WO2022166642A1 - 含氮多环稠环类化合物,其药物组合物、制备方法和用途 - Google Patents

含氮多环稠环类化合物,其药物组合物、制备方法和用途 Download PDF

Info

Publication number
WO2022166642A1
WO2022166642A1 PCT/CN2022/073467 CN2022073467W WO2022166642A1 WO 2022166642 A1 WO2022166642 A1 WO 2022166642A1 CN 2022073467 W CN2022073467 W CN 2022073467W WO 2022166642 A1 WO2022166642 A1 WO 2022166642A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
membered
pyrazolo
mmol
pyridin
Prior art date
Application number
PCT/CN2022/073467
Other languages
English (en)
French (fr)
Inventor
钟俊
朱永宽
刘永波
陈肖虎
刘利彬
陈民春
王皓
高勤
Original Assignee
北京志健金瑞生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 北京志健金瑞生物医药科技有限公司 filed Critical 北京志健金瑞生物医药科技有限公司
Priority to JP2023547651A priority Critical patent/JP2024505711A/ja
Priority to EP22748921.8A priority patent/EP4289428A1/en
Priority to KR1020237028014A priority patent/KR20230136766A/ko
Publication of WO2022166642A1 publication Critical patent/WO2022166642A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic 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
    • C07D471/02Heterocyclic 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
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/439Heterocyclic 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 the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic 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
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic 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
    • C07D487/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to the field of medicinal chemistry, in particular to nitrogen-containing polycyclic fused-ring compounds, pharmaceutical compositions, preparation methods and uses thereof.
  • KIF5B-RET is the most common RET fusion gene in non-small cell lung cancer (Cancer, 2013, 119(8):1486-1494).
  • KIF5B-RET is a fusion gene formed by the chromosomal inversion (p11; q11) of the KIF5B (kinesin family member 5B) gene and the RET gene.
  • KIF5B-RET is low in lung cancer, is more common in non-smokers and adenocarcinoma patients, and is exclusive of other mutations such as EGFR, KRAS, BRAF, ErbB2, EML4-ALK (Genome Res, 2012 , 22(3):436-445).
  • the KIF5B-RET fusion protein contains the motor domain and the coiled-coil domain of KIF5B.
  • the RET tyrosine kinase activity of the fusion protein can be abnormally activated, thereby promoting lung tumorigenesis (Cancer, 2007). 2011, 117(12):2709-2718).
  • KIF5B-RET fusion kinase was confirmed to have significant oncogenic activity both in vitro and in vivo, and the signal transduction pathway of STAT3 may be the main downstream mediator of tumorigenesis .
  • KIF5B-RET regulates the sustained activation of STAT3.
  • KIF5B-RET fusion kinase can bind STAT3 to directly phosphorylate and activate STAT3-Tyr705; it can also mediate activation of STAT3-Tyr705 through JAK/STAT3-dependent pathway and trigger phosphorylation of Ser727 through RAS/RAF/MEK/ERK1 pathway change.
  • RET kinase signaling plays an important role in various human cancers such as thyroid cancer.
  • the RET804V mutation in the gatekeeper residue of RET is an important cause of tumor resistance to currently approved non-selective RET inhibitors such as cabozantinib and vandetanib.
  • One of the important mutations in the extracellular or intracellular domains of RET in isolated familial medullary thyroid carcinoma, namely the V804M mutation in the gatekeeper residue in the ATP-binding site of the kinase results in decreased affinity of existing drugs for the ATP-binding site .
  • the present invention provides a compound represented by the following formula I, its stereoisomer, racemate, tautomer, isotopic label, nitrogen oxide or pharmaceutically acceptable salt:
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , and X 7 are the same or different, and are independently selected from CR 1 or N;
  • X 8 is selected from CR 1 R 1′ or NR 1 ;
  • each R 1 and R 1' are the same or different, independently selected from each other H, halogen, CN, OH, unsubstituted or optionally substituted with one, two or more substituents independently selected from R a
  • A is selected from H, halogen, CN, OH, NH 2 , unsubstituted or optionally substituted with one, two or more substituents independently selected from R b : C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 1-40 alkyloxy, C 2-40 Alkenyloxy, C 2-40 alkynyloxy, C 3-40 cycloalkyloxy, C 3-40 cycloalkenyloxy, C 3-40 cycloalkynyloxy, NR 2 R 3 , - C(O)R 4 , -OCR 5 , -S(O) 2 R 6 , OS(O) 2 R 7 ;
  • D, E are the same or different, and are independently selected from H, halogen, CN, OH, B(OH) 2 , -C(O)R 4 , -OCR 5 , -S(O) 2 R 6 , OS(O ) 2 R 7 , -OR 21 , C(O)OR 22 , -P(O)R 23 R 24 , unsubstituted or optionally substituted by one, two or more substituents independently selected from R c
  • R 21 , R 22 , R 23 , R 24 are the same or different, independently selected from each other from H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R d of the following groups: C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl base, 5-20-membered heteroaryl, 3-20-membered heterocyclic group;
  • G is selected from halogen, unsubstituted or optionally substituted with one, two or more substituents independently selected from R e : 5-20 membered heteroaryl with at least one heteroatom selected from N, at least One heteroatom is selected from 3-20-membered heterocyclic group of N, C 3-40 cycloalkyl-NH-;
  • K is selected from absent, H, halogen, CN, OH, unsubstituted or optionally substituted with one, two or more substituents independently selected from R f of the following groups: C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered heteroaryl base, 3-20 membered heterocyclyl, C 1-40 alkyloxy, C 2-40 alkenyloxy, C 2-40 alkynyloxy, C 3-40 cycloalkyloxy, C 3- 40 cycloalkenyloxy, C 3-40 cycloalkynyloxy, C 6-20 aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, C 6-20 Aryl C 1-40 alkyl, 5-20-membered heteroaryl C 1-40 alkyl, 3
  • Each R 2 is the same or different, independently selected from the following groups of H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R f : C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered hetero Aryl, 3-20 membered heterocyclic group, -C(O)R 4 , -S(O) 2 R 6 ;
  • Each R is the same or different and independently selected from the following groups of H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R: C 1-40 alkyl , C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered hetero Aryl, 3-20 membered heterocyclic group, -C(O)R 4 , -S(O) 2 R 6 ;
  • R and R together with the N atom to which they are attached form the following groups unsubstituted or optionally substituted with one, two or more substituents independently selected from R f : 5-20 membered heteroaryl or 3-20 membered heterocyclyl;
  • Each R is the same or different, independently selected from the following groups of H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R: C 1-40 alkyl , C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered hetero Aryl, 3-20 membered heterocyclyl, C 1-40 alkyloxy, C 2-40 alkenyloxy, C 2-40 alkynyloxy, C 3-40 cycloalkyloxy, C 3 -40 cycloalkenyloxy, C 3-40 cycloalkynyloxy, C 6-20 aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, C 6- 20 -aryl C 1-40 alkyl, 5-20-membered heteroaryl C 1-40 alkyl, 3-20-membere
  • Each R 5 is the same or different, independently selected from the following groups of H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R f : C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered hetero Aryl, 3-20 membered heterocyclyl, C 1-40 alkylcarbonyl, C 2-40 alkenylcarbonyl, C 2-40 alkynylcarbonyl, C 3-40 cycloalkylcarbonyl, C 3-40 cycloalkene carbonyl, C 3-40 cycloalkynylcarbonyl, C 6-20 arylcarbonyl, 5-20-membered heteroarylcarbonyl, 3-20-membered heterocyclylcarbonyl;
  • Each R is the same or different, independently selected from the following groups of H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R: C 1-40 alkyl , C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered hetero Aryl, 3-20 membered heterocyclyl, C 1-40 alkyloxy, C 2-40 alkenyloxy, C 2-40 alkynyloxy, C 3-40 cycloalkyloxy, C 3 -40 cycloalkenyloxy, C 3-40 cycloalkynyloxy, C 6-20 aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, C 6- 20 -aryl C 1-40 alkyl, 5-20-membered heteroaryl C 1-40 alkyl, 3-20-membere
  • Each R 7 is the same or different and is independently selected from the following groups of H, unsubstituted or optionally substituted with one, two or more substituents independently selected from R f : C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20 membered hetero Aryl, 3-20 membered heterocyclic group;
  • n 0, 1, 2, 3, 4, 5, 6, 7 or 8;
  • the following groups are selected substituted by one, two or more substituents independently selected from R g : C 1-40 alkyl, C 2-40 alkenyl, C 2-40 alkynyl, C 3-40 ring Alkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, C 6-20 aryl, 5-20-membered heteroaryl, 3-20-membered heterocyclyl, C 1-40 alkyloxy , C 2-40 alkenyloxy, C 2-40 alkynyloxy, C 3-40 cycloalkyloxy, C 3-40 cycloalkenyloxy, C 3-40 cycloalkynyloxy, C 6-20 -membered aryloxy
  • a cyclic group including but not limited to C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cycloalkynyl, 3-20 membered heterocyclyl, etc.
  • two of the substituents may also form a bridged ring with the cyclic group, wherein the bridge atoms other than the bridgehead atom in the bridged ring may contain 1, 2 , 3, 4 or 5 divalent groups selected from CH 2 , O, NH;
  • two of the substituents may also be unsubstituted or optionally substituted by one, two One or more substituents independently selected from R f of the following groups: unsubstituted or optionally substituted with one, two or more substituents independently selected from R f cyclic groups (including but not Limited to the following groups unsubstituted or optionally substituted with one, two or more substituents independently selected from R f : C 3-40 cycloalkyl, C 3-40 cycloalkenyl, C 3-40 cyclo alkynyl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, etc.).
  • the two substituents Rc may be taken together with the nitrogen atom to which they are commonly linked to form unsubstituted or optionally be independently selected from one, two or more 5-20 membered heteroaryl or 3-20 membered heterocyclyl substituted with substituents of R, eg, unsubstituted or 5, 6 or 7 optionally substituted with one, two or more independently selected from R membered heteroaryl or 3, 4, 5, 6 or 7 membered heterocyclyl which is unsubstituted or optionally substituted with one, two or more independently selected from R;
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 are the same or different, and are independently selected from CR 1 or N; for example, X 1 , X 2 , X 3 , at least one of X 4 , X 5 , X 6 , and X 7 is N, for example, 1, 2, 3, 4, 5, 6 or 7 are N;
  • X 8 is selected from CR 1 R 1′ or NR 1 ;
  • each of R 1 and R 1' is the same or different and independently selected from the group consisting of H, halogen, CN, OH, C 1-6 alkyl, C 3-10 cycloalkyl, C 1- 6 alkyloxy;
  • A is selected from H, halogen, CN, OH, C 1-6 alkyl, C 1-6 alkyloxy;
  • D, E are the same or different, independently selected from H, halogen, CN, unsubstituted or NH optionally substituted with one, two or more substituents independently selected from Rc 2 or -OR 21 , provided that at least one of D and E is not H, for example at least one of D and E is selected from -OR 21 or unsubstituted or optionally by one, two or more independently selected from NH 2 substituted by the substituent of R c ;
  • R 21 is selected from C 1-6 alkyl which is unsubstituted or optionally substituted with one, two or more substituents of R d ;
  • R2 and R3 may together with the attached N atom form the following groups unsubstituted or optionally substituted with one, two or more substituents independently selected from Rf : 5 -20 membered heteroaryl or 3-20 membered heterocyclyl, such as 5, 6 or 7 membered heteroaryl or 3, 4, 5, 6 or 7 membered heterocyclyl;
  • each of R 01 , R 02 , R 03 , R 04 is the same or different, and is independently selected from H, C 1-6 alkyl, C 1-6 alkyloxy;
  • each Ra , Rb , Rc , Rd , Re , Rf is the same or different, independently selected from halogen, NH2 , CN, OH, unsubstituted or optionally by The following groups substituted by one, two or more substituents of R g : C 1-6 alkyl, C 1-6 alkyloxy, C 3-10 cycloalkyl, C 3-10 cycloalkyl Oxy group, C 2-6 alkynyloxy group, 3-8 membered heterocyclic group, 3-8 membered heteroaryl group;
  • each R g is the same or different and is independently selected from OH, halogen or C 3-10 cycloalkyl
  • G is selected from halogen, C 3-10 cycloalkyl, C 3-10 cycloalkyl-NH-, C 6-14 aryl, 5-14 membered heteroaryl, 3-12 membered Heterocyclyl, such as a 6-12 membered heterocyclyl having a monocyclic, bicyclic, tricyclic or bridged ring structure, which may contain 1, 2, 3 or 4 heteroatoms independently selected from N, O and S, provided that is wherein at least one heteroatom is selected from N, eg 1, 2 or 3 heteroatoms are selected from N.
  • K is selected from -C 1-6 alkyl-C 3-10 cycloalkyl, -C 1-6 alkyl-C 6-14 aryl, -C 1-6 alkyl-5 -14-membered heteroaryl, -C 1-6 alkyl-3-10-membered heterocyclyl, -C(O)NH 2 , -C(O)-C 3-10 cycloalkyl, -C(O) -C 6-14 aryl, -C(O)-5-14 membered heteroaryl, -C(O)-3-10 membered heterocyclyl, -C(O)-C 1-6 alkyl-C 3-10 cycloalkyl, -C(O)-C 1-6 alkyl-C 6-14 aryl, -C(O)-C 1-6 alkyl-5-14 membered heteroaryl, -C (O)-C 1-6 alkyl-3-10-membered heterocyclic group, wherein the C 3
  • K is selected from the following groups which are absent, H, OH, unsubstituted or optionally substituted with one, two or more independently selected from Rf : phenyl-C(O)-, phenyl-C (O)-NH-, Phenyl-C(O)-NH-C 1-6 Alkyl-, Phenyloxy-C(O)-NH-, Phenylalkyl-NH-C(O)- , phenylalkyl-C(O)-NH-, pyridyl-C(O)-, pyridyl-C(O)-NH-, pyridyl-C(O)-NH-C 1-6 alkyl -, Pyridyloxy-C(O)-NH-, Pyridylalkyl-NH-C(O)-, Pyridylalkyl-NH-C(O)-, Pyridylalkyl-C(O)-NH-, Pyrrolidinyl-C(
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 are the same or different, and are independently selected from CH or N; for example, X 1 , X 2 , X 3 , at least one of X 4 , X 5 , X 6 , and X 7 is N, for example, 1, 2, 3, 4, 5, 6 or 7 are N;
  • X 8 is selected from NR 1 ;
  • R 01 is methoxy
  • R 02 is H
  • R 03 is methyl, and R 04 is H;
  • R 1 is H
  • A is selected from H, NH 2 , methyl, ethyl, propyl, isopropyl;
  • E is selected from H, NH 2 ;
  • D is selected from the group consisting of halogen, BnO-, H, CN, NH 2 , OCH 3 , COOH, B(OH) 2 ,
  • G is selected from the following groups of F, unsubstituted or optionally substituted with one, two or more substituents independently selected from Re :
  • chemical bonds marked with wavy lines indicate attachment sites to other groups.
  • group G when there are two wavy lines in the group G, either of them can be attached to the ring where X 1 , X 2 , X 3 and X 4 are located, while the other is attached to the group K in the presence of the group K connect.
  • the group G when the ring-forming atoms of the group G include carbon atoms and nitrogen atoms, the group G can pass through one of the carbon atoms and nitrogen atoms (for example, the above-mentioned exemplary groups are marked with a wavy One of the ring-forming atoms of the wire) is connected to the ring in which X 1 , X 2 , X 3 and X 4 are located, and to the group K through the other of a carbon atom and a nitrogen atom.
  • K is selected from the following groups which are absent, H, OH, unsubstituted or optionally substituted with one, two or more substituents independently selected from R f : phenyl- C(O)-, Phenyl-C(O)-NH-, Phenyl-C(O)-NH-C 1-6 Alkyl-, Phenyloxy-C(O)-NH-, Phenyl Alkyl-NH-C(O)-, Phenylalkyl-C(O)-NH-, Pyridyl-C(O)-, Pyridyl-C(O)-NH-, Pyridyl-C(O )-NH-C 1-6 alkyl-, pyridyloxy-C(O)-NH-, pyridylalkyl-NH-C(O)-, pyridylalkyl-NH-C(O)-, pyridylalkyl-NH-C(O)-, pyridy
  • the compound represented by the formula I has the structure represented by the following formula II or III:
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , X 8 , R 01 , R 02 , R 03 , R 04 , A, D, E, G, K, m have Definitions set forth above.
  • the compound shown in the formula I has the structure shown in the following formula IV or V:
  • X 1 , X 2 , X 3 , X 4 , X 5 , R 01 , R 03 , R 04 , A, D, E, G, K have the above-mentioned definitions.
  • the compound is selected from the following compounds:
  • the present invention also provides the preparation method of the compound shown in formula I, comprising the steps:
  • A, D, E, Q, G, K, X 5 , X 6 , X 7 , X 8 have the above definitions; L 1 is selected from a leaving group.
  • the preparation method comprises the steps:
  • A, D, E, Q, G, K, X 5 , X 6 , X 7 , X 8 have the above-mentioned definitions; L 2 is selected from a leaving group.
  • the preparation method comprises the steps:
  • A, D, E, Q, G, K, X 5 , X 6 , X 7 , X 8 have the above-mentioned definitions; L 3 is selected from a leaving group.
  • the preparation method comprises reacting the compound of formula I substituted with a protecting group under the conditions of removing the protecting group to obtain the compound of formula I.
  • the protecting group can be selected from at least one of hydroxyl protecting group, amino protecting group and the like.
  • the present invention also provides the preparation method of the compound shown in formula II, comprising the following steps:
  • A, E, G, K, X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , X 8 , R 21 have the above definitions;
  • D is selected from -OR 21 ;
  • L is selected from a leaving group.
  • the leaving group is selected from OH, halogen or OTf.
  • the reaction is carried out in the presence of a base, for example in the presence of potassium carbonate.
  • the temperature of the reaction is 50-100° C.
  • the reaction time is 1-24 hours.
  • the reaction may be carried out in the presence of an organic solvent such as DMF.
  • the present invention also provides a method for preparing the compound of formula II-1, comprising reacting the compound of formula II-2 to prepare the compound of formula II-1:
  • A, D, E, G, K, X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , and X 8 have the above-mentioned definitions.
  • the reaction is carried out in the presence of hydrazine hydrate, and X 7 is N and X 8 is NH.
  • the reaction is carried out in the presence of an organic solvent such as DMF.
  • the reaction is carried out under heating conditions.
  • the present invention also provides the compound represented by the above formula I-1, formula I-2, formula I-3, formula II-1 or formula II-2.
  • the present invention also provides the use of the compound represented by the formula I-1, the formula I-2, the formula I-3 or the formula II-1 for preparing the compound represented by the formula I.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound selected from formula I, its stereoisomers, racemates, tautomers, isotopic labels, nitrogen oxides or pharmaceutically acceptable compounds At least one of the salts, for example, a therapeutically effective amount selected from the compounds of formula I, its stereoisomers, racemates, tautomers, isotopic labels, nitrogen oxides or pharmaceutically acceptable at least one of salts.
  • the pharmaceutical composition further comprises one, two or more pharmaceutically acceptable adjuvants, such as carriers and/or excipients.
  • the pharmaceutical composition further comprises one or more additional therapeutic agents.
  • the present invention also provides a method for inhibiting cell proliferation in vitro or in vivo, the method comprising mixing cells with an effective amount of the compound of formula I, its stereoisomer, racemate, tautomer, An isotope label, nitrogen oxide, or a pharmaceutically acceptable salt, or a pharmaceutical composition thereof is contacted.
  • the present invention also provides a method for treating diseases mediated by RET gene and/or RET kinase, comprising administering to a patient a therapeutically effective amount of the compound shown in formula I, its stereoisomers, racemates, tautomers at least one of a compound, an isotopic label, a nitrogen oxide, or a pharmaceutically acceptable salt.
  • RET gene or RET when describing RET, RET gene or RET kinase, it means RET gene or RET selected from the group including but not limited to RET-wt, V804M, V804L, V804E, G810R, G810S, G810C, G810V and S904F Kinase.
  • the present invention also provides a method for treating a RET-related disease or disorder in a patient in need of such treatment, the method comprising administering to the patient a therapeutically effective amount of a compound represented by formula I of the present invention, a stereoisomer thereof, a peptide Rotors, tautomers, isotopic labels, nitrogen oxides or pharmaceutically acceptable salts or pharmaceutical compositions thereof.
  • the present invention also provides a method of treating cancer and/or inhibiting metastasis associated with a specific cancer in a patient in need of such treatment, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I of the present invention, its Stereoisomers, racemates, tautomers, isotopic labels, nitrogen oxides or pharmaceutically acceptable salts or pharmaceutical compositions thereof.
  • the present invention also provides a method of treating irritable bowel syndrome (IBS) and/or pain associated with IBS in a patient in need thereof, said method comprising administering to said patient a therapeutically effective amount of a compound of formula I of the present invention
  • IBS irritable bowel syndrome
  • the present invention also provides a method of providing supportive care to a cancer patient, including preventing or minimizing gastrointestinal disorders (eg, diarrhea) associated with treatment, including chemotherapy, comprising administering to the patient a therapeutically effective amount of Formula I of the present invention
  • gastrointestinal disorders eg, diarrhea
  • chemotherapy comprising administering to the patient a therapeutically effective amount of Formula I of the present invention
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomers, racemates, tautomers, isotopic labels, nitrogen oxides, or pharmaceutically acceptable salts in the preparation of medicaments Use in , wherein the medicament is for the treatment of RET kinase mediated diseases, inhibition of RET kinase activity, treatment of cancer and/or inhibition of metastasis associated with a particular cancer, treatment of irritable bowel syndrome (IBS) or IBS-related Pain, providing supportive care to cancer patients, treating RET-related diseases or conditions, reversing or preventing acquired resistance to anticancer drugs, delaying and/or preventing the development of anticancer drug resistance in individuals, developing resistance to anticancer drugs possibility increases.
  • IBS irritable bowel syndrome
  • IBS-related Pain providing supportive care to cancer patients, treating RET-related diseases or conditions, reversing or preventing acquired resistance to anticancer drugs, delaying and/or preventing the development of anticancer drug resistance in individuals, developing resistance to antican
  • the RET-related diseases or disorders are selected from diseases mediated by RET genes and/or RET kinases, wherein RET, RET genes or RET kinases are selected from diseases including but not limited to RET-wt, V804M, V804L, RET genes or RET kinases of V804E, G810R, G810S, G810C, G810V and S904F.
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomer, racemate, tautomer, isotope label, nitrogen oxide, or pharmaceutically acceptable salt for preparation Use in a medicament for the treatment of RET kinase-mediated diseases.
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomer, racemate, tautomer, isotope label, nitrogen oxide, or pharmaceutically acceptable salt for preparation Use in a drug for treating cancer and/or inhibiting metastasis associated with a specific cancer.
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomer, racemate, tautomer, isotope label, nitrogen oxide, or pharmaceutically acceptable salt for preparation Use in a medicament for the treatment of irritable bowel syndrome (IBS) or pain associated with IBS.
  • IBS irritable bowel syndrome
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomer, racemate, tautomer, isotope label, nitrogen oxide, or pharmaceutically acceptable salt for preparation Use in a medicament to provide supportive care to cancer patients, including the prevention or minimization of gastrointestinal disorders, such as diarrhea, associated with treatment, including chemotherapy.
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomer, racemate, tautomer, isotope label, nitrogen oxide, or pharmaceutically acceptable salt for preparation Use in drugs that inhibit RET kinase activity.
  • the present invention also provides at least one of the compound represented by formula I, its stereoisomer, racemate, tautomer, isotope label, nitrogen oxide, or pharmaceutically acceptable salt for preparation Use in a medicament for the treatment of RET-related diseases or disorders.
  • the present invention also provides a method for treating cancer in a patient in need thereof, the method comprising (a) determining whether the cancer is associated with a dysregulation of the expression of the RET gene, RET kinase, or any one thereof or activity or level (eg, RET-related cancer); (b) if the cancer is determined to be associated with a dysregulation of: the expression or activity or level of the RET gene, RET kinase, or any of them (eg, RET-related cancer); cancer), administering to the patient a therapeutically effective amount of a compound shown in formula I, its stereoisomer, racemate, tautomer, isotopic label, nitrogen oxide, or a pharmaceutically acceptable salt thereof at least one, or a pharmaceutical composition thereof.
  • the present invention also provides a method for reversing or preventing acquired resistance to an anticancer drug, the method comprising adding a therapeutically effective amount of the compound represented by formula I, its stereoisomer, racemate, tautomer At least one of isomers, isotopic labels, nitrogen oxides, or pharmaceutically acceptable salts is administered to a patient at risk of developing or having acquired resistance to an anticancer drug.
  • the present invention also provides a method of delaying and/or preventing the development of anticancer drug resistance in an individual, the method comprising administering to the individual an effective amount of a compound of formula I, its stereoisomers, racemates, tautomers At least one of isomers, isotopic labels, nitrogen oxides, or pharmaceutically acceptable salts, before, during, or after administration of an effective amount of the anticancer drug.
  • the present invention also provides a method of treating an individual suffering from cancer and having an increased likelihood of developing resistance to an anticancer drug, comprising concomitantly administering to the individual (a) an effective amount of a compound of formula I, a stereoisomer thereof , at least one of a racemate, a tautomer, an isotopic label, a nitrogen oxide, or a pharmaceutically acceptable salt; and (b) an effective amount of an anticancer drug.
  • the present invention also provides methods of treating an individual with a RET-related cancer having one or more RET inhibitor resistance mutations that increase the resistance of the cancer to those not represented by Formula I
  • Resistance of at least one of the compounds, their stereoisomers, racemates, tautomers, isotopic labels, nitrogen oxides, or pharmaceutically acceptable salts thereof to RET inhibitors e.g., RET -wt, substitutions at amino acid positions 804, 810, 904, eg, V804M, V804L, V804E, G810R, G810S, G810C, G810V, S904F
  • RET inhibitors e.g., RET -wt, substitutions at amino acid positions 804, 810, 904, eg, V804M, V804L, V804E, G810R, G810S, G810C, G810V, S904F
  • the present invention also provides a method of treating an individual with RET-related cancer, the method comprising administering a compound of formula I, a stereoisomer thereof, prior to, during or after administration of another other anticancer drug At least one of a racemate, a tautomer, an isotopic label, a nitrogen oxide, or a pharmaceutically acceptable salt.
  • the present invention provides a method of treating cancer (eg, RET-related cancer) in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound of general formula I, or a pharmaceutically acceptable salt thereof at least one or a pharmaceutical composition thereof.
  • cancer eg, RET-related cancer
  • the cancer eg, RET-associated cancer
  • the cancer is a hematological cancer.
  • the cancer eg, RET-associated cancer
  • the cancer is a solid tumor.
  • the cancer eg, RET-related cancer
  • lung cancer eg, small cell lung cancer or non-small cell lung cancer
  • papillary thyroid cancer medullary thyroid cancer, differentiated Thyroid cancer, recurrent thyroid cancer, refractory differentiated thyroid cancer, lung adenocarcinoma, bronchiolar carcinoma, multiple endocrine tumors type 2A or 2B (MEN2A or MEN2B, respectively), pheochromocytoma, parathyroid hyperplasia, Breast cancer, colorectal cancer (eg metastatic colorectal cancer), papillary renal cell carcinoma, gangliomatosis of the gastrointestinal mucosa, inflammatory myofibroblastic tumor or cervical cancer.
  • lung cancer eg, small cell lung cancer or non-small cell lung cancer
  • papillary thyroid cancer medullary thyroid cancer
  • differentiated Thyroid cancer recurrent thyroid cancer
  • refractory differentiated thyroid cancer lung adenocarcinoma
  • bronchiolar carcinoma multiple endocrine tumors
  • the cancer eg, RET-related cancer
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • juvenile cancer adrenocortical carcinoma, anus carcinoma, appendix cancer, astrocytoma, atypical teratoma/rhabdoid tumor, basal cell carcinoma, cholangiocarcinoma, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumor, primary Kitt lymphoma, carcinoid tumor, cancer of unknown primary, cardiac tumor, cervical cancer, childhood cancer, chordoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myeloproliferative neoplasm, colon cancer , colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, cholangiocarcinoma, ductal
  • the hematological cancer is selected from the group consisting of leukemia, lymphoma (non-Hodgkin lymphoma), Hodgkin disease (also known as Hodgkin lymphoma) and myeloma, e.g., acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic myeloid leukemia Monocytic leukemia (CMML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile monocytic leukemia (JMML), adult T-cell ALL, triple myelodysplastic AML (AML/TMDS), mixed lineage leukemia (MLL), acute lymphoblastic leukemia (ALL), acute myeloid
  • the hematological cancer eg, a hematological cancer that is a RET-related cancer
  • AML or CMML
  • the cancer is a solid tumor.
  • solid tumors eg, solid tumors that are RET-related cancers
  • the cancer is selected from lung cancer, papillary thyroid cancer, medullary thyroid cancer, differentiated thyroid cancer, relapsed thyroid cancer, refractory differentiated thyroid cancer, multiple endocrine tumors type 2A or 2B (respectively MEN2A or MEN2B), pheochromocytoma, parathyroid hyperplasia, breast cancer, colorectal cancer, papillary renal cell carcinoma, gastrointestinal mucosal gangliocytoma and cervical cancer.
  • lung cancer papillary thyroid cancer, medullary thyroid cancer, differentiated thyroid cancer, relapsed thyroid cancer, refractory differentiated thyroid cancer, multiple endocrine tumors type 2A or 2B (respectively MEN2A or MEN2B), pheochromocytoma, parathyroid hyperplasia, breast cancer, colorectal cancer, papillary renal cell carcinoma, gastrointestinal mucosal gangliocytoma and cervical cancer.
  • MEN2A or MEN2B multiple endocrine tumors
  • the patient is a human.
  • the present invention also provides a method for treating irritable bowel syndrome (IBS) in a patient in need thereof, the method comprising (a) determining whether IBS is associated with a disorder of the RET gene, RET kinase, or any of these expression or activity or level of one; (b) if IBS is determined to be associated with a disorder of the RET gene, RET kinase, or the expression or activity or level of any of them, administering to the patient a therapeutically effective amount of Formula I At least one of a compound or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • IBS irritable bowel syndrome
  • the present invention also provides a pharmaceutical combination for treating irritable bowel syndrome (IBS) in a patient in need thereof, comprising administering (a) a compound of formula I, a stereoisomer, a racemate, a tautomer thereof at least one of isomers, isotopic labels, nitrogen oxides, or pharmaceutically acceptable salts, (b) other therapeutic agents, and (c) optionally at least one pharmaceutically acceptable carrier,
  • IBS irritable bowel syndrome
  • the present invention also provides pharmaceutical compositions comprising such combinations.
  • the present invention also provides the use of such a combination in the manufacture of a medicament for the treatment of IBS.
  • the present invention also provides a commercial package or product comprising the combination as a combined preparation for simultaneous, separate or sequential use; and a method of treating IBS in a patient in need thereof.
  • the compounds of the present invention may be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical arts, and they can be administered by a variety of routes, depending on whether local or systemic treatment is desired and the area being treated. May be delivered topically (eg, transdermally, dermally, ocularly, and mucous membranes including intranasal, vaginal, and rectal), pulmonary (eg, by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal), Oral or parenteral administration.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, eg, intrathecal or intracerebroventricular administration.
  • Parenteral administration may be in single bolus form, or may be administered, for example, by a continuous infusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, water, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves, etc. may also be useful.
  • the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of a capsule, sachet, paper or other container.
  • an excipient serves as a diluent, it can be a solid, semi-solid or liquid material serving as a vehicle, carrier or medium for the active ingredient.
  • compositions may be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (solid or dissolved liquid vehicles); ointments, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders containing, for example, up to 10% by weight of the active compound.
  • excipients include lactose, glucose, sucrose, sorbitol, mannitol, starch, acacia, calcium phosphate, alginate, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, poly Vinylpyrrolidone, cellulose, water, syrup and methylcellulose.
  • the formulations may also contain: lubricants such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preservatives such as methyl benzoate and hydroxypropyl benzoate; sweetening and flavoring agents.
  • the compositions of the present invention can be formulated to provide immediate, sustained or delayed release of the active ingredient after administration to a patient by using methods known in the art.
  • compositions may be formulated in unit dosage form, each dosage containing about 5 to 1000 mg, more usually about 100 to 500 mg, of active ingredient.
  • unit dosage form refers to physically discrete units suitable as unitary dosage units for human patients and other mammals, each unit containing a predetermined quantity of activity calculated to produce the desired therapeutic effect in association with a suitable pharmaceutical excipient substance.
  • the effective dose of the active compound can vary widely and is usually administered in a pharmaceutically effective amount.
  • the actual amount of compound administered will generally be determined by the physician according to relevant circumstances, including the condition being treated, the route of administration selected, the actual compound administered; the age, weight and response of the individual patient; severity, etc.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of the compounds of the present invention.
  • these preformulation compositions are referred to as homogeneous, it is meant that the active ingredient is generally uniformly distributed throughout the composition such that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • the solid preformulation is then divided into unit dosage forms of the type described above containing, for example, about 0.1 to 1000 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention may be coated or compounded to provide dosage forms that provide the advantage of prolonged action.
  • a tablet or pill contains an inner-dose and an outer-dose component, the latter being a coated form of the former.
  • the two components can be segregated by an enteric layer, which acts to prevent disintegration in the stomach, allowing the inner component to pass intact through the duodenum or to delay release.
  • enteric layers or coatings such materials including a variety of polymeric acids and mixtures of polymeric acids with such materials such as shellac, cetyl alcohol and cellulose acetate.
  • Liquid forms for oral or injectable administration into which the compounds and compositions of the present invention may be incorporated include aqueous solutions, suitably flavored syrups, aqueous or oily suspensions; and edible oils such as cottonseed oil, sesame oil, coconut oil Flavoured emulsions in oil or peanut oil; and elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions, powders in pharmaceutically acceptable aqueous or organic solvents or mixtures thereof.
  • Liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described above.
  • the compositions are administered by oral or nasal respiratory routes for local or systemic effect.
  • the composition can be nebulized by using an inert gas.
  • the nebulized solution can be inhaled directly from the nebulizing device, or the nebulizing device can be connected to a mask drape or intermittent positive pressure breathing machine.
  • Solutions, suspensions or powder compositions may be administered orally or nasally from a device that delivers the formulation in an appropriate manner.
  • the amount of compound or composition administered to a patient is not fixed and depends on the drug administered, the purpose of administration such as prophylaxis or treatment; the state of the patient, the mode of administration, and the like.
  • the composition may be administered to a patient already suffering from the disease in an amount sufficient to cure or at least partially inhibit the symptoms of the disease and its complications.
  • the effective dose will depend on the disease state being treated and the judgment of the attending clinician, which will depend upon factors such as the severity of the disease, the age, weight and general condition of the patient.
  • composition to be administered to a patient may be in the form of a pharmaceutical composition as described above.
  • These compositions can be sterilized by conventional sterilization techniques or by filter sterilization.
  • Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound formulation is usually 3-11, more preferably 5-9, and most preferably 7-8. It will be appreciated that the use of certain of the foregoing excipients, carriers or stabilizers will result in the formation of pharmaceutical salts.
  • Therapeutic doses of the compounds of the present invention may depend, for example, on the particular use of the treatment, the mode of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the ratio or concentration of a compound of the present invention in a pharmaceutical composition may not be fixed and depends on a variety of factors including dosage, chemical properties (eg, hydrophobicity) and route of administration.
  • the compounds of the present invention may be provided for parenteral administration in physiologically buffered aqueous solutions containing about 0.1 to 10% w/v of the compound. Some typical dosages range from about 1 [mu]g/kg to about 1 g/kg body weight/day.
  • the dose ranges from about 0.01 mg/kg to about 100 mg/kg body weight/day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the general state of health of the particular patient, the relative biological potency of the compound selected, the excipient formulation and its route of administration. Effective doses can be obtained by extrapolation from dose-response curves derived from in vitro or animal model test systems.
  • the numerical ranges recited in this specification and claims are equivalent to at least reciting each specific integer value therein.
  • the numerical range "1-40” is equivalent to reciting each integer value in the numerical range “1-10", ie, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and the numerical range
  • Each integer value in “11-40” is 11, 12, 13, 14, 15, ..., 35, 36, 37, 38, 39, 40. It is to be understood that in the context of one, two or more used herein in describing a substituent, "more” shall refer to an integer > 3, such as 3, 4, 5, 6, 7, 8, 9 or 10 .
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • C 1-40 alkyl is to be understood as preferably denoting a linear or branched saturated monovalent hydrocarbon radical having 1 to 40 carbon atoms.
  • C1-6 alkyl refers to straight and branched chain alkyl groups having 1, 2, 3, 4, 5 or 6 carbon atoms.
  • the alkyl group is, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl , 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-dimethylbutyl or 1,2-dimethylbutyl, etc. or their isomers.
  • C 2-40 alkenyl is to be understood to mean preferably a straight or branched monovalent hydrocarbon group containing one or more double bonds and having 2 to 40 carbon atoms, preferably "C 2-6 alkenyl” .
  • C 2-6 alkenyl is understood to mean preferably a straight or branched monovalent hydrocarbon radical comprising one or more double bonds and having 2, 3, 4, 5 or 6 carbon atoms, in particular 2 or 3 carbon atoms (“C 2-3 alkenyl”), it being understood that where the alkenyl group contains more than one double bond, the double bonds may be separated from each other or conjugated.
  • the alkenyl group is, for example, vinyl, allyl, (E)-2-methylvinyl, (Z)-2-methylvinyl, (E)-but-2-enyl, (Z)- But-2-enyl, (E)-but-1-enyl, (Z)-but-1-enyl, pent-4-enyl, (E)-pent-3-enyl, (Z) -Pent-3-enyl, (E)-pent-2-enyl, (Z)-pent-2-enyl, (E)-pent-1-enyl, (Z)-pent-1-ene base, hex-5-enyl, (E)-hex-4-enyl, (Z)-hex-4-enyl, (E)-hex-3-enyl, (Z)-hex-3- Alkenyl, (E)-hex-2-enyl, (Z)-hex-2-enyl, (E)-hex-1-eny
  • C 2 -40 alkynyl should be understood to mean a straight or branched monovalent hydrocarbon group containing one or more triple bonds and having 2 to 40 carbon atoms, preferably "C 2 -C 6 -alkynyl”".
  • C 2 -C 6 -alkynyl is to be understood as preferably denoting a straight-chain or branched monovalent hydrocarbon radical comprising one or more triple bonds and having 2, 3, 4, 5 or 6 carbon atoms, in particular is 2 or 3 carbon atoms ("C2-C3-alkynyl").
  • Said C2 - C6 -alkynyl is, for example, ethynyl, prop-1-ynyl, prop-2-ynyl, but-1-ynyl, but-2-ynyl, but-3-ynyl, Pent-1-ynyl, pent-2-ynyl, pent-3-ynyl, pent-4-ynyl, hex-1-ynyl, hex-2-ynyl, hex-3-ynyl, hexyl -4-ynyl, hex-5-ynyl, 1-methylprop-2-ynyl, 2-methylbut-3-ynyl, 1-methylbut-3-ynyl, 1-methyl But-2-ynyl, 3-methylbut-1-ynyl, 1-ethylprop-2-ynyl, 3-methylpent-4-ynyl, 2-methylpent-4-ynyl , 1-methylpent-4-ynyl,
  • C 3-40 cycloalkyl is understood to mean a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane having 3 to 40 carbon atoms, preferably “C 3-10 cycloalkyl”.
  • C 3-10 cycloalkyl is understood to mean a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane having 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms.
  • the C 3-10 cycloalkyl group can be a monocyclic hydrocarbon group, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or a bicyclic Hydrocarbyl such as decalin ring.
  • 3-20 membered heterocyclyl means a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane containing a total number of ring atoms of 1-5 heteroatoms independently selected from N, O and S It is a non-aromatic cyclic group of 3-20 (eg, the number of atoms is 3, 4, 5, 6, 7, 8, 9, 10, etc.), preferably "3-10-membered heterocyclic group”.
  • heterocyclyl means a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane containing 1-5, preferably 1-3 heteroatoms independently selected from N, O and S , eg 1, 2, 3 heteroatoms independently selected from N, O and S.
  • the heterocyclyl group can be attached to the remainder of the molecule through any of the carbon atoms or a nitrogen atom, if present.
  • the heterocyclic group may include, but is not limited to: 4-membered rings, such as azetidinyl, oxetanyl (eg, azetidin-1-yl); 5-membered rings, such as Tetrahydrofuranyl, dioxolyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl; or 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithi Alkyl, thiomorpholinyl, piperazinyl, or trithianyl; or a 7-membered ring such as diazepanyl.
  • 4-membered rings such as azetidinyl, oxetanyl (eg, azetidin-1-yl); 5-membered rings, such as Tetrahydrofuranyl, dioxolyl, pyrrolidinyl
  • the heterocyclyl group can be benzo-fused.
  • the heterocyclyl group may be bicyclic, such as, but not limited to, a 5,5 membered ring, such as a hexahydrocyclopento[c]pyrrole-2(1H)-yl ring, or a 5,6 membered bicyclic ring, such as a hexahydropyrrole
  • the [1,2-a]pyrazin-2(1H)-yl ring may be partially unsaturated, i.e.
  • the heterocyclic group is non-aromatic.
  • the carbon atom on the 3-20-membered heterocyclic group can be connected with other groups, or it can be a 3-20-membered heterocyclic group Heterocyclic atoms on the ring are attached to other groups.
  • the 3-20 membered heterocyclic group is selected from piperazinyl
  • the nitrogen atom on the piperazinyl may be attached to other groups.
  • the 3-20-membered heterocyclic group is selected from piperidinyl
  • the nitrogen atom on the piperidinyl ring and the carbon atom in the para position are connected to other groups.
  • C 6-20 aryl group should be understood to preferably mean a monovalent aromatic or partially aromatic monocyclic, bicyclic or tricyclic hydrocarbon ring having 6 to 20 carbon atoms, preferably a "C 6-14 aryl group” .
  • C 6-14 aryl is to be understood as preferably denoting a monovalent aromatic or partially aromatic monocyclic, bicyclic or Tricyclic hydrocarbon rings (“C 6-14 aryl”), especially rings with 6 carbon atoms (“C 6 aryl”), such as phenyl; or biphenyl, or those with 9 carbon atoms a ring (“C 9 aryl”) such as indanyl or indenyl, or a ring having 10 carbon atoms (“C 10 aryl”) such as tetrahydronaphthyl, dihydronaphthyl or naphthyl, Either a ring with 13 carbon atoms (“ C13 aryl”), such as fluorenyl, or a ring with 14 carbon atoms (“ C14 aryl”), such as anthracenyl.
  • the substitution site is not limited,
  • 5-20 membered heteroaryl is understood to include monovalent monocyclic, bicyclic or tricyclic aromatic ring systems having 5 to 20 ring atoms and containing 1 to 5 atoms independently selected from N, O and a heteroatom of S, eg "5-14 membered heteroaryl".
  • the term “5-14 membered heteroaryl” is understood to include monovalent monocyclic, bicyclic or tricyclic aromatic ring systems having 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, especially 5 or 6 or 9 or 10 carbon atoms, and which contain 1 to 5, preferably 1 to 3, heteroatoms each independently selected from N, O and S and, additionally in each case The following can be benzo-fused.
  • heteroaryl is selected from the group consisting of thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiazolyl oxazolyl, thi-4H-pyrazolyl, etc.
  • the 5-20-membered heteroaryl group is connected with other groups to form the compound of the present invention, it may be that the carbon atoms on the 5-20-membered heteroaryl ring are connected to other groups, or it may be a 5-20-membered heteroaryl group. Heteroatoms on the aryl ring are attached to other groups.
  • the 5-20 membered heteroaryl When the 5-20 membered heteroaryl is substituted, it can be mono- or polysubstituted. Moreover, there is no restriction on the substitution site, for example, the hydrogen attached to the carbon atom on the heteroaryl ring may be substituted, or the hydrogen attached to the heteroatom on the heteroaryl ring may be substituted.
  • a heterocyclyl, heteroaryl or heteroarylene group includes all possible isomeric forms thereof, such as positional isomers thereof.
  • thienyl or thienylene includes thien-2-yl, thien-2-yl, thien-3-yl and thien-3 - base; pyrazol-1-yl, pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl.
  • C 1-6 alkyl also apply to C 1-6 alkyloxy, -N(C 1-6 alkyl ) 2 , -NHC 1-6 alkyl or -S(O) 2 -C 1-6 alkyl and the like.
  • the compound represented by formula I can exist in the form of various pharmaceutically acceptable salts. If these compounds have a basic center, they can form acid addition salts; if these compounds have an acidic center, they can form base addition salts; if these compounds contain both an acidic center (such as a carboxyl group) and a basic center (such as amino), it can also form internal salts.
  • the compounds of the present invention may exist in the form of solvates, such as hydrates, wherein the compounds of the present invention comprise a polar solvent as a structural element of the crystal lattice of the compound, in particular, for example, water, methanol or ethanol.
  • a polar solvent as a structural element of the crystal lattice of the compound, in particular, for example, water, methanol or ethanol.
  • the amount of polar solvent, especially water may be present in stoichiometric or non-stoichiometric ratios.
  • the compounds of the present invention may be chiral and thus may exist in various enantiomeric forms. Thus these compounds may exist in racemic or optically active forms.
  • the compounds of the present invention or their intermediates can be separated into enantiomeric compounds by chemical or physical methods well known to those skilled in the art, or used in this form for synthesis. In the case of racemic amines, diastereomers are prepared from the mixture by reaction with an optically active resolving agent.
  • suitable resolving agents are optically active acids such as the R and S forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid, suitable N-protected amino acids such as N- Benzoylproline or N-benzenesulfonylproline) or various optically active camphorsulfonic acids. It is also possible to use optically active resolving agents such as dinitrobenzoylphenylglycine, cellulose triacetate or other carbohydrate derivatives or chiral derivatized methacrylate polymers immobilized on silica gel. Chromatographic enantiomeric resolution is advantageously performed. Suitable eluents for this purpose are aqueous or alcoholic solvent mixtures, eg hexane/isopropanol/acetonitrile.
  • tautomer refers to an isomer of a functional group resulting from the rapid movement of an atom in two positions in a molecule.
  • the compounds of the present invention may exhibit tautomerism.
  • Tautomeric compounds can exist as two or more interconvertible species.
  • Proton tautomers arise from the migration of covalently bonded hydrogen atoms between two atoms.
  • Tautomers generally exist in equilibrium, and attempts to separate individual tautomers usually result in a mixture whose physicochemical properties are consistent with a mixture of compounds. The position of equilibrium depends on the chemical properties within the molecule.
  • the ketone form predominates; in phenols, the enol form predominates.
  • the present invention encompasses all tautomeric forms of the compounds.
  • the corresponding stable isomers can be isolated according to known methods, eg by extraction, filtration or column chromatography.
  • patient refers to any animal including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses or primates, most preferably humans.
  • terapéuticaally effective amount refers to the amount of active compound or drug that a researcher, veterinarian, physician or other clinician is seeking to elicit a biological or medical response in a tissue, system, animal, individual or human
  • Prevention of disease eg, prevention of disease, disorder or condition in individuals who are susceptible to a disease, disorder or condition but have not yet experienced or developed disease pathology or symptoms.
  • Inhibiting a disease eg, inhibiting a disease, disorder or condition (ie preventing further progression of the pathology and/or condition) in an individual who is experiencing or developing the pathology or symptom of the disease, disorder or condition.
  • Alleviating disease eg, alleviating a disease, disorder or condition (ie, reversing the pathology and/or symptoms) in an individual who is experiencing or experiencing the pathology or symptoms of the disease, disorder or condition.
  • the compounds of the present invention can act as highly selective or inhibitory RET inhibitors against, for example, the RET gatekeeper residue mutant RETV804M, the RET solvent front residue mutant G810R and other clinically relevant RET mutants as well as RET-wt Has excellent inhibitory effect.
  • the preferred compounds of the present invention can effectively inhibit the growth of thyroid cancer-derived TT cell lines and Ba/F3 cells transformed by various RET mutants, can block cellular RET autophosphorylation and its downstream pathways, and significantly induce TT cell death.
  • the preferred compounds of the present invention also have good pharmacokinetic properties, and when used as active ingredients, they can be administered to patients in smaller doses, thereby reducing the cost of treatment for patients.
  • Step A N-(1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-4-methylpiperidin-4-yl)-5-fluoro-2-methylbenzamide
  • Step A 2,4,6-Trimethylbenzenesulfonic acid 1-amino-3-bromo-5-methoxypyridine-1-onium
  • Step B 4-Bromo-2-fluoro-6-methoxypyrazolo[1,5-a]pyridine
  • Step C 4-Bromo-2-fluoro-6-methoxypyrazolo[1,5-a]pyridine-3-carbaldehyde
  • Step D 4-Bromo-6-methoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step E 4-Bromo-6-hydroxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step F 4-Bromo-6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step G 6-Ethoxy-4-(6-fluoropyridin-3-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step H (1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine-2- yl)-4-methylpiperidin-4-yl)amino tert-butyl carbonate
  • Step 1 1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-4-methylpiperidine-4-amino hydrochloride
  • Step J 3-Chloro-N-(1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyridin-2-yl)-4-methylpiperidin-4-yl)2-pyridinecarboxamide
  • Step A 4-((tert-Butylcarbonyl)amino)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a ]pyridin-4-yl)pyridin-2-yl)piperidine-4-carboxylate ethyl ester
  • Step B 4-(Amino)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl ) pyridin-2-yl)piperidine-4-carboxylate ethyl ester
  • Step C 4-(2,6-Difluorobenzamide)-1-(5-(6-ethoxy-1hydro-pyrazol[3',4':3,4]pyrazole[1, 5-a]Pyridin-4-yl)pyridin-2-yl)piperidine-4-carboxylate ethyl ester
  • Step D N-(1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-4-(hydroxymethyl)piperidin-4-yl)-2,6-difluorobenzamide
  • Step A 4-(3-Chloromethylpyridineamide)-1-(5-(6-ethoxy-1hydro-pyrazo[3',4':3,4]pyrazo[1,5- a]Pyridin-4-yl)pyridin-2-yl)piperidine-4-carboxylate ethyl ester
  • Step B 3-Chloro-N-(1-(5-(6-ethoxy-1hydro-pyrazol[3',4':3,4]pyrazol[1,5-a]pyridine-4 -yl)pyridin-2-yl)-4-(hydroxymethyl)piperidin-4-yl)-pyridinecarboxamide
  • Step A tert-Butylcarbonyl(1-5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-4-(hydroxymethyl)piperidin-4-yl)carbamate
  • Step B tert-Butylcarbonyl (1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl) Pyridin-2-yl)-4-formylpiperidin-4-yl)carbamate
  • Step C tert-Butylcarbonyl(4-((2-methylamino)methyl)-1-(5-(6-ethoxy-1H-pyrazole[3',4':3,4]pyrazole) [1,5-a]Pyridin-4-yl)pyridin-2-yl)piperidin-4-yl)carbamate
  • Step D 4-((2-Methylamino)methyl)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5- a]Pyridin-4-yl)pyridin-2-yl)piperidin-4-amine hydrochloride
  • Step E 3-Chloro-N-(4-((2methylamino)methyl)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyridine) oxazol[1,5-a]pyridin-4-yl)pyridin-2-yl)piperidin-4-yl)pyridinamide
  • Step A tert-Butylcarbonyl(1-(5-(6-ethoxy-1hydro-pyrazol[3',4':3,4]pyrazol[1,5-a]pyridin-4-yl )pyridin-2-yl)-4-((4-ethylpiperazin-1-yl)methyl)piperidin-4-yl)carbamate
  • Step B 1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2- yl)-4-((4-ethylpiperazin-1-yl)methyl)piperidin-4-amine hydrochloride
  • Step C N-(1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-4-((4-ethylpiperazin-1-yl)methyl)piperidin-4-yl)-2,5-difluorobenzamide
  • Step A tert-Butylcarbonyl(1-(5-(6-ethoxy-1hydro-pyrazol[3',4':3,4]pyrazol[1,5-a]pyridin-4-yl )pyridin-2-yl)-4-(methylmorpholino)piperidin-4-yl)carbamate
  • Step B 1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2- yl)-4-((4-ethylpiperazin-1-yl)methyl)piperidin-4-amine hydrochloride
  • Step C N-(1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-4-((4-ethylpiperazin-1-yl)methyl)piperidin-4-yl)-3-methylbutanamide
  • Step A ((3S,4R)-1-(5-Bromopyridin-2-yl)-4-hydroxypyrrolidin-3-yl)amino tert-butyl carbonate
  • Step B ((3S,4S)-1-(5-Bromopyridin-2-yl)-4-(pyridin-2-yloxy)pyrrolidin-3-yl)amino tert-butyl carbonate
  • Step C ((3S,4S)-1-(5-(6-ethoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3',4':3 ,4]Pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl)-4-(pyridin-2-yloxy)pyrrolidin-3-yl)amino tert-butyl carbonate
  • 1,4-dioxane (1 mL), ((3S,4S)-1-(5-bromopyridin-2-yl)-4-(pyridin-2-yloxy)pyrrolidine were successively added to the reaction flask -3-yl)amino tert-butyl carbonate (60 mg, 0.14 mmol), 6-ethoxy-1-(tetrahydro-2H-pyran-2-yl)-4-(4,4,5,5- Tetramethyl-1,3,2-dioxaboran-2-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine (47.5 mg, 0.12 mmol), potassium carbonate (32.8 mg, 0.23 mmol), tetrakistriphenylphosphine palladium (13.3 mg, 0.006 mmol), water (0.5 mL), and the temperature was raised to 90° C. to react for 16 hours. The reaction solution was poured into water, extracted with dich
  • Step D (3S,4S)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl )pyridin-2-yl)-4-(pyridin-2-yloxy)pyrrolidin-3-amine
  • 6-benzyl-1-thia-6-azaspiro[2.5]octane (4.3 g, 19.6 mmol) and tetrahydrofuran (50 mL) were sequentially added to the reaction flask, the temperature was lowered to 5 °C, and lithium tetrahydroaluminum was added. (1.1 g, 29.4 mmol), the reaction was incubated for 1 hour, and the reaction was complete.
  • Step F 4-(isobutylsulfonyl)-4-methylpiperidine
  • Step G 6-Ethoxy-4-(6-(4-(isobutylsulfonyl)-4-methylpiperidin-1-yl)pyridin-3-yl)-1H-pyrazolo[3' ,4':3,4]pyrazo[1,5-a]pyridine
  • Step A 1-(5-Bromopyridin-2-yl)-4-formyl-N-isobutylpiperidine-4-amide
  • Step B 1-(5-Bromopyridin-2-yl)-4-((dimethylamino)methyl)-N-isobutylpiperidine-4-carboxamide
  • Step C 1-(5-Bromopyridin-2-yl)-4-((dimethylamino)methyl)-N-isobutylpiperidine-4-carboxamide
  • Step A 1-(5-Bromopyridin-2-yl)-4-(azetidin-1-ylmethyl)-N-isobutylpiperidine-4-carboxamide
  • Step B 4-(azetidin-1-ylmethyl)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1 ,5-a]pyridin-4-yl)pyridin-2-yl)-N-isobutylpiperidine-4-amide
  • Step A 1-(5-Bromopyridin-2-yl)-4-(pyrrolidin-1-ylmethyl)-N-isobutylpiperidine-4-carboxamide
  • Step B 4-(Pyrrolidin-1-ylmethyl)-1-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1,5- a]Pyridin-4-yl)pyridin-2-yl)-N-isobutylpiperidine-4-amide
  • Step A ((3R,4S)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyridin-2-yl)-3-hydroxypiperidin-4-yl)amino tert-butyl carbonate
  • Step B ((3R,4S)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyridin-2-yl)-3-hydroxypiperidin-4-yl)amino hydrochloride
  • Step C N-((3R,4S)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine- 4-yl)pyridin-2-yl)-3-hydroxypiperidin-4-yl)-3-methylbutanamide
  • Step A ((3S,4S)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyridin-2-yl)-3-hydroxypiperidin-4-yl)carbamate
  • Step B (3S,4S)-4-amino-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine -4-yl)pyridin-2-yl)piperidin-3-ol
  • Step C 2-Chloro-N-((3S,4S)-1-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1,5- a]Pyridin-4-yl)pyridin-2-yl)-3-hydroxypiperidin-4-yl)-6-fluorobenzamide
  • Step A ((3S,4R)-1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyridin-2-yl)-4-hydroxypiperidin-3-yl)amino tert-butyl carbonate
  • Step B ((3S,4S)-4-(1,3-phthalimide-2-yl)-1-(5-(6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl)piperidin-3yl)amino tert-butyl carbonate
  • Step C ((S,4S)-4-Amino-1-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1,5-a] Pyridin-4-yl)pyridin-2-yl)piperidin-3yl)amino tert-butyl carbonate
  • Step D N-((3S,4S)-3-amino-1-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1,5- a]Pyridin-4-yl)pyridin-2-yl)piperidin-4-yl)-2chloro-6-methylbenzamide
  • Step A (3S,4S)-1-(5-(6-ethoxy-1-(tetrahydro-2hydro-pyran-2-yl)-1hydro-pyrazolo[3',4': 3,4]Pyrazolo[1,5-a]pyridin-4-yl)pyrazin-2-yl)-3-hydroxypiperidin-4-yl)amino tert-butyl carbonate
  • Step B (3S,4S)-4-Amino-1-(5-(6-ethoxy-1hydro-pyrazo[3',4':3,4]pyrazo[1,5-a] Pyridin-4-yl)pyrazin-2-yl)-3-hydroxypiperidine hydrochloride
  • Step C 3-Chloro-N-((3S,4S)-(1-(5-(6-ethoxy-1hydro-pyrazol[3',4':3,4]pyrazol[1, 5-a]Pyridin-4-yl)pyrazin-2-yl)-3-hydroxypiperidin-4-yl)-2-pyridinecarboxamide
  • Step A 2-Chloro-N-((3S,4S)-(1-(5-(6-ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazole[1, 5-a]Pyridin-4-yl)pyrazin-2-yl)-3-hydroxypiperidin-4-yl)-6-fluorobenzamide
  • Step A 4-Bromo-6-ethoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazo[3',4':3,4]pyrazo[1,5 -a]pyridine
  • Step B 6-Ethoxy-1-(tetrahydro-2H-pyran-2-yl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborane -2-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step C 4-(5-Chloropyrazin-2-yl)-6-ethoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3',4': 3,4]Pyrazolo[1,5-a]pyridine
  • Step D (1-(5-(6-Ethoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole[3',4':3,4]pyrazole[ 1,5-a]Pyridin-4-yl)pyrazin-2-yl)-4methylpiperidin-4-yl)amino tert-butyl carbonate
  • Step E (1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyrazine-2 -yl)-4methylpiperidine-4-amino hydrochloride
  • Step F 3-Chloro-N-(1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyrazin-2-yl)-4-methylpiperidin-4-yl)-5-fluoro-2-pyridinecarboxamide
  • Step A N-(1-(5-(6-Ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine oxazin-2-yl)-4methylpiperidin-4-yl)-2,3,6-trifluorobenzamide
  • Step A 2-Chloro-N-(1-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyrazin-2-yl)-4-methylpiperidin-4-yl)-5-fluorobenzamide
  • Step A 3-Chloro-N-(1-(5-(6-ethoxy-1hydro-pyrazol[3',4':3,4]pyrazol[1,5-a]pyridine-4 -yl)pyrazin-2-yl)-4methylpiperidin-4-yl)-2-pyridinecarboxamide
  • Step B Methyl 4-((benzyloxy)methyl)-1-(5-bromopyridin-2-yl)piperidine-4-carboxylate
  • Step D 4-Hydroxymethyl-1-(5-bromopyridin-2-yl)piperidine-4-carboxylic acid
  • Step E 1-(5-Bromopyridin-2-yl)-4-(hydroxymethyl)-N-isobutylpiperidine-4-amide
  • Step F 1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine-2 -yl)-4-(hydroxymethyl)-N-isobutylpiperidine-4-amide
  • Step A 2-Chloro-N-((3S,4S)-(1-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1,5] -a]Pyridin-4-yl)pyrazin-2-yl)-3-hydroxypiperidin-4-yl)-5-fluorobenzamide
  • Step A tert-butyl 4-(N-isopropylsulfonyl)piperidine-1-carbonate
  • Step C 1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-N-isopropylpiperidine-4-sulfonamide
  • Step A tert-butyl 4-hydroxy-4-(benzenesulfonamidemethylene)piperidine-1-carbonate
  • Step C N-((1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-4-hydroxypiperidin-4-yl)methylene)benzenesulfonamide
  • Step A tert-Butyl 4-(pyridine-2-oxy)piperidine-1-carboxylate
  • Step C 6-Ethoxy-4-(6-(4-(pyridin-2-oxy)piperidin-1-yl)pyridin-3-yl)-1H-pyrazolo[3',4': 3,4]Pyrazolo[1,5-a]piperidine
  • Step A tert-butyl 4-((6-methylpyridazin-3-yl)oxy)piperidine-1-carbonate
  • Step C 6-Ethoxy-4-(6-(4-((6-methylpyridazin-3-yl)oxy)piperidin-1-yl)pyridin-3-yl)-1H-pyrazole [3',4':3,4]pyrazole[1,5-a]pyridine
  • Step A 4-(2,5-Difluorobenzamideamide)-1-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1,5] -a]Pyridin-4-yl)pyridin-2-yl)piperidine-4-carboxylate ethyl ester
  • Step B N-(1-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-4-(hydroxymethyl)piperidin-4-yl)-2,5-difluorobenzamide
  • Step A 3-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-3,6-diazabicyclo[3.1.1]heptane-6-carbonate tert-butyl ester
  • Step B 4-(6-(3,6-Diazabicyclo[3.1.1]heptan-6-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3', 4':3,4]pyrazo[1,5-a]pyridine
  • Step C 6-Ethoxy-4-(6-(3-(((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptyl -6-yl)pyridin-3-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step A (3aR,6aS)-5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl ) pyridin-2-yl)hexahydropyridine[3,4-c]pyrrole-2(1H)-carboxylate tert-butyl ester
  • Step B 6-Ethoxy-4-(6-((3aR,6aS)-hexahydropyrido[3,4-c]pyrrol-2(1H)-yl)pyridin-3-yl)-1H- Pyrazo[3',4':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step A (1R,5S)-3-(5-(6-ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4- yl)pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-8-carbonate tert-butyl ester
  • Step B 4-(6-((1R,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)pyridin-3-yl)-6-ethoxy-1H -Pyrazolo[3',4':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C (2-Chloro-6-fluoro)((1R,5S)-3-(5-(6-ethoxy-1H-pyrazo[3',4':3,4]pyrazo[1] ,5-a]pyridin-4-yl)pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octan-8-yl)benzamide
  • Step A 6-Ethoxy-4-(6-((1R,5S)-8-((6-methoxypyridin-3-yl)methyl)-3,8-diazabicyclo[ 3.2.1]Octan-3-yl)pyridin-3-yl)-1H-pyrazolo[3',4':3,4]pyrazo[1,5-a]pyrazole
  • Step A (1R,5S)-8-(5-(6-ethoxy-1H-pyrazol[3',4':3,4]pyrazol[1,5-a]pyridin-4-yl )pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-3-carbonate tert-butyl ester
  • Step B 4-(6-((1R,5S)-3,8-diazabicyclo[3.2.1]octan-8-yl)pyridin-3-yl)-6-ethoxy-1H -Pyrazolo[3',4':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C (2-Chloro-6-fluoro)((1R,5S)-8-(5-(6-ethoxy-1hydro-pyrazole[3',4':3,4]pyrazole[ 1,5-a]pyridin-4-yl)pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octan-3-yl)benzamide
  • Step A 6-Ethoxy-4-(6-((1R,5S)-3-((6-methoxypyridin-3-yl)methyl)-3,8-diazabicyclo[ 3.2.1]Octan-3-yl)pyridin-8-yl)-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyrazole
  • Step A tert-Butyl 5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,5-diazabicyclo[2.2.2]octane-2-carboxylate
  • Step B 4-(6-(2,5-Diazabicyclo[2.2.2]octan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3' ,4':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C (2-Chloro-6-fluorophenyl)-5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a ]pyridin-4-yl)pyridin-2-yl)-2,5-diazabicyclo[2.2.2]octan-2-yl)methanone
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-4,7-diazaspiro[2.5]octane-4-carboxylate tert-butyl ester
  • Step B 4-(6-(4,7-Diazaspiro[2.5]octan-7-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-4,7-diazaspiro[2.5]octan-4-yl)-2-chloro-6-fluorobenzamide
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-3-oxa-7,9-azabicyclo[3.3.1]nonane-9-carboxylate tert-butyl ester
  • Step B 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-3-oxa-7,9-azabicyclo[3.3.1]nonane hydrochloride
  • Step C N-(7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-3-oxa-7,9-azabicyclo[3.3.1]nonan-9-yl)-2-chloro-6-fluorobenzamide
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-9-((6-methoxypyridin-3-yl)methylene)-3-oxa-7,9-azabicyclo[3.3.1]nonane
  • Step A (1R,5S,6S)-3-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4 -yl)pyridin-2-yl)-3-azabicyclo[3.1.0]-6-hexylamino hydrochloride
  • Step B 2-Chloro-N-((1R,5S,6S)-3-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazole[1, 5-a]Pyridin-4-yl)pyridin-2-yl)-3-azabicyclo[3.1.0]hexyl-6-yl)-6-fluorobenzamide
  • Step A (1R,5S,6R)-3-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine-4 -yl)pyridin-2-yl)-3-azabicyclo[3.1.0]-6-hexylamino hydrochloride
  • Step B 2-Chloro-N-((1R,5S,6R)-3-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazole[1, 5-a]Pyridin-4-yl)pyridin-2-yl)-3-azabicyclo[3.1.0]hexyl-6-yl)-6-fluorobenzamide
  • Step A 2-Chloro-6-fluoro-N-(((3aR,5S,6aS)-octahydrocyclopenta[c]pyrrol-5-yl)benzamide hydrochloride
  • Step B 2-Chloro-N-(((3aR,5S,6aS)-2-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1 ,5-a]pyridin-4-yl)pyridin-2-yl)-octahydrocyclopentyl[c]pyrrolyl5-yl)-6-fluorobenzamide
  • Step A 2-Chloro-6-fluoro-N-(((3aR,5R,6aS)-octahydrocyclopenta[c]pyrrol-5-yl)benzamide hydrochloride
  • Step B 2-Chloro-N-(((3aR,5R,6aS)-2-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1 ,5-a]pyridin-4-yl)pyridin-2-yl)-octahydrocyclopentyl[c]pyrrolyl5-yl)-6-fluorobenzamide
  • Step A (2-Chloro-6-fluorophenyl)((3aR,7aS)-octahydro-1H-4,7-epiiminoisoindol-8-yl)methanone hydrochloride
  • Step B (2-Chloro-6-fluorophenyl)((3aR,7aS)-2-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazole [1,5-a]Pyridin-4-yl)pyridin-2-yl)octahydro-1H-4,7-epiiminoisoindol-8-yl)methanone
  • Step A (3aR,7aS)-8-(((6-methoxypyridin-3-yl)methyl)octahydro-1H-4,7-epiaminoisoindole hydrochloride
  • Step B 6-Ethoxy-4-(6-(((3aR,7aS)-8-(((6-methoxypyridin-3-yl)methyl)octahydro-2H-4,7- Epiaminoisoindol-2-yl)pyridin-3-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step A 3-Phenyl-6-ethyl-(1R,5S,6r)-3-azabicyclo[3.1.0]hexane-3,6-carboxylate
  • Step B (1R,5S,6r)-3-azabicyclo[3.1.0]hexane-6-ethyl carbonate
  • Step C (1R,5S,6r)-3-(5-Bromopyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-ethyl carbonate
  • Step D ((1R,5S,6r)-3-(5-bromopyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-yl)methanol
  • Step E tert-Butyl(((1R,5S,6r)-3-(5-bromopyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-yl)methyl)( tert-Butoxycarbonyl)carbamate
  • Step F tert-Butyl(((1R,5S,6r)-3-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5- a]Pyridin-4-yl)pyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-yl)methyl)(tert-butoxycarbonyl)carbamate
  • Step G ((1R,5S,6s)-3-(5-(6-ethoxy-1hydro-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine -4-yl)pyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-yl)methanamine hydrochloride
  • Step H 2-Chloro-N-(((1R,5S,6s)-3-(5-(6-ethoxy-1hydro-pyrazole[3',4':3,4]pyrazole[ 1,5-a]Pyridin-4-yl)pyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-yl)methyl)-6-fluorobenzamide
  • Step A N-(((1R,5S,6s)-3-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a ]pyridin-4-yl)pyridin-2-yl)-3-azabicyclo[3.1.0]hexane-6-yl)methyl)-1-(6-methoxypyridin-3-yl)- N-((6-Methoxypyridin-3-yl)methyl)methanamine
  • Step A tert-Butyl 5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,5-diazaspiro[3.4]octane-5-carboxylate
  • Step B 4-(6-(2,5-diazaspiro[3.4]octan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine
  • Step C (2-Chloro-6-fluorophenyl)-5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a ]pyridin-4-yl)pyridin-2-yl)-2,5-diazaspiro[3.4]octan-5-yl)methanone
  • Step A tert-Butyl 5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,6-diazaspiro[3.4]octane-6-carboxylate
  • Step B 4-(6-(2,6-diazaspiro[3.4]octan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C (2-Chloro-6-fluorophenyl)-5-(5-(6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a ]pyridin-4-yl)pyridin-2-yl)-2,6-diazaspiro[3.4]octan-6-yl)methanone
  • Step A 2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,7-diazaspiro[3.5]nonane-7-carboxylate tert-butyl ester
  • Step B 4-(6-(2,7-Diazaspiro[3.5]nonan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,7-diazaspiro[3.5]nonan-7-yl)-2-chloro-6-fluorobenzamide
  • Step A 2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,6-diazaspiro[3.5]nonane-6-carboxylate tert-butyl ester
  • Step B 4-(6-(2,6-Diazaspiro[3.5]nonan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,6-diazaspiro[3.5]nonan-6-yl)-2-chloro-6-fluorobenzamide
  • Step A 2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,8-diazaspiro[4.5]decane-8-carboxylate tert-butyl ester
  • Step B 4-(6-(2,8-Diazaspiro[4.5]decan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine
  • Step C N-(2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,8-diazaspiro[4.5]decan-8-yl)2-chloro-6-fluorobenzamide
  • Step A 2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,7-diazaspiro[4.5]octane-7-carboxylate tert-butyl ester
  • Step B 4-(6-(2,7-diazaspiro[4.5]octan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,7-diazaspiro[4.5]octan-7-yl)-2-chloro-6-fluorobenzamide
  • Step A 2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,6-diazaspiro[4.5]octane-6-carboxylate tert-butyl ester
  • Step B 4-(6-(2,6-diazaspiro[4.5]octan-2-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(2-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,6-diazaspiro[4.5]octan-6-yl)-2-chloro-6-fluorobenzamide
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-2-yl)pyridin-2-yl )-1,7-diazaspiro[3.5]nonane-1-carboxylate tert-butyl ester
  • Step B 4-(6-(1,7-diazaspiro[3.5]nonan-7-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-1,7-diazaspiro[3.5]nonan-1-yl)-2-chloro-6-fluorobenzamide
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,7-diazaspiro[3.5]nonane-2-carboxylate tert-butyl ester
  • Step B 4-(6-(2,7-Diazaspiro[3.5]nonan-7-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,7-diazaspiro[3.5]nonan-2-yl)-2-chloro-6-fluorobenzamide
  • Step A 8-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-1,8-diazaspiro[4.5]decane-1-carboxylate tert-butyl ester
  • Step B 4-(6-(1,8-Diazaspiro[4.5]decan-8-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(8-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-1,8-diazaspiro[4.5]decan-1-yl)2-chloro-6-fluorobenzamide
  • Step A 8-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,8-diazaspiro[4.5]decane-2-tert-butyl carbonate
  • Step B 4-(6-(2,8-Diazaspiro[4.5]decan-8-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine hydrochloride
  • Step C N-(8-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chloro-6-fluorobenzamide
  • Step A 6-Ethoxy-4-(6-(2-((6-methoxypyridin-3-yl)methylene)-2,8-diazaspiro[4.5]decane- 8-yl)pyridin-3-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,7-diazaspiro[4.5]decane-2-tert-butyl carbonate
  • Step B 4-(6-(2,7-diazaspiro[4.5]decan-7-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine
  • Step C N-(7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,7-diazaspiro[4.5]decan-2-yl)-2-chloro-6-fluorobenzamide
  • Step A 6-Ethoxy-4-(6-(2-((6-methoxypyridin-3-yl)methylene)-2,8-diazaspiro[4.5]decane- 8-yl)pyridin-3-yl)-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine
  • 1,2-Dichloroethane (2 mL), methanol (1 mL), 4-(6-(2,7-diazaspiro[4.5]decan-7-yl)pyridine- 3-yl)-6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine (63 mg, 0.15 mmol), 6-methoxy-3 -Pyridinecarboxaldehyde (21 mg, 0.15 mmol) and acetic acid (2 drops), heated at 45 °C for 1 hour, added sodium triacetoxyborohydride (96 mg, 0.45 mmol) and continued stirring at 45 °C for 16 hours.
  • Step A 7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-1,7-diazaspiro[4.5]decane-1-tert-butyl carbonate
  • Step B 4-(6-(1,7-diazaspiro[4.5]decan-7-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3',4 ':3,4]pyrazo[1,5-a]pyridine
  • Step C N-(7-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine -2-yl)-1,7-diazaspiro[4.5]decan-1-yl)-2-chloro-6-fluorobenzamide
  • Step A 9-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-2,9-diazaspiro[5.5]undecane-2-tert-butyl carbonate
  • Step B 4-(6-(2,9-diazaspiro[5.5]undecan-9-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3', 4':3,4]pyrazo[1,5-a]pyridine
  • Step C N-(9-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-2,9-diazaspiro[5.5]undecan-2-yl)-2-chloro-6-fluorobenzamide
  • 1,2-Dichloroethane (2 mL), methanol (1 mL), 4-(6-(2,9-diazaspiro[5.5]undecan-9-yl)pyridine were added to the reaction flask in sequence -3-yl)-6-ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridine (65 mg, 0.15 mmol), 6-methoxy- 3-Pyridinecarbaldehyde (62 mg, 0.45 mmol) and acetic acid (2 drops) were heated at 45°C for 2 hours, and then sodium triacetoxyborohydride (160 mg, 0.75 mmol) was added to continue stirring at 45°C for 16 hours.
  • Step A 9-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridin-2-yl )-3,9-diazaspiro[5.5]undecane-3-tert-butyl carbonate
  • Step B 4-(6-(3,9-diazaspiro[5.5]undecan-3-yl)pyridin-3-yl)-6-ethoxy-1H-pyrazolo[3', 4':3,4]pyrazo[1,5-a]pyridine
  • Step C N-(9-(5-(6-Ethoxy-1H-pyrazolo[3',4':3,4]pyrazolo[1,5-a]pyridin-4-yl)pyridine- 2-yl)-3,9-diazaspiro[5.5]undecan-3-yl)-2-chloro-6-fluorobenzamide

