![]() six-membered heteroaryl ring pyrrole derivative, method of preparation thereof, and medical uses the
专利摘要:
PYRROL DERIVATIVES OF THE SIX-MEMBER HETEROARYLIC RING, ITS USES AND PREPARATION PROCESSES, AND PHARMACEUTICAL COMPOSITION.The present invention relates to a six-membered heteroaryl ring derivative of pyrrole, to the method of preparation thereof, to medical uses thereof. Specifically, the present invention relates to a novel six-membered heteroaryl ring derivative of pyrrole represented by the formula (I), the method of preparation thereof, a medical composition comprising the derivative, and a therapeutic agent using the same. , and, in particular, to uses such as inhibiting JAK and as an immunosuppressant. The substituents in formula (I) have the definitions given in the descriptive report. 公开号:BR112014014325A2 申请号:R112014014325-0 申请日:2012-12-19 公开日:2021-05-25 发明作者:Xuejun Zhang;Qing Dong;Bonian Liu;Yaoping Zhu;Xiaotao Li;Jiong Lan 申请人:Jiangsu Hengrui Medicine Co.,Ltd.;Shanghai Hengrui Pharmaceutical Co., Ltd.; IPC主号:
专利说明:
[001] [001] The present invention relates to a new six-membered heteroaryl ring type pyrrole derivative, the method of preparation thereof, a medical composition comprising the derivative thereof, and a therapeutic agent using the same, and , in particular, to pharmaceutical uses such as a JAK inhibitor and in the preparation of an immunosuppressant. Background of the Invention [002] [002] Many protein kinases constitute a large family of kinases, which control the transduction of various signals in cells by catalysis. Many diseases are related to abnormal intracellular responses induced by protein kinase regulation, including autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancers and cardiovascular diseases. [003] [003] Janus kinase (JAK) is a type of tyrosine kinases, including four members of JAK1, JAK2, JAK3 and TYK2. JAKs play an important role in signal transduction of a variety of cytokines. JAK1, JAK2 and TYK2 exist in abundance in various tissues and cells, whereas JAK3 is found mainly in lymphocytes. JAK3 can be specifically non-covalently bound to the common γ chain (Fcγ) of the cytokine receptor, whereas JAK1 is bound to a beta chain, both being activated by the cytokines IL-2, IL-4, IL-7 , IL-9, and IL-15. JAK2 plays an important role in the erythropoietin (EPO) signaling pathway, including promoting red blood cell differentiation and activation. [004] [004] Signal transducer and transcription activator (STAT) is a group of cytoplasmic proteins that can be linked to DNA in the regulatory region of the target gene. As downstream substrates of JAKs, STATs can be activated through phosphorylation of tyrosine itself under external signal stimulation, so they can be transferred to the nucleus and regulate gene transcription. [005] [005] The cytokine is bound to the associated receptor, resulting in receptor dimerization, the receptor-coupled JAKs are close to each other and are activated by phosphorylation of interacting tyrosine residues. Activated JAKs catalyze the phosphorylation of tyrosine residues of the receptor itself, thus forming the corresponding "docking sites" for binding STATs to the receptor complex. The SH2 domains of the STATs are linked to phosphotyrosine residues of the receptor, and phosphorylation of the C-terminal tyrosine residues is achieved by the role of the JAKs. Two phosphorylated STAT molecules interact with each other to form homologous/heterologous dimers, which leave the receptor molecules in the cell's nucleus, bind to promoter regions of the target gene, and regulate gene transcription and expression. [006] [006] Many abnormal immune responses, such as autoimmune diseases including allergies, asthma, transplant rejection (allogeneic), rheumatoid arthritis, amyotrophic lateral sclerosis and multiple sclerosis, myeloproliferative disorders, and hematologic malignancies including leukemia and lymphoma, their modulations are associated with the JAK/STAT signaling pathway. [007] [007] A deficiency in JAK3 is associated with a severe combined immunodeficiency immunodeficiency (SCID) phenotype in both rodents and humans. The JAK3-/- mammalian SCID phenotype and lymphoid cell-specific expression of JAK3 are two advantageous properties, which result in JAK3 being a target for an immune suppressor. T cells from mice deficient in JAK3 fail to respond to IL-2, and T cells from mice deficient in JAK1 show a weak response to IL-2. IL-2 plays a critical role in modulating T cells, for example, when the antibody is bound to the extracellular part of the IL-2 receptor, they can effectively prevent transplant rejection. [008] [008] Other animal studies have indicated that JAK3 not only plays a critical role in the maturation of B and T lymphocytes but is also essential to maintain T cell function. The modulation of immune activity through this new mechanism may be useful. for the treatment of T cell proliferative disorders such as transplant rejection and autoimmune diseases. [009] [009] JAK kinase inhibitors, particularly JAK3 kinase inhibitors could prevent T cell activation and prevent graft rejection after transplantation and could also provide therapeutic benefits for other autoimmune disorders. JAK3 is also involved in many biological processes, for example, the proliferation and survival of murine mast cells induced by IL-4 and IL-9 were shown to be dependent on JAK3 and gamma chain signaling (Suzuki et al. , 2000, Blood 96:2172-2180). JAK3 also plays an important role in IgE receptor-mediated mast cell degradation reactions, and JAK3 inhibition also leads to immunosuppression in transplant rejection. JAK3 plays an essential role in IgE receptor-mediated mast cell degradation responses (Malaviya et al., 1999, Biochem. Biophys. Res. Commun. 257:807-813), and it has also been shown to inhibit IgE JAK3 kinase prevents type I hypersensitivity, including anaphylaxis ( Malaviya et al., 1999, J. Biol. Chem. 274:27028-27038 ). It has also been shown that JAK3 inhibition re- [0010] [0010] For JAK2 kinase subtypes, JAK2 kinase - JAK2 V617F (mutation causes abnormal JAK2 kinase activity), a somatic cell gain-of-function mutation has been found in Philadelphia chromosome-negative myeloproliferative neoplasms (Ph) classic, which includes primary thrombocytosis, true polythemia, and primary myelofibrosis, so people have a strong interest in developing JAK2 vectored therapies for these diseases. Some studies have found that in patients suffering from bone marrow fibrosis, the JAK2 kinase mutation was produced in more than 50% of patients in vivo, and disease-related symptoms such as anemia, splenomegaly, and the risk of transformation into acute myeloid leukemia (AML) have been associated with increased activity and the overactive JAK-STAT signaling pathway resulting from the JAK2 gene mutation. Meanwhile, JAK2 activity was abnormally increased in a variety of solid tumors and hematologic tumors (glioblastoma, breast cancer, multiple myeloma, prostate cancer, AML, etc.). Therefore, the development of a selective JAK2 inhibitor for the therapy of myeloproliferative neoplasms and leukemia has great medical value and market potential (estimated to be in the billions of dollars). Recently, a selective JAK2 inhibitor called Ruxolitinib (INCB- [0011] [0011] A series of JAK inhibitors have already been disclosed by some patent applications, which include WO2001042246, WO2002000661, WO2009054941 and WO2011013785 etc. [0012] [0012] Although a number of JAK inhibitors having a function for immune diseases have already been disclosed, there remains a need to develop new compounds with better efficacy. After constant efforts, the present invention offers compounds of formula (I), and finds that compounds having such structure exhibit excellent effects and actions. Invention Summary [0013] [0013] The present invention is intended to provide a compound of formula (I) or pharmaceutically acceptable salts thereof, and tautomers, racemates, mesomers, racemates, enantiomers, and diastereomers thereof, and mixtures thereof, and a pharmaceutically acceptable salt thereof, as well as metabolites, metabolic precursors or prodrugs thereof. The present invention provides a compound of formula (I) having the following structure: 13 14 R R 1 5 R 7 R () N R s 12 [0014] [0014] A is CH or N; [0015] [0015] L is a bond or alkyl; [0016] [0016] R1 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)n C(O)OR15, - OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15, -NR16R17, - OC(O)NR16R17, -NHC(O)NR16R17 and -S(O)mR15, where alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of halogen, hydroxy, cyano, nitro, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)nC (O)OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15, -NR16R17, -OC(O)NR16R17, - NHC(O)NR16R17, -S(O)mR15, -NHC(O)(O)R15 and -NHS(O)mR15; [0017] [0017] R2 or R4 are each independently selected from the group consisting of hydrogen and alkylayl; [0018] [0018] R or R3 are each independently selected from the group consisting of hydrogen, halogen and alkylayl; [0019] [0019] R5 or R6 are each independently selected from the group consisting of hydrogen, alkyl and aryl, wherein the alkyl or aryl are each optionally substituted with one or more groups selected from the group which consists of alkyl and halogen; [0020] [0020] whether R7, R8, R9 or R10 are each independently selected from the group consisting of hydrogen, alkyl, hydroxyalkyl and halogen, or, R7 and R8 or R9 and R10 are taken together to form an oxo group; [0021] [0021] whether R11, R12, R13 or R14 are each independently selected from the group consisting of hydrogen, alkyl and halogen, or, R11 and R12 or R13 and R14 are taken together to form an oxo group; [0022] [0022] R15 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, aryl and heteroaryl, [0023] [0023] R16 or R17 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each one of them, optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, alkoxy, cycloalkyl, heterocyclyl, hydroxyalkyl, alkynyl, aryl, heteroaryl, carboxyl, alkoxycarbonyl and -OR18 ; [0024] [0024] R18 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, hydroxyalkyl, aryl and heteroaryl; [0025] [0025] R19 or R20 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; [0026] [0026] m is 0, 1 or 2; [0027] [0027] n is 0, 1 or 2; [0028] [0028] p is 0, 1 or 2; [0029] q is 0, 1 or 2; [0030] [0030] s is 0, 1 or 2; and [0031] t is 0, 1 or 2; [0032] [0032] In a preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, is selected from a compound of formula (II), or a pharmaceutically acceptable salt thereof: 5 1 7 RR [0033] [0033] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof is selected from a compound of formula (III), or a pharmaceutically acceptable salt thereof: 5 1 7 RR R N [0034] [0034] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, is selected from a compound of formula (IV-a) or (IV-b), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof: [0035] [0035] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, is selected from a compound of formula (Va) or (Vb), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof: 1 1 R R [0036] [0036] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, is selected from a compound of formula (VI-a) or (VI-b), or a pharmaceutically acceptable salt thereof: [0037] [0037] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, is selected from a compound of formula (VII-a) or (VII-b), or a pharmaceutically acceptable salt thereof: 5 R 5 [0038] [0038] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, where L is a bond or alkyl, preferably a bond; said alkyl is preferably -CH 2 - or -CH(CH 3 )-. [0039] [0039] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, where R1 is selected from the group consisting of [0040] [0040] R15 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, nitro, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)nC(O)OR18, -OC(O)R18 , -C(O)R18, -S(O)2R18, -NHC(O)(O)R18, -NHS(O)2R18 and -NR19R20; preferably, R11 is selected from the group consisting of alkyl and cycloalkyl, wherein said alkyl, cycloalkyl or heterocyclyl are each optionally substituted with one or more groups selected from the group consisting of alkyl, hydroxyl , cyano, amino, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)nC(O)OR18, -OC(O)R18, -C(O)R18, -S(O)2R18, - NHC (O)OR18, -NHS(O)2R18 and -NR19R20. [0041] [0041] R16 or R17 are each independently selected from the group consisting of hydrogen, alkyl and heteroaryl; wherein said heteroaryl is optionally substituted with one or more groups selected from the group consisting of alkoxy, cycloalkyl, hydroxyalkyl, alkynyl and -OR18; [0042] [0042] R18 is selected from the group consisting of hydrogen, alkyl and hydroxyalkyl; [0043] [0043] R19 or R20 are each independently selected from the group consisting of hydrogen and alkylayl; [0044] [0044] and n is 0, 1 or 2. [0045] [0045] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantio- [0046] [0046] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, wherein R7, R8, R9 or R10 are each independently selected from the group consisting of hydrogen, alkyl and hydroxyalkyl, preferably hydrogen, methyl or hydroxymethyl, more preferably hydrogen. [0047] [0047] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, where R11, R12, R13 or R14 are each independently hydrogen. [0048] [0048] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, where R11 and R12 or R13 and R14 are taken together to form an oxo group. [0049] [0049] In another preferred embodiment of the invention, the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, where A is N. [0050] The compounds of the present invention include all conformational isomers, for example cis- and trans-isomers; and all of its optical and stereoisomers and mixtures thereof. The compounds of the present invention have asymmetric centers, and therefore there are enantiomeric and diastereomeric isomers. [0051] [0051] A typical compound of the present invention, or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof includes, but is not limited to, the following: Example Structure and Name No. [0052] [0052] In another aspect, this invention provides a compound of formula (IB), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, used as an intermediate to prepare the compound of formula (I), where: 13 14 RR 1 5 R 7 R () NR s 12 8 R ( )p R 11 () R t 2 ( )q [0053] [0053] L is a bond or alkylayl; [0054] [0054] R1 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)n C(O)OR15, - OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15, -NR16R17, - OC(O)NR16R17, -NHC(O)NR16R17 and -S(O)mR15, where alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of halogen, hydroxy, cyano, nitro, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)nC (O)OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15, -NR16R17, -OC(O)NR16R17, - NHC(O)NR16R17, -S(O)mR15, -NHC(O)(O)R15 and -NHS(O)mR15; [0055] [0055] R2 is selected from the group consisting of hydrogen and alkylayl; [0056] [0056] R5 or R6 are each independently selected from the group consisting of hydrogen, alkyl and aryl, wherein the alkyl or aryl are each optionally substituted with one or more groups selected from the group which consists of alkyl and halogen; [0057] [0057] whether R7, R8, R9 or R10 are each independently selected from the group consisting of hydrogen, alkyl, hydroxyalkyl and halogen; or, R7 and R8 or R9 and R10 are taken together to form an oxo group; [0058] [0058] whether R11, R12, R13 or R14 are each independently selected from the group consisting of hydrogen, alkyl and halogen, or, R11 and R12 or R13 and R14 are taken together to form an oxo group; [0059] [0059] R15 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, nitro, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)nC(O)OR18, -OC(O)R18 , -C(O)R18, -C(O)NR19R20, -NHC(O)R18, -NR19R20, -OC(O)NR19R20, -NHC(O)NR19R20, -S(O)mR18, -NHC(O )OR18 and -NHS(O)mR18; [0060] [0060] R16 or R17 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each one of them, optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, alkoxy, cycloalkyl, heterocyclyl. [0061] [0061] R18 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, hydroxyalkyl, aryl and heteroaryl; [0062] [0062] R19 or R20 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; [0063] [0063] m is 0, 1 or 2; [0064] [0064] n is 0, 1 or 2; [0065] [0065] p is 0, 1 or 2; [0066] q is 0, 1 or 2; [0067] [0067] s is 0, 1 or 2; and [0068] t is 0, 1 or 2; [0069] [0069] In another aspect, this invention provides a process for preparing the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, comprising the steps of: 13 14 RR 1 5 R 7 R () NR s 12 ( )p 8 RR ( ) R11 13 14 t XRR 1 2 L ( )q 6 R 5 RRR 7 R () NNR 9 [0070] [0070] the alkaline condition is provided by an organic base and an inorganic base, wherein said organic base includes, but is not limited to, triethylamine, N,N-diisopropylethylamine, n-butyl lithium, tert. butyl potassium, tetrabutylammonium bromide, and said inorganic base include, but are not limited to, sodium hydride, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate or cesium carbonate; [0071] [0071] wherein X is halogen, A, L, R, R1 to R14, p, q, s and t have the definitions given for formula (I); preferably, R1 is tert-butoxycarbonyl. [0072] [0072] In another aspect, this invention provides a process for preparing the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, comprising the steps of: 13 14 RR 13 [0073] [0073] wherein: when R 1 is t-butoxycarbonyl, t-butoxycarbonyl is further optionally removed from the compound of formula (I) to obtain the compound of formula (IC) or a pharmaceutically acceptable salt thereof; [0074] [0074] the reaction solvent includes, but is not limited to: tetrahydrofuran, ethanol, methanol, n-butanol, dichloromethane, 1,4-dioxane or N,N-dimethylformamide; [0075] [0075] the alkaline condition is provided by an organic base and an inorganic base, wherein said organic base includes, but is not limited to, triethylamine, N,N-diisopropylethylamine, n-butyl lithium, tert. butyl potassium, and said inorganic base includes, but is not limited to, sodium hydride, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate or cesium carbonate; [0076] [0076] where A, L, R, R1 to R14, p, q, s and t have the definitions given for formula (I). [0077] [0077] The present invention also offers a pharmaceutical composition, comprising a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutical salt - ceutically acceptable of the same, together with a pharmaceutically acceptable vehicle or excipient. [0078] [0078] The present invention also relates to the use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or of the pharmaceutical composition comprising the same, in the preparation of a medicament for inhibiting JAK kinase; preferably to inhibit JAK1, JAK2 or JAK3. [0079] [0079] The present invention also relates to the use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or of the pharmaceutical composition comprising the same, in the preparation of a medicament for inhibiting JAK kinase; wherein the medicament optionally further contains one or more reagents for regulating the mammalian immune system, anti-cancer agents or anti-inflammatory agents. [0080] [0080] The present invention also relates to the use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt. [0081] [0081] The present invention also relates to the use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or of the pharmaceutical composition comprising the same, in the preparation of a medicament for inhibiting JAK kinase; wherein the medicament optionally further contains one or more reagents for regulating the immune system of mammals, anti-cancer agents or anti-inflammatory agents, wherein said medicament is used for the treatment or prevention of the following disorders or diseases: diseases of the immune system, including those such as organ transplant rejection (eg, allograft rejection and graft versus host disease); autoimmune diseases, including those such as lupus, multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriasis, colitis, Crohn's disease, autoimmune thyroid disease, etc.; skin diseases, including those such as psora, itching, atopic dermatitis, etc.; allergenicity disorders, including those such as asthma, rhinitis, etc.; viral diseases, including those such as hepatitis B, hepatitis C, varicella-zoster virus etc.; diabetes I and diabetes complications; Alzheimer's disease; dry eye; marrow fibrosis; thrombocytosis; polycythemia or leukemia; cancers, including for example, solid tumors (such as prostate cancer, kidney cancer, liver cancer, pancreas cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, breast cancer, thyroid, glioblastoma, melanoma, etc.), blood cancer (such as lymphoma, leukemia, etc.), skin cancer (such as cutaneous T-cell lymphoma, cutaneous B-cell lymphoma), etc. Said mammal is a human being. [0082] [0082] The present invention further relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or composition pharmaceutical comprising the same, for use as a medicine to inhibit JAK kinase. Said JAK kinase is preferably selected from the group consisting of JAK1, JAK2 and JAK3. [0083] [0083] The present invention further relates to the use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or of the composition pharmaceutical comprising the same, for use as a medicine combined further with one or more reagents for regulating the immune system of mammals, anti-cancer agents or anti-inflammatory agents. [0084] [0084] The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the even, for use as a medicine for the treatment or prevention of the following disorders or diseases: diseases of the immune system, including those such as organ transplant rejection (eg, allograft rejection and graft versus host diseases) ; autoimmune diseases, including those such as lupus, multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriasis, colitis, Crohn's