. theta. angle and interplanar spacing d using Cu-K.alpha.radiation are shown in Table 18.

Watch 18

Crystalline form a of the compound 3- ((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile of the present invention has an X-ray powder diffraction pattern substantially as shown in figure 42, a DSC pattern substantially as shown in figure 43, and a TGA pattern substantially as shown in figure 44.

In a further preferred embodiment of the present invention, the above-mentioned crystalline form is a solvent-containing crystalline form, wherein the solvent is selected from the group consisting of water, methanol, acetone, ethyl acetate, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, tert-butanol, 2-butanone, 3-pentanone, N-heptane, ethyl formate, isopropyl acetate, cyclohexane, methyl tert-butyl ether or isopropyl ether.

In a further preferred embodiment of the present invention, wherein the number of solvents is 0.2 to 3, preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3, more preferably 0.5, 1,2 or 3.

The invention also provides a method for preparing the crystal form of the compound shown in the general formula (I), which comprises the following steps:

1) weighing a proper amount of compound, and suspending with a poor solvent, wherein the suspension density is preferably 50-200 mg/mL;

2) shaking the suspension obtained above at a certain temperature for a certain time, wherein the temperature is preferably 0-50 ℃, and the time is preferably 1-10 days;

3) quickly centrifuging the suspension, removing supernatant, and drying to constant weight to obtain a target product;

wherein:

the poor solvent is selected from one or more of acetone, ethyl formate, ethyl acetate, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, chlorobenzene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, ethyl formate, isopropyl acetate, tert-butanol, 2-butanone or 3-pentanone; preferably one or more of ethyl acetate, tetrahydrofuran, acetone, acetonitrile, methanol, ethanol, dichloromethane, isopropyl acetate or isopropanol. Ethyl formate, ethyl formate,

The invention also provides a method for preparing the crystal form of the compound shown in the general formula (I), which comprises the following steps:

1) weighing a proper amount of compound, and dissolving the compound by using a good solvent;

2) adding an anti-solvent into the obtained solution at a certain temperature, and stirring until a solid is separated out, wherein the preferable temperature is 0-25 ℃;

3) quickly centrifuging the suspension, removing supernatant, and drying to constant weight to obtain a target product;

wherein:

the benign solvent is selected from one or more of methanol, acetone, ethyl acetate, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, tert-butanol, 2-butanone or 3-pentanone; one or two of methanol, dimethyl sulfoxide or isobutanol are preferred.

The poor solvent is selected from one or more of dichloromethane, toluene, ethyl formate, ethyl acetate, 2-methyl-tetrahydrofuran, ethanol, n-heptane, 1, 4-dioxane, water, isopropyl acetate, cyclohexane, methyl tert-butyl ether, toluene and isopropyl ether; one or more of water, n-heptane, cyclohexane and methyl tert-butyl ether are preferred.

Another object of the present invention is to provide a pharmaceutical composition comprising a therapeutically effective amount of a crystalline form of the above compound, together with one or more pharmaceutically acceptable carriers, diluents or excipients.

The invention also relates to application of the crystal form of any one of the compounds or the pharmaceutical composition in preparation of drugs for preventing and/or treating JAK kinase-related diseases, wherein the JAK kinase-related diseases are preferably inflammatory diseases and/or tumor diseases.

The invention further relates to the application of the medicine for preventing and/or treating diseases related to JAK kinase, wherein the inflammatory diseases are selected from rheumatoid arthritis, dermatitis, psoriasis and inflammatory bowel diseases; wherein the inflammatory bowel disease is preferably chronic intestinal inflammatory disease, further preferably ulcerative colitis and Crohn's disease;

the tumor disease is selected from myelofibrosis, polycythemia vera, essential thrombocythemia, myeloid leukemia, acute lymphocytic leukemia, ductal carcinoma of breast and non-small cell lung cancer.

Drawings

Figure 1 is a schematic representation of XRPD of compound III free base form a.

Figure 2 is a schematic representation of XRPD of compound III free base form B.

Figures 3-5 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form C.

Fig. 6-8 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form D.

Figures 9-11 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form E.

Fig. 12-14 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form F.

Figure 15 is a schematic representation of XRPD of compound III free base form G.

Figures 16-18 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form H.

Figures 19-21 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form I.

Figures 22-24 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form J.

Figure 25 is a schematic representation of XRPD of compound III free base form K.

Fig. 26-28 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form L.

Fig. 29-31 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form M.

Figure 32 is a schematic representation of XRPD of compound III free base form N.

Figures 33-35 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form O.

Figures 36-38 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form P.

Figures 39-41 are XRPD, DSC, TGA graphic representations of compound III free base crystalline form Q.

Figures 42-44 are XRPD, DSC, TGA graphic representations of compound IV free base crystalline form a.

Detailed Description

Unless stated to the contrary, terms used in the specification and claims have the following meanings.

The term "alkyl" refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 8 carbon atoms, more preferably an alkyl group of 1 to 6 carbon atoms, and most preferably an alkyl group of 1 to 3 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-dimethylpentyl, 2-dimethylhexyl, 3-dimethylpentyl, 2-ethylhexyl, 3-dimethylhexyl, 2-ethylhexyl, 2-dimethylhexyl, 2-ethylhexyl, 2-dimethylhexyl, 2-dimethylhexyl, 2-dimethylhexyl, 2-ethylhexyl, 2-ethyl, 2-2, 2-2, 2-2, or, 2, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof. Alkyl groups may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halo, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxy or carboxylate, preferably methyl, ethyl, isopropyl, tert-butyl, haloalkyl, deuterated alkyl, alkoxy-substituted alkyl, hydroxy-substituted alkyl and cyano-substituted alkyl.

The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 8 carbon atoms, more preferably from 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups, preferably cyclopropyl, cyclobutyl, cyclohexyl, cyclopentyl and cycloheptyl. The cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring, where the ring to which the parent structure is attached is cycloalkyl, non-limiting examples of which include indanyl, tetrahydronaphthyl, benzocycloheptanyl, and the like. Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxy or carboxylate.

The term "heterocyclyl" refers to a saturated or partially unsaturated mono-or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more of the ring atoms is selected from nitrogen, oxygen, or S (O)m(wherein m is an integer from 0 to 2) but excludes the ring moiety of-O-O-, -O-S-, or-S-S-, the remaining ring atoms being carbon. Preferably 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; more preferably from 3 to 8 ring atoms; most preferably from 3 to 8 ring atoms. Non-limiting examples of monocyclic heterocyclic groups include oxetanyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, and the like, with oxetanyl, pyrrolidinonyl, tetrahydrofuranyl, pyrazolidinyl, morpholinyl, piperazinyl, and pyranyl being preferred. Polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups; wherein the heterocyclic groups of the spiro, fused and bridged rings are optionally linked to other groups by single bonds, or further linked to other cycloalkyl, heterocyclic, aryl and heteroaryl groups by any two or more atoms in the ring. The heterocyclyl group may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxoA carboxyl group or a carboxylate group.

The term "aryl" refers to a 6 to 14 membered all carbon monocyclic or fused polycyclic (i.e., rings which share adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. More preferably phenyl. The aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring. The aryl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxy or carboxylate.

The term "heteroaryl" refers to a heteroaromatic system comprising 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl is preferably 5 to 10 membered, more preferably 5 or 6 membered, such as imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, oxadiazole, pyrazinyl and the like, preferably oxazolyl, oxadiazole, tetrazole, triazolyl, thienyl, imidazolyl, pyridyl, pyrazolyl, pyrimidinyl or thiazolyl; more preferred are oxazolyl, oxadiazole, tetrazole, triazolyl, thienyl, pyridyl, thiazolyl, and pyrimidinyl. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring joined to the parent structure is a heteroaryl ring. Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate groups.

The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl), wherein alkyl is as defined above. Alkoxy groups having 1 to 8 carbon atoms are preferred, alkoxy groups having 1 to 6 carbon atoms are more preferred, and alkoxy groups having 1 to 3 carbon atoms are most preferred. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy. Alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxy or carboxylate groups.

"haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.

"haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.

"hydroxyalkyl" refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.

"alkenyl" refers to alkenyl, also known as alkenylene, wherein the alkenyl may be further substituted with other related groups, such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate.

"hydroxy" refers to an-OH group.

"halogen" means fluorine, chlorine, bromine or iodine.

"amino" means-NH2

"cyano" means-CN.

"nitro" means-NO2

"THF" refers to tetrahydrofuran.

"EtOAc" refers to ethyl acetate.

"DMSO" refers to dimethyl sulfoxide.

"LDA" refers to lithium diisopropylamide.

"DMAP" refers to 4-dimethylaminopyridine.

"EtMgBr" refers to ethyl magnesium bromide.

"HOSu" refers to N-hydroxysuccinimide.

"EDCl" refers to 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride.

"IPA" refers to isopropanol.

"MeOH" refers to methanol.

"EtOH" refers to ethanol.

"DMF" refers to N, N-dimethylformamide.

"DIPEA" refers to N, N-diisopropylethylamine.

"HEPES" means 4-hydroxyethylpiperazine ethanesulfonic acid.

Different terms such as "X is selected from A, B or C", "X is selected from A, B and C", "X is A, B or C", "X is A, B and C" and the like all express the same meaning, that is, X can be any one or more of A, B, C.

"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not.

"substituted" means that one or more, preferably up to 5, more preferably 1 to 3, hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.

"stereoisomerism" encompasses geometric isomerism (cis-trans isomerism), optical isomerism, conformational isomerism, and the like.

All hydrogen atoms described in the present invention can be replaced by deuterium, which is an isotope thereof, and any hydrogen atom in the compound of the embodiment related to the present invention can also be replaced by a deuterium atom.

"pharmaceutical composition" means a mixture containing one or more compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof in admixture with other chemical components, as well as other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.

X-ray powder diffraction pattern (XRPD), which refers to the experimentally observed diffraction pattern or parameters derived therefrom, is characterized by peak position (abscissa) and peak intensity (ordinate). Those skilled in the art will appreciate that the experimental error therein will depend on the conditions of the instrument, the sample preparation and the purity of the sample. In particular, it is well known to those skilled in the art that X-ray diffraction patterns will generally vary with the conditions of the instrument, and those skilled in the art will appreciate that a suitable error tolerance for XRPD may be: 2 theta +/-0.5 degrees; 2 theta +/-0.4 degrees; 2 theta +/-0.3 degrees; 2 theta +/-0.2 deg. It is particularly noted that the relative intensities of the X-ray diffraction patterns may also vary with the experimental conditions, so that the order of the peak intensities cannot be considered as the sole or determining factor. In addition, due to the influence of experimental factors such as sample height, an overall shift in peak angle is caused, and a certain shift is usually allowed. Thus, it will be understood by those skilled in the art that any crystalline form having the same or similar characteristic peaks as the inventive profile is within the scope of the invention.

"TGA" refers to a thermogravimetric analysis (TGA) experiment.

"DSC" refers to a Differential Scanning Calorimetry (DSC) experiment.

"HPLC" refers to High Performance Liquid Chromatography (HPLC) experiments.

"PK" refers to Pharmacokinetic (PK) experiments.

The present invention is further described below with reference to examples, which are not intended to limit the scope of the present invention.

Preparation of compounds

The structure of the compounds of the invention is determined by Nuclear Magnetic Resonance (NMR) or/and liquid mass chromatography (LC-MS). NMR chemical shifts (δ) are given in parts per million (ppm). NMR was measured using Bruker AVANCE-400 NMR instrument, and determination solvent is deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated methanol (CD)3OD) and deuterated chloroform (CDCl)3) Internal standard is Tetramethylsilane (TMS).

LC-MS was measured using an Agilent 1200Infinity Series Mass spectrometer. HPLC was measured using an Agilent 1200DAD high pressure liquid chromatograph (Sunfire C18150X 4.6mm column) and a Waters 2695-2996 high pressure liquid chromatograph (Gimini C)18150X 4.6mm column).

The thin layer chromatography silica gel plate adopts a tobacco yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification adopted by TLC is 0.15 mm-0.20 mm, and the specification adopted by the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm. The column chromatography generally uses 200-300 mesh silica gel of the Tibet Huanghai silica gel as a carrier.

The starting materials in the examples of the present invention are known and commercially available, or may be synthesized using or according to methods known in the art.

All reactions of the present invention are carried out under continuous magnetic stirring in a dry nitrogen or argon atmosphere, without specific indication, the solvent is a dry solvent, and the reaction temperature is given in degrees celsius.

Example 1

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 2-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [3,2-d ] pyrimidin-4-amine

To a solution of 2, 4-dichlorothieno [3,2-d ] pyrimidine (205mg,1mmol) in N-methylpyrrolidone (10mL) were added 3-amino-5-methylpyrazole (116mg,1.2mmol), DIPEA (258mg,2mmol) in this order, followed by stirring at 70 ℃ for 1 hour. After completion of the reaction, water (50mL) was added to the reaction mixture, and the precipitated solid was filtered and slurried with ethyl acetate to give the title compound as a pale yellow solid (135mg, 51%).

MS m/z(ESI):266.0[M+H]+.

The second step is that: preparation of tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

To a solution of 2-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [3,2-d ] pyrimidin-4-amine (135mg,0.51mmol) in N-butanol (5mL) were added tert-butyl- (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate (138mg,0.61mmol), DIPEA (129mg,1mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After the reaction was completed, the reaction solution was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 98:2) to obtain the title compound as a pale yellow solid (146mg, 63%).

MS m/z(ESI):456.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (146mg,0.32mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; methanol (10mL) was then added to dissolve it, DIPEA (166mg, 1.28mmol) was added slowly dropwise, stirring at room temperature for 10 minutes, acrylonitrile (25mg,0.48mmol) was added and stirring continued for 2 hours. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (14.4mg, 11%).

1H NMR(400MHz,DMSO)δ12.02(s,1H),9.70(s,1H),7.89(s,1H),6.99(s, 1H),6.44(d,J=59.6Hz,2H),4.14(s,1H),3.29(s,2H),2.62(s,4H),2.22(s,3H), 1.89(s,2H),1.64(dd,J=47.8,17.6Hz,6H).

MS m/z(ESI):409.2[M+H]+.

Example 2

3- ((3-exo) -3- ((7-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 1.

MS m/z(ESI):423.2[M+H]+.

Example 3

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 2-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine

To a solution of 2, 4-dichlorothieno [2,3-d ] pyrimidine (205mg,1mmol) in N-methylpyrrolidone (10mL) were added 3-amino-5-methylpyrazole (116mg,1.2mmol), DIPEA (258mg,2mmol) in this order, followed by stirring at 70 ℃ for 1 hour. After completion of the reaction, water (50mL) was added to the reaction mixture to precipitate a solid, which was filtered and slurried with ethyl acetate to give the title compound as a yellow solid (250mg, 94%).

MS m/z(ESI):266.0[M+H]+.

The second step is that: preparation of tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

To a solution of 2-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (250mg,0.94mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate (256mg,1.13mmol), DIPEA (242mg,1.88mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After the reaction was completed, the reaction solution was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 98:2) to obtain the title compound as a pale yellow solid (200mg, 47%).

MS m/z(ESI):456.1[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (200mg,0.44mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; methanol (10mL) was then added to dissolve it, DIPEA (227mg, 1.76mmol) was added slowly dropwise, stirring was carried out at room temperature for 10 minutes, and acrylonitrile (35mg,0.66mmol) was added followed by stirring for 2 hours. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (31.6mg, 18%).

1H NMR(400MHz,DMSO)δ12.13(s,1H),9.93(s,1H),7.73(s,1H),6.88(d,J =117.2Hz,3H),4.27(s,1H),3.37(s,2H),2.70(s,4H),2.32(s,3H),1.99(s,2H), 1.86-1.61(m,6H).

MS m/z(ESI):409.2[M+H]+.

Example 4

3- ((3-exo) -3- ((7- ((5-methyl-1H-pyrazol-3-yl) amino) thiazolo [4,5-d ] pyrimidin-5-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- ((5-methyl-1H-pyrazol-3-yl) amino) thiazolo [4,5-d ] pyrimidin-5-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 1.

MS m/z(ESI):410.2[M+H]+.

Example 5

3- ((3-exo) -3- ((7- ((5-methyl-1H-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 5-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thiazolo [5,4-d ] pyrimidin-7-amine

To a solution of 5, 7-dichlorothiazolo [5,4-d ] pyrimidine (206mg,1mmol) in dimethylsulfoxide (10mL) was added 3-amino-5-methylpyrazole (116mg,1.2mmol), DIPEA (258mg,2mmol) in this order, followed by stirring at 70 ℃ for 1 hour. After completion of the reaction, water (50mL) was added to the reaction mixture to precipitate a solid, which was filtered and slurried with ethyl acetate to give the title compound as a yellow solid (200mg, 75%).

MS m/z(ESI):267.0[M+H]+.

The second step is that: preparation of tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

To a solution of 5-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thiazolo [5,4-d ] pyrimidin-7-amine (200mg,0.75mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate (204mg,0.9mmol), DIPEA (193mg,1.5mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After the reaction was completed, the reaction solution was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 95:5) to obtain the title compound as a pale yellow solid (74mg, 22%).

MS m/z(ESI):457.1[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (74mg,0.16mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,2mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; methanol (10mL) was then added to dissolve it, DIPEA (83mg, 0.64mmol) was added slowly dropwise, stirring was carried out at room temperature for 10 minutes, and acrylonitrile (9mg,0.24mmol) was added followed by stirring for 2 hours. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (16.3mg, 25%).

1H NMR(400MHz,DMSO)δ12.07(s,1H),9.33(s,1H),8.76(d,J=20.4Hz, 1H),6.96(s,1H),6.55(d,J=12.0Hz,1H),4.14(s,1H),3.31(s,2H),2.61(s,4H), 2.21(s,3H),1.91(s,2H),1.78-1.54(m,6H).