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

式I所示含氮多环稠环类化合物,其药物组合物、制备方法和用途。作为一种高度选择性和非常有效的RET抑制剂,此类化合物对RET看门残基突变体RET V804M突变、RET溶剂前沿残基突变体G810R和其他临床相关RET突变体以及RET-wt均有较强的抑制作用,该化合物还能显著抑制甲状腺癌来源的TT细胞系和各种RET突变体转化的Ba/F3细胞的生长,且抑制作用强于选择性RET抑制剂LOXO-292。此外,该化合物很大程度上阻断细胞RET自磷酸化及其下游通路,并能显著诱导TT细胞死亡。

Description

含氮多环稠环类化合物,其药物组合物、制备方法和用途
本申请要求享有2021年2月8日向中国国家知识产权局提交的申请号为202110172167.6,名称为“含氮多环稠环类化合物,其药物组合物、制备方法和用途”的中国发明专利在先申请的优先权。该在先申请的全文以引用的方式并入本文。
技术领域
本发明涉及药物化学领域,具体涉及含氮多环稠环类化合物,其药物组合物、制备方法和用途。
背景技术
研究发现,多种疾病的发生与RET基因突变有密切联系,包括甲状腺乳头状癌(papillary thyroid carcinoma,PTC)(Cell,1990,60(4):557-563)、甲状腺髓样癌(medullary thyroid carcinoma,MTC)(hyroid,2009,19(6):565-612)、多发性内分泌腺瘤病2型(multiple endocrineneoplasia typeⅡ,MEN2)(Endocr Rev,2006,27(5):535-560)、先天性巨结肠(Proc Natl Acad Sci USA,2000,97(1):268-273)、肺腺癌(Nat Med,2012,18(3):375-377)等。目前只有KIF5B-RET、CCDC6-RET、TRIM33-RET、NCOA4-RET这四种RET融合基因在非小细胞肺癌中被报道,而KIF5B-RET是非小细胞肺癌中最常见的RET融合基因(Cancer,2013,119(8):1486-1494)。KIF5B-RET是KIF5B(kinesin family member 5B)基因和RET基因的染色体倒置(p11;q11)形成的一种融合基因,通过全基因组和转录组测序,第一次在非吸烟韩国人的腺癌中被证实;KIF5B-RET在肺癌中的比例很低,在非吸烟者和腺癌患者中更常见,并与其它突变,如EGFR、KRAS、BRAF、ErbB2、EML4-ALK相排斥(Genome Res,2012,22(3):436-445)。KIF5B-RET融合蛋白包含马达结构域和KIF5B的卷曲螺旋结构域,通过卷曲螺旋结构域的二聚化作用,该融合蛋白的RET酪氨酸激酶活性可异常活化,从而促进肺肿瘤发生(Cancer,2011,117(12):2709-2718)。在Qian等的研究中(Mol Cancer,2014,13:176),KIF5B-RET融合激酶被证实在体外和体内都具有显着的致癌活性,STAT3的信号转导途径可能是肿瘤发生的主要下游介质。有证据显示KIF5B-RET可调节STAT3的持续活化。KIF5B-RET融合激酶可以结合STAT3,直接磷酸化和激活STAT3-Tyr705;它也可以通过JAK/STAT3依赖性途径,介导激活STAT3-Tyr705,并通过RAS/RAF/MEK/ERK1途径触发Ser727的磷酸化。
证明RET融合物在一些癌症中是驱动者,促进了已具有RET抑制活性的多激酶抑制剂(multi-kinase inhibitor)的应用,用于治疗负载RET融合物蛋白质的肿瘤病人。目前尚未有批准药剂可以用来针对性地靶向这一致癌基因,目前RET特异性癌症的治疗方式仅限于多激酶抑制剂和化疗,但这些非特异性治疗临床表现ORR(客观缓释率)不好并且有很大的脱靶毒性。再者,癌症治疗的最大挑战之一是肿瘤细胞对于治疗一定阶段后出现耐药,一旦耐药,病人的治疗方式通常极为有限,且大多数例子中,癌症一直进展、不受抑制。现已发现,在甲状腺癌等多种人类癌症中,RET激酶信号转导起着重要作用。其中RET的看门残基体RET804V突变是导致肿瘤对目前批准的非选择性RET抑制剂(如cabozantinib和vandetanib)耐药的重要原因。分离型家族性甲状腺髓样癌的RET胞外或胞内结构域中的重要突变之一,即激酶ATP结合位点中的看门残基V804M突变,导致现有药物对ATP结合位点亲和力下降。有文献报道(RET Solvent Front Mutations Mediate Acquired Resistance to Selective RET Inhibition in RET-Driven Malignancies,Journal of Thoracic Oncology,2020,Vol.15,No 4,541-549)选择性RET抑制剂LOXO-292(Selpercatinib)虽然可以避免前面提到的RET 804V突变,但在使用该选择性RET抑制剂后,仍会出现其他突变而导致耐药,比如在非小细胞肺癌中会导致激酶ATP结合位点中溶剂前沿的残基G810突变:如G810R,G810S,G801C突变,导致LOXO-292对ATP结合位点下降,从而出现耐药,癌症发生进展,因此,需要开发性能得到改善的RET 突变抑制剂。
发明内容
为改善上述问题,本发明提供一种如下式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐:
Figure PCTCN2022073467-appb-000001
其中,Q选自
Figure PCTCN2022073467-appb-000002
其中,X 1、X 2、X 3、X 4、X 5、X 6、X 7相同或不同,彼此独立地选自CR 1或N;
X 8选自CR 1R 1’或NR 1
其中每一个R 1和R 1’相同或不同,彼此独立地选自H、卤素、CN、OH,无取代或任选被一个、两个或更多个独立选自R a的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
A选自H、卤素、CN、OH,NH 2,无取代或任选被一个、两个或更多个独立选自R b的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
D、E相同或不同,彼此独立地选自H、卤素、CN、OH、B(OH) 2、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7、-O-R 21、C(O)OR 22、-P(O)R 23R 24,无取代或任选被一个、两个或更多个独立选自R c的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、NH 2,条件是D和E中的至少一个不为H,例如D和E中的至少一个选自-O-R 21或D和E中的至少一个选自无取代或任选被一个、两个或更多个独立选自R c的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、NH 2
R 21、R 22、R 23、R 24相同或不同,彼此独立地选自H,无取代或任选被一个、两个或更多个独立选自R d的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基;
G选自卤素、无取代或任选被一个、两个或更多个独立选自R e的取代基取代的下列基团:至少一个杂原子选自N的5-20元杂芳基、至少一个杂原子选自N的3-20元杂环基、C 3-40环烷基-NH-;
K选自不存在、H、卤素、CN、OH,无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔 基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
每一个R 2相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、-C(O)R 4、-S(O) 2R 6
每一个R 3相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、-C(O)R 4、-S(O) 2R 6
或者,R 2和R 3与所连的N原子一起形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:5-20元杂芳基或3-20元杂环基;
每一个R 4相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3
每一个R 5相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基羰基、C 2-40烯基羰基、C 2-40炔基羰基、C 3-40环烷基羰基、C 3-40环烯基羰基、C 3-40环炔基羰基、C 6-20芳基羰基、5-20元杂芳基羰基、3-20元杂环基羰基;
每一个R 6相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3
每一个R 7相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基;
m为0、1、2、3、4、5、6、7或8;
每一个R 01、R 02、R 03、R 04相同或不同,彼此独立地选自H、卤素、CN、OH、SH、氧代(=O)、NO 2,无取代或任选被一个、两个或更多个独立选自R g的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
每一个R a、R b、R c、R d、R e、R f相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2,无取代或任选被一个、两个或更多个独立选自R g的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
每一个R g相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2,无取 代或任选被一个、两个或更多个独立选自R h的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7;或者,当环状基团(包括但不限于C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、3-20元杂环基等)的不同位置被两个或更多个取代基取代时,所述取代基中的两个也可以与所述环状基团形成桥环,其中所述桥环中除桥头原子之外的桥原子可以包含1、2、3、4或5个选自CH 2、O、NH的二价基团;
每一个R h相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2、无取代或任选被一个、两个或更多个独立选自R i的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
每一个R i相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2、无取代或任选被一个、两个或更多个R j的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
或者,当环状基团(包括但不限于C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、3-20元杂环基等)的不同位置被两个或更多个取代基取代时,所述取代基中的两个也可以与所述环状基团形成桥环,其中所述桥环中除桥头原子之外的桥原子可以包含1、2、3、4或5个选自CH 2、O、NH的二价基团;
或者,当一个原子(如碳原子或氮原子)被两个或更多个取代基取代时,所述取代基中的两个也可以与其共同连接的原子形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:无取代或任选被一个、两个或更多个独立选自R f的取代基取代环状基团(包括但不限于无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、5-20元杂芳基、3-20元杂环基等)。
例如,当NH 2被两个取代基R c取代时,所述两个取代基R c可以与它们共同链接的氮原子一起形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的5-20元杂芳基或3-20元杂环基,例如无取代或任选被一个、两个或更多个独立选自R f取代的5、6或7元杂芳基或无取代或任选被一个、两个或更多个独立选自R f取代的3、4、5、6或7元杂环基;
根据本发明的实施方案,X 1、X 2、X 3、X 4、X 5、X 6、X 7相同或不同,彼此独立地选自CR 1或N;例如,X 1,X 2,X 3,X 4、X 5、X 6、X 7中的至少一个为N,例如1、2、3、4、5、6或7个为N;
根据本发明的实施方案,X 8选自CR 1R 1’或NR 1
根据本发明的实施方案,每一个R 1和R 1’相同或不同,彼此独立地选自H、卤素、CN、OH、C 1-6烷基、C 3-10环烷基、C 1-6烷基氧基;
根据本发明的实施方案,A选自H、卤素、CN、OH、C 1-6烷基、C 1-6烷基氧基;
根据本发明的实施方案,D、E相同或不同,彼此独立地选自H、卤素、CN、无取代或任选被一个、两个或更多个独立选自R c的取代基取代的NH 2或-O-R 21,条件是D和E中的至少一个不为H,例如D和E中的至少一个选自-O-R 21或无取代或任选被一个、两个或更多个独立选自R c的取代基取代的NH 2
根据本发明的实施方案,R 21选自无取代或任选被一个、两个或更多个被R d的取代基取 代的C 1-6烷基;
根据本发明的实施方案,R 2和R 3可以与所连的N原子一起形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:5-20元杂芳基或3-20元杂环基,例如5、6或7元杂芳基或3、4、5、6或7元杂环基;
根据本发明的实施方案,每一个R 01、R 02、R 03、R 04相同或不同,彼此独立地选自H、C 1-6烷基、C 1-6烷基氧基;
根据本发明的实施方案,每一个R a、R b、R c、R d、R e、R f相同或不同,彼此独立地选自卤素、NH 2、CN、OH,无取代或任选被一个、两个或更多个R g的取代基取代的下列基团:C 1-6烷基、C 1-6烷基氧基、C 3-10环烷基、C 3-10环烷基氧基、C 2-6炔基氧基、3-8元杂环基、3-8元杂芳基;
根据本发明的实施方案,每一个R g相同或不同,彼此独立地选自OH、卤素或C 3-10环烷基;
根据本发明的实施方案,G选自卤素、C 3-10环烷基、C 3-10环烷基-NH-、C 6-14芳基、5-14元杂芳基、3-12元杂环基,例如6-12元具有单环、双环、三环或桥环结构的杂环基,其可包含1、2、3或4个独立选自N、O和S的杂原子,条件是其中至少一个杂原子选自N,例如1、2或3个杂原子选自N。
根据本发明的实施方案,K选自-C 1-6烷基-C 3-10环烷基、-C 1-6烷基-C 6-14芳基、-C 1-6烷基-5-14元杂芳基、-C 1-6烷基-3-10元杂环基、-C(O)NH 2、-C(O)-C 3-10环烷基、-C(O)-C 6-14芳基、-C(O)-5-14元杂芳基、-C(O)-3-10元杂环基、-C(O)-C 1-6烷基-C 3-10环烷基、-C(O)-C 1-6烷基-C 6-14芳基、-C(O)-C 1-6烷基-5-14元杂芳基、-C(O)-C 1-6烷基-3-10元杂环基,其中所述C 3-10环烷基、C 6-14芳基、5-14元杂芳基、3-10元杂环基、-C(O)-C 3-10环烷基、-C(O)-C 6-14芳基、-C(O)-5-14元杂芳基、-C(O)-3-10元杂环基、-C(O)-C 1-6烷基-C 3-10环烷基、-C(O)-C 1-6烷基-C 6-14芳基、-C(O)-C 1-6烷基-5-14元杂芳基、-C(O)-C 1-6烷基-3-10元杂环基的环上或非环基团,或-C(O)NH 2还任选被一个、两个或更多个选自OH、卤素、CN、C 1-6烷基、C 1-6烷基氧基的基团取代;其中,所述杂环基可以为吡啶基(例如吡啶-2-基、吡啶-3-基、吡啶-4-基、吡啶-5-基、吡啶-6-基),芳基可以为苯基;
或者,K选自不存在、H、OH、无取代或任选被一个、两个或更多个独立选自R f取代的下列基团:苯基-C(O)-、苯基-C(O)-NH-、苯基-C(O)-NH-C 1-6烷基-、苯基氧基-C(O)-NH-、苯基烷基-NH-C(O)-、苯基烷基-C(O)-NH-、吡啶基-C(O)-、吡啶基-C(O)-NH-、吡啶基-C(O)-NH-C 1-6烷基-、吡啶基氧基-C(O)-NH-、吡啶基烷基-NH-C(O)-、吡啶基烷基-C(O)-NH-、吡咯烷基-C(O)-NH-、吡咯烷基氧基-C(O)-NH-、C 1-6烷基-C(O)-、C 1-6烷基-C(O)-NH-、C 1-6烷基氧基-C(O)-NH-、C 3-8环烷基-C(O)-NH-、C 3-8环烷基氧基-C(O)-NH-、吡啶基-NH-C(O)-、C 1-6烷基-NH-C(O)-、C 3-8环烷基-NH-C(O)-、吡啶基氧基-、吡啶基烷基氧基-、吡啶基氧基烷基-、苯基氧基-、苯基烷基氧基-、苯基氧基烷基-、C 1-6烷基-S(O) 2-、C 1-6烷基-S(O) 2-NH-、C 1-6烷基-NH-S(O) 2-、吡啶基C 1-6烷基-、吡啶基-S(O) 2-、吡啶基-C 1-6烷基-S(O) 2-、吡啶基-S(O) 2-NH-、吡啶基-NH-S(O) 2-、吡啶基-C 1-6烷基-NH-、苯基C 1-6烷基-、苯基-S(O) 2-、苯基-C 1-6烷基-S(O) 2-、苯基-S(O) 2-NH-、苯基-NH-S(O) 2-、苯基-C 1-6烷基-NH-、C 1-6烷基氧基-C(O)-、
Figure PCTCN2022073467-appb-000003
根据本发明示例性的实施方案,其中X 1、X 2、X 3、X 4、X 5、X 6、X 7相同或不同,彼此独立地选自CH或N;例如,X 1,X 2,X 3,X 4、X 5、X 6、X 7中的至少一个为N,例如1、2、3、4、5、6或7个为N;
根据本发明示例性的实施方案,X 8选自NR 1
根据本发明示例性的实施方案,R 01为甲氧基;
根据本发明示例性的实施方案,R 02为H;
根据本发明示例性的实施方案,R 03为甲基,R 04为H;
根据本发明示例性的实施方案,R 1为H;
根据本发明示例性的实施方案,A选自H、NH 2、甲基、乙基、丙基、异丙基;
根据本发明示例性的实施方案,E选自H、NH 2
根据本发明示例性的实施方案,D选自如下基团:卤素、BnO-、H、CN、NH 2、OCH 3、COOH、B(OH) 2
Figure PCTCN2022073467-appb-000004
根据本发明示例性的实施方案,G选自F、无取代或任选被一个、两个或更多个独立选自R e的取代基取代的下列基团:
Figure PCTCN2022073467-appb-000005
Figure PCTCN2022073467-appb-000006
Figure PCTCN2022073467-appb-000007
根据本发明示例性的实施方案,当基团G被R e取代时,R e可以取代构成基团G的-CH 2-、-CH=上的H,形成例如选自下列的基团:
Figure PCTCN2022073467-appb-000008
根据本发明的实施方案,除非另有说明,使用波浪线标记的化学键表示与其他基团的连接位点。例如,当基团G中存在两个波浪线时,其中任一个均可与X 1、X 2、X 3和X 4所在的环连接,而另一个则在基团K存在时与基团K连接。
根据本发明示例性的实施方案,当基团G的成环原子包含碳原子和氮原子时,基团G可以通过碳原子和氮原子中的一种(例如上述示例性基团中标记有波浪线的成环原子中的一个)与X 1、X 2、X 3和X 4所在的环连接,并通过碳原子和氮原子中的另一种与基团K连接。
根据本发明示例性的实施方案,K选自不存在、H、OH、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:苯基-C(O)-、苯基-C(O)-NH-、苯基-C(O)-NH-C 1-6烷基-、苯基氧基-C(O)-NH-、苯基烷基-NH-C(O)-、苯基烷基-C(O)-NH-、吡啶基-C(O)-、吡啶基-C(O)-NH-、吡啶基-C(O)-NH-C 1-6烷基-、吡啶基氧基-C(O)-NH-、吡啶基烷基-NH-C(O)-、吡啶基烷基-C(O)-NH-、吡咯烷基-C(O)-NH-、吡咯烷基氧基-C(O)-NH-、C 1-6烷基-C(O)-、C 2-6烯基-C(O)-、C 1-6烷基-C(O)-NH-、C 1-6烷基氧基-C(O)-NH-、C 3-8环烷基-C(O)-NH-、C 3-8环烷基氧基-C(O)-NH-、吡啶基-NH-C(O)-、C 1-6烷基-NH-C(O)-、C 3-8环烷基-NH-C(O)-、吡啶基氧基-、吡啶基烷基氧基-、吡啶基氧基烷基-、苯基氧基-、苯基烷基氧基-、苯基氧基烷基-、C 1-6烷基-S(O) 2-、C 1-6烷基-S(O) 2-NH-、C 1-6烷基-NH-S(O) 2-、吡啶基C 1-6烷基-、吡啶基-S(O) 2-、吡啶基-C 1-6烷基-S(O) 2-、吡啶基-S(O) 2-NH-、吡啶基-NH-S(O) 2-、吡啶基-C 1-6烷基-NH-、苯基C 1-6烷基-、苯基-S(O) 2-、苯基-C 1-6烷基-S(O) 2-、苯基-S(O) 2-NH-、苯基-NH-S(O) 2-、苯基-C 1-6烷基-NH-、C 1-6烷基氧基-C(O)-、
Figure PCTCN2022073467-appb-000009
Figure PCTCN2022073467-appb-000010
根据本发明的实施方案,所述式I所示的化合物具有下式II或III所示的结构:
Figure PCTCN2022073467-appb-000011
其中,X 1、X 2、X 3、X 4、X 5、X 6、X 7、X 8、R 01、R 02、R 03、R 04、A、D、E、G、K、m具有上文所述的定义。
根据本发明的实施方案,所述式I所示的化合物具有下式IV或V所示的结构:
Figure PCTCN2022073467-appb-000012
其中,X 1、X 2、X 3、X 4、X 5、R 01、R 03、R 04、A、D、E、G、K具有上文所述的定义。
根据本发明的实施方案,所述化合物选自如下化合物:
Figure PCTCN2022073467-appb-000013
Figure PCTCN2022073467-appb-000014
Figure PCTCN2022073467-appb-000015
Figure PCTCN2022073467-appb-000016
Figure PCTCN2022073467-appb-000017
Figure PCTCN2022073467-appb-000018
Figure PCTCN2022073467-appb-000019
Figure PCTCN2022073467-appb-000020
Figure PCTCN2022073467-appb-000021
Figure PCTCN2022073467-appb-000022
Figure PCTCN2022073467-appb-000023
Figure PCTCN2022073467-appb-000024
Figure PCTCN2022073467-appb-000025
Figure PCTCN2022073467-appb-000026
Figure PCTCN2022073467-appb-000027
Figure PCTCN2022073467-appb-000028
Figure PCTCN2022073467-appb-000029
Figure PCTCN2022073467-appb-000030
Figure PCTCN2022073467-appb-000031
Figure PCTCN2022073467-appb-000032
Figure PCTCN2022073467-appb-000033
Figure PCTCN2022073467-appb-000034
Figure PCTCN2022073467-appb-000035
Figure PCTCN2022073467-appb-000036
Figure PCTCN2022073467-appb-000037
Figure PCTCN2022073467-appb-000038
本发明还提供式I所示化合物的制备方法,包括如下步骤:
Figure PCTCN2022073467-appb-000039
其中,A、D、E、Q、G、K、X 5、X 6、X 7、X 8具有如上所述的定义;L 1选自离去基团。
或者,所述制备方法包括如下步骤:
Figure PCTCN2022073467-appb-000040
其中,A、D、E、Q、G、K、X 5、X 6、X 7、X 8具有如上所述的定义;L 2选自离去基团。
或者,所述制备方法包括如下步骤:
Figure PCTCN2022073467-appb-000041
其中,A、D、E、Q、G、K、X 5、X 6、X 7、X 8具有如上所述的定义;L 3选自离去基团。
或者,所述制备方法包括将被保护基团取代的式I化合物在脱去所述保护基团的条件下反应,得到式I化合物。其中,根据式I化合物中取代基的不同,所述保护基团可以选自羟基保护基团、氨基保护基团等中的至少一种。
本发明还提供式II所示化合物的制备方法,包括如下步骤:
式II-1化合物与化合物R 21-L反应得到式II化合物,
Figure PCTCN2022073467-appb-000042
其中,A、E、G、K、X 1、X 2、X 3、X 4、X 5、X 6、X 7、X 8、R 21具有如上所述的定义;
D选自-O-R 21;L选自离去基团。
根据本发明的实施方案,所述离去基团选自OH、卤素或OTf。
根据本发明的实施方案,所述反应在碱的存在下进行,例如在碳酸钾的存在下进行。
根据本发明的实施方案,所述反应的温度为50~100℃,反应时间为1~24小时。
根据本发明的实施方案,所述反应可以在有机溶剂(如DMF)的存在下进行。
本发明还提供式II-1化合物的制备方法,包括由式II-2化合物反应制备式II-1化合物:
Figure PCTCN2022073467-appb-000043
其中,A、D、E、G、K、X 1、X 2、X 3、X 4、X 5、X 6、X 7、X 8具有如上所述的定义。
根据本发明的实施方案,所述反应在水合肼的存在下进行,且X 7为N,X 8为NH。
根据本发明的实施方案,所述反应在有机溶剂(如DMF)的存在下进行。
根据本发明的实施方案,所述反应在加热条件下进行。
本发明还提供上述式I-1、式I-2、式I-3、式II-1或式II-2所示的化合物。
本发明还提供式I-1、式I-2、式I-3或式II-1所示的化合物用于制备式I所示化合物的用途。
本发明还提供一种药物组合物,其包含选自式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐中的至少一种,例如治疗有效量的选自式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐中的至少一种。
根据本发明的实施方案,所述药物组合物还包含一种、两种或多种药学上可接受的辅料,如载体和/或赋形剂。
根据本发明的实施方案,所述药物组合物还进一步含有一种或多种额外的治疗剂。
本发明还提供体外或体内抑制细胞增殖的方法,所述方法包括使细胞与有效量的本发明所述式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐,或其药物组合物接触。
本发明还提供一种治疗RET基因和/或RET激酶介导的疾病的方法,包括给予患者治疗有效量的如式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐中的至少一种。
根据本发明的实施方案,当描述RET、RET基因或RET激酶时,其表示选自包括但不限于RET-wt、V804M、V804L、V804E、G810R、G810S、G810C、G810V和S904F的RET基因或RET激酶。
本发明还提供了在有治疗需要的患者中治疗RET相关疾病或病症的方法,所述方法包括 向所述患者施用治疗有效量的本发明式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐或其药物组合物。
本发明还提供了在有治疗需要的患者中治疗癌症和/或抑制与特定癌症相关的转移的方法,所述方法包括向所述患者施用治疗有效量的本发明式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐或其药物组合物。
本发明还提供了在有治疗需要的患者中治疗肠易激综合征(IBS)和/或与IBS相关的疼痛的方法,所述方法包括向所述患者施用治疗有效量的本发明式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐或其药物组合物。
本发明还提供了为癌症患者提供支持护理的方法,包括预防或最小化与治疗(包括化疗治疗)相关的胃肠疾病(例如腹泻),所述方法包括给予患者治疗有效量的本发明式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐或其药物组合物。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备药物中的用途,其中所述药物用于治疗RET激酶介导的疾病、抑制RET激酶活性、治疗癌症和/或抑制与特定癌症相关的转移、治疗肠易激综合征(IBS)或与IBS相关的疼痛、向癌症患者提供支持护理、治疗RET相关疾病或病症、逆转或预防对抗癌药物的获得性抗性、延迟和/或预防个体中抗癌药抗药性发展、对抗癌药物发展抗性的可能性增加。
根据本发明的实施方案,所述RET相关疾病或病症选自RET基因和/或RET激酶介导的疾病,其中RET、RET基因或RET激酶选自包括但不限于RET-wt、V804M、V804L、V804E、G810R、G810S、G810C、G810V和S904F的RET基因或RET激酶。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备用于治疗RET激酶介导的疾病的药物中的用途。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备用于治疗癌症和/或抑制与特定癌症相关的转移的药物中的用途。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备用于治疗肠易激综合征(IBS)或与IBS相关的疼痛的药物中的用途。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备用于向癌症患者提供支持护理的药物中的用途,所述支持护理包括预防或最小化与治疗(包括化疗治疗)相关的胃肠病症,例如腹泻。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备用于抑制RET激酶活性的药物中的用途。
本发明还提供式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备用于治疗RET相关疾病或病症的药物中的用途。
本发明还提供用于在有需要的患者中治疗癌症的方法,所述方法包括(a)确定所述癌症是否与下述的失调有关:RET基因、RET激酶、或其中任何一者的表达或活性或水平(例如,RET相关的癌症);(b)如果确定所述癌症与下述的失调有关:RET基因、RET激酶、或其中任何一者的表达或活性或水平(例如,RET相关的癌症),向患者施用治疗有效量的式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种,或其药物组合物。
本发明还提供了用于逆转或预防对抗癌药物的获得性抗性的方法,所述方法包括将治疗有效量的式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种给予处于对抗癌药物发展或具有获得性抗性的风险的 患者。
本发明还提供延迟和/或预防个体中抗癌药抗药性发展的方法,所述方法包括在个体中施用有效量的式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种,在此之前、期间或之后施用有效量的抗癌药物。
本发明还提供了治疗患有癌症且对抗癌药物发展抗性的可能性增加的个体的方法,其包括对个体伴随施用(a)有效量的式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种;和(b)有效量的抗癌药物。
本发明还提供了治疗患有RET相关癌症的个体的方法,所述癌症具有一种或多种RET抑制剂抗性突变,所述RET抑制剂抗性突变增加所述癌症对不是式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种的RET抑制剂的抗性(例如,RET-wt、在氨基酸位置804,810,904处的取代,例如V804M、V804L、V804E、G810R、G810S、G810C、G810V、S904F),其包括在给予另一种其他的抗癌药物之前、期间或之后,给予式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种。
本发明还提供了治疗患有RET相关癌症的个体的方法,所述方法包括在给予另一种其他的抗癌药物之前、期间或之后,给予式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种。
本发明提供了在有需要的患者中治疗癌症(例如RET相关癌症)的方法,所述方法包括向所述患者施用治疗有效量的通式I的化合物或其或药学上可接受的盐中的至少一种或其药物组合物。
在本发明所述的任何方法或用途的一些实施方案中,癌症(例如RET相关癌症)是血液学癌症。在本发明所述的任何方法或用途的一些实施方案中,癌症(例如RET相关癌症)是实体瘤。在本发明所述的任何方法或用途的一些实施方案中,癌症(例如RET相关癌症)是肺癌(例如,小细胞肺癌或非小细胞肺癌),乳头状甲状腺癌,甲状腺髓样癌,分化型甲状腺癌,复发性甲状腺癌,难治性分化型甲状腺癌,肺腺癌,细支气管肺癌,2A或2B型多发性内分泌肿瘤(分别为MEN2A或MEN2B),嗜铬细胞瘤,甲状旁腺增生,乳腺癌,结直肠癌(例如转移性结肠直肠癌),乳头状肾细胞癌,胃肠粘膜的神经节细胞瘤病,炎性肌纤维母细胞瘤或宫颈癌。在本发明所述的任何方法或用途的一些实施方案中,癌症(例如RET相关癌症)选自:急性淋巴细胞白血病(ALL),急性髓性白血病(AML),青少年癌症,肾上腺皮质癌,肛门癌、阑尾癌,星形细胞瘤,非典型性畸胎瘤/横纹肌样瘤,基底细胞癌,胆管癌,膀胱癌,骨癌,脑干胶质瘤,脑肿瘤,乳腺癌,支气管肿瘤,伯基特淋巴瘤,类癌瘤,未知原发癌,心脏肿瘤,宫颈癌,儿童癌症,脊索瘤,慢性淋巴细胞白血病(CLL),慢性骨髓性白血病(CML),慢性骨髓增殖性肿瘤,结肠癌,结肠直肠癌,颅咽管瘤,皮肤T细胞淋巴瘤,胆管癌,原位导管癌,胚胎性肿瘤,子宫内膜癌,室管膜瘤,食道癌,成感觉神经细胞瘤,尤因肉瘤,颅外生殖细胞肿瘤,性腺外生殖细胞瘤,肝外胆管癌,眼癌,输卵管癌,骨纤维组织细胞瘤,胆囊癌,胃癌,胃肠类癌瘤,胃肠道间质瘤(GIST),生殖细胞瘤,妊娠滋养细胞疾病,神经胶质瘤,多毛细胞瘤,多毛细胞白血病,头颈癌,心脏癌,肝细胞癌,组织细胞增多症,霍奇金淋巴瘤,下咽癌,眼内黑色素瘤,胰岛细胞瘤,胰腺神经内分泌瘤,卡波西肉瘤,肾癌,朗格汉斯细胞组织细胞增多症,喉癌,白血病,唇和口腔癌,肝癌,肺癌,淋巴瘤,巨球蛋白血症,骨噁性纤维组织细胞瘤,骨癌,黑色素瘤,梅克尔细胞癌,间皮瘤,转移性鳞状颈癌,中线状癌,口癌,多发性内分泌瘤综合征,多发性骨髓瘤,真菌病蕈样肉芽肿,骨髓增生异常综合征,骨髓增生异常/骨髓增殖性肿瘤,髓性白血病,骨髓性白血病,多发性骨髓瘤,骨髓增殖性肿瘤,鼻腔和鼻窦癌,鼻咽癌,成神经细胞瘤,非霍奇金淋巴瘤,非小细胞肺癌,口部癌,口腔癌,唇癌,口咽癌,骨肉瘤,卵巢癌,胰腺癌,乳头状瘤病,副神经节瘤,鼻旁窦和鼻腔癌,甲状旁腺癌,阴茎癌,咽癌,嗜铬细胞瘤,垂体癌,浆细胞瘤,胸膜肺胚细胞瘤,妊娠和乳腺癌,原发性中枢神经系统淋巴瘤,原发腹膜癌,前列腺癌,直肠癌,肾细胞癌,视网膜母细胞瘤,横纹肌肉瘤,唾液腺癌,肉瘤,塞扎里综合征,皮肤癌,小细胞肺癌,小肠 癌,软组织肉瘤,鳞状细胞癌,鳞状颈癌,胃癌,T细胞淋巴瘤,睾丸癌,咽喉癌,胸腺瘤和胸腺癌,甲状腺癌症,肾盂和输尿管的移行细胞癌,未知原发癌,尿道癌,子宫癌,子宫肉瘤,阴道癌,外阴癌和威尔姆氏瘤。
在一些实施方案中,血液学癌症(例如,与RET相关的癌症的血液学癌症)选自白血病,淋巴瘤(非霍奇金淋巴瘤),霍奇金病(也称霍奇金淋巴瘤)和骨髓瘤,例如,急性淋巴细胞白血病(ALL),急性骨髓性白血病(AML),急性早幼粒细胞白血病(APL),慢性淋巴细胞白血病(CLL),慢性粒细胞白血病(CML),慢性髓单核细胞白血病(CMML),慢性嗜中性白血病(CNL),急性未分化型白血病(AUL),间变性大细胞淋巴瘤(ALCL),幼淋巴细胞白血病(PML),幼年型单核细胞白血病(JMML),成人T细胞ALL,三联型骨髓发育不良的AML(AML/TMDS),混合谱系白血病(MLL),骨髓增生异常综合征(MDS),骨髓增殖性疾病(MPD)和多发性骨髓瘤(MM)。血液癌症的其它示例包括骨髓增殖性疾病(MPD),如真性红细胞增多症(PV),原发性血小板减少症(ET)和特发性原发性骨髓纤维化(IMF/IPF/PMF)。在一个实施方案中,血液学癌症(例如,作为与RET相关癌症的血液学癌症)是AML或CMML。
在一些实施方案中,癌症(例如RET相关癌症)是实体瘤。实体瘤(例如,作为与RET相关癌症的实体瘤)的示例包括例如甲状腺癌(例如乳头状甲状腺癌,甲状腺髓样癌),肺癌(例如肺腺癌,小细胞肺癌),胰腺癌,胰腺导管癌,乳腺癌,结肠癌,结直肠癌,前列腺癌,肾细胞癌,头颈肿瘤,神经母细胞瘤和黑素瘤。参见,例如,NatureReviewsCancer,2014,14,173-186。
在一些实施方案中,癌症选自肺癌,乳头状甲状腺癌,甲状腺髓样癌,分化的甲状腺癌,复发性甲状腺癌,难治性分化型甲状腺癌,2A或2B型多发性内分泌瘤(分别为MEN2A或MEN2B),嗜铬细胞瘤,甲状旁腺增生,乳腺癌,结直肠癌,乳头状肾细胞癌,胃肠粘膜神经节细胞瘤和宫颈癌。
在一些实施方案中,所述患者是人。
式I化合物及其药学上可接受的盐也可用于治疗RET相关的癌症。
本发明还提供用于在有需要的患者中治疗肠易激综合征(IBS)的方法,所述方法包括(a)确定IBS是否与下述的失调有关:RET基因、RET激酶、或其中任何一者的表达或活性或水平;(b)如果确定IBS与下述的失调有关:RET基因、RET激酶、或其中任何一者的表达或活性或水平,向患者施用治疗有效量的通式I化合物或其或药学上可接受的盐中的至少一种,或其药物组合物。
本发明还提供了用于治疗有需要的患者的肠易激综合征(IBS)的药物组合,其包括施用(a)式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种,(b)其他治疗剂,和(c)任选的至少一种药学上可接受的运载体,用于同时、分开或依次用于治疗IBS,其中式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种的量和其他治疗剂的量在治疗IBS方面共同有效。本发明还提供了包含这种组合的药物组合物。本发明还提供了这种组合在制备用于治疗IBS的药物中的用途。本发明还提供了商业包装或产品,其包含这种组合作为用于同时、单独或顺序使用的组合制剂;并涉及一种治疗有需要的患者的IBS的方法。
作为药物时,可按药物组合物的形式给予本发明化合物。可按药剂领域中熟知的方式制备这些组合物,可通过多种途径给予它们,这取决于是否需要局部或全身治疗和所治疗的区域。可局部(例如,透皮、皮肤、眼和粘膜包括鼻内、阴道和直肠递药)、肺(例如,通过吸入或吹入粉末或气雾剂,包括通过喷雾器;气管内、鼻内)、口服或肠胃外给药。肠胃外给药包括静脉内、动脉内、皮下、腹膜内或肌内注射或输注;或颅内例如鞘内或脑室内给药。可按单次大剂量形式肠胃外给药,或可通过例如连续灌注泵给药。局部给予的药用组合物和制剂可包括透皮贴剂、软膏、洗剂、霜剂、凝胶剂、滴剂、栓剂、喷雾剂、液体剂和散剂。常规药物载体、水、粉末或油性基质、增稠剂等可能是必须的或需要的。包衣避孕套(Coated condoms)、手套等也可以是有用的。
在制备本发明的组合物时,通常将活性成分与赋形剂混合,通过赋形剂稀释或装入例如 胶囊、小药囊、纸或其它容器形式的这种载体内。当赋形剂用作稀释剂时,它可以是固体、半固体或液体物质,用作溶媒、载体或活性成分的介质。因此,组合物可以是以下形式:片剂、丸剂、散剂、锭剂、小药囊、扁囊剂、酏剂、混悬剂、乳剂、溶液剂、糖浆剂、气雾剂(固体或溶于液体溶媒);含例如高达10%重量活性化合物的软膏剂、软和硬明胶胶囊、栓剂、无菌注射溶液和无菌包装粉末。
适宜的赋形剂的某些实例包括乳糖、葡萄糖、蔗糖、山梨醇、甘露醇、淀粉、阿拉伯胶、磷酸钙、藻酸盐、黄蓍胶、明胶、硅酸钙、微晶纤维素、聚乙烯吡咯烷酮、纤维素、水、糖浆和甲基纤维素。制剂还可含有:润滑剂例如滑石粉、硬脂酸镁和矿物油;湿润剂;乳化剂和悬浮剂;防腐剂例如苯甲酸甲酯和苯甲酸羟基丙酯;甜味剂和矫味剂。可通过使用本领域中已知的方法配制本发明组合物,以便在给予患者后提供速释、缓释或延迟释放活性成分的作用。
可按单位剂型配制组合物,每一剂量含约5~1000mg,更通常约100~500mg活性成分。术语“单位剂型”是指物理上分离的适宜作为用于人患者和其它哺乳动物的单一剂量单位,各单位含有与适宜的药物赋形剂混合的经计算可产生所需疗效的预定量的活性物质。
活性化合物的有效剂量的范围可很大,通常按药用有效量给药。但是,可以理解实际给予的化合物的量通常由医师根据相关情况决定,它们包括所治疗的病症、所选择的给药途径、所给予的实际化合物;患者个体的年龄、重量和反应;患者症状的严重程度等。
对于制备固体组合物例如片剂,将主要的活性成分与药物赋形剂混合,形成含本发明化合物的均匀混合物的固体预制剂组合物。当称这些预制剂组合物为均匀时,是指活性成分通常均匀地分布在整个组合物中,致使该组合物可容易地划分为同等有效的单位剂型例如片剂、丸剂和胶囊剂。然后将该固体预制剂划分为上述类型的含例如约0.1~1000mg本发明活性成分的单位剂型。
可将本发明片剂或丸剂包衣或复合,得到提供长效作用优点的剂型。例如,片剂或丸剂含内剂量和外剂量组分,后者是前者的被膜形式。可通过肠溶层将两种组分隔离,肠溶层用于在胃中阻止崩解,以使内组分完整通过十二指肠或延迟释放。多种物质可用于此类肠溶层或包衣剂,此类物质包括多种高分子酸和高分子酸与此类物质如虫胶、鲸蜡醇和醋酸纤维素的混合物。
其中可掺入本发明化合物和组合物,用于口服或注射给药的液体形式包括水溶液、适当矫味的糖浆剂、水或油混悬液;和用食用油例如棉子油、芝麻油、椰子油或花生油矫味的乳剂;以及酏剂和类似的药用溶媒。
用于吸入或吹入的组合物包括溶于药学上可接受的水或有机溶剂或其混合物的溶液剂和混悬液、散剂。液体或固体组合物可含有如上所述适宜的药学上可接受的赋形剂。在某些实施方案中,通过口服或鼻呼吸途径给予组合物,实现局部或全身作用。可通过使用惰性气体,使组合物成雾化。可直接由雾化装置吸入雾化溶液,或雾化装置可与面罩帷或间歇正压呼吸机连接。可通过口服或由按适当方式递送制剂的装置通过鼻给予溶液、混悬液或粉末组合物。
给予患者的化合物或组合物的量不固定,取决于给予的药物、给药的目的例如预防或治疗;患者的状态、给药的方式等。在治疗应用时,可给予已患疾病的患者足够治愈或至少部分抑制疾病及其并发症症状的量的组合物。有效剂量应取决于所治疗的疾病状态和主治临床医师的判断,该判断取决于例如疾病的严重程度、患者的年龄、体重和一般状况等因素。
给予患者的组合物可以是上述药用组合物形式。可通过常规灭菌技术或可过滤灭菌,将这些组合物灭菌。可将水溶液包装原样使用,或冻干,给药前,将冻干制剂与无菌水性载体混合。化合物制剂的pH通常为3~11,更优选5~9,最优选7~8。可以理解,使用某些前述赋形剂、载体或稳定剂会导致形成药物盐。
本发明化合物的治疗剂量可根据例如以下而定:治疗的具体用途、给予化合物的方式、患者的健康和状态,以及签处方医师的判断。本发明化合物在药用组合物中的比例或浓度可不固定,取决于多种因素,它们包括剂量、化学特性(例如疏水性)和给药途径。例如可通过含约0.1~10%w/v该化合物的生理缓冲水溶液提供本发明化合物,用于肠胃外给药。某些 典型剂量范围为约1μg/kg~约1g/kg体重/日。在某些实施方案中,剂量范围为约0.01mg/kg~约100mg/kg体重/日。剂量很可能取决于此类变量,如疾病或病症的种类和发展程度、具体患者的一般健康状态、所选择的化合物的相对生物学效力、赋形剂制剂及其给药途径。可通过由体外或动物模型试验系统导出的剂量-反应曲线外推,得到有效剂量。
术语定义与说明
除非另有说明,本申请说明书和权利要求书中记载的基团和术语定义,包括其作为实例的定义、示例性的定义、优选的定义、表格中记载的定义、实施例中具体化合物的定义等,可以彼此之间任意组合和结合。这样的组合和结合后的基团定义及化合物结构,应当属于本申请说明书记载的范围内。
除非另有说明,本说明书和权利要求书记载的数值范围相当于至少记载了其中每一个具体的整数数值。例如,数值范围“1-40”相当于记载了数值范围“1-10”中的每一个整数数值即1、2、3、4、5、6、7、8、9、10,以及数值范围“11-40”中的每一个整数数值即11、12、13、14、15、......、35、36、37、38、39、40。应当理解,本文在描述取代基时使用的一个、两个或更多个中,“更多个”应当是指≥3的整数,例如3、4、5、6、7、8、9或10。此外,当某些数值范围被定义为“数”时,应当理解为记载了该范围的两个端点、该范围内的每一个整数以及该范围内的每一个小数。例如,“0~10的数”应当理解为不仅记载了0、1、2、3、4、5、6、7、8、9和10的每一个整数,还至少记载了其中每一个整数分别与0.1、0.2、0.3、0.4、0.5、0.6、0.7、0.8、0.9的和。
术语“卤素”表示氟、氯、溴和碘。
术语“C 1-40烷基”应理解为优选表示具有1~40个碳原子的直链或支链饱和一价烃基。例如,“C 1-6烷基”表示具有1、2、3、4、5或6个碳原子的直链和支链烷基。所述烷基是例如甲基、乙基、丙基、丁基、戊基、己基、异丙基、异丁基、仲丁基、叔丁基、异戊基、2-甲基丁基、1-甲基丁基、1-乙基丙基、1,2-二甲基丙基、新戊基、1,1-二甲基丙基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、2-乙基丁基、1-乙基丁基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、2,3-二甲基丁基、1,3-二甲基丁基或1,2-二甲基丁基等或它们的异构体。
术语“C 2-40烯基”应理解为优选表示直连或支链的一价烃基,其包含一个或多个双键并且具有2~40个碳原子,优选“C 2-6烯基”。“C 2-6烯基”应理解为优选表示直连或支链的一价烃基,其包含一个或多个双键并且具有2、3、4、5或6个碳原子,特别是2或3个碳原子(“C 2-3烯基”),应理解,在所述烯基包含多于一个双键的情况下,所述双键可相互分离或者共轭。所述烯基是例如乙烯基、烯丙基、(E)-2-甲基乙烯基、(Z)-2-甲基乙烯基、(E)-丁-2-烯基、(Z)-丁-2-烯基、(E)-丁-1-烯基、(Z)-丁-1-烯基、戊-4-烯基、(E)-戊-3-烯基、(Z)-戊-3-烯基、(E)-戊-2-烯基、(Z)-戊-2-烯基、(E)-戊-1-烯基、(Z)-戊-1-烯基、己-5-烯基、(E)-己-4-烯基、(Z)-己-4-烯基、(E)-己-3-烯基、(Z)-己-3-烯基、(E)-己-2-烯基、(Z)-己-2-烯基、(E)-己-1-烯基、(Z)-己-1-烯基、异丙烯基、2-甲基丙-2-烯基、1-甲基丙-2-烯基、2-甲基丙-1-烯基、(E)-1-甲基丙-1-烯基、(Z)-1-甲基丙-1-烯基、3-甲基丁-3-烯基、2-甲基丁-3-烯基、1-甲基丁-3-烯基、3-甲基丁-2-烯基、(E)-2-甲基丁-2-烯基、(Z)-2-甲基丁-2-烯基、(E)-1-甲基丁-2-烯基、(Z)-1-甲基丁-2-烯基、(E)-3-甲基丁-1-烯基、(Z)-3-甲基丁-1-烯基、(E)-2-甲基丁-1-烯基、(Z)-2-甲基丁-1-烯基、(E)-1-甲基丁-1-烯基、(Z)-1-甲基丁-1-烯基、1,1-二甲基丙-2-烯基、1-乙基丙-1-烯基、1-丙基乙烯基、1-异丙基乙烯基。
术语“C 2- 40炔基”应理解为表示直连或支链的一价烃基,其包含一个或多个三键并且具有2~40个碳原子,优选“C 2-C 6-炔基”。术语“C 2-C 6-炔基”应理解为优选表示直连或支链的一价烃基,其包含一个或多个三键并且具有2、3、4、5或6个碳原子,特别是2或3个碳原子(“C 2-C 3-炔基”)。所述C 2-C 6-炔基是例如乙炔基、丙-1-炔基、丙-2-炔基、丁-1-炔基、丁-2-炔基、丁-3-炔基、戊-1-炔基、戊-2-炔基、戊-3-炔基、戊-4-炔基、己-1-炔基、己-2-炔基、己-3-炔基、己-4-炔基、己-5-炔基、1-甲基丙-2-炔基、2-甲基丁-3-炔基、1-甲基丁-3-炔基、1-甲基丁-2-炔基、3-甲基丁-1-炔基、1-乙基丙-2-炔基、3-甲基戊-4-炔基、2-甲基戊-4-炔基、1-甲基戊-4-炔基、 2-甲基戊-3-炔基、1-甲基戊-3-炔基、4-甲基戊-2-炔基、1-甲基戊-2-炔基、4-甲基戊-1-炔基、3-甲基戊-1-炔基、2-乙基丁-3-炔基、1-乙基丁-3-炔基、1-乙基丁-2-炔基、1-丙基丙-2-炔基、1-异丙基丙-2-炔基、2,2-二甲基丁-3-炔基、1,1-二甲基丁-3-炔基、1,1-二甲基丁-2-炔基或3,3-二甲基丁-1-炔基。特别地,所述炔基是乙炔基、丙-1-炔基或丙-2-炔基。
术语“C 3-40环烷基”应理解为表示饱和的一价单环、双环烃环或桥环烷烃,其具有3~40个碳原子,优选“C 3-10环烷基”。术语“C 3-10环烷基”应理解为表示饱和的一价单环、双环烃环或桥环烷烃,其具有3、4、5、6、7、8、9或10个碳原子。所述C 3-10环烷基可以是单环烃基,如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基或环癸基,或者是双环烃基如十氢化萘环。
术语“3-20元杂环基”意指饱和的一价单环、双环烃环或桥环烷烃,其包含1-5个独立选自N、O和S的杂原子的总成环原子数为3-20(如原子数为3、4、5、6、7、8、9、10等)的非芳族环状基团,优选“3-10元杂环基”。术语“3-10元杂环基”意指饱和的一价单环、双环烃环或桥环烷烃,其包含1-5个,优选1-3个独立选自N、O和S的杂原子,例如1、2、3个独立选自N、O和S的杂原子。所述杂环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,所述杂环基可以包括但不限于:4元环,如氮杂环丁烷基、氧杂环丁烷基(如氮杂环丁烷-1-基);5元环,如四氢呋喃基、二氧杂环戊烯基、吡咯烷基、咪唑烷基、吡唑烷基、吡咯啉基;或6元环,如四氢吡喃基、哌啶基、吗啉基、二噻烷基、硫代吗啉基、哌嗪基或三噻烷基;或7元环,如二氮杂环庚烷基。任选地,所述杂环基可以是苯并稠合的。所述杂环基可以是双环的,例如但不限于5,5元环,如六氢环戊并[c]吡咯-2(1H)-基环,或者5,6元双环,如六氢吡咯并[1,2-a]吡嗪-2(1H)-基环。含氮原子的环可以是部分不饱和的,即它可以包含一个或多个双键,例如但不限于2,5-二氢-1H-吡咯基、4H-[1,3,4]噻二嗪基、4,5-二氢噁唑基或4H-[1,4]噻嗪基,或者,它可以是苯并稠合的,例如但不限于二氢异喹啉基。根据本发明,所述杂环基是无芳香性的。所述3-20元杂环基与其它基团相连构成本发明的化合物时,可以为3-20元杂环基上的碳原子与其它基团相连,也可以为3-20元杂环基环上杂环原子与其它基团相连。例如当3-20元杂环基选自哌嗪基时,可以为哌嗪基上的氮原子与其它基团相连。或当3-20元杂环基选自哌啶基时,可以为哌啶基环上的氮原子和其对位上的碳原子与其它基团相连。
术语“C 6-20芳基”应理解为优选表示具有6~20个碳原子的一价芳香性或部分芳香性的单环、双环或三环烃环,优选“C 6-14芳基”。术语“C 6-14芳基”应理解为优选表示具有6、7、8、9、10、11、12、13或14个碳原子的一价芳香性或部分芳香性的单环、双环或三环烃环(“C 6-14芳基”),特别是具有6个碳原子的环(“C 6芳基”),例如苯基;或联苯基,或者是具有9个碳原子的环(“C 9芳基”),例如茚满基或茚基,或者是具有10个碳原子的环(“C 10芳基”),例如四氢化萘基、二氢萘基或萘基,或者是具有13个碳原子的环(“C 13芳基”),例如芴基,或者是具有14个碳原子的环(“C 14芳基”),例如蒽基。当所述C 6-20芳基被取代时,其可以为单取代或者多取代。并且,对其取代位点没有限制,例如可以为邻位、对位或间位取代。
术语“5-20元杂芳基”应理解为包括这样的一价单环、双环或三环芳族环系:其具有5~20个环原子且包含1-5个独立选自N、O和S的杂原子,例如“5-14元杂芳基”。术语“5-14元杂芳基”应理解为包括这样的一价单环、双环或三环芳族环系:其具有5、6、7、8、9、10、11、12、13或14个环原子,特别是5或6或9或10个碳原子,且其包含1-5个,优选1-3各独立选自N、O和S的杂原子并且,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、噻-4H-吡唑基等以及它们的苯并衍生物,例如苯并呋喃基、苯并噻吩基、苯并噁唑基、苯并异噁唑基、苯并咪唑基、苯并三唑基、吲唑基、吲哚基、异吲哚基等;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,例如喹啉基、喹唑啉基、异喹啉基等;或吖辛因基、吲嗪基、嘌呤基等以及它们的苯并衍生物;或噌啉基、酞嗪基、喹唑啉基、喹喔啉基、萘啶基、蝶啶基、咔唑基、吖啶基、吩嗪基、吩噻嗪基、吩噁嗪基等。当所述5-20元杂芳基与其它基团相连构成本发明的化合物时,可以为5-20 元杂芳基环上的碳原子与其它基团相连,也可以为5-20元杂芳基环上的杂原子与其它基团相连。当所述5-20元杂芳基被取代时,其可以为单取代或者多取代。并且,对其取代位点没有限制,例如可以为杂芳基环上与碳原子相连的氢被取代,或者杂芳基环上与杂原子相连的氢被取代。
除非另有说明,杂环基、杂芳基或亚杂芳基包括其所有可能的异构形式,例如其位置异构体。因此,对于一些说明性的非限制性实例,可以包括在其1-、2-、3-、4-、5-、6-、7-、8-、9-、10-、11-、12-位等(如果存在)中的一个、两个或更多个位置上取代或与其他基团键合的形式,包括吡啶-2-基、亚吡啶-2-基、吡啶-3-基、亚吡啶-3-基、吡啶-4-基和亚吡啶-4-基;噻吩基或亚噻吩基包括噻吩-2-基、亚噻吩-2-基、噻吩-3-基和亚噻吩-3-基;吡唑-1-基、吡唑-3-基、吡唑-4-基、吡唑-5-基。
术语“氧代”是指取代基中的碳原子、氮原子或硫原子被氧化后形成的氧基取代(=O)。
除非另有说明,本文中术语的定义同样适用于包含该术语的基团,例如C 1-6烷基的定义也适用于C 1-6烷基氧基、-N(C 1-6烷基) 2、-NHC 1-6烷基或-S(O) 2-C 1-6烷基等。
本领域技术人员可以理解,式I所示化合物可以以各种药学上可接受的盐的形式存在。如果这些化合物具有碱性中心,则其可以形成酸加成盐;如果这些化合物具有酸性中心,则其可以形成碱加成盐;如果这些化合物既包含酸性中心(例如羧基)又包含碱性中心(例如氨基),则其还可以形成内盐。
本发明的化合物可以溶剂合物(如水合物)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。
根据其分子结构,本发明的化合物可以是手性的,因此可能存在各种对映异构体形式。因而这些化合物可以以消旋体形式或光学活性形式存在。本发明的化合物或其中间体可以通过本领域技术人员公知的化学或物理方法分离为对映异构体化合物,或者以此形式用于合成。在外消旋的胺的情况中,通过与光学活性的拆分试剂反应,从混合物制得非对映异构体。适当的拆分试剂的示例是光学活性的酸,例如R和S形式的酒石酸、二乙酰酒石酸、二苯甲酰酒石酸、扁桃酸、苹果酸、乳酸、适当的N-保护的氨基酸(例如N-苯甲酰脯氨酸或N-苯磺酰基脯氨酸)或各种光学活性的樟脑磺酸。借助光学活性的拆分试剂(例如固定在硅胶上的二硝基苯甲酰基苯基甘氨酸、三乙酸纤维素或其它碳水化合物的衍生物或手性衍生化的异丁烯酸酯聚合物),也可有利地进行色谱对映体拆分。用于此目的的适当的洗脱剂是含水或含醇的溶剂混合物,例如,己烷/异丙醇/乙腈。
术语“互变异构体”是指因分子中某一原子在两个位置迅速移动而产生的官能团异构体。本发明化合物可表现出互变异构现象。互变异构的化合物可以存在两种或多种可相互转化的种类。质子移变互变异构体来自两个原子之间共价键合的氢原子的迁移。互变异构体一般以平衡形式存在,尝试分离单一互变异构体时通常产生一种混合物,其理化性质与化合物的混合物是一致的。平衡的位置取决于分子内的化学特性。例如,在很多脂族醛和酮如乙醛中,酮型占优势;而在酚中,烯醇型占优势。本发明包含化合物的所有互变异构形式。
可以根据已知的方法,例如通过萃取、过滤或柱层析来分离相应的稳定异构体。
术语“患者”是指包括哺乳动物在内的任何动物,优选小鼠、大鼠、其它啮齿类动物、兔、狗、猫、猪、牛、羊、马或灵长类动物,最优选人。
本文中使用的短语“治疗有效量”是指研究人员、兽医、医师或其它临床医师正在组织、系统、动物、个体或人中寻找的引起生物学或医学反应的活性化合物或药物的量,它包括以下一项或多项:(1)预防疾病:例如在易感染疾病、紊乱或病症但尚未经历或出现疾病病理或症状的个体中预防疾病、紊乱或病症。(2)抑制疾病:例如在正经历或出现疾病、紊乱或病症的病理或症状的个体中抑制疾病、紊乱或病症(即阻止病理和/或症状的进一步发展)。(3)缓解疾病:例如在正经历或出现疾病、紊乱或病症的病理或症状的个体中缓解疾病、紊乱或病症(即逆转病理和/或症状)。
有益效果
本发明的化合物可以作为具有高度选择性或抑制作用的RET抑制剂,对例如RET看门残基突变体RET V804M、RET溶剂前沿残基突变体G810R和其他临床相关RET突变体以及RET-wt均有优异的抑制作用。并且,本发明的优选化合物高效抑制甲状腺癌来源的TT细胞系和各种RET突变体转化的Ba/F3细胞的生长,能够阻断细胞RET自磷酸化及其下游通路,并显著诱导TT细胞死亡。此外,本发明的优选化合物还具有良好的药代动力学性质,作为活性成分时可以较小的剂量给患者施用,从而降低患者的治疗成本。
具体实施方式
下文将结合具体实施例对本发明的技术方案做更进一步的详细说明。应当理解,下列实施例仅为示例性地说明和解释本发明,而不应被解释为对本发明保护范围的限制。凡基于本发明上述内容所实现的技术均涵盖在本发明旨在保护的范围内。
除非另有说明,以下实施例中使用的原料和试剂均为市售商品,或者可以通过已知方法制备。
实施例1:化合物1的制备
步骤A:N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌啶-4-基)-5-氟-2-甲基苯甲酰胺
Figure PCTCN2022073467-appb-000044
反应瓶中加入1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌啶-4-氨基盐酸盐(100mg,0.2mmol),5-氟-2-甲基苯甲酸(37mg,0.24mmol),DIEA(129mg,1.0mmol),HATU(76mg,0.2mmol)和2mL DMF,25℃反应12h,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品8.5mg。m/z=528.2[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.58(d,J=2.0Hz,1H),8.51(d,J=2.0Hz,1H),8.01-8.06(m,2H),7.60(s,1H),7.26-7.29(m,2H),7.10-7.17(m,3H),4.15(q,J=6.8Hz,2H),4.05-4.08(m,2H),3.34-3.68(m,2H),2.32-2.37(m,5H),1.60-1.65(m,2H),1.46(s,3H),1.38-1.45(m,3H).
实施例2:化合物3的制备
步骤A:2,4,6-三甲基苯磺酸1-氨基-3-溴-5-甲氧基吡啶-1-鎓
Figure PCTCN2022073467-appb-000045
零度下,向2-[(氨基氧基)磺酰]-1,3,5-三甲基苯(6.8g,31.7mmol)的二氯甲烷(50mL)溶液中加入3-溴-5-甲氧基吡啶(6.0g,32.0mmol),并在零度下继续搅拌3小时,析出大量白色固体;反应完毕后,零度下向反应体系中加入乙醚(50mL)并搅拌10分钟,减压过滤,乙醚冲洗,真空干燥得到产品(15g),无须再次纯化,直接用于下一步反应。m/z=204[M+1] +
步骤B:4-溴-2-氟-6-甲氧基吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000046
室温下,向装有2,4,6-三甲基苯磺酸1-氨基-3-溴-5-甲氧基吡啶-1-鎓(1.0g,2.3mmol)的DMF(30mL)溶液中加入碳酸钾(1.4g,10.0mmol);反应体系冷却至0℃,分批次加入对甲苯磺酸2,2-二氟乙烯基酯(0.5g,2.3mmol),升至室温下搅拌1小时,再90℃下搅拌1小时;反应完毕并冷却至室温,加水淬灭,用乙酸乙酯萃取,合并有机相,并用水洗涤,减压浓缩,柱层析分离得到产品80mg,m/z=245[M+1] +1HNMR(400MHz,CDCl 3)δ8.07(s,1H),6.61(s,1H),6.18(d,1H),3.85(s,3H)。
步骤C:4-溴-2-氟-6-甲氧基吡唑[1,5-a]吡啶-3-甲醛
Figure PCTCN2022073467-appb-000047
零度下,向装有4-溴-2-氟-6-甲氧基吡唑[1,5-a]吡啶(294mg,1.2mmol)的DMF(10mL)溶液中滴加入三氯氧磷(1.0g,6.5mmol),滴完后自然升至室温反应过夜。将反应液倒入100mL冰水中,用2N NaOH溶液调节pH=7,用乙酸乙酯萃取,合并有机相,减压浓缩,柱层析分离得到产品240mg。m/z=273[M+1] +1HNMR(400MHz,DMSO-d 6)δ10.51(s,1H),8.72(d,1H),8.04(d,1H),3.87(s,3H).
步骤D:4-溴-6-甲氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000048
向4-溴-2-氟-6-甲氧基吡唑[1,5-a]吡啶-3-甲醛(273mg,1mmol)的DMF(10.0mL)溶液中加入水合肼(2.0mL),加热至100℃反应12小时。加入水,用乙酸乙酯萃取,合并有机相,并用水洗涤,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得到产品,m/z=267[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.82(s,1H),8.67(d,1H),7.92(s,1H),7.62(d,1H),3.89(s,3H)。
步骤E:4-溴-6-羟基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000049
将4-溴-6-甲氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(267mg,1mmol),DCE(20mL),氯化铝(446mg,3.36mmol)放于100mL单口瓶中,氮气保护,80℃加热搅拌,颜色逐渐变深棕色,反应3小时,TLC检测反应完,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=253[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.69(brs,1H),10.23(s,1H),8.33(d,1H),7.88(s,1H),7.42(d,1H)。
步骤F:4-溴-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000050
向4-溴-6-羟基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(76mg,0.3mmol),的DMF(10.0mL)溶液中加入2-碘乙烷(47mg,0.3mmol),碳酸钾(83mg,0.6mmol),加热至60℃反应12小时。加入水,用乙酸乙酯萃取,合并有机相,并用水洗涤,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得到产品,m/z=281[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.79(s,1H),8.64(d,1H),7.91(s,1H),7.59(d,1H),4.17(t,2H),1.38(t,3H)。
步骤G:6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000051
向4-溴-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(1.0g,3.6mmol)中加入6-氟吡啶-3-硼酸(0.6g,4.3mmol),碳酸钠(1.5g,14mmol),1,4-二氧六环20ml和3ml水,四三苯基膦钯(0.12g,0.1mmol),置换氮气,90℃反应12h,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=298[M+1] +1H NMR(400MHz,DMSO-d6):12.73(s,1H),8.61-8.76(m,2H),8.28(s,1H),8.48(s,1H),7.63(s,1H),4.16(q,2H),1.39(t,J=6.4Hz,3H)。
步骤H:(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000052
反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(500mg,1.7mmol),(4-甲基哌啶-4-基)碳酸叔丁酯(721mg,3.4mmol)、DIEA(650mg,5.1mmol)和10ml DMSO,90℃反应4h,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=492[M+1] +
步骤I:1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌啶-4-氨基盐酸盐
Figure PCTCN2022073467-appb-000053
反应瓶中加入(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌啶-4-基)氨基碳酸叔丁酯(390mg,0.8mmol),5N甲醇盐酸溶液(8ml,40mmol)25℃反应2h,减压浓缩,得到产品,m/z=392[M+1] +
步骤J:3-氯-N-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌啶-4-基)2-吡啶甲酰胺
Figure PCTCN2022073467-appb-000054
反应瓶中加入1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲基哌 啶-4-氨基盐酸盐(100mg,0.2mmol)、3-氯吡啶-2-甲酸(37mg,0.24mmol)、DIEA(85mg,0.66mmol)、HATU(84mg,0.22mmol)和2ml DMF,25℃反应2h,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=531[M+1] +1H NMR(400MHz,DMSO-d6):8.53-8.57(m,3H),8.32(s,1H),8.00-8.10(m,2H),7.61(s,1H),7.50(dd,J1=8.0Hz,J2=2.4Hz,1H),7.28(s,1H),7.16(d,J=8.4Hz,1H),4.15-4.23(m,2H),4.07-4.10(m,2H),3.35-3.42(m,2H),2.33-2.37(m,2H),1.60-1.65(m,2H),1.45-1.48(m,6H)。
实施例3:化合物4的制备
Figure PCTCN2022073467-appb-000055
制备方法同实例1步骤J,m/z=549[M+1] +1H NMR(400MHz,DMSO-d6):12.76(s,1H),8.57-8.62(m,2H),8.51(s,1H),8.30(s,1H),8.19(dd,1H),8.19(dd,1H),7.59(s,1H),7.11(d,1H),4.15-4.22(m,2H),4.06-4.10(m,2H),3.33-3.36(m,2H),2.30-2.33(m,2H),1.58-1.65(m,2H),1.43-1.47(m,6H)。
实施例4:化合物8的制备
Figure PCTCN2022073467-appb-000056
向反应瓶中依次加入2mL DMSO和0.5ml DIEA,3-甲氧基-6-(哌啶-4-氧基)哒嗪(400mg,1.3mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(70mg,0.2mmol),100 oC下反应48小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=487[M+1] +
实施例5:化合物11的制备
步骤A:4-((叔丁基羰基)氨基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯
Figure PCTCN2022073467-appb-000057
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(2.5g,8.4mmol),4-(叔丁基羰基)氨基哌啶-4-羧酸乙酯(2.8g,9.2mmol),DIEA(3.2g,25.2mmol),DMSO(50ml)加入反应瓶,90℃反应12h,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=550[M+1] +
步骤B:4-(氨基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯
Figure PCTCN2022073467-appb-000058
将4-((叔丁基羰基)氨基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(1g,1.8mmol)加入10ml MeOH/HCl(4M),25℃反应4h,旋干溶剂得产品,m/z=450[M+1] +
步骤C:4-(2,6-二氟苯甲酰胺)-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯
Figure PCTCN2022073467-appb-000059
将4-(氨基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(120mg,0.26mmol),2,6-二氟苯甲酸(38mg,0.24mmol),DIEA(100mg,0.78mmol),HATU(108mg,0.28mmol),DMF(2ml)加入反应瓶,25℃反应12h,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=590[M+1] +
步骤D:N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000060
将4-(2,6-二氟苯甲酰胺)-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(30mg,0.05mmol),2ml THF加入反应瓶,0℃下加入LiAlH 4(1.9mg,0.05mmol),反应1h。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=548[M+1] +1H NMR(400MHz,DMSO-d6)δ12.65(s,1H),8.59(d,1H),8.52(d,1H),8.36(s,1H),8.04(dd,1H),7.60(s,1H),7.50(tt,1H),7.27(d,1H),7.22–7.08(m,3H),4.87(t,1H),4.28(d,2H),4.18(q,2H),3.63(d,2H),3.19(t,2H),2.24(d,2H),1.68(td,4.3Hz,2H),1.41(t,3H)。
实施例6:化合物12的制备
步骤A:4-(3-氯甲基吡啶酰胺)-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯
Figure PCTCN2022073467-appb-000061
将4-((叔丁基羰基)氨基)1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯盐酸盐(120mg,0.26mmol),3-氯吡啶甲酸(37mg,0.24mmol),DIEA(100mg,0.78mmol),HATU(108mg,0.28mmol),DMF(2ml)加入反应瓶,25℃反应12h,LCMS监测反应完全。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品80mg,m/z=589.2[M+1] +
步骤B:3-氯-N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)哌啶-4-基)-吡啶甲酰胺
Figure PCTCN2022073467-appb-000062
将4-(3-氯甲基吡啶酰胺)-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(30mg,0.05mmol),2ml THF加入反应瓶,0℃下加入LiAlH 4(1.9mg,0.05mmol),反应1h。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品18mg,m/z=547.2[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.62–8.49(m,3H),8.23(s,1H),8.03(m,2H),7.60(s,1H),7.52(m,1H),7.27(d,1H),7.11(d,1H),4.90(t,1H),4.27–4.07(m,5H),3.66(d,2H),3.18(d,1H),2.29(d,2H),1.72(m,2H),1.40(t,3H).m/z=547.2[M+1] +
实施例7:化合物15的制备
步骤A:叔丁基羰基(1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)哌啶-4-基)氨基甲酸酯
Figure PCTCN2022073467-appb-000063
将4-((叔丁基羰基)氨基)1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(2.7g,5mmol),THF(100ml)加入反应瓶,0℃加入LiBH 4(0.55g,25mmol),室温反应12h。LCMS监测反应完全。将反应液倒入200mL水中,乙酸乙酯萃取,干燥有机相,旋干溶剂,得产品2.1g,m/z=508[M+1] +
步骤B:叔丁基羰基(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲酰基哌啶-4-基)氨基甲酸酯
Figure PCTCN2022073467-appb-000064
将4-((叔丁基羰基)氨基)1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(1g,2.0mmol)加入40ml DCM,0℃加入Dess-Martin(0.93g,2.2mmol)反应,LCMS监测反应完全。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品0.8g,m/z=506[M+1] +
步骤C:叔丁基羰基(4-((2-甲氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-基)氨基甲酸酯
Figure PCTCN2022073467-appb-000065
将叔丁基羰基(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲酰基哌啶-4-基)氨基甲酸酯(505mg,1mmol),二甲胺四氢呋喃溶液(2M/L,3ml,3mmol),NaBH(OAc) 3(424mg,2mmol),DCE(10mL)加入反应瓶,25℃反应12h,LCMS监测反应完全。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品400mg,m/z=534.2[M+1] +
步骤D:4-((2-甲氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-胺盐酸盐
Figure PCTCN2022073467-appb-000066
将叔丁基羰基(4-((2-甲氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-基)氨基甲酸酯(100mg,0.19mmol),2mL甲醇盐酸气加入反应瓶,25℃反应5h。直接旋干溶剂,得产品80mg,m/z=435.2[M+1] +
步骤E:3-氯-N-(4-((2甲氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-基)吡啶酰胺
Figure PCTCN2022073467-appb-000067
将4-((2甲氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-胺(80mg,0.19mmol),3-氯吡啶甲酸(29.8mg,0.19mmol),HATU(72.2mg,0.19mmol),DIEA(73.5mg,0.57mmol),DMF(10mL)加入反应瓶,25℃反应15h。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品60mg,m/z=574.2[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.69(s,1H),8.62-8.49(m,3H),8.28(s,1H),8.08-7.98(m,2H),7.60(s,1H),7.52(dd,J=8.2,4.7Hz,1H),7.27(d,J=2.1Hz,1H),7.10(d,J=9.0Hz,1H),4.26-4.13(m,4H),3.25(d,J=12.4Hz,2H),2.70(s,2H),2.38(d,J=13.2Hz,2H),2.31(s,6H),2.30(d,J=4.2Hz,1H),1.63(td,J=13.0,12.4,4.1Hz,2H),1.41(t,J=6.9Hz,3H),1.24(s,1H),0.85(d,J=7.3Hz,1H),0.5(s,1H)。
实施例8:化合物17的制备
步骤A:叔丁基羰基(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-((4-乙基哌嗪-1-基)甲基)哌啶-4-基)氨基甲酸酯
Figure PCTCN2022073467-appb-000068
将叔丁基羰基(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲酰基哌啶-4-基)氨基甲酸酯(505mg;1mmol),乙基哌嗪(342mg,3mmol),NaBH(OAc) 3(424mg,2mmol),DCE(10mL)加入反应瓶,25℃反应12h,LCMS监测反应完全。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品800mg,m/z=604.4[M+1] +
步骤B:1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-((4-乙基哌嗪-1-基)甲基)哌啶-4-胺盐酸盐
Figure PCTCN2022073467-appb-000069
将叔丁基羰基(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-((4-乙基哌嗪-1-基)甲基)哌啶-4-基)氨基甲酸酯(120mg,0.2mmol),2mL甲醇盐酸气加入反应瓶,25℃反应5h。直接旋干溶剂得产品100mg,m/z=504.2[M+1] +
