CN115124526B - Pyrrolo pyrimidine compound intermediate and preparation method thereof - Google Patents

Pyrrolo pyrimidine compound intermediate and preparation method thereof Download PDF

Info

Publication number
CN115124526B
CN115124526B CN202210519413.5A CN202210519413A CN115124526B CN 115124526 B CN115124526 B CN 115124526B CN 202210519413 A CN202210519413 A CN 202210519413A CN 115124526 B CN115124526 B CN 115124526B
Authority
CN
China
Prior art keywords
formula
compound shown
reaction
compound
solvent
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN202210519413.5A
Other languages
Chinese (zh)
Other versions
CN115124526A (en
Inventor
李永国
王春娟
李磊
贾国慧
舒诗会
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangzhou Jiayue Pharmaceutical Technology Co ltd
Original Assignee
Guangzhou Jiayue Pharmaceutical Technology Co ltd
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 Guangzhou Jiayue Pharmaceutical Technology Co ltd filed Critical Guangzhou Jiayue Pharmaceutical Technology Co ltd
Priority to CN202210519413.5A priority Critical patent/CN115124526B/en
Publication of CN115124526A publication Critical patent/CN115124526A/en
Application granted granted Critical
Publication of CN115124526B publication Critical patent/CN115124526B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C07D471/02Heterocyclic 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C269/06Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups by reactions not involving the formation of carbamate groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/22Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C41/00Preparation of ethers; Preparation of compounds having groups, groups or groups
    • C07C41/01Preparation of ethers
    • C07C41/16Preparation of ethers by reaction of esters of mineral or organic acids with hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/03Ethers having all ether-oxygen atoms bound to acyclic carbon atoms
    • C07C43/14Unsaturated ethers
    • C07C43/164Unsaturated ethers containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • C07F7/1872Preparation; Treatments not provided for in C07F7/20
    • C07F7/188Preparation; Treatments not provided for in C07F7/20 by reactions involving the formation of Si-O linkages
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • C07F7/1872Preparation; Treatments not provided for in C07F7/20
    • C07F7/1892Preparation; Treatments not provided for in C07F7/20 by reactions not provided for in C07F7/1876 - C07F7/1888
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a pyrrolopyrimidine compound intermediate and a preparation method thereof. The pyrrolopyrimidine compound has inhibiting effect on JAK1, JAK2, JAK3 and TYK2 kinase, has characteristic high permeability, has good oral bioavailability in mice, has higher exposure, and is favorable for generating good in-vivo efficacy and can treat arthritis. In addition, the preparation method has the advantages of simple reaction, high yield, simple operation, mild condition, high safety and wide industrial application prospect.

