WO2021018287A1 - Spiroaromatic compound, preparation and application thereof - Google Patents

Spiroaromatic compound, preparation and application thereof Download PDF

Info

Publication number
WO2021018287A1
WO2021018287A1 PCT/CN2020/106214 CN2020106214W WO2021018287A1 WO 2021018287 A1 WO2021018287 A1 WO 2021018287A1 CN 2020106214 W CN2020106214 W CN 2020106214W WO 2021018287 A1 WO2021018287 A1 WO 2021018287A1
Authority
WO
WIPO (PCT)
Prior art keywords
unsubstituted
substituted
halogenated
alkyl
group
Prior art date
Application number
PCT/CN2020/106214
Other languages
French (fr)
Chinese (zh)
Inventor
郑乾刚
曾庆龙
许明
Original Assignee
上海奕拓医药科技有限责任公司
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 上海奕拓医药科技有限责任公司 filed Critical 上海奕拓医药科技有限责任公司
Priority to CN202080053803.7A priority Critical patent/CN114269746A/en
Publication of WO2021018287A1 publication Critical patent/WO2021018287A1/en

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
    • C07D471/10Spiro-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
    • A61K31/495Heterocyclic 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
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring 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
    • A61K31/495Heterocyclic 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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 invention discloses a spiroaromatic ring compound, its pharmaceutically acceptable salt, or its solvate, isotope substitution, prodrug or metabolite.
  • the present invention also provides a method for preparing the compound, a pharmaceutical composition containing the compound, and the use of the compound for preparing a medicine for treating diseases or disorders related to abnormal SHP2 activity.
  • Protein tyrosine phosphatase SHP2 occupies an extremely important position in the process of cell signal transduction, and is a target for the development of treatment of major diseases such as diabetes, autoimmune diseases and cancer.
  • SHP2 is mutated or highly expressed in many diseases, such as Noonan Syndrome, Leopard Syndrome, Juvenile Myelomonocytic Leukemia, Neuroblastoma, Melanoma, Acute Myeloid Leukemia, Breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large cell lymphoma and glioblastoma, etc.
  • SHP2 is involved in multiple tumor cell signal transduction pathways, such as MAPK, JAK/STAT and PI3K/Akt.
  • SHP2 is also responsible for the signal transduction of the PD1-PDL1 immunosuppressive pathway. Therefore, inhibiting the activity of SHP2 can reverse the immunosuppression in the tumor microenvironment.
  • SHP2 is composed of two N-terminal Src homology 2 domains (N-SH2 and C-SH2) and a protein tyrosine phosphatase catalytic domain (PTP).
  • N-SH2 binds to PTP to form a ring structure, thereby hindering the binding of PTP to the substrate, so that the enzyme catalytic activity is inhibited; when the tyrosine of the upstream receptor protein is phosphorylated, N- SH2 binds to it, and the PTP catalytic domain is released to exert phosphatase activity.
  • SHP2 inhibitors are mainly based on allosteric inhibitors in the non-catalytic region, such as some compounds disclosed in WO2015107493A1, WO2016203404A1, WO2016203406A1, WO2017216706A1, WO2017211303A1, CN201710062495, WO2018136265A1WO2018057884, etc.
  • Recent studies have shown that SHP2, as a novel druggable target, has attracted more and more attention. Therefore, there is an urgent need in this field to develop SHP2 inhibitors with novel structure, good biological activity and high druggability.
  • the object of the present invention is to provide a compound of formula I or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the compound or a pharmaceutically acceptable salt thereof, and the compound or the pharmaceutical composition in the prevention and treatment of SHP2 Application in abnormally related diseases or conditions.
  • the first aspect of the present invention provides a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
  • R 1 is selected from H, -halogen, -CN, -OH, -NO 2 , HSO 3 -, unsubstituted or halogenated C1-C6 alkylsulfonyl, unsubstituted or halogenated C1-C6 alkylcarboxy, Unsubstituted or halogenated C1-C6 alkylamino, unsubstituted or halogenated C1-C6 alkyl, unsubstituted or halogenated C1-C6 alkoxy, unsubstituted or halogenated C1-6 alkoxy Carbonyl, unsubstituted or halogenated C1-6 alkylcarbonyl, unsubstituted or halogenated C2-C6 alkenylcarbonyl, unsubstituted or halogenated C1-C6 alkoxy-O-C1-C6 alkyl, unsubstituted Substituted or substituted 3-8 member
  • R 2 is selected from H, -halogen, amino, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl;
  • R 3 is selected from H, substituted or unsubstituted 3-8 membered cycloalkyl, substituted or unsubstituted 3-8 membered heterocyclic group, substituted or unsubstituted 5-10 membered aryl, substituted or unsubstituted 5 -10 membered heteroaryl; said heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
  • X and Y are each independently selected from N or CR 4 ;
  • R 4 is selected from H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, (substituted or unsubstituted C1-C6 Alkyl)SO-, (substituted or unsubstituted C1-C6 alkyl)SO 2 -;
  • the ring may contain 0-4 options Heteroatoms from the following group: N, O or S; the condition is that X and Y cannot be N at the same time;
  • substituted means that one or more hydrogen atoms on the group are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -N (unsubstituted or (Halogenated C1-C6 alkyl) 2 , -CN, unsubstituted or halogenated C1-C8 alkyl, unsubstituted or halogenated C1-C8 alkoxy, unsubstituted or halogenated C1-C8 alkoxy Group-C1-C8 alkyl, unsubstituted or halogenated C3-C8 cycloalkyl, unsubstituted or halogenated C3-C8 cycloalkyl-C1-C8 alkyl, unsubstituted or halogenated C1-C6 alkane Carbonyl, unsubstituted or halogenated C1-D, halogen, -
  • the compound of the present invention or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof is shown in the following formula II:
  • X and Y are each independently selected from N or C;
  • R 4a and R 4b are each independently selected from none, H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, ( Substituted or unsubstituted C1-C6 alkyl) SO-, (substituted or unsubstituted C1-C6 alkyl) SO 2 -;
  • the compound of the present invention or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof is shown in the following formula III :
  • X and Y are independently selected from N or C; X and Y cannot be N at the same time;
  • Ring A is a substituted or unsubstituted 5-10 membered aromatic ring, a substituted or unsubstituted 5-10 membered heteroaromatic ring or a substituted or unsubstituted 5-10 membered heterocyclic group; the ring may contain 0-4 A heteroatom selected from the following group: N, O or S;
  • substituted means that one or more hydrogen atoms on ring A are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -NH (unsubstituted or halogenated C1-C6 alkyl), -N (unsubstituted or halogenated C1-C6 alkyl) 2 , -CN, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy Group, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -;
  • the ring A is selected from: substituted or unsubstituted three-membered (hetero) ring, substituted or unsubstituted four-membered (hetero) ring, substituted or unsubstituted five-membered (hetero) ring , Substituted or unsubstituted six-membered (hetero) ring, substituted or unsubstituted five-membered aromatic (hetero) ring, substituted or unsubstituted six-membered aromatic (hetero) ring, substituted or unsubstituted seven-membered aromatic (hetero) Ring, substituted or unsubstituted five-membered six-membered (hetero) ring, substituted or unsubstituted six-membered (hetero) ring;
  • the heterocyclic ring may contain 1-4 heteroatoms selected from the following group : N, O or S;
  • the ring A is selected from any one of the following groups:
  • the above-mentioned ring A can be optionally substituted, and the substituents are selected from halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1 -C6 alkyl)SO 2 -.
  • the ring A is selected from any one of the following groups:
  • R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, substituted or Unsubstituted benzenesulfonyl, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl, unsubstituted or substituted 5-10 membered heteroaryl group; where the benzenesulfonyl group, carbocyclic group, heterocyclic group;
  • R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, substituted Or an unsubstituted benzenesulfonyl group, and any of the following groups that may be optionally substituted:
  • W is selected from C, O, S or N; wherein, when the benzenesulfonyl group and the optionally substituted ring group are substituted, the number of substituents is 1-4, selected from H, NH 2 , Halogen, halogenated or unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, and (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
  • R 3 is selected from H, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl group and unsubstituted or substituted 5- 10-membered heteroaryl group; when the carbocyclic group, heterocyclic group, aryl group and heteroaryl group are substituted, the number of substituents is 1-4, selected from H, NH 2 , halogen, halogenated Or unsubstituted C3-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N
  • R 3 is selected from H and any of the following cyclic groups which may be optionally substituted:
  • W is selected from O, S or N; wherein, when the cyclic group is substituted, the number of substituents is 1-4, and the substituents are selected from H, NH 2 , halogen, unsubstituted or halogenated C1- C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkyl -NH-.
  • R 2 is selected from H, -halogen, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, is selected from the following structures:
  • the second aspect of the present invention provides a use of the compound of formula I as described in the first aspect of the present invention for:
  • the third aspect of the present invention provides a pharmaceutical composition, the pharmaceutical composition comprising:
  • the fourth aspect of the present invention provides a method for inhibiting the activity of SHP2, said method comprising the steps of: administering an inhibitory effective amount of the compound of formula I as described in the first aspect of the present invention or its pharmaceutically acceptable The accepted salt, or an effective amount of the pharmaceutical composition according to the third aspect of the present invention is administered to the subject of inhibition.
  • the inventors prepared a new type of allosteric inhibitor compound represented by formula I, which can "lock" the self-inhibition with weak SHP2 activity by binding to the non-catalytic region of SHP2 State, so as to achieve the purpose of inhibiting its activity.
  • the compound of the present invention exhibits good biological activity and druggability, and has a good prospect for drug development. It has an inhibitory effect on SHP2 at a very low concentration (which can be as low as 100nM/L). The activity is quite excellent, so it can be used to treat SHP2 related diseases or disorders, such as tumors. Based on the above findings, the inventor completed the present invention.
  • substituent When a substituent is described by a conventional chemical formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the structural formula is written from right to left. For example, -CH 2 O- is equivalent to -OCH 2 -.
  • halogen means fluorine, chlorine, bromine or iodine.
  • Niro refers to -NO 2 .
  • Substituted amino refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, for example, monoalkylamino, dialkylamino, alkyl Amido, aralkylamino, heteroaralkylamino.
  • Carboxy refers to -COOH.
  • alkyl refers to a fully saturated linear or branched hydrocarbon chain group, It consists only of carbon atoms and hydrogen atoms, has, for example, 1 to 12 (preferably 1 to 8, more preferably 1 to 6) carbon atoms, and is connected to the rest of the molecule through a single bond, such as including but not limited to Methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2,2-dimethylpropyl , N-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl and decyl, etc.
  • the term “alkyl” refers to a fully saturated linear or branched hydrocarbon chain group, It consists only of carbon atoms and hydrogen atoms, has, for example, 1 to 12 (preferably
  • heterocyclic group means a group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen and sulfur Stable 3- to 20-membered non-aromatic cyclic group.
  • the heterocyclic group may be a monocyclic, bicyclic, tricyclic or more ring system, which may include a fused ring system, a bridged ring system or a spiro ring system; in the heterocyclic group
  • the nitrogen, carbon, or sulfur atoms of may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclic group may be partially or fully saturated.
  • heterocyclic groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2,7-diaza-spiro[3.5]non Alkyl-7-yl, 2-oxa-6-aza-spiro[3.3]heptane-6-yl, 2,5-diaza-bicyclo[2.2.1]heptan-2-yl, aza Cyclobutanyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxopentyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, Imidazolidinyl, quinazinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, in
  • the nitrogen, carbon or sulfur atoms in the heteroaryl group can be optionally oxidized; the nitrogen atom can be optionally quaternized.
  • the heteroaryl group is preferably a stable 5- to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, and more preferably contains 1 to 4 selected heteroatoms.
  • heteroarylalkyl refers to the above-defined alkyl group substituted by the above-defined heteroaryl group.
  • “optionally” or “optionally” means that the event or condition described later may or may not occur, and the description includes both occurrence and non-occurrence of the event or condition.
  • “optionally substituted aryl group” means that the aryl group is substituted or unsubstituted, and the description includes both substituted aryl groups and unsubstituted aryl groups.
  • pharmaceutically acceptable salt includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to a salt formed with an inorganic acid or an organic acid that can retain the biological effectiveness of the free base without other side effects.
  • Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, etc.
  • organic acid salts include, but are not limited to, formate, acetate, and 2,2-dichloroacetate , Trifluoroacetate, propionate, caproate, caprylate, caprate, undecylenate, glycolate, gluconate, lactate, sebacate, hexanoate Acid salt, glutarate, malonate, oxalate, maleate, succinate, fumarate, tartrate, citrate, palmitate, stearate, oleate , Cinnamate, laurate, malate, glutamate, pyroglutamate, aspartate, benzoate, methanesulfonate,
  • pharmaceutical composition refers to a preparation of a compound of the present invention and a medium generally accepted in the art for delivering a biologically active compound to a mammal (such as a human).
  • the medium includes a pharmaceutically acceptable carrier.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which is conducive to the absorption of the active ingredient and thus the biological activity.
  • pharmaceutically acceptable refers to a substance (such as a carrier or diluent) that does not affect the biological activity or properties of the compound of the present invention, and is relatively non-toxic, that is, the substance can be administered to an individual without causing undesirable biological activity. Reacts or interacts in an undesirable manner with any components included in the composition.
  • the "tumor” of the present invention includes but is not limited to Noonan syndrome, Leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, sarcoma, melanoma, articular chondroma, cholangiomas, leukemia, breast cancer, stomach Intestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, esophageal cancer, pancreatic cancer, lung squamous cell carcinoma, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell carcinoma, cervix Cancer, ovarian cancer, bowel cancer, nasopharyngeal cancer, brain cancer, bone cancer, kidney cancer, oral cancer/head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, anaplastic large cell lymphoma, or glioblast Tumors and other diseases.
  • treatment and other similar synonyms include the following meanings:
  • an effective amount refers to at least one agent or compound that is sufficient to relieve one or more symptoms of the disease or condition being treated after administration The amount.
  • the result can be a reduction and/or alleviation of signs, symptoms or causes, or any other desired changes in the biological system.
  • the "effective amount” for treatment is the amount of the composition containing the compound disclosed herein that is required to provide significant disease relief clinically. Techniques such as dose escalation tests can be used to determine the effective amount suitable for any individual case.
  • administration refers to methods capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, transduodenal routes, parenteral injections (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration.
  • parenteral injections including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion
  • topical administration and rectal administration.
  • Those skilled in the art are familiar with the application techniques that can be used for the compounds and methods described herein, for example in Goodman and Gilman, The Pharmaceutical Basis of Therapeutics, current ed.; Pergamon; and Remington's, Pharmaceutical (current edition), Mack Publishing Co., Those discussed in Easton, Pa.
  • the compounds and compositions discussed herein are administered orally.
  • drug combination refers to drug treatments obtained by mixing or combining more than one active ingredient. It includes fixed and non-fixed combinations of active ingredients.
  • fixed combination refers to the simultaneous administration of at least one compound described herein and at least one synergistic agent to a patient in the form of a single entity or a single dosage form.
  • non-fixed combination refers to the simultaneous administration, combination or sequential administration of at least one compound described herein and at least one synergistic agent to a patient in the form of separate entities. These also apply to cocktail therapy, such as the administration of three or more active ingredients.
  • the functional group of the intermediate compound may need to be protected by an appropriate protecting group.
  • Such functional groups include hydroxyl, amino, mercapto and carboxylic acid.
  • Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl (e.g. tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl) , Tetrahydropyranyl, benzyl, etc.
  • Suitable protecting groups for amino, amidino and guanidino include tert-butoxycarbonyl, benzyloxycarbonyl and the like.
  • Suitable protecting groups for mercapto include -C(O)-R" (wherein R" is an alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like.
  • Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
  • Protecting groups can be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protective groups is detailed in Greene, T.W. and P.G.M. Wuts, Protective Groups in Organi Synthesis, (1999), 4th Ed., Wiley.
  • the protecting group can also be a polymer resin.
  • R 1 is selected from H, -halogen, -CN, -OH, -NO 2 , HSO 3 -, unsubstituted or halogenated C1-C6 alkylsulfonyl, unsubstituted or halogenated C1-C6 alkylcarboxy, Unsubstituted or halogenated C1-C6 alkylamino, unsubstituted or halogenated C1-C6 alkyl, unsubstituted or halogenated C1-C6 alkoxy, unsubstituted or halogenated C1-6 alkoxy Carbonyl, unsubstituted or halogenated C1-6 alkylcarbonyl, unsubstituted or halogenated C2-C6 alkenylcarbonyl, unsubstituted or halogenated C1-C6 alkoxy-O-C1-C6 alkyl, unsubstituted Substituted or substituted 3-8 member
  • R 2 is selected from H, -halogen, amino, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl;
  • R 3 is selected from H, substituted or unsubstituted 3-8 membered cycloalkyl, substituted or unsubstituted 3-8 membered heterocyclic group, substituted or unsubstituted 5-10 membered aryl, substituted or unsubstituted 5 -10 membered heteroaryl; said heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
  • X and Y are each independently selected from N or CR 4 ;
  • R 4 is selected from H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, (substituted or unsubstituted C1-C6 Alkyl)SO-, (substituted or unsubstituted C1-C6 alkyl)SO 2 -;
  • the ring may contain 0-4 heteroatoms selected from the group consisting of N, O or S; the condition is that X and Y cannot be N at the same time;
  • substituted means that one or more hydrogen atoms on the group are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -N (unsubstituted or (Halogenated C1-C6 alkyl) 2 , -CN, unsubstituted or halogenated C1-C8 alkyl, unsubstituted or halogenated C1-C8 alkoxy, unsubstituted or halogenated C1-C8 alkoxy Group-C1-C8 alkyl, unsubstituted or halogenated C3-C8 cycloalkyl, unsubstituted or halogenated C3-C8 cycloalkyl-C1-C8 alkyl, unsubstituted or halogenated C1-C6 alkane Carbonyl group, unsubstituted or halogenated C1
  • the ring formed is any one of the implementations herein The ring A described in the scheme.
  • the present invention provides a compound of Formula II, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
  • X and Y are each independently selected from N or C;
  • R 4a and R 4b are each independently selected from none, H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, ( Substituted or unsubstituted C1-C6 alkyl) SO-, (substituted or unsubstituted C1-C6 alkyl) SO 2 -;
  • the present invention provides a compound of Formula III, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
  • X and Y are independently selected from N or C; X and Y cannot be N at the same time;
  • Ring A is a substituted or unsubstituted 5-10 membered aromatic ring, a substituted or unsubstituted 5-10 membered heteroaromatic ring, a substituted or unsubstituted 5-10 membered heterocyclic ring, or a substituted or unsubstituted 5-10 membered ring Carbocyclic ring; said ring may contain 0-4 heteroatoms selected from the group consisting of N, O or S;
  • substituted means that one or more hydrogen atoms on ring A are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -NH (unsubstituted or halogenated C1-C6 alkyl), -N (unsubstituted or halogenated C1-C6 alkyl) 2 , -CN, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy Group, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -;
  • the ring A is selected from Self-substituted or unsubstituted three-membered (hetero) ring, substituted or unsubstituted four-membered (hetero) ring, substituted or unsubstituted five-membered (hetero) ring, substituted or unsubstituted six-membered (hetero) ring, substituted Or unsubstituted five-membered aromatic (hetero) ring, substituted or unsubstituted six-membered aromatic (hetero) ring, substituted or unsubstituted seven-membered aromatic (hetero) ring, substituted or unsubstituted five-membered and six-membered (hetero) ) Ring, substituted or unsubstituted or unsubstituted
  • the ring A is Choose from any one of the following groups:
  • the compound represented by formula III provided by the present invention or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, more preferably, the ring A Any one selected from the following group:
  • the above-mentioned ring A can be optionally substituted, and the substituents are selected from halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1 -C6 alkyl)SO 2 -.
  • ring A is optionally substituted :
  • the substituent when substituted, may be 1-2 substituents selected from halogen, C1-C6 alkyl and halogenated C1-C6 alkyl.
  • the R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, Substituted or unsubstituted benzenesulfonyl group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or Substituted 5-10 membered heteroaryl group; wherein the benzenesulfonyl group, carbocyclic group
  • R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, A substituted or unsubstituted benzenesulfonyl group, and any of the following groups which may be optionally substituted:
  • W is selected from C, O, S or N; wherein, when the benzenesulfonyl group and the optionally substituted ring group are substituted, the number of substituents is 1-4, selected from H, NH 2 , Halogen, halogenated or unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, and (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
  • R 1 in formula I, II and III is selected from: unsubstituted C1-C6 alkyl-SO 2 -, unsubstituted C2-C6 alkenyl carbonyl, unsubstituted or substituted 3-8 membered heterocycle Group, unsubstituted or substituted 3-8 membered cycloalkyl, unsubstituted or substituted 5-10 membered aryl, unsubstituted or substituted 5-10 membered heteroaryl; wherein, the heteroaryl, cycloalkane When the group, aryl group and heteroaryl group are substituted, the substituents are 1-3 substituents selected from halogen, unsubstituted C1-C6 alkyl and halogenated C1-C6 alkyl.
  • the heterocyclic group, cycloalkyl group, aryl group and heteroaryl group in R 1 may be selected from pyrazinyl, phenyl, azetidinyl, cyclobutanyl, oxetanyl And pyridyl, these groups can be optionally substituted with 1 or 2 C1-C6 alkyl groups.
  • R 2 It is selected from H, halogen, amino and unsubstituted C1-C6 alkyl; more preferably, R 2 is selected from H and amino.
  • the R 3 is selected from H, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl; wherein the ring group can be optionally substituted, the number of the substituent is 1-4; the substituent is selected from H, NH 2 , halogen, halogenated or Unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C
  • R 3 is selected from H
  • W is selected from O, S or N, and the cyclic group can be optionally substituted, the number of substituents is 1-4, and the substituents are selected from H, NH 2 , halogen, unsubstituted or halogenated C1 -C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkyl -NH-.
  • R 3 is selected from 5-10 membered heteroaryl groups optionally substituted by amino and halogen, preferably 5-10 membered nitrogen-containing heteroaryl groups, more preferably pyridyl optionally substituted by amino and halogen, more preferably any Select substituted by amino and halogen:
  • R 1 is selected From unsubstituted C1-C6 alkyl-SO 2 -, unsubstituted C2-C6 alkenyl carbonyl, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered cycloalkyl, Unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl group; wherein, when the heteroaryl group, cycloalkyl group, aryl group and heteroaryl group are substituted, the substituent is 1-3 substituents selected from halogen, unsubstituted C1-C6 alkyl and halogenated C1-C6 alkyl; more preferably, the heterocycl
  • R 2 is selected from H, halogen, amino and unsubstituted C1-C6 alkyl, more preferably selected from H and amino;
  • R 3 is selected from: 5-10 membered heteroaryl optionally substituted by amino and halogen, preferably A 5- to 10-membered nitrogen-containing heteroaryl group is more preferably a pyridyl group optionally substituted with 1 or 2 substituents selected from amino and halogen, and more preferably the following substituted with 2 substituents selected from amino and halogen Group:
  • Ring A is optionally substituted:
  • the substituent when substituted, may be 1-2 substituents selected from halogen, C1-C6 alkyl and halogenated C1-C6 alkyl.
  • R 1 is selected From unsubstituted C1-C6 alkyl-SO 2 -, unsubstituted C2-C6 alkenyl carbonyl, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered cycloalkyl, Unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl group; wherein, when the heteroaryl group, cycloalkyl group, aryl group and heteroaryl group are substituted, the substituent is 1-3 substituents selected from halogen, unsubstituted C1-C6 alkyl and halogenated C1-C6 alkyl; more preferably, the heterocycl
  • the wavy line on the group indicates the position where the group is attached to the rest of the compounds of formula I, II and III.
  • W can be -CH-, -CH 2 -or -NH-, and X or Y can be CH.
  • the compound provided by the present invention or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug, or metabolite thereof is selected from the following structures:
  • the compound of formula I can be prepared by the following route: starting from intermediate A and substituting R 1 X 1 to obtain I-1, removing tert-butoxycarbonyl and tert-butanesulfinyl with acid to obtain intermediate I-2, and finally and intermediate Form I-3 undergoes a substitution reaction to obtain a compound of formula I.
  • the compound of formula I can be prepared by the following route: starting from Intermediate A, the tert-butoxycarbonyl group is removed by trimethylsilyl trifluoromethanesulfonate to obtain 1-4, and then substituted with I-3 to obtain I-5.
  • the benzyloxycarbonyl group is removed with sodium hydroxide aqueous solution to obtain intermediate I-6, and then it is substituted with intermediate R 1 X 1 to obtain I-7, and finally the tert-butylsulfinyl group is removed with acid to obtain the compound of formula I.
  • R 1 , R 2 , R 3 , X and Y are as described above; X 1 and X 2 are halogen.
  • SHP2 has two N-terminal Src homolgy2 domains (N-SH2 and C-SH2), a catalytic domain (PTP) and a C-terminal tail.
  • the two SH2 domains control the subcellular localization and functional regulation of SHP2.
  • the molecule exists in an inactive conformation and inhibits its own activity via a binding network involving residues from the N-SH2 and PTP domains.
  • SHP2 binds to specific tyrosine-phosphorylation sites on docking proteins such as Gab1 and Gab2 via its SH2 domain. This caused a conformational change, which resulted in SHP2 activation.
  • the present invention can also be used to treat other diseases or disorders related to the abnormal activity of SHP2. Therefore, as a preferred embodiment, the present invention relates to a method for treating diseases or conditions selected from the group consisting of: NS, LS, JMML, AML, MDS, B-ALL, neuroblastoma, esophageal cancer, breast cancer, lung cancer, colon cancer , Stomach cancer, head and neck cancer.
  • the SHP2 inhibitor of the present invention can be combined with other pharmacologically active compounds or with two or more other pharmacologically active compounds, especially in the treatment of cancer.
  • the compound of formula (I) according to the present invention or a pharmaceutically acceptable salt thereof can be administered simultaneously, sequentially or separately in combination with one or more substances selected from the group consisting of chemotherapeutic agents, such as mitotic inhibitors, such as Taxanes, vinca alkaloids, paclitaxel, docetaxel, vinblastine, vinblastine, vinorelbine or vinflunine, other anticancer agents such as cisplatin, 5-fluorouracil or 5-fluoro-2 -4(1H,3H)-pyrimidinedione (5FU), flutamide or gemcitabine.
  • chemotherapeutic agents such as mitotic inhibitors, such as Taxanes, vinca alkaloids, paclitaxel, docetaxel, vinblastine, vinblastine, vinorelbine or vinflunine
  • the present invention relates to a method as described above, wherein the compound is administered parenterally.
  • a pharmaceutical composition for treating SHP2 related diseases or disorders is provided.
  • ice bath refers to -5°C to 0°C
  • room temperature refers to 10°C to 30°C
  • reflux temperature generally refers to the reflux temperature of the solvent under normal pressure.
  • the reaction overnight generally means 8-15 hours. In the following examples, where the specific operating temperature is not limited, all are performed at room temperature.
  • the separation and purification of the intermediate and the final product is by normal phase or reverse phase chromatographic column separation or other suitable methods.
  • the normal phase flash chromatography column uses ethyl acetate and n-hexane or methanol and dichloromethane as the mobile phase.
  • Reversed-phase preparative high pressure liquid chromatography (HPLC) uses C18 column and UV 214nm and 254nm to detect, and its mobile phase is A (water and 0.1% formic acid), B (acetonitrile) or mobile phase A (water and 0.1% carbonic acid) Hydrogen ammonium), B (acetonitrile).
  • Step 3 Add compound A-2 (34.6 g, crude product), THF (300 mL), H 2 O (30 mL) and triphenylphosphine (26.5 g, 101.0 mmol) into a dry 500 mL single-neck flask in sequence.
  • Step 10 Add compound A-9 (300 mg, 0.608 mmol), ethanol (3 mL) and 10% aqueous sodium hydroxide solution (3 mL) in a dry 25 mL single-neck flask in sequence.
  • a mixed solvent (10 mL) of DCM/MeOH (2:1) was added to the obtained residue, filtered, and the obtained filtrate was concentrated under reduced pressure.
  • Step 1 Add compound A-9 (330mg, 0.668mmol) and DCM (10mL) in a dry 25mL single-mouth flask, add TMSOTf (298mg, 1.34mmol) dropwise at 0°C, and stir the reaction solution at 20°C2 hour. LCMS detected that the reaction was complete. The reaction solution was quenched by adding methanol (1 mL) and saturated sodium bicarbonate solution (1 mL). The resulting mixture was diluted with water (20 mL) and extracted twice with dichloromethane (20 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain B1-1 (263 mg, crude product) as a colorless oil.
  • TMSOTf 298mg, 1.34mmol
  • Step 3 Add 8-iodoimidazo[1,2-c]pyrimidin-5-ol (0.522g, 2mmol) and phosphorus oxychloride (8mL) into a dry 50mL single-neck flask in sequence, slowly under the protection of nitrogen N,N-diisopropylethylamine (1 mL) was added dropwise, and then the mixture was heated to 120°C and stirred for 5 hours.
  • reaction solution was cooled to room temperature and concentrated in vacuo, and then quenched by adding saturated sodium bicarbonate solution, extracted with ethyl acetate (3 ⁇ 40 mL), dried over anhydrous sodium sulfate, filtered and concentrated, and the residue obtained
  • the product was purified by silica gel chromatography (0 to 30% ethyl acetate: petroleum ether gradient) to give a pale yellow solid 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (B3, 0.360g, yield: 55%).
  • Step 1 Add ammonia (25-28% solution, 10 mL) to 2,4-dichloro-6-methylpyrimidine (8.0 g, 49.08 mmol) in EtOH (60 mL) solution under ice water bath. The reaction was stirred at room temperature for 16 h, and ammonia water (25-28% solution, 5 mL) was added. The reaction was stirred at room temperature for 8 hours, and ammonia water (25-28% solution, 5 mL) was added. Stir at room temperature for 16h.
  • Step 2 Add NIS (3.29 g, 14.63 mmol) to a DMF (15 mL) solution of 2-chloro-6-methylpyrimidin-4-amine (B4-1, 1.4 g, 9.75 mmol). The reaction was stirred at 30°C for 16h. The reaction solution was poured into water, filtered, the filter cake was washed with saturated sodium sulfite solution, and washed with water to obtain a pale yellow solid 2-chloro-5-iodo-6-methylpyrimidin-4-amine (B4-2, 350mg, yield: 13% ).
  • Step 3 Dissolve 2-chloro-5-iodo-6-methylpyrimidin-4-amine (B4-2, 350 mg, 1.30 mmol) in an aqueous solution of chloroacetaldehyde (40 wt.% in H 2 O, 5 mL). Heat to 100°C and stir for 2h. The reaction liquid was cooled to room temperature, filtered, and the filter cake was washed with water to obtain a pale yellow solid 8-iodo-7-methylimidazo[1,2-c]pyrimidin-5-ol (B4-3, 330mg, yield: 67%) .
  • Step 4 Add DIPEA to 8-iodo-7-methylimidazo[1,2-c]pyrimidin-5-ol (B4-3, 320mg, 1.16mmol) dissolved in phosphorus oxychloride (10mL) (226mg, 1.75mmol). Heat to 100°C for 6h. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and EtOAc (30 mL) was added. Wash with saturated aqueous sodium bicarbonate (3 ⁇ 30 mL), saturated brine (3 ⁇ 30 mL), dry with anhydrous sodium sulfate, filter, and concentrate the filtrate to dryness under reduced pressure. The residue obtained is passed through silica gel chromatography (0 to 10%). Gradient methanol: dichloromethane) was purified to obtain 5-chloro-8-iodo-7-methylimidazo[1,2-c]pyrimidine (B4, 135 mg, yield: 40%) as a white solid.
  • Step 1 Add NIS (12.3g, 54.66mmol) to a solution of 4-amino-6-chloro-2-methylthiopyrimidine (8.0g, 45.55mmol) in dichloromethane (150mL) under ice water bath. The reaction was stirred at 30°C for 5 hours, and the reaction was completed. The reaction solution was concentrated under reduced pressure to dryness, then methanol (50mL) was added, poured into saturated sodium sulfite aqueous solution (150mL), filtered, and the filter cake was washed with water to obtain a pale yellow solid 4-amino-6-chloro-5-iodo-2-methylthio Pyrimidine (B5-1, 12.8 g, yield: 93%).
  • Step 2 Add 4-amino-6-chloro-5-iodo-2-methylthiopyrimidine (2.0 g, 6.63 mmol) to a 40 wt.% aqueous solution of chloroacetaldehyde (20 mL). Heat to 100°C for 3 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by silica gel chromatography (0 to 10% gradient methanol: dichloromethane) and washed with ethyl acetate to obtain a yellow solid, 7-chloro-8-iodo-5-(methylthio)imidazo [1,2-c] Pyrimidine (B5-2, 1.9 g, yield: 88%).
  • Step 3 Add 7-chloro-8-iodo-5-(methylthio)imidazo[1,2-c]pyrimidine (B5-2, 1.20g, 3.69mmol) in dichloromethane (20mL) under ice water bath M-CPBA (1.27g, 7.37mmol) was added to the solution. The reaction was stirred at 25°C for 16 hours. The reaction was quenched with ice water, washed with saturated sodium bicarbonate aqueous solution (3 ⁇ 20 mL), saturated brine (3 ⁇ 20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness under reduced pressure.
  • Step 1 Add 3-chloro-4-iodopyridin-2-amine (2.5g, 9.82mmol, 1.0eq), 4,5-bisdiphenylphosphine-9, to a dry 100mL round bottom three-necked flask in sequence 9-dimethylxanthene (341mg, 0.59mmol, 0.06eq), palladium acetate (110mg, 0.49mmol, 0.05eq), DIPEA (3.25mL, 19.6mmol, 2.0q), methyl 3-mercaptopropionate ( 1.19mL, 10.8mmol, 1.1eq) and 1,4-dioxane (32.5mL). Under stirring, replace with nitrogen three times, then heat to 100°C and react for 3 hours.
  • 9-dimethylxanthene 341mg, 0.59mmol, 0.06eq
  • palladium acetate 110mg, 0.49mmol, 0.05eq
  • DIPEA 3.25mL, 19.6mmol, 2.0q
  • reaction solution was cooled to room temperature, diluted with ethyl acetate (50mL) and filtered under reduced pressure. The filter cake was washed with ethyl acetate (25mL), the obtained filtrate was concentrated in vacuo, and the residue obtained was chromatographed on silica gel. Purification by method (0 to 30% ethyl acetate: petroleum ether) to obtain a yellow solid C1-1 (2.0 g, yield: 78%).
  • Step 2 In a dry 100mL round bottom three-necked flask, dissolve C1-1 (2g, 8.11mmol, 1.0eq) in tetrahydrofuran (28mL), under nitrogen protection, add ethanol to the reaction solution dropwise at room temperature Sodium (2.9g, 8.51mmol, 1.05eq, 20%wt), then stirred for one hour. After the completion of the reaction, it was diluted with dichloromethane (60 mL) and sonicated for 5 minutes, filtered under reduced pressure, and the filter cake was dried in vacuum to obtain a yellow solid C1 (1.4 g, yield: 89%).
  • Step 1 Add 2,5-dichloropyrazine (3g, 20.1mmol, 1.0eq), potassium carbonate (2.78g, 20.1mmol, 1.0eq), DMF (25mL) and 3 to a dry 100mL round-bottom flask. -Methyl mercaptopropionate (2.54g, 21.1mmol, 1.05eq), then stirred at 25 degrees Celsius for 18 hours. After the reaction, it was diluted with ethyl acetate (100 mL), washed twice with water (30 mL), then dried over anhydrous sodium sulfate, and filtered. The obtained filtrate was concentrated in vacuo, and the obtained residue was purified by silica gel chromatography (0 to 2.8% ethyl acetate: petroleum ether gradient) to obtain a yellow solid C2-1 (3.68 g, yield: 74%).
  • Step 2 In a dry 100mL round bottom three-necked flask, dissolve C2-1 (3.68g, 15.8mmol, 1.0eq) in tetrahydrofuran (50mL), under nitrogen protection, add dropwise to the reaction solution at room temperature Sodium ethoxide (5.65g, 16.6mmol, 1.05eq, 20%wt), then stirred for one hour. After the reaction was completed, half of the solvent was concentrated under reduced pressure, ether (200 mL) was added to the remaining reaction solution, precipitation was precipitated, and the filter cake was vacuum-dried to obtain a yellow solid C2-2 (2.5 g, yield : 84.2%).
  • Step 3 Under nitrogen protection, sequentially add 3-chloro-4-iodopyridin-2-amine (2g, 7.86mmol, 1.0eq) and 4,5-bis-two to a dry 100mL round bottom three-necked flask. Phenylphosphine-9,9-dimethylxanthene (363mg, 0.63mmol, 0.08eq), Pd 2 (dba) 3 (287mg, 0.31mmol, 0.04eq), DIPEA (2.6mL, 15.7mmol, 2.0eq) ), dioxane (48mL) and C2-2 (1.39g, 8.25mmol, 1.05eq), and then heated to 105 degrees Celsius for 14 hours.
  • Phenylphosphine-9,9-dimethylxanthene (363mg, 0.63mmol, 0.08eq)
  • Pd 2 (dba) 3 (287mg, 0.31mmol, 0.04eq)
  • DIPEA 2.6mL, 15.7
  • Step 2 Add compound 1-1 (40mg, 0.091mmol), methanol (1mL) and dioxane hydrochloride (1mL, 4.0M) to a 25mL round bottom flask in sequence, and the reaction solution was stirred at 20°C for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain yellow solid 1-2 (28 mg, hydrochloride, yield: 100%).
  • Step 3 Add compound 1-2 (28mg, 0.091mmol), acetonitrile (3.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-neck flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (32 mg, 0.118 mmol).
  • Step 1 Add compound A-8 (200mg, 0.407mmol), tetrahydrofuran/water (98:2,5mL) and sodium borohydride (46mg, 1.22mmol) to a 25mL round-bottom flask at -50°C. The solution was heated to 20°C under stirring for 3 hours. The TLC dot plate reaction is complete. The reaction solution was diluted with water (20 mL), and extracted with ethyl acetate (20 mL) three times. The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain A-9 (200 mg, yield: 99.5%) as a colorless oil.
  • Step 2 Add compound A-9 (200mg, 0.405mmol), ethanol (4mL) and 10% sodium hydroxide aqueous solution (2mL) in a dry 25mL single-necked flask in sequence.
  • a mixed solvent (10 mL) of DCM/MeOH (2:1) was added to the obtained residue, filtered, and the obtained filtrate was concentrated under reduced pressure.
  • Step 4 Add compound 2-1 (50 mg, 0.114 mmol), methanol (1 mL), and dioxane hydrochloride (1 mL, 4.0 M) to a 25 mL round bottom flask in sequence, and the reaction solution was stirred at 20° C. for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain a yellow solid 2-2 (35 mg, hydrochloride, yield: 100%).
  • Step 5 Add compound 2-2 (35mg, 0.114mmol), acetonitrile (3.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-neck flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (40 mg, 0.148 mmol).
  • the reaction solution was stirred at 90°C for 10 hours.
  • the TLC dot board shows that new dots are generated.
  • the reaction solution was concentrated under reduced pressure.
  • Step 1 Add compound A (120mg, 0.334mmol), iodobenzene (136mg, 0.668mmol), dioxane (12mL), Pd 2 (dba) 3 (30mg, 0.033mmol) to a 25mL round bottom flask in sequence , 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (39mg, 0.067mmol), and cesium carbonate (326mg, 1.00mmol), the reaction solution was stirred at 100°C under nitrogen protection for 5 hours. The reaction solution was diluted with water (30 mL), and extracted twice with ethyl acetate (30 mL).
  • Step 2 Add compound 3-1 (45mg, 0.103mmol), methanol (2mL) and dioxane hydrochloride (1mL, 4.0M) to a 25mL round bottom flask in sequence, and the reaction solution was stirred at 20°C for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain a yellow solid 3-2 (31 mg, hydrochloride, yield: 100%).
  • Step 3 Add compound 3-2 (31mg, 0.103mmol), acetonitrile (3.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-neck flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (C2, 37 mg, 0.134 mmol).
  • the reaction solution was stirred at 90°C for 7 hours.
  • the reaction solution was concentrated under reduced pressure.
  • Step 2 Add compound 5-1 (20 mg, 0.031 mmol), methanol (1 mL), and dioxane hydrochloride (0.5 mL, 4.0 M) to a round bottom flask in sequence, and the reaction solution was stirred at 20° C. for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure. The obtained residue was prepared and purified by HPLC to obtain 5 as a white solid (9 mg, formate, yield: 60.0%).
  • Step 1 Add compound 3-2 (35mg, 0.115mmol), acetonitrile (3mL), N,N-diisopropylethylamine (0.5mL) and 5-chloro-8-iodine to a dry 10mL single-necked flask.
  • Imidazole [1,2-c] pyrimidine B3, 32 mg, 0.115 mmol was then stirred at 90°C for 7 hours.
  • Boc 2 O 118 mg, 0.542 mmol was added to the reaction solution, and then the reaction was stirred at 50°C for 1 hour.
  • Step 2 Add 2-amino-3-chloropyridine-4-sodium sulfide (14mg, 0.077mmol), compound 9-1 (40mg, 0.070mmol), dioxane (1mL), and Pd to the round bottom flask in sequence 2 (dba) 3 (7mg, 0.008mmol), 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (9mg, 0.016mmol) and N,N-diisopropylethylamine ( 40mg, 0.308mmol), the reaction solution was pumped with nitrogen three times, and then stirred at 100°C for 6 hours.
  • Step 1 Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (A1, 45mg, 0.125mmol) dissolved in 5mL of anhydrous 1,4-dioxane, followed by adding 3-iodo-1-methyl-1H-pyrazole (4.114mg, 0.556mmol), anhydrous potassium tert-butoxide ( 70 mg, 0.624 mmol), Brettphos Pd Gen. 3 (23 mg, 0.0254 mmol), protected by argon, heated to 100°C, and reacted for 5 hours.
  • Step 2 Add (S)-4-(((S)-tert-butylsulfinyl)amino)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazepine Heterosspiro[4.5]decane-8-tert-butyl carboxylic acid (10-1, 34mg, 0.0773mmol) was dissolved in 5mL dichloromethane/methanol (1/1), under argon protection, the ice water was cooled to 0 After adding 4M HCl/1,4-dioxane (0.5 mL, 2.0 mmol), stirring at room temperature for 1 hour, the reaction was complete.
  • Step 3 The crude product (S)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (10-2, 0.0773mmol ) Dissolved in 5mL anhydrous acetonitrile, add DIPEA (0.5mL, 2.926mmol) and 3-chloro-4–((5-chloropyrazin-2-yl)thio)pyridin-2-amine (C2, 28mg, 0.1025mmol). Protected by argon, heated to 90°C and refluxed for 7 hours to complete the reaction.
  • Step 1 Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (A1, 100mg, 0.278mmol) dissolved in 5mL of anhydrous 1,4-dioxane, followed by adding 2-iodopyridine (114mg, 0.556mmol), anhydrous cesium carbonate (272mg, 0.835mmol), 4,5-bis-dioxane Phenylphosphine-9,9-dimethylxanthene (32mg, 0.0553mmol) and Pd 2 (dba) 3 (26mg, 0.0284mmol), protected by argon, heated to 100°C and reacted for 5 hours.
  • 2-iodopyridine 114mg, 0.556mmol
  • anhydrous cesium carbonate 272mg, 0.835mmol
  • Step 2 Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(pyridin-2-yl)-2,8-diazaspiro[4.5]decane- Tert-Butyl 8-carboxylate (11-1, 33mg, 0.07558mmol) was dissolved in 5mL dichloromethane/methanol (1/1), under the protection of argon, the temperature of ice water was cooled to 0 degrees, and 4M HCl/1 was added. 4-Dioxane (0.3 mL, 1.2 mmol) was stirred at room temperature for 1 hour, and the reaction was complete.
  • Step 3 Dissolve the crude (S)-2-(pyridin-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (12-2, 0.08923mmol) in 5mL anhydrous acetonitrile , DIPEA (0.5 mL, 2.926 mmol) and 3-chloro-4-((5-chloropyrazin-2-yl)thio)pyridin-2-amine (7, 32 mg, 0.1172 mmol) were added. Protected by argon, heated to 90° and refluxed for 7 hours, and the reaction was completed.
  • Step 1 Dissolve 3-bromo-6-chloropyrazine-2-amine (208mg, 1.0mmol) and 2-amino-3-chloropyridine-4-thiol (168mg, 1.05mmol) in dioxane ( 10mL), copper iodide (95mg, 0.5mmol) and 1,10-phenanthroline (108mg, 0.6mmol) and DIPEA (260mg, 2mmol) were added. The temperature was raised to 120 degrees and reacted overnight under nitrogen.
  • Step 2 Combine 3-((2-amino-3-chloropyridin-4-yl)thio)-6-chloropyrazine-2-amine (13-1, 45mg, 0.15mmol) and 2-phenyl- 2,8-Diazaspiro[4.5]dec-4-amine hydrochloride (50 mg, 0.16 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (180 mg, 1.47 mmol) was added. The temperature is increased to 110 degrees and the reaction is overnight.
  • Step 1 Combine 5-chloro-8-iodo-7-methylimidazo[1,2-c]pyrimidine (B4, 34mg, 0.12mmol) and 2-phenyl-2,8-diazaspiro[4.5 ] Deca-4-amine hydrochloride (3-2, 35 mg, 0.12 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.2 mL, 1.98 mmol) was added. The temperature was raised to 65°C and reacted for 2h.
  • Step 2 Add 8-(8-iodo-7-methylimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4 -Amine (50mg, 0.10mmol) and sodium 2-amino-3-chloropyridine-4-sulfide (37mg, 0.2mmol) were suspended in dioxane (10mL), DIPEA (0.2mL, 1.21mmol) was added, Xantphos ( 29 mg, 0.05 mmol) and Pd 2 (dba) 3 (45 mg, 0.05 mmol), replaced with argon three times. The temperature was raised to 120°C and reacted for 4 hours.
  • Step 1 To 7-chloro-8-iodo-5-(methylsulfinyl)imidazo[1,2-c]pyrimidine (B5, 222mg, 0.65mmol) and (S)-2-phenyl-2 Cs 2 CO 3 (2.1 g, 6.45 mmol) was added to the suspension of 8-diazaspiro[4.5]decane 4-amine (3-2, 220 mg, 0.72 mmol) in acetonitrile (30 ml). The temperature was raised to 30 degrees and reacted for 1 hour.
  • reaction solution was diluted with water (50ml), extracted with ethyl acetate (2 ⁇ 30ml), the organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to obtain a white solid (S)-8-(7- Chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec4-amine (15-1, 140mg, yield 42.4%).
  • Step 2 To (S)-8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5] Triethylamine (60 mg, 0.56 mmol) and (Boc) 2 O (120 mg, 0.56 mmol) were added to a solution of decyl 4-amine (15-1, 140 mg, 0.28 mmol) in dichloromethane (20 mL), and reacted at room temperature overnight.
  • reaction solution was concentrated and purified by column chromatography to obtain pale yellow solid tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2- Phenyl-2,8-diazaspiro[4.5]dec4-yl)carbamate (15-2, 150 mg, yield 89.8%).
  • Step 3 To tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazepine Spiro[4.5]dec4-yl)carbamate (15-2,130mg, 0.21mmol) in dioxane (120mL) was added with 2-amino-3-chloropyridine-4-sodium thiosulfate ( C1, 115mg, 0.64mmol), cuprous iodide (40mg, 0.21mmol) and o-phenanthroline (20mg, 0.11mmol), replaced with argon three times, heated to 110°C, and reacted overnight.
  • 2-amino-3-chloropyridine-4-sodium thiosulfate C1, 115mg, 0.64mmol
  • cuprous iodide 40mg, 0.21mmol
  • o-phenanthroline 20m
  • Step 4 To tert-butyl (S)-(8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroindole[1,2-c]pyrimidine -5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (15-3, 20mg, 0.03mmol) in dichloromethane (4mL) Trifluoroacetic acid (2mL) was added and reacted for 2 hours at room temperature.
  • Step 1 To 7-chloro-8-iodo-5-(methylsulfinyl)imidazo[1,2-c]pyrimidine (B5, 222mg, 0.65mmol) and (S)-2-phenyl-2 Add Cs 2 CO 3 (2.1g, 6.45mmol) to the suspension of ,8-diazaspiro[4.5]decane 4-amine (3-2, 220mg, 0.72mmol) in acetonitrile (30ml) and raise the temperature to 30°C , Reaction for 1 hour.
  • reaction solution was diluted with water (50ml), extracted with ethyl acetate (2 ⁇ 30ml), the organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to obtain a white solid (S)-8-(7- Chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec4-amine (16-1, 140mg, yield 42.4%).
  • Step 2 To (S)-8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5] Triethylamine (60 mg, 0.56 mmol) and (Boc) 2 O (120 mg, 0.56 mmol) were added to the solution of decyl 4-amine (16-1, 140 mg, 0.28 mmol) in dichloromethane (20 mL), and reacted at room temperature overnight.
  • reaction solution was concentrated and purified by column chromatography to obtain pale yellow solid tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2- Phenyl-2,8-diazaspiro[4.5]dec4-yl)carbamate (16-2, 150 mg, yield 89.8%).
  • Step 3 To tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazepine Spiro[4.5]dec4-yl)carbamate (16-2, 130mg, 0.21mmol) in dioxane (120mL) was added with 2-amino-3-chloropyridine-4-sodium thiosulfate ( C1, 115mg, 0.64mmol), cuprous iodide (40mg, 0.21mmol) and o-phenanthroline (20mg, 0.11mmol), replaced with argon three times, heated to 110°C, and reacted overnight.
  • 2-amino-3-chloropyridine-4-sodium thiosulfate C1, 115mg, 0.64mmol
  • cuprous iodide 40mg, 0.21mmol
  • o-phenanthroline 20m
  • Step 4 Under the protection of argon, add tert-butyl(S)-(8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroindole [1, 2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-3, 30mg, 0.05mmol) in DMF Sodium azide (12mg, 0.19mmol) and potassium carbonate (26mg, 0.19mmol) were added to the mixture, replaced with argon three times, heated to 100°C, and reacted overnight.
  • reaction solution was diluted with ethyl acetate (50mL), followed by water ( 3 ⁇ 50mL) and saturated brine (2 ⁇ 50mL) washed, dried, filtered, and concentrated to obtain a pale yellow solid tert-butyl (S)-(8-(7-amino-8-((2-amino-3-chloropyridine) -4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-4, 20mg).
  • Step 5 To tert-butyl (S)-(8-(7-amino-8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidine- 5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-4, 30mg, 30%, 0.014mmol) in dichloromethane (10mL ) Trifluoroacetic acid (1 mL, 1.35 mmol) was added to the solution and reacted at room temperature for 1 h.
  • reaction solution was concentrated, diluted with dichloromethane (50mL), washed with saturated sodium bicarbonate and saturated brine successively, dried, filtered, concentrated, and purified by Prep-HPLC to obtain a white solid (S)-8-(7-amino-8) -((2-Amino-3-chloropyridinyl-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[ 4.5] Trifluoroacetate of decyl 4-amine (16, 2.5 mg, yield 30.0%).
  • Step 1 To tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylate ( A1, 100mg, 0.28mmol) in 1,4-dioxane (20mL) solution was sequentially added 4-iodobenzonitrile (128mg, 0.56mmol), Cs 2 CO 3 (272mg, 0.42mmol) and Xantphos (32mg , 0.03mmol). Pd 2 (dba) 3 (30 mg, 0.01 mmol) was added after 3 times of vacuum nitrogen replacement. After the addition, the vacuum nitrogen is replaced 3 times. Heat to 110°C for 12h.
  • reaction solution was filtered to remove insoluble materials, and the filtrate was concentrated under reduced pressure to dry to obtain a brown oil, which was purified by silica gel column chromatography to obtain a brown solid tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino group )-2-(4-cyanophenyl)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (17-1, 103 mg, yield: 80.4%).
  • Step 2 Add tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2-(4-cyanophenyl)-2,8-diazepine at 0°C Spiro[4.5]decane-8-t-butyl carboxylate (17-1, 103mg, 0.22mmol) in dichloromethane (5mL) was added dropwise HCl/1,4-dioxane (4.0M, 0.6mL ). Raise to room temperature and react for 1 hour.
  • Step 3 To (S)-4-(4-amino-2,8-diazaspiro[4.5]dec-2-yl)benzonitrile (17-2, 98mg, 0.22mmol) in isopropanol ( (12mL) DIPEA (1143mg, 8.85mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (138mg, 0.49mmol) were added to the solution. The temperature was raised to 70°C for 4 hours.
  • Step 4 To (S)-4-(4-amino-8-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2,8-diazaspiro[4.5]dec- 2-yl)benzonitrile (17-3, 95mg, 0.19mmol) in 1,4-dioxane (15mL) solution was added DIPEA (123mg, 0.28mmol), 2-amino-3-chloropyridine- 4-thiol sodium salt (52mg, 0.28mmol) and Xantphos (33mg, 0.057mmol). Pd 2 (dba) 3 (27 mg, 0.029 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again.
  • Step 1 To tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylate ( A1, 75mg, 0.19mmol) in 1,4-dioxane (15mL) solution was sequentially added Cs 2 CO 3 (191mg, 0.59mmol), 1-fluoro-4-iodobenzene (87mg, 0.39mmol) and Xantphos (24mg, 0.041mmol). Pd 2 (dba) 3 (19 mg, 0.021 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again.
  • Step 2 To tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5] To a solution of tert-butyl decane-8-carboxylate (18-1, 92 mg, 0.20 mmol) in dichloromethane (5 mL) was added dropwise HCl/1,4-dioxane (4.0M, 0.6 mL). React at room temperature for 1.5 hours.
  • Step 3 To (S)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5]dec-4-amine (18-2, 73mg, 0.20mmol) in isopropanol (12mL ) DIPEA (454mg, 3.51mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (65mg, 0.23mmol) were added to the solution. The temperature was raised to 70°C and reacted for 4 hours.
  • LCMS m/z 493 [M+H] + .
  • Step 1 To tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylate ( A1, 70mg, 0.19mmol) in 1,4-dioxane (15mL) solution was sequentially added Cs 2 CO 3 (191mg, 0.59mmol), 2-chloro-4-iodopyridine (94mg, 0.39mmol) and Xantphos (24mg, 0.041mmol). Pd 2 (dba) 3 (19 mg, 0.021 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again.
  • Step 2 To tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazepine [4.5] To a solution of tert-butyl decane-8-carboxylate (19-1, 75 mg, 0.16 mmol) in dichloromethane (5 mL) was added HCl/1,4-dioxane (4.0M, 0.6 mL). React at room temperature for 1.5 hours.
  • Step 3 To (S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-amine (19-2, 75mg, 0.16mmol) in isopropyl DIPEA (420mg, 3.25mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (60mg, 0.21mmol) were added to the alcohol (12mL) solution. The temperature was raised to 70°C and reacted for 4 hours.
  • Step 4 To (S)-2-(2-chloropyridin-4-yl)-8-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2,8-diazepine Spiro[4.5]dec-4-amine (46mg, 0.09mmol) in 1,4-dioxane (15mL) solution was added DIPEA (59mg, 0.46mmol), 2-amino-3-chloropyridine-4- Sodium mercaptan salt (26mg, 0.14mmol) and Xantphos (17mg, 0.029mmol). Pd 2 (dba) 3 (13 mg, 0.015 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again.
  • Step 1 Add 6-amino-2-chloro-5-iodopyrimidine-4-carboxylic acid methyl ester (20-1, 1.0 g, 3.19 mmol) to chloroacetaldehyde (40 wt.% aqueous solution, 12 mL). Heat to 80°C to react for 2 hours. After the reaction solution was cooled to room temperature, it was extracted with ethyl acetate (3 ⁇ 80 mL). The extracts were combined, washed with water (3 ⁇ 80 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness under reduced pressure.
  • Step 2 To (R)-N-((S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methyl Propane-2-sulfinamide (130mg, 0.35mmol) in isopropanol (20mL) was added DIPEA (135mg, 1.05mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine- Methyl 7-carboxylate (20-2, 132 mg, 0.39 mmol). After purging with vacuum nitrogen 3 times, the temperature was raised to 70°C and reacted for 3 hours.
  • Step 3 To methyl 5-((S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazepine Heterosspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7-carboxylate (20-3,200mg, 0.30mmol) of 1,4-dioxane (15mL) DIPEA (114mg, 0.89mmol) and 2-amino-3-chloropyridine-4-thiol sodium salt (108mg, 0.60mmol) were added to the solution in sequence.
  • DIPEA 1,4-dioxane
  • 2-amino-3-chloropyridine-4-thiol sodium salt 108mg, 0.60mmol
  • Step 4 To methyl 8-((2-amino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butylsulfinyl)amino )-2-(2-Chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7- To a solution of carboxylate (20-4, 160mg, 0.23mmol) in dichloromethane (20mL) was added di-tert-butyl dicarbonate (250mg, 1.14mmol), triethylamine (70mg, 0.68mmol) and DMAP (56mg, 0.46mmol).
  • Step 5 To methyl 8-((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butyl Sulfinyl)amino)-2-(2-chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c ] Pyrimidine-7-carboxylate (20-5, 100 mg, 0.11 mmol) and sodium hydroxide (18 mg, 0.45 mmol) in methanol (15 mL) was added with water (3 mL).
  • Step 6 To 8-((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butylsulfinyl) Acyl)amino)-2-(2-chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine To a solution of -7-carboxylic acid (20-6, 90 mg, 0.10 mmol) in tert-butanol (20 mL) was added triethylamine (102 mg, 1.00 mmol) and DPPA (83 mg, 0.30 mmol).
  • Step 7 To tert-butyl (8-(((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)- Tert-Butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)imidazo[1,2-c]pyrimidine -7-yl) carbamate (20-6, 70mg, purity 50.6%, 0.04mmol) in dichloromethane (20 mL) was added HCl/dioxane (3M, 4mL).
  • Step 1 To N-((S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methylpropane-2- DIPEA (145mg, 1.12mmol) and 7-chloro-8-iodo-5-(methylsulfinyl) imidazo[1, sulfinamide (83mg, 0.22mmol) in dichloromethane (20mL) solution were added successively 2-c] A solution of pyrimidine (154 mg, 0.45 mmol) in dichloromethane (3 mL). After 3 times of vacuum nitrogen replacement, the reaction was carried out at room temperature for 2 hours.
  • Step 2 To N-((S)-8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-(2-chloropyridin-4-yl)- 2,8-Diazaspiro[4.5]dec-4-yl)-2-methylpropane-2-sulfenamide (40mg, 0.06mmol) in 1,4-dioxane (15mL) solution DIPEA (0.2 mL, 1.98 mmol), 2-amino-3-chloropyridine-4-thiol sodium salt (18 mg, 0.10 mmol) was added.
  • the compound powder is dissolved in DMSO to make a mother liquor.
  • the compound stock solution was diluted with DMSO in 3-fold gradient, and the same compound was set to 10 different test concentrations. Take 1 ⁇ L of the compound at each concentration point into the well of the detection plate (Corning, Costar 3915), and set 2 parallel replicates for each concentration point.
  • the luminescence cell viability detection kit quantitatively measures intracellular ATP to detect the number of viable cells in culture.
  • the first step Seed MV4-11 cells in a 96-well plate, seed the cells into a 96-well plate at a density of 2500 cells per well, with a volume of 100 ⁇ L per well. Place it in a 37°C, 5% carbon dioxide incubator overnight.
  • Step 2 Compound treatment of cells.
  • the test compound was diluted 3 times, and a total of 8 concentration gradients were set; a certain volume of DMSO or test compound was added to each well, and each concentration was set to 2 replicates, and the final concentration of DMSO was controlled at 0.5%. Place in a 37°C 5% carbon dioxide incubator for 72h.
  • Step 3 Use Luminescent Cell Viability Assay Kit (Promega, G7570) detects the cell viability of the control and treatment groups. Add 50ul CellTiter-Glo to each well, mix well, and incubate at room temperature for 10 minutes. Use EnSpire (Perkin Elmer) to read the signal. The inhibition percentage (%) is calculated by the following formula:
  • Inhibition percentage (%) (1-compound treatment group signal value/DMSO treatment group signal value)*100.
  • CellTiter-Glo R luminescence cell viability detection kit is used to quantitatively determine intracellular ATP to detect the number of viable cells in culture.
  • the first step Seed KYSE520 cells in a 96-well plate, seed the cells in a 96-well plate at a density of 1500 cells per well, with a volume of 100 ⁇ L per well. Place it in a 37°C, 5% carbon dioxide incubator overnight.
  • Step 2 Compound treatment of cells.
  • the test compound was diluted 3 times, and a total of 8 concentration gradients were set; a certain volume of DMSO or test compound was added to each well, and each concentration was set to 2 replicates, and the final concentration of DMSO was controlled at 0.5%. Place in a 37°C 5% carbon dioxide incubator for 72 hours.
  • the following methods were used to determine the pharmacokinetic parameters of the compounds of this application.
  • the healthy male adult rats/mice used in the study were given a single intragastric administration of 5-100 mg/Kg for each group of animals. Fasting is from 10 hours before administration to 4 hours after administration. Blood was collected at different time points after administration, and the plasma content of the compound was determined (LC-MS/MS).
  • Use professional software to analyze (winnonlin) plasma concentration-time relationship and calculate the pharmacokinetic parameters of the compound. The results show that the compound of the present invention has excellent pharmacokinetic properties.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided is a compound with inhibitory activity on SHP2, a preparation method therefor, and a use thereof. Specifically, provided is a compound represented by formula (I), a pharmaceutically acceptable salt thereof, a solvate, an isotopically substituted compound, a polymorph, a prodrug, or a metabolite thereof. The definition of each group is as described in the specification. The compound has high inhibitory activity on SHP2 and therefore can be used to prevent or treat SHP2-related diseases.