步骤C:N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-((4-乙基哌嗪-1-基)甲基)哌啶-4-基)-2,5-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000070
将4-((2甲氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-胺(100mg,0.2mmol,2,5-二氟苯甲酸(31.6mg,0.2mmol),HATU(76.0mg,0.2mmol),DIEA(77.4mg,0.6mmol),DMF(10mL)加入反应瓶,25℃反应15h。将反应液倒入水中,乙酸乙酯萃取,干燥有机相,旋干溶剂,硅胶柱层析纯化得产品67mg,m/z=644.2[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.65(s,1H),8.59(d,J=2.8Hz,1H),8.52(d,J=2.0Hz,1H),8.13(s,1H),8.04(d,J=8.8Hz,1H),7.60(s,1H),7.41-7.29(m,3H),7.26(d,J=2.1Hz,1H),7.10(d,J=9.0Hz,1H),4.19(m,4H),3.32(s,1H),3.27-3.15(m,2H),2.73(s,2H),2.61-2.55(m,7H),2.34(d,J=13.3Hz,4H),1.62(m,2H),1.41(t,J=6.9Hz,3H),0.99(m,3H).m/z=644.2[M+1] +
实施例9:化合物29的制备
步骤A:叔丁基羰基(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(甲基吗啉)哌啶-4-基)氨基甲酸酯
Figure PCTCN2022073467-appb-000071
将叔丁基羰基(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-甲酰基哌啶-4-基)氨基甲酸酯(505mg,1mmol),吗啉(261mg,3mmol),NaBH(OAc) 3(424mg,2mmol), DCE(10mL)加入反应瓶,25℃反应12h,LCMS监测反应完全。将反应液倒入20mL水中,DCM萃取,干燥有机溶剂,旋干溶剂,柱层析纯化产品300mg,m/z=576.2[M+1] +
步骤B:1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-((4-乙基哌嗪-1-基)甲基)哌啶-4-胺盐酸盐
Figure PCTCN2022073467-appb-000072
将叔丁基羰基(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(甲基吗啉)哌啶-4-基)氨基甲酸酯(120mg,0.2mmol),2mL甲醇盐酸气加入反应瓶,25℃反应5h。直接旋干溶剂,得产品90mg,m/z=477.2[M+1] +
步骤C:N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-((4-乙基哌嗪-1-基)甲基)哌啶-4-基)-3-甲基丁酰胺
Figure PCTCN2022073467-appb-000073
将1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(甲基吗啉)哌啶-4-胺(90mg,0.19mmol),异戊酸(20.4mg,0.2mmol),HATU(76.0mg,0.2mmol),DIEA(77.4mg,0.6mmol),DMF(10mL)加入反应瓶,25℃反应15h。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品71mg,m/z=561.2[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.65(s,1H),δ8.58(d,J=2.6Hz,1H),8.52(d,J=2.1Hz,1H),8.36(s,1H),8.03(dd,J=8.9,2.6Hz,1H),7.59(s,1H),7.26(d,J=2.1Hz,1H),7.07(d,J=9.0Hz,1H),4.18(m,3H),3.55(m,4H),3.10(m,2H),2.60(s,2H),2.46(m,5H),2.26(d,J=13.2Hz,2H),2.02(d,J=5.4Hz,2H),1.54(dt,J=12.8,6.7Hz,2H),1.40(m,3H),1.24(s,1H),0.90(d,J=6.2Hz,6H)。
实施例10:化合物31的制备
步骤A:((3S,4R)-1-(5-溴吡啶-2-基)-4-羟基吡咯烷-3-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000074
依次加入5-溴-2-氟吡啶(240mg,1.36mmol),DMSO(2.5mL),碳酸钾(340mg,2.47mmol),((3S,4R)-4-羟基吡咯烷-3-基)氨基碳酸叔丁酯(250mg,1.24mmol),加完升温至90℃反应16小时。反应结束后倒入水中,二氯甲烷萃取,无水硫酸钠干燥后,旋干得到产品390 mg,m/z=358/360[M+1] +
步骤B:((3S,4S)-1-(5-溴吡啶-2-基)-4-(吡啶-2-基氧基)吡咯烷-3-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000075
反应瓶中依次加入THF(20mL),TMAD(260mg,1.51mmol),BuP 3(305mg,1.51mmol),室温搅拌30分钟,滴加((3R,4S)-1-(5-溴吡啶-2-基)-4-羟基吡咯烷-3-基)氨基碳酸叔丁酯(270mg,0.75mmol)的THF(10mL)溶液,滴完后搅拌1分钟,再加入2-羟基吡啶(143mg,1.51mmol)后升温至50℃反应3小时。反应液倒入水中,二氯甲烷萃取,柱层析得产品140mg,m/z=435/437[M+1] +
步骤C:((3S,4S)-1-(5-(6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(吡啶-2-基氧基)吡咯烷-3-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000076
反应瓶中依次加入1,4-二氧六环(1mL),((3S,4S)-1-(5-溴吡啶-2-基)-4-(吡啶-2-基氧基)吡咯烷-3-基)氨基碳酸叔丁酯(60mg,0.14mmol),6-乙氧基-1-(四氢-2H-吡喃-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(47.5mg,0.12mmol),碳酸钾(32.8mg,0.23mmol),四三苯基膦钯(13.3mg,0.006mmol),水(0.5mL),升温至90℃反应16小时。反应液倒入水中,二氯甲烷萃取,柱层析得产品40mg,m/z=641[M+1] +
步骤D:(3S,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(吡啶-2-基氧基)吡咯烷-3-胺
Figure PCTCN2022073467-appb-000077
向50mL的反应瓶中加入((3S,4S)-1-(5-(6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(吡啶-2-基氧基)吡咯烷-3-基)氨基碳酸叔丁酯(40mg,0.06mmol),甲醇(2mL),氯化氢的二氧六环溶液(4M,2mL),室温搅拌2小时。反应液倒入水中,二氯甲烷萃取,柱层析纯化得产品6.9mg,m/z=457[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.63(s,1H),8.55-8.54(m,2H),8.23-8.21(m,1H),7.80-8.02(m,1H),7.71-7.75(m,1H),7.59(s,1H),7.21-7.22(m,1H),7.00-7.04(m,1H),6.81-6.85(m,1H),6.69-6.71(m,1H),5.28-5.29(m,1H),4.14-4.19(m,2H),4.01-4.05(m,1H),3.74-3.78(m,1H),3.61-3.67(m,3H),1.38-1.41(m,3H).
实施例11:化合物33的制备
步骤A:6-苄基-1-氧杂-6-氮杂螺环[2.5]辛烷
Figure PCTCN2022073467-appb-000078
向反应瓶中加入N-苄基-4-哌啶酮(5.7g,30mmol),二甲基亚砜(75mL),搅拌,加入NaH(1.44g,33mmol),反应1小时,加入三甲基碘化亚砜(7.3g,33mmol)。室温反应1小时,加水淬灭,乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩干,得产品5.6g,m/z=204[M+1] +
步骤B:6-苄基-1-硫杂-6-氮杂螺环[2.5]辛烷
Figure PCTCN2022073467-appb-000079
向反应瓶中依次加入6-苄基-1-氧杂-6-氮杂螺环[2.5]辛烷(5.6g,27.6mmol)和甲醇(80mL),搅拌,加入硫脲(2.3g,30.4mmol),40℃反应20小时。反应完全,加入水,乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩干,硅胶柱层析得产品4.63g,m/z=220[M+1] +。步骤C:1-苄基-4-甲基哌啶-4-硫醇
Figure PCTCN2022073467-appb-000080
向反应瓶中依次加入6-苄基-1-硫杂-6-氮杂螺环[2.5]辛烷(4.3g,19.6mmol)和四氢呋喃(50mL),降温至5℃,加入四氢铝锂(1.1g,29.4mmol),保温反应1小时,反应完全。慢慢加入5mL水,加入氢氧化钠饱和溶液调pH值为14,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩干,硅胶柱层析纯化得产品3.8g,m/z=222[M+1] +
步骤D:1-苄基-4-(异丁基硫基)-4-甲基哌啶
Figure PCTCN2022073467-appb-000081
向反应瓶中依次加入1-苄基-4-甲基哌啶-4-硫醇(1.3g,6mmol)和N,N-二甲基甲酰胺30mL,降温至5℃,加入1-溴-2-甲基丙烷(0.9g,6.6mmol),加入碳酸钾(2.5g,18mmol),40℃反应16小时,反应完全。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1.0g,m/z=278[M+1] +
步骤E:1-苄基-4-(异丁基磺酰基)-4-甲基哌啶
Figure PCTCN2022073467-appb-000082
向反应瓶中依次加入1-苄基-4-(异丁基硫基)-4-甲基哌啶(500mg,1.8mmol)和二氯甲烷(6mL),降温至5℃,加入间氯过氧苯甲酸(930mg,5.4mmol),室温反应16小时,反应完全。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品250mg,m/z=310[M+1] +
步骤F:4-(异丁基磺酰基)-4-甲基哌啶
Figure PCTCN2022073467-appb-000083
向反应瓶中依次加入1-苄基-4-(异丁基磺酰基)-4-甲基哌啶(250mg,0.8mmol)和乙醇(10mL),加入10%Pd/C(200mg),置换氢气,室温反应16小时,反应完全。过滤,减压浓缩干,得产品180mg,m/z=220[M+1] +
步骤G:6-乙氧基-4-(6-(4-(异丁基磺酰基)-4-甲基哌啶-1-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000084
向反应瓶中依次加入2mL DMSO和0.5mL DIEA,4-(异丁基磺酰基)-4-甲基哌啶(180mg,0.82mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.34mmol),100℃下反应60小时,反应完全。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品66mg,m/z=497[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.60(d,J=2.5Hz,1H),8.52(d,J=2.1Hz,1H),8.07(dd,J=8.9,2.6Hz,1H),7.59(s,1H),7.28(d,J=2.1Hz,1H),7.13(d,J=8.9Hz,1H),4.43(d,J=13.7Hz,2H),4.18(q,J=6.9Hz,2H),3.16(m,2H),2.96(d,J=6.6Hz,2H),2.25(m,1H),2.01(m,2H),1.72(d,J=13.0Hz,2H),1.53(s,3H),1.40(t,J=6.9Hz,3H),1.08(d,J=6.7Hz,6H).
实施例12:化合物34的制备
Figure PCTCN2022073467-appb-000085
向反应瓶中依次加入2mL DMSO和0.5mL DIEA,4-苄基磺酸基哌啶(140mg,0.51mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(50mg,0.17mmol),100℃下反应20小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品,m/z=517。
实施例13:化合物35的制备
4-((二甲基氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)吡啶-2-基)-N-异丁基哌啶-4-酰胺(化合物APS03035)
步骤A:1-(5-溴吡啶-2-基)-4-甲酰基-N-异丁基哌啶-4-酰胺
Figure PCTCN2022073467-appb-000086
向50mL反应瓶中加入1-(5-溴吡啶-2-基)-4-(羟甲基)-N-异丁基哌啶-4-羧酰胺(170mg,0.46mmol),二氯甲烷(10mL),然后室温下分批加入Dess-Marting(292mg,0.69mmol),室温搅拌0.5小时后,亚硫酸钠水溶液淬灭后,有机相旋干,柱层析分离得到产品210mg,m/z=368/370[M+1] +
步骤B:1-(5-溴吡啶-2-基)-4-((二甲基氨基)甲基)-N-异丁基哌啶-4-羧酰胺
Figure PCTCN2022073467-appb-000087
向50mL反应瓶中加入1-(5-溴吡啶-2-基)-4-甲酰基-N-异丁基哌啶-4-酰胺(70mg,0.19mmol),1,2-二氯甲烷(2mL),二甲胺(25.7mg,0.57mmol),醋酸硼氢化钠(80.8mg,0.38mmol),室温下搅拌16小时后,水淬灭后,浓缩,柱层析分离得到产品50mg,m/z=397/399[M+1] +
步骤C:1-(5-溴吡啶-2-基)-4-((二甲基氨基)甲基)-N-异丁基哌啶-4-羧酰胺
Figure PCTCN2022073467-appb-000088
向反应瓶中依次加入1-(5-溴吡啶-2-基)-4-((二甲基氨基)甲基)-N-异丁基哌啶-4-羧酰胺(50mg,0.13mmol),6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑并[1,5-a]吡啶(56.2mg,0.15mmol),四三苯基磷钯(23mg,0.02mmol),碳酸钾(34.9mg,0.25mmol),水(0.3mL),1,4-二氧六环(0.6mL),加热至90℃反应16小时,反应液倒入水中,二氯甲烷萃取,柱层析得到产品5mg,m/z=519[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.50-8.56(m,2H),8.00-8.03(m,1H),7.83(s,1H),7.58(s,1H),7.25-7.25(m,1H),7.03-7.05(m,1H),4.15-4.20(m,2H),4.04-4.07(m,2H),3.16-3.21(m,2H),2.96(m,2H),2.42(s,1H),2.09-2.17(m,8H),1.75-1.79(m,1H),1.46-1.52(m,2H),1.30-1.40(m,3H),1.21(s,1H),0.80-0.90(m,6H)。
实施例14:化合物36的制备
4-(氮杂环丁烷-1-基甲基)-1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)吡啶-2-基)-N-异丁基哌啶-4-酰胺(化合物APS03036)
步骤A:1-(5-溴吡啶-2-基)-4-(氮杂环丁烷-1-基甲基)-N-异丁基哌啶-4-羧酰胺
Figure PCTCN2022073467-appb-000089
向50mL反应瓶中加入1-(5-溴吡啶-2-基)-4-甲酰基-N-异丁基哌啶-4-酰胺(70mg,0.19mmol),1,2-二氯甲烷(2mL),氮杂环丁胺(32.6mg,0.57mmol),醋酸硼氢化钠(80.8mg,0.38mmol),室温下搅拌16小时后,水淬灭后,有机相旋干,柱层析分离得到产品60mg,m/z=409/411[M+1] +
步骤B:4-(氮杂环丁烷-1-基甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-N-异丁基哌啶-4-酰胺
Figure PCTCN2022073467-appb-000090
向反应瓶中依次加入1-(5-溴吡啶-2-基)-4-(氮杂环丁烷-1-基甲基)-N-异丁基哌啶-4-羧酰胺(60mg,0.15mmol),6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(65.3mg,0.18mmol),四三苯基磷钯(23mg,0.02mmol),碳酸钾(40.6mg,0.29mmol),水(0.35mL),1,4-二氧六环(0.7mL),氮气置换三次,加热至90℃反应16小时,反应液倒入水中,二氯甲烷萃取,柱层析纯化得产品6mg,m/z=531[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.50-8.56(m,2H),7.94-8.03(m,2H),7.71-7.81(m,1H),7.58(s,1H),7.24-7.25(m,1H),7.02-7.04(m,1H),4.14-4.20(m,2H),4.04-4.07(m,2H),3.15-3.20(m,6H),2.93-2.96(m,2H),2.04-2.17(m,2H),1.88-1.96(m,2H),1.74-1.80(m,1H),1.34-1.44(m,5H),0.85-0.88(m,7H)。
实施例15:化合物37的制备
4-(吡咯烷-1-基甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-N-异丁基哌啶-4-酰胺(化合物APS03037)
步骤A:1-(5-溴吡啶-2-基)-4-(吡咯烷-1-基甲基)-N-异丁基哌啶-4-羧酰胺
Figure PCTCN2022073467-appb-000091
向50mL反应瓶中加入1-(5-溴吡啶-2-基)-4-甲酰基-N-异丁基哌啶-4-酰胺(70mg,0.19mmol),1,2-二氯甲烷(2mL),吡咯烷(40.6mg,0.57mmol),醋酸硼氢化钠(80.8mg,0.38mmol),室温下搅拌16小时后,水淬灭后,有机相旋干,柱层析分离产品65mg,m/z=423/425[M+1] +
步骤B:4-(吡咯烷-1-基甲基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-N-异丁基哌啶-4-酰胺
Figure PCTCN2022073467-appb-000092
向反应瓶中依次加入1-(5-溴吡啶-2-基)-4-(吡咯烷-1-基甲基)-N-异丁基哌啶-4-羧酰胺(65mg,0.15mmol),6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(68.3mg,0.18mmol),四三苯基磷钯(23mg,0.02mmol),碳酸钾(42.5mg,0.31mmol),水(0.3mL),1,4-二氧六环(0.7mL),氮气置换三次,加热至90℃反应16小时,反应液倒入水中,二氯甲烷萃取,反相柱层析得产品5mg,m/z=545[M+1] +1H NMR(400MHz, DMSO-d 6)δ12.67(s,1H),8.50-8.56(m,2H),7.71-8.03(m,4H),7.56-7.65(m,2H),7.40-7.41(m,1H),7.24-7.25(m,1H),7.02-7.05(m,1H),4.14-4.20(m,2H),4.04-4.07(m,2H),3.15-3.24(m,2H),2.93-2.96(m,2H),2.51-2.62(m,2H),2.11-2.17(m,2H),1.73-1.79(m,1H),1.63(s,4H),1.38-1.51(m,5H),0.84-0.87(m,6H)。
实施例16:化合物39的制备
Figure PCTCN2022073467-appb-000093
将(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),3-甲基丁酸(78mg,0.77mmol),DIEA(263mg,2.04mmol)和DCM(10mL),HATU(292mg,0.77mmol)置于50mL的反应瓶中,室温反应0.5小时,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品6.4mg,m/z=478[M+1] +1H NMR(400MHz,DMSO-d6)δ12.65(s,1H),8.57(s,1H),8.51(s,1H),8.03(d,1H),7.7(d,1H),7.58(s,1H),7.25(s,1H),7.06(d,1H),5.00(br,1H),4.42-4.44(m,1H),4.14-4.25(m,3H),3.60-3.72(m,1H),3.01-3.07(m,1H),2.81-2.87(m,1H),1.89-1.98(m,4H),1.20-1.45(m,5H),0.80-0.89(m,6H)。
实施例17:化合物40的制备
步骤A:((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000094
反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(1.0g,3.3mmol),((3R,4S)-3-羟基哌啶-4-基)碳酸叔丁酯(720mg,3.3mmol),DIEA(2.2g,10mmol)和20mL DMSO,90℃反应4h,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1.4g,m/z=494[M+1] +
步骤B:((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基盐酸盐
Figure PCTCN2022073467-appb-000095
反应瓶中加入((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁(1.4g,2.8mmol),4N盐酸二氧六环溶液(30mL,120 mmol)25℃反应0.5h,直接减压浓缩干,得到产品1.6g,m/z=394[M+1] +
步骤C:N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-3-甲基丁酰胺
Figure PCTCN2022073467-appb-000096
反应瓶中加入((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基盐酸盐(200mg,0.4mmol),3-甲基丁酸(48mg,0.47mmol),DIEA(166mg,1.29mmol),HATU(179mg,0.47mmol)和2.5mL DMF,25℃反应1h,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品42mg,m/z=478[M+1] +1H NMR(400MHz,DMSO-d6):δ12.64(s,1H),8.37(s,1H),8.50(s,1H),8.00(dd,1H),7.56-7.59(m,2H),7.23(s,1H),7.03(d,1H),4.91(s,1H),4.10-4.27(m,4H),3.92-3.99(m,1H),3.88-3.91(m,1H),3.18-3.21(m,1H),3.10-3.16(m,1H),1.97-2.03(m,3H),1.86-1.94(m,1H),1.56-1.60(m,1H),1.38-1.43(m,3H),0.88-0.94(m,6H)。
实施例18:化合物41的制备
Figure PCTCN2022073467-appb-000097
将(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),1-三氟甲基环丁酸(128mg,0.77mmol),DIEA(263mg,2.04mmol)和DCM(10mL),HATU(292mg,0.77mmol)置于50mL的反应瓶中,室温反应0.5小时,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品9.6mg,m/z=544[M+1] +1H NMR(400MHz,DMSO-d6)δ12.65(s,1H),8.59(s,1H),8.51(s,1H),8.04(d,1H),7.86(d,1H),7.58(s,1H),7.25(s,1H),7.08(d,1H),5.02(br,1H),4.51-4.54(m,1H),4.31-4.34(m,1H),4.18(q,2H),3.80-3.82(m,1H),3.50-3.53(m,1H),2.95-3.01(m,1H),2.73-2.79(m,1H),2.54-2.56(m,1H),2.30-2.33(m,2H),1.78-1.90(m,3H),1.48-1.38(m,4H)。
实施例19:化合物42的制备
Figure PCTCN2022073467-appb-000098
反应瓶中加入((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2- 基)-3-羟基哌啶-4-基)氨基盐酸盐(200mg,0.4mmol),3-氯-2-吡啶甲酸(74mg,0.47mmol)、DIEA(166mg,1.29mmol),HATU(179mg,0.47mmol)和2.5mL DMF,25℃反应1h,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得产品26mg,m/z=533[M+1] +1H NMR(400MHz,DMSO-d6)δ12.64(s,1H),8.54-8.57(m,3H),8.51(s,1H),8.00-8.04(m,2H),7.54-7.59(m,2H),8.19(dd,1H),7.25(s,1H),7.08(d,1H),5.11(s,1H),4.27-4.38(m,2H),4.13-4.17(m,3H),3.91-3.94(m,1H),3.15-3.20(m,1H),1.90-1.98(m,1H),1.73-1.78(m,1H),1.42-1.45(m,3H)。
实施例20:化合物43的制备
Figure PCTCN2022073467-appb-000099
将(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),DIEA(263mg,2.04mmol)和DCM(10mL),2-氯-5-氟苯甲酰氯(98mg,0.51mmol)置于50mL的反应瓶中,0℃下反应0.5小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品24mg,m/z=550[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.57(s,1H),8.52(s,1H),8.46(d,J=8.4Hz,1H),8.07(m,1H),7.53-7.59(m,2H),7.39-7.41(m,1H),7.30-7.35(m,2H),7.11-7.21(m,1H),7.45-7.47(m,1H),4.20-4.29(m,1H),4.14-4.18(m,2H),3.90-3.93(m,1H),3.52-3.55(m,2H),3.14-3.20(m,1H),2.93-2.96(m,1H),1.99-2.02(m,1H),1.49-1.51(m,1H),1.39(t,J=7.2Hz,3H)。
实施例21:化合物44的制备
步骤A:((3S,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基甲酸酯
Figure PCTCN2022073467-appb-000100
室温条件下,向50mL的反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(2g,6.73mmol),((3S,4S)-3-羟基哌啶-4-基)氨基甲酸叔丁酯(1.46g,6.73mmol),10mL DMSO和DIEA(2.61g,20.2mmol),90℃搅拌12小时,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得产品2.7g,m/z=494[M+1] +
步骤B:(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇
Figure PCTCN2022073467-appb-000101
向反应瓶中依次加入叔丁基((3S,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基甲酸酯(2.7g,5.45mmol)和浓度为4M盐酸的1,4-二氧六环溶液,搅拌3小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品2.7g,m/z=394[M+1] +
步骤C:2-氯-N-((3S,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000102
(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),DIEA(263mg,2.04mmol)和DCM(10mL)2-氯-6-氟苯甲酰氯(98mg,0.51mmol)置于50mL的反应瓶中,0℃下反应0.5小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得产品26mg,m/z=550[M+1] +1H NMR(400MHz,DMSO-d6)δ12.65(s,1H),8.69(d,1H),8.58(s,1H),8.57(s,1H),8.02-8.05(m,1H),7.58(s,1H),7.07-7.49(m,5H),5.07(br,1H),4.40-4.44(m,1H),4.14-4.22(m,3H),3.92-3.95(m,1H),3.50-3.51(m,1H),3.16-3.22(m,1H),2.96-3.01(m,1H),2.01-2.11(m,1H),1.37-1.48(m,4H)。
实施例22:化合物45的制备
Figure PCTCN2022073467-appb-000103
(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),DIEA(263mg,2.04mmol)和DCM(10mL),3-(三氟甲基)吡啶-2-甲酰氯(107mg,0.51mmol)置于50mL的反应瓶中,0℃下反应0.5小时,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得产品45mg,m/z=567[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.85-8.86(d,J=4.0Hz,1H),8.62-8.64(d,J=8.0Hz,1H),8.58(s,1H),8.57(s,1H),8.29-8.31(m,1H),8.04-8.07(m,1H),7.72-7.75(m,1H),7.59(s,1H),7.26(s,1H),7.09-7.11(m,1H),5.10(s,1H),4.46-4.50(m,1H),4.26-4.29(m,1H),4.14-4.20(m,2H),3.92-3.96(m,1H),3.54-3.59(m,1H),3.10-3.16(m,1H),2.90-2.95(m,1H),2.01-2.05(m,1H),1.49-1.52(m,1H),1.39(t,J=6.8Hz,3H)。
实施例23:化合物46的制备
Figure PCTCN2022073467-appb-000104
(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),DIEA(263mg,2.04mmol)和DCM(10mL),2-甲基-5-氟苯甲酰氯(78mg,0.51mmol)置于50mL的反应瓶中,0℃下反应0.5小时,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得产品33mg,m/z=530[M+1] +1H NMR(400MHz,DMSO-d6)δ12.65(s,1H),8.58(s,1H),8.57(s,1H),8.21(d,1H),8.04-8.06(m,1H),7.59(s,1H),7.09-7.28(m,5H),5.15(br,1H),4.50-4.54(m,1H),4.31-4.35(m,3H),4.17(q,2H),3.91-3.93(m,1H),3.50-3.54(m,1H),3.04-3.10(m,1H),2.82-2.88(m,1H),2.32(s,3H),1.96-1.98(m,1H),1.23-1.52(m,4H)。
实施例24:化合物48的制备
步骤A:((3S,4R)-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-羟基哌啶-3-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000105
向反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶(1.25g,4.22mmol),DMSO(12mL),DIEA(1.63g,12.66mmol),((3S,4R)-4-羟基哌啶-3-基)氨基碳酸叔丁酯(1.0g,4.64mmol),加热升温至90~95℃搅拌24小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1.2g,m/z=494[M+1] +
步骤B:((3S,4S)-4-(1,3-邻苯二甲亚酰-2-基)-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000106
向反应瓶中加入((3S,4R)-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-羟基哌啶-3-基)氨基碳酸叔丁酯(1.1g,2.22mmol)、THF(10mL)、邻苯二甲亚酰胺(0.65g,4.45mmol)、三苯基膦(1.2g,4.45mmol),0~10℃搅拌30min,滴加入DEAD(0.77g,4.45mmol)的THF(1mL)溶液,升温至室温反应20小时,加水搅拌淬灭,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得产品300mg,m/z=623[M+1] +
步骤C:((S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基) 哌啶-3基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000107
向反应瓶中加入((3S,4S)-4-(1,3-邻苯二甲亚酰-2-基)-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3基)氨基碳酸叔丁酯(300mg,0.48mmol)、乙醇(2mL)、水合肼(1ml),加热至回流1小时,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品100mg,m/z=493[M+1] +
步骤D:N-((3S,4S)-3-氨基-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-基)-2氯-6-甲基苯甲酰胺
Figure PCTCN2022073467-appb-000108
反应瓶A中加入2-甲基-6-氯苯甲酸(51.7mg,0.3mmol),二氯甲烷(2mL)降温至0~10℃,再加入二氯亚砜(36.2mg,0.3mmol),1滴DMF,自然升温至室温搅拌2小时,备用。
反应瓶B中加入((3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3’,4’:3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3基)氨基碳酸叔丁酯(100mg,0.2mmol),2mL二氯甲烷,三乙胺(41mg,0.4mmol)降温至0~10℃,将瓶A中反应液缓慢滴加入反应瓶B,滴完室温搅拌2小时,反应完全。加入2mL水分液,收集有机相,浓缩至干后加入2mL甲醇溶清,降温至0~10℃并加入20%氯化氢甲醇溶液,自然升至室温搅拌20小时,反应完全。浓缩干后加入5mL水溶清,乙酸乙酯萃取2次,收集水相,调节pH至10,二氯甲烷萃取两次,收集有机相,经柱层析纯化得产品18mg, 1HNMR(400MHz,DMSO-d6)δ12.65(s,1H),8.46-8.59(m,3H),8.05-8.08(m,1H),7.55-7.64(m,2H),7.22-7.33(m,4H),7.10-7.12(d,1H),4.50-4.53(m,1H),4.32(m,1H),4.15-4.20(m,2H),3.94(m,1H),3.30(m,1H),3.12(m,1H),2.80-2.91(m,1H),2.31-2.33(m,3H),2.06-2.08(m,1H),1.50-1.58(m,1H),1.34-1.42(m,3H),1.24(s,1H)m/z=545.2[M+1] +
实施例25:化合物50的制备
步骤A:(3S,4S)-1-(5-(6-乙氧基-1-(四氢-2氢-吡喃-2-基)-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000109
反应瓶加入4-(5-氯吡嗪-2-基)-6-乙氧基-1-(四氢-2氢-吡喃-2-基)-1氢-吡唑[3',4':3,4]吡唑 [1,5-a]吡啶(500mg,1.25mmol),((3S,4S)-3-羟基哌啶-4-基)氨基碳酸叔丁酯(542mg,2.5mmol),DIEA(484mg,3.75mmol)和5mL DMSO,80℃反应过夜,反应完全。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品670mg,m/z=579.3[M+1] +
步骤B:(3S,4S)-4-氨基-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶盐酸盐
Figure PCTCN2022073467-appb-000110
反应瓶加入(3S,4S)-1-(5-(6-乙氧基-1-(四氢-2氢-吡喃-2-基)-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯(670mg,1.1mmol),5N甲醇盐酸溶液(10mL,50mmol),25℃反应2h,反应完全,直接减压浓缩干,得到产品710mg,m/z=395.2[M+1] +
步骤C:3-氯-N-((3S,4S)-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶-4-基)-2-吡啶甲酰胺
Figure PCTCN2022073467-appb-000111
反应瓶加入(3S,4S)-4-氨基-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶盐酸盐(80mg,0.16mmol),3-氯吡啶-2-甲酸(38mg,0.24mmol),DIEA(103mg,0.8mmol),HATU(61mg,0.16mmol)和2mL DMF,25℃反应2h,反应完全。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品37.6mg,m/z=534.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.45(brs,1H),8.90(s,1H),8.74(d,J=7.6Hz,1H),8.71(s,1H),8.55(s,1H),8.01(s,1H),7.72(s,1H),7.47(dd,J 1=8.0Hz,J 2=6.0Hz,1H),7.36(d,J=8.0Hz,1H),7.28(t,J=8.8Hz,1H),5.01-5.42(m,1H),4.36-4.41(m,1H),4.17-4.24(m,2H),3.95-4.02(m,1H),3.62-3.71(m,2H),3.12-3.21(m,2H),2.10-2.18(m,1H),1.48-1.60(m,1H),1.39-1.43(m,3H)。
实施例26:化合物51的制备
步骤A:2-氯-N-((3S,4S)-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶-4-基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000112
反应瓶加入(3S,4S)-4-氨基-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶盐酸盐(80mg,0.16mmol),2-氯-6氟苯甲酰氯(46mg,0.24mmol),DIEA(103mg,0.8mmol)和2mL DMF,25℃反应1h,反应完全。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品4.6mg,m/z=551.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.54(brs,1H),8.91(s,1H),8.72(s,1H),8.59(d,J=8.4Hz,1H),8.55(s,1H),8.04(s,1H),8.02(s,1H),7.73(s,1H),7.52(dd,J 1=8.4Hz,J 2=4.8Hz,1H),5.01-5.28(m,1H),4.44-4.52(m,1H),4.25-4.33(m,1H),4.17(q,J=6.8Hz,1H),3.96-4.02(m,1H),3.58-3.67(m,1H),3.30-3.35(m,1H),3.03-3.11(m,1H),2.10-2.18(m,2H),1.56-1.68(m,1H),1.39(t,J=6.8Hz,3H).
实施例27:化合物55的制备
步骤A:4-溴-6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000113
向4-溴-6-羟基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(2.0g,7.1mmol)的四氢呋喃20ml溶液中加入二氢吡喃5ml,对甲苯磺酸(245mg,1.4mmol),加热50℃反应12h,加入水,用乙酸乙酯萃取,合并有机相,并用水洗涤,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得到产品,m/z=365[M+1] +
步骤B:6-乙氧基-1-(四氢-2H-吡喃-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000114
反应瓶加入4-溴-6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(5.4g,14.8mmol),联硼酸频那醇酯(11.3g,44.5mmol),醋酸钾(5.8g,59.3mmol)和100ml 1,4-二氧六环,Pd(dppf)Cl 2(1.0g,1.5mmol)氮气置换后,80℃反应12h,过滤,滤液浓缩后得到产品5.0g,m/z=413[M+1] +
步骤C:4-(5-氯吡嗪-2-基)-6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000115
反应瓶加入6-乙氧基-1-(四氢-2H-吡喃-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(4.5g,10.9mmol),2,5-二氯吡嗪(4.8g,32.6mmol),碳酸钾(4.5g,32.7mmol),1,4-二氧六环(50mL)和水(5mL),Pd(dppf)Cl 2(1.0g,1.5mmol)氮气置换后,80℃反应4h,加入水,用乙酸乙酯萃取,合并有机相,并用水洗涤,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得到产品2.5g,m/z=399[M+1] +
步骤D:(1-(5-(6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4甲基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000116
反应瓶加入4-(5-氯吡嗪-2-基)-6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(1.0g,2.5mmol)、(4-甲基哌啶-4-基)碳酸叔丁酯(1.0g,5.0mmol),DIEA(972mg,7.5mmol)和12mL DMSO,80℃反应12h,加入水,用乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得产品1.3g,m/z=577[M+1] +
步骤E:(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4甲基哌啶-4-氨基盐酸盐
Figure PCTCN2022073467-appb-000117
反应瓶加入(1-(5-(6-乙氧基-1-(四氢-2H-吡喃-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-基)氨基碳酸叔丁酯(1.25g,2.2mmol),5N甲醇盐酸溶液(10mL,50mmol)25℃反应2h,减压浓缩干得产品1.1g,m/z=393[M+1] +
步骤F:3-氯-N-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-基)-5-氟-2-吡啶甲酰胺
Figure PCTCN2022073467-appb-000118
反应瓶加入(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-氨基盐酸盐(150mg,0.3mmol)、3-氯-5-氟吡啶-2-甲酸(63mg,0.36mmol),DIEA(193mg,1.5mmol)、HATU(114mg,0.3mmol)和2mL DMF,25℃反应2h,加入水,用乙酸乙酯萃取,合并有机相,并用水洗涤,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得到产品11mg,m/z=550[M+1] +
实施例28:化合物56的制备
Figure PCTCN2022073467-appb-000119
反应瓶加入(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4甲基哌啶-4-氨基盐酸盐(150mg,0.3mmol),3-氯-5-氟吡啶-2-甲酸(63mg,0.36mmol),DIEA(193mg,1.5mmol),HATU(114mg,0.3mmol)和2mL DMF,25℃反应2h,加入水,用乙酸乙酯萃取,合并有机相,并用水洗涤,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得到产品13mg,m/z=546[M+1] +
实施例29:化合物57的制备
步骤A:N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4甲基哌啶-4-基)-2,3,6-三氟苯甲酰胺
Figure PCTCN2022073467-appb-000120
反应瓶加入(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-氨基盐酸盐(100mg,0.2mmol),2,3,6-三氟苯甲酸(39mg,0.22mmol),DIEA(129mg,1.0mmol),HATU(76mg,0.2mmol)和2mL DMF,25℃反应2h,反应完全。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品24.6mg,m/z=551.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.47(brs,1H),8.91(s,1H),8.73(s,1H),8.60(s,1H),8.55(d, J=2.0Hz,1H),8.02(s,1H),7.72(s,1H),7.55-7.63(m,1H),7.21-7.26(m,1H),4.16-4.24(m,4H),3.30-3.35(m,2H),1.62-1.66(m,2H),1.39-1.44(m,6H)。
实施例30:化合物58的制备
步骤A:2-氯-N-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-基)-5-氟苯甲酰胺
Figure PCTCN2022073467-appb-000121
反应瓶加入(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-氨基盐酸盐(150mg,0.3mmol),2-氯-5-氟苯甲酰氯(63mg,0.36mmol),DIEA(193mg,1.5mmol),HATU(76mg,0.2mmol)和2mL DMF,25℃反应1h,反应完全。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品43.6mg,m/z=549.2[M+1] +1H NMR(400MHz,CD 3OD-d 4)δ8.75(s,1H),8.54(s,1H),8.33(s,1H),8.00(s,1H),7.62(s,1H),7.46-7.49(m,1H),7.17-7.26(m,2H),4.15-4.25(m,4H),3.48-3.55(m,2H),2.44-2.48(m,2H),1.70-1.77(m,2H),1.55(s,3H),1.49(t,J=6.8Hz,3H)。
实施例31:化合物59的制备
步骤A:3-氯-N-(1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4甲基哌啶-4-基)-2-吡啶甲酰胺
Figure PCTCN2022073467-appb-000122
反应瓶加入(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-4-甲基哌啶-4-氨基盐酸盐(100mg,0.2mmol),3-氯-2-吡啶苯甲酸(39mg,0.22mmol),DIEA(129mg,1.0mmol),HATU(76mg,0.2mmol)和2mL DMF,25℃反应2h,反应完全。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品43.5mg,m/z=532.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.47(brs,1H),8.91(s,1H),8.72(s,1H),8.52-8.55(m,2H),8.35(s,1H),8.01-8.03(m,2H),7.72(s,1H),7.50(dd,J 1=8.0Hz,J 2=0.8Hz,1H),4.16-4.22(m,4H),3.34-3.39(m,2H),2.35-2.39(m,2H),1.60-1.66(m,2H),1.56(s,3H),1.39(t,J=6.8Hz,3H).
实施例32:化合物87的制备
1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)-N-异丁基哌啶-4-酰胺(化合物APS03087)
步骤A:1-(5-溴吡啶-2-基)哌啶-4-羧酸甲酯
Figure PCTCN2022073467-appb-000123
向1-(叔丁氧羰基)-4-(甲氧羰基)哌啶-4-羧酸(1.9g,6.6mmol)中,加入氯化氢的1,4-二氧六环(4M,4mL)室温搅拌1小时后,旋干,加入DMSO(10mL),碳酸钾(3.3g,23.3mmol),5-溴-2-氟吡啶(1.4g,7.9mmol),加热至90℃反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1g,m/z=298/300[M+1] +
步骤B:4-((苄氧基)甲基)-1-(5-溴吡啶-2-基)哌啶-4-羧酸甲酯
Figure PCTCN2022073467-appb-000124
向100mL的反应瓶中加入1-(5-溴吡啶-2-基)哌啶-4-羧酸甲酯(1.0g,3.34mmol),四氢呋喃(15mL),降温至-70℃,控温-70±5℃下滴加正丁基锂(2M,1.67mL,3.34mmol),滴完保温搅拌10分钟后,控温-70±5℃下滴加入氯甲氧基苄醚(1.0g,6.68mmol),滴完后,自然升温至室温搅拌2小时,氯化铵水溶液淬灭后,加入二氯甲烷萃取,柱层析分离得产品1.2g。m/z=418/420[M+1] +
步骤C:4-羟甲基-1-(5-溴吡啶-2-基)哌啶-4-羧酸甲酯
Figure PCTCN2022073467-appb-000125
向50mL的反应瓶中加入4-((苄氧基)甲基)-1-(5-溴吡啶-2-基)哌啶-4-羧酸甲酯(1.2g,2.86mmol),二氯甲烷(24mL)和氯化铝(1.5g,11.45mmol),室温搅拌3小时,氢氧化钠调节pH值至7左右,加入二氯甲烷萃取,柱层析分离得到产品0.71g,m/z=328/330[M+1] +
步骤D:4-羟甲基-1-(5-溴吡啶-2-基)哌啶-4-羧酸
Figure PCTCN2022073467-appb-000126
向50mL的反应瓶中加入4-羟甲基-1-(5-溴吡啶-2-基)哌啶-4-羧酸甲酯(0.71g,2.1mmol),四氢呋喃(10mL),甲醇(10mL)和氢氧化锂(181mg,4.31mmol)的水(10mL)溶液,60℃搅拌2小时,浓缩后,盐酸调节pH至5-6,二氯甲烷萃取,浓缩后得到产品315mg,m/z=314/316[M+1] +
步骤E:1-(5-溴吡啶-2-基)-4-(羟甲基)-N-异丁基哌啶-4-酰胺
Figure PCTCN2022073467-appb-000127
向50mL的反应瓶中加入4-羟甲基-1-(5-溴吡啶-2-基)哌啶-4-羧酸(315mg,1mmol),乙腈(3mL),三乙胺(303mg,3mmol),异丁胺(146mg,2.0mmol),再加入HATU(570mg,1.5mmol),室温搅拌0.5小时,反应液倒入水中,二氯甲烷萃取,柱层析得产品270mg,m/z=370/372[M+1] +
步骤F:1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)-N-异丁基哌啶-4-酰胺
Figure PCTCN2022073467-appb-000128
向反应瓶中依次加入1-(5-溴吡啶-2-基)-4-(羟甲基)-N-异丁基哌啶-4-羧酰胺(36.7mg,0.13mmol),6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶(50mg,0.13mmol),四三苯基磷钯(23mg,0.02mmol),碳酸钾(37.3mg,0.27mmol),水(0.25mL),1,4-二氧六环(0.5mL),氮气置换三次,加热至90℃反应16小时,反应液倒入水中,二氯甲烷萃取,柱层析纯化得产品6mg,m/z=492[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.57(s,1H),8.43-8.49(m,2H),7.93-7.96(m,1H),7.51-7.60(m,2H),7.18-7.18(d,1H),6.96-7.02(m,1H),4.83-4.85(m,1H),3.99-4.19(m,4H),3.36-3.37(m,2H),3.11(m,2H),2.86-2.89(m,2H),1.98-2.01(m,2H),1.17-1.71(m,1H),1.43(m,2H),1.36-1.34(m,4H),0.70-0.80(m,6H)。
实施例33:化合物88的制备
((3S,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基甲酸叔丁酯
Figure PCTCN2022073467-appb-000129
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(50mg,0.17mmol),((3S,4S)-3-羟基哌啶-4-基)氨基甲酸叔丁酯(40mg,0.17mmol),DIEA(65mg,0.5mmol),DMSO(3mL)加入反应瓶,90℃反应4h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得产品12mg,m/z=494[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.56(s,1H),8.50(s,1H),8.01(d,J=8.8Hz,1H),7.57(s,1H),7.26(s,1H),7.04(d,J=8.8Hz,1H),6.72(brs,1H),4.98(d,J=4.8Hz,1H),4.43-4.46(m,1H),4.20-4.27(m,1H),4.15(q,J=6.8Hz,2H),3.35-3.41(m,2H),2.96-3.02(m,1H),2.75-2.80(m,1H),1.86-1.88(m,1H),1.35-1.42(m,13H)。
实施例34:化合物89的制备
Figure PCTCN2022073467-appb-000130
(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇(256.5mg,0.51mmol),2,3-二甲基丁酸(60mg,0.51mmol),DIEA(329mg,2.55mmol),HATU(232mg,0.61mmol)和DCM(10mL)置于50mL的反应瓶中,室温反应0.5小时,加入水,用乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩,柱层析分离得产品40mg, m/z=492[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.64(s,1H),8.50-8.60(m,2H),8.05(m,1H),7.65(m,1H),7.60(s,1H),7.10-7.15(m,1H),4.30-4.50(m,1H),4.10-4.30(m,3H),3.70-3.80(m,1H),3.50-3.60(m,2H),3.06-3.39(m,1H),2.75-2.95(m,1H),1.88-1.96(m,2H),1.66-1.70(m,1H),1.33-1.41(m,4H),0.83-0.98(m,9H)。
实施例35:化合物91的制备
步骤A:2-氯-N-((3S,4S)-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶-4-基)-5-氟苯甲酰胺
Figure PCTCN2022073467-appb-000131
反应瓶加入(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡嗪-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),2-氯-5-氟苯甲酰氯(46mg,0.24mmol),DIEA(129mg,1.0mmol)和2mL DMF,25℃反应1h,反应完全。二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品8.5mg,m/z=551.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.47(brs,1H),8.91(s,1H),8.72(s,1H),8.55(s,1H),8.47(d,J=8.0Hz,1H),8.01(s,1H),7.73(s,1H),7.54(dd,J 1=8.8Hz,J 2=4.0Hz,1H),7.40-7.43(m,1H),7.31-7.36(m,1H),5.25(s,1H),4.42-4.46(m,1H),4.28-4.32(m,1H),4.17(t,J=6.8Hz,2H),3.89-3.99(m,1H),3.51-3.57(m,1H),3.25-3.27(m,1H),3.02-3.10(m,1H),2.01-2.08(m,1H),1.48-1.54(m,1H),1.39(t,J=6.8Hz,3H)。
实施例36:化合物101的制备
步骤A:4-(N-异丙基磺酰基)哌啶-1-碳酸叔丁酯
Figure PCTCN2022073467-appb-000132
向反应瓶中加入异丙胺(97mg,1.7mmol),二氯甲烷(10mL),搅拌,加入三乙胺(300mg,3mmol),降温至10℃,加入4–(氯磺酰基)哌啶-1-羧酸叔丁酯(426mg,1.5mmol),反应1小时。加水淬灭,二氯甲烷萃取,无水硫酸钠干燥,过滤,减压浓缩,得产品400mg直接用于下步反应,m/z=307[M+1] +
步骤B:N-异丙基哌啶-4-磺酰胺
Figure PCTCN2022073467-appb-000133
向反应瓶中依次加入4-(N-异丙基磺酰基)哌啶-1-碳酸叔丁酯(400mg,1.3mmol)和HCl的1.4-二氧六环溶液(4M,10mL),室温反应1小时,反应完全。减压浓缩,得产品340mg直接用于下步反应,m/z=207[M+1] +
步骤C:1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-N-异丙基哌啶-4-磺酰胺
Figure PCTCN2022073467-appb-000134
向反应瓶中依次加入DMSO(2mL)和DIEA(0.5mL),N-异丙基哌啶-4-磺酰胺(340mg,1.3mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.3mmol),100℃下反应48小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品46mg,m/z=484[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.60(d,J=2.6Hz,1H),8.53(d,J=2.1Hz,1H),8.06(dd,J=8.9,2.6Hz,1H),7.58(s,1H),7.28(d,J=2.0Hz,1H),7.10(dd,J=19.8,8.4Hz,2H),4.56(d,J=13.3Hz,2H),4.18(q,J=6.9Hz,2H),3.46(q,J=6.7Hz,1H),3.32(s,1H),3.02(t,J=12.2Hz,2H),2.06(d,J=11.3Hz,2H),1.60(qd,J=12.5,4.2Hz,2H),1.40(t,J=6.9Hz,3H),1.13(d,J=6.5Hz,6H).
实施例37:化合物102的制备
步骤A:4-羟基-4-(苯磺酰胺亚甲基)哌啶-1-碳酸叔丁酯
Figure PCTCN2022073467-appb-000135
向反应瓶中加入4-(酰胺基亚甲基)-4-羟基哌啶-1-碳酸叔丁酯(230mg,1mmol),二氯甲烷(10mL),三乙胺(200mg,2mmol),降温至10℃,加入苯甲酰氯(194mg,1.1mmol),反应1小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品300mg,m/z=371[M+1] +
步骤B:N-((4-羟基哌啶-4-基)亚甲基)苯磺酰胺
Figure PCTCN2022073467-appb-000136
向反应瓶中依次加入4-羟基-4-(苯磺酰胺亚甲基)哌啶-1-碳酸叔丁酯(300mg,0.8mmol)和HCl的1,4-二氧六环溶液(4M,10mL),室温反应1小时,反应完全。减压浓缩,得产品320mg直接用于下步反应,m/z=271[M+1] +
步骤C:N-((1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-羟基哌啶-4-基)亚甲基)苯磺酰胺
Figure PCTCN2022073467-appb-000137
向反应瓶中依次加入DMSO(2mL)和DIEA(0.5mL),N-((4-羟基哌啶-4-基)亚甲基)苯磺酰胺(300mg,0.8mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.3mmol),100℃下反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品93mg,m/z=548[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.58(d,J=2.6Hz,1H),8.51(d,J=2.1Hz,1H),8.03(dd,J=8.9,2.6Hz,1H),7.86–7.78(m,2H),7.69–7.53(m,5H),7.26(d,J=2.1Hz,1H),7.08(d,J=9.0Hz,1H),4.60(s, 1H),4.23–4.07(m,4H),3.35–3.24(m,2H),2.72(d,J=6.7Hz,2H),1.60(td,J=12.8,12.1,4.4Hz,2H),1.48(d,J=13.2Hz,2H),1.40(t,J=6.9Hz,3H)。
实施例38:化合物103的制备
步骤A:4-(吡啶-2-氧基)哌啶-1-甲酸叔丁酯
Figure PCTCN2022073467-appb-000138
向反应瓶中加入4-羟基哌啶-1-羧酸叔丁酯(804mg,4mmol),四氢呋喃(10mL),搅拌,降温至10℃,加入NaH(含量60%,192mg,4.8mmol),反应1小时,加入2-氟吡啶(388mg,4mmol)。反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品950mg,m/z=279[M+1] +
步骤B:2-(哌啶-4-氧基)吡啶
Figure PCTCN2022073467-appb-000139
向反应瓶中依次加入4-(吡啶-2-氧基)哌啶-1-甲酸叔丁酯(950mg,3.4mmol)和HCl的1.4-二氧六环溶液(4M,10mL),室温反应1小时,反应完全。减压浓缩干,得产品750mg直接用于下步反应,m/z=179[M+1] +
步骤C:6-乙氧基-4-(6-(4-(吡啶-2-氧基)哌啶-1-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]哌啶
Figure PCTCN2022073467-appb-000140
向反应瓶中依次加入DMSO(2mL)和DIEA(0.5mL),N-((4-羟基哌啶-4-基)甲基)苯磺酰胺(150mg,0.7mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(60mg,0.2mmol),100℃下反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品22mg,m/z=456[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.60(d,J=2.6Hz,1H),8.53(d,J=2.1Hz,1H),8.19(dd,J=5.1,2.0Hz,1H),8.06(dd,J=8.9,2.6Hz,1H),7.72(ddd,J=8.8,7.1,2.1Hz,1H),7.60(s,1H),7.27(d,J=2.1Hz,1H),7.13(d,J=8.9Hz,1H),6.94-7.03(m,1H),6.82(d,J=8.3Hz,1H),5.31(tt,J=8.3,3.9Hz,1H),4.17(dd,J=13.4,6.2Hz,4H),3.45(ddd,J=13.1,9.5,3.2Hz,2H),2.10(dq,J=8.1,3.8Hz,2H),1.71(dtd,J=12.7,9.0,3.7Hz,2H),1.41(t,J=6.9Hz,3H)。
实施例39:化合物104的制备
步骤A:4-((6-甲基哒嗪-3-基)氧)哌啶-1-碳酸叔丁酯
Figure PCTCN2022073467-appb-000141
向反应瓶中加入4-羟基哌啶-1-羧酸叔丁酯(402mg,2mmol),四氢呋喃(10mL),搅拌,降温至10℃,加入NaH(含量60%,96mg,2.4mmol),反应1小时,加入3-氯-6-甲基哒嗪(258mg,2mmol)。反应24小时,加水,乙酸乙酯提取,无水硫酸钠干燥,过滤,减压浓缩干,硅胶柱纯化,得产品250mg,m/z=294[M+1] +
步骤B:3-甲基-6-(吡啶-4-氧基)哒嗪
Figure PCTCN2022073467-appb-000142
向反应瓶中依次加入4-((6-甲基哒嗪-3-基)氧)哌啶-1-碳酸叔丁酯(250mg,0.8mmol)和 HCl的1.4-二氧六环溶液(4M,10mL),室温反应1小时,反应完全。减压浓缩,得产品210mg直接用于下步反应,m/z=194[M+1] +
步骤C:6-乙氧基-4-(6-(4-((6-甲基哒嗪-3-基)氧)哌啶-1-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000143
向反应瓶中依次加入DMSO(2mL)和DIEA(0.5mL),3-甲基-6-(吡啶-4-氧基)哒嗪(210mg,0.8mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(80mg,0.2mmol),100℃下反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品27mg,m/z=471[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.60(d,J=2.5Hz,1H),8.52(d,J=2.1Hz,1H),8.06(dd,J=8.9,2.6Hz,1H),7.60(s,1H),7.51(d,J=9.0Hz,1H),7.27(d,J=2.1Hz,1H),7.12(dd,J=9.0,7.2Hz,2H),5.46(tt,J=8.3,3.8Hz,1H),4.17(p,J=8.6,7.7Hz,4H),3.49(ddd,J=13.0,9.2,3.2Hz,2H),2.53(s,3H),2.21–2.11(m,2H),1.76(dtd,J=12.7,8.9,3.8Hz,2H),1.40(t,J=6.9Hz,3H).
实施例40:化合物107的制备
(3S,4S)-4-((2-氯-6-氟苯)氨基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇
Figure PCTCN2022073467-appb-000144
将(3S,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇盐酸盐(100mg,0.2mmol),1-氯-2-(氯甲基)-3-氟苯(50mg,0.2mmol),Cs 2CO 3(130mg,0.4mmol),DMF(3mL)加入反应瓶,50℃反应12h。将反应液倒入30mL水中,乙酸乙酯萃取,干燥有机相,旋干溶剂,柱层析纯化产品4.6mg,m/z=535[M+1]+。 1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.56(s,1H),8.50(s,1H),8.01(d,J=8.8Hz,1H),7.58(s,1H),7.31-7.42(m,2H),7.22-7.30(m,2H),7.05(d,J=8.8Hz,1H),5.18(d,J=4.0Hz,1H),4.42-4.48(m,1H),4.29-4.35(m,1H),4.14(m,2H),3.81-3.98(m,2H),3.20-3.28(m,2H),2.88-2.98(m,1H),2.66-2.74(m,1H),2.41-2.47(m,2H),2.10-2.20(m,1H),1.37(m,3H)。
实施例41:化合物118的制备
N-((3aR,6aS)-5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)六氢吡醇[3,4-c]吡咯-2(1H)-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000145
将6-乙氧基-4-(6-((3aR,6aS)-六氢吡醇[3,4-c]吡咯-2(1H)-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(200mg,0.4mmol),2-氯-6-氟苯甲酰氯(77mg,0.4mmol),DIEA(258mg,2mmol),DCM(10mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品4.6mg,m/z=546[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.55(s,1H),8.51(s,1H),8.01(d,J=8.8Hz,1H),7.48-7.61(m,2H),7.33-7.47(m,2H),7.25(s,1H),6.70-6.80(m,1H),4.15(q,J=6.8Hz,2H),3.70-3.86(m,3H),3.46-3.53(m,4H),3.06-3.24(m,3H),1.38(t,J=6.8Hz,3H).
实施例42:化合物150的制备
步骤A:4-(2,5-二氟苯酰胺酰胺)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯
Figure PCTCN2022073467-appb-000146
将4-((叔丁基羰基)氨基)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯盐酸盐(120mg,0.26mmol),2,5-二氟苯甲酸(37.9mg,0.24mmol),DIEA(100mg,0.78mmol),HATU(108mg,0.28mmol),DMF(2ml)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品78mg,m/z=590[M+1] +
步骤B:N-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)哌啶-4-基)-2,5-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000147
将4-(2,5-二氟苯酰胺酰胺)-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸乙酯(29.4mg,0.05mmol),2ml THF加入反应瓶,0℃下加入LiAlH 4(1.9mg,0.05mmol),反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析 分离得到产品15mg,m/z=548[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.62–8.56(m,1H),8.52(d,J=2.1Hz,1H),8.08–7.99(m,2H),7.60(s,1H),7.47–7.29(m,3H),7.27(d,J=2.1Hz,1H),7.11(d,J=9.0Hz,1H),4.88(t,J=5.9Hz,1H),4.21(m,2H),3.64(d,J=6.0Hz,2H),3.33(s,2H),3.23(t,J=12.4Hz,2H),2.29(d,J=13.6Hz,2H),1.64(m,2H),1.40(t,J=6.9Hz,3H)。
实施例43:化合物151的制备
步骤A:3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-碳酸叔丁酯
Figure PCTCN2022073467-appb-000148
三口瓶中依次4-溴-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(0.28g,1mmol),1,4-二氧六环(2.8mL),6-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼硼烷-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-3-碳酸叔丁酯(0.52g,1.3mmol),碳酸锂(148mg,2mmol),四三苯基磷钯(23mg,0.02mmol),水(1.4mL),升温至90℃,反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品230mg,m/z=476.2[M+1] +
步骤B:4-(6-(3,6-二氮杂双环[3.1.1]庚烷-6-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000149
三口瓶中依次加入3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-碳酸叔丁酯(200mg,0.42mmol),甲醇(4mL),间苯二甲醚(2.9mg,0.02mmol),加入硫酸(98%,168mg,1.68mmol),升温至55℃反应4小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品300mg,m/z=376.2[M+1] +
步骤C:6-乙氧基-4-(6-(3-(((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-6-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000150
三口瓶中依次加入4-(6-(3,6-二氮杂双环[3.1.1]庚烷-6-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(180mg,0.27mmol),正丙醇(3.6mL),三乙胺(136.2mg,1.35mmol),6-甲氧基-3-吡啶甲醛(111mg,0.81mmol),Pic-BH 3(86.7mg,0.81mmol),保温50℃搅拌24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品70mg,m/z=497[M+1] +1H NMR(400MHz,DMSO-d6)δ12.67(s,1H),8.53-8.56(m,2H),7.99-8.02(m,1H),7.76-7.76(m,1H),7.59(s,1H),7.31-7.32(m,1H),7.19-7.22(m,1H), 6.69-6.71(m,1H),6.49-6.52(m,1H),4.38-4.40(m,2H),4.16-4.21(m,2H),3.74(s,3H),3.56(s,2H),3.21-3.24(m,2H),2.69-2.72(m,2H),2.56-2.60(m,1H),1.75-1.77(d,1H),1.39-1.42(m,3H)。