disease, autoimmune thyroid disease, etc.; skin diseases, including those such as psora, itching, atopic dermatitis, etc.; allergenicity disorders, including those such as asthma, rhinitis, etc.; viral diseases, including those such as hepatitis B, hepatitis C, varicella-zoster virus etc.; diabetes I and diabetes complications; Alzheimer's disease; dry eye; marrow fibrosis; thrombocytosis; polycythemia or leukemia; cancers, including for example solid tumors (such as prostate cancer, kidney cancer, liver cancer, pancreatic cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, melanoma, etc.), blood cancer (such as lymphoma, leukemia, etc.), skin cancer (such as cutaneous T-cell lymphoma, cutaneous B-cell lymphoma) etc. [0085] [0085] The present invention also relates to the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the even, further combined with one or more reagents for regulating the immune system of mammals, anti-cancer agents or anti-inflammatory agents, for use as a medicine for the treatment or prevention of the following disorders or diseases: diseases of the immune system, including those such as organ transplant rejection (eg, allograft rejection and graft-versus-host disease); autoimmune diseases, including those such as lupus, multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriasis, colitis, Crohn's disease, autoimmune thyroid disease, etc.; skin diseases, including those such as psora, itching, atopic dermatitis, etc.; allergenicity disorders, including those such as asthma, rhinitis, etc.; viral diseases, including those such as hepatitis B, hepatitis C, varicella-zoster virus etc.; diabetes I and diabetes complications; Alzheimer's disease; dry eye; marrow fibrosis; thrombocytosis; polycythemia or leukemia; cancers, including for example, solid tumors (such as prostate cancer, kidney cancer, liver cancer, pancreatic cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, breast cancer. thyroid, glioblastoma, melanoma, etc.), blood cancer (such as lymphoma, leukemia, etc.), skin cancer (such as cutaneous T-cell lymphoma, cutaneous B-cell lymphoma) etc. [0086] [0086] In other words, the present invention relates to a method for inhibiting the JAK kinase, comprising the step of administering to the individual in need thereof a therapeutically effective amount of the compounds of formula (I), or tautomers, racemates, enantiomers, diastereoisomers, and mixtures thereof, and pharmaceutically acceptable salts thereof, or a pharmaceutical composition containing the same. Furthermore, compounds of formula (I) or tautomers, racemates, enantiomers, diastereoisomers, and mixtures thereof, and pharmaceutically acceptable salts thereof, or a pharmaceutical composition containing the same, can [0087] [0087] The present invention relates to a method for the treatment or prevention of disorders or diseases of the immune system, comprising the step of administering to the individual in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same; wherein said disorders or diseases of the immune system include those such as organ transplant rejection (e.g. allograft rejection and graft versus host diseases); autoimmune diseases, including those such as lupus, multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriasis, colitis, Crohn's disease, autoimmune thyroid disease, etc.; skin diseases, including those such as psora, pruritus, atopic dermatitis, etc.; allergenicity disorders, including those such as asthma, rhinitis, etc.; viral diseases, including those such as hepatitis B, hepatitis C, varicella-zoster virus etc.; diabetes I and diabetes complications; Alzheimer's disease; dry eye; marrow fibrosis; thrombocytosis; polycythemia or leukemia; cancers, including for example solid tumors (such as prostate cancer, kidney cancer, liver cancer, pancreatic cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, melanoma, etc.), blood cancer (such as lymphoma, leukemia, etc.), skin cancer (such as cutaneous T-cell lymphoma, cutaneous B-cell lymphoma) etc. [0088] [0088] The present invention also relates to a method for the treatment or prevention of disorders or diseases of the immune system. [0089] The compositions of this invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers. Accordingly, the active compounds of this invention may be formulated in various dosage forms for oral or buccal administration, for intranasal, parenteral (eg, intravenous, intramuscular or subcutaneous) or rectal administration, or for administration by inhalation or insufflation. . The compounds of this invention may also be formulated as systematic release dosage forms. [0090] [0090] For oral administration, pharmaceutical compositions, for example, may be formulated as tablets or capsules with pharmaceutically acceptable excipients by conventional means, which excipients include those such as binding agents (e.g. starch, gelatin , polyvinylpyrrolidone or acacia) and fillers (eg lactose, microcrystalline cellulose or calcium phosphate), lubricants (eg magnesium stearate, talc or silica), disintegrants (eg potato starch or starch glycolate sodium) or wetting agents (such as sodium lauryl sulfate). The tablets can be coated by methods well known in the literature. Liquid preparations for oral administration can be solutions, syrups or suspensions, or they can be a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (eg sorbitol syrup, methyl cellulose or hydrogenated edible fats), emulsifying agents (eg lecithin or acacia), non-aqueous vehicles (eg oil almonds, oily esters or ethanol) and preservatives (eg methyl or propyl p-hydroxy benzoate). [0091] [0091] For oral administration, the compositions can be formulated as tablets or lozenges in a conventional manner. [0092] [0092] The active compounds of this invention can be formulated for parenteral administration by injection, including using conventional catheterization techniques or infusion. The injection may be presented in unit dosage form, for example, in ampoules or in multi-capacity containers, with an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for reconstitution with a suitable vehicle, eg sterile pyrogen-free water, before use. [0093] The active compounds of this invention may also be formulated as rectal pharmaceutical compositions such as suppositories or retention enemas, for example, containing conventional suppository bases such as cocoa butter or other glycerides. [0094] [0094] For intranasal administration or for administration by inhalation, the active compounds of the present invention are conveniently delivered in the form of a solution or suspension that is released from a pump-spray type container that is compressed or pumped by the patient, or as an aerosol spray that is delivered from a pressurized container or nebulizer with the use of a suitable propellant, eg, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas. In the case of a pressurized aerosol, the dosage unit can be determined by means of a valve to deliver a regulated amount. The pressurized container or nebulizer can contain a solution or suspension of the active compound. Capsules or cartridges (for example, made from gelatin) for use in an inhaler or insufflator may be formulated containing a powder mix of the present invention and a suitable powder base such as lactose or starch. [0095] The compounds of this invention can be administered in a pharmaceutically acceptable dosage form either alone, or in combination with one or more agents to regulate the mammalian immune system or anti-inflammatory agents. These agents may include, but are not limited to, cyclosporin A (such as Sandimmune® or Neroal®), rapamycin, FK-506 (tacrolimus), leflunomide, deoxyspergualin, mycophenolate salts (eg Cellcept®), azathioprine ( eg Imuran®), daclizumab (eg Zenapax®), OKT3 (eg Orthoclone®), AcGam, aspirin, acetaminophen, ibuprofen, naproxen, meloxicam pyrrole and anti-inflammatory steroids (such as prednisone or dexamethasone). These agents can be administered as part of the same dosage form or as separate dosage forms, by the same route of administration or by different routes of administration, according to traditional pharmaceutical practice based on the same administration schedule or in different administration programs. Detailed Description of the Invention [0096] [0096] Unless otherwise specified, terms used in this specification and claims have the meanings described below. [0097] [0097] "Alkyl" refers to a saturated aliphatic hydrocarbon group including straight chain or branched chain groups of C1-C20. Preferably an alkyl group is an alkyl having 1 to 12 carbon atoms. Representative examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethyl propyl, 2,2-dimethylpropyl, 1-ethyl propyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1, 2-dimethylbutyl, 2,2-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, 2,2-dimethylpentyl, 3,3-dimethylpentyl, [0098] [0098] "Alkenyl" refers to an alkyl already defined above having at least two carbon atoms and at least one carbon-carbon double bond, eg vinyl, 1-propenyl, 2-propenyl, 1-, 2 - or 3-butenyl and tc. The alkenyl group can be substituted or unsubstituted. When substituted, the substituent groups are preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkylayl , aryl, heteroaryl, cycloalkyloxy, heterocyclic alkyloxy, cycloalkylthio, heterocyclic alkylthio, -(CH2)nC(O)OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O )R15, -NR16R17, -OC(O)NR16R17, - NHC(O)NR16R17, -S(O)mR15, -NHC(O)OR15 and -NHS(O)mR15. [0099] [0099] "Alkynyl" refers to an alkyl already defined above having at least two carbon atoms and at least one carbon-carbon triple bond, eg ethynyl, 1-propynyl, 2-propynyl, 1-, 2 - or 3-butynyl and so on, preferably C2-10 alkynyl, more preferably C2-6 alkynyl, and most preferably C2-4 alkynyl. The alkynyl group can be substituted or unsubstituted. When substituted, the substituent groups are preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl , heteroaryl, cycloalkyloxy, heterocyclic alkyloxy, cycloalkylthio, heterocyclic alkylthio, -(CH2)nC(O)OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O )R15, -NR16R17, -OC(O)NR16R17, -NHC(O)NR16R17, -S(O)mR15, -NHC(O)OR15 and -NHS(O)mR15. [00100] [00100] "Cycloalkyl" refers to a saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group and has 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 10 carbon atoms, and even more preferably 3 to 8 carbon atoms or 3 to 6 carbon atoms. Representative examples of monocyclic cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl and etc. Polycyclic cycloalkyl includes cycloalkyl having a spiro ring, a fused ring, or a bridged ring. [00101] [00101] "Spirocycloalkyl" refers to a 5-membered polycyclic group with rings connected through a common carbon atom (called a spiro-atom), in which one or more rings may contain one or more double bonds, but none of the rings have a completely conjugated pi electron system. Preferably a spirocycloalkyl has 6 to 14 members, more preferably 7 to 10 members. According to the common spiro atom number, spirocycloalkyl is divided into monospiro cycloalkyl, di-spirocycloalkyl, or polyspirocycloalkyl, preferably it refers to mono-spirocycloalkyl or di-spirocycloalkyl, more preferably 4-membered mono-spirocycloalkyl/ 4 members, 4 members/5 members, 4 members/6 members, 5 members/5 members, or 5 members/6 members. Representative examples of spirocycloalkyl include, but are not limited to, the following groups: and and . [00102] [00102] "Fused cycloalkyl" refers to a 5- to 20-membered polycyclic hydrocarbon group, in which each ring in the system shares an adjacent pair of carbon atoms with another ring, in which one or more rings may contain a or more double bonds, but none of the rings have a completely conjugated pi electron system. Preferably a fused cycloalkyl group has 6 to 14 members, more preferably 7 to 10 members. According to the number of member rings, fused cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl, preferably it refers to bicyclic or tricyclic fused cycloalkyl, more preferably 5-membered bicyclic fused cycloalkyl /5 members, or 5 members/6 members. Representative examples of fused cycloalkyl include, but are not limited to, the following groups: . and and [00103] [00103] "Bridged-linked cycloalkyl" refers to a 5- to 20-membered polycyclic hydrocarbon group in which every two rings in the system share two unconnected carbon atoms. Said rings may have one or more double bonds, but they do not have the fully conjugated pi electron system. Preferably a bridged cycloalkyl has 6 to 14 members, more preferably 7 to 10 members. According to the number of member rings, the bridged cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, preferably it refers to a bicyclic, tricyclic or polycyclic bridged cycloalkyl tetracyclic, more preferably bicyclic or tricyclic bridged cycloalkyl. Representative examples of bridged cycloalkyl include, but are not limited to, the following groups: and and and . [00104] Said cycloalkyl may be fused to aryl, heteroaryl or to the ring of heterocyclic alkyl, wherein the ring attached to the parent structure is cycloalkyl. Representative examples include, but are not limited to, indanylacetic, tetrahydronaphthalene, benzocidoheptyl, and so forth. Said cycloalkyl may be optionally substituted or unsubstituted. When substituted, the substituent groups are preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl , aryl, heteroaryl, cycloalkyloxy, heterocyclic alkyloxy, cycloalkylthio, heterocyclic alkylthio, an oxo group, - (CH2)nC(O)OR15, -OC(O)R15, -C(O)R15, -C(O) NR16R17, -NHC(O)R15, -NR16R17, -OC(O)NR16R17, -NHC(O)NR16R17, -S(O)mR15, -NHC(O)OR15 and -NHS(O)mR15. [00105] [00105] "Heterocyclyl" refers to a saturated and/or partially unsaturated 3- to 20-membered monocyclic or polycyclic hydrocarbon group having one or more heteroatoms selected from the group consisting of N, O, and S(O)m (where m is 0.1 or 2) as ring atoms, but excluding -OO-, -OS- or -SS- in the ring, the other ring atoms being C. Preferably, the heterocyclyl has 3 to 12 members having 1 to 4 of said heteroatoms; more preferably 3 to 10 members having 1 to 3 of said heteroatoms; more preferably 5 to 6 members having 1 to 2 of said heteroatoms. Representative examples of monocyclic heterocyclyl include, but are not limited to, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, sulfo-morpholinyl, homopiperazinyl, and so on. Polycyclic heterocyclyl includes heterocyclyl having a spiro ring, a fused ring, or a bridged ring. [00106] [00106] "Spiroheterocyclyl" refers to a 5- to 20-membered polycyclic heterocyclyl with rings connected through a common carbon atom (called a spiro-atom), wherein said rings have one or more heteroatoms selected from group consisting of N, O, and S(O)m (where m is 0.1 or 2) as ring atoms, the other ring atoms being C, where one or more rings may contain one or more double bonds, but none of the rings have a completely conjugated pi electron system. Preferably a spiro- [00107] [00107] "Fused heterocyclyl" refers to a 5- to 20-membered polycyclic heterocyclyl group, in which each ring in the system shares a pair of adjacent carbon atoms with the other ring, in which one or more rings may contain one or more double bonds, but none of the rings have a completely conjugated pi electron system, and wherein said rings have one or more heteroatoms selected from the group consisting of N, O, and S(O)p (in that p is 0, 1 or 2) as ring atoms, the other ring atoms being C. Preferably a fused heterocyclyl has 6 to 14 members, more preferably 7 to 10 members. According to the number of member rings, fused heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclyl, and preferably refers to bicyclic or tricyclic fused heterocyclyl, more preferably 5 membered bicyclic fused heterocyclyl/ 5 members, or 5 members/6 members. Representative examples of fused heterocyclyl include, but are not limited to, the following groups: O N N N NO NO H H N N N N N N [00108] [00108] "Bridge-linked heterocyclyl" refers to a 5- to 14-membered polycyclic heterocyclic alkyl group in which each two rings in the system share two disconnected atoms, said rings being one or more double bonds but not having a fully conjugated pi electron system, and wherein said rings have one or more heteroatoms selected from the group consisting of N, O, and S(O)m (where m is 0, 1 or 2) as ring atoms, the other ring atoms being C. Preferably a bridged heterocyclyl has 6 to 14 members, more preferably 7 to 10 members. According to the number of member rings, bridged heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclyl, and preferably refers to bridged heterocyclyl. bicyclic, tricyclic or tetracyclic bridged, more preferably bicyclic or tricyclic bridged heterocyclyl. Representative examples of bridged heterocyclyl include, but are not limited to, the following groups: H N N N N N [00109] [00109] Said heterocyclyl ring can be fused to the aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is heterocyclyl. Representative examples include, but are not limited to, the following groups: H H H O N N N [00110] [00110] Heterocyclyl can be optionally substituted or unsubstituted. When substituted, the substituent groups are preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, alkyl h and - terocyclic, aryl, heteroaryl, cycloalkyloxy, heterocyclic alkyloxy, cycloalkylthio, heterocyclic alkylthio, an oxo group, -(CH2)nC(O)OR15, - OC(O)R15, -C(O)R15, -C(O) NR16R17, -NHC(O)R15, -NR16R17, - OC(O)NR16R17, -NHC(O)NR16R17, -S(O)mR15, -NHC(O)OR15 and - NHS(O)mR15. [00111] [00111] "Aryl" refers to an all-carbon 14-membered monocyclic ring or a polycyclic fused ring (a "fused" ring system means that each ring in the system shares a pair of carbon atoms adjacent to the other ring in the system), and has a system of fully conjugated pi electrons. Preferably aryl is 6 to 10 members, as is phenyl and naphthyl, more preferably phenyl. Said aryl can be fused to the heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is aryl. Representative examples include, but are not limited to, the following groups: O H H N N N N AT THE O O O O H H H N N N N [00112] [00112] The aryl group can be substituted or unsubstituted. When substituted, the substituent groups are preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl , heteroaryl, cycloalkyloxy, heterocyclic alkyloxy, cycloalkylthio, heterocyclic alkylthio, -(CH2)nC(O)OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15 , -NR16R17, -OC(O)NR16R17, -NHC(O)NR16R17, -S(O)mR15, -NHC(O)OR15 and -NHS(O)mR15. [00113] [00113] "Heteroaryl" refers to a heteroaryl system having 1 to 4 heteroatoms selected from the group consisting of O, S and N as ring atoms and having 5 to 14 ring atoms. Preferably a heteroaryl has 5 to 10 members, more preferably 5 or 6 members, for example, thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, triazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrrolyl, N-alkyl pyrroly - la, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, among others. Said heteroaryl can be fused to the aryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is heteroaryl. Representative examples include, but are not limited to, the following groups: the H N N N N N NO NO N N N H [00114] [00114] The heteroaryl group can be substituted or unsubstituted. When substituted, the substituent groups are preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, [00115] [00115] "Alkoxyl" refers to both an -O-(alkyl) and an -O-(unsubstituted cycloalkyl) group, wherein alkyl has the definition given above. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, among others. Alkoxy may optionally be substituted or unsubstituted. When substituted, the substituent is preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, alkyl heterocyclic, aryl, heteroaryl, cycloalkyloxy, heterocyclic alkyloxy, cycloalkylthio, heterocyclic alkylthio, -(CH2)nC(O)OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, - NHC(O)R15, -NR16R17, -OC(O)NR16R17, -NHC(O)NR16R17, -S(O)mR15, -NHC(O)OR15 and -NHS(O)mR15. [00116] [00116] "Bond" refers to a covalent bond using a "—" sign. [00117] [00117] "Hydroxy Alkyl" refers to an alkyl group substituted with a hydroxy group, where alkyl has the definition given above. [00118] [00118] "Hydroxy" refers to an -OH group. [00119] [00119] "Halogen" refers to atoms of fluorine, chlorine, bromine or iodine. [00120] [00120] "Amino" refers to an -NH2 group. [00121] [00121] "Cyan" refers to a -CN group. [00122] [00122] "Nitro" refers to a -NO2 group. [00123] [00123] "An oxo group" refers to an =O group. [00124] [00124] "Carboxyl" refers to a -C(O)OH group. [00125] "Alkoxycarbonyl" refers to a group -C(O)O(alkyl) or (cycloalkyl), wherein alkyl and cycloalkyl have the definitions given above. [00126] [00126] Enantiomer: [00127] [00127] "Optional" or "optionally" means that the event or circumstance described subsequently may, but need not necessarily, occur, and the report includes cases where the event or circumstance may or may not occur. For example, "the heterocyclic group optionally substituted with an alkyl" means that an alkyl group may, but need not necessarily, be present, and the report includes cases where the heterocyclic group is optionally substituted with an alkyl and cases where the heterocyclic group is not substituted with an alkyl. [00128] [00128] "Substituted" refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3 hydrogen atoms independently substituted by a corresponding number of substituents. It goes without saying that the substituents only exist in their possible chemical position. The person skilled in the art will be able to determine whether replacement is possible or impossible without great effort through experience or theory. For example, the combination of an amino or hydroxyl group having free hydrogen and carbon atoms having unsaturated (such as olefinic) bonds may be unstable. [00129] A "pharmaceutical composition" refers to a mixture of one or more of the compounds described in the present invention or physiologically/pharmaceutically acceptable salts or prodrugs thereof and other chemical components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate the administration of a compound to an organism, which is conducive to the absorption of the principle. [00130] "Pharmaceutically acceptable salts" refers to salts of the compounds of the invention, such salts being safe and effective when used in a mammal and having corresponding biological activity. [00131] [00131] N, m and R15 to R17 have the definitions given for the compound of formula (I). Synthesis Method of the Compound of the Present Invention [00132] [00132] To complete the purpose of the present invention, the present invention applies, but without limitation, to the following technical solution: [00133] [00133] A process for preparing the compound of formula (I) of the invention or a pharmaceutically acceptable salt thereof, comprising the following steps: 13 14 R R 1 5 R [00134] reacting the compound of formula (IA) with the compound of formula (IB) under alkaline conditions to obtain the compound of formula (I); [00135] [00135] the alkaline condition is provided by an organic base and an inorganic base, wherein said organic base includes, but is not limited to, triethylamine, N,N-diisopropylethylamine, n-butyl lithium, tert. butyl potassium, and said inorganic base includes, but is not limited to, sodium hydride, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate or cesium carbonate; [00136] [00136] wherein X is halogen, A, R, L, R1 to R14, p, q, s and t have the definitions given for formula (I); preferably, R1 is tert-butoxycarbonyl; preferably L is a bond. [00137] [00137] A process for preparing the compound or the salt of formula (I) of the invention, comprising the following steps: [00138] [00138] when R1 is t-butoxycarbonyl, t-butoxycarbonyl is further optionally removed from the compound of formula (I) to obtain the compound of formula (IC) or a pharmaceutically acceptable salt thereof; reacting the compound of formula (IC) or a pharmaceutically acceptable salt thereof with carboxylic acid, acyl chloride, sulfonyl chloride, carboxylic esters, an ethylene oxide derivative or halogen under alkaline conditions to obtain the compounds of formula ( I); [00139] [00139] the reaction solvent includes, but is not limited to: tetrahydrofuran, ethanol, methanol, n-butanol, dichloromethane, 1,4-dioxane or N,N-dimethylformamide; [00140] [00140] the alkaline condition is provided by an organic base and an inorganic base, wherein said organic base includes, but is not limited to, triethylamine, N,N-diisopropylethylamine, n-butyl lithium, tert. butyl potassium, tetrabutylammonium bromide, and said inorganic base include, but are not limited to, sodium hydride, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate or cesium carbonate; [00141] [00141] wherein A, R, L, R1 to R14, p, q, s and t have the definitions given for formula (I); preferably L is a bond. Preferred Modalities [00142] [00142] The following examples serve to illustrate the invention, but the examples are not to be considered as limiting the scope of the invention. If specific conditions for the experimental method are not specified in the examples of the present invention, they are generally in accordance with conventional conditions or conditions recommended for the raw materials and by the product manufacturer. And reagents without specific source indication are conventional commercially available reagents. [00143] The structure of the compound was identified by NMR and/or MS. NMR chemical shifts (δ) are given at 10-6 (ppm). NMR is determined by a Bruker AVANCE-400 instrument. Solvents were deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDCl3) and deuterated methanol (CD3OD) with tetramethylsilane (TMS) as an internal standard. [00144] [00144] MS was determined by a FIN-NIGAN LCQAd (ESI) mass spectrometer (manufacturer: Thermo, type: Finnigan LCQ advantage MAX). [00145] [00145] HPLC was determined by a spectrometer associated with high pressure liquid chromatography Agilent 1200DAD (chromatographic column Sunfire C18 150×4.6 mm) and a spectrometer associated with high pressure liquid chromatography Waters 2695-2996 high (chromatographic column) Gimini C18 150×4.6 mm). [00146] [00146] The mean kinase inhibition rate and the IC50 value were determined by a microplate reader (BMG company, Germany). [00147] [00147] Thin layer silica gel used a Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate. The size of the plates used in the TLC ranged from 0.15 mm to 0.2 mm, and the size of the plates used in thin layer chromatography for product purification ranged from 0.4 mm to 0.5 mm. [00148] [00148] Column chromatography generally used silica gel Yan- [00149] The known starting material of the invention can be prepared by the conventional synthesis method of the prior art, or can be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc or Dari chemical Company , etc. [00150] [00150] Unless otherwise specified in the examples, the following reactions were placed in an argon atmosphere or in a nitrogen atmosphere. [00151] [00151] The term "argon atmosphere" or "nitrogen atmosphere" refers to the fact that a reaction flask is equipped with a flask having 1 L of argon or nitrogen. [00152] [00152] The term "hydrogen atmosphere" refers to the fact that a reaction flask is equipped with a flask having 1 L of hydrogen. [00153] [00153] The high pressure hydrogenation reactions were carried out with a Parr 3916EKX hydrogenation apparatus and either a clear blue QL-500 hydrogen generator or an HC2-SS hydrogenation apparatus. [00154] [00154] In hydrogenation reactions, the reaction system was usually evacuated and filled with hydrogen, and the above operation was repeated three times. [00155] [00155] The microwave reactions were performed with a CEM Discover-S 908860 microwave reactor. [00156] [00156] Unless otherwise specified in the examples, the solution used in the following reactions refers to an aqueous solution. [00157] [00157] Unless otherwise specified in the examples, the reaction temperature in the following reactions was room temperature. [00158] [00158] Ambient temperature was the most suitable reaction temperature, which ranges from 20 °C to 30 °C. [00159] [00159] The reaction process was monitored by thin layer chromatography (TLC), the developing solvent system included: (A) dichloromethane and methanol system, (B) n-hexane and ethyl acetate system, (C) petroleum ether and ethyl acetate system, (D) acetone. The solvent volume ratio was adjusted according to the polarity of the compounds. [00160] [00160] The elution system for purification of compounds by column chromatography and thin layer chromatography included: (A) dichloromethane and methanol system, (B) n-hexane and ethyl acetate system, (C) dichloromethane system and acetone, and the solvent volume ratio was adjusted according to the polarity of the compounds, and sometimes a little alkaline reagent such as triethylamine or an acidic reagent such as acetic acid could also be added. Example 1 (3aR,5R,6aS/3aS,5S,6aR)-tert-butyl-3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro cyclopenta[c]pyrrole-2(1H)-carboxylate O AT THE N H [00161] Cuprous chloride (770 mg, 4 mmol) was dissolved in 10 mL of tetrahydrofuran. After cooling to -78°C, a 3M solution of methylmagnesium bromide in diethyl ether (30ml,6.7mmol) was added to the reaction mixture. After reacting for 30 minutes, the reaction mixture was warmed to -35°C, followed by the dropwise addition of 10 ml of a solution of tert-butyl 5-oxo-3,3a,4,5-tetra- hydrocyclopenta[c]pyrrole-2(1H)-carboxylate 1a (500 mg, 2.24 mmol) in tetrahydrofuran. After reacting for 30 minutes, the reaction mixture was warmed to room temperature, followed by the addition of 10 mL of a saturated ammonium chloride solution to quench the reaction. The reaction mixture was extracted with ethyl acetate (20 mL×3). The organic phases were combined, dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the crude title product (3aR,6aS/3aS,6aR)-tert-butyl 3a-methyl-5-oxohexahydro-cyclopenta[c]pyrrole-2(1H)- carboxylate 1b (500 mg, brown oil), which was used directly in the next step without further purification. Step 2 (3aR,5R,6aS/3aS,5S,6aR)-tert-Butyl 3a-methyl-5-(methylamino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate [00162] The crude product (3aR,6aS/3aS,6aR)-tert-butyl 3a-methyl-5-oxohexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 1b (200mg, 0, 84 mmol) was dissolved in 5 mL of methanol, followed by the addition of 2 mL of a 37% solution of methyl amine-ethanol and sodium triacetoxyborohydride (532 mg, 2.5 mmol). After reacting for 16 hours, 10 mL of a saturated ammonium chloride solution was added to the reaction mixture to quench the reaction. The reaction mixture was extracted with ethyl acetate (20 mL×3). The organic phases fo- [00163] [00163] MS m/z (ESI): 255.2 [M+1] Step 3 (3aR,5R,6aS/3aS,5S,6aR)-tert-Butyl-3a-methyl-5-(methyl(7H-) pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate [00164] [00164] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (60 mg, 0.39mmol) was dissolved in 5 mL of H2O, followed by the addition of (3aR,5R,6aS/3aS,5S, 6aR)-tert-butyl 3a-methyl-5-(methylamino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 1c (100mg, 0.39mmol) and potassium carbonate (322mg) , 2.34 mmol). After reacting for 16 hours at 100°C, the reaction mixture was extracted with ethyl acetate (10 mL×3). The organic phase was combined, dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product (3aR,5R,6aS/3aS,5S,6aR)-tert-butyl-3a- methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate 1 (40 mg, yield 28.8 %) as a white solid. [00165] [00165] MS m/z (ESI): 372.5[M+1] 1 [00166] [00166] 1 H NMR (400 MHz, CDCl 3 ): 10.38 (s, 1H), 8.30 (s, 1H), 7.05-7.04 (m, 1H), 6.57-6.56 (m, 1H), 5.57-5.52 (m, 1H), 3.56-3.49 (m, 3H), 3.37 (s, 3H), 3.27-3.25 (m) , 1H), 2.25-2.21 (m, 2H), 1.89-1.87 (m, 2H), 1.67-1.65 (m, 2H), 1.49 (s, 9H) ), 0.88-0.86 (m, 2H) Example 2 N-((3aR,5R,6aS/3aS,5S,6aR)-2-(Ethylsulfonyl)-3a-methyl octahydro cyclopenta[c ]pyrrol-5-yl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine O O s N [00167] [00167] (3aR,5R,6aS/3aS,5S,6aR)-tert-Butyl-3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexa- hydrocyclopenta[c]pyrrole-2(1H)-carboxylate 1 (1 g, 2.7 mmol) was dissolved in 15 mL of a 6 M solution of hydrogen chloride in methanol. After reacting for 12 hours, the reaction mixture was concentrated under reduced pressure to obtain the crude title product N-methyl-N-((3aR,5R,6aS)-3a-methyloctahydro-cyclopenta hydrochloride [c]pyrrol-5-yl)-7H-pyrrole[2,3-d]pyrimidin-4-amine 2a (1.2 g, white solid), which was used directly in the next step without further purification. [00168] [00168] MS m/z (ESI): 272.2 [M+1] Step 2 N-((3aR,5R,6aS/3aS,5S,6aR)-2-(Ethylsulfonyl)-3a-methyl octahydro cyclopenta[c]pyrrol-5-yl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine [00169] N-Methyl-N-((3aR,5R,6aS/3aS,5S,6aR)-3a-methyloctahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrole[2] hydrochloride ,3-d]pyrimidin-4-amine 2a (100 mg, 0.37 mmol) and triethylamine (112 mg, 1.11 mmol) were dissolved in 5 mL of tetrahydrofuran, followed by dropwise addition of ethyl sulfonyl chloride (95 mg, 0.74 mmol). After reacting for 16 hours, the reaction mixture was added with 20 mL of H2O, and extracted with ethyl acetate (20 mL×3). The organic phase was combined, dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product N-((3aR,5R,6aS/3aS,5S,6aR) -2-(ethylsulfonyl)-3a-methyl octahydro cyclopenta[c]pyrrol-5-yl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine 2 (28 mg, yield of 21.5%) as a white solid. [00170] [00170] MS m/z (ESI): 364.2 [M+1] 1 [00171] [00171] 1 H NMR (400 MHz, DMSO-d6): 12.56 (s, 1H), 8.25-8.23 (m, 1H), 7.95-7.93 (m, 1H), 7 .21-7.20 (m, 1H), 5.51-5.49 (m, 1H), 4.02 (m, 2H), 3.75-3.58 (m, 6H), 3.38 (s, 3H), 3.22-3.16 (m, 2H), 2.06 (s, 3H), 1.97-1.95 (m, 1H), 1.43-1.41 (m , 3H) Example 3 3-((3aR,5R,6aS/3aS,5S,6aR)-3a-Methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexa -hydrocyclopenta[c]pyrrole-2(1H)-yl)-3-oxopropanenitrile O N N N H [00172] N-Methyl-N-((3aR,5R,6aS/3aS,5S,6aR)-3a-methyloctahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrole[2] hydrochloride ,3-d]pyrimidin-4- [00173] [00173] MS m/z (ESI): 339.1 [M+1] 1 [00174] [00174] 1 H NMR (400 MHz, DMSO-d6): 12.58 (s, 1H), 8.27-8.25 (m, 1H), 7.97-7.95 (m, 1H), 7 .26-7.24 (m, 1H), 5.58-5.53 (m, 1H), 4.33 (s, 2H), 3.75-3.58 (m, 6H), 3.39 (s, 3H), 3.22-3.16 (m, 2H), 2.08 (s, 3H), 1.99-1.97 (m, 1H) Example 4 2-((3aR,5R, 6aS/3aS,5S,6aR)-3a-Methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)- il) acetonitrile N N N H [00175] N-Methyl-N-((3aR,5R,6aS/3aS,5S,6aR)-3a-methyloctahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrole[2] hydrochloride ,3-d]pyrimidin-4-amine 2a (50 mg, 0.18 mmol) was dissolved in 5 mL of acetonitrile, followed by the addition of potassium carbonate (75 mg, 0.54 mmol), bromoacetonitrile1(24 mg, 0.2 mmol) and 5 ml of dichloromethane. After reacting for 16 hours, the reaction mixture was added with a small amount of H2O to abruptly cool the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (10 mL × 3). The organic phase was combined, washed with water (5 ml), brine (5 ml) successively, dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-((3aR,5R,6aS/3aS,5S,6aR) -3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)-yl)acetonitrile 4 (20 mg, 35.1% yield as a white solid. [00176] [00176] MS m/z (ESI): 311.5 [M+1] 1 [00177] [00177] 1 H NMR (400 MHz, DMSO-d6): 11.64 (s, 1H), 8.12-8.10 (m, 1H), 7.14 (s, 1H), 6.54 (s , 1H), 3.86-3.85 (m, 2H), 3.14-3.13 (m, 3H), 2.77-2.75 (m, 1H), 2.62-2.60 (m, 1H), 2.08-2.06 (m, 3H), 1.86-1.80 (m, 1H), 1.70-1.69 (m, 1H), 1.58-1 .55 (m, 1H), 1.30-1.18 (m, 2H), 0.86 (s, 3H) Example 5 (3aR,5s,6aS)-tert-Butyl-(methyl(7H-pyrrole) 2,3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate O H O N N H N N N H O O [00178] (3aR,5R,6aS)-tert-Butyl 5-hydroxyhexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 5a (9 g, 40 mmol) was dissolved in 150 mL of dichloromethane, followed by the addition of sulfonyl methylchloride (4.70 mL, 60 mmol) and triethylamine (11.20 mL, 80 mmol) at 0°C. After reacting for 2 hours at room temperature, 200 ml of a saturated sodium bicarbonate solution were added to the reaction mixture. The aqueous phase and the organic phase were separated. The organic phase was washed with a saturated sodium chloride solution (200 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the title product (3aR,5R,6aS)-tert-butyl 5-((methylsulfonyl)oxy)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 5b (12.00 g, 98.4%) yield as a yellow liquid. Step 2 (3aR,5s,6aS)-tert-Butyl 5-(methylamino) hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate [00179] [00179] (3aR,5R,6aS)-tert-Butyl 5-((methylsulfonyl)oxy)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 5b (60 mg, 0.2 mmol) was dissolved in 10 mL of methanol, followed by the addition of 5 mL of methylamine. After reacting for 16 hours at 40°C, the reaction mixture was concentrated under reduced pressure to obtain the crude title product (3aR,5s,6aS)-tert-butyl 5-(methylamino)hexahydro-cyclo- penta[c]pyrrole-2(1H)-carboxylate 5c (60 mg, brown oil), which was used directly [00180] [00180] MS m/z (ESI): 241.5 [M+1] Step 3 (3aR,5s,6aS)-tert-Butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4) - yl)amino)hexahydro cyclopenta[c]pyrrole-2(1H)-carboxylate [00181] [00181] (3aR,5s,6aS)-tert-Butyl 5-(methylamino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 5c (200mg, 0.8mmol) and 4- chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (127 mg, 0.8 mmol) was dissolved in 5 mL of n-butanol, followed by the addition of triethylamine (168 mg, 1.6 mmol). After reacting for 48 hours at 100°C, the reaction mixture was concentrated under reduced pressure, followed by addition of 10 mL of H2O and 10 mL of ethyl acetate. The aqueous phase and the organic phase were separated. The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparative HPLC to obtain the title product (3aR,5s,6aS)-tert-butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 5 (5 mg, 5.0%) yield as a white solid. [00182] [00182] MS m/z (ESI): 358.5[M+1] 1 [00183] 1 H NMR (400 MHz, CDCl 3 ): 10.07 (s, 1H), 8.31 (s, 1H), 7.50 (s, 1H), 6.55(s, 1H), 5. 58-5.54 (m, 1H), 3.65-3.62 (m, 2H), 3.27-3.23 (m, 5H), 2.86-2.81 (m, 2H), 2.06-2.02 (m, 2H), 1.93-1.91 (m, 2H), 1.49 (s, 6H) Example 6 2-Hydroxy-1-((3aR,5s,6aS) -5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)ethanone O H OH N N H N N N H [00184] [00184] (3aR,5s,6aS)-tert-Butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydrocyclopenta[c]pyrrole-2(1H) -carboxylate 5 (1.5 g, 4.2 mmol) was dissolved in 20 mL of a 1 M solution of hydrogen chloride in methanol. After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure to obtain the crude title product N-methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]-hydrochloride ]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine 6a (1.5 g, brown solid), which was used directly in the next step without further purification. [00185] [00185] MS m/z (ESI): 258.1 [M+1] Step 2 2-Hydroxy-1-((3aR,5s,6aS)-5-(methyl(7H-pyrrole[2,3-d) ]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrol-2(1H)-yl)ethanone [00186] [00186] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.3 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of 2-glycolic acid (26 mg, 0.3 mmol), triethylamine (103 mg, 1.02 mmol) and hexafluorophosphate of O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium (194 mg, 0.45 mmol). After reacting for 16 hours, the reaction mixture was added with 10 mL of H2O. The aqueous phase and the organic phase were separated. The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparatory HPLC to obtain the title product 2-hydroxy-1-((3aR,5s,6aS)-5-(methyl(7H-pyrrole[2,3-d]) pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrol-2(1H)-yl)ethanone 6 (10 mg, 9.7%) yield as a white solid. [00187] [00187] MS m/z (ESI): 316.2 [M+1] 1 [00188] [00188] 1 H NMR (400 MHz, DMSO-d6): 11.60 (s, 1H), 8.09 (s, 1H), 7.11 (s, 1H), 6.53 (s, 1H), 5.47-5.44 (m, 1H), 4.52-4.49 (m, 1H), 4.03-3.99 (m, 2H), 3.60-3.57 (m, 2H) ), 3.24-3.22 (m, 2H), 3.15 (s, 3H), 2.80-2.75 (m, 2H), 2.02-1.94 (m, 2H), 1.78-1.75 (m, 2H) Example 7 2-Methyl-1-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)propan-2-ol H OH N N H N N N [00189] [00189] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of methanol, followed by the addition of 2,2-dimethyl epoxy ethane (42 mg, 0.58 mmol). After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-methyl-1- (( 3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)propan -2-ol 7 (50 mg, yield of [00190] [00190] MS m/z (ESI): 330.2 [M+1] 1 [00191] [00191] 1 H NMR (400 MHz, DMSO-d6): 11.68 (s, 1H), 8.11 (s, 1H), 7.14-713 (m, 1H), 6.60 (s, 1H) ), 5.46 (s, 1H), 5.12 (s, 1H), 3.86 (m, 2H), 3.18 (s, 4H), 3.07-3.01 (m, 3H) , 2.84 (d, 2H), 1.95-1.91 (m, 2H), 1.68 (s, 2H), 1.33-1.18 (m, 6H) Example 8 3-(( 3aR,5s,6aS)-5-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)- 3-oxopropanenitrile O N H N N H N N N H [00192] [00192] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of n-butanol, followed by the addition of 2-ethyl cyanoacetate (77 mg, 0.68 mmol) and DBU (103 mg, 0.68 mmol) . After reacting for 15 hours at 40°C, the reaction mixture was concentrated under reduced pressure. The residue was added with 50 ml of ethyl acetate and washed with a saturated solution of sodium chloride (15 ml×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparative HPLC to obtain the title product 3-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino)hexahydro-cyclo- [00193] [00193] MS m/z (ESI): 325.1 [M+1] 1 [00194] [00194] 1 H NMR (400 MHz, CDCl 3 ): 9.79 (s, 1H), 8.28 (s, 1H), 7.06-7.05 (m, 1H), 6.55-6.54 (m, 1H), 5.68-5.64 (m, 1H), 3.85-3.79 (m, 2H), 3.49 (s, 2H), 3.47-3.37 (m , 2H), 3.27 (s, 3H), 3.08-2.91 (m, 2H), 2.09-2.05 (m, 2H), 1.96-1.94 (m, 2H) ) Example 9 (3aR,5s,6aS)-N-Isopropyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole- 2(1H)-carboxamide O HN N H N H N N N H [00195] [00195] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.39 mmol) was dissolved in 5 mL of N,N-dimethylformamide, followed by dropwise addition of triethylamine (39 mg, 0.39 mmol). After reacting for 30 minutes, the reaction mixture was added with isocyanatopropane (33 mg, 0.39 mmol). After reacting for 16 hours, the reaction mixture was added with 10 ml of H2O and 10 ml of ethyl acetate. The aqueous phase and the organic phase were separated. The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparatory HPLC to obtain [00196] [00196] MS m/z (ESI): 343.3 [M+1] 1 [00197] [00197] 1 H NMR (400 MHz, CDCl 3 ): 11.18 (s, 1H), 8.31 (s, 1H), 7.06 (s, 1H), 6.54 (s, 1H), 5. 63-5.58 (m, 1H), 4.06-4.01 (m, 2H), 3.63-3.58 (m, 2H), 3.26-3.21 (m, 5H), 2.92-2.88 (m, 2H), 2.04-2.01 (m, 2H), 1.94-1.89 (m, 2H), 1.89-1.74 (m, 6H) ) Example 10 (3aR,5S,6aS/3aS,5R,6aR)-tert-Butyl 3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro -cyclopenta[c]pyrrole-2(1H)-carboxylate O AT THE N H [00198] The crude product (3aR,6aS/3aS,6aR)-tert-butyl 3a-methyl-5-oxohexahydro cyclopenta[c]pyrrole-2(1H)-carboxylate 1b (1 g, 4.2 mmol) was dissolved in 10 mL of methanol, followed by the addition of sodium borohydride (320 mg, 8.4 mmol). After reacting for 1 hour, the reaction mixture was poured into 50 mL of a saturated ammonium chloride solution and extracted with ethyl acetate (20 mL×3). The organic phase was combined, washed with H2O (5 mL×3) and a saturated solution of clo. [00199] The crude product (3aR,5R,6aS/3aS,5S,6aR)-tert-butyl 5-hydroxy-3a-methyl-hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate 10a (1 g , 4.2 mmol) was dissolved in 10 mL of dichloromethane, followed by the addition of triethylamine (1.27 g, 12.6 mmol). 5 mL of a solution of sulfonyl methylchloride in dichloromethane (700 mg, 6.2 mmol) was added dropwise to the reaction mixture at 0 °C. After reacting for 16 hours, the reaction mixture was poured into 10 mL of H2O. The aqueous phase and the organic phase were separated. The organic phase was combined and washed with saturated sodium bicarbonate (10 mL×3) and a saturated sodium chloride solution (10 mL×3) successively, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the title product (3aR,5R,6aS/3aS,5S,6aR)-tert-butyl 3a-methyl-5-((methylsulfonyl)oxy)hexahydro-cyclopenta[ c]pyrrole-2(1H)-carboxylate 10b (1.2 g, 92.3% yield) as a pale yellow oil. Step 3 (3aR,5S,6aS/3aS,5R,6aR)-tert-Butyl 3a-methyl-5-(methylamino) hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate [00200] [00200] (3aR,5R,6aS/3aS,5S,6aR)-tert-butyl 3a-Methyl-5-((methylsulfonyl)oxy)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 10b (500 mg, 1.6 mmol) was dissolved in 10 mL of a methyl solution. [00201] The crude product (3aR,5S,6aS/3aS,5R,6aR)-tert-butyl 3a-methyl-5-(methylamino) hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 10c (350mg, 1.37mmol) was dissolved in 10mL of n-butanol, followed by the addition of 4-chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (320mg, 2.06mmol) and triethylamine (410mg, 4.13mmol). After reacting for 16 hours at 100°C, the reaction mixture was cooled to room temperature and poured into 50 mL of H2O, followed by the addition of 20 mL of ethyl acetate. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with ethyl acetate (10 ml). The organic phase was combined, washed with H2O (5 mL×3) and a saturated sodium chloride solution (5 mL×3) successively, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product (3aR,5S,6aS/3aS,5R,6aR)-tert-butyl 3a -methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate (20 mg, yield of 3.9%) as a light yellow oil. [00202] [00202] MS m/z (ESI): 372.2 [M+1] 1 [00203] 1 H NMR (400 MHz, DMSO-d6): 11.61 (s, 1H), 8.09 (s, 1H), 7.10 (s, 1H), 6.53 (s, 1H), 5.47-5.44 (m, 1H), 3.52 (t, 1H), 3.44 (m, 1H), 3.28 (d, 1H), 3.20 (d, 1H), 3 .17 (s, 3H), 2.33-2.30 (m, 1H), 2.13- [00204] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (200 mg, 0.68 mmol) was dissolved in 20 mL of dichloromethane, followed by the addition of triethylamine (206 mg, 2.04 mmol). After reacting for 0.5 hour, the reaction mixture was added with acetyl chloride (53 mg, 0.68 mmol) in drops. After reacting for 16 hours, the reaction mixture was added with 10 mL of H2O. The aqueous phase and the organic phase were separated. The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparative HPLC to obtain the title product 1-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)ethanone 11 (20 mg, 9.8%) yield as a white solid. [00205] [00205] MS m/z (ESI): 300.3 [M+1] 1 [00206] [00206] H NMR (400 MHz, CDCl3): 11.32 (s, 1H), 8.30 (s, 1H), [00207] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) and potassium carbonate (94 mg, 0.68 mmol) were dissolved in 5 mL of 1,4-dioxane. After reacting for 0.5 hour, the reaction mixture was added 2-ethyl bromoacetate (56 mg, 0.34 mmol) dropwise. After reacting for 4 hours, 50 ml of dichloromethane was added to the reaction mixture to dissolve the residue. The reaction mixture was washed with a saturated solution of sodium chloride (15 ml). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product ethyl 2-((3aR,5s,6aS)-5-(methyl(7H) -pyrrole[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)acetate 12 (24 mg, 21.4%) yield as a white solid. [00208] [00208] MS m/z (ESI): 344.4[M+1] 1 [00209] 1 H NMR (400 MHz, DMSO-d6): 11.54 (s, 1H), 8.09 (s, 1H), 7.07-7.06 (m, 1H), 6.76-6 .75 (m, 1H), 5.36-5.34 (m, 1H), 4.11 (q, 2H), 3.28 (s, 2H), 3.08 (s, 3H), 2. 67-2.65 (m, 4H), 1.95-1.93 (m, 2H), 1.63-1.58 (m, 2H), 1.25-1.18 (m, 5H) Example 13 Cyclopropyl((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H) -yl)methanone O H N N H N N N H [00210] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by dropwise addition of triethylamine (69 mg, 0.68 mmol). After reacting for 0.5 hour, the reaction mixture was added with cyclopropyl formyl chloride (39 mg, 0.37 mmol). After reacting for 3 hours, 50 ml of dichloromethane was added to the reaction mixture to dissolve the residue. The reaction mixture was washed with a saturated solution of sodium chloride (15 ml). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the cyclopropyl title product. [00211] [00211] MS m/z (ESI): 326.1 [M+1] 1 [00212] [00212] 1 H NMR (400 MHz, DMSO-d6): 11.60 (s, 1H), 8.09 (s, 1H), 7.11-7.10 (m, 1H), 6.52-6 1.51 (m, 1H), 5.48-5.44 (m, 1H), 3.85-3.83 (m, 1H), 3.55-3.49 (m, 2H), 3.24 -3.21 (m, 1H), 3.15 (s, 3H), 2.93-2.91 (m, 1H), 2.81-2.79 (m, 1H), 2.03-1 .98 (m, 2H), 1.83-1.78 (m, 3H), 0.78-0.71 (m, 4H) Example 14 2-Hydroxy-1-((3aS,5R,6aR/3aS) ,5R,6aR)-3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)- il)ethanone O H OH N N [00213] [00213] (3aR,5S,6aS/3aS,5R,6aR)-tert-Butyl 3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro -cyclopenta[c]pyrrole-2(1H)-carboxylate 10 (1.56 g, 4.2 mmol) was dissolved in 20 mL of a 1 M solution of hydrogen chloride in methanol. After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure to obtain the crude title product N-methyl-N- hydrochloride ((3aR,5S,6aS/3aS,5R,6aR)-3a-methylocta -hydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine 14a (1.5 g, brown solid), which was used directly in the next step without further purification. Step 2 2-Hydroxy-1-((3aS,5R,6aR/3aS,5R,6aR)-3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)ethanone [00214] N-Methyl-N-((3aR,5s,6aS/3aS,5R,6aR)-3a-methyl octahydrocyclopenta[c]pyrrol-5-yl)-7H-pyrrole[2,3a] hydrochloride -d]pyrimidin-4-amine 14a (70 mg, 0.22 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (67 mg, 0.66 mmol). After reacting for 0.5 hour, the reaction mixture was added with 2-glycolic acid (25 mg, 0.33 mmol) and O-(7-azabenzotriazol-1-yl)-N,N,N' hexafluorophosphate, N'-tetramethylformamidinium (210 mg, 0.33 mmol). After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure and purified by preparatory HPLC to obtain the title product 2-hydroxy-1-((3aS,5R,6aR/3aS,5R,6aR) - 3a-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrol-2(1H)-yl)ethanone 14 ( 11 mg, 13.9%) yield as a pale yellow solid. [00215] [00215] MS m/z (ESI): 330.4[M+1] 1 [00216] 1 H NMR (400 MHz, DMSO-d6): 11.62 (s, 1H), 8.09 (s, 1H), 7.12 (s, 1H), 6.54(s, 1H), 5.51 (s, 1H), 4.56 (s, 1H), 4.01-3.66 (m, 2H), 3.64-3.58 (m, 1H), 3.23 (s, 3H), 2.11 (d, 1H), 2.10-2.08 (m, 1H), 1.99-1.97 (m, 1H), 1.76-1.70 (m, 2H) , 1.30-1.19 (m, 5H) Example 15 N-((3aR,5s,6aS)-2-(Cyclopropylsulfonyl)octahydro-cyclopenta[c]pyrrol-5-yl)-N- methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine O O H S N N H N N N H [00217] [00217] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100mg, 0.34mmol) was dissolved in 5mL of dichloromethane, followed by the addition of triethylamine (78mg, 0.7mmol). After reacting for 0.5 hour, the reaction mixture was added dropwise with cyclopropyl sulfonyl chloride (54 mg, 0.4 mmol). After reacting for 16 hours, the reaction mixture was added with 10 mL of H2O. The aqueous phase and the organic phase were separated. The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparatory HPLC to obtain the title product N-((3aR,5s,6aS)-2-(cyclopropylsulfonyl)octahydro-cyclopenta[c]pyrrol-5-yl) - N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine 15 (20 mg, 14.3% yield) as a white solid. [00218] [00218] MS m/z (ESI): 362.3 [M+1] 1 [00219] [00219] 1 H NMR (400 MHz, CDCl 3 ): 10.11 (s, 1H), 8.31 (s, 1H), 7.08 (s, 1H), 6.70 (s, 1H), 5. 52 (s, 1H), 3.57(s, 2H), 3.31-3.25 (m, 4H), 2.95-2.93 (m, 2H), 2.39-2.27 ( m, 2H), 1.25-1.20 (m, 4H), 1.04-1.03 (m, 2H), 0.87-0.86 (m, 2H) Example 16 tert-Butyl (2 -((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H)- il)-2-oxoethyl)carbamate O H H NO N O N H N N N H O [00220] [00220] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (500 mg, 1.7 mmol) was dissolved in 20 mL of dichloromethane, followed by the addition of triethylamine (0.47 mL, 3.4 mmol). After reacting for 1 hour, the reaction mixture was added with 2-(tert-butoxy formamide) acetic acid (0.36 g, 2.04 mmol) and O-(7-azabenzotriazol-1-yl) hexafluorophosphate - N,N,N',N'-tetramethylformamidinium (0.76 g, 2.04 mmol). After reacting for 11 hours, the reaction mixture was added with 100 ml of dichloromethane. The reaction mixture was washed with a saturated solution of sodium chloride (20 ml×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product tert-butyl (2-((3aR,5s,6aS)-5-( methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)-2-oxoethyl)carbamate 16 (320 mg, 45.5% yield) as a pale yellow solid. [00221] [00221] MS m/z (ESI): 415.4 [M+1] 1 [00222] [00222] 1 H NMR (400 MHz, DMSO-d6): 11.60 (s, 1H), 8.10 (s, 1H), 7.13-7.12 (m, 1H), 6.76-6 .75 (m, 1H), 6.54 (s, 1H), 5.49-5.47 (m, 1H), 3.73-3.59 (m, 4H), 3.28-3.20 (m, 2H), 3.16 (s, 3H), 2.91-2.90 (m, [00223] [00223] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100mg, 0.34mmol) was dissolved in 5mL of dichloromethane, followed by the addition of triethylamine (101mg, 1mmol), (S)-2-hydroacrylic acid (46mg, 0.5mmol) ) and O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethylformamidinium hexafluorophosphate (190 mg, 0.5 mmol). After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product (S)-2-hydroxy-1- ((3aR,5R,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl )propan-1-one 17 (13 mg, 11.6% yield) as a pale yellow solid. [00224] [00224] MS m/z (ESI): 330.3 [M+1] 1 [00225] [00225] 1 H NMR (400 MHz, DMSO-d6): 11.61 (s, 1H), 8.09-8.07 (m, 1H), 7.12-7.10 (m, 1H), 6 .53 (s, 1H), 5.46-5.44 (m, 1H), 4.88-4.83 [00226] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (101 mg, 1.02 mmol). After reacting for 0.5 hour, the reaction mixture was added dropwise with 2-methoxyacetic acid (30 mg, 0.33 mmol). After reacting for 16 hours, the reaction mixture was added with 10 mL of H2O. The aqueous phase and the organic phase were separated. The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and purified by preparatory HPLC to obtain the title product 2-methoxy-1-((3aR,5s,6aS)-5 -(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)ethanone 18 (20 mg, yield 17 .9%) as a white solid. [00227] [00227] MS m/z (ESI): 330.3 [M+1] [00228] [00228] 1 H NMR (400 MHz, CDCl 3 ): 10.51 (s, 1H), 8.30 (s, 1H), 7.06 (s, 1H), 6.54 (s, 1H), 5. 66-5.58 (m, 1H), 4.07 (s, 2H), 3.84-3.72 (m, 2H), 3.50-3.46 (m, 4H), 3.45- 3.36 (m, 1H), 3.26 (s, 3H), 2.97-2.86 (m, 2H), 2.08-2.04 (m, 2H), 1.95-1, 93 (m, 2H) Example 19 (R)-2-Hydroxy-1-((3aR,5S,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)propan-1-one O H N oh N H N N N H [00229] [00229] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of (R)-2-hydroacrylic acid (46 mg, 0.5 mmol) and O-(7-azabenzotriazole) Hexafluorophosphate -1-yl)-N,N,N',N'-tetramethylformamidinium (190 mg, 0.5 mmol). After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product (R)-2-hydroxy- 1-((3aR,5S,6aS)-5-(methyl(7H-pyrrole[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H) -yl)propan-1-one 19 (6 mg, 5.4%) yield as a yellow solid. [00230] [00230] MS m/z (ESI): 330.4[M+1] 1 [00231] [00231] 1 H NMR (400 MHz, DMSO-d6): 11.61 (s, 1H), 8.09-8.08 [00232] tert-Butyl (2-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c ]pyrrole-2(1H)-yl)-2-oxoethyl)carbamate 16 (261 mg, 0.63 mmol) was dissolved in 8 mL of dichloromethane, followed by the dropwise addition of 2 mL of trifluoroacetic acid. After reacting for 1 hour, the reaction mixture was added with 10 mL of H2O and extracted with dichloromethane (20 mL×2). A 1 M sodium hydroxide solution was added to the aqueous phase to adjust the pH value to 9, and extracted with dichloromethane (30 ml×5). The organic phase was combined, washed with a saturated sodium chloride solution (15 ml), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-amino-1-((3aR,5s,6aS)-5 -(methyl(7H-pyrrolo[2,3-d]pyrimidin-4- [00233] [00233] MS m/z (ESI): 315.5 [M+1] 1 [00234] [00234] 1 H NMR (400 MHz, DMSO-d6): 11.93 (s, 1H), 8.30 (s, 2H), 8.13 (s, 1H), 7.21-7.20 (m , 1H), 6.63-6.62 (m, 1H), 5.49-5.45 (m, 1H), 3.83-3.65 (m, 4H), 3.40-3.35 (m, 2H), 3.20 (s, 3H), 2.93-2.83 (m, 2H), 2.03-1.97 (m, 2H), 1.83-1.81 (m) , 2H) Example 21 N-(2-((3aR,5s,6aS)-5-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[ c]pyrrole-2(1H)-yl)-2-oxoethyl)methanesulfonamide O H HN N Y O O N H N N N H O O [00235] [00235] 2-Amino-1-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c ]pyrrole-2(1H)-yl)ethanone 20 (60 mg, 0.19 mmol) was dissolved in 10 mL of dichloromethane, followed by dropwise addition of triethylamine (38 mg, 0.38 mmol) and methyl chloride. sulfonyl (24 mg, 0.29 mmol) in an ice bath. After reacting for 2 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product N-(2-((3aR,5s,6aS) )-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)-2-oxoethyl)methanesulfonamide 21 (8 mg, 11.3% yield) as a pale yellow solid. [00236] [00236] MS m/z (ESI): 393.3 [M+1] 1 [00237] [00237] 1 H NMR (400 MHz, DMSO-d6): 11.60 (s, 1H), 8.09 (s, 1H), 7.13-7.10 (m, 2H), 6.55-6 3.53 (m, 1H), 5.50-5.46 (m, 1H), 3.88-3.82 (m, 2H), 3.65-3.60 (m, 2H), 3.32 -3.25 (m, 2H), 3.15 (s, 3H), 2.96 (s, 3H), 2.93-2.73 (m, 2H), 1.99-1.94 (m , 2H), 1.78-1.75 (m, 2H) Example 22 4-((3aR,5s,6aS)-5-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino) hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)-4-oxobutanenitrile O H N N N H N N N H [00238] [00238] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (101 mg, 1 mmol), 3-cyanopropionic acid (37 mg, 0.37 mmol) and hexafluorophosphate. O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethylformamidinium (194 mg, 0.5 mmol). After reacting for 24 hours, the reaction mixture was added with 10 mL of saturated ammonium chloride to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (15 mL×2). The organic phase was combined, washed with a saturated solution of sodium chloride (15 ml), dried over anhydrous magnesium sulphate and filtered. The filtrate was concentrated under reduced pressure. [00239] [00239] MS m/z (ESI): 339.4[M+1] 1 [00240] [00240] 1 H NMR (400 MHz, DMSO-d6): 11.61 (s, 1H), 8.10-8.09 (m, 1H), 7.12-7.11 (m, 1H), 6 .54-6.53 (m, 1H), 3.69-3.57 (m, 2H), 3.36-3.35 (m, 6H), 3.24-3.21 (m, 1H) , 3.16-3.15 (m, 3H), 2.65-2.64 (m, 3H), 1.25-1.24(m, 3H) Example 23 2-Hydroxy-1-((3aR) ,5R,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)ethanone O H OH N N H N N N H O [00241] [00241] (3aR,6aS)-tert-Butyl 5-oxohexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 23a (1.0 g, 4.44 mmol) was dissolved in 20 mL of 2 M methylamine. After reacting for 1 hour, the reaction mixture was added with sodium cyanoborohydride (420 mg, 6.66 mmol) in batches. After reacting for 1 hour, the reaction mixture was concentrated under reduced pressure, followed by addition of 50 ml of dichloromethane to dissolve the residue, and then washed with a saturated solution of sodium chloride (20 ml). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain crude title product (3aR,5R,6aS)-tert-butyl 5-(methylamino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 23b ( 1.23 g, yellow oil), which was used directly in the next step without further purification. [00242] [00242] MS m/z (ESI): 241.3 [M+1] Step 2 (3aR,5R,6aS)-tert-Butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4) -yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate [00243] [00243] (3aR,5R,6aS)-tert-Butyl 5-(methylamino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 23b (1.06 g, 4.41 mmol) was dissolved in 20 mL of n-butanol, followed by the addition of 4-chlorine-7H-pyrrolo[2,3-d]pyrimidine (670 mg, 4.41 mmol) and potassium carbonate (1.22 g, 8.82 mmol). After reacting for 24 hours at 110°C, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography with elution system A to obtain the title product (3aR,5R,6aS )-tert-butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 23c (830 mg , 52.9%) yield as a pale yellow solid. [00244] [00244] MS m/z (ESI): 358.2 [M+1] Step 3 N-Methyl-N-((3aR,5R,6aS)-octahydro-cyclopenta[c]pyrrole-hydrochloride 5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine [00245] [00245] (3aR,5R,6aS)-tert-Butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2 (1H)-carboxylate 23c (830 mg, 2.32 mmol) was dissolved in 10 mL of a 6 M solution of hydrogen chloride in methanol. After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure, washed with anhydrous diethyl ether (20 mL), and dried in vacuo to obtain the title product N-methyl-N-((( 3aR,5R,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine 23d (630 mg, 92.6% yield ) as a gray solid. Step 4 2-Hydroxy-1-((3aR,5R,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c] pyrrole-2(1H)-yl)ethanone [00246] [00246] N-Methyl-N-((3aR,5R,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 23d (100 mg, 0.34 mmol) was dissolved in 5 mL of n-butanol, followed by the addition of 2-hydroxy methyl acetate (61 mg, 0.68 mmol) and 1,8-diazabicyclobicyclo[5 ,4.0]-7-hendecene (103 mg, 0.68 mmol). After reacting for hours at 60°C, the reaction mixture was heated to 60°C for 10 hours. The reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-hydroxy-1-((3aR,5R,6aS)-5- (methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)ethanone 23 (8 mg, yield 7, 5%) as a gray solid. [00247] [00247] MS m/z (ESI): 316.2 [M+1] 1 [00248] [00248] 1 H NMR (400 MHz, DMSO-d6): 11.61 (s, 1H), 8.09 (s, 1H), 7.12 (s, 1H), 6.58 (s, 1H), 5.32-5.30 (m, 1H), 4.52-4.49 (m, 1H), 4.08-3.99 (m, 2H), 3.54-3.51 (m, 2H ), 3.42-3.37 (m, 2H), 3.16 (s, 3H), 2.75-2.63 (m, 2H), 2.02-1.99 (m, 2H), 1.57-1.52 (m, 2H) Example 24 Methyl 2-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)he - xahydro-cyclopenta[c]pyrrole-2(1H)-yl)acetate H O N O N H N N N [00249] [00249] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (101 mg, 1 mmol) and 2-methyl bromoacetate (54 mg, 0.35 mmol). After reacting for 16 hours, the reaction mixture was added with 15 mL of saturated ammonium chloride to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (15 mL×2). The organic phase was combined, washed with a saturated solution of sodium chloride (25 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product methyl 2-((3aR,5s,6aS)-5-(methyl(7H-pyrrole) [2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrol-2(1H)-yl)acetate 24 (6 mg, 5.4%) yield as a solid White. [00250] [00250] MS m/z (ESI): 330.4[M+1] 1 [00251] [00251] 1 H NMR (400 MHz, CDCl 3 ): 10.61 (s, 1H), 8.27-8.26 (m, 1H), 7.10-7.09 (m, 1H), 6.91 (s, 1H), 5.49-5.46 (m, 1H), 3.78 (s, 3H), 3.40-3.33 (m, 2H), 3.23 (s, 3H), 2.90-2.83 (m, 4H), 2.57-2.55 (m, 1H), 2.02-1.94 (m, 2H), 1.83-1.78 (m, 2H) ), 1.47-1.46 (m, 1H) Example 25 [00252] [00252] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (101 mg, 1 mmol) and isopropyl chloroformate (46 mg, 0.37 mmol). After reacting for 16 hours, the reaction mixture was added with 15 mL of saturated ammonium chloride to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (15 mL×2). The organic phase was combined, washed with a saturated sodium chloride solution (15 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product (3aR,5s,6aS)-isopropyl 5-(methyl(7H-pyrrole[2, 3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate 24 (6 mg, 5.1% yield) as a white solid. [00253] [00253] MS m/z (ESI): 344.4[M+1] 1 [00254] 1 H NMR (400 MHz, CDCl 3 ): 10.61 (s, 1H), 8.34 (s, 1H), 7.10-7.09 (m, 1H), 6.60-6.59 (m, 1H), 5.64-5.60 (m, 1H), 5.01-4.95 (m, [00255] [00255] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (101 mg, 1 mmol) and bromoacetonitrile (41 mg, 0.34 mmol). After reacting for 24 hours, the reaction mixture was added with a small amount of a saturated ammonium chloride solution to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (10 mL×3). The organic phase was combined, washed with a saturated solution of ammonium chloride (10 ml) and a saturated solution of sodium chloride (10 ml) successively, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-((3aR,5s,6aS)-5-(methyl(7H-pyrrole[ 2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrol-2(1H)-yl)acetonitrile 26 (10 mg, 9.9%) yield [00256] [00256] MS m/z (ESI): 297.2 [M+1] 1 [00257] [00257] 1 H NMR (400 MHz, CDCl 3 ): 10.43 (s, 1H), 8.33-8.32 (m, 1H), 7.10-7.09 (m, 1H), 6.72 -6.71 (m, 1H), 5.50-5.43 (m, 1H), 3.22 (s, 3H), 2.87-2.86 (m, 4H), 2.73-2 .66 (m, 2H), 2.09-2.04 (m, 2H), 1.85-1.80 (m, 4H) Example 27 3-((3aR,5s,6aS)-5-(Methyl (7H-pyrrole[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2(1H)-yl)propanenitrile N H N N H N N N [00258] [00258] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 5 mL of dichloromethane, followed by the addition of triethylamine (101 mg, 1 mmol) and 3-bromo propionitrile (46 mg, 0.34 mmol). After reacting for 24 hours, the reaction mixture was added with a small amount of a saturated ammonium chloride solution to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (10 mL×3). The organic phase was combined, washed with a saturated solution of ammonium chloride (10 ml) and a saturated solution of sodium chloride (10 ml) successively, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 3-((3aR,5s,6aS)-5-(methyl(7H-pyrrole[ 2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrol-2(1H)-yl)propanenitrile 27 (10 mg, 9.4%) yield as a white solid . [00259] [00259] MS m/z (ESI): 311.3 [M+1] 1 [00260] [00260] H NMR (400 MHz, CDCl3): 10.77 (s, 1H), 8.35-8.34 (m, 1H), 7.12-7.11 (m, 1H), 6.75 -6.74 (m, 1H), 5.50-5.41 (m, 1H), 3.22 (s, 3H), 2.83-2.80 (m, 4H), 2.63-2 .59 (m, 4H), 1.98-1.81 (m, 4H), 1.81-1.78 (m, 2H) Examples 28, 29 N-((3aR,5s,6aS)-2- (4,6-Dichloropyrimidin-2-yl)octahydro-cyclopenta[c]pyrrol-5-yl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine N-( (3aR,5s,6aS)-2-(2,6-Dichloropyrimidin-4-yl)octahydro-cyclopenta[c]pyrrol-5-yl)-N-methyl-7H-pyrrolo[2,3- d]pyrimidin-4-amine Cl Cl [00261] [00261] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (55 mg, 0.2 mmol) was dissolved in 5 mL of ethanol, followed by the addition of triethylamine (52 mg, 0.5 mmol). After reacting for 0.5 hour, the mixture re- [00262] [00262] MS m/z (ESI): 404.3 [M+1] 1 [00263] 1 H NMR (400 MHz, CDCl 3 ): 9.81 (s, 1H), 8.28 (s, 1H), 7.01 (s, 1H), 6.57 (s, 1H), 6. 50 (d, 1H), 5.64-5.60 (m, 1H), 3.90-3.85 (m, 2H), 3.59-3.56 (m, 2H), 3.25 ( s, 3H), 3.04-2.96 (m, 2H), 2.13-2.05 (m, 2H), 1.99-1.95 (m, 2H) 1 [00264] 1 H NMR (400 MHz, CDCl 3 ): 9.97 (s, 1H), 8.28 (s, 1H), 7.04 (s, 1H), 6.52 (d, 1H), 6. 26 (d, 1H), 5.69-5.65 (m, 1H), 3.96-3.92 (m, 1H), 3.68-3.64 (m, 2H), 3.28- 3.24 (m, 4H), 3.06-3.01 (m, 2H), 2.09-2.00 (m, 2H), 1.98-1.95 (m, 2H) Example 30 ( 3aR,5s,6aS)-Methyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate O H O N N H N N N H [00265] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (100 mg, 0.34 mmol) was dissolved in 15 mL of dichloromethane, followed by the addition of methyl chloroformate (44 mg, 0.47 mmol) and 3-triethylamine (59 mg, 0.58 mmol) in one ice bath. After reacting for 16 hours, the reaction mixture was added with a small amount of a saturated sodium bicarbonate solution and extracted with dichloromethane (20 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product (3aR,5s,6aS)-methyl 5-(methyl(7H-pyrrole[ 2,3-d]pyrimidin-4-yl)amino)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate 30 (50 mg, 41.0%) yield as a white solid. [00266] [00266] MS m/z (ESI): 316.3 [M+1] 1 [00267] [00267] 1 H NMR (400 MHz, CDCl 3 ): 10.60 (s, 1H), 8.26 (s, 1H), 7.06-7.05 (m, 1H), 6.54-6.53 (m, 1H), 5.60-5.56 (m, 1H), 3.72 (s, 3H), 3.70-3.65 (m, 2H), 3.32-3.25 (m) , 2H), 3.24 (s, 3H), 2.91-2.85 (m, 2H), 2.06-2.00 (m, 2H), 1.92-1.82 (m, 2H) ) Example 31 2-Hydroxy-1-((1R,5S,6s)-6-((methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)methyl)-3-azabicyclo [3.1.0]hexan-3-yl)ethanone O oh N H H N N N N H O O HN [00268] [00268] (1R,5S,6r)-3-Azabicyclo[3.1.0]hexan-6-ylmethanol 31a (10 g, 88.4 mmol) was dissolved in a mixed solvent of dioxane and H2O (V/V =3/2), followed by the addition of sodium hydroxide (4.2 g, 106 mmol) and di-tert-butyl dicarbonate (28.9 g, 132.6 mmol). After reacting for hours, 50 mL of H2O was added to the reaction mixture. The reaction mixture was extracted with ethyl acetate (150 mL×3). The organic phase was combined, washed with a saturated solution of sodium chloride (50 mL×2), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography over silica gel with elution system C to obtain the title product (1R,5S,6r)-tert-butyl 6- [00269] [00269] Oxalyl chloride (2.41 Ml, 28.1 mmol) was dissolved in 100 ml of dichloromethane, followed by dropwise addition of dimethyl sulfoxide (4.32 ml, 60.8 mmol) at -78°C. After reacting for 30 minutes, (1R,5S,6r)-tert-butyl 6-(hydroxymethyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate 31b (5.0 g, 23 .4 mmol) was added to the reaction mixture. After reacting for 1 hour, triethylamine (16.23 mL, 117 mmol) was added to the reaction mixture. After reacting for 30 minutes at room temperature, 200 ml dichloromethane, 50 ml H2O, 50 ml 1M hydrochloric acid and 50 ml saturated sodium bicarbonate solution were added to the reaction mixture. The aqueous phase and the organic phase were separated, the organic phase was washed with a saturated solution of sodium chloride (50 ml), dried over anhydrous sodium sulphate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography over silica gel with elution system C to obtain the title product (1R,5S)-tert-butyl 6-formyl-3-azabicyclo[3 .1.0]hexane-3-carboxylate 31c (4.12 g, 82.4%) yield as a pale yellow liquid. Step 3 (1R,5S,6s)-tert-Butyl 6-((methylamino)methyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate [00270] [00270] (1R,5S)-tert-Butyl 6-formyl-3-azabicyclo[3.1.0]hexane-3-carboxylate 31c (3.05 g, 14.4 mmol) was dissolved in a 20 mL of a solution of methylamine in methanol. After reacting for 24 hours, sodium cyanoborohydride (1.09 g, 17.3 mmol) was added to the reaction mixture in an ice bath in batches. After reacting for 2 hours at room temperature, 15 ml of a saturated solution of clo- [00271] [00271] MS m/z (ESI): 227.46 [M+1] Step 4 (1R,5S,6s)-tert-Butyl 6-((methyl(7H-pyrrolo[2,3-d]pyrimidin-) 4-yl)amino)methyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate [00272] [00272] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (2.21 g, 14.4 mmol) was dissolved in 320 mL of n-butanol, followed by the addition of (1R,5S,6s )-tert-butyl 6-((methylamino)methyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate 31d (3.25 g, 14.4 mmol) and potassium carbonate (3.97 g , 28.8 mmol). After reacting for 24 hours at 120°C, 150 ml of ethyl acetate and 20 ml of H2O were added to the reaction mixture. The aqueous phase and the organic phase were separated, the organic phase was washed with a saturated solution of sodium chloride (20 ml), dried over anhydrous sodium sulphate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography on silica gel with elution system A to obtain the title product (1R,5S,6s)-tert-butyl 6-((methyl) (7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)methyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate 31e (1.82 g, 36.8% yield ) as a white solid. [00273] [00273] MS m/z (ESI): 344.2 [M+1] Step 5 N-((1R,5S,6r)-3-Azabicyclo[3.1.0]hexan-6-ylmethyl) hydrochloride -N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine [00274] [00274] (1R,5S,6s)-tert-Butyl 6-((methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)methyl)-3-azabicyclo[3.1.0 ]hexane-3-carboxylate 31e (1.72 g, 5.01 mmol) was dissolved in 20 mL of a 1 M solution of hydrogen chloride in methanol. After reacting for 14 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was washed with a mixed solvent of dichloromethane and diethyl ether (V/V=1/1), and dried in vacuo to obtain the product from the title N-((1R,5S,6r)-3-azabicyclo[3.1.0]hexan-6-ylmethyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-hydrochloride amine 31f (1.17 g, 95.9%) yield as a gray solid. [00275] [00275] MS m/z (ESI): 244.46 [M+1] Step 6 2-Hydroxy-1-((1R,5S,6s)-6-((methyl(7H-pyrrole[2,3-) d]pyrimidin-4-yl)amino)methyl)-3-azabicyclo[3.1.0]hexan-3-yl)ethanone [00276] [00276] N-((1R,5S,6r)-3-Azabicyclo[3.1.0]hexan-6-ylmethyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 31f (100 mg, 0.36 mmol) was dissolved in 5 mL of tetrahydrofuran, followed by the addition of triethylamine (111 mg, 1.1 mmol) and O-(7-azabenzotriazol-1-yl) hexafluorophosphate -N,N,N',N'-tetramethylformamidinium (204 mg, 0.54 mmol). After reacting for 30 minutes, 2-glycolic acid (30 mg, 0.39 mmol) was added to the reaction mixture. After reacting for 16 hours, a small amount of a saturated ammonium chloride solution was added to the reaction mixture to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (10 ml×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-hydroxy-1-((1R,5S,6s)-6-((methyl) (7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)methyl)-3-azabicyclo[3.1.0]hexan-3- [00277] [00277] MS m/z (ESI): 300.45 [M-1] 1 [00278] [00278] H NMR (400 MHz, CDCl3): 10.64(s, 1H), 8.27 (s, 1H), 7.12-7.11 (m, 1H), 6.61-6.60 (m, 1H), 5.34-5.33 (m, 1H), 4.09-4.04 (m, 2H), 3.83-3.81 (m, 2H), 3.49 (s , 3H), 1.79-1.72 (m, 2H), 1.53-1.45(m, 2H), 1.31-1.28 (m, 1H), 1.00-0.89 (m, 2H) Example 32 2-((1R,5S,6r)-6-((Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)methyl)-3-a-zabicyclo [3.1.0]hexan-3-yl)acetonitrile N N H H N N N N H [00279] [00279] N-((1R,5S,6r)-3-Azabicyclo[3.1.0]hexan-6-ylmethyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 31f (100 mg, 0.36 mmol) was dissolved in 5 mL acetonitrile, followed by the addition of triethylamine (109 mg, 1.1 mmol) and bromoacetonitrile (47.5 mg, 0.4 mmol). After reacting for 16 hours, a small amount of a saturated solution of ammonium chloride was added to the reaction mixture to quench the reaction. The aqueous phase and the organic phase were separated. The aqueous phase was extracted with dichloromethane (10 ml×3). The organic phase was combined, washed with a saturated solution of sodium chloride (10 ml), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product 2-((1R,5S,6r)-6-((methyl) (7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)methyl)-3-azabicyclo[3.1.0]hexan-3-yl)acetonitrile 32 (15 mg, 14.9% yield ) as a white solid. [00280] [00280] MS m/z (ESI): 283.2 [M+1] 1 [00281] [00281] 1 H NMR (400 MHz, CDCl 3 ): 10.64 (s, 1H), 8.31 (s, 1H), 7.09-7.08 (m, 1H), 6.64-6.63 (m, 1H), 3.76-3.74 (m, 2H), 3.63 (s, 2H), 3.47 (s, 3H), 3.00-2.98 (m, 2H), 2.75-2.73 (m, 2H), 1.57-1.53 (m, 2H), 1.33-1.30 (m, 1H) Example 33 N-(((1R,5S,6s) )-3-(2-Chloropyrimidin-4-yl)-3-azabicyclo[3.1.0]hexan-6-yl)methyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4 -amine N Cl N N H H N N N N [00282] [00282] N-((1R,5S,6r)-3-Azabicyclo[3.1.0]hexan-6-ylmethyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 31f (100 mg, 0.36 mmol) was dissolved in 5 mL of ethanol, followed by the addition of triethylamine (108 mg, 1.07 mmol) and 4,6-dichloropyrimidine (26.6 mg, 0, 17 mmol). After reacting for 16 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain the title product N-(((1R,5S) ,6s)-3-(2-chloropyrimidin-4-yl)-3-azabicyclo[3.1.0]hexan-6-yl)methyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin -4-amine 33 (5 mg, 7.9%) yield as a white solid. [00283] [00283] MS m/z (ESI): 356.1 [M+1] 1 [00284] [00284] 1 H NMR (400 MHz, CDCl 3 ): 10.46 (s, 1H), 8.25 (s, 1H), 7.97 (s, 1H), 7.07 (s, 1H), 6. 59 (s, 1H), 6.13 (s, 1H), 4.03-4.01 (m, 1H), 3.84-3.81 (m, 2H), 3.49-3.47 ( m, 3H), 3.42 (s, 3H), 1.96-1.94 (m, 2H), 1.07-1.05 (m, 1H) Example 34 (3aR,5s,6aS)-N -(3-Methoxy-1,2,4-thiadiazol-5-yl)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[ c]pyrrole-2(1H)-carboxamide O The N N H N Y N H N H N N N H [00285] [00285] 3-Methoxy-1,2,4-thiadiazol-5-amine 34a (500 mg, 3.82 mmol) and phenyl carbon hydrochloride 34b (600 mg, 3.82 mmol) were dissolved in mL of dichloromethane, followed by addition of triethylamine (0.8 mL, 5.73 mmol). After reacting for 16 hours, 30 mL of H2O was added to the reaction mixture to dilute the solution. The aqueous phase and the organic phase were separated, the aqueous phase was extracted with dichloromethane (20 ml×2), and the organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography on silica gel with elution system A to obtain the title product phenyl (3-methoxy-1,2,4-thiadiazole- 5-yl)carbamate 34c (200 mg, 20.8%) yield as a white solid. [00286] [00286] MS m/z (ESI): 252.0 [M+1] Step 2 (3aR,5s,6aS)-N-(3-Methoxy-1,2,4-thiadiazol-5-yl)-5 -(methyl(7H-pyrrole[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxamide [00287] [00287] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (120 mg, 0.47 mmol) was dissolved in 15 mL of tetrahydrofuran, followed by the addition of phenyl(3-methoxy-1,2,4-thiadiazol-5-yl)carbamate 34c (117 mg, 0.47 mmol) and triethylamine (0.13 mL, 0.94 mmol). After reacting for 5 hours at 60°C, the reaction mixture was added with 30 mL of H2O and extracted with dichloromethane (50 mL×3). The organic phase was combined, washed with a saturated sodium chloride solution (50×2 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography on silica gel with elution system A to obtain the title product (3aR,5s,6aS)-N-(3-methoxy -1,2,4-thiadiazol-5-yl)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2 (1H)-carboxamide 34 (50 mg, 25.9%) yield as a white solid. [00288] [00288] MS m/z (ESI): 412.9 [M-1] 1 [00289] [00289] 1 H NMR (400 MHz, DMSO-d6): 11.60(m, 2H), 8.08(s, 1H), 7.06-7.05(m, 1H), 6.53-6 1.51(m, 1H), 5.48-5.44(m, 1H), 3.90(s, 3H), 3.69-3.65(m, 2H), 3.37-3.32 (m, 2H), 3.16(s, 3H), 2.90-2.88(m, 2H), [00290] [00290] (4aS,7aS)-Tetrahydro-1H-cyclopenta[c]pyridin-3,6(2H,4H)-dione 35a (3 g, 19.60 mmol, prepared by a well known method ( "Tetrahedron: Asymmetry, 1997, 8(17), 2893-2904") and N-methyl-1-phenylmethanamine 35b (2.37 g, 19.60 mmol) were dissolved in 5 mL of methanol, followed by the addition of two drops of acetic acid. After stirring and reacting for 3 hours, sodium cyanoborohydride (1.80 g, 29.40 mmol) was added to the reaction mixture. After reacting for 12 hours, the reaction mixture was filtered, and concentrated under reduced pressure and the resulting residue was purified by column chromatography on silica gel with elution system A to obtain the title product (4aS,7aS)-6-(Benzyl(methyl)amino)hexahydro- 1H-cyclopenta[c]pyridin-3(2H)-one 35c (1.99 g, 39.8%) yield as a yellow solid. [00291] [00291] MS m/z (ESI): 259.2 [M+1] Step 2 (4aR,7aS)-N-Benzyl-N-methyloctahydro-1H-cyclopenta[c]pyridin-6-amine [00292] [00292] (4aS,7aS)-6-(Benzyl(methyl)amino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 35c (3.90 g, 15.11 mmol) was dissolved in 150 mL of tetrahydrofuran, followed by the addition of mixed lithium aluminum anhydride (3 g, 78.60 mmol) in batches in an ice bath. After stirring for 48 hours, 10 mL of ice water was added to the reaction mixture. The reaction mixture was filtered, washed with dichloromethane (50 ml) and concentrated under reduced pressure to obtain the crude title product (4aR,7aS)-N-benzyl-N-methyloctahydro-1H-cyclopenta[c]pyridine -6-amine 35d (3.69 g, white solid), which was used directly in the next step without further purification. [00293] [00293] MS m/z (ESI): 245.2 [M+1] Step 3 (4aR,7aS)-tert-Butyl 6-(benzyl(methyl)amino)hexahydro-1H-cyclopenta[c ]pyridin-2(3H)-carboxylate [00294] [00294] (4aR,7aS)-N-Benzyl-N-methyloctahydro-1H-cyclopenta[c]pyridin-6-amine 35d (3.69 g, 15.11 mmol) was dissolved in 50 mL of dichloromethane, followed by the addition of di-tert-butyl dicarbonate (4.94 g, 22.60 mmol) and ethylamine (3.81 g, 37.75 mmol). After stirring for 12 hours, the reaction mixture was added with 20 mL of H2O and extracted with dichloromethane (30 mL×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography on silica gel with elution system A to obtain the title product (4aR,7aS)-tert-butyl 6-(benzyl(methyl) amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate 35e (3.28 g, 63% yield) as a pale yellow solid. [00295] [00295] MS m/z (ESI): 345.3 [M+1] Step 4 (4aR,7aS)-tert-Butyl 6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-2(3H) )-carboxylate [00296] [00296] (4aR,7aS)-tert-Butyl 6-(benzyl(methyl)amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate 35e (2 g, 5.81 mmol) was dissolved in 150 mL of methanol, followed by the addition of palladium hydroxyde on carbon (500 mg, 25%). After the reactor was purged with hydrogen three times, the reaction mixture was stirred for 72 hours, then filtered, washed with methanol (20 mL) and concentrated under reduced pressure to obtain the crude title product (4aR,7aS )-tert-butyl 6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate 35f (1.72 g) as a colorless viscous liquid, which was used directly in the next step without purification later. [00297] [00297] MS m/z (ESI): 255.2 [M+1] Step 5 (4aS,7aR)-tert-Butyl 6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) )amino) hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate [00298] [00298] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (1.78 g, 11.65 mmol) was dissolved in 50 mL of 1,4-dioxane, followed by the addition of (4aR,7aS )-tert-butyl 6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate 35f (2.96 g, 11.65 mmol) and potassium carbonate (3.22 g, 23.30 mmol). After reacting for 48 hours at 110°C, the reaction mixture was concentrated under reduced pressure, followed by the addition of 50 mL of H2O, and extracted with ethyl acetate (30 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography over silica gel with elution system A to obtain the title product (4aS,7aR)-tert-butyl 6-(methyl( 7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate 35g (1.34 g, 31% yield) as one white solid. [00299] [00299] MS m/z (ESI): 372.2 [M+1] Step 6 N-Methyl-N-((4aS,7aR)-octahydro-1H-cyclopenta[c]pyridin-6-hydrochloride il)-7H-pyrrolo[2,3-d]pyrimidin-4-amine [00300] [00300] (4aS,7aR)-tert-Butyl 6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-1H-cyclopenta[c]pyridin-2(3H) )-carboxylate 35g (1.34 g, 3.61 mmol) was dissolved in 15 mL of a 6 M solution of hydrogen chloride in a methanol solution. After reacting for 12 hours, the reaction mixture was concentrated under reduced pressure, followed by the addition of 20 mL of H2O and a 10% sodium hydroxide solution until the pH of the reaction mixture was between 9 and 10. - tion was extracted with dichloromethane (20 mL×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the crude title product N-methyl-N-((4aS,7aR)-octahydro-1H-cyclopenta[c]pyridin-6-yl)-7H-pyrrolo[hydrochloride] 2,3-d]pyrimidin-4-amine 35h (1 g, white solid), which was used directly in the next step without further purification. [00301] [00301] MS m/z (ESI): 272.2 [M+1] Step 7 1-((4aS,7aR)-6-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) )amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H,4H,4aH,5H,6H,7H,7aH)-yl)ethanone [00302] N-Methyl-N-((4aS,7aR)-octahydro-1H-cyclopenta[c]pyridin-6-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine hydrochloride 35h (100 mg, [00303] [00303] MS m/z (ESI): 314.2 [M+1] [00304] [00304] 1 H NMR (400 MHz, CDCl3): δ 11.34 (s, 1H), 8.32 (s, 1H), 7.08 (s, 1H), 6.56 (s, 1H), 5.43-5.45 (m, 1H), 3.88-3.91 (m, 1H), 3.27-3.55 (m, 6H), 2.28-2.30 (m, 2H) ), 1.91-1.96 (m, 6H), 1.58-1.60 (m, 3H) Example 36 (4aS,7aR)-Methyl 6-(methyl(7H-pyrrole[2,3-d) ]pyrimidin-4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxylate O O HN N H N N N H O [00305] N-Methyl-N-((4aS,7aR)-octahydro-1H-cyclopenta[c]pyridin-6-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine hydrochloride 35h (100 mg, [00306] [00306] MS m/z (ESI): 330.3 [M+1] [00307] 1 H NMR (400 MHz, CDCl 3 ): δ 10.57 (s, 1H), 8.24 (s, 1H), 1 7.05 (s, 1H), 6.56 (s, 1H), 5.38-5.39 (m, 1H), 3.70 (s, 3H), 3.61-3.62 (m, 2H), 3.39-3.40 (m, 1H), 3. 23-3.27 (m, 4H), 2.20-2.23 (m, 2H), 2.05-2.09 (m, 1H), 1.83-1.84 (m, 2H), 1.52-1.57 (m, 3H) Example 37 2-Hydroxy-1-((4aS,7aR)-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-1H-cyclopenta[c]pyridin-2(3H,4H,4aH,5H,6H,7H,7aH)-yl)ethanone O HNOH N H N N N H O [00308] [00308] N-Methyl-N-((4aS,7aR)-octahydro-1H- hydrochloride [00309] [00309] MS m/z (ESI): 330.2 [M+1] [00310] 1 H NMR (400 MHz, CDCl 3 ): δ 11.57 (s, 1H), 8.30 (s, 1H), 7.07 (s, 1H), 6.56 (s, 1H), 5.40-5.44 (m, 1H), 4.14-4.18 (m, 1H), 3.34-3.39 (m, 2H), 3.27(s, 3H), 3. 18-3.22(m, 1H), 2.30-2.32 (m, 2H), 1.95-2.13 (m, 3H), 1.66-1.68 (m, 1H), 1.52-1.57 (m, 2H) Example 38 (4aS,7aR)-N-(3-Methoxy-1,2,4-thiadiazol-5-yl)-6-(methyl(7H-pyrrole[2]) ,3-d]pyrimidin-4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H)-carboxamide the H N H N N s AT THE N H N N N H the H [00311] [00311] N-Methyl-N-((4aS,7aR)-octahydro-1H- hydrochloride [00312] [00312] MS m/z (ESI): 429.3 [M+1] 1 [00313] [00313] 1 H NMR (400 MHz, DMSO-d6): 11.63 (s, 1H), 11.61 (s, 1H), 8.08 (s, 1H), 7.12 (d, 1H), 6.58 (d, 1H), 5.26-4.30 (m, 1H), 3.84 (s, 3H), 3.69-3.73 (m, 1H), 3.59-3, 64 (m, 1H), 3.38-3.40 (m, 2H), 3.17 (s, 3H), 2.20-2.23 (m, 2H), 1.82-1.92 ( m, 3H), 1.60-1.65 (m, 2H), 1.40-1.48 (m, 1H) Example 39 3-((4aS,7aR)-6-(Methyl(7H-pyrrole[ 2,3-d]pyrimidin-4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-2(3H,4H,4aH,5H,6H,7H,7aH)-yl)-3-oxopropanenitrile O H N CN N H N N N H O [00314] N-Methyl-N-((4aS,7aR)-octahydro-1H-cyclopenta[c]pyridin-6-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine hydrochloride 35h (100mg, 0.37mmol) was dissolved in 5mL of n-butanol, followed by the addition of 2-ethyl cyanoacetate (83mg, 0.74mmol) and DBU (113mg, 0.74mmol). After reacting for 15 hours at 50°C, the reaction mixture was concentrated under reduced pressure, extracted with ethyl acetate (20 mL×2) and washed with a saturated sodium chloride solution (15 mL×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparative HPLC to obtain the title product 3-((4aS,7aR)-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-1H-cyclopenta[c]pyridin-2(3H,4H,4aH,5H,6H,7H,7aH)-yl)-3-oxopropanenitrile 39 (35 mg, 28.0%) yield as a light pink solid. [00315] [00315] MS m/z (ESI): 339.3 [M+1] [00316] [00316] 1 H NMR (400 MHz, DMSO-d6): δ 11.6 (s, 1H), 8.09 (s, 1H), 1 7.13 (s, 1H), 6.58 (s, 1H ), 5.27-5.29 (m, 1H), 4.01-4.09 (m, 2H), 3.60-3.63 (m, 1H), 3.43-3.48 (m) , 2H), 3.18 (s, 3H), 2.08-2.10 (m, 2H), 1.81-1.88 (m, 3H), 1.23-1.43 (m, 4H) ) Examples 40, 41 (4aR,6R,7aR)-6-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-1H-cyclopenta[c]pyridin-3( 2H)-one (4aR,6S,7aR)-6-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-1H-cyclopenta[c]pyridin-3(2H) )-one H H N H O H N O N H N H N N N N H N N H H H H N H N [00317] [00317] (4aS,7aS)-Tetrahydro-1H-cyclopenta[c]pyridin-3,6(2H,4H)-dione 35a (1 g, 6.54 mmol, prepared by well known methods " Tetrahedron : Asymmetry, 1997, 8 (17), 2893-2904" and methylamine hydrochloride (200 mg, 6.54 mmol) were dissolved in 20 mL of methanol, followed by the addition of two drops of acetic acid. After stirring and reacting for 1 hour, sodium cyanoborohydride (600 mg, 9.68 mmol) was added to the reaction mixture. After reacting for 12 hours, the reaction mixture was filtered, and concentrated under reduced pressure to obtain the title product (4aS ,7aS)-6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 40a (1 g) as a brown oil, which was used directly in the next step without further purification. [00318] [00318] MS m/z (ESI): 169.2 [M+1] Step 2 (4aR,6R,7aR)-6-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino) hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one [00319] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (910 g, 5.95 mmol) was dissolved in 30 mL of H2O, followed by the addition of (4aS,7aS)-6-(methylamino )hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 40a (1 g, 5.95 mmol) and potassium carbonate (1.6 g, 11.59 mmol). After reacting for 12 hours at 100°C, the reaction mixture was concentrated under reduced pressure, added with 50 mL of H2O, and extracted with ethyl acetate (30 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by chiral preparatory HPLC to obtain the title products (4aR,6R,7aR)-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexa -hydro-1H-cyclopenta[c]pyridin-3(2H)-one 40 (15 mg, 0.9% yield) as a white solid and (4aR,6S,7aR)-6-(methyl(7H) - pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 41 (15 mg, 0.9% yield) as a solid White. [00320] [00320] MS m/z (ESI): 286.4 [M+1] [00321] 1 H NMR (400 MHz, CDCl 3 ): δ 11.37 (s, 1H), 8.28 (s, 1H), 7.06 (s, 1H), 6.98 (s, 1H), 6.53 (s, 1H), 5.24-5.27 (m, 1H), 3.41-3.44 (m, 1H), 3.21 (s, 3H), 3.10-3, 13 (m, 1H), 2.56-2.58 (m, 1H), 2.50-2.52 (m, 2H), 2.30-2.31 (m, 1H), 2.07- 2.09 (m, 2H), 1.50-1.64 (m, 2H) [00322] 1 H NMR (400 MHz, CDCl 3 ): δ 11.37 (s, 1H), 8.28 (s, 1H), 7.06 (s, 1H), 6.98 (s, 1H), 6.53 (s, 1H), 5.24-5.27 (m, 1H), 3.41-3.44 (m, 1H), 3.21 (s, 3H), 3.10-3, 13 (m, 1H), 2.56-2.58 (m, 1H), 2.50-2.52 (m, 2H), 2.30-2.31 (m, 1H), 2.07- 2.09 (m, 2H), 1.50-1.64 (m, 2H) Example 42 (4aR,6R,7aR)-2-Methyl-6-(methyl(7H-pyrrole[2,3-d]) pyrimidin-4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one HN O N H N N N H [00323] [00323] In an ice bath, (4aS,7aS)-6-(benzyl(methyl)amino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 35c (400mg, 1.55 mmol) was dissolved in 10 mL of tetrahydrofuran, followed by the addition of sodium hydride (56 mg, 2.33 mmol). After stirring for 30 minutes, the reaction mixture was added with methyl iodide (241 mg, 1.71 mmol). After reacting for 12 hours, the reaction mixture was added with 50 mL of H2O and extracted with ethyl acetate (20 mL×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the crude title product (4aS,7aS)-6-(benzyl(methyl)amino)-2-methyl-hexahydro-1H-cyclopenta[c]pyridin-3(2H) -one 42a (500 mg) as a colorless liquid, which was used directly in the next step without further purification. [00324] [00324] MS m/z (ESI): 273.2 [M+1] Step 2 (4aS,7aS)-2-Methyl-6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-3( 2H)-one [00325] [00325] (4aS,7aS)-6-(Benzyl(methyl)amino)-2-methyl-hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 42a (500 g, 1.84 mmol ) was dissolved in mL methanol, followed by addition of palladium hydroxide on charcoal (125 mg, 25%). After the reactor was purged with hydrogen three times, the reaction mixture was stirred for 6 hours, then filtered, washed with methanol (10 mL) and concentrated under reduced pressure to obtain the crude title product (4aS,7aS )-2-methyl-6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 42b (300 mg) as a colorless oil, which was used directly in the next step without further purification. [00326] [00326] MS m/z (ESI): 183.2 [M+1] Step 3 (4aR,6R,7aR)-2-Methyl-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)amino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one [00327] [00327] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (168 mg, 1.10 mmol) was dissolved in 20 mL of H2O, followed by the addition of (4aS,7aS)-2-methyl- 6-(methylamino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 42b (200mg, 1.10mmol) and potassium carbonate (303mg, 2.20mmol). After reacting for 48 hours at 100°C, the reaction mixture was added with 30 mL of H2O and extracted with ethyl acetate (20 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by preparative HPLC to obtain the title product (4aR,6R,7aR)-2-methyl-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) ) amino)hexahydro-1H-cyclopenta[c]pyridin-3(2H)-one 42 (8 mg, 5% yield) as a white solid. [00328] [00328] MS m/z (ESI): 300.3 [M+1] [00329] 1 H NMR (400 MHz, CDCl 3 ): δ 11.61 (s, 1H), 8.09 (s, 1H), 7.12 (s, 1H), 6.56 (s, 1H), 5.10-5.15 (m, 1H), 3.40-3.44 (m, 1H), 3.15-3.16 (m, 1H), 3.11 (s, 3H), 2, 89 (s, 3H), 2.40-2.44 (m, 3H), 2.14-2.16 (m, 1H), 1.90-1.92 (m, 2H), 1.35- 1.47 (m, 2H) Examples 43, 45 2-Hydroxy-1-((3aS,5R,7aR)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)ami- [00330] [00330] (3aR,7aS)-tert-Butyl 5-oxohexahydro-1H-isoindol-2(3H)-carboxylate 43a (3.77 g, 15.75 mmol, method prepared as well known in the Journal of the American Chemical Society, 2000, 122(44), 10743-10753" and alcoholic methylamine solution (0.73 g, 23.63 mmol) were dissolved in 50 mL methanol. After stirring for 2 hours at 70° C, Sodium cyanoborohydride (2 g, 31.51 mmol) was added to the reaction mixture After reacting for 1 hour at 70°C, the reaction mixture was added 100 mL of H2O and extracted with dichloromethane (30 mL ×3).The organic phase was combined, dried over anhydrous sodium sulphate and filtered.The filtrate was concentrated under reduced pressure to obtain the title product (3aR,7aS)-tert-butyl 5-(methylamino) hexahydro-1H-isoindole-2(3H)-carboxylate 43b (3.2 g, brown oil), which was used directly in the next step without further purification. [00331] [00331] MS m/z (ESI): 255.2 [M+1] Step 2 (3aS,7aR)-tert-Butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) )amino)hexahydro-1H-isoindole-2(3H)-carboxylate [00332] [00332] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (1.93 g, 12.58 mmol) was dissolved in 50 mL of n-butanol, followed by the addition of (3aR,7aS)- tert-butyl 5-(methylamino)hexahydro-1H-isoindole-2(3H)-carboxylate 43b (3.20 g, 12.58 mmol) and potassium carbonate (3.47 g, 25.16 mmol). After reacting for 12 hours at 110°C, the reaction mixture was added with 100 mL of H2O and extracted with ethyl acetate (100 mL×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography over silica gel with elution system A to obtain the title product (3aS,7aR)-tert-butyl 5-(methyl(7H-pyrrole[2, 3-d]pyrimidin-4-yl)amino)hexahydro-1H-isoindole-2(3H)-carboxylate 43c (2.5 g, 53.2% yield) as a white solid. [00333] [00333] MS m/z (ESI): 372.2 [M+1] Step 3 N-Methyl-N-((3aS,7aR)-octahydro-1H-isoindol-5-yl)-7H hydrochloride - pyrrolo[2,3-d]pyrimidin-4-amine [00334] [00334] (3aS,7aR)-tert-Butyl 5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-1H-isoindole-2(3H)-carboxylate 43c (2 g, 5.38 mmol) was dissolved in 15 mL of a 6 M solution of hydrogen chloride in methanol. After reacting for 12 hours, the reaction mixture was concentrated under reduced pressure to obtain the title product N-methyl-N-((3aS,7aR)-octahydro-1H-isoindol-5-yl) hydrochloride -7H-pyrrolo[2,3-d]pyrimidin-4-amine 43d (1.1 g, white solid), which was used directly in the next step without further purification. [00335] [00335] MS m/z (ESI): 272.3 [M+1] Step 4 2-Hydroxy-1-((3aS,5R,7aR)-5-(methyl(7H-pyrrole[2,3-d) ]pyrimidin-4-yl)amino)hexahydro-1H-isoindole-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)ethanone [00336] [00336] 2-Hydroxy-1-((3aS,5S,7aR)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-1H-isoindole-2 (3H,3aH,4H,5H,6H,7H,7aH)-yl)ethanone [00337] N-Methyl-N-((3aS,7aR)-octahydro-1H-isoindol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine 43d hydrochloride (100 mg) , 0.37 mmol) was dissolved in 10 mL of N,N-dimethylformamide, followed by the addition of 2-glycolic acid (33 mg, 0.44 mmol), triethylamine (120 mg, 1.11 mmol) and hexafluorophosphate of O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethylformamidinium (0.21 g, 0.55 mmol). After reacting for 12 hours, the reaction mixture was added with 30 mL of H2O and extracted with ethyl acetate (20 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography over silica gel with elution system A to obtain the title products 2-hydroxy-1-((3aS,5R,7aR)-5-(methyl(7H) -pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-1H-isoindol-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)ethanone 43 (10 mg, 8.3% yield as a white solid and 2-hydroxy-1-((3aS,5S,7aR)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) amino)hexahydro-1H-isoindole-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)ethanone 45 (5 mg, 4.2% yield) as a white solid. [00338] [00338] MS m/z (ESI): 330.3 [M+1] 1 [00339] [00339] 1 H NMR (400 MHz, CD3OD-d4): 8.12 (s, 1H), 7.06 (d, 1H), 6.58 (d, 1H), 4.96-4.99 (m , 1H), 4.60-4.64 (m, 1H), 4.18-4.24 (m, 2H), 3.52-3.57 (m, 1H), 3.48 (s, 3H ), 3.26 (d, 2H), 2.72-2.74 (m, 1H), 2.16-2.26 (m, 1H), 1.96-1.98 (m, 1H), 1.83-1.87 (m, 2H), 1.69-1.72 (m, 1H), 1.44-1.52 (m, 1H), 1.23-1.33 (m, 3H ) [00340] [00340] 1 H NMR (400 MHz, CD3OD-d4): 8.10 (s, 1H), 7.11 (d, 1H), 6.66 (d, 1H), 4.71-4.75 (m , 1H), 4.58-4.62 (m, 1H), 4.10-4.22 (m, 2H), 3.43-3.57 (m, 1H), 3.48 (s, 3H ), 3.26 (d, 2H), 2.50-2.54 (m, 1H), 2.44-2.48 (m, 1H), 1.96-1.98 (m, 2H), 1.58-1.68 (m, 2H), 1.28-1.33 (m, 4H) Example 44 3-((3aS,7aR)-5-(Methyl(7H-pyrrole[2,3-d) ]pyrimidin-4-yl)amino)hexahydro-1H-isoindole-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)-3-oxopropanenitrile H O N N CN H N N N H [00341] N-Methyl-N-((3aS,7aR)-octahydro-1H-isoindol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine hydrochloride 43d (110 mg) , 0.41mmol) was dissolved in 15 mL of N,N-dimethylformamide, followed by the addition of 2-cyanoacetic acid (42 mg, 0.49 mmol), triethylamine (82 mg, 0.82 mmol) and hexafluorophosphate. O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethylformamidinium (231 mg, 0.61 mmol). After stirring for 12 hours, the reaction mixture was added with 15 mL of H2O and extracted with ethyl acetate (20 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography over silica gel with elution system A to obtain the title product 3-((3aS,7aR)-5-(methyl(7H-pyrrole[2,3- d]pyrimidin-4-yl)amino)hexahydro-1H-isoindole-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)-3-oxopropanenitrile 44 (13 mg, yield of 9.5%) as a white solid. [00342] [00342] MS m/z (ESI): 339.3 [M+1] [00343] [00343] 1 H NMR (400 MHz, DMSO-d6): 10.68 (s, 1H), 9.01 (s, 1H), 7.79 (d, 1H), 7.30 (d, 1H), 5.65-4.69 (m, 1H), 4.17-4.43 (m, 6H), 4.02 (s, 3H), 3.31-3.34 (m, 1H), 3. 25-3.28 (m, 1H), 2.61-2.71 (m, 2H), 2.24-2.32 (m, 2H), 1.98-2.05 (m, 2H) Example 46 2-Hydroxy-1-((4aS,8aR)-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)octahydro-isoquinolin-2(1H)- il)ethanone O H N oh N H N N N [00344] (4aR,8aS)-tert-Butyl 6-oxooctahydro-isoquinoline-2(1H)-carboxylate 46a (2.50 g, 9.87 mmol, prepared by a well known method "Journal of the American Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), 1995, 20, 2535-2542") and an alcoholic solution of methylamine (0.92 g, 29.61 mmol) were dissolved in 50 ml of methanol. After stirring for 2 hours at 70°C, sodium cyanoborohydride (1.24 g, 19.74 mmol) was added to the reaction mixture. After reacting for 2 hours at 70°C, the reaction mixture was added with 50 mL of H2O and extracted with dichloromethane (50 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the crude title product (4aR,8aS)-tert-butyl 6-(methylamino)octahydro-isoquinoline-2(1H)-carboxylate 46b (2 g, brown oil) , which was used directly in the next step without further purification. Step 2 (4aS,8aR)-tert-Butyl 6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)octahydro-isoquinoline-2(1H)-carboxylate [00345] [00345] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (1.14 g, 7.45 mmol) was dissolved in 50 mL of 1,4-dioxane, followed by the addition of (4aR,8aS )-tert-butyl 6-(methylamino)octahydro-isoquinoline-2(1H)-carboxylate 46b (2 g, 7.45 mmol) and potassium carbonate (2 g, 14.90 mmol). After reacting for 24 hours at 100°C, the reaction mixture was added with 100 mL of H2O and extracted with ethyl acetate (100 mL×3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography over silica gel with elution system A to obtain the title product (4aS,8aR)-tert-butyl 6-(methyl(7H-pyrrole[2,3-) d]pyrimidin-4-yl)amino)octahydro-isoquinoline-2(1H)-carboxylate 46c (1.4 g, 48.3% yield) as an off-white solid. [00346] [00346] MS m/z (ESI): 386.0 [M+1] Step 3 (4aS,8aR)-N-Methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-hydrochloride il) decahydro-isoquinolin-6-amine [00347] [00347] (4aS,8aR)-tert-Butyl 6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)octahydro-isoquinoline-2(1H)-carboxylate 46c (300 mg, 0.78 mmol) was dissolved in 15 mL of a 6 M solution of hi-chloride. [00348] [00348] MS m/z (ESI): 286.2 [M+1] Step 4 2-Hydroxy-1-((4aS,8aR)-6-(methyl(7H-pyrrolo[2,3-d]pyrimidine) -4-yl)amino)octahydro-isoquinolin-2(1H)-yl)ethanone [00349] ((4aS,8aR)-N-Methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)decahydro-isoquinolin-6-amine hydrochloride 46d (100 mg, 0 .35 mmol) was dissolved in 10 mL of dichloromethane, followed by the addition of glycolic acid (32 mg, 0.42 mmol), triethylamine (106 mg, 1.05 mmol), 1-ethyl-3-(3-) hydrochloride dimethylaminopropyl)carbodiimide (100 mg, 0.53 mmol) and 1-hydroxybenzotriazole (69 mg, 0.53 mmol) After reacting for 12 hours, the reaction mixture was added with 30 mL of H2O and extracted with dichloromethane ( 30 mL×2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by thin layer chromatography with elution system A to obtain the title product 2-hydroxy-1-((4aS,8aR)-6-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)octahydro-isoquinolin-2(1H)- yl)ethanone 46 (15 mg, 12.5% yield) as a white solid. [00350] [00350] MS m/z (ESI): 344.2 [M+1] 1 [00351] [00351] 1 H NMR (400 MHz, CDCl 3 ): 10.87 (s, 1H), 8.28 (s, 1H), 7.06 (d, 1H), 6.55 (d, 1H), 4, 90-4.95 (m, 1H), 4.18 (s, 2H), 3.48-3.51 (m, 1H), 3.30 (s, 3H), 3.12-3.32 ( m, 4H), 1.82-1.85 (m, 2H), 1.70-1.80 (m, 2H), 1.58-1.61 (m, 2H), 1.21-1, 29 (m, 4H) Example 47 [00352] [00352] (3aR,6aS)-Hexahydro-cyclopenta[c]pyrrole-5(1H)-one and 47a (16.80 g, 0.13 mol, prepared by a well known method "Patent EP2246347") were dissolved in 200 mL of dichloromethane in an ice bath, followed by the addition of triethylamine (16.2 mL, 0.16 mol) and the dropwise addition of benzyl chloroformate (25.22 g, 0.15 mol) ). The reaction mixture was warmed to room temperature. After reacting for 3 hours, the reaction mixture was added with 200 mL of H2O and extracted with dichloromethane (100 mL×2). The organic phase was combined, washed with saturated sodium bicarbonate (50 ml), a saturated sodium chloride solution (50 ml), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography over silica gel with elution system B to obtain the title product (3aR,6aS)-benzyl 5-oxohexahydro-cyclopenta[c]pyrrole -2(1H)-carboxylate 47b (18.5 g, yield 54.9%) as a white solid. [00353] [00353] MS m/z (ESI): 260.1 [M+1] Step 2 (3aS,4R,6aS)-Benzyl 4-methyl-5-oxohexahydro-cyclopenta[c]pyrrole-2( 1H)-carboxylate [00354] (3aR,6aS)-Benzyl 5-oxohexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 47b (5 g, 19.28 mmol) was dissolved in 70 mL of tetrahydrofuran a -78°C, followed by dropwise addition of lithium bis(trimethylsilyl)amide (19.7 mL, 19.67 mmol). After stirring for 1 hour at -78°C, the reaction mixture was added with methyl iodide (3.01 g, 21.21 mmol). The reaction mixture was warmed to room temperature. After reacting for 2 hours, the reaction mixture was added with 20 mL of saturated ammonium chloride and extracted with ethyl acetate (50 mL×2). The organic phase was combined, washed with a saturated solution of sodium chloride (20 ml×2), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography on silica gel with elution system B to obtain the title product (3aS,4R,6aS)-benzyl 4-methyl-5-oxohexahydro- cyclopenta[c]pyrrole-2(1H)-carboxylate 47c (1.08 g, 20.5% yield) as a pale yellow slurry. [00355] [00355] MS m/z (ESI): 274.1 [M+1] Step 3 (3aS,4R,5R,6aS)-Benzyl 4-methyl-5-(methylamino)hexahydro-cyclopenta[c ]pyrrole-2(1H)-carboxylate [00356] [00356] (3aS,4R,6aS)-Benzyl 4-methyl-5-oxohexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 47c (1.08 g, 3.95 mmol) was dissolved in 30 ml of alcoholic methylamine solution. After stirring for 24 hours at 50°C, sodium cyanoborohydride (500 mg, 7.90 mmol) was added. [00357] [00357] MS m/z (ESI): 289.2 [M+1] Step 4 (3aS,4R,5S,6aS)-Benzyl 4-methyl-5-(methyl(7H-pyrrole[2,3-d) ]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate [00358] [00358] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine 1d (176 mg, 1.14 mmol) was dissolved in 15 mL of H2O, followed by the addition of (3aS,4R,5R,6aS)- benzyl 4-methyl-5-(methylamino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 47d (300 mg, 1.04 mmol) and potassium carbonate (287 mg, 2.08 mmol) ). After reacting for 24 hours at 100°C, the reaction mixture was added with 10 mL d and H2O and extracted with dichloromethane (20 mL×2). The organic phase was combined, washed with a saturated sodium chloride solution (10 mL×2), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography over silica gel with elution system A to obtain the title product (3aS,4R,5S,6aS)-benzyl 4-methyl-5-(methyl( 7H-pyrrole[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxylate 47e (0.21 g, 49.9%) yield as a bright yellow solid. [00359] [00359] MS m/z (ESI): 406.3 [M+1] Step 5 [00360] [00360] (3aS,4R,5S,6aS)-Benzyl 4-methyl-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclo-penta[c ]pyrrole-2(1H)-carboxylate 47e (280 mg, 0.69 mmol) was dissolved in 20 mL of methanol, followed by the addition of palladium hydroxide on carbon (10 mg, 4%). After the reactor was purged with hydrogen three times, the reaction mixture was stirred for 2 hours, then washed with methanol (10 mL) and concentrated under reduced pressure to obtain the crude title product N-methyl-hydrochloride. N-((3aS,4R,5S,6aS)-4-methyloctahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine 47f (160 mg, light yellow solid), which was used directly in the next step without further purification. [00361] [00361] MS m/z (ESI): 272.3 [M+1] Step 6 1-((3aS,4R,5S,6aS)-4-Methyl-5-(methyl(7H-pyrrole[2,3) -d]pyrimidin-4-yl)amino)-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-2-yl)-2-hydroxy-ethanone [00362] N-Methyl-N-((3aS,4R,5S,6aS)-4-methyloctahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d hydrochloride ]pyrimidin-4-amine 47f (160 mg, 0.59 mmol) was dissolved in 10 mL of N,N-dimethylformamide, followed by the addition of 2-glycolic acid (54 mg, 0.71 mmol), triethylamine (119 mg , 1.18 mmol), 1-ethyl-3-(3-dimethylamino propyl) carbodiimide hydrochloride (169 mg, 0.88 mmol) and 1-hydroxybenzotriazole (119 mg, 0.88 mmol). After reacting for 15 hours, the reaction mixture was concentrated under reduced pressure, added to (20 mL), and then extracted with dichloromethane (30 mL×2). The organic phase was combined, washed with a saturated sodium chloride solution (10 mL×2), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and purified by chiral preparatory HPLC to obtain the title product 21-((3aS,4R,5S,6aS)-4-methyl-5-(methyl(7H-pyrrole[2,3) - d]pyrimidin-4-yl)amino)-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-2-yl)-2-hydroxy-ethanone 47 (32 mg , 16.5% yield) as a white solid. [00363] [00363] MS m/z (ESI): 330.2 [M+1] [00364] [00364] 1 H NMR (400 MHz, DMSO-d6): δ 11.6 (s, 1H), 8.08 (s, 1H), 7.13 (s, 1H), 6.61 (s, 1H ), 4.94-4.96 (m, 1H), 4.47-4.50 (m, 1H), 4.00-4.07 (m, 2H), 3.61-3.63 (m , 1H), 3.41-3.51 (m, 2H), 3.37-3.39 (m, 1H), 3.13 (s, 3H), 2.60-2.70 (m, 1H) ), 2.08-2.18 (m, 3H), 1.50-1.51 (m, 1H), 1.47-1.49 (m, 1H), 0.86-0.89 (m , 3H) Example 48 (3aR,5s,6aS)-N-(3-Ethyl-1,2,4-thiadiazol-5-yl)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxamide The N N H N Y N H N H N N N [00365] [00365] 3-Ethyl-1,2,4-thiadiazol-5-amine 48a (1.29 g, 9.98 mmol, prepared by a well known method "Collection of Czechoslovak Chemical Communications, 1971, 36 , 4091-4098") was dissolved in 50 mL of tetrahydrofuran in an ice bath, followed by a- [00366] [00366] MS m/z (ESI): 250.2 [M+1] Step 2 (3aR,5s,6aS)-N-(3-Ethyl-1,2,4-thiadiazol-5-yl)-5 -(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxamide [00367] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (120 mg, 0.47 mmol) was dissolved in 50 mL of tetrahydrofuran, followed by the addition of phenyl (3-ethyl-1,2,4-thiadiazol-5-yl)carbamate 48b (2 .14 g, 8.58 mmol) and by the dropwise addition of triethylamine (2.4 mL, 17.17 mmol). After reacting for 12 hours at 60°C, the reaction mixture was added with 30 mL of H2O and extracted with dichloromethane (50 mL×3). The organic phase was combined, washed with a saturated sodium chloride solution (50×2 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography over silica gel with elution system A to obtain the title product (3aR,5s,6aS)-N-(3-ethyl -1,2,4-thiadiazol-5-yl)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino) hexahydro-cyclopenta[c]pyrrole-2 (1H)-carboxamide 48 (1.25 g, 81.7%) yield as a white solid. [00368] [00368] MS m/z (ESI): 413.4 [M+1] 1 [00369] [00369] 1 H NMR (400 MHz, DMSO-d6): 11.60 (s, 1H), 11.52 (s, 1H), 8.08 (s, 1H), 7.07-7.02 (m , 1H), 6.55-6.5 (m, 1H), 5.56-5.38 (m, [00370] Sodium thiocyanate (527 mg, 6.50 mmol) was dissolved in mL methanol and placed at -20°C, followed by the addition of cyclopropylcarbamidine hydrochloride (603 mg, 5 mmol) and triethylamine (0 .8 ml, 5.74 mmol). After stirring for 45 minutes, triethylamine (0.7 mL, 5.02 mmol) and an 8% sodium hypochlorite solution (4.2 mL, 5 mmol) were added dropwise to the reaction mixture. After reacting for 2 hours at -20°C, the reaction mixture was warmed to room temperature. After reacting for 12 hours, the reaction mixture was concentrated under reduced pressure, followed by the addition of 35 mL of H2O and extracted with ethyl acetate (30 mL×3). The organic phase was combined, washed with a saturated solution of sodium chloride (50 ml), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain the crude title product 3-cyclopropyl-1,2,4-thiadiazol-5-amine 49a (243 mg, white solid), which was used directly in the next step without further purification. . [00371] [00371] MS m/z (ESI): 142.2 [M+1] Step 2 Phenyl (3-cyclopropyl-1,2,4-thiadiazol-5-yl)carbamate [00372] [00372] 3-Cyclopropyl-1,2,4-thiadiazol-5-amine 49a (212 mg, 1.50 mmol) was dissolved in 5 mL of tetrahydrofuran in an ice bath, followed by the addition of potassium carbonate anhydrous (270 mg, 1.95 mmol) and by dropwise addition of phenyl carbon 34b hydrochloride (246 mg, 1.58 mmol), then the reaction mixture was warmed to room temperature. After reacting for 12 hours, the reaction mixture was filtered and concentrated under reduced pressure to obtain the crude title product phenyl(3-cyclopropyl-1,2,4-thiadiazol-5-yl)carbamate 49b (350 mg, colorless oil ), which was used directly in the next step without further purification. [00373] [00373] MS m/z (ESI): 262.3 [M+1] Step 3 (3aR,5s,6aS)-N-(3-Cyclopropyl-1,2,4-thiadiazol-5-yl)-5 -(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxamide [00374] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (344 mg, 1.34 mmol) was dissolved in 6 mL of tetrahydrofuran, followed by the addition of phenyl (3-cyclopropyl-1,2,4-thiadiazol-5-yl)carbamate 49b (350 mg, 1.34 mmol) and by the dropwise addition of triethylamine (0.6 mL, 4.02 mmol). After reacting for 12 hours at 50°C, the reaction mixture was added with mL of H2O and extracted with dichloromethane (20 mL×3). The organic phase was combined, washed with a saturated sodium chloride solution (20×2 mL), dried over anhydrous sodium sulfate and filtered. The filter [00375] [00375] MS m/z (ESI): 425.4 [M+1] 1 [00376] [00376] 1 H NMR (400 MHz, DMSO-d6): 11.53 (d, 2H), 8.07 (s, 1H), 7.05-7.03 (m, 1H), 6.53-6 5.52 (m, 1H), 5.47-5.43 (m, 1H), 3.70-3.65 (m, 2H), 3.37-3.32 (m, 2H), 3.15 (s, 3H), 2.90-2.89 (m, 2H), 2.12-2.07 (m, 1H), 2.05-1.98 (m, 2H), 1.80-1 .74 (m, 2H), 0.97-0.93 (m, 4H) Example 50 (3aR,5s,6aS)-N-(3-(Hydroxymethyl)-1,2,4-thiadiazol-5-yl )-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxamide O Y N OH H N N N H N H N N N [00377] N-Methyl-N-((3aR,5s,6aS)-octahydro-cyclopenta[c]pyrrol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4 hydrochloride -amine 6a (90 mg, 0.35 mmol) was dissolved in 2 mL of N,N-dimethylformamide, followed by the addition of methyl 5-((phenoxycarbonyl)amino)-1,2,4-thiadiazole-3-carboxylate 50a (97 mg, 0.35 mmol, prepared by a well-known method "patent application WO2004103980") and by the dropwise addition of triethylamine (105 mg, 1.04 mmol). After reacting for 12 hours at 100°C, mL of H2O was added to the reaction mixture, and the reaction mixture was extracted with dichloromethane (10 mL×3). The organic phase was combined, washed with a saturated sodium chloride solution (10×2 ml), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by column chromatography on silica gel with elution system A to obtain the title product methyl 5-((3aR,5s,6aS)-5-(methyl(7H) - pyrrolo[2,3-d]pyrimidin-4-yl)amino)octahydro-cyclopenta[c]pyrrole-2-carboxamido)-1,2,4-thiadiazole-3-carboxylate 50b (99.5 mg, 65.0%) yield as a white solid. [00378] [00378] MS m/z (ESI): 443.4 [M+1] Step 2 (3aR,5s,6aS)-N-(3-(Hydroxymethyl)-1,2,4-thiadiazol-5-yl) -5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)hexahydro-cyclopenta[c]pyrrole-2(1H)-carboxamide [00379] [00379] Methyl 5-((3aR,5s,6aS)-5-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)octahydro-cyclopenta[c]pyrrole- 2-carboxamido)-1,2,4-thiadiazole-3-carboxylate 50b (95 mg, 0.21 mmol) was dissolved in 3 mL of ethanol, followed by the addition of sodium borohydride (49 mg, 1.29 mmol) in batches. After reacting for 2 hours, the reaction mixture was concentrated under reduced pressure, followed by the addition of 10 mL of H2O and extracted with dichloromethane (10 mL×2). The organic phase was combined, washed with a saturated sodium chloride solution (10×2 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by chromate. [00380] [00380] MS m/z (ESI): 415.4 [M+1] 1 [00381] [00381] 1 H NMR (400 MHz, DMSO-d6): 11.56 (s, 1H), 8.08 (s, 1H), 6.99-7.04 (m, 1H), 6.50-6 4.54 (m, 1H), 5.37-5.50 (m, 1H), 4.70 (br.s, 1H), 4.26-4.34 (m, 2H), 3.55-3 .66 (m, 2H), 3.21-3.29 (m, 2H), 3.14 (s, 3H), 2.74-2.85 (m, 2H), 1.92-2.05 (m, 2H), 1.67-1.76 (m, 2H) [00382] The compounds of Examples 51 to 156 were synthesized according to the procedures of Example 1 and Example 2 using appropriate reagents according to the method of synthesis of the compounds of the present invention. [00383] [00383] The Example number, structures and characteristic data are given below: [00384] The in vitro activity of the compounds of the present invention in inhibiting the JAK1 kinase was determined by the following method. [00385] The in vitro kinase assays described below can be used to determine the activity of the compound of the present invention in inhibiting JAK1 kinase. Test compounds were dissolved in dimethyl sulfoxide and diluted with water to the serial concentration gradient required in the experiment. Substrates for JAK1 (Cell Signaling Technology, Catalog number: 1305s) and ATP solution (2 mM) were diluted with water to obtain the final concentration of 20 µM ATP and 1.2 µM of substrate solution. The exact amount of JAK1 kinase (Invitrogen, Catalog number: pv4774 ) was mixed with 4 × buffer (prepared by the user, and comprising 50 mM HEPES, pH 7.3, 125 mM NaCl, 24 mM MgCl2, 1, 25 mM DTT) to a final concentration of 8 ng / µL. Each microplate well [DELFIA® streptavidin coated transparent plate (Perkin Elmer, Catalog number: AAAND-0005)] was added with 17.5 µL of ATP / substrate mixture, 5 µL of aqueous solution of the compound of test (the blank control was added with only 5 µL of pure water), and 7.5 µL of the kinase solution prepared above (the control was added with only 4 × of buffer). Each portion was mixed sufficiently, then incubated at room temperature (27°C) for 50 minutes, washed with wash buffer and dried three times, and then added with an antibody conjugated to HRP [Phospho-Tyrosine mAb mouse (P-Tyr-100) (HRP Conjugate, Cell signaling Technology, Catalog number: 5465)], and incubated for 1 hour. The microplate was washed with wash buffer and dried three times, then TMB (Sigma, Catalog number: T4444) was added and allowed to stain for 15 minutes. Stopping solution (1N sulfuric acid solution) was added to stop the reaction. Absorbance was read in a novostar microplate reader at a wavelength of 450 nm. IC50 values of test compounds can be calculated from data of test compounds to inhibit JAK1 kinase activity at different concentrations. Activity of the compounds of the present invention [00386] [00386] Biochemical activity of the compounds of the present invention was determined by the above assay, and the IC50 values were shown in Table 1 below. [00387] [00387] The in vitro activity of the compounds of the present invention in inhibiting the JAK2 kinase was determined by the following method. [00388] The in vitro kinase assays described below can be used to determine the activity of the compound of the present invention in inhibiting JAK2 kinase. Test compounds were dissolved in dimethyl sulfoxide and diluted with water to the serial concentration gradient required in the experiment. Substrates for JAK2 (cell signaling technology, Catalog number: 1305s) and ATP solution (2 mM) were diluted with water to obtain a final concentration of 20 µM ATP and 1.2 µM substrate solution. The exact amount of JAK2 kinase (Invitrogen, Catalog number: pv4210 ) was mixed with 4 × buffer (prepared by the user, and comprising 50 mM HEPES, pH 7.3.125 mM NaCl, 24 mM MgCl2, 1.25 mM of DTT) to a final concentration of 8 ng / µL. Each microplate well [DELFIA® streptavidin coated transparent plate (Perkin Elmer, Catalog number: AAAND-0005)] was added with 17.5 µL of ATP / substrate mixture, 5 µL of aqueous solution of the compound of test (the blank control was added with only 5 µL of pure water), and 7.5 µL of the so- [00389] [00389] Biochemical activity of the compounds of the present invention was determined by the above assay, and the IC50 values were shown in table 2 below. Table 2. IC50 of compounds of the present invention to inhibit JAK2 kinase activity Example No. IC50 (JAK2/Bio) (nM) 6 45 17 3 22 54 24 77 34 5 35 16 36 75 37 72 38 7 [00390] The in vitro activity of the compounds of the present invention in inhibiting the JAK3 kinase was determined by the following method. [00391] The in vitro kinase assays described below can be used to determine the activity of the compound of the present invention in inhibiting JAK3 kinase. Test compounds were dissolved in dimethyl sulfoxide and diluted with water to the serial concentration gradient required in the experiment. Substrates for JAK3 (cell signaling technology, Catalog number: 1305s) and ATP solution (2 mM) were diluted with water to obtain a final concentration of 20 µM ATP and 1.2 µM substrate solution. The exact amount of JAK3 kinase (Invitrogen, Catalog number: pv3855 ) was mixed with 4 × buffer (prepared by the user, and comprising 50 mM HEPES, pH 7.3.125 mM NaCl, 24 mM MgCl2, 1.25 mM of DTT) to a final concentration of 8 ng / µL. Each microplate well [DELFIA® streptavidin coated transparent plate (Perkin Elmer, Item : AAAND-0005)] was added with 17.5 µL of ATP / substrate mixture, µL of aqueous test compound solution (the control the blank was added with only 5 µL of pure water), and 7.5 µL of the kinase solution prepared above (the control was added with only 4 × of buffer). Each portion was sufficiently mixed, then incubated at room temperature (27°C) for 50 minutes, washed with wash buffer and dried three times, and then added with an antibody conjugated to HRP [Phospho-Tyrosine mouse mAb (P-Tyr-100) (HRP Conjugate, Cell signaling Technology, Catalog number: 5465)], and incubated for 1 hour. The microplate was washed with wash buffer and dried three times, then TMB (Sigma, Catalog number: T4444) was added and allowed to stain for 5 to 15 minutes. Stopping solution (1N sulfuric acid solution) was added to stop the reaction. Absorbance was read in a novostar microplate reader at a wavelength of 450 nm. IC50 values of test compounds can be calculated from data of test compounds to inhibit JAK3 kinase activity at different concentrations. Activity of the compounds of the present invention [00392] [00392] Biochemical activity of the compounds of the present invention was determined by the above assay, and the IC50 values were shown in table 3 below. Table 3. IC50 of compounds of the present invention to inhibit JAK3 kinase activity Example No. IC50 (JAK3/Bio) (nM) 6 11 8 67 17 0.3 31 93 38 40 40 71 49 39 50 201 56 75 68 43 69 20 73 191 84 70 97 110 99 110 111 101 125 39 128 4 129 17 133 90 134 164 [00393] The following in vitro assay was used to determine the activity of the compounds of the present invention to inhibit the proliferation of the human erythroid leukemia cell line TF-1. [00394] The following in vitro cell assay can be used to determine the activity of the test compound to inhibit IL-4 mediated proliferation (said IL-4 can mediate the JAK3 pathway). Activity is represented by the IC50 value, which not only reflects the activity of the test compound to inhibit JAK2 kinase and JAK3 kinase, but also reflects the selectivity of the test compound for JAK2 kinase and JAK3 kinase . [00395] [00395] General assay procedures are given below: first, TF-1 cells (purchased from ATCC, Catalog number: CRL 2003) were seeded in a 96-well cell culture plate at a cell concentration (8000 cells/mL of medium), and 10 ng/mL of IL-4 (Invitrogen, Catalog number: PHC0044) were added to each well. Then a 10 × serial concentration gradient of test compound solutions (10000, 1000, 100, 10.1 and 0.1 nM) were prepared, and then 10 × of compound solutions prepared above were added to 96-well cell culture plates containing IL-4. After the culture plates have been cultivated continuously for 72 hours, the activity of the [00396] [00396] The biological activity of the compounds of the present invention was tested using the assay described above. IC50 values were measured and are shown in table 4 below: Table 4. IC50 of compounds of the present invention to inhibit TF-1 cell proliferation Example No. IC50 (TF-1/ IL-4)/nM 1 196 5 133 6 457 8 530 11 507 17 165 19 711 34 44 37 123 38 160 39 419 40 399 48 27 61 507 73 476 74 546 86 691 94 152 99 44 114 138 121 27 [00397] The following in vitro assay was used to determine the activity of the compounds of the present invention to inhibit the proliferation of T cells. [00398] [00398] The following in vitro cell assay can be used to determine the activity of the test compound to inhibit T cell proliferation. The activity is represented by the IC50 value. [00399] [00399] The general assay procedures are given below: first, the PBMC cell line (purchased from the Shanghai Blood Center) was centrifuged, the supernatant was removed and the cells were counted. Cells were incubated in medium [RPMI-1640 (Hy-clone, Catalog number: SH30809.01B)+10% fetal bovine serum (GIBCO, Catalog number: 10099)+1% Penicillin Streptavidin (GIBCO, Catalog number: 15140 )] with an additional 200 µg of purified functional-grade anti-human CD3 (eBioscience, Catalog number: 16-0037-81) and in a 5% carbon dioxide (CO2) incubator at 37°C for 3 days until reaching a cell concentration of 2×106/ml. After that, the cells were washed 3 times, resuspended, diluted to 2×106/mL, and then added with recombinant human IL-2 (purchased from Peprotech, Catalog number: 200-02) to a concentration of 10 ng /mL, and cultured for another 3 days. The cells [00400] The biological activity of the compounds of the present invention was tested using the assay described above. IC50 values were measured and are shown in table 5 below: Table 5. IC50 of compounds of the present invention for inhibiting T cell proliferation Example No. IC50(T cell)/nM 6 414 8 460 11 705 13 754 14 712 15 853 17 29 19 724 21 892 [00401] [00401] Rats were used as test animals. The compounds of Example 6, Example 17, Example 22, Example 34, Example 35, Example 40, Example 48, Example 49, Example 99, Example 114, Example 121, Example 125, Example 128 and Example 148 were administered intragastrically to rats to determine the drug concentration in the plasma at different time points by the LC/MS/MS method. The pharmacokinetic behavior of the compounds of the present invention was studied and evaluated in rats. [00402] Compounds of Example 6, Example 17, Example 22, Example 34, Example 35, Example 40, Example 48, Example 49, Example 99, Example 114, Example 121, Example 125, Example 128 and Example 148 . [00403] [00403] 56 healthy adult SD rats, half male and half female, purchased from SINO-BRITSH SIPPR/BK LAB. ANIMAL LTD., CO, Certificate No.: SCXK (Shanghai) 2003-0002, were divided into 14 groups, 4 rats in each group. [00404] The exact amount of test compounds was weighed out and 1.0 ml of dimethyl sulfoxide was added to make a suspension at 1.0 mg/ml. [00405] [00405] After an overnight fast, SD rats received intragastrically a dose of 10.0 mg/kg and an administration volume of 10 mL/kg. [00406] [00406] The compounds of Example 6, Example 17, Example 22, Example 34, Example 35, Example 40, Example 48, Example 49, E- [00407] [00407] Determination of the content of test compounds in rat plasma after intragastric administration at different concentrations: 50 µL of rat plasma collected at various time points after administration were added to 50 µL of internal standard solution and 100 µL of methanol and were mixed for 3 minutes by a tumbler. The mixture was centrifuged for 10 minutes at 13,500 rpm. µL of the supernatant was taken from the sample plasma and analyzed by LC-MS/MS. [00408] [00408] The pharmacokinetic parameters of the compounds of the present invention were shown as follows: Example No. Pharmacokinetic Assay (10 mg/kg) Concentration Area Under Half-Life Perclearance Time Dystriplasmic Volume Manency Curve Mean Apparent buoyancy Cmax (ng/ml) AUC (ng/ml*h) t1/2 (h) MRT (h) CL/F (ml/min/kg) Vz/F (ml/kg) Example 6 1227±549 2210±1472 4.01±1.52 2.19±0.60 110±71 34475±20986 Example 17 1482±177 4762±1684 2.03±0.82 3.10±1.04 38.7±13.7 6200±1690 Example 22 2965±2113 6986±4559 2.87±0.70 2.60±0.10 34.6±22.7 9394±8062 Example 34 2668±1449 14881±10395 2.10±0.73 3.81±0.82 18.3±14.2 2760±1530 216/217 Example 35 1220±244 3075±1336 2.11±0.67 3.02±1.33 63.3±28.0 10418±2040 Example 40 1521±317 5228±1169 2.72±1.82 3.27±0.68/ 33.2±7.7 6990±3390 Example 48 2071±1473 9617±7909 2.43±0.75 3.40±0.91 31.9±26.9 7203±8113 Example 49 1061±677 5997±4741 3.53±1.72 5.32±0.96 46.1±35.6 18063±20748 Example 99 2668±1449 14881±10395 2.10±0.73 3.81 ±0.82 18.3±14.2 2760±1530 Example 114 1226±829 2935±2217 2.38±0.67 2.39±0.43 104±88 19115±15952 Example 121 2071±1473 9617±7909 2.43±0.75 3.40±0.91 31.9±26.9 7203±8113 Example 125 1061±677 5997±4741 3.53±1.72 5.32±0.96 46.1± 35.6 18063±20748 Example 128 1423±150 2322±1029 1.62±0.28 1.73±0.50 84.3±37.8 11378±4148 Example 148 1521±317) 5228±1169 2.72 ±1.82 3.27±0.68 33.2±7.7 6990±3390 Conclusion: The compounds of the present invention showed better pharmacokinetic data and significant advantage of pharmacokinetic properties.
权利要求:
Claims (26) [1] 1. Compound, characterized in that it has the formula (I): 13 14 R R 1 5 R 7 R () N R s 12 8 ( )p R R ( ) R11 t 2 L ( )q 6 R R 10 N 9 R R R A 3 R N N 4 (I) R or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, in which: A is CH or N; L is a bond or alkyl; R1 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)n C(O)OR15, - OC(O)R15, -C(O)R15, -C( O)NR16R17, -NHC(O)R15, -NR16R17, - OC(O)NR16R17, -NHC(O)NR16R17 and -S(O)mR15, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are , each optionally substituted with one or more groups selected from the group consisting of halogen, hydroxy, cyano, nitro, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl,-(CH2)nC(O) OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15, -NR16R17, -OC(O)NR16R17, - NHC(O)NR16R17, -S( O)mR15, -NHC(O)(O)R15 and -NHS(O)mR15; R2 or R4 are each independently selected from the group consisting of hydrogen and alkylayl; R or R3 are each independently selected from the group consisting of hydrogen, halogen and alkyl; R5 or R6 are each independently selected. formed from the group consisting of hydrogen, alkyl and aryl, wherein the alkyl or aryl is each optionally substituted with one or more groups selected from the group consisting of alkyl and halogen; whether R7, R8, R9 or R10 are each independently selected from the group consisting of hydrogen, alkyl, hydroxyalkyl and halogen, or, R7 and R8 or R9 and R10 are taken together to form an oxo group ; whether R11, R12, R13 or R14 are each independently selected from the group consisting of hydrogen, alkyl and halogen, or, R11 and R12 or R13 and R14 are taken together to form an oxo group; R15 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, nitro, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH2)nC(O)OR18, -OC(O)R18, -C (O)R18, -C(O)NR19R20, -NHC(O)R18, -NR19R20, -OC(O)NR19R20, -NHC(O)NR19R20, -S(O)mR18, -NHC(O)OR18 and -NHS(O)mR18; R16 or R17 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each of them optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, alkoxy, cycloalkyl, heterocyclyl, hydroxyalkyl, alkynyl, aryl, heteroaryl, carboxyl, alkoxycarbonyl and -OR18; R18 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, hydroxyalkyl, aryl and heteroaryl; R19 or R20 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; m is 0, 1 or 2; n is 0, 1 or 2; p is 0, 1 or 2; q is 0, 1 or 2; s is 0, 1 or 2; and t is 0, 1 or 2. [2] 2. Compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to claim 1, characterized in that it is selected from a compound of formula (II), or a pharmaceutically acceptable salt thereof: 5 1 7 RR R N R 8 ( )p 6 2 ( )q R RL 9 10 N R R R A 3 R N N 4 R(II) in which A, L, R, R 1 to R 10 , p and q are as defined in claim 1. [3] 3. Compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to claim 1 or 2, characterized in that it is selected from a compound of formula (III), or a pharmaceutically acceptable salt thereof: 7 R R R N 8 R ( )p 6 ( )q R L N 10 R R 9 R A 3 R N N H(III) in which A, L, R, R1, R3, R5 to R10, p and q are as defined in claim 1. [4] 4. Compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to claim 1 or 2, characterized in that it is selected from a compound of formula (IV-a) or formula (IV-b), or a pharmaceutically acceptable salt thereof: 5 1 5 1 7 RR 7 RR R N R N 8 8 RR 6 6 R R L L 10 10 N N R R 9 R R R9 R A 3 A 3 R R N N N N H H (IV-a) (IV-b) in which A, L, R, R 1 , R 3 and R 5 to R 10 are as defined in claim 1. [5] 5. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to claim 1 or 2, characterized in that it is selected from a compound of formula (Va) or formula (Vb), or a pharmaceutically acceptable salt thereof: R R N N 5 6 5 6 R R R R L L N N N N N N N N H H (V-a) (V-b) in which R 1 , R 5 , R 6 and L are as defined in claim 1. [6] 6. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to claim 1 or 2, characterized in that is selected from a compound of formula (VI-a) or formula (VI-b), or a pharmaceutically acceptable salt thereof: 1 R 12 1 5 R 11 R 12 7 RNR 5 R 11 R ( )t 7 RNR 8 R ( )t R 8 6 RR 6 L 9 10 R NR RL R N R R R 9 10 A 3 R A 3 N R N N H (VI-a) N (VI-b) H in which A, L, R, R1, R3, R5 to R12 and t are as defined in claim 1. [7] 7. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to claim 1 or 2, characterized in that it is selected from a compound of formula (VII-a) or formula (VII-b), or a pharmaceutically acceptable salt thereof: R 5 R 1 NR 1 ( )t NR L 6 ( )t NR 11 12 L R R R 6 11 12 N R R N N N N H N N H (VII-a) (VII-b) in which R1, R5, R6, R11, R12, L and t are as defined in claim 1. [8] 8. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to any one of claims 1 to 7, characterized in that L it is a connection. [9] 9. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to any one of claims 1 to 8, characterized in that: R1 is selected from the group consisting of alkyl, heteroaryl, -(CH2)nC(O)OR15, -C(O)R15, -C(O)NR16R17 and -S(O)2R15, where the alkyl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of halogen, hydroxyl, cyano and -(CH2)nC(O)OR15; R15 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, nitro, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH2)nC(O)OR18, -OC(O)R18, -C (O)R18, -S(O)2R18, -NHC(O)(O)R18, - NHS(O)2R18 and -NR19R20; preferably, R11 is selected from the group consisting of alkyl and cycloalkyl, wherein the alkyl, cycloalkyl or heterocyclyl are each optionally substituted with one or more groups selected from the group consisting of alkyl, hydroxyl, cyano, amino, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH2)nC(O)OR18, -OC(O)R18, -C(O)R18, -S(O)2R18, -NHC(O)OR18, - NHS(O)2R18 and -NR19R20. R16 or R17 are each independently selected from the group consisting of hydrogen, alkyl and heteroaryl; wherein said heteroaryl is optionally substituted with one or more groups selected from the group consisting of alkoxy, cycloalkyl, hydroxyalkyl, alkynyl and -OR18; R18 is selected from the group consisting of hydrogen, alkyl and hydroxyalkyl; R19 or R20 are each independently selected from the group consisting of hydrogen and alkyl; and n is 0, 1 or 2. [10] 10. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 9, characterized in that R5 or R6 are each independently selected from the group consisting of hydrogen and alkyl, preferably hydrogen or methyl. [11] A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 4, 6 and 8 to 10 , characterized in that R7, R8, R9 or R10 are each independently selected from the group consisting of hydrogen, alkyl and hydroxyalkyl, preferably hydrogen, methyl or hydroxymethyl. [12] 12. Compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1, 6 and 7, characterized in that that R11, R12, R13 or R14 are each independently hydrogen. [13] 13. Compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1, 6 and 7, characterized in that that R11 and R12 or R13 and R14 are taken together to form an oxo group. [14] 14. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof according to any one of claims 1 to 4 and 6, characterized in that that A is N. [15] 15. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 14, characterized in that the compound is selected from the group consisting of: O O O O O N Y N H NO NO NO NO H H H H H H N N N N N N N N N NN NN NN NN NN H H H H H O O O O H OH H OH N O N N H H N N N N H H N H N H N N H N H N N N N N NN NN NN NN NN H H H H H O O O O H O N H H OH S O N N H HN N O N H N H N N H N H N N N N N NN NN NN NN NN H H H H H O O O O O H O H H NH2 H N O H N N N H N NO OH oh H N H N H N H N H N N N N N N NN NN NN NN NN H H H H H O O O H O H N H OH H O H N Y N N N O N N O O O N H N H H N H N H N N N N N N NN NN NN NN NN H H H H H O N Cl Cl H H O N H N N N N N N N Cl H Cl H N N N H H N H N N H N H N N N N N N N N N N N N N N H H N H H H N Cl N N O OH O N N N N O O N H N Y H H H H H N H N H H H N N N N H N H N N N N N NN NN NN NN NN H H H H H O O O O H O H N N H H N H N OH O H N N H N CN s AT THE N H N H N H H H N N N N N N N NN NN NN NN NN H H H H H H H N H N H O the H HO The N N N N H N H N OH N OH N CN H H H N N N N N NN NN NN NN NN H H H H H NO NO YES NO OH The N N H O O H H N Y N N N H N N Y N H OH H H H N N OH H H N H N N N H N H N N N N N N N N N N N N N N N H N H H H H O O O O F F H S F H Y O H Y N N N F H O F H N F N HN N H N H N N H N H N N N N N NN NN NN NN NN H H H H H F OH H F H N H OH H NH H N NO NO F N N H N N H H N H HN N N N N N NN NN NN NN NN H H H H H O N H N N N H OH N H O N H H S N N N N H H N H H N N N H N N N N N NN NN NN NN NN H H H H H F N N H F H N O N H N H Y N N H NO NO F H H H N H H N N H N N N N N N N NN NN NN NN NN H H H H H O O O O N N O HON H OH N N H N N H S H N N N N N Y OH H H H N H H N N H H N N N N N N N NN NN NN NN NN H H H H H O O O The N H O N H H H N O H N N N N N H N H H H H N H N N N N N N N N NN NN NN NN NN H H H H H O O O O N H H H N H N N H NO OH OH H N N N H H H N H N H N N N N HN N N N N N NN NN NN NN NN H H H H H O O O N N O H H H H OH N N H S N N Y N H HN O N oh s H H N H N H N N O N H N N N N N NN NN NN NN NN H H H H H O O F FO the H H OH H H H OH N N F N O N N oh OH H NH H N H N N H N N N N N N NN NN NN NN NN H H H H H O O O O F OH O H H H N N F N O S N H o N oh OH OH H N N N H H H N H N N H N N H N N N N N NN NN NN NN NN H H H H H O F O O F N H H H NO NO NO N F N OH N H H N N N N H H N H H N H N H H N N N N N N N NN NN NN NN NN H H H H H H H NO NO NO NO NO NO NO O O H H H H N N N N H N N N H H N N O H H N H N H N H H N H NO NO N N N N N NN NN NN NN NN H H H H H O O O O N N OH O N N O H F H N N O N H H S H N N N Y N N F H N N F H H N H N H H N H N N H N N NN NN NN NN NN H H H H H The N O N O N O N N O H N H H N N N N N H N N Y N N O H H N N N N N Y H H H H N N H N H H N H N H N N N N N NN NN NN NN NN H H H H H The N O Y Y O Y N O N O H N H N N N H H N N N H N N N N N N N N H H H H H N H N H N H N H N H N N N N N NN NN NN NN NN H H H H H O O O O O H OH H OH H OH N N H H N N O NO HO N H H H H N H F N HN N N N N N N F NN NN NN NN NN H H H H H O O O O O N N N O O H H H H H H H H H NO NO N N N H N H N N N N N N NN NN NN NN NN H H H H H OH O O O S O H H HO N N N N N H H H H H H H H H H H N N N N N N N N N N NN NN NN NN NN H H H H H N O O S O O O O N N N N N H H H H H H H H H H H N N N N N N N N N N NN NN NN NN NN H H H H H N N O N H H O N S N N N O H H N O O N H HN H H N H N N H N N H N N N N N NN NN NN NN NN H H H H H O O H N N N N N H NO H O N N N N Y H N H H N H N O N N H N N N N N NN NN NN NN NN H H H H H THE Y N H OH N N N H N H N N N and H . [16] 16. Compound, characterized by the fact that it has the formula (IB), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof: 13 14 R R 1 5 R 7 R () N R s 12 R 8 ( )p R ( ) R11 t 2 L ( )q 6 R R N 9 R 10 H R (IB) in which: L is a bond or alkylayl; R1 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)n C(O)OR15, - OC(O)R15, -C(O)R15, -C( O)NR16R17, -NHC(O)R15, -NR16R17, - OC(O)NR16R17, -NHC(O)NR16R17 and -S(O)mR15, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are , each optionally substituted with one or more groups selected from the group consisting of halogen, hydroxy, cyano, nitro, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -(CH2)nC(O) OR15, -OC(O)R15, -C(O)R15, -C(O)NR16R17, -NHC(O)R15, -NR16R17, -OC(O)NR16R17, - NHC(O)NR16R17, -S( O)mR15, -NHC(O)(O)R15 and -NHS(O)mR15; R2 is selected from the group consisting of hydrogen and alkylayl; R5 or R6 are each independently selected from the group consisting of hydrogen, alkyl and aryl, wherein the alkyl or aryl are each optionally substituted with one or more groups selected from the group consisting of alkyl and halogen; whether R7, R8, R9 or R10 are each independently selected from the group consisting of hydrogen, alkyl, hydroxyalkyl and halogen, or, R7 and R8 or R9 and R10 are taken together to form an oxo group ; whether R11, R12, R13 or R14 are each independently selected from the group consisting of hydrogen, alkyl and halogen, or, R11 and R12 or R13 and R14 are taken together to form an oxo group; R15 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, nitro, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH2)nC(O)OR18, -OC(O)R18, -C (O)R18, -C(O)NR19R20, -NHC(O)R18, -NR19R20, -OC(O)NR19R20, -NHC(O)NR19R20, -S(O)mR18, -NHC(O)OR18 and -NHS(O)mR18; R16 or R17 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl are each of them optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxy, cyano, amino, alkoxy, cycloalkyl, heterocyclyl, hydroxyalkyl, alkynyl, aryl, heteroaryl, carboxyl, alkoxycarbonyl and -OR18; R18 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, hydroxyalkyl, aryl and heteroaryl; R19 or R20 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; m is 0, 1 or 2; n is 0, 1 or 2; p is 0, 1 or 2; q is 0, 1 or 2; s is 0, 1 or 2; and t is 0, 1 or 2. [17] 17. Process for preparing the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, characterized in that it comprises the steps of: 13 14 RR 1 5 R 7 R () NR s 12 8 ( )p RR ( ) R11 13 14 t XRR 1 2 L ( )q 6 R 5 RRR 7 R () NN 9 R 10 R s RA 3 12 R 11 RR + R 8 ( )p () RNN t ( )q A 3 R 4 R 2 L 6 R R 10 N 9 R N N H R 4 R (IA) (IB) (I) under alkaline conditions, reacting the compound of formula (IA) with the compound of formula (IB) to obtain the compound of formula (I); in which X is halogen, and A, L, R, R1 to R14, p, q, s and t are as defined in claim 1. [18] 18. Process for preparing the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, characterized in that it comprises the steps of: 13 14 R R R R 13 14 1 5 ( ) NH 5 R 7 R 7 R () N R s R 8 12 R 11 s 12 R ( )p () R R ( )p 8 R 11 t () R ( )q t R 2 L 6 R 2 L ( )q 6 10 R 10 R N R 9 R N 9 R R R R A 3 A 3 R R NNNN 4 4 R(IC)R(I) under alkaline conditions, react the compound of formula (IC), or a pharmaceutically acceptable salt thereof, with carboxylic acid, acyl chloride, sulfonyl chloride, carboxylic ester, a derivative of ethylene oxide or halide to obtain the compound of formula (I); in which A, L, R, R1 to R14, p, q, s and t are as defined in claim 1. [19] 19. Pharmaceutical composition, characterized in that it comprises a therapeutically effective amount of the compound of formula (I), or of a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically salt. acceptable term thereof, as defined in any one of claims 1 to 15, and a pharmaceutically acceptable carrier. [20] 20. Use of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, as defined in any one of claims 1 to 15, or the pharmaceutical composition, as defined in claim 19, characterized in that it is in the preparation of a drug to inhibit JAK kinase, preferably to inhibit JAK1, JAK2 or JAK3. [21] 21. Use according to claim 20, characterized in that the drug optionally further contains one or more reagents to regulate the immune system of mammals, anti-cancer agents or anti-inflammatory agents. [22] 22. Use according to claim 20 or 21, characterized in that the drug is useful for the treatment or prevention of the following disorders or diseases: organ transplant rejection, autoimmune diseases, skin diseases , allergenicity disorders, viral diseases, diabetes I and diabetes complications, Alzheimer's disease, dry eye, bone marrow fibrosis; thrombocytosis, polycythemia or cancers. [23] 23. Use according to claim 22, characterized in that organ transplant rejection is allograft rejection or graft versus host disease; autoimmune disease is lupus, multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriasis, ulcerative colitis, Crohn's disease, or autoimmune thyroid disease; the skin disease is psora, itching or atopic dermatitis; the allergenicity disorder is asthma or rhinitis; the viral disease is hepatitis B, hepatitis C, or varicella-zoster virus; cancer is prostate cancer, kidney cancer, liver cancer, pancreatic cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, melanoma, lymphoma, leukemia, cutaneous lymphoma T-cell or cutaneous B-cell lymphoma. [24] 24. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 15, or the pharmaceutical composition, according to claim 19, characterized in that it is for use as a medicine to inhibit JAK kinase, preferably to inhibit JAK1, JAK2 or JAK3. [25] A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 15, or the pharmaceutical composition, according to claim 19, characterized in that it is for use as a medicine combined with one or more reagents to regulate the immune system of mammals, anti-cancer agents or anti-inflammatory agents. [26] 26. A compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, and mixture thereof, and a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 15, or pharmaceutical composition, of according to claim 19, characterized in that it is for use as a medicine for the treatment or prevention of the following disorders or diseases: organ transplant rejection, autoimmune diseases, skin diseases, allergenicity disorders , viral diseases, diabetes I and diabetes complications, Alzheimer's disease, dry eye, bone marrow fibrosis; thrombocytosis, polycythemia or cancers; where organ transplant rejection is allograft rejection or graft versus host disease; the autoimmune disease is lupus, multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriasis, ulcerative colitis, Crohn's disease, or autoimmune thyroid disease; the skin disease is psora, itching or atopic dermatitis; the allergenicity disorder is asthma or rhinitis; the viral disease is hepatitis B, hepatitis C, or varicella-zoster virus; cancer is prostate cancer, kidney cancer, liver cancer, pancreas cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, melanoma, lymphoma, leukemia, cutaneous T-cell lymphoma or cutaneous B-cell lymphoma.
类似技术:
公开号 | 公开日 | 专利标题 US10428074B2|2019-10-01|Pyrrole heteroaryl ring derivative and method of use thereof JP6804524B2|2020-12-23|Bicyclic condensed heteroaryl or aryl compound as IRAK4 modulator CA2944475C|2018-07-10|Bicyclic-fused heteroaryl or aryl compounds AU2008272830B8|2013-12-12|Pyrimidyl cyclopentanes as AKT protein kinase inhibitors JP6165977B2|2017-07-19|Heteroarylpyridone and aza-pyridone amide compounds EA030472B1|2018-08-31|PYRROLO[2,3-d]PYRIMIDINYL, PYRROLO[2,3-b]PYRAZINYL AND PYRROLO[2,3-d]PYRIDINYL ACRYLAMIDES KR101469334B1|2014-12-04|N-9-substituted purine compounds, compositions and methods of use BR112018074621B1|2021-06-08|compounds derived from pyrazolopyrimidine as kinase inhibitors, pharmaceutical composition comprising said compounds and therapeutic use thereof BR112021009880A2|2021-08-17|pyrimidine and derivative of five-membered nitrogen heterocycle, method of preparation thereof and medical uses thereof CA3131293A1|2020-09-03|Jak inhibitor compound and use thereof BR112012011147A2|2021-09-08|COMPOUND, PHARMACEUTICAL COMPOSITION AND USE OF A COMPOUND. BR112021009610A2|2021-08-10|compound, pharmaceutical composition, use of a compound or composition, and, method of inhibiting necrosis, ferroptosis, human rip1 or related indications JP2017508729A|2017-03-30|Furo-3-carboxamide derivatives and methods of use CN113795483A|2021-12-14|Carboxamide-pyrimidine derivatives as SHP2 antagonists CA3080249A1|2019-05-02|Novel pyrazolo-pyrrolo-pyrimidine-dione derivatives as p2x3 inhibitors ES2370973T3|2011-12-26|PIRROLOPIRAZINE QUINASE INHIBITORS.
同族专利:
公开号 | 公开日 US20170044171A1|2017-02-16| TWI601728B|2017-10-11| AU2012357296A1|2014-07-03| HRP20181201T1|2018-10-19| EP2796460A1|2014-10-29| PT2796460T|2018-11-05| WO2013091539A1|2013-06-27| ES2682755T3|2018-09-21| RS57621B1|2018-11-30| CN103415520A|2013-11-27| AU2012357296B2|2017-04-13| CN103415520B|2016-01-20| JP2015500845A|2015-01-08| CN103415520B8|2017-04-05| US9527851B2|2016-12-27| HUE039111T2|2018-12-28| KR20140103300A|2014-08-26| RU2014128307A|2016-02-10| RU2618673C2|2017-05-10| SI2796460T1|2018-10-30| TW201326176A|2013-07-01| JP6075736B2|2017-02-08| US10428074B2|2019-10-01| CY1121161T1|2020-05-29| CA2857977A1|2013-06-27| MX2014007033A|2014-09-16| LT2796460T|2018-10-10| PL2796460T3|2018-12-31| KR102032934B1|2019-10-16| EP2796460B1|2018-07-04| MX358679B|2018-08-31| US20180222912A1|2018-08-09| DK2796460T3|2018-08-27| EP2796460A4|2015-05-20| US20140336207A1|2014-11-13| HK1187615A1|2014-04-11| CA2857977C|2020-10-20|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 JPH0278659A|1988-09-12|1990-03-19|Shionogi & Co Ltd|Compound of azabicycloalkanes| US5521193A|1992-06-24|1996-05-28|G. D. Searle & Co.|Benzimidazole compounds| US5527910A|1992-12-30|1996-06-18|Cheil Foods & Chemicals, Inc.|Pyridone carboxylic acid compounds and their uses for treating infectious diseases caused by bacteria| EA005852B1|1998-06-19|2005-06-30|Пфайзер Продактс Инк.|PYRROLO[2,3-d]PYRIMIDINE COMPOUNDS| OA12118A|1999-12-10|2006-05-04|Pfizer Prod Inc|PyrroloÄ2,3-dÜpyrimidine compounds.| OA12316A|2000-01-24|2006-05-15|Warner Lambert Co|3-Aminoquinazolin-2,4-dione antibacterial agents.| DK1294724T3|2000-06-26|2006-07-17|Pfizer Prod Inc|Pyrrolo [2,3-d] pyrimidine compounds as immunosuppressants| WO2004087142A1|2003-04-04|2004-10-14|Novartis Ag|Quinoline-2-one-derivatives for the treatment of airways diseases| GB0307856D0|2003-04-04|2003-05-14|Novartis Ag|Organic compounds| CA2525945A1|2003-05-21|2004-12-02|Biovitrum Ab|Inhibitors of 11-beta-hydroxy steroid dehydrogenase type i| US7381738B2|2004-02-19|2008-06-03|Bristol-Myers Squibb Company|Substituted bicycloalkylamine derivatives as modulators of chemokine receptor activity| EP1778688A1|2004-07-22|2007-05-02|Glaxo Group Limited|Antibacterial agents| AR054416A1|2004-12-22|2007-06-27|Incyte Corp|PIRROLO [2,3-B] PIRIDIN-4-IL-AMINAS AND PIRROLO [2,3-B] PIRIMIDIN-4-IL-AMINAS AS INHIBITORS OF THE JANUS KINASES. PHARMACEUTICAL COMPOSITIONS.| CA2588220A1|2004-12-23|2006-06-29|Pfizer Products Inc.|Heteroaromatic derivatives useful as anticancer agents| RU2434871C2|2005-02-03|2011-11-27|Вертекс Фармасьютикалз Инкорпорейтед|Pyrrolopyrimidines used as protein kinase inhibitors| CN102127078A|2005-07-14|2011-07-20|安斯泰来制药株式会社|Heterocyclic janus kinase 3 inhibitors| CN101230058A|2007-01-23|2008-07-30|上海恒瑞医药有限公司|Bicycle aza alkyl derivative, preparation method and use in medicine thereof| JP5438028B2|2008-01-23|2014-03-12|ジエンスハンセンファーマセウティカルカンパニーリミテッド|Derivatives of azabicyclooctane, process for producing the same and use thereof as inhibitors of dipeptidyl peptidase IV| WO2009054941A1|2007-10-25|2009-04-30|Merck & Co., Inc.|Therapeutic compounds| TW200944523A|2008-02-08|2009-11-01|Organon Nv|pyrrolo[2,1-a]isoquinolines| EP2419408A4|2009-04-16|2012-09-05|Msd Kk|3-aryl or heteroaryl-substituted indole derivative| TWI466885B|2009-07-31|2015-01-01|Japan Tobacco Inc|Nitrogen-containing spiro cyclic compounds and pharmaceutical use thereof| CN102020643A|2009-09-22|2011-04-20|上海恒瑞医药有限公司|dihydropteridine ketone derivative, and preparation method and medicinal application thereof| EP2632898A4|2010-10-29|2014-04-02|Biogen Idec Inc|Heterocyclic tyrosine kinase inhibitors| EP2686321B1|2011-03-17|2016-11-16|Bristol-Myers Squibb Company|Pyrrolopyridazine jak3 inhibitors and their use for the treatment of inflammatory and autoimmune diseases| EP2721025B1|2011-06-16|2015-11-25|Boehringer Ingelheim International Gmbh|New selective ccr2 antagonists|PE20150371A1|2012-07-17|2015-03-27|Glaxosmithkline Ip No 2 Ltd|INDOLCARBONITRILES AS SELECTIVE MODULATORS OF THE ANDROGEN RECEPTOR| ME03301B|2013-02-22|2019-07-20|Pfizer|Combination of pyrrolo [2, 3 -d]pyrimidine derivatives with one or more additional agents as inhibitors of janus- related kinases | HUE036533T2|2013-06-07|2018-07-30|Jiangsu Hengrui Medicine Co|Bisulfate of janus kinaseinhibitor and preparation method therefor| CN105916843B|2013-09-26|2019-09-20|科登特治疗公司|Octahydro cyclopenta [c] pyroles selective N R2B negative regulator| MA39092B1|2013-12-05|2018-09-28|Pfizer|Pyrrolo [2,3-d] pyrimidinyl, pyrrolo [2,3-b] pyrazinyl and pyrollo [2,3-d] pyridinyl acrylamides| KR101710127B1|2014-08-29|2017-02-27|한화제약주식회사|Substituted N--7H-pyrrolo[2,3-d]pyrimidin-4-amines as Janus kinase inhibitor| JO3579B1|2014-09-26|2020-07-05|Luc Therapeutics Inc|N-alkylaryl-5-oxyaryl- octahydro-cyclopenta[c]pyrrole negative allosteric modulators of nr2b| CN105566327A|2014-10-09|2016-05-11|江苏恒瑞医药股份有限公司|JAK kinase inhibitor bisulfate crystal type I and preparation method thereof| US10023577B2|2014-11-05|2018-07-17|Jiangsu Hengrui Co., Ltd.|Crystalline form of JAK kinase inhibitor bisulfate and a preparation method thereof| CN105712998B|2014-12-05|2019-12-13|上海润诺生物科技有限公司|Azaindole derivatives, preparation method and medical application thereof| WO2016192563A1|2015-05-29|2016-12-08|南京明德新药研发股份有限公司|Janus kinase inhibitor| EP3417861B1|2016-02-19|2020-09-23|Jiangsu Hengrui Medicine Co., Ltd.|Pharmaceutical composition containing jak kinase inhibitor or pharmaceutically acceptable salt thereof| CN107098908B|2016-02-23|2021-01-08|欣凯医药科技有限公司|Preparation method and application of pyrrolopyrimidine compound| CN108884100B|2016-11-23|2021-04-06|江苏恒瑞医药股份有限公司|Preparation method and intermediate of pyrrolo six-membered heteroaromatic ring derivative| CA3047638A1|2016-12-22|2018-06-28|Bayer Cropscience Aktiengesellschaft|Substituted heteroaryl pyrrolones and salts thereof and use thereof as herbicidal active substances| EP3558986A1|2016-12-22|2019-10-30|Bayer CropScience Aktiengesellschaft|Substituted 1,2,4-thiadiazolyl pyrrolones and 1,2,4-thiadiazolyl hydantoines and salts thereof and use thereof as herbicides| WO2018133823A1|2017-01-20|2018-07-26|江苏恒瑞医药股份有限公司|Crystal form of bisulfate of jak kinase inhibitor, and preparation method therefor| WO2019057123A1|2017-09-20|2019-03-28|杭州英创医药科技有限公司|Polycyclic compound acting as ido inhibitor and/or ido-hdac dual inhibitor| WO2019096303A1|2017-11-20|2019-05-23|江苏恒瑞医药股份有限公司|Pharmaceutical composition for topical administration and preparation method therefor| JP2021507894A|2017-12-19|2021-02-25|バイエル、アクチエンゲゼルシャフトBayer Aktiengesellschaft|Substituted N-heterocyclyl- and N-heteroaryl-tetrahydropyrimidinone and salts thereof, and their use as herbicidal active substances| US20210053943A1|2018-03-21|2021-02-25|Xibin Liao|Jak inhibitors| CN112533606A|2018-09-12|2021-03-19|江苏恒瑞医药股份有限公司|Application of JAK kinase inhibitor and EGFR inhibitor in preparation of medicines for treating tumor diseases| CN111205290B|2018-11-22|2021-10-08|江苏恒瑞医药股份有限公司|Crystal form of JAK kinase inhibitor and preparation method thereof| KR20210134738A|2019-03-14|2021-11-10|상하이 시너지 파마슈티컬 사이언시스 코., 엘티디.|JAK inhibitor and its manufacturing method and application in pharmaceutical field| EP3942045A1|2019-03-21|2022-01-26|Onxeo|A dbait molecule in combination with kinase inhibitor for the treatment of cancer| CN110028509B|2019-05-27|2020-10-09|上海勋和医药科技有限公司|Pyrrolopyrimidines as selective JAK2 inhibitors, and synthesis method and application thereof| WO2021089791A1|2019-11-08|2021-05-14|INSERM |Methods for the treatment of cancers that have acquired resistance to kinase inhibitors| WO2021148581A1|2020-01-22|2021-07-29|Onxeo|Novel dbait molecule and its use| WO2022027041A1|2020-07-28|2022-02-03|Arcutis Biotherapeutics, Inc.|Topical formulation containing jak inhibitor and laureth-4|
法律状态:
2018-01-23| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]| 2018-03-27| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]| 2019-10-15| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]| 2019-10-22| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]| 2021-06-01| B350| Update of information on the portal [chapter 15.35 patent gazette]| 2021-12-21| B06A| Patent application procedure suspended [chapter 6.1 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 CN201110434071.9|2011-12-21| CN201110434071|2011-12-21| PCT/CN2012/086922|WO2013091539A1|2011-12-21|2012-12-19|Pyrrole six-membered heteroaryl ring derivative, preparation method therefor, and medicinal uses thereof| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|