Description

一种螺芳环化合物、其制备及应用A spiroaromatic compound, its preparation and application 技术领域Technical field
本发明公开了一种螺芳环化合物、其药学上可接受的盐、或其溶剂化物、同位素取代物、前药或代谢产物。本发明也提供了该类化合物的制备方法、含有该类化合物的药物组合物以及该类化合物用于制备治疗与SHP2活性异常相关疾病或病症的药物的用途。The invention discloses a spiroaromatic ring compound, its pharmaceutically acceptable salt, or its solvate, isotope substitution, prodrug or metabolite. The present invention also provides a method for preparing the compound, a pharmaceutical composition containing the compound, and the use of the compound for preparing a medicine for treating diseases or disorders related to abnormal SHP2 activity.
背景技术Background technique
蛋白酪氨酸磷酸酶SHP2,在细胞信号传导过程中占据及其重要的位置,是开发治疗糖尿病、自身免疫疾病和癌症等重大疾病的靶点。SHP2是在多种疾病中突变或者高表达,如努南综合征(Noonan Syndrome)、豹综合征(Leopard Syndrome)、青少年髓单核细胞白血病、成神经细胞瘤、黑色素瘤、急性髓性白血病、乳腺癌、食道癌、肺癌、结肠癌、头癌、成神经细胞瘤、头颈的鳞状细胞癌、胃癌、间变性大细胞淋巴瘤和成胶质细胞瘤等等。分子生物学研究表明,SHP2参与多个肿瘤细胞信号传导通路,如MAPK、JAK/STAT和PI3K/Akt等。同时,SHP2也负责PD1-PDL1免疫抑制通路的信号传导。因此,抑制SHP2的活性能够在逆转肿瘤微环境中的免疫抑制。Protein tyrosine phosphatase SHP2 occupies an extremely important position in the process of cell signal transduction, and is a target for the development of treatment of major diseases such as diabetes, autoimmune diseases and cancer. SHP2 is mutated or highly expressed in many diseases, such as Noonan Syndrome, Leopard Syndrome, Juvenile Myelomonocytic Leukemia, Neuroblastoma, Melanoma, Acute Myeloid Leukemia, Breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large cell lymphoma and glioblastoma, etc. Molecular biology studies have shown that SHP2 is involved in multiple tumor cell signal transduction pathways, such as MAPK, JAK/STAT and PI3K/Akt. At the same time, SHP2 is also responsible for the signal transduction of the PD1-PDL1 immunosuppressive pathway. Therefore, inhibiting the activity of SHP2 can reverse the immunosuppression in the tumor microenvironment.
SHP2由两个N-末端Src同源2结构域(N-SH2和C-SH2)和一个蛋白酪氨酸磷酸酶催化结构域(PTP)构成。在自抑制状态下,N-SH2与PTP结合形成一个环状结构,从而阻碍PTP与底物的结合,使得酶催化活性被抑制;当上游受体蛋白的酪氨酸被磷酸化后,N-SH2与之相结合,PTP催化域得到释放从而发挥出磷酸酶活性。SHP2 is composed of two N-terminal Src homology 2 domains (N-SH2 and C-SH2) and a protein tyrosine phosphatase catalytic domain (PTP). In the self-inhibition state, N-SH2 binds to PTP to form a ring structure, thereby hindering the binding of PTP to the substrate, so that the enzyme catalytic activity is inhibited; when the tyrosine of the upstream receptor protein is phosphorylated, N- SH2 binds to it, and the PTP catalytic domain is released to exert phosphatase activity.
目前针对SHP2抑制剂的开发,主要以非催化区的变构抑制剂为主,比如WO2015107493A1,WO2016203404A1,WO2016203406A1,WO2017216706A1,WO2017211303A1,CN201710062495,WO2018136265A1WO2018057884等中公开的一些化合物。最近的研究表明SHP2作为一个新颖的可成药靶点,引起了越来越多的关注。因此,本领域急需开发结构新颖、生物活性好,成药性高的SHP2抑制剂。At present, the development of SHP2 inhibitors is mainly based on allosteric inhibitors in the non-catalytic region, such as some compounds disclosed in WO2015107493A1, WO2016203404A1, WO2016203406A1, WO2017216706A1, WO2017211303A1, CN201710062495, WO2018136265A1WO2018057884, etc. Recent studies have shown that SHP2, as a novel druggable target, has attracted more and more attention. Therefore, there is an urgent need in this field to develop SHP2 inhibitors with novel structure, good biological activity and high druggability.
发明内容Summary of the invention
本发明的目的在于提供一种式I化合物或其药学上可接受的盐,含有该化合物或其药学上可接受的盐的药物组合物,以及该化合物或其药物组合物在预防和治疗与SHP2异常 相关的疾病或病症中的应用。The object of the present invention is to provide a compound of formula I or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the compound or a pharmaceutically acceptable salt thereof, and the compound or the pharmaceutical composition in the prevention and treatment of SHP2 Application in abnormally related diseases or conditions.
本发明的第一个方面是提供了一种式I所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物:The first aspect of the present invention provides a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
Figure PCTCN2020106214-appb-000001
Figure PCTCN2020106214-appb-000001
其中,among them,
R 1选自H、-卤素、-CN、-OH、-NO 2、HSO 3-、未取代或卤代的C1-C6烷基磺酰基、未取代或卤代的C1-C6烷基羧基、未取代或卤代的C1-C6烷基氨基、未取代或卤代的C1-C6烷基、未取代或卤代的C1-C6烷氧基、未取代或卤代的C1-6烷氧基羰基、未取代或卤代的C1-6烷基羰基、未取代或卤代的C2-C6烯烃基羰基、未取代或卤代的C1-C6烷氧基-O-C1-C6烷基、未取代或取代的3-8元环烷基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;所述的杂环基或杂芳基包含1-4个选自下组的杂原子:N、O或S; R 1 is selected from H, -halogen, -CN, -OH, -NO 2 , HSO 3 -, unsubstituted or halogenated C1-C6 alkylsulfonyl, unsubstituted or halogenated C1-C6 alkylcarboxy, Unsubstituted or halogenated C1-C6 alkylamino, unsubstituted or halogenated C1-C6 alkyl, unsubstituted or halogenated C1-C6 alkoxy, unsubstituted or halogenated C1-6 alkoxy Carbonyl, unsubstituted or halogenated C1-6 alkylcarbonyl, unsubstituted or halogenated C2-C6 alkenylcarbonyl, unsubstituted or halogenated C1-C6 alkoxy-O-C1-C6 alkyl, unsubstituted Substituted or substituted 3-8 membered cycloalkyl, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl, unsubstituted or substituted 5-10 membered heteroaryl; The heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
R 2选自H、-卤素、氨基、未取代或取代的C1-C6烷基氨基、未取代或取代的C1-C6烷基; R 2 is selected from H, -halogen, amino, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl;
R 3选自H、取代或未取代的3-8元环烷基、取代或未取代的3-8元杂环基、取代或未取代的5-10元芳基、取代或未取代的5-10元杂芳基;所述的杂环基或杂芳基包含1-4个选自下组的杂原子:N、O或S; R 3 is selected from H, substituted or unsubstituted 3-8 membered cycloalkyl, substituted or unsubstituted 3-8 membered heterocyclic group, substituted or unsubstituted 5-10 membered aryl, substituted or unsubstituted 5 -10 membered heteroaryl; said heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
X和Y分别独立地选自N或CR 4X and Y are each independently selected from N or CR 4 ;
R 4选自H、卤素、-CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的氨基、(取代或未取代的C1-C6烷基)SO-、(取代或未取代的C1-C6烷基)SO 2-; R 4 is selected from H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, (substituted or unsubstituted C1-C6 Alkyl)SO-, (substituted or unsubstituted C1-C6 alkyl)SO 2 -;
或者R 4与邻近的
Figure PCTCN2020106214-appb-000002
共同形成一个取代或未取代的5-10元芳环,取代或未取代的5-10元杂芳环或取代或未取代的5-10元杂环;所述环可以包含0-4个选自下组的杂原子:N、O或S;条件是,此时X和Y不能同时为N;
Or R 4 and adjacent
Figure PCTCN2020106214-appb-000002
Together to form a substituted or unsubstituted 5-10 membered aromatic ring, a substituted or unsubstituted 5-10 membered heteroaromatic ring or a substituted or unsubstituted 5-10 membered heterocyclic ring; the ring may contain 0-4 options Heteroatoms from the following group: N, O or S; the condition is that X and Y cannot be N at the same time;
Figure PCTCN2020106214-appb-000003
表示单键或双键;
Figure PCTCN2020106214-appb-000003
Represents a single bond or a double bond;
上述的任一“取代”指基团上的一个或多个氢原子被选自下组的取代基取代:-D、卤素、-OH、-NO 2、-NH 2、-N(未取代或卤代的C1-C6烷基) 2、-CN、未取代或卤代的C1-C8烷基、未取代或卤代的C1-C8烷氧基、未取代或卤代的C1-C8烷氧基-C1-C8烷基、未取代或卤代的C3-C8环烷基、未取代或卤代的C3-C8环烷基-C1-C8烷基、未取代或卤代的C1-C6烷基羰基、未取代或卤代的C1-C6烷氧基羰基、异羟肟酸基、未取代或卤代的C1-C6 烷基巯基、-S(O) 2N(未取代或卤代的C1-C6烷基) 2、-S(O) 2未取代或卤代的C1-C6烷基、-N(未取代或卤代的C1-C6烷基)S(O) 2N(未取代或卤代的C1-C6烷基) 2、-S(O)N(未取代或卤代的C1-C6烷基) 2、-S(O)(未取代或卤代的C1-C6烷基)、-N(未取代或卤代的C1-C6烷基)S(O)N(未取代或卤代的C1-C6烷基) 2、-N(未取代或卤代的C1-C6烷基)S(O)(未取代或卤代的C1-C6烷基)、未取代或卤代的5-8元芳基、未取代或卤代的5-8元杂芳基、未取代或卤代的4-8元饱和杂环或碳环;其中,所述的杂芳基包含1-4个选自下组的杂原子:N、O或S,所述的杂环包含1-4个选自下组的杂原子:N、O或S。 Any of the above "substituted" means that one or more hydrogen atoms on the group are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -N (unsubstituted or (Halogenated C1-C6 alkyl) 2 , -CN, unsubstituted or halogenated C1-C8 alkyl, unsubstituted or halogenated C1-C8 alkoxy, unsubstituted or halogenated C1-C8 alkoxy Group-C1-C8 alkyl, unsubstituted or halogenated C3-C8 cycloalkyl, unsubstituted or halogenated C3-C8 cycloalkyl-C1-C8 alkyl, unsubstituted or halogenated C1-C6 alkane Carbonyl, unsubstituted or halogenated C1-C6 alkoxycarbonyl, hydroxamic acid group, unsubstituted or halogenated C1-C6 alkyl mercapto, -S(O) 2 N (unsubstituted or halogenated C1-C6 alkyl) 2 , -S(O) 2 unsubstituted or halogenated C1-C6 alkyl, -N (unsubstituted or halogenated C1-C6 alkyl) S(O) 2 N (unsubstituted Or halogenated C1-C6 alkyl) 2 , -S(O)N (unsubstituted or halogenated C1-C6 alkyl) 2 , -S(O) (unsubstituted or halogenated C1-C6 alkyl) ), -N (unsubstituted or halogenated C1-C6 alkyl) S(O)N (unsubstituted or halogenated C1-C6 alkyl) 2 , -N (unsubstituted or halogenated C1-C6 alkane Group) S(O) (unsubstituted or halogenated C1-C6 alkyl), unsubstituted or halogenated 5-8 membered aryl, unsubstituted or halogenated 5-8 membered heteroaryl, unsubstituted or A halogenated 4-8 membered saturated heterocyclic ring or carbocyclic ring; wherein the heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S, and the heterocyclic ring contains 1-4 One heteroatom selected from the group: N, O or S.
作为一种优选的实施方式,本发明所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物如下述式II所示:As a preferred embodiment, the compound of the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof is shown in the following formula II:
Figure PCTCN2020106214-appb-000004
Figure PCTCN2020106214-appb-000004
其中,among them,
X和Y分别独立地选自N或C;X and Y are each independently selected from N or C;
R 4a和R 4b分别独立地选自无、H、卤素、-CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的氨基、(取代或未取代的C1-C6烷基)SO-、(取代或未取代的C1-C6烷基)SO 2-; R 4a and R 4b are each independently selected from none, H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, ( Substituted or unsubstituted C1-C6 alkyl) SO-, (substituted or unsubstituted C1-C6 alkyl) SO 2 -;
其他取代基的定义如上所述。The definition of other substituents is as described above.
作为另一种优选的实施方式,本发明所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物如下述式III所示:As another preferred embodiment, the compound of the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof is shown in the following formula III :
Figure PCTCN2020106214-appb-000005
Figure PCTCN2020106214-appb-000005
其中,among them,
X和Y分别独立地选自N或C;X和Y不能同时为N;X and Y are independently selected from N or C; X and Y cannot be N at the same time;
环A为取代或未取代的5-10元芳环,取代或未取代的5-10元杂芳环或取代或未取代的5-10元杂环基;所述的环可以包含0-4个选自下组的杂原子:N、O或S;Ring A is a substituted or unsubstituted 5-10 membered aromatic ring, a substituted or unsubstituted 5-10 membered heteroaromatic ring or a substituted or unsubstituted 5-10 membered heterocyclic group; the ring may contain 0-4 A heteroatom selected from the following group: N, O or S;
上述的“取代”指环A上的一个或多个氢原子被选自下组的取代基取代:-D、卤素、-OH、-NO 2、-NH 2、-NH(未取代或卤代的C1-C6烷基)、-N(未取代或卤代的C1-C6烷基) 2、-CN、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤 代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-; The above-mentioned "substituted" means that one or more hydrogen atoms on ring A are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -NH (unsubstituted or halogenated C1-C6 alkyl), -N (unsubstituted or halogenated C1-C6 alkyl) 2 , -CN, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy Group, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -;
其他取代基的定义如上所述。The definition of other substituents is as described above.
作为进一步优选的实施方式,所述的环A选自:取代或未取代的三元(杂)环、取代或未取代的四元(杂)环、取代或未取代的五元(杂)环、取代或未取代的六元(杂)环、取代或未取代的五元芳(杂)环、取代或未取代的六元芳(杂)环、取代或未取代的七元芳(杂)环、取代或未取代的五元并六元(杂)环、取代或未取代的六元并六元(杂)环;所述的杂环可以包含1-4个选自下组的杂原子:N、O或S;所述的环A可被任意取代,所述取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 As a further preferred embodiment, the ring A is selected from: substituted or unsubstituted three-membered (hetero) ring, substituted or unsubstituted four-membered (hetero) ring, substituted or unsubstituted five-membered (hetero) ring , Substituted or unsubstituted six-membered (hetero) ring, substituted or unsubstituted five-membered aromatic (hetero) ring, substituted or unsubstituted six-membered aromatic (hetero) ring, substituted or unsubstituted seven-membered aromatic (hetero) Ring, substituted or unsubstituted five-membered six-membered (hetero) ring, substituted or unsubstituted six-membered six-membered (hetero) ring; the heterocyclic ring may contain 1-4 heteroatoms selected from the following group : N, O or S; The ring A can be optionally substituted, and the substituent is selected from: halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO- , (Halogenated or unsubstituted C1-C6 alkyl) SO 2 -.
作为更进一步的优选方式,所述环A选自下组任意一个:As a further preferred manner, the ring A is selected from any one of the following groups:
Figure PCTCN2020106214-appb-000006
Figure PCTCN2020106214-appb-000006
Figure PCTCN2020106214-appb-000007
Figure PCTCN2020106214-appb-000007
上述环A可被任意取代,所述取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 The above-mentioned ring A can be optionally substituted, and the substituents are selected from halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1 -C6 alkyl)SO 2 -.
作为更进一步的优选方式,所述环A选自下组任意一个:As a further preferred manner, the ring A is selected from any one of the following groups:
Figure PCTCN2020106214-appb-000008
Figure PCTCN2020106214-appb-000008
上述环A可被任意取代,所述取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 The above-mentioned ring A can be optionally substituted, and the substituents are selected from halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1 -C6 alkyl)SO 2 -.
作为进一步的优选方式,本发明式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 1选自H、卤代或未取代的C1-C6烷基羰基、卤代或未取代的C2-C6烯烃基羰基、(卤代或未取代的C1-C6烷基)SO 2-、取代或未取代的苯磺酰基、未取代或取代的3-8元碳环基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;其中所述苯磺酰 基、碳环基、杂环基、芳基和杂芳基被取代时,取代基的个数为1-4个,选自H、NH 2、卤素、卤代或未取代的C3-C8环烷基、未取代或卤代的C1-C6烷基和(卤代或未取代的C1-C6烷基) 2N-。 As a further preferred mode, in the compounds represented by formulas I, II and III of the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotope substitutions, polymorphs, prodrugs or metabolites thereof, R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, substituted or Unsubstituted benzenesulfonyl, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl, unsubstituted or substituted 5-10 membered heteroaryl group; where the benzenesulfonyl group, carbocyclic group, heterocyclic group, aryl group and heteroaryl group are substituted, the number of substituents is 1-4, selected from H, NH 2 , Halogen, halogenated or unsubstituted C3-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl and (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
作为更进一步的优选方式,本发明式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 1选自H、卤代或未取代的C1-C6烷基羰基、卤代或未取代的C2-C6烯烃基羰基、(卤代或未取代的C1-C6烷基)SO 2-、取代或未取代的苯磺酰基,以及可被任选取代的以下任一基团: As a further preferred mode, the compounds represented by formulas I, II and III of the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, substituted Or an unsubstituted benzenesulfonyl group, and any of the following groups that may be optionally substituted:
Figure PCTCN2020106214-appb-000009
Figure PCTCN2020106214-appb-000009
其中W选自C、O、S或N;其中,所述苯磺酰基以及可被任选取代的环基被取代时,取代基的个数为1-4个,选自H、NH 2、卤素、卤代或未取代的C1-C8环烷基、未取代或卤代的C1-C6烷基和(卤代或未取代的C1-C6烷基) 2N-。 Wherein W is selected from C, O, S or N; wherein, when the benzenesulfonyl group and the optionally substituted ring group are substituted, the number of substituents is 1-4, selected from H, NH 2 , Halogen, halogenated or unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, and (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
作为进一步的优选方式,本发明式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 3选自H、未取代或取代的3-8元碳环基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基和未取代或取代的5-10元杂芳基;其中所述的碳环基、杂环基、芳基和杂芳基被取代时,取代基的个数为1-4个,选自H、NH 2、卤素、卤代或未取代的C3-C8环烷基、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-和卤代或未取代的C1-C6烷基-NH-。 As a further preferred mode, in the compounds represented by formulas I, II and III of the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotope substitutions, polymorphs, prodrugs or metabolites thereof, R 3 is selected from H, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl group and unsubstituted or substituted 5- 10-membered heteroaryl group; when the carbocyclic group, heterocyclic group, aryl group and heteroaryl group are substituted, the number of substituents is 1-4, selected from H, NH 2 , halogen, halogenated Or unsubstituted C3-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkane基-NH-.
作为更进一步的优选方式,本发明式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 3选自H和可被任选取代的以下任一环基: As a further preferred mode, the compounds represented by formulas I, II and III of the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, R 3 is selected from H and any of the following cyclic groups which may be optionally substituted:
Figure PCTCN2020106214-appb-000010
Figure PCTCN2020106214-appb-000010
Figure PCTCN2020106214-appb-000011
Figure PCTCN2020106214-appb-000011
其中W选自O、S或N;其中,所述环基被取代时,取代基的个数为1-4个,取代基选自H、NH 2、卤素、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-和卤代或未取代的C1-C6烷基-NH-。 Wherein W is selected from O, S or N; wherein, when the cyclic group is substituted, the number of substituents is 1-4, and the substituents are selected from H, NH 2 , halogen, unsubstituted or halogenated C1- C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkyl -NH-.
作为更进一步的优选方式,本发明式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 2选自H、-卤素、未取代或取代的C1-C6烷基氨基、未取代或取代的C1-C6烷基。 As a further preferred mode, the compounds represented by formulas I, II and III of the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, R 2 is selected from H, -halogen, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl.
作为进一步的优选方式,本发明所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,选自以下的结构:As a further preferred mode, the compound of the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, is selected from the following structures:
Figure PCTCN2020106214-appb-000012
Figure PCTCN2020106214-appb-000012
Figure PCTCN2020106214-appb-000013
Figure PCTCN2020106214-appb-000013
本发明的第二方面,提供了一种如本发明第一方面所述的式I化合物的用途,用于:The second aspect of the present invention provides a use of the compound of formula I as described in the first aspect of the present invention for:
(a)制备预防或治疗与SHP2活性异常相关的疾病或病症的药物;(a) Preparation of drugs for the prevention or treatment of diseases or disorders related to abnormal SHP2 activity;
(b)制备预防或治疗SHP2-介导的疾病或病症的药物;(b) Preparation of drugs for preventing or treating SHP2-mediated diseases or conditions;
(c)制备抑制SHP2活性的抑制剂药物;(c) Preparation of inhibitor drugs that inhibit SHP2 activity;
(d)体外非治疗性的抑制SHP2活性;(d) Non-therapeutic inhibition of SHP2 activity in vitro;
(e)体外非治疗性的抑制肿瘤细胞增殖;和/或(e) Non-therapeutic inhibition of tumor cell proliferation in vitro; and/or
(f)治疗与SHP2异常相关的疾病或病症。(f) Treatment of diseases or conditions related to abnormal SHP2.
在一个优选的实施方式中,所述的与SHP2活性异常有关的疾病选自下组:癌症;优选为努南综合症、豹综合症、青少年髓单核细胞白血病、成神经细胞瘤、黑色素瘤、急性髓性白血病、乳腺癌、食道癌、肺癌、结肠癌、头癌、成神经细胞瘤、头颈的鳞状细胞 癌、胃癌、间变性大细胞淋巴瘤或成胶质细胞瘤。In a preferred embodiment, the disease related to abnormal SHP2 activity is selected from the following group: cancer; preferably Noonan syndrome, leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma , Acute myeloid leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large cell lymphoma or glioblastoma.
本发明的第三方面,提供了一种药物组合物,所述的药物组合物包括:The third aspect of the present invention provides a pharmaceutical composition, the pharmaceutical composition comprising:
(i)有效量的式I化合物,或其药学上可接受的盐、对映异构体、非对映异构体、互变异构体、溶剂化物、同位素取代物、前药或代谢产物;和(i) An effective amount of the compound of formula I, or a pharmaceutically acceptable salt, enantiomer, diastereomer, tautomer, solvate, isotope substitution, prodrug or metabolite thereof ;with
(ii)药学上可接受的载体。(ii) A pharmaceutically acceptable carrier.
本发明的第四方面,提供了一种抑制SHP2活性的方法,所述的方法包括如下步骤:对抑制对象施用抑制有效量的如本发明第一方面所述的式I化合物或其药学上可接受的盐,或对抑制对象施用有效量的如本发明第三方面所述的药物组合物。The fourth aspect of the present invention provides a method for inhibiting the activity of SHP2, said method comprising the steps of: administering an inhibitory effective amount of the compound of formula I as described in the first aspect of the present invention or its pharmaceutically acceptable The accepted salt, or an effective amount of the pharmaceutical composition according to the third aspect of the present invention is administered to the subject of inhibition.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described in the following (such as the embodiments) can be combined with each other to form a new or preferred technical solution. Due to space limitations, I will not repeat them here.
具体实施方式Detailed ways
本发明人经过长期而深入的研究,制备了一类具有式I所示的新颖的变构抑制剂化合物,其能够通过与SHP2非催化区域的结合并“锁”住SHP2活性很弱的自抑制状态,从而达到抑制其活性的目的。本发明所述的化合物表现出很好的生物活性及可成药性,具有很好的药物开发前景,其在极低浓度(可低至≤100nM/L)下,即对SHP2产生抑制作用,抑制活性相当优异,因而可以用于治疗与SHP2相关的疾病或病症,如肿瘤。基于上述发现,发明人完成了本发明。After long-term and in-depth research, the inventors prepared a new type of allosteric inhibitor compound represented by formula I, which can "lock" the self-inhibition with weak SHP2 activity by binding to the non-catalytic region of SHP2 State, so as to achieve the purpose of inhibiting its activity. The compound of the present invention exhibits good biological activity and druggability, and has a good prospect for drug development. It has an inhibitory effect on SHP2 at a very low concentration (which can be as low as 100nM/L). The activity is quite excellent, so it can be used to treat SHP2 related diseases or disorders, such as tumors. Based on the above findings, the inventor completed the present invention.
术语the term
除非另有定义,否则本文所有科技术语具有的涵义与权利要求主题所属领域技术人员通常理解的涵义相同。除非另有说明,本文全文引用的所有专利、专利申请、公开材料通过引用方式整体并入本文。Unless otherwise defined, all scientific and technological terms herein have the same meanings as commonly understood by those skilled in the art to which the subject of the claims belongs. Unless otherwise specified, all patents, patent applications, and publications cited in this article are incorporated herein by reference in their entirety.
应理解,上述简述和下文的详述为示例性且仅用于解释,而不对本发明主题作任何限制。在本申请中,除非另有具体说明,否则使用单数时也包括复数。必须注意,除非文中另有清楚的说明,否则在本说明书和权利要求书中所用的单数形式包括所指事物的复数形式。还应注意,除非另有说明,否则所用“或”、“或者”表示“和/或”。此外,所用术语“包括”以及其它形式,例如“包含”、“含”和“含有”并非限制性,其可以是开放式、半封闭式和封闭式的。换言之,所述术语也包括“基本上由…构成”、或“由…构成”之义。It should be understood that the above brief description and the following detailed description are exemplary and only used for explanation, without any limitation on the subject of the present invention. In this application, unless specifically stated otherwise, the use of the singular number also includes the plural number. It must be noted that unless clearly stated otherwise in the context, the singular form used in this specification and claims includes the plural form of the things referred to. It should also be noted that the use of "or" and "or" means "and/or" unless stated otherwise. In addition, the term "including" and other forms, such as "including", "including" and "containing" are not restrictive, and they can be open, semi-closed and closed. In other words, the term also includes the meaning of "essentially composed of" or "consisting of".
可在参考文献(包括Carey and Sundberg"ADVANCED ORGANIC CHEMISTRY 4TH  ED."Vols.A(2000)and B(2001),Plenum Press,New York)中找到对标准化学术语的定义。除非另有说明,否则采用本领域技术范围内的常规方法,如质谱、NMR、IR和UV/VIS光谱法和药理学方法。除非提出具体定义,否则本文在分析化学、有机合成化学以及药物和药物化学的有关描述中采用的术语是本领域已知的。可在化学合成、化学分析、药物制备、制剂和递送,以及对患者的治疗中使用标准技术。例如,可利用厂商对试剂盒的使用说明,或者按照本领域公知的方式或本发明的说明来实施反应和进行纯化。通常可根据本说明书中引用和讨论的多个概要性和较具体的文献中的描述,按照本领域熟知的常规方法实施上述技术和方法。在本说明书中,可由本领域技术人员选择基团及其取代基以提供稳定的结构部分和化合物。The definitions of standard chemical terms can be found in references (including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4TH ED." Vols. A (2000) and B (2001), Plenum Press, New York). Unless otherwise specified, conventional methods within the technical scope of the art, such as mass spectrometry, NMR, IR, and UV/VIS spectroscopy and pharmacological methods, are used. Unless specific definitions are provided, the terms used in the descriptions of analytical chemistry, synthetic organic chemistry, and pharmaceuticals and medicinal chemistry herein are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, drug preparation, formulation and delivery, and treatment of patients. For example, the manufacturer's instructions for the use of the kit can be used, or the reaction and purification can be performed in a manner known in the art or the instructions of the present invention. Generally, the above-mentioned techniques and methods can be implemented according to the descriptions in a number of summary and more specific documents cited and discussed in this specification according to conventional methods well known in the art. In this specification, groups and their substituents can be selected by those skilled in the art to provide stable structural parts and compounds.
当通过从左向右书写的常规化学式描述取代基时,该取代基也同样包括从右向左书写结构式时所得到的在化学上等同的取代基。举例而言,-CH 2O-等同于-OCH 2-。 When a substituent is described by a conventional chemical formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the structural formula is written from right to left. For example, -CH 2 O- is equivalent to -OCH 2 -.
本文所用的章节标题仅用于组织文章的目的,而不应被解释为对所述主题的限制。本申请中引用的所有文献或文献部分包括但不限于专利、专利申请、文章、书籍、操作手册和论文,均通过引用方式整体并入本文。The chapter headings used in this article are only for the purpose of organizing the article, and should not be construed as a limitation on the subject. All documents or document parts cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and theses, are incorporated herein by reference in their entirety.
在本文中定义的某些化学基团前面通过简化符号来表示该基团中存在的碳原子总数。例如,C1-C6烷基是指具有总共1至6个碳原子的如下文所定义的烷基。简化符号中的碳原子总数不包括可能存在于所述基团的取代基中的碳。Certain chemical groups defined herein are preceded by simplified symbols to indicate the total number of carbon atoms present in the group. For example, C1-C6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the simplified notation does not include carbons that may be present in the substituents of the group.
除前述以外,当用于本申请的说明书及权利要求书中时,除非另外特别指明,否则以下术语具有如下所示的含义。In addition to the foregoing, when used in the specification and claims of this application, unless otherwise specified, the following terms have the following meanings.
在本申请中,术语“卤素”是指氟、氯、溴或碘。In this application, the term "halogen" means fluorine, chlorine, bromine or iodine.
“羟基”是指-OH基团。"Hydroxy" refers to the -OH group.
“羟基烷基”是指被羟基(-OH)取代的如下文所定义的烷基。"Hydroxyalkyl" refers to an alkyl group as defined below that is substituted with a hydroxyl group (-OH).
“羰基”是指-C(=O)-基团。"Carbonyl" refers to the -C(=O)- group.
“硝基”是指-NO 2"Nitro" refers to -NO 2 .
“氰基”是指-CN。"Cyano" refers to -CN.
“氨基”是指-NH 2"Amino" refers to -NH 2 .
“取代的氨基”是指被一个或两个如下文所定义的烷基、烷基羰基、芳烷基、杂芳烷基取代的氨基,例如,单烷基氨基、二烷基氨基、烷基酰氨基、芳烷基氨基、杂芳烷基氨基。"Substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, for example, monoalkylamino, dialkylamino, alkyl Amido, aralkylamino, heteroaralkylamino.
“羧基”是指-COOH。"Carboxy" refers to -COOH.
在本申请中,作为基团或是其它基团的一部分(例如用在卤素取代的烷基等基团中),术语“烷基”是指完全饱和的直链或支链的烃链基,仅由碳原子和氢原子组成、具有例如 1至12个(优选为1至8个,更优选为1至6个)碳原子,且通过单键与分子的其余部分连接,例如包括但不限于甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、2-甲基丁基、2,2-二甲基丙基、正己基、庚基、2-甲基己基、3-甲基己基、辛基、壬基和癸基等。就本发明而言,术语“烷基”指含有1至8个碳原子的烷基。In this application, as a group or a part of other groups (for example, used in halogen-substituted alkyl groups and other groups), the term "alkyl" refers to a fully saturated linear or branched hydrocarbon chain group, It consists only of carbon atoms and hydrogen atoms, has, for example, 1 to 12 (preferably 1 to 8, more preferably 1 to 6) carbon atoms, and is connected to the rest of the molecule through a single bond, such as including but not limited to Methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2,2-dimethylpropyl , N-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl and decyl, etc. For the purposes of the present invention, the term "alkyl" refers to an alkyl group containing 1 to 8 carbon atoms.
在本申请中,作为基团或是其它基团的一部分,术语“烯基”意指仅由碳原子和氢原子组成、含有至少一个双键、具有例如2至20个(优选为2至10个,更优选为2至6个)碳原子且通过单键与分子的其余部分连接的直链或支链的烃链基团,例如但不限于乙烯基、丙烯基、烯丙基、丁-1-烯基、丁-2-烯基、戊-1-烯基、戊-1,4-二烯基等。In this application, as a group or a part of other groups, the term "alkenyl" means consisting only of carbon atoms and hydrogen atoms, containing at least one double bond, having, for example, 2 to 20 (preferably 2 to 10 One, more preferably 2 to 6) carbon atoms and a straight or branched hydrocarbon chain group connected to the rest of the molecule through a single bond, such as but not limited to vinyl, propenyl, allyl, but- 1-alkenyl, but-2-enyl, pent-1-enyl, pent-1,4-dienyl, etc.
在本申请中,作为基团或是其它基团的一部分,术语“环烃基”或“碳环基”意指仅由碳原子和氢原子组成的稳定的非芳香族单环或多环烃基,其可包括稠合环体系、桥环体系或螺环体系,具有3至15个碳原子,优选具有3至10个碳原子,更优选具有3至8个碳原子,且其为饱和或不饱和并可经由任何适宜的碳原子通过单键与分子的其余部分连接。除非本说明书中另外特别指明,环烃基中的碳原子可以任选地被氧化。示例性的环烃基或碳环基包括C3-C8环烷基。更进一步地,环烃基的实例包括但不限于环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环辛基、1H-茚基、2,3-二氢化茚基、1,2,3,4-四氢-萘基、5,6,7,8-四氢-萘基、8,9-二氢-7H-苯并环庚烯-6-基、6,7,8,9-四氢-5H-苯并环庚烯基、5,6,7,8,9,10-六氢-苯并环辛烯基、芴基、二环[2.2.1]庚基、7,7-二甲基-二环[2.2.1]庚基、二环[2.2.1]庚烯基、二环[2.2.2]辛基、二环[3.1.1]庚基、二环[3.2.1]辛基、二环[2.2.2]辛烯基、二环[3.2.1]辛烯基、金刚烷基、八氢-4,7-亚甲基-1H-茚基和八氢-2,5-亚甲基-并环戊二烯基等。In this application, as a group or a part of other groups, the term "cycloalkyl" or "carbocyclyl" means a stable non-aromatic monocyclic or polycyclic hydrocarbon group composed of only carbon atoms and hydrogen atoms, It may include a fused ring system, a bridged ring system or a spiro ring system, having 3 to 15 carbon atoms, preferably 3 to 10 carbon atoms, more preferably 3 to 8 carbon atoms, and it is saturated or unsaturated It can be connected to the rest of the molecule through a single bond via any suitable carbon atom. Unless specifically indicated otherwise in this specification, the carbon atoms in the cyclic hydrocarbon group may be optionally oxidized. Exemplary cyclic hydrocarbon groups or carbocyclic groups include C3-C8 cycloalkyl groups. Furthermore, examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl , 1H-indenyl, 2,3-indanyl, 1,2,3,4-tetrahydro-naphthyl, 5,6,7,8-tetrahydro-naphthyl, 8,9-dihydro- 7H-benzocyclohepten-6-yl, 6,7,8,9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9,10-hexahydro-benzo ring Octenyl, fluorenyl, bicyclo[2.2.1]heptyl, 7,7-dimethyl-bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, bicyclo[2.2 .2]octyl, bicyclo[3.1.1]heptyl, bicyclo[3.2.1]octyl, bicyclo[2.2.2]octenyl, bicyclo[3.2.1]octenyl, adamantane Group, octahydro-4,7-methylene-1H-indenyl and octahydro-2,5-methylene- and cyclopentadienyl.
在本申请中,作为基团或是其它基团的一部分,术语“杂环基”意指由2至14个碳原子以及1至6个选自氮、磷、氧和硫的杂原子组成的稳定的3元至20元非芳香族环状基团。除非本说明书中另外特别指明,否则杂环基可以为单环、双环、三环或更多环的环体系,其可包括稠合环体系、桥环体系或螺环体系;其杂环基中的氮、碳或硫原子可任选地被氧化;氮原子可任选地被季铵化;且杂环基可为部分或完全饱和。杂环基可以经由碳原子或者杂原子并通过单键与分子其余部分连接。在包含稠环的杂环基中,一个或多个环可以是下文所定义的芳基或杂芳基,条件是与分子其余部分的连接点为非芳香族环原子。就本发明的目的而言,杂环基优选为包含1至3个选自氮、氧和硫的杂原子的稳定的4元至11元非芳香性单环、双环、桥环或螺环基团,更优选为包含1至3个选自氮、氧和硫的杂原子的稳定的4元至8元非芳香性单环、双环、桥环或螺环基团。杂环基的实例包括但不限于:吡咯烷基、吗啉基、哌嗪基、高哌嗪基、哌啶基、硫代吗啉基、2,7-二氮杂-螺[3.5]壬烷-7-基、2-氧杂-6-氮杂-螺[3.3]庚烷-6-基、2,5-二氮杂-双环[2.2.1]庚烷-2-基、氮 杂环丁烷基、吡喃基、四氢吡喃基、噻喃基、四氢呋喃基、噁嗪基、二氧环戊基、四氢异喹啉基、十氢异喹啉基、咪唑啉基、咪唑烷基、喹嗪基、噻唑烷基、异噻唑烷基、异噁唑烷基、二氢吲哚基、八氢吲哚基、八氢异吲哚基、吡咯烷基、吡唑烷基、邻苯二甲酰亚氨基等。In this application, as a group or part of another group, the term "heterocyclic group" means a group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen and sulfur Stable 3- to 20-membered non-aromatic cyclic group. Unless otherwise specified in this specification, the heterocyclic group may be a monocyclic, bicyclic, tricyclic or more ring system, which may include a fused ring system, a bridged ring system or a spiro ring system; in the heterocyclic group The nitrogen, carbon, or sulfur atoms of may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclic group may be partially or fully saturated. The heterocyclic group can be connected to the rest of the molecule via a carbon atom or a heteroatom and through a single bond. In heterocyclic groups containing fused rings, one or more rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the rest of the molecule is a non-aromatic ring atom. For the purpose of the present invention, the heterocyclic group is preferably a stable 4- to 11-membered non-aromatic monocyclic, bicyclic, bridged ring or spirocyclic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur The group is more preferably a stable 4- to 8-membered non-aromatic monocyclic, bicyclic, bridged ring or spirocyclic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclic groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2,7-diaza-spiro[3.5]non Alkyl-7-yl, 2-oxa-6-aza-spiro[3.3]heptane-6-yl, 2,5-diaza-bicyclo[2.2.1]heptan-2-yl, aza Cyclobutanyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxopentyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, Imidazolidinyl, quinazinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, indoline, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl , Phthalimide, etc.
在本申请中,作为基团或是其它基团的一部分,术语“芳基”意指具有6至18个碳原子(优选具有6至10个碳原子)的共轭烃环体系基团。就本发明的目的而言,芳基可以为单环、双环、三环或更多环的环体系,还可以与上文所定义的环烷基或杂环基稠合,条件是芳基经由芳香环上的原子通过单键与分子的其余部分连接。芳基的实例包括但不限于苯基、萘基、蒽基、菲基、芴基、2,3-二氢-1H-异吲哚基、2-苯并噁唑啉酮、2H-1,4-苯并噁嗪-3(4H)-酮-7-基等。In this application, as a group or a part of other groups, the term "aryl" means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms (preferably having 6 to 10 carbon atoms). For the purpose of the present invention, the aryl group can be a monocyclic, bicyclic, tricyclic or more cyclic ring system, and can also be fused with a cycloalkyl or heterocyclic group as defined above, provided that the aryl group passes through The atoms on the aromatic ring are connected to the rest of the molecule through a single bond. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthryl, fluorenyl, 2,3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazine-3(4H)-one-7-yl and the like.
在本申请中,术语“芳基烷基”是指被上文所定义的芳基所取代的上文所定义的烷基。In this application, the term "arylalkyl" refers to the above-defined alkyl group substituted by the above-defined aryl group.
在本申请中,作为基团或是其它基团的一部分,术语“杂芳基”意指环内具有1至15个碳原子(优选具有1至10个碳原子)和1至6个选自氮、氧和硫的杂原子的5元至16元共轭环系基团。除非本说明书中另外特别指明,否则杂芳基可为单环、双环、三环或更多环的环体系,还可以与上文所定义的环烷基或杂环基稠合,条件是杂芳基经由芳香环上的原子通过单键与分子的其余部分连接。杂芳基中的氮、碳或硫原子可任选地被氧化;氮原子可任选地被季铵化。就本发明的目的而言,杂芳基优选为包含1至5个选自氮、氧和硫的杂原子的稳定的5元至12元芳香性基团,更优选为包含1至4个选自氮、氧和硫的杂原子的稳定的5元至10元芳香性基团或者包含1至3个选自氮、氧和硫的杂原子的5元至6元芳香性基团。杂芳基的实例包括但不限于噻吩基、咪唑基、吡唑基、噻唑基、噁唑基、噁二唑基、异噁唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、苯并咪唑基、苯并吡唑基、吲哚基、呋喃基、吡咯基、三唑基、四唑基、三嗪基、吲嗪基、异吲哚基、吲唑基、异吲唑基、嘌呤基、喹啉基、异喹啉基、二氮萘基、萘啶基、喹噁啉基、蝶啶基、咔唑基、咔啉基、菲啶基、菲咯啉基、吖啶基、吩嗪基、异噻唑基、苯并噻唑基、苯并噻吩基、噁三唑基、噌啉基、喹唑啉基、苯硫基、中氮茚基、邻二氮杂菲基、异噁唑基、吩噁嗪基、吩噻嗪基、4,5,6,7-四氢苯并[b]噻吩基、萘并吡啶基、[1,2,4]三唑并[4,3-b]哒嗪、[1,2,4]三唑并[4,3-a]吡嗪、[1,2,4]三唑并[4,3-c]嘧啶、[1,2,4]三唑并[4,3-a]吡啶、咪唑并[1,2-a]吡啶、咪唑并[1,2-b]哒嗪、咪唑并[1,2-a]吡嗪等。In this application, as a group or part of another group, the term "heteroaryl" means having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 nitrogen atoms in the ring. A 5- to 16-membered conjugated ring system group of heteroatoms of, oxygen and sulfur. Unless otherwise specified in this specification, heteroaryl groups can be monocyclic, bicyclic, tricyclic or more cyclic ring systems, and can also be fused with cycloalkyl or heterocyclic groups as defined above, provided that the hetero The aryl group is connected to the rest of the molecule via a single bond through an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group can be optionally oxidized; the nitrogen atom can be optionally quaternized. For the purpose of the present invention, the heteroaryl group is preferably a stable 5- to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, and more preferably contains 1 to 4 selected heteroatoms. A stable 5- to 10-membered aromatic group from heteroatoms of nitrogen, oxygen, and sulfur or a 5- to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, Benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indazinyl, isoindolyl, indazolyl, isoindazolyl , Purinyl, Quinolinyl, Isoquinolinyl, Diazonaphthyl, Naphthyridinyl, Quinoxolinyl, Pteridinyl, Carbazolyl, Carboline, Phenanthridinyl, Phenanthrolinyl, Acridine Group, phenazine group, isothiazolyl, benzothiazolyl, benzothienyl, oxtriazolyl, cinnolinyl, quinazolinyl, phenylthio, indolizinyl, phenanthryl, Isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6,7-tetrahydrobenzo[b]thienyl, naphthopyridyl, [1,2,4]triazolo[4 ,3-b]pyridazine, [1,2,4]triazolo[4,3-a]pyrazine, [1,2,4]triazolo[4,3-c]pyrimidine, [1, 2,4]Triazolo[4,3-a]pyridine, imidazo[1,2-a]pyridine, imidazo[1,2-b]pyridazine, imidazo[1,2-a]pyrazine Wait.
在本申请中,术语“杂芳基烷基”是指被上文所定义的杂芳基所取代的上文所定义的烷基。In this application, the term "heteroarylalkyl" refers to the above-defined alkyl group substituted by the above-defined heteroaryl group.
在本申请中,“任选地”或“任选地”表示随后描述的事件或状况可能发生也可能不发生,且该描述同时包括该事件或状况发生和不发生的情况。例如,“任选地被取代的芳 基”表示芳基被取代或未被取代,且该描述同时包括被取代的芳基与未被取代的芳基。本发明权利要求书和说明书部分所述的“任选地”的取代基选自烷基、烯基、炔基、卤素、卤代烷基、卤代烯基、卤代炔基、氰基、硝基、任选取代的芳基、任选取代的杂芳基、任选取代的环烃基、任选取代的杂环烃基。In this application, "optionally" or "optionally" means that the event or condition described later may or may not occur, and the description includes both occurrence and non-occurrence of the event or condition. For example, "optionally substituted aryl group" means that the aryl group is substituted or unsubstituted, and the description includes both substituted aryl groups and unsubstituted aryl groups. The "optionally" substituents described in the claims and specification of the present invention are selected from alkyl, alkenyl, alkynyl, halogen, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, cyano, nitro , Optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocyclic alkyl.
“SHP2”指“SrcHomolgy-2磷酸酶”,还叫SH-PTP2、SH-PT3、Syp、PTP1D、PTP2C、SAP-2或PTPN11。"SHP2" refers to "SrcHomolgy-2 phosphatase", also known as SH-PTP2, SH-PT3, Syp, PTP1D, PTP2C, SAP-2 or PTPN11.
本文所用术语“部分”、“结构部分”、“化学部分”、“基团”、“化学基团”是指分子中的特定片段或官能团。化学部分通常被认为是嵌入或附加到分子上的化学实体。The terms "part", "structural part", "chemical part", "group", and "chemical group" as used herein refer to specific fragments or functional groups in a molecule. The chemical moiety is generally considered to be a chemical entity embedded or attached to a molecule.
制备/分离个别异构体的常规技术包括由合适的光学纯前体的手性合成,或者使用例如手性高效液相色谱法拆分外消旋体(或盐或衍生物的外消旋体),例如可参见Gerald Gübitz and Martin G.Schmid(Eds.),Chiral Separations,Methods and Protocols,Methods in Molecular Biology,Vol.243,2004;A.M.Stalcup,Chiral Separations,Annu.Rev.Anal.Chem.3:341-63,2010;Fumiss et al.(eds.),VOGEL’S ENCYCLOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5.sup.TH ED.,Longman Scientific and Technical Ltd.,Essex,1991,809-816;Heller,Acc.Chem.Res.1990,23,128。Conventional techniques for preparing/separating individual isomers include chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives using, for example, chiral high performance liquid chromatography) ), for example, see Gerald Gübitz and Martin G. Schmid (Eds.), Chiral Separations, Methods and Protocols, Methods in Molecular Biology, Vol. 243, 2004; AMStalcup, Chiral Separations, Annu. Rev. Anal. Chem. 3 :341-63,2010; Fumis et al.(eds.),VOGEL'S ENCYCLOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5.sup.TH ED.,Longman Scientific and Technical Ltd.,Essex,1991,809-816;Heller,Acc.Chem .Res.1990,23,128.
本发明还包括本发明所述的化合物或其药学上可接受的盐的所有适宜的同位素变体。本发明的化合物或其药学上可接受的盐的同位素变体被定义为其中至少一个原子被具有相同原子数、但原子质量与自然界经常发现的原子质量不同的原子所替换的那些。可以掺入到本发明的化合物及其药学上可接受的盐中的同位素包括但不限于H、C、N和O的同位素,例如 2H、 3H、 11C、 13C、 14C、 15N、 17O、 18O、 35S、 18F、 36Cl和 125I。本发明所述化合物或其药学上可接受的盐的同位素变体可以通过常规技术、采用适宜试剂的适当同位素变体来制备。 The present invention also includes all suitable isotopic variants of the compounds of the present invention or pharmaceutically acceptable salts thereof. Isotopic variants of the compound of the present invention or a pharmaceutically acceptable salt thereof are defined as those in which at least one atom is replaced by an atom having the same atomic number but having an atomic mass different from the atomic mass often found in nature. The isotopes that can be incorporated into the compounds of the present invention and their pharmaceutically acceptable salts include, but are not limited to, isotopes of H, C, N, and O, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 35 S, 18 F, 36 Cl and 125 I. Isotopic variants of the compounds of the present invention or pharmaceutically acceptable salts thereof can be prepared by conventional techniques using appropriate isotopic variants of appropriate reagents.
在本申请中,术语“药学上可接受的盐”包括药学上可接受的酸加成盐和药学上可接受的碱加成盐。In this application, the term "pharmaceutically acceptable salt" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
“药学上可接受的酸加成盐”是指能够保留游离碱的生物有效性而无其它副作用的,与无机酸或有机酸所形成的盐。无机酸盐包括但不限于盐酸盐、氢溴酸盐、硫酸盐、硝酸盐、磷酸盐等;有机酸盐包括但不限于甲酸盐、乙酸盐、2,2-二氯乙酸盐、三氟乙酸盐、丙酸盐、己酸盐、辛酸盐、癸酸盐、十一碳烯酸盐、乙醇酸盐、葡糖酸盐、乳酸盐、癸二酸盐、己二酸盐、戊二酸盐、丙二酸盐、草酸盐、马来酸盐、琥珀酸盐、富马酸盐、酒石酸盐、柠檬酸盐、棕榈酸盐、硬脂酸盐、油酸盐、肉桂酸盐、月桂酸盐、苹果酸盐、谷氨酸盐、焦谷氨酸盐、天冬氨酸盐、苯甲酸盐、甲磺酸盐、苯磺酸盐、对甲苯磺酸盐、海藻酸盐、抗坏血酸盐、水杨酸盐、4-氨基水杨酸盐、萘二磺酸盐等。这些盐可通过本专业已 知的方法制备。"Pharmaceutically acceptable acid addition salt" refers to a salt formed with an inorganic acid or an organic acid that can retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, etc.; organic acid salts include, but are not limited to, formate, acetate, and 2,2-dichloroacetate , Trifluoroacetate, propionate, caproate, caprylate, caprate, undecylenate, glycolate, gluconate, lactate, sebacate, hexanoate Acid salt, glutarate, malonate, oxalate, maleate, succinate, fumarate, tartrate, citrate, palmitate, stearate, oleate , Cinnamate, laurate, malate, glutamate, pyroglutamate, aspartate, benzoate, methanesulfonate, benzenesulfonate, p-toluenesulfonate , Alginate, ascorbate, salicylate, 4-aminosalicylate, naphthalene disulfonate, etc. These salts can be prepared by methods known in the art.
“药学上可接受的碱加成盐”是指能够保持游离酸的生物有效性而无其它副作用的、与无机碱或有机碱所形成的盐。衍生自无机碱的盐包括但不限于钠盐、钾盐、锂盐、铵盐、钙盐、镁盐、铁盐、锌盐、铜盐、锰盐、铝盐等。优选的无机盐为铵盐、钠盐、钾盐、钙盐及镁盐。衍生自有机碱的盐包括但不限于以下的盐:伯胺类、仲胺类及叔胺类,被取代的胺类,包括天然的被取代胺类、环状胺类及碱性离子交换树脂,例如氨、异丙胺、三甲胺、二乙胺、三乙胺、三丙胺、乙醇胺、二乙醇胺、三乙醇胺、二甲基乙醇胺、2-二甲氨基乙醇、2-二乙氨基乙醇、二环己胺、赖氨酸、精氨酸、组氨酸、咖啡因、普鲁卡因、胆碱、甜菜碱、乙二胺、葡萄糖胺、甲基葡萄糖胺、可可碱、嘌呤、哌嗪、哌啶、N-乙基哌啶、聚胺树脂等。优选的有机碱包括异丙胺、二乙胺、乙醇胺、三甲胺、二环己基胺、胆碱及咖啡因。这些盐可通过本专业已知的方法制备。"Pharmaceutically acceptable base addition salt" refers to a salt formed with an inorganic base or an organic base that can maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include but are not limited to the following salts: primary amines, secondary amines and tertiary amines, substituted amines, including natural substituted amines, cyclic amines and basic ion exchange resins , Such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, bicyclic Hexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperazine Pyridine, N-ethylpiperidine, polyamine resin, etc. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. These salts can be prepared by methods known in the art.
在本申请中,“药物组合物”是指本发明化合物与本领域通常接受的用于将生物活性化合物输送至哺乳动物(例如人)的介质的制剂。该介质包括药学上可接受的载体。药物组合物的目的是促进生物体的给药,利于活性成分的吸收进而发挥生物活性。In the present application, "pharmaceutical composition" refers to a preparation of a compound of the present invention and a medium generally accepted in the art for delivering a biologically active compound to a mammal (such as a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to promote the administration of the organism, which is conducive to the absorption of the active ingredient and thus the biological activity.
本文所用术语“药学上可接受的”是指不影响本发明化合物的生物活性或性质的物质(如载体或稀释剂),并且相对无毒,即该物质可施用于个体而不造成不良的生物反应或以不良方式与组合物中包含的任意组分相互作用。The term "pharmaceutically acceptable" as used herein refers to a substance (such as a carrier or diluent) that does not affect the biological activity or properties of the compound of the present invention, and is relatively non-toxic, that is, the substance can be administered to an individual without causing undesirable biological activity. Reacts or interacts in an undesirable manner with any components included in the composition.
在本申请中,“药学上可接受的赋形剂”包括但不限于任何被相关的政府管理部门许可为可接受供人类或家畜使用的佐剂、载体、赋形剂、助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味剂、表面活性剂、润湿剂、分散剂、助悬剂、稳定剂、等渗剂、溶剂或乳化剂。In this application, "pharmaceutically acceptable excipients" include, but are not limited to, any adjuvants, carriers, excipients, glidants, enhancers that are approved by the relevant government administration as acceptable for human or livestock use. Sweeteners, diluents, preservatives, dyes/colorants, flavors, surfactants, wetting agents, dispersants, suspending agents, stabilizers, isotonic agents, solvents or emulsifiers.
本发明所述“肿瘤”包括但不限于努南综合症、豹综合症、青少年髓单核细胞白血病、成神经细胞瘤、肉瘤、黑色素瘤、关节软骨瘤、胆管瘤、白血病、乳腺癌、胃肠间质瘤、组织细胞性淋巴瘤、非小细胞肺癌、小细胞肺癌、食道癌、胰腺癌、肺鳞癌、肺腺癌、乳腺癌、前列腺癌、肝癌、皮肤癌、上皮细胞癌、宫颈癌、卵巢癌、肠癌、鼻咽癌、脑癌、骨癌、肾癌、口腔癌/头癌、成神经细胞瘤、头颈的鳞状细胞癌、间变性大细胞淋巴瘤或成胶质细胞瘤等疾病。The "tumor" of the present invention includes but is not limited to Noonan syndrome, Leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, sarcoma, melanoma, articular chondroma, cholangiomas, leukemia, breast cancer, stomach Intestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, esophageal cancer, pancreatic cancer, lung squamous cell carcinoma, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell carcinoma, cervix Cancer, ovarian cancer, bowel cancer, nasopharyngeal cancer, brain cancer, bone cancer, kidney cancer, oral cancer/head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, anaplastic large cell lymphoma, or glioblast Tumors and other diseases.
本文所用术语“预防的”、“预防”和“防止”包括使病患减少疾病或病症的发生或恶化的可能性。The terms "preventive", "preventing" and "preventing" as used herein include reducing the likelihood of a disease or condition from occurring or worsening.
本文所用的术语“治疗”和其它类似的同义词包括以下含义:As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i)预防疾病或病症在哺乳动物中出现,特别是当这类哺乳动物易患有该疾病或病症,但尚未被诊断为已患有该疾病或病症时;(i) Preventing the occurrence of a disease or condition in a mammal, especially when such a mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii)抑制疾病或病症,即遏制其发展;(ii) Inhibiting the disease or disease, that is, curbing its development;
(iii)缓解疾病或病症,即,使该疾病或病症的状态消退;或者(iii) Alleviate the disease or condition, that is, make the state of the disease or condition subside; or
(iv)减轻该疾病或病症所造成的症状。(iv) Alleviate the symptoms caused by the disease or condition.
本文所使用术语“有效量”、“治疗有效量”或“药学有效量”是指服用后足以在某种程度上缓解所治疗的疾病或病症的一个或多个症状的至少一种药剂或化合物的量。其结果可以为迹象、症状或病因的消减和/或缓解,或生物系统的任何其它所需变化。例如,用于治疗的“有效量”是在临床上提供显著的病症缓解效果所需的包含本文公开化合物的组合物的量。可使用诸如剂量递增试验的技术测定适合于任意个体病例中的有效量。The term "effective amount", "therapeutically effective amount" or "pharmaceutical effective amount" as used herein refers to at least one agent or compound that is sufficient to relieve one or more symptoms of the disease or condition being treated after administration The amount. The result can be a reduction and/or alleviation of signs, symptoms or causes, or any other desired changes in the biological system. For example, the "effective amount" for treatment is the amount of the composition containing the compound disclosed herein that is required to provide significant disease relief clinically. Techniques such as dose escalation tests can be used to determine the effective amount suitable for any individual case.
本文所用术语“服用”、“施用”、“给药”等是指能够将化合物或组合物递送到进行生物作用的所需位点的方法。这些方法包括但不限于口服途径、经十二指肠途径、胃肠外注射(包括静脉内、皮下、腹膜内、肌内、动脉内注射或输注)、局部给药和经直肠给药。本领域技术人员熟知可用于本文所述化合物和方法的施用技术,例如在Goodman and Gilman,The Pharmacological Basis of Therapeutics,current ed.;Pergamon;and Remington’s,Pharmaceutical Sciences(current edition),Mack Publishing Co.,Easton,Pa中讨论的那些。在优选的实施方案中,本文讨论的化合物和组合物通过口服施用。The terms "administration", "administration", "administration" and the like as used herein refer to methods capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, transduodenal routes, parenteral injections (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Those skilled in the art are familiar with the application techniques that can be used for the compounds and methods described herein, for example in Goodman and Gilman, The Pharmaceutical Basis of Therapeutics, current ed.; Pergamon; and Remington's, Pharmaceutical (current edition), Mack Publishing Co., Those discussed in Easton, Pa. In a preferred embodiment, the compounds and compositions discussed herein are administered orally.
本文所使用术语“药物组合”、“药物联用”、“联合用药”、“施用其它治疗”、“施用其它治疗剂”等是指通过混合或组合不止一种活性成分而获得的药物治疗,其包括活性成分的固定和不固定组合。术语“固定组合”是指以单个实体或单个剂型的形式向患者同时施用至少一种本文所述的化合物和至少一种协同药剂。术语“不固定组合”是指以单独实体的形式向患者同时施用、合用或以可变的间隔时间顺次施用至少一种本文所述的化合物和至少一种协同制剂。这些也应用到鸡尾酒疗法中,例如施用三种或更多种活性成分。The terms "drug combination", "drug combination", "combination", "administration of other treatments", "administration of other therapeutic agents" and the like as used herein refer to drug treatments obtained by mixing or combining more than one active ingredient. It includes fixed and non-fixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one synergistic agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, combination or sequential administration of at least one compound described herein and at least one synergistic agent to a patient in the form of separate entities. These also apply to cocktail therapy, such as the administration of three or more active ingredients.
本领域技术人员还应当理解,在下文所述的方法中,中间体化合物官能团可能需要由适当的保护基保护。这样的官能团包括羟基、氨基、巯基及羧酸。合适的羟基保护基包括三烷基甲硅烷基或二芳基烷基甲硅烷基(例如叔丁基二甲基甲硅烷基、叔丁基二苯基甲硅烷基或三甲基甲硅烷基)、四氢吡喃基、苄基等。合适的氨基、脒基及胍基的保护基包括叔丁氧羰基、苄氧羰基等。合适的巯基保护基包括-C(O)-R”(其中R”为烷基、芳基或芳烷基)、对甲氧基苄基、三苯甲基等。合适的羧基保护基包括烷基、芳基或芳烷基酯类。Those skilled in the art should also understand that in the methods described below, the functional group of the intermediate compound may need to be protected by an appropriate protecting group. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl (e.g. tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl) , Tetrahydropyranyl, benzyl, etc. Suitable protecting groups for amino, amidino and guanidino include tert-butoxycarbonyl, benzyloxycarbonyl and the like. Suitable protecting groups for mercapto include -C(O)-R" (wherein R" is an alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
保护基可根据本领域技术人员已知的和如本文所述的标准技术来引入和除去。保护基的使用详述于Greene,T.W.与P.G.M.Wuts,Protective Groups in Organi Synthesis,(1999),4th Ed.,Wiley中。保护基还可为聚合物树脂。Protecting groups can be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protective groups is detailed in Greene, T.W. and P.G.M. Wuts, Protective Groups in Organi Synthesis, (1999), 4th Ed., Wiley. The protecting group can also be a polymer resin.
式I化合物Formula I compound
本发明提供了一种式I所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物:The present invention provides a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
Figure PCTCN2020106214-appb-000014
Figure PCTCN2020106214-appb-000014
其中,among them,
R 1选自H、-卤素、-CN、-OH、-NO 2、HSO 3-、未取代或卤代的C1-C6烷基磺酰基、未取代或卤代的C1-C6烷基羧基、未取代或卤代的C1-C6烷基氨基、未取代或卤代的C1-C6烷基、未取代或卤代的C1-C6烷氧基、未取代或卤代的C1-6烷氧基羰基、未取代或卤代的C1-6烷基羰基、未取代或卤代的C2-C6烯烃基羰基、未取代或卤代的C1-C6烷氧基-O-C1-C6烷基、未取代或取代的3-8元环烷基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;所述的杂环基或杂芳基包含1-4个选自下组的杂原子:N、O或S; R 1 is selected from H, -halogen, -CN, -OH, -NO 2 , HSO 3 -, unsubstituted or halogenated C1-C6 alkylsulfonyl, unsubstituted or halogenated C1-C6 alkylcarboxy, Unsubstituted or halogenated C1-C6 alkylamino, unsubstituted or halogenated C1-C6 alkyl, unsubstituted or halogenated C1-C6 alkoxy, unsubstituted or halogenated C1-6 alkoxy Carbonyl, unsubstituted or halogenated C1-6 alkylcarbonyl, unsubstituted or halogenated C2-C6 alkenylcarbonyl, unsubstituted or halogenated C1-C6 alkoxy-O-C1-C6 alkyl, unsubstituted Substituted or substituted 3-8 membered cycloalkyl, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl, unsubstituted or substituted 5-10 membered heteroaryl; The heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
R 2选自H、-卤素、氨基、未取代或取代的C1-C6烷基氨基、未取代或取代的C1-C6烷基; R 2 is selected from H, -halogen, amino, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl;
R 3选自H、取代或未取代的3-8元环烷基、取代或未取代的3-8元杂环基、取代或未取代的5-10元芳基、取代或未取代的5-10元杂芳基;所述的杂环基或杂芳基包含1-4个选自下组的杂原子:N、O或S; R 3 is selected from H, substituted or unsubstituted 3-8 membered cycloalkyl, substituted or unsubstituted 3-8 membered heterocyclic group, substituted or unsubstituted 5-10 membered aryl, substituted or unsubstituted 5 -10 membered heteroaryl; said heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
X和Y分别独立地选自N或CR 4X and Y are each independently selected from N or CR 4 ;
R 4选自H、卤素、-CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的氨基、(取代或未取代的C1-C6烷基)SO-、(取代或未取代的C1-C6烷基)SO 2-; R 4 is selected from H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, (substituted or unsubstituted C1-C6 Alkyl)SO-, (substituted or unsubstituted C1-C6 alkyl)SO 2 -;
或者R 4与邻近的
Figure PCTCN2020106214-appb-000015
共同形成一个取代或未取代的5-10元芳环,取代或未取代的5-10元杂芳环,取代或未取代的5-10元杂环,或取代或未取代的5-10元碳环;所述环可以包含0-4个选自下组的杂原子:N、O或S;条件是,此时X和Y不能同时为N;
Or R 4 and adjacent
Figure PCTCN2020106214-appb-000015
Together to form a substituted or unsubstituted 5-10 membered aromatic ring, substituted or unsubstituted 5-10 membered heteroaromatic ring, substituted or unsubstituted 5-10 membered heterocyclic ring, or substituted or unsubstituted 5-10 membered ring Carbocyclic ring; the ring may contain 0-4 heteroatoms selected from the group consisting of N, O or S; the condition is that X and Y cannot be N at the same time;
Figure PCTCN2020106214-appb-000016
表示单键或双键;
Figure PCTCN2020106214-appb-000016
Represents a single bond or a double bond;
上述的任一“取代”指基团上的一个或多个氢原子被选自下组的取代基取代:-D、卤素、-OH、-NO 2、-NH 2、-N(未取代或卤代的C1-C6烷基) 2、-CN、未取代或卤代的C1-C8烷基、未取代或卤代的C1-C8烷氧基、未取代或卤代的C1-C8烷氧基-C1-C8烷基、未取代或卤代的C3-C8环烷基、未取代或卤代的C3-C8环烷基-C1-C8烷基、未取代或卤代的C1-C6烷基羰基、未取代或卤代的C1-C6烷氧基羰基、异羟肟酸基、未取代或卤代的C1-C6烷基巯基、-S(O) 2N(未取代或卤代的C1-C6烷基) 2、-S(O) 2未取代或卤代的C1-C6烷基、 -N(未取代或卤代的C1-C6烷基)S(O) 2N(未取代或卤代的C1-C6烷基) 2、-S(O)N(未取代或卤代的C1-C6烷基) 2、-S(O)(未取代或卤代的C1-C6烷基)、-N(未取代或卤代的C1-C6烷基)S(O)N(未取代或卤代的C1-C6烷基) 2、-N(未取代或卤代的C1-C6烷基)S(O)(未取代或卤代的C1-C6烷基)、未取代或卤代的5-8元芳基、未取代或卤代的5-8元杂芳基、未取代或卤代的4-8元饱和杂环或碳环;其中,所述的杂芳基包含1-4个选自下组的杂原子:N、O或S,所述的杂环包含1-4个选自下组的杂原子:N、O或S。 Any of the above "substituted" means that one or more hydrogen atoms on the group are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -N (unsubstituted or (Halogenated C1-C6 alkyl) 2 , -CN, unsubstituted or halogenated C1-C8 alkyl, unsubstituted or halogenated C1-C8 alkoxy, unsubstituted or halogenated C1-C8 alkoxy Group-C1-C8 alkyl, unsubstituted or halogenated C3-C8 cycloalkyl, unsubstituted or halogenated C3-C8 cycloalkyl-C1-C8 alkyl, unsubstituted or halogenated C1-C6 alkane Carbonyl group, unsubstituted or halogenated C1-C6 alkoxycarbonyl, hydroxamic acid group, unsubstituted or halogenated C1-C6 alkyl mercapto group, -S(O) 2 N (unsubstituted or halogenated C1-C6 alkyl) 2 , -S(O) 2 unsubstituted or halogenated C1-C6 alkyl, -N (unsubstituted or halogenated C1-C6 alkyl) S(O) 2 N (unsubstituted Or halogenated C1-C6 alkyl) 2 , -S(O)N (unsubstituted or halogenated C1-C6 alkyl) 2 , -S(O) (unsubstituted or halogenated C1-C6 alkyl) ), -N (unsubstituted or halogenated C1-C6 alkyl) S(O)N (unsubstituted or halogenated C1-C6 alkyl) 2 , -N (unsubstituted or halogenated C1-C6 alkane Group) S(O) (unsubstituted or halogenated C1-C6 alkyl), unsubstituted or halogenated 5-8 membered aryl, unsubstituted or halogenated 5-8 membered heteroaryl, unsubstituted or A halogenated 4-8 membered saturated heterocyclic ring or carbocyclic ring; wherein the heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S, and the heterocyclic ring contains 1-4 One heteroatom selected from the group: N, O or S.
优选地,式I中,当R 4与邻近的
Figure PCTCN2020106214-appb-000017
共同形成一个取代或未取代的5-10元芳环、取代或未取代的5-10元杂芳环、取代或未取代的5-10元杂环时,所形成的环为本文任一实施方案所述的环A。
Preferably, in formula I, when R 4 is adjacent to
Figure PCTCN2020106214-appb-000017
When jointly forming a substituted or unsubstituted 5-10 membered aromatic ring, substituted or unsubstituted 5-10 membered heteroaromatic ring, substituted or unsubstituted 5-10 membered heterocyclic ring, the ring formed is any one of the implementations herein The ring A described in the scheme.
本发明提供了一种如式II所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物:The present invention provides a compound of Formula II, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
Figure PCTCN2020106214-appb-000018
Figure PCTCN2020106214-appb-000018
其中,among them,
X和Y分别独立地选自N或C;X and Y are each independently selected from N or C;
R 4a和R 4b分别独立地选自无、H、卤素、-CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的氨基、(取代或未取代的C1-C6烷基)SO-、(取代或未取代的C1-C6烷基)SO 2-; R 4a and R 4b are each independently selected from none, H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, ( Substituted or unsubstituted C1-C6 alkyl) SO-, (substituted or unsubstituted C1-C6 alkyl) SO 2 -;
其他取代基的定义如上所述。The definition of other substituents is as described above.
本发明提供了一种如式III所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物:The present invention provides a compound of Formula III, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
Figure PCTCN2020106214-appb-000019
Figure PCTCN2020106214-appb-000019
其中,among them,
X和Y分别独立地选自N或C;X和Y不能同时为N;X and Y are independently selected from N or C; X and Y cannot be N at the same time;
环A为取代或未取代的5-10元芳环,取代或未取代的5-10元杂芳环,取代或未取代的5-10元杂环,或取代或未取代的5-10元碳环;所述的环可以包含0-4个选自下组的杂原子:N、O或S;Ring A is a substituted or unsubstituted 5-10 membered aromatic ring, a substituted or unsubstituted 5-10 membered heteroaromatic ring, a substituted or unsubstituted 5-10 membered heterocyclic ring, or a substituted or unsubstituted 5-10 membered ring Carbocyclic ring; said ring may contain 0-4 heteroatoms selected from the group consisting of N, O or S;
上述的“取代”指环A上的一个或多个氢原子被选自下组的取代基取代:-D、卤素、-OH、-NO 2、-NH 2、-NH(未取代或卤代的C1-C6烷基)、-N(未取代或卤代的C1-C6烷基) 2、-CN、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-; The above-mentioned "substituted" means that one or more hydrogen atoms on ring A are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -NH (unsubstituted or halogenated C1-C6 alkyl), -N (unsubstituted or halogenated C1-C6 alkyl) 2 , -CN, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy Group, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -;
其他取代基的定义如上所述。The definition of other substituents is as described above.
本发明提供的式III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,优选地,所述的环A选自取代或未取代的三元(杂)环、取代或未取代的四元(杂)环、取代或未取代的五元(杂)环、取代或未取代的六元(杂)环、取代或未取代的五元芳(杂)环、取代或未取代的六元芳(杂)环、取代或未取代的七元芳(杂)环、取代或未取代的五元并六元(杂)环、取代或未取代的六元并六元(杂)环;所述的杂环可以包含1-4个选自下组的杂原子:N、O或S;所述的环A可被任意取代,所述取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 In the compound represented by formula III provided by the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, preferably, the ring A is selected from Self-substituted or unsubstituted three-membered (hetero) ring, substituted or unsubstituted four-membered (hetero) ring, substituted or unsubstituted five-membered (hetero) ring, substituted or unsubstituted six-membered (hetero) ring, substituted Or unsubstituted five-membered aromatic (hetero) ring, substituted or unsubstituted six-membered aromatic (hetero) ring, substituted or unsubstituted seven-membered aromatic (hetero) ring, substituted or unsubstituted five-membered and six-membered (hetero) ) Ring, substituted or unsubstituted six-membered and six-membered (hetero) ring; the heterocyclic ring may contain 1-4 heteroatoms selected from the group consisting of N, O or S; the ring A may be Optional substitution, the substituent is selected from: halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl , Halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -.
本发明提供的式III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,优选地,所述的环A为选自下组任意一个:In the compound represented by formula III provided by the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, preferably, the ring A is Choose from any one of the following groups:
Figure PCTCN2020106214-appb-000020
Figure PCTCN2020106214-appb-000020
Figure PCTCN2020106214-appb-000021
Figure PCTCN2020106214-appb-000021
上述环A可被任意取代,所述取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 The above-mentioned ring A can be optionally substituted, and the substituents are selected from halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1 -C6 alkyl)SO 2 -.
本发明提供的式III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,更优选地,所述的环A为选自下组任意一个:The compound represented by formula III provided by the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, more preferably, the ring A Any one selected from the following group:
Figure PCTCN2020106214-appb-000022
Figure PCTCN2020106214-appb-000022
上述环A可被任意取代,所述取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 The above-mentioned ring A can be optionally substituted, and the substituents are selected from halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1 -C6 alkyl)SO 2 -.
更优选地,本发明式III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,环A为任选取代的:More preferably, in the compound represented by formula III of the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, ring A is optionally substituted :
Figure PCTCN2020106214-appb-000023
更优选为
Figure PCTCN2020106214-appb-000024
Figure PCTCN2020106214-appb-000023
More preferably
Figure PCTCN2020106214-appb-000024
其中,当被取代时,取代基可以为1-2个选自卤素、C1-C6烷基和卤代C1-C6烷基的取代基。Wherein, when substituted, the substituent may be 1-2 substituents selected from halogen, C1-C6 alkyl and halogenated C1-C6 alkyl.
本发明提供的式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,优选地,所述的R 1选自H、卤代或未取代的C1-C6烷基羰基、卤代或未取代的C2-C6烯烃基羰基、(卤代或未取代的C1-C6烷基)SO 2-、取代或未取代的苯磺酰基、未取代或取代的3-8元杂环基、未取代或取代的3-8元碳环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;其中所述的苯磺酰基、碳环基、杂环基、芳基和杂芳基被取代时,取代基的个数为1-4个,取代基选自H、NH 2、卤素、卤代或未取代的C1-C8环烷基、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-。 The compounds represented by formulas I, II and III provided by the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, preferably, the R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, Substituted or unsubstituted benzenesulfonyl group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or Substituted 5-10 membered heteroaryl group; wherein the benzenesulfonyl group, carbocyclic group, heterocyclic group, aryl group and heteroaryl group are substituted, the number of substituents is 1-4, and the substituents are selected From H, NH 2 , halogen, halogenated or unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
本发明提供的式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,优选地,所述的R 1选自H、卤代或未取代的C1-C6烷基羰基、卤代或未取代的C2-C6烯烃基羰基、(卤代或未取代的C1-C6烷基)SO 2-、取代或未取代的苯磺酰基,以及可被任选取代的以下任一基团: The compounds represented by formulas I, II and III provided by the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, preferably, the R 1 is selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, A substituted or unsubstituted benzenesulfonyl group, and any of the following groups which may be optionally substituted:
Figure PCTCN2020106214-appb-000025
Figure PCTCN2020106214-appb-000025
其中W选自C、O、S或N;其中,所述苯磺酰基以及可被任选取代的环基被取代时,取代基的个数为1-4个,选自H、NH 2、卤素、卤代或未取代的C1-C8环烷基、未取代或卤代的C1-C6烷基和(卤代或未取代的C1-C6烷基) 2N-。 Wherein W is selected from C, O, S or N; wherein, when the benzenesulfonyl group and the optionally substituted ring group are substituted, the number of substituents is 1-4, selected from H, NH 2 , Halogen, halogenated or unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, and (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
优选地,式I、II和III中的R 1选自:未取代的C1-C6烷基-SO 2-,未取代的C2-C6烯烃基羰基,未取代或取代的3-8元杂环基,未取代或取代的3-8元环烷基,未取代或取代的5-10元芳基,未取代或取代的5-10元杂芳基;其中,所述杂芳基、环烷基、芳基和杂芳基被取代时,取代基为1-3个选自卤素、未取代的C1-C6烷基和卤代C1-C6烷基的取代基。更优选地,R 1中的杂环基、环烷基、芳基和杂芳基可选自吡嗪基、苯基、氮杂环丁烷基、环丁烷基、氧杂环丁烷基和吡啶基,这些基团可任选地被1或2个C1-C6烷基取代。 Preferably, R 1 in formula I, II and III is selected from: unsubstituted C1-C6 alkyl-SO 2 -, unsubstituted C2-C6 alkenyl carbonyl, unsubstituted or substituted 3-8 membered heterocycle Group, unsubstituted or substituted 3-8 membered cycloalkyl, unsubstituted or substituted 5-10 membered aryl, unsubstituted or substituted 5-10 membered heteroaryl; wherein, the heteroaryl, cycloalkane When the group, aryl group and heteroaryl group are substituted, the substituents are 1-3 substituents selected from halogen, unsubstituted C1-C6 alkyl and halogenated C1-C6 alkyl. More preferably, the heterocyclic group, cycloalkyl group, aryl group and heteroaryl group in R 1 may be selected from pyrazinyl, phenyl, azetidinyl, cyclobutanyl, oxetanyl And pyridyl, these groups can be optionally substituted with 1 or 2 C1-C6 alkyl groups.
本发明提供的式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,优选地,R 2选自H、卤素、氨基和未取代的C1-C6烷基;更优选地,R 2选自H和氨基。 Among the compounds represented by formulas I, II and III provided by the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, preferably, R 2 It is selected from H, halogen, amino and unsubstituted C1-C6 alkyl; more preferably, R 2 is selected from H and amino.
本发明提供的式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,优选地,所述的R 3选自H、未取代或取代的3-8元杂环基、未取代或取代的3-8元碳环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;其中所述的环基可被任意取代,所述的取代基的个数为1-4个;所述的取代基选自H、NH 2、卤素、卤代或未取代的C1-C8环烷基、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-和卤代或未取代的C1-C6烷基-NH-。 The compounds represented by formulas I, II and III provided by the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, preferably, the R 3 is selected from H, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl; wherein the ring group can be optionally substituted, the number of the substituent is 1-4; the substituent is selected from H, NH 2 , halogen, halogenated or Unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkyl -NH-.
优选地,本发明提供的式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,所述的R 3选自H、 Preferably, the compounds represented by formulas I, II and III provided by the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, R 3 is selected from H,
Figure PCTCN2020106214-appb-000026
Figure PCTCN2020106214-appb-000026
其中W选自O、S或N,且所述的环基可被任意取代,取代基的个数为1-4个,取代基选自H、NH 2、卤素、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-和卤代或未取代的C1-C6烷基-NH-。 Wherein W is selected from O, S or N, and the cyclic group can be optionally substituted, the number of substituents is 1-4, and the substituents are selected from H, NH 2 , halogen, unsubstituted or halogenated C1 -C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkyl -NH-.
更优选地,本发明的式I、II和III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,所述R 3选自任选被氨基和卤素取代的5-10元杂芳基,优选为5-10元含氮杂芳基,更优选为任选被氨基和卤素取代的吡啶基,更优选为任选被氨基和卤素取代的: More preferably, the compounds represented by formulas I, II and III of the present invention, or pharmaceutically acceptable salts thereof, or solvates, isotopic substitutions, polymorphs, prodrugs or metabolites thereof, R 3 is selected from 5-10 membered heteroaryl groups optionally substituted by amino and halogen, preferably 5-10 membered nitrogen-containing heteroaryl groups, more preferably pyridyl optionally substituted by amino and halogen, more preferably any Select substituted by amino and halogen:
Figure PCTCN2020106214-appb-000027
Figure PCTCN2020106214-appb-000027
在特别优选的实施方案中,本发明式III所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 1选自未取代的C1-C6烷基-SO 2-,未取代的C2-C6烯烃基羰基,未取代或取代的3-8元杂环基,未取代或取代的3-8元环烷基,未取代或取代的5-10元芳基,未取代或取代的5-10元杂芳基;其中,所述杂芳基、环烷基、芳基和杂芳基被取代时,取代基为1-3个选自卤素、未取代的C1-C6烷基和卤代C1-C6烷基的取代基;更优选地,R 1中的杂环基、环烷基、芳基和杂芳基可选自吡嗪基、苯基、氮杂环丁烷基、环丁烷基、氧杂环丁烷基和吡啶基,这些基团可任选地被1或2个选自卤素和C1-C6烷基的取代基取代;更优选地,R 1选自任选被1或2个选自卤素和C1-C6烷基的取代基取代的: In a particularly preferred embodiment, the compound represented by formula III of the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, R 1 is selected From unsubstituted C1-C6 alkyl-SO 2 -, unsubstituted C2-C6 alkenyl carbonyl, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered cycloalkyl, Unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl group; wherein, when the heteroaryl group, cycloalkyl group, aryl group and heteroaryl group are substituted, the substituent is 1-3 substituents selected from halogen, unsubstituted C1-C6 alkyl and halogenated C1-C6 alkyl; more preferably, the heterocyclic group, cycloalkyl group, aryl group and heteroaryl group in R 1 Can be selected from pyrazinyl, phenyl, azetidinyl, cyclobutanyl, oxetanyl and pyridinyl, and these groups can be optionally substituted by 1 or 2 selected from halogen and C1- Substituents of C6 alkyl; more preferably, R 1 is selected from optionally substituted with 1 or 2 substituents selected from halogen and C1-C6 alkyl:
Figure PCTCN2020106214-appb-000028
Figure PCTCN2020106214-appb-000028
R 2选自H、卤素、氨基和未取代的C1-C6烷基,更优选选自H和氨基;R 3选自:任选被氨基和卤素取代的5-10元杂芳基,优选为5-10元含氮杂芳基,更优选为任选被1或2个选自氨基和卤素的取代基取代的吡啶基,更优选为被2个选自氨基和卤素的取代基取代的以下基团: R 2 is selected from H, halogen, amino and unsubstituted C1-C6 alkyl, more preferably selected from H and amino; R 3 is selected from: 5-10 membered heteroaryl optionally substituted by amino and halogen, preferably A 5- to 10-membered nitrogen-containing heteroaryl group is more preferably a pyridyl group optionally substituted with 1 or 2 substituents selected from amino and halogen, and more preferably the following substituted with 2 substituents selected from amino and halogen Group:
Figure PCTCN2020106214-appb-000029
Figure PCTCN2020106214-appb-000029
环A为任选取代的:Ring A is optionally substituted:
Figure PCTCN2020106214-appb-000030
Figure PCTCN2020106214-appb-000030
其中,当被取代时,取代基可以为1-2个选自卤素、C1-C6烷基和卤代C1-C6烷基的取代基。Wherein, when substituted, the substituent may be 1-2 substituents selected from halogen, C1-C6 alkyl and halogenated C1-C6 alkyl.
在特别优选的实施方案中,本发明式II所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物中,R 1选自未取代的C1-C6烷基-SO 2-,未取代的C2-C6烯烃基羰基,未取代或取代的3-8元杂环基,未取代或取代的3-8元环烷基,未取代或取代的5-10元芳基,未取代或取代的5-10元杂芳基;其中,所述杂芳基、环烷基、芳基和杂芳基被取代时,取代基为1-3个选自卤素、未取代的C1-C6烷基和卤代C1-C6烷基的取代基;更优选地,R 1中的杂环基、环烷基、芳基和杂芳基可选自吡嗪基、苯基、氮杂环丁烷基、环丁烷基、氧杂环丁烷基和吡啶基,这些基团可任选地被1或2个C1-C6烷基取代;R 2选自H、卤素、氨基和未取代的C1-C6烷基;R 3选自:任选被氨基和卤素取代的5-10元杂芳基,优选为5-10元含氮杂芳基,更优选为任选被氨基和卤素取代的吡啶基;X为CH;Y为N。 In a particularly preferred embodiment, the compound represented by formula II of the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, R 1 is selected From unsubstituted C1-C6 alkyl-SO 2 -, unsubstituted C2-C6 alkenyl carbonyl, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 3-8 membered cycloalkyl, Unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl group; wherein, when the heteroaryl group, cycloalkyl group, aryl group and heteroaryl group are substituted, the substituent is 1-3 substituents selected from halogen, unsubstituted C1-C6 alkyl and halogenated C1-C6 alkyl; more preferably, the heterocyclic group, cycloalkyl group, aryl group and heteroaryl group in R 1 Can be selected from pyrazinyl, phenyl, azetidinyl, cyclobutanyl, oxetanyl and pyridinyl, these groups can be optionally substituted with 1 or 2 C1-C6 alkyl groups ; R 2 is selected from H, halogen, amino and unsubstituted C1-C6 alkyl; R 3 is selected from: 5-10 membered heteroaryl optionally substituted by amino and halogen, preferably 5-10 membered nitrogen-containing hetero Aryl is more preferably pyridyl optionally substituted with amino and halogen; X is CH; Y is N.
本文中,基团上的波浪线表示该基团与式I、II和III化合物余下部分连接的位置。Here, the wavy line on the group indicates the position where the group is attached to the rest of the compounds of formula I, II and III.
应理解,本发明中,W、X和Y的选择应满足键价理论,当未满足键价理论,则以氢原子补足。例如,视不同的环,W可以是-CH-、-CH 2-或-NH-,X或Y可以为CH。 It should be understood that in the present invention, the selection of W, X and Y should satisfy the bond valence theory, and when the bond valence theory is not satisfied, a hydrogen atom is used to make up. For example, depending on the ring, W can be -CH-, -CH 2 -or -NH-, and X or Y can be CH.
本发明提供的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,选自以下结构:The compound provided by the present invention, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug, or metabolite thereof is selected from the following structures:
Figure PCTCN2020106214-appb-000031
Figure PCTCN2020106214-appb-000031
Figure PCTCN2020106214-appb-000032
Figure PCTCN2020106214-appb-000032
式I化合物的制备Preparation of compound of formula I
式I化合物可由如下路线制备得到:由中间体A出发经R 1X 1取代反应得到I-1,用酸脱除叔丁氧羰基和叔丁亚磺酰基得到中间体I-2,最后和中间体I-3进行取代反应得到式I化合物。 The compound of formula I can be prepared by the following route: starting from intermediate A and substituting R 1 X 1 to obtain I-1, removing tert-butoxycarbonyl and tert-butanesulfinyl with acid to obtain intermediate I-2, and finally and intermediate Form I-3 undergoes a substitution reaction to obtain a compound of formula I.
Figure PCTCN2020106214-appb-000033
Figure PCTCN2020106214-appb-000033
或者,式I化合物可由如下路线制备得到:由中间体A出发经三甲基硅三氟甲磺酸酯脱除叔丁氧羰基得到1-4,再和I-3取代反应得到I-5,用氢氧化钠水溶液脱除苄基氧羰 基得到中间体I-6,然后和中间体R 1X 1进行取代反应得到I-7,最后用酸脱除叔丁亚磺酰基得到式I化合物。 Alternatively, the compound of formula I can be prepared by the following route: starting from Intermediate A, the tert-butoxycarbonyl group is removed by trimethylsilyl trifluoromethanesulfonate to obtain 1-4, and then substituted with I-3 to obtain I-5. The benzyloxycarbonyl group is removed with sodium hydroxide aqueous solution to obtain intermediate I-6, and then it is substituted with intermediate R 1 X 1 to obtain I-7, and finally the tert-butylsulfinyl group is removed with acid to obtain the compound of formula I.
Figure PCTCN2020106214-appb-000034
Figure PCTCN2020106214-appb-000034
其中,所述的R 1、R 2、R 3、X、Y如上文所述;X 1和X 2为卤素。 Wherein, the R 1 , R 2 , R 3 , X and Y are as described above; X 1 and X 2 are halogen.
药理学和用途Pharmacology and uses
Src Hommolgy-2磷酸酶(SHP2)是PTPN11基因所编码的蛋白酪氨酸磷酸酶,其促进多种细胞功能,包括增殖、分化、细胞周期维持和迁移。SHP2牵涉在经由Ras-有丝分裂原-激活的蛋白激酶、JAK-STAT或磷酸肌醇3-激酶-AKT途径的信号传导中。SHP2介导受体酪氨酸激酶如ErbB1、ErbB2和c-Met的Erk1和Erk2MAP激酶的激活。Src Hommolgy-2 Phosphatase (SHP2) is a protein tyrosine phosphatase encoded by the PTPN11 gene, which promotes a variety of cell functions, including proliferation, differentiation, cell cycle maintenance and migration. SHP2 is involved in signaling via the Ras-mitogen-activated protein kinase, JAK-STAT, or phosphoinositide 3-kinase-AKT pathway. SHP2 mediates the activation of receptor tyrosine kinases such as ErbB1, ErbB2 and c-Met's Erk1 and Erk2MAP kinases.
SHP2具有两个N-末端Src homolgy2结构域(N-SH2和C-SH2)、催化结构域(PTP)和C-末端尾。所述两个SH2结构域控制SHP2的亚细胞定位和功能调控。该分子以无活性构象存在,经由牵涉来自N-SH2和PTP结构域的残基的结合网络来抑制其自身活性。响应于生长因子的刺激,SHP2经由其SH2结构域结合停靠蛋白上的特定酪氨酸-磷酸化位点如Gab1和Gab2。这引起了构象变化,导致SHP2激活。SHP2 has two N-terminal Src homolgy2 domains (N-SH2 and C-SH2), a catalytic domain (PTP) and a C-terminal tail. The two SH2 domains control the subcellular localization and functional regulation of SHP2. The molecule exists in an inactive conformation and inhibits its own activity via a binding network involving residues from the N-SH2 and PTP domains. In response to growth factor stimulation, SHP2 binds to specific tyrosine-phosphorylation sites on docking proteins such as Gab1 and Gab2 via its SH2 domain. This caused a conformational change, which resulted in SHP2 activation.
已经在多种人类疾病中识别到了PTPN11中的突变,所述疾病例如有努南综合征、豹斑综合征、幼年型粒-单核细胞白血病、成神经细胞瘤、黑素瘤、急性髓样白血病以及乳房、肺和结肠的癌症。SHP2是多种受体酪氨酸激酶、包括血小板衍生生长因子(PDGF-R)、成纤维细胞生长因子(FGF-R)和表皮生长因子(EGF-R)的受体的重要下游信号分子。SHP2还是激活促分裂原活化蛋白(MAP)激酶途径的重要下游信号分子,其可导致细胞转化(癌发展的必要条件)。SHP2的敲减显著抑制了具有SHP2突变或EML4/ALK易位的肺癌细胞系以及EGFR扩增乳癌和食管癌的细胞生长。SHP2还是胃癌、间变型大细胞淋巴癌和成胶质细胞瘤的癌基因的激活下游。Mutations in PTPN11 have been identified in a variety of human diseases, such as Noonan syndrome, Leopard spot syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myeloid Leukemia and cancer of the breast, lung, and colon. SHP2 is an important downstream signaling molecule for a variety of receptor tyrosine kinases, including platelet-derived growth factor (PDGF-R), fibroblast growth factor (FGF-R) and epidermal growth factor (EGF-R) receptors. SHP2 is also an important downstream signaling molecule that activates the mitogen-activated protein (MAP) kinase pathway, which can lead to cell transformation (a necessary condition for cancer development). Knockdown of SHP2 significantly inhibited the growth of lung cancer cell lines with SHP2 mutations or EML4/ALK translocation and EGFR-amplified breast cancer and esophageal cancer. SHP2 is also the downstream activation of oncogenes in gastric cancer, anaplastic large cell lymphoma and glioblastoma.
努南综合征(NS)和豹斑综合征(LS)-PTPNll突变引起LS(多发性色素斑样痣综合征,心电图传导异常,两眼距离过远,肺动脉瓣狭窄,异常生殖器,生长迟缓,感音神经性耳聋)和NS(包括心脏缺陷、颅面骨畸形和身材矮小的先天异常)。这两种障碍是 RAS/RAF/MEK/ERK有丝分裂原活化蛋白激酶途径(正常细胞生长和分化所需)的组分中的种系突变引起的常染色体显性综合征家族的一部分。该途径的异常调控具有深远的影响,特别是对心脏发育具有深远的影响,导致多种异常,包括瓣膜中隔缺陷(valvuloseptal defects)和/或肥厚型心肌病(HCM)。已经确定MAPK信号传导途径的的扰动对于这些障碍而言是重要的,已经在人中识别出沿循该途径的一些候选基因,包括KRAS、NRAS、SOS1、RAF1、BRAF、MEKl、MEK2、SH0C2和CBL中的突变。在NS和LS中最常突变的基因是PTPN11。在~50%的NS病例中和几乎所有的具有NS的某些特征的LS患者中发现了PTPN11(SHP2)的种系突变。对于NS,蛋白质中的Y62D和Y63C是最常见的突变。这两种突变影响SHP2的无催化活性的构象,而不干扰磷酸酶与其磷酸化信号配偶体的结合。Noonan Syndrome (NS) and Leopard Spot Syndrome (LS)-PTPNll mutations cause LS (multiple pigmented nevus syndrome, abnormal electrocardiogram conduction, distance between the eyes, pulmonary valve stenosis, abnormal genitalia, growth retardation, Sensorineural deafness) and NS (including heart defects, craniofacial deformities, and congenital abnormalities of short stature). These two disorders are part of a family of autosomal dominant syndromes caused by germline mutations in components of the RAS/RAF/MEK/ERK mitogen-activated protein kinase pathway (required for normal cell growth and differentiation). Abnormal regulation of this pathway has a profound impact, especially on the development of the heart, leading to a variety of abnormalities, including valvuloseptal defects and/or hypertrophic cardiomyopathy (HCM). It has been determined that the disturbance of the MAPK signaling pathway is important for these disorders, and some candidate genes along this pathway have been identified in humans, including KRAS, NRAS, SOS1, RAF1, BRAF, MEK1, MEK2, SHOC2 and Mutations in CBL. The most frequently mutated gene in NS and LS is PTPN11. Germline mutations of PTPN11 (SHP2) are found in ~50% of NS cases and almost all LS patients with certain characteristics of NS. For NS, Y62D and Y63C in the protein are the most common mutations. These two mutations affect the catalytically inactive conformation of SHP2 without interfering with the binding of phosphatase to its phosphorylation signal partner.
幼年型粒-单核细胞白血病(JMML)—在约35%的JMML(—种儿童期骨髓增生病(MPD))患者中发生PTPN11(SHP2)中的体细胞突变。这些功能获得性突变通常是N-SH2结构域或磷酸酶结构域中的点突变,其阻止催化结构域和N-SH2结构域之间的自我抑制,产生SHP2活性。Juvenile myelomonocytic leukemia (JMML)—Somatic mutations in PTPN11 (SHP2) occur in approximately 35% of JMML (a childhood myelodysplastic (MPD)) patients. These gain-of-function mutations are usually point mutations in the N-SH2 domain or the phosphatase domain, which prevent self-inhibition between the catalytic domain and the N-SH2 domain, resulting in SHP2 activity.
急性髓样白血病——已经在~10%的儿科急性白血病如骨髓发育不良综合征(MDS)、~7%的B细胞急性淋巴母细胞白血病(B-ALL)和~4%的急性髓样白血病(AML)中识别出PTPNll突变。Acute Myeloid Leukemia-Already in ~10% of pediatric acute leukemias such as myelodysplastic syndrome (MDS), ~7% of B-cell acute lymphoblastic leukemia (B-ALL) and ~4% of acute myeloid leukemia PTPN11 mutation was identified in (AML).
NS和白血病突变引起位于由自身抑制SHP2构象中的N-SH2和PTP结构域所形成的界面处的氨基酸的变化,破坏抑制性分子内相互作用,导致催化结构域活动过度。Mutations in NS and leukemia cause changes in amino acids at the interface formed by the N-SH2 and PTP domains in the self-inhibiting SHP2 conformation, disrupting inhibitory intramolecular interactions, leading to excessive activity in the catalytic domain.
SHP2在受体酪氨酸激酶(RTK)信号传导中充当正调节剂。含有RTK改变(EGFR amp,Her2 amp,FGFR amp,Met amp,易位/活化的RTK,即ALK,BCR/ABL)的癌症包括食管癌、乳癌、肺癌、结肠癌、胃癌、神经胶质瘤、头颈癌。 SHP2 acts as a positive regulator in receptor tyrosine kinase (RTK) signaling. Cancers containing RTK changes (EGFR amp , Her2 amp , FGFR amp , Met amp , translocation/activated RTK, namely ALK, BCR/ABL) include esophageal cancer, breast cancer, lung cancer, colon cancer, gastric cancer, glioma, Head and neck cancer.
食管癌(或食道癌)是食管的恶性病。存在多种亚型,主要是鳞状细胞癌(<50%)和腺癌。在食管腺癌和鳞状细胞癌中有高比率的RTK表达。因此,本发明的SHP2抑制剂可用于创新的治疗策略。Esophageal cancer (or esophageal cancer) is a malignant disease of the esophagus. There are many subtypes, mainly squamous cell carcinoma (<50%) and adenocarcinoma. There is a high rate of RTK expression in esophageal adenocarcinoma and squamous cell carcinoma. Therefore, the SHP2 inhibitor of the present invention can be used in innovative treatment strategies.
乳癌是一种重要类型的癌症,并且是女性的主要死因,其中患者对现有药物出现抗性。存在四种主要的乳癌亚型,包括luminal A、luminal B、Her21ik和三阴/Basal-like。三阴乳癌(TNBC)是缺少特定靶向治疗的侵袭性乳癌。表皮生长因子受体I(EGFR)已经显现为TNBC中的有前景的靶标。Her2以及EGFR经由SHP2的抑制可以是乳癌的有前景的治疗。Breast cancer is an important type of cancer and the leading cause of death for women, where patients develop resistance to existing drugs. There are four main subtypes of breast cancer, including luminal A, luminal B, Her21ik, and three-negative/Basal-like. Triple negative breast cancer (TNBC) is an aggressive breast cancer lacking specific targeted therapy. Epidermal growth factor receptor I (EGFR) has emerged as a promising target in TNBC. Inhibition of Her2 and EGFR via SHP2 may be a promising treatment for breast cancer.
肺癌——NSCLC目前是癌症相关死亡率的重要原因。占约85%的肺癌(主要是腺癌和鳞状细胞癌)。虽然细胞毒性化学治疗仍然是治疗的重要部分,但是基于肿瘤中遗传改变 如EGFR和ALK的靶向治疗更可能得益于靶向治疗。Lung cancer-NSCLC is currently an important cause of cancer-related mortality. It accounts for about 85% of lung cancers (mainly adenocarcinoma and squamous cell carcinoma). Although cytotoxic chemotherapy is still an important part of treatment, targeted therapy based on genetic changes in tumors such as EGFR and ALK is more likely to benefit from targeted therapy.
结肠癌——已知约30%至50%的结肠直肠肿瘤具有突变(异常)的KRAS,BRAF突变发生在10至15%的结肠直肠癌中。对于其结肠直肠肿瘤已经被证明过表达EGFR的患者亚群而言,这些患者呈现出对抗-EGFR疗法的有利的临床响应。Colon cancer-Approximately 30% to 50% of colorectal tumors are known to have mutated (abnormal) KRAS, and BRAF mutations occur in 10 to 15% of colorectal cancers. For the subgroup of patients whose colorectal tumors have been shown to overexpress EGFR, these patients present a favorable clinical response to anti-EGFR therapy.
胃癌是最流行的癌症类型之一。酪氨酸激酶的异常表达(如通过胃癌细胞中的异常酪氨酸磷酸化所反映的那样)是本领域已知的。在胃癌中经常扩增三种受体酪氨酸激酶,即c-met(HGF受体)、FGF受体2和erbB2/neu。因此,不同信号途径的破坏可促进不同胃癌类型的进程。Gastric cancer is one of the most popular types of cancer. Abnormal expression of tyrosine kinases (as reflected by abnormal tyrosine phosphorylation in gastric cancer cells) is known in the art. Three receptor tyrosine kinases are often amplified in gastric cancer, namely c-met (HGF receptor), FGF receptor 2 and erbB2/neu. Therefore, the destruction of different signal pathways can promote the progression of different types of gastric cancer.
成神经细胞瘤是发育中的交感神经系统的儿科肿瘤,占约8%的儿童癌症。间变性淋巴瘤激酶(ALK)基因的基因组改变已经被提出促进成神经细胞瘤发病机制。Neuroblastoma is a pediatric tumor of the developing sympathetic nervous system, accounting for about 8% of childhood cancers. Genomic changes in the anaplastic lymphoma kinase (ALK) gene have been proposed to promote the pathogenesis of neuroblastoma.
头和颈鳞状细胞癌(SCCHN)——高水平的EGFR表达与多种癌症、最常见是头和颈鳞状细胞癌(SCCHN)中的预后不良和对放射治疗的抗性相关。EGFR信号阻断导致抑制受体刺激、细胞增殖、侵袭和转移下降。因此,在SCCHN中,EGFR是新抗癌疗法的最佳靶标。Squamous cell carcinoma of the head and neck (SCCHN)-High levels of EGFR expression are associated with poor prognosis and resistance to radiation therapy in many cancers, most commonly squamous cell carcinoma of the head and neck (SCCHN). Blocking of EGFR signal leads to inhibition of receptor stimulation, cell proliferation, invasion and metastasis decline. Therefore, in SCCHN, EGFR is the best target for new anti-cancer therapies.
本发明涉及能够抑制SHP2活性的化合物。本发明还提供了本发明的化合物的制备方法和包含该化合物的药物制剂。本发明的另一方面涉及治疗与SHP2-介导的疾病或病症的方法,该方法包括给需要其的患者施用治疗有效量的如本发明所述的式I化合物的步骤。The present invention relates to compounds capable of inhibiting the activity of SHP2. The present invention also provides a preparation method of the compound of the present invention and a pharmaceutical preparation containing the compound. Another aspect of the present invention relates to a method of treating a disease or condition mediated by SHP2-, which method comprises the step of administering a therapeutically effective amount of a compound of formula I according to the present invention to a patient in need thereof.
在一些实施方式中,本发明涉及如上所述的方法,其中所述的SHP2-介导的疾病或病症是选自但不限于如下的癌症:JMML,AML,MDS,B-ALL,成神经细胞瘤,食管癌,乳癌,肺癌,结肠癌,胃癌,头颈癌。In some embodiments, the present invention relates to the method as described above, wherein the SHP2-mediated disease or disorder is selected from but not limited to the following cancers: JMML, AML, MDS, B-ALL, neuroblasts Tumor, esophageal cancer, breast cancer, lung cancer, colon cancer, stomach cancer, head and neck cancer.
本发明所述的化合物还可用于治疗与SHP2异常活性相关的其它疾病或病症。因此,作为优选的实施方式,本发明涉及治疗选自如下的疾病或病症的方法:NS,LS,JMML,AML,MDS,B-ALL,成神经细胞瘤,食管癌,乳癌,肺癌,结肠癌,胃癌,头颈癌。The compounds of the present invention can also be used to treat other diseases or disorders related to the abnormal activity of SHP2. Therefore, as a preferred embodiment, the present invention relates to a method for treating diseases or conditions selected from the group consisting of: NS, LS, JMML, AML, MDS, B-ALL, neuroblastoma, esophageal cancer, breast cancer, lung cancer, colon cancer , Stomach cancer, head and neck cancer.
本发明所述的SHP2抑制剂可以与其它具有药理活性的化合物或与两种或更多种其它具有药理活性的化合物组合,尤其是在治疗癌症中。例如,本发明所述的式(I)化合物或其药学上可接受的盐可以与一种或多种选自如下的物质组合地同时、依次或分别施用:化疗剂,比如有丝分裂抑制剂,如紫杉烷、长春花生物碱、紫杉醇、多西他赛、长春花新碱、长春花碱、长春瑞滨或长春氟宁,其它抗癌剂如顺铂、5-氟尿嘧啶或5-氟-2-4(1H,3H)-嘧啶二酮(5FU)、氟他胺或吉西他滨。The SHP2 inhibitor of the present invention can be combined with other pharmacologically active compounds or with two or more other pharmacologically active compounds, especially in the treatment of cancer. For example, the compound of formula (I) according to the present invention or a pharmaceutically acceptable salt thereof can be administered simultaneously, sequentially or separately in combination with one or more substances selected from the group consisting of chemotherapeutic agents, such as mitotic inhibitors, such as Taxanes, vinca alkaloids, paclitaxel, docetaxel, vinblastine, vinblastine, vinorelbine or vinflunine, other anticancer agents such as cisplatin, 5-fluorouracil or 5-fluoro-2 -4(1H,3H)-pyrimidinedione (5FU), flutamide or gemcitabine.
一些组合在疗法中可以提供显著的优点,包括协同活性。Some combinations can provide significant advantages in therapy, including synergistic activity.
在一些实施方式中,本发明涉及如上所述的方法,其中所述化合物经胃肠外施用。In some embodiments, the present invention relates to a method as described above, wherein the compound is administered parenterally.
在一些实施方式中,本发明涉及如上所述的方法,其中所述化合物经肌内、静脉内、 皮下、口服、经肺、鞘内、局部或经鼻内施用。In some embodiments, the present invention relates to a method as described above, wherein the compound is administered intramuscularly, intravenously, subcutaneously, orally, pulmonary, intrathecal, topically, or intranasally.
在一些实施方式中,本发明涉及如上所述的方法,其中所述化合物经全身施用。In some embodiments, the present invention relates to a method as described above, wherein the compound is administered systemically.
在一些实施方式中,本发明涉及如上所述的方法,其中所述患者是哺乳动物。In some embodiments, the present invention relates to a method as described above, wherein the patient is a mammal.
在一些实施方式中,本发明涉及如上所述的方法,其中所述患者是灵长类动物。In some embodiments, the present invention relates to a method as described above, wherein the patient is a primate.
在一些实施方式中,本发明涉及如上所述的方法,其中所述患者是人。In some embodiments, the present invention relates to a method as described above, wherein the patient is a human.
在一些实施方式中,本发明涉及治疗SHP2-介导的疾病或病症的方法,该方法包括如下步骤:给需要其的患者施用治疗有效量的化疗剂与如治疗有效量的本发明所述的式I化合物的组合。In some embodiments, the present invention relates to a method of treating SHP2-mediated diseases or conditions, the method comprising the steps of: administering to a patient in need thereof a therapeutically effective amount of a chemotherapeutic agent and a therapeutically effective amount of a chemotherapeutic agent as described in the present invention. Combinations of compounds of formula I.
本发明的主要优点包括:The main advantages of the present invention include:
1.提供了一种如式I所示的化合物。1. A compound of formula I is provided.
2.提供了一种结构新颖的SHP2抑制剂及其制备和应用,所述的抑制剂对SHP2具有较高的抑制活性。2. A novel structure of SHP2 inhibitor and its preparation and application are provided, and the inhibitor has high inhibitory activity on SHP2.
3.提供了一类治疗与SHP2相关疾病或病症的药物组合物。3. A pharmaceutical composition for treating SHP2 related diseases or disorders is provided.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。The present invention will be further described below in conjunction with specific embodiments. It should be understood that these embodiments are only used to illustrate the present invention and not to limit the scope of the present invention. The experimental methods that do not indicate specific conditions in the following examples usually follow the conventional conditions or the conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are calculated by weight.
下述实施例中所用的起始物可由化学品销售商如Aldrich、TCI、Alfa Aesar、毕得、安耐吉等处购得,或者可通过已知的方法来合成。The starting materials used in the following examples can be purchased from chemical vendors such as Aldrich, TCI, Alfa Aesar, Beide, Anaiji, etc., or can be synthesized by known methods.
下述实施例中,冰浴是指-5℃至0℃,室温是指10℃至30℃,回流温度一般是指溶剂在常压下的回流温度。反应过夜一般是指时间为8-15小时。下述实施例中,未限定具体操作温度的,均在室温下进行。In the following examples, ice bath refers to -5°C to 0°C, room temperature refers to 10°C to 30°C, and reflux temperature generally refers to the reflux temperature of the solvent under normal pressure. The reaction overnight generally means 8-15 hours. In the following examples, where the specific operating temperature is not limited, all are performed at room temperature.
下述实施例中,中间体和最终产物的分离提纯是通过正相或反相色谱柱分离或者其它合适的方法。正相快速色谱柱是用乙酸乙酯和正己烷或甲醇和二氯甲烷等作为流动相。反相制备性高压液相色谱(HPLC)是用C18柱并用UV 214nm和254nm来检测,其流动相为A(水和0.1%甲酸)、B(乙腈)或者流动相A(水和0.1%碳酸氢铵)、B(乙腈)。In the following examples, the separation and purification of the intermediate and the final product is by normal phase or reverse phase chromatographic column separation or other suitable methods. The normal phase flash chromatography column uses ethyl acetate and n-hexane or methanol and dichloromethane as the mobile phase. Reversed-phase preparative high pressure liquid chromatography (HPLC) uses C18 column and UV 214nm and 254nm to detect, and its mobile phase is A (water and 0.1% formic acid), B (acetonitrile) or mobile phase A (water and 0.1% carbonic acid) Hydrogen ammonium), B (acetonitrile).
各实施例中:LCMS仪器:Pump Agilent 1260 UV检测器:Agilent 1260 DAD Mass Spectrometer API 3000;In each embodiment: LCMS instrument: Pump Agilent 1260 UV detector: Agilent 1260 DAD Mass Spectrometer API 3000;
层析柱:Waters sunfire C18,4.6×50mm,5um;Chromatography column: Waters sunfire C18, 4.6×50mm, 5um;
流动相:A-H2O(0.1%HCOOH);B-乙腈NMR;Mobile phase: A-H2O (0.1% HCOOH); B-acetonitrile NMR;
仪器:Bruker Ascend 400M(1H NMR:400MHz; 13C NMR:100MHz)。 Instrument: Bruker Ascend 400M (1H NMR: 400 MHz; 13 C NMR: 100 MHz).
中间体A的合成:2-苯基8-(叔丁基)(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-2,8-二甲酸酯Synthesis of Intermediate A: 2-Phenyl 8-(tert-butyl)(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5] Decane-2,8-Dicarboxylate
Figure PCTCN2020106214-appb-000035
Figure PCTCN2020106214-appb-000035
步骤一:在氮气保护下向干燥的1L三口瓶中依次加入1-(叔丁基)4-乙基哌啶-1,4-二羧酸酯(20.0g,77.7mmol),THF(200mL),在-78℃下滴加LDA(2.0M正己烷溶液,46.6mL,93.2mmol),反应液在-40℃下搅拌2小时。用针管将此反应液导入到另一个氮气保护下的-60℃的装有氯乙酰氯(50.0g,442.9mmol)的THF(200mL)溶液的三口瓶中,反应液在搅拌下缓慢升至室温,得到的白色悬浊液在室温下继续搅拌2小时。加入饱和碳酸氢钠溶液和乙酸乙酯,有机相分离,水相用乙酸乙酯继续萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(0至10%梯度的乙酸乙酯/石油醚),得到不纯的黄色油状物A-1(34.0g)。没有进一步纯化直接投入下一步反应。Step 1: Add 1-(tert-butyl)4-ethylpiperidine-1,4-dicarboxylate (20.0g, 77.7mmol), THF (200mL) to a dry 1L three-necked flask under the protection of nitrogen. , LDA (2.0M n-hexane solution, 46.6 mL, 93.2 mmol) was added dropwise at -78°C, and the reaction solution was stirred at -40°C for 2 hours. Use a needle to introduce the reaction solution into another three-necked flask containing chloroacetyl chloride (50.0g, 442.9mmol) in THF (200mL) at -60°C under the protection of nitrogen. The reaction solution is slowly raised to room temperature under stirring. , The resulting white suspension was stirred at room temperature for 2 hours. Saturated sodium bicarbonate solution and ethyl acetate were added, the organic phase was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (0 to 10% ethyl acetate/petroleum ether gradient) to give an impure yellow oil A-1 (34.0 g). There is no further purification directly into the next reaction.
步骤二:向干燥的500mL单口瓶中依次加入化合物A-1(34.0g)、DMF(300mL)和叠氮钠(5.56g,85.5mmol)。反应液在25℃下搅拌3小时。反应液加水稀释,水相用石油醚:乙酸乙酯(1:1)的混合物萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物A-2(34.6g,粗品)。没有进一步纯化直接投入下一步反应。Step 2: Add compound A-1 (34.0 g), DMF (300 mL) and sodium azide (5.56 g, 85.5 mmol) to a dry 500 mL single-neck flask in sequence. The reaction solution was stirred at 25°C for 3 hours. The reaction solution was diluted with water, and the aqueous phase was extracted 3 times with a mixture of petroleum ether: ethyl acetate (1:1). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a yellow oil A-2 (34.6 g, crude product). There is no further purification directly into the next reaction.
步骤三:向干燥的500mL单口瓶中依次加入化合物A-2(34.6g,粗品)、THF(300mL)、 H 2O(30mL)和三苯基膦(26.5g,101.0mmol)。反应液在60℃下搅拌4小时。冷却到室温后,反应液加水稀释,用乙酸乙酯萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/乙酸乙酯=4:1),得到黄色固体A-3(8.0g,收率:38.5%)。 Step 3: Add compound A-2 (34.6 g, crude product), THF (300 mL), H 2 O (30 mL) and triphenylphosphine (26.5 g, 101.0 mmol) into a dry 500 mL single-neck flask in sequence. The reaction solution was stirred at 60°C for 4 hours. After cooling to room temperature, the reaction solution was diluted with water and extracted twice with ethyl acetate. The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/ethyl acetate=4:1) to obtain yellow solid A-3 (8.0 g, yield: 38.5%).
1H NMR(400MHz,CDCl 3)δ6.54(brs,1H),3.91(s,2H),3.85-3.75(m,2H),3.65-3.55(m,2H),1.89–1.76(m,2H),1.74–1.64(m,2H),1.47(s,9H)。 1 H NMR (400MHz, CDCl 3 ) δ6.54 (brs, 1H), 3.91 (s, 2H), 3.85-3.75 (m, 2H), 3.65-3.55 (m, 2H), 1.89-1.76 (m, 2H) ), 1.74–1.64 (m, 2H), 1.47 (s, 9H).
步骤四:向干燥的100mL单口瓶中依次加入化合物A-3(4.0g,14.9mmol)、THF(40mL)和硼氢化钠(282mg,7.45mmol)。反应液在25℃下搅拌1小时。反应完毕后,将反应液过滤。得到的滤液(化合物A-4的THF溶液)直接用于下一步反应。Step 4: Add compound A-3 (4.0 g, 14.9 mmol), THF (40 mL) and sodium borohydride (282 mg, 7.45 mmol) to a dry 100 mL single-neck flask in sequence. The reaction solution was stirred at 25°C for 1 hour. After the reaction is completed, the reaction liquid is filtered. The obtained filtrate (the THF solution of compound A-4) was directly used in the next reaction.
步骤五:在-30℃搅拌下向干燥的100mL单口瓶中依次加入上步得到的化合物A-4的THF溶液(40mL)、硼烷二甲硫醚(22.4mL,44.7mmol,2M四氢呋喃溶液)。反应液在搅拌下1小时内缓慢升至25℃,然后在60℃下搅拌6小时。反应完毕后,向反应液中滴加甲醇(10mL)淬灭,得到的混合物减压浓缩去除挥发物。得到的残留物溶解在甲醇(20mL)中,得到的溶液在70℃下搅拌4小时。将溶液减压浓缩,得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=10:1-1:1),得到白色固体A-5(1.35g,收率:35.3%)。Step 5: Add the THF solution (40mL) of compound A-4 obtained in the previous step and borane dimethyl sulfide (22.4mL, 44.7mmol, 2M tetrahydrofuran solution) into a dry 100mL single-neck flask with stirring at -30℃. . The reaction solution was slowly raised to 25°C within 1 hour under stirring, and then stirred at 60°C for 6 hours. After the reaction was completed, methanol (10 mL) was added dropwise to the reaction solution for quenching, and the resulting mixture was concentrated under reduced pressure to remove volatiles. The obtained residue was dissolved in methanol (20 mL), and the obtained solution was stirred at 70°C for 4 hours. The solution was concentrated under reduced pressure, and the obtained residue was purified by silica gel chromatography (dichloromethane/methanol=10:1-1:1) to obtain white solid A-5 (1.35 g, yield: 35.3%).
1H NMR(400MHz,CDCl 3)δ3.96–3.90(m,1H),3.70–3.56(m,2H),3.30–3.10(m,3H),3.00–2.83(m,3H),1.80–1.70(m,1H),1.60–1.36(m,12H); 1 H NMR (400MHz, CDCl 3 ) δ 3.96-3.90 (m, 1H), 3.70-3.56 (m, 2H), 3.30--3.10 (m, 3H), 3.00-2.83 (m, 3H), 1.80-1.70 (m,1H),1.60–1.36(m,12H);
LCMS:m/z 257.2[M+H] +LCMS: m/z 257.2 [M+H] + .
步骤六:在干燥的100mL单口瓶中依次加入化合物A-5(1.35g,5.27mmol)、THF(14mL)、水(7mL)、碳酸氢钠(2.22g,26.4mmol)和氯甲酸苄酯(1.35g,7.91mmol)。反应液在20℃下搅拌12小时。反应完毕后,反应液加水(60mL)稀释,用乙酸乙酯(60mL)萃取2次。合并的有机相用饱和食盐水(60mL)洗涤2次、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(石油醚/乙酸乙酯=1:1),得到无色油状物A-6(1.40g,收率:68.0%)。Step 6: Add compound A-5 (1.35g, 5.27mmol), THF (14mL), water (7mL), sodium bicarbonate (2.22g, 26.4mmol) and benzyl chloroformate ( 1.35g, 7.91mmol). The reaction solution was stirred at 20°C for 12 hours. After the reaction was completed, the reaction solution was diluted with water (60 mL), and extracted twice with ethyl acetate (60 mL). The combined organic phase was washed twice with saturated brine (60 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:1) to obtain A-6 (1.40 g, yield: 68.0%) as a colorless oil.
1H NMR(400MHz,CDCl 3)δ7.39–7.29(m,5H),5.13(s,2H),4.00–3.90(m,1H),3.85–3.65(m,3H),3.55–3.30(m,3H),3.20–2.90(m,2H),1.78–1.53(m,4H),1.52-1.42(m,10H),1.40–1.32(m,1H); 1 H NMR (400MHz, CDCl 3 ) δ 7.39-7.29 (m, 5H), 5.13 (s, 2H), 4.00-3.90 (m, 1H), 3.85-3.65 (m, 3H), 3.55--3.30 (m ,3H), 3.20–2.90(m,2H), 1.78–1.53(m,4H), 1.52-1.42(m,10H), 1.40–1.32(m,1H);
LCMS:m/z 413.0[M+Na] +LCMS: m/z 413.0 [M+Na] + .
步骤七:在干燥的100mL单口瓶中依次加入化合物A-6(2.30g,5.89mmol)、DCM(50mL)和戴斯-马丁氧化剂(3.75g,8.83mmol)。反应液在25℃下搅拌1小时。TLC板检测反应完毕后,向反应液加入饱和碳酸氢钠溶液(30mL)和饱和硫代硫酸钠溶液(30mL),得到的混合物在25℃下搅拌30分钟。用乙酸乙酯(60mL)萃取2次。合并的有机相用饱和食 盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(石油醚/乙酸乙酯=1:0到2:1),得到无色油状物A-7(2.10g,收率:91.7%)。Step 7: Add compound A-6 (2.30 g, 5.89 mmol), DCM (50 mL) and Dess-Martin oxidant (3.75 g, 8.83 mmol) into a dry 100 mL single-neck flask in sequence. The reaction solution was stirred at 25°C for 1 hour. After the completion of the reaction detected by the TLC plate, saturated sodium bicarbonate solution (30 mL) and saturated sodium thiosulfate solution (30 mL) were added to the reaction solution, and the resulting mixture was stirred at 25° C. for 30 minutes. Extract twice with ethyl acetate (60 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:0 to 2:1) to obtain colorless oil A-7 (2.10 g, yield: 91.7%).
1H NMR(400MHz,CDCl 3)δ7.42–7.30(m,5H),5.18(s,2H),4.00–3.80(m,4H),3.77–3.68(m,2H),3.12–2.96(m,2H),1.80–1.70(m,2H),1.53–1.40(m,11H)。 1 H NMR (400MHz, CDCl 3 ) δ7.42--7.30 (m, 5H), 5.18 (s, 2H), 4.00 - 3.80 (m, 4H), 3.77 - 3.68 (m, 2H), 3.12 - 2.96 (m ,2H), 1.80–1.70(m,2H), 1.53–1.40(m,11H).
步骤八:在干燥的100mL单口瓶中依次加入化合物A-7(1.60g,4.12mmol)、(R)-叔丁基亚磺酰胺(999mg,8.24mmol)、THF(30mL)和钛酸四乙酯(2.83g,12.4mmol)。反应液在20℃下搅拌40小时。反应完毕后,在搅拌下向反应液加入乙酸乙酯(30mL)和饱和氯化钠溶液(6mL),得到的混合物过滤。滤液用乙酸乙酯(40mL)萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(石油醚/乙酸乙酯=1:0到2:1),得到白色固体A-8(1.60g,收率:78.8%)。Step 8: Add compound A-7 (1.60g, 4.12mmol), (R)-tert-butylsulfinamide (999mg, 8.24mmol), THF (30mL) and tetraethyl titanate in sequence in a dry 100mL single-mouth flask Esters (2.83 g, 12.4 mmol). The reaction solution was stirred at 20°C for 40 hours. After the reaction was completed, ethyl acetate (30 mL) and saturated sodium chloride solution (6 mL) were added to the reaction solution under stirring, and the resulting mixture was filtered. The filtrate was extracted 3 times with ethyl acetate (40 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:0 to 2:1) to obtain white solid A-8 (1.60 g, yield: 78.8%).
1H NMR(400MHz,CDCl 3)δ7.41–7.28(m,5H),5.20-5.10(m,2H),4.77–4.67(m,1H),4.49-4,39(m,1H),4.10–3.90(m,2H),3.68–3.55(m,1H),3.53–3.46(m,1H),3.02–2.88(m,2H),1.88–1.77(m,1H),1.76–1.64(m,2H),1.63–1.43(m,11H),1.26(s,9H)。 1 H NMR (400MHz, CDCl 3 ) δ7.41--7.28 (m, 5H), 5.20-5.10 (m, 2H), 4.77-4.67 (m, 1H), 4.49-4, 39 (m, 1H), 4.10 --3.90(m,2H),3.68–3.55(m,1H),3.53–3.46(m,1H),3.02–2.88(m,2H),1.88–1.77(m,1H),1.76–1.64(m, 2H), 1.63-1.43 (m, 11H), 1.26 (s, 9H).
步骤九:在-78℃氮气保护下向干燥的50mL三口瓶中依次加入化合物A-8(500mg,1.02mmol)的THF(10mL)溶液和二异丁基氢化铝溶液(0.89mL,1.33mmol,1.5M)。反应液在-78℃下搅拌0.5小时。反应完毕后,用酒石酸钾钠(20mL)淬灭反应,并在20℃下继续搅拌0.5小时。得到的混合物用乙酸乙酯(30mL)萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(石油醚/乙酸乙酯=1:0-1:1),得到无色油状物A-9(300mg,收率:59.5%)。Step 9: Add compound A-8 (500mg, 1.02mmol) in THF (10mL) solution and diisobutylaluminum hydride solution (0.89mL, 1.33mmol) to a dry 50mL three-necked flask under nitrogen protection at -78℃. 1.5M). The reaction solution was stirred at -78°C for 0.5 hour. After the reaction was completed, the reaction was quenched with potassium sodium tartrate (20 mL), and stirring was continued for 0.5 hour at 20°C. The resulting mixture was extracted 3 times with ethyl acetate (30 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:0-1:1) to obtain A-9 (300 mg, yield: 59.5%) as a colorless oil.
步骤十:在干燥的25mL单口瓶中依次加入化合物A-9(300mg,0.608mmol)、乙醇(3mL)和10%的氢氧化钠水溶液(3mL)。反应液在85℃下搅拌8小时。冷却至室温后,反应液用0.5N稀盐酸调至pH=7,得到的混合物减压浓缩去除挥发物。向得到的残留物中加入DCM/MeOH(2:1)的混合溶剂(10mL),过滤,将得到的滤液减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=1:0-10:1)得到无色油状物A(210mg,收率:95.9%)。Step 10: Add compound A-9 (300 mg, 0.608 mmol), ethanol (3 mL) and 10% aqueous sodium hydroxide solution (3 mL) in a dry 25 mL single-neck flask in sequence. The reaction solution was stirred at 85°C for 8 hours. After cooling to room temperature, the reaction solution was adjusted to pH=7 with 0.5N dilute hydrochloric acid, and the resulting mixture was concentrated under reduced pressure to remove volatiles. A mixed solvent (10 mL) of DCM/MeOH (2:1) was added to the obtained residue, filtered, and the obtained filtrate was concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=1:0-10:1) to obtain a colorless oil A (210 mg, yield: 95.9%).
中间体B1的合成:(R)-N-((S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2,8-二氮杂螺[4.5]癸烷-4-基)-2-甲基丙基-2-亚磺酰胺Synthesis of intermediate B1: (R)-N-((S)-8-(5-((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2, 8-diazaspiro[4.5]decane-4-yl)-2-methylpropyl-2-sulfinamide
Figure PCTCN2020106214-appb-000036
Figure PCTCN2020106214-appb-000036
步骤一:在干燥的25mL单口瓶中依次加入化合物A-9(330mg,0.668mmol),DCM(10mL),在0℃下滴加TMSOTf(298mg,1.34mmol),反应液在20℃下搅拌2小时。LCMS检测反应完全。向反应液中加入甲醇(1mL)和饱和碳酸氢钠溶液(1mL)淬灭。得到的混合物用水(20mL)稀释,用二氯甲烷(20mL)萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩,得到无色油状物B1-1(263mg,粗品)。Step 1: Add compound A-9 (330mg, 0.668mmol) and DCM (10mL) in a dry 25mL single-mouth flask, add TMSOTf (298mg, 1.34mmol) dropwise at 0℃, and stir the reaction solution at 20℃2 hour. LCMS detected that the reaction was complete. The reaction solution was quenched by adding methanol (1 mL) and saturated sodium bicarbonate solution (1 mL). The resulting mixture was diluted with water (20 mL) and extracted twice with dichloromethane (20 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain B1-1 (263 mg, crude product) as a colorless oil.
LCMS:m/z 394.1[M+H] +LCMS: m/z 394.1 [M+H] + .
步骤二:向干燥的25mL单口瓶中依次加入化合物B1-1(263mg,0.668mmol)、乙腈(5.0mL)、N,N-二异丙基乙胺(1.0mL)和3-氯-4-((5-氯吡嗪-2-基)硫基)吡啶-2-胺(C2,182mg,0.668mmol)。反应液在90℃下搅拌10小时。反应液减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=10:1),得到黄色固体B1-2(400mg,收率:95.0%)。Step 2: Add compound B1-1 (263mg, 0.668mmol), acetonitrile (5.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-mouth flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (C2, 182 mg, 0.668 mmol). The reaction solution was stirred at 90°C for 10 hours. The reaction solution was concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=10:1) to obtain a yellow solid B1-2 (400 mg, yield: 95.0%).
LCMS:m/z 630.0[M+H] +LCMS: m/z 630.0 [M+H] + .
步骤三:向干燥的25mL单口瓶中依次加入化合物B1-2(400mg,0.635mmol)、乙醇(9mL)和10%的氢氧化钠水溶液(3mL)。反应液在85℃下搅拌8小时。冷却至室温后,反应液减压浓缩去除挥发物。得到的残留物加水(20mL)稀释,用二氯甲烷和甲醇混合溶剂(10/1,20mL)萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩,得到黄色固体B1(300mg,粗品)。Step 3: Add compound B1-2 (400 mg, 0.635 mmol), ethanol (9 mL) and 10% aqueous sodium hydroxide solution (3 mL) to a dry 25 mL single-necked flask in sequence. The reaction solution was stirred at 85°C for 8 hours. After cooling to room temperature, the reaction solution was concentrated under reduced pressure to remove volatiles. The obtained residue was diluted with water (20 mL), and extracted 3 times with a mixed solvent of dichloromethane and methanol (10/1, 20 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a yellow solid B1 (300 mg, crude product).
LCMS:m/z 496.1[M+H] +LCMS: m/z 496.1 [M+H] + .
中间体B2的合成:5-氯-8-碘-[1,2,4]三氮唑并[4,3-c]嘧啶Synthesis of intermediate B2: 5-chloro-8-iodo-[1,2,4]triazolo[4,3-c]pyrimidine
Figure PCTCN2020106214-appb-000037
Figure PCTCN2020106214-appb-000037
步骤一:向干燥的100mL烧瓶中加入2,4-二氯-5-碘嘧啶(1.096g,4mmol)和20mL无水乙醇。在0℃氮气条件下,向其中缓慢加入80%水合肼(601mg,12mmol)混合物继续搅拌反应1小时。反应完毕后,将混合物过滤并用无水乙醇洗涤得到2-氯-4-肼基-5-碘嘧啶B2-1(850m g,产率78.7%)。Step 1: Add 2,4-dichloro-5-iodopyrimidine (1.096g, 4mmol) and 20mL of absolute ethanol to a dry 100mL flask. Under the condition of nitrogen at 0°C, 80% hydrazine hydrate (601 mg, 12 mmol) was slowly added to the mixture to continue stirring and reacting for 1 hour. After the reaction was completed, the mixture was filtered and washed with absolute ethanol to obtain 2-chloro-4-hydrazino-5-iodopyrimidine B2-1 (850 mg, yield 78.7%).
1H NMR(400MHz,CDCl3)δ8.29(s,1H),6.67(s,1H),4.08(s,2H); 1 H NMR (400MHz, CDCl3) δ 8.29 (s, 1H), 6.67 (s, 1H), 4.08 (s, 2H);
LCMS:m/z 271.1[M+H] +LCMS: m/z 271.1 [M+H] + .
步骤二:向干燥的100mL烧瓶中依次加入2-氯-4-肼基-5-碘嘧啶(810mg,3mmol)和原甲酸三甲酯(10mL)。在氮气条件下,将混合物加热至85℃搅拌反应5小时。反应完毕后,将获得的残留物倒入饱和NaCl溶液(50mL),并使用乙酸乙酯(3×30mL)萃取,饱和食盐水洗涤并混合有机层,经无水硫酸钠干燥,过滤并在减压浓缩得到的残留物通过硅胶色谱法纯化(0至50%梯度的乙酸乙酯/石油醚),得到淡黄色固体5-氯-8-碘-[1,2,4]三唑并[4,3-c]嘧啶B2(420mg,收率:50%)。Step 2: Add 2-chloro-4-hydrazino-5-iodopyrimidine (810 mg, 3 mmol) and trimethyl orthoformate (10 mL) to a dry 100 mL flask in sequence. Under nitrogen, the mixture was heated to 85°C and stirred for 5 hours. After the completion of the reaction, the residue obtained was poured into saturated NaCl solution (50 mL), and extracted with ethyl acetate (3×30 mL), washed with saturated brine and mixed with the organic layer, dried over anhydrous sodium sulfate, filtered and reduced The residue obtained by pressure concentration was purified by silica gel chromatography (0 to 50% ethyl acetate/petroleum ether) to give 5-chloro-8-iodo-[1,2,4]triazolo[4 ,3-c]pyrimidine B2 (420 mg, yield: 50%).
LCMS:m/z 280.9[M+H] +LCMS: m/z 280.9 [M+H] + .
中间体B3的合成:5-氯-8-碘咪唑并[1,2-c]嘧啶Synthesis of intermediate B3: 5-chloro-8-iodoimidazo[1,2-c]pyrimidine
Figure PCTCN2020106214-appb-000038
Figure PCTCN2020106214-appb-000038
步骤一:向干燥的100mL烧瓶中依次加入2,4-二氯-5-碘嘧啶(1.37g,5mmol)和2,2-二甲氧基乙胺(8.4g,10mmol)和无水乙醇(50mL)。然后在0℃氮气条件下,向反应混合物中缓慢滴加三乙胺(1.01g,10mmol)之后将混合物在室温下搅拌反应10小时。反应完毕后,真空浓缩,得到的浓缩物加入15mL的水,并使用二氯甲烷(3×50mL)萃取,饱和食盐水洗涤并混合有机层,经无水硫酸钠干燥,过滤并浓缩得到白色固体2-氯-N-(2,2-二甲氧基乙基)-5-碘嘧啶-4-胺(B3-1,1.46g,收率:85%)。Step 1: Add 2,4-dichloro-5-iodopyrimidine (1.37g, 5mmol) and 2,2-dimethoxyethylamine (8.4g, 10mmol) and absolute ethanol ( 50mL). Then, under nitrogen at 0°C, triethylamine (1.01 g, 10 mmol) was slowly added dropwise to the reaction mixture, and then the mixture was stirred and reacted at room temperature for 10 hours. After the reaction is completed, it is concentrated in vacuo, the obtained concentrate is added with 15 mL of water, and extracted with dichloromethane (3×50 mL), washed with saturated brine and mixed with the organic layer, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a white solid 2-chloro-N-(2,2-dimethoxyethyl)-5-iodopyrimidin-4-amine (B3-1, 1.46 g, yield: 85%).
LC-MS:m/z 344.2[M+H] +LC-MS: m/z 344.2 [M+H] + .
步骤二:向干燥的100mL烧瓶中依次加入2-氯-N-(2,2-二甲氧基乙基)-5-碘嘧啶-4-胺(1.03g,3mmol)和10mL浓硫酸。在氮气条件下,将混合物加热至65℃搅拌反应2小时。反应完毕后,反应液冷却至室温,将混合物缓慢倒入冰水中,然后用4M的NaOH溶液调节pH约到6-7,过滤得到灰白色固体8-碘咪唑并[1,2-c]嘧啶-5-醇(B3-2,0.407g,产率52%)。Step 2: Add 2-chloro-N-(2,2-dimethoxyethyl)-5-iodopyrimidin-4-amine (1.03g, 3mmol) and 10mL concentrated sulfuric acid to a dry 100mL flask in sequence. Under nitrogen, the mixture was heated to 65°C and stirred for 2 hours. After the reaction is complete, the reaction solution is cooled to room temperature, the mixture is slowly poured into ice water, and then the pH is adjusted to about 6-7 with 4M NaOH solution, and the off-white solid 8-iodoimidazo[1,2-c]pyrimidine is obtained by filtration. 5-ol (B3-2, 0.407 g, yield 52%).
1H NMR(400MHz,DMSO-d6)δ11.81(s,1H),7.93(d,J=1.4Hz,1H),7.60(s,1H), 7.40(d,J=1.4Hz,1H);1H NMR(400MHz,DMSO-d6)δ11.81(s,1H),7.93(d,J=1.4Hz,1H),7.60(s,1H), 7.40(d,J=1.4Hz,1H);
LC-MS:m/z 262.2[M+H] +LC-MS: m/z 262.2 [M+H] + .
步骤三:向干燥的50mL单口烧瓶中依次加入8碘咪唑并[1,2-c]嘧啶-5-醇(0.522g,2mmol)和三氯氧磷(8mL),在氮气的保护下,缓慢滴加N,N-二异丙基乙胺(1mL),之后将混合物加热至120℃搅拌5小时。反应完毕后,反应液冷却至室温并将真空浓缩,然后加入饱和碳酸氢钠溶液猝灭,使用乙酸乙酯(3×40mL)萃取,经无水硫酸钠干燥,过滤并浓缩,将得到的残留物通过硅胶色谱法纯化(0至30%梯度的乙酸乙酯:石油醚)得到淡黄色固体5-氯-8-碘咪唑并[1,2-c]嘧啶(B3,0.360g,收率:55%)。Step 3: Add 8-iodoimidazo[1,2-c]pyrimidin-5-ol (0.522g, 2mmol) and phosphorus oxychloride (8mL) into a dry 50mL single-neck flask in sequence, slowly under the protection of nitrogen N,N-diisopropylethylamine (1 mL) was added dropwise, and then the mixture was heated to 120°C and stirred for 5 hours. After the completion of the reaction, the reaction solution was cooled to room temperature and concentrated in vacuo, and then quenched by adding saturated sodium bicarbonate solution, extracted with ethyl acetate (3×40 mL), dried over anhydrous sodium sulfate, filtered and concentrated, and the residue obtained The product was purified by silica gel chromatography (0 to 30% ethyl acetate: petroleum ether gradient) to give a pale yellow solid 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (B3, 0.360g, yield: 55%).
1H NMR(400MHz,DMSO-d6)δ8.24(s,1H),8.20(d,J=1.4Hz,1H),7.81(d,J=1.4Hz,1H); 1 H NMR(400MHz,DMSO-d6)δ8.24(s,1H), 8.20(d,J=1.4Hz,1H), 7.81(d,J=1.4Hz,1H);
LC-MS:m/z 280.1[M+H] +LC-MS: m/z 280.1 [M+H] + .
中间体B4的合成:5-氯-7-甲基-8-碘咪唑并[1,2-c]嘧啶Synthesis of intermediate B4: 5-chloro-7-methyl-8-iodoimidazo[1,2-c]pyrimidine
Figure PCTCN2020106214-appb-000039
Figure PCTCN2020106214-appb-000039
步骤一:冰水浴下向2,4-二氯-6-甲基嘧啶(8.0g,49.08mmol)的EtOH(60mL)溶液中加入氨水(25-28%的溶液,10mL)。室温搅拌反应16h,补加氨水(25-28%的溶液,5mL)。室温搅拌反应8h,补加氨水(25-28%的溶液,5mL)。室温搅拌16h。反应液减压浓缩至干后将得到的残留物通过硅胶色谱法(0至50%梯度的乙酸乙酯:石油醚)纯化得浅黄色固体2-氯-6-甲基嘧啶-4-胺(B4-1,2.4g,收率:34%)。Step 1: Add ammonia (25-28% solution, 10 mL) to 2,4-dichloro-6-methylpyrimidine (8.0 g, 49.08 mmol) in EtOH (60 mL) solution under ice water bath. The reaction was stirred at room temperature for 16 h, and ammonia water (25-28% solution, 5 mL) was added. The reaction was stirred at room temperature for 8 hours, and ammonia water (25-28% solution, 5 mL) was added. Stir at room temperature for 16h. After the reaction solution was concentrated to dryness under reduced pressure, the residue obtained was purified by silica gel chromatography (0 to 50% gradient ethyl acetate: petroleum ether) to obtain a pale yellow solid 2-chloro-6-methylpyrimidin-4-amine ( B4-1, 2.4g, yield: 34%).
1H NMR(400MHz,DMSO-d 6)δ7.25(s,2H),6.22(s,1H),2.18(s,3H)。 1 H NMR (400MHz, DMSO-d 6 ) δ 7.25 (s, 2H), 6.22 (s, 1H), 2.18 (s, 3H).
步骤二:向2-氯-6-甲基嘧啶-4-胺(B4-1,1.4g,9.75mmol)的DMF(15mL)溶液中加入NIS(3.29g,14.63mmol)。30℃搅拌反应16h。反应液倒入水中,过滤,滤饼用饱和亚硫酸钠溶液洗,水洗得淡黄色固体2-氯-5-碘-6-甲基嘧啶-4-胺(B4-2,350mg,收率:13%)。Step 2: Add NIS (3.29 g, 14.63 mmol) to a DMF (15 mL) solution of 2-chloro-6-methylpyrimidin-4-amine (B4-1, 1.4 g, 9.75 mmol). The reaction was stirred at 30°C for 16h. The reaction solution was poured into water, filtered, the filter cake was washed with saturated sodium sulfite solution, and washed with water to obtain a pale yellow solid 2-chloro-5-iodo-6-methylpyrimidin-4-amine (B4-2, 350mg, yield: 13% ).
1H NMR(400MHz,DMSO-d 6)δ7.25(s,2H),6.22(s,1H),2.18(s,3H); 1 H NMR(400MHz,DMSO-d 6 )δ7.25(s,2H), 6.22(s,1H), 2.18(s,3H);
LC-MS:m/z 269.9[M+H] +LC-MS: m/z 269.9 [M+H] + .
步骤三:将2-氯-5-碘-6-甲基嘧啶-4-胺(B4-2,350mg,1.30mmol)溶解在氯乙醛(40wt.%in H 2O,5mL)水溶液中。加热至100℃搅拌反应2h。反应液冷却至室温,过滤,滤饼水洗得淡黄色固体8-碘-7-甲基咪唑并[1,2-c]嘧啶-5-醇(B4-3,330mg,收率:67%)。 Step 3: Dissolve 2-chloro-5-iodo-6-methylpyrimidin-4-amine (B4-2, 350 mg, 1.30 mmol) in an aqueous solution of chloroacetaldehyde (40 wt.% in H 2 O, 5 mL). Heat to 100°C and stir for 2h. The reaction liquid was cooled to room temperature, filtered, and the filter cake was washed with water to obtain a pale yellow solid 8-iodo-7-methylimidazo[1,2-c]pyrimidin-5-ol (B4-3, 330mg, yield: 67%) .
1H NMR(400MHz,DMSO-d 6):δ12.48(br,1H),8.05(s,1H),7.59(s,1H),2.46(s,3H)。 1 H NMR (400MHz, DMSO-d 6 ): δ 12.48 (br, 1H), 8.05 (s, 1H), 7.59 (s, 1H), 2.46 (s, 3H).
步骤四:向8-碘-7-甲基咪唑并[1,2-c]嘧啶-5-醇(B4-3,320mg,1.16mmol)溶解在三氯氧磷(10mL)的溶液中加入DIPEA(226mg,1.75mmol)。加热至100℃反应6h。反应液冷却至室温,减压浓缩,加入EtOAc(30mL)。饱和碳酸氢钠水溶液(3×30mL)洗,饱和盐水(3×30mL)洗,无水硫酸钠干燥,过滤,滤液减压浓缩至干,将得到的残留物通过硅胶色谱法(0至10%梯度的甲醇:二氯甲烷)纯化,得白色固体5-氯-8-碘-7-甲基咪唑并[1,2-c]嘧啶(B4,135mg,收率:40%)。Step 4: Add DIPEA to 8-iodo-7-methylimidazo[1,2-c]pyrimidin-5-ol (B4-3, 320mg, 1.16mmol) dissolved in phosphorus oxychloride (10mL) (226mg, 1.75mmol). Heat to 100°C for 6h. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and EtOAc (30 mL) was added. Wash with saturated aqueous sodium bicarbonate (3×30 mL), saturated brine (3×30 mL), dry with anhydrous sodium sulfate, filter, and concentrate the filtrate to dryness under reduced pressure. The residue obtained is passed through silica gel chromatography (0 to 10%). Gradient methanol: dichloromethane) was purified to obtain 5-chloro-8-iodo-7-methylimidazo[1,2-c]pyrimidine (B4, 135 mg, yield: 40%) as a white solid.
1H NMR(400MHz,DMSO-d 6):δ8.16(d,J=1.2Hz,1H),7.74(d,J=1.2Hz,1H),2.66(s,3H)。 1 H NMR (400MHz, DMSO-d 6 ): δ 8.16 (d, J = 1.2 Hz, 1H), 7.74 (d, J = 1.2 Hz, 1H), 2.66 (s, 3H).
中间体B5的合成:7-氯-8-碘-5-(甲基亚砜基)咪唑并[1,2-c]嘧啶Synthesis of Intermediate B5: 7-chloro-8-iodo-5-(methylsulfoxy)imidazo[1,2-c]pyrimidine
Figure PCTCN2020106214-appb-000040
Figure PCTCN2020106214-appb-000040
步骤一:冰水浴下向4-氨基-6-氯-2-甲硫基嘧啶(8.0g,45.55mmol)的二氯甲烷(150mL)溶液中分批加入NIS(12.3g,54.66mmol)。30℃搅拌反应5小时,反应完毕。反应液减压浓缩至干后加入甲醇(50mL),倒入饱和亚硫酸钠水溶液中(150mL),过滤,滤饼水洗得淡黄色固体4-氨基-6-氯-5-碘-2-甲硫基嘧啶(B5-1,12.8g,收率:93%)。Step 1: Add NIS (12.3g, 54.66mmol) to a solution of 4-amino-6-chloro-2-methylthiopyrimidine (8.0g, 45.55mmol) in dichloromethane (150mL) under ice water bath. The reaction was stirred at 30°C for 5 hours, and the reaction was completed. The reaction solution was concentrated under reduced pressure to dryness, then methanol (50mL) was added, poured into saturated sodium sulfite aqueous solution (150mL), filtered, and the filter cake was washed with water to obtain a pale yellow solid 4-amino-6-chloro-5-iodo-2-methylthio Pyrimidine (B5-1, 12.8 g, yield: 93%).
LC-MS:m/z 301.9[M+H] +LC-MS: m/z 301.9[M+H] + ;
1H NMR(400MHz,DMSO-d 6):δ7.98(br,1H),6.92(br,1H),2.41(s,3H)。 1 H NMR (400MHz, DMSO-d 6 ): δ 7.98 (br, 1H), 6.92 (br, 1H), 2.41 (s, 3H).
步骤二:向氯乙醛40wt.%水溶液(20mL)中加入4-氨基-6-氯-5-碘-2-甲硫基嘧啶(2.0g,6.63mmol)。加热到100℃反应3小时。反应液减压浓缩至干,硅胶色谱法(0至10%梯度的甲醇:二氯甲烷)纯化后乙酸乙酯洗得黄色固体7-氯-8-碘-5-(甲硫基)咪唑并[1,2-c]嘧啶(B5-2,1.9g,收率:88%)。Step 2: Add 4-amino-6-chloro-5-iodo-2-methylthiopyrimidine (2.0 g, 6.63 mmol) to a 40 wt.% aqueous solution of chloroacetaldehyde (20 mL). Heat to 100°C for 3 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by silica gel chromatography (0 to 10% gradient methanol: dichloromethane) and washed with ethyl acetate to obtain a yellow solid, 7-chloro-8-iodo-5-(methylthio)imidazo [1,2-c] Pyrimidine (B5-2, 1.9 g, yield: 88%).
LCMS:m/z 325.8[M+H] +LCMS: m/z 325.8[M+H] + ;
1H NMR(400MHz,DMSO-d 6):δ8.06(d,J=1.2Hz,1H),7.73(d,J=1.2Hz,1H),2.76(s,3H)。 1 H NMR (400MHz, DMSO-d 6 ): δ 8.06 (d, J = 1.2 Hz, 1H), 7.73 (d, J = 1.2 Hz, 1H), 2.76 (s, 3H).
步骤三:冰水浴下向7-氯-8-碘-5-(甲硫基)咪唑并[1,2-c]嘧啶(B5-2,1.20g,3.69mmol)的二氯甲烷(20mL)溶液中加入m-CPBA(1.27g,7.37mmol)。反应25℃搅拌16小时。反应冰水淬灭,饱和碳酸氢钠水溶液洗(3×20mL),饱和盐水洗(3×20mL),无水硫酸钠干燥,过滤,滤液减压浓缩至干,硅胶色谱法(0至10%梯度的甲醇:二氯甲烷)纯化后乙酸乙酯洗得黄色固体7-氯-8-碘-5-(甲基亚磺酰基)咪唑并[1,2-c]嘧啶(B5,550mg,收率:44%)。Step 3: Add 7-chloro-8-iodo-5-(methylthio)imidazo[1,2-c]pyrimidine (B5-2, 1.20g, 3.69mmol) in dichloromethane (20mL) under ice water bath M-CPBA (1.27g, 7.37mmol) was added to the solution. The reaction was stirred at 25°C for 16 hours. The reaction was quenched with ice water, washed with saturated sodium bicarbonate aqueous solution (3×20 mL), saturated brine (3×20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness under reduced pressure. Silica gel chromatography (0 to 10%) Gradient methanol: dichloromethane) and eluted with ethyl acetate to give a yellow solid 7-chloro-8-iodo-5-(methylsulfinyl)imidazo[1,2-c]pyrimidine (B5, 550mg, yield Rate: 44%).
LCMS:m/z 341.8[M+H] +LCMS: m/z 341.8[M+H] + ;
1H NMR(400MHz,DMSO-d 6):δ8.64(d,J=1.2Hz,1H),7.85(d,J=1.2Hz,1H),3.12(s,3H)。 1 H NMR (400MHz, DMSO-d 6 ): δ 8.64 (d, J = 1.2 Hz, 1H), 7.85 (d, J = 1.2 Hz, 1H), 3.12 (s, 3H).
中间体C1的合成:2-氨基-3氯嘧啶-4硫钠Synthesis of Intermediate C1: Sodium 2-amino-3chloropyrimidine-4sulfur
Figure PCTCN2020106214-appb-000041
Figure PCTCN2020106214-appb-000041
步骤一:向干燥的100mL的圆底三口烧瓶中依次加入3-氯-4-碘吡啶-2-胺(2.5g,9.82mmol,1.0eq),4,5-双二苯基膦-9,9-二甲基氧杂蒽(341mg,0.59mmol,0.06eq),醋酸鈀(110mg,0.49mmol,0.05eq),DIPEA(3.25mL,19.6mmol,2.0q),3-巯基丙酸甲酯(1.19mL,10.8mmol,1.1eq)和1,4-二氧六环(32.5mL)。在搅拌的情况下,用氮气置换三次,然后加热到100℃,反应3小时。反应完毕后,反应液冷却至室温,用乙酸乙酯(50mL)稀释并减压抽滤,滤饼用乙酸乙酯(25mL)洗涤,将得到的滤液真空浓缩,将得到的残留物通过硅胶色谱法纯化(0至30%梯度的乙酸乙酯:石油醚)得到黄色固体C1-1(2.0g,收率:78%)。Step 1: Add 3-chloro-4-iodopyridin-2-amine (2.5g, 9.82mmol, 1.0eq), 4,5-bisdiphenylphosphine-9, to a dry 100mL round bottom three-necked flask in sequence 9-dimethylxanthene (341mg, 0.59mmol, 0.06eq), palladium acetate (110mg, 0.49mmol, 0.05eq), DIPEA (3.25mL, 19.6mmol, 2.0q), methyl 3-mercaptopropionate ( 1.19mL, 10.8mmol, 1.1eq) and 1,4-dioxane (32.5mL). Under stirring, replace with nitrogen three times, then heat to 100°C and react for 3 hours. After the completion of the reaction, the reaction solution was cooled to room temperature, diluted with ethyl acetate (50mL) and filtered under reduced pressure. The filter cake was washed with ethyl acetate (25mL), the obtained filtrate was concentrated in vacuo, and the residue obtained was chromatographed on silica gel. Purification by method (0 to 30% ethyl acetate: petroleum ether) to obtain a yellow solid C1-1 (2.0 g, yield: 78%).
1H NMR(400MHz,CDCl 3)δ7.89(d,J=5.4Hz,1H),6.53(d,J=5.5Hz,1H),4.87(s,2H),3.74(s,3H),3.24(t,J=7.5Hz,2H),2.75(t,J=7.5Hz,2H)。 1 H NMR(400MHz,CDCl 3 )δ7.89(d,J=5.4Hz,1H), 6.53(d,J=5.5Hz,1H), 4.87(s,2H), 3.74(s,3H), 3.24 (t, J=7.5 Hz, 2H), 2.75 (t, J=7.5 Hz, 2H).
步骤二:在干燥的100mL的圆底三口烧瓶中,将C1-1(2g,8.11mmol,1.0eq)溶解在四氢呋喃(28mL)里,氮气保护的情况下,室温下向反应液中滴加乙醇钠(2.9g,8.51mmol,1.05eq,20%wt),然后搅拌一小时。反应完毕后,用二氯甲烷(60mL)稀释并超声5分钟,减压抽滤,滤饼真空烘干得到黄色固体C1(1.4g,收率:89%)。Step 2: In a dry 100mL round bottom three-necked flask, dissolve C1-1 (2g, 8.11mmol, 1.0eq) in tetrahydrofuran (28mL), under nitrogen protection, add ethanol to the reaction solution dropwise at room temperature Sodium (2.9g, 8.51mmol, 1.05eq, 20%wt), then stirred for one hour. After the completion of the reaction, it was diluted with dichloromethane (60 mL) and sonicated for 5 minutes, filtered under reduced pressure, and the filter cake was dried in vacuum to obtain a yellow solid C1 (1.4 g, yield: 89%).
中间体C2的合成:3-氯-4-((5-氯吡嗪-2-基)硫基)吡啶-2-胺Synthesis of intermediate C2: 3-chloro-4-((5-chloropyrazin-2-yl)thio)pyridin-2-amine
Figure PCTCN2020106214-appb-000042
Figure PCTCN2020106214-appb-000042
步骤一:向干燥的100mL圆底烧瓶中依次加入2,5-二氯吡嗪(3g,20.1mmol,1.0eq),碳酸钾(2.78g,20.1mmol,1.0eq),DMF(25mL)和3-巯基丙酸甲酯(2.54g,21.1mmol,1.05eq),然后在25摄氏度下搅拌18小时。反应完毕后,用乙酸乙酯(100mL)稀释,水(30mL)洗两遍,然后用无水硫酸钠干燥,过滤。将得到的滤液真空浓缩,将得到的残留物通过硅胶色谱法纯化(0至2.8%梯度的乙酸乙酯:石油醚)得到黄色固体C2-1(3.68g,收率:74%)。Step 1: Add 2,5-dichloropyrazine (3g, 20.1mmol, 1.0eq), potassium carbonate (2.78g, 20.1mmol, 1.0eq), DMF (25mL) and 3 to a dry 100mL round-bottom flask. -Methyl mercaptopropionate (2.54g, 21.1mmol, 1.05eq), then stirred at 25 degrees Celsius for 18 hours. After the reaction, it was diluted with ethyl acetate (100 mL), washed twice with water (30 mL), then dried over anhydrous sodium sulfate, and filtered. The obtained filtrate was concentrated in vacuo, and the obtained residue was purified by silica gel chromatography (0 to 2.8% ethyl acetate: petroleum ether gradient) to obtain a yellow solid C2-1 (3.68 g, yield: 74%).
1H NMR(400MHz,CDCl 3)δ8.38(d,J=1.5Hz,1H),8.22(d,J=1.5Hz,1H),3.71(s,3H),3.41(t,J=7.0Hz,2H),2.76(t,J=7.0Hz,2H)。 1 H NMR(400MHz, CDCl 3 )δ8.38(d,J=1.5Hz,1H), 8.22(d,J=1.5Hz,1H), 3.71(s,3H), 3.41(t,J=7.0Hz , 2H), 2.76 (t, J=7.0 Hz, 2H).
步骤二:在干燥的100mL的圆底三口烧瓶中,将C2-1(3.68g,15.8mmol,1.0eq)溶解在四氢呋喃(50mL)里,氮气保护的情况下,室温下向反应液中滴加乙醇钠(5.65g,16.6mmol,1.05eq,20%wt),然后搅拌一小时。反应完毕后,减压浓缩掉一半体积的溶剂,向剩余的反应液中加入乙醚(200mL),沉淀析出,减压抽滤,滤饼真空烘干得到黄色固体C2-2(2.5g,收率:84.2%)。Step 2: In a dry 100mL round bottom three-necked flask, dissolve C2-1 (3.68g, 15.8mmol, 1.0eq) in tetrahydrofuran (50mL), under nitrogen protection, add dropwise to the reaction solution at room temperature Sodium ethoxide (5.65g, 16.6mmol, 1.05eq, 20%wt), then stirred for one hour. After the reaction was completed, half of the solvent was concentrated under reduced pressure, ether (200 mL) was added to the remaining reaction solution, precipitation was precipitated, and the filter cake was vacuum-dried to obtain a yellow solid C2-2 (2.5 g, yield : 84.2%).
步骤三:在氮气保护条件下,依次向干燥的100mL的圆底三口烧瓶中依次加入3-氯-4-碘吡啶-2-胺(2g,7.86mmol,1.0eq)、4,5-双二苯基膦-9,9-二甲基氧杂蒽(363mg,0.63mmol,0.08eq)、Pd 2(dba) 3(287mg,0.31mmol,0.04eq)、DIPEA(2.6mL,15.7mmol,2.0eq)、二氧六环(48mL)和C2-2(1.39g,8.25mmol,1.05eq),然后加热到105摄氏度反应14小时。反应完全后,冷却至室温,用乙酸乙酯稀释,减压抽滤,滤饼再用乙酸乙酯洗涤。滤液减压浓缩得到的残留物通过硅胶色谱法纯化(0至40%梯度的乙酸乙酯/石油醚),得到黄色固体C2(1.5g,收率:66%)。 Step 3: Under nitrogen protection, sequentially add 3-chloro-4-iodopyridin-2-amine (2g, 7.86mmol, 1.0eq) and 4,5-bis-two to a dry 100mL round bottom three-necked flask. Phenylphosphine-9,9-dimethylxanthene (363mg, 0.63mmol, 0.08eq), Pd 2 (dba) 3 (287mg, 0.31mmol, 0.04eq), DIPEA (2.6mL, 15.7mmol, 2.0eq) ), dioxane (48mL) and C2-2 (1.39g, 8.25mmol, 1.05eq), and then heated to 105 degrees Celsius for 14 hours. After the reaction was completed, it was cooled to room temperature, diluted with ethyl acetate, filtered under reduced pressure, and the filter cake was washed with ethyl acetate. The residue obtained by concentrating the filtrate under reduced pressure was purified by silica gel chromatography (ethyl acetate/petroleum ether with a gradient of 0 to 40%) to obtain a yellow solid C2 (1.5 g, yield: 66%).
1H NMR(400MHz,CDCl 3)δ8.45(d,J=1.4Hz,1H),8.29(d,J=1.4Hz,1H),7.83(d,J=5.3Hz,1H),6.51(d,J=5.3Hz,1H),4.95(s,2H)。 1 H NMR(400MHz,CDCl 3 )δ8.45(d,J=1.4Hz,1H), 8.29(d,J=1.4Hz,1H), 7.83(d,J=5.3Hz,1H), 6.51(d , J=5.3Hz, 1H), 4.95(s, 2H).
实施例一:化合物1的合成Example 1: Synthesis of compound 1
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(甲磺酰基)-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(methylsulfonyl)-2,8-diazepine Spiro[4.5]decane-4-amine
Figure PCTCN2020106214-appb-000043
Figure PCTCN2020106214-appb-000043
步骤一:向25mL的圆底烧瓶中依次加入A(60mg,0.167mmol)、二氯甲烷(5mL)、三乙胺(51mg,0.501mmol)和甲磺酸酐(58mg,0.334mmol),反应液在20℃下搅拌1小时。反应液减压浓缩后加水(10mL)稀释,用乙酸乙酯(10mL)萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=1:0到10:1)得到无色油状物1-1(40mg,收率:54.8%)。Step 1: Add A (60mg, 0.167mmol), dichloromethane (5mL), triethylamine (51mg, 0.501mmol) and methanesulfonic anhydride (58mg, 0.334mmol) to a 25mL round bottom flask in sequence. Stir at 20°C for 1 hour. The reaction solution was concentrated under reduced pressure, diluted with water (10 mL), and extracted 3 times with ethyl acetate (10 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=1:0 to 10:1) to obtain colorless oil 1-1 (40 mg, yield: 54.8%).
LCMS:m/z 438.1[M+H] +LCMS: m/z 438.1 [M+H] + .
步骤二:向25mL的圆底烧瓶中依次加入化合物1-1(40mg,0.091mmol)、甲醇(1mL)和盐酸二氧六环(1mL,4.0M),反应液在20℃下搅拌0.5小时。反应完毕后,将反应液减压浓缩得到黄色固体1-2(28mg,盐酸盐,收率:100%)。Step 2: Add compound 1-1 (40mg, 0.091mmol), methanol (1mL) and dioxane hydrochloride (1mL, 4.0M) to a 25mL round bottom flask in sequence, and the reaction solution was stirred at 20°C for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain yellow solid 1-2 (28 mg, hydrochloride, yield: 100%).
LCMS:m/z 234.1[M+H] +LCMS: m/z 234.1 [M+H] + .
步骤三:向干燥的25mL单口烧瓶中依次加入化合物1-2(28mg,0.091mmol),乙腈(3.0mL),N,N-二异丙基乙胺(1.0mL)和3-氯-4-((5-氯吡嗪-2-基)硫基)吡啶-2-胺(32mg,0.118mmol)。反应液在90℃下搅拌7小时。反应完毕后,反应液减压浓缩。得到的残留物通过薄层层析硅胶板纯化(二氯甲烷/甲醇=10:1)得到30mg不纯的产物。然后用HPLC制备得到白色固体1(17mg,甲酸盐,收率:27.9%)。Step 3: Add compound 1-2 (28mg, 0.091mmol), acetonitrile (3.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-neck flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (32 mg, 0.118 mmol). The reaction solution was stirred at 90°C for 7 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure. The obtained residue was purified by thin-layer chromatography on a silica gel plate (dichloromethane/methanol=10:1) to obtain 30 mg of impure product. Then, a white solid 1 (17 mg, formate, yield: 27.9%) was prepared by HPLC.
1H NMR(400MHz,DMSO-d6)δ8.48(d,J=1.6Hz,1H),8.29(d,J=1.2Hz,1H),8.19(s,1H),7.65(d,J=5.6Hz,1H),6.35(s,2H),5.81(d,J=5.6Hz,1H),4.20–4.06(m,2H),3.55–3.47(m,1H),3.45–3.38(m,1H),3.37–3.25(m,2H),3.20–3.13(m,2H),3.05–2.99(m,1H),2.93(s,3H),1.72–1.60(m,2H),1.53–1.43(m,2H);1H NMR(400MHz,DMSO-d6)δ8.48(d,J=1.6Hz,1H), 8.29(d,J=1.2Hz,1H), 8.19(s,1H), 7.65(d,J=5.6Hz ,1H),6.35(s,2H),5.81(d,J=5.6Hz,1H),4.20–4.06(m,2H),3.55–3.47(m,1H),3.45–3.38(m,1H), 3.37–3.25(m,2H), 3.20–3.13(m,2H), 3.05–2.99(m,1H), 2.93(s,3H), 1.72–1.60(m,2H), 1.53–1.43(m,2H) );
LC-MS:m/z 470.0[M+H]+。LC-MS: m/z 470.0[M+H]+.
实施例二:化合物2的合成Example 2: Synthesis of Compound 2
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-(吡嗪-2-基)-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(pyrazin-2-yl)-2,8- Diazaspiro[4.5]decane-4-amine
Figure PCTCN2020106214-appb-000044
Figure PCTCN2020106214-appb-000044
步骤一:在-50℃下向25mL的圆底烧瓶中依次加入化合物A-8(200mg,0.407mmol)、四氢呋喃/水(98:2,5mL)和硼氢化钠(46mg,1.22mmol),反应液在搅拌下3小时升温至20℃。TLC点板反应完全。反应液加水(20mL)稀释,用乙酸乙酯(20mL)萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩得到无色油状物A-9(200mg,收率:99.5%)。Step 1: Add compound A-8 (200mg, 0.407mmol), tetrahydrofuran/water (98:2,5mL) and sodium borohydride (46mg, 1.22mmol) to a 25mL round-bottom flask at -50°C. The solution was heated to 20°C under stirring for 3 hours. The TLC dot plate reaction is complete. The reaction solution was diluted with water (20 mL), and extracted with ethyl acetate (20 mL) three times. The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain A-9 (200 mg, yield: 99.5%) as a colorless oil.
步骤二:在干燥的25mL单口瓶中依次加入化合物A-9(200mg,0.405mmol)、乙醇(4mL)和10%的氢氧化钠水溶液(2mL)。反应液在85℃下搅拌8小时。冷却至室温后,反应液用0.5N稀盐酸调至pH=7,得到的混合物减压浓缩去除挥发物。向得到的残留物中加入DCM/MeOH(2:1)的混合溶剂(10mL),过滤,将得到的滤液减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=1:0到10:1)得到无色油状物A(110mg,收率:75.3%)。Step 2: Add compound A-9 (200mg, 0.405mmol), ethanol (4mL) and 10% sodium hydroxide aqueous solution (2mL) in a dry 25mL single-necked flask in sequence. The reaction solution was stirred at 85°C for 8 hours. After cooling to room temperature, the reaction solution was adjusted to pH=7 with 0.5N dilute hydrochloric acid, and the resulting mixture was concentrated under reduced pressure to remove volatiles. A mixed solvent (10 mL) of DCM/MeOH (2:1) was added to the obtained residue, filtered, and the obtained filtrate was concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=1:0 to 10:1) to obtain a colorless oil A (110 mg, yield: 75.3%).
LC-MS:m/z 460.2[M+H] +LC-MS: m/z 460.2 [M+H] + .
步骤三:向25mL的圆底烧瓶中依次加入A(110mg,0.306mmol)、乙腈(3mL)、2-氯吡嗪(140mg,1.22mmol)和N,N-二异丙基乙胺(1mL),反应液在90℃下搅拌10小时。反应液减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=1:0到10:1)得到黄色油状物2-1(50mg,收率:37.3%)。Step 3: Add A (110mg, 0.306mmol), acetonitrile (3mL), 2-chloropyrazine (140mg, 1.22mmol) and N,N-diisopropylethylamine (1mL) to a 25mL round bottom flask in sequence The reaction solution was stirred at 90°C for 10 hours. The reaction solution was concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=1:0 to 10:1) to obtain a yellow oil 2-1 (50 mg, yield: 37.3%).
1H NMR(400MHz,CDCl 3)δ8.06–8.02(m,1H),7.88(d,J=1.6Hz,1H),7.84(d,J=2.4Hz,1H),4.20-3.93(m,2H),3.90–3.88(m,1H),3.80–3.60(m,2H),3.55–3.30(m,3H),3.04–2.84(m,2H),1.90–1.75(m,1H),1.72–1.58(m,1H),1.55–1.40(m,11H),1.21(s,9H)。 1 H NMR (400MHz, CDCl 3 ) δ 8.06–8.02 (m, 1H), 7.88 (d, J = 1.6 Hz, 1H), 7.84 (d, J = 2.4 Hz, 1H), 4.20-3.93 (m, 2H), 3.90--3.88 (m, 1H), 3.80 - 3.60 (m, 2H), 3.55 - 3.30 (m, 3H), 3.04 - 2.84 (m, 2H), 1.90 - 1.75 (m, 1H), 1.72-- 1.58 (m, 1H), 1.55-1.40 (m, 11H), 1.21 (s, 9H).
步骤四:向25mL的圆底烧瓶中依次加入化合物2-1(50mg,0.114mmol)、甲醇(1mL)和盐酸二氧六环(1mL,4.0M),反应液在20℃下搅拌0.5小时。反应完毕后,反应液减压浓缩得到黄色固体2-2(35mg,盐酸盐,收率:100%)。Step 4: Add compound 2-1 (50 mg, 0.114 mmol), methanol (1 mL), and dioxane hydrochloride (1 mL, 4.0 M) to a 25 mL round bottom flask in sequence, and the reaction solution was stirred at 20° C. for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain a yellow solid 2-2 (35 mg, hydrochloride, yield: 100%).
步骤五:向干燥的25mL单口烧瓶中依次加入化合物2-2(35mg,0.114mmol),乙腈(3.0mL),N,N-二异丙基乙胺(1.0mL)和3-氯-4-((5-氯吡嗪-2-基)硫基)吡啶-2-胺(40mg,0.148mmol)。反应液在90℃下搅拌10小时。TLC点板显示有新点生成。反应液减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=10:1)得到40mg不纯的产物。然后用HPLC制备得到白色固体2(22mg,甲酸盐,收率:40.7%)。Step 5: Add compound 2-2 (35mg, 0.114mmol), acetonitrile (3.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-neck flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (40 mg, 0.148 mmol). The reaction solution was stirred at 90°C for 10 hours. The TLC dot board shows that new dots are generated. The reaction solution was concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=10:1) to obtain 40 mg of impure product. Then, a white solid 2 (22 mg, formate, yield: 40.7%) was obtained by HPLC preparation.
1H NMR(400MHz,DMSO-d6)δ8.49(d,J=1.2Hz,1H),8.30(d,J=1.6Hz,1H),8.24(s,1H),8.03(dd,J=2.8,1.6Hz,1H),7.98(d,J=1.2Hz,1H),7.78(d,J=2.4Hz,1H),7.66(d,J=5.6Hz,1H),6.35(s,2H),5.82(d,J=5.6Hz,1H),4.28–4.17(m,2H),3.80–3.72(m,1H),3.71–3.64(m,1H),3.50–3.43(m,1H),3.42–3.24(m,4H),1.80–1.65(m,2H),1.58–1.46(m,2H); 1 H NMR(400MHz,DMSO-d6)δ8.49(d,J=1.2Hz,1H), 8.30(d,J=1.6Hz,1H), 8.24(s,1H), 8.03(dd,J=2.8 ,1.6Hz,1H),7.98(d,J=1.2Hz,1H),7.78(d,J=2.4Hz,1H),7.66(d,J=5.6Hz,1H),6.35(s,2H), 5.82(d,J=5.6Hz,1H), 4.28–4.17(m,2H), 3.80–3.72(m,1H), 3.71–3.64(m,1H), 3.50–3.43(m,1H),3.42– 3.24(m,4H), 1.80–1.65(m,2H), 1.58–1.46(m,2H);
LC-MS:m/z 470.0[M+H] +LC-MS: m/z 470.0 [M+H] + .
实施例三:化合物3的合成。Example 3: Synthesis of Compound 3.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-苯基-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-phenyl-2,8-diazaspiro[4.5 ]Decane-4-amine
Figure PCTCN2020106214-appb-000045
Figure PCTCN2020106214-appb-000045
步骤一:向25mL的圆底烧瓶中依次加入化合物A(120mg,0.334mmol)、碘苯(136mg,0.668mmol)、二氧六环(12mL)、Pd 2(dba) 3(30mg,0.033mmol)、4,5-双二苯基膦-9,9-二甲基氧杂蒽(39mg,0.067mmol)、和碳酸铯(326mg,1.00mmol),反应液在氮气保护100℃下搅拌5小时。反应液加水(30mL)稀释,用乙酸乙酯(30mL)萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(石油醚/乙酸乙酯=1:1)得到黄色固体3-1(90mg,收率:62.1%)。 Step 1: Add compound A (120mg, 0.334mmol), iodobenzene (136mg, 0.668mmol), dioxane (12mL), Pd 2 (dba) 3 (30mg, 0.033mmol) to a 25mL round bottom flask in sequence , 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (39mg, 0.067mmol), and cesium carbonate (326mg, 1.00mmol), the reaction solution was stirred at 100°C under nitrogen protection for 5 hours. The reaction solution was diluted with water (30 mL), and extracted twice with ethyl acetate (30 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:1) to obtain a yellow solid 3-1 (90 mg, yield: 62.1%).
LC-MS:m/z 436.1[M+H] +LC-MS: m/z 436.1 [M+H] + .
步骤二:向25mL的圆底烧瓶中依次加入化合物3-1(45mg,0.103mmol)、甲醇(2mL)和盐酸二氧六环(1mL,4.0M),反应液在20℃下搅拌0.5小时。反应完毕后,反应液减压浓缩得到黄色固体3-2(31mg,盐酸盐,收率:100%)。Step 2: Add compound 3-1 (45mg, 0.103mmol), methanol (2mL) and dioxane hydrochloride (1mL, 4.0M) to a 25mL round bottom flask in sequence, and the reaction solution was stirred at 20°C for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain a yellow solid 3-2 (31 mg, hydrochloride, yield: 100%).
LC-MS:m/z 232.1[M+H] +LC-MS: m/z 232.1 [M+H] + .
步骤三:向干燥的25mL单口烧瓶中依次加入化合物3-2(31mg,0.103mmol),乙腈(3.0mL),N,N-二异丙基乙胺(1.0mL)和3-氯-4-((5-氯吡嗪-2-基)硫基)吡啶-2-胺(C2,37mg,0.134mmol)。反应液在90℃下搅拌7小时。反应液减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=5:1)得到30mg不纯的产物。然后用HPLC制备得到白色固体3(23mg,甲酸盐,收率:43.4%)。Step 3: Add compound 3-2 (31mg, 0.103mmol), acetonitrile (3.0mL), N,N-diisopropylethylamine (1.0mL) and 3-chloro-4-into a dry 25mL single-neck flask in sequence ((5-Chloropyrazine-2-yl)thio)pyridin-2-amine (C2, 37 mg, 0.134 mmol). The reaction solution was stirred at 90°C for 7 hours. The reaction solution was concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=5:1) to obtain 30 mg of impure product. Then, a white solid 3 (23 mg, formate, yield: 43.4%) was prepared by HPLC.
1H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.30(d,J=0.8Hz,1H),8.23(s,1H),7.66(d,J=5.2Hz,1H),7.16(t,J=8.0Hz,2H),6.56–6.43(m,3H),6.36(s,2H),5.81(d,J=5.2Hz,1H),4.30–4.16(m,2H),3.59–3.52(m,1H),3.51–3.46(m,1H),3.35–3.24(m,3H),3.23–3.17(m,1H),3.11–3.03(m,1H),1.79–1.65(m,2H),1.54–1.44(m,2H); 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H), 8.30(d,J=0.8Hz,1H), 8.23(s,1H), 7.66(d,J=5.2Hz,1H), 7.16(t,J=8.0Hz,2H),6.56–6.43(m,3H),6.36(s,2H),5.81(d,J=5.2Hz,1H), 4.30–4.16(m,2H),3.59 --3.52(m,1H),3.51–3.46(m,1H), 3.35–3.24(m,3H), 3.23–3.17(m,1H), 3.11–3.03(m,1H), 1.79–1.65(m, 2H),1.54–1.44(m,2H);
LC-MS:m/z 468.0[M+H] +LC-MS: m/z 468.0 [M+H] + .
实施例四:化合物4的合成。Example Four: Synthesis of Compound 4.
(S)-1-(4-氨基-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2,8-二氮杂螺[4.5]癸烷-2-基)丙-2-烯-1-酮(S)-1-(4-Amino-8-(5-((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2,8-diaza spiro [4.5]Decan-2-yl)prop-2-en-1-one
Figure PCTCN2020106214-appb-000046
Figure PCTCN2020106214-appb-000046
步骤一:在-30℃下向圆底烧瓶中依次加入化合物B(90mg,0.181mmol)、二氯甲烷(2mL)、丙烯酸酐(23mg,0.181mmol),反应液在-30℃下搅拌0.5小时。反应完毕后,反应液在0℃下减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=10:1)得到黄色固体4-1(43mg,收率:43.0%)。Step 1: Add compound B (90mg, 0.181mmol), dichloromethane (2mL), and acrylic anhydride (23mg, 0.181mmol) to a round bottom flask at -30℃, and stir the reaction solution at -30℃ for 0.5 hours . After the completion of the reaction, the reaction solution was concentrated under reduced pressure at 0°C. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=10:1) to obtain a yellow solid 4-1 (43 mg, yield: 43.0%).
LC-MS:m/z 550.1[M+H] +LC-MS: m/z 550.1 [M+H] + .
步骤二:向圆底烧瓶中依次加入化合物4-1(43mg,0.078mmol)、甲醇(1mL)和盐酸二氧六环(0.2mL,4.0M),反应液在20℃下搅拌0.5小时。反应完毕后,反应液减压浓缩。得到的残留物通过HPLC制备纯化得到白色固体4(21mg,甲酸盐,收率:55.3%)。Step 2: Add compound 4-1 (43mg, 0.078mmol), methanol (1mL) and dioxane hydrochloride (0.2mL, 4.0M) to a round bottom flask in sequence, and the reaction solution was stirred at 20°C for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure. The obtained residue was prepared and purified by HPLC to obtain 4 as a white solid (21 mg, formate, yield: 55.3%).
1H NMR(400MHz,DMSO-d6)δ8.48(d,J=8.4Hz,1H),8.35–8.25(m,1H),8.23– 8.16(m,1H),6.71–6.49(m,1H),6.34(d,J=8.8Hz,2H),6.20–6.10(m,1H),5.86–5.79(m,1H),5.73–5.64(m,1H),4.30–4.05(m,2H),3.90–3.50(m,4H),3.40–3.10(m,4H),1.76–1.58(m,2H),1.52–1.36(m,2H); 1 H NMR(400MHz,DMSO-d6)δ8.48(d,J=8.4Hz,1H), 8.35–8.25(m,1H), 8.23– 8.16(m,1H), 6.71–6.49(m,1H) ,6.34(d,J=8.8Hz,2H), 6.20–6.10(m,1H), 5.86–5.79(m,1H), 5.73–5.64(m,1H), 4.30–4.05(m,2H), 3.90 –3.50(m,4H), 3.40–3.10(m,4H), 1.76–1.58(m,2H), 1.52–1.36(m,2H);
LC-MS:m/z 446.1[M+H] +LC-MS: m/z 446.1 [M+H] + .
实施例五:化合物5的合成。Example 5: Synthesis of Compound 5.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-(氮杂环丁烷-3-基)-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(azetidin-3-yl)-2 ,8-diazaspiro[4.5]decane-4-amine
Figure PCTCN2020106214-appb-000047
Figure PCTCN2020106214-appb-000047
步骤一:向干燥的10mL单口烧瓶中依次加入化合物B(120mg,0.242mmol)、1-叔丁氧基羰基(Boc)-3-氮杂环丁酮(41mg,0.242mmol)、二氯甲烷(10mL)和三乙酰氧基硼氢化钠(77mg,0.363mmol),然后在25℃下搅拌反应2小时。反应完毕后,加入饱和碳酸氢钠溶液(10mL),用二氯甲烷(15mL)萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(二氯甲烷/甲醇=1:0-10:1)得到黄色固体5-1(90mg,收率:57.0%)。Step 1: Add compound B (120mg, 0.242mmol), 1-tert-butoxycarbonyl (Boc)-3-azetidinone (41mg, 0.242mmol), dichloromethane ( 10mL) and sodium triacetoxyborohydride (77mg, 0.363mmol), and then stirred at 25°C for 2 hours. After the reaction was completed, saturated sodium bicarbonate solution (10 mL) was added, and extraction was performed twice with dichloromethane (15 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (dichloromethane/methanol=1:0-10:1) to obtain a yellow solid 5-1 (90 mg, yield: 57.0%).
LCMS:m/z 651.1[M+H] +LCMS: m/z 651.1 [M+H] + .
步骤二:向圆底烧瓶中依次加入化合物5-1(20mg,0.031mmol)、甲醇(1mL)和盐酸二氧六环(0.5mL,4.0M),反应液在20℃下搅拌0.5小时。反应完毕后,反应液减压浓缩。得到的残留物通过HPLC制备纯化得到白色固体5(9mg,甲酸盐,收率:60.0%)。Step 2: Add compound 5-1 (20 mg, 0.031 mmol), methanol (1 mL), and dioxane hydrochloride (0.5 mL, 4.0 M) to a round bottom flask in sequence, and the reaction solution was stirred at 20° C. for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure. The obtained residue was prepared and purified by HPLC to obtain 5 as a white solid (9 mg, formate, yield: 60.0%).
1H NMR(400MHz,DMSO-d6)δ8.51(d,J=1.2Hz,1H),8.31(s,1H),8.19(s,1H),7.66(d,J=5.2Hz,1H),6.37(s,2H),5.80(d,J=5.6Hz,1H),4.40–4.20(m,2H),4.00–3.80(m,4H),3.65–3.55(m,1H),3.25–3.00(m,6H),2.83–2.75(m,1H),2.69–2.61(m,1H),1.75–1.53(m,4H); 1 H NMR(400MHz,DMSO-d6)δ8.51(d,J=1.2Hz,1H), 8.31(s,1H), 8.19(s,1H), 7.66(d,J=5.2Hz,1H), 6.37(s,2H),5.80(d,J=5.6Hz,1H), 4.40–4.20(m,2H), 4.00–3.80(m,4H), 3.65–3.55(m,1H), 3.25–3.00( m,6H), 2.83–2.75(m,1H), 2.69–2.61(m,1H), 1.75–1.53(m,4H);
LCMS:m/z 447.2[M+H] +LCMS: m/z 447.2 [M+H] + .
实施例六:化合物6的合成。Example 6: Synthesis of Compound 6.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-(1-甲基杂氮环丁烷-3-基)-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(1-methylazacyclobutane-3- Yl)-2,8-diazaspiro[4.5]decane-4-amine
Figure PCTCN2020106214-appb-000048
Figure PCTCN2020106214-appb-000048
步骤一:在0℃下向干燥的10mL单口烧瓶中依次加入化合物5-1(60mg,0.092mmol)、二氯甲烷(3mL)和TMSOTf(41mg,0.184mmol),然后在20℃下搅拌反应2小时。LCMS检测反应完全。向反应液中加入甲醇(0.5mL)和饱和碳酸氢钠溶液(0.5mL)淬灭。得到的混合物用水(15mL)稀释,用二氯甲烷(15mL)萃取3次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩,得到黄色固体6-1(50mg,粗品)。Step 1: Add compound 5-1 (60mg, 0.092mmol), dichloromethane (3mL) and TMSOTf (41mg, 0.184mmol) to a dry 10mL single-necked flask at 0℃, and then stir at 20℃ for reaction 2 hour. LCMS detected that the reaction was complete. The reaction solution was quenched by adding methanol (0.5 mL) and saturated sodium bicarbonate solution (0.5 mL). The resulting mixture was diluted with water (15 mL) and extracted 3 times with dichloromethane (15 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a yellow solid 6-1 (50 mg, crude product).
LCMS:m/z 551.2[M+H] +LCMS: m/z 551.2 [M+H] + .
步骤二:在0℃下,向圆底烧瓶中依次加入化合物6-1(40mg,0.073mmol)、二氯甲烷(6mL)、甲醛(6mg,0.073mmol,37%水溶液)的水(0.1mL)溶液和三乙酰氧基硼氢化钠(31mg,0.146mmol),反应液在0℃下搅拌0.5小时。反应完毕后,加入饱和碳酸氢钠溶液(5mL),用二氯甲烷(6mL)萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩得到黄色固体6-2(18mg,粗品)。Step 2: Add compound 6-1 (40mg, 0.073mmol), dichloromethane (6mL), formaldehyde (6mg, 0.073mmol, 37% aqueous solution) and water (0.1mL) to a round bottom flask at 0°C in sequence The solution and sodium triacetoxyborohydride (31mg, 0.146mmol), the reaction solution was stirred at 0°C for 0.5 hours. After the reaction was completed, saturated sodium bicarbonate solution (5 mL) was added, and extraction was performed twice with dichloromethane (6 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a yellow solid 6-2 (18 mg, crude product).
LCMS:m/z 565.1[M+H] +LCMS: m/z 565.1 [M+H] + .
步骤三:向圆底烧瓶中依次加入化合物6-2(18mg,0.032mmol)、甲醇(1mL)和盐酸二氧六环(0.5mL,4.0M),反应液在20℃下搅拌0.5小时。反应完毕后,反应液减压浓缩。得到的残留物通过HPLC制备纯化得到灰白色固体6(1mg,甲酸盐,收率:6.3%)。Step 3: Add compound 6-2 (18mg, 0.032mmol), methanol (1mL) and dioxane hydrochloride (0.5mL, 4.0M) to the round bottom flask in sequence, and the reaction solution was stirred at 20°C for 0.5 hours. After the completion of the reaction, the reaction solution was concentrated under reduced pressure. The obtained residue was purified by HPLC preparation to obtain an off-white solid 6 (1 mg, formate, yield: 6.3%).
1H NMR(400MHz,DMSO-d6)δ8.47(d,J=1.2Hz,1H),8.32–8.26(m,2H),7.66(d,J=5.6Hz,1H),6.35(s,2H),5.81(d,J=5.6Hz,1H),4.34–4.12(m,2H),3.41–3.34(m,2H),3.22–3.06(m,4H),3.05–2.85(m,4H),2.70–2.60(m,1H),2.30–2.20(m,3H),1.70–1.45(m,4H); 1 H NMR(400MHz,DMSO-d6)δ8.47(d,J=1.2Hz,1H), 8.32–8.26(m,2H), 7.66(d,J=5.6Hz,1H), 6.35(s,2H ), 5.81(d,J=5.6Hz,1H), 4.34–4.12(m,2H),3.41–3.34(m,2H),3.22–3.06(m,4H),3.05–2.85(m,4H), 2.70–2.60(m,1H), 2.30–2.20(m,3H), 1.70–1.45(m,4H);
LCMS:m/z 461.1[M+H] +LCMS: m/z 461.1 [M+H] + .
实施例七:化合物7的合成。Example 7: Synthesis of Compound 7.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-环丁基-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-cyclobutyl-2,8-diazaspiro[ 4.5] Decane-4-amine
Figure PCTCN2020106214-appb-000049
Figure PCTCN2020106214-appb-000049
参照化合物5的合成方法,使用环丁酮代替1-叔丁氧基羰基-3-氮杂环丁酮,进行两步反应得到化合物7。Referring to the synthesis method of compound 5, cyclobutanone was used instead of 1-tert-butoxycarbonyl-3-azetidinone, and compound 7 was obtained by performing a two-step reaction.
1H NMR(400MHz,DMSO-d6)δ8.53(d,J=1.2Hz,1H),8.32(s,1H),8.13(s,1H),7.66(d,J=5.6Hz,1H),6.37(s,2H),5.81(d,J=5.2Hz,1H),4.50–3.50(m,6H),3.25–3.00(m,3H),2.45–2.05(m,4H),2.00–1.50(m,6H),1.18(t,J=7.2Hz,1H); 1 H NMR(400MHz,DMSO-d6)δ8.53(d,J=1.2Hz,1H),8.32(s,1H),8.13(s,1H),7.66(d,J=5.6Hz,1H), 6.37(s,2H),5.81(d,J=5.2Hz,1H),4.50–3.50(m,6H), 3.25–3.00(m,3H), 2.45–2.05(m,4H),2.00–1.50( m, 6H), 1.18 (t, J = 7.2 Hz, 1H);
LCMS:m/z 446.2[M+H] +LCMS: m/z 446.2 [M+H] + .
实施例八:化合物8的合成。Example 8: Synthesis of Compound 8.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-(氧杂环丁烷-3-基)-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(oxetan-3-yl)-2 ,8-diazaspiro[4.5]decane-4-amine
Figure PCTCN2020106214-appb-000050
Figure PCTCN2020106214-appb-000050
参照化合物5的合成方法,使用3-氧杂环丁酮代替1-叔丁氧基羰基-3-氮杂环丁酮,进行两步反应得到化合物8。Referring to the synthesis method of compound 5, 3-oxetanone was used instead of 1-tert-butoxycarbonyl-3-azetidinone, and compound 8 was obtained by performing a two-step reaction.
1H NMR(400MHz,DMSO-d6)δ8.49(d,J=0.8Hz,1H),8.28-8.15(m,2H),7.65(d,J=5.2Hz,1H),6.36(s,2H),5.80(d,J=5.2Hz,1H),4.62–4.53(m,2H),4.51–4.42(m,2H),4.36–4.16(m,2H),3.72–3.62(m,2H),3.21–3.03(m,3H),3.01–2.89(m,1H),2.71–2.63(m,1H),2.60–2.54(m,1H),2.41–2.34(m,1H),1.72–1.50(m,4H); 1 H NMR(400MHz,DMSO-d6)δ8.49(d,J=0.8Hz,1H), 8.28-8.15(m,2H), 7.65(d,J=5.2Hz,1H), 6.36(s,2H ), 5.80(d,J=5.2Hz,1H), 4.62–4.53(m,2H), 4.51–4.42(m,2H), 4.36–4.16(m,2H), 3.72–3.62(m,2H), 3.21--3.03(m,3H), 3.01--2.89(m,1H), 2.71--2.63(m,1H), 2.60--2.54(m,1H), 2.41--2.34(m,1H), 1.72--1.50(m ,4H);
LCMS:m/z 448.2[M+H] +LCMS: m/z 448.2 [M+H] + .
实施例九:化合物9的合成。Example 9: Synthesis of Compound 9.
(S)-8-(8-((2-氨基-3-氯吡啶-4-基)硫代)咪唑[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸烷-4-胺(S)-8-(8-((2-Amino-3-chloropyridin-4-yl)thio)imidazole[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8 -Diazaspiro[4.5]decane-4-amine
Figure PCTCN2020106214-appb-000051
Figure PCTCN2020106214-appb-000051
步骤一:向干燥的10mL单口烧瓶中依次加入化合物3-2(35mg,0.115mmol)、乙腈(3mL)、N,N-二异丙基乙胺(0.5mL)和5-氯-8-碘咪唑[1,2-c]嘧啶(B3,32mg,0.115mmol),然后在90℃下搅拌反应7小时。待反应液冷却到20℃后,向反应液中加入Boc 2O(118mg,0.542mmol),然后在50℃下搅拌反应1小时。将反应液减压浓缩,得到的残留物通过硅胶色谱法纯化(石油醚/乙酸乙酯=1:1)得到黄色固体9-1(40mg,收率:60.6%)。CMS:m/z 575.1[M+H] +Step 1: Add compound 3-2 (35mg, 0.115mmol), acetonitrile (3mL), N,N-diisopropylethylamine (0.5mL) and 5-chloro-8-iodine to a dry 10mL single-necked flask. Imidazole [1,2-c] pyrimidine (B3, 32 mg, 0.115 mmol) was then stirred at 90°C for 7 hours. After the reaction solution was cooled to 20°C, Boc 2 O (118 mg, 0.542 mmol) was added to the reaction solution, and then the reaction was stirred at 50°C for 1 hour. The reaction solution was concentrated under reduced pressure, and the obtained residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:1) to obtain a yellow solid 9-1 (40 mg, yield: 60.6%). CMS: m/z 575.1 [M+H] + .
步骤二:向圆底烧瓶中依次加入2-氨基-3-氯吡啶-4-硫化钠(14mg,0.077mmol)、化合物9-1(40mg,0.070mmol)、二氧六环(1mL)、Pd 2(dba) 3(7mg,0.008mmol)、4,5-双二苯基膦-9,9-二甲基氧杂蒽(9mg,0.016mmol)和N,N-二异丙基乙胺(40mg,0.308mmol),反应液抽换氮气三次,然后在100℃下搅拌6小时。反应完毕后,反应液用水(5mL)稀释,用乙酸乙酯(5mL)萃取2次。合并的有机相用饱和食盐水洗涤、无水硫酸钠干燥、过滤、减压浓缩。得到的残留物通过硅胶色谱法纯化(乙酸乙酯)得到黄色固体9-2(15mg,收率:31.9%)。LCMS:m/z 607.2[M+H] +Step 2: Add 2-amino-3-chloropyridine-4-sodium sulfide (14mg, 0.077mmol), compound 9-1 (40mg, 0.070mmol), dioxane (1mL), and Pd to the round bottom flask in sequence 2 (dba) 3 (7mg, 0.008mmol), 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (9mg, 0.016mmol) and N,N-diisopropylethylamine ( 40mg, 0.308mmol), the reaction solution was pumped with nitrogen three times, and then stirred at 100°C for 6 hours. After the reaction was completed, the reaction solution was diluted with water (5 mL) and extracted twice with ethyl acetate (5 mL). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel chromatography (ethyl acetate) to obtain a yellow solid 9-2 (15 mg, yield: 31.9%). LCMS: m/z 607.2 [M+H] + .
步骤三:向干燥的单口烧瓶中依次加入化合物9-2(15mg,0.025mmol),二氯甲烷(2mL)和三氟乙酸(0.4mL)。反应液在20℃下搅拌1小时。反应液减压浓缩。得到的残留物反应液用饱和碳酸氢钠溶液调至pH=8,用DCM/MeOH(10:1)的混合溶剂(5mL)萃取3次,合并的有机相减压浓缩得到灰白色固体9(7.3mg,收率:57.5%)。Step 3: Add compound 9-2 (15 mg, 0.025 mmol), dichloromethane (2 mL) and trifluoroacetic acid (0.4 mL) to a dry single-neck flask in sequence. The reaction solution was stirred at 20°C for 1 hour. The reaction solution was concentrated under reduced pressure. The resulting residue reaction solution was adjusted to pH=8 with saturated sodium bicarbonate solution, and extracted with a mixed solvent (5 mL) of DCM/MeOH (10:1) for three times. The combined organic phase was concentrated under reduced pressure to obtain an off-white solid 9 (7.3 mg, yield: 57.5%).
1H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.84(s,1H),7.60–7.52(m,2H),7.19–7.12(m,2H),6.60–6.50(m,3H),6.33(brs,2H),5.79(d,J=5.6Hz,1H),3.97–3.85(m,2H),3.56–3.45(m,2H),3.43–3.35(m,2H),3.27–3.21(m,1H),3.20–3.13(m,1H),3.03–2.96(m,1H),2.00–1.87(m,2H),1.60–1.50(m,2H);LCMS:m/z 507.1[M+H] + 1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.84(s,1H),7.60-7.52(m,2H),7.19-7.12(m,2H),6.60-6.50(m, 3H),6.33(brs,2H),5.79(d,J=5.6Hz,1H),3.97–3.85(m,2H),3.56–3.45(m,2H),3.43–3.35(m,2H), 3.27 –3.21(m,1H), 3.20–3.13(m,1H), 3.03–2.96(m,1H), 2.00–1.87(m,2H), 1.60–1.50(m,2H); LCMS: m/z 507.1 [M+H] + .
实施例十:化合物10的合成。Example 10: Synthesis of compound 10.
(S)-8-(5–((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(1-甲基-1H-吡唑-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(1-methyl-1H-pyrazole-3- Yl)-2,8-diazaspiro[4.5]dec-4-amine
Figure PCTCN2020106214-appb-000052
Figure PCTCN2020106214-appb-000052
步骤一:将(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(A1,45mg,0.125mmol)溶于5mL无水1,4-二氧六环中,依次加入3-碘-1-甲基-1H-吡唑(4.114mg,0.556mmol),无水叔丁醇钾(70mg,0.624mmol),Brettphos Pd Gen.3(23mg,0.0254mmol),氩气保护,升温到100℃,反应5小时。将反应液倒至冰水中,乙酸乙酯萃取3次,有机相饱和盐水洗一次,合并有机相,无水硫酸钠干燥,过滤,浓缩,过柱,得(S)-4-(((S)-叔丁基亚磺酰基)氨基)-2-(1-甲基-1H-吡唑-3-基)-2,8-二氮杂螺[4.5]癸烷-8-叔丁酯羧酸(10-1,34mg,收率:61.84%)。LCMS:m/z 437.3[M+H] +Step 1: Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (A1, 45mg, 0.125mmol) dissolved in 5mL of anhydrous 1,4-dioxane, followed by adding 3-iodo-1-methyl-1H-pyrazole (4.114mg, 0.556mmol), anhydrous potassium tert-butoxide ( 70 mg, 0.624 mmol), Brettphos Pd Gen. 3 (23 mg, 0.0254 mmol), protected by argon, heated to 100°C, and reacted for 5 hours. The reaction solution was poured into ice water, extracted with ethyl acetate 3 times, the organic phase was washed once with saturated brine, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and passed through a column to obtain (S)-4-(((S )-Tert-Butylsulfinyl)amino)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazaspiro[4.5]decane-8-tert-butyl carboxy Acid (10-1, 34mg, yield: 61.84%). LCMS: m/z 437.3 [M+H] + .
步骤二:将(S)-4-(((S)-叔丁基亚磺酰基)氨基)-2-(1-甲基-1H-吡唑-3-基)-2,8-二氮杂螺[4.5]癸烷-8-叔丁酯羧酸(10-1,34mg,0.0773mmol)溶于5mL二氯甲烷/甲醇(1/1)中,氩气保护下,冰水降温到0度,加入4M HCl/1,4-二氧六环(0.5mL,2.0mmol),再在室温搅拌1小时,反应完全。将反应液直接旋干,得粗品(S)-2-(1-甲基-1H-吡唑-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(10-2,0.0773mmol),直接投下步。LCMS:m/z 236.3[M+H] +Step 2: Add (S)-4-(((S)-tert-butylsulfinyl)amino)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazepine Heterosspiro[4.5]decane-8-tert-butyl carboxylic acid (10-1, 34mg, 0.0773mmol) was dissolved in 5mL dichloromethane/methanol (1/1), under argon protection, the ice water was cooled to 0 After adding 4M HCl/1,4-dioxane (0.5 mL, 2.0 mmol), stirring at room temperature for 1 hour, the reaction was complete. The reaction solution was directly spin-dried to obtain the crude product (S)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (10- 2, 0.0773mmol), directly cast the step. LCMS: m/z 236.3 [M+H] + .
步骤三:将粗品(S)-2-(1-甲基-1H-吡唑-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(10-2,0.0773mmol)溶于5mL无水乙腈中,加入DIPEA(0.5mL,2.926mmol)与3-氯-4–((5-氯吡嗪-2-基)硫基)吡啶-2-胺(C2,28mg,0.1025mmol)。氩气保护,加热至90℃回流反应7小时,反应完毕。冷却至室温,旋干,爬大板所得纯点再次爬大板,得化合物10,(S)-8-(5–((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(1-甲基-1H-吡唑-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(15mg,收率:41.18%)。Step 3: The crude product (S)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (10-2, 0.0773mmol ) Dissolved in 5mL anhydrous acetonitrile, add DIPEA (0.5mL, 2.926mmol) and 3-chloro-4–((5-chloropyrazin-2-yl)thio)pyridin-2-amine (C2, 28mg, 0.1025mmol). Protected by argon, heated to 90°C and refluxed for 7 hours to complete the reaction. Cool to room temperature, spin dry, and climb the big plate to get the pure spots to climb the big plate again to obtain compound 10, (S)-8-(5—((2-amino-3-chloropyridin-4-yl)thio)pyridine Azin-2-yl)-2-(1-methyl-1H-pyrazol-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (15mg, yield: 41.18%) .
LCMS:m/z 472.5[M+H] + LCMS: m/z 472.5[M+H] +
1H NMR(400MHz,MeOD)δ8.338-8.282(m,2H),7.597(d,J=5.6Hz,1H),7.299(d,J=2.4Hz,1H),5.934(d,J=5.6Hz,1H),5.521(d,J=2.4Hz,1H),4.380-4.304(m,2H),3.702(s,3H),3.621(m,1H),3.546(d,J=9.6Hz,1H),3.264-3.234(m,2H),3,143-3.105(m,1H),1.844-1.747(m,2H),1.664-1.573(m,2H),1.286(s,1H). 1 H NMR(400MHz, MeOD) δ8.338-8.282(m,2H), 7.597(d,J=5.6Hz,1H), 7.299(d,J=2.4Hz,1H), 5.934(d,J=5.6 Hz,1H),5.521(d,J=2.4Hz,1H),4.380-4.304(m,2H),3.702(s,3H),3.621(m,1H),3.546(d,J=9.6Hz,1H ), 3.264-3.234 (m, 2H), 3,143-3.105 (m, 1H), 1.844-1.747 (m, 2H), 1.664-1.573 (m, 2H), 1.286 (s, 1H).
实施例十一:化合物11的合成。Example 11: Synthesis of compound 11.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(吡啶-2-基)-2,8-二氮杂螺[4.5]癸-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(pyridin-2-yl)-2,8-bis Azaspiro[4.5]dec-4-amine
Figure PCTCN2020106214-appb-000053
Figure PCTCN2020106214-appb-000053
步骤一:将(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(A1,100mg,0.278mmol)溶于5mL无水1,4-二氧六环中,依次加入2-碘吡啶(114mg,0.556mmol),无水碳酸铯(272mg,0.835mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(32mg,0.0553mmol)和Pd 2(dba) 3(26mg,0.0284mmol),氩气保护,升温到100℃,反应5小时。将反应液倒至冰水中,乙酸乙酯萃取3次,有机相饱和盐水洗一次,合并有机相,无水硫酸钠干燥,过滤,浓缩,过柱,得(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(吡啶-2-基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(11-1,33mg,收率:27.19%)。LCMS:m/z 437.3[M+H] + Step 1: Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (A1, 100mg, 0.278mmol) dissolved in 5mL of anhydrous 1,4-dioxane, followed by adding 2-iodopyridine (114mg, 0.556mmol), anhydrous cesium carbonate (272mg, 0.835mmol), 4,5-bis-dioxane Phenylphosphine-9,9-dimethylxanthene (32mg, 0.0553mmol) and Pd 2 (dba) 3 (26mg, 0.0284mmol), protected by argon, heated to 100°C and reacted for 5 hours. The reaction solution was poured into ice water, extracted with ethyl acetate 3 times, the organic phase was washed once with saturated brine, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and passed through a column to obtain (S)-4-(((R )-Tert-butylsulfinyl)amino)-2-(pyridin-2-yl)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (11-1, 33mg, Yield: 27.19%). LCMS: m/z 437.3[M+H] +
步骤二:将(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(吡啶-2-基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(11-1,33mg,0.07558mmol)溶于5mL二氯甲烷/甲醇(1/1)中,氩气保护下,冰水降温到0度,加入4M HCl/1,4-二氧六环(0.3mL,1.2mmol),再在室温搅拌1小时,反应完全。将反应液直接旋干,得粗品(S)-2-(吡啶-2-基)-2,8-二氮杂螺[4.5]癸-4-胺(11-2,0.07558mmol),直接投下步。LCMS:m/z 304.2[M+H] +Step 2: Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(pyridin-2-yl)-2,8-diazaspiro[4.5]decane- Tert-Butyl 8-carboxylate (11-1, 33mg, 0.07558mmol) was dissolved in 5mL dichloromethane/methanol (1/1), under the protection of argon, the temperature of ice water was cooled to 0 degrees, and 4M HCl/1 was added. 4-Dioxane (0.3 mL, 1.2 mmol) was stirred at room temperature for 1 hour, and the reaction was complete. The reaction solution was spin-dried directly to obtain crude (S)-2-(pyridin-2-yl)-2,8-diazaspiro[4.5]dec-4-amine (11-2, 0.07558mmol), which was directly cast step. LCMS: m/z 304.2 [M+H] + .
步骤三:将粗品(S)-2-(吡啶-2-基)-2,8-二氮杂螺[4.5]癸-4-胺(11-2,0.07558mmol)溶于5mL无水乙腈中,加入DIPEA(0.5mL,2.926mmol)与3-氯-4-((5-氯吡嗪-2-基) 硫基)吡啶-2-胺(7,27mg,0.09886mmol)。氩气保护,加热至90度回流反应7小时,反应完毕。冷却至室温,旋干,过柱纯化,得化合物(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(吡啶-2-基)-2,8-二氮杂螺[4.5]癸-4-胺(11,9.2mg,收率:25.95%)。Step 3: Dissolve the crude (S)-2-(pyridin-2-yl)-2,8-diazaspiro[4.5]dec-4-amine (11-2, 0.07558mmol) in 5mL anhydrous acetonitrile , DIPEA (0.5 mL, 2.926 mmol) and 3-chloro-4-((5-chloropyrazin-2-yl)thio)pyridin-2-amine (7, 27 mg, 0.09886 mmol) were added. Protected by argon, heated to 90° and refluxed for 7 hours, and the reaction was completed. Cool to room temperature, spin dry, and purify by column to obtain compound (S)-8-(5-((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2- (Pyridin-2-yl)-2,8-diazaspiro[4.5]dec-4-amine (11,9.2 mg, yield: 25.95%).
LCMS:m/z 469.1[M+H] +LCMS: m/z 469.1 [M+H] + ;
1H NMR(400MHz,MeOD)δ8.346(d,J=24.4Hz,2H),8.085(d,J=5.2Hz,1H),7.638-7.591(m,2H),6.723-6.646(m,2H),5.938(d,J=5.6Hz,1H),4.420-4.299(m,2H),4.001-3.956(m,1H),3.800-3.629(m,4H),3.460-3.417(m,1H),1.887-1.749(m,4H),1.287(s,1H)。 1 H NMR (400MHz, MeOD) δ 8.346 (d, J = 24.4 Hz, 2H), 8.085 (d, J = 5.2 Hz, 1H), 7.638-7.591 (m, 2H), 6.723-6.646 (m, 2H) ), 5.938 (d, J = 5.6Hz, 1H), 4.420-4.299 (m, 2H), 4.001-3.956 (m, 1H), 3.800-3.629 (m, 4H), 3.460-3.417 (m, 1H), 1.887-1.749 (m, 4H), 1.287 (s, 1H).
实施例十二:化合物12的合成。Example 12: Synthesis of compound 12.
(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(吡啶-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(S)-8-(5-((2-Amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-(pyridin-3-yl)-2,8-bis Azaspiro[4.5]dec-4-amine
Figure PCTCN2020106214-appb-000054
Figure PCTCN2020106214-appb-000054
步骤一:将(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(A1,50mg,0.139mmol)溶于5mL无水1,4-二氧六环中,依次加入3-碘吡啶(4,57mg,0.278mmol),无水碳酸铯(136mg,0.417mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(17mg,0.0294mmol)和Pd 2(dba) 3(13mg,0.0298mmol),氩气保护,升温到100度,反应5小时,反应完毕。将反应液倒至冰水中,乙酸乙酯萃取3次,有机相饱和盐水洗一次,合并有机相,无水硫酸钠干燥,过滤,浓缩,过柱,得(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(吡啶-3-基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(12-1,39mg,收率:64.26%)。LCMS:m/z 437.3[M+H] +Step 1: Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (A1, 50mg, 0.139mmol) was dissolved in 5mL of anhydrous 1,4-dioxane, followed by the addition of 3-iodopyridine (4,57mg, 0.278mmol), anhydrous cesium carbonate (136mg, 0.417mmol), 4,5- Bisdiphenylphosphine-9,9-dimethylxanthene (17mg, 0.0294mmol) and Pd 2 (dba) 3 (13mg, 0.0298mmol), protected by argon, heated to 100 degrees, reacted for 5 hours, react complete. The reaction solution was poured into ice water, extracted with ethyl acetate 3 times, the organic phase was washed once with saturated brine, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and passed through a column to obtain (S)-4-(((R )-Tert-butylsulfinyl)amino)-2-(pyridin-3-yl)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (12-1, 39mg, Yield: 64.26%). LCMS: m/z 437.3 [M+H] + .
步骤二:将(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(吡啶-3-基)-2,8-二氮杂螺[4.5]癸烷-8-羧酸叔丁酯(12-1,39mg,0.08923mmol)溶于5mL二氯甲烷/甲醇(1/1)中,氩气保护下,冰水降温到0度,加入4M HCl/1,4-二氧六环,再在室温搅拌1小时,反应完全。将反应液直接旋干,得粗品(S)-2-(吡啶-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(12-2, 0.08923mmol),直接投下步。LCMS:m/z 233.5[M+H] +Step 2: Add (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(pyridin-3-yl)-2,8-diazaspiro[4.5]decane- Tert-butyl 8-carboxylate (12-1, 39mg, 0.08923mmol) was dissolved in 5mL of dichloromethane/methanol (1/1), under the protection of argon, the temperature of ice water was cooled to 0 degrees, and 4M HCl/1 was added. 4-Dioxane, then stirred at room temperature for 1 hour, the reaction was complete. The reaction solution was directly spin-dried to obtain the crude product (S)-2-(pyridin-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (12-2, 0.08923mmol), which was cast directly step. LCMS: m/z 233.5 [M+H] + .
步骤三:将粗品(S)-2-(吡啶-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(12-2,0.08923mmol)溶于5mL无水乙腈中,加入DIPEA(0.5mL,2.926mmol)与3-氯-4-((5-氯吡嗪-2-基)硫基)吡啶-2-胺(7,32mg,0.1172mmol)。氩气保护,加热至90度回流反应7小时,反应完毕。冷却至室温,旋干,乙酸乙酯打浆,过滤,制备硅胶板纯化,得(S)-8-(5-((2-氨基-3-氯吡啶-4-基)硫基)吡嗪-2-基)-2-(吡啶-3-基)-2,8-二氮杂螺[4.5]癸-4-胺(化合物12,16.4mg,收率:39.15%)。Step 3: Dissolve the crude (S)-2-(pyridin-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (12-2, 0.08923mmol) in 5mL anhydrous acetonitrile , DIPEA (0.5 mL, 2.926 mmol) and 3-chloro-4-((5-chloropyrazin-2-yl)thio)pyridin-2-amine (7, 32 mg, 0.1172 mmol) were added. Protected by argon, heated to 90° and refluxed for 7 hours, and the reaction was completed. Cool to room temperature, spin dry, beaten with ethyl acetate, filter, and prepare a silica gel plate for purification to obtain (S)-8-(5-((2-amino-3-chloropyridin-4-yl)thio)pyrazine- 2-yl)-2-(pyridin-3-yl)-2,8-diazaspiro[4.5]dec-4-amine (compound 12, 16.4 mg, yield: 39.15%).
LCMS:m/z 469.1[M+H] +LCMS: m/z 469.1 [M+H] + ;
1H NMR(400MHz,MeOD)δ8.382-8.318(m,2H),8.036-7.960(m,2H),7.601(d,J=5.6Hz,1H),7.444-7.411(m,1H),7.310-7.284(m,1H),5.965(d,J=5.6Hz,1H),4.398-4.305(m,2H),3.935-3.907(m,1H),3.849-3.829(m,1H),3.686-3.617(m,2H),3.567-3.533(m,1H),1.879-1.806(m,4H),1.387-1,360(m,2H。 1 H NMR (400MHz, MeOD) δ 8.382-8.318 (m, 2H), 8.036-7.960 (m, 2H), 7.601 (d, J = 5.6 Hz, 1H), 7.444-7.411 (m, 1H), 7.310 -7.284 (m, 1H), 5.965 (d, J = 5.6 Hz, 1H), 4.398-4.305 (m, 2H), 3.935-3.907 (m, 1H), 3.849-3.829 (m, 1H), 3.686-3.617 (m, 2H), 3.567-3.533 (m, 1H), 1.879-1.806 (m, 4H), 1.387-1,360 (m, 2H.
实施例十三:化合物13的合成。Example 13: Synthesis of Compound 13.
(S)-8-(6-氨基-5-(((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(6-Amino-5-(((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-phenyl-2,8-di Azaspiro[4.5]decyl 4-amine
Figure PCTCN2020106214-appb-000055
Figure PCTCN2020106214-appb-000055
步骤一:将3-溴-6-氯吡嗪-2-胺(208mg,1.0mmol)和2-氨基-3-氯吡啶-4-硫醇(168mg,1.05mmol)溶于二氧六环(10mL),加入碘化亚铜(95mg,0.5mmol)和1,10-邻二氮菲(108mg,0.6mmol)和DIPEA(260mg,2mmol)。升温到120度,氮气下反应过夜。加乙酸乙酯(80mL)稀释,饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化(石油醚:乙酸乙酯=1:2)得3-((2-氨基-3-氯吡啶-4-基)硫基)-6-氯吡嗪-2-胺(13-1,238mg,收率:82.6%),为浅黄色固体。LCMS:m/z 287.9[M+H] +Step 1: Dissolve 3-bromo-6-chloropyrazine-2-amine (208mg, 1.0mmol) and 2-amino-3-chloropyridine-4-thiol (168mg, 1.05mmol) in dioxane ( 10mL), copper iodide (95mg, 0.5mmol) and 1,10-phenanthroline (108mg, 0.6mmol) and DIPEA (260mg, 2mmol) were added. The temperature was raised to 120 degrees and reacted overnight under nitrogen. Dilute with ethyl acetate (80 mL), wash with saturated brine (100 mL), dry with anhydrous sodium sulfate, filter, concentrate under reduced pressure, and purify by column chromatography (petroleum ether: ethyl acetate = 1:2) to obtain 3-(( 2-Amino-3-chloropyridin-4-yl)sulfanyl)-6-chloropyrazine-2-amine (13-1, 238 mg, yield: 82.6%), as a pale yellow solid. LCMS: m/z 287.9 [M+H] + .
步骤二:将3-((2-氨基-3-氯吡啶-4-基)硫基)-6-氯吡嗪-2-胺(13-1,45mg,0.15mmol)和2-苯基-2,8-二氮杂螺[4.5]癸-4-胺盐酸盐(50mg,0.16mmol)溶于乙腈(10mL),加入DIPEA(180mg,1.47mmol)。升温到110度,反应过夜。加乙酸乙酯(80mL)稀释,饱和盐水洗涤(100mL),无水硫酸钠干燥,过滤,减压浓缩,硅胶柱层析(二氯甲烷:甲醇=10:1)和Prep-HPLC纯化得8-(6-氨基-5-(((2-氨基-3-氯吡啶-4-基)硫代)吡嗪 -2-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(13,10mg,收率:12.8%)。Step 2: Combine 3-((2-amino-3-chloropyridin-4-yl)thio)-6-chloropyrazine-2-amine (13-1, 45mg, 0.15mmol) and 2-phenyl- 2,8-Diazaspiro[4.5]dec-4-amine hydrochloride (50 mg, 0.16 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (180 mg, 1.47 mmol) was added. The temperature is increased to 110 degrees and the reaction is overnight. Dilute with ethyl acetate (80mL), wash with saturated brine (100mL), dry with anhydrous sodium sulfate, filter, concentrate under reduced pressure, silica gel column chromatography (dichloromethane: methanol = 10:1) and Purp-HPLC to obtain 8 -(6-Amino-5-(((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-phenyl-2,8-diazaspiro[4.5 ] Decane 4-amine (13, 10 mg, yield: 12.8%).
LCMS:m/z 483.5[M+H] +LCMS: m/z 483.5[M+H] + ;
1H NMR(400MHz,MeOD-d 4)δ7.59(t,J=2.6Hz,2H),7.16(t,J=7.8Hz,2H),6.63-6.57(m,3H),5.93(d,J=5.6Hz,1H),4.28-4.23(m,2H),3.65-3.61(m,1H),3.56-3.54(m,1H),3.28-3.24(m,4H),3.13-3.10(m,1H),1.81-1.77(m,2H),1.63-1.58(m,2H)。 1 H NMR(400MHz, MeOD-d 4 )δ7.59(t,J=2.6Hz,2H), 7.16(t,J=7.8Hz,2H), 6.63-6.57(m,3H), 5.93(d, J = 5.6Hz, 1H), 4.28-4.23 (m, 2H), 3.65-3.61 (m, 1H), 3.56-3.54 (m, 1H), 3.28-3.24 (m, 4H), 3.13-3.10 (m, 1H), 1.81-1.77 (m, 2H), 1.63-1.58 (m, 2H).
实施例十四:化合物14的合成。Example 14: Synthesis of compound 14.
(S)-8-(6-氨基-5-(((2-氨基-3-氯吡啶-4-基)硫代)吡嗪-2-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(6-Amino-5-(((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-2-phenyl-2,8-di Azaspiro[4.5]decyl 4-amine
Figure PCTCN2020106214-appb-000056
Figure PCTCN2020106214-appb-000056
步骤一:将5-氯-8-碘-7-甲基咪唑并[1,2-c]嘧啶(B4,34mg,0.12mmol)和2-苯基-2,8-二氮杂螺[4.5]癸-4-胺盐酸盐(3-2,35mg,0.12mmol)溶于乙腈(10mL),加入DIPEA(0.2mL,1.98mmol)。升温到65℃,反应2h。加乙酸乙酯(80mL)稀释,饱和盐水洗涤(100mL),无水硫酸钠干燥,过滤,减压浓缩,经柱层析纯化(二氯甲烷:甲醇=30:1)得浅黄色固体8-(8-碘-7-甲基咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-胺(14-1,50mg,收率:89.3%)。LCMS:m/z 488.9[M+H] +Step 1: Combine 5-chloro-8-iodo-7-methylimidazo[1,2-c]pyrimidine (B4, 34mg, 0.12mmol) and 2-phenyl-2,8-diazaspiro[4.5 ] Deca-4-amine hydrochloride (3-2, 35 mg, 0.12 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.2 mL, 1.98 mmol) was added. The temperature was raised to 65°C and reacted for 2h. Dilute with ethyl acetate (80 mL), wash with saturated brine (100 mL), dry with anhydrous sodium sulfate, filter, concentrate under reduced pressure, and purify by column chromatography (dichloromethane: methanol = 30:1) to obtain a pale yellow solid 8- (8-Iodo-7-methylimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-amine (14-1 , 50mg, yield: 89.3%). LCMS: m/z 488.9 [M+H] + .
步骤二:将8-(8-碘-7-甲基咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-胺(50mg,0.10mmol)和2-氨基-3-氯吡啶-4-硫钠(37mg,0.2mmol)悬浮于二氧六环(10mL),加入DIPEA(0.2mL,1.21mmol),Xantphos(29mg,0.05mmol)和Pd 2(dba) 3(45mg,0.05mmol),氩气置换三次。升温到120℃,反应4h。加乙酸乙酯(80mL)稀释,过滤,减压浓缩,柱层析纯化(二氯甲烷:甲醇=8:1)和prep-HPLC纯化得8-(8-(((2-氨基-3-氯吡啶-4-基)硫基)-7-甲基咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(14,4mg,收率:5.9%)。 Step 2: Add 8-(8-iodo-7-methylimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4 -Amine (50mg, 0.10mmol) and sodium 2-amino-3-chloropyridine-4-sulfide (37mg, 0.2mmol) were suspended in dioxane (10mL), DIPEA (0.2mL, 1.21mmol) was added, Xantphos ( 29 mg, 0.05 mmol) and Pd 2 (dba) 3 (45 mg, 0.05 mmol), replaced with argon three times. The temperature was raised to 120°C and reacted for 4 hours. Diluted with ethyl acetate (80 mL), filtered, concentrated under reduced pressure, purified by column chromatography (dichloromethane: methanol = 8:1) and purified by prep-HPLC to obtain 8-(8-(((2-amino-3- (Chloropyridin-4-yl)thio)-7-methylimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec4- Amine (14, 4 mg, yield: 5.9%).
LCMS:m/z 521.2[M+H] +LCMS: m/z 521.2[M+H] + ;
1H NMR(400MHz,DMSO-d 6)δ8.35(s,3H),7.76(s,1H),7.55(d,J=5.2Hz,1H),7.49(s,1H),7.16(t,J=7.6Hz,2H),6.57-6.52(m,3H),6.31(s,2H),5.71(d,J=5.2Hz,1H),3.90-3.88(m,2H),3.32-3.00(m,7H),2.47(s,3H),1.95-1.92(m,2H),1.58-1.55(m,2H)。 1 H NMR(400MHz,DMSO-d 6 )δ8.35(s,3H),7.76(s,1H),7.55(d,J=5.2Hz,1H),7.49(s,1H),7.16(t, J = 7.6Hz, 2H), 6.57-6.52 (m, 3H), 6.31 (s, 2H), 5.71 (d, J = 5.2Hz, 1H), 3.90-3.88 (m, 2H), 3.32-3.00 (m , 7H), 2.47 (s, 3H), 1.95-1.92 (m, 2H), 1.58-1.55 (m, 2H).
实施例十五:化合物15的合成。Example 15: Synthesis of Compound 15.
(S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫基)-7-氯咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(8-(((2-Amino-3-chloropyridin-4-yl)thio)-7-chloroimidazo[1,2-c]pyrimidin-5-yl)-2- Phenyl-2,8-diazaspiro[4.5]decyl 4-amine
Figure PCTCN2020106214-appb-000057
Figure PCTCN2020106214-appb-000057
步骤一:向7-氯-8-碘-5-(甲基亚磺酰基)咪唑并[1,2-c]嘧啶(B5,222mg,0.65mmol)和(S)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(3-2,220mg,0.72mmol)的乙腈(30ml)悬浊液中加入Cs 2CO 3(2.1g,6.45mmol)。升温到30度,反应1小时。反应液用水(50ml)稀释,乙酸乙酯萃取(2×30ml),有机相用食盐水洗,无水硫酸钠干燥,过滤,浓缩,经柱色谱纯化得白色固体(S)-8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(15-1,140mg,收率42.4%)。 Step 1: To 7-chloro-8-iodo-5-(methylsulfinyl)imidazo[1,2-c]pyrimidine (B5, 222mg, 0.65mmol) and (S)-2-phenyl-2 Cs 2 CO 3 (2.1 g, 6.45 mmol) was added to the suspension of 8-diazaspiro[4.5]decane 4-amine (3-2, 220 mg, 0.72 mmol) in acetonitrile (30 ml). The temperature was raised to 30 degrees and reacted for 1 hour. The reaction solution was diluted with water (50ml), extracted with ethyl acetate (2×30ml), the organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to obtain a white solid (S)-8-(7- Chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec4-amine (15-1, 140mg, yield 42.4%).
LCMS:m/z 509.0[M+H] +LCMS: m/z 509.0 [M+H] + .
步骤二:向(S)-8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(15-1,140mg,0.28mmol)的二氯甲烷(20mL)溶液中加入三乙胺(60mg,0.56mmol)和(Boc) 2O(120mg,0.56mmol),室温反应过夜。将反应液浓缩,经柱层析纯化得浅黄色固体叔丁基(S)-(8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-基)氨基甲酸酯(15-2,150mg,收率89.8%)。 Step 2: To (S)-8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5] Triethylamine (60 mg, 0.56 mmol) and (Boc) 2 O (120 mg, 0.56 mmol) were added to a solution of decyl 4-amine (15-1, 140 mg, 0.28 mmol) in dichloromethane (20 mL), and reacted at room temperature overnight. The reaction solution was concentrated and purified by column chromatography to obtain pale yellow solid tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2- Phenyl-2,8-diazaspiro[4.5]dec4-yl)carbamate (15-2, 150 mg, yield 89.8%).
LCMS:m/z 609.0[M+H] +LCMS: m/z 609.0 [M+H] + .
步骤三:向叔丁基(S)-(8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-基)氨基甲酸酯(15-2,130mg,0.21mmol)的二氧六环(120mL)溶液中加入2-氨基-3-氯吡啶-4-硫代硫酸钠(C1,115mg,0.64mmol),碘化亚铜(40mg,0.21mmol)和邻菲罗啉(20mg,0.11mmol),氩气置换三次,升温到110℃,反应过夜。将反应液浓缩,经柱层析得浅黄色固体叔丁基(S)-(8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)-7-氯吲哚[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-基)氨基甲酸酯(15-3,120mg,收率87.6%)。Step 3: To tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazepine Spiro[4.5]dec4-yl)carbamate (15-2,130mg, 0.21mmol) in dioxane (120mL) was added with 2-amino-3-chloropyridine-4-sodium thiosulfate ( C1, 115mg, 0.64mmol), cuprous iodide (40mg, 0.21mmol) and o-phenanthroline (20mg, 0.11mmol), replaced with argon three times, heated to 110°C, and reacted overnight. The reaction solution was concentrated, and column chromatography was carried out to obtain a pale yellow solid tert-butyl (S)-(8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroindole Indole [1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (15-3, 120mg, yield 87.6%).
LCMS:m/z 641.1[M+H] +LCMS: m/z 641.1 [M+H] + .
步骤四:向叔丁基(S)-(8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)-7-氯吲哚[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-基)氨基甲酸酯(15-3,20mg,0.03mmol)的二氯甲烷(4mL)溶液中加入三氟乙酸(2mL),室温反应2小时。将反应液浓缩,用二氯甲烷(50mL)稀释,饱和碳酸氢钠洗涤,分液,有机相用食盐水洗涤,干燥,过滤,浓缩至干,经制备色谱纯化得白色固体(S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫基)-7-氯咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺的甲酸盐(15,5mg,收率28.0%)。Step 4: To tert-butyl (S)-(8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroindole[1,2-c]pyrimidine -5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (15-3, 20mg, 0.03mmol) in dichloromethane (4mL) Trifluoroacetic acid (2mL) was added and reacted for 2 hours at room temperature. The reaction solution was concentrated, diluted with dichloromethane (50mL), washed with saturated sodium bicarbonate, separated, the organic phase was washed with brine, dried, filtered, and concentrated to Dry, purified by preparative chromatography to obtain a white solid (S)-8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroimidazo[1,2-c] Pyrimidine-5-yl)-2-phenyl-2,8-diazaspiro[4.5]decane 4-amine formate (15,5 mg, yield 28.0%).
LCMS:m/z 541.1[M+H] +LCMS: m/z 541.1[M+H] + ;
1H NMR(400MHz,MeOD-d 4)δ8.55(brs,1H),7.84(s,1H),7.54-7.51(m,2H),7.23-7.20(m,2H),6.71-6.65(m,3H),5.91(d,J=5.2Hz,1H),4.59-4.57(m,1H),4.17-4.09(m,2H),3.75-3.25(m,6H),2.06-1.97(m,2H),1.88-1.77(m,2H)。 1 H NMR (400MHz, MeOD-d 4 ) δ8.55 (brs, 1H), 7.84 (s, 1H), 7.54-7.51 (m, 2H), 7.23-7.20 (m, 2H), 6.71-6.65 (m ,3H),5.91(d,J=5.2Hz,1H),4.59-4.57(m,1H),4.17-4.09(m,2H),3.75-3.25(m,6H),2.06-1.97(m,2H) ), 1.88-1.77 (m, 2H).
实施例十六:化合物16的合成。Example 16: Synthesis of Compound 16.
(S)-8-(7-氨基-8-((2-氨基-3-氯吡啶基-4-基)硫基)咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(7-amino-8-((2-amino-3-chloropyridinyl-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2- Phenyl-2,8-diazaspiro[4.5]decyl 4-amine
Figure PCTCN2020106214-appb-000058
Figure PCTCN2020106214-appb-000058
步骤一:向7-氯-8-碘-5-(甲基亚磺酰基)咪唑并[1,2-c]嘧啶(B5,222mg,0.65mmol)和(S)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(3-2,220mg,0.72mmol)的乙腈(30ml)悬浊液中加入Cs 2CO 3(2.1g,6.45mmol),升温到30度,反应1小时。反应液用水(50ml)稀释,乙酸乙酯萃取(2×30ml),有机相用食盐水洗,无水硫酸钠干燥,过滤,浓缩,经 柱色谱纯化得白色固体(S)-8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(16-1,140mg,收率42.4%)。 Step 1: To 7-chloro-8-iodo-5-(methylsulfinyl)imidazo[1,2-c]pyrimidine (B5, 222mg, 0.65mmol) and (S)-2-phenyl-2 Add Cs 2 CO 3 (2.1g, 6.45mmol) to the suspension of ,8-diazaspiro[4.5]decane 4-amine (3-2, 220mg, 0.72mmol) in acetonitrile (30ml) and raise the temperature to 30°C , Reaction for 1 hour. The reaction solution was diluted with water (50ml), extracted with ethyl acetate (2×30ml), the organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to obtain a white solid (S)-8-(7- Chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec4-amine (16-1, 140mg, yield 42.4%).
LCMS:m/z 509.0[M+H] +LCMS: m/z 509.0 [M+H] + .
步骤二:向(S)-8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺(16-1,140mg,0.28mmol)的二氯甲烷(20mL)溶液中加入三乙胺(60mg,0.56mmol)和(Boc) 2O(120mg,0.56mmol),室温反应过夜。将反应液浓缩,经柱层析纯化得浅黄色固体叔丁基(S)-(8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-基)氨基甲酸酯(16-2,150mg,收率89.8%)。 Step 2: To (S)-8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5] Triethylamine (60 mg, 0.56 mmol) and (Boc) 2 O (120 mg, 0.56 mmol) were added to the solution of decyl 4-amine (16-1, 140 mg, 0.28 mmol) in dichloromethane (20 mL), and reacted at room temperature overnight. The reaction solution was concentrated and purified by column chromatography to obtain pale yellow solid tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2- Phenyl-2,8-diazaspiro[4.5]dec4-yl)carbamate (16-2, 150 mg, yield 89.8%).
LCMS:m/z 609.0[M+H] +LCMS: m/z 609.0 [M+H] + .
步骤三:向叔丁基(S)-(8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-基)氨基甲酸酯(16-2,130mg,0.21mmol)的二氧六环(120mL)溶液中加入2-氨基-3-氯吡啶-4-硫代硫酸钠(C1,115mg,0.64mmol),碘化亚铜(40mg,0.21mmol)和邻菲罗啉(20mg,0.11mmol),氩气置换三次,升温到110℃,反应过夜。将反应液浓缩,经柱层析得浅黄色固体叔丁基(S)-(8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)-7-氯吲哚[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-基)氨基甲酸酯(16-3,120mg,收率87.6%)。Step 3: To tert-butyl (S)-(8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazepine Spiro[4.5]dec4-yl)carbamate (16-2, 130mg, 0.21mmol) in dioxane (120mL) was added with 2-amino-3-chloropyridine-4-sodium thiosulfate ( C1, 115mg, 0.64mmol), cuprous iodide (40mg, 0.21mmol) and o-phenanthroline (20mg, 0.11mmol), replaced with argon three times, heated to 110°C, and reacted overnight. The reaction solution was concentrated, and column chromatography was performed to obtain a pale yellow solid tert-butyl (S)-(8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroindole Indole[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-3, 120mg, yield 87.6%).
LCMS:m/z 641.1[M+H] +LCMS: m/z 641.1 [M+H] + .
步骤四:氩气保护下,向叔丁基(S)-(8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)-7-氯吲哚[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-基)氨基甲酸酯(16-3,30mg,0.05mmol)的DMF溶液中加入叠氮化钠(12mg,0.19mmol)和碳酸钾(26mg,0.19mmol),氩气置换三次,升温到100℃,反应过夜。将反应液加乙酸乙酯(50mL)稀释,依次用水(3×50mL)和饱和盐水(2×50mL)洗涤,干燥,过滤,浓缩得浅黄色固体叔丁基(S)-(8-(7-氨基-8-((2-氨基-3-氯吡啶-4-基)硫代)咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-基)氨基甲酸酯(16-4,20mg)。Step 4: Under the protection of argon, add tert-butyl(S)-(8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroindole [1, 2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-3, 30mg, 0.05mmol) in DMF Sodium azide (12mg, 0.19mmol) and potassium carbonate (26mg, 0.19mmol) were added to the mixture, replaced with argon three times, heated to 100°C, and reacted overnight. The reaction solution was diluted with ethyl acetate (50mL), followed by water ( 3×50mL) and saturated brine (2×50mL) washed, dried, filtered, and concentrated to obtain a pale yellow solid tert-butyl (S)-(8-(7-amino-8-((2-amino-3-chloropyridine) -4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-4, 20mg).
LCMS:m/z 622.2[M+H] +LCMS: m/z 622.2 [M+H] + .
步骤五:向叔丁基(S)-(8-(7-氨基-8-((2-氨基-3-氯吡啶-4-基)硫代)咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸-4-基)氨基甲酸酯(16-4,30mg,30%,0.014mmol)的二氯甲烷(10mL)溶液中加入三氟乙酸(1mL,1.35mmol),室温反应1h。将反应液浓缩,二氯甲烷(50mL)稀释,依次用饱和碳酸氢钠和饱和盐水洗涤,干燥,过滤,浓缩,经Prep-HPLC纯化得白色固体(S)-8-(7-氨基-8-((2-氨基-3-氯吡啶基-4-基)硫基)咪唑并[1,2-c]嘧啶-5-基)-2-苯基-2,8-二氮杂螺[4.5]癸4-胺的三氟乙酸盐(16,2.5mg,收率30.0%)。Step 5: To tert-butyl (S)-(8-(7-amino-8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidine- 5-yl)-2-phenyl-2,8-diazaspiro[4.5]dec-4-yl)carbamate (16-4, 30mg, 30%, 0.014mmol) in dichloromethane (10mL ) Trifluoroacetic acid (1 mL, 1.35 mmol) was added to the solution and reacted at room temperature for 1 h. The reaction solution was concentrated, diluted with dichloromethane (50mL), washed with saturated sodium bicarbonate and saturated brine successively, dried, filtered, concentrated, and purified by Prep-HPLC to obtain a white solid (S)-8-(7-amino-8) -((2-Amino-3-chloropyridinyl-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-phenyl-2,8-diazaspiro[ 4.5] Trifluoroacetate of decyl 4-amine (16, 2.5 mg, yield 30.0%).
LCMS:m/z 522.2[M+H] +LCMS: m/z 522.2[M+H] + ;
1H NMR(400MHz,DMSO-d 6+D 2O):δ7.72(d,J=2.8Hz,1H),7.68(d,J=5.6Hz,1H),7.65(d,J=2.4Hz,1H),7.23(t,J=7.6Hz,2H),6.69(t,J=7.6Hz,1H),6.63(d,J=7.6Hz,2H),6.19(d,J=5.6Hz,1H),3.92-3.86(m,2H),3.82-3.74(m,2H),3.47(s,3H),3.45-3.38(m,2H),1.93-1.88(m,2H),1.82-1.77(m,1H),1.74-1.69(m,1H)。 1 H NMR(400MHz,DMSO-d 6 +D 2 O): δ7.72(d,J=2.8Hz,1H), 7.68(d,J=5.6Hz,1H), 7.65(d,J=2.4Hz ,1H),7.23(t,J=7.6Hz,2H),6.69(t,J=7.6Hz,1H),6.63(d,J=7.6Hz,2H),6.19(d,J=5.6Hz,1H ),3.92-3.86(m,2H),3.82-3.74(m,2H),3.47(s,3H),3.45-3.38(m,2H),1.93-1.88(m,2H),1.82-1.77(m ,1H),1.74-1.69(m,1H).
实施例十七:化合物17的合成。Example 17: Synthesis of Compound 17.
(S)-4-(4-氨基-8-((2-氨基-3-氯吡啶-4-基)硫基)咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]-2-基)苯甲腈(S)-4-(4-Amino-8-((2-amino-3-chloropyridin-4-yl)sulfanyl)imidazo[1,2-c]pyrimidin-5-yl)-2,8 -Diazaspiro[4.5]-2-yl)benzonitrile
Figure PCTCN2020106214-appb-000059
Figure PCTCN2020106214-appb-000059
步骤一:向叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(A1,100mg,0.28mmol)的1,4-二氧六环(20mL)溶液中依次加入4-碘苯甲腈(128mg,0.56mmol),Cs 2CO 3(272mg,0.42mmol)和Xantphos(32mg,0.03mmol)。真空氮气置换3次后加入Pd 2(dba) 3(30mg,0.01mmol)。加毕,真空氮气再置换3次。加热至110℃反应12h。反应液滤除不溶物,滤液减压浓缩干得棕色油状物,经硅胶柱层析纯化得棕黄色固体叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(4-氰基苯基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(17-1,103mg,收率:80.4%)。LCMS:m/z 461.6[M+H] +1H NMR(400MHz,CDCl 3),δppm 7.50(d,J=2.8Hz,2H),7.47(d,J=2.4Hz,2H),4.00~3.97(m,2H),3.79~3.71(m,4H),3.50(d,J=9.2Hz,1H),3.39~3.35(m,2H),3.33(d,J=5.6Hz,2H),3.26(d,J=10.0Hz,2H),1.46(s,9H),1.24(s,9H)。 Step 1: To tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylate ( A1, 100mg, 0.28mmol) in 1,4-dioxane (20mL) solution was sequentially added 4-iodobenzonitrile (128mg, 0.56mmol), Cs 2 CO 3 (272mg, 0.42mmol) and Xantphos (32mg , 0.03mmol). Pd 2 (dba) 3 (30 mg, 0.01 mmol) was added after 3 times of vacuum nitrogen replacement. After the addition, the vacuum nitrogen is replaced 3 times. Heat to 110°C for 12h. The reaction solution was filtered to remove insoluble materials, and the filtrate was concentrated under reduced pressure to dry to obtain a brown oil, which was purified by silica gel column chromatography to obtain a brown solid tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino group )-2-(4-cyanophenyl)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (17-1, 103 mg, yield: 80.4%). LCMS: m/z 461.6[M+H] + ; 1 H NMR(400MHz, CDCl 3 ), δppm 7.50(d,J=2.8Hz,2H), 7.47(d,J=2.4Hz,2H), 4.00~ 3.97(m,2H),3.79~3.71(m,4H), 3.50(d,J=9.2Hz,1H), 3.39~3.35(m,2H),3.33(d,J=5.6Hz,2H), 3.26 (d, J=10.0 Hz, 2H), 1.46 (s, 9H), 1.24 (s, 9H).
步骤二:0℃下向叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(4-氰基苯基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(17-1,103mg,0.22mmol)的二氯甲烷(5mL)溶液中滴加HCl/1,4-二氧六环(4.0M,0.6mL)。升至室温反应1小时。反应液减压浓 缩干得棕黄色固体(S)-4-(4-氨基-2,8-二氮杂螺[4.5]癸-2-基)苯甲腈盐酸盐(17-2,粗品,98mg)。LCMS:m/z 257.5[M+H] +Step 2: Add tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2-(4-cyanophenyl)-2,8-diazepine at 0℃ Spiro[4.5]decane-8-t-butyl carboxylate (17-1, 103mg, 0.22mmol) in dichloromethane (5mL) was added dropwise HCl/1,4-dioxane (4.0M, 0.6mL ). Raise to room temperature and react for 1 hour. The reaction solution was concentrated and dried under reduced pressure to obtain a brown-yellow solid (S)-4-(4-amino-2,8-diazaspiro[4.5]dec-2-yl)benzonitrile hydrochloride (17-2, crude product) , 98mg). LCMS: m/z 257.5 [M+H] + .
步骤三:向(S)-4-(4-氨基-2,8-二氮杂螺[4.5]癸-2-基)苯甲腈(17-2,98mg,0.22mmol)的异丙醇(12mL)溶液中加入DIPEA(1143mg,8.85mmol)和5-氯-8-碘咪唑并[1,2-c]嘧啶(138mg,0.49mmol)。升至70℃反应4小时。反应液减压浓缩干,硅胶柱层析纯化(二氯甲烷/甲醇=50/1)得棕黄色固体(S)-4-(4-氨基-8-(8-碘咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸-2-基)苯甲腈(17-3,98mg,两步收率:87.8%)。LCMS:m/z 500.4[M+H] +1H NMR(400MHz,DMSO-d6),δppm 8.05(s,1H),7.87(s,1H),7.66(s,1H),7.56(d,J=8.8Hz,2H),6.66(d,J=8.8Hz,2H),3.70~3.68(m,3H),3.55(d,J=10.4Hz,1H),3.48~3.45(m,2H),3.38(s,1H),3.24~3.22(m,4H),1.96~1.86(m,2H),1.62~1.56(m,2H)。 Step 3: To (S)-4-(4-amino-2,8-diazaspiro[4.5]dec-2-yl)benzonitrile (17-2, 98mg, 0.22mmol) in isopropanol ( (12mL) DIPEA (1143mg, 8.85mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (138mg, 0.49mmol) were added to the solution. The temperature was raised to 70°C for 4 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=50/1) to obtain a brown-yellow solid (S)-4-(4-amino-8-(8-iodoimidazo[1,2) -c]pyrimidin-5-yl)-2,8-diazaspiro[4.5]dec-2-yl)benzonitrile (17-3, 98 mg, two-step yield: 87.8%). LCMS: m/z 500.4[M+H] + ; 1 H NMR (400MHz, DMSO-d6), δppm 8.05 (s, 1H), 7.87 (s, 1H), 7.66 (s, 1H), 7.56 (d, J = 8.8Hz, 2H), 6.66 (d, J = 8.8Hz, 2H), 3.70 ~ 3.68 (m, 3H), 3.55 (d, J = 10.4 Hz, 1H), 3.48 ~ 3.45 (m, 2H), 3.38(s,1H), 3.24~3.22(m,4H), 1.96~1.86(m,2H), 1.62~1.56(m,2H).
步骤四:向(S)-4-(4-氨基-8-(8-碘咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸-2-基)苯甲腈(17-3,95mg,0.19mmol)的1,4-二氧六环(15mL)溶液中依次加入DIPEA(123mg,0.28mmol),2-氨基-3-氯吡啶-4-硫醇钠盐(52mg,0.28mmol)和Xantphos(33mg,0.057mmol)。真空氮气置换3次后加入Pd 2(dba) 3(27mg,0.029mmol)。再次真空氮气置换3次。升温到100℃反应4小时。反应液用乙酸乙酯(20mL)稀释,过滤,滤液减压浓缩干,经Prep-HPLC纯化得白色固体((S)-4-(4-氨基-8-(8-(((2-氨基-3-氯吡啶基-4-基)硫基)咪唑基[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸-2-基)苯甲腈甲酸盐(化合物17,22mg,收率:20.0%)。LCMS:m/z 532.6[M+H] +1H NMR(400MHz,DMSO-d6),δppm 8.19(s,1H),8.03(s,1H),7.84(d,J=1.6Hz,1H),7.58~7.54(m,4H),6.65(d,J=8.8Hz,2H),6.33(s,2H),5.80(d,J=5.2Hz,1H),3.90~3.87(m,2H),3.65~3.55(m,4H),3.34~3.32(m,2H),3.30(s,1H),3.16~3.08(m,2H),1.96~1.88(m,2H),1.59~1.56(m,2H)。 Step 4: To (S)-4-(4-amino-8-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2,8-diazaspiro[4.5]dec- 2-yl)benzonitrile (17-3, 95mg, 0.19mmol) in 1,4-dioxane (15mL) solution was added DIPEA (123mg, 0.28mmol), 2-amino-3-chloropyridine- 4-thiol sodium salt (52mg, 0.28mmol) and Xantphos (33mg, 0.057mmol). Pd 2 (dba) 3 (27 mg, 0.029 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again. The temperature was raised to 100°C and reacted for 4 hours. The reaction solution was diluted with ethyl acetate (20mL), filtered, the filtrate was concentrated to dryness under reduced pressure, and purified by Prep-HPLC to obtain a white solid ((S)-4-(4-amino-8-(8-(((2-amino) -3-Chloropyridinyl-4-yl)thio)imidazolyl[1,2-c]pyrimidin-5-yl)-2,8-diazaspiro[4.5]dec-2-yl)benzonitrile Formate (compound 17, 22mg, yield: 20.0%). LCMS: m/z 532.6[M+H] + ; 1 H NMR (400MHz, DMSO-d6), δ ppm 8.19 (s, 1H), 8.03 ( s, 1H), 7.84 (d, J = 1.6 Hz, 1H), 7.58 ~ 7.54 (m, 4H), 6.65 (d, J = 8.8 Hz, 2H), 6.33 (s, 2H), 5.80 (d, J =5.2Hz,1H),3.90~3.87(m,2H), 3.65~3.55(m,4H), 3.34~3.32(m,2H), 3.30(s,1H), 3.16~3.08(m,2H), 1.96~1.88(m,2H), 1.59~1.56(m,2H).
实施例十八:化合物18的合成。Example 18: Synthesis of Compound 18.
(S)-8-(8-((2-氨基-3-氯吡啶-4-基)硫基)咪唑并[1,2-c]嘧啶-5-基)-2-(4-氟苯基)-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(8-((2-Amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-(4-fluorobenzene Yl)-2,8-diazaspiro[4.5]decane 4-amine
Figure PCTCN2020106214-appb-000060
Figure PCTCN2020106214-appb-000060
步骤一:向叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(A1,75mg,0.19mmol)的1,4-二氧六环(15mL)溶液中依次加入Cs 2CO 3(191mg,0.59mmol),1-氟-4-碘苯(87mg,0.39mmol)和Xantphos(24mg,0.041mmol)。真空氮气置换3次后加入Pd 2(dba) 3(19mg,0.021mmol)。再次真空氮气置换3次。升温到110℃反应15小时。反应液用乙酸乙酯(20mL)稀释,过滤,滤液减压浓缩至干,硅胶柱层析纯化(二氯甲烷/甲醇=80/1)得棕黄色固体叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(4-氟苯基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(18-1,92mg,收率:97.2%)。LCMS:m/z 454[M+H] +Step 1: To tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylate ( A1, 75mg, 0.19mmol) in 1,4-dioxane (15mL) solution was sequentially added Cs 2 CO 3 (191mg, 0.59mmol), 1-fluoro-4-iodobenzene (87mg, 0.39mmol) and Xantphos (24mg, 0.041mmol). Pd 2 (dba) 3 (19 mg, 0.021 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again. The temperature was raised to 110°C and reacted for 15 hours. The reaction solution was diluted with ethyl acetate (20mL), filtered, the filtrate was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=80/1) to obtain a brown solid tert-butyl (S)-4-( ((R)-tert-Butylsulfinyl)amino)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5]decane-8-carboxylate tert-butyl ester (18-1, 92mg, yield: 97.2%). LCMS: m/z 454 [M+H] + .
步骤二:向叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(4-氟苯基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(18-1,92mg,0.20mmol)的二氯甲烷(5mL)溶液中滴加HCl/1,4-二氧六环(4.0M,0.6mL)。室温反应1.5小时。反应液减压浓缩至干得棕黄色固体(S)-2-(4-氟苯基)-2,8-二氮杂螺[4.5]癸-4-胺盐酸盐(18-2,粗品,73mg)。LCMS:m/z 250[M+H] +Step 2: To tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5] To a solution of tert-butyl decane-8-carboxylate (18-1, 92 mg, 0.20 mmol) in dichloromethane (5 mL) was added dropwise HCl/1,4-dioxane (4.0M, 0.6 mL). React at room temperature for 1.5 hours. The reaction solution was concentrated under reduced pressure to dryness to obtain a brown solid (S)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5]dec-4-amine hydrochloride (18-2, crude product) , 73mg). LCMS: m/z 250 [M+H] + .
步骤三:向(S)-2-(4-氟苯基)-2,8-二氮杂螺[4.5]癸-4-胺(18-2,73mg,0.20mmol)的异丙醇(12mL)溶液中加入DIPEA(454mg,3.51mmol)和5-氯-8-碘咪唑并[1,2-c]嘧啶(65mg,0.23mmol)。升温至70℃反应4小时。反应液减压浓缩干,Prep-TLC(二氯甲烷/甲醇=15/1)纯化得棕黄色固体(S)-2-(4-氟苯基)-8-(8-碘咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸4-胺(18-3,60mg,两步收率:60.1%)。LCMS:m/z 493[M+H] +Step 3: To (S)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5]dec-4-amine (18-2, 73mg, 0.20mmol) in isopropanol (12mL ) DIPEA (454mg, 3.51mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (65mg, 0.23mmol) were added to the solution. The temperature was raised to 70°C and reacted for 4 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by Prep-TLC (dichloromethane/methanol=15/1) to obtain a brown-yellow solid (S)-2-(4-fluorophenyl)-8-(8-iodoimidazo[1 ,2-c]pyrimidin-5-yl)-2,8-diazaspiro[4.5]decane 4-amine (18-3, 60 mg, two-step yield: 60.1%). LCMS: m/z 493 [M+H] + .
步骤四:向(S)-2-(4-氟苯基)-8-(8-碘咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸4-胺(18-3,30mg,0.061mmol)的1,4-二氧六环(15mL)溶液中依次加入DIPEA(39mg,0.30mmol),2-氨基-3-氯吡啶-4-硫醇钠盐(18mg,0.099mmol)和Xantphos(12mg,0.020mmol)。真空氮气置换3次后加入Pd 2(dba) 3(9mg,0.010mmol)。再次真空氮气置换3次。升温到100℃反应4小时。反应液乙酸乙酯(20mL)稀释,过滤,滤液减压浓缩干,Prep-HPLC纯化得白色固体(S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫基) 咪唑并[1,2-c]嘧啶-5-基)-2-(4-氟苯基)-2,8-二氮杂螺[4.5]癸4-胺(化合物18,2.19mg,收率:6.84%)。LCMS:m/z 525[M+H] +1H NMR(400MHz,MeOD-d4):δppm 8.05(s,1H),7.84(d,J=1.6Hz,1H),7.56(d,J=1.6Hz,1H),7.50(d,J=5.6Hz,1H),6.95~6.90(m,2H),6.57~6.53(m,2H),5.89(d,J=5.6Hz,1H),4.05~3.99(m,2H),3.66~3.57(m,2H),3.43~3.32(m,4H),3.14~3.11(m,1H),2.07~1.99(m,2H),1.76~1.67(m,2H); 19F NMR(376MHz,MeOD-d4):δppm-132.72。 Step 4: To (S)-2-(4-fluorophenyl)-8-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2,8-diazaspiro[4.5 ] Decane 4-amine (18-3, 30mg, 0.061mmol) in 1,4-dioxane (15mL) solution was added DIPEA (39mg, 0.30mmol), 2-amino-3-chloropyridine-4- Sodium mercaptan salt (18mg, 0.099mmol) and Xantphos (12mg, 0.020mmol). Pd 2 (dba) 3 (9 mg, 0.010 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again. The temperature was raised to 100°C and reacted for 4 hours. The reaction solution was diluted with ethyl acetate (20 mL), filtered, and the filtrate was concentrated under reduced pressure to dryness, and purified by Prep-HPLC to obtain a white solid (S)-8-(8-(((2-amino-3-chloropyridin-4-yl) Thio) imidazo[1,2-c]pyrimidin-5-yl)-2-(4-fluorophenyl)-2,8-diazaspiro[4.5]dec4-amine (Compound 18, 2.19mg , Yield: 6.84%). LCMS: m/z 525[M+H] + ; 1 H NMR (400MHz, MeOD-d4): δ ppm 8.05 (s, 1H), 7.84 (d, J = 1.6 Hz, 1H ), 7.56 (d, J = 1.6 Hz, 1H), 7.50 (d, J = 5.6 Hz, 1H), 6.95 ~ 6.90 (m, 2H), 6.57 ~ 6.53 (m, 2H), 5.89 (d, J = 5.6Hz, 1H), 4.05~3.99(m,2H), 3.66~3.57(m,2H), 3.43~3.32(m,4H), 3.14~3.11(m,1H), 2.07~1.99(m,2H) , 1.76 ~ 1.67 (m, 2H); 19 F NMR (376 MHz, MeOD-d4): δ ppm-132.72.
实施例十九:化合物19的合成。Example 19: Synthesis of Compound 19.
(S)-8-(8-((2-氨基-3-氯吡啶-4-基)硫基)咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(8-((2-Amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-(2-chloropyridine -4-yl)-2,8-diazaspiro[4.5]decyl 4-amine
Figure PCTCN2020106214-appb-000061
Figure PCTCN2020106214-appb-000061
步骤一:向叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(A1,70mg,0.19mmol)的1,4-二氧六环(15mL)溶液中依次加入Cs 2CO 3(191mg,0.59mmol),2-氯-4-碘吡啶(94mg,0.39mmol)和Xantphos(24mg,0.041mmol)。真空氮气置换3次后加入Pd 2(dba) 3(19mg,0.021mmol)。再次真空氮气置换3次。升温到110℃反应15小时。反应液用乙酸乙酯(20mL)稀释,过滤,滤液减压浓缩干,硅胶柱层析纯化(二氯甲烷/甲醇=80/1)得棕黄色固体叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(19-1,75mg,收率:81.8%)。LCMS:m/z 471.5[M+H] +1H NMR(400MHz,CDCl 3):δppm 8.00(d,J=6.0Hz,1H),8.40(d,J=6.4Hz,1H),6.33~6.31(m,1H),4.00~3.97(m,2H),3.80~3.74(m,1H),3.72~3.68(m,1H),3.46~3.45(m,1H),3.38~3.33(m,2H),3.24(d,J=10.0Hz,1H),2.96~2.89(m,2H),1.81~1.67(m,3H),1.47(s,9H),1.22(s,9H)。 Step 1: To tert-butyl(S)-4-(((R)-tert-butylsulfinyl)amino)-2,8-diazaspiro[4.5]decane-8-carboxylate ( A1, 70mg, 0.19mmol) in 1,4-dioxane (15mL) solution was sequentially added Cs 2 CO 3 (191mg, 0.59mmol), 2-chloro-4-iodopyridine (94mg, 0.39mmol) and Xantphos (24mg, 0.041mmol). Pd 2 (dba) 3 (19 mg, 0.021 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again. The temperature was raised to 110°C and reacted for 15 hours. The reaction solution was diluted with ethyl acetate (20 mL), filtered, and the filtrate was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=80/1) to obtain a brown solid tert-butyl (S)-4-(( (R)-tert-Butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]decane-8-carboxylic acid tert-butyl ester (19- 1,75mg, yield: 81.8%). LCMS: m/z 471.5[M+H] + ; 1 H NMR (400MHz, CDCl 3 ): δppm 8.00(d,J=6.0Hz,1H), 8.40(d,J=6.4Hz,1H), 6.33~ 6.31(m,1H), 4.00~3.97(m,2H), 3.80~3.74(m,1H), 3.72~3.68(m,1H), 3.46~3.45(m,1H), 3.38~3.33(m,2H) ), 3.24 (d, J = 10.0 Hz, 1H), 2.96 to 2.89 (m, 2H), 1.81 to 1.67 (m, 3H), 1.47 (s, 9H), 1.22 (s, 9H).
步骤二:向叔丁基(S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸烷-8-甲酸叔丁酯(19-1,75mg,0.16mmol)的二氯甲烷(5mL)溶液中加入HCl/1,4-二氧六环(4.0M,0.6mL)。室温反应1.5小时。反应液减压浓缩干得 棕黄色固体(S)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-胺盐酸盐(19-2,粗品,77mg)。Step 2: To tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazepine [4.5] To a solution of tert-butyl decane-8-carboxylate (19-1, 75 mg, 0.16 mmol) in dichloromethane (5 mL) was added HCl/1,4-dioxane (4.0M, 0.6 mL). React at room temperature for 1.5 hours. The reaction solution was concentrated and dried under reduced pressure to obtain a brown-yellow solid (S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-amine hydrochloride (19-2 , Crude product, 77mg).
LCMS:m/z 267.4[M+H] + LCMS: m/z 267.4[M+H] +
步骤三:向(S)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-胺(19-2,75mg,0.16mmol)的异丙醇(12mL)溶液中加入DIPEA(420mg,3.25mmol)和5-氯-8-碘咪唑并[1,2-c]嘧啶(60mg,0.21mmol)。升温至70℃反应4小时。反应液减压浓缩干,经Prep-TLC(二氯甲烷/甲醇=15/1)纯化得棕黄色固体(S)-2-(2-氯吡啶-4-基)-8-(8-碘咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸-4-胺(19-3,46mg,两步收率:56.7%)。LCMS:m/z 510.3[M+H] +1H NMR(400MHz,DMSO-d 6):δppm 8.05(s,1H),7.91(d,J=5.6Hz,1H),7.87(s,1H),7.66(s,1H),6.53(s,2H),3.70~3.38(m,8H),3.27~3.18(m,3H),1.92~1.88(m,2H),1.61~1.54(m,2H)。 Step 3: To (S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-amine (19-2, 75mg, 0.16mmol) in isopropyl DIPEA (420mg, 3.25mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (60mg, 0.21mmol) were added to the alcohol (12mL) solution. The temperature was raised to 70°C and reacted for 4 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by Prep-TLC (dichloromethane/methanol=15/1) to obtain a brown solid (S)-2-(2-chloropyridin-4-yl)-8-(8-iodine) Imidazo[1,2-c]pyrimidin-5-yl)-2,8-diazaspiro[4.5]dec-4-amine (19-3, 46mg, two-step yield: 56.7%). LCMS: m/z 510.3[M+H] + ; 1 H NMR (400MHz, DMSO-d 6 ): δppm 8.05(s, 1H), 7.91(d, J=5.6Hz, 1H), 7.87(s, 1H ),7.66(s,1H),6.53(s,2H),3.70~3.38(m,8H),3.27~3.18(m,3H),1.92~1.88(m,2H),1.61~1.54(m,2H) ).
步骤四:向(S)-2-(2-氯吡啶-4-基)-8-(8-碘咪唑并[1,2-c]嘧啶-5-基)-2,8-二氮杂螺[4.5]癸-4-胺(46mg,0.09mmol)的1,4-二氧六环(15mL)溶液中依次加入DIPEA(59mg,0.46mmol),2-氨基-3-氯吡啶-4-硫醇钠盐(26mg,0.14mmol)和Xantphos(17mg,0.029mmol)。真空氮气置换3次后加入Pd 2(dba) 3(13mg,0.015mmol)。再次真空氮气置换3次。升温到100℃反应4小时。反应液用乙酸乙酯(20mL)稀释,过滤,滤液减压浓缩干,经Prep-HPLC纯化得白色固体(S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸4-胺甲酸盐(化合物19,19.83mg,收率:37.4%)。LCMS:m/z 542.6[M+H] +1H NMR(400MHz,MeOD-d 4):δppm8.42(s,1H),8.06(s,1H),7.92(d,J=6.4Hz,1H),7.84(d,J=5.6Hz,1H),7.57(d,J=1.6Hz,1H),7.50(d,J=5.2Hz,1H),6.63(s,1H),6.59(d,J=5.6Hz,1H),5.89~5.86(m,1H),4.05~3.99(m,2H),3.85~3.81(m,1H),3.64~3.53(m,3H),3.48~3.36(m,3H),2.10~2.00(m,2H),1.81~1.74(m,2H)。 Step 4: To (S)-2-(2-chloropyridin-4-yl)-8-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2,8-diazepine Spiro[4.5]dec-4-amine (46mg, 0.09mmol) in 1,4-dioxane (15mL) solution was added DIPEA (59mg, 0.46mmol), 2-amino-3-chloropyridine-4- Sodium mercaptan salt (26mg, 0.14mmol) and Xantphos (17mg, 0.029mmol). Pd 2 (dba) 3 (13 mg, 0.015 mmol) was added after 3 times of vacuum nitrogen replacement. Vacuum nitrogen replacement 3 times again. The temperature was raised to 100°C and reacted for 4 hours. The reaction solution was diluted with ethyl acetate (20 mL), filtered, and the filtrate was concentrated to dryness under reduced pressure, and purified by Prep-HPLC to obtain a white solid (S)-8-(8-(((2-amino-3-chloropyridine-4- (Yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]deca-4-carboxamide Acid salt (compound 19, 19.83 mg, yield: 37.4%). LCMS: m/z 542.6[M+H] + ; 1 H NMR (400MHz, MeOD-d 4 ): δppm8.42(s, 1H), 8.06(s,1H),7.92(d,J=6.4Hz,1H),7.84(d,J=5.6Hz,1H),7.57(d,J=1.6Hz,1H),7.50(d,J=5.2 Hz,1H),6.63(s,1H),6.59(d,J=5.6Hz,1H),5.89~5.86(m,1H),4.05~3.99(m,2H),3.85~3.81(m,1H) , 3.64~3.53(m,3H), 3.48~3.36(m,3H), 2.10~2.00(m,2H), 1.81~1.74(m,2H).
实施例二十:化合物20的合成。Example 20: Synthesis of Compound 20.
(S)-8-(7-氨基-8-((2-氨基-3-氯吡啶基-4-基)硫基)咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(7-amino-8-((2-amino-3-chloropyridinyl-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-2- (2-Chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec4-amine
Figure PCTCN2020106214-appb-000062
Figure PCTCN2020106214-appb-000062
步骤一:向氯乙醛(40wt.%水溶液,12mL)中加入6-氨基-2-氯-5-碘嘧啶-4-羧酸甲酯(20-1,1.0g,3.19mmol)。加热到80℃反应2小时。反应液冷却至室温后乙酸乙酯(3×80mL)萃取,合并萃取液,水洗(3×80mL),无水硫酸钠干燥,过滤,滤液减压浓缩至干,硅胶色谱法(石油醚/乙酸乙酯=3/1)纯化得淡黄色固体5-氯-8-碘咪唑并[1,2-c]嘧啶-7-羧酸甲酯(20-2,537mg,收率:50%)。 1H NMR(400MHz,CDCl 3):δppm 7.98(d,J=1.6Hz,1H),7.93(d,J=1.6Hz,1H),4.04(s,3H)。 Step 1: Add 6-amino-2-chloro-5-iodopyrimidine-4-carboxylic acid methyl ester (20-1, 1.0 g, 3.19 mmol) to chloroacetaldehyde (40 wt.% aqueous solution, 12 mL). Heat to 80°C to react for 2 hours. After the reaction solution was cooled to room temperature, it was extracted with ethyl acetate (3×80 mL). The extracts were combined, washed with water (3×80 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness under reduced pressure. Ethyl ester = 3/1) was purified to give a pale yellow solid 5-chloro-8-iodoimidazo[1,2-c]pyrimidine-7-carboxylic acid methyl ester (20-2,537 mg, yield: 50%). 1 H NMR (400MHz, CDCl 3 ): δ ppm 7.98 (d, J = 1.6 Hz, 1H), 7.93 (d, J = 1.6 Hz, 1H), 4.04 (s, 3H).
步骤二:向(R)-N-((S)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-基)-2-甲基丙烷-2-亚磺酰胺(130mg,0.35mmol)的异丙醇(20mL)溶液中依次加入DIPEA(135mg,1.05mmol)和5-氯-8-碘咪唑并[1,2-c]嘧啶-7-羧酸甲酯(20-2,132mg,0.39mmol)。真空氮气置换3次后升温到70℃反应3小时。反应液减压浓缩干,硅胶柱层析纯化(二氯甲烷/甲醇=25/1)得浅黄色固体甲基5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸盐(20-3,200mg,收率:84.7%)。LCMS:m/z 672.1[M+H] +Step 2: To (R)-N-((S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methyl Propane-2-sulfinamide (130mg, 0.35mmol) in isopropanol (20mL) was added DIPEA (135mg, 1.05mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine- Methyl 7-carboxylate (20-2, 132 mg, 0.39 mmol). After purging with vacuum nitrogen 3 times, the temperature was raised to 70°C and reacted for 3 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=25/1) to obtain a pale yellow solid methyl 5-((S)-4-(((R)-tert-butylsulfinyl) )Amino)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7 -Carboxylate (20-3,200 mg, yield: 84.7%). LCMS: m/z 672.1 [M+H] + .
步骤三:向甲基5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸盐(20-3,200mg,0.30mmol)的1,4-二氧六环(15mL)溶液中依次加入DIPEA(114mg,0.89mmol),2-氨基-3-氯吡啶-4-硫醇钠盐(108mg,0.60mmol)。真空氮气置换3次后加入Xantphos(34mg,0.06mmol)和Pd 2(dba) 3(55mg,0.06mmol)。再次真空氮气置换3次。升温到110℃反应4小时。 反应液用乙酸乙酯(50mL)稀释,过滤,滤液减压浓缩干,经硅胶柱层析纯化(二氯甲烷/甲醇=15/1)得浅黄色固体甲基8-((2-氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶基-4–基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸盐(20-4,160mg,收率:76.3%)。LCMS:m/z 704.1[M+H] +Step 3: To methyl 5-((S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazepine Heterosspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7-carboxylate (20-3,200mg, 0.30mmol) of 1,4-dioxane (15mL) DIPEA (114mg, 0.89mmol) and 2-amino-3-chloropyridine-4-thiol sodium salt (108mg, 0.60mmol) were added to the solution in sequence. After 3 times of vacuum nitrogen replacement, Xantphos (34 mg, 0.06 mmol) and Pd 2 (dba) 3 (55 mg, 0.06 mmol) were added. Vacuum nitrogen replacement 3 times again. The temperature was raised to 110°C to react for 4 hours. The reaction solution was diluted with ethyl acetate (50 mL), filtered, the filtrate was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=15/1) to obtain a pale yellow solid methyl 8-((2-amino- 3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridinyl-4-yl )-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7-carboxylate (20-4,160mg, yield: 76.3% ). LCMS: m/z 704.1 [M+H] + .
步骤四:向甲基8-((2-氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶基-4–基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸盐(20-4,160mg,0.23mmol)的二氯甲烷(20mL)溶液中加入二碳酸二叔丁酯(250mg,1.14mmol),三乙胺(70mg,0.68mmol)和DMAP(56mg,0.46mmol)。室温反应过夜。反应液减压浓缩干,经硅胶柱层析纯化(二氯甲烷/甲醇=30/1)纯化得棕黄色固体甲基8-((2-叔丁氧羰基氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-二叔丁基亚磺酰基)氨基)-2-(2-氯吡啶基-4–基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸盐(20-5,100mg,收率:48.7%)。Step 4: To methyl 8-((2-amino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butylsulfinyl)amino )-2-(2-Chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7- To a solution of carboxylate (20-4, 160mg, 0.23mmol) in dichloromethane (20mL) was added di-tert-butyl dicarbonate (250mg, 1.14mmol), triethylamine (70mg, 0.68mmol) and DMAP (56mg, 0.46mmol). React overnight at room temperature. The reaction solution was concentrated under reduced pressure to dryness, and purified by silica gel column chromatography (dichloromethane/methanol=30/1) to obtain a brown solid methyl 8-((2-tert-butoxycarbonylamino-3-chloropyridine-4- Yl)thio)-5-((S)-4-(((R)-di-tert-butylsulfinyl)amino)-2-(2-chloropyridyl-4-yl)-2,8- Diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine-7-carboxylate (20-5,100 mg, yield: 48.7%).
LCMS:m/z 905.7[M+H] +LCMS: m/z 905.7 [M+H] + .
步骤五:向甲基8-((2-二叔丁氧羰基氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶基-4–基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸盐(20-5,100mg,0.11mmol)和氢氧化钠(18mg,0.45mmol)的甲醇(15mL)溶液中加入水(3mL)。升温到50℃反应1小时,2M稀盐酸中和,减压浓缩。经硅胶柱层析纯化(二氯甲烷/甲醇=8/1)纯化得黄色固体8-((2-二叔丁氧羰基氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚硫酰基)氨基)-2-(2-氯吡啶基-4-基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸(20-6,90mg,收率:91.8%)。LCMS:m/z 890.3[M+H] +Step 5: To methyl 8-((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butyl Sulfinyl)amino)-2-(2-chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c ] Pyrimidine-7-carboxylate (20-5, 100 mg, 0.11 mmol) and sodium hydroxide (18 mg, 0.45 mmol) in methanol (15 mL) was added with water (3 mL). The temperature was raised to 50°C to react for 1 hour, neutralized with 2M dilute hydrochloric acid, and concentrated under reduced pressure. Purified by silica gel column chromatography (dichloromethane/methanol=8/1) to obtain a yellow solid 8-((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-( (S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8 -Yl)-8-iodoimidazo[1,2-c]pyrimidine-7-carboxylic acid (20-6, 90 mg, yield: 91.8%). LCMS: m/z 890.3 [M+H] + .
步骤六:向8-((2-二叔丁氧羰基氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚硫酰基)氨基)-2-(2-氯吡啶基-4-基)-2,8-二氮杂螺[4.5]癸-8-基)-8-碘咪唑并[1,2-c]嘧啶-7-羧酸(20-6,90mg,0.10mmol)的叔丁醇(20mL)溶液中加入三乙胺(102mg,1.00mmol)和DPPA(83mg,0.30mmol)。真空氮气置换3次后升温到100℃反应3小时。反应液减压浓缩干,硅胶柱层析纯化(二氯甲烷/甲醇=25/1)得浅黄色固体叔丁基(8-(((2-二叔丁氧羰基氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-8-基)咪唑并[1,2-c]嘧啶-7-基)氨基甲酸酯(20-7,70mg,纯度50.6%,收率:40.6%)。LCMS:m/z 862.7[M+H] +Step 6: To 8-((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)-tert-butylsulfinyl) Acyl)amino)-2-(2-chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)-8-iodoimidazo[1,2-c]pyrimidine To a solution of -7-carboxylic acid (20-6, 90 mg, 0.10 mmol) in tert-butanol (20 mL) was added triethylamine (102 mg, 1.00 mmol) and DPPA (83 mg, 0.30 mmol). After purging with vacuum nitrogen 3 times, the temperature was raised to 100°C and reacted for 3 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=25/1) to obtain a pale yellow solid tert-butyl (8-(((2-di-tert-butoxycarbonylamino-3-chloropyridine- 4-yl)thio)-5-((S)-4-(((R)-tert-butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8- Diazaspiro[4.5]dec-8-yl)imidazo[1,2-c]pyrimidin-7-yl)carbamate (20-7,70 mg, purity 50.6%, yield: 40.6%). LCMS: m/z 862.7 [M+H] + .
步骤七:向叔丁基(8-(((2-二叔丁氧羰基氨基-3-氯吡啶-4-基)硫基)-5-((S)-4-(((R)-叔丁基亚磺酰基)氨基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-8-基)咪唑并[1,2-c]嘧啶-7-基)氨基甲酸酯(20-6,70mg,纯度50.6%,0.04mmol)的二氯甲烷(20 mL)溶液中加入HCl/二氧六环(3M,4mL)。室温反应1.5小时。反应液用碳酸氢钠溶液调pH到8,以二氯甲烷/甲醇(10/1)萃取(3x100mL),有机相浓缩后经硅胶柱层析纯化(二氯甲烷/甲醇=7/1)和Prep-HPLC得白色固体(S)-8-(7-氨基-8-((2-氨基-3-氯吡啶-4-基)硫基]咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶基-4-基)-2,8-二氮杂螺[4.5]癸4-胺(化合物20,1mg,收率:4.5%)。LCMS:m/z 557.1[M+H] +Step 7: To tert-butyl (8-(((2-di-tert-butoxycarbonylamino-3-chloropyridin-4-yl)sulfanyl)-5-((S)-4-(((R)- Tert-Butylsulfinyl)amino)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-8-yl)imidazo[1,2-c]pyrimidine -7-yl) carbamate (20-6, 70mg, purity 50.6%, 0.04mmol) in dichloromethane (20 mL) was added HCl/dioxane (3M, 4mL). Reaction at room temperature for 1.5 hours The reaction solution was adjusted to pH 8 with sodium bicarbonate solution, extracted with dichloromethane/methanol (10/1) (3x100mL), the organic phase was concentrated and purified by silica gel column chromatography (dichloromethane/methanol=7/1) And Prep-HPLC to obtain white solid (S)-8-(7-amino-8-((2-amino-3-chloropyridin-4-yl)sulfanyl)imidazo[1,2-c]pyrimidine-5 -Yl)-2-(2-chloropyridyl-4-yl)-2,8-diazaspiro[4.5]dec4-amine (Compound 20, 1 mg, yield: 4.5%). LCMS: m/ z 557.1[M+H] + .
1H NMR(400MHz,MeOD-d 4):δppm 7.88(d,J=6.0Hz,1H),7.52(d,J=5.2Hz,1H),7.48(d,J=1.6Hz,1H),7.32~7.20(m,1H),7.19(d,J=1.6Hz,1H),6.57~6.54(m,2H),5.98(d,J=5.6Hz,1H),3.98~3.93(m,2H),3.73~3.69(m,1H),3.64~3.61(m,1H),3.48~3.35(m,2H),3.23~3.12(m,2H),2.04~1.98(m,2H),1.71~1.63(m,2H)。 1 H NMR (400MHz, MeOD-d 4 ): δppm 7.88 (d, J = 6.0 Hz, 1H), 7.52 (d, J = 5.2 Hz, 1H), 7.48 (d, J = 1.6 Hz, 1H), 7.32 ~7.20(m,1H), 7.19(d,J=1.6Hz,1H), 6.57~6.54(m,2H), 5.98(d,J=5.6Hz,1H), 3.98~3.93(m,2H), 3.73~3.69(m,1H), 3.64~3.61(m,1H), 3.48~3.35(m,2H), 3.23~3.12(m,2H), 2.04~1.98(m,2H), 1.71~1.63(m ,2H).
实施例二十一:化合物21的合成。Example 21: Synthesis of Compound 21.
(S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫基)-7-氯咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸4-胺(S)-8-(8-(((2-Amino-3-chloropyridin-4-yl)thio)-7-chloroimidazo[1,2-c]pyrimidin-5-yl)-2- (2-Chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec4-amine
Figure PCTCN2020106214-appb-000063
Figure PCTCN2020106214-appb-000063
步骤一:向N-((S)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-基)-2-甲基丙烷-2-亚磺酰胺(83mg,0.22mmol)的二氯甲烷(20mL)溶液中依次加入DIPEA(145mg,1.12mmol)和7-氯-8-碘-5-(甲基亚磺酰基)咪唑并[1,2-c]嘧啶(154mg,0.45mmol)的二氯甲烷(3mL)溶液。真空氮气置换3次后室温反应2小时。反应液减压浓缩干,硅胶柱层析纯化(二氯甲烷/甲醇=30/1)得浅黄色固体N-((S)-8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-基)-2-甲基丙烷-2-亚磺酰胺(40mg,收率:27.6%)。LCMS:m/z 648.0[M+H] +Step 1: To N-((S)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methylpropane-2- DIPEA (145mg, 1.12mmol) and 7-chloro-8-iodo-5-(methylsulfinyl) imidazo[1, sulfinamide (83mg, 0.22mmol) in dichloromethane (20mL) solution were added successively 2-c] A solution of pyrimidine (154 mg, 0.45 mmol) in dichloromethane (3 mL). After 3 times of vacuum nitrogen replacement, the reaction was carried out at room temperature for 2 hours. The reaction solution was concentrated to dryness under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol=30/1) to obtain light yellow solid N-((S)-8-(7-chloro-8-iodoimidazo[1,2 -c]pyrimidin-5-yl)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methylpropane-2-ylidene Sulfonamide (40 mg, yield: 27.6%). LCMS: m/z 648.0 [M+H] + .
步骤二:向N-((S)-8-(7-氯-8-碘咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-基)-2-甲基丙烷-2-亚磺酰胺(40mg,0.06mmol)的1,4-二氧六环(15mL)溶液中依次加入DIPEA(0.2mL,1.98mmol),2-氨基-3-氯吡啶-4-硫醇钠盐(18mg, 0.10mmol)。真空氮气置换3次后加入Xantphos(8mg,0.01mmol)和Pd 2(dba) 3(16mg,0.02mmol)。再次真空氮气置换3次。升温到110℃反应16小时。反应液用乙酸乙酯(50mL)稀释,过滤,滤液减压浓缩干,经硅胶柱层析纯化(二氯甲烷/甲醇=10/1)得浅黄色固体N-((S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)-7-氯咪唑[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-基)-2-甲基丙烷-2-亚磺酰胺(22mg,收率:52.4%)。LCMS:m/z 682.1[M+H] +Step 2: To N-((S)-8-(7-chloro-8-iodoimidazo[1,2-c]pyrimidin-5-yl)-2-(2-chloropyridin-4-yl)- 2,8-Diazaspiro[4.5]dec-4-yl)-2-methylpropane-2-sulfenamide (40mg, 0.06mmol) in 1,4-dioxane (15mL) solution DIPEA (0.2 mL, 1.98 mmol), 2-amino-3-chloropyridine-4-thiol sodium salt (18 mg, 0.10 mmol) was added. After 3 times of vacuum nitrogen replacement, Xantphos (8 mg, 0.01 mmol) and Pd 2 (dba) 3 (16 mg, 0.02 mmol) were added. Vacuum nitrogen replacement 3 times again. The temperature was raised to 110°C for 16 hours. The reaction solution was diluted with ethyl acetate (50 mL), filtered, and the filtrate was concentrated under reduced pressure to dryness, and purified by silica gel column chromatography (dichloromethane/methanol=10/1) to obtain a pale yellow solid N-((S)-8-( 8-(((2-Amino-3-chloropyridin-4-yl)thio)-7-chloroimidazole[1,2-c]pyrimidin-5-yl)-2-(2-chloropyridine-4- Yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methylpropane-2-sulfinamide (22mg, yield: 52.4%). LCMS: m/z 682.1[M +H] + .
步骤三:向N-((S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫代)-7-氯咪唑[1,2-c]嘧啶-5-基)-2-(2-氯吡啶-4-基)-2,8-二氮杂螺[4.5]癸-4-基)-2-甲基丙烷-2-亚磺酰胺(22mg,0.03mmol)的二氯甲烷(4mL)溶液中加入HCl/二氧六环(1mL)。室温反应1小时。反应液用碳酸氢钠溶液调pH到8,以二氯甲烷/甲醇(10/1)萃取(3×50mL),有机相浓缩后经硅胶柱层析纯化(二氯甲烷/甲醇=7/1)和Prep-HPLC得白色固体(S)-8-(8-(((2-氨基-3-氯吡啶-4-基)硫基)-7-氯咪唑并[1,2-c]嘧啶-5-基)-2-(2-氯吡啶基-4-基)-2,8-二氮杂螺[4.5]癸-4-胺得甲酸盐(2.0mg,收率:9.9%)。LCMS:m/z 578.0[M+H] +1H NMR(400MHz,MeOD-d 4):δppm 8.43(br,1H),7.92(d,J=6.0Hz,1H),7.84(d,J=1.6Hz,1H),7.54(d,J=1.6Hz,1H),7.53(d,J=5.6Hz,1H),6.62(s,1H),8.59~8.58(m,1H),5.92(d,J=6.0Hz,1H),4.15~4.09(m,2H),3.83~3.79(m,1H),3.65~3.46(m,6H),2.09~1.98(m,2H),1.80~1.73(m,2H)。 Step 3: To N-((S)-8-(8-(((2-amino-3-chloropyridin-4-yl)thio)-7-chloroimidazole[1,2-c]pyrimidine-5 -Yl)-2-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]dec-4-yl)-2-methylpropane-2-sulfenamide (22mg, 0.03 HCl/dioxane (1mL) was added to the dichloromethane (4mL) solution of 1 mmol). Reacted at room temperature for 1 hour. The pH of the reaction solution was adjusted to 8 with sodium bicarbonate solution, and then dichloromethane/methanol (10/1) After extraction (3×50mL), the organic phase was concentrated and purified by silica gel column chromatography (dichloromethane/methanol=7/1) and Prep-HPLC to obtain a white solid (S)-8-(8-(((2-amino -3-Chloropyridin-4-yl)thio)-7-chloroimidazo[1,2-c]pyrimidin-5-yl)-2-(2-chloropyridin-4-yl)-2,8 -Diazaspiro[4.5]dec-4-amine to give the formate (2.0mg, yield: 9.9%). LCMS: m/z 578.0[M+H] + . 1 H NMR (400MHz, MeOD-d 4 ): δppm 8.43 (br, 1H), 7.92 (d, J = 6.0 Hz, 1H), 7.84 (d, J = 1.6 Hz, 1H), 7.54 (d, J = 1.6 Hz, 1H), 7.53 (d ,J=5.6Hz,1H),6.62(s,1H),8.59~8.58(m,1H),5.92(d,J=6.0Hz,1H),4.15~4.09(m,2H),3.83~3.79( m,1H), 3.65~3.46(m,6H), 2.09~1.98(m,2H), 1.80~1.73(m,2H).
实施例二十二:化合物22-29的合成。Example 22: Synthesis of Compound 22-29.
采用与实施例二十一相同的方法,以相应的反应物制备得到下述化合物22-29:Using the same method as in Example 21, the following compounds 22-29 were prepared with corresponding reactants:
Figure PCTCN2020106214-appb-000064
Figure PCTCN2020106214-appb-000064
Figure PCTCN2020106214-appb-000065
Figure PCTCN2020106214-appb-000065
本发明公开化合物的生物学功能在酶活性以及细胞水平的测试中得到了证明。比如在SHP2酶活性抑制试验中,本发明公开的化合物能够与达到很强的抑制活性(IC50可达<10nM)。在细胞水平上,本发明公开的化合物不仅可以抑制下游通路激酶ERK的磷酸化水平而且表现出很好的抑制癌细胞的增殖的活性。与SHP099(6-(4-氨基-4-甲基哌啶-1-基)-3-(2,3-二氯苯基)吡嗪-2-胺)对比,本发明的化合物无论在酶学水平上还是在细胞水平上,都体现出了优越的活性。The biological function of the compound disclosed in the present invention has been proved in the test of enzyme activity and cell level. For example, in the SHP2 enzyme activity inhibition test, the compound disclosed in the present invention can achieve strong inhibitory activity (IC50 up to <10 nM). At the cellular level, the compound disclosed in the present invention can not only inhibit the phosphorylation level of downstream pathway kinase ERK, but also exhibit a good activity of inhibiting the proliferation of cancer cells. Compared with SHP099 (6-(4-amino-4-methylpiperidin-1-yl)-3-(2,3-dichlorophenyl)pyrazine-2-amine), the compound of the present invention is no matter in the enzyme Both on the scientific level and on the cellular level have demonstrated superior activity.
测试例一:SHP2酶活测试Test case 1: SHP2 enzyme activity test
化合物粉末溶于DMSO中制成母液。实验时,化合物存贮液用DMSO进行3-倍梯度稀释,同一化合物设置10个不同的测试浓度。取1μL各浓度点的化合物至检测板(Corning,Costar 3915)孔内,每个浓度点设置2个平行重复。以6,8-二氟-4-甲基-7-羟基香豆素磷酸酯(DiFMUP)作为底物,SHP2 E72A催化其水解产生6,8-二氟-4-甲基-7-羟基香豆素(DiFMU),通过PE Enspire多功能读数仪,以358nm为激发波长来检测455nm处的荧光值,确定SHP2的酶活。The compound powder is dissolved in DMSO to make a mother liquor. In the experiment, the compound stock solution was diluted with DMSO in 3-fold gradient, and the same compound was set to 10 different test concentrations. Take 1 μL of the compound at each concentration point into the well of the detection plate (Corning, Costar 3915), and set 2 parallel replicates for each concentration point. Using 6,8-difluoro-4-methyl-7-hydroxycoumarin phosphate (DiFMUP) as a substrate, SHP2 E72A catalyzes its hydrolysis to produce 6,8-difluoro-4-methyl-7-hydroxy Bean (DiFMU), through the PE Enspire multi-function reader, use 358nm as the excitation wavelength to detect the fluorescence value at 455nm to determine the enzyme activity of SHP2.
反应用的SHP2缓冲液的组成为60mmol/L Hepes,PH7.2,75mmol/L NaCl,75mmol/L KCl,1mmol/L EDTA,5mmol/L DTT。筛选体系组成为:SHP2缓冲液、酶SHP2 E76A蛋白、底物DiFMUP和待测化合物。The composition of the SHP2 buffer used in the reaction is 60mmol/L Hepes, PH7.2, 75mmol/L NaCl, 75mmol/L KCl, 1mmol/L EDTA, 5mmol/L DTT. The screening system consists of: SHP2 buffer, enzyme SHP2 E76A protein, substrate DiFMUP and test compound.
IC 50测试方法: IC 50 test method:
96孔筛选板中50ng SHP2 E76A蛋白分别与待测化合物在SHP2缓冲液中反应20min,然后与10uM DiFMUP室温共同孵育20min,用PE Enspire多功能读数仪以358nm为激发光,读取455nm处的光强度。以化合物处理组测出的荧光值比DMSO对照孔的值计算样品对酶活性的抑制率。化合物的IC 50值由Graphpad公司的Prism软件,以抑制率对 抑制剂的浓度非线性拟合计算得到。通过Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))方程拟合出酶活性随化合物浓度变化的曲线。求得各化合物的IC 50值。下表1显示了本发明部分化合物的IC 50值。其中,字母A代表IC 50小于50nM;字母B代表IC 50为50nM至500nM。 In a 96-well screening plate, 50ng SHP2 E76A protein was reacted with the test compound in SHP2 buffer for 20 minutes, and then incubated with 10uM DiFMUP for 20 minutes at room temperature. The PE Enspire multi-function reader was used to excite light at 358nm and read the light at 455nm. strength. The fluorescence value measured by the compound treatment group is compared with the value of the DMSO control well to calculate the inhibition rate of the sample on the enzyme activity. The IC 50 value of the compound was calculated by using the Prism software of Graphpad Company to fit the inhibition rate to the concentration of the inhibitor nonlinearly. The curve of enzyme activity varying with compound concentration was fitted by the equation Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope)). The IC 50 value of each compound was obtained. Table 1 below shows the IC 50 values of some compounds of the present invention. Among them, the letter A represents that the IC 50 is less than 50 nM; the letter B represents that the IC 50 is 50 nM to 500 nM.
表1Table 1
化合物编号Compound number 酶活性(nM)Enzyme activity (nM) 化合物编号Compound number 酶活性(nM)Enzyme activity (nM)
SHP099SHP099 263263 化合物15Compound 15 AA
化合物1Compound 1 AA 化合物16Compound 16 AA
化合物2Compound 2 AA 化合物17Compound 17 AA
化合物3Compound 3 AA 化合物18Compound 18 AA
化合物4Compound 4 AA 化合物19Compound 19 AA
化合物5Compound 5 AA 化合物20Compound 20 AA
化合物6Compound 6 AA 化合物21Compound 21 AA
化合物7Compound 7 BB 化合物22Compound 22 AA
化合物8Compound 8 AA 化合物23Compound 23 AA
化合物9Compound 9 AA 化合物24Compound 24 AA
化合物10Compound 10 AA 化合物25Compound 25 AA
化合物11Compound 11 BB 化合物26Compound 26 AA
化合物12Compound 12 BB 化合物27Compound 27 AA
化合物13Compound 13 AA 化合物28Compound 28 AA
化合物14Compound 14 AA 化合物29Compound 29 AA
测试例二:MV4-11细胞增殖抑制实验Test Example 2: MV4-11 cell proliferation inhibition experiment
通过
Figure PCTCN2020106214-appb-000066
发光法细胞活力检测试剂盒对细胞内ATP进行定量测定来检测培养物中活细胞数目。
by
Figure PCTCN2020106214-appb-000066
The luminescence cell viability detection kit quantitatively measures intracellular ATP to detect the number of viable cells in culture.
第一步:在96孔板种接种MV4-11细胞,以每孔2500个细胞密度接种细胞到96孔板,每孔体积100μL。置于37℃、5%二氧化碳培养箱培养过夜。The first step: Seed MV4-11 cells in a 96-well plate, seed the cells into a 96-well plate at a density of 2500 cells per well, with a volume of 100 μL per well. Place it in a 37°C, 5% carbon dioxide incubator overnight.
第二步:化合物处理细胞。待测化合物进行3倍稀释,共设置8个浓度梯度;每孔分别加入一定体积的DMSO或者待测化合物,每个浓度设置2个重复,DMSO的终浓度控制在0.5%。置于37℃5%二氧化碳培养箱培养72h。Step 2: Compound treatment of cells. The test compound was diluted 3 times, and a total of 8 concentration gradients were set; a certain volume of DMSO or test compound was added to each well, and each concentration was set to 2 replicates, and the final concentration of DMSO was controlled at 0.5%. Place in a 37°C 5% carbon dioxide incubator for 72h.
第三步:使用
Figure PCTCN2020106214-appb-000067
Luminescent Cell Viability Assay试剂盒(Promega,G7570) 检测对照组和处理组细胞活力。每孔加入50ul CellTiter-Glo,混匀,室温孵育10min。使用EnSpire(Perkin Elmer)读取信号。抑制百分率(%)通过以下公式计算获得:
Step 3: Use
Figure PCTCN2020106214-appb-000067
Luminescent Cell Viability Assay Kit (Promega, G7570) detects the cell viability of the control and treatment groups. Add 50ul CellTiter-Glo to each well, mix well, and incubate at room temperature for 10 minutes. Use EnSpire (Perkin Elmer) to read the signal. The inhibition percentage (%) is calculated by the following formula:
抑制百分率(%)=(1-化合物处理组信号值/DMSO处理组信号值)*100。Inhibition percentage (%)=(1-compound treatment group signal value/DMSO treatment group signal value)*100.
结果示于表2中。其中,字母A代表IC 50小于100nM;字母B代表IC 50为100nM至500nM。 The results are shown in Table 2. Among them, the letter A represents that the IC 50 is less than 100 nM; the letter B represents that the IC 50 is 100 nM to 500 nM.
表2:化合物对MV411增殖抑制活性Table 2: Compounds inhibiting the proliferation of MV411
Figure PCTCN2020106214-appb-000068
Figure PCTCN2020106214-appb-000068
测试例三:KYSE520细胞增殖抑制实验Test Example 3: KYSE520 cell proliferation inhibition experiment
通过CellTiter-Glo R发光法细胞活力检测试剂盒对细胞内ATP进行定量测定来检测培养物中活细胞数目。 CellTiter-Glo R luminescence cell viability detection kit is used to quantitatively determine intracellular ATP to detect the number of viable cells in culture.
第一步:在96孔板种接种KYSE520细胞,以每孔1500个细胞密度接种细胞到96孔板,每孔体积100μL。置于37℃、5%二氧化碳培养箱培养过夜。The first step: Seed KYSE520 cells in a 96-well plate, seed the cells in a 96-well plate at a density of 1500 cells per well, with a volume of 100 μL per well. Place it in a 37°C, 5% carbon dioxide incubator overnight.
第二步:化合物处理细胞。待测化合物进行3倍稀释,共设置8个浓度梯度;每孔分别加入一定体积的DMSO或者待测化合物,每个浓度设置2个重复,DMSO的终浓度控制在0.5%。置于37℃5%二氧化碳培养箱培养72h。Step 2: Compound treatment of cells. The test compound was diluted 3 times, and a total of 8 concentration gradients were set; a certain volume of DMSO or test compound was added to each well, and each concentration was set to 2 replicates, and the final concentration of DMSO was controlled at 0.5%. Place in a 37°C 5% carbon dioxide incubator for 72 hours.
第三步:使用CellTiter-Glo R Luminescent Cell Viability Assay试剂盒(Promega,G7570)检测对照组和处理组细胞活力。每孔加入50ul CellTiter-Glo,混匀,室温孵育10min。使用EnSpire(Perkin Elmer)读取信号。抑制百分率(%)通过以下公式计算获得:抑制百分率(%)=(1-化合物处理组信号值/DMSO处理组信号值)*100。结果如下表3所示。 The third step: use CellTiter-Glo R Luminescent Cell Viability Assay Kit (Promega, G7570) to detect the cell viability of the control group and the treatment group. Add 50ul CellTiter-Glo to each well, mix well, and incubate at room temperature for 10 minutes. Use EnSpire (Perkin Elmer) to read the signal. The percentage inhibition (%) is calculated by the following formula: percentage inhibition (%)=(1-compound treatment group signal value/DMSO treatment group signal value)*100. The results are shown in Table 3 below.
其中,字母A代表IC 50小于1000nM;字母B代表IC 50为1000nM至2000nM; Among them, the letter A represents that the IC 50 is less than 1000 nM; the letter B represents that the IC 50 is between 1000 nM and 2000 nM;
表3table 3
化合物编号Compound number KYSE520细胞增殖(nM)KYSE520 cell proliferation (nM)
SHP099SHP099 68696869
化合物7Compound 7 BB
化合物8Compound 8 AA
化合物10Compound 10 AA
化合物16Compound 16 AA
化合物17Compound 17 BB
化合物18Compound 18 AA
化合物19Compound 19 AA
化合物20Compound 20 AA
化合物21Compound 21 AA
测试例四:化合物药代动力学实验Test Example 4: Compound Pharmacokinetic Experiment
采用以下方法测定本申请的化合物药代动力学参数。研究使用的健康雄性成年大鼠/小鼠,每组动物单次灌胃给药5-100mg/Kg。禁食从给药前10小时至给药后4小时。给药后不同时间点后采血,并测定化合物血浆含量(LC-MS/MS)。用专业软件分析(winnonlin)血浆浓度----时间关系,计算化合物的药代动力学参数。结果显示,本发明化合物有着优异的药代动力学性质。The following methods were used to determine the pharmacokinetic parameters of the compounds of this application. The healthy male adult rats/mice used in the study were given a single intragastric administration of 5-100 mg/Kg for each group of animals. Fasting is from 10 hours before administration to 4 hours after administration. Blood was collected at different time points after administration, and the plasma content of the compound was determined (LC-MS/MS). Use professional software to analyze (winnonlin) plasma concentration-time relationship and calculate the pharmacokinetic parameters of the compound. The results show that the compound of the present invention has excellent pharmacokinetic properties.
在本发明中提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。All documents mentioned in the present invention are cited as references in this application, as if each document is individually cited as a reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art can make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of the present application.

Claims (12)

  1. 一种式I所示的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物:A compound represented by Formula I, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof:
    Figure PCTCN2020106214-appb-100001
    Figure PCTCN2020106214-appb-100001
    其中,among them,
    R 1选自H、-卤素、-CN、-OH、-NO 2、HSO 3-、未取代或取代的C1-C6烷基磺酰基、未取代或取代的C1-C6烷基羧基、未取代或取代的C1-C6烷基氨基、未取代或取代的C1-C6烷基、未取代或取代的C1-C6烷氧基、未取代或取代的C1-6烷氧基羰基、未取代或取代的C1-6烷基羰基、未取代或取代的C2-C6烯烃基羰基、未取代或取代的C1-C6烷氧基-O-C1-C6烷基、未取代或取代的3-8元环烷基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;所述的杂环基或杂芳基包含1-4个选自下组的杂原子:N、O或S; R 1 is selected from H, -halogen, -CN, -OH, -NO 2 , HSO 3 -, unsubstituted or substituted C1-C6 alkylsulfonyl, unsubstituted or substituted C1-C6 alkylcarboxy, unsubstituted Or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted C1-C6 alkoxy, unsubstituted or substituted C1-6 alkoxycarbonyl, unsubstituted or substituted C1-6 alkyl carbonyl group, unsubstituted or substituted C2-C6 alkenyl carbonyl group, unsubstituted or substituted C1-C6 alkoxy-O-C1-C6 alkyl group, unsubstituted or substituted 3-8 membered ring Alkyl group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 membered heteroaryl group; the heterocyclic group or heteroaryl group The group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
    R 2选自H、-卤素、氨基、未取代或取代的C1-C6烷基氨基、未取代或取代的C1-C6烷基; R 2 is selected from H, -halogen, amino, unsubstituted or substituted C1-C6 alkylamino, unsubstituted or substituted C1-C6 alkyl;
    R 3选自H、取代或未取代的3-8元环烷基、取代或未取代的3-8元杂环基、取代或未取代的5-10元芳基、取代或未取代的5-10元杂芳基;所述的杂环基或杂芳基包含1-4个选自下组的杂原子:N、O或S; R 3 is selected from H, substituted or unsubstituted 3-8 membered cycloalkyl, substituted or unsubstituted 3-8 membered heterocyclic group, substituted or unsubstituted 5-10 membered aryl, substituted or unsubstituted 5 -10 membered heteroaryl; said heterocyclic group or heteroaryl group contains 1-4 heteroatoms selected from the group consisting of N, O or S;
    X和Y分别独立地选自N或CR 4X and Y are each independently selected from N or CR 4 ;
    R 4选自H、卤素、-CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的氨基、(取代或未取代的C1-C6烷基)SO-、(取代或未取代的C1-C6烷基)SO 2-; R 4 is selected from H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, (substituted or unsubstituted C1-C6 Alkyl)SO-, (substituted or unsubstituted C1-C6 alkyl)SO 2 -;
    或者R 4与邻近的
    Figure PCTCN2020106214-appb-100002
    共同形成一个取代或未取代的5-10元芳环,取代或未取代的5-10元杂芳环,取代或未取代的5-10元杂环基或取代或未取代的5-10元碳环基;所述的环包含0-4个选自下组的杂原子:N、O或S;条件是,此时X和Y不能同时为N;
    Or R 4 and adjacent
    Figure PCTCN2020106214-appb-100002
    Together to form a substituted or unsubstituted 5-10 membered aromatic ring, substituted or unsubstituted 5-10 membered heteroaromatic ring, substituted or unsubstituted 5-10 membered heterocyclic group or substituted or unsubstituted 5-10 membered ring Carbocyclyl; the ring contains 0-4 heteroatoms selected from the group consisting of N, O or S; the condition is that X and Y cannot be N at the same time;
    Figure PCTCN2020106214-appb-100003
    表示单键或双键;
    Figure PCTCN2020106214-appb-100003
    Represents a single bond or a double bond;
    上述的任一“取代”指基团上的一个或多个氢原子被选自下组的取代基取代:-D、卤素、-OH、-NO 2、-NH 2、-NH(未取代或卤代的C1-C6烷基)、-N(未取代或卤代的C1-C6烷基) 2、-CN、未取代或卤代的C1-C8烷基、未取代或卤代的C1-C8烷氧基、未取代或卤 代的C1-C8烷氧基-C1-C8烷基、未取代或卤代的C3-C8环烷基、未取代或卤代的C3-C8环烷基-C1-C8烷基、未取代或卤代的C1-C6烷基羰基、未取代或卤代的C1-C6烷氧基羰基、异羟肟酸基、未取代或卤代的C1-C6烷基巯基、-S(O) 2N(未取代或卤代的C1-C6烷基) 2、-S(O) 2未取代或卤代的C1-C6烷基、-N(未取代或卤代的C1-C6烷基)S(O) 2N(未取代或卤代的C1-C6烷基) 2、-S(O)N(未取代或卤代的C1-C6烷基) 2、-S(O)(未取代或卤代的C1-C6烷基)、-NH(未取代或卤代的C1-C6烷基)S(O)N(未取代或卤代的C1-C6烷基) 2、-N(未取代或卤代的C1-C6烷基) 2S(O)(未取代或卤代的C1-C6烷基)、未取代或卤代的5-8元芳基、未取代或卤代的5-8元杂芳基、未取代或卤代的4-8元饱和杂环、未取代或卤代的4-8元饱和碳环;其中,所述的杂芳基或饱和杂环包含1-4个选自下组的杂原子:N、O或S。 Any of the above "substituted" means that one or more hydrogen atoms on the group are replaced by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -NH (unsubstituted or Halogenated C1-C6 alkyl), -N (unsubstituted or halogenated C1-C6 alkyl) 2 , -CN, unsubstituted or halogenated C1-C8 alkyl, unsubstituted or halogenated C1- C8 alkoxy, unsubstituted or halogenated C1-C8 alkoxy-C1-C8 alkyl, unsubstituted or halogenated C3-C8 cycloalkyl, unsubstituted or halogenated C3-C8 cycloalkyl- C1-C8 alkyl, unsubstituted or halogenated C1-C6 alkylcarbonyl, unsubstituted or halogenated C1-C6 alkoxycarbonyl, hydroxamic acid group, unsubstituted or halogenated C1-C6 alkyl Mercapto, -S(O) 2 N (unsubstituted or halogenated C1-C6 alkyl) 2 , -S(O) 2 unsubstituted or halogenated C1-C6 alkyl, -N (unsubstituted or halogenated C1-C6 alkyl) S(O) 2 N (unsubstituted or halogenated C1-C6 alkyl) 2 , -S(O)N (unsubstituted or halogenated C1-C6 alkyl) 2 ,- S(O) (unsubstituted or halogenated C1-C6 alkyl), -NH (unsubstituted or halogenated C1-C6 alkyl) S(O)N (unsubstituted or halogenated C1-C6 alkyl) ) 2 , -N (unsubstituted or halogenated C1-C6 alkyl) 2 S(O) (unsubstituted or halogenated C1-C6 alkyl), unsubstituted or halogenated 5-8 membered aryl, Unsubstituted or halogenated 5-8 membered heteroaryl, unsubstituted or halogenated 4-8 membered saturated heterocyclic ring, unsubstituted or halogenated 4-8 membered saturated carbocyclic ring; wherein, the heteroaryl group Or the saturated heterocyclic ring contains 1-4 heteroatoms selected from the group consisting of N, O or S.
  2. 如权利要求1所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物如下述式II所示:The compound of claim 1, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof is represented by the following formula II:
    Figure PCTCN2020106214-appb-100004
    Figure PCTCN2020106214-appb-100004
    其中,among them,
    X和Y分别独立地选自N或C;X and Y are each independently selected from N or C;
    R 4a和R 4b分别独立地选自无、H、卤素、-CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的氨基、(取代或未取代的C1-C6烷基)SO-、(取代或未取代的C1-C6烷基)SO 2-; R 4a and R 4b are each independently selected from none, H, halogen, -CN, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted amino, ( Substituted or unsubstituted C1-C6 alkyl) SO-, (substituted or unsubstituted C1-C6 alkyl) SO 2 -;
    其他取代基如权利要求1所述。Other substituents are as described in claim 1.
  3. 如权利要求1所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物如下述式III所示:The compound of claim 1, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof is represented by the following formula III:
    Figure PCTCN2020106214-appb-100005
    Figure PCTCN2020106214-appb-100005
    其中,among them,
    X和Y分别独立地选自N或C;X和Y不能同时为N;X and Y are independently selected from N or C; X and Y cannot be N at the same time;
    所述的环A为取代或未取代的5-10元芳环,取代或未取代的5-10元杂芳环,取代或未取代的5-10元杂环基,或取代或未取代的5-10元碳环基;所述的杂芳环或杂环基包含0-4个选自下组的杂原子:N、O或S;The ring A is a substituted or unsubstituted 5-10 membered aromatic ring, a substituted or unsubstituted 5-10 membered heteroaromatic ring, a substituted or unsubstituted 5-10 membered heterocyclic group, or a substituted or unsubstituted 5-10 membered carbocyclic group; the heteroaromatic ring or heterocyclic group contains 0-4 heteroatoms selected from the group consisting of N, O or S;
    上述的“取代”指环A上的一个或多个氢原子被选自下组的取代基取代:-D、卤素、-OH、-NO 2、-NH 2、-NH(未取代或卤代的C1-C6烷基)、-N(未取代或卤代的C1-C6烷基) 2、-CN、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 The above-mentioned "substituted" means that one or more hydrogen atoms on ring A are substituted by a substituent selected from the following group: -D, halogen, -OH, -NO 2 , -NH 2 , -NH (unsubstituted or halogenated C1-C6 alkyl), -N (unsubstituted or halogenated C1-C6 alkyl) 2 , -CN, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy Group, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -.
    其他取代基如权利要求1所述。Other substituents are as described in claim 1.
  4. 如权利要求3所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,其特征在于,所述的环A选自取代或未取代的三元(杂)环、取代或未取代的四元(杂)环、取代或未取代的五元(杂)环、取代或未取代的六元(杂)环、取代或未取代的五元芳(杂)环、取代或未取代的六元芳(杂)环、取代或未取代的七元芳(杂)环、取代或未取代的五元并六元(杂)环、取代或未取代的六元并六元(杂)环;所述的杂环包含1-4个选自下组的杂原子:N、O或S;所述环A被取代时,取代基选自:卤素、-CN、-OH、-NH 2、(卤代或未取代的C1-C6烷基) 2N-、卤代或未取代的C1-C6烷基、卤代或未取代的C1-C6烷氧基、C1-C6烷基氨基、(卤代或未取代的C1-C6烷基)SO-、(卤代或未取代的C1-C6烷基)SO 2-。 The compound according to claim 3, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug, or metabolite thereof, wherein the ring A is selected from substituted Or unsubstituted three-membered (hetero) ring, substituted or unsubstituted four-membered (hetero) ring, substituted or unsubstituted five-membered (hetero) ring, substituted or unsubstituted six-membered (hetero) ring, substituted or unsubstituted Substituted five-membered aromatic (hetero) ring, substituted or unsubstituted six-membered aromatic (hetero) ring, substituted or unsubstituted seven-membered aromatic (hetero) ring, substituted or unsubstituted five-membered and six-membered (hetero) ring , Substituted or unsubstituted six-membered and six-membered (hetero) ring; the heterocyclic ring contains 1-4 heteroatoms selected from the group consisting of N, O or S; when the ring A is substituted, the substituent Selected from: halogen, -CN, -OH, -NH 2 , (halogenated or unsubstituted C1-C6 alkyl) 2 N-, halogenated or unsubstituted C1-C6 alkyl, halogenated or unsubstituted C1-C6 alkoxy, C1-C6 alkylamino, (halogenated or unsubstituted C1-C6 alkyl) SO-, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -.
  5. 如权利要求1-4中任一项所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,其特征在于,R 1选自H、卤代或未取代的C1-C6烷基羰基、卤代或未取代的C2-C6烯烃基羰基、(卤代或未取代的C1-C6烷基)SO 2-、取代或未取代的苯磺酰基、未取代或取代的3-8元碳环基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;其中,所述苯磺酰基、碳环基、杂环基、芳基和杂芳基被取代时,取代基的个数为1-4个,选自H、NH 2、卤素、卤代或未取代的C1-C8环烷基、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-。 The compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, wherein R 1 Selected from H, halogenated or unsubstituted C1-C6 alkylcarbonyl, halogenated or unsubstituted C2-C6 alkenylcarbonyl, (halogenated or unsubstituted C1-C6 alkyl) SO 2 -, substituted or unsubstituted Substituted benzenesulfonyl group, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5 -10 membered heteroaryl group; wherein, when the benzenesulfonyl group, carbocyclic group, heterocyclic group, aryl group and heteroaryl group are substituted, the number of substituents is 1-4, selected from H, NH 2 , Halogen, halogenated or unsubstituted C1-C8 cycloalkyl, unsubstituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N-.
  6. 如权利要求1-4中任一项所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,其特征在于,R 3选自H、未取代或取代的3-8元碳环基、未取代或取代的3-8元杂环基、未取代或取代的5-10元芳基、未取代或取代的5-10元杂芳基;其中,所述碳环基、杂环基、芳基和杂芳基被取代时,取代基的个数为1-4个,取代基选自H、NH 2、卤素、未取代或卤代的C1-C6烷基、(卤代或未取代的C1-C6烷基) 2N-和卤代或未取代的C1-C6烷基-NH-。 The compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof, wherein R 3 Selected from H, unsubstituted or substituted 3-8 membered carbocyclic group, unsubstituted or substituted 3-8 membered heterocyclic group, unsubstituted or substituted 5-10 membered aryl group, unsubstituted or substituted 5-10 Membered heteroaryl group; wherein, when the carbocyclic group, heterocyclic group, aryl group and heteroaryl group are substituted, the number of substituents is 1-4, and the substituents are selected from H, NH 2 , halogen, and Substituted or halogenated C1-C6 alkyl, (halogenated or unsubstituted C1-C6 alkyl) 2 N- and halogenated or unsubstituted C1-C6 alkyl -NH-.
  7. 如权利要求3所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,其特征在于,所述式III中,The compound of claim 3, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug, or metabolite thereof, wherein in the formula III,
    R 1选自任选卤素的取代基取代的5-10元杂芳基,优选为5-10元含氮杂芳基,更优选为吡啶基; R 1 is selected from 5-10 membered heteroaryl groups substituted with optionally halogen substituents, preferably 5-10 membered nitrogen-containing heteroaryl groups, more preferably pyridyl;
    R 2选自H和氨基; R 2 is selected from H and amino;
    R 3选自任选被1或2个选自NH 2和卤素的取代基取代的5-10元杂芳基,优选为5-10元含氮杂芳基,更优选为吡啶基; R 3 is selected from 5-10 membered heteroaryl groups optionally substituted by 1 or 2 substituents selected from NH 2 and halogen, preferably 5-10 membered nitrogen-containing heteroaryl groups, more preferably pyridyl;
    环A为:Ring A is:
    Figure PCTCN2020106214-appb-100006
    Figure PCTCN2020106214-appb-100006
    其中,X为N,Y为C。Among them, X is N and Y is C.
  8. 如权利要求1所述的化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物,其特征在于所述的化合物选自如下:The compound of claim 1, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug, or metabolite thereof, characterized in that the compound is selected from the following:
    Figure PCTCN2020106214-appb-100007
    Figure PCTCN2020106214-appb-100007
    Figure PCTCN2020106214-appb-100008
    Figure PCTCN2020106214-appb-100008
  9. 如权利要求1-8中任一项所述的式I化合物的用途,其特征在于,用于:The use of the compound of formula I according to any one of claims 1-8, characterized in that it is used for:
    (a)制备预防或治疗与SHP2活性异常相关的疾病或病症的药物;(a) Preparation of drugs for the prevention or treatment of diseases or disorders related to abnormal SHP2 activity;
    (b)制备预防或治疗SHP2-介导的疾病或病症的药物(b) Preparation of drugs for the prevention or treatment of SHP2-mediated diseases or conditions
    (c)制备抑制SHP2活性的抑制剂药物;(c) Preparation of inhibitor drugs that inhibit SHP2 activity;
    (d)体外非治疗性的抑制SHP2活性;(d) Non-therapeutic inhibition of SHP2 activity in vitro;
    (e)体外非治疗性的抑制肿瘤细胞增殖;和/或(e) Non-therapeutic inhibition of tumor cell proliferation in vitro; and/or
    (f)治疗与SHP2异常相关的疾病或病症。(f) Treatment of diseases or conditions related to abnormal SHP2.
  10. 如权利要求9所述的用途,其特征在于,所述的疾病为癌症,包括但不限于努南综合症、豹综合症、青少年髓单核细胞白血病、成神经细胞瘤、黑色素瘤、急性髓性白血病、乳腺癌、食道癌、肺癌、结肠癌、头癌、成神经细胞瘤、头颈的鳞状细胞癌、胃癌、间变性大细胞淋巴瘤或成胶质细胞瘤。The use according to claim 9, wherein the disease is cancer, including but not limited to Noonan syndrome, Leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myeloid Leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large cell lymphoma or glioblastoma.
  11. 一种药物组合物,其特征在于,所述的药物组合物包括:A pharmaceutical composition, characterized in that the pharmaceutical composition comprises:
    (i)有效量的权利要求1-8中任一项所述的式I化合物,或其药学上可接受的盐,或其溶剂化物、同位素取代物、多晶型物、前药或代谢产物;和(i) An effective amount of the compound of formula I according to any one of claims 1-8, or a pharmaceutically acceptable salt thereof, or a solvate, isotope substitution, polymorph, prodrug or metabolite thereof ;with
    (ii)药学上可接受的载体。(ii) A pharmaceutically acceptable carrier.
  12. 一种抑制SHP2活性的方法,其特征在于,包括如下步骤:对抑制对象施用抑制有效量的如权利要求1-8中任一项所述的式I化合物或其要学上可接受的盐,或对抑制对象施用有效量的如权利要求11所述的药物组合物。A method for inhibiting the activity of SHP2, which is characterized by comprising the steps of: administering an inhibitory effective amount of the compound of formula I according to any one of claims 1 to 8 or an acceptable salt thereof, Or administer an effective amount of the pharmaceutical composition according to claim 11 to a subject to be inhibited.
PCT/CN2020/106214 2019-08-01 2020-07-31 Spiroaromatic compound, preparation and application thereof WO2021018287A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080053803.7A CN114269746A (en) 2019-08-01 2020-07-31 Spiro aromatic ring compound, preparation and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910705467.9 2019-08-01
CN201910705467.9A CN112300160A (en) 2019-08-01 2019-08-01 Spiro aromatic ring compound, preparation and application thereof

Publications (1)

Publication Number Publication Date
WO2021018287A1 true WO2021018287A1 (en) 2021-02-04

Family

ID=74229248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/106214 WO2021018287A1 (en) 2019-08-01 2020-07-31 Spiroaromatic compound, preparation and application thereof

Country Status (2)

Country Link
CN (2) CN112300160A (en)
WO (1) WO2021018287A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11179397B2 (en) 2018-10-03 2021-11-23 Gilead Sciences, Inc. Imidazopyrimidine derivatives
WO2021257736A1 (en) 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
WO2022060583A1 (en) 2020-09-03 2022-03-24 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
WO2022060836A1 (en) 2020-09-15 2022-03-24 Revolution Medicines, Inc. Indole derivatives as ras inhibitors in the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022259157A1 (en) 2021-06-09 2022-12-15 Novartis Ag A triple pharmaceutical combination comprising dabrafenib, trametinib and a shp2 inhibitor
WO2022269525A1 (en) 2021-06-23 2022-12-29 Novartis Ag Pharmaceutical combinations comprising a kras g12c inhibitor and uses thereof for the treatment of cancers
WO2023031781A1 (en) 2021-09-01 2023-03-09 Novartis Ag Pharmaceutical combinations comprising a tead inhibitor and uses thereof for the treatment of cancers
US20230102398A1 (en) * 2021-09-27 2023-03-30 New York University Therapeutic targeting of kmt2d mutant lung squamous cell carcinoma through rtk-ras signaling inhibition
WO2023060253A1 (en) 2021-10-08 2023-04-13 Revolution Medicines, Inc. Ras inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112300160A (en) * 2019-08-01 2021-02-02 上海奕拓医药科技有限责任公司 Spiro aromatic ring compound, preparation and application thereof
EP4328228A1 (en) * 2021-05-13 2024-02-28 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Heterocyclic compound for inhibiting shp2 activity, preparation method therefor and use thereof
CN115521305A (en) * 2022-09-20 2022-12-27 中国药科大学 SHP2& NAMPT double-targeting compound and pharmaceutical composition and application thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105916845A (en) * 2014-01-17 2016-08-31 诺华股份有限公司 N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
WO2018136264A1 (en) * 2017-01-23 2018-07-26 Revolution Medicines, Inc. Pyridine compounds as allosteric shp2 inhibitors
WO2018136265A1 (en) * 2017-01-23 2018-07-26 Revolution Medicines, Inc. Bicyclic compounds as allosteric shp2 inhibitors
WO2019075265A1 (en) * 2017-10-12 2019-04-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric shp2 inhibitors
WO2019118909A1 (en) * 2017-12-15 2019-06-20 Revolution Medicines, Inc. Polycyclic compounds as allosteric shp2 inhibitors
CN109983001A (en) * 2016-07-12 2019-07-05 锐新医药公司 Tri- substituted type 3- methylpyrazine of the disubstituted type 3- methylpyrazine of 2,5- and 2,5,6- as allosteric SHP2 inhibitor
CN110156786A (en) * 2018-02-13 2019-08-23 上海青煜医药科技有限公司 Pyrimido cycle compound and its preparation method and application
CN111153901A (en) * 2018-11-07 2020-05-15 如东凌达生物医药科技有限公司 Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2699351T3 (en) * 2014-01-17 2019-02-08 Novartis Ag Derivatives of 1-pyridazin / triazin-3-yl-piper (-azine) / idine / pyrolidine and compositions thereof to inhibit the activity of SHP2
CN112300160A (en) * 2019-08-01 2021-02-02 上海奕拓医药科技有限责任公司 Spiro aromatic ring compound, preparation and application thereof

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105916845A (en) * 2014-01-17 2016-08-31 诺华股份有限公司 N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
CN109983001A (en) * 2016-07-12 2019-07-05 锐新医药公司 Tri- substituted type 3- methylpyrazine of the disubstituted type 3- methylpyrazine of 2,5- and 2,5,6- as allosteric SHP2 inhibitor
WO2018136264A1 (en) * 2017-01-23 2018-07-26 Revolution Medicines, Inc. Pyridine compounds as allosteric shp2 inhibitors
WO2018136265A1 (en) * 2017-01-23 2018-07-26 Revolution Medicines, Inc. Bicyclic compounds as allosteric shp2 inhibitors
WO2019075265A1 (en) * 2017-10-12 2019-04-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric shp2 inhibitors
WO2019118909A1 (en) * 2017-12-15 2019-06-20 Revolution Medicines, Inc. Polycyclic compounds as allosteric shp2 inhibitors
CN110156786A (en) * 2018-02-13 2019-08-23 上海青煜医药科技有限公司 Pyrimido cycle compound and its preparation method and application
CN111153901A (en) * 2018-11-07 2020-05-15 如东凌达生物医药科技有限公司 Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11179397B2 (en) 2018-10-03 2021-11-23 Gilead Sciences, Inc. Imidazopyrimidine derivatives
WO2021257736A1 (en) 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
WO2022060583A1 (en) 2020-09-03 2022-03-24 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
WO2022060836A1 (en) 2020-09-15 2022-03-24 Revolution Medicines, Inc. Indole derivatives as ras inhibitors in the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022259157A1 (en) 2021-06-09 2022-12-15 Novartis Ag A triple pharmaceutical combination comprising dabrafenib, trametinib and a shp2 inhibitor
WO2022269525A1 (en) 2021-06-23 2022-12-29 Novartis Ag Pharmaceutical combinations comprising a kras g12c inhibitor and uses thereof for the treatment of cancers
WO2023031781A1 (en) 2021-09-01 2023-03-09 Novartis Ag Pharmaceutical combinations comprising a tead inhibitor and uses thereof for the treatment of cancers
US20230102398A1 (en) * 2021-09-27 2023-03-30 New York University Therapeutic targeting of kmt2d mutant lung squamous cell carcinoma through rtk-ras signaling inhibition
WO2023060253A1 (en) 2021-10-08 2023-04-13 Revolution Medicines, Inc. Ras inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Also Published As

Publication number Publication date
CN114269746A (en) 2022-04-01
CN112300160A (en) 2021-02-02

Similar Documents

Publication Publication Date Title
WO2021018287A1 (en) Spiroaromatic compound, preparation and application thereof
EP3712151B1 (en) (s)-1&#39;-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4&#39;-piperidine]-5-amine as shp2 inhibitor for the treatment of cancer
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
CN110156786B (en) Pyrimido-cyclic compounds, process for their preparation and their use
WO2021244659A1 (en) Isotope-substituted spiro aromatic ring compound and application thereof
CN110950876B (en) Furanolactam compounds, preparation method and application
KR20200078610A (en) Amino-substituted nitrogen-containing condensed-ring compounds and preparation methods and uses thereof
CN112457326A (en) Aromatic heterocyclic lactam compound, preparation method and application
WO2022089406A1 (en) Nitrogen-containing fused heterocyclic compound, and preparation method therefor and use thereof
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
WO2021083383A1 (en) Nitrogen-containing fused cyclic compound as sting regulator, and preparation method therefor and use thereof
WO2020187123A1 (en) Pyrrole amidopyridone compound, preparation method therefor and use thereof
CN113242858A (en) Preparation and application of protein receptor kinase inhibitor
WO2022037691A1 (en) Aromatic ring-lactam compound, preparation method therefor and use thereof
CN112812118A (en) Preparation and application of protein receptor kinase inhibitor
RU2781100C1 (en) Compounds with spiro and aromatic rings and application thereof
WO2020200154A1 (en) A class of thiophene fused n-heterocyclies, preparation method and use
WO2024104244A1 (en) Oxamide compound, pharmaceutical composition containing same, and preparation method therefor and use thereof
CN116444497A (en) Pyridazinone compound, preparation and application thereof

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: 20846273

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20846273

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 20846273

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 22/09/2022)

122 Ep: pct application non-entry in european phase

Ref document number: 20846273

Country of ref document: EP

Kind code of ref document: A1