实施例44:化合物152的制备
步骤A:(3aR,6aS)-5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)六氢吡醇[3,4-c]吡咯-2(1H)-羧酸叔丁酯
Figure PCTCN2022073467-appb-000151
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(700mg,2.4mmol),(3aR,6aS)-六氢吡醇[3,4-c]吡咯-2(1H)-羧酸叔丁酯(700mg,2.4mmol),DIEA(1.5g,11.6mmol),DMSO(10mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1g,m/z=490[M+1] +
步骤B:6-乙氧基-4-(6-((3aR,6aS)-六氢吡醇[3,4-c]吡咯-2(1H)-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000152
向(3aR,6aS)-5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)六氢吡醇[3,4-c]吡咯-2(1H)-羧酸叔丁酯(1g,2.0mmol)中加入HCl的甲醇溶液(4M,20mL),25℃反应4h,旋干溶剂得产品1.1g,m/z=390[M+1] +
6-乙氧基-4-(6-((3aR,6aS)-5-((6-甲氧基吡啶-3-基)亚甲基)-六氢吡醇[3,4-c]吡咯-2(1H)-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000153
步骤C:将6-乙氧基-4-(6-((3aR,6aS)-六氢吡醇[3,4-c]吡咯-2(1H)-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(200mg,0.4mmol),6-甲氧基-3-吡啶甲醛(165mg,1.2mmol),Et 3N(202mg,2.0mmol),Pic-BH 3(128mg,1.2mmol),正丙醇(10mL)加入反应瓶,40℃反应48h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品67.2mg,m/z=511[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.55(s,1H),8.51(s,1H),8.06(s,1H),8.00(d,J=8.8Hz,1H),7.58-7.64(m,2H),7.23(s,1H),6.72-6.78(m,2H),4.15(q,J=6.8Hz,2H),3.82(s,3H),3.62-3.71(m,2H),3.54(s,2H),3.35-3.41(m,2H),2.88-2.97(m,2H),2.61-2.71(m,2H),2.42-2.44(m,2H),1.38(t,J=6.8Hz,3H).
实施例45:化合物155的制备
步骤A:(1R,5S)-3-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,8-二氮杂二环[3.2.1]辛烷-8-碳酸叔丁酯
Figure PCTCN2022073467-appb-000154
反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(292mg,1.0mmol),3,8-二氮杂二环[3.2.1]辛烷-8-碳酸叔丁酯(250mg,1.2mmol),DIEA(506mg,3.9mmol)和3mL DMSO,95℃反应4h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品208mg,m/z=490[M+1] +
步骤B:4-(6-((1R,5S)-3,8-二氮杂二环[3.2.1]辛烷-3-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000155
反应瓶加入(1R,5S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,8-二氮杂二环[3.2.1]辛烷-8-碳酸叔丁酯(200mg,0.4mmol),5N甲醇盐酸溶液(3ml,15mmol),25℃反应4h,直接减压浓缩得到产品250mg,m/z=390[M+1] +
步骤C:(2-氯-6-氟)((1R,5S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,8-二氮杂二环[3.2.1]辛烷-8-基)苯甲酰胺
Figure PCTCN2022073467-appb-000156
反应瓶中加入4-(6-((1R,5S)-3,8-二氮杂二环[3.2.1]辛烷-3-基)吡啶-3-基)-6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(120mg,0.24mmol),3-氯-6-氟苯甲酰氯(10mg,0.26mmol),DIEA(155mg,1.2mmol),2mL DMF,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品136mg,m/z=546[M+1] +1H NMR(400MHz,CD 3OD-d 3)δ8.58(s,1H),8.31(s,1H),8.01(d,J=8.8Hz,1H),7.65(s,1H),7.49-7.55(m,1H),7.39-7.42(m,1H),7.24-7.30(m,2H),7.00(d,J=8.8Hz,1H),5.03-5.04(m,1H),4.31-4.35(m,1H),4.13-4.20(m,3H),3.92-3.94(m,1H),3.18-3.25(m,1H),3.04-3.07(m,1H),1.92-2.13(m,4H),1.45(t,J=6.8Hz,3H)。
实施例46:化合物156的制备
步骤A:6-乙氧基-4-(6-((1R,5S)-8-((6-甲氧基吡啶-3-基)甲基)-3,8-二氮杂二环[3.2.1]辛烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡唑
Figure PCTCN2022073467-appb-000157
反应瓶中加入4-(6-((1R,5S)-3,8-二氮杂二环[3.2.1]辛烷-3-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(100mg,0.2mmol),6-甲氧基-3-吡啶甲醛(82mg,0.6mmol),TEA(121mg,1.2mmol),2-甲基吡啶硼烷(64mg,0.6mmol),和正丙醇3mL,45℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品37.6mg,m/z=511[M+1] +1H NMR(400MHz,CD 3OD-d 3)δ8.53(s,1H),8.27(s,1H),8.13(s,1H),7.95(d,J=8.8Hz,1H),7.79(d,J=8.4Hz,1H),7.66(s,1H),7.19(s,1H),6.89(d,J=8.8Hz,1H),6.80(d,J=8.4Hz,1H),4.11(q,J=6.8Hz,2H),3.93-3.97(m,2H),3.91(s,3H),3.59(s,2H),3.35-3.38(m,2H),3.15-3.18(m,2H),2.15-2.18(m,2H),1.45(t,J=6.8Hz,3H)。
实施例47:化合物157的制备
步骤A:(1R,5S)-8-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,8-二氮杂二环[3.2.1]辛烷-3-碳酸叔丁酯
Figure PCTCN2022073467-appb-000158
反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(292mg,1.0mmol),3,8-二氮杂二环[3.2.1]辛烷-3-碳酸叔丁酯(250mg,1.2mmol),DIEA(506mg,3.9mmol)和2mL DMSO,120℃反应16h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品220mg,m/z=490[M+1] +
步骤B:4-(6-((1R,5S)-3,8-二氮杂二环[3.2.1]辛烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000159
反应瓶加入(1R,5S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,8-二氮杂二环[3.2.1]辛烷-8-碳酸叔丁酯(200mg,0.4mmol),甲醇3mL,5N甲醇盐酸溶液(3mL,15mmol),25℃反应4h,反应完全,减压浓缩得产品280mg直接用于下步反应,m/z=390[M+1] +
步骤C:(2-氯-6-氟)((1R,5S)-8-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,8-二氮杂二环[3.2.1]辛烷-3-基)苯甲酰胺
Figure PCTCN2022073467-appb-000160
反应瓶中加入4-(6-((1R,5S)-3,8-二氮杂二环[3.2.1]辛烷-8-基)吡啶-3-基)-6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(100mg,0.2mmol),3-氯-6-氟苯甲酰氯(43mg,0.22mmol),DIEA(155mg,1.2mmol),2mL DMF,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品23.2mg,m/z=546[M+1] +1H NMR(400MHz,CD 3OD-d 3)δ8.59(s,1H),8.30(s,1H),8.02(d,J=8.8Hz,1H),7.65(s,1H),7.46-7.52(m,1H),7.42-7.44(m,1H),7.17-7.27(m,2H),7.03(d,J=8.8Hz,1H),4.82-4.87(m,1H),4.63-4.68(m,1H),4.42(t,J=11.2Hz,1H),4.12(q,J=7.2Hz,2H),3.57(t,J=13.6Hz,1H),3.18-2.24(m,1H),1.92-2.18(m,4H),1.45(t,J=6.8Hz,3H).
实施例48:化合物158的制备
步骤A:6-乙氧基-4-(6-((1R,5S)-3-((6-甲氧基吡啶-3-基)甲基)-3,8-二氮杂二环[3.2.1]辛烷-3-基)吡啶-8-基)-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡唑
Figure PCTCN2022073467-appb-000161
反应瓶中加入4-(6-((1R,5S)-3,8-二氮杂二环[3.2.1]辛烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(100mg,0.2mmol),6-甲氧基-3-吡啶甲醛(82mg,0.6mmol),TEA(121mg,1.2mmol),2-甲基吡啶硼烷(64mg,0.6mmol),和正丙醇3mL,45℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品45.5mg,m/z=511[M+1] +1H NMR(400MHz,CD 3OD-d 3)δ8.55(s,1H),8.32(s,1H),8.00-8.10(m,2H),7.68-7.72(m,2H),7.25(s,1H),6.95(d,J=9.2Hz,1H),6.78(d,J=8.8Hz,1H),4.63(s,2H),4.17(q,J=6.8Hz,2H),3.88(s,3H),3.42-3.45(m,2H),2.68-2.72(m,2H),2.42-2.48(m,2H),2.02-2.11(m,2H),1.95-2.00(m,2H),1.46(t,J=6.8Hz,3H).
实施例49:化合物159的制备
步骤A:叔丁基5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,5-二氮杂二环[2.2.2]辛烷-2-羧酸酯
Figure PCTCN2022073467-appb-000162
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(318mg,1.1mmol),叔丁基2,5-二氮杂二环[2.2.2]辛烷-2-羧酸酯(250mg,1.2mmol),DIEA(414mg,3.2mmol),DMSO(5mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品500mg,m/z=490[M+1] +
步骤B:4-(6-(2,5-二氮杂二环[2.2.2]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑 [1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000163
将叔丁基5-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,5-二氮杂二环[2.2.2]辛烷-2-羧酸酯(500mg,1.02mmol)加入5mL MeOH/HCl(4M),25℃反应4h,旋干溶剂得产品500mg直接用于下步反应,m/z=390[M+1] +
步骤C:(2-氯-6-氟苯基)-5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,5-二氮杂二环[2.2.2]辛烷-2-基)甲酮
Figure PCTCN2022073467-appb-000164
将4-(6-(2,5-二氮杂二环[2.2.2]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(500mg,1.18mmol),2-氯-6-氟苯甲酰氯(250mg,1.29mmol),Et 3N(357.5mg,3.54mmol),DMF(5mL)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品412mg,m/z=546[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.59(d,J=2.5Hz,1H),8.52(d,J=1.9Hz,1H),8.07(dd,J=8.9,2.5Hz,1H),7.63-7.58(m,1H),7.58-7.51(m,1H),7.51-7.46(m,1H),7.45-7.36(m,1H),7.25(m,1H),6.81(d,J=8.9Hz,1H),4.18(m,2H),3.85-3.70(m,4H),3.67-3.53(m,2H),2.04-1.95(m,2H),1.90(d,J=9.3Hz,2H),1.40(m,3H)。
实施例50:化合物160的制备
6-乙氧基-4-(6-(5-((6-甲氧基吡啶-3-基)甲基)-2,5-二氮杂二环[2.2.2]辛烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000165
将4-(6-(2,5-二氮杂二环[2.2.2]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(500mg,1.18mmol),6-甲氧基-3-吡啶甲醛(176mg,1.29mmol),Et 3N(357.5mg,3.54mmol),Pic-BH 3(378.8mg,3.54mmol),正丙醇(5mL)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品351mg,m/z=511[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.54(d,J=2.5Hz,1H),8.50(d,J=2.1Hz,1H),8.11(d,J=2.3Hz,1H),8.03(m,1H),7.70(m,1H),7.60(s,1H),7.23(d,J=2.1Hz,1H),6.79(d,J=8.4Hz,1H),6.75(d,J=8.9Hz,1H),4.17(m,2H),3.84(s,3H),3.71(m,2H),3.41(d,J=10.9Hz,1H),3.33(d,J=9.5Hz,3H),2.99(s,1H),2.94-2.80(m,2H),2.08(s,1H),1.85(t,J=8.0Hz,2H),1.40(t,J=6.9Hz,3H)。
实施例51:化合物161的制备
(2-氯-6-氟苯基)(6-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚基-3-基)亚甲基
Figure PCTCN2022073467-appb-000166
反应瓶中依次加入4-(6-(3,6-二氮杂双环[3.1.1]庚烷-6-基)吡啶-3-基)-6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶(120mg,0.18mmol),DMF(2mL),三乙胺(90.5mg,0.9mmol),降温至5℃,滴加入2-氟-5-氯苯甲酰氯(41.5mg,1.25mmol),5℃下搅拌2小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品50mg,m/z=532[M+1] +1H NMR(400MHz,DMSO-d6)δ12.66(s,1H),8.55-8.60(m,2H),8.07(s,1H),7.23-7.51(m,5H),6.86-6.88(m,1H),4.58(s,1H),4.42(s,1H),4.16-4.26(m,3H),3.51-3.82(m,2H),2.77-2.80(m,1H),1.68-1.71(d,1H),1.40-1.42(m,3H).
实施例52:化合物164的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4,7-二氮杂螺环[2.5]辛烷-4-羧酸叔丁酯
Figure PCTCN2022073467-appb-000167
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(700mg,2.4mmol),4,7-二氮杂螺环[2.5]辛烷-4-羧酸叔丁酯(500mg,2.4mmol),DIEA(1.5g,11.6mmol),DMSO(10mL)加入反应瓶,90℃反应12h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品670mg,m/z=491[M+1] +
步骤B:4-(6-(4,7-二氮杂螺环[2.5]辛烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000168
7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4,7-二氮杂螺环[2.5]辛烷-4-羧酸叔丁酯(670mg,1.4mmol)加入HCl的甲醇溶液(4M,20mL),25℃反应4h,旋干溶剂得产品680mg,m/z=391[M+1] +
步骤C:N-(7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4,7-二氮杂螺环[2.5]辛烷-4-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000169
将4-(6-(4,7-二氮杂螺环[2.5]辛烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(200mg,0.4mmol),2氯-6-氟苯甲酰氯(77mg,0.4mmol),DIEA(258mg,2mmol),DCM(10mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品100mg,m/z=546[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.58(s,1H),8.52(s,1H),8.06(d,J=8.8Hz,1H),7.50-7.59(m,2H),7.33-7.48(m,2H),7.26(s,1H),7.05(t,J=11.2Hz,1H),4.05-4.28(m,3H),3.48-3.88(m,4H),3.38-3.46(m,1H),1.38(t,J=6.8Hz,3H),1.02-1.28(m,2H),0.76-0.82(m,1H),0.68-0.75(m,1H).
实施例53:化合物165的制备
6-乙氧基-4-(6-(4-((6-甲氧基吡啶-3-基)亚甲基)-4,7-二氮杂螺环[2.5]辛烷-7-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000170
将4-(6-(4,7-二氮杂螺环[2.5]辛烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(200mg,0.4mmol),6-甲氧基-3-吡啶甲醛(165mg,1.2mmol),Et 3N(202mg,2.0mmol),Pic-BH 3(128mg,1.2mmol),正丙醇(10mL)加入反应瓶,40℃反应48h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品30.8mg,m/z=511[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.57(s,1H),8.51(s,1H),8.09(s,1H),8.02(d,J=8.8Hz,1H),7.62(d,J=8.4Hz,1H),7.57(s,1H),7.25(s,1H),7.02(d,J=8.8Hz,1H),6.78(d,J=8.4Hz,1H),4.15(q,J=6.8Hz,2H),3.86(s,2H),3.84(s,3H),3.62-68(m,2H),3.56(s,2H),2.78-2.86(m,2H),1.38(t,J=7.2Hz,3H),0.60-0.71(m,4H).
实施例54:化合物169的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷-9-羧酸叔丁酯
Figure PCTCN2022073467-appb-000171
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(120mg,0.4mmol)叔丁基3-氧杂-7,9-氮杂双环[3.3.1]壬烷-9-羧酸酯(100mg,0.44mmol),DIEA(155mg,1.2mmol),DMSO(2mL)加入反应瓶,90℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品150mg,m/z=506[M+1] +
步骤B:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷盐酸盐
Figure PCTCN2022073467-appb-000172
将7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷-9-羧酸叔丁酯(150mg,0.3mmol)加入HCl的甲醇溶液(4M,5mL),25℃反应4h,旋干溶剂得产品160mg,m/z=406[M+1] +
步骤C:N-(7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷-9-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000173
将7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷(60mg;0.1mmol),2-氯-6-氟苯甲酰氯(23mg,0.1mmol),Et 3N(50mg,0.5mmol),DMF(2mL)加入反应瓶,25℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品15mg,m/z=562[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.60(d,J=2.5Hz,1H),8.53(d,J=2.1Hz,1H),8.08(dd,J=8.9,2.6Hz,1H),7.60(s,1H),7.53-7.64(m,1H),7.50(d,J=8.3Hz,1H),7.43(t,J=8.6Hz,1H),7.27(d,J=2.0Hz,1H),7.02(d,J=9.0Hz,1H),4.75(s,1H),4.69(d,J=11.5Hz,1H),4.53(t,J=12.6Hz,1H),4.18(q,J=7.0Hz,2H),4.11(d,J=11.5Hz,1H),3.94(dd,J=11.1,5.5Hz,1H),3.76(s,1H),3.62(d,J=13.8Hz,1H),3.32(d,J=11.8Hz,2H),3.15(d,J=12.8Hz,1H),1.40(t,J=6.9Hz,3H).
实施例55:化合物114的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-9-((6-甲氧基吡啶-3-基)亚甲基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷
Figure PCTCN2022073467-appb-000174
将7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氧杂-7,9-氮杂双环[3.3.1]壬烷(100mg,0.2mmol),6-甲氧基-3-吡啶甲醛(83mg,0.6mmol),Pic-BH 3(64mg,0.6mmol),Et 3N(120mg,1.2mmol),正丙醇(5mL)加入反应瓶,45℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品6.5mg,m/z=527[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.61(d,J=2.6Hz,1H),8.52(d,J=2.0Hz,1H),8.18(d,J=2.3Hz,1H),8.06(dd,J=8.9,2.6Hz,1H),7.77(dd,J=8.5,2.4Hz,1H),7.60(s,1H),7.26(d,J=2.1Hz,1H),6.95(d,J=9.0Hz,1H),6.83(d,J=8.5Hz,1H),4.19(q,J=6.9Hz,2H),4.05(d,J=13.1Hz,2H),3.93(s,2H),3.83(d,J=14.8Hz,7H),3.47-3.57(m,2H),2.83(d,J=3.3Hz,2H),1.41(t,J=6.9Hz,3H).
实施例56:化合物149的制备
6-乙氧基-4-(6-(6-(4-(甲磺酰基)苯亚甲基)-3,6-二氮杂二环[3.1.1]庚烷-3-基)-吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000175
将4-(6-(3,6-二氮杂二环[3.1.1]庚烷-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(300mg,0.8mmol),6-甲氧基-3-吡啶甲醛(165mg,1.2mmol),Et 3N(202mg,2.0mmol),Pic-BH 3(128mg,1.2mmol),DMSO(10mL)加入反应瓶,45℃反应48h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品71.4mg,m/z=544[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.65(s,1H),8.52(s,1H),8.11(d,J=8.8Hz,1H),7.87(d,J=8.0Hz,1H),7.58-7.70(m,3H),7.27(s,1H),6.91(d,J=8.8Hz,1H),4.16-4.21(m,2H),3.51-3.83(m,8H),3.20(s,3H),2.58-2.65(m,1H),1.57-1.65(m,1H),1.39-1.42(m,3H).
实施例57:化合物170的制备
2-氯-N-(((1R,5S,6S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己基-6-基)-6-氟苯甲酰胺
步骤A:(1R,5S,6S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]-6-己基氨基盐酸盐
Figure PCTCN2022073467-appb-000176
向反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(311mg,1.05mmol),DMSO(6mL),DIEA(407mg,3.15mmol),((1R,5S,6S)-3-氮杂双环[3.1.0]-6-己基氨基碳酸酯(250mg,1.26mmol),加热至90℃反应24小时,停止反应,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,再加入甲醇(2mL)和1,4-二氧六环/氯化氢(4M,1mL),室温搅拌1小时,直接浓缩得到产品0.4g,m/z=376[M+1] +
步骤B:2-氯-N-((1R,5S,6S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己基-6-基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000177
反应瓶中加入(1R,5S,6S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]-6-己基氨基盐酸盐(200mg,0.41mmol),2mL DMF,三乙胺(208mg,2.06mmol)降温至5℃,将2-氟-5-氯苯甲酰氯缓慢滴加入反应液中,滴完室温搅拌1小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品12mg。 m/z=532[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.94-8.95(m,1H),8.46-8.56(m,2H),8.01-8.04(m,1H),7.52-7.58(m,1H),7.47-7.51(m,1H),7.38-7.40(m,1H),7.29-7.34(m,1H),7.23-7.24(m,1H),6.73-6.75(m,1H),4.15-4.20(m,2H),3.86-3.96(m,2H),3.55-3.58(d,2H),2.66-2.67(m,1H),1.95(s,1H),1.37-1.41(m,3H)。
实施例58:化合物171的制备
(1R,5S,6S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-N-(((6-甲氧基吡啶-3-基)亚甲基)-3-氮杂双环[3.1.0]-6-己基氨基
Figure PCTCN2022073467-appb-000178
反应瓶中加入(1R,5S,6S)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]-6-己基氨基盐酸盐(200mg,0.41mmol),DMSO(4mL),三乙胺(208mg,2.06mmol),6-甲氧基-3-吡啶甲醛(170mg,1.24mmol),然后滴加2-甲基吡啶硼烷(132.4mg,1.24mmol),室温反应1小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品3mg,m/z=497[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.63(s,1H),8.50-8.56(m,2H),7.98-8.08(m,2H),7.57-7.65(m,2H),7.20-7.23(m,1H),6.81-6.87(m,1H),6.61-6.64(m,1H),4.14-4.19(m,2H),3.85-3.90(m,5H),3.60-3.63(m,5H),1.52-1.55(m,2H),1.39-1.46(m,3H),1.20(s,1H)。
实施例59:化合物172的制备
2-氯-N-(((1R,5S,6R)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己基-6-基)-6-氟苯甲酰胺
步骤A:(1R,5S,6R)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]-6-己基氨基盐酸盐
Figure PCTCN2022073467-appb-000179
向反应瓶中加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(311mg,1.05mmol),DMSO(2mL),DIEA(407mg,3.15mmol),((1R,5S,6R)-3-氮杂双环[3.1.0]-6-己基氨基碳酸酯(250mg,1.26mmol),升温至90℃反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,剩余物加入甲醇(2mL)和1,4-二氧六环/氯化氢(4M,2mL),室温搅拌2小时,减压浓缩到产品0.4g直接用于下步反应,m/z=376[M+1] +
步骤B:2-氯-N-((1R,5S,6R)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己基-6-基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000180
反应瓶中加入(1R,5S,6R)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]-6-己基氨基盐酸盐(200mg,0.41mmol),2mL DMF,三乙胺(208mg,2.06mmol),降温至5℃,将2-氟-5-氯苯甲酰氯缓慢滴加入反应液中,滴完室温搅拌1小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品65mg,m/z=532[M+1] +1H NMR(400MHz,DMSO-d6)δ12.64(s,1H),8.63-8.64(m,1H),8.49-8.49(m,2H),7.95-7.97(m,1H),7.54(s,1H),7.35-7.41(m,1H),7.16-7.23(m,3H),6.58-6.60(m,1H),7.23-7.24(m,1H),6.73-6.75(m,1H),4.14-4.19(m,2H),3.70(s,4H),2.92-2.96(m,1H),2.08-2.13(m,2H),1.35-1.45(m,3H)。
实施例60:化合物173的制备
(1R,5S,6R)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-N-(((6-甲氧基吡啶-3-基)亚甲基)-3-氮杂双环[3.1.0]-6-己基氨基
Figure PCTCN2022073467-appb-000181
反应瓶中加入(1R,5S,6R)-3-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]-6-己基氨基盐酸盐(200mg,0.41mmol),正丙醇(4mL),三乙胺(208mg,2.06mmol),6-甲氧基-3-吡啶甲醛(170mg,1.24mmol),2-甲基吡啶硼烷(132.4mg,1.24mol),40℃反应20小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品4.8mg,m/z=497[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.63(s,1H),8.48-8.53(s,2H),7.97-8.01(m,2H),7.51-7.58(m,2H),7.21-7.21(m,1H),6.55-6.63(m,2H),4.14-4.20(m,2H),3.75(s,3H),3.65-3.72(m,4H),3.33-3.35(m,3H),2.22-2.25(m,1H),1.83-1.84(m,2H),1.38-1.41(t,3H)
实施例61:化合物174的制备
2-氯-N-(((3aR,5S,6aS)-2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-八氢环戊基[c]吡咯基5-基)-6-氟苯甲酰胺
步骤A:2-氯-6-氟-N-(((3aR,5S,6aS)-八氢环戊[c]吡咯-5-基)苯甲酰胺盐酸盐
Figure PCTCN2022073467-appb-000182
向反应瓶中加入(3aR,5S,6aS)-5-氨基六氢环戊基[c]吡咯-2(1H)-碳酸叔丁酯盐酸盐(120mg,0.53mmol),二氯甲烷(2mL),三乙胺(107mg,1.06mmol),6-氟-2-氯苯甲酰氯(113mg,0.58mmol),室温搅拌1小时。倒入水,DCM萃取两次,浓缩至不出后,剩余物加入甲醇(1mL)和1,4-二氧六环/氯化氢(4M,2mL)室温搅拌2小时,减压浓缩得到产品140mg直接用于下步反应,m/z=283[M+1] +
步骤B:2-氯-N-(((3aR,5S,6aS)-2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-八氢环戊基[c]吡咯基5-基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000183
反应瓶中加入2-氯-6-氟-N-(((3aR,5S,6aS)-八氢环戊[c]吡咯-5-基)苯甲酰胺盐酸盐(65mg,0.2mmol),2mL DMSO,DIEA(65.7mg,0.51mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(65mg,0.2mmol),加热至90℃反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品30mg,m/z=560[M+1] +1H NMR(400MHz,DMSO-d6)δ12.64(s,1H),8.80-8.82(m,1H),8.50-8.55(m,2H),7.80-8.02(m,1H),7.57(s,1H),7.43-7.48(m,1H),7.35-7.37(m,1H),7.26-7.31(m,1H),7.21-7.22(m,1H),6.72-6.74(m,1H),4.29-4.35(m,1H),4.14-4.19(m,2H),3.59-3.63(m,2H),3.49-3.52(m,2H),2.78-2.80(m,2H),2.32-2.38(m,2H),1.37-1.50(m,5H)。
实施例62:化合物176的制备
2-氯-N-(((3aR,5R,6aS)-2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-八氢环戊基[c]吡咯基5-基)-6-氟苯甲酰胺
步骤A:2-氯-6-氟-N-(((3aR,5R,6aS)-八氢环戊[c]吡咯-5-基)苯甲酰胺盐酸盐
Figure PCTCN2022073467-appb-000184
向反应瓶中加入(3aR,5R,6aS)-5-氨基六氢环戊基[c]吡咯-2(1H)-碳酸叔丁酯盐酸盐(120mg,0.53mmol),二氯甲烷(2mL),三乙胺(107mg,1.06mmol),6-氟-2-氯苯甲酰氯(113mg,0.58mmol),室温搅拌1小时。倒入水,DCM萃取两次,浓缩至不出后,剩余物加入甲醇(1mL)和1,4-二氧六环/氯化氢(4M,2mL)室温搅拌2小时,减压浓缩得到产品170mg直接用于下步反应,m/z=283[M+1] +
步骤B:2-氯-N-(((3aR,5R,6aS)-2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-八氢环戊基[c]吡咯基5-基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000185
反应瓶中加入2-氯-6-氟-N-(((3aR,5R,6aS)-八氢环戊[c]吡咯-5-基)苯甲酰胺盐酸盐(74mg,0.23mmol),2mL DMSO,DIEA(74.8mg,0.58mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(57.4mg,0.19mmol),加热至90℃反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品17mg,m/z=560[M+1] +1H NMR(400MHz,DMSO-d6)δ12.64(s,1H),8.78-8.80(m,1H),8.50-8.59(m,2H),8.00-8.03(m,1H),7.60(s,1H),7.45-7.50(m,1H),7.37-7.38(m,1H),7.28-7.32(m,1H),7.22-7.23(m,1H),6.72-6.75(m,1H),4.37-4.45(m,1H),4.14-4.25(m,2H),3.63-3.67(m,2H),3.40-3.43(m,2H),2.91-2.95(m,2H),1.86-2.01(m,4H),1.38-1.41(m,3H).
实施例63:化合物180的制备
(2-氯-6-氟苯基)((3aR,7aS)-2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)八氢-1H-4,7-表亚氨基异吲哚-8-基)甲酮
步骤A:(2-氯-6-氟苯基)((3aR,7aS)-八氢-1H-4,7-表亚氨基异吲哚-8-基)甲酮盐酸盐
Figure PCTCN2022073467-appb-000186
向反应瓶中加入(3aR,7aS)-八氢-2H-4,7-表亚氨基异吲哚-2-碳酸叔丁酯(100mg,0.35mmol),二氯甲烷(2mL),三乙胺(70mg,0.69mmol),6-氟-2-氯苯甲酰氯(80.3mg,0.42mmol),室温搅拌1小时。倒入水,DCM萃取两次,浓缩至不出后,加入甲醇(1mL)和1,4-二氧六环/氯化氢(4M,2mL),室温搅拌2小时,减压浓缩得到产品50mg直接用于下步反应,m/z=295[M+1] +
步骤B:(2-氯-6-氟苯基)((3aR,7aS)-2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)八氢-1H-4,7-表亚氨基异吲哚-8-基)甲酮
Figure PCTCN2022073467-appb-000187
反应瓶中加入(2-氯-6-氟苯基)((3aR,7aS)-八氢-1H-4,7-表亚氨基异吲哚-8-基)甲酮盐酸盐(50mg,0.15mmol),2mL DMSO,K 2CO 3(69.5mg,0.5mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(37.4mg,0.13mmol),加热至90℃反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品5mg,m/z=572[M+1] +, 1H NMR(400MHz,DMSO-d6)δ12.64(s,1H),8.51-8.57(d,2H),8.04-8.07(m,1H),7.37-7.58(m,5H),7.25(s,1H),6.83-6.85(m,1H),4.73(s,1H),4.15-4.20(m,2H),3.85-3.88(m,1H),3.75-3.78(m,2H),3.03-3.18(m,5H),3.90(s,1H),1.59-1.62(m,4H),1.34-1.41(t,3H),2.78-2.80(m,2H),2.32-2.38(m,2H),1.37-1.50(m,5H)。
实施例64:化合物128的制备
6-乙氧基-4-(6-(((3aR,7aS)-8-(((6-甲氧基吡啶-3-基)甲基)八氢-2H-4,7-表氨基异吲哚-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
步骤A:(3aR,7aS)-8-(((6-甲氧基吡啶-3-基)甲基)八氢-1H-4,7-表氨基异吲哚盐酸盐
Figure PCTCN2022073467-appb-000188
向反应瓶中加入(3aR,7aS)-八氢-2H-4,7-表氨基异吲哚-2-碳酸叔丁酯(100mg,0.35mmol),二氯甲烷(2mL),三乙胺(70mg,0.69mmol),5-(氯甲基)-2-甲氧基吡啶(80.3mg,0.42mmol),50℃反应16小时。倒入水,DCM萃取两次,浓缩至不出后,加入甲醇(1mL)和1,4-二氧六环/氯化氢(4M,2mL)室温搅拌2小时,减压浓缩得到产品30mg直接用于下步反应,m/z=260[M+1] +
步骤B:6-乙氧基-4-(6-(((3aR,7aS)-8-(((6-甲氧基吡啶-3-基)甲基)八氢-2H-4,7-表氨基异吲哚-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000189
反应瓶中加入(3aR,7aS)-8-(((6-甲氧基吡啶-3-基)甲基)八氢-1H-4,7-表氨基异吲哚盐酸盐(30mg,0.10mmol),2mL DMSO,K 2CO 3(69.5mg,0.5mmol),6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(30mg,0.1mmol),加热升温至90℃反应24小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品4mg,m/z=537[M+1] +, 1H NMR(400MHz,DMSO-d6)δ12.65(s,1H),8.51-8.58(m,2H),8.04-8.14(m,2H),7.76(s,1H),7.59(s,1H),7.26(d,1H),6.83-6.85(d,2H),4.15-4.20(m,2H),3.80-3.85(m,5H),3.09-3.18(m,2H),2.89-2.93(m,2H),1.40-1.46(m,4H),1.21-1.29(m,5H),1.18-1.98(m,1H),1.09-1.10(m,1H).
实施例65:化合物183的制备
步骤A:3-苯基-6-乙基-(1R,5S,6r)-3-氮杂双环[3.1.0]己烷-3,6-羧酸酯
Figure PCTCN2022073467-appb-000190
反应瓶中加入2,5-二氢-1H-吡咯-1-碳酸苯酯(5.0g,24.6mmol),Ph 2(OAc) 4(500mg,1.1mmol)和50mL DCE,氮气保护,加热到80℃,将2-重氮乙酸乙酯(14g,123mmol)溶于50mL DCE,4h滴加至反应液,加完后80℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品3.59g,m/z=290[M+1] +
步骤B:(1R,5S,6r)-3-氮杂双环[3.1.0]己烷-6-碳酸乙酯
Figure PCTCN2022073467-appb-000191
反应瓶中加入3-苯基-6-乙基(1R,5S,6r)-3-氮杂双环[3.1.0]己烷-3,6-羧酸酯(3.5g,12mmol),无水甲醇(30mL),钯碳(600mg),氢气置换两次后,25℃反应18h,反应完全,加硅藻土过滤,减压浓缩得到(1R,5S,6R)-3-氮杂双环[3.1.0]己烷-6-碳酸乙酯粗品2.0g直接用于下一步反应,m/z=156[M+1] +
步骤C:(1R,5S,6r)-3-(5-溴吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-碳酸乙酯
Figure PCTCN2022073467-appb-000192
反应瓶加入5-溴-2-氟吡啶(1.1g,6.4mmol),(1R,5S,6r)-3-氮杂双环[3.1.0]己烷-6-碳酸乙酯(2.0g),碳酸钾(2.6g,19mmol)和10mL DMSO,110℃反应3h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品890mg,m/z=311[M+1] +
步骤D:((1R,5S,6r)-3-(5-溴吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲醇
Figure PCTCN2022073467-appb-000193
应瓶加入(1R,5S,6r)-3-(5-溴吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-碳酸乙酯(750mg,2.4mmol),THF(20mL),氮气保护,加入LiBH 4(209mg,9.6mmol),50℃反应5h,补加LiBH 4(156mg,7.2mmol),50℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品340mg,m/z=269[M+1] +
步骤E:叔丁基(((1R,5S,6r)-3-(5-溴吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲基)(叔丁氧羰基)氨基甲酸酯
Figure PCTCN2022073467-appb-000194
应瓶加入((1R,5S,6r)-3-(5-溴吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲醇(340mg,1.2mmol),HN(Boc) 2(412mg,1.9mmol),三苯基膦(660mg,2.5mmol),THF(15mL),氮气保护,然后将偶氮二甲酸二乙酯(438mg,2.5mmol)滴加至反应液,50℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品505mg,m/z=468[M+1] +
步骤F:叔丁基(((1R,5S,6r)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲基)(叔丁氧羰基)氨基甲酸酯
Figure PCTCN2022073467-appb-000195
应瓶加入6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(360mg,1.1mmol),(((1R,5S,6s)-3-(5-溴吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲基)(叔丁氧羰基)氨基甲酸酯(500mg,1.1mmol),碳酸铯(1.0g,3.3mmol),1,4-二氧六环(10mL),水(2mL),氮气置换,加入Pd(dppf)Cl 2(73.1mg,0.1mmol),50℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品330mg,m/z=589[M+1] +
步骤G:((1R,5S,6s)-3-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲胺盐酸盐
Figure PCTCN2022073467-appb-000196
反应瓶加入叔丁基(((1R,5S,6r)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲基)(叔丁氧羰基)氨基甲酸酯(330mg,0.56mmol),4N盐酸-二氧六环溶液(5mL,20mmol),甲醇(1mL),25℃反应16h,反应完全,减压浓缩得产品300mg直接用于下步反应,m/z=389[M+1] +
步骤H:2-氯-N-(((1R,5S,6s)-3-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲基)-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000197
反应瓶中加入((1R,5S,6s)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲胺盐酸盐(120mg,0.3mmol),3-氯-6-氟苯甲酰氯(59mg,0.3mmol),DIEA(232mg,1.8mmol),3mL DMF,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品11.5mg,m/z=546[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.63(brs,1H),8.85-8.88(m,1H),8.50-8.53(m,2H),8.00(d,J=8.8Hz,1H),7.44-7.59(m,2H),7.29-7.42(m,2H),7.21(s,1H),6.68(d,J=8.8Hz,1H),4.14(q,J=6.8Hz,1H),3.74-3.76(m,2H),3.47-3.50(m,2H),3.25-3.28(m,2H),1.75-1.79(m,2H),1.39(t,J=6.8Hz,3H),0.88-0.94(m,1H).
实施例66:化合物184的制备
步骤A:N-(((1R,5S,6s)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲基)-1-(6-甲氧基吡啶-3-基)-N-((6-甲氧基吡啶-3-基)甲基)甲胺
Figure PCTCN2022073467-appb-000198
反应瓶中加入((1R,5S,6s)-3-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-氮杂双环[3.1.0]己烷-6-基)甲胺盐酸盐(170mg,0.44mmol),6-甲氧基-3-吡啶甲醛(178mg,1.3mmol),TEA(212mg,2.1mmol),2-甲基吡啶硼烷(139mg,1.3mmol),和正丙醇10mL,45℃反应18h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品6.4mg,m/z=511[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.66(brs,1H),8.50-8.55(m,2H),8.10(s,1H),8.00(d,J=8.4Hz,1H),7.67-7.71(m,2H),7.57(s,1H),7.21(s,1H),6.61(d,J=8.8Hz,1H),4.14-4.18(m,2H),3.74-3.76(m,2H),3.52-3.61(m,3H),3.37-3.42(m,2H),2.32-2.45(m,2H),1.41-1.48(m,2H),1.39(t,J=6.8Hz,3H),0.80-0.88(m,1H).
实施例67:化合物187的制备
步骤A:叔丁基5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,5-二氮杂螺环[3.4]辛烷-5-羧酸酯
Figure PCTCN2022073467-appb-000199
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(318mg,1.1mmol),叔丁基2,6-二氮杂螺环[3.4]辛烷-6-羧酸酯(254mg,1.2mmol),DIEA(414mg,3.2mmol),DMSO(5mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓 缩,柱层析分离得到产品511mg,m/z=490[M+1] +
步骤B:4-(6-(2,5-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000200
将叔丁基5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-6-羧酸酯(511mg,1.04mmol)加入5mL MeOH/HCl(4M),25℃反应4h减压浓缩得产品500mg直接用于下步反应,m/z=390[M+1] +
步骤C:(2-氯-6-氟苯基)-5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,5-二氮杂螺环[3.4]辛烷-5-基)甲酮
Figure PCTCN2022073467-appb-000201
将4-(6-(2,6-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(500mg,1.18mmol),2-氯-6-氟苯甲酰氯(250mg,1.29mmol),Et 3N(358mg,3.54mmol),DMF(5mL)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品398mg,m/z=546[M+1] +, 1H NMR(400MHz,DMSO-d 6):δ12.65(s,1H),8.57(d,J=2.4Hz,1H),8.52(d,J=2.1Hz,1H),8.06(m,1H),7.59(s,1H),7.53(m,1H),7.49-7.34(m,2H),7.27(d,J=2.1Hz,1H),6.69(d,J=8.6Hz,1H),4.86(t,J=8.4Hz,2H),4.18(q,J=6.9Hz,2H),4.05(dd,J=9.6,7.8Hz,2H),3.23(td,J=6.6,1.9Hz,2H),2.42(t,J=6.8Hz,2H),1.91-1.78(m,2H),1.40(t,J=6.9Hz,3H)。
实施例68:化合物188的制备
6-乙氧基-4-(6-(5-((6-甲氧基吡啶-3-基)甲基)-2,5-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000202
将4-(6-(2,5-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(500mg;1.18mmol),6-甲氧基-3-吡啶甲醛(176mg,1.29mmol),Et 3N(357.5mg,3.54mmol),Pic-BH 3(378.8mg,3.54mmol),正丙醇(5mL)加入反应瓶,25℃反应12h,加水搅 拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品346mg,m/z=511[M+1] +1H NMR(400MHz,DMSO-d 6)12.65(s,1H),8.54(dd,J=16.4,2.2Hz,2H),8.13-8.00(m,2H),7.69(dd,J=8.5,2.4Hz,1H),7.59(s,1H),7.24(d,J=2.1Hz,1H),6.79(d,J=8.4Hz,1H),6.66(d,J=8.6Hz,1H),4.29(d,J=8.9Hz,2H),4.18(m,2H),3.95(d,J=8.9Hz,2H),3.84(s,3H),3.79(s,2H),2.56(t,J=7.1Hz,2H),2.14(dd,J=9.1,6.4Hz,2H),1.71(t,J=7.7Hz,2H),1.40(t,J=6.9Hz,3H)。
实施例69:化合物189的制备
步骤A:叔丁基5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-6-羧酸酯
Figure PCTCN2022073467-appb-000203
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(318mg,1.1mmol),叔丁基2,6-二氮杂螺环[3.4]辛烷-6-羧酸酯(254.4mg,1.2mmol),DIEA(414mg,3.2mmol),DMSO(5mL)加入反应瓶,90℃反应12h加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品511mg,m/z=490[M+1] +
步骤B:4-(6-(2,6-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000204
将叔丁基5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-6-羧酸酯(511mg,1.04mmol)加入5mL MeOH/HCl(4M),25℃反应4h,减压浓缩得产品500mg直接用于下步反应,m/z=390[M+1] +
步骤C:(2-氯-6-氟苯基)-5-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-6-基)甲酮
Figure PCTCN2022073467-appb-000205
将4-(6-(2,6-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(500mg,1.18mmol),2-氯-6-氟苯甲酰氯(250mg,1.29mmol),Et 3N(357.5mg,3.54mmol),DMF(5mL)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品398mg,m/z=546[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.55(dd,J=12.5,2.5Hz,1H),8.52(m,1H),8.03(m,1H),7.61-7.33 (m,4H),7.28-7.20(m,1H),6.64(dd,J=21.1,8.7Hz,1H),4.18(m,2H),4.13-3.91(m,4H),3.89-3.73(m,1H),3.71-3.54(m,1H),3.43(s,1H),3.25(t,J=6.9Hz,1H),2.25(m,2H),1.40(m,3H)。
实施例70:化合物190的制备
6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-2,6-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000206
将4-(6-(2,6-二氮杂螺环[3.4]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(500mg,1.18mmol),6-甲氧基-3-吡啶甲醛(176mg,1.29mmol),Et 3N(357.5mg,3.54mmol),Pic-BH 3(378.8mg,3.54mmol),正丙醇(5mL)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品346mg,m/z=511[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.52(dd,J=8.4,2.3Hz,2H),8.11(s,1H),8.01(dd,J=8.7,2.5Hz,1H),7.69(s,1H),7.56(s,1H),7.22(d,J=2.1Hz,1H),6.81(d,J=8.4Hz,1H),6.59(d,J=8.6Hz,1H),4.17(m,2H),3.97(t,J=6.1Hz,4H),3.84(s,3H),3.06(d,J=7.1Hz,1H),2.77(s,2H),2.58(s,2H),2.12(s,2H),1.39(t,J=6.9Hz,3H),1.19(t,J=7.3Hz,1H)。
实施例71:化合物191的制备
步骤A:2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[3.5]壬烷-7-羧酸叔丁酯
Figure PCTCN2022073467-appb-000207
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(250mg,0.8mmol),叔丁基2,7-二氮杂螺环[3.5]壬烷-7-羧酸酯(230mg,0.9mmol),DIEA(520mg,4mmol),DMSO(2mL)加入反应瓶,90℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品390mg,m/z=504[M+1] +
步骤B:4-(6-(2,7-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000208
将叔丁基2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[3.5]壬烷-7-羧酸酯(390mg,0.7mmol)加入HCl的甲醇溶液(4M,5ml),25℃反应4h, 减压浓缩得产品370mg直接用于下步反应,m/z=404[M+1] +
步骤C:N-(2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[3.5]壬烷-7-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000209
将4-(6-(2,7-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.2mmol),2-氯-6-氟苯甲酰氯(46mg,0.2mmol),Et 3N(100mg,1.0mmol),DMF(2mL)加入反应瓶,25℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品47mg,m/z=560[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.53(dd,J=10.4,2.3Hz,2H),8.03(dd,J=8.6,2.5Hz,1H),7.48-7.59(m,2H),7.33-7.49(m,2H),7.23(d,J=2.1Hz,1H),6.60(d,J=8.7Hz,1H),4.18(q,J=7.0Hz,2H),3.80-3.91(m,4H),3.73(tq,J=13.6,7.4,6.9Hz,2H),3.15-3.26(m,2H),1.71-1.91(m,4H),1.40(t,J=6.9Hz,3H).
实施例72:化合物192的制备
6-乙氧基-4-(6-(7-((6-甲氧基吡啶-3-基)亚甲基)-2,7-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000210
将4-(6-(2,7-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.2mmol),6-甲氧基烟醛(83mg,0.6mmol),Pic-BH 3(64mg,0.6mmol),Et 3N(120mg,1.2mmol),正丙醇(5mL)加入反应瓶,45℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品37mg,m/z=525[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.52(dd,J=7.4,2.3Hz,2H),7.97-8.09(m,2H),7.65(dd,J=8.4,2.4Hz,1H),7.56(s,1H),7.23(d,J=2.1Hz,1H),6.80(d,J=8.4Hz,1H),6.59(d,J=8.7Hz,1H),4.17(q,J=6.9Hz,2H),3.85(s,3H),3.76(s,4H),3.42(s,2H),2.35(s,4H),1.78(s,3H),1.78(d,J=11.2Hz,1H),1.40(t,J=6.9Hz,3H).
实施例73:化合物193的制备
步骤A:2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.5]壬烷-6-羧酸叔丁酯
Figure PCTCN2022073467-appb-000211
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(250mg,0.8mmol),叔丁 基2,6-二氮杂螺环[3.5]壬烷-6-羧酸酯(230mg,0.9mmol),DIEA(520mg,4mmol),DMSO(2mL)加入反应瓶,90℃反应16h加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品370mg,m/z=504[M+1] +
步骤B:4-(6-(2,6-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000212
将叔丁基2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.5]壬烷-6-羧酸酯(370mg,0.7mmol)加入HCl的甲醇溶液(4M,5mL),25℃反应4h,减压浓缩得产品370mg直接用于下步反应,m/z=404[M+1] +
步骤C:N-(2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.5]壬烷-6-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000213
将4-(6-(2,6-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg;0.2mmol),2-氯-6-氟苯甲酰氯(46mg,0.2mmol),Et 3N(100mg,1.0mmol),DMF(2mL)加入反应瓶,25℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品36mg,m/z=560[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.59-8.48(m,2H),8.03(dd,J=21.8,8.6,2.5Hz,1H),7.41-7.62(m,3H),7.34-7.43(m,1H),7.23(dd,J=17.9,2.1Hz,1H),6.68(d,J=8.7Hz,1H),4.12-4.23(m,2H),4.08(d,J=12.8Hz,1H),3.90(dd,J=11.8,8.2Hz,1H),3.79(dq,J=8.0,4.1,2.8Hz,3H),3.70(s,1H),3.44(s,1H),3.13-3.22(m,1H),1.84-1.98(m,2H),1.59(d,J=28.4Hz,4H),1.40(td,J=7.0,3.5Hz,3H).
实施例74:化合物194的制备
6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-2,6-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000214
将4-(6-(2,6-二氮杂螺环[3.5]壬烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg;0.2mmol),6-甲氧基-3-吡啶甲醛(83mg,0.6mmol),Pic-BH 3(64mg,0.6mmol),Et 3N(120mg,1.2mmol),正丙醇(5mL)加入反应瓶,45℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品32mg,m/z=525[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.51(t,J=1.9Hz,2H),8.08(d,J=2.3Hz,1H),7.99(dd,J=8.7,2.5Hz,1H),7.67(dd,J=8.5,2.4Hz,1H),7.56(s,1H),7.22(d,J=2.1Hz,1H),6.81(d,J=8.4Hz,1H),6.57(d,J=8.7Hz,1H),4.17(q,J=6.9Hz,2H),3.83(s,3H),3.70(s, 4H),3.47(s,2H),2.28-2.34(m,4H),1.52-1.69(m,4H),1.40(t,J=6.9Hz,3H).
实施例75:化合物195的制备
步骤A:2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,8-二氮杂螺环[4.5]癸烷-8-羧酸叔丁酯
Figure PCTCN2022073467-appb-000215
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(250mg,0.8mmol),2,8-二氮杂螺环[4.5]癸烷-8-羧酸叔丁酯(240mg,1.0mmol),DIEA(520mg,4mmol),DMSO(2mL)加入反应瓶,90℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品360mg,m/z=518[M+1] +
步骤B:4-(6-(2,8-二氮杂螺环[4.5]癸烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000216
将2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,8-二氮杂螺环[4.5]癸烷-8-羧酸叔丁酯(360mg,0.7mmol)加入HCl的甲醇溶液(4M,5mL),25℃反应4h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品370mg,m/z=418[M+1] +
步骤C:N-(2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,8-二氮杂螺环[4.5]癸烷-8-基)2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000217
将4-(6-(2,8-二氮杂螺环[4.5]癸烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.2mmol),2-氯-6-氟苯甲酰氯(46mg,0.2mmol),Et 3N(100mg,1.0mmol),DMF(2mL)加入反应瓶,25℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品55mg,m/z=574[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.53(dd,J=19.9,2.3Hz,2H),8.01(dd,J=8.8,2.5Hz,1H),7.57(s,1H),7.52(td,J=8.2,6.2Hz,1H),7.45(d,J=8.0Hz,1H),7.37(t,J=8.5Hz,1H),7.22(d,J=2.1Hz,1H),6.70(d,J=8.8Hz,1H),4.18(q,J=7.0Hz,2H),3.78-3.92(m,1H),3.69(dtd,J=13.4,8.7,8.1,3.8Hz,1H),3.57(q,J=6.9Hz,2H),3.40-3.55(m,2H),3.27(q,J=4.9Hz,2H),1.97(dddd,J=15.5,12.3,9.2,5.0Hz,2H),1.46-1.74(m,4H),1.40(t,J=6.9Hz,3H).
实施例76:化合物196的制备
6-乙氧基-4-(6-(8-((6-甲氧基吡啶-3-基)亚甲基)-2,8-二氮杂螺环[4.5]癸烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000218
将4-(6-(2,8-二氮杂螺环[4.5]癸烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(100mg,0.2mmol),6-甲氧基-3-吡啶甲醛(83mg,0.6mmol),Pic-BH 3(64mg,0.6mmol),Et3N(120mg,1.2mmol),正丙醇(5mL)加入反应瓶,45℃反应16h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品15mg,m/z=539[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.52(dd,J=16.8,2.3Hz,2H),8.07(s,1H),8.00(dd,J=8.8,2.5Hz,1H),7.65(dd,J=8.3,2.4Hz,1H),7.58(s,1H),7.22(d,J=2.1Hz,1H),6.80(d,J=8.4Hz,1H),6.67(d,J=8.8Hz,1H),4.17(q,J=6.9Hz,2H),3.84(s,3H),3.53(t,J=6.9Hz,2H),3.45(s,3H),3.33(s,2H),3.27(s,1H),2.51(m,2H),2.21-2.42(m,3H),1.87(t,J=7.0Hz,2H),1.60(s,4H),1.40(t,J=7.0Hz,3H).
实施例77:化合物197的制备
步骤A:2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[4.5]辛烷-7-羧酸叔丁酯
Figure PCTCN2022073467-appb-000219
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(310mg,1.04mmol),2,7-二氮杂螺环[4.5]辛烷-7-羧酸叔丁酯(250mg,1.04mmol),DIEA(430mg,3.12mmol),DMSO(5mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品350mg,m/z=518[M+1] +
步骤B:4-(6-(2,7-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000220
2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[4.5]辛烷-7-羧酸叔丁酯(350mg,0.676mmol)加入,HCl的甲醇溶液(4M,10mL),25℃反应4h,减压浓缩得产品320mg直接用于下步反应,m/z=418[M+1] +
步骤C:N-(2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂 螺环[4.5]辛烷-7-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000221
将4-(6-(2,7-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(160mg,0.3mmol),2-氯-6-氟苯甲酰氯(58mg,0.3mmol),DIEA(194mg,1.5mmol),DCM(5mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品48.3mg,m/z=573[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.63(brs,1H),8.57(s,1H),8.50(s,1H),8.02(d,J=8.8Hz,1H),7.32-7.59(m,4H),7.23(s,1H),6.68(d,J=8.8Hz,1H),4.15-4.20(m,2H),3.45-3.73(m,4H),3.01-3.39(m,4H),1.45-2.13(m,6H),1.37-1.41(m,3H)。
实施例78:化合物198的制备
6-乙氧基-4-(6-(7-((6-甲氧基吡啶-3-基)亚甲基)-2,7-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000222
将4-(6-(2,7-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(160mg,0.3mmol),6-甲氧基-3-吡啶甲醛(124mg,0.9mmol),Et 3N(152mg,1.5mmol),正丙醇(10mL)加入反应瓶,45℃反应48h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品16.0mg,m/z=539[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.52(dd,J=17.8,2.3Hz,2H),7.95-8.07(m,2H),7.62(dd,J=8.4,2.4Hz,1H),7.59(s,1H),7.22(d,J=2.1Hz,1H),6.68(dd,J=27.0,8.6Hz,2H),4.18(q,J=7.0Hz,2H),3.79(s,3H),3.55(d,J=10.4Hz,2H),3.29-3.50(m,3H),3.25(d,J=10.7Hz,1H),2.46(s,1H),2.29(s,2H),2.15(s,1H),1.97(s,1H),1.78(dt,J=12.2,8.0Hz,1H),1.56(d,J=14.6Hz,3H),1.40(m,4H).
实施例79:化合物199的制备
步骤A:2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[4.5]辛烷-6-羧酸叔丁酯
Figure PCTCN2022073467-appb-000223
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(125mg,0.4mmol),2,6-二氮杂螺环[4.5]辛烷-6-羧酸叔丁酯(100mg,0.4mmol),DIEA(1.5g,2.0mmol),DMSO(3mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品200mg,m/z=518[M+1] +
步骤B:4-(6-(2,6-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000224
2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[4.5]辛烷-6-羧酸叔丁酯(200mg,0.39mmol)加入HCl的1,4-二氧六环溶液(4M,9mL),25℃反应4h。减压浓缩得产品170mg直接用于下步反应,m/z=418[M+1] +
步骤C:N-(2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[4.5]辛烷-6-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000225
将4-(6-(2,6-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(85mg,0.16mmol),2-氯-6-氟苯甲酰氯(32mg,0.16mmol),DIEA(125mg,1mmol),DCM(3mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品60mg,m/z=574[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.58(d,J=2.5Hz,1H),8.51(d,J=2.1Hz,1H),8.05(dt,J=8.9,1.7Hz,1H),7.61(s,1H),7.31-7.55(m,3H),7.24(d,J=2.1Hz,1H),6.75(dd,J=8.7,4.7Hz,1H),4.18(q,J=6.9Hz,2H),4.07(dd,J=18.3,7.7Hz,2H),3.71(s,1H),3.54(s,1H),2.83-2.70(m,1H),2.51-2.65(m,1H),1.46-1.93(m,8H),1.41(t,J=6.9Hz,3H).
实施例80:化合物200的制备
6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-2,6-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000226
将4-(6-(2,6-二氮杂螺环[4.5]辛烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(85mg,0.16mmol),6-甲氧基-3-吡啶甲醛(66mg,0.48mmol),Et 3N(81mg,0.8 mmol),Pic-BH 3(52mg,0.48mmol),DMSO(3mL)加入反应瓶,45℃反应72h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品21.3mg,m/z=539[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.56(d,J=2.4Hz,1H),8.51(d,J=2.1Hz,1H),7.99-8.10(m,2H),7.67(dd,J=8.5,2.4Hz,1H),7.58(s,1H),7.23(d,J=2.1Hz,1H),6.76(dd,J=12.8,8.6Hz,2H),4.18(q,J=6.9Hz,2H),3.82(s,3H),3.73(m,2H),3.53(dd,J=14.8,9.0Hz,4H),3.31(s,2H),2.44(s,2H),1.70(d,J=5.9Hz,2H),1.56-1.62(m,2H),1.45(d,J=11.8Hz,2H),1.40(t,J=6.9Hz,3H).
实施例81:化合物201的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-2-基)吡啶-2-基)-1,7-二氮杂螺环[3.5]壬烷-1-羧酸叔丁酯
Figure PCTCN2022073467-appb-000227
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(300mg,1mmol),1,7-二氮杂螺环[3.5]壬烷-1-羧酸叔丁酯(226mg,1mmol),DIEA(645mg,5mmol),DMSO(3mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品497mg,m/z=504[M+1] +
步骤B:4-(6-(1,7-二氮杂螺环[3.5]壬烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000228
7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-2-基)吡啶-2-基)-1,7-二氮杂螺环[3.5]壬烷-1-羧酸叔丁酯(497mg,1mmol)加入HCl的1,4-二氧六环溶液(4M,10mL),25℃反应4h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品470mg,m/z=404[M+1] +
步骤C:N-(7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,7-二氮杂螺环[3.5]壬烷-1-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000229
将4-(6-(1,7-二氮杂螺环[3.5]壬烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(50mg,0.1mmol),2-氯-6-氟苯甲酰氯(19mg,0.1mmol),DIEA(65mg,0.5mmol),DCM(3mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品5.1mg,m/z=560[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.72(t,J=5.7Hz,1H),8.60(dd,J=6.1,2.6Hz,1H),8.52(t,J=2.5Hz,1H),8.11-8.02(m,1H),7.60(s,1H),7.57-7.41(m,1H),7.46-7.24(m,2H),7.02(d,J=9.0Hz,1H), 4.18(q,J=6.9Hz,2H),4.03(s,2H),3.83(t,J=5.6Hz,2H),3.41(q,J=6.6Hz,2H),3.00(t,J=12.6Hz,1H),2.36-2.18(m,4H),1.98(d,J=12.6Hz,1H),1.40(t,J=7.0Hz,3H)。
实施例82:化合物202的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[3.5]壬烷-2-羧酸叔丁酯
Figure PCTCN2022073467-appb-000230
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(600mg,2mmol),2,7-二氮杂螺环[3.5]壬烷-2-羧酸叔丁酯(100mg,2mmol),DIEA(1.3g,10mmol),DMSO(10mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品840mg,m/z=504[M+1] +
步骤B:4-(6-(2,7-二氮杂螺环[3.5]壬烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000231
7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[3.5]壬烷-2-羧酸叔丁酯(840mg,1.67mmol)加入HCl的1,4-二氧六环溶液(4M,20mL),25℃反应4h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品730mg,m/z=404[M+1] +
步骤C:N-(7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[3.5]壬烷-2-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000232
将4-(6-(2,7-二氮杂螺环[3.5]壬烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(50mg,0.1mmol),2-氯-6-氟苯甲酰氯(19mg,0.1mmol),DIEA(65mg,0.5mmol),DCM(3mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品19.2mg,m/z=560[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.57(s,1H),8.51(s,1H),8.03(d,J=8.8Hz,1H),7.57(s,1H),7.51-7.55(m,1H),7.44(d,J=8.0Hz,1H),7.35-7.39(m,1H),7.25(s,1H),7.10(d,J=8.8Hz,1H),4.14-4.20(m,2H),3.89(s,2H),3.50-3.71(m,6H),1.70-1.89(m,4H),1.38-1.43(m,3H).
实施例83:化合物203的制备
6-乙氧基-4-(6-(2-((6-甲氧基吡啶-3-基)亚甲基)-2,7-二氮杂螺环[3.5]辛烷-7-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000233
将4-(6-(2,7-二氮杂螺环[3.5]壬烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(100mg,0.19mmol),6-甲氧基-3-吡啶甲醛(80mg,0.58mmol),Et 3N(98mg,0.97mmol),Pic-BH 3(63mg,0.58mmol),DMSO(3mL)加入反应瓶,45℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品6.1mg,m/z=525[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.56(s,1H),8.50(s,1H),8.06(s,1H),8.02(d,J=8.8Hz,1H),7.62(d,J=8.8Hz,1H),7.56(s,1H),7.24(s,1H),7.06(d,J=8.0Hz,1H),6.78(d,J=8.0Hz,1H),4.14-4.19(m,2H),3.83(s,3H),3.48-3.67(m,6H),2.92-3.09(m,4H),1.65-1.81(m,4H),1.38-1.43(m,3H).
实施例84:化合物206的制备
步骤A:8-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,8-二氮杂螺环[4.5]癸烷-1-羧酸叔丁酯
Figure PCTCN2022073467-appb-000234
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(150mg,0.5mmol),1,8-二氮杂螺环[4.5]癸烷-1-羧酸叔丁酯(120mg,0.5mmol),DIEA(323mg,2.5mmol),DMSO(3mL)加入反应瓶,90℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品181mg,m/z=518[M+1] +
步骤B:4-(6-(1,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000235
8-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,8-二氮杂螺环[4.5]癸烷-1-羧酸叔丁酯(181mg,0.35mmol)加入HCl的1,4-二氧六环溶液(4M,5mL),25℃反应4h,减压浓缩得产品150mg直接用于下步反应,m/z=418[M+1] +
步骤C:N-(8-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,8-二氮杂螺环[4.5]癸烷-1-基)2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000236
将4-(6-(1,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐,2-氯-6-氟苯甲酰氯(18mg,0.1mmol),DIEA(65mg,0.5mmol),DCM(3mL)加入反应瓶,25℃反应1h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品2.6mg,m/z=574[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.60(s,1H),8.52(s,1H),8.04(d,J=8.8Hz,1H),7.60(s,1H),7.55-7.63(m,2H),7.27-7.38(m,2H),7.12(s,J=11.2Hz,1H),4.48-4.56(m,2H),4.17(q,J=6.8Hz,2H),3.16-3.24(m,2H),2.89-3.08(m,4H),2.12-2.20(m,2H),1.80-1.87(m,2H),1.52-1.58(m,2H),1.38(t,J=6.8Hz,3H).
实施例85:化合物207的制备
6-乙氧基-4-(6-(1-((6-甲氧基吡啶-3-基)亚甲基)-1,8-二氮杂螺环[4.5]辛烷-8-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000237
将4-(6-(1,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐(100mg,0.19mmol),6-甲氧基-3-吡啶甲醛(80mg,0.57mmol),Et 3N(98mg,0.8mmol),Pic-BH 3(63mg,0.57mmol),DMSO(3mL)加入反应瓶,45℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品23.8mg,m/z=539[M+1] +, 1H-NMR(400MHz,DMSO-d 6):δ12.65(brs,1H),8.58(s,1H),8.51(s,1H),7.99-8.05(m,2H),7.54-7.63(m,2H),7.25(s,1H),7.09(d,J=8.8Hz,1H),6.75(d,J=8.8Hz,1H),4.46-4.55(m,2H),4.15-4.20(m,2H),3.81(s,3H),3.50(s,2H),2.93-3.00(m,2H),2.55-3.59(m,2H),1.85-1.92(m,2H),1.65-1.84(m,4H),1.40-1.51(m,2H),1.39-1.41(m,3H).
实施例86:化合物208的制备
步骤A:8-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,8-二氮杂螺环[4.5]癸烷-2-碳酸叔丁酯
Figure PCTCN2022073467-appb-000238
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(297mg,1.0mmol),2,8-二氮杂螺环[4.5]癸烷-2-羧酸叔丁酯(240mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(4mL)。升温100℃反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品510mg,m/z=518[M+1] +
步骤B:4-(6-(2,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶盐酸盐
Figure PCTCN2022073467-appb-000239
向反应瓶中依次加入甲醇(5mL),9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,9-二氮杂螺环[5.5]十一烷-3-碳酸叔丁酯(510mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(4.0mL,24.0mmol)。室温反应30min,直接减压浓缩得到产品320mg,m/z=418[M+1] +
步骤C:N-(8-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,8-二氮杂螺环[4.5]癸烷-2-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000240
向反应瓶中依次加入4-(6-(2,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(63mg,0.15mmol),DMF(1mL)和二异丙基乙基胺(58mg,0.45mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(29mg,0.15mmol)的DMF(0.2mL)溶液,室温反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品23mg,m/z=574[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.55(s,1H),8.50(s,1H),8.00-8.04(m,1H),7.35-7.56(m,4H),7.23-7.26(m,1H),7.04-7.08(m,1H),4.16-4.18(m,2H),3.52-3.79(m,6H),3.20-3.26(m,1H),3.06-3.10(m,1H),1.50-1.71(m,4H),1.38-1.43(m,3H).
实施例87:化合物209的制备
步骤A:6-乙氧基-4-(6-(2-((6-甲氧基吡啶-3-基)亚甲基)-2,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000241
向反应瓶中依次加入1,2-二氯乙烷(2mL),甲醇(1mL),4-(6-(2,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(63mg,0.15mmol),6-甲氧基烟醛(21mg,0.15mmol),三乙酰氧基硼氢化钠(96mg,0.45mmol),升温45℃反应1小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品24mg,m/z=539[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.56(s,1H),8.51(s,1H),8.02-8.15(m,1H),8.02(d,J=8.8Hz,1H),7.65-7.78(m,1H),7.57(s,1H),7.24(s,1H),7.06(d,J=8.8Hz,1H),6.76-6.89(m,1H),4.14-4.20(m,2H),3.84(s,3H),3.50-3.75(m,6H),3.30-3.50(m,3H),1.91(s,3H),1.50-1.79(m,6H),1.38-1.41(m,3H).
实施例88:化合物210的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[4.5]癸烷-2-碳酸叔丁酯
Figure PCTCN2022073467-appb-000242
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(150mg,0.5mmol),2,7-二氮杂螺环[4.5]癸烷-2-羧酸叔丁酯半草酸盐(250mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(4mL),升温100℃反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品525mg,m/z=518[M+1] +
步骤B:4-(6-(2,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000243
向反应瓶中依次加入甲醇(5mL),7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[4.5]癸烷-2-碳酸叔丁酯(525mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(4.0mL,24.0mmol)。室温搅拌反应30min,加水搅拌淬灭,用1N NaOH调pH=10后用二氯甲烷萃取,无水硫酸钠干燥后浓缩得产品301mg直接用于下步反应,m/z=418[M+1] +
步骤C:N-(7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,7-二氮杂螺环[4.5]癸烷-2-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000244
向反应瓶中依次加入4-(6-(2,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(63mg,0.15mmol),DMF(1mL)和二异丙基乙基胺(58mg,0.45mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(29mg,0.15mmol)的DMF(0.2mL)溶液,室温搅拌反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品23mg,m/z=574[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.50-8.55(m,2H),7.97-8.04(m,1H),7.30-7.61(m,4H),7.25(s,1H),7.01-7.12(m,1H),4.16-4.18(m,2H),3.33-3.70(m,6H),3.22-3.28(m,1H),3.12-3.19(m,1H),1.52-2.01(m,6H),1.38-1.43(m,3H).
实施例89:化合物211的制备
步骤A:6-乙氧基-4-(6-(2-((6-甲氧基吡啶-3-基)亚甲基)-2,8-二氮杂螺环[4.5]癸烷-8-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000245
向反应瓶中依次加入1,2-二氯乙烷(2mL),甲醇(1mL),4-(6-(2,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(63mg,0.15mmol),6-甲氧基-3-吡啶甲醛(21mg,0.15mmol)和醋酸(2滴),升温45℃反应1小时后加入三乙酰氧基硼氢化钠(96mg,0.45mmol)继续45℃搅拌16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品16mg,m/z=539[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.53(s,1H),8.51(s,1H),8.02-8.15(m,1H),7.98(d,J=8.8Hz,1H),7.62-7.75(m,1H),7.58(s,1H),7.24(s,1H),7.00(d,J=8.8Hz,1H),6.69-6.79(m,1H),4.15-4.20(m,2H),3.76(s,3H),3.34-3.75(m,8H),3.16-3.18(m,1H),1.91(s,1H),1.49-1.68(m,6H),1.38-1.42(m,3H)。
实施例90:化合物212的制备
步骤A:7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,7-二氮杂螺环[4.5]癸烷-1-碳酸叔丁酯
Figure PCTCN2022073467-appb-000246
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(297mg,1.0mmol),1,7-二氮杂螺环[4.5]癸烷-1-羧酸叔丁酯(240mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(4mL)。反应液升温100℃反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品500mg,m/z=518[M+1] +
步骤B:4-(6-(1,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000247
向反应瓶中依次加入甲醇(5mL),7-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,7-二氮杂螺环[4.5]癸烷-1-碳酸叔丁酯(500mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(4.0mL,24.0mmol)。室温搅拌反应30min。浓缩后加10mL水稀释,用1N NaOH调pH=10后用二氯甲烷(30mL)萃取两次,萃取液经饱和食盐水洗涤(10mL),无水硫酸钠干燥后浓缩得产品310mg,m/z=418[M+1] +
步骤C:N-(7-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-1,7-二氮杂螺环[4.5]癸烷-1-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000248
向反应瓶中依次加入4-(6-(1,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(63mg,0.15mmol),DMF(1mL)和二异丙基乙基胺(58mg,0.45mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(29mg,0.15mmol)的DMF(0.2mL)溶液,室温搅拌反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品27mg,m/z=574[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.56(s,1H),8.51(s,1H),8.03(d,J=8.8Hz,1H),7.60(s,1H),7.36-7.52(m,3H),7.26(s,1H),7.12-7.16(m,1H),4.43-4.46(m,2H),4.15-4.18(m,2H),3.86-3.91(m,1H),3.20-3.26(m,2H),3.00-3.10(m,1H),2.85-2.96(m,1H),2.05-2.13(m,1H),1.71-1.92(m,4H),1.61-1.70(m,2H),1.38-1.42(m,3H)。
实施例91:化合物213的制备
6-乙氧基-4-(6-(1-((6-甲氧基吡啶-3-基)亚甲基)-1,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000249
向反应瓶中依次加入4-(6-(1,7-二氮杂螺环[4.5]癸烷-7-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(42mg,0.1mmol),DMF(1mL),碳酸钾(40mg,0.3mmol)和5-(氯甲基)-2-甲氧基吡啶(16mg,0.1mmol),升温60℃搅拌反应25小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品4.1mg,m/z=539[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.55(s,1H),8.50(s,1H),8.07(s,1H),8.01(d,J=8.8Hz,1H),7.66(d,J=8.8Hz,1H),7.59(s,1H),7.25(s,1H),7.07(d,J=8.8Hz,1H),6.78(d,J=8.8Hz,1H),4.39-4.43(m,1H),4.27(d,J=12.0Hz,1H),4.15-4.20(m,2H),3.84(s,3H),3.67-3.79(m,2H),2.93(d,J=12.0Hz,1H),2.58-2.83(m,3H),1.50-1.91(m,8H), 1.38-1.41(m,3H)。
实施例92:化合物214的制备
步骤A:9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,9-二氮杂螺环[5.5]十一烷-2-碳酸叔丁酯
Figure PCTCN2022073467-appb-000250
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(297mg,1.0mmol),2,9-二氮杂螺环[5.5]十一烷-2-羧酸叔丁酯(254mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(4mL)。反应液升温100℃反应16小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品540mg,m/z=532[M+1] +
步骤B:4-(6-(2,9-二氮杂螺环[5.5]十一烷-9-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000251
向反应瓶中依次加入甲醇(5mL),9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,9-二氮杂螺环[5.5]十一烷-2-碳酸叔丁酯(540mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(2.0mL,12.0mmol)。室温搅拌反应30min。浓缩后加水(3mL)中,用1N NaOH调pH=10后用二氯甲烷萃取两次(30mLX2),萃取液经饱和食盐水洗(10mL),无水硫酸钠干燥后浓缩得产品345mg,m/z=432[M+1] +
步骤C:N-(9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,9-二氮杂螺环[5.5]十一烷-2-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000252
向反应瓶中依次加入4-(6-(2,9-二氮杂螺环[5.5]十一烷-9-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(130mg,0.3mmol),DMF(2mL)和二异丙基乙基胺(117mg,0.9mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(58mg,0.3mmol)的DMF(0.2mL)溶液,室温搅拌反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱 层析分离得到产品45mg,m/z=588[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.59(s,1H),8.53(s,1H),8.04(d,J=8.8Hz,1H),7.60(s,1H),7.36-7.56(m,3H),7.27(s,1H),7.10(d,J=8.8Hz,1H),4.14-4.20(m,2H),3.80-3.95(m,2H),3.48-3.72(m,4H),3.15-3.28(m,2H),1.47-1.77(m,8H),1.38-1.42(m,3H)。
实施例93:化合物215的制备
6-乙氧基-4-(6-(2-((6-甲氧基吡啶-3-基)亚甲基)-2,9-二氮杂螺环[5.5]十一烷-9-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000253
向反应瓶中依次加入1,2-二氯乙烷(2mL),甲醇(1mL),4-(6-(2,9-二氮杂螺环[5.5]十一烷-9-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(65mg,0.15mmol),6-甲氧基-3-吡啶甲醛(62mg,0.45mmol)和醋酸(2滴),升温45℃反应2小时后加入三乙酰氧基硼氢化钠(160mg,0.75mmol)继续45℃搅拌16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品17mg,m/z=553[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.54(s,1H),8.50(s,1H),8.05(s,1H),8.00(d,J=8.8Hz,1H),7.65(d,J=8.8Hz,1H),7.57(s,1H),7.24(s,1H),7.01(d,J=8.8Hz,1H),6.79(d,J=8.8Hz,1H),4.14-4.20(m,2H),3.82(s,3H),3.55-3.67(m,2H),3.42-3.50(m,2H),3.40(s,1H),3.16-3.18(m,1H),2.10-2.42(m,4H),1.30-1.60(m,11H)。
实施例94:化合物216的制备
步骤A:9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,9-二氮杂螺环[5.5]十一烷-3-碳酸叔丁酯
Figure PCTCN2022073467-appb-000254
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(297mg,1.0mmol),3,9-二氮杂螺环[5.5]十一烷-3-羧酸叔丁酯(254mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(3mL)。反应液升温100℃反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品540mg,m/z=532[M+1] +
步骤B:4-(6-(3,9-二氮杂螺环[5.5]十一烷-3-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000255
向反应瓶中依次加入甲醇(5mL),9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,9-二氮杂螺环[5.5]十一烷-3-碳酸叔丁酯(540mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(4.0mL,24.0mmol)。室温搅拌反应30min。加入水(3mL),用1NNaOH调pH=10后用二氯甲烷萃取两次(15mL),萃取液经饱和食盐水洗(10mL),无水硫酸钠干燥后浓缩得产品355mg,m/z=432[M+1] +
步骤C:N-(9-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,9-二氮杂螺环[5.5]十一烷-3-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000256
向反应瓶中依次加入4-(6-(3,9-二氮杂螺环[5.5]十一烷-3-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(130mg,0.3mmol),DMF(2mL)和二异丙基乙基胺(117mg,0.9mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(58mg,0.3mmol)的DMF(0.2mL)溶液,室温搅拌反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品62.9mg,m/z=588[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.54(s,1H),8.50(s,1H),7.98(d,J=8.8Hz,1H),7.57(s,1H),7.48(d,J=8.8Hz,1H),7.48(d,J=8.8Hz,1H),7.33-7.36(m,1H),7.25(s,1H),7.13(d,J=8.8Hz,1H),4.14-4.20(m,2H),3.59-3.76(m,6H),3.15-3.29(m,2H),1.46-1.64(m,8H),1.37-1.41(m,3H)。
实施例95:化合物217的制备
6-乙氧基-4-(6-(9-((6-甲氧基吡啶-3-基)亚甲基)-3,9-二氮杂螺环[5.5]十一烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000257
向反应瓶中依次加入4-(6-(3,9-二氮杂螺环[5.5]十一烷-3-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(43mg,0.1mmol),DMF(1mL),碳酸钾(35mg,0.3mmol)和5-(氯甲基)-2-甲氧基吡啶(16mg,0.1mmol),室温搅拌反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品12.0mg,m/z=553[M+1] +, 1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.56(s,1H),8.50(s,1H),8.04(s,1H),8.01(d,J=8.8Hz,1H),7.63(d,J=8.8Hz,1H),7.57(s,1H),7.24(s,1H),7.02(d,J=8.8Hz,1H),6.79(d,J=8.8Hz,1H),4.14-4.19(m,2H),3.82(s,3H),3.58-3.65(m,4H),3.42(s,2H),3.28-3.41(m,4H), 1.41-1.59(m,8H),1.37-1.41(m,3H)。
实施例96:化合物218的制备
步骤A:2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,9-二氮杂螺环[5.5]十一烷-9-碳酸叔丁酯
Figure PCTCN2022073467-appb-000258
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(297mg,1.0mmol),2,9-二氮杂螺环[5.5]十一烷-9-羧酸叔丁酯(254mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(3mL)。反应液升温100℃反应16小时。冷却后倒入水(20mL)搅拌30分钟。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品550mg,m/z=532[M+1] +
步骤B:4-(6-(2,9-二氮杂螺环[5.5]十一烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000259
向反应瓶中依次加入甲醇(5mL),2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,9-二氮杂螺环[5.5]十一烷-9-碳酸叔丁酯(550mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(4.0mL,24.0mmol),室温搅拌反应30min。加入水(3mL),用1NNaOH调pH=10后用二氯甲烷萃取两次(30mL),萃取液经饱和食盐水洗(10mL),无水硫酸钠干燥后浓缩得产品355mg,m/z=432[M+1] +
步骤C:N-(2-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,9-二氮杂螺环[5.5]十一烷-9-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000260
向反应瓶中依次加入4-(6-(2,9-二氮杂螺环[5.5]十一烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(130mg,0.3mmol),DMF(2mL)和二异丙基乙基胺(119mg,0.9 mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(58mg,0.3mmol)的DMF(0.2mL)溶液,室温反应2小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品62.9mg,m/z=588[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.54(s,1H),8.50(s,1H),7.98-8.01(m,1H),7.57(s,1H),7.33-7.50(m,3H),7.25(s,1H),7.11-7.15(m,3H),4.14-4.20(m,2H),3.75-3.90(m,1H),3.59-3.73(m,5H),3.25-3.32(m,1H),3.10-3.20(m,1H),1.30-1.68(m,11H).
实施例97:化合物219的制备
6-乙氧基-4-(6-(9-((6-甲氧基吡啶-3-基)亚甲基)-2,9-二氮杂螺环[5.5]十一烷-2-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000261
向反应瓶中依次加入1,2-二氯乙烷(2mL),甲醇(1mL),4-(6-(2,9-二氮杂螺环[5.5]十一烷-2-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(65mg,0.15mmol),6-甲氧基-3-吡啶甲醛(24mg,0.17mmol)和醋酸(2滴),升温45℃反应2小时后加入三乙酰氧基硼氢化钠(96mg,0.45mmol)继续45℃搅拌16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品15.8mg,m/z=553[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.55(s,1H),8.50(s,1H),8.04-8.18(m,1H),8.00(d,J=8.8Hz,1H),7.65-7.73(m,1H),7.58(s,1H),7.25(s,1H),7.03-7.15(m,1H),6.75-6.90(m,1H),4.14-4.20(m,2H),3.83(s,3H),3.30-3.65(m,8H),3.17(s,1H),1.91(s,1H),1.38-1.68(m,11H)。
实施例98:化合物220的制备
步骤A:6-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-2-碳酸叔丁酯
Figure PCTCN2022073467-appb-000262
向反应瓶中依次加入6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(297mg,1.0mmol),2,6-二氮杂螺环[3.4]辛烷-2-羧酸叔丁酯(212mg,1.0mmol),二异丙基乙基胺(390mg,3.0mmol)和DMSO(4mL)。反应液升温100℃反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品520mg,m/z=490[M+1] +
步骤B:4-(6-(2,6-二氮杂螺环[3.4]辛烷-6-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000263
向反应瓶中依次加入甲醇(4mL),6-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-2-碳酸叔丁酯(520mg,1.0mmol)和6N HCl的1,4-二氧六环溶液(2.0mL,12.0mmol),室温搅拌反应30min。加入水(10mL),用1N NaOH调pH=10后用二氯甲烷萃取两次(40mL),萃取液经无水硫酸钠干燥后浓缩得产品310mg,m/z=390[M+1] +
步骤C:N-(6-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2,6-二氮杂螺环[3.4]辛烷-2-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000264
向反应瓶中依次加入4-(6-(2,6-二氮杂螺环[3.4]辛烷-6-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(78mg,0.2mmol),DMF(2mL)和二异丙基乙基胺(78mg,0.6mmol)。冰浴冷却至5℃滴加2-氯-6-氟苯甲酰氯(40mg,0.2mmol)的DMF(0.2mL)溶液,室温搅拌反应16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品29mg,m/z=546[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.55(s,1H),8.50(s,1H),8.01(d,J=8.8Hz,1H),7.50-7.56(m,2H),7.44(m,1H),7.35-7.39(m,1H),7.22(s,1H),6.68(d,J=8.8Hz,1H),4.11-4.19(m,4H),3.85-3.94(m,2H),3.65-3.78(m,2H),3.45-3.60(m,2H),2.16-2.33(m,2H),1.33-1.45(m,3H).
实施例99:化合物221的制备
6-乙氧基-4-(6-(2-((6-甲氧基吡啶-3-基)亚甲基)-2,6-二氮杂螺环[3.4]辛烷-6-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000265
向反应瓶中依次加入1,2-二氯乙烷(2mL),甲醇(1mL),4-(6-(2,6-二氮杂螺环[3.4]辛烷-6-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(60mg,0.15mmol),6-甲氧基-3-吡啶甲醛(62mg,0.45mmol)和醋酸(2滴),升温45℃反应2小时后加入三乙酰氧基硼氢化钠(160mg,0.75mmol)继续45℃搅拌16小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品29mg,m/z=511[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.54(s,1H),8.50(s,1H),8.08(s,1H),8.00(d,J=8.8Hz,1H),7.64(d,J=8.8Hz,1H),7.57(s,1H),7.22(s,1H),6.78(d,J=8.8Hz,1H),6.66(d,J=8.8Hz,1H),4.14-4.19(m,2H),3.83(s,3H),3.40-3.68(m,6H),3.15-3.30(m,3H),2.15-2.19(m,2H),1.91(s,1H),1.37-1.44(m,3H).
实施例100:化合物227的制备
4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-(丙基-2-炔-1-苯氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000266
4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-醇(46.8mg,0.1mmol),炔丙基氯(8.2mg,0.11mmol),Cs 2CO 3(48mg,0.15mmol)和DMF(2mL)置于10mL的反应瓶中,升温至50℃反应5小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品9.4mg,m/z=507[M+1] +, 1H NMR(400MHz,DMSO-d 6):δ12.69(s,1H),8.60-8.66(m,2H),8.1-8.13(m,2H),7.60-7.78(m,2H),7.34(s,1H),6.77-6.94(m,2H),5.00(s,2H),3.70-3.86(m,7H),3.30-3.32(m,7H).
实施例101:化合物228的制备
甲基2-((4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-基)醚)乙酸酯
Figure PCTCN2022073467-appb-000267
4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-醇(70mg,0.15mmol),氯乙酸甲酯(19mg,0.18mmol),Cs 2CO 3(73mg,0.225mmol)和DMF(5mL)置于10mL的反应瓶中,升温至50℃反应3小时,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品29.6mg,m/z=541[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.68(s,1H),8.60-8.66(m,2H),8.10-8.16(m,2H),7.64-7.72(m,2H),7.35(s,1H),6.91-6.93(m,1H),6.76-6.79(m,1H),5.0(s,2H),3.80-3.97(m,4H),3.62-3.80(m,6H),3.42-3.62(m,4H),3.31-3.33(m,1H),2.51-2.67(m,1H)
实施例102:化合物229的制备
步骤A:乙基1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(((6-甲氧基吡啶-3-基)甲基)氨)哌啶-4-羧酸乙酯
Figure PCTCN2022073467-appb-000268
将乙基4-氨基-1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-羧酸盐(449mg,1.0mmol),6-甲氧基烟醛(164mg,1.2mmol),Et 3N(303mg,3.0mmol),正丙醇(5ml)加入反应瓶,25℃反应12h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品268mg,m/z=571[M+1] +
步骤B:(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(((6-甲氧基吡啶-3-基)甲基)氨)哌啶-4-基)甲醇
Figure PCTCN2022073467-appb-000269
将乙基1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(((6-甲氧基吡啶-3-基)甲基)氨)哌啶-4-羧酸乙酯(268mg,0.47mmol),5mL THF加入反应瓶,0℃下加入LiAlH 4(19.6mg,0.51mmol),反应1h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品101mg,m/z=529[M+1] +, 1H NMR(400MHz,DMSO-d 6)δ12.66(s,1H),8.58(s,1H),8.52(d,J=2.0Hz,1H),8.15(s,1H),8.03(d,J=9.1Hz,1H),7.76(d,J=8.4Hz,1H),7.60(m,2H),7.26(d,J=2.1Hz,1H),7.08(s,1H),6.81(s,1H),4.66(s,1H),4.18(m,2H),3.99(s,2H),3.83(s,3H),3.64(s,2H),2.89(m,3H),1.65(s,2H),1.53(s,2H),1.40(t,J=6.9Hz,3H),0.90(d,J=6.7Hz,1H)。
实施例103:化合物232的制备
步骤A:4-(6-(6-((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-三氟甲基磺酸酯
Figure PCTCN2022073467-appb-000270
反应瓶中加入4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-醇(0.936g,2mmol),DIEA(0.516g,4mmol)和10mL DMSO。冰浴下,加入1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(0.714g,2mmol),然后室温反应2h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1.0g,m/z=601[M+1] +
步骤B:4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000271
反应瓶中加入4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-三氟甲基磺酸酯(300mg,0.5mmol),(双(频那醇合)二硼(381mg,1.5mmol),Pd(dppf)Cl 2(73mg,0.1mmol),醋酸钾(245mg,2.5mmol)和10mL 1,4二氧六环,90℃反应4h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品178mg,m/z=579[M+H] +
步骤C:4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-基)硼酸
Figure PCTCN2022073467-appb-000272
反应瓶中加入(4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(180mg,0.31mmol),NaIO 4(200mg,0.93mmol)和THF(8mL)/水(2mL),25℃反应0.5h;1N HCl(3mL)加入到上述反应液中,搅拌过夜。反应液倒入100mL水中,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩得到产品55mg,m/z=496[M+1] +
步骤D:6-(氮杂环丁基-1-基)-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000273
反应瓶中加入4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-基)硼酸(50mg,0.1mmol),杂氮环丁烷(114mg,2mmol),醋酸铜(37mg,0.2mmol),吡啶(45mg,0.5mmol),4A分子筛(2g)和5mL DMF,50℃反应12h,过滤,滤液倒入20mL水中,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩得到产品20mg,m/z=508[M+1] +
实施例104:化合物233的制备
N-((3R,4S)-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇的合成
Figure PCTCN2022073467-appb-000274
将(3S,4S)-4-氨基-1-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇盐酸盐(100mg,0.2mmol),2,6-二氟苯甲酸(32mg,0.2mmol),DIEA(129mg,1.0mmol),HATU(76mg,0.2mmol),DMF(3mL)加入反应瓶,25℃反应1小时。将反应液倒入30mL水中,EA萃取,干燥有机相,旋干溶剂,柱层析纯化(DCM:MeOH=10:1)得产品5.0mg,m/z=534[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.65(s,1H),8.61(d,J=8.0Hz,1H),8.59(s,1H),8.57(s,1H),8.02-8.05(m,1H),7.58(s,1H),7.07-7.49(m,5H),5.07(br, 1H),4.40-4.44(m,1H),4.14-4.22(m,3H),3.92-3.95(m,1H),3.50-3.51(m,1H),3.16-3.22(m,1H),2.96-3.01(m,1H),2.01-2.11(m,1H),1.37-1.48(m,4H)。
实施例105:化合物234的制备
步骤A:(1-(5-溴吡啶-2-基)-4-((二甲氨基)甲基)哌啶-4-基)氨基甲酸叔丁酯
Figure PCTCN2022073467-appb-000275
向50mL反应瓶中加入(1-(5-溴吡啶-2-基)-4-甲酰基哌啶-4-基)氨基甲酸叔丁酯(200mg,0.52mmol),1,2-二氯甲烷(2mL),二甲胺(0.78mL,1.56mmol),醋酸硼氢化钠(220mg,1.04mmol),室温下搅拌16小时后,加水淬灭后,浓缩,柱层析分离得到产品177mg,m/z=413/415[M+1] +
步骤B:1-(5-溴吡啶-2-基)-4-((二甲基氨基)甲基)哌啶-4-胺盐酸盐
Figure PCTCN2022073467-appb-000276
将(1-(5-溴吡啶-2-基)-4-((二甲氨基)甲基)哌啶-4-)氨基甲酸叔丁酯(177mg,0.43mmol),甲醇盐酸气(4M,2mL)加入反应瓶中,25℃反应5h。直接旋干得产品250mg,m/z=313/315[M+1] +
步骤C:N-(1-(1-溴吡啶-2-基)-4-((二甲基氨基)甲基)哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000277
将1-(5-溴吡啶-2-基)-4-((二甲基氨基)甲基)哌啶-4-胺盐酸盐(250mg,0.8mmol),2,6-二氟苯甲酸(152mg,0.96mmol),HATU(334mg,0.88mmol),DIEA(309mg,2.4mmol),DMF(2mL)加入反应瓶,25℃反应15h。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品117mg,m/z=453/455[M+1] +
步骤D:N-(4-((二甲基氨基)甲基)-1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000278
向反应瓶中依次加入N-(1-(1-溴吡啶-2-基)-4-((二甲基氨基)甲基)哌啶-4-基)-2,6-二氟苯甲酰胺(117mg,0.35mmol),6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑[3',4':3,4]吡唑并[1,5-a]吡啶(161mg,0.35mmol),碳酸铯(326mg,1.0mmol),水(1mL), 1,4-二氧六环(5mL),Pd(dppf)Cl 2(25mg,0.035mmol),加热至90℃反应16小时,反应液倒入水中,二氯甲烷萃取,柱层析分离得到产品50mg,m/z=575[M+1] +, 1H NMR(400MHz,DMSO-d 6):δ12.64(s,1H),8.61-8.56(m,1H),8.51(m,1H),8.44(s,1H),8.04(m,1H),7.59(s,1H),7.49(m,1H),7.26(m,1H),7.21-7.06(m,3H),4.22(m,2H),4.18(m,2H),3.17(m,2H),2.67(s,2H),2.34(m,2H),2.29(s,6H),1.57(m,2H),1.40(m,3H)。
实施例106:化合物237的制备
(4-(6-(6-(((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-6-基)二甲基氧化膦
Figure PCTCN2022073467-appb-000279
向反应瓶中依次加入4-(6-(6-(((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-6-基三氟甲磺酸盐(30mg,0.05mmol),二甲基氧化磷(9.7mg,0.125mmol),醋酸钯(0.56mg,0.0025mmol),磷酸钾(11mg,0.055mmol),DMF(1mL),XantPhos(29mg,0.05mmol),在氮气保护条件下,加热至150℃反应2小时,反应液倒入水中,二氯甲烷萃取,柱层析分离得到产品19mg,m/z=529[M+1] +1HNMR(400MHz,DMSO-d 6):12.90(s,1H),9.02(m,1H),8.70(m,1H),8.20–8.08(m,2H),7.83–7.65(m,3H),6.95(m,1H),6.79(m,1H),3.83(m,5H),3.70(m,2H),3.60(s,2H),3.54(s,2H),3.34(s,1H),1.85(s,3H),1.82(s,3H),1.60(m,1H)。
实施例107:化合物238的制备
步骤A:4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-羟基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶
Figure PCTCN2022073467-appb-000280
将6-((4-甲氧基)苄基)-4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-羟基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶(1g,1.7mmol),DCM(5mL)加入反应瓶,0℃条件下滴加TFA(5mL),室温反应4h。直接旋干溶剂得产品1.55g,m/z=469[M+1] +
步骤B:4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-三氟甲烷磺烷基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶
Figure PCTCN2022073467-appb-000281
向反应瓶中依次加入4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-羟基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶(1.5g,3.2mmol),N-苯基双(三氟甲磺酰)亚胺(1.26g,3.5mmol),DIEA(2g,16mmol),DMF(10mL)。25℃反应4h,LCMS监测反应完全,将反应液倒入30mL水中,EA萃取,干燥有机溶剂,浓缩,硅胶柱纯化分离(DCM:MeOH=10:1)得产品810mg,m/z=601[M+1] +
步骤C:4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶
Figure PCTCN2022073467-appb-000282
依次将4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-三氟甲烷磺烷基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶(810mg,0.19mmol),联硼酸频那醇酯(377mg,1.49mmol),KOAc(400mg,4.05mmol),Pd(dppf)Cl 2(100mg,0.14mmol),1,4-二氧六环(10mL)加入反应瓶,N 2置换3次,95℃反应12h,LCMS监测反应完全。反应液直接进行硅胶柱纯化(DCM:MeOH=10:1)分离得产品510mg,m/z=579[M+1] +
步骤D:(4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-6-基)硼酸
Figure PCTCN2022073467-appb-000283
将4-(6-(6((6-甲氧基吡啶-3-基)亚甲基)-3,6-二氮杂环[3.1.1]庚烷-3-基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶(110mg,0.19mmol),THF(5mL)加入反应瓶,搅拌,向反应瓶中加高碘酸钠(41mg,0.19mmol),搅拌0.5h后,加HCl的水溶液(1N,3mL),25℃反应4h。反应液加硫代硫酸钠水溶液淬灭,二氯甲烷萃取,无水硫酸钠干燥,浓缩,硅胶柱纯化分离得产品38.7mg,m/z=497[M+1] +1H-NMR(400MHz,DMSO-d 6):δ12.69(s,1H),9.00-9.20(m,1H),8.63-8.69(m,1H),8.46-8.50(m,1H),8.05-8.11(m,2H),7.59(s,1H),7.28(m,1H),7.13(m,1H),6.53-6.90(m,1H),3.82-3.88(m,5H),3.66-3.72(m,4H),3.50-3.58(m,3H),1.60(m,1H)。
实施例108:化合物239的制备
4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-羧酸
Figure PCTCN2022073467-appb-000284
向反应瓶中依次加入MeOH(3mL)和NaOH水溶液(5M,3mL),4-(6-(6-(((6-甲氧基 吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-腈(15mg,0.03mmol),50℃下反应16小时,反应完全。加HCl水溶液(1M)调pH=7,减压浓缩,柱层析分离得到产品8mg,m/z=497[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.79(brs,1H),9.11(s,1H),8.64(s,1H),8.11(s,1H),8.09(s,1H),7.84(s,1H),7.73-7.69(m,2H),6.92(d,J=8.4Hz,1H),6.77(d,J=8.4Hz,1H),3.86-3.75(m,5H),3.72-3.68(m,2H),3.66-3.51(m,4H),1.60(d,J=8.4Hz,1H).
实施例109:化合物240的制备
步骤A:2-((4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-6-基)氧基)乙酸乙酯
Figure PCTCN2022073467-appb-000285
向反应瓶中加入4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-6-羟基(100mg,0.21mmol),碳酸铯(138.8mg,0.42mmol),N,N-二甲基甲酰胺(3mL),搅拌均匀,然后加入氯乙酸乙酯(39.1mg,0.32mmol)室温反应10小时,加水淬灭,乙酸乙酯萃取,硅胶柱层析分离得产品60mg,m/z=555.2[M+1] +
步骤B:2-((4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-6-基)氧基)乙酸
Figure PCTCN2022073467-appb-000286
向反应瓶中依次加入2-((4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-6-基)氧基)乙酸乙酯(60mg,0.11mmol),甲醇(1mL),四氢呋喃(1mL),水(1mL),氢氧化锂(9mg,0.22mmol),搅拌,60℃反应3小时。反应完全,浓缩后,加入2mL水,盐酸调节pH=6析出产品,过滤,干燥得产品20mg,m/z=527.1[M+1] +1H-NMR(400MHz,DMSO-d 6):δ12.66(s,1H),8.67(d,J=2.4Hz,1H),8.56(d,J=2.4Hz,1H),8.27(s,1H),8.15(m,1H),7.80(s,1H),7.62(s,1H),7.34(s,1H),6.95(d,J=8.8Hz,1H),6.85(s,1H),4.89(s,2H),3.85(m,7H),3.56(s,2H),3.50(s,2H),2.51(s,1H),1.79(s,1H).
实施例110:化合物242的制备
4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-丙烯酰酯
Figure PCTCN2022073467-appb-000287
反应瓶中加入4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂二环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-醇(156mg,0.33mmol)和3mL NMP。冰浴条件下,加入NaH(60%,20mg,0.49mmol),搅拌30分钟。然后在冰浴条件下,缓慢加入丙烯酰氯(45mg,0.49mmol),反应0.5h,反应完全。反应液倒入30mL冰水中,二氯甲烷萃取,硫酸钠干燥,减压浓缩,硅胶柱纯化得产品10mg,m/z=523[M+1] +, 1H NMR(400MHz,DMSO-d 6):δ12.80(s,1H),8.99(s,1H),8.66(s,1H),8.10-8.13(m,2H),7.68-7.72(m,2H),7.52(s,1H),6.92-6.94(m,1H),6.76-6.78(m,1H),6.60-6.65(m,1H),6.47-6.53(m,1H),6.22-6.25(m,1H),3.76-3.82(m,6H),3.53-3.69(m,7H)。
实施例111:化合物244的制备
4-(5-(6-乙氧基-1-甲基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)1-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-2基)甲醇
Figure PCTCN2022073467-appb-000288
将4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)1-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-2基)甲醇(50mg,0.1mmol),K 2CO 3(41mg,0.3mmol),CH 3I(16mg,0.1mmol),DMF(3mL)加入反应瓶,25℃反应1小时。将反应液倒入30mL水中,EA萃取,干燥有机相,旋干溶剂,柱层析纯化得产品9.8mg,m/z=529[M+1] +, 1H-NMR(400MHz,DMSO-d 6):δ8.57(s,1H),8.51(s,1H),8.09(s,1H),8.02(d,J=8.8Hz,1H),7.62(d,J=8.4Hz,1H),7.57(s,1H),7.25(s,1H),7.02(d,J=8.8Hz,1H),6.78(d,J=8.4Hz,1H),4.74-4.77(m,1H),4.10-4.20(m,3H),3.99-4.00(m,1H),3.84-3.90(m,3H),3.79-3.84(m,6H),3.53-3.54(m,1H),3.16-3.38(m,2H),2.67-2.68(m,1H),2.26-2.33(m,1H),1.40(t,J=7.2Hz,3H).
实施例112:化合物245的制备
步骤A:4-(6-(3-(氯甲基)-4-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-1基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶
Figure PCTCN2022073467-appb-000289
将4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)1-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-2基)甲醇(100mg,0.2mmol),DMF(2mg,0.02mmol),SOCl 2(1mL)加入反应瓶,80℃反应4h。LCMS监测反应完全,将反应液倒入20mL冰水中,4M NaOH 的水溶液调节pH=7,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品50mg,m/z=533[M+1] +
步骤B:1-4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)1-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-2基)–N,N-二甲基甲基甲胺
Figure PCTCN2022073467-appb-000290
将4-(6-(3-(氯甲基)-4-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-1-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶(50mg,0.1mmol),二甲胺的四氢呋喃溶液(1M,0.1mL,0.1mmol),DMF(2mL)加入反应瓶,80℃反应1h。将反应液倒入20mL水中,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品22.8mg,m/z=542[M+1] +, 1H-NMR(400MHz,DMSO-d 6):δ12.64(brs,1H),8.57(s,1H),8.51(s,1H),8.09(s,1H),8.02(d,J=8.8Hz,1H),7.62(d,J=8.4Hz,1H),7.57(s,1H),7.25(s,1H),7.02(d,J=8.8Hz,1H),6.78(d,J=8.4Hz,1H),4.74-4.77(m,1H),4.10-4.20(m,3H),3.99-4.00(m,1H),3.79-3.84(m,6H),3.53-3.54(m,1H),3.16-3.38(m,2H),2.67-2.68(m,1H),2.49-2.51(m,6H),2.26-2.33(m,1H),1.36(t,J=7.2Hz,3H)。
实施例113:化合物246的制备
步骤A:4-(6-(6-(((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-基三氟甲磺酸盐
Figure PCTCN2022073467-appb-000291
向反应瓶中依次加入DMSO(5mL),DIEA(1mL),4-(6-(6-(((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-6-醇(510mg,1.09mmol),然后分批加入1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰基)甲磺酰胺(389mg,1.09mmol),室温下反应2小时。加水搅拌淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品500mg,m/z=601[M+1] +1HNMR(400MHz,DMSO-d 6):δ13.00(brs,1H),9.51(d,J=2.0Hz,1H),8.68(s,1H),8.14(d,J=8.0Hz,1H),8.11(s,1H),7.79-7.70(m,3H),6.95(d,J=8.8Hz,1H),6.77(d,J=8.4Hz,1H),3.76-3.91(m,5H),3.47-3.75(m,6H),3.31(s,2H),1.61-1.59(m,1H).
实施例114:化合物247的制备
步骤A:7-溴-6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶
Figure PCTCN2022073467-appb-000292
向反应瓶中加入6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶(5g,10.06mmol),N,N-二甲基甲酰胺(50mL),搅拌溶解,然后加入NBS(2.15g,12.08mmol),室温反应1小时,加水淬灭,乙酸乙酯萃取,硅胶柱层析分离得产品1.5g,m/z=575/577[M+1] +
步骤B:7-氨基-6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶
Figure PCTCN2022073467-appb-000293
向反应瓶中依次加入7-溴-6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶(200mg,0.35mmol),二甲基亚砜(3mL),碘化亚铜(200mg),氨水(1mL),氮气置换,90℃反应13小时。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品5.2mg,m/z=512.1[M+1] +, 1H-NMR(400MHz,DMSO-d 6):δ12.66(s,1H),8.57(s,1H),8.10(s,1H),8.01(m,1H),7.70(m,1H),7.62(s,1H),7.44(s,1H),6.87(m,1H),6.78(m,1H),6.51(s,2H),4.22-4.16(q,J=8Hz,2H),3.83(s,3H),3.78-3.70(m,5H),3.54(m,4H),1.70(m,1H),1.41-1.38(m,3H).
实施例115:化合物248的制备
步骤A:3-溴-6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶
Figure PCTCN2022073467-appb-000294
向反应瓶中加入6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶(5g,10.06mmol),N,N-二甲基甲酰胺(50mL),搅拌溶解,室温下缓慢加入NBS(2.15g,12.08mmol),反应1小时,加水淬灭,乙酸乙酯萃取,硅胶柱层析分离得产品1g,m/z=575/577[M+1] +
步骤B:3-氰基-6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶
Figure PCTCN2022073467-appb-000295
向反应瓶中依次加入3-溴-6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚-3-基)吡啶-3-基)-1H-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶(100mg,0.17mmol),N,N-二甲基乙酰胺(1mL),水(0.5ml),亚铁氰化钾(32mg,0.09mmol),XantPhos(20mg, 0.03mmol),氯化烯丙基钯(II)二聚体(6.3mg,0.017mmol),氮气置换,90℃反应13小时。加水和乙酸乙酯萃取,硅胶柱层析分离得产品2mg,m/z=522[M+1] +1H-NMR(400MHz,DMSO-d 6):δ12.85(s,1H),8.69(s,1H),8.15-8.10(m,2H),7.71-7.57(m,3H),6.98-6.92(m,1H),6.79-6.77(d,J=8.4Hz,1H),4.38-4.33(q,J=6.8Hz,2H),3.83-3.77(m,5H),3.70-3.53(m,7H),1.61-1.59(d,J=8.4Hz,1H),1.41-1.40(t,J=3.2Hz,3H)。
实施例116:化合物249的制备
步骤A:4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2-(羟甲基)哌嗪-1-羧酸叔丁酯
Figure PCTCN2022073467-appb-000296
将6-乙氧基-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(1.4g,4.7mmol),2-(羟甲基)哌嗪-1-羧酸叔丁酯(1g,4.7mmol),DIEA(1.8g,14.1mmol),DMSO(15mL)加入反应瓶,90℃反应12h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品1.6g,m/z=494[M+1] +
步骤B:4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2-(羟甲基)哌嗪盐酸盐
Figure PCTCN2022073467-appb-000297
4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2-(羟甲基)哌嗪-1-羧酸叔丁酯(1.6g,3.2mmol)加入到HCl的1,4-二氧六环溶液(4M,20mL),25℃反应4h,直接旋干溶剂得产品1.7g,m/z=394[M+1] +
步骤C:4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)1-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-2基)甲醇
Figure PCTCN2022073467-appb-000298
将4-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-2-(羟甲基)哌嗪盐酸盐(100mg,0.19mmol),6-甲氧基-3-吡啶甲醛(80mg,0.57mmol),Et 3N(98mg,0.8mmol),Pic-BH 3(63mg,0.57mmol),DMSO(3mL)加入反应瓶,45℃反应12h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品10.1mg,m/z=515[M+1] +1H-NMR(400MHz,DMSO-d 6):δ12.64(brs,1H),8.57(s,1H),8.51(s,1H),8.09(s,1H),8.02(d,J=8.8Hz,1H),7.62(d,J=8.4Hz,1H),7.57(s,1H),7.25(s,1H),7.02(d,J=8.8Hz,1H),6.78 (d,J=8.4Hz,1H),4.74-4.77(m,1H),4.10-4.20(m,3H),3.99-4.00(m,1H),3.79-3.84(m,6H),3.53-3.54(m,1H),3.16-3.38(m,2H),2.67-2.68(m,1H),2.26-2.33(m,1H),1.39(t,J=7.2Hz,3H).
实施例117:化合物250的制备
1-(3-(5-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3,6-氮杂二环[3.1.1]庚烷-6-基)丙-2-烯-1-酮
Figure PCTCN2022073467-appb-000299
反应瓶中加入4-(6-(3,6-二氮杂双环[3.1.1]庚基-3-基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶硫酸盐(150mg,0.2mmol),DIEA(193mg,1.5mmol)和DMF(2mL),冰浴下加入丙烯酰氯(22mg,0.25mmol),然后25℃反应2h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品15.2mg,m/z=430[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.60(d,J=2.4Hz,1H),8.51(s,1H),8.05(dd,J 1=2.4Hz,J 2=8.8Hz,1H),7.60(s,1H),7.24(s,1H),6.83(d,J=8.8Hz,1H),6.44-6.51(m,1H),6.10(d,J=16.8Hz,1H),5.67(d,J=10.4Hz,1H),4.87-4.88(m,1H),4.53-4.54(m,1H),4.15(q,J=6.8Hz,2H),3.98-4.04(m,1H),3.75-3.78(m,2H),3.59-3.68(m,1H),2.71-2.77(m,1H),1.65-1.68(m,1H),1.39(t,J=6.8Hz,3H)。
实施例118:化合物251的制备
6-乙氧基-4-(6-(3-(甲氧基甲基)-4-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-1基)哌啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶
Figure PCTCN2022073467-appb-000300
将4-(6-(3-(氯甲基)-4-((6-甲氧基吡啶-3-基)亚甲基)哌嗪-1基)吡啶-3-基)-6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶(150mg,0.28mmol),甲醇钠(17mg,0.31mmol),MeOH(2mL)加入反应瓶,25℃反应1h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品6.5mg,m/z=529[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.57(s,1H),8.51(s,1H),7.99-8.10(m,2H),7.57-7.71(m,2H),7.26(s,1H),7.04(d,J=8.8Hz,1H),6.77(d,J=8.4Hz,1H),4.20-4.24(m,2H),4.02-4.18(m,2H),3.91-3.98(m,4H),3.82-3.84(m,2H),3.30-3.33(m,6H),3.21-3.22(m,1H),2.49-2.51(m,2H),1.39(t,J=7.2Hz,3H).
实施例119:化合物258和化合物259的制备
化合物258:(1S,4R)-4-(6-乙氧基-1氢-吡唑并[3’,4’:3,4]吡唑并[1,5-a]吡啶-4-基)-N-((S)-1-(6-(4–氟-1氢-吡唑-1-基)吡啶-3-基)乙基)-1-甲氧基环己烷-1-羧酰胺
Figure PCTCN2022073467-appb-000301
化合物259:(1R,4S)-4-(6-乙氧基-1氢-吡唑并[3',4':3,4]吡唑并[1,5-a]吡啶-4-基)-N-((S)-1-(6-(4-氟-1氢-吡唑-1-基)吡啶-3-基)乙基)-1-甲氧基环己烷-1-羧酰胺
Figure PCTCN2022073467-appb-000302
步骤A:叔丁基-(S)-(1-(6-溴吡啶-3-基)乙基)氨基甲酸酯
Figure PCTCN2022073467-appb-000303
向反应瓶中加入(S)-1-(6-溴吡啶-3-基)乙-1-胺(210mg,1.05mmol),Boc酸酐(273mg,1.25mmol),TEA(318mg,3.15mmol),乙腈(10mL),室温反应4h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品300mg,m/z=301[M+1] +
步骤B:(S)-(1-(6-(4-(4-氟-1氢-吡唑-1-基)吡啶-3-基)乙基)氨基甲酸叔丁酯
Figure PCTCN2022073467-appb-000304
向反应瓶中加入叔丁基-(S)-(1-(6-溴吡啶-3-基)乙基)氨基甲酸酯(290mg,0.96mmol),4-氟-1氢-吡唑(249mg,29.3mmol),N 1,N 2-二甲基环己烷-1,2-二胺(341mg,2.4mmol),碘化亚铜(183mg,0.96mmol),乙腈(5mL),108℃反应4h,然后降至室温,加入50mL水和50mL乙酸乙酯,萃取分液,有机相用50mL饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩,柱层析分离(EA/PE=20%)得到产品245mg,m/z=307[M+1] +
步骤C:(S)-(1-(6-(4-(4-氟-1氢-吡唑-1-基)吡啶-3-基)乙-1-胺盐酸盐
Figure PCTCN2022073467-appb-000305
向反应瓶中加入(S)-(1-(6-(4-(4-氟-1氢-吡唑-1-基)吡啶-3-基)乙基)氨基甲酸叔丁酯(240mg,0.78mmol),4M盐酸/二氧六环溶液(10mL),室温反应2h,反应完全,减压浓缩得到产品180mg,m/z=207[M+1] +
步骤D:1-甲氧基-4-(((三氟甲基)磺酰基)氧基)环己-3-烯-1-羧酸甲酯
Figure PCTCN2022073467-appb-000306
反应瓶中加入1-甲氧基-4-氧代环己烷-1-甲酸甲酯(680mg,3.6mmol),THF(15mL),然后在-70℃到-80℃内温下,将LDA(2N,2.7mL)滴加入反应液,保持该温度区间反应1h。将1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰基)甲磺酰胺(1.5g,4.3mmol)溶于10mL四氢呋喃,然后滴加入上述反应液,滴加完后,反应液自然升到室温反应2h,加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品820mg。
步骤E:4-(6-乙氧基-1氢-吡唑[3'.4':3,4]吡唑[1,5-a]吡啶-4-基)-1-甲氧基环己-3-烯-1-羧酸甲酯
Figure PCTCN2022073467-appb-000307
反应瓶中加入6-乙氧基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(938mg,2.9mmol),1-甲氧基-4-(((三氟甲基)磺酰基)氧基)环己-3-烯-1-羧酸甲酯(1.0g,3.1mmol),碳酸铯(1.8g,5.7mmol),Pd(dppf)Cl 2(21mg,0.28mmol),1,4-二氧六环(25mL),水(5mL),氮气置换2min,加热100℃反应6h。加水搅拌淬灭,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,柱层析分离得到产品700mg,m/z=371[M+1] +
步骤F:4-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)-1-甲氧基环己烷-1-甲酸甲酯
Figure PCTCN2022073467-appb-000308
反应瓶中加入4-(6-乙氧基-1氢-吡唑[3'.4':3,4]吡唑[1,5-a]吡啶-4-基)-1-甲氧基环己-3-烯-1-羧酸甲酯(420mg,1.1mmol),无水乙醇(30mL),氮气置换,加入Pd(OH) 2(100mg),再用氢气置换,保持氢气环境80℃反应48h,反应完全。降温,过滤,用20mL乙醇淋洗,浓缩,柱层析纯化得到产品330mg,m/z=373[M+1] +
步骤G:4-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)-1-甲氧基环己烷-1-甲酸
Figure PCTCN2022073467-appb-000309
反应瓶中加入4-(6-乙氧基-1氢-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)-1-甲氧基环己烷-1-甲酸甲酯(320mg,0.8mmol),THF(10mL),甲醇(2mL),将氢氧化锂(81mg,3.4mmol)溶于1mL水中,加入上述反应体系,60℃反应18h,反应完全。减压浓缩得到产品280mg,m/z=359[M+1] +
步骤H:(S)-4-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)-N-(1-(6-(4-氟-1氢-吡唑-1-基)吡啶-3-基)乙基)-1-甲氧基环己烷-1-羧酰胺
反应瓶中加入4-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)-1-甲氧基环己烷-1-甲酸(250mg,0.7mmol),(S)-(1-(6-(4-(4-氟-1氢-吡唑-1-基)吡啶-3-基)乙-1-胺盐酸盐(180mg,0.7mmol),PyBop(400mg,0.77mmol),DIEA(271mg,2.1mmol),DMF(10mL),室温反应12h,反应完全。10mL饱和碳酸氢钠淬灭,50mL水稀释,乙酸乙酯(3x 30mL)萃取,合并有机相,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析分离得产品340mg,经 过SFC手性分离,得到化合物258(104.2mg),m/z=547[M+1] +1H NMR(400MHz,DMSO-d 6):δ12.55(brs,1H),8.68(d,J=4.4Hz,1H),8.55(d,J=8.4Hz,1H),8.45(s,1H),8.37(s,1H),8.00(d,J=8.4Hz,1H),7.94-7.88(m,3H),6.99(s,1H),5.06(t,J=7.2Hz,1H),4.08(q,J=7.2Hz,2H),3.21(s,3H),3.07-2.96(m,1H),2.09-1.86(m,5H),1.84-1.72(m,3H),1.48(d,J=7.2Hz,3H),1.34(t,J=7.2Hz,3H).和化合物259(75.8mg),m/z=547[M+1] +1HNMR(400MHz,DMSO-d 6):δ12.50(brs,1H),8.65(d,J=4.4Hz,1H),8.59(d,J=8.4Hz,1H),8.45(s,1H),8.35(s,1H),8.00(d,J=8.4Hz,1H),7.92-7.86(m,3H),6.83(s,1H),5.15(t,J=7.6Hz,1H),4.04(q,J=7.2Hz,2H),3.11(s,3H),3.05-2.93(m,1H),2.37-2.32(m,1H),1.94-1.72(m,5H),1.70-1.62(m,2H),1.48(d,J=7.2Hz,3H),1.32(t,J=7.2Hz,3H)。
实施例120:化合物267的制备
N-((3R,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
步骤A:4-溴-6-(2-氟乙氧基)-2-氟吡唑[1,5-a]吡啶-3-甲醛
Figure PCTCN2022073467-appb-000310
将4-溴-6-羟基-2-氟吡啶[1,5-a]吡啶-3-甲醛(5g,19.3mmol),K 2CO 3(5.3g,38.6mmol),1-氟-2-溴乙烷(2.7g,21.2mmol),DMF(50mL)加入反应瓶,65℃条件下反应2小时。LCMS监测反应完全,将反应液倒入冰水中,二氯甲烷萃取,柱层析纯化得到产品2.65g,m/z=305,307[M+1] +
步骤B:4-溴-6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000311
将4-溴-6-(2-氟乙氧基)-2-氟吡唑[1,5-a]吡啶-3-甲醛(2.55g,13.6mmol),水合肼(85%,4.0g,68.1mmol),DMSO(50mL)加入反应瓶,130℃反应12小时。LCMS监测反应完全,将反应液倒入水中,EA萃取,干燥有机相,过滤,减压旋干溶剂,柱层析纯化得到产品1.6g,m/z=299,301[M+1] +
步骤C:6-(2-氟乙氧基)-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000312
将4-溴-6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(1.6g,5.3mmol),6-氟吡啶-3-硼酸(0.75g,5.9mmol),Cs 2CO 3(3.49g,10.7mmol),Pd(dppf)Cl 2(390mg,0.5mmol),二氧六环(20mL),水(5mL)加入反应瓶,氮气置换3次,90℃反应16小时。LCMS监测反应完全,将反应液倒入水中,EA萃取,干燥有机相,减压浓缩,柱层析纯化得到产品837mg,m/z=316[M+1] +
步骤D:((3R,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000313
反应瓶中加入6-(2-氟乙氧基)-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(410mg,1.3mmol),((3R,4S)-3-羟基哌啶-4-基)碳酸叔丁酯(309mg,1.4mmol),DIEA(840mg,6.5mmol)和DMSO(5mL),90℃反应40h。冷却到室温,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品420mg,m/z=512[M+1] +
步骤E:(3R,4S)-4-氨基-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐
Figure PCTCN2022073467-appb-000314
反应瓶中加入((3R,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯(420mg,0.8mmol),4N盐酸-二氧六环溶液(8mL),25℃反应1h,反应完全,减压浓缩干,得到产品400mg,m/z=412[M+1] +
步骤F:N-((3R,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000315
将(3R,4S)-4-氨基-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(400mg,0.9mmol),2,6-二氟苯甲酸(151mg,1.0mmol),DIEA(512mg,4.3mmol),HATU(363mg,1.0mmol)和DMF(5mL),25℃反应2h,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品161.5mg,m/z=552[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.68(s,1H),8.57-8.63(m,3H),8.02-8.05(m,1H),7.70(m,1H),7.48-7.61(m,1H),7.32-7.33(m,1H),7.11-7.15(m,3H),5.01-5.01(m,1H),4.87-4.89(m,1H),4.75-4.77(m,1H),4.38-4.46(m,2H),4.17-4.21(m,3H),3.94-3.94(m,1H),3.36-3.41(m,1H),3.11-3.14(m,1H),1.93-1.96(m,1H),1.69-1.72(m,1H)。
实施例121:化合物268的制备
N-((3R,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
步骤A:4-溴-6-(2,2-二氟乙氧基)-2-氟吡唑[1,5-a]吡啶-3-甲醛
Figure PCTCN2022073467-appb-000316
将4-溴-6-羟基-2-氟吡啶[1,5-a]吡啶-3-甲醛(15g,54mmol),K 2CO 3(14.9g,108mmol),1,1-二氟-2-碘乙烷(11.4g,60mmol),DMF(140mL)加入反应瓶,65℃反应24小时。LCMS监测反应完全,将反应液倒入水中,EA萃取,干燥有机相,减压浓缩,柱层析纯化得到产品4.4g,m/z=323,325[M+1] +
步骤B:4-溴-6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000317
将4-溴-6-(2,2-二氟乙氧基)-2-氟吡唑[1,5-a]吡啶-3-甲醛(4.4g,13.6mmol),水合肼(85%,4.0g,68.1mmol),DMSO(50mL)加入反应瓶,130℃反应12小时。LCMS监测反应完全,将反应液倒入水中,EA萃取,干燥有机相,减压浓缩,柱层析纯化得到产品3.4g,m/z=317,319[M+1] +
步骤C:6-(2,2-二氟乙氧基)-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000318
将4-溴-6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(2.9g,9.1mmol),6-氟吡啶-3-硼酸(1.4g,10.0mmol),Cs 2CO 3(6.0g,18.3mmol),Pd(dppf)Cl 2(670mg,0.9mmol),二氧六环(34mL),水(7mL)加入反应瓶,氮气置换3次,90℃反应2小时。LCMS监测反应完全,将反应液倒入水中,EA萃取,干燥有机相,减压浓缩,柱层析纯化得到产品1.6g,m/z=334[M+1] +
步骤D:((3R,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000319
反应瓶中加入6-(2,2-二氟乙氧基)-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(1g,3.0mmol),((3R,4S)-3-羟基哌啶-4-基)碳酸叔丁酯(700mg,3.3mmol),DIEA(1900mg,15.0mmol)和DMSO(10mL),90℃反应40h。冷却到室温,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品1g,m/z=530[M+1] +
步骤E:(3R,4S)-4-氨基-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐
Figure PCTCN2022073467-appb-000320
反应瓶中加入((3R,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯(1g,1.9mmol),4N盐酸-二氧六环溶液(20mL),25℃反应1h,反应完全,减压浓缩干,得到产品1.1g,m/z=430[M+1] +
步骤F:N-((3R,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000321
将(3R,4S)-4-氨基-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(400mg,0.9mmol),2,6-二氟苯甲酸(163mg,1.0mmol),DIEA(553mg,4.3mmol),HATU(392mg,1.0mmol)和DMF(5mL),25℃反应2h,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品115.4mg,m/z=570[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.71(s,1H),8.57-8.69(m,3H),8.01-8.04(m,1H),7.50-7.62(m,1H),7.34-7.35(m,1H),7.12-7.17(m,1H),7.06-7.08(m,3H),6.35-6.62(m,1H),5.00-5.00(m,1H),4.47-4.56(m,2H),4.20-4.20(m,3H),3.94-3.94(m,1H),3.36-3.44(m,1H),3.24-3.29(m,1H),1.93-1.96(m,1H),1.71-1.72(m,1H)。
实施例122:化合物269的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯磺酰胺
Figure PCTCN2022073467-appb-000322
反应瓶加入(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),2,6-二氟苯甲酰氯(43mg,0.2mmol),DIEA(129mg,1.0mmol)和DCM(10mL),25℃反应1h,反应完全。倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品15.5mg,m/z=570.2[M+1]+。 1HNMR(400MHz,DMSO-d 6):δ12.64(brs,1H),8.54-8.50(m,2H),8.02-7.96(m,2H),7.70(t,J=6.4Hz,1H),7.56(s,1H),7.36-7.22(m,3H),7.00(d,J=9.2Hz,1H),4.92(s,1H),4.28-4.08(m,4H),3.72-3.70(m,1H),3.68-3.59(m,1H),3.45-3.41(m,1H),3.20-3.14(m,1H),1.90-1.85(m,1H),1.52-1.48(m,1H),1.39(t,J=6.8Hz,3H)。
实施例123:化合物273的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,5-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000323
向反应瓶中依次加入2,5-二氟苯甲酸(158mg,1.0mmol)和二氯甲烷(3mL),冰浴冷却至5℃,依次滴加草酰氯(139mg,1.1mmol)和催化量的DMF(4mg,0.05mmol)。加毕,反应液室温搅拌2小时。反应液浓缩后用DMF(5mL)稀释后,取1mL溶液于冰浴下,滴加到(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol)和二异丙基乙基胺(129mg,1.0mmol)的DMF(4mL)溶液中。反应液室温搅拌反应2小时。反应液经LCMS监测基本反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品45mg,m/z=534[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.56(s,1H),8.55(s,1H),7.99-8.07(m,2H),7.59(s,1H),7.49-7.53(m,1H),7.36-7.42(m,2H),7.25(s,1H),7.07(d,J=9.2Hz,1H),5.11(d,J=4.4Hz,1H),4.15-4.38(m,5H),3.90-3.99(m,1H),3.28-3.35(m,1H),3.11-3.21(m,1H),1.89-2.01(m,1H),1.67-1.79(m,1H),1.36-1.48(m,3H)。
实施例124:化合物274的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,3,6-三氟苯甲酰胺
Figure PCTCN2022073467-appb-000324
向反应瓶中依次加入2,3,6-三氟苯甲酸(176mg,1.0mmol)和二氯甲烷(3mL),冰浴冷却至5℃,然后依次滴加草酰氯(139mg,1.1mmol)和催化量的DMF(4mg,0.05mmol)。加毕,反应液室温搅拌2小时。反应液浓缩后,用DMF(5mL)稀释后,取1mL溶液于冰浴下,滴加到(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol)和二异丙基乙基胺(129mg,1.0mmol)的DMF(4mL)溶液中。反应液室温搅拌反应2小时。反应液经LCMS监测基本反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品35mg,m/z=552[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.75(d,J=8.0Hz,1H),8.56(s,1H),8.51(s,1H),8.02(d,J=8.0Hz,1H),7.50-7.61(m,2H),7.15-7.26(m,2H),7.07(d,J=8.0Hz,1H),5.11(d,J=4.4Hz,1H),4.15-4.38(m,5H),3.90-3.99(m,1H),3.21-3.41(m,2H),1.89-2.01(m,1H),1.67-1.79(m,1H),1.38-1.42(m,3H)。
实施例125:化合物275的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000325
向反应瓶中依次加入(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),DMF(2mL)和二异丙基乙基胺(129mg,1.0mmol)。冰浴冷却至5℃,滴加2-氯-6-氟苯甲酰氯(39mg,0.2mmol)的DMF(0.2mL)溶液,室温搅拌反应2小时。反应液经LCMS监测基本反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品47mg,m/z=550[M+1] +1H-NMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.60(d,J=8.0Hz,1H),8.56(s,1H),8.51(s,1H),8.60(d,J=8.0Hz,1H),7.58(s,1H),7.45-7.48(m,1H),7.36(d,J=8.0Hz,1H),7.21-7.28(m,2H),7.07(d,J=9.2Hz,1H),5.11(d,J=4.4Hz,1H),4.15-4.38(m,5H),3.90-3.99(m,1H),3.21-3.35(m,1H),3.31-3.47(m,1H),1.89-2.01(m,1H),1.67-1.79(m,1H),1.36-1.48(m,3H)。
实施例126:化合物276的制备
2-氯-N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-5-氟苯甲酰胺
Figure PCTCN2022073467-appb-000326
将(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑并[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.25mmol),5-氯-2-氟苯甲酰氯(52mg,0.25mmol),DIEA(194mg,1.80mmol),DMF(3mL)加入反应瓶,25℃反应1h。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品18mg,m/z=550[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.55(s,1H),8.51(s,1H),8.33(d,J=7.6Hz,1H),8.00(d,J=8.8Hz,1H),7.58-7.53(m,2H),7.42-7.30(m,2H),7.25(s,1H),7.06(d,J=8.0Hz,1H),4.96(s,1H),4.33-4.13(m,5H),3.97-3.93(m,1H),3.42-3.40(m,1H),3.24-3.18(m,1H),1.99-1.89(m,1H),1.75-1.66(m,1H),1.39(t,J=6.8Hz,3H)。
实施例127:化合物277的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)-吡啶-2–基)-3-羟基哌啶-4-基)-5-氟-2-甲基苯甲酰胺
Figure PCTCN2022073467-appb-000327
将(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.25mmol),5-氟-2-甲基苯甲酰氯(44mg,0.25mmol),DIEA(194mg,1.80mmol),DMF(3mL)加入反应瓶,25℃反应1h。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品30mg,m/z=530[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.55(s,1H),8.51(s,1H),8.09(d,J=7.6Hz,1H),7.99(d,J=8.8Hz,1H),7.58(s,1H),7.30-7.15(m,4H),7.06(d,J=9.2Hz,1H),4.95(s,1H),4.34-4.14(m,5H),3.97-3.93(m,1H),3.42-3.40(m,1H),3.24-3.18(m,1H),2.33(s,3H),1.99-1.90(m,1H),1.75-1.66(m,1H),1.39(t,J=6.8Hz,3H)。
实施例128:化合物278的制备
2-氯-N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基))吡啶-2-基)-3-羟基哌啶-4-基)-6-甲基苯甲酰胺
Figure PCTCN2022073467-appb-000328
将(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.25mmol),2-氯-6-甲基苯甲酸(42mg,0.25mmol),DIEA(194mg,1.80mmol),HATU(95mg,0.25mmol),DMF(3mL)加入反应瓶,25℃反应1h。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品28mg,m/z=546[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.55(s,1H),8.51(s,1H),8.32(d,J=8.0Hz,1H),7.99(d,J=8.8Hz,1H),7.58(s,1H),7.30-7.20(m,4H),7.05(d,J=9.2Hz,1H),4.89(s,1H),4.24-4.13(m,5H),3.97-3.93(m,1H),3.42-3.40(m,1H),3.24-3.18(m,1H),2.33(s,3H),1.99-1.90(m,1H),1.75-1.68(m,1H),1.39(t,J=6.8Hz,3H)。
实施例129:化合物279的制备
N-((3R,4S)-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2-三氟甲基苯甲酰胺
Figure PCTCN2022073467-appb-000329
反应瓶加入(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),2-三氟甲基苯甲酰氯(41mg,0.2mmol),DIEA(129mg,1.0mmol)和10mL DCM,25℃反应1h,反应完全。倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品28.2mg,淡黄色固体,m/z=566.2[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.56(s,1H),8.51(s,1H),8.27(d,J=8.0Hz,1H),8.01(d,J=8.8Hz,1H),7.79-7.41(m,2H),7.66-7.56(m,3H),7.21(s,1H),7.06(d,J=8.0Hz,1H),4.93(s,1H),4.42-4.12(m,5H),4.97-4.93(m,1H),3.42-3.40(m,1H),3.28-3.19(m,1H),1.98-1.89(m,1H),1.75-1.68(m,1H),1.39(t,J=6.8Hz,3H)。
实施例130:化合物280的制备
2-氰基-N-((3R,4S)-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-苯甲酰胺
Figure PCTCN2022073467-appb-000330
反应瓶加入(3R,4S)-4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),2-氰基苯甲酰氯(33mg,0.2mmol),DIEA(129mg,1.0mmol)和10mL DCM,25℃反应1h,反应完全。倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品12.4mg,淡黄色固体,m/z=523.2[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),10.27(s,1H),8.58-8.51(m,2H),8.20(d,J=6.8Hz,1H),8.01(d,J=8.8Hz,1H),7.85-7.72(m,3H),7.59(s,1H),7.26(s,1H),7.09(d,J=8.4Hz,1H),6.05(s,1H),4.85-4.51(m,2H),4.27-4.15(m,4H),3.22-3.20(m,1H),3.08-2.98(m,2H),1.75-1.68(m,1H),1.39(t,J=6.8Hz,3H)。
实施例131:化合物281的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-1-(三氟甲基)环丁烷-1-甲酰胺
Figure PCTCN2022073467-appb-000331
将(3R,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(120.6mg,0.24mmol),DMF(2mL),DIEA(154.8mg,1.2mmol),1-三氟环丁烷-1-羧酸(33.6mg,0.2mmol),依次加入反应瓶,室温下加入HATU(114mg,0.3mmol),室温反应1h。LCMS监测反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品55mg,m/z=544.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.55-8.50(m,2H),8.01-7.98(m,1H),7.58(s,1H),7.50-7.48(d,1H),7.24-7.23(m,1H),7.07-7.05(m,1H),5.01(s,1H),4.42-4.32(m,2H),4.20-4.15(m,3H),4.01-3.98(m,1H),3.88(s,1H),3.35-3.05(m,2H),2.37-2.34(m,2H),1.96-1.83(m,4H),1.61-1.58(m,1H),1.43-1.38(t,3H)。
实施例132:化合物282的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-1-氟环丙烷-1-甲酰胺
Figure PCTCN2022073467-appb-000332
将(3R,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(120.6mg,0.24mmol),DMF(2mL),DIEA(154.8mg,1.2mmol),1-氟环丙烷-1-羧酸(20.8mg,0.2mmol),依次加入反应瓶,室温下加入HATU(114mg,0.3mmol),室温反应1h。LCMS监测反应完全。将反应液倒入水中,乙酸乙酯萃取,干燥有机相,减压 浓缩,柱层析纯化得到产品6mg,m/z=480.1[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.55-8.50(m,2H),8.01-7.98(m,1H),7.58(s,1H),7.50-7.48(d,1H),7.24-7.23(m,1H),7.07-7.05(m,1H),5.01(s,1H),4.20-4.15(m,3H),3.60-3.58(m,1H),3.37-3.04(m,4H),1.70-1.85(m,2H),1.43-1.38(t,3H),1.00-1.25(m,4H)。
实施例133:化合物283的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,3-二甲基丁酰胺
Figure PCTCN2022073467-appb-000333
将(3R,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(238mg,0.51mmol),2,3-二甲基丁酸(60mg,0.51mmol),DIEA(329mg,2.55mmol),HATU(232mg,0.61mmol),DMF(5mL)加入反应瓶,25℃反应1小时。LCMS监测反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品25mg,m/z=492.3[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.64(brs,1H),8.50-8.54(m,2H),7.98-8.01(m,1H),7.58(s,1H),7.48-7.49(m,1H),7.24(m,1H),7.00-7.02(m,1H),4.87-4.93(m,1H),4.16-4.18(m,4H),3.80-3.94(m,2H),3.10-3.20(m,1H),2.00-2.10(m,1H),1.50-1.90(m,3H),1.40(t,J=6.8Hz,3H),0.84-0.98(m,10H)。
实施例134:化合物284的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-3-(三氟甲基)吡啶甲酰胺
Figure PCTCN2022073467-appb-000334
将(3R,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(150mg,0.30mmol),DMF(2mL),DIEA(154.8mg,1.2mmol),3-三氟甲基-2-羧酸吡啶(38mg,0.2mmol),依次加入反应瓶,室温下加入HATU(114mg,0.3mmol),室温反应1h。LCMS监测反应完全。将反应液倒入水中,乙酸乙酯萃取,干燥有机相,减压浓缩,柱层析纯化得到产品6mg,m/z=567[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.65(s,1H),8.55-8.50(m,3H),8.01-7.98(m,2H),7.58(s,1H),7.50-7.48(d,2H),7.24-7.23(m,1H),7.07-7.05(m,1H),5.01(s,1H),4.20-4.15(m,3H),4.01-3.98(m,1H),3.88(s,1H),3.35-3.05(m,2H),2.37-2.34(m,2H),1.61-1.58(m,1H),1.43-1.38(t,3H)。
实施例135:化合物285的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-羟基哌啶-4-基)-3-氯-5-氟吡啶甲酰胺
Figure PCTCN2022073467-appb-000335
将(3R,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),3-氯-5-氟吡啶甲酸(32mg,0.2mmol),DIEA(129mg,1.0mmol),HATU(76mg,0.2mmol),DMF(3mL)加入反应瓶,25℃反应1小时。LCMS监测反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品41.6mg,m/z=551[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.63(s,1H),8.55(s,1H),8.51-8.49(m,2H),8.32(d,J=8.0Hz,1H),8.00(d,J=8.8Hz,1H),7.92-7.87(m,1H),7.73-7.69(m,1H),7.60(s,1H),7.25(s,1H),7.07(d,J=9.2Hz,1H),5.25(s,1H),4.45-4.36(m,2H),4.20-4.14(m,3H),3.95-3.93(m,1H),3.33-3.30(m,1H),3.15-3.10(m,1H),1.95-1.92(m,1H),1.78-1.74(m,1H),1.38(t,J=6.8Hz,3H)。
实施例136:化合物286的制备
N-((3R,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-羟基哌啶-4-基)-3-氟吡啶甲酰胺
Figure PCTCN2022073467-appb-000336
将(3R,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(100mg,0.2mmol),3-氟吡啶甲酸(32mg,0.2mmol),DIEA(129mg,1.0mmol),HATU(76mg,0.2mmol),DMF(3mL)加入反应瓶,25℃反应1小时。LCMS监测反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品39.9mg,m/z=517[M+1] +1HNMR(400MHz,DMSO-d 6)δ12.65(brs,1H),8.63(s,1H),8.55(s,1H),8.51(s,1H),8.35(d,J=8.0Hz,1H),8.20(d,J=8.8Hz,1H),8.00(d,J=8.8Hz,1H),7.60(s,1H),7.24(s,1H),7.07(d,J=9.2Hz,1H),5.14(s,1H),4.34-4.27(m,2H),4.20-4.15(m,2H),3.95-3.93(m,1H),3.42-3.40(m,1H),3.20-3.15(m,1H),1.99-1.90(m,1H),1.78-1.70(m,1H),1.39(t,J=6.8Hz,3H)。
实施例137:化合物291的制备
N-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)哌啶-4-基)-2,5-二氟苯甲酰胺
步骤A:4-(2,5-二氟苯甲酰胺)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-甲酸乙酯
Figure PCTCN2022073467-appb-000337
将乙基4-氨基-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-甲酸乙酯(120mg,0.26mmol),2,5-二氟苯甲酸(38mg,0.24mmol),DIEA(100mg,0.78mmol),HATU(108mg,0.28mmol),DMF(2mL)加入反应瓶,25℃反应12h,LCMS监测反应完全。加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品100mg,m/z=590[M+1] +
步骤B:N-(1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-4-(羟甲基)哌啶-4-基)-2,5-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000338
将4-(2,5-二氟苯甲酰胺)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-4-甲酸乙酯(30mg,0.05mmol),2mL THF加入反应瓶,0℃下加入LiAlH 4(1.9mg,0.05mmol),反应1h。LCMS监测反应完全,加水搅拌淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,减压浓缩,柱层析分离得到产品20mg,m/z=548[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.61(s,1H),8.55(d,J=2.4Hz,1H),8.48(d,J=2.4Hz,1H),8.32(s,1H),8.04(dd,J=8.8,2.6Hz,1H),7.60(s,1H),7.50(s,1H),7.27(d,J=2.4Hz,1H),7.25-7.18(m,3H),4.85(t,J=5.6Hz,1H),4.20(d,J=13.4Hz,2H),4.15(q,J=7.6Hz,2H),3.58(d,J=5.6Hz,2H),3.15(t,J=12.4Hz,2H),2.20(d,J=13.4Hz,2H),1.68(td,J=13.4,4.4Hz,2H),1.38(t,J=6.8Hz,3H)。
实施例138:化合物292的制备
N-((3R,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-羟基哌啶-4-基)-2-氯-6-氟苯甲酰胺
Figure PCTCN2022073467-appb-000339
将(3R,4S)-4-氨基-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇盐酸盐(100mg,0.2mmol),DCM(3mL),DIEA(138mg,1.0mmol),2-氯-6-氟苯甲酰氯(41mg,0.2mmol)加入反应瓶,0℃反应15min,LCMS监测反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品43.5mg,m/z=586[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.70(s,1H),8.57-8.69(m,3H),8.01-8.02(m,1H),7.61-7.61(m,1H),7.45-7.47(m,1H),7.28-7.37(m,3H),7.06-7.06(m,1H),6.34-6.62(m,1H),4.94-4.95(m,1H),4.48-4.52(m,2H),4.19-4.20(m,3H),3.93-3.93(m,1H),3.42-3.45(m,1H),3.27-3.34(m,1H),1.94-1.97(m,1H),1.70-7.73(m,1H)。
实施例139:化合物293的制备
N-((3R,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-羟基哌啶-4-基)-2-氯-5-氟苯甲酰胺
Figure PCTCN2022073467-appb-000340
将(3R,4S)-4-氨基-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)哌啶-3-醇盐酸盐(100mg,0.2mmol),DCM(3mL),DIEA(138mg,1.0mmol),2-氯-5-氟苯甲酰氯(41mg,0.2mmol)加入反应瓶,0℃反应15min,LCMS监测反应完全,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品11.0mg,m/z=586[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.70(s,1H),8.68-8.69(m,1H),8.57-8.58(m,1H),8.34-8.36(m,1H),8.01-8.04(m,1H),7.53-7.61(m,1H),7.39-7.41(m,1H),7.33-7.35(m,3H),7.07-7.09(m,1H),6.35-6.62(m,1H),4.97-4.98(m,1H),4.47-4.56(m,2H),4.14-4.25(m,3H),3.96-3.96(m,1H),3.40-3.40(m,1H),3.22-3.37(m,1H),1.94-1.94(m,1H),1.67-1.71(m,1H)。
实施例140:化合物294的制备
N-((3S,4S)-1-(5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000341
将(3S,4S)-4-氨基-1-5-(6-乙氧基-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶盐酸盐(200mg,0.51mmol),DMF(2mL),DIEA(197mg,1.5mmol),2,6-二氟苯甲酸(88.4mg,0.56mmol),依次加入反应瓶,室温下加入HATU(290mg,0.76mmol),室温反应1h。LCMS监测反应完全。将反应液倒入水中,乙酸乙酯萃取,干燥有机相,减压浓缩,柱层析纯化得到产品7mg,m/z=534[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.64(s,1H),8.61-8.50(m,3H),8.02-7.99(m,1H),7.60(s,1H),7.52-7.47(m,1H),7.24(s,1H),7.17-7.12(m,2H),7.07-7.05(d,1H),4.97(s,1H),4.24-4.15(m,5H),3.93(s,1H),3.44-3.43(d,J=1.6Hz,1H),3.28-3.26(d,1H),1.95-1.93(m,1H),1.71-1.68(m,1H),1.43(t,3H)。
实施例141:化合物295的制备
N-((3S,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
步骤A:((3S,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000342
反应瓶中加入6-(2-氟乙氧基)-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(410mg,1.3mmol),((3S,4S)-3-羟基哌啶-4-基)碳酸叔丁酯(309mg,1.4mmol),DIEA(840mg,6.5mmol)和DMSO(5mL),90℃反应40h。反应液冷却到室温,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品430mg,m/z=512[M+1] +
步骤B:((3S,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基盐酸盐
Figure PCTCN2022073467-appb-000343
反应瓶中加入((3S,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯(430mg,0.8mmol),4N盐酸-二氧六环溶液(8mL),25℃反应0.5h,反应完全,减压浓缩干,得到产品320mg,m/z=412[M+1] +
步骤C:N-((3S,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000344
将((3S,4S)-1-(5-(6-(2-氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基盐酸盐(320mg,0.8mmol),2,6-二氟苯甲酸(110mg,0.8mmol),DIEA(502mg,4.0mmol),HATU(325mg,0.9mmol)和DMF(5mL),25℃反应2h,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品144.2mg,m/z=552[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.68(s,1H),8.60-9.00(m,3H),8.05-8.07(m,1H),7.61(m,1H),7.50-7.50(m,1H),7.32-7.33(m,1H),7.09-7.16(m,3H),5.11-5.11(m,1H),4.76-4.88(m,1H),4.76-4.46(m,1H),4.38-4.39(m,3H),4.24-4.27(m,1H),3.94-3.95(m,1H),3.49-3.50(m,1H),3.16-3.36(m,1H),2.91-2.97(m,1H),2.04-2.07(m,1H),1.45-1.48(m,1H)。
实施例142:化合物296的制备
N-((3S,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
步骤A:((3S,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯
Figure PCTCN2022073467-appb-000345
反应瓶中加入6-(2,2-二氟乙氧基)-4-(6-氟吡啶-3-基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶(600mg,1.8mmol),((3S,4S)-3-羟基哌啶-4-基)碳酸叔丁酯(430mg,2.0mmol),DIEA(1160mg,9.0mmol)和DMSO(6mL),90℃反应40h。冷却到室温,倒入水中,二氯甲烷萃取, 干燥有机相,减压浓缩,柱层析纯化得到产品600mg,m/z=530[M+1] +
步骤B:((3S,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基盐酸盐
Figure PCTCN2022073467-appb-000346
反应瓶中加入((3S,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基碳酸叔丁酯(600mg,1.1mmol),4N盐酸-二氧六环溶液(10mL),25℃反应0.5h,反应完全,减压浓缩干,得到产品600mg,m/z=430[M+1] +
步骤C:N-((3S,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)-2,6-二氟苯甲酰胺
Figure PCTCN2022073467-appb-000347
将((3S,4S)-1-(5-(6-(2,2-二氟乙氧基)-1H-吡唑[3',4':3,4]吡唑[1,5-a]吡啶-4-基)吡啶-2-基)-3-羟基哌啶-4-基)氨基盐酸盐(400mg,0.9mmol),2,6-二氟苯甲酸(163mg,1.0mmol),DIEA(553mg,4.3mmol),HATU(392mg,1.0mmol)和DMF(5mL),25℃反应2h,倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩,柱层析纯化得到产品111.2mg,m/z=570[M+1] +1H NMR(400MHz,DMSO-d 6)δ12.71(s,1H),8.60-8.70(m,3H),8.05-8.08(m,1H),7.61-7.61(m,1H),7.50-7.50(m,1H),7.36-7.37(m,1H),7.11-7.18(m,3H),6.34-6.62(m,1H),5.11-5.12(m,1H),4.48-4.52(m,3H),4.24-4.27(m,1H),3.94-3.95(m,1H),3.48-3.51(m,1H),3.13-3.18(m,1H),2.94-2.97(m,1H),2.04-2.08(m,1H),1.45-1.47(m,1H)。
实施例143:化合物297的制备
6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶
步骤A:2-(5-溴-3-氯吡啶-2-基)-2-氰基乙酸乙酯
Figure PCTCN2022073467-appb-000348
反应瓶中加入5-溴-2,3-二氯吡啶(45.4g,0.2mol),氰基乙酸乙酯(45.2g,0.4mol),碳酸铯(195g,0.6mol),DMSO(500mL),90℃反应15h,LCMS监控反应完全。将反应液倒入水中,二氯甲烷萃取,干燥有机相,减压浓缩得到产品50g直接用于下步反应,m/z=303[M+1] +
步骤B:2-(5-溴-3-氯吡啶-2-基)乙腈
Figure PCTCN2022073467-appb-000349
反应瓶中加入2-(5-溴-3-氯吡啶-2-基)-2-氰基乙酸乙酯(60g,0.2mol),氯化钠(3.5g,0.06mol),DMSO(500mL),160℃反应2h,LCMS监控反应完全。将反应液倒入水中,二氯 甲烷萃取,干燥有机相,减压浓缩得到产品40g直接用于下步反应,m/z=231[M+1] +
步骤C:2,4,6-三甲基苯磺酸1-氨基-5-溴-3-氯-2-(氰甲基)吡啶-1-鎓盐
Figure PCTCN2022073467-appb-000350
零度下,向2-[(氨基氧基)磺酰]-1,3,5-三甲基苯(6.8g,31.7mmol)的二氯甲烷(50mL)溶液中加入2-(5-溴-3-氯吡啶-2-基)乙腈(7.4g,32.0mmol),并在零度下继续搅拌3小时,析出大量白色固体。反应完毕后,零度下向反应体系中加入乙醚(50mL)并搅拌10分钟,减压过滤,乙醚冲洗,真空干燥得到产品15g,无须再次纯化,直接用于下一步反应。m/z=246[M+1] +
步骤D:6-溴-4-氯吡唑[1,5-a]吡啶-2-胺
Figure PCTCN2022073467-appb-000351
零度下,向装有2,4,6-三甲基苯磺酸1-氨基-5-溴-3-氯-2-(氰甲基)吡啶-1-鎓盐(1.0g,2.2mmol)的DMF(30mL)溶液中加入三乙胺(1.1g,11.0mmol),升至室温下搅拌12小时,反应完毕,加食盐水淬灭,用乙酸乙酯萃取,合并有机相,并用食盐水洗涤,减压浓缩,柱层析分离得到产品350mg,m/z=246[M+1] +
步骤E:1-(2-氨基-6-溴-4-氯吡唑[1,5-a]吡啶-3-基)-2-氯乙基-1-酮
Figure PCTCN2022073467-appb-000352
向反应瓶中加入6-溴-4-氯吡唑[1,5-a]吡啶-2-胺(2.5g,10.0mmol),氯乙腈(1.5g,20.0mmol),三氯化硼(12mL,12.0mmol),三氯化铝(2.7g,20.0mmol),氯苯(20mL),120℃反应12小时,加入稀盐酸(1N,30mL),100℃反应1小时,反应完毕,用乙酸乙酯萃取,合并有机相,并用水洗涤,减压浓缩,柱层析分离得到产品1.5g,m/z=322[M+1] +
步骤F:1-(2-氨基-6-溴-4-氯吡唑[1,5-a]吡啶-3-基)-2-氯乙基-1-醇
Figure PCTCN2022073467-appb-000353
反应瓶中加入1-(2-氨基-6-溴-4-氯吡唑[1,5-a]吡啶-3-基)-2-氯乙基-1-酮(2.0g,6.2mmol),二氧六环(20mL),水(2mL),反应体系冷却至0℃,分批次加入硼氢化钠(0.3g,7.9mmol),升至室温,反应5小时。反应完毕,加水淬灭,用乙酸乙酯萃取,合并有机相,并用水洗涤,减压浓缩,柱层析分离得到产品1.0g,m/z=324[M+1] +
步骤G:6-溴-4-氯-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000354
向反应瓶中加入1-(2-氨基-6-溴-4-氯吡唑[1,5-a]吡啶-3-基)-2-氯乙基-1-醇(323mg,1.0mmol),四氢呋喃(10mL),反应体系冷却至0℃,缓慢滴加异丙基氯化镁(1.5mL,3.0mmol),80℃反应1小时,反应完毕,加饱和氯化铵水溶液淬灭,用乙酸乙酯萃取,合并有机相,并用水洗涤,减压浓缩,柱层析分离得到产品178mg,m/z=270[M+1] +
步骤H:4-氯-6-(4,4,5,5-四甲基-1,3,2-二恶硼烷-2-基)-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000355
向6-溴-4-氯-1H-吡咯并[2',3':3,4]吡唑[1,5-a]吡啶(650mg,2.4mmol)中加入联硼酸频那醇酯(736mg,2.9mmol),醋酸钾(705mg,7.2mmol)和1,4-二氧六环(10mL),Pd(dppf)Cl 2(175mg,0.24mmol)氮气置换后,80℃反应12h,过滤,滤液浓缩后柱层析分离得到产品523mg,m/z=318[M+1] +
步骤I:4-氯-1H-吡唑[2',3':3,4]吡唑[1,5-a]吡啶-6-羟基
Figure PCTCN2022073467-appb-000356
向反应瓶中加入4-氯-6-(4,4,5,5-四甲基-1,3,2-二恶硼烷-2-基)-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶(634mg,2.0mmol),四氢呋喃(10mL),然后反应体系冷却至0℃,缓慢滴加双氧水(340mg,10.0mmol),0℃反应1小时,反应完毕,加饱和偏重亚硫酸钠水溶液淬灭,用乙酸乙酯萃取,合并有机相,并用水洗涤,减压浓缩,柱层析分离得到产品500mg,m/z=208[M+1] +
步骤J:4-氯-6-乙氧基-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000357
向反应瓶中加入4-氯-1H-吡唑[2',3':3,4]吡唑[1,5-a]吡啶-6-羟基(416mg,2.0mmol),碘乙烷(374mg,2.4mmol),碳酸钾(552mg,4.0mmol),DMF(5mL),反应体系升温至50℃,反应5小时,反应完毕,加水淬灭,用乙酸乙酯萃取,合并有机相,并用水洗涤,减压浓缩,柱层析分离得到产品400mg,m/z=236[M+1] +
步骤J:6-乙氧基-4-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶
Figure PCTCN2022073467-appb-000358
向反应瓶加入4-氯-6-乙氧基-1H-吡咯[2',3':3,4]吡唑[1,5-a]吡啶(235mg,1.0mmol),6-((6-甲氧基吡啶-3-基)甲基)-3-(5-(4,4,5,5-四甲基-1,3,2-二氧杂环戊烷-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷(422mg,1.0mmol),Xphos(95.2mg,0.2mmol),醋酸钯(22.4mg,0.1mmol),磷酸钾(424mg,2.0mmol),1,4-二氧六环(10mL)和水(2mL),氮气置换后,100℃反应12h,过滤,滤液浓缩后柱层析分离得到产品250mg,m/z=496[M+1] +1H NMR(400MHz,DMSO-d 6)δ10.81(s,1H),8.61(d,J=2.4Hz,1H),8.33(d,J=2.1Hz,1H),8.11-7.99(m,2H),7.69(d,J=8.7Hz,1H),7.01-6.92(m,2H),6.86(d,J=8.9Hz,1H),6.76(d,J=8.5Hz,1H),6.04(d,J=3.2Hz,1H),4.10(q,J=6.9Hz,2H),3.81(m,5H),3.68(s,2H),3.52(s,2H),3.31(s,2H),1.58(s,1H),1.37(t,J=7.0Hz,3H)。
通过上述方法或参考上述方法,共制备得到化合物1-化合物265,其化学结构式和质谱数据汇总于表1。
表1:实施例化合物1-实施例化合物265的化学结构和质谱数据
Figure PCTCN2022073467-appb-000359
Figure PCTCN2022073467-appb-000360
Figure PCTCN2022073467-appb-000361
Figure PCTCN2022073467-appb-000362
Figure PCTCN2022073467-appb-000363
Figure PCTCN2022073467-appb-000364
Figure PCTCN2022073467-appb-000365
Figure PCTCN2022073467-appb-000366
Figure PCTCN2022073467-appb-000367
Figure PCTCN2022073467-appb-000368
Figure PCTCN2022073467-appb-000369
Figure PCTCN2022073467-appb-000370
Figure PCTCN2022073467-appb-000371
Figure PCTCN2022073467-appb-000372
Figure PCTCN2022073467-appb-000373
Figure PCTCN2022073467-appb-000374
Figure PCTCN2022073467-appb-000375
Figure PCTCN2022073467-appb-000376
Figure PCTCN2022073467-appb-000377
Figure PCTCN2022073467-appb-000378
Figure PCTCN2022073467-appb-000379
Figure PCTCN2022073467-appb-000380
Figure PCTCN2022073467-appb-000381
Figure PCTCN2022073467-appb-000382
Figure PCTCN2022073467-appb-000383
Figure PCTCN2022073467-appb-000384
Figure PCTCN2022073467-appb-000385
Figure PCTCN2022073467-appb-000386
Figure PCTCN2022073467-appb-000387
Figure PCTCN2022073467-appb-000388
Figure PCTCN2022073467-appb-000389
Figure PCTCN2022073467-appb-000390
Figure PCTCN2022073467-appb-000391
Figure PCTCN2022073467-appb-000392
Figure PCTCN2022073467-appb-000393
Figure PCTCN2022073467-appb-000394
Figure PCTCN2022073467-appb-000395
Figure PCTCN2022073467-appb-000396
Figure PCTCN2022073467-appb-000397
Figure PCTCN2022073467-appb-000398
Figure PCTCN2022073467-appb-000399
Figure PCTCN2022073467-appb-000400
Figure PCTCN2022073467-appb-000401
Figure PCTCN2022073467-appb-000402
Figure PCTCN2022073467-appb-000403
Figure PCTCN2022073467-appb-000404
Figure PCTCN2022073467-appb-000405
Figure PCTCN2022073467-appb-000406
Figure PCTCN2022073467-appb-000407
生物活性测试例
测试例1
转染重组基因(RET)是一个已经确认的原癌基因。它编码的单次跨膜受体酪氨酸激酶,是许多组织和细胞类型的发育,成熟和维持所必需的。在正常条件下,神经胶质细胞系衍生的神经营养因子(GDNF)家族配体与细胞表面上的RET的结合导致细胞内酪氨酸残基的二聚化和自磷酸化。这反过来导致下游RAS-MAPK,PI3K-AKT和磷脂酶Cγ(PLCγ)通路的激活,并增加细胞存活和增殖。激活RET突变的实例包括C634W,M918T和关守突变,V804L、V804M、G810R。
该试验将肽底物和单一专有单克隆抗体与HTRF技术相结合,HTRF技术是一种高灵敏度和稳定的技术,用于检测蛋白质的分子相互作用。酶将底物磷酸化,然后Eu标记的抗体结合磷酸化底物,链霉抗生物素蛋白-XL665结合所有底物。TR-FRET信号由HTRF原理产生。一旦添加抑制剂(受试化合物),就获得较弱的TR-FRET信号。据此,评估抑制效果。
激酶活性测试试剂和耗材
Figure PCTCN2022073467-appb-000408
Figure PCTCN2022073467-appb-000409
1.2配制溶液
将所有的化合物都溶解在DMSO中的配制成10mM的储液。
用DMSO将卡博替尼(Cabozantinib)从10mM和1mM分别梯度稀释3倍,共10个浓度。
其他化合物从10mM的原液用DMSO梯度稀释3倍,共10个浓度。
制备1000×阳性对照(1mM Cabozantinib和0.2mM Staurosporine)和1000×阴性对照(100%DMSO)。
在平板振荡器上振荡5分钟。
1.3制备1x激酶缓冲液
将4体积蒸馏水加入1体积酶缓冲液5X;5mM MgCl 2;1mM DTT。
1.4筛选方法
a)将10nl化合物稀释液转移到测试板的每个孔中;
b)在1000g下将化合物板离心1分钟。
c)密封测试板。
d)制备1x激酶缓冲液中的5X Ret-wt(0.2ng/μl)和5X Ret V804M(0.5ng/μl)和2.5X RET G810R(2.5ng/μl)。
e)加入2μl的5X Ret-wt或2μl的Ret V804L或2μl的RET G810R至384孔测试板。
f)将4μl的1x激酶缓冲液加入测试板的每个孔中,在1000g下离心样品板30秒,在室温放置10分钟。
g)制备激酶缓冲液中的5x TK-底物-生物素(5μM)和激酶缓冲液中的5x ATP(50μM)的溶液。
h)通过加入2μl STK-底物-生物素和2μlATP(步骤g中制备的)开始反应。
i)在1000g下离心样品板30秒。密封测试板,室温放置30分钟。
j)制备在HTRF检测缓冲液中的4X Sa-XL 665(250nM)。
k)将5μl的Sa-XL 665和5μl的TK-antibody-Cryptate(在步骤i中制备的)加入到测试板的每个孔中。
l)在1000g下离心30秒,室温放置1小时。
m)在Envision 2104读板仪或BioTek酶标仪上读取615nm/620nm(G810R)(Cryptate)和665nm(XL665)的荧光信号值。
1.5数据分析
计算每个孔的比率(665nm/615nm)。
抑制率%通过如下方式计算:
抑制率%=[1-(受试化合物荧光信号值—阳性对照荧光信号值)/(阴性对照平均比率—阳性对照平均比率)]*100%
比率:由测得的荧光信号值产生
阳性对照平均比率为样品板中阳性对照(20μM Cabozantinib)的平均比率;
阴性对照平均比率为样品板中阴性对照(0.1%DMSO)的平均比率。
利用一些非线性拟合公式来得到化合物的IC50(半数抑制浓度):用GraphPad 6.0软件进行数据分析。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*Hill Slope))
X:化合物浓度Log值Y:抑制率(%inhibition)
Z’因子技术方程:
Z’=1-3(SDmin+SDmax)/(AVEmax-AVEmin)
Min为阳性对照药20μM Cabozantinib Ratio(665/620nM*10000)至,Max为阴性对照药DMSO Ration(665/620nM*10000)。
SD为标准误差,AVE为Ration(665/620nM*10000)平均值。
实施例化合物的激酶结果如表2、表3和表4所示:
表2
Figure PCTCN2022073467-appb-000410
表3
Figure PCTCN2022073467-appb-000411
Figure PCTCN2022073467-appb-000412
表4
Figure PCTCN2022073467-appb-000413
Figure PCTCN2022073467-appb-000414
Figure PCTCN2022073467-appb-000415
Figure PCTCN2022073467-appb-000416
上述激酶和细胞测试结果表明,本发明化合物,尤其是上述实施例化合物对于RET-wt、RET V804M和RET G810R均具有优异的抑制活性。其中,化合物3,化合物4,化合物11,化合物39,化合物40,化合物41,化合物42,化合物44,化合物46和化合物88对RET-wt和RET V804M的抑制活性均显著优于Cabozantinib(卡博替尼)。化合物3,化合物4,化合物11,化合物12,化合物15,化合物17,化合物31,化合物33,化合物34,化合物39,化合物40,化合物41,化合物42,化合物43,化合物44,化合物46,化合物48,化合物51,化合物55,化合物59,化合物88,化合物89,化合物91,化合物101,化合物102,化合物103,化合物104,化合物107,化合物114,化合物118,化合物149,化合物150,化合物152,化合物155,化合物156,化合物157,化合物158,化合物159,化合物160,化合物165,化合物169,化合物170,化合物172,化合物173,化合物174,化合物176,化合物180,化合物183,化合物188,化合物189,化合物193,化合物201,化合物208,化合物220,化合物227,化合物228,化合物229,化合物233,化合物234,化合物235,化合物239,化合物240,化合物242,化合物245,化合物248,化合物249,化合物258,化合物267,化合物268,化合物269,化合物273,化合物274,化合物275,化合物276,化合物277,化合物278,化合物279,化合物280,化合物281,化合物282,化合物283,化合物284,化合物285,化合物286,化合物291,化合物292,化合物293,化合物294,化合物295,化合物296,化合物297对RET G810R的抑制活性还显著优于Cabozantinib(卡博替尼)和Selpercatinib(塞尔帕替尼,LOXO-292)。
尤其是化合物3,化合物4,化合物11,化合物12,化合物15,化合物17,化合物31,化合物33,化合物40,化合物41,化合物42,化合物43,化合物44,化合物46,化合物48,化合物89,化合物91,化合物102,化合物114,化合物174,化合物233,化合物234, 化合物235,化合物267,化合物268,化合物269,化合物273,化合物274,化合物275,化合物276,化合物277,化合物278,化合物279,化合物280,化合物281,化合物282,化合物291,化合物292,化合物293,化合物295,化合物296,化合物297对RET G810R的抑制活性比Selpercatinib(塞尔帕替尼,LOXO-292)活性高10倍以上。因此本发明的化合物对选择性RET抑制剂如Selpercatinib(塞尔帕替尼,LOXO-292)治疗后产生的溶剂前沿G810耐药突变有显著效果,有望用于治疗选择性RET抑制剂发生耐药后的患者。
上文对本发明示例性的实施方案进行了说明。但是应当理解,本发明的保护范围不拘囿于上述示例性的实施方案。在本发明的精神和原则之内,所作出的任何修改、等同替换、改进等,均应包含在本发明的保护范围之内。

Claims (10)

  1. 一种如下式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐:
    Figure PCTCN2022073467-appb-100001
    其中,Q选自
    Figure PCTCN2022073467-appb-100002
    X 1、X 2、X 3、X 4、X 5、X 6、X 7相同或不同,彼此独立地选自CR 1或N;
    X 8选自CR 1R 1’或NR 1
    其中每一个R 1和R 1’相同或不同,彼此独立地选自H、卤素、CN、OH,无取代或任选被一个、两个或更多个独立选自R a的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    A选自H、卤素、CN、OH,NH 2,无取代或任选被一个、两个或更多个独立选自R b的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    D、E相同或不同,彼此独立地选自H、卤素、CN、OH、B(OH) 2、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7、-O-R 21、C(O)OR 22、-P(O)R 23R 24,无取代或任选被一个、两个或更多个独立选自R c的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、NH 2,条件是D和E中的至少一个不为H,例如D和E中的至少一个选自-O-R 21或D和E中的至少一个选自无取代或任选被一个、两个或更多个独立选自R c的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、NH 2
    R 21、R 22、R 23、R 24相同或不同,彼此独立地选自H,无取代或任选被一个、两个或更多个独立选自R d的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基;
    G选自卤素、无取代或任选被一个、两个或更多个独立选自R e的取代基取代的下列基团:至少一个杂原子选自N的5-20元杂芳基、至少一个杂原子选自N的3-20元杂环基、C 3-40环烷基-NH-;;
    K选自不存在、H、卤素、CN、OH,无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40 环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    每一个R 2相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、-C(O)R 4、-S(O) 2R 6
    每一个R 3相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、-C(O)R 4、-S(O) 2R 6
    或者,R 2和R 3与所连的N原子一起形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:5-20元杂芳基或3-20元杂环基;
    每一个R 4相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3
    每一个R 5相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基羰基、C 2-40烯基羰基、C 2-40炔基羰基、C 3-40环烷基羰基、C 3-40环烯基羰基、C 3-40环炔基羰基、C 6-20芳基羰基、5-20元杂芳基羰基、3-20元杂环基羰基;
    每一个R 6相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3
    每一个R 7相同或不同,彼此独立地选自H、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基;
    m为0、1、2、3、4、5、6、7或8;
    每一个R 01、R 02、R 03、R 04相同或不同,彼此独立地选自H、卤素、CN、OH、SH、氧代(=O)、NO 2,无取代或任选被一个、两个或更多个独立选自R g的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    每一个R a、R b、R c、R d、R e、R f相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2,无取代或任选被一个、两个或更多个独立选自R g的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、 C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    每一个R g相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2,无取代或任选被一个、两个或更多个独立选自R h的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7;或者,当环状基团(包括但不限于C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、3-20元杂环基等)的不同位置被两个或更多个取代基取代时,所述取代基中的两个也可以与所述环状基团形成桥环,其中所述桥环中除桥头原子之外的桥原子可以包含1、2、3、4或5个选自CH 2、O、NH的二价基团;
    每一个R h相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2、无取代或任选被一个、两个或更多个独立选自R i的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    每一个R i相同或不同,彼此独立地选自卤素、CN、OH、SH、氧代(=O)、NO 2、无取代或任选被一个、两个或更多个R j的取代基取代的下列基团:C 1-40烷基、C 2-40烯基、C 2-40炔基、C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、C 6-20芳基、5-20元杂芳基、3-20元杂环基、C 1-40烷基氧基、C 2-40烯基氧基、C 2-40炔基氧基、C 3-40环烷基氧基、C 3-40环烯基氧基、C 3-40环炔基氧基、C 6-20芳基氧基、5-20元杂芳基氧基、3-20元杂环基氧基、C 6-20芳基C 1-40烷基、5-20元杂芳基C 1-40烷基、3-20元杂环基C 1-40烷基、-NR 2R 3、-C(O)R 4、-OCR 5、-S(O) 2R 6、OS(O) 2R 7
    或者,当环状基团(包括但不限于C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、3-20元杂环基等)的不同位置被两个或更多个取代基取代时,所述取代基中的两个也可以与所述环状基团形成桥环,其中所述桥环中除桥头原子之外的桥原子可以包含1、2、3、4或5个选自CH 2、O、NH的二价基团;
    或者,当一个原子(如碳原子或氮原子)被两个或更多个取代基取代时,所述取代基中的两个也可以与其共同连接的原子形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:无取代或任选被一个、两个或更多个独立选自R f的取代基取代环状基团(包括但不限于无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:C 3-40环烷基、C 3-40环烯基、C 3-40环炔基、5-20元杂芳基、3-20元杂环基等)。
  2. 根据权利要求1所述的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐,其特征在于,X 1、X 2、X 3、X 4、X 5、X 6、X 7相同或不同,彼此独立地选自CR 1或N;例如,X 1,X 2,X 3,X 4、X 5、X 6、X 7中的至少一个为N,;
    X 8选自CR 1R 1’或NR 1
    每一个R 1和R 1’相同或不同,彼此独立地选自H、卤素、CN、OH、C 1-6烷基、C 3-10环烷基、C 1-6烷基氧基;
    A选自H、卤素、CN、OH、C 1-6烷基、C 1-6烷基氧基;
    D、E相同或不同,彼此独立地选自H、卤素、CN、无取代或任选被一个、两个或更多个独立选自R c的取代基取代的NH 2或-O-R 21,条件是D和E中的至少一个不为H,例如D 和E中的至少一个选自-O-R 21或无取代或任选被一个、两个或更多个独立选自R c的取代基取代的NH 2
    R 21选自无取代或任选被一个、两个或更多个被R d的取代基取代的C 1-6烷基;
    R 2和R 3与所连的N原子一起形成无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:5-20元杂芳基或3-20元杂环基,例如5、6或7元杂芳基或3、4、5、6或7元杂环基;
    每一个R 01、R 02、R 03、R 04相同或不同,彼此独立地选自H、C 1-6烷基、C 1-6烷基氧基;
    每一个R a、R b、R c、R d、R e、R f相同或不同,彼此独立地选自卤素、NH 2、CN、OH,无取代或任选被一个、两个或更多个R g的取代基取代的下列基团:C 1-6烷基、C 1-6烷基氧基、C 3-10环烷基、C 3-10环烷基氧基、C 2-6炔基氧基、3-8元杂环基、3-8元杂芳基;
    每一个R g相同或不同,彼此独立地选自OH、卤素或C 3-10环烷基;
    G选自卤素、C 3-10环烷基、C 3-10环烷基-NH-、C 6-14芳基、5-14元杂芳基、3-12元杂环基,例如6-12元具有单环、双环、三环或桥环结构的杂环基,其可包含1、2、3或4个独立选自N、O和S的杂原子,条件是其中至少一个杂原子选自N,例如1、2或3个杂原子选自N;
    K选自-C 1-6烷基-C 3-10环烷基、-C 1-6烷基-C 6-14芳基、-C 1-6烷基-5-14元杂芳基、-C 1-6烷基-3-10元杂环基、-C(O)NH 2、-C(O)-C 3-10环烷基、-C(O)-C 6-14芳基、-C(O)-5-14元杂芳基、-C(O)-3-10元杂环基、-C(O)-C 1-6烷基-C 3-10环烷基、-C(O)-C 1-6烷基-C 6-14芳基、-C(O)-C 1-6烷基-5-14元杂芳基、-C(O)-C 1-6烷基-3-10元杂环基,其中所述C 3-10环烷基、C 6-14芳基、5-14元杂芳基、3-10元杂环基、-C(O)-C 3-10环烷基、-C(O)-C 6-14芳基、-C(O)-5-14元杂芳基、-C(O)-3-10元杂环基、-C(O)-C 1-6烷基-C 3-10环烷基、-C(O)-C 1-6烷基-C 6-14芳基、-C(O)-C 1-6烷基-5-14元杂芳基、-C(O)-C 1-6烷基-3-10元杂环基的环上或非环基团,或-C(O)NH 2还任选被一个、两个或更多个选自OH、卤素、CN、C 1-6烷基、C 1-6烷基氧基的基团取代;其中,所述杂环基可以为吡啶基(例如吡啶-2-基、吡啶-3-基、吡啶-4-基、吡啶-5-基、吡啶-6-基),芳基可以为苯基;
    或者,K选自不存在、H、OH、无取代或任选被一个、两个或更多个独立选自R f取代的下列基团:苯基-C(O)-、苯基-C(O)-NH-、苯基-C(O)-NH-C 1-6烷基-、苯基氧基-C(O)-NH-、苯基烷基-NH-C(O)-、苯基烷基-C(O)-NH-、吡啶基-C(O)-、吡啶基-C(O)-NH-、吡啶基-C(O)-NH-C 1-6烷基-、吡啶基氧基-C(O)-NH-、吡啶基烷基-NH-C(O)-、吡啶基烷基-C(O)-NH-、吡咯烷基-C(O)-NH-、吡咯烷基氧基-C(O)-NH-、C 1-6烷基-C(O)-、C 1-6烷基-C(O)-NH-、C 1-6烷基氧基-C(O)-NH-、C 3-8环烷基-C(O)-NH-、C 3-8环烷基氧基-C(O)-NH-、吡啶基-NH-C(O)-、C 1-6烷基-NH-C(O)-、C 3-8环烷基-NH-C(O)-、吡啶基氧基-、吡啶基烷基氧基-、吡啶基氧基烷基-、苯基氧基-、苯基烷基氧基-、苯基氧基烷基-、C 1-6烷基-S(O) 2-、C 1-6烷基-S(O) 2-NH-、C 1-6烷基-NH-S(O) 2-、吡啶基C 1-6烷基-、吡啶基-S(O) 2-、吡啶基-C 1-6烷基-S(O) 2-、吡啶基-S(O) 2-NH-、吡啶基-NH-S(O) 2-、吡啶基-C 1-6烷基-NH-、苯基C 1-6烷基-、苯基-S(O) 2-、苯基-C 1-6烷基-S(O) 2-、苯基-S(O) 2-NH-、苯基-NH-S(O) 2-、苯基-C 1-6烷基-NH-、C 1-6烷基氧基-C(O)-、
    Figure PCTCN2022073467-appb-100003
  3. 根据权利要求1或2所述的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐,其特征在于,其中X 1、X 2、X 3、X 4、X 5、X 6、X 7相同或不同,彼此独立地选自CH或N;例如,X 1,X 2,X 3,X 4、X 5、X 6、X 7中的至少一个为N;
    X 8选自NR 1
    R 01为甲氧基;
    R 02为H;
    R 03为甲基;
    R 04为H;
    R 1为H;
    A选自H、NH 2、甲基、乙基、丙基、异丙基;
    E选自H、NH 2
    D选自如下基团:卤素、BnO-、H、CN、NH 2、OCH 3、COOH、B(OH) 2
    Figure PCTCN2022073467-appb-100004
    G选自F、无取代或任选被一个、两个或更多个独立选自R e的取代基取代的下列基团:
    Figure PCTCN2022073467-appb-100005
    Figure PCTCN2022073467-appb-100006
    优选地,当基团G被R e取代时,R e可以取代构成基团G的-CH 2-、-CH=上的H,形成例如选自下列的基团:
    Figure PCTCN2022073467-appb-100007
    K选自不存在、H、OH、无取代或任选被一个、两个或更多个独立选自R f的取代基取代的下列基团:苯基-C(O)-、苯基-C(O)-NH-、苯基-C(O)-NH-C 1-6烷基-、苯基氧基-C(O)-NH-、苯基烷基-NH-C(O)-、苯基烷基-C(O)-NH-、吡啶基-C(O)-、吡啶基-C(O)-NH-、吡啶基-C(O)-NH-C 1-6烷基-、吡啶基氧基-C(O)-NH-、吡啶基烷基-NH-C(O)-、吡啶基烷基-C(O)-NH-、吡咯烷基-C(O)-NH-、吡咯烷基氧基-C(O)-NH-、C 1-6烷基-C(O)-、C 2-6烯基-C(O)-、C、C 1-6烷基-C(O)-NH-、C 1-6烷基氧基-C(O)-NH-、C 3-8环烷基-C(O)-NH-、C 3-8环烷基氧基-C(O)-NH-、吡啶基-NH-C(O)-、C 1-6烷基-NH-C(O)-、C 3-8环烷基-NH-C(O)-、吡啶基氧基-、吡啶基烷基氧基-、吡啶基氧基烷基-、苯基氧基-、苯基烷基氧基-、苯基氧基烷基-、C 1-6烷基-S(O) 2-、C 1-6烷基-S(O) 2-NH-、C 1-6烷基-NH-S(O) 2-、吡啶基C 1-6烷基-、吡啶基-S(O) 2-、吡啶基-C 1-6烷基-S(O) 2-、吡啶基-S(O) 2-NH-、吡啶基-NH-S(O) 2-、吡啶基-C 1-6烷基-NH-、苯基C 1-6烷基-、苯基-S(O) 2-、苯基-C 1-6烷基-S(O) 2-、苯基-S(O) 2-NH-、苯基-NH-S(O) 2-、苯基-C 1-6烷基-NH-、C 1-6烷基氧基-C(O)-、
    Figure PCTCN2022073467-appb-100008
    Figure PCTCN2022073467-appb-100009
    优选地,所述式I所示的化合物具有下式II、III、IV或V所示的结构:
    Figure PCTCN2022073467-appb-100010
    其中,X 1、X 2、X 3、X 4、X 5、X 6、X 7、X 8、R 01、R 02、R 03、R 04、A、D、E、G、K、m具有权利要求1-3任一项所述的定义。
  4. 根据权利要求1-3任一项所述的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐,其特征在于,式I化合物选自如下化合物:
    Figure PCTCN2022073467-appb-100011
    Figure PCTCN2022073467-appb-100012
    Figure PCTCN2022073467-appb-100013
    Figure PCTCN2022073467-appb-100014
    Figure PCTCN2022073467-appb-100015
    Figure PCTCN2022073467-appb-100016
    Figure PCTCN2022073467-appb-100017
    Figure PCTCN2022073467-appb-100018
    Figure PCTCN2022073467-appb-100019
    Figure PCTCN2022073467-appb-100020
    Figure PCTCN2022073467-appb-100021
    Figure PCTCN2022073467-appb-100022
    Figure PCTCN2022073467-appb-100023
    Figure PCTCN2022073467-appb-100024
    Figure PCTCN2022073467-appb-100025
    Figure PCTCN2022073467-appb-100026
    Figure PCTCN2022073467-appb-100027
    Figure PCTCN2022073467-appb-100028
    Figure PCTCN2022073467-appb-100029
    Figure PCTCN2022073467-appb-100030
    Figure PCTCN2022073467-appb-100031
    Figure PCTCN2022073467-appb-100032
    Figure PCTCN2022073467-appb-100033
    Figure PCTCN2022073467-appb-100034
    Figure PCTCN2022073467-appb-100035
    Figure PCTCN2022073467-appb-100036
    Figure PCTCN2022073467-appb-100037
    Figure PCTCN2022073467-appb-100038
  5. 权利要求1-4任一项所述式I化合物的制备方法,其特征在于,包括如下步骤:
    Figure PCTCN2022073467-appb-100039
    其中,A、D、E、Q、G、K、X 5、X 6、X 7、X 8具有权利要求1-4任一项所述的定义;L 1选自离去基团;
    或者,所述制备方法包括如下步骤:
    Figure PCTCN2022073467-appb-100040
    其中,A、D、E、Q、G、K、X 5、X 6、X 7、X 8具有权利要求1-4任一项所述的定义;L 2选自离去基团;
    或者,所述制备方法包括如下步骤:
    Figure PCTCN2022073467-appb-100041
    其中,A、D、E、Q、G、K、X 5、X 6、X 7、X 8具有权利要求1-4任一项所述的定义;L 3选自离去基团;
    或者,所述制备方法包括将被保护基团取代的式I化合物在脱去所述保护基团的条件下反应,得到式I化合物。
  6. 式II化合物的制备方法,其中所述制备方法包括如下步骤:式II-1化合物与化合物R 21-L反应得到式II化合物:
    Figure PCTCN2022073467-appb-100042
    其中,A、E、G、K、X 1、X 2、X 3、X 4、X 5、X 6、X 7、X 8、R 21具有权利要求1-4任一项所述的定义;D选自-O-R 21;L选自离去基团;L选自离去基团;
    优选地,式II-1化合物的制备方法包括由式II-2化合物反应制备式II-1化合物:
    Figure PCTCN2022073467-appb-100043
    其中,A、D、E、G、K、X 1、X 2、X 3、X 4、X 5、X 6、X 7、X 8具有权利要求1-4任一项所述的定义。
  7. 一种化合物,其选自权利要求5中式I-1、式I-2、式I-3所示的化合物,或权利要求6中式II-1或式II-2所示的化合物。
  8. 权利要求7所述的化合物用于制备权利要求1-4任一项所述式I所示化合物的用途。
  9. 一种药物组合物,其包含选自式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐中的至少一种,例如治疗有效量的选自权利要求1-4任一项所述式I所示的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物或药学上可接受的盐中的至少一种;
    优选地,所述药物组合物还包含一种、两种或多种药学上可接受的辅料,如载体和/或赋形剂。
  10. 权利要求1-4任一项所述的化合物、其立体异构体、消旋体、互变异构体、同位素标记物、氮氧化物、或药学上可接受的盐中的至少一种在制备药物中的用途,其中所述药物用于治疗RET激酶介导的疾病、抑制RET激酶活性、治疗癌症和/或抑制与特定癌症相关的转移、治疗肠易激综合征(IBS)或与IBS相关的疼痛、向癌症患者提供支持护理、治疗RET相关疾病或病症、逆转或预防对抗癌药物的获得性抗性、延迟和/或预防个体中抗癌药抗药性发展、对抗癌药物发展抗性的可能性增加;
    优选地,所述支持护理包括预防或最小化与治疗(包括化疗治疗)相关的胃肠病症,例如腹泻;
    优选地,所述RET相关疾病或病症选自RET基因和/或RET激酶介导的疾病,其中RET、RET基因或RET激酶选自包括但不限于RET-wt、V804M、V804L、V804E、G810R、G810S、G810C、G810V和S904F的RET基因或RET激酶;
    优选地,所述癌症选自血液学癌症或实体瘤。
PCT/CN2022/073467 2021-02-08 2022-01-24 含氮多环稠环类化合物,其药物组合物、制备方法和用途 WO2022166642A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2023547651A JP2024505711A (ja) 2021-02-08 2022-01-24 窒素含有多環式縮合環系化合物、その医薬組成物、製造方法及び用途
EP22748921.8A EP4289428A1 (en) 2021-02-08 2022-01-24 Nitrogen-containing polycyclic fused ring compound, pharmaceutical composition thereof, preparation method therefor, and use thereof
KR1020237028014A KR20230136766A (ko) 2021-02-08 2022-01-24 질소 함유 다환 축합 고리계 화합물, 이의 약학 조성물,제조 방법 및 용도