Description

Pyrrolo pyrimidine compound intermediate and preparation method thereof
The present application is a divisional application of 202011408385.7. Filing date of the original application: 12 months 04 in 2020, application number: 202011408385.7, the invention creation name: pyrrolo pyrimidine compound intermediate and a preparation method thereof.
Technical Field
The invention relates to the technical field of medicines, in particular to a pyrrolopyrimidine compound intermediate serving as a JAK inhibitor and a preparation method thereof.
Background
Janus kinase (JAKs) is a cytoplasmic tyrosine kinase that can transmit cytokine signals. From membrane receptors to STAT transcription factors. JAK families contain four members, JAK1, JAK2, JAK3 and TYK2. The JAK-STAT pathway transmits extracellular signals from a variety of cytokines, growth factors, and hormones to the nucleus and is responsible for the expression of thousands of protein-encoding genes. The JAK-STAT pathway converts extracellular signals into transcriptional responses involving several steps 1) when cytokine receptors on the cell surface change configuration upon binding to their respective cytokine ligands, resulting in dimerization of the receptor molecules, which brings the receptor-coupled JAK kinases into proximity and activation by interactive tyrosine phosphorylation. 2) Tyrosine residues on the catalytic receptor undergo phosphorylation modification upon JAK activation, and these phosphorylated tyrosine sites then form "docking sites" with the surrounding amino acid sequence, with STAT proteins containing the SH2 domain recruited to this "docking site". 3) Finally, kinase JAK catalyzes the phosphorylation modification of STAT proteins bound to the receptor, and activated STAT proteins leave the receptor and form dimers, which are then transferred into the nucleus for transcriptional regulation of specific genes. JAK-STAT intracellular signaling serves interferon, most interleukins, and a variety of cytokines and endocrine factors, such as EPO, TPO, GH, OSM, LIF, CNTF, GM-CSF, and PRL (Vainchenker W.E T al (2008).
JAK-1, JAK-2 and TYK-2 are expressed in various tissue cells of the human body, JAK-3 is mainly expressed in various hematopoietic tissue cells, and is mainly present in bone marrow cells, thymus cells, NK cells, and activated B lymphocytes, T lymphocytes. JAK1 binds to IL-10, IL-19, IL-20, IL-22, IL-26, IL-28, IFN- α, IFN- γ, IL-6 in gp130 family, other receptors containing yc, etc. JAK1 has become a novel target in the field of diseases such as immunity, inflammation, cancer and the like. JAK2 plays an important role in the regulation of various receptor signaling including IL-3, IL-5, gm-CSF in the EPO, GH, PRL, IFN- γ and βc families. One base mutation on the JAK2 gene in humans, JAK2V617F, is closely related to the occurrence of Polycythemia Vera (PV), essential Thrombocythemia (ET), idiopathic Myelofibrosis (IMF), chronic Myelogenous Leukemia (CML), etc. in myeloproliferative diseases. JAK3 regulates cell signaling by binding to the gamma co-chain (yc) in cytokine receptor complexes such as IL-2, IL-4, IL-7, IL-9, IL-15, IL-21. JAK3 or yc mutations can both lead to severe combined immunodeficiency. Abnormal JAK3 activity is manifested by a massive decrease in T cells and NK cells, loss of B cell function, and serious effects on normal biological functions of the immune system and the like. Based on their functional characteristics and specific tissue distribution, JAK3 has become an attractive drug target for immune system related diseases. TYK2 is the 1 st member of the JAK family and is activated by a variety of receptors such as IFNs, IL-10, IL-6, IL-12, IL-23, IL-27, and the like. In mice, a deficiency in TYK2 function causes a defect in the signaling pathway of various cytokine receptors, which in turn leads to viral infection, a decrease in antibacterial immune function, and an increased likelihood of pulmonary infection (John J.O' Shea,2004,Nature Reviews Drug Discovery 3,555-564). Different JAK family members selectively bind to different cytokine receptors, conferring signal transduction specificity, and thus exerting different physiological effects in a manner that allows JAK inhibitors to be applied relatively specifically for disease treatment. For example, IL-2 or IL-4 receptors bind JAK1 and JAK3 along with the common gamma chain, while type I receptors with the same beta chain bind JAK 2. Type I receptors of gp130 (glycoprotein 130) and type I receptors activated by heterodimeric cytokines preferentially bind JAK1/2 and TYK2. Type I receptors activated by hormone-like cytokines bind to and activate JAK2 kinase. Type II receptors for interferon bind to JAK1 and TYK2, while receptors for the IL-10 cytokine family bind to JAK1/2 and TYK2. The specific binding of the cytokines and their receptors to various JAK family members triggers different physiological actions, providing the possibility for the treatment of different diseases. JAK1 was heterodimerized with other JAKs to transduce cytokine-driven pro-inflammatory signaling. Thus, inhibition of JAK1 and/or other JAKs is expected to be of therapeutic benefit for a range of inflammatory disorders and other diseases driven by JAK-mediated signal transduction (Daniella M.Schwartz,2017,Nature Reviews Drug Discovery 16,843-862.)
Disclosure of Invention
The technical problem to be solved by the invention is to overcome the defect of single type of the existing JAK inhibitor, and the preparation method of the pyrrolopyrimidine compound has poor safety and is not beneficial to the mass production. The invention provides a pyrrolopyrimidine compound intermediate and a preparation method thereof, wherein the pyrrolopyrimidine compound has an inhibiting effect on JAK1, JAK2, JAK3 and TYK2 kinase, and the preparation method has the advantages of simple reaction, high yield, simple operation, mild condition, high safety and wide industrial application prospect.
The invention solves the technical problems through the following technical proposal.
The invention provides a compound shown as a formula III,
wherein,
T 1 CH or N;
D 1 is O or CH 2
R 1 H, C of a shape of H, C 1-3 Alkyl, boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, wherein the C 1-3 Alkyl is optionally substituted with 1, 2 or 3R a Substitution;
R 2 is H or C 1-3 Alkyl, wherein the C 1-3 Alkyl is optionally substituted with 1, 2 or 3R b Substitution;
R a 、R b are respectively and independently selected from F, cl, br, I and NH 2
R 4 Selected from H, MOM, bn, THP, tr, ac, bzPiv, TMS, TES, TBS and TBDPS;
but is not
In one embodiment, in the compound shown in formula III, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
T 1 CH.
In one embodiment, in the compound shown in formula III, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
D 1 is CH 2
In one embodiment, in the compound shown in formula III, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
R 1 h or Boc.
In one embodiment, in the compound shown in formula III, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
R 2 h.
In one embodiment, in the compound shown in formula III, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
R 4 is H, bn, TBS or TBDPS.
In one embodiment, the compound of formula III is any one of the following:
the invention also provides a compound shown in the formula IV,
wherein,
R 1 and R is 4 Is defined as above;
R 5 is CH 2 OH, COOEt or COOMe.
In one embodiment, in the compound shown in formula iv, some groups may be defined as follows, and the remaining groups may be defined as described in any one of the embodiments above:
R 1 boc.
In one embodiment, in the compound shown in formula iv, some groups may be defined as follows, and the remaining groups may be defined as described in any one of the embodiments above:
R 4 is H, bn, TBS or TBDPS.
In one embodiment, in the compound shown in formula iv, some groups may be defined as follows, and the remaining groups may be defined as described in any one of the embodiments above:
R 5 is CH 2 OH or COOEt.
In one embodiment, the compound of formula iv is any one of the following:
the invention also provides a compound shown as a formula V,
wherein,
R 1 、R 4 and R is 5 Is defined as above.
In one embodiment, in the compound shown in formula v, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
R 1 boc.
In one embodiment, in the compound shown in formula v, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
R 4 is H, bn, TBS or TBDPS.
In one embodiment, in the compound shown in formula v, some groups may be defined as follows, and the remaining groups may be defined as in any one of the embodiments above:
R 5 is COOEt.
In one embodiment, the compound of formula V is any one of the following:
the invention also provides a compound shown in a formula VI,
R 4 as defined above, bn, TBS or TBDPS are preferred.
The invention also provides a preparation method of the compound shown in the formula III, which is any one of the following schemes:
scheme one:
when the compound shown in the formula III is a compound shown in the formula III-5, the preparation method comprises the following steps:
in a solvent, in the presence of tributylphosphine and azodicarbonyl dipiperidine (ADDP), carrying out a cyclization reaction shown in the following formula IV to obtain a compound shown in the formula III-5;
scheme II:
when the compound shown in the formula III is a compound shown in the formula III-6, the preparation method comprises the following steps:
in a solvent, carrying out deprotection reaction on a compound shown as a formula III-5 in the presence of a deprotection reagent to obtain a compound shown as a formula III-6;
scheme III:
when the compound shown in the formula III is a compound shown in the formula III-7, the preparation method comprises the following steps:
in a solvent, in the presence of alkali and a protective reagent, carrying out amino protection reaction on a compound shown as a formula III-6 to obtain a compound shown as a formula III-7;
In the compounds of formula III-5: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 4 MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS; d (D) 1 Is CH 2 ,T 1 And R is 2 Is defined as above;
in the compounds of formula IV: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 4 For MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS, R 5 Is CH 2 OH;
In the compounds of formula III-6: d (D) 1 Is CH 2 ,T 1 、R 2 And R is 4 Is defined as above;
in the compounds of formula III-7: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, D 1 Is CH 2 ,T 1 、R 2 And R is 4 Is defined as above.
In the cyclization reaction, the solvent may be a solvent conventional in the art, preferably one or more of halogenated hydrocarbon solvents, ether solvents, alkane solvents and aromatic hydrocarbon solvents, more preferably CH 2 Cl 2 、CHCl 3 、CCl 4 One or more of tetrahydrofuran, diethyl ether, petroleum ether, n-hexane, benzene and toluene, most preferably toluene.
In the cyclization reaction, the compound shown in the formula III-5 is preferably Correspondingly, the compounds of formula IV are each +.>
In the cyclization reaction, the molar ratio of tributylphosphine to the compound shown in the formula IV can be 1:1-4:1, preferably 1.5:1-2.5:1, more preferably 2.0:1.
In the cyclization reaction, the molar ratio of the azodicarbonyl dipiperidine to the compound of formula IV may be 1:1 to 3:1, preferably 1.7:1 to 2.5:1, more preferably 2.2:1.
In the cyclization reaction, the concentration of the compound shown in the formula IV in the solvent can be the concentration which is conventional in the art of the cyclization reaction, preferably 0.1mol/L to 1.0mol/L, more preferably 0.12mol/L or 0.14mol/L.
In the cyclization reaction, the reaction temperature may be a temperature conventional in the art for such cyclization reaction, preferably-10 to 50 ℃, more preferably 0-5 ℃ to 10 to 30 ℃.
In the above-mentioned cyclization reaction, the reaction is terminated by the disappearance or absence of the compound represented by the formula IV, and the reaction time is preferably 1 to 30 hours, more preferably 12 to 20 hours.
The cyclization reaction can be performed in an inert atmosphere, and the inert atmosphere can be nitrogen or argon.
The cyclization reaction can be performed by sequentially adding azodicarbonyl dipiperidine and tributylphosphine into a system of the compound shown in the formula IV and the solvent at 0-5 ℃ and performing the cyclization reaction at 10-30 ℃ to obtain the compound shown in the formula III-5.
The cyclization reaction may further comprise a post-treatment after completion, and the post-treatment preferably comprises the following steps: after the cyclization reaction is finished, filtering and concentrating to obtain the compound shown in the formula III-5.
In the deprotection reaction, the compound of formula III-5 is preferably Accordingly, the compound represented by formula III-6 is +.>
In the deprotection reaction, the solvent may be a solvent conventionally used in such deprotection reaction in the art, preferably an alcoholic solvent, more preferably one or more of methanol, ethanol and isopropanol.
