WO2017097224A1 - 氮杂环丁烷衍生物、其制备方法及用途 - Google Patents
氮杂环丁烷衍生物、其制备方法及用途 Download PDFInfo
- Publication number
- WO2017097224A1 WO2017097224A1 PCT/CN2016/109036 CN2016109036W WO2017097224A1 WO 2017097224 A1 WO2017097224 A1 WO 2017097224A1 CN 2016109036 W CN2016109036 W CN 2016109036W WO 2017097224 A1 WO2017097224 A1 WO 2017097224A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- reaction
- pyrrolo
- azetidin
- mmol
- Prior art date
Links
- 0 *[n]1c2nc(Cl)nc(Cl)c2cc1 Chemical compound *[n]1c2nc(Cl)nc(Cl)c2cc1 0.000 description 7
- MYTMNNCDIQLYEV-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3nc(C#N)n2)c1)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3nc(C#N)n2)c1)(=O)=O MYTMNNCDIQLYEV-UHFFFAOYSA-N 0.000 description 2
- HQUIOHSYUKWGOM-UHFFFAOYSA-N CCS(N(C1)CC1=CC#N)(=O)=O Chemical compound CCS(N(C1)CC1=CC#N)(=O)=O HQUIOHSYUKWGOM-UHFFFAOYSA-N 0.000 description 2
- QGCBQWVSXZPFIO-UHFFFAOYSA-N CC(C)C1(C)OB(c2c[nH]nc2)OC1(C)C Chemical compound CC(C)C1(C)OB(c2c[nH]nc2)OC1(C)C QGCBQWVSXZPFIO-UHFFFAOYSA-N 0.000 description 1
- ZDQIXJDZKHFAJT-UHFFFAOYSA-N CC(C)CS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CC(C)CS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O ZDQIXJDZKHFAJT-UHFFFAOYSA-N 0.000 description 1
- HIIYTBVTZSJMSM-UHFFFAOYSA-N CC(C)S(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CC(C)S(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O HIIYTBVTZSJMSM-UHFFFAOYSA-N 0.000 description 1
- HMPKISDTEVDGSO-UHFFFAOYSA-N CC(C)[Si+](C(C)C)(C(C)C)[n]1cc(B2OC(C)(C)C(C)(C)O2)cc1 Chemical compound CC(C)[Si+](C(C)C)(C(C)C)[n]1cc(B2OC(C)(C)C(C)(C)O2)cc1 HMPKISDTEVDGSO-UHFFFAOYSA-N 0.000 description 1
- ROEHREGDMNNAOY-XYOKQWHBSA-N CC/C(/c1c(c(Cl)c[nH]2)c2ncn1)=C\N(C(CC#N)(C1)CN1S(CC)(=O)=O)N Chemical compound CC/C(/c1c(c(Cl)c[nH]2)c2ncn1)=C\N(C(CC#N)(C1)CN1S(CC)(=O)=O)N ROEHREGDMNNAOY-XYOKQWHBSA-N 0.000 description 1
- KLSXACKMKSRIDU-UHFFFAOYSA-N CC1(CC1)S(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CC1(CC1)S(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O KLSXACKMKSRIDU-UHFFFAOYSA-N 0.000 description 1
- IBOWTTRZVLYQNA-UHFFFAOYSA-N CCC(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)=O Chemical compound CCC(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)=O IBOWTTRZVLYQNA-UHFFFAOYSA-N 0.000 description 1
- QTTVFBMPHNQQGE-UHFFFAOYSA-N CCCCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(O)=O Chemical compound CCCCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(O)=O QTTVFBMPHNQQGE-UHFFFAOYSA-N 0.000 description 1
- MTRRZGZVHTYSCU-UHFFFAOYSA-N CCCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CCCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O MTRRZGZVHTYSCU-UHFFFAOYSA-N 0.000 description 1
- URYHTJUZJKTDDB-UHFFFAOYSA-N CCN(C1)CC1(CC#N)[n]1ncc(-c2c3nc[nH]c3ncn2)c1 Chemical compound CCN(C1)CC1(CC#N)[n]1ncc(-c2c3nc[nH]c3ncn2)c1 URYHTJUZJKTDDB-UHFFFAOYSA-N 0.000 description 1
- AIWPXJIFJMOIFE-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n](cc1)cc1-c1c(cc[nH]2)c2ncc1)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n](cc1)cc1-c1c(cc[nH]2)c2ncc1)(=O)=O AIWPXJIFJMOIFE-UHFFFAOYSA-N 0.000 description 1
- XLJXDCQWYYQDQT-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n](cc1)cc1-c1c(cc[nH]2)c2ncc1C#N)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n](cc1)cc1-c1c(cc[nH]2)c2ncc1C#N)(=O)=O XLJXDCQWYYQDQT-UHFFFAOYSA-N 0.000 description 1
- NIWWCVQLIOVITA-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n]1cnc(-c2c(cc[nH]3)c3ncn2)c1)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n]1cnc(-c2c(cc[nH]3)c3ncn2)c1)(=O)=O NIWWCVQLIOVITA-UHFFFAOYSA-N 0.000 description 1
- QLUVNBXAVDKOTB-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n]1nc(C)c(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n]1nc(C)c(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O QLUVNBXAVDKOTB-UHFFFAOYSA-N 0.000 description 1
- OTEICFLBCWCABS-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc([nH]3)Cl)c3ncn2)c1)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc([nH]3)Cl)c3ncn2)c1)(=O)=O OTEICFLBCWCABS-UHFFFAOYSA-N 0.000 description 1
- CKXWJAPUCSPQHF-UHFFFAOYSA-N CCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CCS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O CKXWJAPUCSPQHF-UHFFFAOYSA-N 0.000 description 1
- CYNXBSSBVIGQCC-UHFFFAOYSA-N CCS(N(C1)CC1(CF)[n]1ncc(-c2c(cc[nH]3)c3ncc2C#N)c1)(=C)=O Chemical compound CCS(N(C1)CC1(CF)[n]1ncc(-c2c(cc[nH]3)c3ncc2C#N)c1)(=C)=O CYNXBSSBVIGQCC-UHFFFAOYSA-N 0.000 description 1
- SCENKFBJGKNNHX-LBPRGKRZSA-N CCS(N(C1)CC1([C@@H](C)C#N)[n]1ncc(-c2ncnc3c2cc[nH]3)c1)(=O)=O Chemical compound CCS(N(C1)CC1([C@@H](C)C#N)[n]1ncc(-c2ncnc3c2cc[nH]3)c1)(=O)=O SCENKFBJGKNNHX-LBPRGKRZSA-N 0.000 description 1
- FSLKDQDNHNMMQO-ZDUSSCGKSA-N CC[C@H](C)S(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2C(N)=O)c1)(=O)=O Chemical compound CC[C@H](C)S(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2C(N)=O)c1)(=O)=O FSLKDQDNHNMMQO-ZDUSSCGKSA-N 0.000 description 1
- DUGFRSWSRSEYCH-UHFFFAOYSA-N CS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O Chemical compound CS(N(C1)CC1(CC#N)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1)(=O)=O DUGFRSWSRSEYCH-UHFFFAOYSA-N 0.000 description 1
- GHXBPCSSQOKKGB-UHFFFAOYSA-N Clc1c(cc[nH]2)c2nc(Cl)n1 Chemical compound Clc1c(cc[nH]2)c2nc(Cl)n1 GHXBPCSSQOKKGB-UHFFFAOYSA-N 0.000 description 1
- JHDITXGBIGFJTG-UHFFFAOYSA-N N#CCC(C1)(CN1C(C1CC1)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 Chemical compound N#CCC(C1)(CN1C(C1CC1)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 JHDITXGBIGFJTG-UHFFFAOYSA-N 0.000 description 1
- ARPQOFJMHYOADI-UHFFFAOYSA-N N#CCC(C1)(CN1C(c1ccccc1)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 Chemical compound N#CCC(C1)(CN1C(c1ccccc1)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 ARPQOFJMHYOADI-UHFFFAOYSA-N 0.000 description 1
- WCLJHVYTJKESBY-UHFFFAOYSA-N N#CCC(C1)(CN1S(C1CC1)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 Chemical compound N#CCC(C1)(CN1S(C1CC1)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 WCLJHVYTJKESBY-UHFFFAOYSA-N 0.000 description 1
- KYVZBLQQLBOCDU-UHFFFAOYSA-N N#CCC(C1)(CN1S(CCC(F)(F)F)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 Chemical compound N#CCC(C1)(CN1S(CCC(F)(F)F)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 KYVZBLQQLBOCDU-UHFFFAOYSA-N 0.000 description 1
- FUROSCLCECVZNR-UHFFFAOYSA-N N#CCC(C1)(CN1S(c1ccccc1)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 Chemical compound N#CCC(C1)(CN1S(c1ccccc1)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2)c1 FUROSCLCECVZNR-UHFFFAOYSA-N 0.000 description 1
- UMALZZVSLVGHFK-UHFFFAOYSA-N N#CCC(C1)(CN1S(c1ccccc1)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2C#N)c1 Chemical compound N#CCC(C1)(CN1S(c1ccccc1)(=O)=O)[n]1ncc(-c2c(cc[nH]3)c3ncc2C#N)c1 UMALZZVSLVGHFK-UHFFFAOYSA-N 0.000 description 1
- NXHIMYIDESVDMD-UHFFFAOYSA-N N#CCC1(CNC1)[n]1ncc(-c2ccnc3c2cc[nH]3)c1 Chemical compound N#CCC1(CNC1)[n]1ncc(-c2ccnc3c2cc[nH]3)c1 NXHIMYIDESVDMD-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/4353—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
- A61K31/437—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/34—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
- A61K31/343—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
- A61K38/1793—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
Definitions
- the present invention relates to an azetidine derivative as a Janus kinase (JAK) inhibitor, a pharmaceutical composition comprising the same, a process for the preparation thereof, and JAK correlation thereof in the treatment including, for example, inflammatory diseases, autoimmune diseases, and cancer Use in disease.
- JAK Janus kinase
- Protein kinases are a series of structurally related enzymes that are primarily responsible for the control of intracellular signal transduction processes. They exert their physiological functions by catalyzing the phosphorylation of proteins (or substrates) to regulate the cellular activities of substrates, including regulation of a variety of important biological processes such as cell growth, survival and differentiation, organ formation and morphogenesis. , new blood vessel formation, tissue repair and regeneration. Many diseases are associated with abnormal intracellular responses triggered by protein kinase regulation.
- JAK is a non-receptor tyrosine protein kinase that is a member of the protein kinase family.
- the molecular weight of JAK is about 120-140 kDa.
- the JAK family has four members: JAK1 (also known as Janus kinase-1), JAK2 (also known as Janus kinase-2), JAK3 (also known as Janus kinase-3), and TYK2 (also known as protein-tyrosine).
- Acid kinase 2 also known as Janus kinase-1
- JAK2 also known as Janus kinase-2
- JAK3 also known as Janus kinase-3
- TYK2 also known as protein-tyrosine
- Acid kinase 2 also known as protein-tyrosine.
- These kinases play their part through interactions with cytokines and cytokine receptors (see Rodig S. et al., "D
- JAK plays a key role in cytokine signaling.
- Downstream substrates for JAK family kinases include signal transduction and transcriptional activation (STAT) proteins.
- STAT signal transduction and transcriptional activation
- Abnormal JAK/STAT signaling is associated with many diseases, such as diseases of the immune system (such as organ transplant rejection), autoimmune diseases (such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I) Diabetes, lupus, psoriasis), allergic conditions (such as asthma, food allergies, atopic dermatitis and rhinitis), skin diseases (such as psoriasis, atopic dermatitis, rash), solid and hematological malignancies ( Such as prostate cancer, kidney cancer, liver cancer, pancreatic cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, leukemia, lymphoma, multiple myeloma) and my
- Blocking signal transduction at the JAK level provides a new approach to the treatment of JAK-related diseases such as inflammatory diseases, autoimmune diseases, myeloproliferative diseases, and cancer. Inhibition of JAK is also expected to have therapeutic benefit in patients suffering from skin immune disorders such as psoriasis and skin sensitization. Therefore, it is desirable to develop novel JAK inhibitors to more effectively treat JAK-related diseases.
- JAK-related diseases such as inflammatory diseases, autoimmune diseases, myeloproliferative diseases, and cancer.
- Inhibition of JAK is also expected to have therapeutic benefit in patients suffering from skin immune disorders such as psoriasis and skin sensitization. Therefore, it is desirable to develop novel JAK inhibitors to more effectively treat JAK-related diseases.
- interleukin IL-6 interleukin-6
- IL-15 interferon
- IFN interferon
- GM-CSF granulocyte-macrophage colony-stimulating factor
- JAK inhibitors For JAK inhibitors, some studies have been conducted (see, for example, Peter Norman, “Selective JAK inhibitors in development for rheumatoid arthritis", Expert Opin. Investig. Drugs, 2014, 23(8), 1067-77). Among them, Baricitinib is a drug candidate for the treatment of rheumatoid arthritis, and several phase III clinical studies have been conducted in the United States (see CN102026999). Tofacitinib is currently the only JAK1 and JAK3 selective inhibitor for the treatment of rheumatoid arthritis marketed by the FDA in the United States (see Kremer, J.
- tolfatinib has a short half-life in humans and needs to be taken twice daily and cannot be combined with a disease-modifying anti-rheumatic drug (DMARD) such as methotrexate. Therefore, it is desirable to develop JAK inhibitors with better JAK selectivity and pharmacokinetic properties that can be combined with DMARDs (eg, methotrexate) to provide better efficacy and reduce adverse effects in patients.
- DMARD disease-modifying anti-rheumatic drug
- the invention provides a compound of formula I, a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, as a JAK inhibitor:
- R 1 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclic, C 6-14 aryl, 5-14 membered heteroaryl, C 7-20 aralkyl, C(O)R 10 and S(O) 2 R 11 ;
- R 2 and R 3 are each independently selected from the group consisting of H, CN, halogen, and C 1-6 alkyl;
- R 4 and R 5 are each independently selected from the group consisting of H, halogen, and CN;
- X is selected from N and CR 6 ;
- Y is selected from N and CR 9 ;
- Z is selected from N and CR 7 ;
- W is selected from N and CR 8 ;
- R 6 , R 7 , R 8 and R 9 are each independently selected from the group consisting of H, halogen, CN, C 1-6 alkyl, C 1-6 alkoxy and C(O)NR 12 R 13 ;
- R 10 and R 11 are each independently selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclic, C 6-14 aryl, 5-14 membered heteroaryl, C 7 -20 aralkyl and NR 12 R 13 ;
- R 12 and R 13 are each independently selected from H and C 1-6 alkyl
- alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl and aralkyl groups are each optionally substituted by 1, 2 or 3 independently selected from the group consisting of halogen, CN and C 1-4 alkyl. Substituted.
- Another aspect of the invention provides a pharmaceutical composition
- a pharmaceutical composition comprising a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, and one or more A pharmaceutically acceptable carrier, and optionally further comprising one or more additional agents for treating a JAK-associated disease.
- a further aspect of the invention provides a method of treating a JAK-associated disease, which comprises The individual is administered a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug or pharmaceutical composition thereof, and optionally includes Individuals in need thereof are additionally administered other drugs for treating JAK-related diseases.
- a further aspect of the invention provides a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug or pharmaceutical composition thereof, for the manufacture of a JAK-associated disease Use in medicine.
- a further aspect of the invention provides a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug or pharmaceutical composition thereof, for use in the treatment of JAK correlation disease.
- a further aspect of the invention provides a method of preparing a compound of the invention, the method comprising:
- halogen is selected from the group consisting of fluorine, chlorine, bromine and iodine, and the remaining groups and substituents are as defined above;
- Compound b is reacted with a suitable reagent in the presence of a base under the catalysis of a palladium catalyst to form compound c;
- Compound d is reacted under Lewis acid catalysis to give a compound of formula I; or compound d is first reacted under acid catalysis, and the resulting product is then treated and reacted in the presence of a base to provide a compound of formula I.
- a further aspect of the invention provides a method of preparing a compound of the invention, the method comprising:
- halogen is selected from the group consisting of fluorine, chlorine, bromine and iodine
- R 14 and R 15 are each independently selected from H and C 1-6 alkyl, or R 14 and R 15 together with the atom to which they are attached constitute a 5- or 6-membered ring system;
- Compound b is reacted with a suitable reagent in the presence of a base under the catalysis of a palladium catalyst to form compound c';
- the compounds of the present invention have various advantages including: excellent JAK kinase inhibitory activity, better selectivity for JAK1 and JAK2 kinases, and more favorable pharmacokinetic properties. Good patient compliance, can be combined with other drugs, and better safety.
- the invention provides a compound of Formula I, a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof:
- R 1 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclic, C 6-14 aryl, 5-14 membered heteroaryl, C 7-20 aralkyl, C(O)R 10 and S(O) 2 R 11 ;
- R 2 and R 3 are each independently selected from the group consisting of H, CN, halogen, and C 1-6 alkyl;
- R 4 and R 5 are each independently selected from the group consisting of H, halogen, and CN;
- X is selected from N and CR 6 ;
- Y is selected from N and CR 9 ;
- Z is selected from N and CR 7 ;
- W is selected from N and CR 8 ;
- R 6 , R 7 , R 8 and R 9 are each independently selected from the group consisting of H, halogen, CN, C 1-6 alkyl, C 1-6 alkoxy and C(O)NR 12 R 13 ;
- R 10 and R 11 are each independently selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclic, C 6-14 aryl, 5-14 membered heteroaryl, C 7 -20 aralkyl and NR 12 R 13 ;
- R 12 and R 13 are each independently selected from H and C 1-6 alkyl
- alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl and aralkyl groups are each optionally substituted by 1, 2 or 3 independently selected from the group consisting of halogen, CN and C 1-4 alkyl. Substituted.
- R 1 is selected from the group consisting of C(O)R 10 and S(O) 2 R 11 .
- R 2 and R 3 are each independently selected from the group consisting of H, CN, F and methyl.
- R 4 and R 5 are each independently selected from the group consisting of H, F, Cl, and CN.
- X, Y, Z or W are each independently selected from N and CH.
- R 6 , R 7 , R 8 and R 9 are each independently selected from the group consisting of H, F, Cl, CN, methyl, ethyl, methoxy and C(O)NH 2 .
- R 10 and R 11 are each independently selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, cyclopropyl, n-butyl, isobutyl, tert-butyl, and aziridine.
- R 12 and R 13 are each independently selected from the group consisting of H, methyl and ethyl.
- the present invention encompasses compounds of formula I which result from the arbitrary combination of the above preferred groups.
- the compound of Formula I or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, is a compound of Formula II or a pharmaceutical thereof Acceptable salts, stereoisomers, polymorphs, solvates, metabolites or prodrugs:
- the compound of Formula II, or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof is a compound of Formula III or a pharmaceutical thereof
- W or Z is N; or W is CR 8 , Z is CR 7 , and R 2 , R 4 , R 5 and R 8 are not H at the same time.
- the compound of Formula II, or a pharmaceutically acceptable salt thereof, A stereoisomer, polymorph, solvate, metabolite or prodrug is a compound of formula IV or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite thereof or Prodrug,
- R 1 is selected from the group consisting of C(O)R 10 and S(O) 2 R 11 ;
- R 10 and R 11 are each independently selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclic, C 6-14 aryl, 5-14 membered heteroaryl, C 7 a -20 aralkyl group and NR 12 R 13 , wherein the alkyl group, cycloalkyl group, heterocyclic group, aryl group, heteroaryl group and aralkyl group are each independently selected from 1, 2 or 3 independently Substituents for halogen, CN and C 1-4 alkyl groups.
- the compound of Formula IV or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, is a compound of Formula V or a pharmaceutical thereof
- the compound of Formula V or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, is a compound of Formula VI or a pharmaceutical thereof
- the compound of the invention is a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, the compound of the invention being selected from the group consisting of:
- the invention relates to a method of preparing a compound of the invention, the method comprising:
- halogen is selected from the group consisting of fluorine, chlorine, bromine and iodine, and the remaining groups and substituents are as defined above;
- Compound a and SEMCl are reacted in a polar aprotic solvent (for example, DMF or DMSO) in a polar aprotic solvent (for example, DMF or DMSO) at 0 ° C to room temperature for 3 to 24 hours to form a compound.
- a polar aprotic solvent for example, DMF or DMSO
- a polar aprotic solvent for example, DMF or DMSO
- Compound b is catalyzed by a palladium catalyst (eg, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(OAc) 2 ) in the presence of a base such as sodium carbonate, potassium carbonate, potassium phosphate or potassium acetate, And reacting with a borate compound for 5-24 hours under inert gas protection and room temperature to 100 ° C to form compound c, the reaction can be carried out in a solvent (for example, a mixed solvent of dioxane and water);
- a solvent for example, a mixed solvent of dioxane and water
- Compound c is reacted with an acrylonitrile compound in the presence of a base (for example, DBU, DMAP, potassium carbonate or triethylamine) at 0 ° C to room temperature for 18-24 hours to form compound d, which may be in a solvent ( For example, in acetonitrile, acetone, N,N-dimethylformamide or dichloromethane);
- a base for example, DBU, DMAP, potassium carbonate or triethylamine
- Compound d is catalyzed by a Lewis acid (for example lithium tetrafluoroborate) at room temperature to 100 ° C for 18-24 hours to give a compound of formula I, which can be carried out in a solvent such as acetonitrile; or compound d first It is catalyzed by an acid (for example, trifluoroacetic acid or a Lewis acid (for example, boron trifluoride diethyl ether)) (for example, in a solvent such as dichloromethane) at 0 ° C to room temperature for 3-8 hours, and then the obtained product is obtained.
- a Lewis acid for example lithium tetrafluoroborate
- an acid for example, trifluoroacetic acid or a Lewis acid (for example, boron trifluoride diethyl ether)
- the invention relates to a method of preparing a compound of the invention, the method comprising:
- halogen is selected from the group consisting of fluorine, chlorine, bromine and iodine, and the remaining groups and substituents are as defined above; the reaction conditions are the same as those described above.
- the invention relates to a method of preparing a compound of the invention, the method comprising:
- the halogen is selected from the group consisting of fluorine, chlorine, bromine and iodine.
- R 14 and R 15 are each independently selected from H and C 1-6 alkyl, or R 14 and R 15 together with the atom to which they are attached form a 5- or 6-membered ring system, and R 14 and R 15 are attached thereto.
- the atoms preferably together form the following groups:
- Compound b is catalyzed by a palladium catalyst (eg, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(OAc) 2 ) in the presence of a base such as sodium carbonate, potassium carbonate, potassium phosphate or potassium acetate,
- a palladium catalyst eg, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(OAc) 2
- the compound c' is formed by reacting with a borate compound at an inert gas atmosphere and at room temperature to 100 ° C for 5 to 24 hours, and the reaction can be carried out in a solvent such as dioxane, water or a mixed solvent thereof;
- Compound c' is catalyzed by a palladium catalyst (for example Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(OAc) 2 ) in the presence of a base such as sodium carbonate, potassium carbonate, potassium phosphate or potassium acetate And reacting with a borate compound for 5-24 hours under inert gas protection and at room temperature to 100 ° C to form a compound d', which can be carried out in a solvent (for example, a mixed solvent of dioxane and water);
- a solvent for example, a mixed solvent of dioxane and water
- Compound d' is reacted with an acrylonitrile compound in the presence of a base (for example, DBU, DMAP, potassium carbonate or triethylamine) at 0 ° C to room temperature for 18-24 hours to form compound e'.
- a base for example, DBU, DMAP, potassium carbonate or triethylamine
- a solvent such as acetonitrile, acetone, N,N-dimethylformamide or dichloromethane;
- Compound e' is reacted at room temperature to 100 ° C under the catalysis of a Lewis acid such as lithium tetrafluoroborate. 18-24 hours to give a compound of formula I, the reaction can be carried out in a solvent such as acetonitrile; or compound e' is first catalyzed by an acid such as trifluoroacetic acid or a Lewis acid (for example boron trifluoride etherate) (for example in a solvent such as dichloromethane), reacting at 0 ° C to room temperature for 3-8 hours, and then subjecting the resulting product to treatment, optionally in the presence of a base such as sodium hydroxide, aqueous ammonia or sodium carbonate. The reaction is carried out in a solvent such as tetrahydrofuran, methanol or ethanol at 0 ° C to room temperature for 10-24 hours to give a compound of formula I.
- a solvent such as tetrahydrofuran, methanol or ethanol at
- the invention relates to a method of preparing a compound of the invention, the method comprising:
- halogen is selected from the group consisting of fluorine, chlorine, bromine and iodine, and the remaining groups and substituents are as defined above; the reaction conditions are the same as those described above.
- alkyl is defined to include saturated aliphatic hydrocarbons, including straight-chain and branched chains.
- an alkyl group has from 1 to 6, such as from 1 to 4 carbon atoms.
- C1-6 alkyl refers to a linear or branched group of 1 to 6 carbon atoms (eg, methyl, ethyl, n-propyl, isopropyl, n-butyl) Base, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl), which is optionally substituted by one or more (such as 1 to 3) suitable substituents such as halogen (eg CF 3 , C 2 F 5 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 Cl or —CH 2 CH 2 CF 3 , etc.).
- suitable substituents such as halogen (eg CF 3 , C 2 F 5 , CHF 2 , CH 2 F,
- C 1-4 alkyl refers to a linear or branched aliphatic hydrocarbon chain of 1 to 4 carbon atoms (ie methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, Sec-butyl or tert-butyl).
- alkoxy refers to a linear, branched or cyclic saturated monovalent hydrocarbon radical of the formula -O-alkyl, wherein the term “alkyl” is as defined above or as defined below.
- Cycloalkyl such as methoxy, ethoxy, n-propoxy, isopropoxy, cyclopropoxy, n-butoxy, isobutoxy, tert-butoxy, sec-butoxy, Cyclobutoxy, pentyloxy, isopentyloxy or n-hexyloxy, or isomers thereof.
- cycloalkyl refers to a saturated or unsaturated, non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (eg, a monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl, Cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, or bicyclic, including spiro, fused or bridged systems (such as bicyclo [1.1.1] pentyl, bicyclo [2.2.1] heptyl, bicyclo [ 3.2.1] Octyl or bicyclo [5.2.0] anthracenyl, decalinyl, etc.), which is optionally substituted by one or more (such as 1 to 3) suitable substituents.
- bicyclic hydrocarbon ring eg, a monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl, Cyclohex
- C 3-10 cycloalkyl refers to a saturated or unsaturated, non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring of 3 to 10 ring-forming carbon atoms ( For example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or bicyclo[1.1.1]pentyl), which is optionally substituted by one or more (such as 1 to 3) suitable substituents, such as A substituted cyclopropyl group.
- the 3-10 membered heterocyclic group is a group having 3 to 10 carbon atoms and a hetero atom in the ring, such as, but not limited to, an oxiranyl group, an aziridine group, an azetidinyl group ( Azetidinyl), oxetanyl, tetrahydrofuranyl, dioxolinyl, pyrrolidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, tetrahydropyridyl Oryl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl or trithianyl.
- an oxiranyl group an aziridine group
- an azetidinyl group Azetidinyl
- oxetanyl tetrahydrofuranyl
- dioxolinyl pyrrolidinyl
- aryl refers to an all-carbon monocyclic or fused-ring polycyclic aromatic group having a conjugated pi-electron system.
- C6-14 aryl means an aromatic group containing from 6 to 14 carbon atoms, such as phenyl or naphthyl.
- the aryl group is optionally substituted with one or more (such as 1 to 3) suitable substituents.
- heteroaryl refers to a monovalent monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 8, 9, 10, 11, 12, 13 or 14 ring atoms, In particular 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atoms, and it contains at least one hetero atom which may be the same or different (the hetero atom is, for example, oxygen, nitrogen or sulfur), and additionally In each case it may be benzofused.
- the heteroaryl is selected from the group consisting of thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thia Diazolyl, etc. and their benzo derivatives; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl and the like, and their benzo derivatives.
- aralkyl preferably denotes an aryl-substituted alkyl group, wherein the aryl group and the alkyl group are as defined herein.
- the aryl group can have from 6 to 14 carbon atoms and the alkyl group can have from 1 to 6 carbon atoms.
- Exemplary aralkyl groups include, but are not limited to, benzyl, phenylethyl, phenylpropyl, phenylbutyl.
- halo or halogen group, as used herein, is defined to include F, Cl, Br or I.
- substituted means that one or more (eg, one, two, three or four) hydrogens on the designated atom are replaced by the selection of the indicated group, provided that the specified atom is not exceeded.
- the normal valence of the current case and the substitution form a stable compound. Combinations of substituents and/or variables are permissible only if such combinations form stable compounds.
- optionally substituted refers to being optionally substituted with a particular group, radical or moiety.
- the compounds of the invention may also comprise one or more (e.g., one, two, three or four) isotopic substitutions.
- H may be in any isotopic form, including 1 H, 2 H (D or ⁇ ) and 3 H (T or ⁇ );
- C may be in any isotopic form, including 12 C, 13 C and 14 C;
- O can be any isotopic form, including 16 O and 18 O.
- stereoisomer denotes an isomer formed by at least one asymmetric center.
- a compound having one or more (eg, one, two, three or four) asymmetric centers which can give racemates, racemic mixtures, single enantiomers, diastereomers Body mixture and individual diastereomers.
- Specific individual molecules can also exist as geometric isomers (cis/trans).
- the compounds of the invention may exist as mixtures (often referred to as tautomers) of two or more different forms in a rapidly balanced structure.
- Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers Wait.
- the invention encompasses all possible crystalline forms or polymorphs of the compounds of the invention, which may be a single polymorph or a mixture of more than one polymorph in any ratio.
- compositions of the invention may exist in free form for treatment or, where appropriate, in the form of their pharmaceutically acceptable derivatives.
- pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, solvates, metabolites or prodrugs which, after administration to a patient in need thereof, can be directly or The compounds of the invention or their metabolites or residues are provided indirectly.
- the pharmaceutically acceptable salts of the compounds of the present invention include the acid addition salts and base addition salts thereof.
- Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include aspartate, bicarbonate/carbonate, hydrogen sulfate, borate, camphorsulfonate, citrate, cyclamate, ethanedisulfonate, ethanesulfonate , fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, hydroiodide/iodide, isethionate, lactate, methyl sulfate, Naphthylate, 2-naphthalenesulfonate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/phosphoric acid Hydrogen salts, pyroglutamate, sugar diacid salts, stearates, tannins and xinofoate.
- Suitable base addition salts are formed from bases which form non-toxic salts. Examples include aluminum salts, arginine salts, benzathine penicillin salts, calcium salts, choline salts, diethylamine salts, diethanolamine salts, glycinates, lysine salts, magnesium salts, meglumine salts, ethanolamine salts, Potassium salt, sodium salt, tromethamine salt and zinc salt.
- the compound of the present invention may exist in the form of a hydrate or a solvate, wherein the compound of the present invention contains a polar solvent as a structural element of the crystal lattice of the compound, particularly such as water, methanol or ethanol.
- a polar solvent as a structural element of the crystal lattice of the compound, particularly such as water, methanol or ethanol.
- the amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric ratios.
- metabolites of the compounds of the invention i.e., compounds formed in vivo upon administration of the drug.
- Prodrugs of the invention may be, for example, by the use of certain groups known to those skilled in the art (e.g., "pro-moieties” as described in H. Bundgaard's Design of Prodrugs (Elsevier, 1985)) It is produced by substituting a suitable functional group present in the compound of formula I.
- compositions and methods of treatment are provided.
- Another aspect of the invention provides a pharmaceutical composition
- a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, polymorph, solvate, metabolite or prodrug thereof, and a One or more pharmaceutically acceptable carriers, and optionally further comprising one or more additional agents for treating JAK-associated diseases.
- a further aspect of the invention provides a method of treating a JAK-associated disease comprising administering to a subject in need thereof a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, polymorph thereof, a solvate, metabolite or prodrug or pharmaceutical composition.
- JAK-related diseases such as, but not limited to, diseases of the immune system (such as organ transplant rejection), autoimmune diseases (such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis). , psoriatic arthritis, type 1 diabetes, lupus, psoriasis), allergic conditions (such as asthma, food allergies, atopic dermatitis and rhinitis), skin diseases (such as psoriasis, atopic dermatitis, rash) , solid and hematological malignancies (eg prostate cancer, kidney cancer, liver cancer, pancreatic cancer, stomach cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, leukemia, lymphoma, multiple myeloma) and Myeloproliferative disorders (including polycythemia, idiopathic thrombocytosis, chronic spontaneous bone marrow fibrosis, extramedullary metaplasi
- diseases of the immune system such as organ transplant rejection
- pharmaceutically acceptable carrier refers to a diluent, adjuvant, excipient or vehicle with which the therapeutic agent is administered, and which is suitable for contacting humans and/or within the scope of sound medical judgment. Or the tissues of other animals without excessive toxicity, irritation, allergic reactions, or other problems or complications corresponding to a reasonable benefit/risk ratio.
- Pharmaceutically acceptable carriers that can be used in the pharmaceutical compositions of the present invention include, but are not limited to, sterile liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, minerals. Oil, sesame oil, etc. Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. It is also possible to use physiological saline and an aqueous solution of glucose and glycerin as a liquid carrier, particularly for injection.
- Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, skimmed milk powder, glycerin, propylene glycol, water, Ethanol and the like.
- the composition may also contain minor amounts of wetting agents, emulsifying agents or pH buffering agents as needed.
- Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like. Examples of suitable pharmaceutically acceptable carriers are as described in Remington's Pharmaceutical Sciences (1990).
- compositions of the invention may act systemically and/or locally.
- they may be administered in a suitable route, for example by injection, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal administration; or by oral, buccal, nasal, transmucosal, topical, It is administered in the form of an ophthalmic preparation or by inhalation.
- compositions of the invention may be administered in a suitable dosage form.
- the dosage forms include, but are not limited to, tablets, capsules, troches, hard candy, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions. Injectable solutions, elixirs, syrups.
- terapéuticaally effective amount refers to an amount of a compound that, to a certain extent, relieves one or more symptoms of the condition being treated after administration.
- the dosing regimen can be adjusted to provide the optimal desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as indicated by the urgent need for treatment. It is noted that the dose value can vary with the type and severity of the condition to be alleviated and can include single or multiple doses. It is to be further understood that for any particular individual, the particular dosage regimen will be adjusted over time according to the individual needs and the professional judgment of the person administering the composition or the composition of the supervised composition.
- an effective dose will be from about 0.0001 to about 50 mg per kg body weight per day, for example from about 0.01 to about 10 mg/kg/day (single or divided doses). For a 70 kg person, this would add up to about 0.007 mg/day to about 3500 mg/day, such as from about 0.7 mg/day to about 700 mg/day.
- a dose level that is not higher than the lower limit of the aforementioned range may be sufficient, while in other cases, a larger dose may still be employed without causing any harmful side effects, provided that the larger The dose is divided into several smaller doses to be administered throughout the day.
- the amount or amount of the compound of the present invention in the pharmaceutical composition may be from about 0.01 mg to about 1000 mg, suitably from 0.1 to 500 mg, preferably from 0.5 to 300 mg, more preferably from 1 to 150 mg, particularly preferably from 1 to 50 mg, for example, 1.5 mg, 2 mg, 4 mg, 10 mg, 25 mg, and the like.
- treating means reversing, alleviating, inhibiting the progression of a condition or condition to which such a term applies or one or more symptoms of such a condition or condition, or Prevention of such a condition or condition or one or more symptoms of such condition or condition.
- “Individual” as used herein includes human or non-human animals.
- Exemplary human individuals include a human individual (referred to as a patient) or a normal individual having a disease, such as the disease described herein.
- “Non-human animals” in the present invention include all vertebrates, such as non-mammals (eg, birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals, and/or domesticated animals (eg, sheep, dogs). , cats, cows, pigs, etc.).
- the pharmaceutical compositions of the present invention may further comprise one or more additional therapeutic or prophylactic agents including, but not limited to, chemotherapeutic or anti-proliferative agents, anti-inflammatory agents, immunomodulators, or Immunosuppressants, neurotrophic factors, agents for treating cardiovascular diseases, agents for treating destructive bone disorders, agents for treating liver diseases, antiviral agents, agents for treating blood diseases, for treating diabetes An agent or agent for treating an immunodeficiency disease.
- the one or more additional therapeutic or prophylactic agents are selected from the group consisting of ezetuzumab, sodium mycophenolate, etanercept, methotrexate and the like.