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110172167 2021-02-08
CN202110172167.6 2021-02-08

Publications (1)

Publication Number Publication Date
WO2022166642A1 true WO2022166642A1 (zh) 2022-08-11

Family

ID=82740872

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/073467 WO2022166642A1 (zh) 2021-02-08 2022-01-24 含氮多环稠环类化合物,其药物组合物、制备方法和用途

Country Status (6)

Country Link
EP (1) EP4289428A1 (zh)
JP (1) JP2024505711A (zh)
KR (1) KR20230136766A (zh)
CN (1) CN114907338B (zh)
TW (1) TWI818424B (zh)
WO (1) WO2022166642A1 (zh)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021023209A1 (zh) * 2019-08-05 2021-02-11 北京志健金瑞生物医药科技有限公司 含氮多环稠环类化合物,其药物组合物、制备方法和用途
TWI818424B (zh) * 2021-02-08 2023-10-11 大陸商北京志健金瑞生物醫藥科技有限公司 含氮多環稠環類化合物,其藥物組合物、製備方法和用途

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090042876A1 (en) * 2005-07-01 2009-02-12 Pierre Fabre Medicament Pyrroloquinoline Derivatives And Their Use As Protein Kinases Inhibitors
CN108349969A (zh) * 2015-07-16 2018-07-31 阵列生物制药公司 作为RET激酶抑制剂的取代的吡唑并[1,5-a]吡啶化合物
CN110267960A (zh) * 2017-01-18 2019-09-20 阿雷生物药品公司 作为RET激酶抑制剂的取代的吡唑并[1,5-a]吡嗪化合物
CN110382494A (zh) * 2016-10-10 2019-10-25 阿雷生物药品公司 经取代的吡唑并[1,5-a]吡啶化合物作为ret激酶抑制剂
WO2020064009A1 (zh) * 2018-09-30 2020-04-02 北京志健金瑞生物医药科技有限公司 取代吡唑稠环类衍生物及其制备方法和应用
CN111285874A (zh) * 2018-12-07 2020-06-16 广东东阳光药业有限公司 Ret抑制剂、其药物组合物及其用途
WO2020228756A1 (zh) * 2019-05-14 2020-11-19 上海翰森生物医药科技有限公司 含二并环类衍生物抑制剂、其制备方法和应用
WO2020233641A1 (zh) * 2019-05-20 2020-11-26 浙江同源康医药股份有限公司 用作ret激酶抑制剂的化合物及其应用
WO2021023209A1 (zh) * 2019-08-05 2021-02-11 北京志健金瑞生物医药科技有限公司 含氮多环稠环类化合物,其药物组合物、制备方法和用途

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI818424B (zh) * 2021-02-08 2023-10-11 大陸商北京志健金瑞生物醫藥科技有限公司 含氮多環稠環類化合物,其藥物組合物、製備方法和用途
CN115594678A (zh) * 2021-07-09 2023-01-13 北京志健金瑞生物医药科技有限公司(Cn) 含氮多环稠环类化合物的制备方法及其中间体和用途

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090042876A1 (en) * 2005-07-01 2009-02-12 Pierre Fabre Medicament Pyrroloquinoline Derivatives And Their Use As Protein Kinases Inhibitors
CN108349969A (zh) * 2015-07-16 2018-07-31 阵列生物制药公司 作为RET激酶抑制剂的取代的吡唑并[1,5-a]吡啶化合物
CN110382494A (zh) * 2016-10-10 2019-10-25 阿雷生物药品公司 经取代的吡唑并[1,5-a]吡啶化合物作为ret激酶抑制剂
CN110267960A (zh) * 2017-01-18 2019-09-20 阿雷生物药品公司 作为RET激酶抑制剂的取代的吡唑并[1,5-a]吡嗪化合物
WO2020064009A1 (zh) * 2018-09-30 2020-04-02 北京志健金瑞生物医药科技有限公司 取代吡唑稠环类衍生物及其制备方法和应用
CN111285874A (zh) * 2018-12-07 2020-06-16 广东东阳光药业有限公司 Ret抑制剂、其药物组合物及其用途
WO2020228756A1 (zh) * 2019-05-14 2020-11-19 上海翰森生物医药科技有限公司 含二并环类衍生物抑制剂、其制备方法和应用
WO2020233641A1 (zh) * 2019-05-20 2020-11-26 浙江同源康医药股份有限公司 用作ret激酶抑制剂的化合物及其应用
WO2021023209A1 (zh) * 2019-08-05 2021-02-11 北京志健金瑞生物医药科技有限公司 含氮多环稠环类化合物,其药物组合物、制备方法和用途

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
"RET Solvent Front Mutations Mediate Acquired Resistance to Selective RET Inhibition in RET-Driven Malignancies", JOURNAL OF THORACIC ONCOLOGY, vol. 15, no. 4, 2020, pages 541 - 549
CANCER, vol. 117, no. 12, 2011, pages 2709 - 2718
CANCER,, vol. 119, no. 8, 2013, pages 1486 - 1494
CELL, vol. 60, no. 4, 1990, pages 557 - 563
ENDOCR REV, vol. 27, no. 5, 2006, pages 535 - 560
GENOME RES, vol. 22, no. 3, 2012, pages 436 - 445
MEDULLARY THYROID CARCINOMA (MTC, vol. 19, no. 6, 2009, pages 565 - 612
NAT MED, vol. 18, no. 3, 2012, pages 375 - 377
NATURE REVIEWS CANCER, vol. 14, 2014, pages 173 - 186
PROC NATL ACAD SCI USA, vol. 97, no. 1, 2000, pages 268 - 273
QIAN ET AL., MOL CANCER, vol. 13, 2014, pages 176

Also Published As

Publication number Publication date
TW202237607A (zh) 2022-10-01
KR20230136766A (ko) 2023-09-26
CN114907338B (zh) 2024-02-06
TWI818424B (zh) 2023-10-11
EP4289428A1 (en) 2023-12-13
JP2024505711A (ja) 2024-02-07
CN114907338A (zh) 2022-08-16

Similar Documents

Publication Publication Date Title
WO2020216371A1 (zh) Egfr抑制剂及其应用
CN108513574B (zh) N-磺酰化吡唑并[3,4-b]吡啶-6-甲酰胺和使用方法
CN107857755B (zh) 作为trka激酶抑制剂的n-吡咯烷基、n′-吡唑基-脲、硫脲、胍和氰基胍化合物
CN109970743B (zh) 为jak抑制剂的5-氯-2-二氟甲氧基苯基吡唑并嘧啶化合物
CN110818724B (zh) 吡啶酮和氮杂吡啶酮化合物及使用方法
TWI667233B (zh) 新穎吲唑羧醯胺,其製備方法、含彼等之醫藥製劑及其製造醫藥之用途
TW202016109A (zh) 作為hpk1抑制劑的吡咯並[2,3-b]吡啶或吡咯並[2,3-b]吡嗪及其用途
CN110964008B (zh) 取代吡唑稠环类衍生物及其制备方法和应用
TWI773651B (zh) 醫藥化合物
CN108368116B (zh) Janus激酶抑制剂、其组合物及用途
CN114456176A (zh) 作为tam抑制剂的吡咯并三嗪化合物
TWI726888B (zh) 核受體調節劑
WO2021143680A1 (zh) 杂芳基类衍生物及其制备方法和用途
CN110114343B (zh) 吡唑并嘧啶化合物及其使用方法
TW201130835A (en) Imidazopyridine derivatives as JAK inhibitors
JP2018515612A (ja) 置換キノキサリン誘導体
CN102695710A (zh) 三环吡唑胺衍生物
CN108473478A (zh) TBK/IKKε抑制剂化合物及其用途
WO2022166642A1 (zh) 含氮多环稠环类化合物,其药物组合物、制备方法和用途
WO2020143763A1 (zh) 卤代烯丙基胺类化合物及其应用
WO2021023209A1 (zh) 含氮多环稠环类化合物,其药物组合物、制备方法和用途
WO2022222963A1 (zh) 含咪唑稠环类衍生物、其制备方法及其在医药上的应用
TW202304918A (zh) 經取代之1-芳基-1'-雜芳基化合物、經取代之1,1'-聯雜芳基化合物及其使用方法
WO2021233236A1 (zh) 取代的杂芳基化合物及其组合物和用途
WO2023174406A1 (zh) 含氮杂环类衍生物抑制剂、其制备方法和应用

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: 22748921

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2023547651

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 18264687

Country of ref document: US

ENP Entry into the national phase

Ref document number: 20237028014

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020237028014

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022748921

Country of ref document: EP

Effective date: 20230908