In the deprotection reaction, the deprotection reagent may be HCl.
In the deprotection reaction, the mass ratio of the deprotection reagent to the solvent is preferably 1:1 to 1:4, more preferably 1:2.3.
In the deprotection reaction, the concentration of the compound represented by the formula III-5 in the solvent may be a concentration conventional in the art for such deprotection reaction, preferably 1mol/L to 5mol/L, more preferably 2.6mol/L.
In the deprotection reaction, the reaction temperature may be a temperature conventional in the art for such deprotection reaction, preferably-10 to 50 ℃, more preferably 0-5 ℃ to 10 to 30 ℃.
In the deprotection reaction, the deprotection reaction takes the disappearance or no longer reaction of the compound shown as the formula III-5 as a reaction end point, and the reaction time is preferably 2 to 10 hours, more preferably 4 to 8 hours.
The deprotection reaction may further comprise a post-treatment after completion, preferably comprising the steps of: and after the deprotection reaction is finished, absolute ethyl alcohol is added, stirring is carried out, filtering is carried out, and the hydrochloride of the compound shown in the formula III-6 is obtained after concentration.
In the amino-protecting reaction, the compound of formula III-7 is preferablyCorrespondingly, the compound shown as the formula III-6 is +.>
In the amino protection reaction, the solvent can be a solvent which is conventional in the amino protection reaction in the field, preferably an alcohol solvent and/or water, wherein the alcohol solvent is one or more of methanol, ethanol and isopropanol, and the solvent is more preferably methanol and water; for example methanol and water in a volume ratio of 15:49.
In the amino protection reaction, the protecting agent may be Boc anhydride.
In the amino protection reaction, the base may be a base conventionally used in the art for such amino protection reaction, preferably an alkali metal carbonate, more preferably sodium carbonate.
In the amino protection reaction, the molar ratio of the compound shown in the formula III-6 to the base may be a ratio conventional in the art for such amino protection reactions, preferably 1:1 to 1:1.5, more preferably 1:1.1.
In the amino protection reaction, the molar ratio of the compound shown in the formula III-6 to the protecting reagent can be a ratio conventional in the amino protection reaction in the field, preferably 1:1 to 1:1.8, such as 1:1.4.
In the amino-protecting reaction, the concentration of the compound represented by the formula III-6 in the solvent may be a concentration conventional in the art for such amino-protecting reaction, preferably 0.1mol/L to 1.0mol/L, more preferably 0.5mol/L.
In the amino-protecting reaction, the reaction temperature may be a temperature conventional in the art for such amino-protecting reaction, preferably-10 to 50 ℃, more preferably 0-5 ℃ to 10 to 30 ℃.
In the amino-protecting reaction, the amino-protecting reaction takes the compound shown in the formula III-6 as a reaction end point, and the reaction time is preferably 5-20 hours, more preferably 6-10 hours.
The amino protection reaction may further comprise a post-treatment after completion, said post-treatment preferably comprising the steps of: after the amino protection reaction is finished, adding dichloromethane, stirring, standing, layering and concentrating to obtain the compound shown in the formula III-7.
In the preparation method, when the compound shown in the formula IV is a compound shown in the formula IV-4, the preparation method of the compound shown in the formula III-5 further comprises the preparation method of the compound shown in the formula IV-4, which comprises the following steps:
in a solvent, in the presence of a reducing agent and an initiator, carrying out the reduction reaction of the compound shown as the formula IV-3 to obtain a compound shown as the formula IV-4;
in the compounds of formula IV-3: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 4 MOM, bn, THP, tr, ac, bz is a,Piv, TMS, TES, TBS or TBDPS;
in the compounds of formula IV-4: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 4 MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS.
In the reduction reaction, the compound shown in the formula IV-3 is preferably Correspondingly, the compounds shown as the formula IV-4 are respectively
In the reduction reaction, the reducing agent may be sodium borohydride.
In the reduction reaction, the solvent may be a solvent conventionally used in such reduction reaction in the art, preferably one or more of an ether solvent, a halogenated hydrocarbon solvent, an ether solvent and an alcohol solvent, preferably one or more of chloroform, tetrahydrofuran, diethyl ether, methyl tert-butyl ether (MTBE), ethanol, methanol and isopropanol, more preferably methyl tert-butyl ether.
In the reduction reaction, the initiator may be an alcoholic solvent, preferably methanol, ethanol or isopropanol, more preferably methanol.
In the reduction reaction, the molar ratio of the compound represented by the formula IV-3 to the reducing agent may be a ratio conventional in this type of reduction reaction in the art, preferably 1:1.1 to 1:1.8, more preferably 1:1.4 or 1:1.6.
In the reduction reaction, the concentration of the compound represented by the formula IV-3 in the solvent may be a concentration conventional in the art for such reduction reaction, preferably 0.5mol/L to 5mol/L, more preferably 0.8mol/L to 1.1mol/L.
In the reduction reaction, the mass ratio of the reducing agent to the initiator may be in a ratio conventional in this type of reduction reaction in the art, preferably 1:3 to 1:5, more preferably 1:4.
In the reduction reaction, the reaction temperature may be a temperature conventional in the art for such reduction reaction, preferably-10 to 60 ℃, more preferably 0 to 5 ℃ or 10 to 30 ℃.
In the reduction reaction, the reduction reaction is carried out with the compound of formula IV-3 disappearing or no longer reacting as the reaction end point, and the reaction time is preferably 5 to 30 hours, more preferably 4 hours.
The reduction reaction may be carried out in an inert atmosphere, which may be nitrogen or argon.
The reduction reaction can be performed by sequentially adding the reducing agent and the initiator into a system of the compound shown in the formula IV-3 and the solvent at the temperature of 0-5 ℃ and then performing the reduction reaction at the temperature of 10-30 ℃ to obtain the compound shown in the formula IV-4.
The reduction reaction may further comprise a post-treatment after completion, and the post-treatment preferably comprises the following steps: quenching (e.g., sequentially adding water and ammonium chloride), layering, and concentrating the organic phase to obtain the compound shown in formula IV-4.
In the preparation method, when the compound shown in the formula IV is a compound shown in the formula IV-4, the preparation method of the compound shown in the formula III-5 can further comprise the preparation method of the compound shown in the formula IV-3, and the preparation method comprises the following steps:
1) In a solvent, in the presence of alkali, carrying out ring-opening reaction on a compound shown in a formula VI and a compound shown in a formula VII to obtain a compound shown in a formula V;
2) In a solvent, in the presence of hydrazine hydrate, carrying out a cyclization reaction shown in the following formula V to obtain a compound shown in the formula IV-3;
In the compounds of formula VI: r is R 4 MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS;
in the compounds of formula VII: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 5 Is COOEt or COOMe;
in the compounds of formula V: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 4 For MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS, R 5 Is COOEt or COOMe;
in the compounds of formula IV-3: r is R 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB, R 4 For MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS, R 5 Is COOEt or COOMe.
In said step 1), said compound of formula VI is preferably The compound shown in formula VII is preferably +.>
In the step 1), the solvent may be a solvent conventional in the art for such ring-opening reaction, preferably one or more of an alkane solvent, an haloalkane solvent, an ether solvent and an aromatic solvent, more preferably CH 2 Cl 2 、CHCl 3 、CCl 4 One or more of tetrahydrofuran, diethyl ether, petroleum ether, n-hexane, benzene and toluene, most preferably tetrahydrofuran.
In the step 1), the base may be butyllithium.
In said step 1), the molar ratio of said compound of formula VI to said compound of formula VII may be in a ratio conventional in the art for such ring-opening reactions, preferably 1:1 to 2:1, more preferably 1.7:1.
In the step 1), the concentration of the compound of formula VII in the solvent may be a concentration conventional in the art for such ring-opening reaction, preferably 0.1mol/L to 1.0mol/L, more preferably 0.51mol/L.
In the step 1), the molar ratio of the compound represented by formula VII to the base may be 1:3 to 1:8, more preferably 1:5.5.
In said step 1), the reaction temperature may be a temperature conventional in the art for such ring-opening reactions, preferably 35 to 70 ℃, more preferably 40 to 50 ℃.
In the step 1), the ring-opening reaction takes the disappearance or no longer reaction of the compound shown in the formula VII as a reaction end point, and the reaction time is preferably 1 to 5 hours, more preferably 4 hours.
In the step 1), the ring-opening reaction may be performed in an inert atmosphere, and the inert atmosphere may be nitrogen or argon.
The step 1) may further comprise a post-treatment, preferably comprising the steps of: after the ring-opening reaction is finished, adding water, adding acid, adding MTBE and layering to obtain an organic phase containing the compound shown as the formula V, and directly carrying out the reaction in the step 2). The acid is preferably ammonium chloride.
In said step 2), said compound of formula V is preferably Correspondingly, the compound shown as the formula IV-3 is
In said step 2), said solvent may be a solvent conventional in the art for such cyclization reactions, preferably an ether solvent, more preferably MTBE.
The molar ratio of the compound of formula VII to the hydrazine hydrate may be from 1:1 to 1:2, more preferably 1:1.3.
The molar ratio of the compound of formula V to the hydrazine hydrate may be 1:1 to 2:1, preferably 1.6:1.
In said step 2), the reaction temperature may be a temperature conventional in the art for such cyclization reactions, preferably-10 to 40 ℃, more preferably 0 to 5 ℃ or 20 to 30 ℃.
In the step 2), the cyclization reaction takes the disappearance or no longer reaction of the compound shown in the formula V as a reaction end point, and the reaction time is preferably 1 to 10 hours, more preferably 4 hours.
The step 2) may further comprise a post-treatment, preferably comprising the steps of: after the cyclization reaction is completed, adding water, layering, and concentrating an organic phase to obtain the compound shown in the formula IV-3.
When the compound shown in formula IV is a compound shown in formula IV-4, the above-mentioned preparation method of the compound shown in formula III-5 may further include the preparation method of the compound shown in formula VI, which includes the following steps:
In a solvent, in the presence of alkali, carrying out substitution reaction on a compound shown in a formula VIII and a compound shown in a formula IX to obtain a compound shown in a formula VI;
R 4 MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS;
x is halogen.
In the substitution reaction, the solvent may be a solvent conventional in the art for such substitution reaction, preferably a halogenated hydrocarbon solvent, more preferably dichloromethane.
In the substitution reaction, the base may be a base conventional in the art for such substitution reaction, preferably imidazole.
In the substitution reaction, X is preferably fluorine, chlorine, bromine or iodine, more preferably chlorine.
In the substitution reaction, the compound shown in the formula IX is preferably TBSCI, bnCI or TBDPSCI; correspondingly, the compound shown as the formula VIII is
In the substitution reaction, the molar ratio of the compound of formula VIII to the compound of formula IX may be a ratio conventional in substitution reactions of this type in the art, preferably 1:0.8 to 1:1.5, more preferably 1:0.93 to 1:1.1.
in the substitution reaction, the molar ratio of the compound of formula VIII to the base may be a ratio conventional in substitution reactions of this type in the art, preferably 1:1.2 to 1:2, more preferably 1:1.5.
In the substitution reaction, the concentration of the compound represented by formula VIII in the solvent may be a concentration conventional in the substitution reaction of this type in the art, preferably 0.1mol/L to 5mol/L, more preferably 1mol/L to 2.3mol/L.