Example 6

1- (((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

The first step is as follows: preparation of (3- ((2-chlorothieno [2,3-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol

2, 4-dichlorothieno [2,3-d ] pyrimidine (100mg, 0.49mmol), (3-amino-1H-pyrazol-5-yl) methanol (55mg, 0.49mmol), DIPEA (190mg, 1.47mmol) and the reaction mixture were added to N' N-dimethylformamide (2mL), and the reaction mixture was stirred at 70 ℃ overnight. Concentrated under reduced pressure, and the resulting crude product was isolated and purified by flash silica gel column chromatography to give the title compound as a yellow solid (100mg, 73%).

MS m/z(ESI):282.0[M+H]+.

The second step is that: preparation of tert-butyl (3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

Adding (3- ((2-chlorothieno [2,3-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol (100mg,0.36 mmol), tert-butyl (3-exo) -3- (methylamino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (135mg, 0.53mmol) and DIPEA (140mg,1.08mmol) into n-butanol (2.5mL), uniformly mixing, reacting at the temperature of 150 ℃ for 10 hours by using a microwave, cooling to room temperature, concentrating the reaction liquid under reduced pressure, and separating and purifying the obtained crude product by using a flash silica gel column chromatography to obtain a white solid (70mg, 39%).

MS m/z(ESI):500.1[M+H]+.

The third step: preparation of 1- (((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Dioxane hydrochloride (4N,2.5mL) was slowly added dropwise to a methanol (10mL) solution of tert-butyl (3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (70mg, 0.14mmol), the mixture was reacted at room temperature for 2 hours, the reaction mixture was concentrated under reduced pressure, the crude product was dissolved in DMF (5mL), DIPEA (0.3mL) and 3-cyanoazetidine-1-sulfonyl chloride (22mg,0.12mmol) were added in an ice-water bath at 0 ℃ and the mixture was reacted at room temperature overnight. The reaction mixture was concentrated under reduced pressure and purified by prep-HPLC to give the title compound as a white solid (9.7mg, 13%)

1H NMR(400MHz,DMSO-d6)δ9.81(s,1H),7.68(d,J=4.4Hz,1H),7.04(d,J =6.0Hz,1H),6.52-6.54(m,1H),5.53-5.55(m,1H),5.33-5.35(m,1H),4.44(d,J=5.2 Hz,2H),4.05-4.01(m,4H),3.94-3.90(m,2H),382-3.79(m,1H),2.89(d,J=8.4Hz, 3H),2.08-1.68(m,11H).

MS m/z(ESI):544.1[M+H]+.

Example 7

1- (((3-exo) -3- ((7- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- (((3-exo) -3- ((7- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile reference example 6.

MS m/z(ESI):545.2[M+H]+.

Example 8

1- (((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (200mg, 0.439mmol) was dissolved in a solution of 4M HCl in 1, 4-epoxyhexacyclic ring (20mL), and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (1.45 mL, 8.78mmol) and 3-cyanoazetidine-1-sulfonyl chloride (95mg, 0.527mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 16.5 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (70mg, 32%).

1H NMR(400MHz,MeOD-d4)δ7.37(d,J=6.0Hz,1H),6.94(d,J=6.0 Hz,1H),6.25(s,1H),4.44-4.34(m,1H),4.26(s,2H),4.16(t,J=8.5Hz, 2H),4.12-4.05(m,2H),3.57(ddd,J=15.3,8.7,6.5Hz,1H),2.31(s,3H), 2.23-2.10(m,4H),2.01(d,J=7.4Hz,2H),1.73(dd,J=18.2,7.1Hz,2H).

MS m/z(ESI):500.1[M+H]+.

Example 9

3- ((3-exo) -3- (((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino)) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then it was dissolved in N, N-dimethylformamide (10mL), DIPEA (108mg, 0.84mmol) was added slowly dropwise, stirred at room temperature for 10 minutes, and after addition of 3-nitriloazetidine-1-sulfonyl chloride (45mg, 0.25mmol), stirring at room temperature was continued overnight. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (14.4mg, 13%).

1H NMR(400MHz,DMSO)δ=12.02(s,1H),9.81(s,1H),7.61(s,1H), 6.90(s,1H),6.59(d,J=57.6Hz,2H),4.74(s,1H),3.96(t,J=8.4Hz,2H), 3.85(dd,J=16.8Hz,6.4,4H),3.75-3.67(m,1H),2.14(s,3H),2.00(d,J= 8.4Hz,2H),1.87-1.60(m,8H).

MS m/z(ESI):514.1[M+H]+.

Example 10

1- (((3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

The first step is as follows: preparation of tert-butyl- (3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

To a solution of 2-chloro-N- (5-methyl-1 h-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (250mg,0.94mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3- (methylamino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (287mg, 1.13mmol), DIPEA (242mg,1.88mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After completion of the reaction, the reaction mixture was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 98:2) to obtain the title compound as a pale white solid (228mg, 50%).

MS m/z(ESI):484.2[M+H]+.

The second step is that: preparation of 1- (((3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl- (3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then it was dissolved in N, N-dimethylformamide (10mL), DIPEA (108mg, 0.84mmol) was added slowly dropwise, stirred at room temperature for 10 minutes, and after addition of 3-nitriloazetidine-1-sulfonyl chloride (45mg, 0.25mmol), stirring at room temperature was continued overnight. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (46.0mg, 42%).

1H NMR(400MHz,DMSO)δ=12.09(s,1H),9.79(s,1H),7.68(d,J=6.0 Hz,1H),7.02(d,J=6.0Hz,1H),6.43(s,1H),5.77(s,1H),3.98(dt,J= 14.4,8.4Hz,6H),3.84-3.74(m,1H),2.90(s,3H),2.22(s,3H),2.13-1.61(m, 10H).

MS m/z(ESI):528.2[M+H]+.

Example 11

1- (((3-exo) -3- (methyl (6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

The first step of reaction: preparation of tert-butyl (3-exo) -3- (methyl (6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

2-chloro-6-methyl-N- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (150mg, 0.536mmol) and tert-butyl (3-exo) -3- (methylamino) -8-azabicyclo [3.2.1] octane-8-carboxylate (257mg, 1.072mmol) were added to N-butanol (10mL) and heated to 170 ℃ with a microwave synthesizer for 8 hours. The solvent was removed by concentration under reduced pressure, and the residue was dissolved in methylene chloride, washed successively with a saturated aqueous solution of sodium hydrogencarbonate and saturated aqueous solution of sodium chloride, and the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated, and isolated by silica gel column chromatography to give the title compound (73mg, 28%).

MS m/z(ESI):484.2[M+H]+.

The second step of reaction: preparation of 1- (((3-exo) -3- (methyl (6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- (methyl (6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (73mg, 0.151mmol) was dissolved in 4M HCl in 1, 4-epoxyhexacyclic ring (20mL) and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residual solid was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (0.75mL, 4.53mmol) and 3-cyanoazetidine-1-sulfonyl chloride (30mg, 0.166mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 4.5 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by prep-HPLC to give the title compound (31.5mg, 40%).

1H NMR(400MHz,DMSO-d6)δ12.06(s,1H),9.66(s,1H),7.35(s,1H), 6.48(s,1H),5.31-5.15(m,1H),4.18(d,J=1.0Hz,2H),4.06(t,J=8.6Hz, 2H),4.00-3.91(m,2H),3.80(ddd,J=12.8,8.9,6.5Hz,1H),2.90(s,3H), 2.40(s,3H),2.22(s,3H),2.07-1.99(m,2H),1.95(dd,J=18.2,7.0Hz,2H), 1.88-1.79(m,2H),1.62(dd,J=11.8,4.1Hz,2H).

MS m/z(ESI):528.2[M+H]+.

Example 12

1- (((3-exo) -3- (methyl (4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

The first step of reaction: preparation of tert-butyl (3-exo) -3- (methyl (4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

2-chloro-N- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (100mg, 0.376mmol) and tert-butyl (3-exo) -3- (methylamino) -8-azabicyclo [3.2.1] octane-8-carboxylate (181mg, 0.752 mmol) were added to N-butanol (3mL), and heated to 170 ℃ with a microwave synthesizer for 18 hours. The solvent was removed by concentration under reduced pressure, and the residue was used directly in the next reaction.

MS m/z(ESI):470.2[M+H]+.

The second step of reaction: preparation of 1- (((3-exo) -3- (methyl (4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- (methyl (4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate was dissolved in 4M HCl in 1, 4-epoxyhexacyclic compound (20mL), the reaction was stirred at room temperature for 30 minutes, the solvent was removed by concentration under reduced pressure, and the residue was chromatographically separated by a reverse phase column to give 117mg of a white solid.

The above white solid was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 deg.C, DIPEA (0.14mL, 0.632mmol) and 3-cyanoazetidine-1-sulfonyl chloride (57mg, 0.316 mmol) were added sequentially, and the reaction mixture was stirred at 0 deg.C for 17 hours. The solvent was removed by concentration under reduced pressure and the residue was isolated by prep-HPLC to give the title compound (16.4mg, 10%).

1H NMR(400MHz,MeOD-d4)δ7.37(d,J=5.9Hz,1H),6.98(d,J=5.7 Hz,1H),6.40(s,1H),5.40-5.28(m,1H),4.31-4.24(m,2H),4.17(t,J=8.5 Hz,2H),4.11-4.04(m,2H),3.57(ddd,J=15.4,8.9,6.7Hz,1H),3.04(s, 3H),2.31(s,3H),2.17(dd,J=8.6,3.3Hz,2H),2.11-2.01(m,2H),2.00-1.92 (m,2H),1.75(ddd,J=10.8,4.3,2.7Hz,2H).

MS m/z(ESI):514.1[M+H]+.

Example 13

2- (dimethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ethan-1-one

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.32mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, dimethylglycine (24mg, 0.23mmol) was added and stirring was continued for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (17.1mg, 18%).

1H NMR(400MHz,DMSO)δ=12.00(s,1H),9.80(s,1H),7.61(s,1H), 6.98-6.45(m,3H),4.76(s,1H),4.59(s,1H),4.27(s,1H),3.30(s,6H),3.05 (s,2H),2.16(s,3H),2.14(s,2H),2.07-1.92(m,2H),1.86-1.40(m,6H).

MS m/z(ESI):455.2[M+H]+.

Example 14

2- (dimethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (40mg, 0.09mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,2mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (51mg, 0.13mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (46mg, 0.36mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, dimethylglycine (10mg,0.1 mmol) was added and stirring was continued for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (4.4mg, 11%).

1H NMR(400MHz,DMSO)δ=12.08(s,1H),9.88(s,1H),7.65(s,1H), 7.11-6.46(m,3H),4.47(d,J=30.0Hz,3H),3.06(s,2H),2.21(s,9H),2.04- 1.66(m,6H),1.62-1.44(m,2H).

MS m/z(ESI):441.2[M+H]+.

Example 15

1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) -2-morpholinoethan-1-one

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (40mg, 0.09mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,2mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; 2- (7-Benzotolyltriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (51mg, 0.13mmol) was then added and dissolved in N, N-dimethylformamide (5mL), DIPEA (46mg, 0.36mmol) was added slowly dropwise, stirring was carried out for 10 minutes in an ice-water bath, and 2-morpholinoacetic acid (14.5mg, 0.1mmol) was added and stirring was continued for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (7.8mg, 18%).

1H NMR(400MHz,DMSO)δ=12.07(s,1H),9.88(s,1H),7.66(s,1H), 7.11-6.49(m,3H),4.48(d,J=26.4Hz,3H),3.60(s,4H),3.17(s,2H),2.46 (s,4H),2.23(s,3H),1.98(s,2H),1.90-1.45(m,6H).

MS m/z(ESI):483.2[M+H]+.

Example 16

1- ((3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) -2-morpholinoethan-1-one

Preparation of 1- ((3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) -2-morpholinoethan-1-one refers to example 244.

1H NMR(400MHz,DMSO-d6)δ=9.85(s,1H),8.22(s,1H),7.68(d,J=6.0Hz, 1H),7.02(d,J=6.0Hz,1H),6.53(s,1H),5.37(s,1H),4.54(d,J=16.4Hz,2H),3.58 (d,J=4.0Hz,4H),3.04(d,J=13.2Hz,2H),2.85(s,3H),2.45(s,4H),2.23(s,3H), 2.03-1.97(m,2H),1.87-1.59(m,6H).

MS m/z(ESI):497.2[M+H]+.

Example 17

1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2-morpholinoethane-1-one

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, and 2-morpholinoacetic acid (33mg, 0.23mmol) was added followed by stirring for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (18.0mg, 17%).

1H NMR(400MHz,DMSO)δ=12.08(s,1H),9.87(s,1H),7.68(s,1H), 7.07-6.53(m,3H),4.83(s,1H),4.65(s,1H),4.37(s,1H),3.59(d,J=4.0Hz, 4H),3.12(dd,J=25.2,12.4Hz,2H),2.39(s,4H),2.21(s,3H),2.11-1.51(m, 10H).

MS m/z(ESI):497.2[M+H]+.

Example 18

1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (methylamino) -ethan-1-one

The first step is as follows: preparation of tert-butylmethyl (2- ((3-exo) -3- ((4- ((5-methyl-1-hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2-carbonylethyl) carbamate

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (200mg, 0.42mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N, 10mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; 2- (7-Benzobenzotriazol-N, N, N ', N' -tetramethyluronium hexafluorophosphate) (240mg, 0.64mmol) was then added and dissolved in N, N-dimethylformamide (5mL), DIPEA (216mg, 1.68mmol) was added slowly dropwise, stirring was carried out for 10 minutes in an ice water bath, and stirring was continued for 1 hour after the addition of N- (tert-butoxycarbonyl) -N-methylglycine (87mg, 0.46 mmol). After completion of the reaction, the reaction mixture was extracted with dichloromethane (15mL × 3), washed with saturated aqueous sodium chloride (15mL × 3), the organic phase was collected, dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 95:5) to obtain the title compound as a white solid (205mg, 90%).

MS m/z(ESI):541.2[M+H]+.

The second step is that: preparation of 1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (methylamino) -ethan-1-one

Tert-butylmethyl (2- ((3-exo) -3- ((4- ((5-methyl-1 h-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2-carbonylethyl) carbamate (205mg, 0.38mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N, 10mL), stirred at room temperature for 30 minutes, then ammonia water (10mL) was added dropwise to the reaction solution in an ice-water bath, and then the reaction solution was concentrated under reduced pressure to give the title compound as a white solid (37.6mg, 22%) by prep-HPLC.

1H NMR(400MHz,DMSO)δ=12.10(s,1H),9.88(s,1H),7.68(s,1H), 6.96(s,1H),6.61(s,2H),4.84(s,1H),4.69(s,1H),4.12(s,1H),2.29(s, 3H),2.20(s,3H),2.15-1.96(m,3H),1.87-1.47(m,10H).

MS m/z(ESI):441.2[M+H]+.

Example 19

((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((R) -pyrrolidin-2-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((R) -pyrrolidin-2-yl) methanone reference example 18.

1H NMR(400MHz,DMSO-d6)δ=12.08(s,1H),9.89(s,1H),7.66(s,1H), 7.06-6.51(m,3H),4.55-4.35(m,3H),3.73(s,1H),3.01(s,1H),2.64(d,J= 6.8Hz,2H),2.23(s,3H),2.10-1.43(m,12H).

MS m/z(ESI):453.1[M+H]+.

Example 20

((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((S) -pyrrolidin-2-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((S) -pyrrolidin-2-yl) methanone reference example 18.

1H NMR(400MHz,DMSO-d6)δ=12.11(s,1H),9.90(s,1H),7.67(s,1H), 5.98-6.54(m,3H),4.58-4.35(m,3H),4.09-4.02(m,1H),3.11(s,1H),2.97- 2.64(m,2H),2.23(s,3H),2.10-1.37(m,10H).

MS m/z(ESI):453.1[M+H]+.

Example 21

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((R) -morpholin-3-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((R) -morpholin-3-yl) methanone reference example 18.

MS m/z(ESI):483.2[M+H]+.

Example 22

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((R) -pyrrolidin-2-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ((R) -pyrrolidin-2-yl) methanone reference example 18.

MS m/z(ESI):467.2[M+H]+.

Example 23

2- ((2-methoxyethyl) amino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ethan-1-one

Preparation of 2- ((2-methoxyethyl) amino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) ethan-1-one reference example 18.

MS m/z(ESI):485.2[M+H]+.

Example 24

1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- ((pyridin-3-ylmethyl) amino) ethan-1-one

Preparation of 1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- ((pyridin-3-ylmethyl) amino) ethan-1-one reference example 18.

MS m/z(ESI):518.2[M+H]+.

Example 25

2- ((4-methoxybenzyl) amino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Preparation of 2- ((4-methoxybenzyl) amino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one reference example 18.

MS m/z(ESI):533.2[M+H]+.

Example 26

2- (ethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Preparation of 2- (ethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one reference example 18.

MS m/z(ESI):441.2[M+H]+.

Example 27

2- (cyclopropylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Preparation of 2- (cyclopropylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one reference example 18.

MS m/z(ESI):453.2[M+H]+.

Example 28

1- ((3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (4-methylpiperazin-1-yl) ethan-1-one

Tert-butyl- (3-exo) -3- (methyl (4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then, 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirring was performed for 10 minutes in an ice-water bath, and 2- (4-methylpiperazin-1-yl) acetic acid (36mg, 0.23mmol) was added followed by stirring for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (43.8mg, 40%).

1H NMR(400MHz,DMSO)δ=12.07(s,1H),9.79(s,1H),7.68(d,J=6.0 Hz,1H),7.02(d,J=6.0Hz,1H),6.46(s,1H),5.81(s,1H),4.71(s,1H), 4.39(s,1H),3.22(d,J=12.8Hz,1H),3.06(d,J=12.8Hz,1H),2.85(s, 3H),2.40(s,8H),2.22(s,3H),2.17(s,3H),2.12-2.02(m 2H),1.90-1.61(m, 8H).

MS m/z(ESI):424.2[M+H]+.

Example 29

1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (4-methylpiperazin-1-yl) ethan-1-one

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then, 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirring was performed for 10 minutes in an ice-water bath, and 2- (4-methylpiperazin-1-yl) acetic acid (36mg, 0.23mmol) was added followed by stirring for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (25.2 mg, 24%).