- the structure of the compound was confirmed by nuclear magnetic resonance spectroscopy ( 1 H NMR) or mass spectrometry (MS).
- reaction was monitored by thin layer chromatography (TLC) or LC-MS using a solvent system of dichloromethane and methanol, n-hexane and ethyl acetate, petroleum ether and ethyl acetate.
- TLC thin layer chromatography
- LC-MS LC-MS using a solvent system of dichloromethane and methanol, n-hexane and ethyl acetate, petroleum ether and ethyl acetate.
- the microwave reaction was carried out using a Biotage Initiator + microwave reactor.
- Column chromatography generally uses 200 to 300 mesh silica gel (Qingdao Ocean) as the stationary phase.
- the system of the eluent includes: dichloromethane and methanol systems and n-hexane and ethyl acetate systems.
- the temperature of the reaction was room temperature (20 ° C to 30 ° C) unless otherwise specified.
- the reagents used in the present application were purchased from companies such as Acros Organics, Aldrich Chemical Company or Tiber Chemical.
- Example 1 4-(1-(3-(Cyanomethyl)-1-((1-methylcyclopropyl)sulfonyl)azetidin-3-yl)-1H-pyrazole-4 -yl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (1)
- Step 5 4-(1-(3-(Cyanomethyl)-1-((1-methylcyclopropyl)sulfonyl)azetidin-3-yl)-1H-pyrazole-4 -yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (1 g)
- Step 6 4-(1-(3-(Cyanomethyl)-1-((1-methylcyclopropyl)sulfonyl)azetidin-3-yl)-1H-pyrazole-4 -yl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (1)
- the clear oil obtained in the first step was dissolved in dichloromethane (10 mL), and the system was placed in an ice bath, then triethylamine was slowly added to a system pH of 9, and then DMAP (8 mg, 0.06 mmol) was added. After stirring for 5 min in an ice-bath, methanesulfonyl chloride (0.3 mL, 3.9 mmol) was dissolved in dichloromethane (2 mL), and slowly added dropwise to the reaction system, and reacted for 30 min.
- reaction solution was extracted, and the organic phase was washed with an aqueous solution of citric acid, and the organic phase was dried over anhydrous sodium sulfate, and then evaporated to dryness to give a solid, and the solid was pulverized with petroleum ether to give 2-(1-(methylsulfonyl) nitrogen heterocycle.
- butane-3-ylidene) acetonitrile (2c) (423mg, as a white solid, yield 82%), MS (ESI, m / z): 173 [m + H] +.
- the third step 4-(1-(3-(cyanomethyl)-1-(methylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (2e)
- Step 4 2-(3-(3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)nitrogen Heterocyclobutane-3-yl)acetonitrile (3)
- the third step 2-(1-(ethylsulfonyl)-3-(4-(3-((2-(trimethylsilyl)ethoxy)methyl)-3H-imidazo[4] ,5-b]pyridin-7-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (4d)
- the fourth step 2-(3-(4-(3H-imidazo[4,5-b]pyridin-7-yl)-1H-pyrazol-1-yl)-1-(ethyl sulfonate) Acyl)azetidin-3-yl)acetonitrile (4)
- the third step 4-(1-(3-(cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (5d)
- 6-Bromo-9-((2-(trimethylsilyl)ethoxy)methyl)-9H-indole (990 mg) and 4-pyrazoleboronic acid pinacol ester at room temperature (780 mg) was dissolved in dioxane (14 mL), then water (4 mL) and EtOAc. Pd(dppf)Cl 2 (222 mg) was added under nitrogen. The reaction system was transferred to a 95 ° C oil bath and allowed to react overnight with stirring. After the substrate was substantially disappeared by TLC, the reaction was quenched by water and extracted with EA.
- the third step 2-(1-(ethylsulfonyl)-3-(4-(9-((2-(trimethylsilyl))ethoxy)methyl)-9H-indole-6- -1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (6d)
- the third step 4-(1-(3-(cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-amide (7d)
- Step 5 4-(1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrazole-4- -1H-pyrrolo[2,3-b]pyridine-5-amide (7)
- Step 4 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)nitrogen Heterocyclobutane-3-yl)acetonitrile (8)
- Step 2 2-(3-(4-(5-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d] Pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)azetidin-3-yl)acetonitrile (9c)
- the third step 2-(3-(4-(5-chloro-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(ethyl) Sulfonyl)azetidin-3-yl)acetonitrile (9)
- Step 5 4-(1H-pyrazol-4-yl)-7-(2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine (12g)
- Step 6 2-(1-(ethylsulfonyl)-3-(4-(7-(2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2, 3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)propanenitrile (12h)
- Step 7 2-(3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)nitrogen Heterocyclobutane-3-yl)propanenitrile (12)
- the synthesized compound (12) was subjected to chiral separation to obtain two isomers (retention times were 6.9 minutes (compound 12-1) and 8.4 minutes (compound 12-2), respectively), and their structures were respectively
- Step 5 3-(Cyanomethyl)-N,N-dimethyl-3-(4-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H- Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-1-sulfonamide (14g)
- Step 6 3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-3-(cyanomethyl)-N,N- Dimethylazetidin-1-sulfonamide (14)
- Step 5 2-(1-propionyl-3-(4-(1-((2-(trimethylsilyl))ethoxy)methyl)-1H-pyrrolo[2,3-b Pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (15g)
- Step 5 2-(1-(cyclopropylcarbonyl)-3-(4-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2 ,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (16g)
- the second step 4-(1-(3-(cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrrol-3-yl)-1-(( 2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (17e)
- the third step 4-(1-(3-(cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrrol-3-yl)-1H-pyrrole [2,3-b]pyridine-5-carbonitrile (17)
- the clear oil obtained in the first step was dissolved in dichloromethane (10 mL), and the reaction was placed in an ice bath, then triethylamine was slowly added until the pH of the reaction reached 9 and then DMAP (8 mg, 0.06) was added. (mmol), the reaction was stirred in an ice bath for 5 min. Then, isopropylsulfonyl chloride (0.44 mL, 3.9 mmol) was dissolved in dichloromethane (2 mL), which was slowly added dropwise to the reaction system, and reacted for 30 min in an ice bath, and then the mixture was extracted with ethyl acetate.
- the third step 2-(1-(isopropylsulfonyl)-3-(4-(1-((2-(trimethylsilyl))ethoxy)methyl)-1H-pyrrolo[ 2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (18e)
- Step 4 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(methylsulfonyl)nitrogen Heterocyclobutane-3-yl)acetonitrile (19)
- 2,4-Dichloro-7H-pyrrolo[2,3-d]pyrimidine (20a) 500 mg, 2.66 mmol
- DMF 5 mL
- the mixture was cooled to below 5 ° C in an ice salt bath.
- sodium hydride 60 wt%, 138 mg, 3.46 mmol
- Step 5 4-(1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-7H-pyrrole And [2,3-d]pyrimidine-2-carbonitrile (20)
- the third step 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(phenylsulfonyl)nitrogen Heterocyclobutane-3-yl)acetonitrile (21)
- the third step 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(propylsulfonyl)nitrogen Heterocyclobutane-3-yl)acetonitrile (23)
- Second step 2-(1-(isobutylsulfonyl)-3-(4-(1-((2-(trimethylsilyl))ethoxy)methyl)-1H-pyrrolo[ 2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (25c)
- the third step 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(isobutylsulfonyl) Azetidin-3-yl)acetonitrile (25)
- Second step 4-(1-(3-(cyanomethyl)-1-(phenylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (27d)
- the third step 4-(1-(3-(cyanomethyl)-1-(phenylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H-pyrrole And [2,3-b]pyridine-5-carbonitrile (27)
- Second step 2-(1-(ethylsulfonyl)-3-(3-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2 ,3-b]pyridin-4-yl)-1H-pyrrol-1-yl)azetidin-3-yl)acetonitrile (28e)
- Step 5 3-(4-(5-Cyano-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine- 4-yl)-1H-pyrazol-1-yl)-3-(fluoromethyl)azetidin-1-carboxylic acid tert-butyl ester (29 g)
- Step 6 4-(1-(3-(fluoromethyl)azetidin-3-yl)1H-pyrazol-4-yl)-1-((2-(trimethylsilyl)) Ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile hydrochloride (29h)
- Step 7 4-(1-(3-(fluoromethyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine -5-carbonitrile (29i)
- Step 8 4-(1-(1-(ethylsulfonyl)-3-(fluoromethyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H-pyrrole And [2,3-b]pyridine-5-carbonitrile (29)
- the white solid 30b (500 mg) obtained in the first step was added to dichloromethane (20 mL), and (3,3,3-trifluoropropyl)sulfonyl chloride (1.126 g) and TEA (1.16). g) 15 mL of DCM solution. The reaction solution was allowed to react under stirring for 5 hours. After the completion of the reaction, water (10 mL) was added, and the mixture was stirred for 10 minutes. The organic phase was separated and concentrated. MS (ESI, m/z): 495[M+H] + .
- the fourth step 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(3,3,3- Trifluoropropylsulfonyl)azetidin-3-yl)acetonitrile (30)
- Step 5 2-(1-((1-methylcyclopropyl)sulfonyl)-3-(4-(1-((2-(trimethylsilyl)ethoxy)methyl)) -1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (31 g)
- Step 6 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-pyrazol-1-yl)-1-((1-methylcyclopropyl) Sulfonyl)azetidin-3-yl)acetonitrile (31)
- the oil obtained in the first step was dissolved in 14 mL of dichloromethane under ice bath, 14 mL of trifluoroacetic acid was added, the reaction solution was warmed to room temperature, stirred for 1 hour, and after the reaction was completed, 20 mL of ice water was added.
- NCS 1.2 g, 9 mmol
- Second step 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)aza Cyclobutane-3-yl)propanenitrile (33)
- Second step 3-(4-(5-cyano-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine- 4-yl)-1H-pyrazol-1-yl)-3-(cyanomethyl)-N,N-dimethylazetidine-1-sulfonate Amide (34c)
- Step 4 3-(4-(5-Cyano-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-3-(cyanomethyl) -N,N-dimethylazetidin-1-sulfonamide (34)
- Compound (35b) (crude, transparent oil) was obtained as described in the first step of Example 2 and used directly in the next step.
- the third step 2-(1-(pyrrolidin-1-ylsulfonyl)-3-(4-(1-((2-(trimethylsilyl))ethoxy)methyl)-1H- Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile (35e)
- Step 4 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(pyrrolidin-1-yl) Sulfonyl)azetidin-3-yl)acetonitrile (35)
- Example 36 1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-4-(1H-pyrrolo[2,3-b]pyridine -4-yl)-1H-pyrazole-3-carbonitrile (36)
- Second step 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-((2-(trimethylsilyl)) Oxy)methyl)-1H-pyrrolo[2,3-b]pyridine (36c)
- the third step 4-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyridyl Oxazole-3-carbonitrile (36e)
- Step 5 1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-4-(1H-pyrrolo[2,3-b]pyridine -4-yl)-1H-pyrazole-3-carbonitrile (36)
- the fifth step 4-(1-(3-(cyanomethyl)-1-propionylazetidin-3-yl)-1H-pyrazol-4-yl)-1-((2-( Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (37g)
- Step 6 4-(1-(3-(Cyanomethyl)-1-propionylazetidin-3-yl)-1H-pyrazol-4-yl)-1H-pyrrolo[2, 3-b]pyridine-5-carbonitrile (37)
- Step 5 4-(1-(3-(Cyanomethyl)-1-(cyclopropanoyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (38 g)
- Step 6 4-(1-(3-(Cyanomethyl)-1-(cyclopropanoyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H-pyrrole And [2,3-b]pyridine-5-carbonitrile (38)
- Step 3 4-(7-((2-(Trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrrole -2-formaldehyde (39d)
- Step 4 4-(7-((2-(Trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrrole -2-carbonitrile (39e)
- Step 5 1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-4-(7-((2-(trimethylsilane) Ethyl)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrrole-2-carbonitrile (39f)
- Step 6 1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-4-(7H-pyrrolo[2,3-d]pyrimidine -4-yl)-1H-pyrrole-2-carbonitrile (39)
- Second step 4-(1-(3-(cyanomethyl)-1-(propylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H- Pyrrolo[2,3-b]pyridine-5-carbonitrile (40)
- the organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated, and the residue was purified by column chromatography to give intermediate product (200mg) .
- the intermediate solid was dissolved in THF (20 mL), and a 1 mol/L sodium hydroxide solution was added dropwise to adjust the pH of the reaction system to about 10, and the reaction solution was stirred at room temperature. After the reaction was completed, the reaction solution was extracted with EA.
- the third step 1-(3-(cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-4-(7H-pyrrolo[2,3-d]pyrimidine 4-yl)-1H-pyrrole-3-carbonitrile (60)
- Step 5 1-(3-(Cyanomethyl)-1-(ethylsulfonyl)azetidin-3-yl)-4-(1H-pyrrolo[2,3-b]pyridine -4-yl)-1H-pyrrole-2-carbonitrile (61)
- Second step 4-(1-(3-(cyanomethyl)-1-(isopropylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1- ((2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (43d)
- the third step 4-(1-(3-(cyanomethyl)-1-(isopropylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H- Pyrrolo[2,3-b]pyridine-5-carbonitrile (43)
- Example 44 4-(1-(3-(Cyanomethyl)-1-((3,3,3-trifluoropropyl)sulfonyl)azetidin-3-yl)-1H-pyridinium Zin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (44)
- the white solid (44b) (500 mg) obtained in the first step was added to dichloromethane (20 mL), (3,3,3-trifluoropropyl)sulfonyl chloride (1.126 g) was added and the system was placed on ice Under a water bath, TEA (1.16 g) diluted with DCM (15 mL) was slowly added dropwise to the reaction system. After the addition was completed, the reaction was stirred for 5 hours. Water (10 mL) was added to the reaction system, stirred for 10 minutes, and the organic phase was separated. The aqueous phase was back-extracted once with DCM (30 mL). The organic phases were combined and concentrated to dryness. MS (ESI, m/z): 495[M+H] + .
- the third step 4-(1-(3-(cyanomethyl)-1-((3,3,3-trifluoropropyl)sulfonyl)azetidin-3-yl)-1H-pyridyl) Zin-4-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (44e)
- Step 5 4-(1-(3-(Cyanomethyl)-1-((3,3,3-trifluoropropyl)sulfonyl)azetidin-3-yl)-1H-pyridyl) Zin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (44)
- Step 5 4-(1H-pyrazol-4-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b] Pyridine-5-carbonitrile (45g)
- Step 6 4-(1-(3-(1-Cyanoethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1 -((2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile (45h)
- Step 7 4-(1-(3-(1-cyanoethyl)-1-(ethylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-1H -pyrrolo[2,3-b]pyridine-5-carbonitrile (45)
- the compound synthesized in the sixth step (45h) was isolated by chiral chromatography to give stereoisomer (45h-1) (retention time: 2.7 min) and stereoisomer (45h-2) (retention time: 3.2 min) After removing the protecting groups from the stereoisomers (45h-1) and (45h-2), respectively, the compound (45-1) and the compound (45-2) are obtained:
- Example 46 2-(1-(ethylsulfonyl)-3-(4-(5-fluoro-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazole-1 -yl)azetidin-3-yl)acetonitrile hydrochloride (52-HCl)
- the third step 4-bromo-1-p-toluenesulfonyl-1H-pyrazol-3-amine (48d)
- the fourth step 4-bromo-3-chloro-1-p-toluenesulfonyl-1H-pyrazole (48e)
- Step 5 4-(3-Chloro-1H-pyrazol-4-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2, 3-b]pyridine (48g)
- Step 6 2-(3-(3-Chloro-4-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b] Pyridin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)azetidin-3-yl)acetonitrile (48h)
- Step 7 2-(3-(3-Chloro-4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(ethyl) Sulfonyl)azetidin-3-yl)acetonitrile (48)
- the third step 2-(1-(ethylsulfonyl)-3-(3-methyl-4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazole- 1-yl)azetidin-3-yl)acetonitrile (49)
- Step 3 2-(3-(4-(5-Chloro-1-(2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b] Pyridin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)azetidin-3-yl)acetonitrile (50d)
- Step 4 2-(3-(4-(5-Chloro-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(ethyl) Sulfonyl)azetidin-3-yl)acetonitrile (50)
- Example 54 2-(3-(4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)-1-(isobutylsulfonyl) Azetidin-3-yl)acetonitrile hydrochloride (25-HCl)
- the third step 2-(1-(ethylsulfonyl)-3-(3-methyl-4-(7-((2-(trimethylsilyl)ethoxy)methyl)-7H) -pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrrol-1-yl)azetidin-3-yl)acetonitrile (56d)
- Step 4 2-(1-(ethylsulfonyl)-3-(3-methyl-4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrrole-1 -yl)azetidin-3-yl)acetonitrile (56)
- Test Example 1 JAK1, JAK2, JAK3 and TYK2 enzyme activity inhibition assay
- JAK1, JAK2, JAK3 and TYK2 enzymes purchased from Life Technologies;
- Substrate GFP-STAT1 purchased from Life Technologies;
- test compound, enzyme, substrate and ATP were diluted to the desired concentration with assay buffer (40 mM Tris-HCl pH 7.4, 20 mM MgCl 2 , 0.1% BSA), and the first three were added to the multiwell plate and mixed evenly. Incubate at room temperature. Add ATP to initiate the kinase reaction and incubate at room temperature. Add ADP-Glo reagent and incubate at room temperature. Add the kinase assay reagent and incubate at room temperature. The luminescence intensity of each test group was measured, and the half-inhibitory concentration IC 50 value was calculated. The results are shown in Table 1-1 and Table 1-2.
- TYK2 enzyme a method of adding Tb-anti-pSTAT1, incubated at room temperature, using the detected LanthaScreen TR-FRET, to calculate the median value of the inhibition concentration 50 IC.
- Table 1-3 The results are shown in Table 1-3.
- Table 1-1 Example Compound inhibitory concentration IC 50 values of JAK1 and JAK2 activity
- the compounds of the present invention have a good inhibitory effect on JAK1 and JAK2.
- Table 1-2 Example Compound inhibitory concentration IC 50 values of JAK3 activity
- the compounds of the present invention are significantly more inhibitory against JAK1 and JAK2 than JAK3, and the compounds of the present invention have good selectivity for JAK1/2 kinase.
- Table 1-3 IC 50 values of the compounds of the examples against TYK2 enzyme activity inhibition
- the compounds of the present invention are significantly more inhibitory to JAK1 and JAK2 than to TYK2, and the compounds of the present invention have good selectivity for JAK1/2 kinase.
- the 50% inhibitory concentration (IC 50 ) of the compound of the present invention for hERG was more than 30 ⁇ M. Therefore, the compound of the present invention has no inhibitory effect on hERG and has no safety hazard leading to prolongation of the QT interval of the heart.
- CYP450 is the most important enzyme system in drug metabolism, and enzymes involved in metabolism interact with drugs, the most important of which are CYP1A2, CYP2D6 and CYP3A4.
- CYP450 enzymes in the test the measurement of the inhibitory activity of compounds using the enzyme CYP1A2 and CYP3A4 P450-Glo TM CYP1A2 screening system.
- the CYP2D6Cyan screening kit measures the inhibitory activity of the compound on the CYP2D6 enzyme. The test results are shown in Table 2-2:
- the 50% inhibitory concentration (IC 50 ) of the tested compounds for CYP1A2, CYP3A4 and CYP2D6 was greater than 10 ⁇ M. Therefore, the compound of the present invention has no significant inhibitory effect on CYP1A2, CYP2D6 and CYP3A4, and does not cause a safety problem due to the metabolic interaction between different drugs due to inhibition of the enzyme.
- IV intravenous
- PO intragastric
- the dose of IV is 1 mg/kg
- the dose of PO is 0.5-5 mg/kg, respectively.
- the vehicle system is 10% DMSO: 10% solutol: 80% normal saline or 5% DMSO: 5% solutol: 90% normal saline or 10% DMSO: 60% PEG: 30% saline.
- Blood was collected at different time points after administration. Plasma samples were treated with acetonitrile precipitation protein for LC-MS/MS analysis.
- IV intravenous
- Table 3-1 Compound exposure after IV dose of 1 mg/kg to rats (AUC last )
- AUC last Area under the curve at all time points (0-24 hours).
- Table 3-2 Compound half-life (T 1/2 ) after IV dose of 1 mg/kg to rats
- the compound of the present invention has a better exposure amount (AUC last ) and a longer half-life (T 1/2 ) at a dose of 1 mg/kg and an IV administration route relative to Baricitinib, so that the compound of the present invention has Significant pharmacokinetic advantages.
- AUC last Area under the curve at all time points (0-24 hours).
- the compound of the present invention has a bioavailability (F%) of more than 70% and a half-life (T 1/2 ) of 2-4 hours at an administration dose of 0.5-5 mg/kg and a PO administration route, which is significantly better than Baricitinib ( Under the same conditions, the F% was 55% and the T 1/2 was 1.58 hours). Therefore, the compounds of the invention have significant pharmacokinetic advantages.
- test compound was administered to the male rhesus monkey by intravenous (IV) to examine the pharmacokinetic characteristics of the test compound.
- IV dose was 0.5 mg/kg.
- Blood was collected at various time points after IV administration, and plasma samples were treated with acetonitrile precipitation protein for LC-MS/MS analysis.
- the pharmacokinetic parameters were calculated using the non-compartment model using WinNonlin 6.3 software.
- test compound was administered to male rhesus monkeys by gavage (PO) to examine the pharmacokinetic characteristics of the test compound.
- the dose of PO administered is 1 mg/kg.
- Blood was collected at different time points after PO administration. Plasma samples were treated with acetonitrile precipitation protein for LC-MS/MS analysis.
- the pharmacokinetic parameters were calculated using the non-compartment model using WinNonlin 6.3 software.
- AUC last Area under the curve at all time points (0-24 hours).
- the compound of the present invention for example, compound 8 had a better exposure (AUC last ) and a longer half-life (T 1/2 ) than the Baricitinib at a dose of 1 mg/kg and a route of PO administration, and was orally administered.
- the bioavailability (F%) is higher, so the compounds of the invention (e.g., compound 8) have significant pharmacokinetic advantages.
- Test Example 4 In vivo efficacy in a rat collagen-induced arthritis (CIA) model
- Bovine type II collagen and incomplete Freund's adjuvant 1:1 mixed emulsion were intradermally injected into the back and tail roots of Lewis rats. Rat foot thickness, foot volume were measured after 14 days and arthritis scores were performed. D0 before administration, grouped according to the volume of the foot, the test compound was intragastrically administered, the vehicle was 5% DMSO + 5% solutol, administered once a day for two weeks, measuring the thickness of the foot and the volume of the rat, and performing Arthritis score. The test results are shown in Tables 5-1, 5-2 and 5-3 below.
- Compound 8 caused a significant reduction in rat foot thickness over Baricitinib at the same dose of Compound 8 and Baricitinib as the number of days of administration increased.
- Compound 8 caused a significant reduction in rat foot volume over Baricitinib at the same dose of Compound 8 and Baricitinib as the number of days of administration increased.
- Compound 8 reduced the mean arthritis score significantly better than Baricitinib at the same dose of Compound 8 and Baricitinib (the lower the average arthritis score, the more effective the compound is. Improve arthritis symptoms in model animals).
- test results show that the compound of the present invention at a dose of 10 mg/kg can effectively improve the symptoms and scores of the model animals compared to the vehicle group; at the same dose, the compound 8 of the present invention is improved over Baricitinib (control). More obvious. Other compounds of the invention have similar results.
- a tablet containing the following ingredients is prepared.
- compound 5 microcrystalline cellulose and croscarmellose sodium were mixed, and then the mixture was lubricated with magnesium stearate and compressed into tablets.
- a particle-filled capsule containing the active ingredient compound 8 was prepared.
- Compound 8 and lactose were sieved through a 60 mesh sieve.
- the corn starch was sieved through a 120 mesh sieve. These were mixed, and the HPC-L solution was added to the mixed powder, kneaded, granulated, and dried. After the resulting dried granules were sized, they were filled (150 mg) into a No. 4 hard gelatin capsule.
- a granule containing the following ingredients was prepared.
- Compound 31 and lactose were sieved through a 60 mesh sieve.
- the corn starch was sieved through a 120 mesh sieve. They were mixed using a V-type mixer. A low-viscosity hydroxypropylcellulose aqueous solution is added to the mixed powder, and kneading, granulation (extrusion granulation, pore diameter: 0.5 to 1 mm), and drying are carried out. The obtained dried granules were sieved with a shaking sieve (12/60 mesh) to obtain granules.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Rheumatology (AREA)
- Immunology (AREA)
- Epidemiology (AREA)
- Pain & Pain Management (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Physical Education & Sports Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Nitrogen Condensed Heterocyclic Rings (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
本发明涉及作为Janus激酶(JAK)抑制剂的氮杂环丁烷衍生物、包含其的药物组合物、其制备方法及其在治疗包括例如炎性疾病、自身免疫病以及癌症等的JAK相关性疾病中的用途。
Description
本发明涉及作为Janus激酶(JAK)抑制剂的氮杂环丁烷衍生物、包含其的药物组合物、其制备方法及其在治疗包括例如炎性疾病、自身免疫病以及癌症等的JAK相关性疾病中的用途。
发明背景
蛋白激酶是一系列结构上相关的酶,其主要负责细胞内信号转导过程的控制。它们通过催化蛋白(或底物)的磷酸化从而调节底物的细胞活性来发挥它们的生理功能,包括调节各种各样重要的生物过程,如细胞生长、存活和分化、器官形成和形态发生、新血管形成、组织修复和再生。诸多疾病与蛋白激酶调节所引发的细胞内应答异常有关。
JAK是一种非受体型酪氨酸蛋白激酶,属于蛋白激酶家族成员。JAK的分子量为约120-140kDa。在哺乳动物中,JAK家族共有四个成员:JAK1(亦称Janus激酶-1)、JAK2(亦称Janus激酶-2)、JAK3(亦称Janus激酶-3)和TYK2(亦称蛋白质-酪氨酸激酶2)。这些激酶通过与细胞因子和细胞因子受体的相互作用来发挥自己的作用(参见Rodig S.等人,“Disruption of the Jak1gene demonstratesobligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses”,Cell,1998,93(3),373-83)。
JAK在细胞因子信号转导中起着关键作用。JAK家族激酶的下游底物包括信号转导和转录激活(STAT)蛋白。JAK/STAT信号转导异常与许多疾病有关,如免疫系统的疾病(如器官移植排斥反应)、自身免疫病(如多发性硬化、类风湿性关节炎、少年关节炎、牛皮癣关节炎、I型糖尿病、狼疮、银屑病)、变态反应性病症(如哮喘、食物过敏、特应性皮炎和鼻炎)、皮肤病(如银屑病、特应性皮炎、皮疹)、实体和血液恶性肿瘤(如前列腺癌、肾癌、肝癌、胰腺癌、胃癌、乳腺癌、肺癌、头颈部癌、甲状腺癌、恶性胶质瘤、白血病、淋巴瘤、多发性骨髓瘤)以及骨髓增生障碍(包括红细胞增多症、特发性
血小板增多症、慢性自发性骨髓纤维变性、伴有骨髓纤维变性的骨髓外化生、慢性髓细胞白血病、慢性粒-单核细胞型白血病、慢性嗜酸细胞白血病、嗜酸细胞增多综合征和系统性肥大细胞病)。
在JAK水平下阻断信号转导为炎性疾病、自身免疫病、骨髓增殖性疾病和癌症等JAK相关性疾病的治疗提供新的途径。预计JAK的抑制在患有皮肤免疫疾病(如银屑病和皮肤敏化)的患者中也具有治疗益处。因此,期望研发新型JAK抑制剂,以更有效地治疗JAK相关性疾病。
譬如,类风湿性关节炎滑膜组织中白介素IL-6、IL-15、干扰素(IFN)、粒细胞-巨噬细胞集落刺激因子(GM-CSF)等表达水平显著升高,在病症的发生发展过程中发挥重要作用,且上述细胞因子均通过JAK-STAT信号转导通路发挥作用。因此,针对性阻断JAK-STAT通路可以达到改善类风湿性关节炎病理生理过程的目的(参见Joel M.K.等人,Arthritis Rheum.2009,60,1859-1905)。
对于JAK抑制剂,目前已进行了一些研究(参见例如Peter Norman,“Selective JAK inhibitors in development for rheumatoid arthritis”,Expert Opin.Investig.Drugs,2014,23(8),1067-77)。其中,Baricitinib是用于治疗类风湿性关节炎的候选药物,在美国进行多个III期临床研究(参见CN102026999)。托法替尼(Tofacitinib)是目前唯一被FDA批准在美国市场上销售的用于治疗类风湿性关节炎的JAK1和JAK3选择性抑制剂(参见Kremer,J.等人,“The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis:Results of a double-blind,placebo-controlled phase IIa trial of three dosage levels of CP-690,550versus placebo”,Arthritis&Rheumatism,2009,60(7),1895-1905)。然而,患者服用托法替尼后会产生一些不良反应,例如可能的严重感染及增加的癌症和心脏衰竭风险(FDA将严重感染与恶性肿瘤列入黑框警告)。这些不良反应可能是由于托法替尼对JAK3的抑制作用所导致。另外,托法替尼在人体中的半衰期较短,需要每日服用两次,且不能将其与改善病情的抗风湿药(DMARD)(例如甲氨蝶呤)联用。因此,期望开发具有更好的JAK选择性和药物代谢动力学性质,可与DMARD(例如甲氨蝶呤)联用的JAK抑制剂,以提供更好的疗效并减少患者的不良反应。
发明概述
一方面,本发明提供作为JAK抑制剂的式I的化合物,其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药:
其中:
R1选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基、C(O)R10和S(O)2R11;
R2和R3各自独立地选自H、CN、卤素和C1-6烷基;
R4和R5各自独立地选自H、卤素和CN;
X选自N和CR6;
Y选自N和CR9;
Z选自N和CR7;
W选自N和CR8;
R6、R7、R8和R9各自独立地选自H、卤素、CN、C1-6烷基、C1-6烷氧基和C(O)NR12R13;
R10和R11各自独立地选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基和NR12R13;
R12和R13各自独立地选自H和C1-6烷基;
其中上述烷基、环烷基、杂环基、芳基、杂芳基和芳烷基各自任选地被1、2或3个独立地选自卤素、CN和C1-4烷基的取代基取代。
本发明另一方面提供药物组合物,其包含本发明的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药以及一种或多种药学上可接受的载体,并任选地进一步包含一种或多种治疗JAK相关性疾病的其它药物。
本发明再一方面提供治疗JAK相关性疾病的方法,其包括向需要其的
个体给药治疗有效量的本发明的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药或者药物组合物,并任选地包括向需要其的个体另外给药治疗JAK相关性疾病的其它药物。
本发明再一方面提供本发明的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药或者药物组合物在制备治疗JAK相关性疾病的药物中的用途。
本发明再一方面提供本发明的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药或者药物组合物,其用于治疗JAK相关性疾病。
本发明再一方面提供制备本发明的化合物的方法,所述方法包括:
其中卤素选自氟、氯、溴和碘,其余各基团和取代基如上文中所定义;
其中:
化合物a与SEMCl在碱的存在下,于极性非质子溶剂中反应,生成化合物b;
化合物b在碱的存在下,在钯催化剂催化下,与适当的试剂反应,生成化合物c;
化合物c在碱的存在下,与适当的试剂反应,生成化合物d;
化合物d在路易斯酸催化下反应,得到式I的化合物;或者化合物d首先在酸催化下反应,然后将得到的产物经处理后在碱存在下反应,得到式I的化合物。
本发明再一方面提供制备本发明的化合物的方法,所述方法包括:
其中卤素选自氟、氯、溴和碘;
R14和R15各自独立地选自H和C1-6烷基,或者R14和R15连同其所连接的原子共同构成5元或6元环系;并且
其余各基团和取代基如上文中所定义;
其中
化合物a与SEMCl在碱的存在下,于极性非质子溶剂中反应,生成化合物b;
化合物b在碱的存在下,在钯催化剂催化下,与适当的试剂反应,生成化合物c’;
化合物c’在碱的存在下,在钯催化剂催化下,与适当的试剂反应,生成化合物d’;
化合物d’在碱的存在下,与适当的试剂反应,生成化合物e’;
化合物e’在路易斯酸催化下反应,得到式I的化合物;或者化合物e’首先在酸催化下反应,然后将得到的产物经处理后在碱存在下反应,得到式I的化合物。
与现有技术中的JAK抑制剂相比,本发明的化合物具有多种优点,包括:优异的JAK激酶抑制活性,对JAK1和JAK2激酶具有更好的选择性,更有利的药物代谢动力学性质,良好的患者顺应性,可与其它药物联用,以及更佳的安全性等。
发明详述
化合物和制备方法
在一个实施方案中,本发明提供式I的化合物,其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药:
其中:
R1选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基、C(O)R10和S(O)2R11;
R2和R3各自独立地选自H、CN、卤素和C1-6烷基;
R4和R5各自独立地选自H、卤素和CN;
X选自N和CR6;
Y选自N和CR9;
Z选自N和CR7;
W选自N和CR8;
R6、R7、R8和R9各自独立地选自H、卤素、CN、C1-6烷基、C1-6烷氧基和C(O)NR12R13;
R10和R11各自独立地选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基和NR12R13;
R12和R13各自独立地选自H和C1-6烷基;
其中上述烷基、环烷基、杂环基、芳基、杂芳基和芳烷基各自任选地被1、2或3个独立地选自卤素、CN和C1-4烷基的取代基取代。
在一优选的实施方案中,R1选自C(O)R10和S(O)2R11。
在一优选的实施方案中,R2和R3各自独立地选自H、CN、F和甲基。
在一优选的实施方案中,R4和R5各自独立地选自H、F、Cl和CN。
在一优选的实施方案中,X、Y、Z或W各自独立地选自N和CH。
在一优选的实施方案中,R6、R7、R8和R9各自独立地选自H、F、Cl、CN、甲基、乙基、甲氧基和C(O)NH2。
在一优选的实施方案中,R10和R11各自独立地选自甲基、乙基、正丙基、异丙基、环丙基、正丁基、异丁基、叔丁基、氮丙啶基、吡咯烷基、苯基、苄基和N(CH3)2,其中上述基团各自任选地被1、2或3个独立地选自F、CN和甲基的取代基取代。
在一优选的实施方案中,R12和R13各自独立地选自H、甲基和乙基。
本发明涵盖对上述优选基团进行任意组合所得的式I的化合物。
在一优选的实施方案中,所述式I的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药为式II的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药:
其中各基团和取代基如上述式I中所定义。
在一优选的实施方案中,所述式II的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药为式III的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药,
其中:
W或Z之一为N;或者W为CR8,Z为CR7,并且R2、R4、R5和R8不同时为H。
在一优选的实施方案中,所述式II的化合物或其药学上可接受的盐、
立体异构体、多晶型物、溶剂合物、代谢物或前药为式IV的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药,
其中:
R1选自C(O)R10和S(O)2R11;
R10和R11各自独立地选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基和NR12R13,其中所述烷基、环烷基、杂环基、芳基、杂芳基和芳烷基各自任选地被1、2或3个独立地选自卤素、CN和C1-4烷基的取代基取代。
在一优选的实施方案中,所述式IV的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药为式V的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药,
其中各基团和取代基如上述式I中所定义。
在一优选的实施方案中,所述式V的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药为式VI的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药,
其中各基团和取代基如上式I中所定义。
在一优选的实施方案中,本发明的化合物其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药,本发明的化合物选自:
在一优选的实施方案中,本发明涉及制备本发明的化合物的方法,所述方法包括:
其中卤素选自氟、氯、溴和碘,其余各基团和取代基如上文中所定义;
其中
化合物a与SEMCl在碱(例如NaH或LDA)的存在下,于极性非质子溶剂(例如DMF或DMSO)中,在0℃至室温下反应3-24小时,生成化合物
b;
化合物b在碱(例如碳酸钠、碳酸钾、磷酸钾或乙酸钾)的存在下,在钯催化剂(例如Pd(PPh3)4、Pd(dppf)Cl2或Pd(OAc)2)催化下,于惰性气体保护和室温至100℃下,与硼酸酯类化合物反应5-24小时,生成化合物c,所述反应可在溶剂(例如二氧六环和水的混合溶剂)中进行;
化合物c在碱(例如DBU、DMAP、碳酸钾或三乙胺)的存在下,在0℃至室温下,与丙烯腈类化合物反应18-24小时,生成化合物d,所述反应可在溶剂(例如乙腈、丙酮、N,N-二甲基甲酰胺或二氯甲烷)中进行;
化合物d在路易斯酸(例如四氟硼酸锂)催化下,在室温至100℃下反应18-24小时,得到式I的化合物,所述反应可在溶剂(例如乙腈)中进行;或者化合物d首先在酸(例如三氟乙酸或路易斯酸(例如三氟化硼乙醚))催化下(例如在如二氯甲烷的溶剂中),在0℃至室温下反应3-8小时,然后将得到的产物经处理后在碱(例如氢氧化钠、氨水或碳酸钠)存在下任选地在溶剂(如四氢呋喃、甲醇或乙醇)中,于0℃至室温反应10-24小时,得到式I的化合物。
在一优选的实施方案中,本发明涉及制备本发明的化合物的方法,所述方法包括:
其中卤素选自氟、氯、溴和碘,其余各基团和取代基如上文中所定义;反应条件与上文中所述条件相同。
在另一优选的实施方案中,本发明涉及制备本发明的化合物的方法,所述方法包括:
其中卤素选自氟、氯、溴和碘,
其余各基团和取代基如上文中所定义;
其中
化合物a与SEMCl在碱(例如NaH或LDA)的存在下,于极性非质子溶剂(例如DMF或DMSO)中,在0℃至室温下反应3-24小时,生成化合物b;
化合物b在碱(例如碳酸钠、碳酸钾、磷酸钾或乙酸钾)的存在下,在钯催化剂(例如Pd(PPh3)4、Pd(dppf)Cl2或Pd(OAc)2)催化下,于惰性气体保护和室温至100℃下,与硼酸酯类化合物反应5-24小时,生成化合物c’,所述反应可在溶剂(例如二氧六环、水或其混合溶剂)中进行;
化合物c’在碱(例如碳酸钠、碳酸钾、磷酸钾或乙酸钾)的存在下,在钯催化剂(例如Pd(PPh3)4、Pd(dppf)Cl2或Pd(OAc)2)催化下,于惰性气体保护和室温至100℃下,与硼酸酯类化合物反应5-24小时,生成化合物d’,所述反应可在溶剂(例如二氧六环和水的混合溶剂)中进行;
化合物d’在碱(例如DBU、DMAP、碳酸钾或三乙胺)的存在下,在0℃至室温下,与丙烯腈类化合物反应18-24小时,生成化合物e’,所述反应可在溶剂(例如乙腈、丙酮、N,N-二甲基甲酰胺或二氯甲烷)中进行;
化合物e’在路易斯酸(例如四氟硼酸锂)催化下,在室温至100℃下反应
18-24小时,得到式I的化合物,所述反应可在溶剂(例如乙腈)中进行;或者化合物e’首先在酸(例如三氟乙酸或路易斯酸(例如三氟化硼乙醚))催化下(例如在如二氯甲烷的溶剂中),在0℃至室温下反应3-8小时,然后将得到的产物经处理后在碱(例如氢氧化钠、氨水或碳酸钠)存在下任选地在溶剂(如四氢呋喃、甲醇或乙醇)中,于0℃至室温反应10-24小时,得到式I的化合物。
在一优选的实施方案中,本发明涉及制备本发明的化合物的方法,所述方法包括:
其中卤素选自氟、氯、溴和碘,其余各基团和取代基如上文中所定义;反应条件与上文中所述条件相同。
除非在下文中另有定义,本文中所用的所有技术术语和科学术语的含义意图与本领域技术人员通常所理解的相同。提及本文中使用的技术意图指在本领域中通常所理解的技术,包括那些对本领域技术人员显而易见的技术的变化或等效技术的替换。虽然相信以下术语对于本领域技术人员很好理解,但仍然阐述以下定义以更好地解释本发明。
术语“包括”、“包含”、“具有”、“含有”或“涉及”及其在本文中的其它变体形式为包含性的(inclusive)或开放式的,且不排除其它未列举的元素或方法步骤。
如本文中所使用,术语“烷基”定义为包括饱和脂肪族烃,所述饱和脂肪族烃包括直链及支链。在一些实施方案中,烷基具有1至6个,例如1至4个碳原子。例如,如本文中所使用,术语“C1-6烷基”指1至6个碳原子
的线性或支化的基团(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基),其任选地被1或多个(诸如1至3个)适合的取代基如卤素取代(例如CF3、C2F5、CHF2、CH2F、CH2CF3、CH2Cl或-CH2CH2CF3等)。术语“C1-4烷基”指1至4个碳原子的线性或支化的脂肪族烃链(即甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基)。
如本文中所使用,术语“烷氧基”指式-O-烷基的线性的、支化的或环状的饱和一价烃基,其中术语“烷基”如上所定义或者为下文所定义的“环烷基”,例如甲氧基、乙氧基、正丙氧基、异丙氧基、环丙氧基、正丁氧基、异丁氧基、叔丁氧基、仲丁氧基、环丁氧基、戊氧基、异戊氧基或正己氧基,或它们的异构体。
如本文中所使用,术语“环烷基”指饱和或不饱和的非芳族单环或多环(诸如双环)烃环(例如单环,诸如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基,或双环,包括螺环、稠合或桥连系统(诸如双环[1.1.1]戊基、双环[2.2.1]庚基、双环[3.2.1]辛基或双环[5.2.0]壬基、十氢化萘基等),其任选地被1或多个(诸如1至3个)适合的取代基取代。所述环烷基具有3至15个碳原子。例如,术语“C3-10环烷基”指3至10个成环碳原子的饱和或不饱和的非芳族单环或多环(诸如双环)烃环(例如环丙基、环丁基、环戊基、环己基或双环[1.1.1]戊基),其任选地被1或多个(诸如1至3个)适合的取代基取代,例如甲基取代的环丙基。
如本文中所使用,术语“杂环基”指饱和或不饱和的一价单环或双环基团,其在环中具有2、3、4、5、6、7、8或9个碳原子和一个或多个(例如一个、两个、三个或四个)选自C(=O)、O、S、S(=O)、S(=O)2和NRa的含杂原子的基团,其中Ra表示氢原子或C1-6烷基或卤代-C1-6烷基;所述杂环烷基可以通过所述碳原子中的任一个或氮原子(如果存在的话)与分子的其余部分连接。特别地,3-10元杂环基为在环中具有3-10个碳原子及杂原子的基团,例如但不限于环氧乙烷基、氮丙啶基、氮杂环丁烷基(azetidinyl)、氧杂环丁烷基(oxetanyl)、四氢呋喃基、二氧杂环戊烯基(dioxolinyl)、吡咯烷基、吡咯烷酮基、咪唑烷基、吡唑烷基、吡咯啉基、四氢吡喃基、哌啶基、吗啉基、二噻烷基(dithianyl)、硫吗啉基、哌嗪基或
三噻烷基(trithianyl)。
如本文中所使用,术语“芳基”指具有共轭π电子系统的全碳单环或稠合环多环芳族基团。例如,如本文中所使用,术语“C6-14芳基”意指含有6至14个碳原子的芳族基团,诸如苯基或萘基。芳基任选地被1或多个(诸如1至3个)适合的取代基取代。
如本文中所使用,术语“杂芳基”指一价单环、双环或三环芳族环系统,其具有5、6、8、9、10、11、12、13或14个环原子,特别是1或2或3或4或5或6或9或10个碳原子,且其包含至少一个可以相同或不同的杂原子(所述杂原子是例如氧、氮或硫),并且,另外在每一种情况下可为苯并稠合的。特别地,杂芳基选自噻吩基、呋喃基、吡咯基、噁唑基、噻唑基、咪唑基、吡唑基、异噁唑基、异噻唑基、噁二唑基、三唑基、噻二唑基、等以及它们的苯并衍生物;或吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基等,以及它们的苯并衍生物。
术语“芳烷基”优选表示芳基取代的烷基,其中所述芳基和所述烷基如本文中所定义。通常,所述芳基可具有6-14个碳原子,并且所述烷基可具有1-6个碳原子。示例性芳烷基包括但不限于苄基、苯基乙基、苯基丙基、苯基丁基。
如本文中所使用,术语“卤代”或“卤素”基团定义为包括F、Cl、Br或I。
术语“取代的”指所指定的原子上的一个或多个(例如一个、两个、三个或四个)氢被从所指出的基团的选择代替,条件是未超过所指定的原子在当前情况下的正常原子价并且所述取代形成稳定的化合物。取代基和/或变量的组合仅仅当这种组合形成稳定的化合物时才是允许的。
术语“任选地取代”指任选地被特定的基团、原子团或部分取代。
当取代基的键显示为穿过环中连接两个原子的键时,则这样的取代基可键连至该可取代的环中的任一成环原子。
本发明的化合物还可以包含一个或多个(例如一个、两个、三个或四个)同位素置换。例如,在所述化合物中,H可是任何同位素形式,包括1H、2H(D或氘)和3H(T或氚);C可是任何同位素形式,包括12C、13C和14C;O可是任何同位素形式,包括16O和18O等。
术语“立体异构体”表示由于至少一个不对称中心形成的异构体。在具
有一个或多个(例如一个、两个、三个或四个)不对称中心的化合物中,其可产生外消旋体、外消旋混合物、单一对映异构体、非对映异构体混合物和单独的非对映异构体。特定个别分子也可以几何异构体(顺式/反式)存在。类似地,本发明的化合物可以两种或更多种处于快速平衡的结构不同的形式的混合物(通常称作互变异构体)存在。互变异构体的代表性实例包括酮-烯醇互变异构体、苯酚-酮互变异构体、亚硝基-肟互变异构体、亚胺-烯胺互变异构体等。要理解,本申请的范围涵盖所有这样的以任意比例(例如60%、65%、70%、75%、80%、85%、90%、95%、96%、97%、98%、99%)的异构体或其混合物。
本发明涵盖本发明的化合物的所有可能的结晶形式或多晶型物,其可为单一多晶型物或多于一种多晶型物的任意比例的混合物。
还应当理解,本发明的某些化合物可以游离形式存在用于治疗,或适当时,以其药学上可接受的衍生物形式存在。根据在本发明中,药学上可接受的衍生物包括但不限于,药学上可接受的盐、溶剂合物、代谢物或前药,在将它们向需要其的患者给药后,能够直接或间接提供本发明的化合物或其代谢物或残余物。
本发明的化合物的药学上可接受的盐包括其酸加成盐及碱加成盐。
适合的酸加成盐由形成无毒盐的酸来形成。实例包括天冬氨酸盐、碳酸氢盐/碳酸盐、硫酸氢盐、硼酸盐、樟脑磺酸盐、柠檬酸盐、环己氨磺酸盐、乙二磺酸盐、乙磺酸盐、延胡索酸盐、葡庚糖酸盐、葡糖酸盐、葡糖醛酸盐、六氟磷酸盐、氢碘酸盐/碘化物、羟乙基磺酸盐、乳酸盐、甲基硫酸盐、萘甲酸盐(naphthylate)、2-萘磺酸盐、烟酸盐、硝酸盐、乳清酸盐、草酸盐、棕榈酸盐、双羟萘酸盐、磷酸盐/磷酸氢盐/磷酸二氢盐、焦谷氨酸盐、糖二酸盐、硬脂酸盐、单宁酸盐及昔萘酸盐(xinofoate)。
适合的碱加成盐由形成无毒盐的碱来形成。实例包括铝盐、精氨酸盐、苄星青霉素盐、钙盐、胆碱盐、二乙胺盐、二乙醇胺盐、甘氨酸盐、赖氨酸盐、镁盐、葡甲胺盐、乙醇胺盐、钾盐、钠盐、氨丁三醇盐及锌盐。
适合的盐的综述参见Stahl及Wermuth的“Handbook of Pharmaceutical Salts:Properties,Selection,and Use”(Wiley-VCH,2002)。用于制备本发明的化合物的药学上可接受的盐的方法为本领域技术人员已知的。
本发明的化合物可以水合物或溶剂合物的形式存在,其中本发明的化合物包含作为所述化合物晶格的结构要素的极性溶剂,特别是例如水、甲醇或乙醇。极性溶剂特别是水的量可以化学计量比或非化学计量比存在。
在本发明的范围内还包括本发明的化合物的代谢物,即在给药药物时体内形成的化合物。
本发明的前药可例如通过用本领域技术人员已知的某些基团(例如在H.Bundgaard的Design of Prodrugs(Elsevier,1985)中所描述的“前基团(pro-moieties)”)来替换式I的化合物中存在的适当官能团来产生。
药物组合物和治疗方法
本发明另一方面提供药物组合物,其包含治疗有效量的本发明的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药以及一种或多种药学上可接受的载体,并任选地进一步包含一种或多种治疗JAK相关性疾病的其它药物。
本发明再一方面提供治疗JAK相关性疾病的方法,其包括向需要其的个体给药治疗有效量的本发明的化合物或其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药或者药物组合物。
本发明的药物组合物和方法可用于治疗JAK相关性疾病,例如但不限于免疫系统的疾病(如器官移植排斥反应)、自身免疫病(如多发性硬化、类风湿性关节炎、少年关节炎、牛皮癣关节炎、I型糖尿病、狼疮、银屑病)、变态反应性病症(如哮喘、食物过敏、特应性皮炎和鼻炎)、皮肤病(如银屑病、特应性皮炎、皮疹)、实体和血液恶性肿瘤(如前列腺癌、肾癌、肝癌、胰腺癌、胃癌、乳腺癌、肺癌、头颈部癌、甲状腺癌、恶性胶质瘤、白血病、淋巴瘤、多发性骨髓瘤)以及骨髓增生障碍(包括红细胞增多症、特发性血小板增多症、慢性自发性骨髓纤维变性、伴有骨髓纤维变性的骨髓外化生、慢性髓细胞白血病、慢性粒-单核细胞型白血病、慢性嗜酸细胞白血病、嗜酸细胞增多综合征和系统性肥大细胞病)。特别地,本发明的化合物用于治疗包括例如炎性疾病、自身免疫病以及癌症的JAK相关性疾病。更特别地,本发明的化合物用于治疗类风湿性关节炎。
本发明中“药学上可接受的载体”是指与治疗剂一同给药的稀释剂、辅剂、赋形剂或媒介物,并且其在合理的医学判断的范围内适于接触人类和/
或其它动物的组织而没有过度的毒性、刺激、过敏反应或与合理的益处/风险比相应的其它问题或并发症。
在本发明的药物组合物中可使用的药学上可接受的载体包括但不限于无菌液体,例如水和油,包括那些石油、动物、植物或合成来源的油,例如花生油、大豆油、矿物油、芝麻油等。当所述药物组合物通过静脉内给药时,水是示例性载体。还可以使用生理盐水和葡萄糖及甘油水溶液作为液体载体,特别是用于注射液。适合的药物赋形剂包括淀粉、葡萄糖、乳糖、蔗糖、明胶、麦芽糖、白垩、硅胶、硬脂酸钠、单硬脂酸甘油酯、滑石、氯化钠、脱脂奶粉、甘油、丙二醇、水、乙醇等。所述组合物还可以视需要包含少量的湿润剂、乳化剂或pH缓冲剂。口服制剂可以包含标准载体,如药物级的甘露醇、乳糖、淀粉、硬脂酸镁、糖精钠、纤维素、碳酸镁等。适合的药学上可接受的载体的实例如在Remington’s Pharmaceutical Sciences(1990)中所述。
本发明的组合物可以系统地作用和/或局部地作用。为此目的,它们可以适合的途径给药,例如通过注射、静脉内、动脉内、皮下、腹膜内、肌内或经皮给药;或通过口服、含服、经鼻、透粘膜、局部、以眼用制剂的形式或通过吸入给药。
对于这些给药途径,可以适合的剂型给药本发明的组合物。
所述剂型包括但不限于片剂、胶囊剂、锭剂、硬糖剂、散剂、喷雾剂、乳膏剂、软膏剂、栓剂、凝胶剂、糊剂、洗剂、软膏剂、水性混悬剂、可注射溶液剂、酏剂、糖浆剂。
如本文中所使用的术语“治疗有效量”指被给药后会在一定程度上缓解所治疗病症的一或多种症状的化合物的量。
可调整给药方案以提供最佳所需响应。例如,可给药单次推注,可随时间给药数个分剂量,或可如治疗情况的急需所表明而按比例减少或增加剂量。要注意,剂量值可随要减轻的病况的类型及严重性而变化,且可包括单次或多次剂量。要进一步理解,对于任何特定个体,具体的给药方案应根据个体需要及给药组合物或监督组合物的给药的人员的专业判断来随时间调整。
所给药的本发明的化合物的量会取决于所治疗的个体、病症或病况的
严重性、给药的速率、化合物的处置及处方医师的判断。一般而言,有效剂量在每日每kg体重约0.0001至约50mg,例如约0.01至约10mg/kg/日(单次或分次给药)。对70kg的人而言,这会合计为约0.007mg/日至约3500mg/日,例如约0.7mg/日至约700mg/日。在一些情况下,不高于前述范围的下限的剂量水平可以是足够的,而在其它情况下,仍可在不引起任何有害副作用的情况下采用较大剂量,条件是首先将所述较大剂量分成数个较小剂量以在一整天中给药。
本发明的化合物在药物组合物中的含量或用量可以是约0.01mg至约1000mg,适合地是0.1-500mg,优选0.5-300mg,更优选1-150mg,特别优选1-50mg,例如1.5mg、2mg、4mg、10mg、25mg等。
除非另外说明,否则如本文中所使用,术语“治疗(treating)”意指逆转、减轻、抑制这样的术语所应用的病症或病况或者这样的病症或病况的一或多种症状的进展,或预防这样的病症或病况或者这样的病症或病况的一或多种症状。
如本文所使用的“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
在一些实施方案中,本发明的药物组合物还可以包含一种或多种另外的治疗剂或预防剂,其包括但不限于:化学治疗剂或抗增殖剂、抗炎剂、免疫调节剂或免疫抑制剂、神经营养因子、用于治疗心血管病的药剂、用于治疗破坏性骨障碍的药剂、用于治疗肝病的药剂、抗病毒剂、用于治疗血液病的药剂、用于治疗糖尿病的药剂或用于治疗免疫缺陷性疾病的药剂。特别地,所述一种或多种另外的治疗剂或预防剂选自依法利珠单抗、霉酚酸钠、依那西普和甲氨蝶呤等。
以下结合实施例进一步描述本发明,但提供这些实施例并非意图限制本发明的范围。
本发明中的缩写具有以下含义:
缩写 | 含义 |
ACN | 乙腈 |
BF3C4H10O | 三氟化硼乙醚 |
n-BuOH | 正丁醇 |
CDCl3 | 氘代氯仿 |
CD3OD | 氘代甲醇 |
CH3CN | 乙腈 |
m-CPBA | 间氯过氧苯甲酸 |
DAST | 二乙氨基三氟化硫 |
DBU | 1,8-二氮杂二环十一碳-7-烯 |
DCM | 二氯甲烷 |
DIEA/DIPEA | N,N-二异丙基乙胺 |
DMAP | 4-二甲氨基吡啶 |
DMF | N,N-二甲基甲酰胺 |
DMSO | 二甲基亚砜 |
EA | 乙酸乙酯 |
EtOH | 乙醇 |
h | 小时 |
HCl | 盐酸 |
H2O | 水 |
H2O2 | 过氧化氢 |
HPLC,LC | 高效液相色谱 |
K2CO3 | 碳酸钾 |
KOH | 氢氧化钾 |
LDA | 二异丙基氨基锂 |
LHMDS | 双(三甲基硅基)氨基锂 |
LiBF4 | 四氟硼酸锂 |
MeOH | 甲醇 |
min | 分钟 |
MS | 质谱 |
NaH | 氢化钠 |
Na2CO3 | 碳酸钠 |
NaOH | 氢氧化钠 |
NCS | N-氯代丁二酰亚胺 |
NH4OH | 氨水 |
NMP | N-甲基吡咯烷酮 |
NMR | 核磁共振 |
PE | 石油醚 |
Pd(dppf)Cl2 | 1,1-双(二苯基膦)二茂铁二氯化钯 |
Pd(PPh3)4 | 四(三苯基膦)钯 |
Pd(OAc)2 | 乙酸钯 |
rt | 室温 |
Rt | 保留时间 |
SEM- | [2-(三甲基甲硅烷基)乙氧基]甲基- |
SEMCl | [2-(三甲基甲硅烷基)乙氧基]甲基氯 |
TBAF | 四丁基氟化胺 |
TFA | 三氟乙酸 |
THF | 四氢呋喃 |
TLC | 薄层色谱法 |
化合物的结构通过核磁共振波谱(1H NMR)或质谱(MS)来确证。
反应的监测采用薄层色谱法(TLC)或LC-MS,使用的展开剂体系有:二氯甲烷和甲醇体系、正己烷和乙酸乙酯体系、石油醚和乙酸乙酯体系。
微波反应使用BiotageInitiator+微波反应器进行。
柱色谱法一般使用200~300目硅胶(青岛海洋)为固定相。洗脱剂的体系包括:二氯甲烷和甲醇体系和正己烷和乙酸乙酯体系。
在以下实施例中,如无特殊说明,反应的温度为室温(20℃~30℃)。
本申请中所使用的试剂购自Acros Organics、Aldrich Chemical Company或特伯化学等公司。
实施例1:4-(1-(3-(氰甲基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(1)
第一步:4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(1b)
在室温下,将4-氯-5-氰基-7-氮杂吲哚(1a)(1.92g,10.76mmol)和DMF(22mL)加入到100mL三颈瓶中,用N2保护。将该混合物在冰盐浴中冷却至5℃以下,待反应液搅拌均匀后,向瓶内分批加入氢化钠(60wt%,560mg,13.98mmol),保持体系温度不高于10℃。搅拌1小时后,向体系内缓慢滴加[2-(三甲基甲硅烷基)乙氧基]甲基氯(2.33g,13.98mmol),保持温度不高于5℃,继续搅拌2小时。通过薄层色谱法监测反应,反应结束后将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,用硅胶柱色谱法纯化,得到4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(1b)(4.30g,收率:86%,白色固体)。MS(ESI,m/z):308.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(1c)
在室温下,依次将化合物1b(200mg,0.65mmol)、4-吡唑硼酸频哪醇酯(189mg,0.98mmol)、碳酸钾(225mg,1.63mmol)溶液(2mL)和1,4-二氧六环(8mL)加入到50mL反应瓶中,用N2保护。待反应液搅拌均匀,加入Pd(dppf)Cl2(50mg,0.065mmol),用N2保护。将体系加热至95℃,在回流下反应过夜,通过薄层色谱法监测,反应结束后将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯
并[2,3-b]吡啶(1c)(134mg,收率:61%,黄色固体)。MS(ESI,m/z):340.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(1e)
将化合物1d(1.0g,5.15mmol)和4M盐酸二氧六环溶液(10mL)加入到50mL反应瓶中,用Ar保护,在冰浴下搅拌反应2.5h,逐渐析出白色固体,通过薄层色谱法监测反应,反应结束后将反应液抽滤,用无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(1e)(600mg,收率:90.0%,白色固体),将其直接用于下一步反应。
第四步:2-(1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(1f)
在冰浴下,依次将化合物1e(200mg,1.54mmol)、二氯甲烷(10mL)、三乙胺(1.3mL,9.24mmol)和DMAP(3.8mg,0.03mmol)加入到50mL反应瓶中,搅拌均匀后,向反应体系内缓慢滴加1-甲基环丙基-1-磺酰氯(357mg,2.31mmol)的二氯甲烷溶液(10mL),加毕后搅拌1h,将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸溶液、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到2-(1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(1f)(272mg,收率:83.4%,褐色固体)。MS(ESI,m/z):213.1[M+H]+。
第五步:4-(1-(3-(氰甲基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(1g)
将化合物1f(150mg,0.71mmol)、化合物1c(235mg,0.70mmol)和乙腈(25mL)加入到50mL反应瓶中,将反应液搅拌均匀,随后加入DBU(130mg,0.85mmol),将上述反应液在室温下反应1h,通过薄层色谱法监测,反应结束后,将反应液用水淬灭,在减压下浓缩反应液,用硅胶柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(1g)(179mg,收率:46.5%,白色泡状固体)。MS(ESI,m/z):552.2[M+H]+。
第六步:4-(1-(3-(氰甲基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(1)
在室温下,将化合物1g(179mg,0.33mmol)与TFA/DCM(V:V=1:2,8.5
mL)的混合液加入到50mL反应瓶中,用Ar保护,在室温搅拌3h,LC-MS监测反应结束后,将反应液加水淬灭,用二氯甲烷萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到羟甲基中间体(92mg),并将其置于250mL反应瓶中,加入四氢呋喃(15mL)使样品全部溶解,随后加入1M NaOH,调节pH为10-11,在室温下搅拌24h,反应结束后,将体系内的四氢呋喃蒸除,析出类白色固体,抽滤,滤饼用水洗涤,真空干燥后得到4-(1-(3-(氰甲基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(1)(70mg,收率:40.1%,类白色固体)。1H NMR(400MHz,DMSO-d6)δ:8.83(s,1H),8.65(s,1H),8.34(s,1H),7.78(d,J=3.48Hz,1H),6.89(d,J=3.44Hz,1H),4.59(d,J=8.92Hz,2H),4.27(d,J=8.92Hz,2H),3.73(s,2H),1.49(s,3H),1.22(t,2H),0.93(t,2H),.MS(ESI,m/z):422.1[M+H]+。
实施例2:4-(1-(3-(氰甲基)-1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2)
第一步:2-(氮杂环丁烷-3-亚基)乙腈三氟乙酸盐(2b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(2a)(583mg,3mmol)溶于二氯甲烷(18mL)及三氟乙酸(6mL)的混合液中,在室温下搅拌30min,旋干得透明油状物,将其直接用于下一步反应。
第二步:2-(1-(甲基磺酰基)氮杂环丁烷-3-亚基)乙腈(2c)
将第一步所得的透明油状物溶于二氯甲烷(10mL)中,并将体系置于冰浴下,然后缓慢加入三乙胺至体系pH为9,再加入DMAP(8mg,0.06mmol),在冰浴下搅拌5min后,将甲基磺酰氯(0.3mL,3.9mmol)溶于二氯甲烷(2
mL)中,并缓慢滴加入反应体系,在冰浴下反应30min后,用乙酸乙酯萃取反应液,将有机相用柠檬酸的水溶液洗涤,用无水硫酸钠干燥有机相后,旋干得固体,将固体用石油醚打浆得到2-(1-(甲基磺酰基)氮杂环丁烷-3-亚基)乙腈(2c)(423mg,白色固体,收率为82%),MS(ESI,m/z):173[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2e)
将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2d)(200mg,0.58mmol)和2-(1-(甲基磺酰基)氮杂环丁烷-3-亚基)乙腈(2c)(150mg,0.87mmol)溶于乙腈(10mL)中,并向反应体系中加入DBU(0.16mL),在室温下搅拌过夜,反应结束后,旋干,通过硅胶柱色谱法纯化到4-(1-(3-(氰甲基)-1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2e)(270mg,白色固体,收率为89.5%)。MS(ESI,m/z):512[M+H]+。
第四步:4-(1-(3-(氰甲基)-1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2)
将4-(1-(3-(氰甲基)-1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2e)(270mg,0.52mmol)溶于二氯甲烷(7mL)中,并将反应体系置于冰浴中,加入三氟乙酸(7mL),使整个反应过程中温度小于10℃,TLC显示反应结束后,旋干,加入四氢呋喃(10mL),用1N的氢氧化钠调节体系pH为10,反应结束后,旋干,TLC纯化得到4-(1-(3-(氰甲基)-1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(2)(84mg,白色固体,收率为43%)。1H NMR(400MHz,DMSO-d6)δ:12.42(s,1H),8.82(s,1H),8.63(s,1H),8.32(s,1H),7.76(d,1H,J=3.6Hz),6.89(d,1H,J=3.6Hz),4.61(d,2H,J=9.2Hz),4.29(d,2H,J=9.2Hz),3.70(s,2H),3.14(s,3H).MS(ESI,m/z):382[M+H]+。
实施例3:2-(3-(3-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(3)
第一步:4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(3b)
在冰盐浴冷却下,向4-氯吡咯并嘧啶(3a)(1.0g,6.5mmol)在DMF(15mL)中的溶液分两批加入氢化钠(340mg,60%),保持反应物温度不高于10℃,在氮气保护下搅拌1h。通过注射器缓慢加入SEMCl(1.4g,8.5mmol),保持温度不高于10℃。使反应物温热至室温,并搅拌过夜。用水淬灭反应液,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过快速制备色谱法(PE:EA=19:1)纯化,得到4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(3b)(1.834g,油状产物),收率:97%。MS(ESI,m/z):284[M+H]+。
第二步:4-(1H-吡唑-3-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(3c)
在室温下,于氮气气氛下,将4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(3b)(500mg)和1H-吡唑-3-硼酸(135mg)在二氧六环(10mL)中溶解,然后加入水(5mL)和碳酸钾(414mg),在室温下搅拌10min。于氮气气氛下,加入Pd(PPh3)4(116mg)。将反应体系移至95℃油浴中,在搅拌下反应过夜。TLC检测,底物基本消失,加入水淬灭,用EA萃取,无水硫酸钠干燥有机相。通过快速制备色谱法(PE:EA=1:1)纯化,得到4-(1H-吡唑-3-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(3c)(50mg,固体),收率:9%。MS(ESI,m/z):316[M+H]+。
第三步:2-(1-(乙基磺酰基)-3-(3-(7-((2-(三甲基甲硅烷基)乙氧基)甲
基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(3d)
在室温下,向4-(1H-吡唑-3-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(3c)(45mg)和2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(35mg)的混合物中加入乙腈(2mL),得到反应液。然后加入DBU(25mg),将反应物在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化,得到2-(1-(乙基磺酰基)-3-(3-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(3d)(75mg,棕色产物)。收率:105%(含部分溶剂)。MS(ESI,m/z):502[M+H]+。
第四步:2-(3-(3-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(3)
在室温下,将2-(1-(乙基磺酰基)-3-(3-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(3d)(75mg,0.15mmol)在乙腈(2.5mL)中溶解,加入水(0.5mL),最后加入四氟硼酸锂(140mg),将反应体系移至80℃油浴中,搅拌过夜。将反应物通过加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化(EA:MeOH=15:1),得到2-(3-(3-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(3)(40mg,白色固体),收率:73%。MS(ESI,m/z):372[M+H]+。1H NMR(DMSO-d6,400MHz)δ12.12(s,1H),8.76(s,1H),8.27(d,J=2.8Hz,1H),7.58-7.60(m,1H),7.17(d,J=2.4Hz,2H),4.57(d,J=8.8Hz,2H),4.26(d,J=9.2Hz,2H),3.72(s,2H),3.21-3.26(m,2H),1.21-1.25(m,3H).
实施例4:2-(3-(4-(3H-咪唑并[4,5-b]吡啶-7-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(4)
第一步:7-氯-3-(2-(三甲基甲硅烷基)乙氧基)甲基-3H-咪唑并[4,5-b]吡啶(4b)
在冰盐浴冷却下,向7-氯-3H-咪唑并[4,5-b]吡啶(4a)(1.0g,6.5mmol)在DMF(15mL)中的溶液分两批加入氢化钠(340mg,60%),保持反应物温度不高于10℃,在氮气保护下搅拌1h。通过注射器缓慢加入SEMCl(1.4g,8.5mmol),保持温度不高于10℃。使反应物温热至室温,并搅拌过夜。用水淬灭反应液,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过快速制备色谱法(PE:EA=4:1)纯化,得到7-氯-3-(2-(三甲基甲硅烷基)乙氧基)甲基-3H-咪唑并[4,5-b]吡啶(4b)(1.01g,油状产物),收率:55%。MS(ESI,m/z):284[M+H]+。
第二步:7-(1H-吡唑-4-基)-3-((2-(三甲基甲硅烷基)乙氧基)甲基)-3H-咪唑并[4,5-b]吡啶(4c)
在室温下,于氮气气氛下,将7-氯-3-(2-(三甲基甲硅烷基)乙氧基)甲基-3H-咪唑并[4,5-b]吡啶(4b)(400mg)和4-吡唑硼酸频哪醇酯(383mg)在二氧六环(14mL)中溶解,然后加入水(4mL)和碳酸钾(390mg),在室温下搅拌10min。于氮气保护下,加入Pd(dppf)Cl2(103mg)。将反应体系移至95℃油浴中,在搅拌下反应过夜。TLC检测,底物基本消失,加入水淬灭,用EA萃取,无水硫酸钠干燥有机相。通过快速制备色谱法(PE:EA=3:7)纯化,得到7-(1H-吡唑-4-基)-3-((2-(三甲基甲硅烷基)乙氧基)甲基)-3H-咪唑并[4,5-b]吡啶(4c)(175mg,棕色固体),收率:39%.MS(ESI,m/z):316[M+H]+。
第三步:2-(1-(乙基磺酰基)-3-(4-(3-((2-(三甲基甲硅烷基)乙氧基)甲基)-3H-咪唑并[4,5-b]吡啶-7-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(4d)
在室温下,向7-(1H-吡唑-4-基)-3-((2-(三甲基甲硅烷基)乙氧基)甲基)-3H-咪唑并[4,5-b]吡啶(4c)(175mg)和2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(103mg)的混合物中加入乙腈(6mL),得到反应液。然后加入DBU(100mg),将反应物在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化(DCM:MeOH=20:1),得到2-(1-(乙基磺酰基)-3-(4-(3-((2-(三甲基甲硅烷基)乙氧基)甲基)-3H-咪唑并[4,5-b]吡啶-7-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(4d)(214mg,油状产物)。收率77%.MS(ESI,m/z):502[M+H]+。
第四步:2-(3-(4-(3H-咪唑并[4,5-b]吡啶-7-基)-1H-吡唑-1-基)-1-(乙基磺
酰基)氮杂环丁烷-3-基)乙腈(4)
在室温下,将2-(1-(乙基磺酰基)-3-(4-(3-((2-(三甲基甲硅烷基)乙氧基)甲基)-3H-咪唑并[4,5-b]吡啶-7-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(4d)(210mg,0.40mmol)在乙腈(8mL)中溶解,加入水(1mL),最后加入四氟硼酸锂(373mg),将反应体系移至80℃油浴中,搅拌过夜。将反应物通过加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化,得到2-(3-(4-(3H-咪唑并[4,5-b]吡啶-7-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(4)(110mg,白色固体),收率:71%。MS(ESI,m/z):372[M+H]+。1H NMR(DMSO-d6,400MHz)δ13.15(s,1H),9.06(s,1H),8.64(s,1H),8.47(s,1H),8.31(d,J=4.4Hz,1H),7.57(d,J=5.2Hz,1H),4.52(d,J=9.2Hz,2H),4.26(d,J=9.2Hz,2H),3.68(s,2H),3.22-3.28(m,2H),1.23-1.27(m,3H).
实施例5:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(5)
第一步:4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(5b)
在室温下,将4-氯-5-氰基-7-氮杂吲哚(5a)(1.92g,10.76mmol)和DMF(22mL)加入到100mL三颈瓶中,用N2保护。将反应物在冰盐浴中冷却至5℃以下,在将反应液搅拌均匀后,向瓶内分批加入氢化钠(60wt%,560mg,
13.98mmol),保持反应物温度始终不高于10℃。搅拌1小时后,向反应物中缓慢滴加2-(三甲基甲硅烷基)乙氧甲基氯(2.33g,13.98mmol),保持温度始终不高于5℃,继续搅拌2小时。通过薄层色谱法监测反应,反应结束后将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(5b)(4.30g,收率:86%,白色固体)。MS(ESI,m/z):308[M+H]+。
第二步:4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(5c)
在室温下,将4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(5b)(200mg,0.65mmol)、4-吡唑硼酸频哪醇酯(189mg,0.98mmol)、碳酸钾(225mg,1.63mmol)溶液(2mL)和二氧六环(8mL)依次加入到50mL反应瓶中,用N2保护。在将反应液搅拌均匀后,加入Pd(dppf)Cl2(50mg,0.065mmol),用N2保护。将反应物加热至95℃,并在回流下反应过夜,通过薄层色谱法监测反应,在反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(5c)(134mg,收率:61%,黄色固体)。MS(ESI,m/z):340[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(5d)
依次将4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(5c)(259mg,0.76mmol)、2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(155mg,0.84mmol)和乙腈(10mL)加入到50mL反应瓶中,然后加入DBU(119mg,0.84mmol),在室温下反应2小时。在通过薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(5d)(305mg,收率:76%,白色固体)。MS(ESI,m/z):526[M+H]+。
第四步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(5)
在室温下,将4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(5d)(260mg,0.49mmol)与TFA/DCM(1:1)(8mL)的混合液加入到50mL反应瓶中,在Ar保护下反应2.5小时。在通过薄层色谱法监测反应结束后,将反应物在减压下浓缩,得到黄色油状物。然后将所得的黄色油状物直接溶解于四氢呋喃(10mL)中,在搅拌均匀后,加入1M氢氧化钠溶液,以调节反应物的pH至约10,反应0.5小时。在通过薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化得到目标产物4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(5)(65mg,收率:35%,白色固体)。1H NMR(400MHz,DMSO-d6)δ12.41(s,1H),8.82(d,J=0.8Hz,1H),8.63(s,1H),8.32(d,J=0.8Hz,1H),7.76(dd,J=3.6,2.2Hz,1H),6.88(dd,J=3.7,1.5Hz,1H),4.59(d,J=9.1Hz,2H),4.27(d,J=9.1Hz,2H),3.70(s,2H),3.25(q,J=7.3Hz,2H),1.25(t,J=7.3Hz,3H)ppm.MS(ESI,m/z):396[M+H]+。
实施例6:2-(3-(4-(9H-嘌呤-6-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(6)
第一步:6-溴-9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤(6b)
在冰盐浴冷却下,向6-溴嘌呤(6a)(2.0g,10.0mmol)在DMF(15mL)
中的溶液分三批加入氢化钠(522mg,60%),保持反应物温度不高于10℃,在氮气保护下搅拌1h。缓慢加入SEMCl(2.2g,13.0mmol),保持温度不高于10℃。将反应物温热至室温,并搅拌过夜。用水淬灭反应液,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过快速制备色谱法(PE:EA=17:1)纯化,得到6-溴-9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤(6b)(1.9g,固体产物),收率:58%。MS m/z:329[M+1]+。
第二步:6-(1H-吡唑-4-基)-9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤(6c)
在室温下,将6-溴-9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤(6b)(990mg)和4-吡唑硼酸频哪醇酯(780mg)在二氧六环(14mL)中溶解,然后加入水(4mL)和碳酸钾(1.1g),用氮气保护,将反应物在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(222mg)。将反应体系移至95℃油浴中,并在搅拌下反应过夜。通过TLC检测,在底物基本消失后,将反应物通过加入水淬灭,用EA萃取,无水硫酸钠干燥有机相。通过快速制备色谱法(PE:EA=2:3)纯化,得到6-(1H-吡唑-4-基)-9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤(6c)(400mg,棕色固体),收率:42%。MS m/z:317[M+1]+。
第三步:2-(1-(乙基磺酰基)-3-(4-(9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤-6-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(6d)
在室温下,向6-(1H-吡唑-4-基)-9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤(6c)(200mg)和2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(118mg)的混合物中加入乙腈(10mL),得到反应液。然后加入DBU(120mg),将反应物在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化(DCM:MeOH=10:1),得到2-(1-(乙基磺酰基)-3-(4-(9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤-6-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(6d)(289mg,油状产物)。收率91%。MS m/z:503[M+1]+。
第四步:2-(3-(4-(9H-嘌呤-6-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(6)
在室温下,将2-(1-(乙基磺酰基)-3-(4-(9-((2-(三甲基甲硅烷基)乙氧基)甲基)-9H-嘌呤-6-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(6d)(289mg,0.58mmol)在乙腈(8mL)中溶解,然后加入水(1mL),最后加入四氟硼酸锂(540
mg),将反应物移至80℃油浴锅中,搅拌过夜。将反应物通过加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化,得到2-(3-(4-(9H-嘌呤-6-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(6)(175mg,白色固体),收率:81%。MS m/z:373[M+1]+。1H NMR(400MHz,DMSO-d6)δ13.54(s,1H),9.08(s,1H),8.84(s,1H),8.65(s,1H),8.62(s,1H),4.56(d,J=9.1Hz,2H),4.26(d,J=9.0Hz,2H),3.71(s,2H),3.25(q,J=7.3Hz,2H),1.25(t,J=7.3Hz,3H).
实施例7:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7)
第一步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(7b)
在室温下,于氩气气氛下,向4-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(7a)(500mg,1.76mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1H-吡唑(410mg,2.1mmol)和K2CO3(490mg,4.4mmol)在二氧六环:H2O=500mL:500mL中的溶液加入Pd(PPh3)4(100mg,0.176mmol),用氩气保护,升温至100℃,并反应过夜。TLC(PE:EA=1:1)检测,反应完毕。加入水(500mL),用乙酸乙酯萃取(100mL*3),合并乙酸乙酯层,用饱和盐水洗涤,无水硫酸钠干燥,浓缩,并通过柱色谱法纯化,得到白色固体形式的4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(7b)(500mg,收率:83%)。MS(ESI,m/z):340[M+H]+。
第二步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7c)
在室温下,向4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(7b)(200mg,0.58mmol)在甲醇(2mL)中的溶液依次加入H2O2(30%,1mL)、氢氧化钠水溶液(1M,1mL),然后升温,并反应16h。通过TLC(PE:EA=1:3)检测,反应完毕。加入水(10mL),然后用乙酸乙酯萃取(10mL*3),合并乙酸乙酯层,用饱和盐水洗涤,无水硫酸钠干燥,浓缩,得到白色固体形式的4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7c)(130mg,收率:63%)。MS(ESI,m/z):358[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7d)
在室温下,向4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7c)(130mg,0.36mmol)在乙腈(3mL)中的溶液依次加入2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(71.8mg,0.3mmoL)和DBU(60mg,0.39mmol),然后在室温下反应2h。TLC检测,反应完毕。加入水(10mL),然后用乙酸乙酯萃取(10mL*3),合并乙酸乙酯层,用饱和盐水洗涤,无水硫酸钠干燥,浓缩得到无色油状液体形式的4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7d)(130mg,收率:78%).MS(ESI,m/z):544[M+H]+。
第四步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7e)
在室温下,向4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7d)(130mg,0.23mmol)在DCM(1mL)中的溶液加入TFA(1mL),然后在室温下反应0.5h。TLC检测,原料基本消失。直接干燥浓缩,通过制备硅胶板纯化,以得到无色油状液体形式的4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7e)(60mg,含有少量脱羟甲基产物),将其直接用于下一步反应。MS(ESI,m/z):444[M+H]+。
第五步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-
基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7)
在室温下,向4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7e)(60mg,0.135mmol)在THF(2mL)中的溶液加入氢氧化钠水溶液(1M,1.3mL),然后在室温下反应1h。TLC检测,原料基本消失。然后加入水(10mL),用EA萃取(10mL*3),合并EA层,用饱和盐水洗涤,无水硫酸钠干燥,浓缩得到白色固体形式的4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-酰胺(7)(20mg,34%)。1H NMR(400MHz,氯仿-d)δ9.39(s,1H),8.00(d,J=1.5Hz,1H),7.36(d,J=7.5Hz,1H),7.22(d,J=1.5Hz,1H),6.68(s,2H),6.50(d,J=7.5Hz,1H),4.02(d,J=12.3Hz,2H),3.79(d,J=12.3Hz,2H),3.45(q,J=8.0Hz,2H),2.77(s,2H),1.39(t,J=8.0Hz,3H).MS(ESI,m/z):414[M+H]+。
实施例8:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(8)
第一步:4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(8b)
将4-溴-7-氮杂吲哚(8a)(10g,50.7mmol)和DMF(100mL)加入到250mL三颈瓶中,通过冰盐浴冷却将反应物温度降至-10℃以下,用N2保护,将反应液搅拌均匀,然后在1h内向反应物中分批加入氢化钠(60%,2.64g,54.4mmol),保持温度不超过-5℃。将反应物搅拌1小时,然后向反应物中滴加2-(三甲基甲硅烷基)乙氧甲基氯,保持温度不超过10℃,约1.5h滴加
完毕,将反应物搅拌1小时。薄层色谱法监测反应,原料基本消失。将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(8b)(14.57g,收率:88.0%,黄色液体)。MS(ESI,m/z):326.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(8c)
将化合物4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(8b)(4g,12.3mmol)、4-吡唑硼酸频哪醇酯(3.86g,19.9mmol)和二氧六环(300mL)依次加入到500mL反应瓶中,然后加入碳酸钾(4.58g,33.1mmol)溶液(60mL),将反应液搅拌均匀,随后加入Pd(dppf)Cl2(0.97g,1.33mmol),用N2保护,将反应物加热至95℃,并在回流下反应过夜,薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(8c)(1.67g,收率:43.5%,黄色固体)。MS(ESI,m/z):314.2[M+H]+。
第三步:2-(1-(乙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(8d)
将化合物4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(8c)(200mg,0.64mmol)、2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(118mg,0.64mmol)和乙腈(15mL)加入到50mL反应瓶中,将反应液搅拌均匀,然后加入DBU(116mg,0.76mmol),将上述反应液在室温下反应1h,薄层色谱法监测反应结束后,将反应液用水淬灭,在减压下浓缩反应液,用硅胶柱色谱法纯化,得到化合物2-(1-(乙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(8d)(285mg,收率:89.1%,黄色固体)。MS(ESI,m/z):500.2[M+H]+。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(8)
在室温下,将化合物2-(1-(乙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)
乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(8d)(345mg,0.69mmol)与TFA/DCM(1:1)(6mL)的混合溶液加入到50mL反应瓶中,在Ar保护下,于室温下搅拌1h。LC-MS监测反应结束后,用乙酸乙酯萃取,用无水硫酸钠干燥,在减压下浓缩,得到黄色油状物,在室温下加入四氢呋喃(7.6mL),搅拌均匀后,加入1M氢氧化钠溶液(7.6mL),在室温下搅拌2h。薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用TLC色谱法纯化,得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(8)(40mg,收率:15.7%,黄色固体)。1H NMR(400MHz,DMSO-d6)δ:11.72(s,1H),8.78(s,1H),8.35(s,1H),8.21(d,J=5.00Hz,1H),7.54(t,J=3.04Hz,1H),7.34(d,J=5.00Hz,1H),6.90(dd,J1=3.62Hz,J2=1.84Hz,1H),4.59(d,J=9.02Hz,2H),4.24(d,J=9.02Hz,2H),3.68(s,2H),3.24(q,2H),1.25(t,J=7.29Hz,3H).MS(ESI,m/z):370.1[M+H]+。
实施例9:2-(3-(4-(5-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(9)
第一步:2-(1-(乙基磺酰基)-3-(4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(9b)
在室温下,向4-(1H-吡唑-4-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(9a)(214mg)和2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(126mg)的混合物中加入乙腈(10mL),得到反应溶液。然后加入DBU(120mg),在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化,得到2-(1-(乙基磺酰基)-3-(4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(9b)(327mg,固体产物),收率77%。MS m/z:502[M+1]+。
第二步:2-(3-(4-(5-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(9c)
在室温下,向2-(1-(乙基磺酰基)-3-(4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(9b)(200mg,0.4mmol)中加入DMF(5mL),随后加入NCS(70mg,0.52mmol),搅拌使其溶解。使反应在30℃油浴中反应过夜。TLC检测,底物完全消失。用乙酸乙酯萃取,饱和食盐水洗涤,合并有机相,用无水硫酸钠干燥。浓缩有机相,通过制备硅胶板纯化,得到2-(3-(4-(5-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(9c)(140mg,固体),收率:65%。MS m/z:536[M+1]+。
第三步:2-(3-(4-(5-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(9)
在室温下,将2-(3-(4-(5-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(9c)(120mg,0.23mmol)在乙腈(8mL)中溶解,然后加入水(1mL),最后加入四氟硼酸锂(210mg),将反应体系移至80℃油浴锅中,在搅拌下反应过夜。加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化,得到2-(3-(4-(5-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(9)(57mg,白色固体),收率:63%。MS m/z:406[M+1]+。1H NMR(400MHz,DMSO-d6)δ12.58(s,1H),8.78(d,J=6.1Hz,2H),8.28(s,1H),7.84(d,J=2.6Hz,1H),4.53(d,J=9.0Hz,2H),4.24(d,J=9.0Hz,2H),3.70(s,2H),3.24(q,J=7.3Hz,2H),1.25(d,J=7.3Hz,3H).
实施例10:2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(10)
第一步:4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(10b)
在冰盐浴冷却下,向4-氯吡咯并嘧啶(10a)(3.0g,19.53mmol)在DMF(15mL)中的溶液分两批加入氢化钠(1.1g,60%),保持反应物温度不高于10℃,在氮气保护下,搅拌1h。用注射器缓慢加入SEMCl(4.2g,25.4mmol),保持温度不高于10℃。将反应物温热至室温,并搅拌过夜。用水淬灭反应液,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过快速制备色谱法纯化,得到4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(10b)(4.65g,油状产物),收率:84%。MS(ESI,m/z):284[M+H]+。
第二步:N,N-二甲基-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-磺酰胺(10c)
在室温下,将4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(10b)(150mg)和1-N,N-二甲氨基磺酰基-4-咪唑硼酸频哪醇酯(210mg)在二氧六环(10mL)中溶解,然后加入水(2mL)和碳酸钾(150mg),用氮气保护,在室温下搅拌10min。在氮气保护下,加入Pd(dppf)Cl2(45mg)。将反应物移至95℃油浴中,并搅拌反应过夜。TLC检测,底物基本消失。加入水淬灭,用EA萃取,无水硫酸钠干燥有机相。通过快速制备色谱法纯化,得到N,N-二甲基-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-磺酰胺(10c)(38mg,棕色固体),收率:17%。MS(ESI,m/z):423[M+H]+。
第三步:4-(1H-咪唑-4-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(10d)
在室温下,将N,N-二甲基-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-磺酰胺(10c)(38mg)在乙醇(3mL)中溶解,然后加入氢氧化钾的水溶液(2%,12mL),将反应物移至105℃油浴中,在搅拌下回流5h。停止加热,自然冷却,用乙酸乙酯萃取,无水硫酸钠干燥有机相。将有机相旋转蒸发,得到粗产品形式的4-(1H-咪唑-4-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(10d)(30mg,纯度为80%),将其在未纯化下直接用于下一步反应。MS(ESI,m/z):316[M+H]+。
第四步:2-(1-(乙基磺酰基)-3-(4-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-基)氮杂环丁烷-3-基)乙腈(10e)
在室温下,向4-(1H-咪唑-4-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(10d)(30mg)和2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(20mg)的混合物中加入乙腈(2mL),得到浑浊的反应液。然后加入DBU(20mg),在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化,得到2-(1-(乙基磺酰基)-3-(4-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-基)氮杂环丁烷-3-基)乙腈(10e)(13mg,棕色产物)。MS(ESI,m/z):502[M+H]+。
第五步:2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(10)
在室温下,将2-(1-(乙基磺酰基)-3-(4-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-基)氮杂环丁烷-3-基)乙腈(10e)(13mg,0.003mmol)在乙腈(2.5mL)中溶解,加入水(0.5mL),最后加入四氟硼酸锂(30mg)。将反应物移至80℃油浴锅中,在搅拌下反应过夜。加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化,得到2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-咪唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(10)(8mg,白色固体),收率:83%。MS(ESI,m/z):372[M+H]+。1H NMR(DMSO-d6,400MHz)δ12.01(s,1H),8.67(s,1H),8.33(d,J=1.2Hz,1H),8.23(d,J=1.6Hz,1H),7.51-7.53(m,1H),7.21-7.22(m,1H),4.49(d,J=9.6Hz,2H),4.26(d,J=9.2Hz,2H),3.67(s,2H),3.21-3.26(m,2H),1.21-1.15(m,3H).
实施例11:2-(3-(4-(6-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(11)
第一步:4,6-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(11b)
在冰盐浴冷却下,向4,6-二氯-7H-吡咯并[2,3-d]嘧啶(11a)(1.0g,5.32mmol)在DMF(10mL)中的溶液分两批加入氢化钠(213mg,60%),保持反应物温度不高于10℃,在氮气保护下搅拌1h。用注射器缓慢加入SEMCl(1.2g,7.20mmol),保持温度不高于10℃。将反应物温热至室温,并搅拌过夜。用水淬灭反应液,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过快速制备色谱法纯化,得到4,6-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(11b)(1.0g,油状产物),收率:60%。MS m/z:318[M+1]+。
第二步:6-氯-4-(1H-吡唑-4-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(11c)
在室温下,将4,6-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(11b)(358mg)和4-吡唑硼酸频哪醇酯(327mg)在二氧六环(10mL)中溶解,然后加入水(2mL)和碳酸钾(150mg),用氮气保护,在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(82mg)。将反应物移至95℃油浴锅中,并搅拌反应过夜。TLC检测,底物基本消失。加入水淬灭,用EA萃取,无水硫酸钠干燥有机相。通过快速制备色谱法纯化,得到6-氯-4-(1H-吡唑-4-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(11c)(161mg,棕色油状物),收率:41%。MS m/z:350[M+1]+。
第三步:2-(3-(4-(6-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(11d)
在室温下,向6-氯-4-(1H-吡唑-4-基)-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(11c)(160mg)和2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(85mg)的混合物中加入乙腈(10mL),得到反应液。然后加入DBU(100mg),将反应物在室温下搅拌过夜。将反应物浓缩,通过快速制备色谱法纯化,得到2-(3-(4-(6-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(11d)(177mg,固体产物)。MS m/z:536[M+1]+。
第四步:2-(3-(4-(6-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙
基磺酰基)氮杂环丁烷-3-基)乙腈(11)
在室温下,将2-(3-(4-(6-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(11d)(177mg,0.33mmol)在乙腈(10mL)中溶解,加入水(1mL),最后加入四氟硼酸锂(310mg)。将反应物移至80℃油浴锅中,在搅拌下过夜。加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化(DCM:MeOH=10:1),得到2-(3-(4-(6-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(11)(70mg,白色固体),收率:53%。MS m/z:406[M+1]+。1H NMR(400MHz,DMSO-d6)δ13.04(s,1H),8.93(s,1H),8.72(s,1H),8.47(s,1H),7.22(s,1H),4.60(d,J=9.1Hz,2H),4.24(d,J=9.1Hz,2H),3.69(s,2H),3.24(q,J=7.3Hz,2H),1.25(d,J=7.4Hz,3H).
实施例12:2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)丙腈(12)
第一步:3-(1-氰基亚乙基)氮杂环丁烷-1-甲酸叔丁酯(12b)
在冰浴条件下,向(1-氰乙基)膦酸二乙酯(12a)(220mg,1.15mmol)在四氢呋喃(10mL)中的溶液加入氢化钠(70mg,60%),搅拌40min后,反应物变为粉红色,向反应物中加入3-氧代氮杂环丁烷-1-甲酸叔丁酯(394mg,2.30mmol),反应物缓慢变澄清。将反应物缓慢升温至室温,在搅拌下过夜。
用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板分离纯化,最终得到3-(1-氰基亚乙基)氮杂环丁烷-1-甲酸叔丁酯(12b)(180mg,油状产物),收率:75%。MS(ESI,m/z):153[M+H]+。
第二步:2-(氮杂环丁烷-3-亚基)丙腈盐酸盐(12c)
在室温下,向3-(1-氰基亚乙基)氮杂环丁烷-1-甲酸叔丁酯(12b)(180mg,0.87mmol)中加入4M盐酸的二氧六环溶液(5mL),使反应物在搅拌下溶解。在室温下搅拌1h,LC-MS检测,底物完全消失。停止搅拌,将反应体系中的溶剂旋转蒸发,用乙醚打浆,过滤得到2-(氮杂环丁烷-3-亚基)丙腈盐酸盐(12c)(115mg,固体),收率:92%。MS(ESI,m/z):109[M+H]+。
第三步:2-(1-(乙基磺酰基)氮杂环丁烷-3-亚基)丙腈(12d)
在冰浴环境中,将2-(氮杂环丁烷-3-亚基)丙腈盐酸盐(12c)(108mg,0.75mmol)在乙腈(10mL)中溶解,然后加入DIEA(340mg),在冰浴下搅拌15min。向反应物中缓慢加入乙基磺酰氯(145mg,1.12mmol),保持反应物温度不高于5℃。在乙基磺酰氯加入完成后,缓慢升至室温,将反应物在搅拌下过夜。LC-MS检测,底物基本消失。通过制备板纯化(PE:EA=2:1),最终获得2-(1-(乙基磺酰基)氮杂环丁烷-3-亚基)丙腈(12d)(92mg,油状产物),收率:61%。MS(ESI,m/z):201[M+H]+。
第四步:4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(12f)
在冰盐浴冷却下,向4-氯吡咯并嘧啶(12e)(600mg,3.9mmol)在DMF(4mL)中的溶液分两批加入氢化钠(203mg,60%),保持反应物温度不高于10℃,在氮气保护下搅拌1h。用注射器缓慢加入SEMCl(846mg,5.07mmol),保持反应物温度不高于10℃。将反应物温热至室温,并搅拌过夜。用水淬灭反应液,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过快速制备色谱法(PE:EA=19:1)纯化,得到4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(12f)(800mg,油状产物),收率:72%。MS(ESI,m/z):284[M+H]+。
第五步:4-(1H-吡唑-4-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(12g)
在室温下,将4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并
[2,3-d]嘧啶(12f)(800mg,2.82mmol)和4-吡唑硼酸频哪醇酯(842mg,4.34mmol)在二氧六环(10mL)中溶解,然后加入水(2mL)和碳酸钾(857mg,6.2mmol),用氮气保护,在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(227mg,0.31mmol)。将反应物移至95℃油浴锅中,在搅拌下反应过夜。TLC检测,底物基本消失。加入水淬灭,用EA萃取,用无水硫酸钠干燥有机相。通过快速制备色谱法(PE:EA=2:3)纯化,得到4-(1H-吡唑-4-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(12g)(240mg,棕色固体),收率:27%。MS(ESI,m/z):316[M+H]+。
第六步:2-(1-(乙基磺酰基)-3-(4-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)丙腈(12h)
在室温下,向4-(1H-吡唑-4-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(12g)(159mg,0.51mmol)和2-(1-(乙基磺酰基)氮杂环丁烷-3-亚基)丙腈(12d)(102mg,0.51mmol)的混合物中加入乙腈(10mL),得到浑浊的反应液。然后加入DBU(100mg),在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化,得到2-(1-(乙基磺酰基)-3-(4-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)丙腈(12h)(160mg,棕色产物),收率:67.5%。MS(ESI,m/z):516[M+H]+。
第七步:2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)丙腈(12)
室温下,将2-(1-(乙基磺酰基)-3-(4-(7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)丙腈(12h)(160mg,0.31mmol)在乙腈(9mL)中溶解,加入水(1mL),最后加入四氟硼酸锂(583mg,6.21mmol),将反应物移至80℃油浴锅中,搅拌过夜。加入水淬灭,用EA萃取,无水硫酸钠干燥,浓缩有机相,通过制备硅胶板纯化,得到2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)丙腈(12)(80mg,白色固体),收率:66%。MS(ESI,m/z):386[M+H]+。1H NMR(DMSO-d6,400MHz)δ12.16(s,1H),8.94(s,1H),8.71(s,1H),8.47(s,1H),7.62-7.64(m,1H),7.08-7.09(m,1H),4.56-4.62(m,2H),4.33-4.37(m,2H),3.92-3.97(m,1H),3.19-3.25(m,2H),1.19-1.25(m,3H),
1.17-1.18(m,3H).
将合成的化合物(12)通过手性分离获得两个异构体(保留时间分别为6.9分钟(化合物12-1)和8.4分钟(化合物12-2)),其结构分别为
(R)-2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)丙腈(12-1)MS(ESI,m/z):386[M+H]+。1H NMR(DMSO-d6,400MHz)δ12.16(s,1H),8.94(s,1H),8.71(s,1H),8.47(s,1H),7.62-7.64(m,1H),7.08-7.09(m,1H),4.56-4.62(m,2H),4.33-4.37(m,2H),3.92-3.97(m,1H),3.19-3.25(m,2H),1.19-1.25(m,3H),1.17-1.18(m,3H);和
(S)-2-(3-(4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)丙腈(12-2)MS(ESI,m/z):386[M+H]+。1H NMR(DMSO-d6,400MHz)δ12.16(s,1H),8.94(s,1H),8.71(s,1H),8.47(s,1H),7.62-7.64(m,1H),7.08-7.09(m,1H),4.56-4.62(m,2H),4.33-4.37(m,2H),3.92-3.97(m,1H),3.19-3.25(m,2H),1.19-1.25(m,3H),1.17-1.18(m,3H).
实施例13:2-(1-(乙基磺酰基)-3-(3-甲基-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(13)
根据以上合成路线,采用与实施例8类似的操作,制备得到化合物(13)(100mg,油状产物)。收率30%。MS m/z:385[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.72(s,1H),8.46(s,1H),8.23(d,J=4.9Hz,1H),7.50(t,J=2.8,1H),7.10(d,J=4.9Hz,1H),6.64(dd,J=3.2,1.6Hz,1H),4.56(d,J=9.0Hz,2H),4.20(d,J=9.0Hz,2H),3.63(s,2H),3.24(q,J=7.3Hz,2H),2.39(s,3H),1.25(t,J=7.3Hz,3H).
实施例14:3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(14)
第一步:4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(14b)
将4-溴-7-氮杂吲哚(14a)(10g,50.7mmol)和DMF(100mL)加入到250mL三颈瓶中,用冰盐浴将反应物温度降至-10℃以下,用N2保护,将反应液搅拌均匀后,在1h内向反应液中分批加入氢化钠(60%,2.64g,54.4mmol),保持反应物温度不超过-5℃。搅拌反应1小时,然后向反应物中滴加2-(三甲基甲硅烷基)乙氧甲基氯,保持反应物温度不超过10℃,约1.5h滴毕,在搅拌下反应1小时,薄层色谱法监测反应,原料基本消失。将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(14b)(14.57g,收率:88.0%,黄色油状物)。MS(ESI,m/z):326.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡
咯并[2,3-b]吡啶(14c)
依次将化合物14b(4g,12.3mmol)、4-吡唑硼酸频哪醇酯(3.86g,19.9mmol)和二氧六环(300mL)加入到500mL反应瓶中,然后加入碳酸钾(4.58g,33.1mmol)溶液(60mL),将反应液搅拌均匀,然后加入Pd(dppf)Cl2(0.97g,1.33mmol),用N2保护,将反应物加热至95℃,在回流下反应过夜,薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(14c)(1.67g,收率:43.5%,黄色固体)。MS(ESI,m/z):314.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(14e)
将化合物14d(1.0g,5.15mmol)和4M盐酸的二氧六环溶液(10mL)加入到50mL反应瓶中,用Ar保护,在冰浴下搅拌反应物2.5h,逐渐析出白色固体。薄层色谱法监测反应,原料基本消失。将反应物抽滤,用无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(14e)(600mg,收率:90.0%,白色固体),将其直接用于下一步反应。
第四步:3-(氰基亚甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(14f)
在冰浴下,依次将化合物14e(300mg,2.31mmol)、二氯甲烷(15mL)、三乙胺(1.6mL,11.55mmol)和DMAP(5.7mg,0.05mmol)加入到50mL反应瓶中,搅拌均匀后,向反应物中缓慢滴加二甲氨基磺酰氯(431mg,3.00mmol)在二氯甲烷(15mL)中的溶液,加入完毕后将反应物搅拌1h。将反应物加水淬灭,用二氯甲烷萃取,然后分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到3-(氰基亚甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(14f)(311mg,收率:67.0%,褐色固体)。MS(ESI,m/z):201.1[M+H]+。
第五步:3-(氰甲基)-N,N-二甲基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-磺酰胺(14g)
将化合物14f(192mg,0.96mmol)、化合物14c(300mg,0.96mmol)和乙腈(22mL)加入到50mL反应瓶中,将反应液搅拌均匀,然后加入DBU(174mg,1.15mmol),将反应液在室温下搅拌1h。薄层色谱法监测反应结束后,将反应液用水淬灭,在减压下浓缩反应液,用硅胶柱色谱法纯化,
得到3-(氰甲基)-N,N-二甲基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-磺酰胺(14g)(387mg,收率:78.5%,黄色固体)。MS(ESI,m/z):515.2[M+H]+。
第六步:3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(14)
在室温下,将化合物14g(242mg,0.75mmol)与TFA/DCM(V:V=1:1.5)(14.4mL)的混合液加入到50mL反应瓶中,在Ar保护下,于室温下搅拌1h,LC-MS监测反应结束后,在冰浴下用浓氨水将反应物调节至碱性(pH=9-10),搅拌过夜。LC-MS监测反应结束后,将反应物用二氯甲烷萃取,无水硫酸钠干燥,在减压下浓缩,用TLC色谱法纯化,得到3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(14)(87mg,收率:15.7%,黄色固体)。1H NMR(400MHz,DMSO-d6)δ:11.72(s,1H),8.77(s,1H),8.34(s,1H),8.21(d,J=4.92Hz,1H),7.54(t,J=2.60Hz,1H),7.34(d,J=4.96Hz,1H),6.90(dd,J1=3.64Hz,J2=1.88Hz,1H),4.52(d,J=8.84Hz,2H),4.18(d,J=8.88Hz,2H),3.66(s,2H),2.79(s,6H).MS(ESI,m/z):385.1[M+H]+。
实施例15:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-丙酰基氮杂环丁烷-3-基)乙腈(15)
第一步:4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]
吡啶(15b)
将4-溴-7-氮杂吲哚(15a)(10g,50.7mmol)和DMF(100mL)加入到250mL三颈瓶中,用冰盐浴将反应物温度降至-10℃以下,用N2保护,将反应液搅拌均匀后,向反应物中分批加入氢化钠(60%,2.64g,54.4mmol),保持反应物温度不超过-5℃,1小时后加入完毕,搅拌反应1小时后,继续向反应物中滴加2-(三甲基甲硅烷基)乙氧甲基氯,保持温度不超过10℃,约1.5h滴加完毕。搅拌反应1小时,通过薄层色谱法监测反应,原料基本消失。将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(15b)(14.57g,收率:88.0%,黄色油状物)。MS(ESI,m/z):326.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(15c)
依次将化合物15b(4g,12.3mmol)、4-吡唑硼酸频哪醇酯(3.86g,19.9mmol)、二氧六环(300mL)加入到500mL反应瓶中,然后加入碳酸钾(4.58g,33.1mmol)溶液(60mL),将反应液搅拌均匀,然后加入Pd(dppf)Cl2(0.97g,1.33mmol),用N2保护,将反应物加热至95℃,在回流下反应过夜,通过薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(15c)(1.67g,收率:43.5%,黄色固体)。MS(ESI,m/z):314.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(15e)
将化合物15d(1.0g,5.15mmol)和4M盐酸的二氧六环溶液(10mL)加入到50mL反应瓶中,用Ar保护,在冰浴下搅拌反应2.5h,逐渐析出白色固体。通过薄层色谱法监测反应,原料基本消失后,将反应物抽滤,用无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(15e)(600mg,收率:90.0%,白色固体),将其直接用于下一步反应。
第四步:2-(1-丙酰基氮杂环丁烷-3-亚基)乙腈(15f)
在冰浴下,依次将化合物15e(160mg,1.23mmol)、二氯甲烷(8mL)、三乙胺(0.8mL,5.78mmol)和DMAP(2.5mg,0.02mmol)加入到50mL反应
瓶中,将其搅拌均匀后,向反应体系内缓慢滴加丙酰氯(134mg,1.45mmol)在二氯甲烷(8mL)中的溶液,加入完毕后搅拌1h,将反应液用水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到2-(1-丙酰基氮杂环丁烷-3-亚基)乙腈(15f)(116mg,收率:80.2%,黄色油状物)。MS(ESI,m/z):150.1[M+H]+。
第五步:2-(1-丙酰基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(15g)
将化合物15f(116mg,0.77mmol)、化合物15c(243mg,0.77mmol)和乙腈(18mL)加入到50mL反应瓶中,将反应液搅拌均匀,然后加入DBU(141mg,0.93mmol),将上述反应液在室温下搅拌1h,通过薄层色谱法监测反应结束后,将反应液用水淬灭,在减压下浓缩反应液,用硅胶柱色谱法纯化,得到2-(1-丙酰基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(15g)(230mg,收率:64.4%,黄色固体)。MS(ESI,m/z):464.2[M+H]+。
第六步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-丙酰基氮杂环丁烷-3-基)乙腈(15)
在室温下,将化合物15g(230mg,0.50mmol)与TFA/DCM(V:V=1:1.5)(15.0mL)的混合液加入到50mL反应瓶中,在Ar保护下,于室温下搅拌1h,LC-MS监测反应结束后,在冰浴下用浓氨水将反应物调节至碱性(pH=9-10),将反应物搅拌过夜,LC-MS监测反应结束后,用二氯甲烷萃取,无水硫酸钠干燥,在减压下浓缩,用TLC色谱法纯化,得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-丙酰基氮杂环丁烷-3-基)乙腈(15)(38mg,收率:23.0%,淡黄色固体)。1H NMR(400MHz,DMSO-d6)δ:11.71(s,1H),8.77(s,1H),8.32(s,1H),8.20(d,J=5.00Hz,1H),7.53(t,J=2.48Hz,1H),7.33(d,J=5.00Hz,1H),6.89(dd,J1=3.56Hz,J2=1.88Hz,1H),4.77(d,J=9.32Hz,1H),4.77(d,J=9.88Hz,2H),4.21(d,J=10.32Hz,1H),3.67(s,2H),2.15(q,2H),0.99(t,J=7.48Hz,3H).MS(ESI,m/z):334.1[M+H]+。
实施例16:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(环丙基羰基)氮杂环丁烷-3-基)乙腈(16)
第一步:4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(16b)
将4-溴-7-氮杂吲哚(16a)(10g,50.7mmol)和DMF(100mL)加入到250mL三颈瓶中,用冰盐浴将反应物温度降至-10℃以下,用N2保护,将反应液搅拌均匀后,向反应物中分批加入氢化钠(60%,2.64g,54.4mmol),保持反应物温度不超过-5℃,1小时加入完毕。搅拌反应1小时后,向反应物中滴加2-(三甲基甲硅烷基)乙氧甲基氯,保持反应物温度不超过10℃,约1.5h滴加完毕,搅拌反应1小时。通过薄层色谱法监测反应原料基本消失后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(16b)(14.57g,收率:88.0%,黄色油状物)。MS(ESI,m/z):326.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(16c)
将化合物16b(4g,12.3mmol)、4-吡唑硼酸频哪醇酯(3.86g,19.9mmol)和二氧六环(300mL)依次加入到500mL反应瓶中,然后加入碳酸钾(4.58g,33.1mmol)溶液(60mL),将反应液搅拌均匀,然后加入Pd(dppf)Cl2(0.97g,1.33mmol),用N2保护,加热至95℃,在回流下反应过夜。通过薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到化合物4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(16c)(1.67
g,收率:43.5%,黄色固体)。MS(ESI,m/z):314.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(16e)
将化合物16d(1.0g,5.15mmol)和4M盐酸的二氧六环溶液(10mL)加入到50mL反应瓶中,用Ar保护,在冰浴下搅拌反应物2.5h,逐渐析出白色固体。通过薄层色谱法监测反应原料基本消失后,将反应物抽滤,用无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(16e)(600mg,收率:90.0%,白色固体,将其直接用于下一步反应。
第四步:2-(1-(环丙基羰基)氮杂环丁烷-3-亚基)乙腈(16f)
在冰浴下,依次将化合物16e(160mg,1.23mmol)、二氯甲烷(8mL)、三乙胺(0.8mL,5.78mmol)和DMAP(2.5mg,0.02mmol)加入到50mL反应瓶中,搅拌均匀后,向反应物中缓慢滴加环丙甲酰氯(151mg,1.45mmol)在二氯甲烷(8mL)中的溶液,加入完毕后搅拌1h。将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到2-(1-(环丙基羰基)氮杂环丁烷-3-亚基)乙腈(16f)(130mg,收率:83.2%,类白色固体)。MS(ESI,m/z):162.1[M+H]+。
第五步:2-(1-(环丙基羰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(16g)
将化合物16f(130mg,0.80mmol)、化合物16c(250mg,0.80mmol)和乙腈(19mL)加入到50mL反应瓶中,将反应液搅拌均匀,然后加入DBU(183mg,1.20mmol),将所得的反应液在室温下搅拌1h。通过薄层色谱法监测反应结束后,将反应液用水淬灭,在减压下浓缩反应液,用硅胶柱色谱法纯化,得到2-(1-(环丙基羰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(16g)(243mg,收率:63.8%,黄色固体)。MS(ESI,m/z):476.2[M+H]+。
第六步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(环丙基羰基)氮杂环丁烷-3-基)乙腈(16)
在室温下,将化合物16g(243mg,0.51mmol)与TFA/DCM(V:V=1:1.5)(17.3mL)的混合液加入到50mL反应瓶中,用Ar保护,在室温下搅拌1h,LC-MS监测反应结束后,在冰浴下用浓氨水将反应物调节至碱性
(pH=9-10),搅拌过夜。LC-MS监测反应结束后,将反应物用二氯甲烷萃取,无水硫酸钠干燥,在减压下浓缩,用TLC色谱法纯化,得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(环丙基羰基)氮杂环丁烷-3-基)乙腈(16)(39mg,收率:22.2%,类白色固体)。1H NMR(400MHz,DMSO-d6)δ:11.71(s,1H),8.79(s,1H),8.34(s,1H),8.20(d,J=4.96Hz,1H),7.53(t,J=2.96Hz,1H),7.34(d,J=5.04Hz,1H),6.89(dd,J1=3.60Hz,J2=1.84Hz,1H),4.90(d,J=9.40Hz,1H),4.63(d,J=9.40Hz,1H),4.50(d,J=10.36Hz,1H),4.23(d,J=10.40Hz,1H),3.72(s,2H),1.63(q,1H),0.76(t,J=7.20Hz,4H).MS(ESI,m/z):346.2[M+H]+。
实施例17:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡咯-3-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17)
第一步:4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17c)
将4-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17a)(154mg,0.5mmol)、3-(4,4,5,5-四甲基-1,3-二氧杂环戊硼烷-2-基)-1-(三异丙基甲硅烷基)-1H-吡咯(17b)(262mg,0.75mmol)、PdCl2(dppf)(37mg,0.05mmol)和碳酸钾(173mg,1.25mmol)加入50mL的三颈瓶中,并加入二氧六环(5mL)及水(1mL),用氩气保护,将反应液加热至90℃,反应18h,TLC显示反应结束后,通过硅藻土抽滤,将滤液旋转蒸发至干燥,柱色谱法纯化得到4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17c)(142mg,白色固体,收率为84%)。
MS(ESI,m/z):339[M+H]+。
第二步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17e)
将4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17c)(142mg,0.42mmol)和2-(1-(乙基磺酰基)氮杂环丁烷-3-亚基)乙腈(17d)(85mg,0.46mmol)在乙腈(5mL)中溶解,然后加入DBU(0.12mL)。在室温下搅拌,TLC显示反应结束后,将反应物旋转蒸发至干燥,柱色谱法纯化得到4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17e)(200mg,乳白色油状物,收率为91%)。MS(ESI,m/z):525[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡咯-3-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17)
将4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17e)(110mg,0.21mmol)在二氯甲烷(2.6mL)中溶解,并将反应物置于冰浴下,然后加入三氟乙酸(2.6mL),在整个反应过程期间保持温度在10℃以下,TLC显示反应结束后,用碳酸钠水溶液调节pH为9,用乙酸乙酯萃取,旋转蒸发至干燥后得到灰白色固体,将其用95%的乙醇溶解,并向其中加入碳酸钠(334mg,3.15mmol),在室温搅拌,在反应结束后,将反应物旋转蒸发至干燥,加入水溶解其中的碳酸钠,抽滤得到固体。用正己烷洗涤固体,得到4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡咯-3-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(17)(53mg,白色固体,收率为64%)。1H NMR(400MHz,DMSO-d6)δ:12.27(s,1H),8.54(s,1H),7.74(s,1H),7.67(s,1H),7.28(s,1H),6.89(s,1H),6.85(s,1H),4.45(d,2H,J=8.4Hz),4.24(d,2H,J=8.8Hz),3.59(s,2H),3.23(q,2H,J=6.4Hz),1.24(t,3H,J=6.4Hz).MS(ESI,m/z):395[M+H]+。
实施例18:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)乙腈(18)
第一步:2-(氮杂环丁烷-3-亚基)乙腈三氟乙酸盐(18b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(18a)(583mg,3mmol)在二氯甲烷(18mL)及三氟乙酸(6mL)的混合液中溶解,在室温下搅拌30min,旋转蒸发至干燥得透明油状物,将其直接用于下一步反应。
第二步:2-(1-(异丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(18c)
将第一步所得的透明油状物在二氯甲烷(10mL)中溶解,并将反应物置于冰浴下,然后缓慢加入三乙胺,至反应物的pH达到9,然后加入DMAP(8mg,0.06mmol),将反应物在冰浴下搅拌5min。然后将异丙基磺酰氯(0.44mL,3.9mmol)在二氯甲烷(2mL)中溶解,并将其缓慢滴加入反应体系中,在冰浴下反应30min,然后用乙酸乙酯萃取反应液,将有机相用柠檬酸的水溶液洗涤,无水硫酸钠干燥有机相,然后旋转蒸发至干燥,以得到固体,将固体用石油醚打浆,得到2-(1-(异丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(18c)(450mg,白色固体,收率为75%),MS(ESI,m/z):201[M+H]+。
第三步:2-(1-(异丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(18e)
将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(18d)(200mg,0.64mmol)和2-(1-(异丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(18c)(192mg,0.96mmol)在乙腈(10mL)中溶解,并向反应物中加入DBU(0.16mL),将反应物在室温下搅拌过夜。TLC显示反应结束后,将反应物旋转蒸发至干燥,通过硅胶柱色谱法纯化得到2-(1-(异丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-
基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(18e)(310mg,乳白色油状物,收率为94%)。MS(ESI,m/z):515[M+H]+。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)乙腈(18)
将2-(1-(异丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(18e)(310mg,0.6mmol)在二氯甲烷(7mL)中溶解,并将反应物置于冰浴中,加入三氟乙酸(7mL),在整个反应过程中保持温度小于10℃。TLC显示反应结束后,用碳酸钠的水溶液调节反应物的pH为9,然后用乙酸乙酯萃取,旋转蒸发至干燥后,将固体在95%乙醇中溶解,并向反应物中加入碳酸钠(954mg,9mmol),将其在室温下搅拌过夜,旋转蒸发至干燥,通过硅胶柱色谱法纯化得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)乙腈(18)(190mg,淡黄色固体,收率为82%),1H NMR(400MHz,DMSO-d6)δ:11.77(s,1H),8.78(s,1H),8.35(s,1H),8.21(d,1H,J=5.2Hz),7.55(t,1H,J=3.2Hz),7.35(d,1H,J=4.8Hz),6.91(dd,1H,J=3.2Hz,J=2.0Hz),4.58(d,2H,J=8.8Hz),4.21(d,2H,J=8.8Hz),3.68(s,2H),3.38-3.31(m,1H),1.28(s,3H),1.26(s,3H).MS(ESI,m/z):385[M+H]+。
实施例19:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(甲基磺酰基)氮杂环丁烷-3-基)乙腈(19)
第一步:2-(氮杂环丁烷-3-亚基)乙腈三氟乙酸盐(19b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(19a)(583mg,3mmol)在二氯甲烷(18mL)及三氟乙酸(6mL)的混合液中溶解,在室温下搅拌30min,旋转蒸发至干燥得到透明油状物,将其直接用于下一步反应。
第二步:2-(1-(甲基磺酰基)氮杂环丁烷-3-亚基)乙腈(19c)
将第一步所得的透明油状物在二氯甲烷(10mL)中溶解,并将反应物置于冰浴中,然后缓慢加入三乙胺,至反应物的pH为9,然后加入DMAP(8mg,0.06mmol),将反应物在冰浴下搅拌5min,然后将甲基磺酰氯(0.3mL,3.9mmol)在二氯甲烷(2mL)中溶解,并将其缓慢滴加入反应物中。在冰浴下反应30min后,用乙酸乙酯萃取反应液,将有机相用柠檬酸的水溶液洗涤,无水硫酸钠干燥有机相,然后旋转蒸发至干燥得到固体,将固体用石油醚打浆,得到2-(1-(甲基磺酰基)氮杂环丁烷-3-亚基)乙腈(19c)(423mg,白色固体,收率为82%),MS(ESI,m/z):173[M+H]+。
第三步:2-(1-(甲基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(19e)
将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(19d)(252mg,0.8mmol)、2-(1-(甲基磺酰基)氮杂环丁烷-3-亚基)乙腈(19c)(207mg,1.2mmol)在乙腈(10mL)中溶解,并向反应物中加入DBU(0.16mL)。将反应物在室温下搅拌过夜,TLC显示反应结束后,将反应物旋转蒸发至干燥,硅胶柱色谱法纯化,得到2-(1-(甲基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(19e)(320mg,乳白色油状物,收率为82%)。MS(ESI,m/z):487[M+H]+。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(甲基磺酰基)氮杂环丁烷-3-基)乙腈(19)
将2-(1-(甲基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(19e)(150mg,0.3mmol)在二氯甲烷(3mL)中溶解,并将反应物置于冰浴中,然后加入三氟乙酸(3mL),在整个反应过程中保持温度小于10℃。TLC显示反应结束后,将反应物置于冰浴中,用氨水将反应物的pH调节至10,反应22h后,用乙酸乙酯萃取,旋转蒸发至干燥,硅胶制备板上纯化,得到2-(3-(4-(1H-
吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(甲基磺酰基)氮杂环丁烷-3-基)乙腈(19)(40mg,淡黄色固体,收率为36.6%)。1H NMR(400MHz,DMSO-d6)δ:11.72(s,1H),8.78(s,1H),8.35(s,1H),8.21(d,1H,J=4.8Hz),7.54(t,1H,J=3.2Hz),7.34(d,1H,J=5.2Hz),6.90(dd,1H,J=3.2Hz,J=2.0Hz),4.60(d,2H,J=9.2Hz),4.27(d,2H,J=9.6Hz),3.67(s,2H),3.14(s,3H).MS(ESI,m/z):357[M+H]+。
实施例20:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-甲腈(20)
第一步:2,4-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20b)
在室温下,将2,4-二氯-7H-吡咯并[2,3-d]嘧啶(20a)(500mg,2.66mmol)和DMF(5mL)加入到50mL三颈瓶中,用N2保护。将该混合物在冰盐浴中冷却至5℃以下,在反应液搅拌均匀后,向反应物中分批加入氢化钠(60wt%,138mg,3.46mmol),保持反应物温度始终不高于10℃。搅拌1小时后,向反应物中缓慢滴加2-(三甲基甲硅烷基)乙氧甲基氯(577mg,3.46mmol),保持反应物温度始终不高于5℃,继续搅拌2小时。通过薄层色谱法监测反应,在原料基本消失后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到2,4-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20b)(823mg,收率:97%,淡黄色液体)。MS(ESI,m/z):318[M+H]+。
第二步:2-氯-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲
基)-7H-吡咯并[2,3-d]嘧啶(20c)
在室温下,依次将2,4-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20b)(823mg,2.58mmol)、4-吡唑硼酸频哪醇酯(752mg,3.88mmol)、碳酸钾(891mg,6.45mmol)溶液(4mL)和二氧六环(24mL)加入到100mL反应瓶中,用N2保护。将反应液搅拌均匀,然后加入Pd(dppf)Cl2(189mg,0.26mmol),用N2保护。将反应物加热至95℃,在回流下反应过夜,通过薄层色谱法监测,在原料反应完后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到2-氯-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20c)(500mg,收率:55%,黄色固体)。MS(ESI,m/z):350[M+H]+。
第三步:2-氰基-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20d)
依次将2-氯-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20c)(320mg,0.91mmol)、氰化锌(53mg,0.46mmol)、Pd(dppf)Cl2(168mg,0.23mmol)和DMF(15mL)加入到50mL密封管中,用Ar气保护,将反应物加热至110℃,并反应24小时。通过薄层色谱法监测,在原料反应完后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到2-氰基-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20d)(219mg,收率:71%,淡黄色固体)。MS(ESI,m/z):341[M+H]+。
第四步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-甲腈(20e)
依次将2-氰基-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(20d)(219mg,0.64mmol)、2-[1-(乙基磺酰基)-3-氮杂环丁亚基]乙腈(131mg,0.71mmol)和乙腈(10mL)加入到50mL反应瓶中,然后加入DBU(108mg,0.76mmol),在室温下反应2小时。通过薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)
甲基)-7H-吡咯并[2,3-d]嘧啶-2-甲腈(20e)(280mg,收率:83%,白色固体)。MS(ESI,m/z):527[M+H]+。
第五步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-甲腈(20)
在室温下,将4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-甲腈(20e)(218mg,0.41mmol)与TFA/DCM(1:1)(8mL)的混合液加入到50mL反应瓶中,在Ar保护下反应2.5小时。在薄层色谱法监测反应结束后,在减压下浓缩,得黄色油状物。然后将所得的黄色油状物直接在四氢呋喃(10mL)中溶解,在搅拌均匀后,加入1M氢氧化钠溶液将反应物的pH调节至约10,反应0.5小时。在薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化得到目标产物4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-甲腈(20)(106mg,收率:65%,白色固体)。
1H NMR(400MHz,DMSO-d6)δ12.83(s,1H),9.04(s,1H),8.57(s,1H),7.98(d,J=3.5Hz,1H),7.30(d,J=3.5Hz,1H),4.61(d,J=9.2Hz,2H),4.25(d,J=9.1Hz,2H),3.70(s,2H),3.24(q,J=7.3Hz,2H),1.24(t,J=7.3Hz,3H)ppm.MS(ESI,m/z):397[M+H]+。
实施例21:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(苯基磺酰基)氮杂环丁烷-3-基)乙腈(21)
第一步:2-(1-(苯基磺酰基)氮杂环丁烷-3-亚基)乙腈(21b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(21a)(583mg,3.00mmol)和TFA/DCM(1/3)(8mL)加入到50mL反应瓶中,在室温下搅拌半小时。在薄层色谱法显示所有起始原料基本消失后,将反应混合物在减压下浓缩至干燥。然后,将所得粗产物在DCM(5mL)中溶解。在冰浴下,缓慢滴加三乙胺调节体系pH至约8,然后加入DMAP(7mg,0.06mmol),随后缓慢滴加苯磺酰氯(795mg,4.5mmol)。将所得反应混合物逐渐升至室温,并在室温下继续搅拌半小时。在LC-MS显示反应结束后,将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到2-(1-(苯基磺酰基)氮杂环丁烷-3-亚基)乙腈(21b)(650mg,收率:90%,褐色固体)。MS(ESI,m/z):235[M+H]+。
第二步:2-(1-(苯基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(21d)
将2-(1-(苯基磺酰基)氮杂环丁烷-3-亚基)乙腈(21b)(164mg,0.70mmol)、4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(21c)(200mg,0.64mmol)和乙腈(8mL)加入到50mL反应瓶中,然后加入DBU(106mg,0.70mmol),将上述反应液在室温下反应2小时。在薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到2-(1-(苯基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(21d)(317mg,收率:89%,白色固体)。MS(ESI,m/z):549[M+H]+。
第三步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(苯基磺酰基)氮杂环丁烷-3-基)乙腈(21)
在冰浴下,将2-(1-(苯基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(21d)(317mg,0.58mmol)与TFA/DCM(1:1)(8mL)的混合液加入到50mL反应瓶中,在Ar保护下反应2.5小时。在薄层色谱法监测反应结束后,在减压下浓缩,得黄色油状物。然后将所得黄色油状物直接在四氢呋喃(10mL)中溶解,在搅拌均匀后,加入1M氢氧化钠溶液,调节反应物pH至约10,反应2小时。在薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠
干燥,在减压下浓缩,通过薄层色谱法纯化得到目标产物2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(苯基磺酰基)氮杂环丁烷-3-基)乙腈(21)(58mg,收率:24%,白色固体)。1H NMR(400MHz,DMSO-d6)δ11.71(s,1H),8.48(s,1H),8.18(d,J=5.0Hz,1H),8.14(s,1H),7.89–7.84(m,2H),7.68–7.58(m,3H),7.53(dd,J=3.5,2.5Hz,1H),7.23(d,J=5.0Hz,1H),6.79(dd,J=3.5,1.9Hz,1H),4.37(d,J=9.6Hz,2H),4.21(d,J=9.6Hz,2H),3.52(s,2H)ppm.MS(ESI,m/z):419[M+H]+。
实施例22:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(苯甲酰基)氮杂环丁烷-3-基)乙腈(22)
第一步:2-(1-(苯甲酰基)氮杂环丁烷-3-亚基)乙腈(22b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(22a)(583mg,3.00mmol)和TFA/DCM(1/3)(8mL)加入到50mL反应瓶中,在室温下搅拌半小时,在薄层色谱法显示所有起始原料基本消失后,将反应混合物在减压下浓缩至干燥。然后,将所得的粗产物在DCM(5mL)中溶解。在冰浴下,缓慢滴加三乙胺,调节反应物pH至约8,然后加入DMAP(7mg,0.06mmol),随后缓慢滴加苯甲酰氯(633mg,4.5mmol)。将所得的反应混合物逐渐温热至室温,并在室温下继续搅拌半小时。在LC-MS显示反应结束后,将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到2-(1-(苯甲酰基)氮杂环丁烷-3-亚基)乙腈(22b)(560mg,收率:91%,褐色固体)。MS(ESI,m/z):199[M+H]+。
第二步:2-(1-(苯甲酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲
基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(22d)
将2-(1-(苯甲酰基)氮杂环丁烷-3-亚基)乙腈(22b)(139mg,0.70mmol)、4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(22c)(200mg,0.64mmol)和乙腈(8mL)加入到50mL反应瓶中,然后加入DBU(106mg,0.70mmol),将所得的反应液在室温下反应2小时。在薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过硅胶柱色谱法纯化,得到2-(1-(苯甲酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(22d)(311mg,收率:93%,白色固体)。MS(ESI,m/z):513[M+H]+。
第三步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(苯甲酰基)氮杂环丁烷-3-基)乙腈(22)
在冰浴下,将2-(1-(苯甲酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(22d)(311mg,0.58mmol)与TFA/DCM(1:1)(8mL)的混合液加入到50mL反应瓶中,在Ar保护下反应2.5小时。在薄层色谱法监测反应结束后,在减压下浓缩,得到黄色油状物。然后将所得的黄色油状物直接在四氢呋喃(10mL)中溶解,在搅拌均匀后,加入1M氢氧化钠溶液,调节反应物的pH至约10,反应2小时。在薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,通过薄层色谱法纯化得到目标产物2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(苯甲酰基)氮杂环丁烷-3-基)乙腈(22)(104mg,收率:47%,白色固体)。1H NMR(400MHz,DMSO-d6)δ11.72(s,1H),8.82(s,1H),8.34(s,1H),8.20(d,J=5.0Hz,1H),7.77–7.66(m,2H),7.65–7.47(m,4H),7.34(d,J=5.0Hz,1H),6.91(dd,J=3.5,1.8Hz,1H),5.04(d,J=9.7Hz,1H),4.67(dd,J=10.5,4.8Hz,2H),4.45(d,J=10.9Hz,1H),3.71(s,2H)ppm.MS(ESI,m/z):383[M+H]+。
实施例23:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(丙基磺酰基)氮杂环丁烷-3-基)乙腈(23)
第一步:2-(1-(丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(23b)
向3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(23a)(600mg)中加入TFA/DCM(1/2,8mL),将其在室温下搅拌半小时。在TLC监测起始原料基本消失后,将反应混合物在减压下浓缩至干燥。然后向粗产物加入DCM(10mL)中。在冰浴下,缓慢滴加三乙胺,调节反应物的pH至8,然后加入DMAP(29mg),随后缓慢滴加丙磺酰氯(1.02g)。将所得反应混合物逐渐温热至室温,并在室温下继续搅拌半小时。在LC-MS显示反应结束后,将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到粗品2-(1-(丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(23b)(930mg,棕色固体)。MS m/z:201[M+1]+。
第二步:2-(1-(丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(23c)
在室温下,向4-(1H-吡唑-4-基)-1-((2-((三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(200mg)和2-(1-(丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(23b)(153mg)中加入乙腈(10mL),得到浑浊的反应液。然后加入DBU(194mg),在室温下搅拌过夜。将反应液浓缩,通过制备硅胶板纯化(DCM:EA=1:3),得到2-(1-(丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(23c)(328mg,白色固体)。收率98%。MS m/z:515[M+1]+。
第三步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(丙基磺酰基)氮杂环丁烷-3-基)乙腈(23)
在冰浴下,向2-(1-(丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(23c)(312mg)中加入TFA/DCM(1:2,12mL),反应2.5小时。在TLC监测反应结束后,将反应物在减压下浓缩,得黄色油状物。在冰浴下,将所得黄色油状物直接在95%乙醇(20mL)中溶解,然后加入适量的碳酸钠,使pH=8,
反应2小时,有大量的固体析出。在TLC监测反应结束后,加入水和乙酸乙酯,在固体完全溶解后,用EA萃取,无水硫酸钠干燥,在减压下浓缩,用制备硅胶板纯化(DCM:EA=1:5),得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(丙基磺酰基)氮杂环丁烷-3-基)乙腈(23)(90mg,白色固体),收率:39%。MS m/z:385[M+1]+。1H NMR(DMSO-d6,400MHz)δ11.72(s,1H),8.78(s,1H),8.35(s,1H),8.20(d,J=4.0Hz,1H),7.53-7.55(m,1H),7.33(d,J=4.0Hz,1H),6.89-6.91(m,1H),4.58(d,J=8.0Hz,2H),4.24(d,J=4.0Hz,2H),3.67(s,2H),3.22-3.25(m,2H),1.68-1.78(m,2H),1.00(t,J=8.0Hz,3H).
实施例24:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(丁基磺酰基)氮杂环丁烷-3-基)乙腈(24)
第一步:2-(1-(丁基磺酰基)氮杂环丁烷-3-亚基)乙腈(24b)
向3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(24a)(600mg)中加入TFA/DCM(1/2,8mL),在室温下搅拌半小时。在TLC监测起始原料基本消失后,将该反应混合物在减压下浓缩至干燥。然后向粗产物加入DCM(10mL)。在冰浴下,缓慢滴加三乙胺,调节反应物pH=8,然后加入DMAP(29mg),随后缓慢滴加丁磺酰氯(1.02g)。将所得的反应混合物逐渐温热至室温,并在室温下继续搅拌半小时。在LC-MS显示反应结束后,将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,在减压下浓缩得到粗品2-(1-(丁基磺酰基)氮杂环丁烷-3-亚基)乙腈(24b)(930mg,棕色固体)。MS m/z:215[M+1]+。
第二步:2-(1-(丁基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(24c)
在室温下,向4-(1H-吡唑-4-基)-1-((2-((三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(200mg)和2-(1-(丁基磺酰基)氮杂环丁烷-3-亚基)乙腈(24b)(300mg)中加入乙腈(10mL),得到浑浊的反应液。然后加入DBU
(194mg),在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化(DCM:EA=1:4),得到2-(1-(丁基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(24c)(329mg,白色固体)。收率98%。MS m/z:529[M+1]+。
第三步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(丁基磺酰基)氮杂环丁烷-3-基)乙腈(24)
在冰浴下,向2-(1-(丁基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(24c)(328mg)中加入TFA/DCM(1:2,12mL),反应2.5小时。在TLC监测反应结束后,在减压下浓缩,得到黄色油状物。在冰浴下,将所得的黄色油状物直接在95%乙醇(20mL)中溶解,然后加入适量的碳酸钠,使pH=8,反应2小时,有大量的固体析出。在TLC监测反应结束后,加入水和乙酸乙酯,在固体完全溶解后,用EA萃取,无水硫酸钠干燥,在减压下浓缩,用制备硅胶板纯化(DCM:EA=1:5),得到2-(3-((4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(丁基磺酰基)氮杂环丁烷-3-基)乙腈(24)(100mg,白色固体),收率:40%。MS m/z:399[M+1]+。1H NMR(DMSO-d6,400MHz)δ11.72(s,1H),8.78(s,1H),8.34(s,1H),8.20(d,J=4.0Hz,1H),7.53-7.54(m,1H),7.33(d,J=4.0Hz,1H),6.89-6.90(m,1H),4.58(d,J=12.0Hz,2H),4.24(d,J=12.0Hz,2H),3.66(s,2H),3.22-3.26(m,2H),1.63-1.71(m,2H),1.35-1.45(m,2H),0.89(t,J=8.0Hz,3H).
实施例25:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丁基磺酰基)氮杂环丁烷-3-基)乙腈(25)
第一步:2-(1-(异丁基磺酰基)氮杂环丁烷-3-亚基)乙腈(25b)
在室温下,向3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(25a)(583mg,3.00mmol)加入TFA/DCM(1:2,8mL),将反应液搅拌半小时,监测反应结束后,将该反应混合物在减压下浓缩得粗产物。然后,将粗产物溶于DCM(10mL)中。在冰浴下,滴加三乙胺调节体系pH=8后,再依次加入DMAP
(7mg,0.06mmol)和异丁磺酰氯(633mg,4.5mmol)。将反应液在室温下搅拌半小时。检测反应结束后加水淬灭,二氯甲烷萃取,收集有机相,将其分别用水、柠檬酸、饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品2-(1-(异丁基磺酰基)氮杂环丁烷-3-亚基)乙腈(25b)(600mg,白色固体),收率:93%。MS m/z:215[M+1]+。
第二步:2-(1-(异丁基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(25c)
在室温下,向4-(1H-吡唑-4-基)-1-((2-((三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(150mg,0.48mmol)和2-(1-(异丁基磺酰基)氮杂环丁烷-3-亚基)乙腈(25b)(113mg,0.53mmol)的体系中加入乙腈(10mL)和DBU(80mg,0.53mmol)。将反应液在室温下搅拌过夜。检测反应结束后,将反应液浓缩,浓缩残余物经柱色谱法纯化得到2-(1-(异丁基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(25c)(242mg,白色固体)。收率98%。MS m/z:529[M+1]+。
第三步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丁基磺酰基)氮杂环丁烷-3-基)乙腈(25)
在冰浴下,向2-(1-(异丁基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(25c)(242mg,0.46mmol)中加入TFA/DCM(1:2,12mL),反应2.5小时。监测反应结束后,减压浓缩,得黄色油状物粗产品。在冰浴下,将所得粗产品溶于95%乙醇(20mL)中,再加入适量的碳酸钠,使pH=8,将反应液继续搅拌2小时。监测反应结束后,加入水和乙酸乙酯,EA萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,浓缩残余物,经柱色谱法纯化得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丁基磺酰基)氮杂环丁烷-3-基)乙腈(25)(90mg,白色固体),收率:49%。MS m/z:399[M+1]+。1H NMR(400MHz,DMSO)δ11.72(s,1H),8.78(s,1H),8.35(s,1H),8.21(d,J=5.0Hz,1H),7.56-7.53(m,1H),7.34(d,J=5.0Hz,1H),6.90(dd,J=3.5,1.8Hz,1H),4.58(d,J=9.2Hz,2H),4.25(d,J=9.2Hz,2H),3.66(s,2H),3.16(d,J=6.6Hz,2H),2.20-2.10(m,1H),1.04(d,J=6.7Hz,6H).