In the substitution reaction, the reaction temperature may be a temperature conventional in the art for such substitution reaction, preferably-10 to 40 ℃, more preferably-2 to 5 ℃.
In the substitution reaction, the substitution reaction is carried out with the compound of formula VIII disappearing or no longer reacting as the reaction end point, and the reaction time is preferably 1 to 10 hours, more preferably 3 hours.
The substitution reaction may further comprise a post-treatment after completion, said post-treatment preferably comprising the steps of: adding methanol, adding water, layering, and concentrating the organic phase to obtain the compound shown in formula VI.
In the second scheme, the compound shown in the formula III-5 can be prepared by adopting the method in the first scheme.
In the third scheme, the compound shown in the formula III-6 can be prepared by adopting the method of the second scheme.
The invention also provides a preparation method of the compound shown in the formula X, which comprises the following steps: in a solvent, in the presence of lithium aluminum hydride, carrying out ester reduction reaction on a compound shown as a formula III-7 to obtain a compound shown as a formula X;
/>
In the compound shown in the formula III-7, R 1 Boc, fmoc, cbz, alloc or Teoc, R 2 、D 1 、T 1 And R is 4 Is defined as above for the compounds of formula III-7.
In the ester group reduction reaction, the compound shown in the formula III-7 is preferablyThe compound of formula X is preferably +.>
In the ester group reduction reaction, the solvent may be a solvent conventional in the art for such ester group reduction reaction, preferably an ether solvent, more preferably tetrahydrofuran.
In the ester group reduction reaction, the molar ratio of the compound shown in the formula III-7 to the lithium aluminum hydride may be 1:3 to 1:5, and more preferably 1:4.
In the ester group reduction reaction, the concentration of the compound represented by the formula III-7 in the solvent may be 0.1mol/L to 1.0mol/L, preferably 0.2mol/L.
In the ester group reduction reaction, the reaction temperature may be-10℃to 50℃and more preferably 0 to 5℃or 50 ℃.
In the ester group reduction reaction, the ester group reduction reaction takes the compound shown in the formula III-7 as a reaction end point, and the reaction time is preferably 1 to 10 hours, more preferably 3 hours.
The ester group reduction reaction may further comprise a post-treatment after completion, and the post-treatment preferably comprises the following steps: adding water, adding sodium hydroxide aqueous solution (preferably 15% sodium hydroxide aqueous solution by mass fraction), and filtering to obtain the compound shown in formula X.
The preparation method of the compound shown in the formula X can further comprise the following steps:
in a solvent, in the presence of alkali and a protective reagent, carrying out amino protection reaction on a compound shown as a formula III-6 to obtain a compound shown as a formula III-7;
the conditions and steps of the amino protection reaction are as described above;
in the compound as shown in the formula III-7, R 1 Boc, fmoc, cbz, alloc or Teoc, R 2 、D 1 、T 1 And R is 4 Is defined as above for the compounds of formula III-7;
the definition of the compound shown in the formula III-6 is shown in the above.
The preparation method of the compound shown in the formula X can further comprise the following steps:
in a solvent, carrying out deprotection reaction on a compound shown as a formula III-5 in the presence of a deprotection reagent to obtain a compound shown as a formula III-6;
the definition of the compound shown in the formula III-5 and the compound shown in the formula III-6 is shown in the above;
the conditions and steps of the deprotection reaction are as described above.
The preparation method of the compound shown in the formula X can further comprise the following steps:
in a solvent, in the presence of tributylphosphine and azodicarbonyl dipiperidine (ADDP), carrying out a cyclization reaction shown in the following formula IV to obtain a compound shown in the formula III-5;
The definition of the compound shown in the formula IV and the compound shown in the formula III-5 is shown in the above;
the conditions and steps of the cyclization reaction are as described above.
The preparation method of the compound shown in the formula X can further comprise the following steps:
in a solvent, in the presence of a reducing agent and an initiator, carrying out the reduction reaction of the compound shown as the formula IV-3 to obtain a compound shown as the formula IV-4;
the definition of the compound shown in the formula IV-3 and the compound shown in the formula IV-4 is shown in the above;
the conditions and steps of the reduction reaction are as described above.
The preparation method of the compound shown in the formula X can further comprise the following steps:
1) In a solvent, in the presence of alkali, carrying out ring-opening reaction on a compound shown in a formula VI and a compound shown in a formula VII to obtain a compound shown in a formula V;
2) In a solvent, in the presence of hydrazine hydrate, carrying out a cyclization reaction shown in the following formula V to obtain a compound shown in the formula IV-3;
the definition of the compound shown in the formula VI, the compound shown in the formula VII, the compound shown in the formula V and the compound shown in the formula IV-3 is as above;
The conditions and steps of the ring opening reaction and the ring forming reaction are as described above.
The preparation method of the compound shown in the formula X can further comprise the following steps:
in a solvent, in the presence of alkali, carrying out substitution reaction on a compound shown in a formula VIII and a compound shown in a formula IX to obtain a compound shown in a formula VI;
the definition of the compounds of formula VIII, of the compounds of formula IX and of the compounds of formula VI is as described above.
The conditions and steps of the substitution reaction are as described above.
The invention provides a preparation method of the compound shown in the formula IV-4, which comprises the following steps:
in a solvent, in the presence of a reducing agent and an initiator, carrying out the reduction reaction of the compound shown as the formula IV-3 to obtain a compound shown as the formula IV-4;
the definition of the compound shown in the formula IV-3 and the compound shown in the formula IV-4 is shown in the above;
the conditions and steps of the reduction reaction are as described above.
The invention provides a preparation method of the compound shown in the formula IV-3, which comprises the following steps:
1) In a solvent, in the presence of alkali, carrying out ring-opening reaction on a compound shown in a formula VI and a compound shown in a formula VII to obtain a compound shown in a formula V;
2) In a solvent, in the presence of hydrazine hydrate, carrying out a cyclization reaction shown in the following formula V to obtain a compound shown in the formula IV-3;
the definition of the compound shown in the formula VI, the compound shown in the formula VII, the compound shown in the formula V and the compound shown in the formula IV-3 is as above;
the conditions and steps of the ring opening reaction and the ring forming reaction are as described above.
The invention provides a preparation method of a compound shown in a formula VI, which comprises the following steps:
in a solvent, in the presence of alkali, carrying out substitution reaction on a compound shown in a formula VIII and a compound shown in a formula IX to obtain a compound shown in a formula VI;
the definition of the compound shown in formula VIII, the compound shown in formula IX and the compound shown in formula VI is as described above;
the conditions and steps of the substitution reaction are as described above.
Definition and description:
the following terms or phrases used herein are intended to have the following meanings unless otherwise indicated. A particular phrase or terminology, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense. When trade names are presented herein, it is intended to refer to their corresponding commercial products or active ingredients thereof.
Intermediate compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthetic methods, and equivalent alternatives well known to those skilled in the art, preferred embodiments including but not limited to the examples of the present invention.
The chemical reactions of the embodiments of the present invention are accomplished in a suitable solvent that is compatible with the chemical changes of the present invention and the reagents and materials required therefor. In order to obtain the compounds of the present invention, it is sometimes necessary for a person skilled in the art to modify or select the synthesis steps or reaction schemes on the basis of the embodiments already present.
The present invention will be specifically described by the following examples, which are not meant to limit the present invention in any way.
In the present invention, boc: t-butoxycarbonyl; cbz: a benzyloxycarbonyl group; fmoc: fluorenylmethoxycarbonyl; alloc: allyloxycarbonyl; teoc: trimethylsiloxycarbonyl; pht: phthaloyl; tos: p-toluenesulfonyl; tfa: trifluoroacetyl; trt: a trityl group; dmb:2, 4-dimethoxybenzyl; PMB: p-methoxybenzyl; MOM: methoxymethylene, bn: benzyl, THP: tetrahydropyranyl, tr: trityl, ac; acetyl, bz: benzoyl, piv: pivaloyl, TMS: trimethylsilyl, TES: triethylsilyl, TBS: t-butyldimethylsilyl, TBDPS: tertiary butyl diphenyl silyl, ms: methanesulfonyl, ts: p-toluenesulfonyl.
The term "alkyl" refers to a straight or branched chain alkyl group having the indicated number of carbon atoms.
All solvents used in the present invention are commercially available and can be used without further purification. The reaction is generally carried out under inert nitrogen in an anhydrous solvent.
The above preferred conditions can be arbitrarily combined on the basis of not deviating from the common knowledge in the art, and thus, each preferred embodiment of the present invention can be obtained.
The reagents and materials used in the present invention are commercially available.
The invention has the positive progress effects that: the pyrrolopyrimidine compound has an inhibiting effect on JAK1, JAK2, JAk3 and TYK2 kinase, and the preparation method has the advantages of simple reaction, high yield, simple operation, mild condition, safety, controllability and wide industrial application prospect.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limiting in any way. The present invention has been described in detail herein, and specific embodiments thereof are also disclosed, it will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the invention without departing from the spirit and scope of the invention. The experimental methods, in which specific conditions are not noted in the following examples, were selected according to conventional methods and conditions, or according to the commercial specifications.
Example 1
The preparation method of the compound 8 comprises the following steps:
step 1:
DCM (220 g) is added into a reactor, stirring is opened, TBSCl (61.5 g) is then added, stirring and clearing are carried out, a compound (21.8 g) shown in a formula (2-1) is added, imidazole (39.7 g) is introduced, nitrogen is introduced, the temperature is raised to-2-5 ℃, the reaction is kept for 3 hours, 3.1g of methanol is added after the reaction is finished, stirring is carried out for 0.5 hour, 110.0g of water is added, stirring is carried out for 0.5 hour, standing, layering and organic phase concentration are carried out, thus obtaining a product 1 with the yield of 95.5% and the purity of 97.5%;
step 2:
adding the product 1 (72.3 g) obtained in the step 1 and THF (430 g) into a reactor, stirring, introducing nitrogen, heating to 40-50 ℃, preserving heat and reacting for 1h, then adding butyllithium (88 g), stirring for 1h, adding the compound (63.8 g) of the formula (1-0), continuing to react for 4h in a heat-preserving way, adding water (480 g) and ammonium chloride (30 g) after the reaction is finished, stirring for 0.5h, adding MTBE (200 g), stirring for 0.5h, standing, layering, and collecting an organic phase to obtain a product 2 solution; 1 H NMR(400MHz,CDCl 3 )δ=4.971(s,1H),4.33(s,2H),4.177-4.144(m,H),4.100-4.039(m,2H),2.621-2.525(m,2H),2.054-1.912(m,2H),1.312(s,9H),1.172-1.127(m,3H),0.781(s,9H),0.001(s,6H)。
step 3:
adding 80% hydrazine hydrate (19.8 g) (80% hydrazine hydrate is an aqueous solvent with the mass fraction of 80% hydrazine hydrate) into the reactor in the step 2, maintaining the temperature at 0-5 ℃, heating to 20-30 ℃ after the dripping is finished, preserving heat for 4h, adding water (400.0 g) after the reaction is finished, stirring for 0.5h, standing, layering, and concentrating an organic phase to obtain a product 3; yield 89.5%, purity 87.4%; 1 H NMR(400MHz,CDCl 3 )δ=6.018(s,1H),5.366-5.345(m,1H),4.746(s,2H),4.348-4.321(m,1H),4.212-4.142(m,2H),2.764-2.705(m,2H),2.143-2.110(m,2H),1.462(s,9H),1.309-1.253(m,3H),0.961-0.891(m,9H),0.139-0.082(m,6H)。
Step 4:
adding a product 3 (220 g) and MTBE (450 g) into a reactor, introducing nitrogen, cooling to 0-5 ℃, slowly adding sodium borohydride (30 g), then adding methanol (120 g), heating to room temperature, preserving heat for 20h, adding water (480 g) and ammonium chloride (40 g) after the reaction is finished, stirring for 0.5h, standing, layering, concentrating an organic phase to obtain a product 4, wherein the yield is 90.9%, and the purity is 86.4%; 1 H NMR(400MHz,CDCl 3 )δ=6.003(s,1H),5.321(s,1H),5.100-5.078(m,1H),4.738(s,2H),4.321(m,1H),4.212-4.142(m,2H),2.809-2.646(m,2H),1.861-1.826(m,2H),1.491-1.453(s,9H),0.958-0.887(s,9H),0.133-0.110(s,6H);
step 5:
adding a product 4 (40 g) and THF (630 g) into a reactor, introducing nitrogen, cooling to 0-5 ℃, adding tributylphosphine (40 g), stirring, adding ADDP (56 g) in batches, heating to room temperature, preserving heat for 20h, filtering after reaction, concentrating to obtain a product 5 with a yield of 85.8% and a purity of 53.3%, 1 H NMR(400MHz,CDCl 3 )δ=6.007(s,1H),4.876-4.860(m,1H),4.617(m,2H),4.331-4.240(m,2H),3.948-3.901(m,1H),2.878-2.848(m,2H),2.067-2.027(m,2H),1.463(s,9H);
step 6:
adding 30% (HCl/EtOH, 180 g) into a reactor, cooling to 0-5 ℃, then adding a product 5 (160 g), heating to room temperature, preserving heat for 4h, adding absolute ethyl alcohol (300 g) after the reaction is finished, stirring for 12 h, filtering,drying to obtain a product 6, 1 H NMR(400MHz,CDCl 3 )δ=5.870(s,1H),4.320-4.085(s,2H),3.686-3.637(m,2H),2.813-2.771(m,1H),2.630-2.308(m,2H),1.933-1.918(m,3H),1.628-1.594(m,2H);
step 7:
adding a product 6 (32 g) and water (320 g) into a reactor, stirring a sodium carbonate solution (the mass fraction is 10%,180 g), cooling to 0-5 ℃, then dropwise adding a Boc anhydride/methanol solution (50 g of Boc anhydride and 150g of methanol), heating to room temperature after the dropwise adding is finished, carrying out heat preservation reaction for 10 hours, adding dichloromethane (300 g) after the reaction is finished, stirring for 0.5 hour, standing, layering, and concentrating an organic phase to obtain a product 7, wherein the yield is 43.2%, and the purity is 98.5%;
Step 8:
THF (90 ml) is added into a reactor, nitrogen is introduced, the temperature is reduced to 0-5 ℃, then lithium aluminum hydride (3.4 g) is added, the temperature is raised to 50 ℃, then 7/THF solution (6 g/30 ml) is added dropwise, the temperature is raised to 70 ℃ after the completion of the dropwise addition, the reaction is kept for 3 hours, the temperature is reduced to 5-10 ℃ after the completion of the reaction, water (30 g) is added, stirring is carried out, sodium hydroxide aqueous solution (15% by mass, 4 g) is added, anhydrous sodium sulphate (7 g) is added, stirring is carried out for 3 hours, filtering and drying are carried out, and the product 8 is obtained, wherein the yield is 80.9%, and the purity is 98.8%.
Yield and purity of example 1:
sequence number Step 1 Step 2 and 3 Step 4 Step 5 Step 6 and 7 Step 8
Yield (%) 95.5 89.5 90.9 85.8 43.2 80.9
Purity (%) 97.5 87.4 86.4 53.3 98.5 98.8
Example 2
Steps 1-7 are the same as in example 1, differing from step 8, and specifically as follows:
LAH (0.1 mol) was added to the reactor at 0 ℃, then product 7 (23 g)/THF (400 mL) was added and warmed to 70 ℃, incubated for 2h, cooled to 0 ℃, water (6.6 mL) was added dropwise to the mixture, naOH aqueous solution (15%, 6.6 mL) and water (19.8 mL) were added, stirred at room temperature for 30 minutes, filtered, washed with DCM/MeOH (10:1) (300 ml×3), concentrated to give product 8 in a yield of 50.4%, purity of 97.6%.
Example 3
Step 1:
DCM (220 g) is added into a reactor, stirring is opened, bnCl (51.6 g) is then added, stirring and clearing are carried out, a compound (21.8 g) shown in a formula (2-1) is added, imidazole (39.7 g) is introduced, nitrogen is introduced, the temperature is raised to-2-5 ℃, the reaction is kept for 3 hours, 110.0g of water is added after the reaction is finished, stirring is carried out for 0.5 hour, standing, layering and organic phase concentration are carried out, and the product 2-1 is obtained, the yield is 95.5%, and the purity is 97.3%;
Step 2:
adding the product 2-1 (62.3 g), THF (430 g) and the compound (63.8 g) shown in the formula (1-0) obtained in the step (1) into a reactor, stirring, introducing nitrogen, heating to 40-50 ℃, preserving heat for reaction for 1h, then adding butyl lithium (88 g), continuing to preserve heat for reaction for 4h, adding water (480 g) and ammonium chloride (30 g) after the reaction is finished, stirring for 0.5h, adding MTBE (200 g), stirring for 0.5h, standing, layering, and collecting an organic phase to obtain a product 2-2 solution; 1 H NMR(400MHz,CDCl 3 )δ=7.1-7.5(m,5H),4.971(s,1H),4.5(s,2H)4.33(s,2H),4.177-4.144(m,H),4.100-4.039(m,2H),2.621-2.525(m,2H),2.054-1.912(m,2H),1.312(s,9H),1.172-1.127(m,3H);
step 3:
adding 80% hydrazine hydrate (19.8 g) dropwise into the reactor in the step 2, keeping the temperature at 0-5 ℃, heating to 20-30 ℃ after the dropwise addition is finished, carrying out heat preservation reaction for 4 hours, adding water (400.0 g) after the dropwise addition is finished, stirring for 0.5 hours, standing, layering, concentrating an organic phase to obtain a product 2-3, wherein the yield is 89.3%, and the purity is 87.5%;
step 4:
adding 2-3 (236 g) and MTBE (400 g) into a reactor, introducing nitrogen, cooling to 0-5 ℃, slowly adding sodium borohydride (30 g), then adding methanol (120 g), heating to room temperature, preserving heat for reaction for 20h, adding water (480 g) and ammonium chloride (40 g) after the reaction is finished, stirring for 0.5h, standing, layering, concentrating an organic phase, and obtaining 2-4 products, wherein the yield is 90.9%;
step 5:
adding 2-4 (37.6 g) and THF (630 g) into a reactor, introducing nitrogen, cooling to 0-5 ℃, adding tributylphosphine (40 g), stirring, adding ADDP (56 g) in batches, heating to room temperature, preserving heat for reaction for 20h, filtering after the reaction is finished, concentrating to obtain 2-5, and obtaining 85.4% yield; 1 H NMR(400MHz,CDCl 3 )δ=7.6-7.7(m,1H),7.1-7.5(m,4H),6.018(s,1H),5.366-5.345(m,1H),4.746(s,2H),4.5(s,2H),4.348-4.321(m,1H),4.212-4.142(m,2H),2.764-2.705(m,2H),2.143-2.110(m,2H),1.462(s,9H),1.309-1.253(m,3H);
Step 6:
adding 30% (HCl/EtOH, 180 g) into a reactor, cooling to 0-5 ℃, then adding 2-5 (150.4 g) of a product, heating to room temperature, carrying out heat preservation reaction for 4h, cooling to room temperature after the reaction is finished, filtering, and drying to obtain 2-6 of the product;
example 4
Step 1:
DCM (220 g) is added into a reactor, stirring is opened, TBDPSCl (112.3 g) is then added, stirring and clearing are carried out, a compound (21.8 g) shown in a formula (2-0) is added, imidazole (39.7 g) is introduced, nitrogen is introduced, the temperature is raised to-2-5 ℃, the reaction is kept for 3 hours, 110.0g of water is added after the reaction is finished, stirring is carried out for 0.5 hours, standing, layering and organic phase concentration are carried out, and the product 3-1 is obtained, and the yield is 95.6%;
step 2:
adding the product 3-1 (125.1 g), THF (430 g) and the compound (63.8 g) shown in the formula (1-0) obtained in the step (1) into a reactor, stirring, introducing nitrogen, heating to 40-50 ℃, preserving heat for reaction for 1h, then adding butyl lithium (88 g), continuing to preserve heat for reaction for 4h, adding water (480 g) and ammonium chloride (30 g) after the reaction is finished, stirring for 0.5h, adding MTBE (200 g), stirring for 0.5h, standing, layering, and collecting an organic phase to obtain a product 3-2 solution; 1 H NMR(400MHz,CDCl 3 )δ=7.3-7.5(m,2H),6.9-7.3(m,8H)4.971(s,1H),4.33(s,2H),4.177-4.144(m,H),4.100-4.039(m,2H),2.621-2.525(m,2H),2.054-1.912(m,2H),1.312(s,9H),1.172-1.127(m,3H),0.781(s,9H);
step 3:
adding 80% hydrazine hydrate (19.8 g) dropwise into the reactor in the step 2, keeping the temperature at 0-5 ℃, heating to 20-30 ℃ after the dropwise addition is finished, carrying out heat preservation reaction for 4 hours, adding water (400.0 g) after the dropwise addition is finished, stirring for 0.5 hours, standing, layering, concentrating an organic phase to obtain a product 3-3, and obtaining the yield of 89.0%;
Step 4:
adding 3-3 (322.6 g) and MTBE (400 g) into a reactor, introducing nitrogen, cooling to 0-5 ℃, slowly adding sodium borohydride (30 g), then adding methanol (120 g), heating to room temperature, preserving heat for reaction for 20h, adding water (480 g) and ammonium chloride (40 g) after the reaction is finished, stirring for 0.5h, standing, layering, concentrating an organic phase to obtain 3-4 of a product, and obtaining 90.9% of yield;
step 5:
adding 3-4 (53 g) and THF (630 g) into a reactor, introducing nitrogen, cooling to 0-5 ℃, adding tributylphosphine (40 g), stirring, adding ADDP (56 g) in batches, heating to room temperature, preserving heat for reaction for 20h, filtering after the reaction is finished, concentrating to obtain 3-5, and obtaining 85.6% yield; 1 H NMR(400MHz,CDCl 3 )δ=7.3-7.5(m,2H),6.9-7.3(m,8H),6.018(s,1H),5.366-5.345(m,1H),4.746(s,2H),4.348-4.321(m,1H),4.212-4.142(m,2H),2.764-2.705(m,2H),2.143-2.110(m,2H),1.462(s,9H),1.309-1.253(m,3H),0.961-0.891(m,9H);
step 6:
30% (HCl/EtOH, 180 g) is added into a reactor, the temperature is reduced to 0-5 ℃, then 3-5 (211 g) is added into the reactor, the temperature is raised to room temperature, the reaction is kept for 4h, the temperature is reduced to room temperature after the reaction is finished, and the product 3-6 is obtained after filtration and drying.
Example 5
The synthesis method of compound 12 is as follows
Step 9:
adding the product 8 (60 g), the compound (107.0 g) of the formula (7-1), DMSO (600 ml) and DIPEA (85.6 g) in the example 1 into a reactor, heating to 100-110 ℃, keeping the temperature and stirring for 4 hours, cooling to 20-30 ℃, adding water (150 ml), stirring for 1 hour, filtering, and drying to obtain a product 9 with the yield of 91% and the purity of 98%;
Step 10:
dichloromethane (200 g), a product 9 (15 g), 2, 6-tetramethylpiperidine-nitrogen-oxide (TEMPO, 0.52 g) and iodobenzene diacetate (11.7 g) are added into a reactor to react for 10 hours at room temperature, triethylamine (12 g) is added after the reaction is finished, stirring is carried out for 1 hour, filtering and drying are carried out, and a product 10 with the purity of 98% and the yield of 95% is obtained;
step 11:
to the reactor were added product 10 (73 g), aqueous ammonia (25 mL), iodine (12.4 g) THF (730 mL), stirred at room temperature for 12h, ethyl acetate (100 mL) was added after the reaction was completed, stirred for 0.5h, and the mixture was allowed to stand, separate layers, and concentrate the organic phase to give product 11, purity: 98.2% Compound 10:0.2%; the yield thereof was found to be 96%;
step 12:
the reactor was charged with 11 (11 g), THF (100 ml) and TBAF (0.1 mol), the temperature was raised to 70℃and the reaction was continued for 12 hours, after the completion of the reaction, the temperature was lowered to room temperature, and 10% NaHCO was added 3 Aqueous solution (100 ml) and the mixture was stirred at 20 ℃ for 2h, filtered and dried to give product 12, purity 98.6%, yield 86%; 1 H NMR(400MHz,DMSO-d 6 )δ=12.80-12.56(m,1H),8.43-8.32(m,1H),7.49-7.38(m,1H),7.01-6.88(m,1H),6.88-6.74(m,1H),5.31-5.17(m,1H),4.52-4.36(m,2H),3.45-3.41(m,3H),3.13-3.04(m,1H),3.01-2.91(m,1H),2.38-2.29(m,1H),2.17-2.09(m,1H)。
comparative example 1
Step a: (4-methoxyphenyl) methanol (50 g) was added to a DMF (500 mL) suspension of sodium hydride (15.92 g) at 0℃and after stirring for 0.5 hours, 3-bromoprop-1-yne (59.19 g) was slowly added to the reaction system, the resulting solution was stirred at 0℃for 2.5 hours and at 25℃for 16 hours, saturated aqueous ammonium chloride solution (500 mL) was added to the reaction solution, the aqueous phase was extracted with ethyl acetate (500 mL. 3), the combined organic phase was washed with water (200 mL. Times.2) and brine (200 mL. Times.1), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, the resulting residue was purified by column chromatography (petroleum ether/ethyl acetate=1/0-1/1), 1 H NMR(400MHz,DMSO-d6)δ=7.27-7.22(m,2H),6.93-6.89(m,2H),4.44(s,1H),4.46-4.41(m,1H),4.12(d,J=2.4Hz,2H),3.74(s,2H),3.77-3.72(m,1H),3.46(t,J=2.4Hz,1H),3.48-3.45(m,1H),3.48-3.45(m,1H),3.48-3.45(m,1H),3.48-3.44(m,1H);
Step b: to a solution of 5-bromo-2-iodo-pyridine (50 g) in THF (500 mL) at 25 ℃ was added compound B (34.14 g), cuprous iodide (3.35 g), piperidine (44.99 g) and bis (triphenylphosphine) palladium (II) (6.18 g), the reaction system was replaced three times with nitrogen, the resulting solution was stirred at 25 ℃ for 16 hours, the reaction solution was filtered with celite to obtain a filtrate, after concentrating under reduced pressure, the residue was dissolved with 800mL of ethyl acetate, then washed sequentially with 300mL of water and 300mL of brine, and then the organic phase was dried with anhydrous sodium sulfate, filtered, concentrated under reduced pressure to obtain a residue which was purified by column chromatography (silica, petroleum ether/ethyl acetate=1/0-30/1) to obtain compound C in 73.9% yield; 1 H NMR(400MHz,CDCl3-d)δ=8.61(d,J=1.8Hz,1H),7.75(dd,J=2.4,8.4Hz,1H),7.32-7.28(m,1H),7.32-7.26(m,1H),7.27(s,1H),6.89-6.84(m,2H),4.58(s,2H),4.35(s,2H),3.79-3.76(m,1H),3.77(s,2H)。