1H NMR(400MHz,DMSO-d6)δ=12.07(s,1H),9.92(s,1H),7.68(s,1H), 6.96-6.61(m,3H),4.85(s,1H),4.65(s,1H),4.37(s,1H),3.10(s,2H),2.37 (s,8H),2.21(s,3H),2.14(s,3H),2.09-1.99(m,2H),1.97-1.46(m,8H).

MS m/z(ESI):510.2[M+H]+.

Example 30

1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (piperazin-1-yl) ethan-1-one

Preparation of 1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (piperazin-1-yl) ethan-1-one reference example 18.

MS m/z(ESI):496.2[M+H]+.

Example 31

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) (pyridin-2-yl) methanone

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, pyridine-2-carboxylic acid (28mg, 0.23mmol) was added and stirring was continued for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (27.3mg, 21%).

1H NMR(400MHz,DMSO)δ=12.07(s,1H),9.86(s,1H),8.59(d,J=4.4 Hz,1H),7.94(td,J=7.7,1.6Hz,1H),7.73-7.44(m,3H),7.05-6.50(m,3H), 4.84(d,J=28.0Hz,2H),3.94(s,1H),2.21(s,3H),2.18-1.59(m,10H).

MS m/z(ESI):475.1[M+H]+.

Example 32

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) (pyridin-3-yl) methanone

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, pyridine-3-carboxylic acid (28mg, 0.23mmol) was added and stirring was continued for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (28.6mg, 22%).

1H NMR(400MHz,DMSO-d6)δ=12.07(s,1H),9.87(s,1H),8.71-8.58(m, 2H),7.84(d,J=7.6Hz,1H),7.69(d,J=6.0Hz,1H),7.51(dd,J=7.6,4.8 Hz,1H),6.96(d,J=5.2Hz,1H),6.68-6.50(m,2H),4.83(d,J=39.2Hz, 2H),3.78(s,1H),2.21(s,3H),2.13-1.61(m,10H).

MS m/z(ESI):475.1[M+H]+.

Example 33

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) (pyridin-4-yl) methanone

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, pyridine-4-carboxylic acid (28mg, 0.23mmol) was added and stirring was continued for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (34.5mg, 27%).

1H NMR(400MHz,DMSO-d6)δ=12.07(s,1H),9.87(s,1H),8.69(d,J =6.0Hz,2H),7.69(d,J=5.6Hz,1H),7.39(d,J=5.6Hz,2H),6.97(d,J =6.0Hz,1H),6.57(d,J=7.6Hz,2H),4.82(d,J=41.6Hz,2H),3.68(s, 1H),2.21(s,3H),2.14-1.59(m,10H).

MS m/z(ESI):475.1[M+H]+.

Example 34

(1-methyl-1 hydro-imidazol-2-yl) ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) methanone

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, and 1-methyl-1 h-imidazole-2-carboxylic acid (29mg, 0.23mmol) was added followed by stirring for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (15.0 mg, 12%).

1H NMR(400MHz,DMSO-d6)δ=12.08(s,1H),9.87(s,1H),7.68(s,1H), 7.29(s,1H),6.97(s,2H),6.65(s,2H),4.91-4.80(m,3H),3.77(s,3H),2.22 (s,3H),2.14-1.60(m,10H).

MS m/z(ESI):478.2[M+H]+.

Example 35

(1-methyl-1-hydro-imidazol-4-yl) ((3-exo) -3- ((4- ((5-methyl-1-hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) methanone

Preparation of (1-methyl-1-hydro-imidazol-4-yl) ((3-exo) -3- ((4- ((5-methyl-1-hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) methanone reference example 21

1H NMR(400MHz,DMSO-d6)δ=12.08(s,1H),9.86(s,1H),7.70-7.60(m,3H),6.95(s,1H),6.65(s,1H),5.57(s,1H),4.82(d,J=65.2Hz,3H),3.68 (s,3H),2.22(s,3H),2.13-1.58(s,10H).

MS m/z(ESI):478.1[M+H]+.

Example 36

N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -8 (pyridin-3 ylsulfonyl) -8-azabicyclo [ 3.2.1)]Octane-3-yl) thieno [2,3-d]Pyrimidine-2, 4-diamines

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (40mg, 0.09mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,2mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; n, N-dimethylformamide (5mL) was then added to dissolve it, DIPEA (46mg, 0.36mmol) was added slowly dropwise, stirring was carried out for 10 minutes in an ice-water bath, and stirring was continued for 1 hour after addition of pyridine-3-sulfonyl chloride (18mg, 0.1 mmol). The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (5.6mg, 13%).

1H NMR(400MHz,DMSO)δ=12.01(s,1H),9.86(s,1H),9.05(s,1H), 8.87(d,J=4.4Hz,1H),8.31(d,J=8.0Hz,1H),7.65(dd,J=7.8Hz,5.0, 2H),6.97(s,1H),6.78(s,1H),6.54(s,1H),4.33(s,2H),3.17(d,J=5.2 Hz,1H),2.14(s,3H),1.99(s,2H),1.76-1.56(m,4H),1.35-1.26(m,2H).

MS m/z(ESI):497.1[M+H]+.

Example 37

N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -8- (pyridin-2-ylsulfonyl) -8-azabicyclo [3.2.1] octan-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine

Preparation of N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -8- (pyridin-2-ylsulfonyl) -8-azabicyclo [3.2.1] octan-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine reference example 23.

1H NMR(400MHz,CD3OD:CDCl3,v/v=1:2)δ8.70(d,J=4.6Hz,1H),8.00 (dt,J=8.0,4.6Hz,2H),7.60(ddd,J=6.8,4.8,1.8Hz,1H),7.35(d,J=6.0Hz,1H), 6.92(d,J=6.0Hz,1H),6.19(s,1H),4.43(s,2H),4.40-4.32(m,1H),2.27(s,3H), 2.15(ddd,J=12.7,5.3,2.6Hz,2H),1.88-1.81(m,2H),1.80-1.70(m,2H),1.62(dd,J =8.6,4.7Hz,2H).

MS m/z(ESI):497.1[M+H]+.

Example 38

N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -9- (pyridin-2-ylsulfonyl) -9-azabicyclo [3.3.1] nonan-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.213mmol) was dispersed in 4M HCl 1, 4-epoxyhexacyclic (15mL), the reaction was stirred at room temperature for 60 minutes, the solvent was removed by concentration under reduced pressure, the residual solid was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 deg.C, DIPEA (1.05mL, 6.39mmol) and pyridine-2-sulfonyl chloride (40mg, 0.224mmol) were added in that order, and the reaction mixture was stirred at 0 deg.C for 2.5 hours. The solvent was removed by concentration under reduced pressure and the residue was isolated by prep-HPLC to give the title compound as a white solid (12.4mg, 25%).

1H NMR(400MHz,DMSO-d6)δ12.07(s,1H),9.85(s,1H),8.78(d,J=4.0Hz, 1H),8.08(td,J=7.7,1.4Hz,1H),7.96(d,J=7.8Hz,1H),7.67(dd,J=6.7,4.7Hz, 2H),6.95(s,1H),6.59(d,J=30.3Hz,2H),4.85-4.71(m,1H),4.18(s,2H),2.17(s, 3H),2.05(dd,J=12.8,4.9Hz,3H),1.68(d,J=2.6Hz,7H).

MS m/z(ESI):511.1[M+H]+.

Example 39

N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -9- (pyridin-3-ylsulfonyl) -9-azabicyclo [3.3.1] nonan-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine

Preparation of N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -9- (pyridin-3-ylsulfonyl) -9-azabicyclo [3.3.1] nonan-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine reference example 38.

1H NMR(400MHz,CD3OD:CDCl3,v/v=1:1)δ9.11(s,1H),8.84(d,J=3.9Hz, 1H),8.29(d,J=8.2Hz,1H),7.70-7.62(m,1H),7.39(d,J=5.9Hz,1H),6.96(d,J=5.8Hz,1H),6.62(s,1H),5.05-4.90(m,1H),4.34(d,J=2.8Hz,2H),2.55-2.19(m, 5H),2.19-1.61(m,8H).

MS m/z(ESI):511.1[M+H]+.

Example 40

N2- ((3-exo) -9- ((1-methyl-1H-imidazol-2-yl) sulfonyl) -9-azabicyclo [3.3.1] nonan-3-yl) -N4- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine

Preparation of N2- ((3-exo) -9- ((1-methyl-1H-imidazol-2-yl) sulfonyl) -9-azabicyclo [3.3.1] nonan-3-yl) -N4- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidine-2, 4-diamine reference example 38.

1H NMR(400MHz,DMSO-d6)δ12.05(s,1H),9.86(s,1H),7.67(d,J= 2.9Hz,1H),7.45(s,1H),7.08(s,1H),6.96(d,J=4.9Hz,1H),6.73-6.47(m, 2H),4.88-4.74(m,1H),4.12(s,2H),3.87(s,3H),2.19(s,3H),2.09(ddd, J=5.5,5.1,1.0Hz,3H),1.91-1.58(m,7H).

MS m/z(ESI):514.1[M+H]+.

EXAMPLE 41

N, N-dimethyl-2- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) acetamide

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then N, N-dimethylformamide (5mL) was added to dissolve it, DIPEA (108mg, 0.84mmol) was added slowly dropwise, stirring was carried out for 10 minutes in an ice-water bath, and stirring was continued for 1 hour after the addition of 2-bromo-N, N-dimethylacetamide (38mg, 0.23 mmol). The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (16.6mg, 17%).

1H NMR(400MHz,DMSO)δ=12.00(s,1H),9.80(s,1H),7.63(s,1H), 6.74(d,J=128.0Hz,3H),4.62(s,1H),3.42(s,2H),3.04(s,3H),2.86(s, 2H),2.77(s,3H),2.16(s,3H),1.96-1.47(m,10H).

MS m/z(ESI):455.2[M+H]+.

Example 42

N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -9- (pyridin-2-ylmethyl) -9-azabicyclo [3.3.1]Nonan-3-yl) thieno [2,3-d]Pyrimidine-2, 4-diamines

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then, this was dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, and the mixture was stirred for 10 minutes in an ice-water bath, and after adding 2- (chloromethyl) pyridine hydrochloride (38mg, 0.23mmol), the mixture was heated to 70 ℃ and stirred overnight. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (20.8mg, 22%).

1H NMR(400MHz,DMSO-d6)δ=12.06(s,1H),9.85(s,1H),8.47(d,J =4.0Hz,1H),7.81-7.48(m,3H),7.33-6.52(m,4H),4.74(s,1H),3.92(s, 2H),2.89(s,2H),2.23(d,J=13.6Hz,3H),2.08-1.50(m,10H).

MS m/z(ESI):461.1[M+H]+.

Example 43

N2- ((3-exo) -9- ((1-methyl-1-hydro-imidazol-2-yl) methyl) -9-azabicyclo [3.3.1]Nonan-3-yl) -N4- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d]Pyrimidine-2, 4-diamines

N2- ((3-exo) -9- ((1-methyl-1-hydro-imidazol-2-yl) methyl) -9-azabicyclo [3.3.1]Nonan-3-yl) -N4- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d]Preparation of pyrimidine-2, 4-diamine reference example 42.

1H NMR(400MHz,DMSO-d6)δ=12.06(s,1H),9.84(s,1H),7.67(s,1H),7.08(s,1H),6.95(s,1H),6.74-6.55(m,3H),4.69(s,1H),3.91(s,2H),3.69 (s,3H),2.84(s,2H),2.20(s,3H),2.01-1.66(m,10H).

MS m/z(ESI):464.2[M+H]+.

Example 44

3- ((3-exo)) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of (3- ((2-chlorothieno [2,3-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol

2, 4-dichlorothieno [2,3-d ] pyrimidine (100mg, 0.49mmol), (3-amino-1H-pyrazol-5-yl) methanol (55mg, 0.49mmol), DIPEA (190mg, 1.47mmol) and the reaction mixture were added to N' N-dimethylformamide (2mL), and the reaction mixture was stirred at 70 ℃ overnight. Concentrated under reduced pressure, and the resulting crude product was isolated and purified by flash silica gel column chromatography to give the title compound as a yellow solid (100mg, 73%).

MS m/z(ESI):282.0[M+H]+.

The second step is that: preparation of tert-butyl (3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

(3- ((2-chlorothieno [2,3-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol (100mg,0.36 mmol), N-Boc-exo-3-aminotropane acetate (113mg, 0.40mmol) and DIPEA (140mg,1.08mmol) were added to N-butanol (2.5mL), the reaction mixture was uniformly mixed, and then reacted under microwave heating at 150 ℃ for 10 hours, cooled to room temperature, concentrated under reduced pressure, and the resulting crude product was isolated and purified by flash chromatography to give the title compound as a pale yellow solid (60mg, 35%).

MS m/z(ESI):472.0[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (60mg, 0.13mmol) was dissolved in methanol (10mL), followed by slowly adding dioxane hydrochloride (4N,2.5mL) to the reaction mixture, reacting at room temperature for 2 hours, concentrating under reduced pressure, adding a mixture of methanol (15mL), DIPEA (0.5mL) and acrylonitrile (1mL) to the resulting crude product, reacting at room temperature for 2 hours, concentrating under reduced pressure, and isolating and purifying by prep-HPLC to give the title compound as a white solid (11.6mg, 21%).

1H NMR(400MHz,CD3OD)δ7.39(dd,J=6.0Hz,1H),6.99(dd,J=5.6 Hz,1H),6.02-6.04(m,1H),4.60(s,2H),4.21-4.24(m,1H),3.45-3.42(m,2H), 2.83(s,2H),2.69-2.65(m,2H),2.08-1.91(m,6H),1.69(t,J=12.4Hz,2H).

MS m/z(ESI):425.1[M+H]+.

Example 45

3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference is made to example 44.

MS m/z(ESI):439.2[M+H]+.

Example 46

3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 44.

MS m/z(ESI):453.2[M+H]+.

Example 47

1- (((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- (((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile reference example 44.

MS m/z(ESI):530.2[M+H]+.

Example 48

3- ((3-exo) -3- ((6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of tert-butyl- (3-exo) -3- ((6-methyl-4- ((5-methyl-1-hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

To a solution of 2-chloro-6-methyl-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (200mg, 0.72mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate (194mg, 0.86mmol), DIPEA (186mg, 1.44mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After the reaction was completed, the reaction solution was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 98:2) to obtain the title compound as a pale yellow solid (124mg, 37%).

MS m/z(ESI):470.2[M+H]+.

The second step is that: preparation of 3- ((3-exo) -3- ((6-methyl-4- ((5-methyl-1-hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Dissolving tert-butyl- (3-exo) -3- ((6-methyl-4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (124mg, 0.26mmol) in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirring at room temperature for 30 minutes, and concentrating the reaction solution; methanol (10mL) was then added to dissolve it, DIPEA (137mg, 1.06mmol) was added slowly dropwise, stirring at room temperature for 10 minutes, acrylonitrile (21mg, 0.39mmol) was added and stirring continued for 2 hours. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (12.7mg, 12%).

1H NMR(400MHz,DMSO)δ=9.70(s,1H),7.30(s,1H),6.59(s,3H),4.15(s, 1H),3.29(s,2H),2.61(s,4H),2.39(s,3H),2.22(s,3H),1.90(s,2H),1.78-1.50(m, 6H).

MS m/z(ESI):423.2[M+H]+.

Example 49

3- ((3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

The first step is as follows: preparation of 5-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thiazolo [5,4-d ] pyrimidin-7-amine

To a solution of 5, 7-dichlorothiazolo [5,4-d ] pyrimidine (206mg,1mmol) in dimethylsulfoxide (10mL) was added 3-amino-5-methylpyrazole (116mg,1.2mmol), DIPEA (258mg,2mmol) in this order, followed by stirring at 70 ℃ for one hour. After completion of the reaction, water (50mL) was added to the reaction mixture to precipitate a solid, which was filtered and slurried with ethyl acetate to give the title compound as a yellow solid (200mg, 75%).

MS m/z(ESI):267.0[M+H]+.

The second step is that: preparation of tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

To a solution of 5-chloro-N- (5-methyl-1 h-pyrazol-3-yl) thiazolo [5,4-d ] pyrimidin-7-amine (200mg,0.75mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3-amino-9-azabicyclo [3.3.1] nonane-9-carboxylate (216mg, 0.9mmol), DIPEA (193mg,1.5mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After the reaction was completed, the reaction solution was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 95:5) to obtain the title compound as a pale yellow solid (232mg, 66%).

MS m/z(ESI):471.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Tert-butyl- (3-exo) -3- ((7- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thiazolo [5,4-d ] pyrimidin-5-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (232mg, 0.49mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; methanol (10mL) was then added to dissolve it, DIPEA (127mg, 0.98mmol) was added slowly dropwise, stirring at room temperature for 10 minutes, acrylonitrile (39mg, 0.74mmol) was added and stirring continued for 2 hours. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a pale yellow solid (63mg, 30%).

1H NMR(400MHz,DMSO)δ=12.06(s,1H),9.29(s,1H),8.76(d,J= 18.8Hz,1H),6.92(d,J=7.2Hz,1H),6.57(s,1H),4.67(s,1H),3.31(s, 2H),2.58(t,J=6.2Hz,4H),2.19(s,3H),2.00-1.65(m,10H).

MS m/z(ESI):424.2[M+H]+.

Example 50

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) oxo) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) oxo) -8-azabicyclo [3.2.1] octane-8-carboxylate

NaH (120mg, 3.01mmol, 60%) was added portionwise to a solution of tert-butyl (3-exo) -3-hydroxy-8-azabicyclo [3.2.1] octane-8-carboxylate (427mg, 1.88mmol) in N, N-dimethylformamide (2mL) at room temperature, stirred for 5 minutes at room temperature, then a solution of 2-chloro-N- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (100mg, 0.376mmol) in N, N-dimethylformamide (1mL) was added dropwise, and the mixture was heated to 120 ℃ under nitrogen and stirred for 2 hours. The reaction solution was cooled to room temperature, and then poured into ice water (10mL) and stirred for 10 minutes, followed by filtration, the filtrate was extracted with ethyl acetate, the organic phases were combined, washed with a saturated aqueous sodium chloride solution, the organic phase was collected, dried over anhydrous sodium sulfate, filtered, the organic solvent was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography to give the title compound as a yellow oil (149mg, 87%).