实施例26:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(环丙基磺酰基)氮杂环丁烷-3-基)乙腈(26)
第一步:2-(氮杂环丁烷-3-亚基)乙腈三氟乙酸盐(26b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(26a)(583mg,3mmol)溶于18mL二氯甲烷及6mL三氟乙酸的混合液中,在室温下搅拌30min,旋干得透明油状物,将其直接用于下一步。
第二步:2-(1-(环丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(26c)
在冰浴下,将第一步所得的透明油状物溶于10mL的二氯甲烷中,加入三乙胺,至体系pH为9,再加入DMAP(8mg,0.06mmol)和环丙基磺酰氯(0.44mL,3.9mmol)的2mL二氯甲烷溶液,将反应液搅拌30min。检测反应结束后,用乙酸乙酯萃取反应液,将有机相用柠檬酸的水溶液洗涤,无水硫酸钠干燥,过滤,浓缩得到2-(1-(环丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(26c)(450mg,白色固体,收率为79%),MS(ESI,m/z):199[M+H]+。
第三步:2-(1-(环丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(26e)
将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(26d)(200mg,0.64mmol)、2-(1-(环丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(26c)(192mg,0.96mmol)溶于10mL乙腈中,加入0.16mL DBU,将反应液在室温下搅拌过夜。检测反应结束后,将反应液浓缩,浓缩残余物,经柱色谱法纯化得到2-(1-(环丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(26e)(293mg,乳白色油状物,收率为90%)。MS(ESI,m/z):513[M+H]+。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(环丙基磺酰基)氮杂环丁烷-3-基)乙腈(26)
在冰浴下,将2-(1-(环丙基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(26e)(293mg,0.57mmol)溶于6mL的二氯甲烷中,加入6mL的三氟乙酸,将反应液搅拌1小时。检测反应结束后,加入碳酸钠的水溶液调节体系pH为9,再用乙酸乙酯萃取,收集有机相干燥,过滤,浓缩得固体。用95%乙醇溶解固体,再加入碳酸钠(907mg,8.55mmol)。将反应液在室温下搅拌过夜后,柱色谱法纯化得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(环丙基磺酰基)氮杂环丁烷-3-基)乙腈(26)(133mg,白色固体,收率为61%)。1H NMR(400MHz,DMSO-d6)δ:11.72(s,1H),8.79(s,1H),8.34(s,1H),8.20(d,1H,J=4.8Hz),7.54(d,1H,J=3.6Hz),7.34(d,1H,J=5.2Hz),6.89(d,1H,J=3.6Hz),4.64(d,2H,J=9.2Hz),4.28(d,2H,J=9.2Hz),3.67(s,2H),2.88-2.81(m,1H),1.07-1.02(m,2H),1.01-0.97(m,2H).MS(ESI,m/z):383[M+H]+。
实施例27:4-(1-(3-(氰甲基)-1-(苯基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(27)
第一步:2-(1-(苯基磺酰基)氮杂环丁烷-3-亚基)乙腈(27b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(27a)(583mg,3.00mmol)和TFA/DCM(1:3,8mL)加入到反应瓶中,在室温下搅拌半小时,检测反应结束后,将该反应混合物在减压下浓缩得粗产物。然后,将所得粗产物
溶于DCM(10mL)中。在冰浴下,缓慢滴加三乙胺,调节体系pH值约等于8后,再依次加入DMAP(7mg,0.06mmol)和苯磺酰氯(795mg,4.5mmol)。将反应在室温下搅拌半小时。检测反应结束后加水淬灭,二氯甲烷萃取,收集有机相,将其分别用水、柠檬酸、饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到2-(1-(苯基磺酰基)氮杂环丁烷-3-亚基)乙腈(27b)(650mg,收率:90%,褐色固体)。MS(ESI,m/z):235[M+H]+。
第二步:4-(1-(3-(氰甲基)-1-(苯基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(27d)
在室温下,将2-(1-(苯基磺酰基)氮杂环丁烷-3-亚基)乙腈(27b)(114mg,0.48mmol)、4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(27c)(150mg,0.44mmol)和乙腈8mL加入到反应瓶中,然后加入DBU(73mg,0.48mmol),将反应液搅拌2小时。监测反应结束后加水淬灭,用乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,浓缩残余物经柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-(苯基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(27d)(223mg,收率:89%,白色固体)。MS(ESI,m/z):574[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-(苯基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(27)
在冰浴下,将2-(1-(苯基磺酰基)-3-(5-氰基-4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(27d)(223mg,0.39mmol)与TFA/DCM(1:1,6mL)的混合物加入到反应瓶中,在氩气保护下搅拌2.5小时。监测反应结束后,减压浓缩,得黄色油状物粗产品。然后将所得粗产品溶于四氢呋喃(10mL)中,待搅拌均匀后,加入1M氢氧化钠溶液调节体系pH约等于10,将反应液搅拌0.5小时。监测反应结束后,加入乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,浓缩残余物经柱色谱法纯化得到目标产物4-(1-(3-(氰甲基)-1-(苯基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(27)(100mg,收率:58%,白色固体)。1H NMR(400MHz,DMSO)δ12.40(s,1H),8.61(s,1H),8.53(s,1H),8.10(s,1H),7.89-7.82(m,2H),7.76(d,
J=3.5Hz,1H),7.70-7.56(m,3H),6.75(d,J=3.5Hz,1H),4.40(d,J=9.8Hz,2H),4.25(d,J=9.8Hz,2H),3.54(s,2H).MS(ESI,m/z):444[M+H]+。
实施例28:2-(3-(3-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(28)
第一步:4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(28c)
将4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(28a)(326mg,1mmol)、3-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1-(三异丙基硅烷基)-1H-吡咯(28b)(524mg,1.5mmol)、PdCl2(dppf)(74mg,0.1mmol)和碳酸钾(346mg,2.5mmol)置于反应瓶中,加入10mL二氧六环及2mL水,氩气保护后,将反应液加热至90℃,搅拌22h。检测反应结束后,将反应液抽滤,滤液浓缩,柱色谱法纯化分别得到4-(1-(三异丙基硅烷基)-1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶及4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(28c),将4-(1-(三异丙基硅烷基)-1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶在1M的四丁基氟化铵的四氢呋喃溶液中脱硅保护基得到4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(28c),总共得到283mg,白色固体,收率为90%,MS(ESI,m/z):314[M+H]+。
第二步:2-(1-(乙基磺酰基)-3-(3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-1-基)氮杂环丁烷-3-基)乙腈(28e)
将4-(1H-吡咯-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(28c)(283mg,0.9mmol)、2-(1-(乙基磺酰基)氮杂环丁烷-3-亚基)乙腈(28d)(183mg,0.99mmol)溶于10mL乙腈中,再加入0.3mL DBU,在室温下搅拌过夜,将反应液浓缩,柱色谱法纯化得到2-(1-(乙基磺酰基)-3-(3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-1-基)氮杂环丁烷-3-基)乙腈(28e)(240mg,乳白色油状物,收率为53%)。MS(ESI,m/z):500[M+H]+。
第三步:2-(3-(3-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(28)
在冰浴下,将2-(1-(乙基磺酰基)-3-(3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-1-基)氮杂环丁烷-3-基)乙腈(28e)(240mg,0.48mmol)溶于5mL的二氯甲烷中,加入5mL的三氟乙酸,将反应液搅拌1小时,检测反应结束后,加入Na2CO3水溶液调节pH为9,乙酸乙酯萃取,收集有机相旋干得浅黄色固体,再用95%的乙醇溶解固体,加入碳酸钠(763mg,7.2mmol),室温搅拌,待反应结束后,浓缩,柱色谱法纯化得到2-(3-(3-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(28)(83mg,浅黄色固体,收率为46%。)。1H NMR(400MHz,DMSO-d6)δ:11.56(s,1H),8.13(d,1H,J=5.2Hz),7.70(s,1H),7.45(t,1H,J=2.8Hz),7.20(d,1H,J=4.8Hz),7.16(t,1H,J=2.4Hz),6.84-6.83(m,1H),6.78-6.77(m,1H),4.44(d,2H,J=9.2Hz),4.20(d,2H,J=8.8Hz),3.56(s,2H),3.22(q,2H,J=7.2Hz),1.23(t,3H,J=7.2Hz).MS(ESI,m/z):370[M+H]+。
实施例29:4-(1-(1-(乙基磺酰基)-3-(氟甲基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(29)
第一步:氟甲基苯基硫醚(29b)
在冰水浴下,向反应瓶中依次加入DCM(50mL)、三氯化锑(0.9g)、苯甲硫醚(29a)(5.0mL)和DAST(13.0g),将反应液搅拌6小时。检测反应结束后浓缩反应液,将浓缩残余物经快速柱色谱法纯化得到黄色油状物4.6g,纯度94%,收率80%。MS(ESI,m/z):143[M+H]+。
第二步:氟甲基苯基砜(29c)
在冰水浴下,将第一步所得的氟甲基苯基硫醚(29b)(4.0g)溶于DCM(160mL)中,分批加入间氯过氧苯甲酸(16.5g),将反应液搅拌过夜。检测反应结束后加入10%碳酸钾溶液(100mL)和DCM(100mL),通过硅藻土过滤,DCM萃取,合并有机相,用饱和氯化钠洗涤,无水硫酸钠干燥,过滤,浓缩,将浓缩残余物经柱色谱法纯化,得无色油状物3.3g,纯度91%,收率67%。MS(ESI,m/z):175[M+H]+。
第三步:3-(氟(苯磺酰基)亚甲基)氮杂环丁烷-1-甲酸叔丁酯(29d)
在氮气保护下,向反应瓶中加入THF(10mL)、氟甲基苯基砜(29c)(0.5g)、氯磷酸二乙酯(0.5g)。体系降温至-78℃,滴加LHMDS(6.2mL),维持体系温度-78℃以下反应1小时。用THF(2mL)溶解3-氧代氮杂环丁烷-1-甲酸叔丁酯(0.378g),将其滴加至反应体系,将反应液升温至室温,反应2小时。检测反应结束后将反应液倒入饱和氯化铵溶液(50mL)中,EA萃取,合并有机相,用饱和食盐水洗涤,硫酸钠干燥,过滤,浓缩滤液,将浓缩残余物经柱色谱法纯化,得到3-(氟(苯磺酰基)亚甲基)氮杂环丁烷-1-甲酸叔丁酯(29d),无色油状物565mg,纯度91.6%。MS(ESI,m/z):328[M+H]+。
第四步:3-(4-(5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)3-(氟(苯磺酰基)甲基)氮杂环丁烷-1-甲酸叔丁酯(29f)
向反应瓶中加入乙腈(6mL)、3-(氟(苯磺酰基)亚甲基)氮杂环丁烷-1-甲酸叔丁酯(29d)(550mg)、4-(1H-吡唑-4-基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(29e)(439mg)和DBU(196mg),搅拌反应2.5小时,检测反应结束后浓缩反应液,浓缩残余物经柱色谱法纯化,得到标题产品(类白色固体730mg,纯度95%,收率85%)。MS(ESI,m/z):667[M+H]+。
第五步:3-(4-(5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氟甲基)氮杂环丁烷-1-甲酸叔丁酯(29g)
在氮气保护下,向反应瓶中加入3-(4-(5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氟(苯磺酰基)甲基)氮杂环丁烷-1-甲酸叔丁酯(29f)(500mg)、甲醇(12.5mL)和无水磷酸氢二钠(2.13g),将体系降温至-20℃,加入钠汞齐(3.35g),维持体系反应温度-20℃~0℃,搅拌1.5小时,检测反应结束后向反应体系加入EA(100mL),通过硅藻土过滤,将滤液倒入饱和氯化铵溶液(150mL)中,分离有机相,用饱和食盐水洗涤,硫酸钠干燥,过滤,浓缩滤液,TLC制备板纯化,得到标题产品(29g,61mg,黄色固体,收率15%)。MS(ESI,m/z):527[M+H]+。
第六步:4-(1-(3-(氟甲基)氮杂环丁烷-3-基)1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈盐酸盐(29h)
向反应瓶中加入3-(4-(5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氟甲基)氮杂环丁烷-1-甲酸叔丁酯(29g)(20mg)、1,4-二氧六环(2mL)和1,4-二氧六环/氯化氢(1mL),将反应液室温搅拌1小时后,浓缩反应液除去溶剂得粗品,将其直接用于下一步。MS(ESI,m/z):427[M+H]+。
第七步:4-(1-(3-(氟甲基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(29i)
向反应瓶中加入第六步所得的粗品、DCM(4mL)和TFA(1.5mL),使反应液在搅拌下反应1.5小时,检测反应结束后浓缩反应液,向残渣加入氨水,调节pH为9~10,并搅拌2小时,再加入2N HCl调节pH为1~2,用DCM萃取杂质,向水相加入碳酸钾溶液调节pH为9~10,用DCM萃取,合并有机相,硫酸钠干燥,过滤,浓缩滤液,得标题产品(29i,6mg),将其直接用于下一步。MS(ESI,m/z):297[M+H]+。
第八步:4-(1-(1-(乙基磺酰基)-3-(氟甲基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(29)
在室温下,向反应瓶中加入THF(3mL)、29i(6mg)、DIPEA(13mg)和乙基磺酰氯(9mg),将反应液搅拌1小时,检测反应结束后浓缩反应液,TLC制备板纯化得黄色标题化合物(固体,7mg)
1H NMR(400MHz,CDCl3)δ:9.69(s,1H),8.58(s,1H),8.30(s,1H),8.23(s,1H),7.48(s,1H),6.76(s,1H),5.06(s,1H),4.94(s,1H),4.64(d,J=9.76Hz,2H),4.28(d,J=9.00Hz,2H),3.08-3.14(m,2H),1.41-1.45(t,J=7.08Hz,3H)。MS(ESI,m/z):389[M+H]+。
实施例30:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(3,3,3-三氟丙基磺酰基)氮杂环丁烷-3-基)乙腈(30)
第一步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(30b)
在冰水浴下,将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(2.0g)和1,4-二氧六环/HCl(20mL)加入到反应瓶中,将反应液温热至室温,并搅拌2小时,检测反应结束后,过滤反应液,滤饼用甲基叔丁基醚洗涤,干燥得白色固体2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(30b)(1.11g,收率82.4%)。MS(ESI,m/z):95[M+H]+。
第二步:2-(1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(30c)
在冰水浴下,将第一步所得的白色固体30b(500mg)加入到二氯甲烷(20mL)中,加入(3,3,3-三氟丙基)磺酰氯(1.126g)和TEA(1.16g)的15mL DCM溶液。使反应液在搅拌下反应5小时,检测反应结束后加入水(10mL),搅拌10分钟,分出有机相浓缩,将残渣直接用于下一步反应。MS(ESI,m/z):255[M+H]+。
第三步:2-(1-(3,3,3-三氟丙基)磺酰基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(30e)
将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(30d)(600mg)、2-(1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-亚基)
乙腈(30c)(485mg,粗品)溶于乙腈(50mL)中,加入DBU(1mL),在室温下搅拌过夜,检测反应结束后,将反应液浓缩,浓缩残余物经柱色谱法纯化得到2-(1-(3,3,3-三氟丙基)磺酰基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(30e),黄色固体165mg。MS(ESI,m/z):569[M+H]+。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(3,3,3-三氟丙基磺酰基)氮杂环丁烷-3-基)乙腈(30)
在冰浴下,将2-(1-(3,3,3-三氟丙基)磺酰基-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(30e)(120mg)溶于二氯甲烷(8mL)中,加入三氟乙酸(4.8mL),将反应液搅拌3小时。检测反应结束后,加入氨水溶液调节体系pH为10,搅拌反应过夜。将反应液用(DCM/MeOH=10:1)萃取,合并有机相,无水硫酸钠干燥,过滤,浓缩,TLC制备板纯化得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(3,3,3-三氟丙基磺酰基)氮杂环丁烷-3-基)乙腈(30)(70mg,淡黄色固体,收率为60.5%),1H NMR(400MHz,CDCl3)δ9.34(s,1H),8.31(d,J=5.24Hz,1H),8.11(d,J=3.72Hz,2H),7.39(d,J=2.84Hz,1H),7.18(d,J=5.04Hz,1H),6.70(d,J=3.56Hz,1H),4.71(d,J=9.52Hz,2H),4.30(d,J=9.20Hz,2H),3.39(s,2H),3.29(m,2H),2.68(m,2H)。MS(ESI,m/z):439[M+H]+。
实施例31:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-吡唑-1-基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)乙腈(31)
第一步:4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(31b)
在冰盐浴下,将4-溴-7-氮杂吲哚(31a)(10g,50.7mmol)和DMF(100mL)加入到反应瓶中,Ar保护后向反应液中分批加入氢化钠(60%,2.64g,54.4mmol)和[2-(三甲基甲硅烷基)乙氧基]甲基氯,使反应液在搅拌下反应1小时。检测反应结束后加水淬灭,乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,浓缩残余物经柱色谱法纯化,得到化合物4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(31b)(14.57g,收率:88.0%,黄色油状物)。MS(ESI,m/z):326.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(31c)
依次将化合物31b(4g,12.3mmol)、4-吡唑硼酸频哪醇酯(3.86g,19.9mmol)和1,4-二氧六环(300mL)加入到反应瓶中,加入碳酸钾(4.58g,33.1mmol)溶液(60mL)和Pd(dppf)Cl2(0.97g,1.33mmol),将反应液用Ar保护后加热至95℃,搅拌过夜。检测反应结束后加水淬灭,乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,得到化合物4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(31c)(1.67g,收率:43.5%,黄色固体)。MS(ESI,m/z):314.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(31e)
在冰浴下,将化合物31d(1.0g,5.15mmol)和4M盐酸二氧六环溶液(10mL)加入到反应瓶中,Ar保护后搅拌反应2.5h。检测反应结束后过滤反应液,无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(31e)(600mg,收率:90.0%,白色固体),将其直接用于下一步。
第四步:2-(1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(31f)
在冰浴下,依次将化合物31e(200mg,1.54mmol)、二氯甲烷(10mL)、三乙胺(1.3mL,9.24mmol)和DMAP(3.8mg,0.03mmol)加入到反应瓶中,缓慢滴加1-甲基环丙基-1-磺酰氯(357mg,2.31mmol)的二氯甲烷溶液(10mL),将反应液搅拌1h,检测反应结束后加水淬灭,二氯甲烷萃取,收集有机相,将其分别用水、柠檬酸、饱和食盐水洗涤,无水硫酸钠干燥,过
滤,减压浓缩得到2-(1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(31f)(272mg,收率:83.4%,褐色固体)。MS(ESI,m/z):213.1[M+H]+。
第五步:2-(1-((1-甲基环丙基)磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(31g)
将化合物31f(140mg,0.66mmol)、化合物31c(197mg,0.63mmol)和乙腈(20mL)加入到反应瓶中,加入DBU(115mg,0.76mmol),将上述反应液在室温下搅拌1h。检测反应结束后加水淬灭,在减压下浓缩反应液,将浓缩残余物经柱色谱法纯化,得到2-(1-((1-甲基环丙基)磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(31g)(233mg,收率:70.4%,黄色泡状固体)。MS(ESI,m/z):526.2[M+H]+。
第六步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-吡唑-1-基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)乙腈(31)
室温下,将化合物31g(233mg,0.44mmol)与TFA/DCM(V:V=1:2)(11.2mL)的混合物加入到反应瓶中,Ar保护,室温搅拌1.5h。监测反应结束后,将反应液加水淬灭,二氯甲烷萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,得到羟甲基中间体(145mg),并将其置于反应瓶中,依次加入无水乙醇(35mL)、蒸馏水(4mL)和无水碳酸钠(721mg,6.8mmol),调节pH为9,在室温下搅拌过夜,将反应液抽滤,将滤饼用水洗涤,干燥后得到粗品,将粗品通过柱色谱法纯化得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-吡唑-1-基)-1-((1-甲基环丙基)磺酰基)氮杂环丁烷-3-基)乙腈(31)(70mg,收率:40.1%,类白色固体)。1H NMR(400MHz,DMSO-d6)δ:11.70(s,1H),8.76(s,1H),8.33(s,1H),8.19(d,J=5.00Hz,1H),7.52(t,J=2.84Hz,1H),7.32(d,J=5.00Hz,1H),6.88(dd,J1=3.64Hz,J2=1.88Hz,1H),4.56(d,J=8.80Hz,2H),4.20(d,J=8.84Hz,2H),3.66(s,2H),1.45(s,3H),1.18(t,2H),0.89(t,2H),.MS(ESI,m/z):396.1[M+H]+。
实施例32:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(叔丁基磺酰基)氮杂环丁烷-3-基)乙腈(32)
第一步:3-(氰基亚甲基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-甲酸叔丁酯(32c)
室温下,将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(32a)(500mg,1.6mmol)、3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(32b)(464mg,2.38mmol)溶于15mL的乙腈中,加入0.4mL DBU后,将反应液在搅拌下过夜。检测反应结束后,浓缩反应液,将浓缩残余物经柱色谱法纯化得到3-(氰基亚甲基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-甲酸叔丁酯(32c)(680mg,淡黄色油状物,收率为84%)。MS(ESI,m/z):509[M+H]+。
第二步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-氮杂环丁烷-3-基)乙腈(32d)
冰浴下,将第一步得到的油状物在14mL的二氯甲烷中溶解,加入14mL的三氟乙酸,将反应液温热至室温,搅拌1小时,检测反应结束后,加入20mL的冰水,用二氯甲烷萃取,将水相用氨水调节pH至10,将溶液搅拌过夜后,用二氯甲烷/甲醇(10:1)萃取,收集有机相干燥,过滤,浓缩得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-氮杂环丁烷-3-基)乙腈的粗品,MS(ESI,m/z):279[M+H]+。
第三步:叔丁基磺酰氯(32f)
冰盐浴下,将NCS(1.2g,9mmol)溶于15mL的二氯甲烷中,然后将1mL的叔丁基硫醇用5mL的二氯甲烷稀释后,缓慢滴加至反应体系中,将反应液搅拌3h,用二氯甲烷将反应液稀释至总体积为30mL,将该反应液直接用于下一步。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(叔丁基磺酰基)氮杂环丁烷-3-基)乙腈(32)
在冰浴下,将第二步得到的2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡
唑-1-基)-氮杂环丁烷-3-基)乙腈(32d)粗品(273mg)溶于15mL的1,4-二氧六环溶液中,加入0.7mL的三乙胺,将反应液搅拌10min后,将第三步得到的叔丁基磺酰氯(32f)的二氯甲烷溶液(5mL)滴加至反应体系,将反应液温热至室温,继续继续搅拌2小时,检测反应结束后,将反应液浓缩并通过TLC制备板纯化得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(叔丁基磺酰基)氮杂环丁烷-3-基)乙腈(32)(64mg,淡黄色固体),1H NMR(400MHz,CDCl3)δ:10.67(s,1H),8.35(d,1H,J=4.8Hz),8.18(s,1H),8.13(s,1H),7.47(d,1H,J=4.4Hz),7.24(d,1H,J=5.2Hz),6.76(d,1H,J=3.6Hz),4.12(d,2H,J=9.2Hz),3.88(d,2H,J=9.2Hz),3.41(s,2H),1.42(s,9H).MS(ESI,m/z):399[M+H]+。
实施例33:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙磺酰基)氮杂环丁烷-3-基)丙腈(33)
第一步:2-(1-(乙磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶基)-1H-吡唑基)氮杂环丁烷-3-基)丙腈(33c)
在室温下,向4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(33a)(160mg,0.51mmol)和2-(1-(乙磺酰基)氮杂环丁烷-3-亚基)丙腈(33)(102mg,0.51mmol)的体系中加入乙腈(10mL)和DBU(100mg),将反应液在室温下搅拌过夜。检测反应结束后将反应液浓缩,将浓缩残余物经柱色谱法纯化得到2-(1-(乙磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶基)-1H-吡唑-1-基)氮杂环丁烷-3-基)丙腈(33c)(170mg,棕色油状产物),收率:65%。MS m/z:515[M+1].
第二步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙磺酰基)氮杂环丁烷-3-基)丙腈(33)
在室温下,用乙腈(9mL)溶解2-(1-(乙磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶基)-1H-吡唑-1-基)氮杂环丁烷-3-基)丙腈(33c)(170mg,0.33mmol),加入水(1mL)和四氟硼酸锂(583mg,6.21
mmol),将反应液升温至80℃,并搅拌过夜。检测反应结束后加入水淬灭,用EA萃取,收集有机相,将其用无水硫酸钠干燥,过滤,浓缩有机相,将浓缩残余物经柱色谱法纯化,得到2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙磺酰基)氮杂环丁烷-3-基)丙腈(33)(50mg,灰色固体),收率:40%。MS m/z:385[M+1]+。1H NMR(400MHz,DMSO-d6)δ11.71(s,1H),8.76(s,1H),8.33(s,1H),8.20(d,J=5.00Hz,1H),7.65-7.49(m,1H),7.34(d,J=4.99Hz,1H),6.89(dd,J=3.58,1.85Hz,1H),4.57(dd,J=14.48,9.37Hz,2H),4.35(dd,J=9.36,6.46Hz,2H),3.92(q,J=6.99Hz,1H),3.22(q,J=7.32Hz,2H),1.23(t,J=7.33Hz,3H),1.15(d,J=6.96Hz,3H).
实施例34:3-(4-(5-氰基-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34)
第一步:3-(氰基亚甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34b)
冰水浴下,将2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(34a)(400mg)和DCM(20mL)加入到反应瓶中,加入DMAP(8mg)和TEA(1548mg),并搅拌均匀。将用DCM(20mL)稀释N,N-二甲基磺酰氯(571mg)缓慢滴加至反应体系,将反应液搅拌反应5小时。检测反应结束后加入水(20mL)和DCM(20mL),分离有机相,依次用柠檬水和饱和氯化钠洗涤,无水硫酸钠干燥,过滤,浓缩,得到粗品(34b,596mg),将其在未进行纯化下直接用于下一步反应。MS(ESI,m/z):202[M+H]+。
第二步:3-(4-(5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰基甲基)-N,N-二甲基氮杂环丁烷-1-磺
酰胺(34c)
将第一步所得的粗品3-(氰基亚甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34b)(240mg)溶于乙腈(150mL)中,加入4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(200mg)和DBU(0.5mL),使反应液在搅拌下反应2小时。检测反应结束后浓缩反应液,将浓缩残余物经TLC制备板纯化,得白色粘稠油状物(34c,200mg),收率:63%。MS(ESI,m/z):541[M+H]+。
第三步:3-(4-(5-氰基-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34d)
将3-(4-(5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰基甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34c)(200mg)溶于DCM(6mL)中,向反应体系滴加TFA(2mL),使反应液在搅拌下反应3小时,检测反应结束后浓缩反应液,将浓缩残余物经TLC制备板纯化得到白色固体(34d,84mg)。MS(ESI,m/z):441[M+H]+。
第四步:3-(4-(5-氰基-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34)
将3-(4-(5-氰基-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-3-(氰甲基)-N,N-二甲基氮杂环丁烷-1-磺酰胺(34d)(84mg)溶于乙醇(150mL)中,加入水(15mL)和碳酸钠(403mg),将反应液在搅拌下过夜。检测反应结束后浓缩反应液,得到白色固体,加入水(30mL)过滤,将滤饼用(DCM/MeOH=1:1)(80mL)溶解,无水硫酸钠干燥,过滤,浓缩,干燥后,得到类白色固体(34)(53mg,收率为68%)。
1H NMR(400MHz,DMSO)δ12.40(s,1H),8.81(s,1H),8.63(s,1H),8.32(s,1H),7.75(d,J=3.36Hz,1H),6.88(d,J=3.40Hz,1H),4.52(d,J=8.96Hz,2H),4.22(d,J=8.96Hz,2H),3.69(s,2H),2.79(s,6H).MS(ESI,m/z):411[M+H]+。
实施例35:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(吡咯烷-1-基磺酰基)氮杂环丁烷-3-基)乙腈(35)
第一步:2-(氮杂环丁烷-3-亚基)乙腈三氟乙酸盐(35b)
如实施例2第一步中所述制备得到化合物(35b)(粗品,透明油状物),将其直接用于下一步反应。
第二步:2-(1-(吡咯烷-1-基磺酰基)氮杂环丁烷-3-亚基)乙腈(35c)
将化合物(35b)(780mg,4mmol)在二氯甲烷(10mL)中溶解,并将反应物置于冰浴下,然后缓慢加入三乙胺,直至反应物的pH达到9。然后加入DMAP(8mg,0.06mmol),将反应物在冰浴下搅拌5min。然后将吡咯烷-1-磺酰氯(0.44mL,3.90mmol)在二氯甲烷(2mL)中溶解,并将其缓慢滴加入反应体系中,在冰浴下反应30min。然后用乙酸乙酯萃取反应液,将有机相用柠檬酸的水溶液洗涤,无水硫酸钠干燥有机相,然后旋转蒸发至干燥,得到固体,将固体用石油醚研磨,过滤后得到化合物(35c)(505mg,白色固体),收率:74%,MS(ESI,m/z):228[M+H]+。
第三步:2-(1-(吡咯烷-1-基磺酰基)-3-(4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(35e)
将化合物(35d)(200mg,0.64mmol)和化合物(35c)(218mg,0.96mmol)在乙腈(10mL)中溶解,并向反应物中加入DBU(0.16mL),将反应物在室温下搅拌过夜。TLC显示反应结束后,将反应物旋转蒸发至干燥,通过硅胶柱色谱法纯化得到化合物(35e)(329mg,乳白色油状物),收率:95%。MS(ESI,m/z):542[M+H]+。
第四步:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(吡咯烷-1-基磺酰基)氮杂环丁烷-3-基)乙腈(35)
将化合物(35e)(329mg,0.61mmol)在二氯甲烷(7mL)中溶解,并将反应物置于冰浴中,加入三氟乙酸(7mL),在整个反应过程中保持温度小于10℃。TLC显示反应结束后,用碳酸钠的水溶液调节反应物的pH为9,然后用乙酸乙酯萃取,旋转蒸发至干燥后,将固体在95%乙醇中溶解,并向反应物中加入碳酸钠(954mg,9mmol),将其在室温下搅拌过夜,旋转蒸发至干燥,通过硅胶柱色谱法纯化得到化合物(35)(188mg,白色固体),收率:75%。MS(ESI,m/z):412[M+H]+。
1H NMR(400MHz,DMSO-d6)δ11.73(s,1H),8.76(s,1H),8.34(s,1H),8.21(d,1H,J=5.2Hz),7.54(s,1H),7.34(d,1H,J=4.8Hz),6.89(s,1H),4.52(d,2H,J=8.8Hz),4.17(d,2H,J=8.8Hz),3.65(s,2H),3.26(s,4H),1.84(s,4H).
实施例36:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-3-甲腈(36)
第一步:4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(36b)
如实施例8第一步中所述制备化合物(36b)(14.57g,黄色液体),收率:88%。MS(ESI,m/z):326[M+H]+。
第二步:4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(36c)
在室温下,依次将化合物(36b)(10g,27.60mmol)、联硼酸频那醇酯(8.41g,33.10mmol)、醋酸钾(2.70g,82.80mmol)和二氧六环(200mL)加入到500mL反应瓶中,用N2保护。待反应液搅拌均匀,在N2保护下加入Pd(dppf)Cl2(1.00g,1.38mmol)。将体系加热至90℃,反应3h。通过薄层色谱法监测,反应结束后将反应液加水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,得到化合物(36c)粗品,将其直接用于下一步反应。MS(ESI,m/z):375[M+H]+。
第三步:4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-3-甲腈(36e)
在室温下,依次将化合物(36c)(1.63g,4.36mmol)、4-溴-1H-吡唑-3-甲腈(36d)(500mg,2.91mmol)、碳酸钾(1.00g,7.28mmol)溶液(10mL)和1,4-二氧六环(50mL)加入到150mL反应瓶中,用N2保护。待反应液搅拌均匀,在N2保护下加入Pd(dppf)Cl2(213mg,0.29mmol)。将体系加热至100℃,反应过夜。通过薄层色谱法监测,反应结束后将反应液加水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(36e)(255mg,白色固体),收率:26%。MS(ESI,m/z):340[M+H]+。
第四步:4-(1-(3-(氰甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-(3-氰基吡唑)-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(36f)
在室温下,向化合物(36e)(189mg,0.56mmol)在乙腈(2mL)中的溶液依次加入2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(521mg,2.80mmoL)和DBU(127mg,0.84mmol),反应过夜。通过薄层色谱法监测,反应结束后将反应液加水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(36f)(140mg,淡黄色固体),收率:48%。MS(ESI,m/z):526[M+H]+。
第五步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-3-甲腈(36)
在室温下,将化合物(36f)(147mg,0.28mmol)在TFA/DCM(1:1)(4mL)
中的溶液加入到25mL反应瓶中,在氩气保护下,于室温下搅拌1h。LC-MS监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,得到黄色油状物;再在室温下向其中加入四氢呋喃(5mL),搅拌均匀后,加入1M氢氧化钠溶液将体系pH值调节至约等于10,在室温下搅拌2h。待薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(36)(10mg,白色固体),收率:10%。MS(ESI,m/z):396[M+H]+。
1H NMR(400MHz,DMSO-d6)δ11.95(s,1H),8.96(s,1H),8.35(s,1H),7.64(s,1H),7.35(s,1H),6.77(s,1H),4.62(d,J=9.2Hz,2H),4.29(d,J=9.2Hz,2H),3.74(s,2H),3.25(q,J=7.3Hz,2H),1.23(t,J=7.3Hz,3H).
实施例37:4-(1-(3-(氰甲基)-1-丙酰基氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(37)
第一步:4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(37b)
在室温下,将4-氯-5-氰基-7-氮杂吲哚(37a)(1.92g,10.76mmol)和DMF(22mL)加入到反应瓶中,N2保护后分批加入氢化钠(60wt%,560mg,13.98mmol)和[2-(三甲基甲硅烷基)乙氧基]甲基氯(2.33g,13.98mmol),将反应液搅拌2小时。监测反应结束后加水淬灭,乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,
得到4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(37b)(4.30g,收率:86%,白色固体)。MS(ESI,m/z):307.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(37c)
在室温下,依次将化合物37b(200mg,0.65mmol)、4-吡唑硼酸频哪醇酯(189mg,0.98mmol)、碳酸钾(225mg,1.63mmol)溶液(2mL)、1,4-二氧六环(8mL)加入到反应瓶中,N2保护后加入Pd(dppf)Cl2(50mg,0.065mmol)。将反应液加热至95℃,并搅拌过夜。检测反应结束后加水淬灭,乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,得到4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(37c)(134mg,收率:61%,黄色固体)。MS(ESI,m/z):339.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(37e)
将化合物37d(1.0g,5.15mmol)和4M盐酸二氧六环溶液(10mL)加入到反应瓶中,将反应液中用Ar保护后搅拌2.5h。检测反应结束后将反应液过滤,无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(37e)(600mg,收率:90.0%,白色固体),将其直接用于下一步。
第四步:2-(1-丙酰基氮杂环丁烷-3-亚基)乙腈(37f)
在冰浴下,依次将化合物37e(200mg,1.54mmol)、二氯甲烷(10mL)、三乙胺(1.3mL,9.24mmol)和DMAP(3.8mg,0.03mmol)加入到反应瓶中,搅拌均匀后,向反应体系内滴加丙酰氯(212mg,2.3mmol)的二氯甲烷溶液(10mL),将反应液搅拌1h。检测反应结束后加水淬灭,二氯甲烷萃取,收集有机相,将其分别用水、柠檬酸、饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到2-(1-丙酰基氮杂环丁烷-3-亚基)乙腈(37f)(140mg,收率:60.6%,褐色固体)。MS(ESI,m/z):150.1[M+H]+。
第五步:4-(1-(3-(氰甲基)-1-丙酰基氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(37g)
将化合物37f(107mg,0.71mmol)、化合物37c(200mg,0.59mmol)和乙腈(15mL)加入到反应瓶中,再加入DBU(183mg,0.71mmol)。将上述反应液在室温下搅拌1h。检测反应结束后加水淬灭,减压浓缩反应液,
将浓缩残余物经柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-丙酰基氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(37g)(202mg,收率:70.0%,白色泡状固体)。MS(ESI,m/z):489.2[M+H]+。
第六步:4-(1-(3-(氰甲基)-1-丙酰基氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(37)
室温下,将化合物37g(202mg,0.44mmol)与TFA/DCM(V:V=1:2)(9.75mL)的混合液加入到反应瓶中,在Ar保护下于室温下搅拌2h。监测反应结束后,将反应液加水淬灭,二氯甲烷萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,得到羟甲基中间体(137mg,收率:85.3%,白色固体)。将上述羟甲基中间体置于反应瓶中,加入无水乙醇(90mL)、蒸馏水(10mL)和无水碳酸钠(746mg,7.04mmol),在室温下搅拌过夜,检测反应结束后浓缩,抽滤,滤饼用水洗涤,干燥后得到化合物37(73mg,收率:57.6%,白色固体)。1H NMR(400MHz,DMSO-d6)δ:12.40(s,1H),8.79(s,1H),8.62(s,1H),8.29(s,1H),7.75(d,J=3.32Hz,1H),6.88(d,J=3.40Hz,1H),4.79(d,J=9.88Hz,1H),4.49(m,2H),4.24(d,J=9.80Hz,1H),3.69(s,2H),2.15(q,2H),0.99(t,J=7.44Hz,3H).MS(ESI,m/z):359.1[M+H]+。
实施例38:4-(1-(3-(氰甲基)-1-(环丙甲酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(38)
第一步:4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(38b)
在室温下,将4-氯-5-氰基-7-氮杂吲哚(38a)(1.92g,10.76mmol)和DMF(22mL)加入到反应瓶中,N2保护后将该混合物冷却至5℃以下,分批加入氢化钠(60wt%,560mg,13.98mmol)和[2-(三甲基甲硅烷基)乙氧基]甲基氯(2.33g,13.98mmol),将反应液继续搅拌2小时。检测反应结束后加水淬灭,乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,经柱色谱法纯化,得到4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(38b)(4.30g,收率:86%,白色固体)。MS(ESI,m/z):307.1[M+H]+。
第二步:4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(38c)
在室温下,依次将化合物38b(200mg,0.65mmol)、4-吡唑硼酸频哪醇酯(189mg,0.98mmol)、碳酸钾(225mg,1.63mmol)溶液(2mL)和1,4-二氧六环(8mL)加入到反应瓶中,N2保护后加入Pd(dppf)Cl2(50mg,0.065mmol)。将反应液加热至95℃,并搅拌过夜。检测反应结束后加水淬灭,用乙酸乙酯萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,得到4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(38c)(134mg,收率:61%,黄色固体)。MS(ESI,m/z):339.2[M+H]+。
第三步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(38e)
在冰浴下,将化合物38d(1.0g,5.15mmol)和4M盐酸二氧六环溶液(10mL)加入到反应瓶中,Ar保护后搅拌反应2.5h。检测反应结束后过滤反应液,用无水乙醚洗涤滤饼,干燥后得到2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(38e)(600mg,收率:90.0%,白色固体),将其直接用于下一步。
第四步:2-(1-(环丙基甲酰基)氮杂环丁烷-3-亚基)乙腈(38f)
在冰浴下,依次将化合物38e(200mg,1.54mmol)、二氯甲烷(10mL)、三乙胺(1.3mL,9.24mmol)和DMAP(3.8mg,0.03mmol)加入到反应瓶中,向反应体系内缓慢滴加环丙甲酰氯(240mg,2.30mmol)的二氯甲烷溶液(10mL),将反应液搅拌1h。检测反应结束后加水淬灭,二氯甲烷萃取,收集有机相,将其分别用水、柠檬酸、饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到2-(1-(环丙基甲酰基)氮杂环丁烷-3-亚基)乙腈(38f)(223mg,收率:89.9%,褐色固体)。MS(ESI,m/z):162.1[M+H]+。
第五步:4-(1-(3-(氰甲基)-1-(环丙甲酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(38g)
将化合物38f(115mg,0.71mmol)、化合物38c(200mg,0.59mmol)和乙腈(15mL)加入到反应瓶中,继续加入DBU(183mg,0.71mmol)。将上述反应液在室温下搅拌1h。检测反应结束后加水淬灭,减压浓缩反应液,将浓缩残余物经柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-(环丙甲酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(38g)(236mg,收率:79.8%,白色固体)。MS(ESI,m/z):501.2[M+H]+。
第六步:4-(1-(3-(氰甲基)-1-(环丙甲酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(38)
在室温下,将化合物38g(236mg,0.47mmol)与TFA/DCM(V:V=1:2)(11.38mL)的混合物加入到反应瓶中,在Ar保护下于室温下搅拌2h。检测反应结束后加水淬灭,用二氯甲烷萃取,收集有机相,将其用无水硫酸钠干燥,过滤,减压浓缩,将浓缩残余物经柱色谱法纯化,得到羟甲基中间体(157mg,收率:83.1%,白色固体)。将上述羟甲基中间体置于反应瓶中,加入无水乙醇(100mL)、蒸馏水(11mL)和无水碳酸钠(830mg,7.83mmol),将反应液在室温下搅拌过夜。检测反应结束后过滤,将滤饼用水洗涤,干燥后得到化合物38(101mg,收率:70.0%,白色固体)。1H NMR(400MHz,DMSO-d6)δ:12.40(s,1H),8.81(s,1H),8.63(s,1H),8.31(s,1H),7.75(d,J=3.60Hz,1H),6.88(d,J=3.52Hz,1H),4.92(d,J=9.04Hz,1H),4.66(d,J=9.20Hz,1H),4.49(d,J=10.76Hz,1H),4.66(d,J=10.32Hz,1H),3.73(s,2H),1.63(m,1H),0.75(s,4H).MS(ESI,m/z):371.1[M+H]+。
实施例39:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-2-甲腈(39)
第一步:4-溴-2-甲酰基-1H-吡咯-1-羧酸叔丁酯(39b)
在室温下,用DCM(100mL)溶解4-溴-1H-吡咯-2-甲醛(39a)(7.40g,43mmol)和二碳酸二叔丁酯(9.40g),然后加入Et3N(8.80mL)和4-二甲氨基吡啶(263mg),在室温下搅拌4h。TLC监测(PE:EA=5:1)底物消失后,将反应体系倒入柠檬酸水溶液中,用DCM萃取。合并有机相,用无水硫酸钠干燥,旋干溶剂得到化合物(39b)(11.58g,棕色固体),收率:99%。MS m/z:274[M+1]+。
第二步:2-甲酰基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1H-吡咯-1-羧酸叔丁酯(39c)
将化合物(39b)(11.58g,0.04mol)、联硼酸频那醇酯(21.54g,0.08mol)、乙酸钾(12.47g,0.12mol)和Pd(dppf)Cl2(2.08g)置于250mL单口瓶中,用氮气置换,最后加入二氧六环(120mL),用氮气保护。将反应体系置于100℃的油浴锅中,搅拌反应5h。TLC监测(PE:EA=6:1)底物消失后,过滤移除不溶物,将滤液浓缩后用EA溶解,通过快速制备色谱法(PE:EA=10:1)纯化,得到化合物(39c)(8.70g,黄色油状物),收率:67%。MS m/z:266[M-55]+。
第三步:4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-2-甲醛(39d)
将4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶
(2.50g,8.80mmol)、化合物(39c)(4.60g)和碳酸钾(3.10g,22.08mmol)置于100mL烧瓶中,加入二氧六环(50mL)和水(6mL),搅拌均匀,再加入Pd(dppf)Cl2(370mg),用氮气置换2-3次,将烧瓶放于100℃的油浴锅中进行反应。反应过夜,通过LC-MS确认底物消失后,通过硅藻土过滤移除不溶物,旋干溶剂,用EA溶解后,用水洗涤,最终获得有机相,用无水硫酸钠干燥。通过快速制备色谱法(PE:EA=3:2)纯化,获得化合物(39d)(1.67g,黄色固体),收率:56%。MS m/z:243[M+1]+。
第四步:4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-2-甲腈(39e)
用NMP(15mL)溶解化合物(39d)(1.67g,5mmol)和盐酸羟胺(520mg,7mmol),在125℃油浴锅中搅拌反应18h。LC-MS监测反应完成后,将反应体系冷却至室温,倒入柠檬酸水溶液中,用EA萃取,合并有机相,用无水硫酸钠干燥,通过快速制备色谱法(PE:EA=3:2)纯化,得到化合物(39e)(1.65g,油状产物),收率:97%。MS m/z:340[M+1]+。
第五步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-2-甲腈(39f)
将化合物(39e)(1.65g,4.86mmol)和2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(1.36g,7.30mmol)加入乙腈(50mL)中,使反应物在溶剂中分散,最后加入叔丁基醇钾(1.37g,12.17mmol),将反应物在室温下搅拌24h。TLC监测(PE:EA=1:1)反应完成后,加水淬灭,用EA萃取产物,无水硫酸钠干燥有机相,浓缩有机相,通过快速制备色谱法(PE:EA=1:1)纯化,得到化合物(39f)(1.30g,黄色油状物),收率:51%。MS m/z:526[M+1]+。
第六步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-2-甲腈(39)
在室温下,用二氯甲烷(20mL)溶解化合物(39f)(1.3g,2.48mmol),待体系在搅拌下澄清后,加入三氟化硼乙醚溶液,搅拌反应3h。LC-MS监测底物消失后,旋干溶剂后,加入20mL甲醇,缓慢滴加1N NaOH水溶液,调节pH值至10左右,在室温下搅拌反应,过夜。经分离纯化,得到化合物(39)(120mg,白色固体),收率:55%。MS m/z:410[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.17(s,1H),8.71(s,1H),8.22(d,J=1.7Hz,1H),7.96(d,J=1.7Hz,1H),7.63(d,J=3.6Hz,1H),7.10(d,J=3.6Hz,1H),4.71(d,J=9.2Hz,2H),4.25(d,J=9.3Hz,2H),3.66(s,2H),3.27(q,J=7.3Hz,2H),1.25(t,J=7.3Hz,3H).