step c: stirring the solution obtained by the compound C (1 g) at the temperature of 0 ℃ for 16 hours at the temperature of 25 ℃, then slowly adding 32mL of tert-butyl methyl ether into the system under the stirring at the temperature of 0 ℃, slowly separating out a large amount of off-white solid, filtering and drying to obtain the compound D with the yield of 80%; 1 H NMR(400MHz,CHLOROFORM-d)δ=9.68-9.61(m,1H),7.87-7.79(m,1H),7.45-7.40(m,1H),7.22-7.19(m,2H),6.85-6.77(m,2H),6.76-6.69(m,2H),4.51-4.44(m,2H),4.40-4.32(m,2H),3.78-3.71(m,3H),2.64-2.58(m,6H),2.18-2.13(m,3H)。
step d: silver carbonate (10.07 g) was added to a DMF (160 mL) solution of compound D (10 g) at 25 ℃ and the resulting solution was stirred at 40 ℃ for 16 hours, the reaction solution was filtered with celite to obtain a filtrate, after concentrating under reduced pressure, the residue was dissolved with 200mL of ethyl acetate, then washed with 100mL of water and 100mL of brine in this order, and then the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure to obtain a residue which was purified by column chromatography (silica, petroleum ether/ethyl acetate=1/0-5/1) to obtain compound E in 82% yield; 1 H NMR(400MHz,CDCl 3 -d)δ=8.50-8.46(m,1H),8.50-8.46(m,1H),7.34-7.27(m,1H),7.26-7.21(m,2H),7.10-7.06(m,1H),6.84-6.78(m,2H),6.50-6.46(m,1H),4.67-4.60(m,2H),4.51-4.46(m,2H),3.75-3.71(m,3H);
Step e: cesium carbonate (8.82 g), 4, 5-bis-diphenylphosphine-9, 9-dimethyloxaxanthene (783.26 mg) and tris (dibenzylideneacetone) dipalladium (619.79 mg) were added sequentially to a DME (30 mL) solution of compound E (4.7 g) and t-butyl N-methylcarbamate (5.33 g) at 25 ℃, the resulting solution was stirred at 100 ℃ for 16 hours, the reaction solution was filtered with celite to give a filtrate, the filter cake was washed with 100mL of ethyl acetate, the combined organic phases were washed with 100mL of water and 100mL of brine in this order, and then the organic phases were dried with anhydrous sodium sulfate, filtered, concentrated under reduced pressure to give a residue which was purified by column chromatography (silica, petroleum ether/ethyl acetate=1/0-20/1), to give compound F in a yield of 47.3%; MS (ESI) calculated C 22 H 27 N 3 O 4 397, measurement 398[ M+H ]] +
Step f: to a solution of Compound F (4 g) in EtOH (60 mL) at 25℃under nitrogen gas was added platinum dioxide (933.33 mg) as a reaction system H 2 Three substitutions were made and the resulting solution was taken up in H 2 Stirring at 70 ℃ for 72 hours under the pressure of (3 MPa), filtering the reaction solution by using diatomite to obtain filtrate, and concentrating under reduced pressure to obtain a compound G with the yield of 62%; MS (ESI) calculated C 22 H 31 N 3 O 4 401, measurement 402[ M+H ]] +1 H NMR(400MHz,CDCl 3 -d)δ=7.32-7.27(m,2H),6.91-6.85(m,2H),6.07-5.98(m,1H),4.53-4.50(m,2H),4.49-4.46(m,2H),4.32-4.24(m,1H),4.00-3.90(m,1H),3.82-3.79(m,3H),3.82-3.78(m,3H),3.84-3.78(m,3H),3.04-2.93(m,1H),2.88-2.82(m,3H),2.82-2.74(m,1H),2.05-1.89(m,2H),1.52-1.46(m,9H),1.50-1.45(m,9H),1.43-1.35(m,1H);
Step g: trifluoroacetic acid (12.33G) was added to a solution of compound G (4G) in DCM (40 mL) at 0 ℃, and the resulting solution was stirred at 25 ℃ for 2 hours, followed by concentration under reduced pressure to give compound H in 66% yield; MS (ESI) calculated C 9 H 15 N 3 O181, measurement 182[ M+H ]] +
In comparison of example 1 with comparative example 1 in the present invention, example 1 has the following advantages:
1. compound D in the route of comparative example 1 was prone to explosion, and had a problem of safety;
2. compound G in the route of comparative example 1 is not a chiral isolated compound, and API was prepared from compound G as a starting material, and the final step requires a chiral column for separation, which is not conducive to mass production.
Biological Activity test
Experimental example 1: in vitro Activity test of Jak1, jak2, jak3, tyk2 kinase
Experimental materials
Recombinant human JAK1, JAK2, JAK3, tyk2 protease, major instruments and reagents are all supplied by Eurofins company in the united kingdom
Experimental method
JAK2, JAK3 and TYK2 dilutions: 20mM 3- (N-morpholino) propanesulfonic acid (MOPS), 1mM EDTA,0.01%Brij-35.5% glycerol, 0.1% beta-mercaptoethanol, 1mg/mL BSA; JAK1 dilution: 20mM TRIS,0.2mM EDTA,0.1% beta-mercaptoethanol, 0.01% Brij-35.5% glycerol. All compounds were prepared as 100% DMSO solutions and reached 50-fold final assay concentration. The test compounds were subjected to 3-fold concentration gradient dilutions at a final concentration of 10 μm to 0.001 μm for a total of 9 concentrations, with a DMSO content of 2% in the assay reaction. The working stock of the compound was added to the assay wells as the first component of the reaction, and the remaining components were then added according to the protocol detailed in the assay below.
JAK1 (h) enzymatic reaction
JAK1 (h) with 20mM Tris/HCl pH7.5,0.2mM EDTA,500 mu M MGEEPLYWSFPAKKK,10mM magnesium acetate and [ gamma ] 33 P]ATP (activity and concentration as formulated) is incubated together. The reaction was started by adding the Mg/ATP mixture and stopped by adding 0.5% phosphoric acid after incubation for 40 minutes at room temperature. Then 10 μl of the reaction was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol over 4 minutes, dried and flash counted.
JAK2 (h) enzyme reaction
JAK2 (h) and 8mM MOPS pH 7.0,0.2mM EDTA,100 mu M KTFCGTPEYLAPEVRREPRILSEE EQEMFRDFDYIADWC,10mM magnesium acetate and [ gamma- ] 33 P]ATP (activity and concentration as formulated) is incubated together. Adding Mg/ATP mixtureAfter incubation at room temperature for 40 minutes, the reaction was started and terminated by adding 0.5% strength phosphoric acid. Then 10 μl of the reaction was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol over 4 minutes, dried and flash counted.
JAK3 (h) enzymatic reaction
JAK3 (h) and 8mM MOPS pH 7.0,0.2mM EDTA,500 mu M GGEEEEYFELVKKKK,10mM magnesium acetate and [ gamma- ] 33 P]ATP (activity and concentration as formulated) is incubated together. The reaction was started by adding the Mg/ATP mixture and stopped by adding 0.5% phosphoric acid after incubation for 40 minutes at room temperature. Then 10 μl of the reaction was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol over 4 minutes, dried and flash counted.
TYK2 (h) enzyme reaction
TYK2 (h) and 8mM MOPS pH 7.0,0.2mM EDTA,250 mu M GGMEDIYFEFMGGKKK,10mM magnesium acetate and [ gamma- ] 33 P]ATP (activity and concentration as formulated) is incubated together. The reaction was started by adding the Mg/ATP mixture and stopped by adding 0.5% phosphoric acid after incubation for 40 minutes at room temperature. Then 10 μl of the reaction was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol over 4 minutes, dried and flash counted.
Data analysis
IC 50 The results were analyzed by the xlit 5 (formula 205) from IDBS, see in particular table 1.
TABLE 1 in vitro screening test results for the compounds of the invention
Conclusion: the compounds of the invention exhibit good selective inhibition of JAK1 and/or JAK2 in vitro activity assays of kinase 4 subtypes JAK1, JAK2, JAk and TYK 2.
Experimental example 2: permeability test
Experimental materials
The transport buffer is HBSS (hanks balanced salt solution) and 10mm HEPES (N- (2-hydroxyethyl) piperazine-N' - (2-ethanesulfonic acid) solution), and the pH value is 7.40+/-0.05; caco-2 cells were purchased from ATCC.
Experimental method
Caco-2 cells at 1X 10 5 Cells/cm 2 The medium was inoculated into a 96-well BD insert plate of polyethylene film (PET) and refreshed every 4-5 days until day 21-28, forming a confluent cell monolayer. Test compounds were bi-directionally tested at 2. Mu.M and were double-well. Digoxin is bi-directional 10 μm, nadolol and metoprolol are bi-directional 2 μm. The final DMSO concentration was adjusted to less than 1%. The plates were incubated in a CO2 incubator at 37.+ -. 1 ℃ for 2 hours, at saturated humidity, with 5% CO2 without shaking. After all samples were mixed with acetonitrile containing internal standard, the cores were separated at 4000 rpm for 10 minutes, and then 100 μl of the supernatant was diluted with 100 μl of distilled water for LC/MS analysis. The initial solution of the test sample, the solution of the test sample and the concentration of the control sample in the solution of the test sample are quantified by the LC/MS/MS method and by the peak area ratio of the analyte/the internal standard substance. After the transport assay, the integrity of the Caco-2 cell monolayer was determined by fluorescein rejection and the apparent permeability coefficient and efflux were calculated.
Experimental results
The experimental results are shown in Table 2-1:
table 2-1 permeability of compound 12
Conclusion: the compound has characteristic high permeability, and is favorable for realizing good target tissue concentration and oral bioavailability.
Note that: ND: no detection was made.
Experimental example 3: pharmacokinetic (PK) test
The clear solutions obtained after dissolution of the test compounds were administered to male mice (C57 BL/6) or rats (SD) in vivo (overnight fast, 7-8 weeks old) via tail vein injection and gavage, respectively. After administration of the test compound, the intravenous group (1 mg/kg) was followed by the intragastric group (3 mg/kg) at 0.117,0.333,1,2,4,7 and 24 hoursAt 0.25,0.5,1,2,4,8 and 24 hours, plasma was obtained after blood collection from the mandibular vein and centrifugation, respectively. The blood concentration was determined by LC-MS/MS method using WinNonlin TM Version 6.3 pharmacokinetic software, the relevant pharmacokinetic parameters were calculated by a non-compartmental model linear log trapezium method. The test results were as follows:
TABLE 3-1 PK test results of Compound 12 in mice
PK parameters Results
T 1/2 (hr) 1.89
C max (nM) 6000
AUC 0-inf (nM.hr) 12765
Bioavailability(%) a 88.4
Note that: t (T) 1/2 : half-life period; c (C) max : peak concentration;
AUC 0-inf : area under the plasma concentration-time curve from time 0 to extrapolation to infinity;
bioavailability: bioavailability.
Conclusion: the compound of the invention has good oral bioavailability in mice, and higher exposure is beneficial to generating good in vivo efficacy.
Experimental example 4: in vivo efficacy study of rat adjuvant-induced arthritis (AIA)
The experimental process comprises the following steps:
the effect of the compounds of the invention in the treatment of arthritis was verified using a rat adjuvant arthritis model. Female, 160-180 g Lewis rats were anesthetized with isoflurane and injected subcutaneously with 0.1ml Mycobacterium tuberculosis suspension in the left hind leg. The rats were then grouped and given the corresponding test compounds 13 days after molding, e.g., different doses were given (specific doses are shown in Table 4-2, test compound 12 was dissolved in [ 5% DMSO,95% (12% SBE-. Beta. -CD), 0.5% MC) ] mixed vehicle and female Lewis rats were given orally 2 times daily (number of test animals per dose group is 8). Two weeks of continuous dosing, during which time the rat status was observed, the foot volume swelling was recorded and scored, and the scoring criteria are shown in table 4-1.
TABLE 4-1 clinical scoring criteria for arthritis
Experimental results:
the compound 12 two dose treatment groups have remarkable relieving effect on the weight reduction trend of animals caused by morbidity, and the low and medium dose groups (3 mg/kg and 10 mg/kg) have remarkable differences from the solvent control group in 20 days, and show good weight recovery effect. Compound 12 inhibited the elevation of arthritis clinical scores and foot volumes, and this inhibition was dose dependent. The 1210mg/kg effect of the compound was most pronounced (significant difference compared to the solvent control group starting from 15 days). The average arthritis clinical score for this group decreased from 6 points at the 13 th day peak to 1.4 points at the 27 th day of the experimental endpoint and was significantly different from the solvent control group.
TABLE 4-2 area under clinical score inhibition (AUC)
Conclusion: compound 12 of the present invention showed significant therapeutic effects (inhibition ratio P <0.0001 compared to vehicle control) at doses (3 mg/kg and 10 mg/kg), and compound 12 of the present invention showed positive correlation of effects at good doses (3 mg/kg and 10 mg/kg).
While particular embodiments of the present invention have been described above, it will be appreciated by those skilled in the art that these are merely illustrative, and that many changes and modifications may be made to these embodiments without departing from the principles and spirit of the invention. Accordingly, the scope of the invention is defined by the appended claims.

