WO2023005928A1 - 8-氧-3-氮杂二环[3.2.1]辛烷类化合物或其盐及其制备方法和用途 - Google Patents

8-氧-3-氮杂二环[3.2.1]辛烷类化合物或其盐及其制备方法和用途 Download PDF

Info

Publication number
WO2023005928A1
WO2023005928A1 PCT/CN2022/107932 CN2022107932W WO2023005928A1 WO 2023005928 A1 WO2023005928 A1 WO 2023005928A1 CN 2022107932 W CN2022107932 W CN 2022107932W WO 2023005928 A1 WO2023005928 A1 WO 2023005928A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
oxa
cancer
azabicyclo
Prior art date
Application number
PCT/CN2022/107932
Other languages
English (en)
French (fr)
Inventor
程耀邦
黄亚飞
周娟
王永辉
陈伟
Original Assignee
上海辉启生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海辉启生物医药科技有限公司 filed Critical 上海辉启生物医药科技有限公司
Priority to KR1020247006386A priority Critical patent/KR20240041354A/ko
Priority to CA3227713A priority patent/CA3227713A1/en
Priority to AU2022316931A priority patent/AU2022316931A1/en
Priority to IL310412A priority patent/IL310412A/en
Publication of WO2023005928A1 publication Critical patent/WO2023005928A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to the field of medicinal chemistry, in particular to a group of 8-oxa-3-azabicyclo[3.2.1]octane compounds used as ATR inhibitors, their preparation methods, their pharmaceutical compositions and their Use for treating or preventing ATR-related diseases.
  • DDR DNA Damage Response
  • ATR ataxia telangiectasia mutated and Rad3-associated kinase
  • ATM ataxia telangiectasia mutated kinase
  • DNA-PK DNA-dependent protein kinase
  • ATR is a member of the phosphatidylinositol kinase-related kinase (PIKK) protein family, and its main target is CHK1.
  • PIKK phosphatidylinositol kinase-related kinase
  • ATR inhibition Compared with healthy proliferating cells, tumor cells have higher DNA damage and replication stress. To survive replication and maintain cell division, they are more dependent on ATR for DNA repair. Therefore, by inhibiting ATR, the repair function of tumor cells can be inhibited, leading to increased DNA damage and replication stress that cannot be repaired, and eventually leading to tumor cell death, while not affecting or less affecting healthy proliferating cells, which constitutes ATR inhibition.
  • ATR inhibition has been considered as an important cancer treatment approach in recent years.
  • Standard cancer therapies such as radiation or chemotherapy work by inducing DNA damage, which is particularly toxic to proliferating cells, and the presence of DNA damage repair mechanisms limits the efficacy of these therapies, resulting in resistance to chemotherapy or radiation agents sex.
  • ATR increasing replication stress and increasing DNA damage, it can enhance the sensitivity of tumor cells to these DNA damage-induced therapies, help to overcome the drug resistance caused by radiotherapy or chemotherapy drugs due to damage repair, and can be used for gene therapy
  • the dose of chemotherapy or radiotherapy should be reduced, thereby reducing the toxicity to the blood and gastrointestinal organ systems.
  • ATR inhibitors can be used to increase replication stress and induce tumor cell death. Indeed, studies have revealed that ATR inhibitors are synthetically lethal against p53-mutant tumors or tumors that have lost ATM function and are synergistic when combined with a variety of replication stress/DNA damage-inducing chemotherapeutic agents, such as platinum, ionizing radiation and PARP inhibitors.
  • ATR inhibition can also prevent the occurrence of cancer, because ATR is also an important member of the DNA damage checkpoint, and ATR inhibition will limit the expansion of original cancer cells caused by oncogene activation.
  • ATR inhibitors have been developed (eg WO2017202748, CN111848605A, WO2020087170, WO2020049017). However, it is still very challenging to develop new potent ATR inhibitors.
  • the high identity of the PIKK kinase family to related lipid kinases such as phosphatidylinositol 3-kinase (PI3KS) as well as mTOR increases the risk of inhibition of other kinases, which may increase toxicity or counteract the therapeutic effect of ATR inhibition; in addition, some ATR
  • the application of inhibitors is also limited by physicochemical properties, pharmacokinetic properties, and drug-drug interactions.
  • the present inventors have identified through research that the compounds of the present invention exhibit satisfactory ATR inhibitory activity, and show good performance in in vivo and/or in vitro pharmacokinetic experiments, indicating improved druggability and improved bioavailability Spend. Therefore, the compound of the present invention can not only achieve the purpose of preventing or treating diseases related to ATR, but also the prepared medicine is expected to have improved absorption, improved curative effect at the same dose, or provide the same curative effect at a lower dose and/or Reduce possible side effects.
  • the present invention also provides the use of the compound of the present invention in the preparation of a medicament for the prevention or treatment of diseases associated with ATR, the pharmaceutical composition comprising the compound, and the prevention and/or treatment of ATR-related diseases by administering the compound. way of disease.
  • compounds of the formula (I) of the present invention are provided, their stereoisomers, mutuals, which have ATR inhibitory activity and are used as medicines, especially as ATR inhibitors for the treatment or prevention of ATR-related diseases.
  • a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition may additionally comprise additional therapeutically active ingredients suitable for use in combination with the compounds of the invention.
  • a pharmaceutical combination such as a kit, comprising a compound of the invention and a further active agent.
  • the use of the compound of the present invention or a pharmaceutical composition comprising it for preventing or treating ATR-related diseases in mammals, especially humans.
  • a method of inhibiting ATR in vivo or in vitro comprising contacting said ATR with an effective amount of a compound of the invention.
  • a method for preventing or treating an ATR-related disease in an individual comprising administering an effective amount of the compound of the present invention described herein or a medicament comprising it combination.
  • treating means administering one or more compounds of the invention described herein to a subject, such as a mammal, such as a human, suffering from, or having symptoms of, the disease, for Curing, alleviating, alleviating or affecting the disease or the symptoms of the disease.
  • a subject such as a mammal, such as a human
  • said disease is an ATR-associated disease as defined below, especially a tumor or cancer.
  • prophylaxis as used herein is well known in the art and is administered to a subject, such as a mammal, such as a human, suspected of suffering from or susceptible to an ATR-associated disease as defined herein, especially cancer or tumor
  • a subject such as a mammal, such as a human
  • ATR-associated disease as defined herein, especially cancer or tumor
  • One or more of the compounds of the invention described herein results in a reduced risk of developing a defined disease.
  • prevention encompasses the use of the compounds of the invention prior to the diagnosis or determination of any clinical and/or pathological symptoms.
  • inhibitor and “reduce” or any variation of these terms, as used herein, refer to the ability of a biologically active agent to reduce the signaling activity of a target of interest by interacting directly or indirectly with the target, and refer to Any measurable reduction or complete inhibition of the activity of a target of interest.
  • the activity e.g., ATR activity
  • the activity may be reduced by about or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50% compared to normal conditions. %, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more, or any range derivable therein.
  • selective inhibition refers to the ability of a bioactive agent to preferentially reduce signaling activity of a target of interest over off-target signaling activity by interacting directly or indirectly with the target. As far as the compound of the present invention is concerned, it can selectively inhibit the activity of ATR, thereby reducing Simultaneously acting on other kinases may cause toxicity or offset the inhibitory effect of ATR.
  • the present invention has at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55% ATR as compared to another specific kinase , 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more, or any range of better activity derivable therein, or with another At least 1-, 2-, 3-, 4-, 5-, 10-, 25-, 50-, 100-, 250-, or 500-fold better activity on ATR than the activity of a particular kinase.
  • cancer refers to neoplastic cell growth and proliferation, whether malignant or benign, and to all precancerous and cancerous cells and tissues.
  • the cancer or tumor includes but not limited to colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma Carcinoma, adenocarcinoma, sweat gland cancer, sebaceous gland cancer, lung cancer, leukemia, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, thyroid cancer, kidney cancer, uterine cancer, pemphigus cancer, liver cancer, auditory nerve tumor, oligodendroglioma, meningioma, neuroblastoma, eye cancer.
  • an "anticancer effect” or “antitumor effect” as described herein includes, but is not limited to, effects on response rate, time to disease progression, and survival rate.
  • the antitumor effects of the compounds of the present invention and their medical uses and methods include, but are not limited to, inhibiting tumor growth, delaying tumor growth, regressing tumors, shrinking tumors, prolonging tumor regrowth after cessation of treatment, slowing down disease progression, and also including preventing tumorigenesis .
  • the term "therapeutically effective amount” means an amount sufficient to alleviate or completely relieve the symptoms or other deleterious effects of a disorder; reverse, completely stop or slow the progression of a disorder; or reduce the risk of a worsening of a disorder when administered to an individual to treat a disease.
  • the “effective amount” may vary depending on the compound, the disease and its severity, and the age, weight, etc. of the individual to be treated.
  • the term "individual” as used herein includes a human or a non-human animal.
  • exemplary human subjects include human subjects suffering from a disease (eg, a disease described herein) (referred to as a patient) or normal subjects.
  • Non-human animals in the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, , cats, cows, pigs, etc.).
  • ATR-associated disease refers to a disease in which ATR activity promotes the occurrence and development of the disease, or inhibition of ATR will reduce the incidence of the disease, reduce or eliminate the disease symptoms.
  • ATR-associated disease preferably refers to an ATR-mediated disease, more preferably cancer or tumor.
  • ATR kinase inhibitors should be of therapeutic or prophylactic value for hematological malignancies such as leukemia (including chronic lymphocytic leukemia, acute lymphocytic leukemia, acute myelogenous leukemia, and chronic myelogenous leukemia), Multiple myeloma, lymphoid malignancies (such as lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma), myelodysplastic syndromes, and solid tumors such as carcinomas and sarcomas and their metastases, such as breast cancer, Lung cancer (non-small cell lung cancer, small cell lung cancer, squamous cell carcinoma, bronchoalveolar carcinoma), central nervous system tumors (eg, glioma, dysembryonic dysplastic neuroepithelial tumor, glioblastoma multiforme, mixed Glioma, medulloblastoma, retinoblastom
  • the compounds of the present invention are especially useful for the treatment of patients with lung cancer, prostate cancer, melanoma, ovarian cancer, breast cancer, endometrial cancer, renal cancer, gastric cancer, sarcoma, head and neck cancer, central nervous system tumors and their metastases, and patients with Patients with acute myeloid leukemia.
  • composition or “pharmaceutical preparation” as used herein refers to a compound comprising one or more compounds of formula (I) of the present invention or its stereoisomers, tautomers, stable isotope derivatives, pharmaceutically
  • composition of acceptable salt or solvate and pharmaceutically acceptable excipient or carrier generally accepted in the art, and may be in solid, semi-solid, liquid or gaseous form.
  • the term "pharmaceutical combination" as used herein means that the compounds of the present invention may be combined with other active agents for the purposes of the present invention.
  • the other active agent may be one or more additional compounds of the invention, or may be a second or additional (e.g. a third) compound which is compatible with the compounds of the invention, i.e. does not adversely affect each other, or has complementary activities. ) compound.
  • Such active agents are suitably present in combination in amounts effective to achieve the intended purpose.
  • the other active agent may be co-administered with the compound of the present invention in a single pharmaceutical composition, or administered separately from the compound of the present invention in separate discrete units, eg, in kit form, which when administered separately may be carried out simultaneously or sequentially. The sequential administration may be close or distant in time.
  • the term "pharmaceutically acceptable” means approved or may be approved by the corresponding agency of each country, or listed in the generally accepted pharmacopoeia for use in animals, and more particularly in humans, or when administered in appropriate amounts to animals, such as humans. Molecular entities and compositions that do not produce adverse, allergic or other untoward reactions.
  • the term "pharmaceutically acceptable excipient or carrier” means one or more compatible solid or liquid filler or gel substances, which are pharmacologically inactive, compatible with the other ingredients in the composition and should be acceptable for administration to warm-blooded animals such as humans, for use as carriers or vehicles in administration forms for the compounds of the present invention, examples of which include, but are not limited to, cellulose and its derivatives (such as carboxymethylcellulose Sodium, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as magnesium stearate), calcium sulfate, vegetable oil, polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween category), wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, etc.
  • cellulose and its derivatives such as carboxymethylcellulose Sodium, cellulose acetate
  • the term "pharmaceutically acceptable salt” means a salt of a compound of the present invention that is pharmaceutically acceptable and possesses the desired pharmacological activity of the parent compound. Specifically, such salts are non-toxic and may be inorganic acid addition salts or organic acid addition salts and base addition salts.
  • such salts include: (1) acid addition salts formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, etc.; or acid addition salts formed with organic acids, the Organic acids such as acetic, propionic, caproic, glycolic, pyruvic, lactic, malonic, succinic, malic, maleic, fumaric, tartaric, citric, benzoic, cinnamic, almond acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptanoic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butyl Acetic acid, lauryl sulfate, gluconic acid, glutamic acid, hydroxynaphthoic acid,
  • stereoisomer refers to an isomer formed by at least one asymmetric center. In compounds having one or more, for example 1, 2, 3 or 4, asymmetric centers, racemic mixtures, single enantiomers, diastereomers and individual diastereomers may arise Construct. Certain molecules may also exist as geometric isomers (cis/trans). Compounds of the invention may also exist as mixtures of two or more structurally distinct forms in rapid equilibrium, commonly referred to as tautomers, representative examples include keto-enol, phenol-keto, nitroso- Oxime tautomers, etc. It should be understood that the scope of the present invention covers all such isomers and mixtures thereof in any proportion, such as 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97% , 98%, 99%.
  • solvate refers to solvent addition forms containing stoichiometric or non-stoichiometric amounts of solvent, including any solvated form of the compounds of the invention, including for example solvates with water, such as hydrates, Or a solvate with an organic solvent, such as methanol, ethanol or acetonitrile, ie as methanolate, ethanolate or acetonitrile, respectively; or in any polymorphic form. It should be understood that such solvates of the compounds of the invention also include solvates of the pharmaceutically acceptable salts of the compounds of the invention.
  • prodrug means a compound having a cleavable group and becoming a pharmaceutically active compound of the present invention in vivo by solvolysis or under physiological conditions.
  • Prodrugs include acid derivatives well known in the art, such as esters prepared by reacting the parent acid with a suitable alcohol, or amides prepared by reacting the parent acid compound with a substituted or unsubstituted amine, or anhydrides or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acid groups pendant on the compounds of the invention are particularly useful prodrugs. Specific such prodrugs are C 1-8 alkyl, C 2-8 alkenyl, optionally substituted C 6-10 aryl and (C 6- 10 aryl)-(C 1- 4 alkyl) esters.
  • isotopic variation refers to a compound that contains unnatural proportions of isotopes at one or more atoms that make up the compound.
  • Compounds of the present invention are those that contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute the compounds, resulting in isotopic variations, whether radioactive or not, that are intended to be encompassed within the scope of the present invention.
  • isotopes that may be incorporated into the compounds of the present invention, and pharmaceutically acceptable salts thereof, include, but are not limited to, isotopes of hydrogen (e.g., 2H, 3H); isotopes of carbon (e.g., 11C, 13C, and 14C); isotopes of chlorine (such as 36Cl); isotopes of fluorine (such as 18F); isotopes of iodine (such as 123I and 125I); isotopes of nitrogen (such as 13N and 15N); isotopes of oxygen (such as 15O, 17O and 18O); 32P); and sulfur isotopes (eg 35S).
  • isotopic variations of the compounds of the invention may generally be prepared by conventional methods using the appropriate isotopic variation of suitable reagents.
  • compound structural formula herein Represents an aromatic ring, that is, the selection of A 1 to A 5 makes the formed ring satisfy the valence bond theory of an aromatic ring, and is chemically feasible and stable.
  • the structure fragments used in this article The bond indicated to cross it is the bond by which the structural fragment connects to the rest of the molecule.
  • halo or halogen as used herein means fluorine (F), chlorine (Cl), bromine (Br) and iodine (I). Preferred halo is fluorine or chlorine.
  • halogen-substituted groups is intended to include monohalogenated or polyhalogenated groups in which one or more (eg 2, 3, 4, 5 or 6) identical or different halogen substituents One or more (eg 2, 3, 4, 5 or 6) hydrogens in a group.
  • cyano as used herein means the group -CN.
  • hydroxyl refers to -OH.
  • alkyl refers to a linear or branched saturated hydrocarbon group composed of carbon atoms and hydrogen atoms. Specifically, the alkyl group has 1-10, such as 1 to 6, 1 to 5, 1 to 4, 1 to 3 or 1 to 2 carbon atoms.
  • C 1 -C 6 alkyl refers to a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms, examples of which are methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl or tert-butyl), pentyl (including n-pentyl, isopentyl, neopentyl), n-hexyl, 2-methylpentyl, etc.
  • Certain alkyl groups have 1 to 3 carbon atoms.
  • alkoxy means the group -O-alkyl, wherein alkyl has the meaning described herein.
  • the term includes the group -OC 1-6 alkyl, more specifically -OC 1-3 alkyl.
  • Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy, isopropoxy), butoxy (including n-butoxy, isobutoxy, tert-butoxy), pentyloxy (including n-pentyloxy, isopentyloxy, neopentyloxy), hexyloxy (including n-hexyloxy, isohexyloxy) and the like.
  • Certain alkoxy groups have 1 to 3 carbon atoms.
  • alkylene used herein alone or in combination with other groups refers to a saturated straight-chain or straight-chain divalent hydrocarbon group.
  • C 1-3 alkylene refers to an alkylene group having 1 to 3 carbon atoms, such as methylene, ethylene, propylene, 1-methylethylene, 2-methyl Ethylene, etc.
  • cycloalkyl refers to a monocyclic, fused polycyclic, bridged polycyclic or spiro non-aromatic saturated monovalent hydrocarbon ring structure having the indicated number of ring atoms.
  • Cycloalkyl can have from 3 to 12 carbon atoms (i.e. C 3 -C 12 cycloalkyl), such as C 3-10 cycloalkyl, C 3-8 cycloalkyl, C 3-6 cycloalkyl, C 5 -6 cycloalkyl.
  • Suitable cycloalkyl groups include, but are not limited to, monocyclic structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; or polycyclic (e.g., bicyclic) structures, including spiro Ring, fused or bridged systems such as bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, spiro[3.4]octyl, bicyclo[3.1.1]hexyl, bicyclo[3.1. 1] heptyl or bicyclo [3.2.1] octyl, etc.
  • monocyclic structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl
  • polycyclic (e.g., bicyclic) structures including spir
  • aryl as used herein means a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom in an aromatic ring system. Specifically, aryl refers to a monocyclic or fused polycyclic aromatic ring structure having the indicated number of ring atoms. In particular, the term includes groups comprising 6 to 14, eg 6 to 10, preferably 6 ring members. Particular aryl groups include phenyl and naphthyl, the most specific aryl group being phenyl.
  • substituted or “substituted” means that one or more (eg, 1, 2, 3, or 4) hydrogens on the designated atom are replaced by the designated group, provided that it does not exceed Where the indicated atoms are bonded at their normal valences at the current situation and form stable compounds, combinations of substituents and variables are permissible only if such combinations form stable compounds.
  • the term "optionally substituted”, unless otherwise indicated, means that a group may be unsubstituted or replaced by one or more (eg 0, 1, 2, 3, 4 or 5 or more)
  • the listed substituents are substituted, wherein the substituents may be the same or different.
  • Cn-n+m or Cn - Cm in the definition of the compounds of the present invention includes various situations of n to n+m carbons, for example, C 1-6 includes C 1 , C 2 , C 3 , C 4 , C 5 and C 6 , including any range from n to n+m, for example, C 0-6 includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 0- 1. C 0-2 , C 0-3 , C 0-4 , C 0-5 , C 1-2 , C 1-3 , C 1-4 , C 2-3 etc.
  • n-membered to n+m-membered in the definition of the compound of the present invention means that the number of ring atoms can be any one between n and n+m, and also includes any range of n-n+m-membered.
  • the stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates and prodrugs are as described above in the Definitions section.
  • the compound of the present invention is a free form of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof; most preferably a free form of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • Certain compounds of the present invention may exist in polymorphic or amorphous forms which are also within the scope of the present invention.
  • the compound of formula (I) may be in the form of a co-crystal with another chemical entity, and this description includes all such co-crystals.
  • the compounds of the invention may exist as individual enantiomers or as mixtures of enantiomers, and those skilled in the art will be able to determine which isomeric forms of the compounds of the invention are stable and feasible.
  • a compound of formula (I), or a pharmaceutically acceptable salt thereof which is an enantiomeric excess (%ee) >95, >98% or >99% of a single enantiomer.
  • the single enantiomer is present in >99% enantiomeric excess (%ee).
  • the compounds of the invention also encompass possible N-oxides, and those skilled in the art will be able to determine stable and viable N-oxides of the compounds of the invention.
  • the compounds of the present invention also encompass metabolites of the compounds of the present invention, that is, substances formed in vivo by oxidation, reduction, hydrolysis, amidation, esterification, etc. when the compounds of the present invention are administered, which can be identified by techniques known in the art.
  • the present invention provides a compound of formula (I), its stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate:
  • a 1 , A 2 and A 5 are each independently C or N;
  • a 3 and A 4 are each independently CR 4 , N or NR 5 ;
  • X is O, C(R6 )2 or NR7 ;
  • Y is N or CR 8 ;
  • R 1 , R 2 and R 3 are each independently H, -OH, oxo, halogen, CN, -C 1-6 alkyl or -OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally Substituted by one or more halogens or hydroxy groups; or R 1 and R 2 are joined together to form a C 1-3 alkylene bridge;
  • R 4 is H, oxo, halogen or -C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted by one or more halogen or hydroxyl;
  • R 5 is H or -C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted by one or more halogens;
  • R 6 are each independently H, halogen, CN, -OH, -NH 2 , -NH-C 1-6 alkyl, -N(C 1-6 alkyl) 2 , -C 1-6 alkyl, - OC 1-6 alkyl, -C(O)-C 1-6 alkyl, -C(O)-C 3-6 cycloalkyl, -SO 2 -C 1-6 alkyl, -SO 2 -C 3-6 cycloalkyl, -SO-C 1-6 alkyl , -SO-C 3-6 cycloalkyl, C 6-10 aryl or C 3-6 cycloalkyl, where -C 1-6 Alkyl, C 6-10 aryl or C 3-6 cycloalkyl are optionally substituted by one or more halogen, hydroxy, -OC 1-6 alkyl, -C 1-6 alkyl, or halogen or hydroxy C 1-6 alkyl substitution;
  • R 7 is H, -C 1-6 alkyl, -C(O)-C 1-6 alkyl, -C(O)-C 3-6 cycloalkyl, -SO 2 -C 1-6 alkyl , -SO 2 -C 3-6 cycloalkyl, -SO-C 1-6 alkyl or -SO-C 3-6 cycloalkyl, wherein -C 1-6 alkyl or C 3-6 cycloalkane
  • the group is optionally substituted by one or more halogen, hydroxyl, -OC 1-6 alkyl, -C 1-6 alkyl, or halogen or hydroxy substituted C 1-6 alkyl;
  • R 8 is H, -OH or halogen
  • n and m are each independently an integer of 0 to 4.
  • At least two of A 1 , A 2 , A 3 , A 4 and A 5 are N or NR 5 and the remainder are C or CR 4 ; preferably two of them are N or NR 5 , the rest are C or CR 4 .
  • the six- to five-membered heteroaryl moiety comprising A 1 to A 5 has a structure selected from:
  • the six-membered and five-membered heteroaryl moiety comprising A 1 to A 5 is selected from:
  • the six-membered and five-membered heteroaryl moiety comprising A 1 to A 5 is preferably
  • the six-membered and five-membered heteroaryl moiety comprising A 1 to A 5 is preferably
  • the six-membered and five-membered heteroaryl moiety comprising A 1 to A 5 is preferably
  • the six-membered and five-membered heteroaryl moiety comprising A 1 to A 5 is preferably
  • R4 is H.
  • R4 is oxo or halo.
  • R 4 is -C 1-6 alkyl, optionally substituted by one or more halogen or hydroxy; such as but not limited to methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl or tert-butyl), pentyl (including n-pentyl, isopentyl, neopentyl), n-hexyl, 2-Methylpentyl, -CH 2 F, -CHF 2 , -CF 3 , -C 2 F 5 , -CH 2 CF 3 , -CH 2 Cl, -CH 2 CH 2 CF 3 , -CH(CF 3 ) 2 , —CH 2 OH or —CH 2 CH 2 OH.
  • R 4 is -C 1-3 alkyl, eg methyl, ethyl
  • R4 is preferably H.
  • R 5 is H.
  • R 5 is -C 1-6 alkyl, optionally substituted by one or more halogens; such as but not limited to methyl, ethyl, propyl (including n-propyl base and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl or tert-butyl), pentyl (including n-pentyl, isopentyl, neopentyl), n-hexyl, 2- Methylpentyl, -CH 2 F, -CHF 2 , -CF 3 , -C 2 F 5 , -CH 2 CF 3 , -CH 2 Cl, -CH 2 CH 2 CF 3 or -CH(CF 3 ) 2 .
  • halogens such as but not limited to methyl, ethyl, propyl (including n-propyl base and isopropyl), butyl (including n-butyl, iso
  • R 5 is preferably C 1-3 alkyl, such as methyl, ethyl, propyl, isopropyl, most preferably methyl.
  • X is O.
  • X is C(R 6 ) 2 , wherein each R 6 is H.
  • X is C(R 6 ) 2 , wherein one of R 6 is H and the other is selected from halogen, CN, -OH, -C 1-6 alkyl, - OC 1-6 alkyl, -C(O)-C 1-6 alkyl, -C(O)-C 3-6 cycloalkyl, -SO 2 -C 1-6 alkyl, -SO 2 -C 3- 6 cycloalkyl, -SO-C 1-6 alkyl, -SO-C 3-6 cycloalkyl, C 6-10 aryl or C 3-6 cycloalkyl, where -C 1-6 Alkyl, C 6-10 aryl or C 3-6 cycloalkyl are optionally substituted by one or more halogen, hydroxy, -OC 1-6 alkyl, -C 1-6 alkyl, or halogen or hydroxy -C 1-6 alkyl substitution.
  • X is C(R 6 ) 2 , wherein one of R 6 is H and the other is selected from halogen such as fluorine, chlorine, bromine, iodine.
  • X is C(R 6 ) 2 , wherein one of R 6 is H and the other is OH, CN, NH 2 , -NH-C 1-6 alkyl, -N(C 1-6 alkyl) 2 , such as but not limited to OH, CN, -NH 2 , -NH-CH 3 , -NH-CH 2 -CH 3 , -N(CH 3 ) 2 , -N( CH 2 -CH 3 ) 2 , -N(CH 3 )(CH 2 -CH 3 ).
  • X is C(R 6 ) 2 , wherein one of R 6 is H and the other is -C 1-6 alkyl, optionally replaced by one or more halogen , hydroxyl or -OC 1-6 alkyl substitution, such as but not limited to methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl or tert-butyl), pentyl (including n-pentyl, isopentyl, neopentyl), n-hexyl, 2-methylpentyl, -CH 2 F, -CHF 2 , -CF 3 , -C 2 F 5.
  • R 6 is H and the other is -C 1-6 alkyl, optionally replaced by one or more halogen , hydroxyl or -OC 1-6 alkyl substitution, such as but not limited to methyl, ethyl, propy
  • X is C(R 6 ) 2 , wherein one of R 6 is H, and the other is -OC 1-6 alkyl, wherein -C 1-6 alkyl Optionally substituted with one or more halogen, hydroxy, or -OC 1-6 alkyl, such as but not limited to -OCH 3 , -OCH 2 CH 3 , -OCF 3 , -OCH 2 CF 3 , -OCH 2 CH 2 OH , -OCH2CH2OCH3 .
  • X is C(R 6 ) 2 , wherein one of R 6 is H and the other is -C(O)-C 1-6 alkyl, -C(O )-C 3-6 cycloalkyl, -SO 2 -C 1-6 alkyl, -SO 2 -C 3-6 cycloalkyl, -SO-C 1-6 alkyl, -SO-C 3-6 Cycloalkyl, C 6-10 aryl or C 3-6 cycloalkyl, such as but not limited to -C(O)-CH 3 , -C(O)-CH 2 CH 3 , C(O)-CH 2 CH 2 CH 3 , -C(O)-CH(CH 3 ) 2 , -C(O)-cyclopropyl, -C(O)-cyclopentyl, -C(O)-cyclohexyl, -SO 2 -CH 3 , -SO 2 -CH 2 CH
  • X is C(R 6 ) 2 , wherein each R 6 is independently -C 1-6 alkyl, halogen, CN, -OH, -NH 2 , -NH -C 1-6 alkyl, -N(C 1-6 alkyl) 2 , -OC 1-6 alkyl, wherein -C 1-6 alkyl is optionally replaced by one or more halogen, hydroxyl or - OC 1-6 alkyl substitution;
  • R 6 are halogen at the same time, C 1-6 alkyl optionally substituted by (such as halogen) at the same time, one is halogen and the other is said optionally substituted by (such as halogen) C 1-6 alkyl, one is OH or -OC 1-6 alkyl and the other is said C 1-6 alkyl optionally substituted by (eg halogen), one is NH 2 , -NH-C 1-6 alkyl or -N(C 1-6 alkyl) 2 and the other is said optionally substituted (eg halogen) C 1-6 alkyl, one is CN and the other is said optionally C 1-6 alkyl substituted by (eg halogen);
  • X is for example but not limited to CF 2 , CCl 2 , CBr 2 , CFCl, C(CH 3 ) 2 , C(CH 2 CH 3 ) 2 , C(CH 3 )(CH 2 CH 3 ), C(CF 3 )( CF 3 ), C(CH 3 )(CF 3 ), C(CH 3 )(CH 2 OH), C(CH 3 )(CH 2 OCH 3 ), C(CH 3 )(F), C(CH 3 )(OCH 3 ), C(CH 3 )(OH), C(CH 3 )(NH 2 ), -C(CH 3 )(NHCH 3 )-.
  • X is C(R 6 ) 2 , wherein each R 6 is independently selected from H or halogen, for example both are H, or both are halogen, for example both are F.
  • X is NR7 and R7 is H.
  • X is NR 7
  • R 7 is -C 1-6 alkyl, optionally substituted by one or more halogen, hydroxy, or -OC 1-6 alkyl,
  • X is NR 7
  • R 7 is -C(O)-C 1-6 alkyl, -C(O)-C 3-6 cycloalkyl, -SO 2 -C 1-6 alkyl, -SO 2 -C 3-6 cycloalkyl, -SO-C 1-6 alkyl or -SO-C 3-6 cycloalkyl, such as but not limited to -C(O )-CH 3 , -C(O)-CH 2 CH 3 , C(O)-CH 2 CH 2 CH 3 , -C(O)-CH(CH 3 ) 2 , -C(O)-cyclopropyl , -C(O)-cyclobutyl, -C(O)-cyclopentyl, -C(O)-cyclohexyl, -SO 2 -CH 3 , -SO 2 -CH 2 CH 3 , SO 2 -CH 2 CH 2 CH 3
  • X is NR 7 , R 7 is selected from H, -C 1-6 alkyl and -C(O)-C 1-6 alkyl, preferably -C 1- 6 alkyl, or preferably -C(O)-C 1-6 alkyl, such as but not limited to methyl, ethyl, propyl, isopropyl, -C(O)-CH 3 , -C(O) -CH2CH3, C(O ) -CH2CH2CH3 or -C (O)-CH ( CH3 ) 2 .
  • X is preferably -O-, -NH-, -N(C 1-6 alkyl)-, -CH 2 -, -C(halogen) 2 -, for example - O-, -NH-, -N(CH 3 )-, -CH 2 -, -C(F) 2 -.
  • X is preferably -O-, -N(C 1-6 alkyl)-, -N(CO-C 1-6 alkyl)-, -CH 2 -, -CH(C 1-6 alkyl)-, -C(C 1-6 alkyl) 2 - or -C(halogen) 2 -, for example -O-, -N(CH 3 )-, -N(CO -CH 3 )-, -CH 2 -, -C(F) 2 -.
  • Y is N.
  • Y is CR8 , wherein R8 is H.
  • Y is CR8 , wherein R8 is OH.
  • Y is CR 8 , wherein R 8 is halogen, eg fluorine, chlorine, bromine, iodine, preferably F.
  • Y is preferably N, or CR8 , wherein R8 is OH; more preferably Y is N.
  • the six-membered ring comprising X and Y selected from Preferably the six membered ring comprising X and Y is selected from More preferably the six membered ring comprising X and Y is selected from R 6 , R 7 and R 8 respectively have the meanings defined in each of the corresponding embodiments above, preferably R 6 is selected from H, halogen and C 1-6 alkyl, R 7 is selected from C 1-6 alkyl and -CO- C 1-6 alkyl.
  • R 1 and R 2 are each independently H.
  • R and R are each independently -OH , oxo, halogen, CN, -C 1-6 alkyl or -OC 1-6 alkyl, wherein C 1-6 alkyl is optionally substituted by one or more halogen or hydroxyl, such as but not limited to -OH, oxo, fluorine, chlorine, bromine, iodine, CN, -CH 3 , -CH 2 CH 3 , -O -CH 3 , -O-CH 2 CH 3 , -CF 3 , -CH 2 CF 3 , -CH 2 OH, -CH 2 CH 2 OH, preferably -CH 3 , -CF 3 or -CH 2 CH 3 , most -CH3 is preferred.
  • R 1 and R 2 are joined together to form a C 1-3 alkylene bridge, eg -CH 2 -, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -.
  • R 1 and R 2 are connected together to form a C 1-3 alkylene bridge, such as but not limited to:
  • Y is N and X is O.
  • R 1 and R 2 are each independently H, -CH 3 , -CH 2 CH 3 , or R 1 and R 2 are linked together to form a C 1-3 alkylene A base bridge, preferably a C 2 ethylene bridge.
  • m and n are both zero.
  • one of m and n is 0 and the other is 1, at this time R 1 or R 2 is not hydrogen, and may be attached to the ortho position of Y or X on the ring
  • the ortho position is preferably connected to the ortho position of Y.
  • R 1 or R 2 is C 1-6 alkyl, such as but not limited to -CH 3 , -CH 2 CH 3 or -CH(CH 3 ) 2 , preferably R 1 or R 2 is -CH 3 , attached to
  • the ortho-position of Y or the ortho-position of X is preferably connected to the ortho-position of Y.
  • R 1 or R 2 is not hydrogen, and can be connected to the ortho position of Y, X on the ring at the same time
  • R 1 or R 2 are each independently C 1-6 alkyl, such as but not limited to -CH 3 , -CH 2 CH 3 or -CH 2 -CH 2 -CH 3 , preferably R 1 or R 2 is - CH 3 , which is connected to the adjacent position of Y at the same time.
  • one of m and n is 0, the other is 3 or 4, at this time R 1 or R 2 is not hydrogen, the R 1 or R 2 are for example but not limited to - CH 3 , -CH 2 CH 3 or -CH 2 -CH 2 -CH 3 , for example, R 1 or R 2 is C 1-6 alkyl, preferably R 1 or R 2 is -CH 3 .
  • m and n are both 1 , at this time R and R are not hydrogen , each independently can be connected to the ortho position of Y or the ortho position of X on the ring, for example Both are connected to the ortho position of Y, or are connected to the ortho position of X, or are respectively connected to the ortho position of Y and the ortho position of X ; preferably R and R are C 1-6 alkyl , more preferably R and R 2 is -CH 3 , both are connected to the ortho-position of Y, or both are connected to the ortho-position of X, or are respectively connected to the ortho-position of Y and the ortho-position of X.
  • m and n are both 1 , at this time R and R are not hydrogen , and are connected to, preferably simultaneously connected to, R and R at the ortho - position of Y and the ortho-position of X 2 are linked together to form a C 1-3 alkylene bridge, eg -CH 2 -, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -.
  • m is 1 and n is 2, or m is 1 and n is 3, or m is 1 and n is 4, or m is 2 and n is 2, or m is 2 and n is 3, or m is 2 and n is 4, or m is 3 and n is 4, or m is 4 and n is 4 , wherein for example R and R are C 1-6 alkyl , preferably R 1 and R 2 are -CH 3 .
  • one of m and n is 0, the other is 1, R 1 or R 2 are each independently C 1-6 alkyl, optionally replaced by one or more halogen Substituted, connected to the ortho-position of Y or the ortho-position of X;
  • R 1 or R 2 are each independently C 1-6 alkyl, optionally substituted by one or more halogens, connected to the ortho position of Y, ortho to X or the adjacent positions connected to X and Y respectively;
  • m and n are both 1 and R 1 and R 2 are each independently C 1-6 alkyl, optionally substituted by one or more halogens, both are connected to the ortho position of Y on the ring, and both are connected to the ortho position of X bit, or respectively connected to the ortho-position of Y and the ortho-position of X.
  • one of m and n is 0, the other is 1, R 1 or R 2 is C 1-6 alkyl, and is connected to the ortho position of Y or to the ortho.
  • one of m and n is 0, the other is 1, R 1 or R 2 is C 1-6 alkyl, optionally substituted by halogen, and is attached to Y Ortho position; or one of m and n is 0, the other is 2, R 1 or R 2 are each independently C 1-6 alkyl, optionally substituted by halogen, all connected to the ortho position of Y.
  • R and/or R may be in R or S configuration, preferably R configuration, where chemically feasible.
  • R3 is H.
  • R3 is halogen, such as fluorine, chlorine, bromine, iodine, preferably fluorine or chlorine.
  • the compound of the present invention covers each of the above independent embodiments or each of the specific exemplified embodiments, and also encompasses any combination or subcombination of the above-mentioned individual embodiments or specific exemplified embodiments, and also covers the above Any combination of any preferred or exemplary embodiments constitutes an embodiment.
  • the six-membered and five-membered heteroaryl moiety comprising A 1 to A 5 is selected from:
  • the six-membered ring comprising X and Y is selected from
  • R 1 and R 2 are each independently H or -C 1-6 alkyl, wherein C 1-6 alkyl is optionally substituted by one or more halogens; or R 1 and R 2 are linked together to form C 1- 3 alkylene bridges;
  • R 3 is H or halogen
  • R4 is H ;
  • R 5 is H or -C 1-6 alkyl
  • R 6 are each independently H, halogen, -C 1-6 alkyl or -OC 1-6 alkyl, wherein -C 1-6 alkyl is optionally substituted by one or more halogens;
  • R 7 is H or -C 1-6 alkyl, wherein -C 1-6 alkyl is optionally substituted by one or more halogens;
  • R 8 is H, -OH or halogen
  • n and m are each independently an integer of 0 to 2.
  • the six- and five-membered heteroaryl moieties comprising A 1 to A 5 are selected from
  • the six membered ring comprising X and Y is selected from
  • the six membered ring comprising X and Y is
  • a six-membered ring containing X and Y is wherein R 1 is each independently C 1-6 alkyl, optionally substituted by halogen, n is selected from 0, 1 or 2; or a six-membered ring comprising X and Y R 1 and R 2 are joined together to form a C 1-3 alkylene bridge, e.g.
  • the six membered ring comprising X and Y is for example a six-membered ring containing X and Y is Wherein R 1 is each independently C 1-6 alkyl, optionally substituted by halogen, n is selected from 1 or 2, R 7 is selected from C 1-6 alkyl or -CO-C 1-6 alkyl.
  • the six membered ring comprising X and Y is for example a six-membered ring containing X and Y is Wherein R 1 is each independently C 1-6 alkyl, optionally substituted by halogen, n is selected from 0 or 1, R 6 is selected from H, halogen or C 1-6 alkyl substituted by halogen.
  • the six membered ring comprising X and Y is selected from
  • the six membered ring comprising X and Y is selected from
  • R and R are each independently H.
  • R 1 and R 2 are each independently -C 1-6 alkyl, optionally substituted with one or more halogens, for example -CH 3 , -CH 2 CH 3 , -CH 3 , -CH 2 CH 3 , - CH2CH2CH3 , -CH ( CH3 )2 , -CF3 or -CH2CF3 , most preferably -CH3 .
  • R 1 and R 2 are joined together to form a C 1-3 alkylene bridge, preferably a C 2 alkylene bridge.
  • R3 is H.
  • R 5 is -C 1-6 alkyl, for example -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , most -CH3 is preferred.
  • each R6 is independently H or halogen, preferably H or F.
  • R 7 is H, or -C 1-6 alkyl, eg -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , most preferably -CH3 .
  • R8 is -OH.
  • one of n and m is 0 and the other is 1
  • R or R is attached to the ortho position of Y or the ortho position of X on the ring, preferably attached to the ortho position of Y
  • R 1 or R 2 is C 1-6 alkyl, preferably R 1 or R 2 is -CH 3 , connected to the ortho position of Y or the ortho position of X, preferably connected to the ortho position of Y.
  • R 1 or R 2 are simultaneously connected to the ortho-position of Y, the ortho-position of X, or respectively connected to the ortho-position of Y on the ring.
  • the ortho-position and the ortho-position of X are preferably connected to the ortho-position of Y at the same time.
  • R 1 or R 2 is C 1-6 alkyl, preferably R 1 or R 2 is -CH 3 , and is connected to the ortho position of Y at the same time.
  • both n and m are 1 , and each of R and R can be independently connected to the ortho position of Y on the ring, or the ortho position of X, for example, both are connected to the ortho position of Y.
  • R1 and R2 are C 1-6 alkyl , more preferably R 1 and R 2 are -CH 3 , both are connected to the ortho-position of Y, or both are connected to the ortho-position of X, or are respectively connected to the ortho-position of Y and the ortho-position of X, preferably both are connected to the ortho-position of Y;
  • R 1 and R 2 connected to, preferably simultaneously connected to, the Y ortho position and the X ortho position are linked together to form a C 1-3 alkylene bridge, such as -CH 2 -, -CH 2 CH 2 -, -CH 2 CH2CH2- .
  • R and/or R may be in the R or S configuration, preferably the R configuration, where chemically feasible.
  • Specific embodiments of the compounds of the present invention include the following specific compounds, their stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates,
  • each preferred embodiment of the compound of formula (I) as defined herein is preferred, more preferably the Specific compounds listed.
  • ATR kinase is known to play a role in tumorigenesis as well as in a variety of other diseases.
  • the above-mentioned compounds of formula (I) are capable of potently inhibiting ATR kinase and thus have utility as anti-proliferative, apoptotic and/or anti-invasive drugs in the suppression and/or treatment of solid and/or liquid tumor diseases value.
  • the compounds of the present invention are expected to be useful in the prophylaxis or treatment of those tumors which are sensitive to inhibition of ATR.
  • the compounds of the present invention are expected to be useful in the prophylaxis or treatment of those tumors which are mediated solely or in part by ATR.
  • the compounds of the present invention can effectively inhibit the activity of ATR kinase and tumor cell lines, and can achieve one or more of the following technical effects:
  • IC50 is in the range of 0.1nM ⁇ 1 ⁇ M, preferably in the range of 0.1nM ⁇ 0.5 ⁇ M, preferably in the range of 0.1nM ⁇ 0.1 ⁇ M, more preferably in the range of 0.1nM ⁇ 50nM, 0.1 nM ⁇ 20nM, as verified in Activity Example 1; and/or
  • ⁇ Has good pharmacokinetic properties, such as longer t 1/2 , so that, for example, the dosing interval can be increased, and the half-life can be longer, so that patients have better compliance, as verified in Example 3 ;and / or
  • Good safety profile such as membrane permeability, P450 (reduced risk of drug interaction), lower toxicity and/or fewer side effects; and/or
  • the present invention also provides the following technical solutions in various aspects.
  • the invention provides compounds of the invention for use as a medicament, especially as an ATR inhibitor, more especially as an anticancer or antineoplastic agent.
  • the present invention provides compounds of the present invention for use in the treatment and/or prevention of ATR-associated diseases.
  • the present invention provides a method for treating and/or preventing ATR from promoting the occurrence and development of said disease, or inhibiting ATR will reduce the incidence of disease, reduce or eliminate disease symptoms.
  • Compounds, diseases such as tumors or cancers including but not limited to: hematological malignancies such as leukemia (including chronic lymphocytic leukemia, acute lymphocytic leukemia, acute myelogenous leukemia and chronic myelogenous leukemia), multiple myeloma , malignancies of the lymphatic system (such as lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma), myelodysplastic syndromes, and solid tumors such as carcinomas and sarcomas and their metastases, such as breast cancer, lung cancer (non-small cell lung cancer, small cell lung cancer, squamous cell carcinoma, bronchoalveolar carcinoma), central nervous system tumors (eg, glioma, dyse
  • the present invention provides a method for treating lung cancer, prostate cancer, melanoma, ovarian cancer, breast cancer, endometrial cancer, kidney cancer, gastric cancer, sarcoma, head and neck cancer, central nervous system tumors and metastases thereof, and patients with A compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or a solvate thereof in a patient with acute myeloid leukemia.
  • the compounds of the invention may be formulated into pharmaceutical compositions according to standard pharmaceutical practice for therapeutic or prophylactic purposes.
  • drugs with better pharmacokinetic properties and higher bioavailability can be prepared from the compounds of the present invention.
  • the present invention provides a pharmaceutical composition comprising the above-mentioned compound of the present invention and a pharmaceutically acceptable excipient.
  • excipients to include in a particular composition will depend on factors such as the mode of administration and the form of the composition being provided. Suitable pharmaceutically acceptable excipients are well known to those skilled in the art and are described, for example, in Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems.
  • diluents such as glucose, lactose or mannitol
  • carriers pH adjusters, buffers, sweeteners, fillers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives additives, antioxidants, opacifiers, glidants, processing aids, coloring agents, flavoring agents, flavoring agents, and other known additives.
  • diluents such as glucose, lactose or mannitol
  • carriers pH adjusters, buffers, sweeteners, fillers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives additives, antioxidants, opacifiers, glidants, processing aids, coloring agents, flavoring agents, flavoring agents, and other known additives.
  • compositions of the present invention can be formulated by techniques known to those skilled in the art, such as those disclosed in Remington's Pharmaceutical Sciences 20th Edition.
  • compositions of the invention can be administered in standard manner.
  • suitable modes of administration include oral, intravenous, rectal, parenteral, topical, transdermal, ophthalmic, nasal, buccal or pulmonary (inhalation) administration, wherein parenteral infusions include intramuscular, intravenous, intraarterial, peritoneal Administered intradermally or subcutaneously.
  • the compounds of the present invention can be formulated by methods known in the art, for example as tablets, capsules, syrups, powders, granules, aqueous or oily solutions or suspensions, (lipid) emulsions, dispersible powders, suppositories, Ointments, creams, drops, aerosols, dry powder preparations and sterile injectable aqueous or oily solutions or suspensions.
  • the size of the prophylactic or therapeutic dose of a compound of the invention will vary depending on a number of factors including the individual being treated, the severity of the disorder or condition, the rate of administration, disposition of the compound, and the judgment of the prescribing physician.
  • an effective amount is that amount sufficient to ameliorate or alleviate symptoms associated with the disease.
  • Such amounts may be administered as a single dose, or may be administered according to an effective treatment regimen.
  • the effective dose is about 0.0001 to about 5000 mg per kg body weight per day, for example about 0.01 to about 1000 mg/kg/day (single or divided administration).
  • the content or dosage of the compound of the present invention in the pharmaceutical composition can be about 0.01 mg to about 1000 mg, suitably 0.1-500 mg, preferably 0.5-300 mg, more preferably 1-150 mg, especially preferably 1-50 mg, Such as 1.5 mg, 2 mg, 4 mg, 10 mg, 25 mg, etc.; correspondingly, the pharmaceutical composition of the present invention will contain 0.05 to 99% w/w (percentage by weight), such as 0.05 to 80% w/w, such as 0.10 to 70% w/w, eg 0.10 to 50% w/w of the compound of the invention, all weight percentages being based on the total composition. It will be understood that it may be necessary to use dosages outside these limits in some cases.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention and one or more pharmaceutically acceptable excipients, the composition being formulated for oral administration.
  • the composition may be presented in unit dosage form, for example in the form of a tablet, capsule or oral liquid formulation.
  • Such unit dosage forms may contain from 0.1 mg to 1 g, for example 5 mg to 250 mg, of a compound of the invention as active ingredient.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention and one or more pharmaceutically acceptable excipients, the composition being formulated for topical administration.
  • Topical administration may be in the form of, for example, creams, lotions, ointments or transdermal patches, wherein the concentration of the compound of the invention may be from about 0.01 to 100 mg per gram of carrier.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for administration by inhalation.
  • Administration by inhalation can be by oral inhalation or intranasal administration.
  • the compounds of the present invention may be effectively used in the present invention in daily doses, for example up to 500 ⁇ g, such as 0.1-50 ⁇ g, 0.1-40 ⁇ g, 0.1-30 ⁇ g, 0.1-20 ⁇ g or 0.1-10 ⁇ g of the present invention compound.
  • compositions of the present invention for oral inhalation may be formulated as dry powders, suspensions (in liquids or gases) or solutions (in liquids), and may be in any suitable form and using any suitable inhaler device known in the art.
  • Administration including, for example, metered dose inhalers (MDIs), dry powder inhalers (DPIs), nebulizers, and soft mist inhalers.
  • MDIs metered dose inhalers
  • DPIs dry powder inhalers
  • nebulizers nebulizers
  • soft mist inhalers soft mist inhalers.
  • Multi-chambered devices can be used to deliver a compound of the present description and one or more other active ingredients, when present.
  • compositions of the invention may additionally comprise additional therapeutically active ingredients suitable for use in combination with the compounds of the invention.
  • therapeutically active ingredients suitable for administration in combination with the compounds of the present invention may be known anticancer drugs, especially other anticancer drugs related to DNA damage and repair mechanisms, including PARP inhibitors, HDAC inhibitors and the like.
  • Other therapeutically active ingredients suitable for administration in combination with the compounds of the present invention may also be selected from anticancer drugs related to cell division checkpoints, including ChK1/2 inhibitors, CDK4/6 inhibitors, ATM/ATR inhibitors.
  • Other known anticancer drugs that can be used in combination include alkylating agents, topoisomerase I/II inhibitors, RNA/DNA antimetabolites, antimitotic agents, antibody drugs, kinase inhibitors, and the like.
  • the compound of the invention and at least one known anticancer drug may be administered as a single pharmaceutical composition, or separately, simultaneously or sequentially as separate entities, eg, as a kit.
  • the compounds of the invention may also be administered as bioconjugates.
  • the bioconjugate is composed of the compound of the present invention and at least one known therapeutically active antibody such as Herceptin or MabThera, or growth factors such as EGF or FGF, or cytokines such as interleukin 2 or 4 or any cell surface Combined molecular composition.
  • the antibodies and other molecules function by delivering the compounds of the invention to their targets, while also enhancing the therapeutic activity of the antibodies or other molecules.
  • Compounds of the invention may also be used in combination therapy with radiation therapy, which may be administered at the same or different times.
  • the above-mentioned pharmaceutical composition provided by the present invention can be used for preventing or treating the above-defined ATR-related diseases in mammals such as human subjects.
  • the compounds of the present invention can be used in methods for treating ATR-related diseases in animals, especially mammals such as humans.
  • the present invention provides a method of modulating, especially inhibiting, the kinase activity of ATR, said method comprising contacting a cell with a compound of the invention as hereinbefore described to modulate, especially inhibit, the activity of ATR in the cell.
  • the present invention also correspondingly provides a method for inhibiting abnormal cell growth in a mammal, comprising administering to the mammal a therapeutically effective amount of the compound of the present invention, or a pharmaceutical composition comprising the compound of the present invention.
  • the present invention provides a method for preventing or treating a disease associated with ATR (for example, a disease that can be treated or prevented by ATR inhibition), the method comprising administering to an individual in need thereof an effective amount of the present invention as described above.
  • a compound of the invention or a pharmaceutical composition of the invention comprising the same.
  • the present invention provides the use of the compound of the present invention as described above or a pharmaceutical composition comprising it, for inhibiting ATR activity, or for treating and/or preventing diseases associated with ATR, for example by inhibiting ATR A disease that can be treated or prevented.
  • the present invention also provides the use of the above-mentioned compound of the present invention or the pharmaceutical composition containing it in the preparation of medicines, especially the use of medicines with ATR kinase inhibitor activity.
  • the present invention provides a compound of the present invention as described above or a pharmaceutical composition comprising it in the preparation of a medicament for treating or preventing a disease associated with ATR, for example a disease treatable or preventable by inhibition of ATR kinase Use, wherein said compound or pharmaceutical composition is optionally combined with one or more chemotherapy or immunotherapy.
  • the present invention also provides a method for preparing the compound of formula (I), and the general synthetic scheme for synthesizing the compound of the present invention is illustrated below.
  • suitable reaction conditions are known to the person skilled in the art or can be determined routinely.
  • the raw materials and reagents used in the preparation of these compounds are generally commercially available, or can be prepared by the methods below, methods similar to those given below, or methods known in the art.
  • Starting materials and intermediates in the synthetic reaction schemes can be isolated and purified, if necessary, by conventional techniques including, but not limited to, filtration, distillation, crystallization, chromatography, and the like.
  • the materials can be characterized using conventional methods including physical constants and spectral data.
  • Step 1 heating the compound of formula (I-1) and amine in the presence of a base such as DIEA in a solvent such as NMP to obtain a compound of formula (I-2);
  • Step 2 reacting the compound of formula (I-2) with amine in the presence of a base such as DIEA and heating in a solvent such as NMP to obtain a compound of formula (I-3);
  • Step 3 reacting the compound of formula (I-3) with an iodination reagent such as NIS in a solvent such as ACN at room temperature to obtain the compound of formula (I-4);
  • Step 4 The compound of formula (I-4) is coupled by Suzuki, in the presence of a coupling agent such as Pd(dtbpf)Cl 2 /H 3 PO 4 , and heated in a solvent such as dioxane/water to obtain formula (I- 5) compounds; and
  • a coupling agent such as Pd(dtbpf)Cl 2 /H 3 PO 4
  • a solvent such as dioxane/water
  • Step 5 The compound of formula (I-5) is deprotected under the action of acid to obtain the compound of formula (I).
  • Step 1 the compound of formula (II-1) reacts with an iodination reagent such as NIS in the presence of an acid such as TFA in a solvent such as CHCl3 at room temperature to obtain a compound of formula (II-2);
  • an iodination reagent such as NIS
  • an acid such as TFA
  • a solvent such as CHCl3
  • Step 2 reacting the compound of formula (II-2) with amine in the presence of a base such as DIEA and heating in a solvent such as THF to obtain a compound of formula (II-3);
  • Step 3 the compound of formula (II-3) is coupled by Suzuki, in the presence of a coupling agent such as Pd(dtbpf)Cl 2 /H 3 PO 4 , and heated in a solvent such as dioxane/water to obtain formula (II- 4) compound;
  • a coupling agent such as Pd(dtbpf)Cl 2 /H 3 PO 4
  • a solvent such as dioxane/water
  • Step 4 reacting the compound of formula (II-4) with amine under the action of a catalyst such as RuPhos-G2, in the presence of a base such as cesium carbonate, and heating in a solvent such as toluene to obtain a compound of formula (II-4);
  • a catalyst such as RuPhos-G2
  • a base such as cesium carbonate
  • Step 5 the compound of formula (II-5) is deprotected under the action of acid such as HCl to obtain the compound of formula (II);
  • R 1 , R 2 , and X are as defined above for general formula (I):
  • Step 1 the compound of formula (III-1) reacts with a chlorination reagent such as POCl3 by heating to obtain the compound of formula (III-2);
  • Step 2 reacting the compound of formula (III-2) with amine in the presence of a base such as K 2 CO 3 in a solvent such as DMF at about room temperature to obtain a compound of formula (III-3);
  • Step 3 reacting the compound of formula (III-3) with amine in the presence of a base such as DIEA and heating in a solvent such as DMF to obtain a compound of formula (III-4);
  • Step 4 The compound of formula (III-4) reacts with a cyanating agent such as Zn(CN) 2 , in the presence of a catalyst such as Pd 2 (dba) 3 /DPPF, and in the presence of a solvent such as DMF, to obtain formula (III-5 ) compounds;
  • a cyanating agent such as Zn(CN) 2
  • a catalyst such as Pd 2 (dba) 3 /DPPF
  • a solvent such as DMF
  • Step 5 the compound of formula (III-5) is reacted in a solvent such as MeOH/THF in the presence of a catalyst such as Raney nickel/ammonia water to obtain a compound of formula (III-6) through reduction reaction;
  • Step 6 the compound of formula (III-6) is reacted in a solvent such as DME in the presence of a condensing agent such as HATU/DIEA to obtain a compound of formula (III-7) through condensation reaction;
  • Step 7 The compound of formula (III-7) is ring-closed by heating eg at 100-150° C. in the presence of POCl 3 to obtain the compound of formula (III).
  • Step 1 the compound of formula (IV-1) reacts with a formate such as ethyl formate in a solvent such as THF under the action of a base such as LDA at low temperature to obtain a compound of formula (IV-2);
  • Step 2 reacting the compound of formula (IV-2) with 3-hydrazine-1H-pyrazole in a solvent such as ethanol at room temperature to obtain the compound of formula (IV-3);
  • Step 3 ring-closing reaction of the compound of formula (IV-3) in a solvent such as NMP at elevated temperature such as reflux to obtain the compound of formula (IV-4);
  • Step 4 reacting the compound of formula (IV-4) with amine in a solvent such as DMSO by heating to obtain the compound of formula (IV-5);
  • Step 5 Add a protecting group to the compound of formula (IV-5) with a protecting reagent such as DHP/TsOH in a solvent such as DCM at room temperature to obtain a compound of formula (IV-6);
  • a protecting reagent such as DHP/TsOH in a solvent such as DCM
  • Step 6 the compound of formula (IV-6) is heated in a solvent such as NMP in the presence of a coupling agent such as Pd(dtbpf)Cl 2 /CsCO 3 , and coupled with an amine to obtain a compound of formula (IV-7);
  • a solvent such as NMP
  • a coupling agent such as Pd(dtbpf)Cl 2 /CsCO 3
  • Step 7 The compound of formula (IV-7) is deprotected to obtain the compound of formula (IV).
  • the experimental materials and reagents used in the following examples can be obtained from commercial sources, prepared according to methods in the prior art or prepared according to methods similar to those disclosed in the present application unless otherwise specified.
  • the raw materials used in the present invention are all commercially available raw materials, which can be used directly without further purification, wherein the 5,7-dichloropyrazolo[1,5-A]pyrimidine used in the following examples was purchased from Shanghai Haohong Biomedical Technology Co., Ltd. (Leyan, CAS: 57489-77-7, batch number: Ld102321002), 8-bromo-6-chloroimidazo[1,2-B]pyridazine was purchased from Shaoyuan Technology ( Shanghai) Co., Ltd.
  • column chromatography adopts silica gel (300-400 mesh) produced by Rushan Sun Desiccant Co., Ltd.; thin-layer chromatography adopts GF254 (0.25 mm); nuclear magnetic resonance chromatography (NMR) uses Varian- 400 nuclear magnetic resonance instrument for determination; liquid mass spectrometry (LC/MS) uses Agilent TechnologiESI 6120 liquid mass spectrometry instrument.
  • Step 4 3-(7-((R)-3-Methylmorpholino)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) Synthesis of pyrazolo[1,5-a]pyrimidin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 5 3-(7-((R)-3-Methylmorpholino)-3-(1H-pyrazol-3-yl)pyrazolo[1,5-a]pyrimidin-5-yl) Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 4 3-(8-((R)-3-Methylmorpholino)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) Synthesis of imidazo[1,2-b]pyridazin-6-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 5 3-(8-((R)-3-methylmorpholino)-3-(1H-pyrazol-3-yl)imidazo[1,2-b]pyridazin-6-yl) Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 4 6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-((R)-3-methylmorpholino)pyridazine-3-carba Nitrile synthesis
  • Step 5 (6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-((R)-3-methylmorpholino)pyridazine-3- base) synthesis of methylamine
  • Step 6 N-((6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-((R)-3-methylmorpholino)pyridazine- Synthesis of 3-yl)methyl)-1H-pyrazole 5-carboxamide
  • Step 7 3-(4-((R)-3-Methylmorpholino)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl) Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • the reaction solution was slowly added dropwise to a saturated sodium bicarbonate solution at 0°C, and then adjusted to pH 9 with a saturated sodium bicarbonate solution at 0°C, then washed with EA (200mL ⁇ 3 ) extraction, the combined organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue obtained was reversed-phase preparative chromatography twice (acetonitrile/water contained 0.05% ammonia) and (acetonitrile/water contained 0.05% formic acid) Separation and purification gave the target compound (1.50g, yield 22.2%, green solid).
  • 3-aminopyrazole (1.00g, 12.0mmol) was added to 6mol/L hydrochloric acid aqueous solution (7mL), the mixture was cooled to -5°C, and 1mol/L sodium nitrite aqueous solution (12mL, 12.0 mmol), the reaction solution was stirred and reacted at room temperature for 2 hours.
  • a concentrated hydrochloric acid solution 24mL of stannous chloride dihydrate (5.43g, 24.1mmol) was added to the reaction mixture, and the reaction solution was stirred at room temperature for 1 hour, then concentrated under reduced pressure to obtain the crude product of the target compound (9.00g, Crude product, bright yellow solid).
  • Step 2 Synthesis of 3-((2-(1H-pyrazolyl-3-yl)hydrazino)methyl)-2,6-difluoro-4-iodopyridine
  • Step 5 3-(4-Iodo-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)-1H-pyrazol[3,4-b] Synthesis of pyridin-6-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 6 3-(4-((R)-3-Methylmorpholino)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) Synthesis of -1H-pyrazol[3,4-b]pyridin-6-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 7 3-(4-((R)-3-Methylmorpholino)-1-(1H-pyrazol-3-yl)-1H-pyrazol[3,4-b]pyridine-6- Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 4-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-6-(8-oxa-3-azabicyclo[3.2.1]octane- Synthesis of 3-yl)pyridazine-3-carbonitrile
  • Step 4 (4-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-6-(8-oxa-3-azabicyclo[3.2.1]octane Synthesis of -3-yl)pyridazin-3-yl)methanamine
  • Step 5 N-((4-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-6-(8-oxa-3-azabicyclo[3.2.1 Synthesis of ]octane-3-yl)pyridazin-3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 6 3-(4-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-7-(1H-pyrazol-3-yl)imidazol[1,5- b] Synthesis of pyridazin-2-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 4 Synthesis of (6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-morpholinopyridazin-3-yl)methanamine
  • Step 5 N-((6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-morpholinopyridazin-3-yl)methyl)-1H -Synthesis of pyrazole-5-carboxamide
  • Step 7 3-(4-Morpholino-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl)-8-oxa-3-aza Synthesis of Bicyclo[3.2.1]octane
  • Example 8 8-(2-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-7-(1H-pyrazol-3-yl)imidazol[1,5 -b]pyridazin-4-yl)-3- Oxa-8-azabicyclo[3.2.1]octane
  • LC-MS (ESI) m/z 259.9 [M+H] + , 261.9 [M+2+H] + .
  • Step 2 8-(6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-chloropyrazin-4-yl)-3-oxa-8- Synthesis of azabicyclo[3.2.1]octane
  • LC-MS (ESI) m/z 337.0 [M+H] + .
  • LC-MS (ESI) m/z 328.0 [M+H] + .
  • Step 4 (6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(3-oxa-8-azabicyclo[3.2.1]octane Synthesis of -8-yl)pyridazin-3-yl)methanamine
  • Step 5 N-((6-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-4-(3-oxa-8-azabicyclo[3.2.1 Synthesis of ]octane-8-yl)pyridazin-3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 6 8-(2-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-7-(1H-pyrazol-3-yl)imidazol[1,5- b] Synthesis of pyridazin-4-yl)-3-oxa-8-azabicyclo[3.2.1]octane
  • LC-MS (ESI) m/z 346.9 [M+H] + .
  • LC-MS (ESI) m/z 424.2 [M+H] + .
  • LC-MS (ESI) m/z 415.0 [M+H] + .
  • Step 4 (3R)-4-(3-(Aminomethyl)-6-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)pyridazin-4-yl) Synthesis of tert-butyl 3-methylpiperazine-1-carboxylate
  • Step 6 3-(4-((R)-2-Methylpiperazin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazine-2 Synthesis of -yl)-8-oxa-3-azabicyclo[3.2.1]octane formate
  • Step 1 Synthesis of tert-butyl 4-(3,6-dichloropyridazin-4-yl)piperazine-1-carboxylate
  • Step 2 4-(6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-chloropyridazin-4-yl)piperazine-1-carboxylic acid Synthesis of tert-butyl ester
  • Step 3 4-(6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-cyanopyridazin-4-yl)piperazine-1-carboxy Synthesis of tert-butyl ester
  • LC-MS (ESI) m/z 401.1 [M+H] + .
  • Step 4 4-(3-(Aminomethyl)-6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)pyridazin-4-yl)piperazine-1- Synthesis of tert-Butyl Carboxylate
  • Step 5 4-(3-((1H-pyrazole-5-carboxamide)methyl)-6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)pyridazine Synthesis of -4-yl)piperazine-1-carboxylate tert-butyl ester
  • Step 6 3-(4-(Piperazin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl)-8-oxa Synthesis of -3-azabicyclo[3.2.1]octane formate
  • Step 2 6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-((R)-2,4-dimethylpiperazin-1-yl) Synthesis of pyridazine-3-carbonitrile
  • Step 3 (6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-((R)-2,4-dimethylpiperazin-1-yl ) Synthesis of pyridazin-3-yl)methylamine
  • Step 4 N-((6-(8-Oxy-3-azabicyclo[3.2.1]octan-3-yl)-4-((R)-2,4-Dimethylpiperazine- Synthesis of 1-yl)pyrazin-3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 5 3-(3-Chloro-4-((R)-2,4-dimethylpiperazin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5 -b] Synthesis of pyridazin-2-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 6 3-(4-((R)-2,4-Dimethylpiperazin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridine Synthesis of oxazin-2-yl)-8-oxa-3-azabicyclo[3.2.1]octane carboxylate
  • Step 2 3-(4-iodo-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)-1H-pyrazolo[3,4-b Synthesis of ]pyridin-6-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 3-(6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-1-(1-(tetrahydro-2H-pyran-2-yl) Synthesis of -1H-pyrazol-3-yl)-1H-pyrazolyl[3,4-b]pyridin-4-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 4 3-(6-(8-Oxa-3-azabicyclo[3.2.1]octane-3-yl)-1-(1H-pyrazol-3-yl)-1H-pyrazol[ Synthesis of 3,4-b]pyridin-4-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 1 Synthesis of methyl 4-amino-1-methyl-1H-pyrazole-5-carboxylate
  • Step 2 4-((1-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)ethylidene)amino)-1-methyl-1H-pyrazole-5 -Synthesis of methyl formate
  • Step 3 5-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-1-methyl-1H-pyrazolo[4,3-b]pyridine-7- Alcohol synthesis
  • Step 4 3-(7-Chloro-1-methyl-1H-pyrazolo[4,3-b]pyridin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane Alkane
  • Step 5 3-(7-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-1-methyl-1H-pyrazolo[4,3-b]pyridine Synthesis of -5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 6 3-(7-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-bromo-1-methyl-1H-pyrazolo[4,3 -b] Synthesis of pyridin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 7 3-(7-(8-oxa-3-azabicyclo[3.2.1]octyl)-1-methyl-3-(1-(tetrahydro-2H-pyran-2-yl Synthesis of )-1H-pyrazol-3-yl)-1H-pyrazolyl[4,3-b]pyridin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 8 3-(7-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-1-methyl-3-(1H-pyrazol-3-yl)- Synthesis of 1H-pyrazolo[4,3-b]pyridin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-(4,4-difluoropiperidin-1-yl)pyridazine-3-carba Nitrile synthesis
  • Step 4 (6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-(4,4-difluoropiperidin-1-yl)pyridazine-3- base) synthesis of methylamine
  • Step 5 N-((6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-(4,4-difluoropiperidin-1-yl)pyridazine Synthesis of -3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 6 3-(4-(4,4-Difluoropiperidin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl Synthesis of )-8-oxa-3-azabicyclo[3.2.1]octane
  • reaction solution was slowly added dropwise to a saturated sodium bicarbonate solution at 0°C, and then the pH was adjusted to 9 with a saturated sodium bicarbonate solution at 0°C, and then washed with EA (40.0 mL ⁇ 3) extraction, the combined organic layer was dried with anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was separated and purified by reverse-phase preparative chromatography (acetonitrile/water containing 0.05% formic acid) to obtain the target compound (127.46 mg, recovered rate 40.3%, gray solid).
  • LC-MS (ESI) m/z 416.1 [M+H] + .
  • Step 4 (6-(8-Oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-((S)-3-methylmorpholino)pyridazin-3-yl ) Synthesis of methylamine
  • Step 6 3-(4-((S)-3-methylmorpholino)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl) Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 6-(8-Oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-(cis-2,6-dimethylmorpholino)pyridazine-3-carba Nitrile synthesis
  • Step 4 (6-(8-Oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-(cis-2,6-dimethylmorpholino)pyridazine-3- base) synthesis of methylamine
  • Step 5 N-((6-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-4-(cis-2,6-dimethylmorpholino)pyridazine Synthesis of -3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 6 3-(4-(cis-2,6-dimethylmorpholino)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl Synthesis of )-8-oxa-3-azabicyclo[3.2.1]octane
  • LC-MS (ESI) m/z 232.1 [M+H] + .
  • Step 2 Synthesis of 3-(6-chloro-5-(piperidin-1-yl)pyridazin-3-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 Synthesis of 6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(piperidin-1-yl)pyridazine-3-carbonitrile
  • LC-MS (ESI) m/z 300.2 [M+H] + .
  • Step 4 (6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(piperidin-1-yl)pyridazin-3-yl)methanamine synthesis
  • Step 6 3-(4-(Piperidin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl)-8-oxa Synthesis of -3-azabicyclo[3.2.1]octane
  • LC-MS (ESI) m/z 380.1 [M+H] + .
  • LC-MS (ESI) m/z 248.1 [M+H] + .
  • LC-MS (ESI) m/z 225.1 [M+H] + .
  • Step 3 Synthesis of 6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(2-methylmorpholino)pyridazine-3-carbonitrile
  • LC-MS (ESI) m/z 316.0 [M+H] + .
  • LC-MS (ESI) m/z 396.1 [M+H] + .
  • Step 3 3-(1-Methyl-7-((R)-3-methylmorpholino)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole Synthesis of -3-yl)-1H-pyrazolo[4,3-b]pyridin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 4 3-(1-Methyl-7-((R)-3-methylmorpholino)-3-(1H-pyrazol-3-yl)-1H-pyrazolo[4,3- b] Synthesis of pyridin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 2 3-(6-Chloro-5-(3,3-dimethylmorpholino)pyridazin-3-yl)-8-oxa-3-azabicyclo[3.2.1]octane synthesis
  • LC-MS (ESI) m/z 330.2 [M+H] + .
  • Step 4 (6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(3,3-dimethylmorpholino)pyridazin-3-yl ) Synthesis of methylamine
  • Example 23 4-(5-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-3-(1H-pyrazol-5-yl )pyrazolo[1,5-a]pyrimidine -7-yl)tetrahydro-2H-pyran-4-ol
  • LC-MS (ESI) m/z 231.1 [M+H] + .
  • Step 2 4-(5-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octan-3-yl)pyrazol[1,5-a]pyrimidine-7- base) synthesis of tetrahydro-2H-pyran-4-ol
  • Step 3 4-(5-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-iodopyrazol[1,5-a] Synthesis of pyrimidin-7-yl)tetrahydro-2H-pyran-4-ol
  • Step 4 4-(5-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-3-(1-(tetrahydro-2H-pyridine Synthesis of pyran-2-yl)-1H-pyrazol-5-yl)pyrazol[1,5-a]pyrimidin-7-yl)tetrahydro-2H-pyran-4-ol
  • Step 5 4-(5-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-3-(1H-pyrazol-5-yl) Synthesis of pyrazol[1,5-a]pyrimidin-7-yl)tetrahydro-2H-pyran-4-ol
  • LC-MS (ESI) m/z 311.2 [M+H] + .
  • Step 3 3-(3-iodo-7-(1-methyl-1H-pyrazol-5-yl)pyrazolo[1,5-a]pyrimidin-5-yl)-8-oxa-3 -Synthesis of azabicyclo[3.2.1]octane
  • Step 4 3-(7-(1-Methyl-1H-pyrazol-5-yl)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3- Synthesis of yl)pyrazolo[1,5-a]pyrimidin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 5 3-(7-(1-Methyl-1H-pyrazol-5-yl)-3-(1H-pyrazol-3-yl)pyrazolo[1,5-a]pyrimidine-5- Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 6-Chloro-8-(1-methyl-1H-pyrazol-5-yl)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3 -Synthesis of imidazo[1,2-b]pyridazine
  • Step 4 3-(8-(1-Methyl-1H-pyrazol-5-yl)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3- Synthesis of imidazo[1,2-b]pyridazin-6-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 5 3-(8-(1-Methyl-1H-pyrazol-5-yl)-3-(1H-pyrazol-3-yl)imidazo[1,2-b]pyridazine-6- Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 1 3-(7-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)pyrazolo[1,5-a]pyrimidin-5-yl)-8- Synthesis of Oxa-3-azabicyclo[3.2.1]octane
  • Step 2 3-(7-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-iodopyrazol[1,5-a]pyrimidin-5-yl) Synthesis of -8-oxa-3-azabicyclo[3.2.1]octane
  • Step 3 3-(7-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-(1-(tetrahydro-2H-pyran-2-yl) Synthesis of -1H-pyrazol-5-yl)pyrazol[1,5-a]pyrimidin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 4 3-(7-(8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-3-(1H-pyrazol-5-yl)pyrazol[1,5 Synthesis of -a]pyrimidin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Example 29 1-((R)-4-(2-((1R,S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-7-(1H -pyrazol-3-yl)imidazol[1,5- b] pyridazin-4-yl)-3-methylpiperazin-1-yl)ethane-1-one
  • Step 3 Synthesis of (R)-1-(4-(3,6-dichloropyridazin-4-yl)-3-methylpiperazin-1-yl)ethan-1-one
  • Step 4 1-((3R)-4-(6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-3-chloropyridazin-4-yl)- Synthesis of 3-methylpiperazin-1-yl)ethan-1-one
  • Step 5 4-((R)-4-Acetyl-2-methylpiperazin-1-yl)-6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl ) Synthesis of pyridazine-3-carbonitrile
  • LC-MS (ESI) m/z 357.0 [M+H] + .
  • Step 6 1-((3R)-4-(3-(Aminomethyl)-6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)pyridazine-4 Synthesis of -yl)-3-methylpiperazin-1-yl)ethan-1-one
  • Step 7 N-((4-((R)-4-acetyl-2-methylpiperazin-1-yl)-6-(8-oxa-3-azabicyclo[3.2.1]octane Synthesis of alk-3-yl)pyridazin-3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 8 1-((R)-4-(2-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-7-(1H- Synthesis of pyrazol-3-yl)imidazol[1,5-b]pyridazin-4-yl)-3-methylpiperazin-1-yl)ethan-1-one
  • LC-MS (ESI) m/z 359.0 [M+H] + .
  • Step 4 6-(8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(4,4-difluoro-2-methylpiperidin-1-yl) Synthesis of pyridazine-3-carbonitrile
  • LC-MS (ESI) m/z 350.1 [M+H] + .
  • Step 5 (6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(4,4-difluoro-2-methylpiperidin-1-yl ) Synthesis of pyridazin-3-yl)methylamine
  • Step 6 N-((6-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-(4,4-difluoro-2-methylpiperidine- Synthesis of 1-yl)pyridazin-3-yl)methyl)-1H-pyrazole-5-carboxamide
  • Step 7 3-(4-(-4,4-Difluoro-2-methylpiperidin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b] Synthesis of pyridazin-2-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Examples 31 and 32 3-(4-((R)-4,4-difluoro-2-methylpiperidin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[ 1,5-b]pyridazin-2-yl)-8-oxa-3-azabicyclo[3.2.1]octane, and 3-(4-((S)-4,4-difluoro- 2-Methylpiperidin-1-yl)-7-(1H-pyrazol-3-yl)imidazo[1,5-b]pyridazin-2-yl)-8-oxa-3-aza Bicyclo[3.2.1]octane
  • Example 34 1-((R)-4-(6-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-1-(1H -pyrazol-3-yl)-1H- Pyrazol[3,4-b]pyridin-4-yl)-3-methylpiperazin-1-yl)ethan-1-one
  • Step 1 1-((R)-4-(6-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-1-(1- (Tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)-1H-pyrazolyl[3,4-b]pyridin-4-yl)-3-methylpiperazine- Synthesis of 1-yl)ethan-1-one
  • Step 2 1-((R)-4-(6-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-1-(1H- Synthesis of pyrazol-3-yl)-1H-pyrazol[3,4-b]pyridin-4-yl)-3-methylpiperazin-1-yl)ethan-1-one
  • Step 1 (4-(6-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-1-(1-(tetrahydro-2H- Synthesis of pyran-2-yl)-1H-pyrazol-3-yl)-1H-pyrazol[3,4-b]pyridin-4-yl)morpholin-3-yl)methanol
  • Step 2 (4-(6-((1R,5S)-8-oxa-3-azabicyclo[3.2.1]octane-3-yl)-1-(1H-pyrazol-3-yl Synthesis of )-1H-pyrazol[3,4-b]pyridin-4-yl)morpholin-3-yl)methanol
  • Step 4 3-(7-(3,3-Dimethylmorpholino)-3-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)pyridine Synthesis of Azol[1,5-a]pyrimidin-5-yl)-8-oxa-3-azabicyclo[3.2.1]octane
  • Step 5 3-(7-(3,3-Dimethylmorpholino)-3-(1H-pyrazol-5-yl)pyrazol[1,5-a]pyrimidin-5-yl)-8 -Synthesis of oxa-3-azabicyclo[3.2.1]octane
  • the homogeneous time-resolved fluorescence technique from Cisbio was used in this experiment to test.
  • test compound solutions Prepare the solutions of the compounds of the examples shown in the table below using DMSO as a solvent, and the stock solution is usually 10 mM.
  • the maximum initial concentration was 3 ⁇ M
  • DMSO was serially diluted 3 times
  • a total of 10 concentrations were added to the corresponding wells of a 384-well plate (geriner bio-one, Cat#784075)
  • the final concentrations were 3000, 1000, 333, 111, 37, 12.3, 4.12, 1.37, 0.457, 0.152nM, add the corresponding amount of DMSO to other wells to be used as negative control wells or positive control wells respectively.
  • anti-phospho-p53-Eu acts as an energy donor to specifically bind to the phosphorylation site on p53
  • anti-GST-d2 acts as an energy
  • the receptor can specifically bind to the GST tag carried on p53. If excited by a laser with a certain wavelength (the wavelength of the excitation light in this experiment is 340nm), the energy donor can emit light with a wavelength of 615nm. Linked to p53), energy transfer can occur between the energy donor and the energy acceptor, so that the energy acceptor emits emission light with a wavelength of 665nm. Use a plate reader to detect the two emitted lights and calculate the ratio of the two signals at 665nm and 615nm, and calculate the IC 50 of the sample to be tested by drawing and calculating.
  • HTRF detection buffer (Cisbio, Cat#62SDBRDF, Lot#17A), as shown in the following table:
  • Detection Reagent source Dilution factor Mother liquor concentration anti-phospho-p53-Eu Cisbio,Cat#61P08KAZ 400 400 units anti-GST-d2 Cisbio, Cat#61GSTDLB 200 200 units
  • Inhibition rate of single well 1-(signal value of single well-average signal value of positive control)/(average signal value of negative control-average signal value of positive control)
  • the inhibition rate of compound pores can be calculated.
  • Inhibition rate minimum response+(compound concentration ⁇ curve slope)*(highest response-minimum response)/(compound concentration ⁇ curve slope+half inhibitory concentration ⁇ curve slope)
  • ATR IC50 (nM) compound number ATR IC50 (nM) compound number.
  • ATR IC50 (nM) 1 17.4 16 21.9 2 40.3 17 32.0 3 5.9 18 50.8 4 20.3 26 57.7 5 8.2 27 149.0 6 13.2 20 1.4 7 21.4 twenty three 85.5

Abstract

提供一组用做ATR抑制剂的式(I)的8-氧杂-3-氮杂二环[3.2.1]辛烷化合物、其制备方法、包含其的药物组合物以及它们用于治疗或预防ATR相关疾病的用途。

Description

8-氧-3-氮杂二环[3.2.1]辛烷类化合物或其盐及其制备方法和用途
交叉引用
本申请要求申请日为2021年7月27日的中国发明专利申请202110860428.3、申请日为2022年7月1日的中国发明专利申请202210779302.8的优先权。
技术领域
本发明涉及药物化学领域,具体地涉及一组用做ATR抑制剂的8-氧杂-3-氮杂二环[3.2.1]辛烷化合物、其制备方法、包含其的药物组合物以及它们用于治疗或预防ATR相关疾病的用途。
背景技术
由于环境因素如紫外线辐射、X射线和内源性因素如活性氧,人体细胞持续暴露于DNA损伤事件。在癌症细胞中,由于受到致癌基因驱动子驱动或受到DNA损伤药物或电离辐射(IR)的外源性诱导,存在较高水平的DNA复制应激(癌症的标志),持续的复制应激导致DNA断裂,使得肿瘤细胞经历较高的DNA损伤,所述DNA损伤和复制应激构成基因组不稳定性的主要来源。
DNA损伤如果足够高且不被修复,将会对细胞产生高度毒性,导致细胞死亡。为了确保真核细胞基因组的完整性,已经进化形成一组统称为DDR(DNA损伤响应)的生物学通路,用以识别、传递信号并修复DNA损伤。ATR(共济失调毛细血管扩张突变和Rad3相关激酶)、ATM(共济失调毛细血管扩张突变激酶)和DNA-PK(DNA依赖性蛋白激酶)是DDR的关键组分,它们通过响应于不同的DNA损伤而发挥修复DNA损伤的功能,其中ATM和DNA-PK主要响应于DNA双链断裂,ATR主要响应于复制应激。
ATR属于磷脂酰肌醇激酶相关激酶(PIKK)蛋白家族的成员,其主要靶标是CHK1。当ATR被多种DNA损伤、尤其是复制应激激活后,ATR通过CHK1的磷酸化,传递DNA损伤信号,使细胞周期停滞在S期或G2/M中,进行损伤修复,使细胞缓解复制应激压力,从而在去除应激压力后重新开始复制。
相比之于健康的增殖细胞,肿瘤细胞中具有较高的DNA损伤和复制应激,若要复制存活,维持细胞的分裂,更依赖于ATR进行DNA修复。因此,通过抑制ATR,可以抑制肿瘤细胞的修复功能,导致DNA损伤和复制应激增加且不能被修复,最终导致肿瘤细胞死亡,同时不影响或较少地影响健康的增殖细胞,这构成ATR抑制剂用于癌症治疗的基础,也使得ATR抑制在近年来已经被认为是重要的癌症治疗途径。
标准癌症疗法如放疗或化疗通过诱发DNA损伤来发挥治疗功效,这对增殖性细胞特别具有毒性,而DNA损伤修复机制的存在会限制这些疗法的功效,从而会产生针对化疗或放疗药物的耐药性。通过抑制ATR,增加复制应激、增加DNA损伤,可增强肿瘤细胞对这些DNA 损伤诱导性疗法的敏感性,有助于克服放疗或化疗药物因损伤修复导致的耐药性,可以用于治疗基因突变或化疗耐药的肿瘤患者,降低化疗或放疗的剂量,从而降低对血液和胃肠道器官系统的毒性。
因此,对于复制应激增加的癌细胞或其他DNA损伤修复通路活性受损或缺陷的癌细胞,可以用ATR抑制剂增加复制应激,诱导肿瘤细胞死亡。的确,研究已揭示,ATR抑制剂对p53突变肿瘤或丧失ATM功能的肿瘤具有合成致死活性,当与多种复制应激/DNA损伤诱导性化疗剂联合使用时具有协同作用,例如铂、电离辐射以及PARP抑制剂。
此外,ATR抑制还可以预防癌症的发生,因为ATR也是DNA损伤检查点的重要成员,ATR抑制会限制癌基因激活导致的原癌细胞的扩增。
近年来,已经开发出一些ATR抑制剂(例如WO2017202748、CN111848605A、WO2020087170、WO2020049017)。但是,开发新的强效ATR抑制剂仍然是非常具有挑战性的。PIKK激酶家族与相关脂质激酶如磷脂酰肌醇3-激酶(PI3KS)以及mTOR的高度同一性增加了抑制其他激酶的风险,这可能会增加毒性或抵消ATR抑制的治疗效果;此外,一些ATR抑制剂的应用还受限于物理化学性质、药动学性质以及药物相互作用。
因此,本领域仍然需要新的选择性ATR抑制剂,其具有增强的ATR抑制活性,特别是具有改进的物理化学性质、改善的代谢稳定性、改善的药动学性质(口服可利用的)、和/或CYP450抑制最小化的ATR抑制剂。
发明简述
本发明人通过研究已经鉴定,本发明的化合物显示令人满意的ATR抑制活性,而且在体内和/或体外药代动力学实验中显示良好的性能,预示着改进的成药性和改进的生物利用度。因此,本发明化合物不仅可实现用于预防或治疗与ATR相关疾病的目的,而且所制备的药物有望具有改善的吸收、在同等剂量下疗效提高、或以更低的剂量提供相同疗效和/或降低可能的副作用。由此,本发明还提供了本发明化合物在制备用于预防或治疗与ATR相关疾病的药物中的用途、包含所述化合物的药物组合物和通过施用所述化合物预防和/或治疗与ATR相关疾病的方法。
因此,在本发明的一方面,提供式(I)化合物、其立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物:
Figure PCTCN2022107932-appb-000001
其中各个基团的定义如发明详述部分所述。
在本发明的另一方面,提供了具有ATR抑制活性、用作药物、尤其是用作ATR抑制剂用于治疗或预防ATR相关疾病的本发明式(I)化合物、其立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物。
在本发明的另一方面,提供了包含本发明化合物和药学上可接受的赋形剂的药物组合物。在一个具体的方面,药物组合物可以另外包含适合与本发明化合物组合使用的另外的治疗活性成分。在一个具体的方面,提供了包含本发明化合物和另外的活性剂的药物组合产品,例如试剂盒。
在本发明的另一方面,提供了本发明的化合物或包含其的药物组合物用于在哺乳动物、特别是人中预防或治疗与ATR相关疾病的用途。
在本发明的另一方面,提供了在体内或体外抑制ATR的方法,该方法包括将有效量的本发明化合物与所述ATR相接触。
在本发明的另一方面,提供了在个体、例如哺乳动物、特别是人中预防或治疗与ATR相关疾病的方法,该方法包括施用有效量的本文所述的本发明化合物或包含其的药物组合物。
在本发明的另一方面,提供了上述本发明的化合物或包含其的药物组合物在制备用于预防或治疗ATR相关疾病的药物中的用途。
在另外的方面,提供了用于合成本发明化合物的方法,其中代表性的合成方案和途径在下文描述。
通过阅读随后的详细描述,本发明的其它目的和优点对于本领域技术人员将变得显而易见。
发明详述
定义
除非另外指出,说明书和权利要求书中使用的各个术语具有以下所示含义。在特定的术语或短语没有特别定义的情况下,不应该被认为是不确定或不清楚的,而是应该按照本领域的普通含义、并结合本文上下文理解。
本文定义的许多基团都是任选被取代的,该定义部分所给出的取代基列表仅仅是示例性的,而非穷举,也不意欲限制本说明书和权利要求书中其他部分所定义的取代基。
本文所用的术语“治疗”是指给患有所述疾病、或者具有所述疾病的症状的受试者、例如哺乳动物、例如人施用一种或多种本文所述的本发明化合物,用以治愈、缓解、减轻或影响所述疾病或所述疾病的症状。在本发明具体的实施方案中,所述疾病是下文所定义的ATR相关的疾病、尤其是肿瘤或癌症。
本文所用的术语“预防”在本领域中是众所周知的,是给怀疑患上或易感于如本文所定 义的ATR相关疾病、尤其是癌症或肿瘤的受试者、例如哺乳动物、例如人施用一种或多种本文所述的本发明化合物,使得罹患所定义疾病的风险降低。术语“预防”包含在诊断或确定任何临床和/或病理症状以前使用本发明的化合物。
本文所用的术语“抑制”和“降低”或这些术语的任何变体,是指生物活性剂的能力,其通过直接或间接与靶点相互作用,降低目标靶点的信号传导活性,且是指目标靶点活性的任何可以测量的减少或完全抑制。例如,与正常情况相比,可以是活性(例如ATR活性)降低量约或至少约5%、10%、15%、20%、25%、30%、35%、40%、45%、50%、55%、60%、65%、70%、75%、80%、85%、90%、95%、99%或更多、或其中可衍生的任何范围。
本文所用的术语“选择性抑制”是指生物活性剂的能力,其通过直接或间接与靶点相互作用,相比脱靶的信号活性,优先降低目标靶点的信号传导活性。就本发明化合物而言,相对于具有高度同一性的其他激酶如PIKK激酶家族与相关脂质激酶如磷脂酰肌醇3-激酶(PI3KS)以及mTOR,其能够选择性抑制ATR的活性,从而减少同时作用于其他激酶可能导致的毒性或对ATR抑制效果的抵消。例如,与对另一种特定激酶相比,本发明对ATR具有至少5%、10%、15%、20%、25%、30%、35%、40%、45%、50%、55%、60%、65%、70%、75%、80%、85%、90%、95%、99%或更多、或其中可衍生的任何范围的更好活性的抑制,或与对另一种特定激酶的活性相比,对ATR具有至少1-、2-、3-、4-、5-、10-、25-、50-、100-、250-或500-倍的更好活性。
本文所用的术语“癌症”或“肿瘤”是指赘生性细胞生长和增殖,无论是恶性的还是良性的,和所有的癌前期细胞和癌细胞和组织。对本发明的化合物、方法、药物组合物、药物组合及用途而言,所述癌症或肿瘤包括但不限于结肠癌、胰腺癌、乳腺癌、卵巢癌、前列腺癌、鳞状细胞癌、基底层细胞癌、腺癌、汗腺癌、皮脂腺癌、肺癌、白血病、膀胱癌、胃癌、子宫颈癌、睾丸癌、皮肤癌、直肠癌、甲状腺癌、肾癌、子宫癌、天疱疮癌、肝癌、听神经瘤、少突神经胶质瘤、脑(脊)膜瘤、成神经细胞瘤、眼癌。
相应地,本文所述的“抗癌作用”或“抗肿瘤作用”包括但不限于对响应速率、疾病进展的时间及存活率的作用。本发明化合物及其医药用途和方法的抗肿瘤作用包括但不限于抑制肿瘤生长、延迟肿瘤生长、消退肿瘤、使肿瘤萎缩、治疗停止后肿瘤再生长时间延长、减缓疾病进展,也包括预防肿瘤发生。
本文所使用的术语“治疗有效量”意指当向个体施用以治疗疾病时足以减轻或完全缓解病症的症状或其它有害作用;逆转、完全停止或减缓病症的进展;或降低病症恶化的风险的量,“有效量”可视化合物、疾病及其严重程度及待治疗的个体的年龄、体重等而变化。
如本文所使用的术语“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
本文所用的术语“ATR相关疾病”是指ATR活性对所述疾病的发生和发展起到促进作用,或抑制ATR将降低疾病的发生率、减少或消除疾病病状的疾病。对于本发明而言,“ATR相关疾病”优选指的是ATR介导的疾病,更进一步优选癌症或肿瘤。如本文所述,ATR激酶抑制剂对于下列疾病应该具有治疗或预防价值:血液恶性肿瘤,例如白血病(包括慢性淋巴细胞性白血病、急性淋巴细胞性白血病、急性髓性白血病和慢性骨髓性白血病)、多发性骨髓瘤、淋巴系统恶性肿瘤(例如淋巴瘤、霍奇金病、非霍奇金淋巴瘤)、骨髓增生异常综合症,以及实体瘤、例如癌和肉瘤及其转移灶,例如乳腺癌、肺癌(非小细胞肺癌、小细胞肺癌、鳞状细胞癌、支气管肺泡癌)、中枢神经系统肿瘤(例如胶质瘤、胚胎期发育不良性神经上皮肿瘤、多形性成胶质细胞瘤、混合胶质瘤、成髓细胞瘤、成视网膜细胞瘤、成神经细胞瘤、生殖细胞瘤及畸胎瘤)、胃肠道癌(例如胃癌、食管癌、肝癌、胆管癌、结直肠癌、小肠癌、胰腺癌)、皮肤癌、黑色素瘤、甲状腺癌、骨癌、头颈癌、唾液腺癌、前列腺癌、睾丸癌、卵巢癌、子宫颈癌、子宫癌、子宫内膜癌、外阴癌、膀胱癌、肾癌、鳞状细胞癌、肉瘤(例如骨肉瘤、软骨肉瘤、平滑肌肉瘤、软组织肉瘤、尤因式肉瘤、胃肠组织癌、胃肠基质肿瘤、卡波西氏肉瘤),以及儿科癌症(例如横纹肌肉瘤和成神经细胞瘤)。
本发明的化合物尤其可用于治疗患有肺癌、前列腺癌、黑色素瘤、卵巢癌、乳腺癌、子宫内膜癌、肾癌、胃癌、肉瘤、头颈癌、中枢神经系统肿瘤及其转移灶、以及患有急性髓性白血病的患者。
本文所用的术语“药物组合物”或“药物制剂”是指包含一种或多种本发明式(I)化合物或者其立体异构体、互变异构体、稳定的同位素衍生物、药学上可接受的盐或溶剂合物和在本领域中通常接受的可药用赋形剂或载体的组合物,且可以为固体、半固体、液体或气态形式。
本文所用的术语“药物组合”是指本发明化合物可与其它活性剂组合用于实现本发明的目的。所述其他活性剂可以是一种或多种另外的本发明化合物,或可以是与本发明化合物相容即不会相互不利影响、或具有互补活性的第二种或另外的(例如第三种)化合物。这类活性剂以达到预期目的的有效量适宜地组合存在。所述其他活性剂可以与本发明化合物在单一药物组合物中共同施用,或与本发明化合物处于不同的离散单元中分别施用,例如以试剂盒形式施用,当分别施用时可以同时或相继进行。所述相继施用在时间上可以是接近或隔远的。
本文所用的术语“药学可接受的”意指由各个国家的相应机构批准的或可由其批准,或列于用于动物且更具体地人类的一般公认药典中,或当向动物例如人类适量施用时不会产生不利、过敏或其它不良反应的分子实体和组合物。
本文所用的术语“药学上可接受的赋形剂或载体”是指一种或多种相容性固体或液体填料或凝胶物质,其药理学上无活性,与组合物中的其它成分相容,并且应是对温血动物如人给药可接受的,用作本发明化合物在施用形式中的载体或介质,其实例包括但不限于纤维素及其衍生物(如羧甲基纤维素钠、醋酸纤维素等)、明胶、滑石、固体润滑剂(如硬脂酸镁)、硫酸钙、植物油、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温类)、润湿剂(如 十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂等。
本文所用的术语“药学可接受的盐”意指药学上可接受且具有母体化合物所需药理学活性的本发明化合物的盐。具体地,此类盐无毒,可为无机酸加成盐或有机酸加成盐及碱加成盐。具体地,此类盐包括:(1)与无机酸形成的酸加成盐,该无机酸如盐酸、氢溴酸、硫酸、硝酸、磷酸等;或与有机酸形成的酸加成盐,该有机酸如乙酸、丙酸、己酸、乙醇酸、丙酮酸、乳酸、丙二酸、丁二酸、苹果酸、马来酸、富马酸、酒石酸、柠檬酸、苯甲酸、肉桂酸、扁桃酸、甲磺酸、乙磺酸、苯磺酸、2-萘磺酸、4-甲苯磺酸、樟脑磺酸、葡糖庚酸、3-苯基丙酸、三甲基乙酸、叔丁基乙酸、月桂基硫酸、葡萄糖酸、谷氨酸、羟基萘甲酸、水杨酸、硬脂酸、黏康酸等;或(2)当存在于母体化合物中的酸性质子经金属离子如碱金属离子、碱土金属离子或铝离子置换时、或与有机碱如乙醇胺、二乙醇胺、三乙醇胺、N-甲基葡糖胺等配位时形成的盐。本领域技术人员将知道制备药用盐的一般原理和技术,例如Berge等,Pharm ScL,66,1-19.(1977)中所述的那些。
本文所用的术语“立体异构体”是指由至少一个不对称中心形成的异构体。在具有一个或多个、例如1、2、3或4个不对称中心的化合物中,可产生外消旋混合物、单一对映异构体、非对映异构体和单独的非对映异构体。特定分子也可以以几何异构体(顺式/反式)存在。本发明化合物还可以以两种或更多种处于快速平衡的结构不同的形式的混合物存在,通常称为互变异构体,代表性实例包括酮-烯醇、苯酚-酮、亚硝基-肟互变异构体等。应当理解,本发明范围涵盖所有这样的异构体以及其任意比例的混合物,例如60%、65%、70%、75%、80%、85%、90%、95%、96%、97%、98%、99%。
本文所用的术语“溶剂合物”是指包含化学计量的或非化学计量的溶剂的溶剂加成形式,包括本发明化合物的任何溶剂化形式,包括例如与水的溶剂合物,例如水合物,或与有机溶剂的溶剂合物,例如甲醇、乙醇或乙腈,即分别作为甲醇化物、乙醇化物或乙腈化物;或为任何多晶型物的形式。应当理解的是,本发明化合物的这类溶剂合物还包括本发明化合物的药学上可接受盐的溶剂合物。
本文所用的术语“前药”意指具有可裂解基团且通过溶剂分解或在生理条件下变成在体内具有药学活性的本发明化合物的化合物。前药包括本领域熟知的酸衍生物,如通过母体酸与适合醇反应制备的酯,或通过母体酸化合物与取代或未取代的胺反应制备的酰胺,或酸酐或混合酸酐。衍生自本发明化合物侧接的酸基的简单脂族或芳族酯、酰胺及酸酐为尤其适用的前药。特定的此类前药为本发明化合物的C 1-8烷基、C 2-8烯基、任选被取代的C 6-10芳基及(C 6- 10芳基)-(C 1-4烷基)酯。
本文所用的术语“同位素变体”是指构成该化合物的一或多个原子上含有非天然比例同位素的化合物。本发明的化合物即可在构成化合物的一个或多个原子上包含非天然比例的原子同位素,从而形成同位素变化形式,其无论是否具有放射性,都旨在涵盖在本发明的范围内。可以掺入本发明化合物中的同位素及其药学上可接受的盐的实例包括(但不限于)氢的同位素(例如2H、3H);碳的同位素(例如11C、13C及14C);氯的同位素(例如36Cl);氟的同位 素(例如18F);碘的同位素(例如123I及125I);氮的同位素(例如13N及15N);氧的同位素(例如15O、17O及18O);磷的同位素(例如32P);及硫的同位素(例如35S)。应当理解,本发明化合物的同位素变化形式通常可以通过常规方法、使用适合试剂的适当同位素变化形式来制备。
本文通式(I)化合物结构式中的
Figure PCTCN2022107932-appb-000002
表示芳族环,即A 1至A 5的选择使得所形成的环满足芳族环的价键理论,且在化学上可行及稳定。
本文化合物结构式或结构片段中使用的
Figure PCTCN2022107932-appb-000003
表示存在立体异构体,且表示不对称中心的绝对构型,在本发明所提供的化合物或中间体的命名中通常以R或S表示。当存在于外消旋混合物中时,该实及虚锲形符号定义的是相对立体化学,而非绝对立体化学。
本文所涉及结构片段中使用的
Figure PCTCN2022107932-appb-000004
指示与其交叉的键是结构片段连接于分子其余部分的键。
本文所用的术语“卤代”或“卤素”意指氟(F)、氯(Cl)、溴(Br)及碘(I)。优选的卤代为氟或氯。
文所用的术语“被卤素取代的”基团旨在包括单卤代或多卤代基团,其中一个或多个(例如2、3、4、5或6个)相同或不同的卤素取代基团中的一个或多个(例如2、3、4、5或6个)氢。
本文所用的术语“氰基”意指基团-CN。
如本文中所使用的术语“羟基”指-OH。
如本文中所使用的术语“氧代”指=O。
如本文中所使用的术语“烷基”指由碳原子和氢原子组成的直链或支链的饱和烃基团。具体地,烷基具有1-10个,例如1至6个、1至5个、1至4个、1至3个或1至2个碳原子。例如,如本文中所使用的术语“C 1-C 6烷基”指具有1至6个碳原子的直链或支链的饱和烃基团,其实例例如甲基、乙基、丙基(包括正丙基和异丙基)、丁基(包括正丁基、异丁基、仲丁基或叔丁基)、戊基(包括正戊基、异戊基、新戊基)、正己基、2-甲基戊基等。特定的烷基具有1至3个碳原子。
本文所用的术语“烷氧基”意指基团-O-烷基,其中烷基具有本文所述的含义。具体地,该术语包括基团-O-C 1-6烷基,更具体的-O-C 1-3烷基。烷氧基的代表性实例包括但不限于甲氧基、乙氧基、丙氧基(包括正丙氧基、异丙氧基)、丁氧基(包括正丁氧基、异丁氧基、叔丁氧基)、戊氧基(包括正戊氧基、异戊氧基、新戊氧基)、己氧基(包括正己氧基、异己氧基)等。特定的烷氧基具有1至3个碳原子。
本文所用的术语“亚烷基”在单独或与其他基团组合使用式是指饱和的直链或直链二价烃基。例如术语“C 1-3亚烷基”是指具有1至3个碳原子的亚烷基,例如亚甲基、亚乙基、亚 丙基、1-甲基亚乙基、2-甲基亚乙基等。
本文所用的术语“环烷基”指具有指定环原子数的单环、稠合多环、桥接多环或螺环非芳族饱和单价烃环结构。环烷基可具有3至12个碳原子(即C 3-C 12环烷基),例如C 3-10环烷基,C 3- 8环烷基,C 3-6环烷基,C 5-6环烷基。适合的环烷基的实例包括但不限于单环结构,如环丙基、环丁基、环戊基、环己基、环庚基或环辛基;或多环(例如双环)结构,包括螺环、稠合或桥连系统,如双环[1.1.1]戊基、双环[2.2.1]庚基、螺[3.4]辛烷基、双环[3.1.1]己烷基、双环[3.1.1]庚基或双环[3.2.1]辛基等。
本文所用的术语“芳基”意指通过自芳族环系统中的单个碳原子移除一个氢原子而衍生的单价芳族烃基。具体地,芳基系指具有指定环原子数的单环或稠合多环芳族环结构。具体地,该术语包括包含6至14个、例如6至10个、优选6个环成员的基团。特定的芳基包括苯基及萘基,最具体的芳基为苯基。
本文所用的术语“取代”或“取代的”是指是指所指定的原子上的一个或多个(例如1、2、3或4个)氢被所指定的基团代替,条件是未超出所指定原子在当前情况下的正常价键并且形成稳定的化合物,取代基和变量的组合仅当这种组合形成稳定的化合物时才是允许的。
本文所用的术语“任选取代的”,除非另外指出,表示基团可以是未取代的或被一个或多个(例如0、1、2、3、4或5或更多)对该基团所列的取代基取代,其中所述取代基可以相同或不同。
除非另有规定,本发明化合物定义中的C n-n+m或C n-C m包括n至n+m个碳的各种情况,例如C 1-6包括C 1、C 2、C 3、C 4、C 5和C 6,也包括n至n+m中的任何一个范围,例如C 0-6包括C 1、C 2、C 3、C 4、C 5、C 6、C 0-1、C 0-2、C 0-3、C 0-4、C 0-5、C 1-2、C 1-3、C 1-4、C 2-3等。同理,本发明化合物定义中的n元至n+m元表示环原子数可以为n至n+m之间的任何一个,也包括n至n+m元的任何一个范围。
如在本说明书和随后的权利要求书中所使用的,词语“包含”、“包括”和“含有”意指“包括但不限于”,并且不排除例如其他添加剂、成分、整数或步骤。应理解的是,该术语包括“由所述成分、步骤或条件组成”或“基本上由所述成分、步骤或条件组成”的技术方案。
应理解,当本文描述本发明化合物、包含其的药物组合物、药物组合以及相关的用途和方法时所涉及的剂量,是基于游离形式的重量,而非基于其任何盐、水合物或溶剂化物等,除非说明书中另外定义。
本发明化合物
本申请通篇使用的术语“发明的化合物”和“本发明的化合物”等,除非另外指出,涵盖本文各个实施方案及其具体或优选实施方式中定义的式(I)化合物、它们的立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物,以及前药。所述立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物以及前药如上文定义部分所 描述。优选地,本发明化合物为式(I)化合物的游离形式或其药学上可接受的盐或溶剂合物;最优选为式(I)化合物的游离形式或其药学上可接受的盐。
本发明的某些化合物可以以多晶型或无定形形式存在,它们也落入本发明的范围内。当为固体结晶形式时,式(I)化合物可以是与另一种化学实体的共晶体形式,并且本说明书包括所有这些共晶体。
在存在手性中心时,本发明的化合物可以以单独的对映异构体或对映体混合物形式存在,本领域技术人员能够确定稳定和可行的本发明化合物的异构体形式。根据一个实施方案,提供了式(I)化合物或其药学上可接受的盐,其是对映体过量(%ee)>95、>98%或>99%的单一对映体。优选地,单一对映异构体以>99%的对映异构体过量(%ee)存在。
本发明化合物还涵盖可能存在的N-氧化物,本领域技术人员能够确定稳定和可行的本发明化合物的N-氧化物。本发明化合物还涵盖本发明化合物的代谢物,即给药本发明化合物时在体内通过氧化、还原、水解、酰胺化、酯化等形成的物质,其可通过本领域公知的技术进行鉴定。
具体地,一方面,本发明提供式(I)化合物、其立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物:
Figure PCTCN2022107932-appb-000005
其中
A 1、A 2和A 5各自独立地是C或N;
A 3和A 4各自独立地是CR 4、N或NR 5
X是O、C(R 6) 2或NR 7
Y是N或CR 8
R 1、R 2和R 3各自独立地是H、-OH、氧代、卤素、CN、-C 1-6烷基或-O-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素或羟基取代;或R 1和R 2连接在一起形成C 1-3亚烷基桥;
R 4是H、氧代、卤素或-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素或羟基取代;
R 5是H或-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素取代;
R 6各自独立地是H、卤素、CN、-OH、-NH 2、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-C 1-6烷基、-O-C 1-6烷基、-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3-6环烷基、-SO-C 1- 6烷基、-SO-C 3-6环烷基、C 6-10芳基或C 3-6环烷基,其中的-C 1-6烷基、C 6-10芳基或C 3-6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的C 1-6烷基取代;
R 7是H、-C 1-6烷基、-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3-6环烷基、-SO-C 1-6烷基或-SO-C 3-6环烷基,其中的-C 1-6烷基或C 3-6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的C 1-6烷基取代;
R 8是H、-OH或卤素;
n和m各自独立地是0至4的整数。
在一种式(I)化合物的实施方案中,A 1、A 2、A 3、A 4和A 5中至少两个是N或NR 5,其余是C或CR 4;优选其中两个是N或NR 5,其余是C或CR 4
在一种式(I)化合物的实施方案中,包含A 1~A 5的六元并五元杂芳基部分具有选自以下的结构:
Figure PCTCN2022107932-appb-000006
在一种式(I)化合物的实施方案中,包含A 1~A 5的六元并五元杂芳基部分选自:
Figure PCTCN2022107932-appb-000007
Figure PCTCN2022107932-appb-000008
优选选自
Figure PCTCN2022107932-appb-000009
Figure PCTCN2022107932-appb-000010
更优选
Figure PCTCN2022107932-appb-000011
Figure PCTCN2022107932-appb-000012
在一种式(I)化合物的实施方案中,包含A 1~A 5的六元并五元杂芳基部分优选是
Figure PCTCN2022107932-appb-000013
在一种式(I)化合物的实施方案中,包含A 1~A 5的六元并五元杂芳基部分优选是
Figure PCTCN2022107932-appb-000014
在一种式(I)化合物的实施方案中,包含A 1~A 5的六元并五元杂芳基部分优选是
Figure PCTCN2022107932-appb-000015
在一种式(I)化合物的实施方案中,包含A 1~A 5的六元并五元杂芳基部分优选是
Figure PCTCN2022107932-appb-000016
在一种式(I)化合物的实施方案中,R 4是H。
在一种式(I)化合物的实施方案中,R 4是氧代或卤素。
在一种式(I)化合物的实施方案中,R 4是-C 1-6烷基,任选被一个或多个卤素或羟基取代; 例如但不限于甲基、乙基、丙基(包括正丙基和异丙基)、丁基(包括正丁基、异丁基、仲丁基或叔丁基)、戊基(包括正戊基、异戊基、新戊基)、正己基、2-甲基戊基、-CH 2F、-CHF 2、-CF 3、-C 2F 5、-CH 2CF 3、-CH 2Cl、-CH 2CH 2CF 3、-CH(CF 3) 2、-CH 2OH或-CH 2CH 2OH。优选R 4是-C 1-3烷基,例如甲基、乙基、丙基、异丙基,最优选甲基。
在前述式(I)化合物的实施方案中,R 4优选是H。
在一种式(I)化合物的实施方案中,R 5是H。
在一种式(I)化合物的实施方案中,R 5是-C 1-6烷基,任选被一个或多个卤素取代;例如但不限于甲基、乙基、丙基(包括正丙基和异丙基)、丁基(包括正丁基、异丁基、仲丁基或叔丁基)、戊基(包括正戊基、异戊基、新戊基)、正己基、2-甲基戊基、-CH 2F、-CHF 2、-CF 3、-C 2F 5、-CH 2CF 3、-CH 2Cl、-CH 2CH 2CF 3或-CH(CF 3) 2
在前述式(I)化合物的实施方案中,R 5优选是C 1-3烷基,例如甲基、乙基、丙基、异丙基,最优选甲基。
在一种式(I)化合物的实施方案中,X是O。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6各自为H。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6之一为H,另一个选自卤素、CN、-OH、-C 1-6烷基、-O-C 1-6烷基、-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3- 6环烷基、-SO-C 1-6烷基、-SO-C 3-6环烷基、C 6-10芳基或C 3-6环烷基,其中的-C 1-6烷基、C 6-10芳基或C 3-6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的-C 1-6烷基取代。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6之一为H,另一个选自卤素,例如氟、氯、溴、碘。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6之一为H,另一个为OH、CN、NH 2、-NH-C 1-6烷基、-N(C 1-6烷基) 2,例如但不限于OH、CN、-NH 2、-NH-CH 3、-NH-CH 2-CH 3、-N(CH 3) 2、-N(CH 2-CH 3) 2、-N(CH 3)(CH 2-CH 3)。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6之一为H,另一个为-C 1-6烷基,任选地被一个或多个卤素、羟基或-O-C 1-6烷基取代,例如但不限于甲基、乙基、丙基(包括正丙基和异丙基)、丁基(包括正丁基、异丁基、仲丁基或叔丁基)、戊基(包括正戊基、异戊基、新戊基)、正己基、2-甲基戊基、-CH 2F、-CHF 2、-CF 3、-C 2F 5、-CH 2CF 3、-CH 2Cl、-CH 2CH 2CF 3、-CH(CF 3) 2、-CH 2OH、-CH 2CH 2OH、-CH 2OCH 3和-CH 2OCH 2CH 3
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6之一为H,另一个为-O-C 1-6烷基,其中的-C 1-6烷基任选地被一个或多个卤素、羟基或-O-C 1-6烷基取代,例如但不限于-OCH 3、-OCH 2CH 3、-OCF 3、-OCH 2CF 3、-OCH 2CH 2OH、-OCH 2CH 2OCH 3
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6之一为H,另一个为-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3-6环烷基、-SO-C 1-6烷基、-SO-C 3-6环烷基、 C 6-10芳基或C 3-6环烷基,例如但不限于-C(O)-CH 3、-C(O)-CH 2CH 3、C(O)-CH 2CH 2CH 3、-C(O)-CH(CH 3) 2、-C(O)-环丙基、-C(O)-环戊基、-C(O)-环己基、-SO 2-CH 3、-SO 2-CH 2CH 3、SO 2-CH 2CH 2CH 3、SO 2-CH(CH 3) 2、-SO 2-环丙基、-SO 2-环戊基、-SO-CH 3、-SO-CH 2CH 3、SO-CH 2CH 2CH 3、SO-CH(CH 3) 2、-SO-环丙基、-SO-环戊基、苯基、环丙基、环丁基、环戊基或环己基,其中的-C 1-6烷基、C 6-10芳基或C 3-6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的-C 1-6烷基取代,取代基例如但不限于氟、氯、溴、碘、-OH、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3) 2、-O-CH 3、-O-CH 2CH 3、-O-CH 2CH 2CH 3、-O CH(CH 3) 2、-CF 3、-CH 2CF 3、-CH 2OH、-CH 2CH 2OH。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6各自独立地为-C 1-6烷基、卤素、CN、-OH、-NH 2、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-O-C 1-6烷基,其中的-C 1-6烷基任选地被一个或多个卤素、羟基或-O-C 1-6烷基取代;
例如但不限于,两个R 6同时为卤素、同时为所述任选被(例如卤素)取代的C 1-6烷基、一个为卤素且另一个为所述任选被(例如卤素)取代的C 1-6烷基、一个为OH或-O-C 1-6烷基且另一个为所述任选被(例如卤素)取代的C 1-6烷基、一个为NH 2、-NH-C 1-6烷基或-N(C 1-6烷基) 2且另一个为所述任选被(例如卤素)取代的C 1-6烷基、一个为CN且另一个为所述任选被(例如卤素)取代的C 1-6烷基;
X例如但不限于CF 2、CCl 2、CBr 2、CFCl、C(CH 3) 2、C(CH 2CH 3) 2、C(CH 3)(CH 2CH 3)、C(CF 3)(CF 3)、C(CH 3)(CF 3)、C(CH 3)(CH 2OH)、C(CH 3)(CH 2OCH 3)、C(CH 3)(F)、C(CH 3)(OCH 3)、C(CH 3)(OH)、C(CH 3)(NH 2)、-C(CH 3)(NHCH 3)-。
在一种式(I)化合物的实施方案中,X是C(R 6) 2,其中R 6各自独立地选自H或卤素,例如均为H,或均为卤素,例如均为F。
在一种式(I)化合物的实施方案中,X是NR 7,R 7是H。
在一种式(I)化合物的实施方案中,X是NR 7,R 7是-C 1-6烷基,任选地被一个或多个卤素、羟基或-O-C 1-6烷基取代,例如但不限于甲基、乙基、丙基(包括正丙基和异丙基)、丁基(包括正丁基、异丁基、仲丁基或叔丁基)、戊基(包括正戊基、异戊基、新戊基)、正己基、2-甲基戊基、-CH 2F、-CHF 2、-CF 3、-C 2F 5、-CH 2CF 3、-CH 2Cl、-CH 2CH 2CF 3、-CH(CF 3) 2、-CH 2OH、-CH 2CH 2OH、-CH 2OCH 3和-CH 2OCH 2CH 3;优选R 7是甲基、乙基、丙基或异丙基。
在一种式(I)化合物的实施方案中,X是NR 7,R 7是-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3-6环烷基、-SO-C 1-6烷基或-SO-C 3-6环烷基,例如但不限于-C(O)-CH 3、-C(O)-CH 2CH 3、C(O)-CH 2CH 2CH 3、-C(O)-CH(CH 3) 2、-C(O)-环丙基、-C(O)-环丁基、-C(O)-环戊基、-C(O)-环己基、-SO 2-CH 3、-SO 2-CH 2CH 3、SO 2-CH 2CH 2CH 3、SO 2-CH(CH 3) 2、-SO 2-环丙基、-SO 2-环丁基、-SO 2-环戊基、-SO 2-环己基、-SO-CH 3、-SO-CH 2CH 3、SO-CH 2CH 2CH 3、SO-CH(CH 3) 2、-SO-环丙基、-SO-环丁基、-SO-环戊基或-SO-环己基,其中的各个-C 1-6烷基或C 3- 6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的C 1- 6烷基取代,取代基例如但不限于氟、氯、溴、碘、-OH、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、- CH(CH 3) 2、-O-CH 3、-O-CH 2CH 3、-O-CH 2CH 2CH 3、-O CH(CH 3) 2、-CF 3、-CH 2CF 3、-CH 2OH、-CH 2CH 2OH、-CH 2OCH 3和-CH 2OCH 2CH 3
在一种式(I)化合物的实施方案中,X是NR 7,R 7选自H、-C 1-6烷基和-C(O)-C 1-6烷基,优选-C 1-6烷基,或优选-C(O)-C 1-6烷基,例如但不限于甲基、乙基、丙基、异丙基、-C(O)-CH 3、-C(O)-CH 2CH 3、C(O)-CH 2CH 2CH 3或-C(O)-CH(CH 3) 2
在前述式(I)化合物的实施方案中,X优选是-O-、-NH-、-N(C 1-6烷基)-、-CH 2-、-C(卤素) 2-,例如-O-、-NH-、-N(CH 3)-、-CH 2-、-C(F) 2-。
在前述式(I)化合物的实施方案中,X优选是-O-、-N(C 1-6烷基)-、-N(CO-C 1-6烷基)-、-CH 2-、-CH(C 1-6烷基)-、-C(C 1-6烷基) 2-或-C(卤素) 2-,例如-O-、-N(CH 3)-、-N(CO-CH 3)-、-CH 2-、-C(F) 2-。
在一种式(I)化合物的实施方案中,Y是N。
在一种式(I)化合物的实施方案中,Y是CR 8,其中R 8是H。
在一种式(I)化合物的实施方案中,Y是CR 8,其中R 8是OH。
在一种式(I)化合物的实施方案中,Y是CR 8,其中R 8是卤素,例如氟、氯、溴、碘,优选F。
在前述式(I)化合物的实施方案中,Y优选是N,或是CR 8,其中R 8是OH;更优选Y是N。
在一种式(I)化合物的实施方案中,包含X和Y的六元环
Figure PCTCN2022107932-appb-000017
选自
Figure PCTCN2022107932-appb-000018
Figure PCTCN2022107932-appb-000019
优选包含X和Y的六元环选自
Figure PCTCN2022107932-appb-000020
更优选包含X和Y的六元环选自
Figure PCTCN2022107932-appb-000021
R 6、R 7和R 8分别具有上述各个相应实施方案所定义的含义,优选R 6选自H、卤素和C 1-6烷基,R 7选自C 1-6烷基和-CO-C 1-6烷基。
在一种式(I)化合物的实施方案中,R 1和R 2各自独立地是H。
在一种式(I)化合物的实施方案中,R 1和R 2各自独立地是-OH、氧代、卤素、CN、-C 1-6烷基或-O-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素或羟基取代,例如但不限于-OH、 氧代、氟、氯、溴、碘、CN、-CH 3、-CH 2CH 3、-O-CH 3、-O-CH 2CH 3、-CF 3、-CH 2CF 3、-CH 2OH、-CH 2CH 2OH,优选-CH 3、-CF 3或-CH 2CH 3,最优选-CH 3
在一种式(I)化合物的实施方案中,R 1和R 2连接在一起形成C 1-3亚烷基桥,例如-CH 2-、-CH 2CH 2-、-CH 2CH 2CH 2-。
在一种式(I)化合物的实施方案中,R 1和R 2连接在一起形成C 1-3亚烷基桥,桥接方式例如但不限于:
Figure PCTCN2022107932-appb-000022
优选
Figure PCTCN2022107932-appb-000023
Figure PCTCN2022107932-appb-000024
其中优选Y是N且X是O。
在前述式(I)化合物的实施方案中,优选R 1和R 2各自独立地是H、-CH 3、-CH 2CH 3,或R 1和R 2连接在一起形成C 1-3亚烷基桥,优选C 2亚乙基桥。
在一种式(I)化合物的实施方案中,m和n均为0。
在一种式(I)化合物的实施方案中,m和n之一为0,另一个为1,此时R 1或R 2不为氢,且可连接于环上Y的邻位或X的邻位,优选连接于Y的邻位。例如,R 1或R 2是C 1-6烷基,例如但不限于-CH 3、-CH 2CH 3或-CH(CH 3) 2,优选R 1或R 2是-CH 3,连接于Y的邻位或X的邻位,优选连接于Y的邻位。
在一种式(I)化合物的实施方案中,m和n之一为0,另一个为2,此时R 1或R 2不为氢,且可同时连接于环上Y的邻位、X的邻位、或分别连接于Y的邻位和X的邻位,优选同时连接于Y的邻位。例如,R 1或R 2各自独立地是C 1-6烷基,例如但不限于-CH 3、-CH 2CH 3或-CH 2-CH 2-CH 3,优选R 1或R 2是-CH 3,同时连接于Y的邻位。
在一种式(I)化合物的实施方案中,m和n之一为0,另一个为3或4,此时R 1或R 2不为氢,该R 1或R 2例如但不限于-CH 3、-CH 2CH 3或-CH 2-CH 2-CH 3,例如该R 1或R 2是C 1-6烷基,优选R 1或R 2是-CH 3
在一种式(I)化合物的实施方案中,m和n均为1,此时R 1和R 2不为氢,各自独立地可连接于环上Y的邻位或X的邻位,例如均连接于Y的邻位,或均连接于X的邻位,或分别连接于Y的邻位和X的邻位;优选R 1和R 2是C 1-6烷基,更优选R 1和R 2是-CH 3,均连接于Y的邻位,或均连接于X的邻位,或分别连接于Y的邻位和X的邻位。
在一种式(I)化合物的实施方案中,m和n均为1,此时R 1和R 2不为氢,连接于、优选同时连接于Y邻位和X邻位的R 1和R 2连接在一起形成C 1-3亚烷基桥,例如-CH 2-、-CH 2CH 2-、-CH 2CH 2CH 2-。
在一种式(I)化合物的实施方案中,m为1且n为2,或m为1且n为3,或m为1且n为4,或m为2且n为2,或m为2且n为3,或m为2且n为4,或m为3且n为4,或 m为4且n为4,其中例如R 1和R 2是C 1-6烷基,优选R 1和R 2是-CH 3
在一种式(I)化合物的实施方案中,m和n之一为0,另一个为1,R 1或R 2各自独立地是C 1-6烷基,任选被一个或多个卤素取代,连接于Y的邻位或X的邻位;
或m和n之一为0,另一个为2,R 1或R 2各自独立地是C 1-6烷基,任选被一个或多个卤素取代,连接于Y的邻位、X的邻位或分别连接于X和Y的邻位;
或m和n均为1且R 1和R 2各自独立地是C 1-6烷基,任选被一个或多个卤素取代,均连接于环上Y的邻位、均连接于X的邻位,或分别连接于Y的邻位和X的邻位。
在一种式(I)化合物的实施方式中,优选m和n之一为0,另一个为1,R 1或R 2是C 1-6烷基,且连接于Y的邻位或X的邻位。
在一种式(I)化合物的实施方案中,m和n之一为0,另一个为1,R 1或R 2是C 1-6烷基,任选被卤素取代,且连接于Y的邻位;或m和n之一为0,另一个为2,R 1或R 2各自独立地是C 1-6烷基,任选被卤素取代,均连接于Y的邻位。
在前述式(I)化合物的实施方案中,在化学上可行的情况下,R 1和/或R 2可以呈R或S构型,优选R构型。
在一种式(I)化合物的实施方案中,其中包含X和Y的六元环例如但不限于:
Figure PCTCN2022107932-appb-000025
优选
Figure PCTCN2022107932-appb-000026
Figure PCTCN2022107932-appb-000027
更优选
Figure PCTCN2022107932-appb-000028
在一种式(I)化合物的实施方案中,R 3是H。
在一种式(I)化合物的实施方案中,R 3是卤素,例如氟、氯、溴、碘,优选氟或氯。
需要说明的是,本发明的化合物涵盖以上各个独立的实施方案或各个具体示例的实施方式,还涵盖上述各个实施方案或具体示例的实施方式的任何组合或亚组合构成的实施方案,也涵盖以上任何优选或示例的实施方案的任何组合所构成的实施方案。
在一种式(I)化合物的实施方案中,
Figure PCTCN2022107932-appb-000029
其中,
包含A 1~A 5的六元并五元杂芳基部分选自:
Figure PCTCN2022107932-appb-000030
包含X和Y的六元环选自
Figure PCTCN2022107932-appb-000031
Figure PCTCN2022107932-appb-000032
R 1和R 2各自独立地是H或-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素取代;或R 1和R 2连接在一起形成C 1-3亚烷基桥;
R 3是H或卤素;
R 4是H;
R 5是H或-C 1-6烷基;
R 6各自独立地是H、卤素、-C 1-6烷基或-O-C 1-6烷基,其中的-C 1-6烷基任选地被一个或多个卤素取代;
R 7是H或-C 1-6烷基,其中的-C 1-6烷基任选地被一个或多个卤素取代;
R 8是H、-OH或卤素;
n和m各自独立地是0至2的整数。
在该实施方案的一种优选实施方式中,包含A 1~A 5的六元并五元杂芳基部分选自
Figure PCTCN2022107932-appb-000033
在该实施方案的一种优选实施方式中,包含X和Y的六元环选自
Figure PCTCN2022107932-appb-000034
Figure PCTCN2022107932-appb-000035
在该实施方案的一种优选实施方式中,包含X和Y的六元环是
Figure PCTCN2022107932-appb-000036
例如包含X和Y的六元环是
Figure PCTCN2022107932-appb-000037
其中R 1各自独立地是C 1-6烷基,任选被卤素取代,n选自0、1或2;或包含X和Y的六元环
Figure PCTCN2022107932-appb-000038
的R 1和R 2连接在一起形成C 1-3亚烷基桥,例如
Figure PCTCN2022107932-appb-000039
在该实施方案的一种优选实施方式中,包含X和Y的六元环是
Figure PCTCN2022107932-appb-000040
例如包含X和Y的六元环是
Figure PCTCN2022107932-appb-000041
其中R 1各自独立地是C 1-6烷基,任选被卤素取代,n选自1或2,R 7选自C 1-6烷基或-CO-C 1-6烷基。
在该实施方案的一种优选实施方式中,包含X和Y的六元环是
Figure PCTCN2022107932-appb-000042
例如包含X和Y的六元环是
Figure PCTCN2022107932-appb-000043
其中R 1各自独立地是C 1-6烷基,任选被卤素取代,n选自0或1,R 6选自H、卤素或被卤素取代的C 1-6烷基。
在该实施方案的一种优选实施方式中,包含X和Y的六元环选自
Figure PCTCN2022107932-appb-000044
在该实施方案的一种优选实施方式中,包含X和Y的六元环选自
Figure PCTCN2022107932-appb-000045
在该实施方案的上述优选实施方式中,R 1和R 2各自独立地是H。
在该实施方案的上述优选实施方式中,R 1和R 2各自独立地是-C 1-6烷基,任选被一个或多个卤素取代,例如-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3) 2、-CF 3或-CH 2CF 3,最优选-CH 3
在该实施方案的一种优选实施方式中,R 1和R 2连接在一起形成C 1-3亚烷基桥,优选C 2亚烷基桥。
在该实施方案的上述优选实施方式中,R 3是H。
在该实施方案的上述优选实施方式中,R 5是-C 1-6烷基,例如-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3) 2,最优选-CH 3
在该实施方案的上述优选实施方式中,R 6各自独立地是H或卤素,优选H或F。
在该实施方案的上述优选实施方式中,R 7是H,或-C 1-6烷基,例如-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3) 2,最优选-CH 3
在该实施方案的上述优选实施方式中,R 8是-OH。
在该实施方案的上述优选实施方式中,n和m之一为0,另一个为1,R 1或R 2连接于环上Y的邻位或X的邻位,优选连接于Y的邻位;例如,R 1或R 2是C 1-6烷基,优选R 1或R 2是-CH 3,连接于Y的邻位或X的邻位,优选连接于Y的邻位。
在该实施方案的上述优选实施方式中,n和m之一为0,另一个为2,R 1或R 2同时连接于环上Y的邻位、X的邻位、或分别连接于Y的邻位和X的邻位,优选同时连接于Y的邻位。例如,R 1或R 2是C 1-6烷基,优选R 1或R 2是-CH 3,同时连接于Y的邻位。
在该实施方案的上述优选实施方式中,n和m均为1,且R 1和R 2各自独立地可连接于环 上Y的邻位,或X的邻位,例如均连接于Y的邻位,或均连接于X的邻位,或分别连接于Y的邻位和X的邻位,优选均连接于Y的邻位,优选R 1和R 2是C 1-6烷基,更优选R 1和R 2是-CH 3,均连接于Y的邻位,或均连接于X的邻位,或分别连接于Y的邻位和X的邻位,优选均连接于Y的邻位;或连接于、优选同时连接于Y邻位和X邻位的R 1和R 2连接在一起形成C 1-3亚烷基桥,例如-CH 2-、-CH 2CH 2-、-CH 2CH 2CH 2-。
在该实施方案的前述优选实施方式中,在化学上可行的情况下,R 1和/或R 2可以呈R或S构型,优选R构型。
需要说明的是,本发明的化合物涵盖以上任何优选或示例的实施方案的任何组合所构成的实施方案。
本发明的化合物的具体实施方式包括以下具体化合物、其立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物,
Figure PCTCN2022107932-appb-000046
Figure PCTCN2022107932-appb-000047
对于下文所述用作药物的本发明化合物、本发明的预防或治疗方法、药物组合物、药物组合或用途而言,优选本文所定义的式(I)化合物的各个优选实施方式,更优选所列的具体化合物。
发明的有益效果
如前文所述,已知ATR激酶在肿瘤发生以及多种其它疾病中发挥作用。我们已令人惊讶地发现,上述式(I)化合物能够强效抑制ATR激酶,从而在遏制和/或治疗实体和/或液体肿瘤疾病方面具有作为抗增生、凋亡和/或抗侵袭药物的价值。特别地,预期本发明化合物可用于 预防或治疗那些对ATR的抑制敏感的肿瘤。另外,预期本发明化合物有用于预防或治疗那些由ATR单独或部分介导的肿瘤。
具体地,经研究发现,本发明的化合物可有效抑制ATR激酶及肿瘤细胞株活性,能够实现以下一种或多种技术效果:
●高的ATR激酶抑制活性:在激酶ATR抑制测定实验中显示IC50在0.1nM~1μM范围,优选0.1nM~0.5μM范围,优选在0.1nM~0.1μM范围,更优选0.1nM~50nM范围、0.1nM~20nM,如活性实施例1所验证;和/或
●高的LOVO细胞株增殖抑制活性,如活性实施例2所验证;和/或
●具有良好的药物代谢动力学性质,例如具有更长的t 1/2,从而例如可以加大给药间隔,更长的半衰期,使患者具有更好的依从性,如活性实施例3所验证;和/或
●具有改善的AUC0-t数据,具有更好的成药性,更高的生物利用度,如下文活性实施例4所验证;和/或
●良好的安全性,如透膜性、P450(减少的药物相互作用风险)、较低的毒性和/或较少的副作用;和/或
●优异的理化性质,如溶解度、物理和/或化学稳定性,如活性实施例5所验证。
基于以上本发明化合物的有益效果,本发明还提供以下各个方面的技术方案。
用于治疗或用作药物的本发明化合物
一方面,本发明提供用作药物、尤其是用作ATR抑制剂、更尤其是用作抗癌剂或抗肿瘤剂的本发明化合物。
另一方面,本发明提供用于治疗和/或预防与ATR相关疾病的本发明化合物。
在具体的实施方式中,本发明提供用于治疗和/或预防ATR对所述疾病的发生和发展起到促进作用或抑制ATR将降低疾病的发生率、减少或消除疾病病状的疾病的本发明化合物,所述疾病例如肿瘤或癌症,包括但不限于:血液恶性肿瘤,例如白血病(包括慢性淋巴细胞性白血病、急性淋巴细胞性白血病、急性髓性白血病和慢性骨髓性白血病)、多发性骨髓瘤、淋巴系统恶性肿瘤(例如淋巴瘤、霍奇金病、非霍奇金淋巴瘤)、骨髓增生异常综合症,以及实体瘤、例如癌和肉瘤及其转移灶,例如乳腺癌、肺癌(非小细胞肺癌、小细胞肺癌、鳞状细胞癌、支气管肺泡癌)、中枢神经系统肿瘤(例如胶质瘤、胚胎期发育不良性神经上皮肿瘤、多形性成胶质细胞瘤、混合胶质瘤、成髓细胞瘤、成视网膜细胞瘤、成神经细胞瘤、生殖细胞瘤及畸胎瘤)、胃肠道癌(例如胃癌、食管癌、肝癌、胆管癌、结直肠癌、小肠癌、胰腺癌)、皮肤癌、黑色素瘤、甲状腺癌、骨癌、头颈癌、唾液腺癌、前列腺癌、睾丸癌、卵巢癌、子宫颈癌、子宫癌、子宫内膜癌、外阴癌、膀胱癌、肾癌、鳞状细胞癌、肉瘤(例如骨肉瘤、软骨肉瘤、平滑肌肉瘤、软组织肉瘤、尤因式肉瘤、胃肠组织癌、胃肠基质肿瘤、卡波西氏肉瘤),以及儿科癌症(例如横纹肌肉瘤和成神经细胞瘤)。
本发明尤其提供可用于治疗患有肺癌、前列腺癌、黑色素瘤、卵巢癌、乳腺癌、子宫内膜癌、肾癌、胃癌、肉瘤、头颈癌、中枢神经系统肿瘤及其转移灶、以及患有急性髓性白血病的患者的式(I)化合物、其立体异构体、互变异构体、稳定的同位素变体、药学上可接受的盐或溶剂合物。
药物组合物及其施用
另一方面,为了实现治疗或预防目的,可以将本发明化合物根据标准药学实践配制为药物组合物。同时,基于本发明化合物良好的药物代谢动力学性质、改善的AUC0-last、良好的成药性,由本发明化合物可制备具有更好的药动学性质、更高生物利用度的药物。
因此,本发明提供一种药物组合物,其包含上述本发明化合物和药学可接受的赋形剂。
选择包含在特定组合物中的赋形剂将取决于多种因素、例如给药方式和所提供的组合物的形式。合适的药学可接受的赋形剂是本领域技术人员熟知的且描述于例如Ansel,Howard C.,等,Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems.Philadelphia:Lippincott,Williams&Wilkins,2004中,包括例如佐剂、稀释剂(例如葡萄糖、乳糖或甘露醇)、载体、pH调节剂、缓冲剂、甜味剂、填充剂、稳定剂、表面活性剂、润湿剂、润滑剂、乳化剂、悬浮剂、防腐剂、抗氧化剂、遮光剂、助流剂、加工助剂、着色剂、加香剂、调味剂、其它已知添加剂。
本发明的药物组合物可以通过本领域技术人员已知的技术来配制,如在Remington’s Pharmaceutical Sciences第20版中公开的技术。
本发明的药物组合物可以以标准方式施用。例如,合适的施用方式包括口服、静脉内、直肠、肠胃外、局部、经皮、眼、鼻、颊或肺(吸入)给药,其中肠胃外输注包括肌肉、静脉内、动脉内、腹膜内或皮下施用。为了这些目的,本发明的化合物可以通过本领域已知的方法配制成例如片剂、胶囊、糖浆、粉末、颗粒、水性或油性溶液或悬浮液、(脂质)乳剂、可分散粉末、栓剂、软膏、乳膏、滴剂、气溶胶、干粉制剂和无菌可注射水性或油性溶液或悬浮液的形式。
本发明化合物的预防或治疗剂量的大小将根据一系列因素而变化,包括所治疗的个体、病症或病况的严重性、给药的速率、化合物的处置及处方医师的判断。对于特定疾病的治疗,有效量是足以改善或减轻与疾病有关的症状的药量。这样的药量可作为单一剂量施用,或者可依据有效的治疗方案施用。一般而言,有效剂量在每日每kg体重约0.0001至约5000mg,例如约0.01至约1000mg/kg/日(单次或分次给药)。对70kg的人而言,这会合计为约0.007mg/日至约7000mg/日,例如约0.7mg/日至约1500mg/日。根据给药模式,本发明化合物在药物组合物中的含量或用量可以是约0.01mg至约1000mg,适合地是0.1-500mg,优选0.5-300mg,更优选1-150mg,特别优选1-50mg,例如1.5mg、2mg、4mg、10mg、25mg等;相应地,本发明的药物组合物将包含0.05至99%w/w(重量百分比),例如0.05至80%w/w,例如0.10至 70%w/w,例如0.10至50%w/w的本发明化合物,所有重量百分比均基于总组合物。应当理解,可能有必要在某些情况下使用超出这些限制的剂量。
在一种具体实施方式中,本发明提供了一种药物组合物,其包含本发明化合物和一种或多种药学可接受的赋形剂,该组合物被配制用于口服施用。该组合物可以以单位剂型提供,例如以片剂、胶囊或口服液体制剂的形式。这样的单位剂型可以含有0.1mg至1g,例如5mg至250mg的本发明化合物作为活性成分。
在一种具体实施方式中,本发明提供了一种药物组合物,其包含本发明化合物和一种或多种药学可接受的赋形剂,该组合物被配制用于局部施用。局部施用可以是以例如乳膏剂、洗剂、软膏剂或透皮贴剂的形式,其中本发明化合物的浓度可以是每克载体约0.01至100mg。
在一种具体实施方式中,本发明提供了一种药物组合物,其包含本发明化合物和一种或多种药学可接受的赋形剂,该组合物被配制用于吸入施用。吸入施用可以通过口服吸入,也可以通过鼻内施用。当通过口服吸入施用时,本发明的化合物可以以每日剂量有效地用于本发明,例如至多500μg,如0.1-50μg、0.1-40μg、0.1-30μg、0.1-20μg或0.1-10μg的本发明化合物。口服吸入的本发明药物组合物可以配制成干粉、悬浮液(在液体或气体中)或溶液(在液体中),且可以以任何合适的形式和使用任何本领域已知的合适的吸入器装置施用,包括例如定量吸入器(MDI)、干粉吸入器(DPI)、喷雾器和软雾吸入器。多室装置可用于递送本说明书的化合物和一种或多种其它活性成分(当存在时)。
在一个具体的实施方式中,本发明的药物组合物可以另外包含适合与本发明化合物组合使用的另外的治疗活性成分。
适合于与本发明化合物组合施用的其他治疗活性成分可以是已知的抗癌药物,特别是其他与DNA损伤和修复机制相关的抗癌药物,包括PARP抑制剂、HDAC抑制剂等。适合于与本发明化合物组合施用的其他治疗活性成分还可以选自与细胞分裂检查点有关的抗癌药物,包括ChK1/2抑制剂、CDK4/6抑制剂、ATM/ATR抑制剂。其他可用于联合使用的已知抗癌药包括烷化剂、拓扑异构酶I/II抑制剂、RNA/DNA抗代谢物、抗有丝分裂剂、抗体药物、激酶抑制剂等。对于联合施用而言,本发明化合物与至少一种已知的抗癌药可作为单一药物组合物施用,也可以作为分离的实体分开、同时或依次施用,例如作为试剂盒施用。
本发明化合物还可以作为生物偶联体施用。该生物偶联体由本发明化合物与至少一种已知有治疗活性的抗体如赫赛汀或美罗华、或生长因子如EGF或FGF、或细胞激素如白细胞介素2或4或任意能与细胞表面结合的分子组成。该抗体和其他分子可把本发明化合物递送至其靶点发挥作用,同时也可提高所述抗体或其他分子的治疗活性。
本发明化合物还可以与放射疗法联合治疗,二者可在相同或不同的时间施用。
本发明提供的上述药物组合物,可用于在例如哺乳动物如人个体中预防或治疗如上所定义的与ATR相关的疾病。
治疗方法和用途
基于上述本发明化合物具有的有益效果,本发明化合物可用于治疗动物、特别是哺乳动物例如人的ATR相关疾病的方法中。
因此,另一方面,本发明提供了调节、尤其是抑制ATR激酶活性的方法,所述方法包括使细胞与如前所述的本发明化合物相接触以调节、尤其是抑制细胞中ATR的活性。
基于同样的性质,本发明还相应地提供一种抑制哺乳动物中异常细胞生长的方法,包括给所述哺乳动物施用治疗有效量的本发明的化合物、或包含本发明化合物的药物组合物。
另一方面,本发明提供了预防或治疗与ATR相关的疾病(例如通过ATR抑制可治疗或预防的疾病)的方法,所述方法包括向需要其的个体施用有效量的如前所述的本发明化合物或包含其的本发明药物组合物。
另一方面,本发明提供了如前所述的本发明化合物或包含其的药物组合物的用途,用于抑制ATR活性,或者用于治疗和/或预防与ATR相关的疾病、例如通过ATR抑制可治疗或预防的疾病。
另一方面,本发明还提供了如前所述的本发明化合物或包含其的药物组合物在制备药物中的用途、尤其是具有ATR激酶抑制剂活性的药物中的应用。
另一方面,本发明提供如前所述的本发明化合物或包含其的药物组合物在制备用于治疗或预防与ATR相关的疾病、例如通过ATR激酶抑制可治疗或预防的疾病的药物中的用途,其中所述化合物或药物组合物任选地与一种或多种化学治疗或免疫治疗联合。
本发明化合物的制备方法
本发明还提供了制备式(I)化合物的方法,下文举例说明了合成本发明化合物的通用合成方案。对于各反应步骤而言,适当的反应条件是本领域技术人员已知的或可以常规确定的。如果没有特别说明,在制备这些化合物中使用的原料和试剂通常可商购获得,或者可以通过下文的方法、与下文给出的方法类似的方法或本领域已知的方法制得。如果需要,合成反应流程中的原料和中间体可以采用常规技术进行分离和纯化,所述技术包括但不限于过滤、蒸馏、结晶、色谱法等。所述材料可以采用包括物理常数和波谱数据在内的常规方法表征。
合成方案1:
Figure PCTCN2022107932-appb-000048
如方案1所示例,本发明化合物可通过包含以下步骤的方法合成:
步骤1:将式(I-1)化合物与胺在碱如DIEA存在下、在溶剂如NMP中加热反应,得到式(I-2)化合物;
步骤2:式(I-2)化合物与胺在碱如DIEA存在下、在溶剂如NMP中加热反应,得到式(I-3)化合物;
步骤3:式(I-3)化合物与碘化试剂如NIS、在溶剂如ACN中于室温反应,得到式(I-4)化合物;
步骤4:式(I-4)化合物通过Suzuki偶联、在偶联剂如Pd(dtbpf)Cl 2/H 3PO 4存在下、在溶剂如二噁烷/水中加热反应,得到式(I-5)化合物;和
步骤5:式(I-5)化合物在酸的作用下脱保护基,得到式(I)化合物。
合成方案2:
Figure PCTCN2022107932-appb-000049
如方案2所示例,本发明化合物可通过包含以下步骤的方法合成:
步骤1:式(II-1)化合物与碘化试剂如NIS、在酸如TFA存在下、在溶剂如CHCl 3中于室温反应,得到式(II-2)化合物;
步骤2:式(II-2)化合物与胺在碱如DIEA存在下、在溶剂如THF中加热反应,得到式(II-3)化合物;
步骤3:式(II-3)化合物通过Suzuki偶联、在偶联剂如Pd(dtbpf)Cl 2/H 3PO 4存在下、在溶剂如二噁烷/水中加热反应,得到式(II-4)化合物;
步骤4:式(II-4)化合物与胺在催化剂如RuPhos-G2作用下、在碱如碳酸铯存在下、在溶剂如甲苯中加热反应,得到式(II-4)化合物;
步骤5:式(II-5)化合物在酸如HCl的作用下脱保护基,得到式(II)化合物;
合成方案3:
Figure PCTCN2022107932-appb-000050
其中R 1、R 2、X如上文对通式(I)所定义:
如方案3所示例,本发明化合物可通过包含以下步骤的方法合成:
步骤1:式(III-1)化合物与氯化试剂如POCl 3加热反应,得到式(III-2)化合物;
步骤2:式(III-2)化合物与胺在碱如K 2CO 3存在下、在溶剂如DMF中于约室温反应,得到式(III-3)化合物;
步骤3:式(III-3)化合物与胺在碱如DIEA存在下、在溶剂如DMF中加热反应,得到式(III-4)化合物;
步骤4:式(III-4)化合物与氰化剂如Zn(CN) 2、在催化剂如Pd 2(dba) 3/DPPF存在下、在溶剂如DMF存在下加热反应,得到式(III-5)化合物;
步骤5:式(III-5)化合物通过还原反应、在催化剂如雷尼镍/氨水存在下、在溶剂如MeOH/THF中反应,得到式(III-6)化合物;
步骤6:式(III-6)化合物通过缩合反应、在缩合剂如HATU/DIEA存在下、在溶剂如DME中反应,得到式(III-7)化合物;
步骤7:式(III-7)化合物在POCl 3存在下加热如在100-150℃关环,得到式(III)化合物。
合成方案4:
Figure PCTCN2022107932-appb-000051
如方案4所示例,本发明化合物可通过包含以下步骤的方法合成:
步骤1:式(IV-1)化合物在碱如LDA的作用下、在溶剂如THF中与甲酸酯如甲酸乙酯在低温反应,得到式(IV-2)化合物;
步骤2:式(IV-2)化合物与3-肼-1H-吡唑在溶剂如乙醇中于室温反应,得到式(IV-3)化合物;
步骤3:式(IV-3)化合物在溶剂如NMP中、在升高的温度如回流下关环反应,得到式(IV-4)化合物;
步骤4:式(IV-4)化合物与胺在溶剂如DMSO中加热反应,得到式(IV-5)化合物;
步骤5:式(IV-5)化合物用保护试剂如DHP/TsOH、在溶剂如DCM中于室温加保护基,得到式(IV-6)化合物;
步骤6:式(IV-6)化合物在偶联剂如Pd(dtbpf)Cl 2/CsCO 3存在下、在溶剂如NMP中加热,与胺偶联,得到式(IV-7)化合物;
步骤7:式(IV-7)化合物脱保护基,得到式(IV)化合物。
上述合成方案只是列举了本发明中部分化合物的制备方法。本发明的化合物或者其立体异构体、互变异构体、稳定的同位素衍生物、药学上可接受的盐或溶剂合物可以通过多种方法、包括上文给出的方法、实施例中给出的方法或与之类似的方法、由本领域普通技术人员在上述合成方案的基础上、结合本领域的常规技术适当改变而制备得到。
具体实施方式
以下结合具体实施例,对本发明的技术方案做进一步的描述,但是本发明的保护范围并不限于这些实施例。凡是不背离本发明构思的改变或等同替代均包括在本发明的保护范围之 内。
下列实施例中未注明具体条件的实验方法,通常按照这类反应的常规条件,或按照制造厂商所建议的条件。下列实施例中未注明手性中心构型时,意味着可以以单独的对映异构体或对映体混合物形式存在,且本领域技术人员能够确定化合物的稳定和可行的异构体形式。除非另外说明,否则百分比和份数分别是重量百分比和重量份数。除非另外说明,否则液体的比为体积比,本发明使用的温度均为摄氏度℃。
以下实施例中所用的实验材料和试剂如无特别说明均可从市售渠道获得、依据现有技术的方法制得或根据与本申请公开的类似的方法制得。除非另有说明,本发明使用的原料都是市售原料,无需进一步纯化可以直接使用,其中以下实施例中使用的5,7-二氯吡唑并[1,5-A]嘧啶购自上海皓鸿生物医药科技有限公司(乐研,CAS:57489-77-7,批次号:Ld102321002),8-溴-6-氯咪唑并[1,2-B]哒嗪购自韶远科技(上海)有限公司(CAS:933190-51-3,批次号:J140-1157-27),8-氧杂-3-氮杂二环[3.2.1]辛烷盐酸盐购自上海毕得医药科技有限公司(CAS:54745-74-3,批次号:BGX458)。
本申请使用的缩写具有本领域通常理解的含义,除非说明书中另外清楚定义。在下面列出说明书中使用的缩写的含义:
Pd 2(dba) 3 三二亚苄基丙酮二钯
Pd(dtbpf)Cl 2 二氯[1,1'-双(二叔丁基膦)二茂铁钯(II)
DPPF 1,1'-双(二苯基膦)二茂铁
RuPhosPdG2 氯(2-二环己基膦基-2',6'-二异丙氧基-1,1'-联苯基)(2-氨基-1,1'-联苯-2-基)钯(II)
HATU 2-(7-偶氮苯并三氮唑)-N,N,N’,N’,-四甲基脲六氟磷酸盐
DIEA N,N-二异丙基乙胺
DCM 二氯甲烷
EA 乙酸乙酯
PE 石油醚
NIS N-碘代丁二酰亚胺
DMF N,N-二甲基甲酰胺
ACN 乙腈
DHP 3,4-二氢-2H-吡喃
NMP N-甲基吡咯烷酮
LC-MS 液相色谱质谱联用
ESI 电喷雾离子化
m/z 质荷比
HPLC 高效液相色谱
合成实施例
本发明提供的目标化合物制备方法中,柱层析色谱采用乳山太阳干燥剂有限公司生产的硅胶(300-400目);薄层色谱采用GF254(0.25毫米);核磁共振色谱(NMR)使用Varian-400核磁共振仪测定;液质联用(LC/MS)使用Agilent TechnologiESI 6120液质联用仪。
此外,凡涉及易氧化或易水解的原料的所有操作都在氮气保护下进行。。
当本发明化合物结构与化合物名称不一致时,通常以结构式所示为准,除非通过上下文可以确定化合物名称正确。
实施例1:3-(7-((R)-3-甲基吗啉代)-3-(1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双 环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000052
步骤1:(R)-4-(5-氯吡唑并[1,5-a]嘧啶-7-基)-3-甲基吗啉的合成
Figure PCTCN2022107932-appb-000053
5,7-二氯吡唑[1,5-a]嘧啶(5.0g,26.6mmol)、(R)-3-甲基吗啉(8.07g,79.8mmol)、DIEA(10.31g,79.8mmol)的NMP(30.0mL)溶液在100℃下搅拌0.5小时后,停止反应。将反应混合物冷却至室温后DCM(150mL)稀释,用饱和食盐水(30.0mL×5)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1-1:2),得到目标化合物(6.50g,收率96.7%,黄色固体)。LC-MS(ESI)m/z 253.1[M+H] +1H NMR(400MHz,CDCl 3)δ8.01(d,J=2.3Hz,1H),6.50(d,J=2.3Hz,1H),6.04(s,1H),5.22–5.08(m,1H),4.13–3.95(m,2H),3.89–3.80(m,1H),3.80–3.75(m,1H),3.74–3.68(m,1H),3.67–3.62(m,1H),1.30(d,J=6.8Hz,3H)。
步骤2:3-(7-((R)-3-甲基吗啉代)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000054
向(R)-4-(5-氯吡唑并[1,5-a]嘧啶-7-基)-3-甲基吗啉(200mg,0.791mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(268mg,1.79mmol)的NMP(15.0mL)溶液中加入DIEA(306mg,2.37mmol)。反应混合液在氮气保护下140℃微波反应器中反应2小时,停止反应。然后向反应液中加入EA(35.0mL),用饱和食盐水(30.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1),得到目标化合物(120mg,产率46.0%,黄色油状物)。LC-MS(ESI)m/z 330.1[M+H] +
步骤3:3-(3-碘-7-((R)-3-甲基吗啉代)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000055
向3-(7-((R)-3-甲基吗啉代)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(120mg,0.364mmol)的乙腈(5.00mL)溶液中加入NIS(82mg,0.364mmol),反应混合液在室温下搅拌反应0.5小时,停止反应后。然后加水(10.0mL),再用EA(20.0mL×3)萃取,合并有机相,用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1),得到目标化合物(100mg,产率60.3%,黄色固体)。LC-MS(ESI)m/z 456.0[M+H] +1H NMR(400MHz,CDCl 3)δ7.85(s,1H),5.38(s,1H),5.10(s,1H),4.51(s,2H),4.08–3.91(m,3H),3.90–3.80(m,2H),3.74(d,J=11.6Hz,1H),3.68–3.57(m,1H),3.40–3.15(m,3H),2.03–1.96(m,2H),1.93–1.83(m,2H),1.25–1.14(m,3H)。
步骤4:3-(7-((R)-3-甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000056
氮气保护下,向3-(3-碘-7-((R)-3-甲基吗啉代)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(100mg,0.219mmol)和1-(四氢-2H-吡喃-2-基)-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(91.6mg,0.329mmol)的1,4-二氧六环(5.00mL)和水(1.00mL)的溶液中加入Pd(dtbpf)Cl 2(14.0mg,0.0219mmol)和磷酸钾(139mg,0.658mmol),反应混合液在60℃下搅拌反应16小时后,停止反应。然后加水(30.0mL),再用EA(20.0mL×3)萃取,合并有机相用饱和食盐水(25.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(100mg,收率94.9%,黄色固体)。LC-MS(ESI)m/z 480.1 [M+H] +
步骤5:3-(7-((R)-3-甲基吗啉代)-3-(1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000057
室温下,向反应瓶中加入3-(7-((R)-3-甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(100mg,0.208mmol)和盐酸乙酸乙酯溶液(5.00mL,3M),反应混合液在室温下搅拌反应2小时,停止反应。减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(75.8mg,产率91.9%,淡黄色固体)。LC-MS(ESI)m/z 396.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ12.62(Brs,1H),8.23(s,1H),7.64–7.47(m,1H),6.77–6.63(m,1H),5.77(s,1H),5.15–5.07(m,1H),4.49–4.44(m,2H),4.19–4.10(m,1H),4.09–4.01(m,1H),3.96–3.91(m,1H),3.85–3.80(m,1H),3.70–3.63(m,2H),3.57–3.50(m,1H),3.40–3.34(m,1H),3.14–3.07(m,2H),1.88–1.81(m,2H),1.79–1.72(m,2H),1.10(d,J=6.8Hz,3H)。
实施例2:3-(8-((R)-3-甲基吗啉代)-3-(1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双 环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000058
步骤1:(8-溴-6-氯-3-碘咪唑并[1,2-b]哒嗪的合成
Figure PCTCN2022107932-appb-000059
向8-溴-6-氯咪唑并[1,2-b]哒嗪(500mg,2.15mmol)的三氯甲烷(10.0mL)和三氟乙酸(1.00mL)溶液中加入NIS(484mg,2.15mmol),反应混合液在室温下搅拌反应2小时,停止反应。然后加入饱和碳酸氢钠溶液(25.0mL),再用EA(30.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(500mg,产率64.8%,黄色固体)。LC-MS(ESI)m/z 357.7,359.7 [M+H] +1H NMR(400MHz,CDCl 3)δ8.07(s,1H),7.56(s,1H)。
步骤2:(R)-4-(6-氯-3-碘咪唑并[1,2-b]哒嗪-8-基)-3-甲基吗啉的合成
Figure PCTCN2022107932-appb-000060
向8-溴-6-氯-3-碘咪唑并[1,2-b]哒嗪(500mg,1.40mmol)和(R)-3-甲基吗啡啉(211mg,2.09mmol)的四氢呋喃(10.0mL)溶液中加入DIEA(541mg,4.19mmol)。反应混合液在氮气保护下100℃搅拌反应2小时,停止反应。然后将反应液加入EA(35.0mL)中,用饱和食盐水(35mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(500mg,产率94.6%,黄色油状物)。LC-MS(ESI)m/z 378.8[M+H] +
步骤3:(3R)-4-(6-氯-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-8-基)-3-甲基吗啉的合成
Figure PCTCN2022107932-appb-000061
氮气保护下,向(R)-4-(6-氯-3-碘咪唑并[1,2-b]哒嗪-8-基)-3-甲基吗啉(400mg,1.06mmol)和1-(四氢-2H-吡喃-2-基)-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(293mg,1.06mmol)的1,4-二氧六环(10.0mL)和水(2.00mL)的溶液中加入Pd(dtbpf)Cl 2(67.3mg,0.105mmol)和磷酸钾(448mg,2.11mmol),反应混合液在室温下搅拌反应16小时,停止反应。然后加水(25.0mL),再用EA(30.0mL×3)萃取,合并有机相用饱和食盐水(25.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(320mg,收率75.1%,黄色固体)。LC-MS(ESI)m/z 403.1[M+H] +
步骤4:3-(8-((R)-3-甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000062
氮气保护下,向(3R)-4-(6-氯-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-8-基)-3-甲基吗啉(300mg,0.744mmol)和RuPhosPdG2(58.1mg,0.0744mmol)的甲苯(15.0mL)溶液中加入碳酸铯(727mg,2.23mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(168mg,1.12 mmol),反应混合液在110℃下搅拌反应16小时,停止反应。然后加水(25.0mL),再用EA(25.0mL×3)萃取,合并有机相用饱和食盐水(25.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(100mg,收率28.0%,黄色固体)。LC-MS(ESI)m/z 480.2[M+H] +
步骤5:3-(8-((R)-3-甲基吗啉代)-3-(1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000063
室温下,向反应瓶中加入3-(8-((R)-3-甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(100mg,0.208mmol)和盐酸的EA溶液(10.0mL,3M),反应混合液在室温下搅拌反应2小时,停止反应。减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(38.7mg,产率47.0%,白色固体)。LC-MS(ESI)m/z 396.3[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.33–12.85(m,1H),7.84–7.72(m,1H),7.14–6.86(m,1H),5.98(s,1H),5.63–5.33(m,1H),4.51–4.43(m,2H),4.15–3.99(m,1H),4.00–3.93(m,1H),3.81–3.71(m,4H),3.66–3.57(m,1H),3.39–3.34(m,1H),3.30–3.25(m,1H),3.07–2.99(m,2H),1.90–1.81(m,4H),1.15(d,J=6.7Hz,3H)。
实施例3:3-(4-((R)-3-甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双 环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000064
步骤1:4-溴-3,6-二氯哒嗪的合成
Figure PCTCN2022107932-appb-000065
将4-溴-1,2-二氢哒嗪-3,6-二酮(5.50g,28.8mmol)溶于三氯氧磷(35.0mL),反应混合液加热至100℃反应16小时,停止反应。待反应液冷却至室温后,将其慢慢倒入冰水中,然后调节pH至7左右,再用EA(100mL×3)萃取,有机相用饱和食盐水(50.0mL×3)洗涤后经无水硫 酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=20:1-5:1),得到目标化合物(5.00g,产率76.2%,淡黄色固体)。LC-MS(ESI)m/z 227.0[M+H] +
步骤2:(R)-4-(3,6-二氯哒嗪-4-基)-3-甲基吗啉的合成
Figure PCTCN2022107932-appb-000066
向4-溴-3,6-二氯哒嗪(2.73g,12.0mmol)和(R)-3-甲基吗啉(2.40g,23.7mmol)的DMF(37.0mL)溶液中加入碳酸钾(6.59g,47.7mmol),氮气保护下,反应混合液在30℃下搅拌反应16小时后,停止反应。加入水(45.0mL),然后用EA(60.0mL×3)萃取,有机相用饱和食盐水(45.0mL×3)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-2:1),得到目标化合物(2.36g,收率79.4%,白色固体)。LC-MS(ESI)m/z 248.1[M+H] +
步骤3:3-(6-氯-5-((R)-3-甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000067
向(R)-4-(3,6-二氯哒嗪-4-基)-3-甲基吗啉(1.44g,5.80mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(1.30g,8.68mmol)的NMP(26.0mL)溶液中加入DIEA(2.26g,17.5mmol),反应混合液在145℃下搅拌16小时后,停止反应。将反应混合物冷却至室温后,加入水(25.0mL),用EA(30.0mL×2)萃取,有机相用饱和食盐水(25.0mL×3)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:1),得到目标化合物(1.50g,收率79.6%,棕色固体)。LC-MS(ESI)m/z 325.2[M+H] +
步骤4:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000068
向3-(6-氯-5-((R)-3-甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.49g,4.59 mmol)的水(8滴)和DMF(40.0mL)混合溶液中依次加入氰化锌(1.08g,9.18mmol)、DPPF(510mg,0.918mmol)和Pd2(dba)3(421mg,0.459mmol)。氮气保护下,反应混合液在145℃搅拌反应过夜后,停止反应。待反应液冷却至室温后加入水(30.0mL),然后用EA(35.0mL×3)萃取,有机相用饱和食盐水(30.0mL×3)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:2),得到目标化合物(1.10g,收率76.0%,棕色固体)。LC-MS(ESI)m/z 316.3[M+H] +
步骤5:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000069
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-甲腈(1.03g,3.27mmol)的四氢呋喃(30.0mL)溶液中依次加入氨水(5.00mL)和雷尼镍(~1.15g),氢气置换三次后反应混合物在氢气氛围下室温反应过夜,停止反应。将反应混合物过滤除去固体残渣、滤液减压浓缩,得到目标化合物(1.00g,收率95.9%,棕色固体)。LC-MS(ESI)m/z 320.3[M+H] +
步骤6:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑5-甲酰胺的合成
Figure PCTCN2022107932-appb-000070
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-基)甲胺(1.20g,3.74mmol)和1H-吡唑-5-甲酸(505mg,4.49mmol)的DMF(28.0mL)溶液中加入HATU(2.85g,7.48mmol)和DIEA(1.19g,9.35mmol),反应混合物在室温下反应4小时,停止反应。将反应混合物加水(15.0mL)稀释,然后用DCM:甲醇=10:1(20.0mL×3)萃取,合并有机相,用饱和食盐水(15.0mL×4)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=50:1-10:1),得到目标化合物(660mg,收率42.7%,白色固体)。LC-MS(ESI)m/z 414.2[M+H] +
步骤7:3-(4-((R)-3-甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000071
将N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑5-甲酰胺(660mg,1.60mmol)溶于三氯氧磷(23.0mL)溶液中,反应混合物在145℃下反应3小时后,停止反应。待反应液冷却至室温后,将反应液减压浓缩除去多余三氯氧磷,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)分离纯化,得到目标化合物(36.8mg,收率5.83%,白色固体)。LC-MS(ESI)m/z 396.3[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.22(brs,1H),7.76–7.54(m,2H),7.11-7.04(m,1H),5.79(s,1H),4.50–4.42(m,2H),4.38–4.30(m,1H),3.99–3.92(m,1H),3.85–3.75(m,3H),3.74–3.68(m,1H),3.65–3.57(m,1H),3.50–3.38(m,2H),3.11–3.00(m,2H),1.90–1.78(m,4H),1.15(d,J=6.6Hz,3H)。
实施例4:3-(3-氯-4-((R)-3-甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮 杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000072
向N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((R)-3-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑5-甲酰胺(6.50g,15.72mmol)的乙腈(100mL)溶液中加入三氯氧磷(25.0mL),反应混合物在90℃下反应1.5小时后,停止反应。待反应混合液冷却至室温后,将反应液慢慢滴加到0℃的饱和碳酸氢钠溶液中,然后在0℃下用饱和碳酸氢钠溶液调节pH至9后,用EA(200mL×3)萃取,合并的有机相用无水硫酸钠干燥,过滤、滤液减压浓缩,所得残留物经两次反相制备色谱(乙腈/水含0.05%氨水)和(乙腈/水含0.05%甲酸)分离纯化,得到目标化合物(1.50g,收率22.2%,绿色固体)。LC-MS(ESI)m/z 430.2[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.74(s,1H),7.64(s,1H),7.17(s,1H),4.50–4.40(m,2H),4.10–4.01(m,1H),4.00–3.91(m,2H),3.84–3.71(m,3H),3.66–3.60(m,1H),3.42–3.35(m,2H),3.34-3.32(m,1H),2.97(d,J=11.8Hz,1H),2.38–2.31(m,1H),2.16–2.08(m,1H),2.01–1.91(m,2H),1.23(d,J=6.5Hz,3H)。
实施例5:3-(4-((R)-3-甲基吗啉代)-1-(1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂 双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000073
中间体3-肼基-1H-吡唑的合成
Figure PCTCN2022107932-appb-000074
将3-氨基吡唑(1.00g,12.0mmol)加入到6mol/L的盐酸水溶液(7mL)中,混合液降温至-5℃,向反应混合物中加入1mol/L的亚硝酸钠水溶液(12mL,12.0mmol),反应液在室温下搅拌反应2小时。向反应混合物中加入氯化亚锡二水化合物(5.43g,24.1mmol)的浓盐酸溶液(24mL),反应液在室温下搅拌1小时后,减压浓缩,得到目标化合物粗产品(9.00g,粗品,亮黄色固体)。
步骤1:2,6-二氟-4-碘-3-吡啶甲醛的合成
Figure PCTCN2022107932-appb-000075
将2,6-二氟-4-碘吡啶(5.00g,20.7mmol)加入到装有无水四氢呋喃(75mL)的三口瓶中。所得混合物在氮气保护下降温至-78℃,加入二异丙基氨基锂(2mol/L的四氢呋喃溶液)(12.5mL,24.9mmol),搅拌1小时后,缓慢加入甲酸乙酯(2.31g,31.1mmol),继续在-78℃下搅拌30分钟后,TLC点板检测原料反应完全,向反应液中加入甲酸(1.91g,41.5mmol),在-78℃搅拌10分钟后,加入EA(25mL),然后升温至0℃,加入水(30mL)。停止搅拌,加入额外的EA(25mL)进行萃取,饱和食盐水洗洗涤,无水硫酸钠干燥后,减压除去溶剂,残余物经硅胶柱分离纯化(PE:EA=5:1),得到目标化合物(3.54g,收率63.4%,黄色固体)。
步骤2:3-((2-(1H-吡唑基-3-基)肼基)甲基)-2,6-二氟-4-碘吡啶的合成
Figure PCTCN2022107932-appb-000076
将2,6-二氟-4-碘-3-吡啶甲醛(1.00g,3.72mmol)和3-肼基-1H-吡唑(9.00g,粗品)加入到95%乙醇水溶液(20mL)中。所得混合物氮气保护下室温搅拌3小时,减压除去溶剂,残余物加入EA(30mL)并搅拌均匀,向搅拌均匀的悬浊液中缓慢滴加饱和碳酸氢钠溶液并剧烈搅拌, 使混合液至碱性,继续搅拌15分钟后,用EA萃取,饱和食盐水洗涤,无水硫酸钠干燥,减压蒸馏后经硅胶柱分离纯化(PE:EA=1:2),得到目标化合物(1.40g,粗品,黄色固体)。LC-MS(ESI)m/z:349.9[M+H] +
步骤3:6-氟-4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶的合成
Figure PCTCN2022107932-appb-000077
将3-((2-(1H-吡唑基-3-基)肼基)甲基)-2,6-二氟-4-碘吡啶(1.40g,4.01mmol)溶解于NMP(19mL)中。反应混合物在加热回流下搅拌反应1小时。LC-MS监测反应结束后,将反应混合物滴加到水(50mL)中,析出棕色固体,混合液在室温下搅拌10分钟后降至0℃,继续搅拌10分钟,悬浊液用抽滤漏斗抽滤,收集固体,水相用EA(10mL×2)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩,得到粗产品。经制备板分离纯化(硅胶,EA:PE=1:1)得到目标化合物(838mg,两步收率68.5%,棕黄色固体)。LC-MS(ESI)m/z:329.9[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.09(s,1H),8.27(s,1H),7.93(t,J=2.0Hz,1H),7.68(d,J=1.2Hz,1H),6.66(t,J=2.2Hz,1H)。
步骤4:3-(4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000078
将6-氟-4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶(538mg,1.63mmol)溶解到二甲基亚砜(3mL)中。所得混合物氮气保护下室温搅拌均匀,加入8-氧杂-3-氮杂二环[3.2.1]辛烷盐酸盐(367mg,2.45mmol),反应液升温至120℃搅拌45分钟,LC-MS监测反应结束后,将反应混合物滴加到水(30mL)中,析出棕黄色固体,混合液在室温下搅拌10分钟后降至0℃,继续搅拌10分钟,悬浊液用抽滤漏斗抽滤,收集固体,水相用EA(10mL×2)萃取。残余物经硅胶柱分离纯化(PE:EA=1:2),得到目标化合物(300mg,收率43.5%,黄色固体)。LC-MS(ESI)m/z:423.0[M+H] +
步骤5:3-(4-碘-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000079
将3-(4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(300mg,0.711mmol)、DHP(120mg,1.42mmol)、对甲苯磺酸一水合物(14mg,0.071mmol)依次加入到DCM(5mL)中。反应混合物在室温下搅拌反应16小时。LC-MS监测反应结束后,向反应混合物中加入水(10mL),用EA(10mL×2)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品。经制备板分离纯化(硅胶,EA:PE=1:2),得到目标化合物(330mg,收率91.7%,无色油状物)。LC-MS(ESI)m/z 507.0[M+H] +
步骤6:3-(4-((R)-3-甲基吗啉代)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000080
将3-(4-碘-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(160mg,0.316mmol)、(R)-3-甲基吗啉(64mg,0.632mmol)、RuPhosPdG2(25mg,0.032mmol)、碳酸铯(309mg,0.948mmol)加入到NMP(2mL)中。所得混合物氮气保护下110℃搅拌2小时,LC-MS监测反应结束后,向反应混合物中加入水(10mL),用EA(10mL×2)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品,经硅胶柱分离纯化(PE:EA=1:2),得到目标化合物(110mg,收率72.6%,无色油状物)。LC-MS(ESI)m/z:480.2[M+H] +
步骤7:3-(4-((R)-3-甲基吗啉代)-1-(1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000081
在3-(4-((R)-3-甲基吗啉代)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(110mg,2.4mmol)的甲醇(2mL)溶液中,加入4mol/L的盐酸1,4-二氧六环溶液(2mL)。所得混合物氮气保护下室温搅拌2小时,减压除去溶剂,残 余物经prep-HPLC制备柱分离纯化,得到目标化合物(12.2mg,收率13.4%,白色固体)。LC-MS(ESI)m/z:396.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ8.20(s,1H),7.80(d,J=2.3Hz,1H),6.80(d,J=2.2Hz,1H),5.82(s,1H),4.52–4.42(m,2H),4.42–4.26(m,1H),4.01–3.84(m,3H),3.80–3.69(m,2H),3.65–3.55(m,2H),3.39–3.27(m,1H),3.07–2.97(m,2H),1.87–1.79(m,2H),1.80–1.70(m,2H),1.16(d,J=6.6Hz,3H).
实施例6:3-(4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-7-(1H-吡唑-3-基)咪唑[1,5-b]哒嗪-2-基)-8- 氧杂-3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000082
步骤1:3-(3,6-二氯哒嗪-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000083
向4-溴-3,6-二氯哒嗪(1.00g,4.39mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(745mg,4.98mmol)的DMF(15.0mL)溶液中加入碳酸钾(1.21g,8.78mmol)。反应混合液在氮气保护下室温下搅拌反应16小时,停止反应。然后将反应液加入EA(35.0mL)中,用饱和食盐水(35.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(700mg,产率61.3%,黄色油状物)。LC-MS(ESI)m/z 260.1[M+H] +
步骤2:3-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯吡啶-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000084
向3-(3,6-二氯哒嗪-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(700mg,2.69mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(609mg,4.07mmol)的NMP(15.0mL)溶液中加入DIEA(1.04g,8.07mmol)。反应混合液在氮气保护下145℃搅拌反应16小时,停止反应。然后将反应液加入EA(35.0mL)中,用饱和食盐水(25.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残 留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(550mg,产率60.7%,黄色固体)。LC-MS(ESI)m/z 337.0[M+H] +
步骤3:4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000085
向3-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯吡啶-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(550mg,1.63mmol)和氰化锌(383mg,3.27mmol)的DMF(10.0mL)溶液中加入DPPF(181mg,0.326mmol)和Pd 2(dba) 3(149mg,0.163mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用EA(25.0mL),用饱和食盐水(20.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(350mg,产率65.5%,黄色固体)。LC-MS(ESI)m/z 328.2[M+H] +
步骤4:(4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000086
向4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-甲腈(350mg,1.07mmol)的四氢呋喃(10.0mL)溶液中加入雷尼镍(~314mg,5.35mmol)。反应混合液在氢气环境下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(300mg,收率84.7%,黄色油状物)。LC-MS(ESI)m/z 332.2[M+H] +
步骤5:N-((4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000087
向(4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-基)甲胺(300mg,0.905mmol)和1H-吡唑-5-甲酸(101mg,0.905mmol)的四氢呋喃(10.0mL)溶液中加入HATU(413mg,1.09mmol)和DIEA(351mg,2.72mmol)。反应混合液在室温下反应1小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=0:1),得到目标化合物(200mg,收率51.9%,黄色油状物)。LC-MS(ESI)m/z 426.0[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.27(s,1H),8.30(s,1H),7.83(s,1H),6.65(s,1H),6.46(s,1H),4.60(d,J=5.1Hz,2H),4.46–4.37(m,4H),3.93–3.86(m,2H),3.00–2.96(m,6H),2.05–2.02(m,2H),1.89–1.80(m,4H),1.76–1.73(m,2H)。
步骤6:3-(4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-7-(1H-吡唑-3-基)咪唑[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000088
室温下向反应瓶中加入N-((4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(200mg,0.470mmol)和三氯氧磷(10.0mL)。反应混合液在100℃下反应1小时,停止反应。将反应液慢慢加入到饱和碳酸氢钠水溶液并用饱和碳酸氢钠水溶液调节pH=7,然后混合液用EA(20.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(21.3mg,产率11.1%,白色固体)。LC-MS(ESI)m/z 408.3[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.39–12.85(s,1H),7.72–7.55(m,2H),7.07(d,J=1.6Hz,1H),5.72(s,1H),4.49-4.42(m,4H),3.81–3.71(m,4H),3.23–3.19(m,2H),3.07–3.01(m,2H),1.91–1.80(m,8H)。
实施例7:3-(4-吗啉代-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛
Figure PCTCN2022107932-appb-000089
步骤1:4-(3,6-二氯哒嗪-4-基)吗啉的合成
Figure PCTCN2022107932-appb-000090
向4-溴-3,6-二氯哒嗪(700mg,3.07mmol)和吗啉(535mg,6.14mmol)的DMF(15.0mL)溶液中加入碳酸钾(1.69g,12.3mmol),氮气保护下,反应混合液在30℃下搅拌反应12小时后,停止反应。加入水(10.0mL),然后用EA(15.0mL×3)萃取,有机相用饱和食盐水(15.0mL×4)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:1),得到目标化合物(700mg,收率97.3%,白色固体)。LC-MS(ESI)m/z 234.0[M+H] +
步骤2:3-(6-氯-5-吗啉代吡啶-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000091
向4-(3,6-二氯哒嗪-4-基)吗啉(700mg,2.99mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(671mg,4.49mmol)的NMP(10.0mL)溶液中加入DIEA(1.16g,8.97mmol),反应混合液在145℃下搅拌16小时后,停止反应。将反应混合物冷却至室温后,加入水(15.0mL),用EA(20.0mL×3)萃取,合并有机相,然后用水(15.0mL×2)和饱和食盐水(15.0mL×3)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1-1:2),得到目标化合物(800mg,收率86.1%,白色固体)。LC-MS(ESI)m/z 311.2[M+H] +
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-吗啉代哒嗪--3-甲腈的合成
Figure PCTCN2022107932-appb-000092
向3-(6-氯-5-吗啉代哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(600mg,1.93mmol)的水(5滴)DMF(10.0mL)混合溶液中依次加入氰化锌(453mg,3.86mmol)、DPPF(214mg,0.386mmol)和Pd 2(dba) 3(177mg,0.193mmol)。氮气保护下,反应混合液在145℃搅拌反应16小时后,停止反应。待反应液冷却至室温后加入水(15.0mL),然后用EA(20.0mL×3)萃取,合并有机相用水(15.0mL×2)和饱和食盐水(10.0mL×2)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:3),得到目标化合物(360mg,收率61.9%,棕色 固体)。LC-MS(ESI)m/z 302.3[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-吗啉代哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000093
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-吗啉代哒嗪-3-甲腈(360mg,1.19mmol)的四氢呋喃(20.0mL)溶液中依次加入氨水(2.00mL)和雷尼镍(~300mg),氢气置换三次后反应混合物在氢气氛围下室温反应过夜,停止反应。将反应混合物过滤除去固体残渣、滤液减压浓缩,得到目标化合物(320mg,收率87.7%,棕色固体)。LC-MS(ESI)m/z 306.1[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-吗啉代哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000094
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-吗啉代哒嗪-3-基)甲胺(320mg,1.05mmol)和1H-吡唑-5-甲酸(118mg,1.05mmol)的四氢呋喃(13.0mL)溶液中加入HATU(599mg,1.58mmol)和DIEA(271mg,2.10mmol),反应混合物在室温下反应2小时,停止反应。反应混合物减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=100:1-30:1),得到目标化合物(170mg,收率40.6%,棕色固体)。LC-MS(ESI)m/z 400.3[M+H] +
步骤6:3-(3-氯-4-吗啉代-7-(1H-吡唑基-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000095
向N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-吗啉代吡啶-3-基)甲基)-1H-吡唑-5-甲酰胺(170mg,0.426mmol)的乙腈(4.00mL)溶液中加入三氯氧磷(1.00mL),反应混合物在90℃下反应1小时后,停止反应。待反应混合液冷却至室温后,将反应液慢慢滴加到0℃的饱和 碳酸氢钠溶液中,然后在0℃下用饱和碳酸氢钠溶液调节pH至9后,用EA(15.0mL×3)萃取,合并的有机相用无水硫酸钠干燥,过滤、滤液减压浓缩,所得残留物经硅胶板(DCM:甲醇=20:1)分离纯化,得到目标化合物(80.0mg,收率45.2%,黄色固体)。LC-MS(ESI)m/z 416.1[M+H] +
步骤7:3-(4-吗啉代-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000096
向3-(3-氯-4-吗啉代-7-(1H-吡唑基-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(80.0mg,0.192mmol)的甲醇(3.00mL)和四氢呋喃(3.00mL)混合溶液中加入钯碳(50.0mg),氢气置换三次后反应混合物在氢气氛围下50℃反应过夜后,停止反应。将反应混合物过滤除去固体残渣、滤液减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)分离纯化,得到目标化合物(4.60mg,收率6.27%,白色固体)。LC-MS(ESI)m/z 382.1[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.83–7.64(m,1H),7.60–7.43(m,1H),7.25–7.02(m,1H),5.84(s,1H),4.55-4.45(m,2H),3.94–3.79(m,6H),3.51–3.43(m,4H),3.23–3.14(m,2H),2.03–1.93(m,4H)。
实施例8:8-(2-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-7-(1H-吡唑-3-基)咪唑[1,5-b]哒嗪-4-基)-3- 氧杂-8-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000097
步骤1:8-(3,6-二氯哒嗪-4-基)-3-氧杂-8-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000098
向4-溴-3,6-二氯哒嗪(1.00g,4.39mmol)和3-氧杂-8-氮杂双环[3.2.1]辛烷盐酸盐(985mg,6.58mmol)的DMF(15.0mL)溶液中加入碳酸钾(1.82g,13.2mmol)。反应混合液在氮气保护下室温下搅拌反应16小时,停止反应。然后将反应液用EA(45.0mL)稀释,用饱和食盐水(35.0 mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(1.00g,产率87.6%,黄色油状物)。LC-MS(ESI)m/z 259.9[M+H] +,261.9[M+2+H] +1H NMR(400MHz,CDCl 3)δ6.72(s,1H),4.44–4.33(m,2H),3.91–3.82(m,2H),3.72–3.62(m,2H),2.21–2.12(m,2H),2.06–1.95(m,2H)。
步骤2:8-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯吡嗪-4-基)-3-氧杂-8-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000099
向8-(3,6-二氯哒嗪-4-基)-3-氧杂-8-氮杂双环[3.2.1]辛烷(1.00g,3.84mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(1.15g,7.69mmol)的NMP(15.0mL)溶液中加入DIEA(1.49g,11.5mmol)。反应混合液在氮气保护下145℃下搅拌反应16小时,停止反应。然后将反应液用EA(45.0mL)稀释,用饱和食盐水(35.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(770mg,产率59.5%,黄色固体)。LC-MS(ESI)m/z 337.0[M+H] +
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)吡啶-3-甲腈的合成
Figure PCTCN2022107932-appb-000100
向8-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯吡嗪-4-基)-3-氧杂-8-氮杂双环[3.2.1]辛烷(750mg,2.23mmol)和氰化锌(523mg,4.45mmol)的DMF(15.0mL)溶液中加入DPPF(247mg,0.445mmol)和Pd 2(dba) 3(204mg,0.222mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用EA(35.0mL)稀释,用饱和食盐水(30.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(600mg,产率82.3%,黄色固体)。LC-MS(ESI)m/z 328.0[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000101
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)吡啶-3-甲腈(600mg,1.83mmol)的四氢呋喃(10.0mL)溶液中加入雷尼镍(~323mg,5.50mmol)。反应混合液在氢气氛围下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(480mg,产率79.0%,黄色油状物)。LC-MS(ESI)m/z 332.1[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000102
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)哒嗪-3-基)甲胺(480mg,1.45mmol)和1H-吡唑-5-甲酸(146mg,1.30mmol)的四氢呋喃(10.0mL)溶液中加入HATU(661mg,1.74mmol)和DIEA(562mg,4.34mmol)。反应混合液在室温下反应1小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=0:1),得到目标化合物(400mg,产率64.9%,黄色油状物)。LC-MS(ESI)m/z 426.0[M+H] +
步骤6:8-(2-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-7-(1H-吡唑-3-基)咪唑[1,5-b]哒嗪-4-基)-3-氧杂-8-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000103
室温下,向反应瓶中加入N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(100mg,0.235mmol)和三氯氧磷(5.00mL)。反应混合液在100℃下反应1小时,停止反应。冰浴下将反应液加入饱和碳酸氢钠水溶液中至pH=7,然后混合液用EA(20.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,减压浓缩,所得残留物经反相制备色谱法(乙腈/水含0.05%甲酸)纯 化,得到目标化合物(10.5mg,产率11.0%,白色固体)。LC-MS(ESI)m/z 408.3[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.70(s,1H),7.58(s,1H),7.13(s,1H),5.78(s,1H),4.56–4.48(m,4H),3.90–3.80(m,4H),3.67–3.61(m,2H),3.23–3.14(m,2H),2.18–2.09(m,4H),2.01–1.94(m,4H)。
实施例9:3-(4-((R)-2-甲基哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂 双环[3.2.1]辛烷甲酸盐
Figure PCTCN2022107932-appb-000104
步骤1:(R)-4-(3,6-二氯哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000105
向4-溴-3,6-二氯哒嗪(2.00g,8.78mmol)和(R)-3-甲基哌嗪-1-羧酸叔丁酯(2.64g,13.2mmol)的DMF(25.0mL)溶液中加入碳酸钾(2.43g,17.6mmol)。反应混合液在氮气保护下室温下搅拌反应16小时,停止反应。然后将反应液用EA(55.0mL)稀释,用饱和食盐水(35.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(1.60g,产率52.5%,黄色固体)。LC-MS(ESI)m/z 346.9[M+H] +1H NMR(400MHz,CDCl 3)δ6.83(s,1H),4.25–4.08(m,2H),3.89–3.79(m,1H),3.46–3.28(m,2H),3.23–3.02(m,2H),1.48(s,9H),1.12(d,J=6.6Hz,3H)。
步骤2:(3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000106
向(R)-4-(3,6-二氯哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯(1.60g,4.61mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(1.38g,9.22mmol)的NMP(15.0mL)溶液中加入DIEA(1.79g,13.8mmol)。反应混合液在氮气保护下145℃下搅拌反应16小时,停止反应。然后将反应液用 EA(55.0mL)稀释,用饱和食盐水(35.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(1.20g,产率61.4%,黄色固体)。LC-MS(ESI)m/z 424.2[M+H] +1H NMR(400MHz,CDCl 3)δ6.11(s,1H),4.54–4.49(m,2H),4.14–3.93(m,2H),3.86–3.81(m,1H),3.77–3.65(m,2H),3.44–3.31(m,2H),3.26–3.10(m,3H),2.90–2.84(m,1H),2.03–1.95(m,2H),1.90–1.82(m,2H),1.48(s,9H),1.02(d,J=6.5Hz,3H)。
步骤3:(3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氰基哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000107
(3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯吡嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯(1.20g,2.83mmol)和氰化锌(665mg,5.66mmol)的DMF(20.0mL)溶液中加入DPPF(314mg,0.556mmol)和Pd 2(dba) 3(259mg,0.283mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用EA(45.0mL)稀释,用饱和食盐水(30.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(1.00g,产率85.2%,黄色固体)。LC-MS(ESI)m/z 415.0[M+H] +
步骤4:(3R)-4-(3-(氨基甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000108
向(3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氰基吡啶-4-基)-3-甲基哌嗪-1-羧酸叔丁酯(500mg,1.21mmol)的四氢呋喃(10.0mL)溶液中加入雷尼镍(~212mg,3.62mmol,)。反应混合液在氢气氛围下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(400mg,收率79.2%,黄色油状物)。LC-MS(ESI)m/z 419.1[M+H] +
步骤5:(3R)-4-(3-((1H-吡唑-5-甲酰胺)甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)吡啶-4-基)-3-甲基哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000109
向(3R)-4-(3-(氨基甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯(400mg,0.956mmol)和1H-吡唑-5-甲酸(96.4mg,0.860mmol)的四氢呋喃(10.0mL)溶液中加入HATU(436mg,1.15mmol)和DIEA(371mg,2.87mmol)。反应混合液在室温下反应1小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=0:1),得到目标化合物(300mg,收率61.2%,黄色油状物)。LC-MS(ESI)m/z 513.1[M+H] +
步骤6:3-(4-((R)-2-甲基哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷甲酸盐的合成
Figure PCTCN2022107932-appb-000110
室温下,向反应瓶中加入(3R)-4-(3-((1H-吡唑-5-甲酰胺)甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)吡啶-4-基)-3-甲基哌嗪-1-羧酸叔丁酯(100mg,0.195mmol)和三氯氧磷(5.00mL)。反应混合液在100℃下反应1小时,停止反应。冰浴下将反应液加入饱和碳酸氢钠水溶液中至pH=7,然后混合液用EA(20.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,减压浓缩,所得残留物经反相制备色谱法(乙腈/水含0.05%甲酸)纯化,得到目标化合物(8.50mg,产率11.0%,白色固体)。LC-MS(ESI)m/z 395.3[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.72(s,1H),7.59(s,1H),7.14(s,1H),5.93(s,1H),4.54–4.49(m,2H),3.93–3.86(m,4H),3.84–3.81(m,1H),3.73–3.59(m,2H),3.53–3.44(m,2H),3.23–3.17(m,2H),2.01–1.93(m,4H),1.32(d,J=6.9Hz,3H)。
实施例10:3-(4-(哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1] 辛烷甲酸盐
Figure PCTCN2022107932-appb-000111
步骤1:4-(3,6-二氯哒嗪-4-基)哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000112
向4-溴-3,6-二氯哒嗪(1.00g,4.39mmol)和叔丁基哌嗪-1-羧酸酯(1.23g,6.58mmol)的DMF溶液(12.0mL)中加入碳酸钾(1.82g,13.2mmol),反应混合液在室温下搅拌反应16小时,停止反应。然后加水(35.0mL),再用EA(30.0mL×3)萃取,合并有机相用饱和食盐水(30.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(1.50g,产率100%,黄色固体)。LC-MS(ESI)m/z 334.5[M+2+H] +
步骤2:4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯哒嗪--4-基)哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000113
向4-(3,6-二氯哒嗪-4-基)哌嗪-1-羧酸叔丁酯(1.48g,4.44mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(997mg,6.66mmol)的NMP溶液(20.0mL)中加入DIEA(1.72g,13.3mmol),反应混合液在145℃下搅拌反应16小时,停止反应。然后加水(35.0mL),再用EA(30.0mL×3)萃取,合并有机相用饱和食盐水(30.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经层析板分离纯化(PE:EA=5:1),得到目标化合物(1.20g,产率65.9%,黄色固体)。LC-MS(ESI)m/z 410[M+H] +
步骤3:4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氰基哒嗪--4-基)哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000114
向4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯哒嗪--4-基)哌嗪-1-羧酸叔丁酯(1.10g,2.68mmol)和氰化锌(630mg,5.37mmol)的DMF(20.0mL)溶液中加入DPPF(297mg,0.537mmol)和Pd 2(dba) 3(246mg,0.268mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用EA(30.0mL)稀释,用饱和食盐水(25.0mL×3)洗涤后用无水硫 酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(600mg,产率55.8%,黄色固体)。LC-MS(ESI)m/z 401.1[M+H] +1H NMR(400MHz,CDCl 3)δ5.89(s,1H),4.54–4.50(m,2H),4.01–3.93(m,2H),3.64–3.61(m,4H),3.35–3.26(m,6H),2.04–1.99(m,2H),1.85–1.79(m,2H),1.48(s,9H)。
步骤4:4-(3-(氨基甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)哒嗪-4-基)哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000115
向4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氰基哒嗪-4-基)哌嗪-1-羧酸叔丁酯(600mg,1.50mmol)的四氢呋喃(12.0mL)溶液中加入雷尼镍(439mg,7.49mmol)和氨水(5滴)。反应混合液在氢气氛围下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(530mg,产率87.5%,黄色油状物)。LC-MS(ESI)m/z 405.1[M+H] +
步骤5:4-(3-((1H-吡唑-5-甲酰胺)甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)哒嗪-4-基)哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000116
氮气保护下,向4-(3-(氨基甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)哒嗪-4-基)哌嗪-1-羧酸叔丁酯(480mg,1.19mmol)和1H-吡唑-5-甲酸(119mg,1.07mmol)的四氢呋喃溶液(10.0mL)中加入HATU(541mg,1.42mmol)和DIEA(306mg,2.37mmol),反应混合液在室温下搅拌反应1小时,停止反应。然后加水(25.0mL),再用EA(20.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经层析板分离纯化(DCM:甲醇=10:1),得到目标化合物(500mg,产率84.5%,黄色固体)。LC-MS(ESI)m/z 499.2[M+H] +
步骤6:3-(4-(哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷甲酸盐的合成
Figure PCTCN2022107932-appb-000117
室温下向反应瓶中加入4-(3-((1H-吡唑-5-甲酰胺)甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)哒嗪-4-基)哌嗪-1-羧酸叔丁酯(150mg,0.301mmol)和三氯氧磷(15.0mL)。反应混合液在100℃下反应1小时,停止反应。冰浴下将反应液加入饱和碳酸氢钠水溶液中至pH=7,然后混合液用EA(25.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(5.66mg,产率4.94%,白色固体)。LC-MS(ESI)m/z 381.0[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.72(s,1H),7.55(s,1H),7.14(s,1H),5.94(s,1H),4.53–4.48(m,2H),3.88–3.82(m,2H),3.70–3.63(m,4H),3.41–3.36(m,4H),3.23–3.17(m,2H),2.01–1.92(m,4H)。
实施例11:3-(4-((R)-2,4-二甲基哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂- 3-氮杂双环[3.2.1]辛烷甲酸盐
Figure PCTCN2022107932-appb-000118
步骤1:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2-甲基哌嗪-1-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000119
室温下,向反应瓶中加入(3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氰基吡啶-4-基)-3-甲基哌嗪-1-羧酸叔丁基酯(500mg,1.21mmol)和盐酸EA(10.0mL,3M),反应混合液在室温下搅拌反应2小时,停止反应。减压浓缩,所得残留物即为目标化合物(400mg,产率94.5%,白色固体)。LC-MS(ESI)m/z 315.1[M+H] +
步骤2:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2,4-二甲基哌嗪-1-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000120
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2-甲基哌嗪-1-基)哒嗪-3-甲腈(400mg,1.27mmol)和多聚甲醛(144mg,3.82mmol)的甲醇(10.0mL)溶液中加入硼氢化钠(115mg,3.82mmol),反应混合液在室温下搅拌反应1小时,停止反应。然后加水(20.0mL),再用EA(20.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(400mg,产率95.7%,白色固体)。LC-MS(ESI)m/z 329.2[M+H] +
步骤3:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2,4-二甲基哌嗪-1-基)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000121
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2,4-二甲基哌嗪-1-基)哒嗪-3-甲腈(400mg,1.22mmol)的四氢呋喃(10.0mL)溶液中加入雷尼镍(214mg,3.65mmol)。反应混合液在氢气氛围下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(300mg,收率74.1%,黄色油状物)。LC-MS(ESI)m/z 333.1[M+H] +
步骤4:N-((6-(8-氧基-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2,4-二甲基哌嗪-1-基)吡嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000122
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2,4-二甲基哌嗪-1-基)哒嗪-3-基)甲胺(300mg,0.902mmol)和1H-吡唑-5-甲酸(91.0mg,0.812mmol)的四氢呋喃(10.0mL)溶液中加入HATU(412mg,1.08mmol)和DIEA(350mg,2.71mmol)。反应混合液在室温下反应1小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(EA=100%),得到目标化合物(200mg,收率51.9%,黄色油状物)。LC-MS(ESI)m/z 427.2[M+H] +
步骤5:3-(3-氯-4-((R)-2,4-二甲基哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000123
室温下向反应瓶中加入N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-((R)-2,4-二甲基哌嗪-1-基)吡嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(100mg,0.234mmol)和三氯氧磷(5.00mL)。反应混合液在100℃下反应1小时,停止反应。冰浴下,将反应液加入饱和碳酸氢钠水溶液中至pH=7,然后混合液用EA(20.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,减压浓缩,所得残留即为目标化合物(100mg,产率96.3%,白色固体)。LC-MS(ESI)m/z 443.3[M+H] +
步骤6:3-(4-((R)-2,4-二甲基哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷甲酸盐的合成
Figure PCTCN2022107932-appb-000124
向3-(3-氯-4-((R)-2,4-二甲基哌嗪-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(100mg,0.226mmol)的甲醇(10.0mL)溶液中加入钯碳(53.5mg,5%)。反应混合液在氢气氛围下60℃反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(12.5mg,产率13.6%,白色固体)。LC-MS(ESI)m/z 409.3[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.72(s,1H),7.63–7.44(m,1H),7.26–6.99(m,1H),5.84(s,1H),4.52–4.49(m,2H),3.95–3.75(m,3H),3.63–3.49(m,2H),3.22–3.11(m,3H),3.06–2.98(m,1H),2.81–2.73(m,1H),2.62–2.54(m,1H),2.52(s,3H),2.00–1.91(m,4H),1.28(d,J=6.4Hz,3H)。
实施例12:3-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-4- 基)-8-氧杂-3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000125
中间体6-氟-4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶的合成参考实施例5
步骤1:3-(4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000126
将6-氟-4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶(718mg,2.18mmol)溶解到二甲基亚砜(7mL)中。所得混合物氮气保护下室温搅拌均匀,加入8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(326mg,2.18mmol),反应液升温至120℃搅拌45分钟,LC-MS监测反应结束后,将反应混合物滴加到水(30mL)中,析出棕黄色固体,混合液在室温下搅拌10分钟后降至0℃,继续搅拌10分钟,悬浊液用抽滤漏斗抽滤,收集固体,水相用EA(10mL×2)萃取,所得粗产物经硅胶柱分离纯化(PE:EA=1:2),得到目标化合物(215mg,收率23.3%,黄色固体)。LC-MS(ESI)m/z:423.0[M+H] +
步骤2:3-(4-碘-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000127
将3-(4-碘-1-(1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(215mg,0.509mmol)、DHP(86mg,1.02mmol)、对甲苯磺酸一水合物(10mg,0.051mmol)依次加入到DCM(2mL)中。反应混合物在室温下搅拌反应过夜。LC-MS监测反应结束后,向反应混合物中加入水(5mL),用EA(10mL×2)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品。经制备板分离纯化(硅胶,EA:PE=1:2),得到目标化合物(210mg,收率81.4%,黄色固体)。LC-MS(ESI)m/z:507.1[M+H] +
步骤3:3-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑基[3,4-b]吡啶-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000128
将3-(4-碘-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑并[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(210mg,0.415mmol)、8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(124mg,0.829mmol)、RuPhosPdG2(32mg,0.042mmol)、碳酸铯(405mg,1.24mmol)加入到1,4-二氧六环(2mL)中。所得混合物氮气保护下110℃搅拌2小时,LC-MS监测反应结束后,向反应混合物中加入水(10mL),用EA(10mL×2)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品,经硅胶柱分离纯化(PE:EA=1:1),得到目标化合物(55mg,收率27.0%,棕色油状物)。LC-MS(ESI)m/z:492.2[M+H] +
步骤4:3-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000129
在3-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑基[3,4-b]吡啶-4-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(55mg,0.112mmol)的甲醇(2mL)溶液中,加入4mol/L的盐酸1,4-二氧六环溶液(2mL)。所得混合物氮气保护下室温搅拌2小时,减压除去溶剂,残余物经prep-HPLC制备柱分离纯化,得到目标化合物(3.3mg,收率7.24%,白色固体)。LC-MS(ESI)m/z:408.2[M+H] +1H NMR(400MHz,MeOD)δ8.31-8.23(m,1H),7.90-7.77(m,1H),6.80(s,1H),4.63–4.49(m,4H),3.92(d,J=12.2Hz,2H),3.81(d,J=11.9Hz,2H),3.48(d,J=11.8Hz,2H),3.35(d,J=12.0Hz,2H),2.09–1.99(m,4H),1.99–1.89(m,4H).
实施例13:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-甲基-3-(1H-吡唑-3-基)-1H-吡唑并[4,3- b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000130
中间体1-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000131
将8-氧杂-3-氮杂二环[3.2.1]辛烷盐酸盐(3.20g,32.6mmol)加入到DCM溶液(60mL)中,混合液降温至0℃,向反应混合物中加入碳酸钾(28.2g,204mmol),反应液在0℃下搅拌反应30分钟后,向反应混合物中加入乙酰氯(8mL,114mmol),反应液在室温下搅拌16小时后,TLC点板监测反应完成(EA:PE=1:1,磷钼酸显色),抽滤除去固体后,减压浓缩,得到目标化合物(3.50g,粗品,黄色油状物)。
步骤1:4-氨基-1-甲基-1H-吡唑-5-甲酸甲酯的合成
Figure PCTCN2022107932-appb-000132
将1-甲基-4-硝基-1H-吡唑-5-甲酸甲酯(5.00g,27.0mmol)加入到甲醇(200mL)中,向反应混合液中加入10%钯碳(100mg),反应液用氢气置换3次后,在氢气氛围中,室温下搅拌反应过夜,LC-MS监测反应完成,垫硅藻土过滤,滤液减压浓缩,得到粗品目标化合物(4.50g,粗品,蓝紫色固体)。LC-MS(ESI)m/z:156.2[M+H] +.
步骤2:4-((1-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)亚乙基)氨基)-1-甲基-1H-吡唑-5-甲酸甲酯的合成
Figure PCTCN2022107932-appb-000133
将4-氨基-1-甲基-1H-吡唑-5-甲酸甲酯(3.48g,22.4mmol)加入到1,2-二氯乙烷溶液(35mL)中,混合液降温至0℃,向反应混合物中加入1-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)乙烷-1-酮(3.48g,22.4mmol),反应液在0℃下搅拌反应10分钟后,向反应混合物中加入三氯氧磷(6.25mL,67.3mmol),反应液在80℃下搅拌0.5小时后,LC-MS监测反应完成,减压浓缩并通过硅胶柱分离(PE:EA=1:1),得到目标化合物(3.56g,产率54.3%,黄色油状物)。LC-MS(ESI)m/z:293.1[M+H] +.
步骤3:5-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-甲基-1H-吡唑并[4,3-b]吡啶-7-醇的合成
Figure PCTCN2022107932-appb-000134
将4-((1-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)亚乙基)氨基)-1-甲基-1H-吡唑-5-甲酸甲酯(3.56g,12.2mmol)溶于DMF(70mL)中,于0℃下缓慢加入1M的双三甲基硅基胺基锂四氢呋喃溶液(36.5mL)。所得混合物在氮气保护下0℃搅拌1小时,LC-MS监测反应后,加入适量氯化铵水溶液淬灭,减压蒸馏后经硅胶柱分离纯化(DCM:MeOH=20:1-10:1),得到目标化合物(2.16g,产率68.1%,黄色油状物)。LC-MS(ESI)m/z:261.2[M+H] +
步骤4:3-(7-氯-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的
Figure PCTCN2022107932-appb-000135
将5-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-甲基-1H-吡唑并[4,3-b]吡啶-7-醇(2.16g,8.30mmol)溶解于乙腈(22mL)中。室温搅拌半小时后,向反应混合物中加入三氯氧磷(2.3mL,24.9mmol),并且在80℃下搅拌反应过夜。LC-MS监测反应结束后,将反应混合物旋干,经硅胶柱分离纯化(硅胶,EA:PE=1:2),得到目标化合物(2.28g,收率98.6%,黄色固体)。LC-MS(ESI)m/z:279.1[M+H] +. 1H NMR(400MHz,DMSO-d 6)δ7.89(s,1H),7.10(s,1H),4.43-4.39(m,2H),4.17(s,3H),3.82(d,J=12.2Hz,2H),2.95(d,J=12.2Hz,2H),1.85–1.76(m,2H),1.79–1.69(m,2H).
步骤5:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000136
将3-(7-氯-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.20g,4.50mmol)溶解到N-甲基吡咯烷酮(5mL)中,加入8-氧杂双环[3.2.1]辛烷8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(537mg,3.59mmol),RuphosPdG2(139mg,0.179mmol),以及碳酸铯(1.75g,5.38mmol)。所得混合物氮气保护下110℃搅拌2小时,LC-MS监测反应结束后,将反应混合物 加入到水(50mL)中,水相用EA(20mL×2)萃取,饱和食盐水(10mL)洗涤。有机相旋干后经硅胶柱分离纯化(DCM:MeOH=20:1),得到目标化合物(245mg,收率38.4%,棕色固体)。LC-MS(ESI)m/z:356.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ7.78(s,1H),6.43(s,1H),4.46–4.30(m,4H),4.15(s,3H),3.79(d,J=12.1Hz,2H),3.10(d,J=11.4Hz,2H),2.99(d,J=11.4Hz,2H),2.90(d,J=11.7Hz,2H),2.13–2.03(m,2H),1.94–1.86(m,2H),1.83–1.71(m,4H)。
步骤6:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-溴-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000137
将3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(95mg,0.267mmol)溶到DCM(5mL)中。反应混合物在氮气保护下加入N-溴代丁二酰亚胺(48mg,0.267mmol),反应液在室温下搅拌10分钟。LC-MS监测反应结束后,向反应混合物中加入水(5mL),用DCM(5mL×3)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品。经制备硅胶板分离纯化(EA:PE=1:2)目标化合物3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-溴-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(67mg,收率57.7%,黄色固体)。LC-MS(ESI)m/z:434.1[M+H] +1H NMR(400MHz,DMSO-d 6)δ6.49(s,1H),4.46–4.36(m,4H),4.12(s,3H),3.87(d,J=12.3Hz,2H),3.10(d,J=11.5Hz,2H),3.04–2.89(m,4H),2.11–2.02(m,2H),1.97–1.85(m,2H),1.83–1.71(m,4H)。
步骤7:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛基)-1-甲基-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑基[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000138
将3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-溴-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(67mg,0.154mmol)溶于1,4-二氧六环(1mL)和水(0.2mL)中,加入1-(四氢-2H-吡喃-2-基)-3-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2-基)-1H-吡唑(215mg,0.772mmol)、Pd(dtbpf)Cl 2(10mg,0.015mmol)和磷酸钾(98mg,0.463mmol)。所得混合物氮气保护下110℃搅拌2小时,LC-MS监测反应结束后,向反应混合物中加入水(5mL),用EA(10mL×2)萃 取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品,经硅胶柱分离纯化(PE:EA=1:2),得到目标化合物(53mg,收率67.9%,棕色油状物)。LC-MS(ESI)m/z:506.2[M+H] +
步骤8:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-甲基-3-(1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000139
在3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛基)-1-甲基-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑基[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(53mg,0.105mmol)的甲醇(2mL)溶液中,加入4mol/L的盐酸1,4-二氧六环溶液(2mL)。所得混合物氮气保护下室温搅拌2小时,LC-MS监测反应结束后,减压除去溶剂,残余物经prep-HPLC制备色谱柱分离纯化,得到目标化合物(2.9mg,收率6.57%,白色固体)。LC-MS(ESI)m/z:422.2[M+H] +1H NMR(400MHz,MeOD)δ7.91-7.75(m,1H),6.90-6.77(m,1H),6.41(s,1H),4.67–4.57(m,2H),4.57–4.49(m,2H),4.27(s,3H),3.81(d,J=11.8Hz,2H),3.58(d,J=12.3Hz,2H),3.48(d,J=11.8Hz,2H),3.37(d,J=12.2Hz,2H),2.22–2.04(m,4H),2.04–1.91(m,4H)。
实施例14:3-(4-(4,4-二氟哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂 双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000140
步骤1:3,6-二氯-4-(4,4-二氟哌啶-1-基)哒嗪的合成
Figure PCTCN2022107932-appb-000141
向4-溴-3,6-二氯哒嗪(1.00g,4.39mmol)和4,4-二氟哌啶(797mg,6.58mmol)的DMF(15.0mL)溶液中加入碳酸钾(1.82g,13.2mmol),氮气保护下,反应混合液在室温下搅拌反应16小 时后,停止反应。加入水(20.0mL),然后用EA(25.0mL×3)萃取,合并有机相用水(20.0mL×2)和饱和食盐水(20.0mL×3)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=10:1-3:1),得到目标化合物(1.10g,收率93.5%,白色固体)。LC-MS(ESI)m/z 268.0[M+H] +
步骤2:3-(6-氯-5-(4,4-二氟哌啶-1-基)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000142
向3,6-二氯-4-(4,4-二氟哌啶-1-基)哒嗪(1.10g,4.10mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(921mg,6.16mmol)的NMP(23.0mL)溶液中加入DIEA(1.59g,12.3mmol),反应混合液在145℃下搅拌16小时后,停止反应。将反应混合物冷却至室温后,加入水(25.0mL),用EA(30.0mL×3)萃取,合并有机相,然后用水(30.0mL×2)和饱和食盐水(30.0mL×2)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=10:1-1:1),得到目标化合物(1.20g,收率84.8%,白色固体)。LC-MS(ESI)m/z 345.0[M+H] +
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(4,4-二氟哌啶-1-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000143
向3-(6-氯-5-(4,4-二氟哌啶-1-基)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(700mg,2.03mmol)的水(3滴)的DMF(15.0mL)混合溶液中依次加入氰化锌(476mg,4.05mmol)、DPPF(225mg,0.406mmol)和Pd 2(dba) 3(186mg,0.203mmol)。氮气保护下,反应混合液在145℃搅拌反应16小时后,停止反应。待反应液冷却至室温后加入水(20.0mL),然后用EA(25.0mL×3)萃取,合并有机相用水(20.0mL×2)和饱和食盐水(20.0mL×2)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1-1:3),得到目标化合物(450mg,收率66.1%,棕色固体)。LC-MS(ESI)m/z 336.3[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(4,4-二氟哌啶-1-基)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000144
向6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(4,4-二氟哌啶-1-基)哒嗪-3-甲腈(450mg,1.34mmol)的四氢呋喃(20.0mL)溶液中依次加入氨水(2.00mL)和雷尼镍(400mg),氢气置换三次后反应混合物在氢气氛围下室温反应过夜,停止反应。将反应混合物过滤除去固体残渣、滤液经减压浓缩、真空干燥,得到目标化合物(450mg,收率98.8%,棕色固体)。LC-MS(ESI)m/z 340.2[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(4,4-二氟哌啶-1-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000145
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(4,4-二氟哌啶-1-基)哒嗪-3-基)甲胺(450mg,1.33mmol)和1H-吡唑-5-甲酸(134mg,1.20mmol)的四氢呋喃(20.0mL)溶液中加入HATU(605mg,1.60mmol)和DIEA(343mg,2.66mmol),反应混合物在室温下反应1小时后,向反应液中加入氢氧化钠(1.60mL,1M)溶液,继续搅拌反应1小时,停止反应。将反应混合物加水(25.0mL)稀释,然后用(DCM:甲醇)=10:1(30.0mL×3)萃取,有机相用饱和食盐水(30.0mL)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=100:1-30:1),得到目标化合物(330mg,收率57.4%,棕色固体)。LC-MS(ESI)m/z 434.3[M+H] +
步骤6:3-(4-(4,4-二氟哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000146
将N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(4,4-二氟哌啶-1-基)哒嗪-3-基)甲基)-1H-吡 唑-5-甲酰胺(330mg,0.761mmol)溶于三氯氧磷(12.0mL)溶液中,反应混合物在120℃下反应1小时后,停止反应。待反应混合液冷却至室温后,将反应液慢慢滴加到0℃的饱和碳酸氢钠溶液中,然后在0℃下用饱和碳酸氢钠溶液调节pH至9后,用EA(40.0mL×3)萃取,合并的有机层用无水硫酸钠干燥,过滤、滤液减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)分离纯化,得到目标化合物(127.46mg,收率40.3%,灰色固体)。LC-MS(ESI)m/z416.1[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.16(Brs,1H),7.71–7.64(m,1H),7.62(s,1H),7.12-7.03(m,1H),5.93(s,1H),4.47(s,2H),3.82(d,J=12.2Hz,2H),3.65–3.59(m,4H),3.06(d,J=11.1Hz,2H),2.22–2.11(m,4H),1.87–1.81(m,4H)。
实施例15:3-(4-((S)-3-甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双 环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000147
步骤1:(S)-4-(3,6-二氯哒 -4-基)-3-甲基吗啉的合成
Figure PCTCN2022107932-appb-000148
向3,4,6-三氯哒嗪(2.00g,10.9mmol)和(S)-3-甲基吗啉(1.65g,16.3mmol)的NMP(20.0mL)溶液中加入碳酸钾(4.52g,32.7mmol),氮气保护下,反应混合液在室温下搅拌反应16小时后,停止反应。加入水(25.0mL),然后用EA(30.0mL×3)萃取,合并有机相用水(20.0mL×2)和饱和食盐水(20.0mL×3)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=10:1-3:1),得到目标化合物(1.70g,收率62.8%,淡黄色固体)。LC-MS(ESI)m/z 247.9[M+H] +
步骤2:3-(6-氯-5-((S)-3-甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000149
向(S)-4-(3,6-二氯哒 -4-基)-3-甲基吗啉(900mg,3.63mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(814mg,5.44mmol)的NMP(15.0mL)溶液中加入DIEA(1.41g,10.9mmol),反应混合液在145℃下搅拌16小时后,停止反应。将反应混合物冷却至室温后,加入水(20.0mL),用EA(25.0mL×3)萃取,合并有机相,然后用水(20.0mL×2)和饱和食盐水(20.0mL×2)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:1),得到目标化合物(770mg,收率65.4%,白色固体)。LC-MS(ESI)m/z 325.3[M+H] +
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((S)-3-甲基吗啉代)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000150
向3-(6-氯-5-((S)-3-甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(770mg,2.37mmol)的水(3滴)和DMF(15.0mL)混合溶液中依次加入氰化锌(557mg,4.74mmol)、DPPF(263mg,0.474mmol)和Pd 2(dba) 3(217mg,0.237mmol)。氮气保护下,反应混合液在145℃搅拌反应16小时后,停止反应。待反应液冷却至室温后加入水(20.0mL),然后用EA(25.0mL×3)萃取,合并有机相用水(20.0mL×2)和饱和食盐水(20.0mL×2)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1-1:4),得到目标化合物(670mg,收率89.6%,棕色固体)。LC-MS(ESI)m/z 316.1[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((S)-3-甲基吗啉代)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000151
向6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((S)-3-甲基吗啉代)哒嗪-3-甲腈(670mg,2.12mmol)的四氢呋喃(15.0mL)溶液中依次加入氨水(1.50mL)和雷尼镍(500mg),氢气置换三次后反应混合物在氢气氛围下室温反应过夜,停止反应。将反应混合物过滤除去固体残渣、滤液经减压浓缩、真空干燥,得到目标化合物(630mg,收率92.8%,棕色固体)。LC-MS(ESI)m/z320.3[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((S)-3-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑5-甲酰胺的合成
Figure PCTCN2022107932-appb-000152
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((S)-3-甲基吗啉代)哒嗪-3-基)甲胺(330mg,1.03mmol)和1H-吡唑-5-甲酸(116mg,1.03mmol)的四氢呋喃(15.0mL)溶液中加入HATU(471mg,1.24mmol)和DIEA(267mg,2.06mmol),反应混合物在室温下反应1小时后,向反应液中加入氢氧化钠(1mol/L)溶液,继续搅拌反应1小时,停止反应。将反应混合物加水(20.0mL)稀释,然后用(DCM:甲醇)=10:1(25.0mL×2)萃取,有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=100:1-20:1),得到目标化合物(275mg,收率64.4%,棕色固体)。LC-MS(ESI)m/z 414.3[M+H] +
步骤6:3-(4-((S)-3-甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000153
将N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-((S)-3-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑5-甲酰胺(275mg,0.665mmol)溶于三氯氧磷(12.0mL)溶液中,反应混合物在120℃下反应1小时后,停止反应。待反应混合液冷却至室温后,将反应液慢慢滴加到0℃的饱和碳酸氢钠溶液中,然后在0℃下用饱和碳酸氢钠溶液调节pH至9后,用EA(30.0mL×3)萃取,合并的有机层用无水硫酸钠干燥,过滤、滤液减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)分离纯化,得到目标化合物(112.12mg,收率42.6%,白色固体)。LC-MS(ESI)m/z 396.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.20(Brs,1H),7.72–7.61(m,2H),7.08(s,1H),5.78(s,1H),4.49–4.43(m,2H),4.38–4.31(m,1H),3.99–3.92(m,1H),3.84–3.75(m,3H),3.74–3.68(m,1H),3.65–3.57(m,1H),3.49–3.39(m,2H),3.10–3.02(m,2H),1.88–1.80(m,4H),1.15(d,J=6.6Hz,3H)。
实施例16:3-(4-顺-2,6-二甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮 杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000154
步骤1:4-(3,6-二氯哒嗪-4-基)-顺-2,6-二甲基吗啉的合成
Figure PCTCN2022107932-appb-000155
向3,4,6-三氯哒嗪(3.00g,16.4mmol)和顺-2,6-二甲基吗啉(3.01g,26.1mmol)的NMP(30.0mL)溶液中加入碳酸钾(6.78g,49.1mmol),氮气保护下,反应混合液在室温下搅拌反应16小时后,停止反应。加入水(35.0mL),然后用EA(40.0mL×3)萃取,合并有机相用水(30.0mL×2)和饱和食盐水(30.0mL×2)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=10:1-3:1),得到目标化合物(3.70g,收率86.3%,白色固体)。LC-MS(ESI)m/z 262.2[M+H] +
步骤2:3-(6-氯-5-(顺-2,6-二甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000156
向4-(3,6-二氯哒-4-基)-顺2,6-二甲基吗啉(1.00g,3.81mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(913mg,6.10mmol)的NMP(15.0mL)溶液中加入DIEA(1.48g,11.4mmol),反应混合液在145℃下搅拌16小时后,停止反应。将反应混合物冷却至室温后,加入水(25.0mL),用EA(30.0mL×3)萃取,合并有机相,然后用水(25.0mL×2)和饱和食盐水(25.0mL×2)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:1),得到目标化合物(1.00g,收率77.3%,棕色固体)。LC-MS(ESI)m/z 339.3[M+H] +
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(顺-2,6-二甲基吗啉代)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000157
向3-(6-氯-5-(顺-2,6-二甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.00g,2.95mmol)的水(5滴)的DMF(20.0mL)混合溶液中依次加入氰化锌(695mg,5.92mmol)、DPPF(328mg,0.592mmol)和Pd 2(dba) 3(271mg,0.295mmol)。氮气保护下,反应混合液在145℃搅拌反应16小时后,停止反应。待反应液冷却至室温后加入水(20.0mL),然后用EA(25.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL×4)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:3),得到目标化合物(860mg,收率88.5%,棕色固体)。LC-MS(ESI)m/z 330.3[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(顺-2,6-二甲基吗啉代)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000158
向6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(顺-2,6-二甲基吗啉代)哒嗪-3-甲腈(860mg,2.61mmol)的四氢呋喃(20.0mL)溶液中依次加入氨水(2.00mL)和雷尼镍(~600mg),氢气置换三次后反应混合物在氢气氛围下室温反应过夜,停止反应。将反应混合物过滤除去固体残渣、滤液经减压浓缩、真空干燥,得到目标化合物(710mg,收率81.6%,棕色固体)。LC-MS(ESI)m/z334.3[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(顺-2,6-二甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000159
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(顺-2,6-二甲基吗啉代)哒嗪-3-基)甲胺(350mg,1.05mmol)和1H-吡唑-5-甲酸(118mg,1.05mmol)的四氢呋喃(15.0mL)溶液中加入HATU(479mg,1.26mmol)和DIEA(271mg,2.10mmol),反应混合物在室温下反应1小时后,向反应液中 加入氢氧化钠(1mol/L)溶液,继续搅拌反应1小时,停止反应。将反应混合物加水(20.0mL)稀释,然后用(DCM:甲醇)=10:1(25.0mL×3)萃取,有机相用饱和食盐水(20.0mL×2)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(甲醇:DCM=5%),得到目标化合物(280mg,收率62.4%,棕色固体)。LC-MS(ESI)m/z 428.4[M+H] +
步骤6:3-(4-(顺-2,6-二甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000160
将N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛-3-基)-4-(顺-2,6-二甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(150mg,0.351mmol)溶于三氯氧磷(6.00mL)溶液中,反应混合物在120℃下反应1小时后,停止反应。待反应混合液冷却至室温后,将反应液慢慢滴加到0℃的饱和碳酸氢钠溶液中,然后在0℃下用饱和碳酸氢钠溶液调节pH至9后,用EA(30.0mL×3)萃取,合并的有机层用无水硫酸钠干燥,过滤、滤液减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)分离纯化,得到目标化合物(54.42mg,收率37.9%,白色固体)。LC-MS(ESI)m/z410.4[M+H] +1H NMR(400MHz,DMSO-d 6)δ13.12(Brs,1H),7.68-7.62(m,2H),7.07(d,J=1.8Hz,1H),5.85(s,1H),4.47(s,2H),3.90–3.74(m,6H),3.09–3.02(m,2H),2.66–2.57(m,2H),1.88–1.81(m,4H),1.19(d,J=6.2Hz,6H)。
实施例17:3-(4-(哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1] 辛烷
Figure PCTCN2022107932-appb-000161
步骤1:3,6-二氯-4-(哌啶-1-基)哒嗪的合成
Figure PCTCN2022107932-appb-000162
向3,4,6-三氯哒嗪(2.00g,10.9mmol)和哌啶(1.39g,16.4mmol)的NMP(25.0mL)溶液中加入碳酸钾(3.01g,21.8mmol)。反应混合液在氮气保护下室温下搅拌反应16小时,停止反应。然后将反应液用EA(45.0mL)稀释,用饱和食盐水(45.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(2.30g,产率90.9%,黄色固体)。LC-MS(ESI)m/z 232.1[M+H] +
步骤2:3-(6-氯-5-(哌啶-1-基)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000163
向3,6-二氯-4-(哌啶-1-基)哒嗪(3.00g,12.9mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(3.87g,25.8mmol)的NMP溶液(60.0mL)中加入DIEA(6.75mL,38.7mmol),反应混合液在145℃下搅拌反应16小时,停止反应。然后加水(50.0mL),再用EA(40.0mL×3)萃取,合并有机相用饱和食盐水(30.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经层析柱分离纯化(PE:EA=5:1),得到目标化合物(1.20g,产率30.1%,黄色固体)。LC-MS(ESI)m/z 309.1[M+H] +1H NMR(400MHz,CDCl 3)δ6.12(s,1H),4.52–4.46(m,2H),3.82–3.75(m,2H),3.20–3.18(m,1H),3.18–3.15(m,1H),3.13–3.07(m,4H),2.01–1.94(m,2H),1.88–1.82(m,2H),1.76–1.71(m,4H),1.66–1.58(m,2H)。
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(哌啶-1-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000164
向3-(6-氯-5-(哌啶-1-基)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.20g,3.89mmol)和氰化锌(912mg,7.77mmol)DMF(25.0mL)溶液中加入DPPF(431mg,0.777mmol)和Pd 2(dba) 3(356mg,0.388mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用EA(40.0mL)稀释,用饱和食盐水(35.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(1.20g,产率100%,黄色固体)。LC-MS(ESI)m/z 300.2[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(哌啶-1-基)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000165
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(哌啶-1-基)哒嗪-3-甲腈(1.2g,4.01mmol)的四氢呋喃(20.0mL)溶液中加入雷尼镍(~1.18g,20.0mmol)和氨水(5滴)。反应混合液在氢气氛围下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(1.10g,收率90.4%,黄色油状物)。LC-MS(ESI)m/z 304.1[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(哌啶-1-基)吡啶-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000166
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(哌啶-1-基)哒嗪-3-基)甲胺(500mg,1.65mmol)和1H-吡唑-5-甲酸(166mg,1.48mmol)的四氢呋喃溶液(12.0mL)中加入HATU(752mg,1.98mmol)和DIEA(639mg,4.94mmol),反应混合液在室温下搅拌反应45分钟,停止反应。向反应液中滴加氢氧化钠溶液搅拌2分钟,减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=10:1),得到目标化合物(300mg,产率45.8%,黄色固体)。LC-MS(ESI)m/z 398.2[M+H] +
步骤6:3-(4-(哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000167
室温下向反应瓶中加入N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(哌啶-1-基)吡啶-3-基)甲基)-1H-吡唑-5-甲酰胺(150mg,0.327mmol)和三氯氧磷(9.00mL)。反应混合液在100℃下反应1小时,停止反应。将反应液滴加到饱和碳酸氢钠水溶液中并调节pH=7,然后混合液用EA(20.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(51.7mg,产率36.1%,白色固体)。LC-MS(ESI)m/z 380.1[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.69(s,1H),7.47 (s,1H),7.10(s,1H),5.77(s,1H),4.51–4.48(m,2H),3.83–3.79(m,2H),3.52–3.48(m,4H),3.21–3.16(m,2H),1.99–1.95(m,4H),1.79–1.74(m,6H)。
实施例18:3-(4-(2-甲基吗啉代)-7-(1H-吡唑基-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双 环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000168
步骤1:4-(3,6-二氯哒嗪-4-基)-2-甲基吗啉的合成
Figure PCTCN2022107932-appb-000169
向3,4,6-三氯哒嗪(2.00g,10.9mmol)和2-甲基吗啉(1.32g,13.1mmol)的NMP(25.0mL)溶液中加入碳酸钾(3.01g,21.8mmol)。反应混合液在氮气保护下室温下搅拌反应16小时,停止反应。然后将反应液用EA(45.0mL)稀释,用饱和食盐水(45.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(2.30g,产率85.0%,黄色固体)。LC-MS(ESI)m/z 248.1[M+H] +
步骤2:3-(6-氯-5-(2-甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000170
向4-(3,6-二氯哒嗪-4-基)-2-甲基吗啉(2.30g,9.27mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(2.08g,13.9mmol)的NMP(25.0mL)溶液中加入DIEA(2.40g,18.5mmol)。反应混合液在氮气保护下145℃搅拌反应16小时,停止反应。然后将反应液用EA(45.0mL)稀释,用饱和食盐水(45.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(2.00g,产率76.4%,黄色固体)。LC-MS(ESI)m/z 225.1[M+H] +1H NMR(400MHz,CDCl 3)δ6.13(s,1H),4.55–4.48(m,2H),4.01–3.94(m,1H),3.89–3.80(m,4H),3.50–3.43(m,2H),3.25–3.19(m,2H),2.89–2.80(m,1H),2.57–2.49(m,1H),2.04–1.96(m,2H),1.91–1.82(m,2H),1.24(d,J=6.3Hz,3H)。
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(2-甲基吗啉代)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000171
向3-(6-氯-5-(2-甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.80g,5.54mmol)和氰化锌(1.30g,11.1mmol)DMF(35.0mL)溶液中加入DPPF(614mg,1.11mmol)和Pd 2(dba) 3(507mg,0.554mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用EA(55.0mL)稀释,用饱和食盐水(40.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(1.50g,产率85.8%,黄色固体)。LC-MS(ESI)m/z 316.0[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(2-甲基吗啉代)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000172
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(2-甲基吗啉代)哒嗪-3-甲腈(1.50g,4.76mmol)的四氢呋喃(30.0mL)溶液中加入雷尼镍(~1.40g,23.8mmol)。反应混合液在氢气氛围下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=10:1),得到目标化合物(1.50g,收率98.7%,黄色固体)。LC-MS(ESI)m/z 320.0[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(2-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000173
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(2-甲基吗啉代)哒嗪-3-基)甲胺(500mg,1.57mmol)和1H-吡唑-5-甲酸(158mg,1.41mmol)的四氢呋喃(10.0mL)溶液中加入HATU(714mg,1.88mmol)和DIEA(607mg,4.70mmol)。反应混合液在室温下反应1小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=0:1),得到目标化合物(500 mg,收率77.2%,黄色固体)。LC-MS(ESI)m/z 414.0[M+H] +
步骤6:3-(4-(2-甲基吗啉代)-7-(1H-吡唑基-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000174
室温下,向反应瓶中加入N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(2-甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(200mg,0.484mmol)和三氯氧磷(6.00mL)。反应混合液在100℃下反应1小时,停止反应。将反应液滴加到饱和碳酸氢钠水溶液中并调节pH=7,然后混合液用EA(30.0mL×3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥、过滤,减压浓缩,减压浓缩,所得残留物经反相制备色谱(乙腈/水含0.05%甲酸)纯化,得到目标化合物(82.0mg,产率42.9%,白色固体)。LC-MS(ESI)m/z 396.1[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.86–7.63(m,1H),7.62–7.41(m,1H),7.13(s,1H),5.84(s,1H),4.54–4.47(m,2H),4.04–3.98(m,1H),3.87–3.78(m,6H),3.22–3.16(m,2H),3.13–3.04(m,1H),2.79–2.70(m,1H),2.01–1.93(m,4H),1.25(d,J=6.1Hz,3H)。
实施例19:3-(1-甲基-7-((R)-3-甲基吗啉代)-3-(1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧 杂-3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000175
中间体3-(7-氯-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成参考实施例13
步骤1:3-(3-溴-7-氯-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000176
将3-(7-氯-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(200mg, 0.718mmol)溶到DCM(5mL)中。反应混合物在氮气保护下加入N-溴代丁二酰亚胺(128mg,0.718mmol),反应液在室温下搅拌10分钟。LC-MS监测反应结束后,向反应混合物中加入水(5mL),用DCM(5mL×3)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品。经硅胶柱分离纯化(EA:PE=1:4),得到目标化合物(241mg,收率93.9%,黄色固体)。LC-MS(ESI)m/z:357.1[M+H] +1H NMR(400MHz,DMSO-d 6)δ7.20(s,1H),4.45–4.36(m,2H),4.20(s,3H),3.87(d,J=12.4Hz,2H),2.99(d,J=12.4Hz,2H),1.86–1.76(m,2H),1.75–1.69(m,2H).
步骤2:3-(7-氯-1-甲基-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000177
将3-(3-溴-7-氯-1-甲基-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(201mg,0.562mmol)溶于1,4-二氧六环(2mL)和水(0.2mL)中,加入1-(四氢-2H-吡喃-2-基)-3-(4,4,5,5-四甲基-1,3,2-二氧硼烷-2-基)-1H-吡唑(313mg,1.12mmol)、Pd(dtbpf)Cl 2(36.3mg,0.056mmol)和磷酸钾(358mg,1.69mmol)。所得混合物氮气保护下40℃搅拌2小时,LC-MS监测反应结束后,向反应混合物中加入水(5mL),用EA(10mL×2)萃取。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品,经硅胶柱分离纯化(PE:EA=1:2),得到目标化合物(102mg,收率42.3%,棕色油状物)。LC-MS(ESI)m/z:429.2[M+H] +
步骤3:3-(1-甲基-7-((R)-3-甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000178
将3-(7-氯-1-甲基-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(80mg,0.187mmol)溶解到N-甲基吡咯烷酮(1mL)中,加入(3R)-3-甲基吗啉(37.7mg,0.373mmol),RuphosPdG2(14.5mg,0.019mmol),碳酸铯(182mg,0.560mmol)以及碘化亚铜(35.5mg,0.187mmol)。所得混合物氮气保护下110℃搅拌2小时,LC-MS监测反应结束后,将反应混合物加水(10mL)中,水相用EA(20mL×2)萃取,饱和食盐水(10mL)洗涤。有机相干燥旋干后经硅胶柱分离纯化(PE:EA=1:1),得到目标化合物(39mg,收 率42.4%,棕色固体)。LC-MS(ESI)m/z:494.3[M+H] +
步骤4:3-(1-甲基-7-((R)-3-甲基吗啉代)-3-(1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000179
在3-(1-甲基-7-((R)-3-甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑并[4,3-b]吡啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(39.0mg,0.079mmol)的甲醇(2mL)溶液中,加入4mol/L的盐酸1,4-二氧六环溶液(2mL)。所得混合物氮气保护下室温搅拌2小时,LC-MS监测反应结束后,减压除去溶剂,残余物经prep-HPLC制备色谱柱分离纯化,得到目标化合物(4.2mg,收率13.0%,白色固体)。LC-MS(ESI)m/z:410.2[M+H] +1H NMR(400MHz,CD 3OD)δ7.82-7.73(m,1H),6.89-6.85(m,1H),6.50(s,1H),4.26(s,3H),4.07–3.94(m,2H),3.91–3.78(m,4H),3.74–3.64(m,1H),3.65–3.55(m,1H),3.51–3.37(m,3H),3.17–3.02(m,2H),2.09–2.00(m,2H),2.02–1.93(m,2H),1.17(d,J=6.4Hz,3H).
实施例20:3-(4-(3,3-二甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂 双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000180
步骤1:4-(3,6-二氯哒嗪-4-基)-3,3-二甲基吗啉的合成
Figure PCTCN2022107932-appb-000181
向3,4,6-三氯吡啶(7.64g,41.7mmol)的N-甲基吡咯烷酮溶液(30.0mL)中加入3,3-二甲基吗啉(4.00g,34.7mmol)和DIEA(13.5g,104mmol),反应混合液在氮气保护下100℃搅拌反应16小时,停止反应。然后将反应液用EA(70.0mL)稀释,用饱和食盐水(50.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1)得到目标化合物(2.00g,产率22.0%,黄色固体)。LC-MS(ESI)m/z 262.1[M+H] +1H NMR(400MHz,CDCl 3)δ7.13(s,1H),3.90–3.86(m,2H),3.47(s,2H),3.32–3.29(m,2H),1.32(s,6H)。
步骤2:3-(6-氯-5-(3,3-二甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000182
向4-(3,6-二氯哒嗪-4-基)-3,3-二甲基吗啉(2.50g,9.54mmol)和(1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(2.14g,14.3mmol)的N-甲基吡咯烷酮(30.0mL)溶液中加入DIEA(3.70g,28.6mmol)。反应混合液在氮气保护下145℃搅拌反应16小时,停止反应。然后将反应液用水(50.0mL)稀释,用乙酸乙酯(40.0mL×3)萃取,合并有机相用饱和食盐水(30.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经层析柱分离纯化(石油醚:乙酸乙酯=1:1)得到目标化合物(2.50g,产率77.4%,黄色固体)。LC-MS(ESI)m/z 339.1[M+H] +1H NMR(400MHz,CDCl 3)δ6.49(s,1H),4.54–4.51(m,2H),3.87–3.84(m,2H),3.79(d,J=12.1Hz,2H),3.47(s,2H),3.26–3.24(m,1H),3.23–3.19(m,3H),2.04–1.99(m,2H),1.90–1.85(m,2H),1.27(s,6H)。
步骤3:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3,3-二甲基吗啉代)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000183
向3-(6-氯-5-(3,3-二甲基吗啉代)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.40g,4.13mmol)和氰化锌(970mg,8.26mmol)的N,N-二甲基甲酰胺(30.0mL)溶液中加入2-(二苯基膦酰基)环戊二烯基铁(458mg,0.826mmol)和三二亚苄基丙酮二钯(378mg,0.413mmol)。反应混合液在氮气保护下135℃搅拌反应16小时,停止反应。然后将反应液用乙酸乙酯(50.0mL)稀释,用饱和食盐水(30.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(800mg,产率58.8%,黄色固体)。LC-MS(ESI)m/z 330.2[M+H] +
步骤4:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3,3-二甲基吗啉代)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000184
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3,3-二甲基吗啉代)哒嗪-3-甲腈(800mg,2.43mmol)的四氢呋喃(15.00mL)溶液中加入雷尼镍(428mg,7.29mmol)和氨水(1.00mL)。反应混合液在氢气环绕下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合(500mg,收率61.7%,黄色油状物)。LC-MS(ESI)m/z 334.1[M+H] +
步骤5:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3,3-二甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000185
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3,3-二甲基吗啉代)哒嗪-3-基)甲胺(500mg,1.50mmol)和1H-吡唑-5-羧酸(151mg,1.35mmol)的四氢呋喃溶液(15.0mL)中加入2-(3H-[1,2,3]三唑并[4,5-b]吡啶-3-基)-1,1,3,3-四甲基六氟磷酸脲(684mg,1.80mmol)和DIEA(581mg,4.50mmol),反应混合液在室温下搅拌反应1小时,停止反应。然后加水(30.0mL),再用乙酸乙酯(25.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=10:1)得到目标化合物(300mg,产率46.8%,黄色固体)。LC-MS(ESI)m/z 428.4[M+H] +1H NMR(400MHz,CDCl 3)δ9.16(s,1H),7.60–7.56(m,1H),6.81–6.77(m,1H),6.65(s,1H),5.29(s,1H),4.91–4.78(m,2H),4.55–4.50(m,2H),3.97–3.89(m,2H),3.84–3.76(m,2H),3.55(s,2H),3.26–3.21(m,2H),3.19–3.09(m,2H),2.04–1.99(m,2H),1.91–1.85(m,2H),1.16(s,6H)。
步骤6:3-(4-(3,3-二甲基吗啉代)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000186
室温下向N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(3,3-二甲基吗啉代)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(250mg,0.585mmol)的乙腈(5.00mL)溶液中加入三氯氧磷(6.00mL)。反应混合液在100℃下反应1小时,停止反应。将反应液减压浓缩,所得残留物经反相制备(乙腈/水含0.05%甲酸)纯化得到目标化合物(58.9mg,产率24.6%,白色固体)。LC-MS(ESI)m/z410.1[M+H] +1H NMR(400MHz,CDCl 3)δ7.75–7.69(m,1H),7.44(s,1H),7.09–7.05(m,1H),5.99(s,1H),4.60–4.54(m,2H),3.94–3.89(m,2H),3.67–3.62(m,2H),3.52(s,2H),3.50–3.46(m,2H),3.36–3.31(m,2H),2.09–2.02(m,2H),1.96–1.89(m,2H),1.35(s,6H)。
实施例23:4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1H-吡唑-5-基)吡唑[1,5-a]嘧啶 -7-基)四氢-2H-吡喃-4-醇
Figure PCTCN2022107932-appb-000187
步骤1:3-(吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000188
向5-氯吡唑并[1,5-a]嘧啶(5.00g,32.6mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(7.31g,48.8mmol)的N-甲基吡咯烷酮(60.0mL)溶液中加入DIEA(12.6g,97.7mmol)。反应混合液在氮气保护下100℃搅拌反应1小时,停止反应。然后将反应液用乙酸乙酯(85.0mL)稀释,用饱和食盐水(55mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1)得到目标化合物(3.60g,产率48.0%,黄色固体)。LC-MS(ESI)m/z 231.1[M+H] +1H NMR(400MHz,CDCl 3)δ8.33(d,J=7.8Hz,1H),7.88(d,J=2.0Hz,1H),6.26(d,J=7.8Hz,1H),6.13(d,J=1.9Hz,1H),4.57–4.47(m,2H),3.92(d,J=12.5Hz,2H),3.27(dd,J=12.6,2.4Hz,2H),2.06–1.94(m,2H),1.87–1.74(m,2H).
步骤2:4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇的合成
Figure PCTCN2022107932-appb-000189
氮气氛围下,将(1R,5S)-3-(吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.50g,6.51mmol)的四氢呋喃(20.0mL)溶液降温至-78℃。然后滴加正丁基锂(8.14mL,13.0mmol,1.6M),反应混合液在-78℃搅拌反应1小时,再缓慢滴加四氢-4H-吡喃-4-酮(1.30g,13.0mmol),反应液室温搅拌过夜,停止反应。加入氯化铵水溶液(45.0mL)稀释,再用EA(45.0mL×3)萃取,合并有机相用饱和食盐水(35.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(500mg,收率23.2%,黄色固体)。LC-MS(ESI)m/z 331.1[M+H] +1H NMR(400MHz,CDCl 3)δ7.86(d,J=2.3Hz,1H),6.21(d,J= 2.2Hz,1H),6.15(s,1H),4.56–4.50(m,2H),4.31–4.27(m,2H),4.23–4.19(m,2H),3.77–3.76(m,2H),3.33–3.26(m,2H),2.27–2.20(m,2H),2.12–2.07(m,2H),2.03–1.96(m,2H),1.88–1.82(m,2H).
步骤3:4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-碘吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇的合成
Figure PCTCN2022107932-appb-000190
向4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇(400mg,1.21mmol)的乙腈(10.0mL)溶液降中加入N-碘代丁二酰亚胺(272mg,1.21mmol),反应混合液在室温下搅拌反应1小时,停止反应。然后加水(35.0mL)稀释,再用EA(30.0mL×3)萃取,合并有机相用饱和食盐水(30.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(400mg,产率72.4%,黄色固体)。LC-MS(ESI)m/z 457.1[M+H] +
步骤4:4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇的合成
Figure PCTCN2022107932-appb-000191
氮气保护下向4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-碘吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇(400mg,0.877mmol)和1-(四氢-2H-吡喃-2-基)-5-(4,4,5,5-四甲基-1,3,2-二氧苯并呋喃-2-基)-1H-吡唑(366mg,1.31mmol)的1,4-二氧六环(10.0mL)和水(2.00mL)的溶液降中加入二氯[1,1'-双(二叔丁基膦)二茂铁钯(55.9mg,0.0877mmol)和磷酸钾(558mg,2.63mmol),反应混合液在100℃下搅拌反应16小时,停止反应。然后加水(35.0mL)稀释,再用EA(35.0mL×3)萃取,合并有机相用饱和食盐水(35.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(150mg,收率35.6%,黄色固体)。LC-MS(ESI)m/z 481.4[M+H] +
步骤5:4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1H-吡唑-5-基)吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇的合成
Figure PCTCN2022107932-appb-000192
室温下向反应瓶中加入4-(5-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡唑[1,5-a]嘧啶-7-基)四氢-2H-吡喃-4-醇(150mg,0.312mmol)和盐酸乙酸乙酯(10.0mL,3M),反应混合液在室温下搅拌反应2小时,停止反应。减压浓缩,所得残留物经反相制备(乙腈/水含0.05%甲酸)纯化得到目标化合物(66.6mg,产率53.8%,白色固体)。LC-MS(ESI)m/z 397.2[M+H] +1H NMR(400MHz,CDCl 3)δ8.21(s,1H),7.64(s,1H),6.56(s,1H),6.20(s,1H),4.59–4.55(m,2H),4.11–4.05(m,2H),4.04–3.93(m,2H),3.93–3.87(m,2H),3.40–3.34(m,2H),2.26–2.20(m,2H),2.13–2.02(m,4H),1.88–1.80(m,2H).
实施例26:3-(7-(1-甲基-1H-吡唑-5-基)-3-(1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮 杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000193
步骤1:5-氯-7-(1-甲基-1H-吡唑-5-基)吡唑并[1,5-a]嘧啶的合成
Figure PCTCN2022107932-appb-000194
室温下,在氮气保护下向5,7-二氯吡唑[1,5-a]嘧啶(1.00g,5.32mmol)和1-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(1.33g,6.38mmol)的水(1.00mL)和二氧六环(4.00mL)混合溶液中加入碳酸钠(1.69g,16.0mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(340mg,0.532mmol)。反应混合液在氮气保护下90℃搅拌反应10小时后,停止反应。待反应液冷却后,加入水(20.0mL),再用EA(30.0mL×2)萃取,合并有机相用,饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥、减压浓缩,所得残留物经正相层析柱分离纯化(EA:PE=1:1),得到粗产品(370mg,产率29.7%,黄色固体)。LC-MS(ESI)m/z 233.9[M+H] +
步骤2:3-(7-(1-甲基-1H-吡唑-5-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000195
室温下向5-氯-7-(1-甲基-1H-吡唑-5-基)吡唑并[1,5-a]嘧啶(370mg,1.58mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(358mg,2.39mmol)的NMP(15.0mL)溶液中加入DIEA(613mg,4.75mmol)。反应混合液在氮气保护下140℃微波反应器中反应2小时,停止反应。然后将反应液用EA(35.0mL)稀释,用饱和食盐水(30.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(260mg,产率52.9%,黄色油状物)。LC-MS(ESI)m/z 311.2[M+H] +1H NMR(400MHz,CDCl 3)δ7.92(d,J=2.2Hz,1H),7.61(d,J=1.9Hz,1H),6.63(d,J=1.9Hz,1H),6.30(s,1H),6.23(d,J=2.2Hz,1H),4.55–4.50(m,2H),3.97–3.90(m,2H),3.88(s,3H),3.33–3.27(m,2H),2.04–1.98(m,2H),1.90–1.82(m,2H)。
步骤3:3-(3-碘-7-(1-甲基-1H-吡唑-5-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000196
向3-(7-(1-甲基-1H-吡唑-5-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(260mg,0.837mmol)的乙腈(10.0mL)溶液中加入NIS(188mg,0.837mmol),反应混合液在室温下搅拌反应3小时,停止反应。然后加水(15.0mL),再用EA(20.0mL×3)萃取,合并有机相用饱和食盐水(15.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(300mg,产率82.1%,黄色固体)。LC-MS(ESI)m/z 436.9[M+H] +1H NMR(400MHz,CDCl 3)δ7.90(s,1H),7.62(d,J=1.9Hz,1H),6.61(d,J=1.9Hz,1H),6.30(s,1H),4.57–4.52(m,2H),4.11–3.89(m,2H),3.85(s,3H),3.37–3.30(m,2H),2.05–1.99(m,2H),1.90–1.82(m,2H)。
步骤4:3-(7-(1-甲基-1H-吡唑-5-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000197
氮气保护下向3-(3-碘-7-(1-甲基-1H-吡唑-5-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(300mg,0.687mmol)和1-(四氢-2H-吡喃-2-基)-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(286mg,1.03mmol)的1,4-二氧六环(10.0mL)和水(2.00mL)的溶液中加入Pd(dtbpf)Cl 2(43.8mg,0.0687mmol)和磷酸钾(437mg,0.2.06mmol),反应混合液在60℃下搅拌反应16小时,停止反应。然后加水(30.0mL)稀释,再用EA(30.0mL×3)萃取,合并有机相用饱和食盐水(25.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(150mg,收率47.3%,黄色固体)。LC-MS(ESI)m/z461.1[M+H] +1H NMR(400MHz,CDCl 3)δ8.16(s,1H),7.68(d,J=1.5Hz,1H),7.64(d,J=1.9Hz,1H),6.71–6.63(m,2H),6.38(s,1H),5.51–5.39(m,1H),4.60–4.51(m,2H),4.09–3.95(m,2H),3.93(s,3H),3.72–3.61(m,1H),3.39–3.30(m,2H),2.72–2.59(m,1H),2.16–2.09(m,1H),2.02–1.96(m,2H),1.88–1.77(m,3H),1.64–1.55(m,2H)。
步骤5:3-(7-(1-甲基-1H-吡唑-5-基)-3-(1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000198
室温下向反应瓶中加入3-(7-(1-甲基-1H-吡唑-5-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(150mg,0.325mmol)和盐酸的EA溶液(5.00mL,3M),反应混合液在室温下搅拌反应2小时,停止反应。减压浓缩,所得残留物经反相制备(乙腈/水含0.05%甲酸)纯化,得到目标化合物(82.68mg,产率67.4%,淡黄色固体)。LC-MS(ESI)m/z 377.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ12.89–12.58(m,1H),8.38–8.19(m,1H),7.80–7.45(m,2H),6.89–6.68(m,3H),4.52–4.46(m,2H),4.31–4.03(m,2H),3.85(s,3H),3.23–3.15(m,2H),1.89–1.74(m,4H)。
实施例27:3-(8-(1-甲基-1H-吡唑-5-基)-3-(1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-6-基)-8-氧杂-3-氮 杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000199
步骤1:8-溴-6-氯-3-碘咪唑并[1,2-b]哒嗪的合成
Figure PCTCN2022107932-appb-000200
向8-溴-6-氯咪唑并[1,2-b]哒嗪(2.00g,8.60mmol)的乙腈(30.0mL)溶液中加入NIS(1.94g,8.60mmol),反应混合液在室温下搅拌反应2小时,停止反应。然后加水(45.0mL),再用EA(40.0mL×3)萃取,合并有机相用饱和食盐水(40.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1),得到目标化合物(3.00g,产率97.3%,黄色固体)。LC-MS(ESI)m/z 357.8[M+H] +,359.8[M+2+H] +
步骤2:8-溴-6-氯-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑并[1,2-b]哒嗪的合成
Figure PCTCN2022107932-appb-000201
氮气保护下,向8-溴-6-氯-3-碘咪唑并[1,2-b]哒嗪(2.00g,5.58mmol)和1-(四氢-2H-吡喃-2-基)-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(1.55g,5.58mmol)的1,4-二氧六环(20.0mL)和水(4.00mL)的溶液中加入四三苯基膦钯(644mg,0.558mmol)和磷酸钾(3.55g,16.7mmol),反应混合液在室温下搅拌反应16小时,停止反应。然后加水(45.0mL),再用EA(45.0mL×3)萃取,合并有机相用饱和食盐水(45.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=3:1),得到目标化合物(400mg,产率18.7%,黄色固体)。LC-MS(ESI)m/z 382.0[M+H] +,384.0[M+2+H] +1H NMR(400MHz,CDCl 3)δ8.10(s,1H),7.73(s,1H),7.46(s,1H),6.91(s,1H),5.32–5.27(m,1H),4.11–4.06(m,1H),3.68–3.62(m,1H),2.65–2.56(m,1H),2.02–1.97(m,2H),1.77–1.70(m,1H),1.63–1.57(m,2H)。
步骤3:6-氯-8-(1-甲基-1H-吡唑-5-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑[1,2-b]哒嗪的合成
Figure PCTCN2022107932-appb-000202
氮气保护下,向8-溴-6-氯-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑并[1,2-b]哒嗪(400mg,1.05mmol)和1-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(217mg,1.05mmol)的1,4-二氧六环(10.0mL)和水(2.00mL)的溶液中加入1,1-双(二苯基磷)二茂铁氯化钯(76.5mg,0.105mmol)和磷酸钾(666mg,3.14mmol),反应混合液在60℃下搅拌反应2小时,停止反应。然后加水(25.0mL),再用EA(25.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(150mg,产率37.4%,黄色固体)。LC-MS(ESI)m/z 384.0[M+H] +1H NMR(400MHz,CDCl 3)δ8.10(s,1H),7.73(s,1H),7.46(s,1H),6.91(s,1H),5.32–5.27(m,1H),4.11–4.06(m,1H),3.68–3.62(m,1H),2.65–2.56(m,1H),2.02–1.97(m,2H),1.77–1.70(m,1H),1.63–1.57(m,2H)。
步骤4:3-(8-(1-甲基-1H-吡唑-5-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000203
氮气保护下,向6-氯-8-(1-甲基-1H-吡唑-5-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑[1,2-b]哒嗪(150mg,0.391mmol)和(2-氨基-2-[1,1'-联苯]-2-基)(二环己基(2,6-二异丙氧基-2-[1,1'-联苯]-2-基)-膦酰基)钯(II)氯化物(39.1mg,0.0391mmol)的甲苯(6.00mL)的溶液中加入碳酸铯(382mg,1.17mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(66.3mg,0.443mmol),反应混合液在110℃下搅拌反应16小时,停止反应。然后加水(15.0mL),再用EA(15.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1),得到目标化合物(60.0mg,产率33.3%,黄色固体)。LC-MS(ESI)m/z 461.2[M+H] +
步骤5:3-(8-(1-甲基-1H-吡唑-5-基)-3-(1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000204
室温下,向反应瓶中加入3-(8-(1-甲基-1H-吡唑-5-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)咪唑并[1,2-b]哒嗪-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(60.0mg,0.130mmol)和盐酸的EA溶液(5.00mL,3M),反应混合液在室温下搅拌反应2小时,停止反应。减压浓缩,所得残 留物经反相制备(乙腈/水含0.05%甲酸)纯化,得到目标化合物(18.0mg,产率36.7%,白色固体)。LC-MS(ESI)m/z 377.2[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.98(s,1H),7.82–7.74(m,1H),7.63(d,J=1.9Hz,1H),7.24–7.18(m,1H),7.17(s,1H),6.71(d,J=1.9Hz,1H),4.58–4.52(m,2H),3.93(s,4H),3.90–3.88(m,1H),3.30–3.28(m,1H),3.28–3.25(m,1H),2.04–1.97(m,4H).
实施例28:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)- 8-氧杂-3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000205
步骤1:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000206
5,7-二氯吡唑[1,5-a]嘧啶(1.00g,5.32mmol)和(1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(2.39g,16.0mmol)的N-甲基吡咯烷酮(15.0mL)溶液中加入DIEA(2.06g,16.0mmol),反应混合液在145℃下搅拌反应16小时,停止反应。待反应混合物冷却至室温后,加入水(20.0mL)稀释,用EA(25.0mL×3)萃取,合并有机相并用水(20.0mL×3)和饱和食盐水(20.0mL×3)洗涤后用无水硫酸钠干燥、过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=10:1-1:1)得到目标化合物(550mg,收率30.3%,白色固体)。LC-MS(ESI)m/z 342.3[M+H] +
步骤2:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-碘吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000207
向3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)吡唑并[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(550mg,1.61mmol)的乙腈(20.0mL)溶液中加入N-碘代丁二酰亚胺(399mg,1.77 mmol)。反应混合液在室温下搅拌反应2小时后,停止反应。将反应混合物减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:1)得到目标化合物(500mg,收率66.4%,黄色固体)。LC-MS(ESI)m/z 468.3[M+H] +1H NMR(400MHz,DMSO-d 6)δ7.90(s,1H),5.70(s,1H),4.45–4.38(m,4H),4.08–3.98(m,4H),3.09–3.00(m,4H),2.07–1.98(m,2H),1.90–1.78(m,4H),1.75–1.65(m,2H).
步骤3:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000208
向3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-碘吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(300mg,0.642mmol)和1-(四氢-2H-吡喃-2-基)-5-(4,4,5,5-四甲基-1,3,2-二氧苯并呋喃-2-基)-1H-吡唑(268mg,0.963mmol)的水(1.00mL)和1,4-二氧六环(5.00mL)混合溶液中加入碳酸钾(177mg,1.28mmol)、[1,1'-双(二叔丁基膦)二茂铁]二氯化钯(42.0mg,0.0642mmol)。反应混合液在氮气保护下60℃搅拌反应8小时后,停止反应。待反应液冷却至室温后加入水(10.0mL),然后用EA(15.0mL×3)萃取,有机相用饱和食盐水(15.0mL×2)洗涤后经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1-1:3)得到目标化合物(150mg,收率47.5%,黄色固体)。LC-MS(ESI)m/z 492.4[M+H] +
步骤4:3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000209
3-(7-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(120mg,0.244mmol)的氯化氢甲醇溶液(3.0M,3.00mL)在室温下搅拌反应1小时,停止反应。将反应混合物减压浓缩,所得残留物经反相制备(乙腈/水含0.05%甲酸)分离纯化得到目标化合物(33.96mg,收率34.1%,白色固体)。LC-MS(ESI)m/z 408.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ12.64(Brs,1H),8.23(s,1H),7.55(s,1H),6.69(s,1H),5.72(s,1H),4.49–4.41(m,4H),4.16–4.04(m,4H),3.09(d,J=10.9Hz,4H),2.11–2.01(m,2H),1.92–1.80(m,4H),1.77–1.68(m,2H)。
实施例29:1-((R)-4-(2-((1R,S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-7-(1H-吡唑-3-基)咪唑[1,5- b]哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮
Figure PCTCN2022107932-appb-000210
步骤1:(R)-4-(3,6-二氯哒嗪-4-基)-3-甲基哌嗪-1-羧酸叔丁酯的合成
Figure PCTCN2022107932-appb-000211
向(R)-3-甲基哌嗪-1-羧酸叔丁酯(5.00g,24.9mmol)的N-甲基吡咯烷酮(35.0mL)溶液降中加入3,4,6-三氯哒嗪(5.49g,29.9mmol)和DIEA(9.68g,74.9mmol),反应混合液在氮气保护下室温搅拌反应16小时,停止反应。然后加水(60.0mL)稀释,再用乙酸乙酯(50.0mL×3)萃取,再用饱和食盐水(40.0mL)洗涤有机相后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(石油醚:乙酸乙酯=5:1)得到目标化合物(3.50g,产率40.4%,黄色固体)。LC-MS(ESI)m/z 346.9[M+H] +1H NMR(400MHz,CDCl 3)δ6.83(s,1H),4.21–4.16(m,1H),4.14–4.08(m,1H),3.83(d,J=13.1Hz,1H),3.42–3.29(m,2H),3.13–3.09(m,2H),1.47(s,9H),1.12(d,J=6.6Hz,3H).
步骤2:(R)-3,6-二氯-4-(2-甲基哌嗪-1-基)哒嗪的合成
Figure PCTCN2022107932-appb-000212
向反应瓶中加入叔丁基(R)-4-(3,6-二氯哒嗪-4-基)-3-甲基哌嗪-1-羧酸酯(3.50g,10.1mmol)和盐酸/乙酸乙酯(8.00mL,4M),反应混合液在室温下搅拌反应5小时,停止反应。将反应液减压浓缩,得到目标化合物(2.30g,产率80.5%,黄色固体)。LC-MS(ESI)m/z 247.0[M+H] +
步骤3:(R)-1-(4-(3,6-二氯哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000213
向(R)-3,6-二氯-4-(2-甲基哌嗪-1-基)哒嗪盐酸盐(2.30g,8.11mmol)的二氯甲烷(20.0mL)溶液中加入乙酰氯(0.955g,12.2mmol)和三乙胺(2.46g,24.3mmol),反应混合液在室温下搅拌反应16小时,停止反应。然后加水(50.0mL)稀释,再用乙酸乙酯(45.0mL×3)萃取,再用饱和食盐水(30.0mL)洗涤有机相后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经层析板分离纯化(石油醚:乙酸乙酯=5:1)得到目标化合物(2.00g,产率85.2%,黄色固体)。LC-MS(ESI)m/z 288.9[M+H] +1H NMR(400MHz,CDCl 3)δ6.85(s,1H),4.75–4.49(m,1H),4.36–4.15(m,2H),3.47–3.30(m,2H),3.23–2.94(m,2H),2.17–2.10(m,3H),1.19–1.12(m,3H)。
步骤4:1-((3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000214
向(R)-1-(4-(3,6-二氯哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮(2.00g,6.92mmol)的N-甲基甲吡咯烷酮(25.0mL)溶液降中加入(1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(1.55g,10.4mmol)和N,N-二异丙基乙胺(2.68g,20.7mmol),反应混合液在145℃下搅拌反应16小时,停止反应。然后加水(50.0mL)稀释,再用乙酸乙酯(45.0mL×3)萃取,再用饱和食盐水(40.0mL)洗涤有机相后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(1.80g,产率71.1%,黄色固体)。LC-MS(ESI)m/z 366.3[M+H] +1H NMR(400MHz,CDCl 3)δ6.15–6.08(m,1H),4.54–4.44(m,3H),3.97–3.82(m,2H),3.78–3.64(m,2H),3.58–3.47(m,2H),3.25–3.18(m,2H),2.20–2.13(m,2H),2.13–2.10(m,2H),2.02–1.97(m,2H),1.89–1.81(m,3H),1.06–1.01(m,3H)。
步骤5:4-((R)-4-乙酰-2-甲基哌嗪-1-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000215
向1-((3R)-4-(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-3-氯哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮(1.8g,4.92mmol)和氰化锌(1.16g,9.84mmol)的DMF(30.0mL)溶液中加入2-(二苯基膦酰基)环戊二烯基铁(546mg,0.984mmol)和三二亚苄基丙酮二钯(450mg,0.492mmol)。反应混合液在氮气保护下135℃搅拌反应16小时,停止反应。然后将反应液用EA(40.0mL)稀释,用饱和食盐水(35mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=10:1)得到目标化合物(1.05g,产率59.9%,黄色固体)。LC-MS(ESI)m/z 357.0[M+H] +1H NMR(400MHz,CDCl 3)δ5.95–5.84(m,1H),4.74–4.61(m,1H),4.53(s,2H),4.30–4.18(m,1H),4.07–3.83(m,2H),3.66(s,1H),3.54–3.41(m,1H),3.36–3.17(m,4H),3.03–2.89(m,1H),2.18–2.10(m,3H),2.03–1.98(m,2H),1.87–1.79(m,2H),1.19–1.12(m,3H)。
步骤6:1-((3R)-4-(3-(氨基甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000216
向4-((R)-4-乙酰-2-甲基哌嗪-1-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-甲腈(1.10g,3.09mmol)的四氢呋喃(15.0mL)溶液中加入雷尼镍(543mg,9.26mmol)和氨水(1.00mL)。反应混合液在氢气环绕下室温反应5小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合物(660mg,收率59.3%,黄色油状物)。LC-MS(ESI)m/z 361.0[M+H] +
步骤7:N-((4-((R)-4-乙酰基-2-甲基哌嗪-1-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000217
向1-((3R)-4-(3-(氨基甲基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮(360mg,0.999mmol)和1H-吡唑-5-羧酸(101mg,0.899mmol)的四氢呋喃溶液(10.0mL)中加入2-(3H-[1,2,3]三唑并[4,5-b]吡啶-3-基)-1,1,3,3-四甲基六氟磷酸脲(0.456mg,1.20mmol)和DIEA(387mg,3.00mmol),反应混合液在室温下搅拌反应1小时,停止反应。然后加水(30.0mL),再用EA(25.0mL×3)萃取,合并有机相用饱和食盐水(20.0mL)洗涤后经无水硫酸钠干燥,过滤,减压浓缩,所得残留物经柱层析分离纯化(DCM:甲醇=10:1)得到目标化合物(170mg,产率37.4%,黄色固体)。LC-MS(ESI)m/z 455.0[M+H] +
步骤8:1-((R)-4-(2-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-7-(1H-吡唑-3-基)咪唑[1,5-b]哒嗪-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000218
室温下,向反应瓶中加入N-((4-((R)-4-乙酰基-2-甲基哌嗪-1-基)-6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(170mg,0.374mmol)和三氯氧磷(6.00mL)。反应混合液在100℃下反应1小时,停止反应。将反应液减压浓缩,所得残留物经反相制备(乙腈/水含0.05%甲酸)纯化得到目标化合物(6.68mg,产率4.09%,白色固体)。LC-MS(ESI)m/z 437.3[M+H] +1H NMR(400MHz,MeOD-d 4)δ7.83–7.51(m,2H),7.13(s,1H),5.80(s,1H),4.55–4.48(m,3H),3.87–3.80(m,2H),3.74–3.42(m,4H),3.30–3.05(m,4H),2.24-2.12(m,3H),2.04–1.93(m,4H),1.26-1.13(m,3H)。
实施例30:3-(4-(4,4-二氟-2-甲基哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂- 3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000219
步骤1:1-(3,6-二氯哒嗪-4-基)-2-甲基哌啶-4-酮的合成
Figure PCTCN2022107932-appb-000220
向2-甲基哌啶-4-酮盐酸盐(3.00g,20.1mmol)和3,4,6-三氯哒嗪(4.41g,24.1mmol)的N-甲基吡咯烷酮(50.0mL)溶液中加入DIEA(6.09g,60.2mmol)。反应混合液在氮气保护下60℃搅拌反应16小时,停止反应。然后将反应液用乙酸乙酯(100mL)稀释,用饱和食盐水(60.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=5:1)得到目标化合物(3.00g,产率57.5%,黄色固体)。LC-MS(ESI)m/z 260.0[M+H] +
步骤2:3,6-二氯-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪的合成
Figure PCTCN2022107932-appb-000221
0℃且氮气保护下向1-(3,6-二氯哒嗪-4-基)-2-甲基哌啶-4-酮(2.8g,10.7mmol)的二氯甲烷(50.0mL)溶液中加入二乙胺基三氟化硫(2.60g,16.2mmol)。反应混合液在氮气保护下0℃下搅拌反应1小时,停止反应。然后将反应液减压浓缩,所得残留物即为目标化合物(1.80g,产率59.3%,黄色油状物)。LC-MS(ESI)m/z 281.9[M+H] +
步骤3:3-(6-氯-5-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000222
向3,6-二氯-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪(1.80g,6.38mmol)和8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(1.43g,9.57mmol)的N-甲基吡咯烷酮(50.0mL)溶液中加入DIEA(2.47g,19.1mmol)。反应混合液在氮气保护下145℃搅拌反应16小时,停止反应。然后将反应液用乙酸乙酯(85.0mL)稀释,用饱和食盐水(55.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(1.80g,产率78.6%,黄色固体)。LC-MS(ESI)m/z 359.0[M+H] +1H NMR(400MHz,CDCl 3)δ6.23(s,1H),4.54–4.50(m,2H),4.09–3.96(m,1H),3.86–3.74(m,2H),3.47–3.37(m,1H),3.26–3.19(m,2H),3.13–2.99(m,1H),2.32–2.23(m,1H),2.23–2.08(m,2H),2.04–1.97(m,3H),1.90–1.84(m,2H),1.14(d,J=16.6,6.7Hz,3H)。
步骤4:6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-甲腈的合成
Figure PCTCN2022107932-appb-000223
向3-(6-氯-5-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1.8g,5.02mmol)和氰化锌(1.18g,10.0mmol)的N,N-二甲基甲酰胺(30.0mL)溶液中加入2-(二苯基膦酰基)环戊二烯基铁(556mg,1.00mmol)和三二亚苄基丙酮二钯(459mg,0.501mmol)。反应混合液在氮气保护下140℃搅拌反应16小时,停止反应。然后将反应液用乙酸乙酯(65.0mL)稀释,用饱和食盐水(40.0mL×3)洗涤后用无水硫酸钠干燥、过滤,减压浓缩,所得残留物经柱层析分离纯化(PE:EA=1:1)得到目标化合物(1.50g,产率85.6%,黄色固体)。LC-MS(ESI)m/z 350.1[M+H] +1H NMR(400MHz,CDCl 3)δ5.93(s,1H),4.53–4.50(m,2H),4.42–4.33(m,1H),4.01–3.90(m,2H),3.51–3.42(m,2H),3.31–3.26(m,2H),2.29–2.15(m,2H),2.11–1.99(m,4H),1.84–1.80(m,2H),1.24(dd,J=6.9,1.7Hz,3H)。
步骤5:(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-基)甲胺的合成
Figure PCTCN2022107932-appb-000224
向6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-甲腈(1.50 g,4.29mmol)的四氢呋喃(30.0mL)溶液中加入雷尼镍(1.26g,21.5mmol)。反应混合液在氢气环绕下室温反应16小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物即为目标化合(1.20g,产率79.1%,黄色油状物)。LC-MS(ESI)m/z 354.0[M+H] +
步骤6:N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺的合成
Figure PCTCN2022107932-appb-000225
向(6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-基)甲胺(1.20g,3.40mmol)和1H-吡唑-5-羧酸(342mg,3.06mmol)的四氢呋喃(30.0mL)溶液中加入2-(3H-[1,2,3]三唑并[4,5-b]吡啶-3-基)-1,1,3,3-四甲基六氟磷酸脲(1.55g,4.07mmol,)和DIEA(1.32g,10.2mmol)。反应混合液在室温下反应1小时,停止反应。然后将反应液过滤,减压浓缩,所得残留物经柱层析分离纯化(EA)得到目标化合物(1.00g,产率65.8%,黄色固体)。LC-MS(ESI)m/z 448.0[M+H] +1H NMR(400MHz,CDCl 3)δ8.76(s,1H),7.59(d,J=2.1Hz,1H),6.80(d,J=2.2Hz,1H),6.44(s,1H),4.87–4.72(m,2H),4.55–4.50(m,2H),3.86–3.77(m,2H),3.49(s,2H),3.38–3.31(m,1H),3.26–3.21(m,2H),2.35–2.22(m,2H),2.03–1.99(m,2H),1.90–1.84(m,2H),1.50–1.47(m,2H),1.02(d,J=6.2Hz,3H)。
步骤7:3-(4-(-4,4-二氟-2-甲基哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000226
室温下向反应瓶中加入N-((6-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-4-(4,4-二氟-2-甲基哌啶-1-基)哒嗪-3-基)甲基)-1H-吡唑-5-甲酰胺(200mg,0.447mmol)、三氯氧磷(685mg,4.47mmol)和乙腈(5mL)。反应混合液在100℃下反应1小时,停止反应。将反应液减压浓缩,所得残留物经反相制备(乙腈/水含0.05%甲酸)纯化得到目标化合物(110mg,产率57.3%,白色固体)。LC-MS(ESI)m/z 430.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ7.81–7.59(m,2H),7.10(s,1H),5.92(s,1H),4.71–4.59(m,1H),4.47(s,2H),3.88–3.79(m,3H),3.47–3.40(m,1H),3.07(d,J=11.1Hz,2H),2.44–2.28(m,1H),2.22–2.04(m,3H),1.89-1.81(m,4H),1.21(d,J= 5.6Hz,3H)。
实施例31和32:3-(4-((R)-4,4-二氟-2-甲基哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷,和3-(4-((S)-4,4-二氟-2-甲基哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷
Figure PCTCN2022107932-appb-000227
取50mg 3-(4-(-4,4-二氟-2-甲基哌啶-1-基)-7-(1H-吡唑-3-基)咪唑并[1,5-b]哒嗪-2-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷消旋体进行(Waters SFC 150(室温,100bar,214nm)和250*25mm10mm DAICELCHIRALPAKIG(超临界二氧化碳:MEOH(+0.1%7.0mol/l Ammonia in MEOH),45:55,3.2min,100mL/min))手性拆分,分别得到先洗脱出来(保留时间较小)的P1(14.40mg,e.e.100.00%),和后洗脱出来(保留时间较大)的P2(14.37mg,e.e.99.07%)。LC-MS(ESI)m/z 430.2[M+H] +
LC-MS(ESI)m/z 430.2[M+H]+。(P1):1H NMR(400MHz,MeOD-d4)δ7.71(s,1H),7.55(s,1H),7.13(s,1H),5.88(s,1H),4.71–4.63(m,1H),4.50(s,2H),3.87–3.76(m,3H),3.58–3.50(m,1H),3.24–3.16(m,2H),2.44–2.28(m,1H),2.24–2.08(m,3H),2.01–1.94(m,4H),1.30(d,J=6.9Hz,3H);
(P2): 1H NMR(400MHz,MeOD-d 4)δ7.70(s,1H),7.54(s,1H),7.13(s,1H),5.88(s,1H),4.71–4.63(m,1H),4.50(s,2H),3.86–3.76(m,3H),3.58–3.46(m,1H),3.22–3.17(m,2H),2.44–2.27(m,1H),2.25–2.08(m,3H),2.02–1.94(m,4H),1.30(d,J=7.3Hz,3H)。
实施例34:1-((R)-4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1H-吡唑-3-基)-1H- 吡唑[3,4-b]吡啶-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮
Figure PCTCN2022107932-appb-000228
步骤1:1-((R)-4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑基[3,4-b]吡啶-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000229
将(1R,5S)-3-(4-碘代-1-(1-(四氢-2H-吡喃-2-基-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(1g,1.97mmol)、(R)-1-(3-甲基哌嗪-1-基)乙烷-1-酮(561mg,3.95mmol)、RuphosG2(155mg,0.2mmol)、碳酸铯(1.93g,5.91mmol)依次加入到NMP(10mL)中,反应液在微波150度反应1小时,反应毕,待反应液冷却后,加入水(20mL),用EA(20mL×3)萃取,合并有机相用水和饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品,粗产品经反相柱分离纯化(MeOH:H 2O=5%:95%-95%:5%)得到目标化合物(60mg,收率6.0%)。LC-MS(ESI)m/z 521.2[M+H] +
步骤2:1-((R)-4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮的合成
Figure PCTCN2022107932-appb-000230
1-((R)-4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑基[3,4-b]吡啶-4-基)-3-甲基哌嗪-1-基)乙烷-1-酮(60mg,0.12mmol)加入到6M盐酸水溶液/四氢呋喃(1mL/2mL)中,室温搅拌过夜。反应结束后,用饱和碳酸氢钠水溶液调pH>8,用EA(20mL×3)萃取,合并有机层,减压浓缩得到粗产品,粗产品用EA(2mL)洗涤,所得固体为目标产物(7mg,收率14.0%,绿色固体)。LC-MS(ESI)m/z 437.3[M+H] +1H NMR(400MHz,CDCl 3)δ7.97(s,1H),7.66(s,1H),6.82(s,1H),5.63(s,1H),4.62–4.49(m,2H),4.46–4.33(m,1H),4.01–3.81(m,2H),3.74(s,2H),3.66–3.45(m,2H),3.46–3.34(m,1H),3.34–3.18(m,2H),3.18–3.03(m,1H),2.24–2.10(m,3H),2.05–1.95(m,2H),1.92–1.83(m,2H),1.25–1.17(m,3H).
实施例35:(4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1H-吡唑-3-基)-1H-吡唑[3,4- b]吡啶-4-基)吗啉-3-基)甲醇
Figure PCTCN2022107932-appb-000231
步骤1:(4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-4-基)吗啉-3-基)甲醇的合成
Figure PCTCN2022107932-appb-000232
将(1R,5S)-3-(4-碘代-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-6-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(300mg,0.6mmol)、吗啉-3-基甲醇(144mg,1.2mmol)、RuphosPdG2(48mg,0.06mmol)、碳酸铯(582mg,1.8mmol)依次加入到NMP(12mL),微波150℃搅拌反应1小时。反应结束后,用EA(20mL×3)萃取,合并有机层,水(20mL×5)洗涤,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品,粗产品经硅胶柱分离纯化(DCM:MeOH=30:1)得到目标化合物(29mg,收率9.7%,白色固体)。LC-MS(ESI)m/z:496.3[M+H] +
步骤2:(4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-4-基)吗啉-3-基)甲醇的合成
Figure PCTCN2022107932-appb-000233
将(4-(6-((1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-1-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-3-基)-1H-吡唑[3,4-b]吡啶-4-基)吗啉-3-基)甲醇(29mg,0.06mmol)加入到6M盐酸水溶液/四氢呋喃(2mL/4mL),50℃搅拌过夜。反应结束后,用饱和碳酸氢钠水溶液调pH>8,用EA(20mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品。硅胶柱分离纯化(DCM:MeOH=15:1)得到目标产物(10mg,收率40.5%,白色固体)。LC-MS(ESI)m/z:412.2。 1H NMR(400MHz,CDCl 3)δ8.02(s,1H),7.61(d,J=1.5Hz,1H),6.80(s,1H),5.69(s,1H),4.57–4.47(m,2H),4.21(d,J=11.7Hz,1H),4.17–4.09(m,1H),4.10–3.98(m,2H),3.91–3.79(m,4H),3.80–3.69(m,1H),3.53(d,J=5.8Hz,2H),3.28–3.18(m,2H),2.06–1.91(m,3H),1.91–1.79(m,2H).
实施例36:3-(7-(3,3-二甲基吗啉代)-3-(1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环 [3.2.1]辛烷
Figure PCTCN2022107932-appb-000234
步骤1:4-(5-氯吡唑并[1,5-a]嘧啶-7-基)-3,3-二甲基吗啉的合成
Figure PCTCN2022107932-appb-000235
将5,7-二氯吡唑[1,5-a]嘧啶(540mg,2.9mmol)、3,3-二甲基吗啉盐酸盐(1.0g,8.6mmol)、DIEA(1.1g,8.6mmol)加入到NMP(15mL)中。所得混合物氮气保护下微波100℃反应0.5小时。反应毕,向反应混合物中加入水(15mL),用EA(20mL×3)萃取。合并有机相用水(30mL×5)和饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品,粗产品经硅胶柱分离纯化(PE:EA=7:3),得到目标化合物(0.9g,收率116.3%,白色固体)。LC-MS(ESI)m/z:267.0[M+H] +
步骤2:3-(7-(3,3-二甲基吗啉代)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000236
将4-(5-氯吡唑并[1,5-a]嘧啶-7-基)-3,3-二甲基吗啉(200mg,0.8mmol)、(1R,5S)-8-氧杂-3-氮杂双环[3.2.1]辛烷盐酸盐(225mg,1.5mmol)、DIEA(290mg,2.3mmol)依次加入到NMP(10mL)中。反应混合物在微波仪内140℃下反应2小时。反应毕,向反应混合物中加入水(15mL),用EA(20mL×3)萃取,合并有机相用,水洗(30mL×5)和饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品。硅胶柱分离纯化(PE:EA=57%:43%)得到目标产物(202mg,收率78.6%,白色固体)。
步骤3:3-(7-(3,3-二甲基吗啉代)-3-碘吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000237
将3-(7-(3,3-二甲基吗啉代)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(202mg,0.6mmol)、N-碘代丁二酰亚胺(135mg,0.6mmol)加入到乙腈(5mL),室温搅拌0.5小时。反应结束后,减压浓缩得到粗产品,粗产品经硅胶柱分离纯化(PE:EA=1:1)得到目标化合物(224mg,收率79.7%,白色固体)。
步骤4:3-(7-(3,3-二甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000238
将3-(7-(3,3-二甲基吗啉代)-3-碘吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(172mg,0.4mmol)、1-(四氢-2H-吡喃-2-基)-5-(4,4,5,5-四甲基-1,3,2-二氧苯并呋喃-2-基)-1H-吡唑(153mg,0.6mmol)、磷酸钾(233mg,1.1mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(24mg,0.04mmol)依次加入到二氧六环/水(10mL/2mL)中。反应混合物在80℃油浴下反应1小时。反应结束后,向反应混合物中加入水(15mL),用EA(20mL×3)萃取,合并有机相用水(30mL×5)和饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到粗产品,粗产品经硅胶柱分离纯化(PE:EA=1:4)得到目标化合物(68mg,收率37.2%,白色固体)。LC-MS(ESI)m/z:494.0[M+H] +
步骤5:3-(7-(3,3-二甲基吗啉代)-3-(1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷的合成
Figure PCTCN2022107932-appb-000239
将3-(7-(3,3-二甲基吗啉代)-3-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡唑[1,5-a]嘧啶-5-基)-8-氧杂-3-氮杂双环[3.2.1]辛烷(68mg,0.1mmol)加入到6M盐酸水溶液/四氢呋喃(2mL/4 mL)中,50℃搅拌反应1小时。反应结束后,用饱和碳酸氢钠水溶液调pH>8,用EA(20mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品,粗产品经硅胶柱分离纯化(DCM/MeOH=30/1)得到目标化合物(38mg,收率67.9%,白色固体)。LC-MS(ESI)m/z:410.2[M+H] +1H NMR(400MHz,DMSO-d 6)δ8.21(s,1H),7.57(s,1H),6.70(s,1H),5.97(s,1H),4.54-4.40(m,2H),4.05(d,J=11.3Hz,2H),3.88-3.75(m,2H),3.59-3.50(m,2H),3.45(s,2H),3.12(d,J=10.4Hz,2H),1.90-1.69(m,4H),1.32(s,6H).
活性实施例
如无特别说明,以下活性实施例中所用的实验材料、试剂、操作和方法均可从市售渠道获得或基于现有技术容易地获知或制备。
活性实施例1:激酶ATR抑制测定
本实验采用Cisbio公司的均相时间分辨荧光技术
Figure PCTCN2022107932-appb-000240
进行检测。
实验方法:如下配制缓冲液:
名称 来源 终浓度 母液浓度 体积(uL)
HEPES(mM) Gibco,Cat#15630-080 25 1000 75
Brij35 Sigma,Cat#9002-92-0 0.01% 1% 30
BSA(mg/mL) Sigma,Cat#B2064-50G 1 100 30
DTT(mM) Sigma,Cat#D0632-10G 5 1000 15
甘油 Sigma,Cat#G5516-500ML 1% 50% 60
MnCl 2(mM) Sigma,Cat#7773-01-5 10 1000 30
H 2O       2760
共计(uL)       3000
配制供试化合物溶液。将下表所示实施例化合物用DMSO作为溶剂配制溶液,母液通常为10mM。本实验中最大起始浓度为3μM,DMSO 3倍系列稀释,共10个浓度,加入至相应的384孔板(geriner bio-one,Cat#784075)的孔中,终浓度分别为3000、1000、333、111、37、12.3、4.12、1.37、0.457、0.152nM,在另外的孔中添加相应量的DMSO,待分别用作阴性对照孔或阳性对照孔。
将酶与底物用上述制备的缓冲液分开溶解并稀释于两管中,浓度如下表:
名称 来源 加入平板后的终浓度 配制浓度 母液浓度
ATR(A管) eurofins,Cat#14-953M 30nM 60nM 410.26nM
p53(B管) eurofins,Cat#14-953M 40nM 80nM 9720nM
ATP(B管) Sigma,Cat#R0441 150nM 300nM 10000nM
使用电动多通道移液器以每孔5uL的体积将A管的ATR溶液添加至实验板的化合物孔以及阴性对照孔中,在阳性对照孔中添加同等体积的上述制备的缓冲液(仅介质,不加酶)。使用离心机以1000转每分钟的转速离心一分钟,随后将实验板置于恒温孵育箱中以25℃孵育15分 钟。之后用同样的方法将B管的底物混合溶液添加5uL至实验板的阳性对照、阴性对照以及化合物孔中,离心并以25℃孵育90分钟。
反应过程中,当ATR将底物p53磷酸化以后加入两种抗体,其中anti-phospho-p53-Eu作为能量供体能与p53上的磷酸化位点特异性结合,而anti-GST-d2作为能量受体能与p53上携带的GST标签特异性结合。若使用一定波长的激光(本实验的激发光波长为340nm)激发,能量供体能发射出615nm波长的发射光,同时当能量供体与能量受体的空间距离足够接近时(即两个抗体同时连接在p53上),能量供体与能量受体之间能发生能量转移,使得能量受体发射出665nm波长的发射光。使用读板机对两个发射光进行检测并求出665nm与615nm两种信号的比值,通过作图和计算即可求出待测样品的IC 50
检测时,将两种抗体稀释于HTRF检测缓冲液(Cisbio,Cat#62SDBRDF,Lot#17A)中,如下表:
检测试剂 来源 稀释倍数 母液浓度
anti-phospho-p53-Eu Cisbio,Cat#61P08KAZ 400 400单位
anti-GST-d2 Cisbio,Cat#61GSTDLB 200 200单位
使用电动多通道移液器以每孔10uL的体积将检测溶液添加至实验板的阳性对照、阴性对照以及化合物孔中,离心并以4℃孵育过夜,使用读板机Envision 2104读数。
使用内插法计算化合物抑制率:分别计算阳性对照和阴性对照的平均值,使用公式:
单孔抑制率=1-(单孔信号值-阳性对照信号平均值)/(阴性对照信号平均值-阳性对照信号平均值)
即可计算出化合物孔的抑制率。使用四参数罗吉斯方程曲线作出化合物抑制曲线:将化合物浓度转化成以10为底的对数,将浓度和抑制率导入XLfit软件中,公式为:
抑制率=最低响应+(化合物浓度^曲线坡度)*(最高响应-最低响应)/(化合物浓度^曲线坡度+半抑制浓度^曲线坡度)
从而得出各个化合物对酶活性的IC 50值。
表1 ATR激酶抑制活性
化合物编号. ATR IC 50(nM) 化合物编号. ATR IC 50(nM)
1 17.4 16 21.9
2 40.3 17 32.0
3 5.9 18 50.8
4 20.3 26 57.7
5 8.2 27 149.0
6 13.2 20 1.4
7 21.4 23 85.5
8 18.4 29 4.5
9 79.2 30 5.2
10 144.7 31 3.9
11 22.4 32 5.6
12 16.9 34 7.6
13 18.0 35 15.3
14 8.0 36 6.2
15 22.0    
活性实施例2:细胞增殖试验
采用CellTiter-Glo TM活细胞检测试剂盒,测试了本发明化合物对细胞增殖的抑制作用。该试剂盒采用萤光素酶作检测物,发光过程中萤光素酶需要ATP的参与,向细胞培养基中加入CellTiter-Glo TM试剂,测量发光值,光信号和体系中ATP量成正比,而ATP又和活细胞数正相关,由此确定细胞的增殖活性。
细胞培养和接种:收获处于对数生长期的细胞LOVO(人结肠癌细胞)(ATCC CCL-229),并采用血小板计数器进行细胞计数。用台盼蓝排斥法检测细胞活力,确保细胞活力在90%以上。分别添加90μL细胞悬液(RPMI1640+10%胎牛血清)至96孔透明平底黑壁板(Thermo,165305)中,调整细胞浓度到3000/孔/90ul。将96孔板中的细胞置于37℃、5%CO 2、95%湿度条件下培养过夜(Thermo,Model 3100 Series)。
先用DMSO作为溶剂配制10mM的待测化合物母液,然后用PBS稀释100倍,配制成10倍终浓度的溶液,最高浓度为100μM,在接种有细胞的96孔板中每孔加入10μL待测化合物溶液,即又稀释10倍,浓度达到最终终浓度10μM。将待测化合物终浓度从10μM开始,3倍系列稀释,共9个浓度,每个3个复孔。将已加入待测化合物和细胞的96孔板置于37℃、5%CO 2、95%湿度条件下继续培养96小时,之后进行CellTiter-Glo分析。
融化CellTiter-Glo试剂(
Figure PCTCN2022107932-appb-000241
Luminescent Cell Viability Assay,Promega,G7572)并平衡细胞板至室温30分钟。每孔加入等体积的CellTiter-Glo溶液,在定轨摇床上振动5分钟使细胞裂解。将细胞板放置于室温20分钟以稳定冷光信号,用SpectraMax多标记微孔板检测仪(MD,M3)读取冷光值。
使用GraphPad Prism 7.0软件分析数据,利用非线性S曲线回归来拟合数据得出剂量-效应曲线,并由此计算IC 50值。
细胞存活率(%)=(Lum 待测药-Lum 培养液对照)/(Lum 细胞对照-Lum 培养液对照)×100%
表2细胞增殖实验结果
化合物编号. LOVO IC 50(μM) 化合物编号. LOVO IC 50(μM)
1 0.304 16 0.310
2 0.479 19 0.620
3 0.067 20 0.039
4 0.140 26 1.268
5 0.384 29 0.488
6 0.097 31 0.158
8 0.243 32 0.156
9 0.592 34 0.424
11 2.642 35 0.522
14 0.419 36 0.148
15 0.186    
活性实施例3:人和小鼠肝微粒体代谢稳定性实验
根据本领域常规的体外代谢稳定性研究的标准方法,例如(Kerns,Edward H.and Di Li(2008).Drug-like Properties:Concepts,Structure Design and Methods:from ADME to Toxicity Optimization.San Diego:Academic Press;Di,Li等人,Optimization of a Higher Throughput Microsomal Stability Screening Assay for Profiling Drug Discovery Candidates,J.Biomol.Screen.2003,8(4),453.)中所述的方法,类似地如下进行本发明化合物的肝微粒体代谢稳定性试验。
Figure PCTCN2022107932-appb-000242
将肝微粒体(蛋白浓度为0.56mg/mL)加入1μM化合物工作液(由10mM DMSO储备液用100%乙腈稀释到100μM,有机相含量:99%ACN,1%DMSO),37℃预孵育10min后,加入辅助因子(NADPH)(由氯化镁溶液配制)启动反应。孵育适当时间(如5、10、20、30和60分钟)后取样,加入适当终止液(含有200ng/mL甲苯磺丁脲和200ng/mL拉贝洛尔的冰乙腈(即4℃的乙腈)),终止反应。
样品处理(n=1):各加合适样品,涡旋后高速离心,取上清液,采用HPLC-MS/MS对底物进行检测。把0min时间点峰面积作为100%。其他时间点的峰面积转换为百分剩余量,各时间点的百分剩余量的自然对数对孵育时间作图,求算出斜率(-k),然后按固有清除率(Clint)= (k*孵育液体积)/肝微粒体质量,计算出Clint(μL/min/mg)及化合物半衰期(T 1/2,min)。结果见表3。
表3.人和小鼠肝微粒体代谢稳定性实验结果
Figure PCTCN2022107932-appb-000243
活性实施例4:本发明化合物在小鼠中的药代动力学(PK)测定
每个化合物的PK测定方法如下:6只CD-1小鼠(上海灵畅生物科技有限公司)分为两组,每组3只。其中一组通过静脉(IV)给药,剂量为1mg/kg,溶媒为5%DMSO/95%(20%Captisol);一组通过口服(Po)灌胃给药,剂量为5mg/kg,溶媒为1%HPMC。每一组在给药后0、0.083、0.25、0.5、1、2、4、6、8、24h分别通过小腿隐静脉采血。将约40μL血液收集到含EDTA-K2的抗凝管中。在收集完成后迅速将采血管倒置至少5次,以确保混合均匀,然后放置在冰上。采集到的各时间点血液在4℃/8000rpm离心5分钟以获得血浆。另取1.5mL离心管,标记化合物名称,动物编号,时间点,将血浆转移至该管中。血浆保存在-80℃直至分析。
采用UPLC-MS/MS方法测定血浆中化合物浓度,用Phoenix WinNolin 6.4药代动力学软件对所得数据进行药代动力学参数计算。
具体实验结果如下,结果表明化合物药代吸收较好,具有药代动力学优势。
表4.实施例化合物体内PK结果
Figure PCTCN2022107932-appb-000244
活性实施例5:本发明化合物在FaSSIF中平衡溶解度测定
根据本领域技术人员熟知的、本领域常规的溶解度测定标准方法,例如(Kerns,Edward H.and Di Li(2008).Drug-like Properties:Concepts,Structure Design and Methods:from ADME to Toxicity Optimization.San Diego:Academic Press)中所述的方法,以FaSSIF(pH6.5)(模拟人餐前饥饿状态下的小肠肠液)为试验系统,如下考察本发明化合物的溶解性质。
将每种化合物的样品粉末分别在容量瓶中称重,加入450μL pH值6.5 FaSSIF溶液得到过饱和混悬液,对样品进行至少2分钟的涡流,在振动筛上以800转/分的速度将容量瓶摇24小时。然后4000rpm离心20分钟。将压缩后的滤液加入高效液相色谱系统中,用标准曲线法计算其浓度,结果见表5。
快速模拟肠液FaSSIF的组成是:0.056%(w/v)卵磷脂,0.161%(w/v)牛磺胆酸钠,0.39%(w/v)磷酸钾,0.77%(w/v)氯化钾,去离子化H 2O,pH 6.5。
表5.本发明化合物在FaSSIF中的溶解度
实施例 FaSSIF pH=6.5(μg/mL)
1 1140.8
3 1144.7
BAY-1895344 33.4
上述实验结果表明,与对照相比,本发明的实施例化合物出乎意料地具有显著更高的溶解性,从而具有更好的成药性。
上述实验中使用的参比化合物BAY-1895344的结构式是
Figure PCTCN2022107932-appb-000245
(购自上海前衍生物科技有限公司,CAS NO.1876467-74-1),或可按照WO2016020320实施例111的方法制备和表征。
本领域技术人员将了解,上文描述本质上为示例性及说明性的,且欲说明本发明及其优选实施方案。通过常规实验,技术人员将了解可作出明显修正及变化而不悖离本发明的精神。在随附申请专利范围内的所有此类修正欲包括于其中。因此,本发明意欲并非由上述描述而是由以下权利要求范围及其等效物定义。
本说明书中所引用的所有公开出版物以引用方式并入本文中。

Claims (26)

  1. 式(I)化合物或其药学上可接受的盐或异构体,
    Figure PCTCN2022107932-appb-100001
    其中
    A 1、A 2和A 5各自独立地是C或N;
    A 3和A 4各自独立地是CR 4、N或NR 5
    X是O、C(R 6) 2或NR 7
    Y是N或CR 8
    R 1、R 2和R 3各自独立地是H、-OH、氧代、卤素、CN、-C 1-6烷基或-O-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素或羟基取代;或R 1和R 2连接在一起形成C 1-3亚烷基桥;
    R 4是H、氧代、卤素或-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素或羟基取代;
    R 5是H或-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素取代;
    R 6各自独立地是H、卤素、CN、-OH、-NH 2、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-C 1-6烷基、-O-C 1-6烷基、-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3-6环烷基、-SO-C 1- 6烷基、-SO-C 3-6环烷基、C 6-10芳基或C 3-6环烷基,其中的-C 1-6烷基、C 6-10芳基或C 3-6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的-C 1-6烷基取代;
    R 7是H、-C 1-6烷基、-C(O)-C 1-6烷基、-C(O)-C 3-6环烷基、-SO 2-C 1-6烷基、-SO 2-C 3-6环烷基、-SO-C 1-6烷基或-SO-C 3-6环烷基,其中的-C 1-6烷基或C 3-6环烷基任选地被一个或多个卤素、羟基、-O-C 1-6烷基、-C 1-6烷基、或卤素或羟基取代的-C 1-6烷基取代;
    R 8是H、-OH或卤素;
    n和m各自独立地是0至4的整数。
  2. 根据权利要求1的式(I)化合物或其药学上可接受的盐或异构体,其中A 1、A 2、A 3、A 4和A 5中至少两个是N或NR 5,其余是C或CR 4;优选其中两个是N或NR 5,其余是C或CR 4
  3. 根据权利要求1或2的式(I)化合物或其药学上可接受的盐或异构体,其中包含A 1~A 5的六元并五元杂芳基部分具有选自以下的结构:
    Figure PCTCN2022107932-appb-100002
    优选
    Figure PCTCN2022107932-appb-100003
  4. 根据权利要求1-3任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 4是H,R 5是H或C 1-6烷基。
  5. 根据权利要求1-4任一项的式(I)化合物或其药学上可接受的盐或异构体,其中包含X和Y的六元环
    Figure PCTCN2022107932-appb-100004
    选自
    Figure PCTCN2022107932-appb-100005
    Figure PCTCN2022107932-appb-100006
  6. 根据权利要求1-4任一项的式(I)化合物或其药学上可接受的盐或异构体,其中X选自-O-、-NH-、-N(C 1-6烷基)-、-CH 2-、-C(卤素) 2
  7. 根据权利要求1-6任一项的式(I)化合物或其药学上可接受的盐或异构体,其中Y是N或CR 8,其中R 8是OH。
  8. 根据权利要求1-7任一项的式(I)化合物或其药学上可接受的盐或异构体,其中m和n之一为0,另一个为1,R 1或R 2各自独立地是C 1-6烷基,任选被一个或多个卤素取代,连接于Y的邻位或X的邻位;或
    m和n之一为0,另一个为2,R 1或R 2各自独立地是C 1-6烷基,任选被一个或多个卤素取代,连接于Y的邻位、X的邻位或分别连接于X和Y的邻位;或
    m和n均为1,R 1和R 2各自独立地是C 1-6烷基,任选被一个或多个卤素取代,均连接于环上Y的邻位、均连接于X的邻位,或分别连接于Y的邻位和X的邻位;
    优选m和n之一为0,另一个为1,R 1或R 2是C 1-6烷基,且连接于Y的邻位或X的邻位。
  9. 根据权利要求1-7任一项的式(I)化合物或其药学上可接受的盐或异构体,其中m=1且n=1,R 1和R 2分别连接于Y邻位且一起形成C 1-3亚烷基桥,优选C 2亚烷基桥;或R 1和R 2分别连接于X邻位且一起形成C 1-3亚烷基桥,优选C 2亚烷基桥。
  10. 根据权利要求1-9任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 3是H。
  11. 根据权利要求1的式(I)化合物或其药学上可接受的盐或异构体,其中
    包含A 1~A 5的六元并五元杂芳基部分选自:
    Figure PCTCN2022107932-appb-100007
    包含X和Y的六元环选自
    Figure PCTCN2022107932-appb-100008
    Figure PCTCN2022107932-appb-100009
    R 1和R 2各自独立地是H或-C 1-6烷基,其中的C 1-6烷基任选被一个或多个卤素取代;或R 1和R 2连接在一起形成C 1-3亚烷基桥;
    R 3是H或卤素;
    R 4是H;
    R 5是H或-C 1-6烷基;
    R 6各自独立地是H或卤素、-C 1-6烷基或-O-C 1-6烷基,其中的-C 1-6烷基任选地被一个或多个卤素取代;
    R 7是H或-C 1-6烷基,其中的-C 1-6烷基任选地被一个或多个卤素取代;
    R 8是H、-OH或卤素;
    n和m各自独立地是0至2的整数。
  12. 根据权利要求11的式(I)化合物或其药学上可接受的盐或异构体,其中包含A 1~A 5的 六元并五元杂芳基部分选自
    Figure PCTCN2022107932-appb-100010
  13. 根据权利要求11或12的式(I)化合物或其药学上可接受的盐或异构体,其中包含X和Y的六元环
    Figure PCTCN2022107932-appb-100011
    选自
    Figure PCTCN2022107932-appb-100012
  14. 根据权利要求11-13任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 1和R 2各自独立地是H或-C 1-6烷基。
  15. 根据权利要求11-14任一项的式(I)化合物或其药学上可接受的盐或异构体,其中n和m之一为0,另一个为1,R 1或R 2连接于Y的邻位或X的邻位;或n和m之一为0,另一个为2,R 1或R 2同时连接于环上Y的邻位、X的邻位、或分别连接于Y的邻位和X的邻位,优选均连接于Y的邻位;或n和m均为1,R 1和R 2各自独立地连接于环上Y的邻位,或X的邻位,优选均连接于X的邻位;或同时连接于Y邻位或X邻位的R 1和R 2连接在一起形成C 1-3亚烷基桥,优选C 2亚烷基桥。
  16. 根据权利要求11-15任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 3是H。
  17. 根据权利要求11-16任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 5是-C 1-6烷基,优选-CH 3
  18. 根据权利要求11-16任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 6各自独立地是H或卤素,优选H或F。
  19. 根据权利要求11-16任一项的式(I)化合物或其药学上可接受的盐或异构体,其中R 7是H或-C 1-6烷基,优选-CH 3
  20. 化合物或其药学上可接受的盐或异构体,所述化合物选自
    Figure PCTCN2022107932-appb-100013
    Figure PCTCN2022107932-appb-100014
    Figure PCTCN2022107932-appb-100015
  21. 药物组合物,包含权利要求1至20任一项的化合物或其药学上可接受的盐或异构体,以及任选的一种或多种药学上可接受的载体。
  22. 根据权利要求21的药物组合物,其还包含至少一种另外的药物活性成分。
  23. 根据权利要求1至20任一项的化合物或其药学上可接受的盐或异构体或根据权利要求21-22任一项的药物组合物用于治疗或预防与ATR激酶相关疾病的用途。
  24. 根据权利要求1至20任一项的化合物或其药学上可接受的盐或异构体或根据权利要求21-22任一项的药物组合物在制备用于治疗或预防与ATR激酶相关疾病的药物中的用途。
  25. 根据权利要求23或24的用途,其中与ATR激酶相关疾病选自血液恶性肿瘤,例如白血病(包括慢性淋巴细胞性白血病、急性淋巴细胞性白血病、急性髓性白血病和慢性骨髓性白血病)、多发性骨髓瘤、淋巴系统恶性肿瘤(例如淋巴瘤、霍奇金病、非霍奇金淋巴瘤)、骨髓增生异常综合症,以及实体瘤、例如癌和肉瘤及其转移灶,例如乳腺癌、肺癌(非小细胞肺癌、小细胞肺癌、鳞状细胞癌、支气管肺泡癌)、中枢神经系统肿瘤(例如胶质瘤、胚胎期发育不良性神经上皮肿瘤、多形性成胶质细胞瘤、混合胶质瘤、成髓细胞瘤、成视网膜细胞瘤、成神经细胞瘤、生殖细胞瘤及畸胎瘤)、胃肠道癌(例如胃癌、食管癌、肝癌、胆管癌、结直肠癌、小肠癌、胰腺癌)、皮肤癌、黑色素瘤、甲状腺癌、骨癌、头颈癌、唾液腺癌、前列腺癌、睾丸癌、卵巢癌、子宫颈癌、子宫癌、子宫内膜癌、外阴癌、膀胱癌、肾癌、鳞状细胞癌、肉瘤(例如骨肉瘤、软骨肉瘤、平滑肌肉瘤、软组织肉瘤、尤因式肉瘤、胃肠组织癌、胃肠基质肿瘤、卡波西氏肉瘤),以及儿科癌症(例如横纹肌肉瘤和成神经细胞瘤)。
  26. 根据权利要求25的用途,其中与ATR激酶相关疾病选自肺癌、前列腺癌、黑色素瘤、卵巢癌、乳腺癌、子宫内膜癌、肾癌、胃癌、肉瘤、头颈癌、中枢神经系统肿瘤及其转移灶、以及急性髓性白血病。
PCT/CN2022/107932 2021-07-27 2022-07-26 8-氧-3-氮杂二环[3.2.1]辛烷类化合物或其盐及其制备方法和用途 WO2023005928A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
KR1020247006386A KR20240041354A (ko) 2021-07-27 2022-07-26 8-옥소-3-아자비시클로[3.2.1]옥탄계 화합물 또는 이의 염 및 이의 제조 방법과 용도
CA3227713A CA3227713A1 (en) 2021-07-27 2022-07-26 8-oxa-3-azabicyclo[3.2.1]octane compounds or salt thereof, and preparation method and use thereof
AU2022316931A AU2022316931A1 (en) 2021-07-27 2022-07-26 8-oxo-3-azabicyclo[3.2.1]octane compound or salt thereof, and preparation method therefor and use thereof
IL310412A IL310412A (en) 2021-07-27 2022-07-26 8-oxa-3-aza-dicyclo[3.2.1]octane compounds or their salt, method of preparation, and their use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202110860428 2021-07-27
CN202110860428.3 2021-07-27
CN202210779302.8 2022-07-01
CN202210779302 2022-07-01

Publications (1)

Publication Number Publication Date
WO2023005928A1 true WO2023005928A1 (zh) 2023-02-02

Family

ID=85060847

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/107932 WO2023005928A1 (zh) 2021-07-27 2022-07-26 8-氧-3-氮杂二环[3.2.1]辛烷类化合物或其盐及其制备方法和用途

Country Status (7)

Country Link
KR (1) KR20240041354A (zh)
CN (1) CN115677730A (zh)
AU (1) AU2022316931A1 (zh)
CA (1) CA3227713A1 (zh)
IL (1) IL310412A (zh)
TW (1) TW202321256A (zh)
WO (1) WO2023005928A1 (zh)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016020320A1 (en) 2014-08-04 2016-02-11 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines
WO2017202748A1 (en) 2016-05-24 2017-11-30 Merck Patent Gmbh Tricyclic heterocylic derivatives
WO2020049017A1 (en) 2018-09-07 2020-03-12 Merck Patent Gmbh 5-morpholin-4-yl-pyrazolo[4,3-b]pyridine derivatives
WO2020087170A1 (en) 2018-10-30 2020-05-07 Repare Therapeutics Inc. Compounds, pharmaceutical compositions, and methods of preparing compounds and of their use as atr kinase inhibitors
CN111848605A (zh) 2019-04-30 2020-10-30 四川科伦博泰生物医药股份有限公司 一种取代吡啶并[3,4-b]吡嗪-2(1H)-酮化合物、其制备方法和用途
CN112142744A (zh) * 2019-06-28 2020-12-29 上海瑛派药业有限公司 取代的稠合杂芳双环化合物作为激酶抑制剂及其应用
WO2021012049A1 (en) * 2019-07-22 2021-01-28 Repare Therapeutics Inc. Substituted 2-morpholinopyridine derivatives as atr kinase inhibitors
WO2022028598A1 (en) * 2020-08-07 2022-02-10 Shanghai Antengene Corporation Limited Atr inhibitors and uses thereof

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016020320A1 (en) 2014-08-04 2016-02-11 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines
WO2017202748A1 (en) 2016-05-24 2017-11-30 Merck Patent Gmbh Tricyclic heterocylic derivatives
WO2020049017A1 (en) 2018-09-07 2020-03-12 Merck Patent Gmbh 5-morpholin-4-yl-pyrazolo[4,3-b]pyridine derivatives
WO2020087170A1 (en) 2018-10-30 2020-05-07 Repare Therapeutics Inc. Compounds, pharmaceutical compositions, and methods of preparing compounds and of their use as atr kinase inhibitors
CN111848605A (zh) 2019-04-30 2020-10-30 四川科伦博泰生物医药股份有限公司 一种取代吡啶并[3,4-b]吡嗪-2(1H)-酮化合物、其制备方法和用途
CN112142744A (zh) * 2019-06-28 2020-12-29 上海瑛派药业有限公司 取代的稠合杂芳双环化合物作为激酶抑制剂及其应用
WO2021012049A1 (en) * 2019-07-22 2021-01-28 Repare Therapeutics Inc. Substituted 2-morpholinopyridine derivatives as atr kinase inhibitors
WO2022028598A1 (en) * 2020-08-07 2022-02-10 Shanghai Antengene Corporation Limited Atr inhibitors and uses thereof

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANSEL, HOWARD C ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, LIPPINCOTT, WILLIAMS & WILKINS
BERGE ET AL., PHARM SCL, vol. 66, 1977, pages 1 - 19
DI, LI ET AL.: "Optimization of a Higher Throughput Microsomal Stability Screening Assay for Profiling Drug Discovery Candidates", J. BIOMOL. SCREEN, vol. 8, no. 4, 2003, pages 453, XP002670661, DOI: 10.1177/1087057103255988
KERNS, EDWARD HDI LI: "Drug-like Properties: Concepts, Structure Design and Methods: from ADME to Toxicity Optimization", 2008, ACADEMIC PRESS
no. 1876467-74-1

Also Published As

Publication number Publication date
CA3227713A1 (en) 2023-02-02
TW202321256A (zh) 2023-06-01
KR20240041354A (ko) 2024-03-29
IL310412A (en) 2024-03-01
AU2022316931A1 (en) 2024-03-14
CN115677730A (zh) 2023-02-03

Similar Documents

Publication Publication Date Title
TWI431001B (zh) 作為圓滑蛋白(smo)抑制劑的有機化合物
CN111635408B (zh) 三唑并-嘧啶化合物和其用途
CN103288833B (zh) 作为激酶抑制剂的咪唑并三嗪和咪唑并嘧啶
EP4074713A1 (en) 1'-(imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine derivatives and related compounds as shp2 inhibitors for the treatment of cancer
WO2018113584A1 (zh) Fgfr4抑制剂、其制备方法与药学上的应用
WO2015127872A1 (zh) 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2015101293A1 (zh) 激酶抑制剂及其用途
CN114423753A (zh) 作为cd38抑制剂的杂双环酰胺
WO2022170952A1 (zh) 一种作为sos1抑制剂的多环哒嗪酮类衍生物、其制备方法及用途
CN115427035A (zh) Enl/af9 yeats抑制剂
WO2022012409A1 (zh) 一种rock抑制剂及其制备方法和用途
WO2022063212A1 (zh) 嘧啶基衍生物、其制备方法及其用途
CN115260187A (zh) 吡啶酮化合物及其用途
WO2022199662A1 (zh) 一种多环化合物及其应用
CN112300173B (zh) 一类含氮多环类化合物、制备方法和用途
TW201927787A (zh) 吡咯並三嗪化合物及抑制tam激酶之方法
WO2023036252A1 (zh) 吡咯并嘧啶类或吡咯并吡啶类衍生物及其医药用途
CN108264511B (zh) 杂环类衍生物及其制备方法和其在医药上的用途
WO2023005928A1 (zh) 8-氧-3-氮杂二环[3.2.1]辛烷类化合物或其盐及其制备方法和用途
WO2022222911A1 (zh) 嘧啶酮化合物及其用途
JP7216105B2 (ja) Erkキナーゼ阻害活性を有する化合物及びその使用
WO2021136354A1 (zh) 联苯类衍生物抑制剂、其制备方法和应用
WO2020207419A1 (zh) 哌嗪酰胺衍生物,其制备方法及其在医药上的用途
WO2023083362A1 (zh) 一种治疗肿瘤的药物
WO2023165581A1 (zh) 一种吡啶类衍生物及其用途

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: 22848533

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 310412

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 18292437

Country of ref document: US

Ref document number: 3227713

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112024001633

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 202490178

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20247006386

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2022316931

Country of ref document: AU

Ref document number: 1020247006386

Country of ref document: KR

Ref document number: AU2022316931

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2022848533

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022848533

Country of ref document: EP

Effective date: 20240227

ENP Entry into the national phase

Ref document number: 2022316931

Country of ref document: AU

Date of ref document: 20220726

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112024001633

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20240126