The second step is that: preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) oxo) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) oxo) -8-azabicyclo [3.2.1] octane-8-carboxylate (77mg, 0.169mmol) was dissolved in methanol (2mL), 4M HCl 1, 4-dioxane (2mL) was added thereto with stirring at room temperature, the reaction mixture was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (1mL), DIPEA (109mg, 0.844mmol) and acrylonitrile (45mg, 0.844mmol) were successively added thereto, and the resulting reaction mixture was further stirred at room temperature for 1 hour. After the reaction solution was concentrated under reduced pressure, the residue was preliminarily isolated and purified by silica gel chromatography and further isolated and purified by preparative TLC to give the title compound as a gray solid (7mg, 10%).

1H NMR(400MHz,CD3OD)δ7.50(d,J=6.1Hz,1H),7.22(d,J=5.9Hz, 1H),6.51(s,1H),5.43-5.26(m,1H),3.44-3.37(m,2H),2.78(t,J=6.9Hz, 2H),2.62(t,J=6.9Hz,2H),2.33(s,3H),2.12-2.00(m,4H),1.86-1.74(m, 4H).

MS m/z(ESI):410.1[M+H]+.

Example 51

3- ((3-exo) -3- ((6- (methoxymethyl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((6- (methoxymethyl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 48.

MS m/z(ESI):453.2[M+H]+.

Example 52

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6-morpholinothieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6-morpholinothieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 48.

MS m/z(ESI):494.2[M+H]+.

Example 53

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6- (morpholinomethyl) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6- (morpholinomethyl) thieno [2,3-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 48.

MS m/z(ESI):508.3[M+H]+.

Example 54

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6- ((4-methylpiperazin-1-yl) methyl) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6- ((4-methylpiperazin-1-yl) methyl) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 48.

MS m/z(ESI):535.3[M+H]+.

Example 55

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6- (pyridin-3-ylthio) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -6- (pyridin-3-ylthio) thieno [2,3-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 48.

MS m/z(ESI):532.2[M+H]+.

Example 56

3- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.22mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then it was dissolved in N, N-dimethylformamide (10mL), DIPEA (108mg, 0.84mmol) was added slowly dropwise, stirred at room temperature for 10 minutes, and after addition of 3-nitriloazetidine-1-sulfonyl chloride (45mg, 0.25mmol), stirring at room temperature was continued overnight. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (23.2 mg, 21%).

1H NMR(400MHz,DMSO-d6)δ=12.07(s,1H),9.74(s,1H),7.90(s,1H), 7.00(s,1H),6.54(s,2H),4.27(s,1H),4.13(s,2H),4.04(t,J=8.4Hz,2H), 3.98-3.89(m,2H),3.80(dd,J=15.2,6.0Hz,1H),2.23(s,3H),1.99(s,4H), 1.84(d,J=7.2Hz,2H),1.63(s,2H).

MS m/z(ESI):500.1[M+H]+.

Example 57

1- (((3-exo) -3- ((6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- (((3-exo) -3- ((6-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile reference example 38.

1H NMR(400MHz,DMSO-d6)δ12.02(s,1H),9.64(s,1H),6.77-6.45(m, 3H),4.25-4.23(m,1H),4.12(s,2H),4.06-4.02(m,2H),3.95-3.88(m,2H), 3.83-3.77(m,1H),2.24-2.21(m,4H),1.99-1.98(m,5H),1.84-1.81(m,2H), 1.64-1.59(m,3H).

MS m/z(ESI):513.1[M+H]+.

Example 58

2- (dimethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Preparation of 2- (dimethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one reference example 56.

1H NMR(400MHz,DMSO-d6)δ=12.06(s,1H),9.72(s,1H),7.89(s,1H), 6.99(s,1H),6.49(d,J=58.8Hz,2H),4.59-4.28(m,3H),3.04(s,2H),2.15 (s,9H),1.98-1.80(m,6H),1.59-1.45(m,2H).

MS m/z(ESI):441.1[M+H]+.

Example 59

1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2-morpholinoethane-1-one

The first step is as follows: preparation of tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

To a solution of 2-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [3,2-d ] pyrimidin-4-amine (250mg,0.94mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3-amino-9-azabicyclo [3.3.1] nonane-9-carboxylate (271mg, 1.13mmol), DIPEA (242mg,1.88mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After completion of the reaction, the reaction mixture was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 98:2) to obtain the title compound as a pale white solid (150mg, 34%).

MS m/z(ESI):470.1[M+H]+.

The second step is that: preparation of tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then 2- (7-benzotriazol oxide) -N, N' -tetramethyluronium hexafluorophosphate (120mg, 0.31mmol) was added and dissolved in N, N-dimethylformamide (5mL), DIPEA (108mg, 0.84mmol) was slowly added dropwise, stirred for 10 minutes in an ice-water bath, and 2-morpholinoacetic acid (33mg, 0.23mmol) was added followed by stirring for 1 hour. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (17.8mg, 17%).

1H NMR(400MHz,DMSO-d6)δ=11.99(s,1H),9.69(s,1H),7.84(s,1H), 7.07-6.23(m,3H),4.77(s,1H),4.58(s,1H),4.30(s,1H),3.52(d,J=4.0Hz, 4H),3.10-3.01(m,2H),2.32(s,3H),2.14(s,2H),2.09-1.39(m,10H).

MS m/z(ESI):497.1[M+H]+.

Example 60

1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (methylamino) ethan-1-one

Preparation of 1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) -2- (methylamino) ethan-1-one reference example 18.

MS m/z(ESI):441.2[M+H]+.

Example 61

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) (pyridin-2-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) (pyridin-2-yl) methanone reference example 59.

MS m/z(ESI):475.2[M+H]+.

Example 62

(1-methyl-1H-imidazol-2-yl) ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) methanone

Preparation of (1-methyl-1H-imidazol-2-yl) ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) methanone reference example 59.

MS m/z(ESI):478.2[M+H]+.

Example 63

2- (dimethylamino) -1- ((1R,3R,5S) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Preparation of 2- (dimethylamino) -1- ((1R,3R,5S) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one reference example 13.

MS m/z(ESI):441.2[M+H]+.

Example 64

N, N-dimethyl-2- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) acetamide

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then N, N-dimethylformamide (5mL) was added to dissolve it, DIPEA (108mg, 0.84mmol) was added slowly dropwise, stirring was carried out for 10 minutes in an ice-water bath, and stirring was continued for 1 hour after the addition of 2-bromo-N, N-dimethylacetamide (38mg, 0.23 mmol). The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (16.4mg, 17%).

1H NMR(400MHz,DMSO-d6)δ=12.05(s,1H),9.74(s,1H),7.90(d,J =4.0Hz,1H),7.14-6.30(m,3H),4.15(s,1H),3.32-3.23(m,4H),3.08(s, 3H),2.83(s,3H),2.22(s,3H),1.97(s,2H),1.82-1.55(m,6H).

MS m/z(ESI):441.1[M+H]+.

Example 65

3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) -6-methylthieno [3, 2-d-pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

The first step is as follows: preparation of (3- ((2-chloro-6-methylthiophen [3,2-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol

2, 4-dichloro-6-methylthioeno [3,2-d ] pyrimidine (200mg, 0.91mmol), (3-amino-1H-pyrazol-5-yl) methanol (120mg, 1.09mmol), and DIPEA (350mg, 2.73mmol) were dissolved in N, N-dimethylformamide (10mL), mixed uniformly, and reacted at 70 ℃ overnight. After cooling to room temperature, the reaction mixture was extracted with water (30mL) and ethyl acetate (20 mL. times.3), the organic phases were combined and concentrated under reduced pressure, and the crude product was isolated and purified by flash column chromatography on silica gel to give the title compound as a white solid (200mg, 75%).

MS m/z(ESI):296.0[M+H]+.

The second step is that: preparation of tert-butyl (3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) -6-methylthieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

Adding (3- ((2-chloro-6-methylthieno [3,2-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol (150mg, 0.51mmol), tert-butyl (3-exo) -3-amino-9-azabicyclo [3.3.1] nonane-9-carboxylate oxalate (200mg, 0.61mmol) and DIPEA (200mg, 1.53mmol) into n-butanol (3mL), uniformly mixing, reacting for 8 hours under the condition of microwave heating at 165 ℃, cooling to room temperature, concentrating the reaction solution under reduced pressure, and using the obtained crude product (200mg) as an impure product for the next reaction.

MS m/z(ESI):500.1[M+H]+.

The third step: preparation of (3- ((2- (((3-exo) -9-azabicyclo [3.3.1] nonan-3-yl) amino) -6-methylthio [3,2-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol

To a solution of tert-butyl (3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) -6-methylthieno [3,2-d ] pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (200mg, 0.40mmol) in methanol (10mL) was slowly added dropwise dioxane hydrochloride (4N, 5mL), the reaction solution was allowed to react at room temperature for 3 hours, concentrated under reduced pressure, and the resulting crude product was isolated and purified by prep-HPLC to give the title compound as a yellow solid (100mg, 63%).

MS m/z(ESI):400.1[M+H]+.

The fourth step: preparation of 3- ((3-exo) -3- ((4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) -6-methylthieno [3, 2-d-pyrimidin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

(3- ((2- (((3-exo) -9-azabicyclo [3.3.1] nonan-3-yl) amino) -6-methylthiophene [3,2-d ] pyrimidin-4-yl) amino) -1H-pyrazol-5-yl) methanol (100mg, 0.25mmol), acrylonitrile (0.2mL), and DIPEA (0.1mL) were added to methanol (10mL), and after uniform mixing, the mixture was reacted at room temperature for 1 hour, and concentrated under reduced pressure, and the resulting crude product was isolated and purified by prep-HPLC to give the title compound as a white solid (11.7mg, 10%).

1H NMR(400MHz,DMSO-d6)δ12.28(s,1H),10.12(s,1H),7.09-6.64(m, 2H),6.29-6.23(s,1H),5.22-4.94(m,1H),4.67-4.37(m,3H),2.95(s,2H), 2.85-2.81(m,2H),2.70-2.57(m,5H),1.95-1.49(m,10H).

MS m/z(ESI):453.2[M+H]+.

Example 66

3- (cis-5- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrol-2 (1H) -yl) propionitrile

The first step is as follows: preparation of tert-butyl cis-5- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate

2-chloro-N- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (100mg, 0.376mmol), tert-butyl cis-5-aminohexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (102mg, 0.452 mmol) and DIPEA (146mg, 1.13mmol) were added to NMP (1mL), and the mixture was heated to 160 ℃ with a microwave under nitrogen protection for 8 hours. The reaction was cooled to room temperature and then poured into ice water (10mL) and stirred for 10 min, filtered, the filter cake was washed with water (15mL) and dried in vacuo to give the title compound as a yellow solid (171mg, crude).

The second step is that: preparation of 3- (cis-5- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrol-2 (1H) -yl) propionitrile

Tert-butylcis-5- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (86mg, 0.188mmol) was dissolved in methanol (2mL), 4M HCl 1, 4-dioxane (2mL) was added with stirring at room temperature, the reaction was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (2mL), DIPEA (121mg, 0.938mmol) and acrylonitrile (15mg, 0.282mmol) were sequentially added, and the resulting reaction mixture was stirred at room temperature for further 16 hours. The reaction was diluted with DCM (20mL), washed with water (5mL), the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and the residue was isolated and purified by silica gel chromatography to give the title compound as a white solid (17mg, 22%).

1H NMR(400MHz,CD3OD)δ7.36(d,J=6.0Hz,1H),7.02-6.86(m,1H), 6.54(s,0.6H),5.80(s,0.4H),4.34-4.09(m,1H),2.86-2.72(m,4H),2.73-2.57 (m,4H),2.40-2.19(m,7H),1.57-1.37(m,2H).

MS m/z(ESI):409.1[M+H]+.

Example 67

3- (cis-5- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrol-2 (1 hydro) -yl) propionitrile

The first step is as follows: preparation of tert-butyl-cis-5- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1 hydro) -carboxylate

To a solution of 2-chloro-N- (5-methyl-1-hydro-pyrazol-3-yl) thieno [3,2-d ] pyrimidin-4-amine (100mg, 0.38mmol) in N-butanol (5mL) were added tert-butyl-cis-5-aminohexahydrocyclopenta [ c ] pyrrole-2 (1-hydro) -carboxylate (102mg, 0.45mmol), DIPEA (98mg, 0.76mmol) in this order, followed by stirring at 160 ℃ for 15 hours under microwave conditions. After the reaction was completed, the reaction solution was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 95:5) to obtain the title compound as a pale yellow solid (80mg, 46%).

MS m/z(ESI):456.2[M+H]+.

The second step is that: preparation of 3- (cis-5- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrol-2 (1 hydro) -yl) propionitrile

Dissolving tert-butyl-cis-5- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1 hydro) -carboxylate (80mg, 0.18mmol) in 1, 4-epoxy hexacyclic solution (4.0N,2mL), stirring at room temperature for 30 minutes, and concentrating the reaction solution; methanol (5mL) was then added to dissolve it, DIPEA (93mg, 0.72mmol) was added slowly dropwise, stirring at room temperature for 10 minutes, acrylonitrile (14mg, 0.27mmol) was added and stirring continued for 2 hours. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (26.3mg, 37%).

1H NMR(400MHz,DMSO-d6)δ=12.30(s,1H),9.92(s,1H),7.90(s,1H), 7.51-6.25(m,3H),4.11(s,1H),2.66(dd,J=13.6,7.2Hz,6H),2.22(s,8H), 1.31(s,3H).

MS m/z(ESI):409.1[M+H]+.

Example 68

3- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) propionitrile

The first step is as follows: preparation of tert-butyl 4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate

2-chloro-N- (5-methyl-1H-pyrazol-3-yl) thieno [2,3-d ] pyrimidin-4-amine (100mg, 0.376mmol), 1-BOC-4-aminopiperidine (108mg, 0.539mmol) and DIPEA (146mg, 1.13mmol) were added to NMP (1mL), and the mixture was heated to 130 ℃ by microwave heating under nitrogen protection for 16 hours. The reaction was cooled to room temperature and then poured into ice water (10mL) and stirred for 10 min, filtered, the filter cake was washed with water (5mL) and dried in vacuo to give the title compound as a yellow solid (100mg, crude).

The second step is that: preparation of 3- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) propionitrile

Tert-butyl 4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (100mg, 0.233mmol) was dissolved in methanol (2mL), 4M HCl in ethyl acetate (2mL) was added with stirring at room temperature, the reaction mixture was stirred at room temperature for 2 hours, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (2mL), DIPEA (150mg, 1.17mmol) and acrylonitrile (62mg, 1.17mmol) were sequentially added, and the resulting reaction mixture was further stirred at room temperature for 1 hour. The reaction was diluted with DCM (20mL), washed with water (5mL), the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and the residue was isolated and purified by silica gel chromatography to give the title compound as a white solid (18mg, 20%).

1H NMR(400MHz,CD3OD)δ7.37(d,J=6.0Hz,1H),6.97(d,J=6.1Hz, 1H),6.55(s,0.5H),5.81(s,0.5H),3.92-3.74(m,1H),3.04-2.88(m,2H),2.81- 2.57(m,4H),2.44-2.15(m,5H),2.14-1.97(m,2H),1.73-1.52(m,2H).

MS m/z(ESI):383.1[M+H]+.

Example 69

1- ((4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- ((4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) sulfonyl) azetidine-3-carbonitrile reference example 8.

1H NMR(400MHz,CD3OD)δ7.38(d,J=6.0Hz,1H),6.99(d,J=6.0Hz, 1H),6.49(s,0.5H),5.83(s,0.5H),4.20-4.10(m,2H),4.07-3.99(m,2H),3.99- 3.89(m,1H),3.77-3.61(m,3H),3.09-2.99(m,2H),2.28(s,3H),2.17-2.06(m, 2H),1.67-1.51(m,2H).

MS m/z(ESI):474.0[M+H]+.

Example 70

1- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) -2- (methylamino) ethan-1-one

Preparation of 1- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) -2- (methylamino) ethan-1-one reference example 18.

MS m/z(ESI):401.2[M+H]+.

Example 71

1- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) -2-morpholinoethan-1-one

Preparation of 1- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) -2-morpholinoethan-1-one reference example 17.

MS m/z(ESI):457.2[M+H]+.

Example 72

(4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) (pyridin-2-yl) methanone

Preparation of (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) piperidin-1-yl) (pyridin-2-yl) methanone reference example 31.

MS m/z(ESI):435.2[M+H]+.

Example 73

3- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) propionitrile

Preparation of 3- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) propionitrile reference example 1.

MS m/z(ESI):383.2[M+H]+.

Example 74

1- ((4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- ((4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) sulfonyl) azetidine-3-carbonitrile reference example 1.

MS m/z(ESI):474.1[M+H]+.

Example 75

1- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) -2-morpholinoethan-1-one

Preparation of 1- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) -2-morpholinoethan-1-one reference example 17.

MS m/z(ESI):457.2[M+H]+.

Example 76

(4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) (pyridin-2-yl) methanone

Preparation of (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) piperidin-1-yl) (pyridin-2-yl) methanone reference example 31.

MS m/z(ESI):435.2[M+H]+.

Example 77

3- (endo-6- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -3-azabicyclo [3.1.0] hex-3-yl) propionitrile

Preparation of 3- (endo-6- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [2,3-d ] pyrimidin-2-yl) amino) -3-azabicyclo [3.1.0] hex-3-yl) propionitrile reference example 3.

MS m/z(ESI):381.2[M+H]+.

Example 78

3- (endo-6- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -3-azabicyclo [3.1.0] hex-3-yl) propionitrile

Preparation of 3- (endo-6- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) thieno [3,2-d ] pyrimidin-2-yl) amino) -3-azabicyclo [3.1.0] hex-3-yl) propionitrile reference example 1.

MS m/z(ESI):381.2[M+H]+.

Example 79

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 2-chloro-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine

2, 4-dichloroquinazoline (199mg,1.0mmol), 5-methyl-1H-pyrazol-3-amine (99mg,1.02mmol) and triethylamine (213mg,2.1mmol) were added to anhydrous ethanol (5mL), and the mixture was stirred at room temperature for 18 hours. The reaction mixture was concentrated under reduced pressure, and the obtained solid was suspended in water-ethanol (v \ v ═ 9:1,20mL), filtered, and the obtained solid was washed with petroleum ether and dried to obtain the title compound (240mg, 92%).

MS m/z(ESI):260.1,262.1[M+H]+.

The second step is that: tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

2-chloro-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine (40mg,0.154mmol) and tert-butyl (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate (70mg,0.308mmol) were added to N-butanol (3mL), and after stirring well at room temperature, the reaction was carried out at 150 ℃ for 4 hours with a microwave. The solvent was removed by concentration under reduced pressure, and the residue was purified by silica gel column chromatography to give the crude title compound (120mg) which was used directly in the next reaction.

MS m/z(ESI):450.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Crude tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (120mg,0.154mmol) was dissolved in methanol (3mL), 4M HCl in 1, 4-dioxane (10mL) was added with stirring at room temperature, the resulting reaction mixture was stirred at room temperature for 30 minutes, the solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous methanol (10mL), diisopropylethylamine (0.51mL,3.08mmol) and acrylonitrile (10mg,0.154mmol) were added sequentially with stirring at room temperature, the resulting reaction mixture was further stirred at room temperature for 2.5 hours, the solvent was removed by concentration under reduced pressure, the residue was separated and purified by silica gel column chromatography and reverse phase HPLC sequentially to give the title compound (6.0mg, 10%).

1H NMR(400MHz,CD3OD)δ8.04(d,J=8.1Hz,1H),7.58(t,J=7.5Hz,1H), 7.39(s,1H),7.16(t,J=7.5Hz,1H),6.62(s,1H),4.35(s,1H),3.37(s,2H),2.76(t,J =6.9Hz,2H),2.62(t,J=6.9Hz,2H),2.31(s,3H),2.16-1.74(m,6H),1.67(t,J=11.7 Hz,2H).

MS m/z(ESI):403.2[M+H]+.

Example 80

3- ((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 2-chloro-7-methoxy-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine

2, 4-dichloro-7-methoxyquinazoline (500mg, 2.18mmol), 5-methyl-1H-pyrazol-3-amine (223 mg, 2.29mmol) and DIPEA (592mg, 4.58mmol) were added to absolute ethanol (10mL), and the mixture was stirred at room temperature for 3 days. The reaction was filtered, and the filter cake was washed with acetonitrile (5mL) and dried in vacuo to give the title compound as a white solid (355mg, 56%).

MS m/z(ESI):290.1[M+H]+.

The second step is that: preparation of tert-butyl (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

The compounds 2-chloro-7-methoxy-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine (355mg, 1.23 mmol), tert-butyl (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate acetate (421mg, 1.47mmol) and DIPEA (475mg, 3.68mmol) were mixed in N-butanol (7mL), and the mixture was heated to 150 ℃ with a microwave and stirred for 4 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give the title compound as a white solid (259mg, 44%).

MS m/z(ESI):480.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (259mg, 0.540mmol) was dissolved in methanol (3mL), 4M HCl 1, 4-dioxane (4mL) was added with stirring at room temperature, the reaction was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (3mL), DIPEA (349mg, 2.70mmol) and acrylonitrile (43mg, 0.810mmol) were sequentially added, and the resulting reaction mixture was stirred at room temperature for 0.5 hour. The reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was isolated and purified by silica gel column chromatography to give the title compound as a white solid (76.8mg, 33%).

1H NMR(400MHz,Methanol-d4)δ8.12(s,1H),7.14-6.75(m,2H),6.50(s, 1H),4.50-4.21(m,1H),3.92(s,3H),3.41(s,2H),2.91-2.55(m,4H),2.34(s, 3H),2.14-1.50(m,7H),1.40-1.23(m,1H).

MS m/z(ESI):433.2[M+H]+.

Example 81

3- ((3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 7-bromo-2-chloro-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine

7-bromo-2, 4-dichloroquinazoline (3.36g, 12.1mmol), 5-methyl-1H-pyrazol-3-amine (1.29g, 13.3mmol) and TEA (2.57g, 25.4mmol) were each added to anhydrous ethanol (67mL), and the mixture was stirred at room temperature for 16 hours. The reaction was filtered, and the filter cake was washed with absolute ethanol (20mL) and dried in vacuo to give the title compound as a white solid (4.17g, 100%).

The second step is that: preparation of tert-butyl (3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

The compounds 7-bromo-2-chloro-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine (500mg, 1.48 mmol), tert-butyl (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate acetate (465mg, 1.62mmol) and DIPEA (591mg, 4.58mmol) were mixed in NMP (5mL), and the mixture was heated by microwave to 130 ℃ and stirred for reaction for 4 hours. The reaction mixture was cooled to room temperature, poured into 25mL of ice water, and stirred for 30 minutes. The mixture was filtered and the filter cake was washed with acetonitrile (2mL) and dried under reduced pressure to give the title compound as a grey solid (877mg, 100%).

The third step: preparation of 3- ((3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (220mg, 0.416mmol) was dissolved in methanol (2mL), 4M HCl 1, 4-dioxane (2mL) was added with stirring at room temperature, the reaction was stirred at room temperature for 2 hours, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (2mL), DIPEA (269mg, 2.08 mmol) and acrylonitrile (66mg, 1.25mmol) were sequentially added, and the resulting reaction mixture was stirred at room temperature for 1 hour. The reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was isolated and purified by silica gel column chromatography to give the title compound as a white solid (17.4mg, 9%).

1H NMR(400MHz,CD3OD)δ7.94(d,J=8.8Hz,1H),7.74-7.37(m,1H), 7.24(dd,J=8.9,2.0Hz,1H),6.59(s,0.8H),5.92(s,0.2H),4.51-4.12(m, 1H),3.42-3.35(m,2H),2.75(t,J=6.9Hz,2H),2.62(t,J=6.9Hz,2H),2.31 (s,3H),2.09-1.61(m,8H).

MS m/z(ESI):481.1[M+H]+.

Example 82

3- ((3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of 2, 7-dichloro-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine

2,4, 7-trichloroquinazoline (2.0g, 8.58mmol), 5-methyl-1H-pyrazol-3-amine (915mg, 9.42 mmol) and TEA (1.82g, 18.0mmol) were added to anhydrous ethanol (40mL), and the mixture was stirred at room temperature for 16 hours. The reaction was filtered, and the filter cake was washed with anhydrous ethanol (5mL) and dried in vacuo to give the title compound as a white solid (2.5g, 99%).

MS m/z(ESI):294.0[M+H]+.

The second step is that: preparation of tert-butyl (3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

The compounds 2, 7-dichloro-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine (500mg, 1.70mmol), tert-butyl (3-exo) -3-amino-8-azabicyclo [3.2.1] octane-8-carboxylate acetate (535mg, 1.87mmol) and DIPEA (681mg, 5.27mmol) were mixed in NMP (7mL), and the mixture was heated to 180 ℃ with a microwave and stirred for reaction for 2 hours. After the reaction solution was cooled to room temperature, it was added to ice water and stirred, and the precipitated solid was filtered. The filter cake was washed with water, dried in vacuo and then isolated and purified by silica gel column chromatography to give the title compound as a white solid (405mg, 49%).

MS m/z(ESI):484.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (405mg, 0.837mmol) was dissolved in methanol (4mL), 4M HCl 1, 4-dioxane (2.5mL) was added with stirring at room temperature, the reaction mixture was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (4mL), DIPEA (486mg, 3.77mmol) and acrylonitrile (53mg, 1.00mmol) were sequentially added, and the resulting reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was subjected to preliminary separation and purification by silica gel column chromatography and then separation and purification by prep-HPLC to give the title compound as a white solid (40mg, 11%).

1H NMR(400MHz,Methanol-d4)δ8.02(d,J=8.8Hz,1H),7.54-7.22(m, 1H),7.14-7.07(m,1H),6.71-6.49(m,0.6H),6.05-5.76(m,0.4H),4.44-4.17(m, 1H),3.40-3.35(m,2H),2.75(t,J=7.0Hz,2H),2.62(t,J=6.9Hz,2H),2.46- 2.12(m,3H),2.07-2.00(m,2H),1.96-1.75(m,4H),1.71-1.61(m,2H).

MS m/z(ESI):437.2[M+H]+.

Example 83

3- ((3-exo) -3- ((7-fluoro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7-fluoro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 82.

MS m/z(ESI):421.2[M+H]+.

Example 84

3- ((3-exo) -3- ((5-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((5-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

1H NMR(400MHz,DMSO-d6)δ12.24(s,1H),9.62(s,1H),7.45(t,J= 8.0Hz,1H),7.33-6.92(m,3H),6.92-6.55(m,1H),4.32-4.15(m,1H),3.33- 3.25(m,2H),2.68-2.56(m,4H),2.26(s,3H),2.00-1.85(m,2H),1.83-1.54(m, 6H).

MS m/z(ESI):437.2[M+H]+.

Example 85

3- ((3-exo) -3- ((8-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((8-methyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

1H NMR(400MHz,Methanol-d4)δ7.86(d,1H),7.44(d,J=7.1Hz,1H), 7.03(t,J=7.7Hz,1H),6.70-6.54(m,0.6H),5.96-5.84(m,0.4H),4.49-4.32(m, 1H),3.46-3.36(m,2H),2.75(t,J=6.9Hz,2H),2.62(t,J=6.9Hz,2H),2.48 (s,3H),2.38-2.17(m,3H),2.10-1.89(m,4H),1.88-1.77(m,2H),1.64(t,J =12.0Hz,2H).

MS m/z(ESI):417.2[M+H]+.

Example 86

3- ((3-exo) -3- ((8-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((8-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

1H NMR(400MHz,Methanol-d4)δ8.01(s,1H),7.69(d,J=7.6Hz,1H), 7.09(s,1H),6.83-5.77(m,1H),4.52-4.26(m,1H),3.57-3.36(m,2H),2.94- 2.71(m,2H),2.71-2.53(m,2H),2.32(s,3H),2.19-1.49(m,8H).

MS m/z(ESI):437.2[M+H]+.

Example 87

3- ((3-exo) -3- ((6-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((6-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

1H NMR(400MHz,Methanol-d4)δ8.15(d,J=2.3Hz,1H),7.54(d,J=8.8 Hz,1H),7.49-7.21(m,1H),6.72-6.46(m,0.6H),6.08-5.75(m,0.4H),4.46- 4.20(m,1H),3.41-3.36(m,2H),2.76(t,J=7.0Hz,2H),2.62(t,J=7.0Hz, 2H),2.42-2.22(m,3H),2.08-2.01(m,2H),1.97-1.77(m,4H),1.72-1.61(m, 2H).

MS m/z(ESI):437.2[M+H]+.

Example 87

3- ((3-exo) -3- ((6-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((6-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

1H NMR(400MHz,Methanol-d4)δ8.15(d,J=2.3Hz,1H),7.54(d,J=8.8 Hz,1H),7.49-7.21(m,1H),6.72-6.46(m,0.6H),6.08-5.75(m,0.4H),4.46- 4.20(m,1H),3.41-3.36(m,2H),2.76(t,J=7.0Hz,2H),2.62(t,J=7.0Hz, 2H),2.42-2.22(m,3H),2.08-2.01(m,2H),1.97-1.77(m,4H),1.72-1.61(m, 2H).

MS m/z(ESI):437.2[M+H]+.

Example 88

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

Reacting tert-butyl (3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1]Octane-8-carboxylate (400mg, 0.758mmol), 3-pyridineboronic acid (187mg, 1.52mmol), Pd (dppf) Cl2(110mg, 0.152mmol), cesium carbonate (740g, 2.27mmol) were added to dioxane (8mL) respectively) And water (0.8mL) was added to the mixture, and the mixture was heated to 100 ℃ under nitrogen and stirred for 1 hour. After the reaction solution was concentrated, the residue was separated and purified by silica gel column chromatography to give the title compound as a pale yellow colloid (160mg, 40%).

The second step is that: preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (160mg, 0.302mmol) was dissolved in methanol (4mL), 4M HCl 1, 4-dioxane (4mL) was added with stirring at room temperature, the reaction mixture was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (2mL), DIPEA (195 mg, 1.51mmol) and acrylonitrile (48mg, 0.906mmol) were sequentially added, and the resulting reaction mixture was further stirred at room temperature for 2 hours. After the reaction, concentration under reduced pressure, preliminary separation and purification of the residue by silica gel column chromatography gave a gray solid, which was slurried with N, N-dimethylformamide/acetonitrile (2mL/4mL), the filtered solid was then slurried with N, N-dimethylformamide/acetonitrile (1.1mL/2.2mL), the solid was filtered off and dried under vacuum to give the title compound as a white solid (49mg, 34%).

1H NMR(400MHz,CD3OD)δ8.90(d,J=2.3Hz,1H),8.59(dd,J=4.9, 1.6Hz,1H),8.31-8.08(m,2H),7.84-7.40(m,3H),6.63(s,0.8H),5.94(s, 0.2H),4.49-4.26(m,1H),3.45-3.37(m,2H),2.77(t,J=6.9Hz,2H),2.63(t, J=6.9Hz,2H),2.34(s,3H),2.13-1.63(m,8H).

MS m/z(ESI):480.2[M+H]+.

Example 89

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-4-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-4-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 88.

1H NMR(400MHz,CD3OD)δ8.73-8.58(m,2H),8.21(d,J=8.5Hz,1H), 7.95-7.64(m,3H),7.59-7.49(m,1H),6.64(s,1H),4.49-4.22(m,1H),3.45- 3.35(m,2H),2.77(t,J=7.0Hz,2H),2.63(t,J=7.0Hz,2H),2.33(s,3H), 2.16-1.58(m,8H).

MS m/z(ESI):480.2[M+H]+.

Example 90

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-2-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7-fluoro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 88.

MS m/z(ESI):480.3[M+H]+.

Example 91

3- ((3-exo) -3- ((7- (5-methoxypyridin-3-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (5-methoxypyridin-3-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 88.

1H NMR(400MHz,CD3OD)δ8.54-8.41(m,1H),8.27(d,J=2.7Hz,1H), 8.17(d,J=8.5Hz,1H),7.88-7.56(m,2H),7.47(dd,J=8.5,1.8Hz,1H), 6.62(s,1H),4.49-4.25(m,1H),3.97(s,3H),3.44-3.37(m,2H),2.76(t,J=7.0Hz,2H),2.63(t,J=7.0Hz,2H),2.32(s,3H),2.10-1.64(m,8H).

MS m/z(ESI):510.2[M+H]+.

Example 92

3- ((3-exo) -3- ((7- (6-methoxypyridin-3-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (6-methoxypyridin-3-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 88.

MS m/z(ESI):510.3[M+H]+.

Example 93

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-phenylquinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-phenylquinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 88.

1H NMR(400MHz,DMSO-d6Small amount of CD3OD)δ8.47-8.23(m,1H),7.88-7.69 (m,2H),7.67-7.25(m,5H),6.92-6.62(m,0.8H),5.88(s,0.2H),4.41-4.20(m, 1H),3.58(s,2H),2.76-2.57(m,4H),2.38-2.11(m,3H),2.06-1.47(m,8H).

MS m/z(ESI):479.3[M+H]+.

Example 94

3- ((3-exo) -3- ((7- (1-cyclopropyl-1H-pyrazol-4-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of tert-butyl (3-exo) -3- ((7- (1-cyclopropyl-1H-pyrazol-4-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

Tert-butyl (3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.189mmol), (1-cyclopropyl-1H-pyrazol-4-yl) boronic acid (35mg, 0.227mmol), cesium carbonate (185mg, 0.567mmol), chlorine (2-dicyclohexylphosphino-2, 4, 6-triisopropyl-1, 1-biphenyl) [2- (2-amino-1, 1-biphenyl) ] palladium (II) (X-Phos Pd G2) (15mg, 0.0189mmol) were added to a mixed solvent of dioxane (2mL) and water (0.4mL), respectively, after three times of nitrogen replacement, the reaction mixture was heated to 100 ℃ and stirred for 2 hours. The reaction solution was cooled and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give the title compound as a brown oil (60mg, 57%).

The second step is that: preparation of 3- ((3-exo) -3- ((7- (1-cyclopropyl-1H-pyrazol-4-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((7- (1-cyclopropyl-1H-pyrazol-4-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (60mg, 0.108mmol) was dissolved in methanol (2mL), 4M HCl 1, 4-dioxane (2mL) was added with stirring at room temperature, and the reaction was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (1mL), DIPEA (70mg, 0.542mmol) and acrylonitrile (17mg, 0.324mmol) were sequentially added, and the resulting reaction mixture was further stirred at room temperature for reaction for 16 hours. The reaction was diluted with DCM (30mL), washed with water (10mL), concentrated under reduced pressure and the residue was initially isolated and purified by silica gel chromatography to give the title compound as a grey solid (20mg, 36%).

1H NMR(400MHz,CD3OD)δ8.20(s,1H),8.06(d,J=8.3Hz,1H), 7.93(s,1H),7.72-7.34(m,2H),6.60(s,1H),4.45-4.23(m,1H),3.78-3.67(m, 1H),3.43-3.36(m,2H),2.76(t,J=6.8Hz,2H),2.63(t,J=6.8Hz,2H),2.32 (s,3H),2.09-1.64(m,8H),1.24-1.00(m,4H).

MS m/z(ESI):509.2[M+H]+.

Example 95

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (1-methyl-1H-pyrazol-4-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (1-methyl-1H-pyrazol-4-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 94.

1H NMR(400MHz,CD3OD)δ8.09(s,1H),8.03(d,J=8.5Hz,1H),7.93 (s,1H),7.64-7.43(m,1H),7.38(dd,J=8.6,1.7Hz,1H),6.62(s,0.8H), 5.92(s,0.2H),4.42-4.28(m,1H),3.95(s,3H),3.42-3.36(m,2H),2.76(t,J =7.0Hz,2H),2.63(t,J=7.0Hz,2H),2.32(s,3H),2.08-2.01(m,2H),2.00- 1.80(m,4H),1.77-1.62(m,2H).

MS m/z(ESI):483.2[M+H]+.

Example 96

3- ((3-exo) -3- ((7- (1- (2-fluoroethyl) -1H-pyrazol-4-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (1- (2-fluoroethyl) -1H-pyrazol-4-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 94.

MS m/z(ESI):515.3[M+H]+.

Example 97

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (thiazol-4-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (thiazol-4-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 94.

MS m/z(ESI):486.2[M+H]+.

Example 98

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 88.

MS m/z(ESI):494.3[M+H]+.

Example 99

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (1-methyl-1H-pyrazol-4-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (1-methyl-1H-pyrazol-4-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 88.

MS m/z(ESI):497.3[M+H]+.

Example 100

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-morpholinoquinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

The first step is as follows: preparation of tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-morpholinoquinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

Reacting tert-butyl (3-exo) -3- ((7-bromo-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1]Octane-8-carboxylate (300mg, 0.568mmol), morpholine (494mg, 5.68 mmol), Pd2(dba)3(104mg, 0.114mmol), DavePhos (90mg, 0.227mmol), t-BuONa (109mg, 1.14mmol) were added to dioxane (6mL) respectively and stirred at 100 ℃ under nitrogen. The reaction solution was cooled to room temperatureAfter dilution with ethyl acetate (20mL), the mixture was washed with water (20mL) and saturated aqueous sodium chloride (10mL), the organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography to give the title compound as a pale yellow oil (66mg, 22%).

The second step is that: preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-morpholinoquinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-morpholinoquinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (66mg, 0.123mmol) was dissolved in methanol (2mL), 4M HCl 1, 4-dioxane (2mL) was added with stirring at room temperature, the reaction was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, the residue was redissolved in anhydrous methanol (1mL), DIPEA (80mg, 0.617mmol) and acrylonitrile (20mg, 0.369mmol) were sequentially added, and the resulting reaction mixture was stirred at room temperature for 2 hours. After the reaction solution was concentrated under reduced pressure, the residue was preliminarily isolated and purified by silica gel chromatography and further isolated and purified by preparative TLC to give the title compound as a gray solid (12mg, 20%).

1H NMR(400MHz,CD3OD)δ8.06(d,J=9.3Hz,1H),7.08(d,J=9.4Hz, 1H),6.68(s,1H),6.49(s,1H),4.46-4.28(m,1H),3.97-3.73(m,4H),3.52- 3.36(m,6H),2.73(t,J=6.7Hz,2H),2.62(t,J=6.7Hz,2H),2.34(s,3H), 2.11-1.61(m,8H).

MS m/z(ESI):488.2[M+H]+.

Example 101

3- ((3-exo) -3- ((7- (3-methoxyazetidin-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (3-methoxyazetidin-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):488.3[M+H]+.

Example 102

3- ((3-exo) -3- ((7- (4-methoxypiperidin-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (4-methoxypiperidin-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 100.

MS m/z(ESI):516.3[M+H]+.

Example 103

3- ((3-exo) -3- ((7- (4- (dimethylamino) piperidin-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (4- (dimethylamino) piperidin-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):529.3[M+H]+.

Example 104

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyrrolidin-1-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyrrolidin-1-yl) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):472.3[M+H]+.

Example 105

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (methylamino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (methylamino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):432.3[M+H]+.

Example 106

3- ((3-exo) -3- ((7- (methyl (oxetan-3-ylmethyl) amino) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (methyl (oxetan-3-ylmethyl) amino) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):502.3[M+H]+.

Example 107

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (((1-methylazetidin-3-yl) methyl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (((1-methylazetidin-3-yl) methyl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):501.3[M+H]+.

Example 108

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (((tetrahydro-2H-pyran-4-yl) methyl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (((tetrahydro-2H-pyran-4-yl) methyl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):516.3[M+H]+.

Example 109

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (((1-methylpiperidin-4-yl) methyl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (((1-methylpiperidin-4-yl) methyl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):529.3[M+H]+.

Example 110

3- ((3-exo) -3- ((7- (methyl (pyridin-3-ylmethyl) amino) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (methyl (pyridin-3-ylmethyl) amino) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 100.

MS m/z(ESI):523.3[M+H]+.

Example 111

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-morpholinoquinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7-morpholinoquinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 100.

MS m/z(ESI):502.3[M+H]+.

Example 112

3- ((3-exo) -3- ((7- (1H-imidazol-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (1H-imidazol-1-yl) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 100.

1H NMR(400MHz,CD3OD)δ8.29(s,1H),8.21(d,J=8.9Hz,1H), 7.70(s,1H),7.64-7.26(m,2H),7.19(s,1H),6.62(s,0.8H),5.93(s,0.2H), 4.47-4.22(m,1H),3.41-3.36(m,2H),2.76(t,J=6.9Hz,2H),2.63(t,J=6.9 Hz,2H),2.33(s,3H),2.08-2.01(m,2H),2.00-1.79(m,4H),1.74-1.63(m, 2H).

MS m/z(ESI):469.2[M+H]+.

Example 113

3- ((3-exo) -3- ((7- (2-methoxyethoxy) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7- (2-methoxyethoxy) -4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

MS m/z(ESI):477.3[M+H]+.

Example 114

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (oxetan-3-ylmethoxy) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (oxetan-3-ylmethoxy) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 80.

MS m/z(ESI):503.3[M+H]+.

Example 115

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- ((1-methylazetidin-3-yl) methoxy) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- ((1-methylazetidin-3-yl) methoxy) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 80.

MS m/z(ESI):502.3[M+H]+.

Example 116

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-ylmethoxy) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (pyridin-3-ylmethoxy) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 80.

MS m/z(ESI):510.3[M+H]+.

Example 117

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- ((1-methylazetidin-3-yl) oxo) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- ((1-methylazetidin-3-yl) oxo) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference is made to example 80.

MS m/z(ESI):488.3[M+H]+.

Example 118

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- ((1-methylpiperidin-4-yl) oxo) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- ((1-methylpiperidin-4-yl) oxo) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 80.

MS m/z(ESI):516.3[M+H]+.

Example 119

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (morpholinomethyl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (morpholinomethyl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 80.

MS m/z(ESI):516.3[M+H]+.

Example 120

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (1-methylazetidin-3-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (1-methylazetidin-3-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 80.

MS m/z(ESI):486.3[M+H]+.

Example 121

3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (oxetan-3-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) -7- (oxetan-3-yl) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile reference example 80.

MS m/z(ESI):473.3[M+H]+.

Example 122

1- (((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.222mmol) was dissolved in a 4M HCl solution of 1, 4-epoxyhexacyclic ring (10mL) and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (0.73mL, 4.44mmol) and 3-cyanoazetidine-1-sulfonyl chloride (44mg, 0.244mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 5 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (59.3mg, 54%).

1H NMR(400MHz,CD3OD)δ8.12(s,1H),7.50(s,1H),7.29(d,J=44.1 Hz,1H),7.06(s,1H),6.58(s,1H),4.36(s,1H),4.12(s,2H),4.03(t,J= 8.4Hz,2H),3.90(t,J=7.0Hz,2H),3.69-3.57(m,1H),2.20(s,3H),2.10- 1.51(m,8H).

MS m/z(ESI):494.2[M+H]+.

Example 123

1- (((3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (174mg, 0.36mmol) was dissolved in a solution of 4M HCl in 1, 4-epoxyhexacyclic ring (20mL), and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (1.19 mL, 7.2mmol) and 3-cyanoazetidine-1-sulfonyl chloride (78mg, 0.432mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 16.5 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (17.7mg, 9%).

1H NMR(400MHz,MeOD-d4)δ8.02(s,1H),7.42(s,1H),7.20(s,1H), 6.57(s,1H),4.51-4.40(m,1H),4.27(s,2H),4.17(t,J=8.5Hz,2H),4.13- 4.05(m,2H),3.64-3.53(m,1H),2.34(s,3H),2.16(s,4H),1.98(d,J=42.2 Hz,2H),1.76(t,J=11.9Hz,2H).

MS m/z(ESI):528.2[M+H]+.

Example 124

1- (((3-exo) -3- ((7-fluoro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- (((3-exo) -3- ((7-fluoro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile reference example 122.

1H NMR(400MHz,CD3OD)δ8.11(dd,J=9.1,6.1Hz,1H),7.21-6.82(m, 2H),6.56(s,0.8H),5.88(s,0.2H),4.58-4.34(m,1H),4.29-4.19(m,2H),4.17- 4.08(m,2H),4.06-3.96(m,2H),3.72-3.58(m,1H),2.31(s,3H),2.18-1.85(m, 6H),1.82-1.66(m,2H).

MS m/z(ESI):512.1[M+H]+.

Example 125

1- (((3-exo) -3- ((7-cyclopropyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- (((3-exo) -3- ((7-cyclopropyl-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile reference example 122.

1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.26-6.93(m,2H),6.50(s, 1H),4.58-4.37(m,1H),4.31-4.18(m,2H),4.18-4.07(m,2H),4.06-3.95(m, 2H),3.71-3.58(m,1H),2.32(s,3H),2.17-1.71(m,8H),1.34-1.25(m,1H), 1.19-1.03(m,2H),0.94-0.75(m,2H).

MS m/z(ESI):534.1[M+H]+.

Example 126

3- ((3-exo) -3- (((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

The first step is as follows: preparation of tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

To a solution of 2-chloro-N- (5-methyl-1 hydro-pyrazol-3-yl) quinazolin-4-amine (200mg, 0.77mmol) in N-butanol (10mL) were added tert-butyl- (3-exo) -3-amino-9-azabicyclo [3.3.1] nonane-9-carboxylate (222mg, 0.92mmol), DIPEA (199mg, 1.54mmol) in this order, followed by stirring at 170 ℃ for 4 hours under microwave conditions. After completion of the reaction, the reaction mixture was extracted with ethyl acetate (15mL × 3), washed with a saturated aqueous solution of sodium chloride (15mL × 3), the organic phase was collected and dried over anhydrous sodium sulfate, filtered, and the organic phase was concentrated under reduced pressure, and the obtained product was separated and purified by silica gel column chromatography (dichloromethane: methanol ═ 95:5) to obtain the title compound as a white solid (275mg, 77%).

MS m/z(ESI):464.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- (((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl- (3-exo) -3- ((4- ((5-methyl-1 hydro-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (100mg, 0.21mmol) was dissolved in 1, 4-epoxyhexacyclic solution (4.0N,5mL), stirred at room temperature for 30 minutes and then the reaction solution was concentrated; then it was dissolved in N, N-dimethylformamide (10mL), DIPEA (108mg, 0.84mmol) was added slowly dropwise, stirred at room temperature for 10 minutes, and after addition of 3-nitriloazetidine-1-sulfonyl chloride (45mg, 0.25mmol), stirring at room temperature was continued overnight. The reaction was concentrated under reduced pressure, and the resulting product was subjected to prep-HPLC to give the title compound as a white solid (30.5mg, 29%).

1H NMR(400MHz,DMSO)δ=12.09(s,1H),10.04(s,1H),8.24(s,1H), 7.45(s,1H),7.33-6.42(m,4H),4.79(s,1H),4.01-3.79(m,6H),3.74-3.67(m, 1H),2.15(s,3H),2.09-1.57(m,10H).

MS m/z(ESI):508.2[M+H]+.

Example 127

1- (((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

The first step of reaction: preparation of 2-chloro-7-methoxy-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine

2, 4-dichloro-7-methoxyquinazoline (497mg, 2.17mmol), 5-methyl-1H-pyrazol-3-amine (221 mg, 2.28mmol) and DIPEA (0.75mL, 4.56mmol) were added to anhydrous ethanol (10mL), stirred at room temperature for 24 hours, and then heated to 50 ℃ for reaction for 5 hours. The solvent was removed by concentration under reduced pressure, the residue was washed with a mixed solvent of ethanol-water (v/v ═ 1: 9, 20mL), and the residue was dried under reduced pressure to give the title compound (509mg, 81%).

MS m/z(ESI):290.0[M+H]+.

The second step of reaction: preparation of tert-butyl (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

2-chloro-7-methoxy-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine (150mg, 0.518 mmol), tert-butyl (3-exo) -3- (methylamino) -8-azabicyclo [3.2.1] octane-8-carboxylate (249mg, 1.036mmol), and DIPEA (0.43mL, 2.59mmol) were added to N-butanol (3mL), and heated to 170 ℃ with a microwave synthesizer for 6 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase column chromatography to give the title compound (193mg, 75%).

MS m/z(ESI):494.2[M+H]+.

The third step of reaction: preparation of 1- (((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octan-8-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (193mg, 0.39mmol) was dissolved in 4M HCl 1, 4-epoxyhexacyclic compound (20mL), the reaction was stirred at room temperature for 60 minutes, the solvent was removed by concentration under reduced pressure, the residual solid was dissolved in anhydrous N, N-dimethylformamide (10mL), the reaction mixture was cooled to 0 ℃ and DIPEA (1.93mL, 11.7mmol) and 3-cyanoazetidine-1-sulfonyl chloride (71mg, 0.39mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 4 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by prep-HPLC to give the title compound (97mg, 46%).

1H NMR(400MHz,DMSO-d6)δ12.11(s,1H),9.77(s,1H),8.23(d,J= 8.9Hz,1H),6.72(s,1H),6.67(d,J=8.8Hz,1H),6.48(s,1H),5.40-5.25(m, 1H),4.18(s,2H),4.06(t,J=8.6Hz,2H),4.01-3.92(m,2H),3.87-3.74(m, 4H),2.96(s,3H),2.23(s,3H),2.06-1.89(m,4H),1.83(d,J=5.8Hz,2H), 1.61(d,J=11.2Hz,2H).

MS m/z(ESI):538.2[M+H]+.

Example 128

1- (((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

The first step of reaction: tert-butyl (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate

2-chloro-7-methoxy-N- (5-methyl-1H-pyrazol-3-yl) quinazolin-4-amine (50mg, 0.173mmol) and tert-butyl (3-exo) -3-amino-9-azabicyclo [3.3.1] nonane-9-carboxylate oxalate (171mg, 0.518 mmol) were added to N-butanol (10mL), heated to 170 ℃ by a microwave synthesizer for 8 hours, concentrated under reduced pressure to remove the solvent, and the residue was separated by silica gel column chromatography to give the title compound (68mg, 80%).

MS m/z(ESI):494.2[M+H]+.

The second step of reaction: 1- (((3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) sulfonyl) azetidine-3-carbonitrile

Tert-butyl (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (68mg, 0.138mmol) was dissolved in 4M HCl in 1, 4-epoxyhexacyclic ring (15mL) and the reaction was stirred at room temperature for 60 minutes. The solvent was removed by concentration under reduced pressure, the residual solid was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (0.68mL, 4.14mmol) and 3-cyanoazetidine-1-sulfonyl chloride (25mg, 0.138mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 8 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by prep-HPLC to give the title compound (6.9mg, 9%).

1H NMR(400MHz,DMSO-d6)δ12.10(s,1H),9.83(s,1H),8.24(d,J= 7.3Hz,1H),6.65(dd,J=29.7,20.4Hz,4H),4.83(s,1H),4.02(t,J=8.5 Hz,2H),3.92(dd,J=14.9,8.4Hz,4H),3.87-3.73(m,4H),2.21(s,3H),2.04(d,J=4.3Hz,3H),1.92-1.68(m,7H).

MS m/z(ESI):538.2[M+H]+.

Example 129

Tert-butyl (3-exo) -3- ((7-chloro-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate was dispersed in dichloromethane (2mL), a 4M HCl solution of 1, 4-epoxyhexacyclic ring (20mL) was added, and the reaction mixture was stirred at room temperature for reaction for 1.5 hours. The solvent was removed by concentration under reduced pressure, and the residue was dried under reduced pressure on an oil pump for 10 minutes. The resulting crude product was dissolved in anhydrous N, N-dimethylformamide (8mL), cooled to 0 ℃ in an ice-water bath, and DIPEA (1.2mL, 7.1mmol), dimethylglycine (0.31mL, 4.72mmol) and HATU (118mg, 0.31mmol) were added sequentially with stirring, and the resulting reaction mixture was stirred at 0 ℃ for 60 minutes. The solvent was removed by concentration under reduced pressure and the crude product was isolated by prep-HPLC to give the title compound as a white solid (20.7mg, 21%).

1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.33(s,1H),7.31(s,1H), 7.17(s,1H),7.13-6.86(m,2H),6.60(s,1H),4.52(s,1H),4.42(d,J=3.4Hz, 2H),3.16(s,2H),2.38-2.12(m,9H),2.05-1.94(m,2H),1.93-1.73(m,4H), 1.63-1.46(m,2H).

MS m/z(ESI):469.1[M+H]+.

Example 130

2- (dimethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Preparation of 2- (dimethylamino) -1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one reference example 129.

1H NMR(400MHz,DMSO-d6)δ12.21(s,1H),10.13(s,1H),8.33(s,1H), 7.54(t,J=7.5Hz,1H),7.27(d,J=30.8Hz,1H),7.08(s,1H),6.78(s,1H), 6.61(s,1H),4.56(d,J=6.1Hz,1H),4.48(s,1H),4.32(d,J=5.3Hz,1H), 3.65(dd,J=32.9,14.8Hz,2H),2.53(s,6H),2.25(s,3H),1.94(ddd,J=36.8, 20.0,10.6Hz,6H),1.56(dd,J=19.2,9.5Hz,2H).

MS m/z(ESI):435.2[M+H]+.

Example 131

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) (pyridin-2-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) (pyridin-2-yl) methanone reference example 129.

1H NMR(400MHz,DMSO-d6)δ12.29(s,1H),10.80(s,1H),10.33(s, 1H),8.61(d,J=4.3Hz,1H),8.52-8.24(m,1H),7.96(td,J=7.8,1.7Hz, 1H),7.73(d,J=7.8Hz,1H),7.71-7.57(m,1H),7.52(ddd,J=7.5,4.9,1.0 Hz,1H),7.39(d,J=18.4Hz,1H),7.21(d,J=39.2Hz,1H),6.60(s,1H), 4.74(s,1H),4.69-4.44(m,2H),2.27(s,3H),2.12-1.69(m,8H).

MS m/z(ESI):455.2[M+H]+.

Example 132

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) (pyridin-3-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) (pyridin-3-yl) methanone reference example 129.

1H NMR(400MHz,DMSO-d6)δ12.25(s,1H),10.36(s,1H),10.05(s,1H),8.78-8.62(m,2H),8.34(d,J=29.6Hz,1H),7.90(d,J=7.0Hz,1H), 7.62-7.48(m,2H),7.31(dd,J=19.3,8.1Hz,1H),7.12(s,1H),6.60(s,1H), 4.68(d,J=4.8Hz,1H),4.53(d,J=9.2Hz,1H),4.02(d,J=3.1Hz,1H), 2.25(s,3H),2.16-1.48(m,8H).

MS m/z(ESI):455.2[M+H]+.

Example 133

((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) (pyridin-4-yl) methanone

Preparation of ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) (pyridin-4-yl) methanone reference example 129.

1H NMR(400MHz,DMSO-d6)δ12.23(s,1H),10.21(s,1H),8.70(d,J= 5.5Hz,2H),8.32(dd,J=28.3,8.1Hz,1H),7.59-7.51(m,1H),7.45(d,J= 1.8Hz,2H),7.36-7.22(m,1H),7.09(t,J=7.4Hz,1H),6.86(s,1H),6.59 (s,1H),4.67(d,J=4.9Hz,1H),4.61-4.44(m,1H),3.94(d,J=1.9Hz,1H), 2.24(s,3H),2.09-1.53(m,8H).

MS m/z(ESI):455.2[M+H]+.

Example 134

2, 2-difluoro-1- ((3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) ethan-1-one

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.222mmol) was dissolved in a 4M HCl solution of 1, 4-epoxyhexacyclic ring (10mL) and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃, DIPEA (0.73mL, 4.44mmol) was added, and after mixing well, a mixture of difluoroacetic acid (0.023mL, 0.233mmol) and HATU (169mg, 4.44mmol) (previously dissolved in 1mL of dry N, N-dimethylformamide and reacted for 10 minutes), and the reaction mixture was stirred at 0 ℃ for 1 hour. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (48.9mg, 52%).

1H NMR(400MHz,MeOD-d4)δ7.99(d,J=7.7Hz,1H),7.56-7.44(m, 1H),7.40-7.19(m,1H),7.13-7.03(m,1H),6.55(d,J=4.7Hz,1H),6.33(t, J=53.6Hz,1H),4.57(s,2H),4.46-4.40(m,1H),2.18(d,J=33.6Hz,3H), 2.09-1.75(m,6H),1.56(t,J=12.1Hz,2H).

MS m/z(ESI):428.1[M+H]+.

Example 135

N4- (5-methyl-1H-pyrazol-3-yl) -N2- ((3-exo) -8- (pyridin-3-ylsulfonyl) -8-azabicyclo [3.2.1] octan-3-yl) quinazoline-2, 4-diamine

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.222mmol) was dissolved in a 4M HCl solution of 1, 4-epoxyhexacyclic ring (10mL) and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (0.73mL, 4.44mmol) and 3-pyridinesulfonyl chloride hydrochloride (50mg, 0.233mmol) were added in this order, and the reaction mixture was stirred at 0 ℃ for 0.5 hour. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (20.5mg, 19%).

1H NMR(400MHz,DMSO-d6)δ8.98(d,J=2.0Hz,1H),8.76(dd,J=4.8, 1.4Hz,1H),8.27-8.19(m,1H),8.14-8.01(m,1H),7.60-7.44(m,2H),7.29(ddd, J=15.0,9.9,4.2Hz,1H),7.07(t,J=7.4Hz,1H),6.58-6.39(m,1H),4.30 (dd,J=6.0,2.6Hz,3H),2.17(s,3H),2.08-1.95(m,2H),1.74(dd,J=16.7, 6.2Hz,2H),1.64(dd,J=17.3,6.7Hz,2H),1.43-1.32(m,2H).

MS m/z(ESI):491.1[M+H]+.

Example 136

N2- ((3-exo) -8- ((2-methoxyethyl) sulfonyl) -8-azabicyclo [3.2.1] octan-3-yl) -N4- (5-methyl-1H-pyrazol-3-yl) quinazoline-2, 4-diamine

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.222mmol) was dissolved in a 4M HCl solution of 1, 4-epoxyhexacyclic ring (10mL) and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (10mL), cooled to 0 ℃ and DIPEA (0.73mL, 4.44mmol) and 2-methoxyethane-1-sulfonyl chloride (37mg, 0.233mmol) were added in that order, and the reaction mixture was stirred at 0 ℃ for 2 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (25.1mg, 43%).

1H NMR(400MHz,DMSO-d6)δ8.31(s,1H),7.64(s,1H),7.41(s,1H), 7.24(dd,J=23.8,8.5Hz,1H),6.61(s,1H),4.49-4.43(m,1H),4.26(s,2H), 3.76(t,J=6.2Hz,2H),3.43-3.29(m,5H),2.32(s,3H),2.11-1.86(m,6H), 1.74(t,J=13.5Hz,2H).

MS m/z(ESI):472.2[M+H]+.

Example 137

N2- ((3-exo) -8- (2-fluoroethyl) -8-azabicyclo [3.2.1] octan-3-yl) -N4- (5-methyl-1H-pyrazol-3-yl) quinazoline-2, 4-diamine

Tert-butyl (3-exo) -3- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (100mg, 0.222mmol) was dissolved in a 4M HCl solution of 1, 4-epoxyhexacyclic ring (10mL) and the reaction was stirred at room temperature for 30 minutes. The solvent was removed by concentration under reduced pressure, the residue was dissolved in anhydrous N, N-dimethylformamide (5mL), anhydrous potassium carbonate (184mg, 1.33mmol) and 1-bromo-2-fluoroethane (50mg, 0.233mmol) were added in this order, and the reaction mixture was stirred at 40 ℃ for 19 hours. The solvent was removed by concentration under reduced pressure, and the residue was isolated by reverse phase HPLC to give the title compound (27.3mg, 31%).

1H NMR(400MHz,DMSO-d6)δ8.15(d,J=17.8Hz,1H),7.56(s,1H), 7.35(d,J=44.5Hz,1H),7.11(s,1H),6.71(s,1H),4.64-4.45(m,2H),4.33- 4.19(m,1H),3.35(s,2H),2.91-2.68(m,2H),2.32(s,3H),2.11-1.56(m,8H).

MS m/z(ESI):396.2[M+H]+.

Example 138

3- ((3-exo) -3- ((7-chloro-4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

The first step is as follows: preparation of (3- ((2, 7-dichloroquinazolin-4-yl) amino) -1H-pyrazol-5-yl) methanol

2,4, 7-trichloroquinazoline (300mg, 1.29mmol), (3-amino-1H-pyrazol-5-yl) methanol (180 mg, 1.55mmol), and DIPEA (500mg, 3.87mmol) were added to 1, 4-dioxane (5mL), and the mixture was uniformly mixed and reacted at room temperature overnight. Concentrated under reduced pressure, the crude product was taken up in methanol (5mL), filtered and the solid dried to give the title compound as a white solid (350mg, 87%)

MS m/z(ESI):310.0[M+H]+.

The second step is that: preparation of tert-butyl (3-exo) -3- ((7-chloro-4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate

Adding (3- ((2, 7-dichloroquinazolin-4-yl) amino) -1H-pyrazol-5-yl) methanol (150mg, 0.49mmol), tert-butyl (3-exo) -3- (methylamino) -9-azabicyclo [3.3.1] nonane-9-carboxylate (150mg, 0.58 mmol) and DIPEA (190mg, 1.47mmol) into n-butanol (2mL), uniformly mixing, reacting at microwave 150 ℃ for 10 hours, cooling to room temperature, concentrating the reaction solution under reduced pressure, and separating and purifying the obtained crude product by using flash silica gel column chromatography to obtain a target product which is a white solid (140mg, 55%).

MS m/z(ESI):528.2[M+H]+.

The third step: preparation of 3- ((3-exo) -3- ((7-chloro-4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -9-azabicyclo [3.3.1] nonan-9-yl) propionitrile

To a solution of tert-butyl (3-exo) -3- ((7-chloro-4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylate (140mg, 0.27mmol) in methanol (10mL), dioxane hydrochloride (4N, 2mL) was slowly added dropwise, the reaction was carried out at room temperature for 2 hours, the reaction mixture was concentrated under reduced pressure, the resulting crude product was dissolved in methanol (15mL), DIPEA (0.5mL), acrylonitrile (25mg, 0.46mmol) and the reaction was carried out at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure and isolated and purified by prep-HPLC to give the title compound as a white solid (22mg, 20%).

1H NMR(400MHz,DMSO-d6)δ12.41(s,1H),10.02(s,1H),8.35(d,J= 8.4Hz,1H),7.28(s,1H),7.06(d,J=8.4Hz,1H),6.52-6.54(m,1H),5.53- 5.55(m,1H),5.25(s,1H),4.46(t,J=5.2Hz,2H),3.31-2.87(m,7H),2.66- 2.59(m,2H),2.08-1.87(m,5H),1.60-1.41(m,5H).

MS m/z(ESI):481.2[M+H]+.

Example 139

3- ((3-exo) -3- ((7-chloro-4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile

Preparation of 3- ((3-exo) -3- ((7-chloro-4- ((5- (hydroxymethyl) -1H-pyrazol-3-yl) amino) quinazolin-2-yl) (methyl) amino) -8-azabicyclo [3.2.1] octan-8-yl) propionitrile reference example 138.

H NMR(400MHz,DMSO-d6)δ12.45(s,1H),10.08(d,J=8.8Hz,1H),8.36(d,J=8.8Hz,1H),7.29(s,1H),7.07(d,J=8.8Hz,1H),6.62-6.54(m, 1H),5.27-5.11(m,2H),4.50(d,J=5.6Hz,2H),3.31-2.27(m,2H),2.94(d, J=16.0Hz,3H),2.67-2.58(m,4H),1.92-1.81(m,4H),1.71-1.62(m,2H), 1.39-1.23(m,2H).

MS m/z(ESI):467.2[M+H]+.

Example 140

3- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) piperidin-1-yl) propionitrile

Preparation of 3- (4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) piperidin-1-yl) propionitrile reference example 68.

1H NMR(400MHz,CD3OD:CDCl3,v/v=1:1)δ8.03(d,J=8.1Hz,1H), 7.59(d,J=7.9Hz,1H),7.44(s,1H),7.20(t,J=7.4Hz,1H),6.63(s,1H), 5.92(s,1H),4.01-3.87(m,1H),2.98(d,J=11.6Hz,2H),2.77(t,J=6.9Hz, 2H),2.64(t,J=6.9Hz,2H),2.45-2.22(m,5H),2.19-2.07(m,2H),1.65(td, J=14.0,3.4Hz,2H).

MS m/z(ESI):377.1[M+H]+.

Example 141

1- ((4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) piperidin-1-yl) sulfonyl) azetidine-3-carbonitrile

Preparation of 1- ((4- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) piperidin-1-yl) sulfonyl) azetidine-3-carbonitrile reference example 122.

1H NMR(400MHz,CD3OD:CDCl3,v/v=1:1)δ8.04(d,J=8.1Hz,1H), 7.65-7.59(m,1H),7.45(d,J=8.2Hz,1H),7.22(t,J=7.5Hz,1H),6.31(s, 1H),4.17(t,J=8.3Hz,2H),4.12-4.01(m,3H),3.74(d,J=12.7Hz,2H), 3.61(ddd,J=15.1,8.7,6.4Hz,1H),3.06(t,J=11.3Hz,2H),2.32(s,3H), 2.21-2.11(m,2H),1.64(td,J=13.6,3.3Hz,2H).

MS m/z(ESI):468.1[M+H]+.

Example 142

3- (endo-6- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -3-azabicyclo [3.1.0] hex-3-yl) propionitrile

Preparation of 3- (endo-6- ((4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -3-azabicyclo [3.1.0] hex-3-yl) propionitrile reference example 79.

MS m/z(ESI):375.2[M+H]+.

Second, evaluation of biological test

The present invention is further described and explained below in conjunction with test examples, which are not intended to limit the scope of the present invention.

Test example 1 measurement of inhibitory Effect of the Compound of the present invention on JAK kinase Activity

Purpose of the experiment: the purpose of this test example was to test the activity of compounds on inhibition of JAK kinase activity.

An experimental instrument: the centrifuge (5702R) is purchased from Eppendorf company, the pipettor is purchased from Eppendorf or Rainin company, and the microplate reader is purchased from BioTek company in the United states and is a SynergyH1 full-function microplate reader.

The experimental method comprises the following steps: the experiment adopts a fluorescence resonance energy transfer (TR-FRET) method to test the inhibition effect of the compound on the JAK kinase activity, and obtains the half inhibition concentration IC of the compound on the JAK kinase activity50

The specific experimental operations were as follows:

the kinase reaction was performed in white 384-well plates (PerkinElmer) with 1-5. mu.L of DMSO and ddH per well2O diluted compounds of different concentrations, 1-5. mu.L of the corresponding vehicle was added to the positive control wells, followed by 1-5. mu.L of kinase buffer (HEPES 50-250mM, MgCl) per well25-20mM, etc.) diluted 0.1-20 nM JAK kinase solution, adding 1-5 uL of kinase buffer solution into a negative control hole, adding 1-5 ul of substrate mixed solution containing polypeptide substrate and ATP, incubating at room temperature for 0.5-5 hours, adding 10ul of EDTA and test solution containing labeled antibodyAnd (3) incubating the test solution at room temperature for 1-24 hours, measuring fluorescence signal values of about 620nm and 665nm of each plate hole by using a BioTek Synergy H1 enzyme labeling instrument, and calculating the inhibition rate according to the fluorescence signal values. Obtaining the IC of the compound by curve fitting according to the inhibition rates of different concentrations50

The experimental data processing method comprises the following steps:

percent inhibition data {% inhibition 100- [ (test compound value-negative control value) for wells treated with compound was calculated by counting the percent inhibition data for wells treated with compound over positive control wells (DMSO control wells) and negative control wells (no kinase added) on the plate]V (positive control value-negative control value) × 100 }. IC was calculated using GraphPad prism to fit different concentrations and corresponding percent inhibition data to a 4-parameter nonlinear logistic formula50The value is obtained.

And (4) experimental conclusion:

the above scheme shows that the compounds of the examples shown in the present invention show the biological activities in the JAK1/2/3/TYK2 kinase activity assay as shown in table 19 below.

Watch 19

From the above table, it can be seen that: the compounds of the above examples can obviously inhibit the enzymatic activity of JAK1/2/3/TYK2 kinase, and some compounds show strong inhibition effect on JAK1/2/3/TYK2 kinase (NA indicates no detection).

Test example 2 measurement of inhibitory Effect of the Compound of the present invention on the JAK-STAT Signal pathway of cells

Purpose of the experiment:

the purpose of this test example was to test the activity of compounds on inhibition of the JAK-STAT signaling pathway in cells.

An experimental instrument:

microplate shaker (88880024) available from Thermo ScientificTMCompany(s)

Centrifuge (5702R) from Eppendorf

Pipettes were purchased from Eppendorf Inc

The microplate reader is purchased from BioTek company of America, and is a SynergyH1 full-function microplate reader.

The experimental method comprises the following steps:

the experiment adopts a U266 cell line, activates a JAK-STAT signal channel through INF-alpha stimulation, detects the inhibition activity of a compound on the phosphorylation of downstream STAT3 of the compound, and obtains the half inhibition concentration IC of the compound on the activity of the JAK-STAT signal channel50

The specific experimental operations were as follows:

3-12 μ L of U266 fine particles are spread in 384-well detection plates, the number of cells in each well is 100-300K, 2 μ L of the compound solution diluted in gradient is added, and the incubation is performed for 2 hours with shaking at 350rpm at room temperature. After 2 hours, 2. mu.L of INF-. alpha.was added to the solution at a final concentration of 1000U/mL, and the mixture was shaken at room temperature for 15 minutes. 2-5. mu.L (5X) of LANCE Ultra Lysis Buffer 2 solution was added thereto, and the mixture was shaken at room temperature for 2 hours. After 2 hours, 5. mu.L of a final concentration of 0.5nM LANCE Ultra Eu-labeled Anti-STAT3 antibody (PerkinElmer) and a final concentration of 5nM LANCE Ultra high-labeled Anti-STAT3 antibody (PerkinElmer) solution were added and incubated overnight at room temperature. Measuring 665nm fluorescence signal value of each plate hole by a microplate reader, calculating inhibition rate through the fluorescence signal value, and obtaining IC of the compound through curve fitting according to the inhibition rates of different concentrations50

The experimental data processing method comprises the following steps:

by positive control wells on the plate (DMSO control)Wells) and negative control wells (no cells) the percent inhibition data {% inhibition 100 {% inhibition for wells treated with compound (test compound value-negative control value)]V (positive control value-negative control value) × 100 }. IC was calculated using GraphPad prism to fit different concentrations and corresponding percent inhibition data to a 4-parameter nonlinear logistic formula50The value is obtained.

And (4) experimental conclusion:

the above scheme shows that the compound of the embodiment shown in the invention has the following biological activity on JAK-STAT signal pathway of U266 cells in the following table 20.

Watch 20

From the above table, it can be seen that: the compound of the above example has obvious inhibition effect on the JAK-STAT signal pathway activity of human myeloma cell U266.

Test 3, Balb/C mouse pharmacokinetic assay

1. The research aims are as follows:

compound example 1, example 8, example 9, example 15, example 17, example 18, example 28, example 31, example 33, example 34, example 38, example 48, example 49, example 59, example 67, example 68, example 80, example 81, example 82, example 88, example 100, example 122, and example 123 were studied using Balb/C mice as test animals, pharmacokinetic behavior in mice (plasma and colon, ileal tissue) after oral administration at a dose of 5mg/kg was analyzed for compounds with superior PK by analyzing drug concentrations in the colon and ileum, and colon/ileum drug concentration, colon/plasma drug concentration ratio for further studies.

2. Test protocol

2.1 test drugs:

examples 1, 8, 9, 15, 17, 18, 28, 31, 33, 34, 38, 48, 49, 59, 67, 68, 80, 81, 82, 88, 100, 122, and 123 of the present invention were prepared by oneself.

2.2 test animals:

Balb/C Mouse group was 12 per group, male, Shanghai Jitsie laboratory animals Ltd, animal production license number (SCXK 2013) 0006N 0.311620400001794).

2.3 administration:

Balb/C mice were 12 per group, male; p.o. after fasting overnight, the dose was 5mg/kg and the administration volume was 10 mL/kg.

2.4 sample collection:

mice were dosed with CO before and after dosing at 0, 0.5, 1,2, 3, 5 and 7 hours2Sacrifice, blood sampling 0.2mL of the heart, placing in EDTA-K2Centrifuging at 4 deg.C and 6000rpm for 6 min in a test tube to separate plasma, and storing at-80 deg.C; the ileum is taken near the cecum and is about 4-5cm long; the colon is also taken near the cecum end, about 2-3cm in length, taken out, weighed, placed in a 2mL centrifuge tube, and stored at-80 ℃.

2.5 sample treatment:

1) plasma samples 40uL added 160uL acetonitrile precipitation, after mixing 3500 x g centrifugal 5 ~ 20 minutes.

2) Plasma and intestinal homogenate samples 30. mu.L were precipitated by adding 90. mu.L acetonitrile containing internal standard (100ng/mL), mixed and centrifuged at 13000rpm for 8 minutes.

3) 70uL of the treated supernatant solution was added to 70uL of water, vortexed and mixed for 10 minutes, and then 20 uL of the supernatant solution was subjected to LC/MS/MS analysis for the concentration of the test compound, and the LC/MS/MS analyzer: AB Sciex API 4000 Qtrap.

2.6 liquid phase analysis

● liquid phase conditions: shimadzu LC-20AD pump

● column chromatography: agilent ZORBAX XDB-C18 (50X 2.1mm,3.5 μm) mobile phase:

the solution A is 0.1% formic acid water solution, and the solution B is acetonitrile

Flow rate: 0.4mL/min

Elution time: 0-4.0 min, eluent as follows:

3. test results and analysis

The main pharmacokinetic parameters were calculated using WinNonlin 6.1, and the results of the mouse pharmacokinetic experiments are shown in table 21:

TABLE 21

NA indicates no detection or no detection (limit of detection of blood concentration is 1ng/ml, C is detected in bloodmaxWhen the NA is NA, the NA in the blood detection index is not detected; when blood is detected CmaxWhen the content is higher than the limit of quantitation by 1ng/ml, the NA in the blood detection index is not detected; NA in tissues (colon and ileum) indicates not detected).

And (4) experimental conclusion:

as can be seen from the results of the mouse Pharmacokinetic (PK) experiments in the table: the compounds of the examples of the invention showed good exposure levels in the colon and ileum, area under the time curve of plasma drug concentration (AUC) and maximum plasma drug concentration (C)max) All reach the screening standard; and the colon/ileum drug concentration and colon/plasma drug concentration ratio of the compound is high, and good selectivity is shown.

Test 4, in vivo efficacy test procedure and results

4.1 purpose of experiment:

the compounds of the examples were evaluated for efficacy in a dss (dextran sulfate sodium) induced colitis model in C57BL/6 mice.

4.2. Experiment main material

4.2.1 instruments

1. Balance Mettler toledo AL104

2. Balance TP-602

4.2.2 reagents

1. Dextran Sodium Sulfate (DSS): MP Biomedicals, LLC, Solon, Ohio, cat No.:

160110

2. cyclosporine (CsA): norwalk, switzerland, batch number: S0033A

3. Sodium carboxymethylcellulose: chemical reagents of national drug group Co Ltd

4. Tween 80: sigma, cat No.: 8CBM 513V

4.2.3 Experimental animals

TABLE 22

Animal species and strains: C57BL/6
sex, age/body weight: female, 6-8 weeks old/18-20 g
The supplier: Shanghai Slac Experimental Animal Co.,Ltd.

4.3. experimental procedure

4.3.1 grouping

Animals were randomized into groups based on animal weight on day-1 using BioBook software to ensure similar weight values for each group of animals to reduce bias, and the grouping and dosing schedule are shown in the table below.

Grouping and dosing regimens

TABLE 23

a, the solvent is 0.5 percent of CMC-Na +1 percent of Tween 80

b, at intervals of 8 hours

4.3.2 Experimental procedures

1. Reagent preparation

Drinking water containing DSS: an appropriate amount of DSS powder was dissolved in autoclaved drinking water to prepare a 2% DSS solution.

2. Induction of enteritis

On day-1, animals were divided into 12 groups of 10 animals on average. (specific grouping scheme see Table 1)

Starting on day 0, 9:00 to day 6, 9:00, mice in groups 2 to 9 were given 2% DSS-containing aqueous solution for 6 days (from day 0 to day 6), after which the mice were given free water for 3 days (from day 6, 9:00 to day 9 before necropsy). The day of molding was counted as 0 day. The DSS aqueous solution is wrapped by tin foil paper to ensure light resistance. The DSS aqueous solution was replaced every 2 days.

Group 1 mice were free to drink normal water for 9 days (from day 0, 9:00 to day 9 before necropsy).

3. Administration of drugs

The specific dosages, routes of administration and times of administration are referenced in the table above.

4.4 measurement

1) Body weight

The frequency of recording was once a day.

2) Daily disease index (DAI)

The frequency of recording was once a day, rated on 4 scales according to the following criteria:

weight change (0, less than or equal to 1%; 1, 1-5%; 2, 5-10%; 3, 10-15%; 4, > 15%);

bloody stools (0, negative; 4, positive);

stool score (0, Normal; 2, loose stool; 4, diarrhea)

The daily disease index value (DAI) was obtained by dividing the sum of the scores of the above 3 sections by 3. The DAI-time (day) curve was plotted against the daily DAI score and the peak area under the curve (AUC) was calculated. The DAI AUC decrease ratio was calculated as the administration group compared with the Vehicle group, and the calculation formula was (DAI AUC)Administration set-DAI AUCVehicle) /DAI AUCVehicle×100%

4.5. The experimental results are as follows:

watch 24

4.6. Conclusion of the experiment

On a DSS-induced C57BL/6 mouse colitis model, the compounds of the above examples can obviously reduce the daily disease index (DAI) and have obvious drug effects.

III, research on crystal forms

1.1 Experimental instruments

1.1.1 some parameters of the physicochemical measuring instrument

TABLE 25

1.2 Instrument and conditions for liquid phase analysis

1.2.1 instruments and devices

Watch 26

Name of instrument Model number
Analytical balance Sartorius BSA224S-CW
Water purifier Milli-Q Plus,Millipore
High performance liquid chromatograph Agilent1260
Pump and method of operating the same Agilent G1311B
Sample injector G1329B
Column oven G1316A
Detector G1315D

1.2.2 chromatographic conditions

A chromatographic column: ZORBAX (SB-C8, 3.5 μm, 4.6X 75mm)

Flow rate: 1.5mL/min

Column temperature: 40 deg.C

Detection wavelength: 242nm

Sample introduction volume: 5.0. mu.L

Operating time: 15min

Diluent agent: ACN-Water (v/v, 1:1)

Mobile phase: a: water (0.05% trifluoroacetic acid); b: acetonitrile (0.05% trifluoroacetic acid)

Watch 27

T(min) A(%) B(%)
0.00 95 5
8.00 60 40
12.00 10 90
12.10 95 5
15.00 95 5

1. Preparation of different crystal forms

1.1 preparation of Compound III free base form A

Weighing 1000mg of hydrochloride of free base of compound III into a 40mL glass bottle, adding 7mL of methanol, heating and stirring at 50 ℃, adding 1M HCl hydrochloric acid solution to dissolve and clear, filtering, adding 1M NaOH solution until precipitation is separated out (about pH 10), stirring at room temperature overnight, filtering, washing with water, and drying the solid at 50 ℃ in vacuum to obtain the free base crystal form A. Upon detection and analysis, it has the following XRPD pattern as shown in fig. 1.

1.2 preparation of Compound III free base form B

Weighing 1000mg of hydrochloride of compound III free base into a 40mL glass bottle, adding 6.67mL of methanol, heating and stirring at 50 ℃, adding 1M HCl hydrochloric acid solution to dissolve and clear, filtering, adding 1M NaOH solution until precipitation is generated (pH is about 10), stirring at room temperature overnight, filtering, washing with water, and drying the solid at 50 ℃ in vacuum to obtain the free base crystal form B. Upon detection and analysis, it has the following XRPD pattern as shown in fig. 2.

1.3 preparation of Compound III free base form C

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of acetonitrile, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form C. By detection analysis, it has the following XRPD pattern as shown in figure 3, DSC pattern as shown in figure 4 and TGA pattern as shown in figure 5.

1.4 preparation of Compound III free base form D

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of ethyl acetate, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form D. By detection analysis, it has the following XRPD pattern as shown in figure 6, DSC pattern as shown in figure 7 and TGA pattern as shown in figure 8.

1.5 preparation of Compound III free base form E

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of ethanol, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form E. By detection analysis, it has the following XRPD pattern as shown in figure 9, DSC pattern as shown in figure 10 and TGA pattern as shown in figure 11.

1.6 preparation of Compound III free base form F

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of 3-pentanone, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form F. By detection analysis, it has the following XRPD pattern as shown in figure 12, DSC pattern as shown in figure 13 and TGA pattern as shown in figure 14.

1.7 preparation of Compound III free base form G

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of toluene, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form G. Upon detection analysis, it has the following XRPD pattern as shown in fig. 15.

1.8 preparation of Compound III free base form H

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of isopropanol, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form H. By detection analysis, it has the following XRPD pattern as shown in figure 16, DSC pattern as shown in figure 17 and TGA pattern as shown in figure 18.

1.9 preparation of Compound III free base form I

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of 2-butanol, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form I. By detection analysis, it has the following XRPD pattern as shown in figure 19, DSC pattern as shown in figure 20 and TGA pattern as shown in figure 21.

1.10 preparation of Compound III free base form J

Weighing 10mg of compound III free alkali crystal form B into a 2mL glass bottle, adding 200 μ l of 2-butanone, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free alkali crystal form J. By detection analysis, it has the following XRPD pattern as shown in figure 22, DSC pattern as shown in figure 23 and TGA pattern as shown in figure 24.

1.11 preparation of Compound III free base form K

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of 2-methyl-tetrahydrofuran, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form K. Upon detection analysis, it has the following XRPD pattern as shown in fig. 25.

1.12 preparation of Compound III free base form L

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ L of 1-propanol, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form L. By detection analysis, it has the following XRPD pattern as shown in figure 26, DSC pattern as shown in figure 27 and TGA pattern as shown in figure 28.

1.13 preparation of Compound III free base form M

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of isopropyl acetate, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form M. By detection analysis, it has the following XRPD pattern as shown in figure 29, DSC pattern as shown in figure 30 and TGA pattern as shown in figure 31.

1.14 preparation of Compound III free base crystalline form N

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of chlorobenzene, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form N. Upon detection analysis, it has the following XRPD pattern as shown in fig. 32.

1.15 preparation of Compound III free base form O

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of ethyl formate, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form O. By detection analysis, it has the following XRPD pattern as shown in figure 33, DSC pattern as shown in figure 34 and TGA pattern as shown in figure 35.

1.16 preparation of Compound III free base form P

Weighing 10mg of compound III free base crystal form B into a 2mL glass bottle, adding 200 μ l of methanol, pulping at 50 ℃ for 1 week, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form P. By detection analysis, it has the following XRPD pattern as shown in figure 36, DSC pattern as shown in figure 37 and TGA pattern as shown in figure 38.

1.17 preparation of Compound III free base form Q

Weighing 10mg of compound III free base crystal form A into a 2mL glass bottle, adding 200 μ l of methanol, pulping at 50 ℃ overnight, centrifuging, removing supernatant, and drying the residual solid in a 50 ℃ vacuum drying oven to constant weight to obtain free base crystal form Q. By detection analysis, it has the following XRPD pattern as shown in figure 39, DSC pattern as shown in figure 40 and TGA pattern as shown in figure 41.

1.18 preparation of Compound IV free base form A

Weighing 9.4g of (3-exo) -3- ((7-methoxy-4- ((5-methyl-1H-pyrazol-3-yl) amino) quinazolin-2-yl) amino) -8-azabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester, dissolving the tert-butyl ester in methanol (47mL), adding 4M HCl 1, 4-dioxane (94mL) at the temperature of 10-20 ℃ under stirring, reacting for 1-2 hours at the temperature of 20-30 ℃, filtering the reaction solution after the reaction is finished, washing a filter cake by using methanol (20mL), directly using the solid for the next reaction, dissolving the solid in methanol (56mL), dropwise adding DIPEA (10g, 0.0784mol) at the temperature of 10-20 ℃, adjusting the temperature of the reaction solution to be 20-30 ℃ after the dropwise adding is finished, and acrylonitrile (3.1g, 0.0588 mol) was added dropwise to the reaction solution, and reacted at a temperature of 20 to 30 ℃ for 2 hours. The reaction solution was filtered, and the filter cake was washed with 15mL of methanol and dried to give a white solid of the compound of formula IV (7.6 g). The detected and analyzed sample has the following XRPD pattern as shown in figure 42, DSC pattern as shown in figure 43 and TGA pattern as shown in figure 44.

2. Solid stability test

2.1 purpose of experiment:

and (3) investigating the physical and chemical stability of the compound III crystal form A, the compound III crystal form C, the compound III crystal form D, the compound III crystal form Q and the compound IV crystal form A under the conditions of 5000lx of illumination, high temperature of 60 ℃, high humidity of 92.5% RH and high temperature of 50 ℃ of 75% RH, and providing a basis for crystal form screening and compound storage.

2.2 protocol:

taking about 2mg of each of the compound III crystal form A, the compound III crystal form C, the compound III crystal form D, the compound III crystal form Q and the compound IV crystal form A, inspecting for 5 days and 10 days under the conditions of illumination of 5000lx, high temperature of 60 ℃, high humidity of 92.5 percent RH and high temperature of 50 ℃ and 75 percent RH, measuring the content by using an HPLC (high performance liquid chromatography) and external standard method, and calculating the change of related substances by using a chromatographic peak area normalization method.

2.3 Experimental results:

the results of physicochemical stability of different crystal forms of free alkali are as follows:

watch 28

The data show that the compound III crystal form C is relatively stable under the conditions of high temperature, high humidity and high temperature and high humidity.

3 hygroscopicity test

3.1 purpose of the experiment

And inspecting the hygroscopicity of the free alkali crystal form A of the compound III, the free alkali crystal form C of the compound III and the free alkali crystal form A of the compound IV under different relative humidity conditions, and providing a basis for screening and storing the crystal forms of the compounds.

3.2 Experimental protocol:

and (3) placing the crystal form A of the free base of the compound III, the crystal form C of the free base of the compound III and the crystal form A of the free base of the compound IV in saturated vapor with different relative humidity to ensure that the compound and the vapor reach dynamic balance, and calculating the percentage of moisture absorption weight gain of the compound after the balance.

3.3 Experimental results:

and (3) carrying out moisture absorption and moisture desorption circulation on the free alkali crystal form A of the compound III for 2 times under the condition of 0-95% relative humidity, wherein an XRPD spectrogram of the free alkali crystal form A is not changed, namely the crystal form is not transformed.

Compound III free base form C absorbs about 0.717% weight gain at RH 80% with a slight hygroscopicity. After the moisture absorption and moisture desorption cycle is carried out for 2 times under the condition of 0-95% relative humidity, the XRPD spectrogram of the free alkali crystal form C is not changed, namely the crystal form is not transformed.

Compound IV free base form a absorbs moisture and gains weight of about 0.5354% at RH 80% with a slight hygroscopic property. After the moisture absorption and moisture desorption cycle is carried out for 2 times under the condition of 0-95% relative humidity, the XRPD spectrogram of the free alkali crystal form A is not changed, namely the crystal form is not transformed.

4. Solubility experiments in different media

4.1 purpose of the experiment

The solubility of the compound III free base crystal form A and the compound IV free base crystal form A in media such as different pH media, artificial simulated gastric fluid (FaSSGF), fasting artificial simulated intestinal fluid (FaSSIF), non-fasting artificial simulated intestinal fluid (FeSSIF), fasting artificial simulated colon fluid (FaSSCoF), non-fasting artificial simulated colon fluid (FeSSCoF) and pure water is compared, and a basis is provided for the evaluation of druggability.

4.2 protocol:

suspending about 1mg of compound III free base form a and about 1mg of compound IV free base form a in different media for 6 hours, respectively, and determining thermodynamic solubility of the compound at 37 ℃ by HPLC, external standard method.

4.3 Experimental results: as shown in Table 29

Watch 29

From the above experimental results, the solubility of the crystal form satisfies the drug concentration required for local treatment.

5. Polycrystal screening and stable crystal form confirmation experiment

5.1 purpose of experiment:

and finding a relatively stable compound crystal form through polycrystal screening.

5.2 protocol:

selecting an organic solvent and water with certain solubility, suspending different crystal forms in a solvent system, stirring and pulping at room temperature for 1 week, centrifuging, removing a supernatant, drying the solid at 50 ℃ in vacuum (-0.1Mpa) overnight, and measuring XRPD, DSC and TGA of the solid for comparison.

5.3 Experimental results:

the method comprises the following steps of beating, changing a crystallization solvent, a crystallization mode and the like to obtain 17 crystal forms, namely a compound III crystal form A, a compound III crystal form B, a compound III crystal form C, a compound III crystal form D, a compound III crystal form E, a compound III crystal form F, a compound III crystal form G, a compound III crystal form H, a compound III crystal form I, a compound III crystal form J, a compound III crystal form K, a compound III crystal form L, a compound III crystal form M, a compound III crystal form N, a compound III crystal form O, a compound III crystal form P and a compound IV free base crystal form A. By comparing DSC spectra of different crystal forms, the thermodynamic stability of the compound III crystal form C and the compound III crystal form I can be judged to be better.

197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:一种C5格氏试剂的制备方法

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!