Claims (9)

1. A compound of the formula V,
wherein,
R 1 boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB;
R 4 selected from MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS and TBDPS;
R 5 is COOEt or COOMe;
but the compound of formula V is not
2. The compound of formula V according to claim 1, wherein R 1 Boc.
3. The compound of formula V according to claim 1, wherein R 4 Be Bn, TBS or TBDPS.
4. The compound of formula V according to claim 1, wherein R 5 Is COOEt.
5. As claimed inThe compound shown in the formula V in the claim 1, wherein the compound shown in the formula V is any one of the following compounds:or->
6. A process for the preparation of a compound of formula V, characterized in that: in a solvent, in the presence of alkali, carrying out ring opening reaction on a compound shown in a formula VI and a compound shown in a formula VII to obtain a compound shown in a formula V
Wherein R is 1 Boc, fmoc, cbz, alloc, teoc, pht, tos, tfa, trt, dmb or PMB;
R 4 MOM, bn, THP, tr, ac, bz, piv, TMS, TES, TBS or TBDPS;
R 5 is COOEt or COOMe;
but the compound of formula V is not
7. The process for the preparation of a compound of formula V according to claim 6, wherein one or more of the following conditions are satisfied:
(1) The compound shown in the formula VI isOr->The compound shown in formula VII is +.>
(2) The solvent is one or more of alkane solvent, halogenated alkane solvent, ether solvent and aromatic hydrocarbon solvent;
(3) The alkali is butyl lithium;
(4) The molar ratio of the compound shown in the formula VI to the compound shown in the formula VII is 1:1-2:1;
(5) The concentration of the compound shown in the formula VII in the solvent is 0.1mol/L to 1.0mol/L;
(6) The molar ratio of the compound shown in the formula VII to the alkali is 1:3-1:8;
(7) The temperature of the reaction is 35-70 ℃;
(8) The reaction time of the ring-opening reaction is 1-5 hours;
(9) The ring-opening reaction is carried out in an inert atmosphere, and the inert atmosphere is nitrogen or argon;
(10) The preparation method of the compound shown as the formula V further comprises post-treatment, wherein the post-treatment comprises the following steps: after the ring-opening reaction is finished, water and acid are added, MTBE is added, and the organic phase containing the compound shown in the formula V is obtained through layering.
8. A process for the preparation of a compound of formula V according to claim 7, which satisfies one or more of the following conditions:
(1) The solvent is CH 2 Cl 2 、CHCl 3 、CCl 4 One or more of tetrahydrofuran, diethyl ether, petroleum ether, n-hexane, benzene and toluene;
(2) The molar ratio of the compound shown in the formula VI to the compound shown in the formula VII is 1.7:1;
(3) The concentration of the compound shown in the formula VII in the solvent is 0.51 mol/L;
(4) The molar ratio of the compound shown in the formula VII to the alkali is 1:5.5;
(5) The temperature of the reaction is 40-50 ℃;
(6) The reaction time of the ring-opening reaction is 4 hours.
9. A process for the preparation of a compound of formula V according to claim 8, wherein one or more of the following conditions are met:
(1) The solvent is tetrahydrofuran;
(2) In the post-treatment step, the acid is ammonium chloride.
CN202210519413.5A 2020-12-04 2020-12-04 Pyrrolo pyrimidine compound intermediate and preparation method thereof Active CN115124526B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210519413.5A CN115124526B (en) 2020-12-04 2020-12-04 Pyrrolo pyrimidine compound intermediate and preparation method thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011408385.7A CN113372343B (en) 2020-12-04 2020-12-04 Pyrrolopyrimidine compound intermediate and preparation method thereof
CN202210519413.5A CN115124526B (en) 2020-12-04 2020-12-04 Pyrrolo pyrimidine compound intermediate and preparation method thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202011408385.7A Division CN113372343B (en) 2020-12-04 2020-12-04 Pyrrolopyrimidine compound intermediate and preparation method thereof