实施例40:4-(1-(3-(氰基甲基)-1-(丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40)
第一步:4-(1-(3-(氰基甲基)-1-(丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-(1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40c)
在室温下,向4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40a)(220mg,0.64mmol)和2-(1-(丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(40a)(192mg,0.96mmol)的混合物中加入乙腈(20mL)和DBU(200mg),将反应液在室温下搅拌过夜。检测反应结束后,将反应物浓缩,将浓缩残余物经TLC制备板纯化,得到4-(1-(3-(氰基甲基)-1-(丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-(1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40c)(290mg,棕色产物),收率:83%。MS m/z:540[M+1]+。
第二步:4-(1-(3-(氰基甲基)-1-(丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40)
在室温下,用DCM(12mL)溶解4-(1-(3-(氰基甲基)-1-(丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-(1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40c)(290mg,0.54mmol),在冰浴条件下加入TFA(6mL),将反应液缓慢升温至室温,并搅拌过夜。检测反应结束后用饱和碳酸钠调节pH值为7左右,EA萃取,收集有机相,将其用无水硫酸钠干燥,过滤,浓缩,浓缩残余物经柱色谱法,得到中间产物固体(200mg)。将中间产物固体用THF(20mL)溶解,滴加1mol/L的氢氧化钠溶液,使反应体系
pH值为10左右,将反应液在室温下搅拌,检测反应结束后将反应液用EA萃取,收集有机相,将其用无水硫酸钠干燥,过滤,浓缩得到4-(1-(3-(氰基甲基)-1-(丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(40)(120mg,白色固体),收率:55%。MS m/z:410[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.40(s,1H),8.81(s,1H),8.63(s,1H),8.32(s,1H),7.75(d,J=3.48Hz,1H),6.88(d,J=3.56Hz,1H),4.58(d,J=9.28Hz,2H),4.27(d,J=9.28Hz,2H),3.69(s,2H),3.22(t,J=7.66Hz,2H),1.68-1.77(m,2H),0.99(t,J=7.40Hz,3H).
实施例41:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-3-甲腈(60)
第一步:4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-3-甲腈(60b)
在室温下,用二氧六环(6mL)溶解4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(60a)(268mg,0.95mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1-(三异丙基甲硅烷基)-1H-吡咯-3-甲腈(532mg,1.50mmol),然后加入水(0.6mL)和七水磷酸钾(801mg),用氮气置换,在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(77mg)。将反应体系移至90℃油浴锅中搅拌反应过夜。TLC检测,底物消失,将反应液缓慢倒入冰水中淬灭,用EA萃取,无水硫酸钠干燥有机相。通过硅胶制备板
(PE:EA=1:1)纯化,得到化合物(60b)(235mg,白色固体),收率:73%。MS m/z:340[M+1]+。
第二步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-3-甲腈(60c)
在室温下,向化合物(60b)(200mg,0.59mmol)和2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(164mg)的体系中加入乙腈(10mL),反应液浑浊。再加入DBU(269mg),反应液变澄清,在室温下搅拌过夜。将反应液浓缩,加入DCM溶解,通过硅胶制备板(PE:EA=1:2)纯化,得到化合物(60c)(232mg,淡黄色固体),收率:75%。MS m/z:526[M+1]+。
第三步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-3-甲腈(60)
向化合物(60c)(232mg,0.44mmol)中加入DCM(10mL)和三氟化硼乙醚(188mg),在室温下搅拌2h。LC-MS检测底物消失后,旋干溶剂,得到油状物,再加入MeOH(10mg)使其完全溶解,接着滴入1mol/L的NaOH水溶液,调节其pH值至10左右,在室温下搅拌4h。LC-MS检测反应完成。减压浓缩,移除大部分MeOH,加水后析出固体,过滤得到固体,通过反相制备色谱法纯化,得到化合物(60)(80mg,白色固体)。收率46%。MS m/z:396[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.20(s,1H),8.76(s,1H),8.18(d,J=2.4Hz,1H),8.06(d,J=2.4Hz,1H),7.65(t,J=2.9Hz,1H),7.05–6.98(m,1H),4.54(d,J=9.4Hz,2H),4.25(d,J=9.4Hz,2H),3.65(s,2H),3.23(q,J=7.3Hz,2H),1.24(t,J=7.3Hz,3H).
实施例42:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-2-甲腈(61)
第一步:4-溴-1H-吡咯-2-甲腈(61b)
用NMP(10mL)溶解4-溴-1H-吡咯-2-甲醛(61a)(2.00g,11.49mmol)和盐酸羟胺(1.30g,18.39mmol),在125℃油浴锅中搅拌反应18h。LC-MS监测反应完成后,将反应体系冷至室温,倒入柠檬酸水溶液中,用EA萃取,合并有机相,用无水硫酸钠干燥,通过快速制备色谱法(PE:EA=9:1)纯化,得到化合物(61b)(1.18g,棕色固体),收率:60%。MS m/z:170[M+1]+。
第二步:4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(61d)
将4-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(61c)(1.00g,3.06mmol)、联硼酸频那醇酯(1.50g,6.11mmol)、乙酸钾(898mg,9.17mmol)和Pd(dppf)Cl2(125mg)置于100mL单口瓶中,用氮气置换,最后加入二氧六环(12mL)。将反应体系置于100℃的油浴锅中,搅拌反应3h。TLC监测(PE:EA=10:1)底物消失后,过滤掉不溶物,浓缩后用EA溶解,通过快速制备色谱法(PE:EA=9:1)纯化,得到化合物(61d)(1.58g,黄色油状物),其中含有少量溶剂。MS m/z:375[M+1]+。
第三步:4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-2-甲腈(61e)
将化合物(61b)(500mg,2.94mmol)、化合物(61d)(1.60g)和碳酸钾(1.1g)置于100mL烧瓶中,加入二氧六环(20mL)和水(3mL),搅拌均匀,再加入Pd(dppf)Cl2(241mg),用氮气置换2-3次,将烧瓶放于100℃的油浴锅中进行反应。反应过夜,通过LC-MS监测底物消失后,通过硅藻土过滤移除不溶物,旋干溶剂,用EA溶解后,加水冲洗,最终获得有机相,用无水硫酸钠干燥,通过快速制备色谱法(PE:EA=2:1)纯化,获得化合物(61e)(260mg,固体),收率:26%。MS m/z:339[M+1]+。
第四步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-2-甲腈(61f)
向化合物(61e)(260mg,0.77mmol)和2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(215mg,1.15mmol)中加入DMF(20mL),使反应物在溶剂中分散,最后加入碳酸钾(319mg,2.31mmol),在室温下搅拌过夜。LC-MS监测底物消失后,将反应液倒入水中,用EA萃取,无水硫酸钠干燥有机相,浓缩有机相,通过硅胶制备板(PE:EA=1:1)纯化,得到化合物(61f)(159mg,油状物),收率:39%。MS m/z:525[M+1]+。
第五步:1-(3-(氰基甲基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯-2-甲腈(61)
在室温下,用二氯甲烷(5mL)溶解化合物(61f)(159mg,0.30mmol),待反应液在搅拌下澄清后,加入三氟化硼乙醚溶液(130mg),搅拌反应2h。LC-MS监测底物消失后,旋干溶剂,加入5mL甲醇,缓慢滴加1N NaOH水溶液,调节pH值至10左右,室温搅拌反应,过夜。LC-MS监测反应完成后,浓缩掉大部分甲醇,加水使固体分散,过滤,将固体通过反相制备色谱法纯化,得到化合物(61)(20mg,白色固体),收率:17%。MS m/z:395[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.72(s,1H),8.20(d,J=5.0Hz,1H),8.04(d,J=1.8Hz,1H),7.81(d,J=1.8Hz,1H),7.53(t,J=3.0Hz,1H),7.29(d,J=5.0Hz,1H),6.89(dd,J=3.6,1.8Hz,1H),4.70(d,J=8.9Hz,2H),4.25(d,J=8.8Hz,2H),3.65(s,2H),3.28(q,J=7.4Hz,2H),1.25(t,J=7.3Hz,3H).
实施例43:4-(1-(3-(氰甲基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(43)
第一步:2-(1-(异丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(43b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(43a)(583mg,3.00mmol)和TFA/DCM(1/3,8mL)加入到50mL反应瓶中,在室温下搅拌半小时,当薄层色谱法显示所有起始原料基本消失后,将该反应混合物在减压下浓缩至干燥。然后,将所得粗产物溶于DCM(10mL)中。冰浴下,缓慢滴加三乙胺调节体系pH值约等于8后,先加入DMAP(7mg,0.06mmol),随后缓慢滴加异丙基磺酰氯(705mg,4.5mmol)。将所得反应混合物逐渐升至室温,并在室温下继续搅拌半小时。待LC-MS显示反应完全时,将反应液加水淬灭,用二氯甲烷萃取,分别用水、柠檬酸、饱和食盐水洗涤有机相,无水硫酸钠干燥,减压浓缩得到2-(1-(异丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(43b)(600mg,收率:93%,褐色固体)。MS(ESI,m/z):201[M+H]+。
第二步:4-(1-(3-(氰甲基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(43d)
将2-(1-(异丙基磺酰基)氮杂环丁烷-3-亚基)乙腈(43b)(220mg,0.65mmol)、4-(1H-吡唑-4-基)-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(43c)(220mg,0.59mmol)和乙腈(8mL)加入到50mL反应瓶中,随后加入DBU(99mg,0.65mmol),将上述反应液在室温下反应2小时。待薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃
取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-(1-(3-(氰甲基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(43d)(290mg,收率:91%,淡黄色固体)。MS(ESI,m/z):540[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(43)
在冰浴下,将4-(1-(3-(氰甲基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(43d)(290mg,0.54mmol)与TFA/DCM(1:1,6mL)的混合液加入到25mL反应瓶中,Ar保护,反应2.5小时。待薄层色谱法监测反应结束后,在减压下浓缩,得黄色油状物。然后将所得粗产品直接溶于四氢呋喃中,待搅拌均匀后,加入1M氢氧化钠溶液调节体系pH约等于10,反应0.5小时。通过薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用薄层色谱法纯化得到目标产物4-(1-(3-(氰甲基)-1-(异丙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(43)(105mg,收率:48%,白色固体)。
1H NMR(1MHz,DMSO-d6)δ12.41(s,1H),8.80(s,1H),8.63(s,1H),8.31(s,1H),7.75(s,1H),6.88(s,1H),4.57(d,J=8.8Hz,2H),4.25(d,J=8.8Hz,2H),3.70(s,2H),1.28(s,3H),1.26(s,3H).MS(ESI,m/z):410[M+H]+。
实施例44:4-(1-(3-(氰甲基)-1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44)
第一步:2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(44b)
将3-(氰基亚甲基)氮杂环丁烷-1-甲酸叔丁酯(2.0g)加入到反应瓶中,用冰水浴降温,向反应体系滴加1,4-二氧六环/HCl(20mL),加毕,撤除冰浴,搅拌反应2小时,体系析出大量白色固体,过滤反应体系,将滤饼用甲基叔丁基醚洗涤,干燥得白色固体2-(氮杂环丁烷-3-亚基)乙腈盐酸盐(44b)(1.15g,收率85.4%)。MS(ESI,m/z):95[M+H]+。
第二步:2-(1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(44c)
将第一步所得的白色固体(44b)(500mg)加入到二氯甲烷(20mL)中,加入(3,3,3-三氟丙基)磺酰氯(1.126g),并将体系置于冰水浴下,将用DCM(15mL)稀释的TEA(1.16g)缓慢滴加至反应体系,加毕,搅拌反应5小时,往反应体系中加入水(10mL),搅拌10分钟,分离有机相,将水相用DCM反萃取1次(30mL),合并有机相,浓缩至干燥,将其直接用于下一步反应。MS(ESI,m/z):255[M+H]+。
第三步:4-(1-(3-(氰甲基)-1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44e)
将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44d)(350mg)和2-(1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-亚基)乙腈(44c)(940mg,粗品)溶于乙腈(15mL)中,并在反应体系中加入DBU(1mL),在室温下搅拌过夜,TLC显示反应结束后,旋干,通过硅胶柱色谱法纯化得到黄色固体(44e,380mg)。MS(ESI,m/z):594[M+H]+。
第四步:4-(1-(3-(氰甲基)-1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44f)
将4-(1-(3-(氰甲基)-1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44e)(350g)溶于二氯甲烷(8mL)中,并将反应体系置于冰浴中,加入三氟乙酸,反应约3小时。TLC显示反应结束后,浓缩反应液,TLC纯化得白色固体(44f,160mg)。MS(ESI,m/z):494[M+H]+。
第五步:4-(1-(3-(氰甲基)-1-((3,3,3-三氟丙基)磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44)
往反应瓶中加入4-(1-(3-(氰甲基)-1-((3,3,3-三氟丙基)磺酰基)氮杂环丁
烷-3-基)-1H-吡唑-4-基)-1-(羟甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(44f)(160mg)和乙醇(200mL),通过超声助溶,然后加入水(20mL)和碳酸钠(678mg),搅拌反应过夜。浓缩反应液,析出大量固体,过滤,水洗,真空干燥,得白色固体(44,65mg)。1H NMR(400MHz,DMSO)δ12.42(s,1H),8.81(s,1H),8.63(s,1H),8.32(s,1H),7.75(s,1H),6.88(s,1H),4.66(m,J=8.92Hz,2H),4.35(m,J=8.96Hz,2H),3.72(s,2H),3.59(m,3H),2.76(m,2H).MS(ESI,m/z):464[M+H]+。
实施例45:4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45)
第一步:3-(1-氰基亚乙基)氮杂环丁烷-1-甲酸叔丁酯(45b)
在冰浴条件下,向(1-氰乙基)膦酸二乙酯(45a)(2g,11mmol)的四氢呋喃(100mL)溶液中加入氢化钠(880mg,60%),搅拌40min后,体系变为粉红色,向反应体系中加入3-氧代氮杂环丁烷-1-甲酸叔丁酯(3.8g,22mmol),体系缓慢变为澄清。将体系缓慢升温至室温,搅拌过夜。用水淬灭,乙酸乙酯萃取,无水硫酸钠干燥,浓缩有机相,最终得到3-(1-氰基亚乙基)氮杂环丁烷-1-甲酸叔丁酯(45b)(3.95g,淡黄色油状产物),将其直接用于下一步反应。MS(ESI,m/z):209[M+H]+。
第二步:2-(氮杂环丁烷-3-亚基)丙腈盐酸盐(45c)
在室温下,向3-(1-氰基亚乙基)氮杂环丁烷-1-甲酸叔丁酯(45b)(3.8g,18.3mmol)中加入4M盐酸的二氧六环溶液(40mL),将反应物搅拌溶解。在
室温下搅拌1h,LC-MS检测,底物消失。停止搅拌,将体系中的溶剂旋干,用乙醚打浆,过滤得到2-(氮杂环丁烷-3-亚基)丙腈盐酸盐(45c)(2.24g,收率:85%,白色固体)。MS(ESI,m/z):109[M+H]+。
第三步:2-(1-(乙磺酰基)氮杂环丁烷-3-亚基)丙腈(45d)
在冰浴环境中,用100mL的乙腈溶解2-(氮杂环丁烷-3-亚基)丙腈盐酸盐(45c)(2.24g,15.5mmol),随后加入DIEA(7.0g),冰浴搅拌15min。向体系中缓慢加入乙基磺酰氯(3.98g,31mmol),使其温度不高于5℃。待底物加完后,缓慢升至室温,将反应液搅拌过夜。LC-MS检测,底物消失。通过柱色谱法纯化(PE:EA=2:1),最终获得2-(1-(乙磺酰基)氮杂环丁烷-3-亚基)丙腈(45d)(1.2g,收率:65%,淡黄色油状产物)。MS(ESI,m/z):201[M+H]+。
第四步:4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(45f)
在室温下,将4-氯-5-氰基-7-氮杂吲哚(45e)(1.92g,10.76mmol)和DMF(22mL)加入到100mL三颈瓶中,N2保护。将该混合物在冰盐浴中冷却至5℃以下,待反应液搅拌均匀后,向瓶内分批加入氢化钠(60wt%,560mg,13.98mmol),保持体系温度始终不高于10℃。搅拌1小时后,向体系内缓慢滴加2-(三甲基甲硅烷基)乙氧甲基氯(2.33g,13.98mmol),保持温度始终不高于5℃,继续搅拌2小时。通过薄层色谱法监测反应,待原料基本消失后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(45f)(4.30g,收率:86%,白色固体)。MS(ESI,m/z):308[M+H]+。
第五步:4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45g)
在室温下,依次将4-氯-5-氰基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(45f)(200mg,0.65mmol)、4-吡唑硼酸频哪醇酯(189mg,0.98mmol)、碳酸钾(225mg,1.63mmol)溶液(2mL)和1,4-二氧六环(8mL)加入到50mL反应瓶中,用N2保护。将反应液搅拌均匀,随后加入Pd(dppf)Cl2(50mg,0.065mmol),用N2保护。将体系加热至95℃,回流反
应过夜,通过薄层色谱法监测,待原料反应完后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45g)(134mg,收率:61%,黄色固体)。MS(ESI,m/z):340[M+H]+。
第六步:4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45h)
依次将4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45g)(600mg,1.77mmol)、2-(1-(乙磺酰基)氮杂环丁烷-3-亚基)丙腈(45d)(388mg,1.94mmol)和乙腈(20mL)加入到50mL反应瓶中,随后加入DBU(277mg,1.94mmol),在室温下反应2小时。待薄层色谱法监测反应结束后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化,得到4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45h)(851mg,收率:89%,淡黄色固体)。MS(ESI,m/z):540[M+H]+。
第七步:4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45)
在室温下,将4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45h)(851mg,1.58mmol)与TFA/DCM(1:1,20mL)的混合液加入到50mL反应瓶中,Ar保护,反应2.5小时。待薄层色谱法监测反应结束后,减压浓缩,得黄色油状物。然后将所得粗产品直接溶于四氢呋喃(20mL)中,待搅拌均匀后,加入1M氢氧化钠溶液调节体系pH约等于10,反应0.5小时。薄层色谱法监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,在减压下浓缩,用硅胶柱色谱法纯化得到目标产物4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45)(305mg,收率:48%,白色固体)。
1H NMR(400MHz,DMSO-d6)δ12.44(s,1H),8.82(s,1H),8.66(s,1H),
8.32(s,1H),7.78(d,J=5.00Hz,1H),6.87(d,J=5.00Hz,1H),4.59(dd,J=14.48,9.37Hz,2H),4.40(dd,J=9.36,6.46Hz,2H),3.98(q,J=6.99Hz,1H),3.25(q,J=7.32Hz,2H),1.26(t,J=7.33Hz,3H),1.20(d,J=6.96Hz,3H).MS(ESI,m/z):410[M+H]+。
将第六步中合成的化合物(45h)通过手性色谱法分离得到立体异构体(45h-1)(保留时间:2.7分钟)和立体异构体(45h-2)(保留时间:3.2分钟),将立体异构体(45h-1)和(45h-2)分别脱除保护基后获得化合物(45-1)和化合物(45-2):
(R)-4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45-1):MS(ESI,m/z):410[M+H]+。1H NMR(400MHz,DMSO-d6)δ12.44(s,1H),8.82(s,1H),8.66(s,1H),8.32(s,1H),7.78(d,J=5.00Hz,1H),6.87(d,J=5.00Hz,1H),4.59(dd,J=14.48,9.37Hz,2H),4.40(dd,J=9.36,6.46Hz,2H),3.98(q,J=6.99Hz,1H),3.25(q,J=7.32Hz,2H),1.26(t,J=7.33Hz,3H),1.20(d,J=6.96Hz,3H);和
(S)-4-(1-(3-(1-氰乙基)-1-(乙基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(45-2):MS(ESI,m/z):410[M+H]+。1H NMR(400MHz,DMSO-d6)δ12.44(s,1H),8.82(s,1H),8.66(s,1H),8.32(s,1H),7.78(d,J=5.00Hz,1H),6.87(d,J=5.00Hz,1H),4.59(dd,J=14.48,9.37Hz,2H),4.40(dd,J=9.36,6.46Hz,2H),3.98(q,J=6.99Hz,1H),3.25(q,J=7.32Hz,2H),1.26(t,J=7.33Hz,3H),1.20(d,J=6.96Hz,3H)。
实施例46:2-(1-(乙基磺酰基)-3-(4-(5-氟-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈盐酸盐(52-HCl)
将实施例52中获得的化合物(52)(60mg)在混合溶剂(EA/EtOH=1,15mL)中分散,缓慢滴加1mol/L的盐酸在乙酸乙酯中的溶液,待反应液澄清后,停止滴加,在室温下搅拌10min,旋干有机溶剂,加水(30mL)使固体分散,冻干后得到化合物(52)的盐酸盐(52-HCl)(65mg,白色固体)。MS m/z:389[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.90(s,1H),8.73(s,1H),8.27(t,J=3.2Hz,2H),7.64(t,J=3.0Hz,1H),6.90(dd,J=3.6,1.8Hz,1H),4.60(d,J=9.1Hz,2H),4.25(d,J=9.1Hz,2H),3.69(s,2H),3.24(q,J=7.3Hz,2H),1.24(t,J=7.3Hz,3H).
实施例47:2-(3-(3-氯-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(59)
根据以上合成路线,采用与实施例41类似的操作,制备得到化合物(59)(98mg,白色固体)。收率59%。MS m/z:405[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.10(s,1H),8.73(s,1H),7.74(d,J=2.7Hz,1H),7.57(t,J=2.9Hz,1H),7.42(d,J=2.7Hz,1H),6.82(dd,J=3.6,1.8Hz,1H),4.47(d,J=9.2Hz,2H),4.20(d,J=9.2Hz,2H),3.59(s,2H),3.23
(q,J=7.3Hz,2H),1.24(t,J=7.3Hz,3H).
实施例48:2-(3-(3-氯-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(48)
第一步:4-溴-3-硝基-1H-吡唑(48b)
在室温下,将水(15mL)加入到3-硝基-1H-吡唑(48a)(1g,8.90mmol)与NBS(1.73g,9.70mmol)的混合物中,搅拌过夜。通过薄层色谱法监测反应完成后,将反应液加水稀释,用乙酸乙酯萃取,饱和氯化钠水洗,无水硫酸钠干燥,减压浓缩,旋干得到化合物(48b)(1.66g,收率:93%,白色固体)。MS(ESI,m/z):192[M+H]+。
第二步:4-溴-3-硝基-1-对甲苯磺酰基-1H-吡唑(48c)
在室温下,将化合物(48b)(500mg,2.60mmol)、对甲苯磺酰氯(595mg,3.10mmol)和二氯甲烷(10mL)加入到圆底烧瓶中,然后逐滴加入吡啶(1.3mL),反应3h。通过薄层色谱法监测反应完成后,将反应液用饱和碳酸氢钠溶液淬灭,用二氯甲烷萃取,饱和氯化钠水洗,无水硫酸钠干燥,减压浓缩,旋干得到化合物(48c)(652mg,收率:73%,白色固体)。MS(ESI,m/z):346[M+H]+。
第三步:4-溴-1-对甲苯磺酰基-1H-吡唑-3-胺(48d)
在室温下,依次将化合物(48c)(650mg,1.90mmol)、铁粉(526mg,9.40mmol)、氯化铵(502mg,9.40mmol)、乙醇(20mL)和水(20mL)加入到100mL圆底烧瓶中,加热至55℃反应2h。通过薄层色谱法监测反应完成后,通
过硅藻土过滤移除铁粉,然后旋转蒸发移除部分乙醇后,用乙酸乙酯萃取,饱和氯化钠水洗,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(48d)(526mg,收率:89%,淡黄色固体)。MS(ESI,m/z):316[M+H]+。
第四步:4-溴-3-氯-1-对甲苯磺酰基-1H-吡唑(48e)
将亚硝酸叔丁酯(255mg,2.50mmol)、氯化铜(338mg,2.00mmol)和乙腈(10mL)置于50mL三口烧瓶中,在N2保护下,于冰浴下搅拌。然后将溶于乙腈(5mL)中的化合物(48d)(521mg,1.70mmol)逐滴加入到上述烧瓶中,继续反应3h。通过薄层色谱法监测反应完成后,通过硅藻土过滤移除氯化铜,然后旋转蒸发移除部分乙腈后,用乙酸乙酯萃取,饱和氯化钠水洗,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(48e)(280mg,淡黄色固体),收率:51%。MS(ESI,m/z):335[M+H]+。
第五步:4-(3-氯-1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(48g)
在室温下,N2保护下,依次将化合物(48e)(260mg,0.80mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(435mg,1.20mmol)、碳酸钾(268mg,1.90mmol)溶液(3mL)和二氧六环(15mL)加入到50mL反应瓶中。待反应液搅拌均匀后,在N2保护下加入Pd(dppf)Cl2(57mg,0.08mmol)。将体系加热至100℃,反应过夜。通过薄层色谱法监测反应完成后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(48g)(159mg,白色固体),收率:59%。MS(ESI,m/z):349[M+H]+。
第六步:2-(3-(3-氯-4-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(48h)
在室温下,向化合物(48g)(159mg,0.46mmol)在乙腈(5mL)中的溶液依次加入2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(169mg,0.91mmoL)和DBU(105mg,0.69mmol),反应3h。通过薄层色谱法监测反应完成后,将反应液用水淬灭,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(48h)(177mg,白色固体),收率:73%。MS(ESI,m/z):535[M+H]+。
第七步:2-(3-(3-氯-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(48)
在室温下,将化合物(48h)(110mg,0.21mmol)在TFA/DCM(1:1)(4mL)中的溶液加入到25mL反应瓶中,在Ar保护,于冰浴下搅拌1h。LC-MS监测反应结束后,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,得到黄色油状物;然后在室温下加入四氢呋喃(3mL),搅拌均匀后,加入1M氢氧化钠溶液将反应液pH值调节至约等于10,继续搅拌2h。在薄层色谱法监测反应完成后,用乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,通过硅胶柱色谱法纯化,得到化合物(48)(47mg,白色固体),收率:55%。MS(ESI,m/z):405[M+H]+。
1H NMR(400MHz,DMSO-d6)δ11.83(s,1H),8.70(s,1H),8.28(d,J=5.0Hz,1H),7.58–7.55(m,1H),7.29(d,J=5.0Hz,1H),6.67(dd,J=3.3,1.8Hz,1H),4.57(d,J=9.2Hz,2H),4.24(d,J=9.2Hz,2H),3.68(s,2H),3.25(q,J=7.3Hz,2H),1.24(t,J=7.3Hz,3H).
实施例49:2-(1-(乙基磺酰基)-3-(3-甲基-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(49)
第一步:4-(3-甲基-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(49b)
在室温下,用二氧六环(30mL)溶解4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(49a)(1.00g,3.52mmol)和3-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1H-吡唑(1.10g,5.28mmol),然后加入水(4
mL)和碳酸钾(1.20g),用氮气置换,在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(140mg)。将反应体系移至90℃油浴锅中搅拌反应过夜。TLC检测底物消失后,将反应液缓慢倒入冰水中淬灭,用EA萃取,用无水硫酸钠干燥有机相。通过快速制备色谱法(PE:EA=1:1)纯化,得到化合物(49b)(200mg,黄色固体),收率:18%。MS m/z:330[M+1]+。
第二步:2-(1-(乙基磺酰基)-3-(3-甲基-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(49c)
在室温下,向化合物(49b)(200mg,0.59mmol)和2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(112mg)中加入乙腈(15mL),反应液浑浊,然后加入DBU(249mg),将其在室温下搅拌过夜。将反应物浓缩,通过制备硅胶板纯化(DCM:EA=2:1),得到化合物(49c)(220mg,白色固体)。收率73%。MS m/z:516[M+1]+。
第三步:2-(1-(乙基磺酰基)-3-(3-甲基-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(49)
向化合物(49c)(220mg,0.43mmol)中加入DCM(10mL)和三氟化硼乙醚(182mg),在室温下搅拌4h。LC-MS检测底物消失后,旋干溶剂,得到油状物,再加入MeOH(9mg)使其完全溶解,然后加入1mol/L的NaOH水溶液,调节其pH值至10左右,在室温下搅拌过夜。LC-MS检测反应完成后,将反应物减压浓缩,移除大部分MeOH,加水分散后析出固体,过滤得到固体,通过反相制备色谱法纯化,得到化合物(49)(114mg,白色固体)。收率69%。MS m/z:386[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.12(s,1H),8.74(s,2H),7.60(d,J=3.6Hz,1H),6.96(d,J=3.6Hz,1H),4.60(d,J=9.1Hz,2H),4.21(d,J=9.2Hz,2H),3.65(s,2H),3.24(q,J=7.3Hz,2H),2.60(s,3H),1.25(t,J=7.3Hz,3H).
实施例50:2-(3-(4-(5-氯-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(50)
第一步:4-溴-5-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(50b)
用DMF(3mL)溶解4-溴-5-氯-1H-吡咯并[2,3-b]吡啶(50a)(200mg,0.86mmol),用氮气置换,将其至于冰盐浴中,使溶液温度降至0℃以下,随后加入NaH(60%,80mg),搅拌30min。最后加入SEMCl(190mg,1.12mmol),待温度稳定后,移至室温搅拌反应4h。TLC监测(PE:EA=10:1)底物消失后,用冰水淬灭反应体系,通过EA萃取,合并有机相,用无水硫酸钠干燥,过滤,浓缩,通过制备硅胶板(PA:EA=20:1)纯化,得到化合物(50b)(275mg,油状产物),收率:89%。MS m/z:361[M+1]+。
第二步:5-氯-4-(1H-吡唑-4-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶(50c)
在室温下,用二氧六环(10mL)溶解化合物(50b)(275mg,0.76mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1H-吡唑(296mg,1.52mmol),然后加入水(1.5mL)和碳酸钾(316mg,2.29mmol),用氮气置换,在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(80mg)。将反应体系移至90℃油浴锅中搅拌反应过夜。TLC检测底物消失后,将反应液缓慢倒入冰水中淬灭,用EA萃取,无水硫酸钠干燥有机相。通过快速制备色谱法(PE:EA=2:1)纯化,得到化合物(50c)(130mg,黄色固体),收率:49%。MS m/z:349[M+1]+。
第三步:2-(3-(4-(5-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(50d)
在室温下,向化合物(50c)(130mg,0.37mmol)和2-[1-(乙基磺酰基)氮
杂环丁烷-3-亚基]乙腈(76mg,0.41mmol)中加入乙腈(3mL),反应液浑浊。然后加入DBU(170mg),体系变澄清,在室温下搅拌过夜。减压浓缩反应液,通过制备硅胶板(PE:EA=2:1)纯化,得到化合物(50d)(141mg,油状物)。收率71%。MS m/z:535[M+1]+。
第四步:2-(3-(4-(5-氯-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈(50)
向化合物(50d)(141mg,0.26mmol)中加入DCM(3mL)和三氟化硼乙醚(112mg),在室温下搅拌2h。LC-MS检测底物消失后,将溶剂旋转蒸发至干燥,得到油状物,然后加入MeOH(3mL)使其完全溶解,接着滴入1mol/L的NaOH水溶液,调节其pH值至10左右,在室温下搅拌4h。LC-MS检测反应完成。减压浓缩移除大部分MeOH,加水分散后析出固体,过滤得到固体,通过反相制备色谱法纯化,得到化合物(50)(54mg,白色固体)。收率51%。MS m/z:405[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.97(s,1H),8.66(s,1H),8.29(s,1H),8.19(s,1H),7.60(d,J=2.6Hz,1H),6.68–6.61(m,1H),4.58(d,J=9.1Hz,2H),4.25(d,J=9.1Hz,2H),3.68(s,2H),3.24(q,J=7.3Hz,2H),1.25(t,J=7.3Hz,3H).
实施例51:2-(1-(乙基磺酰基)-3-(4-(5-甲基-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(51)
根据以上反应路线,用4-氯-5-甲基-1H-吡咯并[2,3-b]吡啶(51a)为起始原料,采用与实施例50类似的操作,制备得到化合物(51)(11mg,白色固
体)。收率32%。MS m/z:385[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.54(s,1H),8.49(s,1H),8.12(s,1H),8.05(s,1H),7.42(t,J=2.4Hz,1H),6.50(d,J=1.7Hz,1H),4.58(d,J=9.1Hz,2H),4.24(d,J=9.1Hz,2H),3.67(s,2H),3.24(q,J=7.4Hz,2H),2.42(s,3H),1.25(t,J=7.3Hz,3H).
实施例52:2-(1-(乙基磺酰基)-3-(4-(5-氟-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-氮杂环丁烷-3-基)乙腈(52)
根据以上反应路线,用4-氯-5-氟-1H-吡咯并[2,3-b]吡啶(52a)为起始原料,采用与实施例50类似的操作,制备得到得到化合物(52)(90mg,白色固体)。收率20%。MS m/z:389[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.88(s,1H),8.73(s,1H),8.28–8.25(m,2H),7.64(d,J=3.4Hz,1H),6.90(d,J=3.5Hz,1H),4.60(d,J=9.1Hz,2H),4.25(d,J=9.1Hz,2H),3.69(s,2H),3.24(q,J=7.3Hz,2H),1.25(t,J=7.3Hz,3H).
实施例53:2-(1-(乙基磺酰基)-3-(3-甲基-4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈盐酸盐(13-HCl)
采用与实施例46中类似的操作,由实施例13中合成的化合物(13)(300
mg)制备化合物(13)的盐酸盐(13-HCl)(336mg,白色固体)。MS m/z:385[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.85(s,1H),8.73(s,1H),8.45(d,J=6.0Hz,1H),7.78(dd,J=3.6,2.3Hz,1H),7.52(d,J=6.0Hz,1H),7.01(dd,J=3.6,1.6Hz,1H),4.58(d,J=9.1Hz,2H),4.23(d,J=9.1Hz,2H),3.67(s,2H),3.24(q,J=7.3Hz,2H),2.47(s,3H),1.25(t,J=7.3Hz,3H).
实施例54:2-(3-(4-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(异丁基磺酰基)氮杂环丁烷-3-基)乙腈盐酸盐(25-HCl)
采用与实施例46中类似的操作,由实施例25中合成的化合物(25)(30mg)制备化合物(25)的盐酸盐(25-HCl)(33mg,白色固体)。MS m/z:399[M+1]+。
1H NMR(400MHz,DMSO-d6)δ12.61(s,1H),9.04(s,1H),8.55(s,1H),8.40(d,J=5.8Hz,1H),7.75(dd,J=3.6,2.2Hz,1H),7.68(d,J=5.8Hz,1H),7.21(dd,J=3.5,1.5Hz,1H),4.59(d,J=9.2Hz,2H),4.28(d,J=9.1Hz,2H),3.69(s,2H),3.16(d,J=6.8Hz,2H),2.20-2.10(m,1H),1.04(d,J=6.7Hz,6H).
实施例55:2-(3-(4-(5-氯-1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑-1-基)-1-(乙基磺酰基)氮杂环丁烷-3-基)乙腈盐酸盐(50-HCl)
采用与实施例46中类似的操作,由实施例50中合成的化合物(50)(50mg)制备化合物(50)的盐酸盐(50-HCl)(55mg,白色固体)。MS m/z:
405[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.97(s,1H),8.66(s,1H),8.29(s,1H),8.19(s,1H),7.63–7.57(m,1H),6.64(dd,J=3.5,1.9Hz,1H),4.58(d,J=9.1Hz,2H),4.25(d,J=9.1Hz,2H),3.69(s,2H),3.25(q,J=7.3Hz,2H),1.25(t,J=7.3Hz,3H).
实施例56:2-(1-(乙基磺酰基)-3-(3-甲基-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-1-基)氮杂环丁烷-3-基)乙腈(56)
第一步:4-(4-甲基-1-(三异丙基甲硅烷基)-1H-吡咯-3-基)-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(56b)
在室温下,用二氧六环(4mL)溶解4-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(56a)(150mg,0.53mmol)和3-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-1-(三异丙基甲硅烷基)-1H-吡咯(289mg,0.80mmol),再加入水(0.5mL)和七水磷酸钾(448mg),用氮气置换,在室温下搅拌10min。在氮气保护下加入Pd(dppf)Cl2(50mg)。将反应体系移至90℃油浴锅中搅拌反应过夜。TLC检测底物消失后,将反应液缓慢倒入冰水中淬灭,用EA萃取,无水硫酸钠干燥有机相。通过硅胶制备板(PE:EA=10:1)纯化,得到化合物(56b)(190mg,油状物),收率:74%。MS m/z:485[M+1]+。
第二步:4-(4-甲基-1H-吡咯-3-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基-7H-吡咯并[2,3-d]嘧啶(56c)
用四氢呋喃(2mL)溶解化合物(56b)(190mg),加入四丁基氟化胺,在室
温下搅拌30min。TLC监测(PE:EA=6:1)底物消失后,将反应体系倒入水中淬灭,用EA萃取产物,合并有机相,无水硫酸钠干燥,过滤,浓缩,通过硅胶制备板(PE:EA=3:1)纯化,得到化合物(56c)(128mg,淡黄色固体),收率:99%。MS m/z:329[M+1]+。
第三步:2-(1-(乙基磺酰基)-3-(3-甲基-4-(7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-1-基)氮杂环丁烷-3-基)乙腈(56d)
在室温下,向化合物(56c)(108mg,0.33mmol)和2-[1-(乙基磺酰基)氮杂环丁烷-3-亚基]乙腈(92mg)中加入乙腈(5mL),反应液浑浊。然后加入DBU(150mg),在室温下搅拌过夜。将反应液浓缩,加入DCM溶解,通过硅胶制备板(PE:EA=1:1)纯化,得到化合物(56d)(88mg,黄色固体),收率:53%。MS m/z:515[M+1]+。
第四步:2-(1-(乙基磺酰基)-3-(3-甲基-4-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯-1-基)氮杂环丁烷-3-基)乙腈(56)
向化合物(56d)(88mg,0.17mmol)加入DCM(3mL)和三氟化硼乙醚(80mg),在室温下搅拌2h。LC-MS检测底物消失后,将溶剂旋转蒸发至干燥,得到油状物,再加入MeOH(3mg)使其完全溶解,接着滴入1mol/L的NaOH水溶液,调节其pH值至10左右,在室温下搅拌4h。LC-MS检测反应完成后,将反应液减压浓缩移除大部分MeOH,加水分散析出固体,过滤得到固体,通过反相制备色谱法纯化,得到化合物(56)(20mg,白色固体)。收率31%。MS m/z:385[M+1]+。
1H NMR(400MHz,DMSO-d6)δ11.98(s,1H),8.68(s,1H),7.70(d,J=2.4Hz,1H),7.51(dd,J=3.6,2.2Hz,1H),6.95(dd,J=2.5,1.2Hz,1H),6.89(dd,J=3.6,1.6Hz,1H),4.44(d,J=9.2Hz,2H),4.19(d,J=9.0Hz,2H),3.55(s,2H),3.22(q,J=7.3Hz,2H),2.38(s,3H),1.24(t,J=7.3Hz,3H).
下列化合物参考上述实施例相应制备方法制备:
生物学测试
试验例1:JAK1、JAK2、JAK3和TYK2酶活抑制检测
1)试验材料
JAK1、JAK2、JAK3和TYK2酶:购自Life Technologies;
底物GFP-STAT1:购自Life Technologies;
2)试验方法
用检测缓冲液(40mM Tris-HCl pH 7.4,20mM MgC12,0.1%BSA)将待测化合物、酶、底物和ATP稀释至所需浓度,将前三者加入多孔板中,混合均匀后在室温下孵育。加入ATP启动激酶反应,在室温下孵育。加入ADP-Glo试剂,在室温下孵育。加入激酶检测试剂,在室温下孵育。检测各试验组发光强度,计算半数抑制浓度IC50值。结果如表1-1和表1-2所示。
对于TYK2酶,加入Tb-anti-pSTAT1,在室温下孵育,使用LanthaScreen的方法检测TR-FRET,计算半数抑制浓度IC50值。结果如表1-3所示。
表1-1:实施例的化合物对JAK1和JAK2酶活抑制浓度IC50值
由表1-1可见,本发明的化合物对于JAK1和JAK2具有很好的抑制作用。
表1-2:实施例的化合物对JAK3酶活抑制浓度IC50值
由表1-1和1-2可见,本发明的化合物对JAK1和JAK2的抑制性明显强于对JAK3的抑制性,本发明的化合物对JAK1/2激酶具有良好的选择性。
表1-3:实施例的化合物对TYK2酶活抑制浓度IC50值
由表1-1和1-3可见,本发明的化合物对JAK1和JAK2的抑制性明显强于对TYK2的抑制性,本发明的化合物对JAK1/2激酶具有良好的选择性。
试验例2:安全性试验
采用PredictorTMhERG Fluorescence Polarization Assay,检测待测化合物对hERG钾离子通道的作用,测试浓度为3、10和30μM。试验结果如表2-1中所示:
表2-1:hERG实验结果:
化合物 | IC50(μM) |
5 | >30 |
8 | >30 |
14 | >30 |
17 | >30 |
19 | >30 |
23 | >30 |
25 | >30 |
26 | >30 |
30 | >30 |
31 | >30 |
12-2 | >30 |
43 | >30 |
1 | >30 |
2 | >30 |
13 | >30 |
35 | >30 |
45-1 | >30 |
45-2 | >30 |
39 | >30 |
48 | >30 |
49 | >30 |
50 | >30 |
52 | >30 |
50-HCl | >30 |
52-HCl | >30 |
由表2-1的试验数据可知,在该项试验中,本发明的化合物对于hERG的50%抑制浓度(IC50)均大于30μM。因此,本发明的化合物对hERG无抑制作用,无导致心脏QT间期延长的安全性隐患。
CYP450是药物代谢中最重要的酶系统,参与代谢的酶与药物相互作用,其中最主要的是CYP1A2、CYP2D6和CYP3A4。在CYP450酶测试中,采用P450-GloTMCYP1A2筛选系统测定化合物对CYP1A2和CYP3A4酶的抑制活性。采用CYP2D6Cyan筛选试剂盒测定化合物对CYP2D6酶的抑制活性。测试结果如表2-2中所示:
表2-2:测试结果
由表2-2的数据可知,在该项试验中,所测试的化合物对于CYP1A2、CYP3A4和CYP2D6的50%抑制浓度(IC50)均大于10μM。因此,本发明的化合物对CYP1A2、CYP2D6和CYP3A4无明显抑制作用,不会引起由于对所述酶的抑制导致不同药物之间代谢相互影响而产生的安全性问题。
试验例3:药代谢动力学研究
大鼠:
通过静脉(IV)和灌胃(PO)途径向雄性SD大鼠给药待测化合物,考察待测化合物的药物代谢动力学特点。IV的给药剂量是1mg/kg,PO的给药剂量分别是0.5-5mg/kg。溶媒系统为10%DMSO:10%solutol:80%生理盐水或者5%DMSO:5%solutol:90%生理盐水或者10%DMSO:60%PEG:
30%生理盐水。在给药后的不同时间点收集血液。将血浆样品经乙腈沉淀蛋白处理后进行LC-MS/MS分析。
静脉(IV)给药结果见表3-1、3-2。
表3-1:向大鼠IV给药剂量1mg/kg后的化合物暴露量(AUClast)
AUClast:所有时间点(0-24小时)的曲线下面积。
表3-2:向大鼠IV给药剂量1mg/kg后的化合物半衰期(T1/2)
结果表明:相对于Baricitinib,在1mg/kg给药剂量、IV给药途径下,本发明的化合物暴露量(AUClast)更好,半衰期(T1/2)更长,所以本发明的化合物具有明显的药物代谢动力学优势。
向大鼠灌胃(PO)给药后的结果见表4-1。
表4-1:向大鼠灌胃给药剂量5mg/kg后的药代动力学参数
AUClast:所有时间点(0-24小时)的曲线下面积。
结果表明:相对于Baricitinib,在5mg/kg给药剂量、PO给药途径下,本发明的化合物暴露量(AUClast)更好。在0.5-5mg/kg给药剂量、PO给药途径下,本发明的化合物的生物利用度(F%)大于70%,半衰期(T1/2)为2-4小时,明显优于Baricitinib(相同条件下,其F%为55%,T1/2为1.58小时)。所以本发明的化合物具有明显的药物代谢动力学优势。
恒河猴:
通过静脉(IV)向雄性恒河猴给药待测化合物,考察待测化合物的药代动力学特点。IV给药剂量是0.5mg/kg。在IV给药后的不同时间点收集血液,将血浆样品经乙腈沉淀蛋白处理后进行LC-MS/MS分析。应用WinNonlin6.3软件,采用非房室模型计算药代动力学参数。
结果表明:在0.5mg/kg给药剂量、IV给药途径下,本发明的化合物8的半衰期为5.29h,Baricitinib的半衰期为1.38h,化合物8半衰期更长,所以本发明的化合物8具有明显的药物代谢动力学优势。本发明的其它化合物具有类似的优势。
通过灌胃(PO)向雄性恒河猴给药待测化合物,考察待测化合物的药代动力学特点。PO的给药剂量是1mg/kg。PO给药后不同时间点收集血液。将血浆样品经乙腈沉淀蛋白处理后进行LC-MS/MS分析。应用WinNonlin6.3软件,采用非房室模型计算药代动力学参数。
结果如下所示:
AUClast:所有时间点(0-24小时)的曲线下面积。
结果表明:相对于Baricitinib,在1mg/kg给药剂量、PO给药途径下,本发明的化合物(例如化合物8)暴露量(AUClast)更好,半衰期(T1/2)更长,口服生物利用度(F%)更高,所以本发明的化合物(例如化合物8)具有明显的药物代谢动力学优势。
试验例4.在大鼠胶原诱导关节炎(CIA)模型中的体内药效
向Lewis大鼠的背部及尾根部3点皮内注射牛II型胶原和不完全弗氏佐剂1:1混合乳剂。14天后测量大鼠足厚度、足体积,并进行关节炎评分。给药前为D0,按足体积分组,灌胃给予待测化合物,溶媒为5%DMSO+5%solutol,每天给药1次,给药两周,测量大鼠足厚度、足体积,并进行关节炎评分。试验结果如下表5-1、5-2和5-3中所示。
表5-1:足厚度变化
由上表可见,随着给药天数的增加,在同等剂量的化合物8和Baricitinib下,化合物8使得大鼠足厚度的减小明显优于Baricitinib。
表5-2:足体积变化
由上表可见,随着给药天数的增加,在同等剂量的化合物8和Baricitinib下,化合物8使得大鼠足体积的减小明显优于Baricitinib。
表5-3:关节炎评分
由上表可见,随着给药天数的增加,在同等剂量的化合物8和Baricitinib下,化合物8使得平均关节炎评分减小明显优于Baricitinib(平均关节炎评分越低,表明化合物越能有效的改善模型动物的关节炎症状)。
综上,试验结果表明,与溶媒组相比,10mg/kg剂量下的本发明的化合物能有效的改善模型动物的症状及评分;在同等剂量下,本发明的化合物8较Baricitinib(对照)改善更明显。本发明的其他化合物具有类似的结果。
制剂实施例
制剂实施例1.片剂
作为口服药物组合物的具体实施方式,制备包含以下成分的片剂。
成分:
首先,将化合物5、微晶纤维素和交联羧甲基纤维素钠混合,然后用硬脂酸镁将混合物润滑并压为片剂。
制剂实施例2.胶囊剂
制备含有活性成分化合物8的颗粒填充的胶囊剂。
成分:
使化合物8、乳糖通过60筛目的筛进行筛分。使玉米淀粉通过120筛目的筛进行筛分。将它们混合,在混合粉末中添加HPC-L溶液,进行捏合、造粒、干燥。将所得的干燥颗粒整粒后,将其(150mg)填充到4号硬明胶胶囊中。
制剂实施例3.颗粒剂
制备含有以下成分的颗粒剂。
成分:
将化合物31和乳糖通过60网目的筛进行筛分。将玉米淀粉通过120网目的筛进行筛分。将它们利用V型混合机混合。在混合粉末中添加低粘度羟丙基纤维素水溶液,进行捏合、造粒(挤压造粒,孔径0.5~1mm)、干燥的步骤。将所得的干燥颗粒用振荡筛(12/60筛目)过筛,得到颗粒剂。
除本文中描述的那些外,根据前述描述,本发明的多种修改对本领域技术人员而言会是显而易见的。这样的修改也意图落入所附权利要求书的范围内。本申请中所引用的各参考文献(包括所有专利、专利申请、期刊文章、书籍及任何其它公开)均以其整体援引加入本文。
Claims (17)
- 式I的化合物,其药学上可接受的盐、立体异构体、多晶型物、溶剂合物、代谢物或前药:其中:R1选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基、C(O)R10和S(O)2R11;R2和R3各自独立地选自H、CN、卤素和C1-6烷基;R4和R5各自独立地选自H、卤素和CN;X选自N和CR6;Y选自N和CR9;Z选自N和CR7;W选自N和CR8;R6、R7、R8和R9各自独立地选自H、卤素、CN、C1-6烷基、C1-6烷氧基和C(O)NR12R13;R10和R11各自独立地选自C1-6烷基、C3-10环烷基、3-10元杂环基、C6-14芳基、5-14元杂芳基、C7-20芳烷基和NR12R13;R12和R13各自独立地选自H和C1-6烷基;其中上述烷基、环烷基、杂环基、芳基、杂芳基和芳烷基各自任选地被1、2或3个独立地选自卤素、CN和C1-4烷基的取代基取代。
- 药物组合物,其包含治疗有效量的权利要求1-7中任一项的化合物以及一种或多种药学上可接受的载体。
- 权利要求8的药物组合物,其中所述治疗有效量为约0.01mg至约1000mg,适合地是0.1-500mg,优选0.5-300mg,更优选1-150mg,特别优选1-50mg,例如1.5mg、2mg、4mg、10mg或25mg。
- 权利要求8或9的药物组合物,其进一步包含一种或多种治疗JAK相关性疾病的其它药物。
- 权利要求10的药物组合物,其中所述治疗JAK相关性疾病的其它药物为选自依法利珠单抗、霉酚酸钠、依那西普和甲氨蝶呤中的种多多种。
- 权利要求1-7中任一项的化合物或权利要求8-11中任一项的药物组合物在制备治疗JAK相关性疾病的药物中的用途。
- 权利要求12的用途,其中所述JAK相关性疾病选自炎性疾病、 自身免疫病或癌症,更特别地,所述疾病为类风湿性关节炎。
- 治疗JAK相关性疾病的方法,其包括向需要其的个体给药治疗有效量的权利要求1-7中任一项的化合物或权利要求8-11中任一项的药物组合物。
- 权利要求14的方法,其中所述JAK相关性疾病选自炎性疾病、自身免疫病和癌症,更特别地,所述疾病为类风湿性关节炎。
- 制备权利要求1-7中任一项的化合物的方法,所述方法包括:其中卤素选自氟、氯、溴和碘,R14和R15各自独立地选自H和C1-6烷基,或者R14和R15连同其所连接的原子共同构成5元或6元环系;其余各基团和取代基如权利要求1-7中任一项所定义;其中化合物a与SEMCl在碱的存在下,于极性非质子溶剂中反应,生成化合物b;化合物b在碱的存在下,在钯催化剂催化下,与适当的试剂反应,生成化合物c’;化合物c’在碱的存在下,在钯催化剂催化下,与适当的试剂反应,生成化合物d’;化合物d’在碱的存在下,与适当的试剂反应,生成化合物e’;化合物e’在路易斯酸催化下反应,得到式I的化合物;或者化合物e’首先在酸催化下反应,然后将得到的产物经处理后在碱存在下反应,得到式I的化合物。
Priority Applications (10)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2018518690A JP6860267B2 (ja) | 2015-12-11 | 2016-12-08 | アゼチジン誘導体、その調製方法、及びその使用 |
DK16872413.6T DK3360878T3 (da) | 2015-12-11 | 2016-12-08 | Azetidinderivat, fremstillingsfremgangsmåde derfor og anvendelse deraf |
US15/767,508 US10159662B2 (en) | 2015-12-11 | 2016-12-08 | Azetidine derivative, preparation method therefor, and use thereof |
EP16872413.6A EP3360878B9 (en) | 2015-12-11 | 2016-12-08 | Azetidine derivative, preparation method therefor, and use thereof |
CN201680059396.4A CN108137603B (zh) | 2015-12-11 | 2016-12-08 | 氮杂环丁烷衍生物、其制备方法及用途 |
ES16872413T ES2830446T3 (es) | 2015-12-11 | 2016-12-08 | Derivado de azetidina, método de preparación del mismo y uso del mismo |
HK18109969.8A HK1250707B (zh) | 2015-12-11 | 2018-08-02 | 氮雜環丁烷衍生物、其製備方法及用途 |
HK18110823.2A HK1251551A1 (zh) | 2015-12-11 | 2018-08-22 | 氮雜環丁烷衍生物、其製備方法及用途 |
US16/169,780 US20190060291A1 (en) | 2015-12-11 | 2018-10-24 | Azetidine derivative, preparation method therefor, and use thereof |
US16/745,206 US11045455B2 (en) | 2015-12-11 | 2020-01-16 | Azetidine derivative, preparation method therefor, and use thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201510924946.1 | 2015-12-11 | ||
CN201510924946 | 2015-12-11 |
Related Child Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US15/767,508 A-371-Of-International US10159662B2 (en) | 2015-12-11 | 2016-12-08 | Azetidine derivative, preparation method therefor, and use thereof |
US16/169,780 Division US20190060291A1 (en) | 2015-12-11 | 2018-10-24 | Azetidine derivative, preparation method therefor, and use thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2017097224A1 true WO2017097224A1 (zh) | 2017-06-15 |
Family
ID=59012693
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2016/109036 WO2017097224A1 (zh) | 2015-12-11 | 2016-12-08 | 氮杂环丁烷衍生物、其制备方法及用途 |
Country Status (8)
Country | Link |
---|---|
US (3) | US10159662B2 (zh) |
EP (1) | EP3360878B9 (zh) |
JP (1) | JP6860267B2 (zh) |
CN (1) | CN108137603B (zh) |
DK (1) | DK3360878T3 (zh) |
ES (1) | ES2830446T3 (zh) |
HK (2) | HK1250707B (zh) |
WO (1) | WO2017097224A1 (zh) |
Cited By (19)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2018133875A1 (zh) * | 2017-01-23 | 2018-07-26 | 上海长森药业有限公司 | Jak酶抑制剂及其制备方法和用途 |
WO2018223859A1 (zh) * | 2017-06-07 | 2018-12-13 | 四川科伦博泰生物医药股份有限公司 | 氮杂环丁烷衍生物的固体形式及其制备方法和用途 |
WO2019090158A1 (en) * | 2017-11-03 | 2019-05-09 | Aclaris Therapeutics, Inc. | Substituted pyrrolopyrimidine jak inhibitors and methods of making and using the same |
CN109867675A (zh) * | 2017-12-01 | 2019-06-11 | 北京普祺医药科技有限公司 | 一种吡咯并嘧啶衍生的化合物、药物组合物以及其用途 |
CN110028432A (zh) * | 2019-05-22 | 2019-07-19 | 南京合巨药业有限公司 | 一种(3-氨基-氮杂环丁烷-1-基)-环丙基-甲酮的制备方法 |
JP2019156839A (ja) * | 2018-03-12 | 2019-09-19 | アッヴィ・インコーポレイテッド | チロシンキナーゼ2媒介性シグナル伝達の阻害剤 |
CN111320633A (zh) * | 2018-12-14 | 2020-06-23 | 中国医药研究开发中心有限公司 | 吡咯/咪唑并六元杂芳环类化合物及其制备方法和医药用途 |
WO2020173364A1 (zh) * | 2019-02-27 | 2020-09-03 | 四川科伦博泰生物医药股份有限公司 | 以氮杂环丁烷衍生物为活性成分的口服药物组合物、其制备方法及用途 |
WO2020188015A1 (en) | 2019-03-21 | 2020-09-24 | Onxeo | A dbait molecule in combination with kinase inhibitor for the treatment of cancer |
US10800775B2 (en) | 2017-11-03 | 2020-10-13 | Aclaris Therapeutics, Inc. | Pyrazolyl pyrrolo[2,3-b]pyrmidine-5-carboxylate analogs and methods of making the same |
WO2021089791A1 (en) | 2019-11-08 | 2021-05-14 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Methods for the treatment of cancers that have acquired resistance to kinase inhibitors |
US11021482B2 (en) | 2018-08-10 | 2021-06-01 | Adaris Therapeutics, Inc. | Pyrrolopyrimidine ITK inhibitors |
WO2021148581A1 (en) | 2020-01-22 | 2021-07-29 | Onxeo | Novel dbait molecule and its use |
CN114174294A (zh) * | 2019-05-28 | 2022-03-11 | 人类制药有限公司 | 用于抑制janus激酶1的新化合物 |
US11420966B2 (en) | 2019-05-02 | 2022-08-23 | Aclaris Therapeutics, Inc. | Substituted pyrrolopyridines as JAK inhibitors |
CN116444529A (zh) * | 2023-06-16 | 2023-07-18 | 北京科翔中升医药科技有限公司 | 一种氘代氮杂环丁烷类jak抑制剂及其用途 |
WO2023174314A1 (zh) | 2022-03-18 | 2023-09-21 | 四川科伦博泰生物医药股份有限公司 | 氮杂环丁烷衍生物的用途及治疗方法 |
WO2023226822A1 (zh) * | 2022-05-26 | 2023-11-30 | 四川科伦博泰生物医药股份有限公司 | 氮杂环丁烷衍生物治疗病毒感染的方法及用途 |
CN114174294B (zh) * | 2019-05-28 | 2024-10-25 | 人类制药有限公司 | 用于抑制janus激酶1的新化合物 |
Families Citing this family (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP3360878B9 (en) * | 2015-12-11 | 2021-05-05 | Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. | Azetidine derivative, preparation method therefor, and use thereof |
CN111518096B (zh) * | 2019-02-02 | 2022-02-11 | 江苏威凯尔医药科技有限公司 | 两面神激酶jak家族抑制剂及其制备和应用 |
WO2024148184A1 (en) * | 2023-01-04 | 2024-07-11 | Aerie Pharmaceuticals, Inc. | Compounds and processes for the preparation of jak inhibitors |
WO2024153147A1 (zh) * | 2023-01-18 | 2024-07-25 | 北京普祺医药科技股份有限公司 | 一种氮杂环烷烃化合物、药物组合物及其用途 |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2010039939A1 (en) * | 2008-10-02 | 2010-04-08 | Incyte Corporation | Janus kinase inhibitors for treatment of dry eye and other eye related diseases |
CN102026999A (zh) | 2008-03-11 | 2011-04-20 | 因塞特公司 | 作为jak抑制剂的氮杂环丁烷和环丁烷衍生物 |
WO2013173506A2 (en) * | 2012-05-16 | 2013-11-21 | Rigel Pharmaceuticals, Inc. | Method of treating muscular degradation |
RU2601410C1 (ru) * | 2015-11-13 | 2016-11-10 | ЗАО "Р-Фарм" | {3-[(7H-ПИРРОЛО[2,3-d]ПИРИМИДИН-4-ИЛ)АЗОЛИЛ]АЗЕТИДИН-3-ИЛ}АЦЕТОНИТРИЛЫ В КАЧЕСТВЕ ИНГИБИТОРОВ ЯНУС КИНАЗ |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2354140A1 (en) | 2005-05-20 | 2011-08-10 | Vertex Pharmaceuticals Incorporated | Pyrrolopyridines useful as inhibitors of protein kinase |
AU2011224484A1 (en) | 2010-03-10 | 2012-09-27 | Incyte Holdings Corporation | Piperidin-4-yl azetidine derivatives as JAK1 inhibitors |
AR086983A1 (es) | 2011-06-20 | 2014-02-05 | Incyte Corp | Derivados de azetidinil fenil, piridil o pirazinil carboxamida como inhibidores de jak |
PL3129021T3 (pl) | 2014-04-08 | 2021-05-31 | Incyte Corporation | Leczenie nowotworów złośliwych z komórek b za pomocą skojarzenia inhibitora jak i pi3k |
TWI748941B (zh) | 2015-02-27 | 2021-12-11 | 美商英塞特公司 | Pi3k抑制劑之鹽及製備方法 |
EP3360878B9 (en) * | 2015-12-11 | 2021-05-05 | Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. | Azetidine derivative, preparation method therefor, and use thereof |
-
2016
- 2016-12-08 EP EP16872413.6A patent/EP3360878B9/en active Active
- 2016-12-08 JP JP2018518690A patent/JP6860267B2/ja active Active
- 2016-12-08 CN CN201680059396.4A patent/CN108137603B/zh active Active
- 2016-12-08 DK DK16872413.6T patent/DK3360878T3/da active
- 2016-12-08 US US15/767,508 patent/US10159662B2/en active Active
- 2016-12-08 ES ES16872413T patent/ES2830446T3/es active Active
- 2016-12-08 WO PCT/CN2016/109036 patent/WO2017097224A1/zh active Application Filing
-
2018
- 2018-08-02 HK HK18109969.8A patent/HK1250707B/zh unknown
- 2018-08-22 HK HK18110823.2A patent/HK1251551A1/zh unknown
- 2018-10-24 US US16/169,780 patent/US20190060291A1/en not_active Abandoned
-
2020
- 2020-01-16 US US16/745,206 patent/US11045455B2/en active Active
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102026999A (zh) | 2008-03-11 | 2011-04-20 | 因塞特公司 | 作为jak抑制剂的氮杂环丁烷和环丁烷衍生物 |
WO2010039939A1 (en) * | 2008-10-02 | 2010-04-08 | Incyte Corporation | Janus kinase inhibitors for treatment of dry eye and other eye related diseases |
WO2013173506A2 (en) * | 2012-05-16 | 2013-11-21 | Rigel Pharmaceuticals, Inc. | Method of treating muscular degradation |
RU2601410C1 (ru) * | 2015-11-13 | 2016-11-10 | ЗАО "Р-Фарм" | {3-[(7H-ПИРРОЛО[2,3-d]ПИРИМИДИН-4-ИЛ)АЗОЛИЛ]АЗЕТИДИН-3-ИЛ}АЦЕТОНИТРИЛЫ В КАЧЕСТВЕ ИНГИБИТОРОВ ЯНУС КИНАЗ |
Non-Patent Citations (4)
Title |
---|
JOEL M.K. ET AL., ARTHRITIS RHEUM., vol. 60, 2009, pages 1859 - 1905 |
KREMER, J. ET AL.: "The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo-controlled phase Ila trial of three dosage levels of CP-690, 550 versus placebo", ARTHRITIS & RHEUMATISM,, vol. 60, no. 7, 2009, pages 1895 - 1905 |
PETER NORMAN: "Selective JAK inhibitors in development for rheumatoid arthritis", EXPERT OPIN. INVESTIG. DRUGS, vol. 23, no. 8, 2014, pages 1067 - 77 |
RODIG S. ET AL.: "Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses", CELL, vol. 93, no. 3, 1998, pages 373 - 83 |
Cited By (30)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108341820B (zh) * | 2017-01-23 | 2020-10-16 | 上海长森药业有限公司 | Jak酶抑制剂及其制备方法和用途 |
CN108341820A (zh) * | 2017-01-23 | 2018-07-31 | 上海长森药业有限公司 | Jak酶抑制剂及其制备方法和用途 |
US11524961B2 (en) | 2017-01-23 | 2022-12-13 | Shanghai Longwood Biopharmaceuticals Co., Ltd. | JAK kinase inhibitor and preparation method and use thereof |
WO2018133875A1 (zh) * | 2017-01-23 | 2018-07-26 | 上海长森药业有限公司 | Jak酶抑制剂及其制备方法和用途 |
AU2018209579B2 (en) * | 2017-01-23 | 2020-12-24 | Shanghai Longwood Biopharmaceuticals Co., Ltd. | JAK kinase inhibitor and preparation method and use thereof |
WO2018223859A1 (zh) * | 2017-06-07 | 2018-12-13 | 四川科伦博泰生物医药股份有限公司 | 氮杂环丁烷衍生物的固体形式及其制备方法和用途 |
US10981906B2 (en) | 2017-11-03 | 2021-04-20 | Aclaris Therapeutics, Inc. | Substituted pyrrolopyridine JAK inhibitors and methods of making and using the same |
US10800775B2 (en) | 2017-11-03 | 2020-10-13 | Aclaris Therapeutics, Inc. | Pyrazolyl pyrrolo[2,3-b]pyrmidine-5-carboxylate analogs and methods of making the same |
WO2019090158A1 (en) * | 2017-11-03 | 2019-05-09 | Aclaris Therapeutics, Inc. | Substituted pyrrolopyrimidine jak inhibitors and methods of making and using the same |
US11739086B2 (en) | 2017-11-03 | 2023-08-29 | Aclaris Therapeutics, Inc. | Substituted pyrrolopyridine JAK inhibitors and methods of making and using the same |
CN109867675B (zh) * | 2017-12-01 | 2021-01-19 | 北京普祺医药科技有限公司 | 一种吡咯并嘧啶衍生的化合物、药物组合物以及其用途 |
CN109867675A (zh) * | 2017-12-01 | 2019-06-11 | 北京普祺医药科技有限公司 | 一种吡咯并嘧啶衍生的化合物、药物组合物以及其用途 |
JP2019156839A (ja) * | 2018-03-12 | 2019-09-19 | アッヴィ・インコーポレイテッド | チロシンキナーゼ2媒介性シグナル伝達の阻害剤 |
US11021482B2 (en) | 2018-08-10 | 2021-06-01 | Adaris Therapeutics, Inc. | Pyrrolopyrimidine ITK inhibitors |
US11820775B2 (en) | 2018-08-10 | 2023-11-21 | Aclaris Therapeutics, Inc. | Pyrrolopyrimidine ITK inhibitors |
CN111320633A (zh) * | 2018-12-14 | 2020-06-23 | 中国医药研究开发中心有限公司 | 吡咯/咪唑并六元杂芳环类化合物及其制备方法和医药用途 |
CN113508114B (zh) * | 2019-02-27 | 2024-03-26 | 四川科伦博泰生物医药股份有限公司 | 以氮杂环丁烷衍生物为活性成分的口服药物组合物、其制备方法及用途 |
CN113508114A (zh) * | 2019-02-27 | 2021-10-15 | 四川科伦博泰生物医药股份有限公司 | 以氮杂环丁烷衍生物为活性成分的口服药物组合物、其制备方法及用途 |
WO2020173364A1 (zh) * | 2019-02-27 | 2020-09-03 | 四川科伦博泰生物医药股份有限公司 | 以氮杂环丁烷衍生物为活性成分的口服药物组合物、其制备方法及用途 |
WO2020188015A1 (en) | 2019-03-21 | 2020-09-24 | Onxeo | A dbait molecule in combination with kinase inhibitor for the treatment of cancer |
US11420966B2 (en) | 2019-05-02 | 2022-08-23 | Aclaris Therapeutics, Inc. | Substituted pyrrolopyridines as JAK inhibitors |
CN110028432A (zh) * | 2019-05-22 | 2019-07-19 | 南京合巨药业有限公司 | 一种(3-氨基-氮杂环丁烷-1-基)-环丙基-甲酮的制备方法 |
CN114174294A (zh) * | 2019-05-28 | 2022-03-11 | 人类制药有限公司 | 用于抑制janus激酶1的新化合物 |
CN114174294B (zh) * | 2019-05-28 | 2024-10-25 | 人类制药有限公司 | 用于抑制janus激酶1的新化合物 |
WO2021089791A1 (en) | 2019-11-08 | 2021-05-14 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Methods for the treatment of cancers that have acquired resistance to kinase inhibitors |
WO2021148581A1 (en) | 2020-01-22 | 2021-07-29 | Onxeo | Novel dbait molecule and its use |
WO2023174314A1 (zh) | 2022-03-18 | 2023-09-21 | 四川科伦博泰生物医药股份有限公司 | 氮杂环丁烷衍生物的用途及治疗方法 |
WO2023226822A1 (zh) * | 2022-05-26 | 2023-11-30 | 四川科伦博泰生物医药股份有限公司 | 氮杂环丁烷衍生物治疗病毒感染的方法及用途 |
CN116444529B (zh) * | 2023-06-16 | 2023-12-05 | 药康众拓(江苏)医药科技有限公司 | 一种氘代氮杂环丁烷类jak抑制剂及其用途 |
CN116444529A (zh) * | 2023-06-16 | 2023-07-18 | 北京科翔中升医药科技有限公司 | 一种氘代氮杂环丁烷类jak抑制剂及其用途 |
Also Published As
Publication number | Publication date |
---|---|
EP3360878A4 (en) | 2019-03-06 |
ES2830446T3 (es) | 2021-06-03 |
CN108137603B (zh) | 2019-10-18 |
CN108137603A (zh) | 2018-06-08 |
HK1251551A1 (zh) | 2019-02-01 |
HK1250707B (zh) | 2020-07-03 |
EP3360878A1 (en) | 2018-08-15 |
US20190060291A1 (en) | 2019-02-28 |
EP3360878B1 (en) | 2020-09-16 |
US20200147062A1 (en) | 2020-05-14 |
DK3360878T3 (da) | 2020-11-09 |
US11045455B2 (en) | 2021-06-29 |
JP6860267B2 (ja) | 2021-04-14 |
JP2018536634A (ja) | 2018-12-13 |
EP3360878B9 (en) | 2021-05-05 |
US20180289680A1 (en) | 2018-10-11 |
US10159662B2 (en) | 2018-12-25 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2017097224A1 (zh) | 氮杂环丁烷衍生物、其制备方法及用途 | |
KR101216055B1 (ko) | 야누스 키나아제 억제제로서의 헤테로아릴 치환된 피롤로[2,3-b]피리딘 및 피롤로[2,3-b]피리미딘 | |
WO2018166493A1 (zh) | 杂芳基并[4,3-c]嘧啶-5-胺类衍生物、其制备方法及其在医药上的应用 | |
CA3131293A1 (en) | Jak inhibitor compound and use thereof | |
KR101438293B1 (ko) | Jak의 매크로사이클릭 억제자 | |
JP2023538060A (ja) | 二環化合物、それを含む組成物、及びそれらの使用 | |
WO2021042890A1 (zh) | 杂环化合物及其作为Trk激酶抑制剂的应用 | |
WO2019228341A1 (zh) | 含三并环类衍生物抑制剂、其制备方法和应用 | |
KR20220140710A (ko) | 트리아졸로피리다진 유도체, 이의 제조 방법, 약물 조성물 및 용도 | |
CN114466850A (zh) | [1,2,4]三唑并[1,5-c]喹唑啉-5-胺 | |
WO2021249913A9 (en) | 2'-(quinolin-3-yl)-5',6'-dihydrospiro[azetidine-3,4'-pyrrolo[1,2-b]pyrazole]-1-carboxylate derivatives and related compounds as map4k1 (hpk1) inhibitors for the treatment of cancer | |
EP3526212B1 (en) | Substituted 6-(1h-pyrazol-1-yl)pyrimidin-4-amine derivatives and uses thereof | |
CN113966336B (zh) | 三环类化合物及其用途 | |
CN109535132B (zh) | 2-取代吡唑氨基-4-取代氨基-5-嘧啶甲酰胺类化合物、组合物及其应用 | |
TWI750855B (zh) | 嘧啶醯胺化合物及其用途 | |
CN114907407A (zh) | 细胞周期蛋白依赖性激酶7(cdk7)的抑制剂 | |
WO2021249417A1 (zh) | 杂环化合物及其衍生物 | |
CN112812105A (zh) | 一种氨基吡啶基氧基吡唑类衍生物及其制备方法和应用 | |
TW202313035A (zh) | Jak2抑制劑及其使用方法 | |
CN116249696A (zh) | 嘧啶酮类化合物及其用途 | |
CN116262739A (zh) | 含氮芳基衍生物调节剂、其制备方法和应用 | |
CN116745299A (zh) | 噻吩并嘧啶衍生物 | |
CN117136052A (zh) | Cdk抑制剂和其使用方法 | |
CN118525018A (zh) | 一种联炔类化合物及其应用 | |
CN113563341A (zh) | 作为Trk抑制剂的取代的吡唑并[1,5-a]嘧啶化合物 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 16872413 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 15767508 Country of ref document: US Ref document number: 2018518690 Country of ref document: JP |
|
NENP | Non-entry into the national phase |
Ref country code: DE |