Publications (2)

Publication Number Publication Date
CN115124526A CN115124526A (en) 2022-09-30
CN115124526B true CN115124526B (en) 2024-01-30

Family

ID=77569151

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202011408385.7A Active CN113372343B (en) 2020-12-04 2020-12-04 Pyrrolopyrimidine compound intermediate and preparation method thereof
CN202210519413.5A Active CN115124526B (en) 2020-12-04 2020-12-04 Pyrrolo pyrimidine compound intermediate and preparation method thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202011408385.7A Active CN113372343B (en) 2020-12-04 2020-12-04 Pyrrolopyrimidine compound intermediate and preparation method thereof

Country Status (1)

Country Link
CN (2) CN113372343B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113372366B (en) * 2020-12-04 2022-08-30 广州嘉越医药科技有限公司 Salt of pyrrolopyrimidine compound, crystal form and application thereof
WO2022242768A1 (en) * 2021-05-21 2022-11-24 广州嘉越医药科技有限公司 Use of pyrrolopyrimidine compound

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016116025A1 (en) * 2015-01-20 2016-07-28 南京明德新药研发股份有限公司 Jak inhibitor
WO2016192563A1 (en) * 2015-05-29 2016-12-08 南京明德新药研发股份有限公司 Janus kinase inhibitor
CN107805259A (en) * 2017-10-31 2018-03-16 无锡福祈制药有限公司 A kind of azolopyrimidines

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3981769A4 (en) * 2019-06-05 2023-07-19 Guangzhou Joyo Pharmatech Co., Ltd Pyrrolopyrimidine compound and use thereof
KR20220107018A (en) * 2019-11-27 2022-08-01 치아타이 티안큉 파마수티컬 그룹 주식회사 Triheterocyclic compounds as JAK inhibitors and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016116025A1 (en) * 2015-01-20 2016-07-28 南京明德新药研发股份有限公司 Jak inhibitor
WO2016192563A1 (en) * 2015-05-29 2016-12-08 南京明德新药研发股份有限公司 Janus kinase inhibitor
CN107805259A (en) * 2017-10-31 2018-03-16 无锡福祈制药有限公司 A kind of azolopyrimidines

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
"Reactivity of N-Boc-Protected Amino-Ynones in the Presence of Zinc Chloride:Formation of Acetylenic Cyclic Imines and Their Palladium Complexes";Huy-Dinh Vu等;《Eur. J. Org. Chem.》;4868–4875页 *
"Synthesis and Cycloaromatization of a Cyclic Enyne-Allene Prodrug";Tefsit Bekele等;《J. Org. Chem.》;第68卷;第8471-8479页 *

Also Published As

Publication number Publication date
CN115124526A (en) 2022-09-30
CN113372343B (en) 2022-07-29
CN113372343A (en) 2021-09-10

Similar Documents

Publication Publication Date Title
US10323037B2 (en) Aminopyridazinone compounds as protein kinase inhibitors
CN111362975B (en) Naphthyridine compounds as JAK kinase inhibitors
EP3409673B1 (en) Pyrrolopyrimidine five-membered azacyclic derivative and application thereof
US20100204187A1 (en) Purine Derivatives
CN115124526B (en) Pyrrolo pyrimidine compound intermediate and preparation method thereof
JP7261428B2 (en) Pyrrolopyrimidine class compound and use thereof
KR20000068010A (en) Azolo Triazines and Pyrimidines
WO2017162215A1 (en) Substituted pyrrolopyrimidine cdk inhibitor, pharmaceutical composition containing same and use thereof
KR20190057069A (en) Novel JAK1 selective inhibitors and uses thereof
US20230017539A1 (en) Tri-heterocyclic compound as jak inhibitor, and use thereof
US20220144844A1 (en) Selective dihydropyrrolopyrimidine jak2 inhibitors
US20240002404A1 (en) Salt of pyrrolopyrimidine compound, crystalline form thereof, and application thereof
US20240132515A1 (en) Toxin molecule suitable for antibody-drug conjugate
CN114591333B (en) Preparation method of pyrrolopyrimidine compound
US20080033001A1 (en) Derivatives of Variolin B
CN115066425B (en) Application of JAK inhibitor in preparation of medicines for treating JAK kinase related diseases
TWI830024B (en) Tri-heterocyclic compounds as jak inhibitors and applications thereof
CN112209933B (en) BTK inhibitors containing 4-azacycloheptane
KR20230141050A (en) Polyheterocyclic derivatives containing pyrimidine and use thereof
CN117295739A (en) Crystal and salt of tri-heterocyclic compound and application thereof
CN116283993A (en) Pyrimidine compound and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant