WO2022095960A1 - 用于癌症治疗的KRas抑制剂 - Google Patents

用于癌症治疗的KRas抑制剂 Download PDF

Info

Publication number
WO2022095960A1
WO2022095960A1 PCT/CN2021/128977 CN2021128977W WO2022095960A1 WO 2022095960 A1 WO2022095960 A1 WO 2022095960A1 CN 2021128977 W CN2021128977 W CN 2021128977W WO 2022095960 A1 WO2022095960 A1 WO 2022095960A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
alkyl
halogen
compound
formula
Prior art date
Application number
PCT/CN2021/128977
Other languages
English (en)
French (fr)
Inventor
尚尔昌
仲伯禹
张彦涛
宋光琳
汪瑞祥
唐冰清
胡旭波
Original Assignee
泰励生物科技(上海)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
Priority claimed from CN202111280310.XA external-priority patent/CN114920738A/zh
Application filed by 泰励生物科技(上海)有限公司 filed Critical 泰励生物科技(上海)有限公司
Priority to US18/035,341 priority Critical patent/US20230406849A1/en
Priority to EP21888652.1A priority patent/EP4242207A1/en
Priority to JP2023526934A priority patent/JP2023547522A/ja
Priority to CN202180074854.2A priority patent/CN116867779A/zh
Publication of WO2022095960A1 publication Critical patent/WO2022095960A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • Ras the rat sarcoma oncogene homolog
  • Ras represents a group of closely related monomeric globular proteins belonging to the GTPases protein family.
  • Ras is activated by growth factors and various other extracellular signals, and is responsible for regulating functions such as cell growth, survival, migration, and differentiation.
  • These regulatory functions of Ras are carried out by switching between the GDP-bound state and the GTP-bound state, a "molecular switch" (Alamgeer et al., Current Opin Pharmacol. 2013, 13:394-401).
  • Ras bound to GDP is an inactive form, in a dormant or off state, when the signaling system is turned off, and is activated when it is exposed to some growth-promoting stimuli, such as it can be induced by guanine nucleotide exchange factor (GEF).
  • GEF guanine nucleotide exchange factor
  • GDP is released and combined with GTP, the result is that Ras is "turned on” and converted into the active form of Ras, which recruits and activates various downstream effectors, carries out signal transmission, and can transmit cell surface signals to the cytoplasm, thereby Controls numerous key cellular processes such as differentiation, survival and proliferation (Zhi Tan et al., Mini-Reviews in Medicinal Chemistry, 2016, 16, 345-357).
  • Ras tumor protein is a very attractive anticancer drug target widely accepted in the pharmaceutical field.
  • Ras has long been regarded as an "undruggable" target in the industry.
  • the present inventors have found through research that the compounds of formula (I) as defined herein, isomers thereof or their pharmaceutically acceptable salts or solvates are effective Ras mutant, especially KRas mutant protein inhibitors, capable of inhibiting cellular Ras mutations in Ras, especially KRas activity, can be used for the treatment or prevention of diseases or disorders mediated by or benefited from Ras mutations, especially KRas muteins, especially by inhibiting Ras mutations, especially KRas muteins. KRas mutant protein to inhibit abnormal cell proliferation, thereby treating or preventing tumors or cancer.
  • a first aspect of the present invention provides a compound of formula (I), an isomer thereof or a pharmaceutically acceptable salt or solvate thereof,
  • R b is at each occurrence independently selected from H, halogen, CN or C 1-6 alkyl optionally substituted with halogen or CN;
  • E is selected from halogen, -OR d or -N(R d ) 2 -, wherein R d is each independently H or C 1-6 alkyl optionally substituted with halogen;
  • R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R 8 , A, R b each have the meaning given above for the compounds of formula (I).
  • a second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or formula (II) of the present invention, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof.
  • a fifth aspect of the present invention provides a compound of formula (I) or formula (II), an isomer thereof or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition comprising the same of the present invention in the manufacture of a therapeutic and Use in a medicament for the prevention of diseases mediated by Ras mutations, especially KRas mutations, preferably KRas G12C, KRas G12D or KRas G13D mutations, most preferably KRas G12C mutations.
  • a sixth aspect of the present invention provides methods of treating and/or preventing diseases mediated by Ras mutations, especially KRas mutations, preferably KRas G12C, KRas G12D or KRas G13D mutations, most preferably KRas G12C mutations, comprising administering to a subject in need thereof A therapeutically effective amount of a compound of formula (I) or formula (II) of the present invention, an isomer thereof, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising the same.
  • a seventh aspect of the present invention provides a process for the preparation of a compound of formula (I) or formula (II) of the present invention, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof.
  • An eighth aspect of the present invention provides a pharmaceutical combination comprising a compound of formula (I) or formula (II) of the present invention, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof and one or more other drugs active agent.
  • Ras mutant or “Ras mutein” as used herein refers to a protein encoded and expressed by a Ras gene in which one or more codons are mutated, typically including, but not limited to, glycine at codon 12 of Ras, Ras proteins with mutated glycine at codon 13 or glutamine at codon 61, such as mutated HRas, NRas or KRas. These residues are located in the active site of Ras, and their mutation can impair the intrinsic or GAP-catalyzed GTPase activity of Ras, resulting in the persistence of GTP-bound Ras.
  • Ras mutant or “Ras mutein” are used interchangeably and generally refer to a mutated HRas, NRas or KRas, such as, but not limited to, HRas-G12C (glycine at codon G12 to half Mutation of cystine), NRas-G12C, KRas-G12C, KRas-G12D (mutation of glycine to aspartate at codon G12), KRas-G13D (mutation of glycine to aspartate at codon G13) ; particularly refers to KRas muteins, more particularly to KRas-G12C muteins, KRas-G12D muteins, KRas-G13D muteins, most particularly to KRas-G12C muteins.
  • HRas-G12C glycine at codon G12 to half Mutation of cystine
  • NRas-G12C glycine at
  • treating refers to the administration of one or more compounds of formula (I) as described herein, to a subject, eg, a mammal, eg, a human, having the disease, or a symptom of the disease. Isomers thereof or their pharmaceutically acceptable salts or solvates for curing, alleviating, alleviating or affecting the disease or symptoms of the disease.
  • the disease is a Ras mutation mediated disease, in particular a tumor or cancer, as defined below.
  • prevention as used herein is well known in the art and is administered to a subject, eg a mammal, suspected of suffering from or susceptible to a Ras mutation-mediated disease, especially cancer or tumor, as defined herein,
  • a subject eg a mammal
  • administration to a human of one or more compounds of formula (I) described herein, isomers thereof, or pharmaceutically acceptable salts or solvates thereof results in a reduced risk of developing a defined disease.
  • prevention encompasses the use of the compounds of the present invention prior to the diagnosis or determination of any clinical and/or pathological symptoms.
  • inhibitor and “reduce”, or any variants of these terms, as used herein, refer to the ability of a biologically active agent, by directly or indirectly interacting with the target, to reduce the signaling activity of a target of interest, and refer to Any measurable reduction or complete inhibition of the activity of the target of interest.
  • the activity eg, KRas activity
  • the activity can be reduced by an amount of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, as compared to normal %, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more, or any range derivable therefrom.
  • selective inhibition refers to the ability of a biologically active agent to preferentially reduce the signaling activity of a target of interest over off-target signaling activity by interacting directly or indirectly with the target.
  • the compound of formula (I) of the present invention has the ability to selectively inhibit G12 or G13 mutation of KRas, HRas or NRas protein relative to various mutations in one or more codons of Ras protein, such as G12C mutation , G12D mutation and G13D mutation, preferably the ability to selectively inhibit the G12C mutation of the KRas protein.
  • the invention has at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50% for a specific Ras mutation compared to another specific Ras mutation , 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more, or any range of better activity from which it can be derived, or with At least 1-, 2-, 3-, 4-, 5-, 10-, 25-, 50-, 100- for a specific Ras mutation (eg KRas-G12C) compared to activity against another specific Ras mutation , 250- or 500-fold better activity.
  • a specific Ras mutation eg KRas-G12C
  • Ras mutation-mediated disease refers to a disease in which Ras mutation contributes to the occurrence and progression of the disease, or in which inhibition of Ras mutation will reduce the incidence, reduce or eliminate disease symptoms.
  • Ras mutation mediated disease preferably refers to a KRas mutation mediated disease, most preferably a KRas-G12C mediated disease, still more preferably cancer or tumor.
  • the term “cancer” or “tumor” refers to abnormal cell growth and proliferation, whether malignant or benign, and all precancerous cells and cancer cells and tissues.
  • the cancer or tumor includes, but is not limited to, lung adenocarcinoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer cancer, anal region cancer, stomach cancer, colon cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, esophageal cancer, small bowel cancer, endocrine system cancer, thyroid cancer, Parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, prostate cancer, chronic or acute leukemia, lymphocytic lymphoma, bladder cancer, kidney or ureter cancer, renal cell cancer, renal pelvis cancer, central
  • the cancer or tumor is associated with a Ras mutation, especially a KRas mutation, preferably a KRas G12C, KRas G12D or KRas G13D mutation, most preferably a KRas G12C mutation, including but not limited to the tumor types described above and their Preferred range.
  • a KRas mutation preferably a KRas G12C, KRas G12D or KRas G13D mutation, most preferably a KRas G12C mutation, including but not limited to the tumor types described above and their Preferred range.
  • Particularly preferred tumors of the present invention include lung cancer, lung adenocarcinoma, colon cancer, rectal cancer, pancreatic cancer, endometrial cancer, bile duct cancer, leukemia and ovarian cancer.
  • the terms "subject”, “individual” or “patient” as used herein refer to vertebrates.
  • the vertebrate is a mammal.
  • Mammals include, but are not limited to, farm animals (eg, cattle), sport animals, pets (eg, guinea pigs, cats, dogs, rabbits, and horses), primates, mice, and rats.
  • the mammal is a human.
  • the term "therapeutically effective amount” refers to an amount or dosage that is generally sufficient to produce a beneficial therapeutic effect in a patient with a cancer or tumor in need of treatment. Those skilled in the art can determine the effective amount or dosage of the active ingredient in the present invention by conventional methods and in combination with conventional influencing factors.
  • pharmaceutical combination means that a compound of the present invention may be used in combination with other active agents for the purposes of the present invention.
  • the other active agent may be one or more additional compounds of the present invention, or may be a second or additional (eg, third) compound that is compatible with, that is, does not adversely affect each other, or has complementary activities.
  • compounds such as these active agents are known to modulate other biologically active pathways, or to modulate different components of the biologically active pathways involved in the compounds of the invention, or even to overlap with the biological targets of the compounds of the invention.
  • Such active agents are suitably combined in amounts effective to achieve the intended purpose.
  • the other active agents 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, either simultaneously or sequentially when administered separately.
  • the sequential administrations may be close or distant in time.
  • other active agents that can be used in combination with the compounds of the present invention include, but are not limited to, chemotherapeutic agents, therapeutic antibodies, and radiation therapy, such as alkylating agents, antimetabolites, cell cycle inhibitors, mitotic inhibitors, topoisomerase inhibitors agents, antihormonal drugs, angiogenesis inhibitors or cytotoxic agents.
  • pharmaceutically acceptable means molecular entities and compositions that do not produce adverse, allergic or other untoward reactions when administered in appropriate amounts to animals such as humans.
  • the term “pharmaceutically acceptable salts” refers to those salts, including acid addition salts and base addition salts, that retain the biological effectiveness and properties of the parent compound and are not biologically or otherwise undesirable.
  • “Pharmaceutically acceptable acid addition salts” can be formed from compounds having free bases with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, etc.
  • the organic acid can be selected from aliphatic , cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic organic acids, such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, Maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, pamoic acid, Phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, salicylic acid, etc.
  • organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid,
  • “Pharmaceutically acceptable base addition salts” include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like, as well as those derived from pharmaceutically acceptable bases.
  • organic non-toxic bases including but not limited to primary, secondary and tertiary amines, substituted ammonium including naturally occurring substituted amines, cyclic amines and basic ion exchange resins such as ammonia, isopropylamine, trimethylamine, Diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, tromethamine, dicyclohexylamine, lysine, arginine, histamine Acid, Caffeine, Procaine, Hebamine, Choline, Betaine, Ethylenediamine, Glucosamine, Methylglucamine, Triethanolamine, Theobromine, Purine, Piperazine, Piperidine, N- Ethyl piperidine, polyamine resin, etc.
  • basic ion exchange resins such as ammonia, isopropylamine, trimethylamine, Diethylamine, trieth
  • isomer refers to any stereoisomer, enantiomeric mixture, including racemates, diastereomeric mixtures, geometric isomers, enantiomeric mixtures, which may exist in a compound in structure Isomers and/or tautomers. Methods for the determination and separation of the stereochemistry of such isomers are well known to those skilled in the art (S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E.
  • R and S represent the absolute configuration of the chiral center, and the determination of the absolute configuration is this well known to those skilled in the art.
  • solvate refers to a solvent addition form comprising a stoichiometric or non-stoichiometric amount of solvent, including any solvated form of a compound of the present invention, including, for example, a solvate with water, such as a hydrate, or a solvate with an organic solvent such as methanol, ethanol or acetonitrile, ie as methanolate, ethanolate or acetonitrile, respectively; or in the form of any polymorph. It should be understood that such solvates of the compounds of the present invention also include solvates of pharmaceutically acceptable salts of the compounds of the present invention.
  • isotopic variant refers to a compound that contains an unnatural proportion of isotopes at one or more of the atoms that make up the compound.
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms constituting the compounds, thereby forming isotopic variations of the compounds of the present invention or pharmaceutically acceptable salts thereof, whether radioactive or not, are intended to are encompassed within the scope of the present invention.
  • isotopes and pharmaceutically acceptable salts thereof that may be incorporated into the compounds of the present invention include, for example, 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, 31 P, 32 P, 35 S , 18 F and 36 Cl.
  • isotopic variations of the compounds of the present invention and their pharmaceutically acceptable salts can generally be prepared by conventional methods using appropriate isotopic variations of suitable reagents.
  • Certain isotopic variations of the compounds of the invention, and pharmaceutically acceptable salts thereof, such as those into which radioactive isotopes (eg, 3 H or 14 C) are incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated, ie, 3H , and carbon- 14 , or14C, isotopes are particularly preferred due to ease of preparation and detectability.
  • substitution with isotopes such as deuterium, ie, 2 H may provide certain therapeutic advantages due to greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements, and is therefore preferred in certain circumstances.
  • compounds of the invention substituted with positron emitting isotopes eg 11 C, 18 F, 15 O and 13 N
  • PET positron tomography
  • metabolite means a product produced by metabolism of a particular compound in vivo. Such products may, for example, result from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Identification and analysis of metabolite products is performed in a manner well known to those skilled in the art.
  • prodrug refers to a compound that can be converted under physiological conditions or by solvolysis to a biologically active compound described herein, eg, a compound of formula (I).
  • prodrug refers to a precursor of a pharmaceutically acceptable biologically active compound.
  • the prodrug is inactive when administered to a subject, but is converted to the active compound in vivo, eg, by hydrolysis.
  • Prodrug compounds generally offer the advantages of solubility, histocompatibility or delayed release in mammalian organisms (see, eg, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, pp. 21-24 (Elsevier , Amsterdam).
  • prodrugs can be found in Higuchi, T. et al., ACS Symposium Series, Vol. 14, and "Bioreversible Carriers in Drug Design", Edward B. Roche, USA Pharmaceutical Association & Pergamon Press, 1987, which is incorporated herein by reference in its entirety.
  • the term "prodrug” is also meant to include any covalently bonded carrier that is released in vivo when such prodrug is administered to a mammalian subject Active compounds.
  • Prodrugs of active compounds as described herein are generally prepared by modifying functional groups present in the active compound so that the modification can be cleaved to the parent active compound in routine practice or in vivo.
  • Prodrugs include such Compounds in which a hydroxy, amino or sulfhydryl group is bonded to any group that cleaves to form a free hydroxy, free amino or free sulfhydryl group when the prodrug is administered to a mammal.
  • Examples of prodrugs include, but are not limited to, hydroxy functional acetate esters , formate and benzoate derivatives, or acetamide, formamide, and benzamide derivatives of the amine functional group in the active compound.
  • prodrugs include phosphate-containing prodrugs, boronate ester prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, Prodrugs of beta-lactam, optionally substituted phenoxyacetamide-containing prodrugs, or optionally substituted phenylacetamide-containing prodrugs, and 5-fluorocytosine and 5-fluorouridine prodrugs.
  • pharmaceutically acceptable excipient or carrier refers to one or more compatible solid or liquid filler or gelling substances, suitable for human use, of sufficient purity and sufficiently low toxicity , whose examples include but are not limited to cellulose and its derivatives (such as sodium carboxymethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as magnesium stearate), calcium sulfate, vegetable oils, polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween), wetting agents (such as sodium lauryl sulfate), colorants, flavors, stabilizers, antioxidants, preservatives Wait.
  • cellulose and its derivatives such as sodium carboxymethyl cellulose, cellulose acetate, etc.
  • gelatin talc
  • solid lubricants such as magnesium stearate
  • calcium sulfate such as magnesium stearate
  • vegetable oils such as prop
  • halogen or "halo” as used herein means F, Cl, Br or I.
  • halogen-substituted group is intended to include mono- or polyhalogenated groups in which one or more of the same or different halogens replace one or more hydrogens in the group.
  • alkyl refers to a straight or branched chain saturated hydrocarbon group consisting of carbon atoms and hydrogen atoms. Specifically, the alkyl group has 1 to 10, eg, 1 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 carbon atoms.
  • Ci - C6 alkyl refers to a straight or branched chain 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.
  • Particular alkyl groups have 1 to 3 carbon atoms.
  • alkoxy means the group -O-alkyl, wherein alkyl has the meaning set forth herein. Specifically, the term includes the groups -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. Particular alkoxy groups have 1 to 3 carbon atoms.
  • alkylthio refers to an -S-alkyl group, wherein the alkyl group is as defined above for "alkyl”. Specifically, the term includes the groups -SC 1-6 alkyl, more specifically -SC 1-3 alkyl.
  • alkylthio include, but are not limited to, methylthio, ethylthio, propylthio (including n-propylthio, isopropylthio), butylthio (including n-butylthio, isobutylthio, tert-butylthio), pentylthio (including n-pentylthio, isopentylthio, neopentylthio), hexylthio (including n-hexylthio, isohexylthio) and the like.
  • Particular alkylthio groups have 1 to 3 carbon atoms.
  • halogen-substituted C1 - C6 alkyl refers to the C1 - C6 alkyl groups described above, wherein one or more (eg 1, 2, 3, 4 or 5) ) hydrogen atoms are replaced by halogens. It will be understood by those skilled in the art that when there is more than one halogen substituent, the halogens may be the same or different, and may be located on the same or different C atoms.
  • halogen substituted C1 - C6 alkyl are eg -CH2F , -CHF2 , -CF3 , -CCl3 , -C2F5 , -C2Cl5 , -CH2CF3 , -CH 2 Cl, -CH 2 CH 2 CF 3 or -CF(CF 3 ) 2 , etc.
  • cycloalkyl refers to a monocyclic, fused polycyclic, bridged polycyclic or spirocyclic non-aromatic saturated monovalent hydrocarbon ring structure having the specified number of ring atoms. Cycloalkyl groups may have 3 to 12 carbon atoms (ie C3 - C12 cycloalkyl), eg 3 to 10, 3 to 8, 3 to 7, 3 to 6, 5 to 6 carbon atoms .
  • Suitable cycloalkyl groups include, but are not limited to, monocyclic structures, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; or polycyclic (eg, 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 and the like.
  • monocyclic structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl
  • polycyclic (eg, bicyclic) structures including spiro Ring,
  • cycloalkenyl means a monocyclic, fused polycyclic, bridged polycyclic, or spirocyclic non-aromatic unsaturated hydrocarbon ring structure having the specified number of ring atoms, comprising at least one (eg, 1, 2, or 3) carbon-carbon double bonds.
  • Cycloalkenyl groups may have 3 to 12 carbon atoms (ie, C3 - C12 cycloalkenyl groups), such as 3 to 10, 3 to 8, 3 to 7, 3 to 6, 5 to 6 carbon atoms .
  • Suitable cycloalkenyl groups include, but are not limited to, monocyclic structures such as cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, cycloheptene cycloheptadienyl, cycloheptatrienyl, or cyclooctenyl.
  • heterocycloalkyl as used herein means a monocyclic, fused, monocyclic, fused ring comprising one or more (eg 1, 2, 3 or 4) heteroatoms independently selected from O, N and S and the specified number of ring atoms
  • Heterocycloalkyl may have 3 to 12 ring members (may be referred to as 3-12 membered heterocycloalkyl), for example 3 to 10 ring members, 3 to 8 ring members, 3 to 7 ring members, 4 to 7 ring members, 4 to 6 ring members, 5 to 6 ring members.
  • Heterocycloalkyl groups typically contain up to 4 (eg, 1, 2, 3, or 4) heteroatoms. Examples of suitable heterocycloalkyl groups include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl (eg, 1-pyrrolidinyl, 2-pyrrolidinyl, and 3-pyrrolidinyl).
  • -pyrrolidinyl tetrahydrofuranyl (eg 1-tetrahydrofuranyl, 2-tetrahydrofuranyl and 3-tetrahydrofuranyl), tetrahydrothienyl (eg 1-tetrahydrothienyl, 2-tetrahydrothienyl and 3-tetrahydrothienyl) thienyl), piperidinyl (such as 1-piperidinyl, 2-piperidinyl, 3-piperidinyl and 4-piperidinyl), tetrahydropyranyl (such as 4-tetrahydropyranyl), Tetrahydrothiopyranyl (eg 4-tetrahydrothiopyranyl), morpholinyl (eg morpholino), thiomorpholinyl, dioxanyl, piperazinyl or azepanyl, diazepine Cycloheptyl groups such as 1,4-diazacycloheptyl, 3,6-
  • heterocycloalkyl groups such as It should be understood that structures having an asymmetric center encompass their racemic and/or single enantiomeric forms, such as representable and / or
  • an optionally substituted group as used herein, unless otherwise indicated, means that a group may be unsubstituted or be derivatized by one or more (eg, 0, 1, 2, 3, 4, or 5 or more), or wherein derivatizable any range) for that group is substituted with the substituents listed for that group, wherein the substituents may be the same or different.
  • an optionally substituted group has 1 substituent.
  • an optionally substituted group has 2 substituents.
  • an optionally substituted group has 3 substituents.
  • an optionally substituted group has 4 substituents.
  • an optionally substituted group has 5 substituents.
  • the present invention also covers N-oxides of compounds of formula (I) or formula (II), provided that these compounds contain basic nitrogen atoms, such as those present in nitrogen-containing heterocycles. Certain compounds of the present invention may exist in polymorphic or amorphous forms and are therefore also within the scope of the present invention.
  • X is selected from a bond, -O- or -NH-;
  • A is N.
  • A is CR a , wherein R a is F or Cl, preferably Cl.
  • R1 is selected from H or halogen.
  • both R2 and R3 are H.
  • Rb is H.
  • R and R is C1-6 alkyl , preferably C1-3 alkyl, substituted by CN; in specific embodiments, there is one R b and R b is cyanomethyl; in a more specific embodiment, there is one R b and R b is cyanomethyl, and the attachment on the piperazine ring is
  • -XR4 is not H.
  • -XR4 is halogen; in specific embodiments, -XR4 is Cl or F.
  • -XR 4 is -C 0-6 alkyl-phenyl, preferably -C 0-3 alkyl-phenyl, more preferably phenyl having The meanings given above for the embodiments, preferred or more preferred embodiments of phenyl as R 4 .
  • -XR 4 is -OC 0-6 alkyl-phenyl, preferably -OC 0-3 alkyl-phenyl, more preferably -O-phenyl, wherein benzene Radical has the meanings given above for the embodiment, preferred or more preferred embodiment given for phenyl as R 4 .
  • -XR 4 is -C 0-6 alkyl-C 3-6 cycloalkyl, preferably -C 0-3 alkyl-C 3-6 cycloalkyl, More preferred are C 3-6 cycloalkyl groups having the meanings given above for the embodiments, preferred or more preferred embodiments as cycloalkyl in R 4 .
  • 1-tetrahydrofuranyl hydrothienyl, 2-tetrahydrothienyl and 3-tetrahydrothienyl), piperidinyl (such as 1-piperidinyl, 2-piperidinyl, 3-piperidinyl and 4-piperidinyl), tetra Hydropyranyl (eg 4-tetrahydropyranyl), tetrahydrothiopyranyl (eg 4-tetrahydrothiopyranyl), morpholinyl (eg morpholino), thiomorpholinyl, dioxanyl or piperazinyl.
  • piperidinyl such as 1-piperidinyl, 2-piperidinyl, 3-piperidinyl and 4-piperidinyl
  • tetra Hydropyranyl eg 4-tetrahydropyranyl
  • tetrahydrothiopyranyl tetrahydrothiopyranyl
  • morpholinyl eg morpholino
  • R 4 is selected from Most preferably R4 is selected from
  • exemplary substituents include, but are not limited to: F, Cl , Br, I, -CH3 , -CH2CH3 , -CH2CH2CH3 , -CH ( CH3 ) CH3 , -CH2CH( CH3 ) CH3 , -CH2CH2CH2CH3 , -C ( CH3 ) ( CH3 ) ( CH3 ) , -CH2F , -CHF2 , -CF3 , -CH 2 CF 3 , -CH 2 CH 2 CF 3 , -CH(CF 3 )CH 3 , -CH 2 CH(CF 3 )CH 3 , -CH 2 CH 2 CH 2 CF 3 , -C(CF 3 ) ( CH3 )( CH3 ) , -CH2OH , -CH2CH2OH , -CH2CH2CH2OH , -CH2CH ( OH ) CH3 , -CH2CH ( OH )
  • the 5-6 membered heteroaryl in R is selected from the group consisting of pyrrolyl, furanyl, thienyl, imidazolyl, furazanyl, oxazolyl, oxadiazolyl , oxtriazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrazolyl, triazolyl and tetrazolyl, pyridyl, pyrazinyl, pyridazinyl, pyrimidinyl and triazinyl.
  • R 4 is selected from Most preferably, R4 is selected from
  • -XR4 is -C0-6alkyl -5-6 membered heteroatoms containing 1, 2 or 3 heteroatoms independently selected from N, O or S Aryl, preferably -C 0-3 alkyl - 5-6 membered heteroaryl containing 1, 2 or 3 heteroatoms independently selected from N, O or S, more preferably 1, 2 or 3 independently 5-6 membered heteroaryl radicals selected from heteroatoms of N, O or S, said heteroaryl radicals having the meanings given above for the embodiments, preferred or more preferred embodiments given as heteroaryl in R4 .
  • -XR4 is -OC0-6alkyl -5-6 membered heteroatoms containing 1, 2 or 3 heteroatoms independently selected from N, O or S Aryl, preferably -OC 0-3 alkyl - 5-6 membered heteroaryl containing 1, 2 or 3 heteroatoms independently selected from N, O or S, more preferably 1, 2 or 3 independently 5-6 membered heteroaryl radicals selected from heteroatoms of N, O or S, said heteroaryl radicals having the meanings given above for the embodiments, preferred or more preferred embodiments given as heteroaryl in R4 .
  • -XR4 is -NH- C0-6alkyl -5-6 containing 1, 2 or 3 heteroatoms independently selected from N, O or S Membered heteroaryl, preferably -NH-C 0-3 alkyl - 5-6 membered heteroaryl containing 1, 2 or 3 heteroatoms independently selected from N, O or S, more preferably 1, 2 or 3 heteroatoms independently selected from N, O or S heteroatoms of 5-6 membered heteroaryl having the embodiments, preferred or more preferred embodiments given above for the heteroaryl as R the meaning of the way.
  • -( CRcRc ) 0-6 - SRc , - ( CRcRc ) 0-6- (CO) 0-1 as substituents of R4 -OR c , -(CR c R c ) 0-6 -(CO) 0-1 -N(R c ) 2 are each preferably -(CR c R c ) 0-6 -SR c , -(CR c R c ) 0-6 -OR c , -(CR c R c ) 0-6 -N(R c ) 2 ; more preferably -(CR c R c ) 0-3 -SR c , -(CR c R c ) 0-3 -OR c , -(CR c R c ) 0-3 -N(R c ) 2 .
  • E is halogen, preferably F.
  • E is -OR d and R d is C 1-6 alkyl optionally substituted with halogen, preferably C 1-6 alkyl, such as but not limited to methyl, Ethyl, propyl, isopropyl, trifluoromethyl, difluoromethyl, trifluoroethyl and the like.
  • R5 is Among them, at most two of Z, G, Y, B, and D are not C. Exemplary R5 includes, but is not limited to
  • R 5 is Among them, at most two of Z, Y, B, and D are not C.
  • Exemplary R5 includes, but is not limited to In a more preferred embodiment, R 5 is Most preferably R5 is
  • R 5 is more preferred
  • At least one of R6, R7 and R8 is halogen, preferably F.
  • R6 is F
  • R7 and R8 are H.
  • At least one of R6, R7 and R8 is halogen substituted C1-6 alkyl , preferably -CF3 .
  • R c is H or C 1-6 alkyl.
  • the present invention also provides compounds of formula (II), isomers, pharmaceutically acceptable salts or solvates thereof.
  • R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R 8 , A, E, R b each have the meaning given above for the compounds of formula (I).
  • A is selected from CR a or N, wherein R a is selected from halogen;
  • R 1 , R 2 and R 3 are each independently selected from H, halogen or C 1-6 alkyl;
  • R b is at each occurrence independently selected from H, halogen, CN or C 1-6 alkyl optionally substituted with halogen or CN;
  • X is selected from a bond, -O- or -NH-;
  • R 4 is selected from H, halogen, C 1-6 alkyl, -C 0-3 alkyl-C 6-10 aryl, -C 0-3 alkyl-C 3-8 cycloalkyl, -C 0- 3 alkyl-C 3-8 cycloalkenyl, -C 0-3 alkyl - 3-8 membered heterocycloalkyl containing 1, 2 or 3 independent heteroatoms selected from N, O or S, - C 0-3 alkyl - 3-8 membered heterocycloalkenyl containing 1, 2 or 3 independent heteroatoms selected from N, O or S, -C 0-3 alkyl - containing 1, 2 or 3 5-10 membered heteroaryl independently selected from heteroatoms of N, O or S, wherein the alkyl, aryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl and heterocycloalkenyl Aryl is optionally substituted with 1, 2 or
  • R c is independently at each occurrence selected from H or C 1-6 alkyl optionally substituted with halogen;
  • E is selected from halogen, -OR d or -N(R d ) 2 -, wherein R d is each independently H or C 1-6 alkyl optionally substituted with halogen;
  • R 6 , R 7 and R 8 are each independently selected from H, halogen and C 1-6 alkyl optionally substituted with halogen;
  • A is CR a , wherein R a is selected from F or Cl, preferably Cl.
  • R b is H.
  • Rb is other than H and there is either one Rb or two Rbs present.
  • -XR4 is H.
  • -XR4 is not H.
  • At least one of R6, R7 and R8 is halogen, preferably F ; more preferably R6 is F, and R7 and R8 are H.
  • E is halogen, preferably F.
  • E is -OR d and R d is C 1-6 alkyl optionally substituted with halogen, preferably C 1-6 alkyl, such as but not limited to methyl group, ethyl, propyl, isopropyltrifluoromethyl, difluoromethyl, trifluoroethyl and the like.
  • R4 is selected from H, halogen, C1-6 alkyl, phenyl, C3-6 cycloalkyl, containing 1, 2 or 3 independent 4-6 membered heterocycloalkyl with heteroatoms selected from N, O or S and 5-6 membered heteroaryl containing 1, 2 or 3 independent heteroatoms selected from N, O or S, wherein the The alkyl, aryl, cycloalkyl, heterocycloalkyl and heteroaryl groups are optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1-6 optionally substituted with halogen Alkyl, -( CRcRc ) 0-6 - ORc and -( CRcRc ) 0-6 -N( Rc ) 2 where attached to aryl, cycloalkyl, cycloalkenyl, hetero -(CR c R c ) 0-6 -N(R c ) 2 on cyclo
  • R4 is selected from halogen, preferably F or Cl.
  • R4 is selected from C1-6 alkyl, such as methyl, ethyl, propyl, isopropyl, etc., optionally independently selected from the following 1, 2 or 3 group substitutions: halogen, C 1-6 alkyl optionally substituted with halogen, -(CR c R c ) 0-6 -OR c and -(CR c R c ) 0-6 - N(R c ) 2 .
  • R 4 is selected from phenyl, optionally halogen, C 1-6 alkyl optionally substituted with halogen, -(CR c R c ) 0-6 -OR c and -(CR c R c ) 0-6 -N(R c ) 2 are substituted.
  • R4 is selected from C3-6cycloalkyl , optionally substituted with 1, 2 or 3 groups independently selected from halogen, optionally Halogen-substituted C 1-6 alkyl, -(CR c R c ) 0-6 -OR c and -(CR c R c ) 0-6 -N(R c ) 2 .
  • R4 is selected from 4-6 membered heterocycloalkyl containing 1, 2 or 3 independent heteroatoms selected from N, O or S, optionally Substituted with 1 , 2 or 3 groups independently selected from halogen, C 1-6 alkyl optionally substituted with halogen, -(CR c R c ) 0-6 -OR c and -(CR c R c ) 0-6 -N(R c ) 2 .
  • R4 is selected from 5-6 membered heteroaryl containing 1, 2 or 3 independent heteroatoms selected from N, O or S, optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1-6 alkyl optionally substituted with halogen, -(CR c R c ) 0-6 -OR c and -(CR c R c c ) 0-6 -N(R c ) 2 .
  • R c is independently selected at each occurrence from H or C 1-6 alkyl.
  • the present invention also provides embodiments of compounds of formula (II) wherein each of R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R 8 , A, E, R b is of formula ( I) Meaning in the given embodiment, preferred, more preferred or most preferred embodiment of the compound.
  • the compound of the present invention covers each of the above independent embodiments or specific embodiments, and also covers the above-mentioned embodiments or embodiments formed by any combination or sub-combination of specific embodiments, and also covers any of the above preferred or example embodiments Any combination of the above examples constitutes an embodiment.
  • Specific embodiments of the compounds of the present invention include the following specific compounds or isomers, pharmaceutically acceptable salts or solvates thereof,
  • the compounds defined herein above and various embodiments thereof are inhibitors of Ras mutations, especially KRas mutations, including mutations at codons G12, G13 and Q61, eg, G12C mutation, G12D mutation, G13D mutation.
  • the compounds of the present invention especially those specifically exemplified in the context of this document, show proliferation-inhibitory activity in cellular assays against Ras-mutated, especially KRas G12C-mutated cells, as shown in the Activity Examples section below.
  • the compounds of the present invention can be used for the treatment or prevention of diseases mediated by Ras mutations, preferably KRas mutations, most preferably KRas G12C mutations, such as diseases or conditions that can be treated by inhibiting Ras mutations, preferably KRas mutations, most preferably KRas G12C mutations, Or a disease or disorder in which Ras mutation, preferably KRas mutation, most preferably KRas G12C mutation activity plays a role or is implicated, in particular to treat or prevent tumors or cancer by inhibiting Ras mutation, preferably KRas mutation, most preferably KRas G12C mutation.
  • some compounds of the present invention also show inhibitory activity against KRas G12D mutation, and other compounds of the present invention show inhibitory activity against KRas G13D mutation.
  • the compounds as defined herein and their various embodiments, especially the example compounds have improved structural patterns compared to KRas muteins existing in the prior art, which inhibit the agent that retains comparable or enhanced, even significantly enhanced KRas mutant proteins and related cancer cell proliferation-inhibiting activity; has a different spectrum of biological activity for new indications; has improved metabolic stability, resulting in more Good pharmacokinetic properties; and improved physical and chemical properties, so that it has good druggability, such as easier absorption in the body.
  • the present invention provides a compound of the present invention, an isomer thereof, or a pharmaceutically acceptable salt or solvate thereof for use as a medicament.
  • the present invention provides compounds of the present invention, isomers thereof or pharmaceutically acceptable salts or solvates thereof for use in the treatment and/or prevention of diseases mediated by Ras mutations, preferably KRas mutations.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or formula (II) as defined above, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof, together with a pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition of the present invention can be used for the treatment or prevention of diseases mediated by Ras mutations, especially KRas mutations, such as diseases mediated by KRas G12C, KRas G12D or KRas G13D mutations, such as tumors or cancers.
  • 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 present invention are in accordance with good medical practice. Factors to be considered in this context include the particular disorder being treated, the particular mammal being treated, the clinical situation of the individual patient, the cause of the disorder, the location of drug delivery, the method of administration, the schedule of administration, and other factors well known to medical practitioners. Optimal dosage levels and frequency of administration of the pharmaceutical compositions of the present invention will be determined by clinical trials required in the art of pharmacy.
  • daily doses for oral administration will range from about 0.001 mg to about 100 mg per kg of patient body weight, often 0.01 mg to about 50 mg per kg body weight, such as 0.1 to 10 mg per kg body weight, preferably about 0.01 to about 35 mg per kg body weight kg body weight in single or divided doses.
  • a suitable dosage range is from about 0.07 to about 7000 mg/day, preferably from about 0.7 to about 2500 mg/day. It will be understood that it may be necessary in certain circumstances to use doses above these limits.
  • compositions of the present invention may be administered in any suitable manner, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal, inhalation and epidural and intranasal, and if topical treatment is desired, intralesional administration can also be employed.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration.
  • oral administration is employed.
  • compositions of the present invention may be administered in any convenient form of administration, such as tablets, powders, capsules, lozenges, granules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches Wait.
  • compositions may contain conventional components in pharmaceutical formulations, such as diluents (eg, glucose, lactose or mannitol), carriers, pH adjusters, buffers, sweeteners, fillers, stabilizers, surfactants, Wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, perfuming agents, flavoring agents, other known additives, and other active agents.
  • diluents eg, glucose, lactose or mannitol
  • carriers pH adjusters, buffers, sweeteners, fillers, stabilizers, surfactants, Wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, perfuming agents, flavoring agents, other known additives, and other active agents.
  • Suitable carriers and excipients are well known
  • the compounds of formula (I) or formula (II) of the present invention and the compounds of various embodiments thereof, especially the compounds specifically prepared and characterized in the Examples, show resistance to Ras mutations, especially KRas mutations, For example the inhibitory effect of KRas G12C, KRas G12D or KRas G13D mutations.
  • the present invention provides a method for inhibiting Ras mutation, especially KRas mutation, preferably KRas G12C, KRas G12D or KRas G13D mutation, most preferably KRas G12C mutation in a cell, comprising making the cell with the formula ( I) or a compound of formula (II), its isomers or their pharmaceutically acceptable salts or solvates are contacted to inhibit Ras mutation, especially KRas mutation, preferably KRas G12C, KRas G12D or KRas G13D mutation, most The activity of the KRas G12C mutation is preferred.
  • the present invention also accordingly provides a method for inhibiting abnormal cell growth in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or formula (II) of the present invention, its isoform Constituents or their pharmaceutically acceptable salts or solvates, or pharmaceutical combinations comprising the compounds of formula (I) or (II) of the present invention, their isomers or their pharmaceutically acceptable salts or solvates thing.
  • the present invention provides methods for the treatment and/or prevention of diseases mediated by Ras mutations, especially KRas mutations, preferably KRas G12C, KRas G12D or KRas G13D mutations, most preferably KRas G12C mutations, including administering a therapeutically effective amount of a compound of formula (I) or formula (II) of the present invention, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof, or a compound of formula (I) or formula ( II) Pharmaceutical compositions of the compounds, their isomers or their pharmaceutically acceptable salts or solvates.
  • the present invention provides a compound of formula (I) or formula (II) of the present invention, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof, or a compound of formula (I) or formula comprising the present invention (II)
  • a pharmaceutical composition of a compound, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof for inhibiting Ras mutation, especially KRas mutation, preferably KRas G12C, KRas G12D or KRas G13D mutation in cells , most preferably the KRas G12C mutation, or for inhibiting abnormal cell growth in mammals, or for the treatment and/or prevention of mutations mediated by Ras mutations, especially KRas mutations, preferably KRas G12C, KRas G12D or KRas G13D, most preferably KRas G12C mutations induced disease.
  • the present invention provides a compound of formula (I) or formula (II) of the present invention, an isomer thereof or a pharmaceutically acceptable salt or solvate thereof, or a compound of formula (I) or formula comprising the present invention (II)
  • Pharmaceutical compositions of the compounds, their isomers or their pharmaceutically acceptable salts or solvates are prepared for use in the treatment and/or prophylaxis of mutations caused by Ras, especially KRas mutations, preferably KRas G12C, KRas G12D or KRas Use in the medicament of a disease mediated by a G13D mutation, most preferably a KRas G12C mutation.
  • the abnormal cell growth or the disease mediated by Ras mutation especially KRas mutation, preferably KRas G12C, KRas G12D or KRas G13D, most preferably KRas G12C mutation, especially refers to of cancer or tumor.
  • Exemplary such cancers or tumors include, but are not limited to, lung cancer, lung adenocarcinoma, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, anal region cancer , gastric cancer, colon cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, esophageal cancer, small bowel cancer, endocrine system cancer, thyroid cancer, parathyroid cancer, Adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, prostate cancer, chronic or acute leukemia, lymphocytic lymphoma, bladder cancer, kidney or ureter cancer, renal cell carcinoma, renal pelvis cancer, central nervous system tumor (CNS), primary CNS lymphoma, spinal tumor, brainstem glioma, or pituitary adenoma.
  • CNS central nervous
  • the abnormal cell growth or the disease mediated by Ras mutation is preferably selected from lung adenocarcinoma, lung cancer. , colon cancer, rectal cancer, pancreatic cancer, bile duct cancer, endometrial cancer, ovarian cancer, leukemia; most preferably selected from lung adenocarcinoma, colon cancer, rectal cancer, pancreatic cancer, bile duct cancer.
  • the present invention provides the above-mentioned various methods and uses for the treatment or prevention of cancer or tumor by inhibiting the KRas-G12C mutation.
  • the present invention provides the above-mentioned methods and technical solutions for the treatment or prevention of lung adenocarcinoma, colon cancer, rectal cancer, pancreatic cancer and bile duct cancer by inhibiting KRas-G12C mutation.
  • the other active agent may be one or more additional compounds of the present invention, or may be a second or additional (eg, third) compound that is compatible with, that is, does not adversely affect each other, or has complementary activities.
  • these active agents may be compounds known to modulate other biologically active pathways, or may be compounds that modulate different components of the biologically active pathways involved in the compounds of the invention, or even biological targets related to the compounds of the invention overlapping compounds.
  • the compounds of the present invention and other active agents may be (i) prior to sending the combination product to a physician (eg, in the case of a kit comprising a compound of the present invention and an additional drug); (ii) immediately prior to administration by The physician himself (or under the direction of the physician); (iii) by the patient himself, eg, during sequential administration of the compound of the invention and the other active agent together into the combination therapy.
  • a physician eg, in the case of a kit comprising a compound of the present invention and an additional drug
  • immediately prior to administration by The physician himself (or under the direction of the physician) or by the patient himself, eg, during sequential administration of the compound of the invention and the other active agent together into the combination therapy.
  • the abnormal cell growth involved therein or is caused by Ras mutation, especially KRas mutation, preferably KRas G12C, KRas G12D or KRas G13D, most preferably KRas G12C Mutation-mediated diseases are as defined above for the methods and uses of the invention.
  • Step D tert-Butyl (4-bromo-7-fluorobenzo[d]thiazol-2-yl)carbamate
  • intermediate Int B2 The synthesis of intermediate Int B2 was carried out as described in intermediate Int B1, substituting (R)-2-methylpiperazine-1-carboxylate tert-butyl ester with piperazine-1-carboxylate tert-butyl ester. LCMS (m/z): 503.2 (M+H).
  • intermediate Int C2 The synthesis of intermediate Int C2 was carried out as described in Intermediate Int C1 using 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinolin-4-yl)piperazine-1 - tert-butyl carboxylate (Intermediate B2) in place of (R)-4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinolin-4-yl)-2-methyl Piperazine-1-carboxylate tert-butyl ester (Intermediate B1). LCMS (m/z): 459.2, 471.2 (M+H).
  • Step B 4-Amino-6-chloro-5-fluoronicotinic acid ethyl ester
  • Step C 6-Chloro-4-(2-cyanoacetamido)-5-fluoronicotinic acid ethyl ester
  • Step D 7-Chloro-8-fluoro-2,4-dihydroxy-1,6-naphthyridine-3-carbonitrile
  • 6-Chloro-4-(2-cyanoacetamido)-5-fluoronicotinic acid ethyl ester 300 mg, 1.05 mmol was dissolved in THF (10 mL) and stirred under an ice bath for 2 min, followed by tert-butanol Potassium (236 mg, 2.10 mmol) was slowly added to the reaction solution. Under the condition that the ice bath naturally returned to room temperature, the mixture was stirred for 1 h. After the reaction was detected by LCMS, the reaction was added dropwise to a stirred saturated NH 4 Cl (50 mL) solution, and the pH of the mixed solution was adjusted to 7 with 1N hydrochloric acid.
  • Step E 2,4,7-Trichloro-8-fluoro-1,6-naphthyridine-3-carbonitrile
  • intermediate Int E1 The synthesis of intermediate Int E1 was carried out as described in intermediate Int B1, substituting 2,4,7-trichloro-8-fluoro-1,6-naphthyridine-3-carbonitrile (intermediate Int D) for 7- Bromo-2,4,6-trichloro-8-fluoroquinoline-3-carbonitrile (Intermediate Int A) with piperazine-1-carboxylate tert-butyl in place of (R)-2-methylpiperazine - 1-Carboxylic acid tert-butyl ester. LCMS (m/z): 426.3 (M+H).
  • intermediate Int F was carried out as described in intermediate Int B1, substituting (S)-2(cyanomethyl)piperazine-1-carboxylic acid benzyl ester acid salt for (R)-2-methylpiperidine tert-butyl oxazine-1-carboxylate.
  • LCMS m/z: 576.3, 578.3 (M+H).
  • Step A (R)-4-(7-Bromo-6-chloro-3-cyano-8-fluoro-2-(((3R,4R)-4-methoxy-1-methylpyrrolidine- 3-yl)oxy)quinolin-4-yl)-2-methylpiperazine-1-carboxylate tert-butyl ester
  • Step B (2R)-4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-8 -Fluoro-2-(((3R,4R)-4-methoxy-1-methylpyrrolidin-3-yl)oxy)quinolin-4-yl)-2-methylpiperazine-1- tert-butyl carboxylate
  • Step C 7-(2-Amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8-fluoro-2-((((3R,4R)-4-methoxy-1 -Methylpyrrolidin-3-yl)oxy)-4-((R)-3-methylpiperazin-1-yl)quinoline-3-carbonitrile
  • Step D 4-((R)-4-Acryloyl-3-methylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6 -Chloro-8-fluoro-2-(((3R,4R)-4-methoxy-1-methylpyrrolidin-3-yl)oxy)quinoline-3-carbonitrile
  • Example 2 The synthesis of Example 2 was carried out as described in Example 1, using 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinolin-4-yl)piperazine in step A - 1-Carboxylic acid tert-butyl ester (Intermediate B2) in place of (R)-4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinolin-4-yl)-2- Methylpiperazine-1-carboxylate tert-butyl ester (Intermediate B1).
  • Step A 4-(7-Bromo-6-chloro-3-cyano-2-(3-((dimethylamino)methyl)phenyl)-8-fluoroquinolin-4-yl)piperazine- 1-Carboxylic acid tert-butyl ester
  • Step B to Step D 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-2-( 3-((dimethylamino)methyl)phenyl)-8-fluoroquinoline-3-carbonitrile
  • Example 3 The subsequent synthesis of Example 3 was carried out as described in the synthesis of Example 1, using 4-(7-bromo-6-chloro-3-cyano-2-(3-((dimethylamino)methyl)benzene in step B) (R)-4-(7-bromo-6-chloro-3-cyano-8-fluoro-2- (((3R,4R)-4-Methoxy-1-methylpyrrolidin-3-yl)oxy)quinolin-4-yl)-2-methylpiperazine-1-carboxylate tert-butyl ester.
  • Example 4 The synthesis of Example 4 was carried out as described in Example 3, using (1-(2-(dimethylamino)ethyl)-1H-pyrazol-5-yl)boronic acid in step A instead of (3-(( Dimethylamino)methyl)phenyl)boronic acid.
  • Example 5 The synthesis of Example 5 was carried out as described in Example 3, using (3-(1-(dimethylamino)ethyl)phenyl)boronic acid in step A instead of (3-((dimethylamino)methyl) phenyl) boronic acid.
  • Example 6 The synthesis of Example 6 was carried out as described in Example 3, using (R)-4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinoline-4- yl)-2-methylpiperazine-1-carboxylate tert-butyl ester (Intermediate Int B1) in place of 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinoline- 4-yl)piperazine-1-carboxylate tert-butyl ester (Intermediate Int B2).
  • Example 6 The synthesis of Example 6 was carried out as described in Example 3, using (R)-4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinoline-4- yl)-2-methylpiperazine-1-carboxylate tert-butyl ester (Intermediate Int B1) in place of 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinoline- 4-yl)piperazine-1-carboxylate tert-butyl ester (Intermediate Int B2) and substituting (1-(2-(dimethylamino)ethyl)-1H-pyrazol-5-yl)boronic acid for (3 -((dimethylamino)methyl)phenyl)boronic acid.
  • Step A (2R)-4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-8 - Fluoroquinolin-4-yl)-2-methylpiperazine-1-carboxylate tert-butyl ester
  • Step B 7-(2-Amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8-fluoro-4-((R)-3-methylpiperazin-1-yl ) quinoline-3-carbonitrile
  • Step B to Step D 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8-fluoro -2-(2-Methyl-1,2,3,4-tetrahydroisoquinolin-5-yl)quinoline-3-carbonitrile
  • Example 12 The synthesis of Example 12 was carried out as described in Example 8, using (5-fluoro-2-((4-methoxybenzyl)amino)quinolin-8-yl)boronic acid in step A instead of (2- ((tert-Butoxycarbonyl)amino)-7-fluorobenzothiazol-4-yl)boronic acid.
  • LCMS (m/z): 519.1 (M+H).
  • Example 13 The synthesis of Example 13 was carried out as described in Example 1, using 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinolin-4-yl)piperazine in Step A - 1-Carboxylic acid tert-butyl ester (Intermediate Int B2) in place of (R)-4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinolin-4-yl)-2 - Methylpiperazine-1-carboxylate tert-butyl ester (Intermediate Int B1) and replacing the NaH mixture of (3R,4R)-4-methoxy-1-methylpyrrolidin-3-ol with sodium methoxide .
  • Example 14 was carried out as described in Example 3, using (1-methyl-1H-pyrazol-5-yl)boronic acid in step A instead of (3-((dimethylamino)methyl)phenyl ) boric acid.
  • 19 F NMR (376 MHz, DMSO) ⁇ -112.07, -118.03. LCMS (m/z): 591.4 (M+H).
  • Example 15 The synthesis of Example 15 was carried out as described in Example 3, using (1,3-dimethyl-1H-pyrazol-4-yl)boronic acid in step A instead of (3-((dimethylamino)methyl) ) phenyl) boronic acid.
  • Step A tert-Butyl 4-(7-Bromo-6-chloro-3-cyano-8-fluoro-2-methylquinolin-4-yl)piperazine-1-carboxylate
  • Step B to Step D 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8-fluoro -2-Methylquinoline-3-carbonitrile
  • Example 17 was carried out as described in the synthesis of Example 1, using 4-(7-bromo-6-chloro-3-cyano-8-fluoro-2-methylquinolin-4-yl) in step B Piperazine-1-carboxylate tert-butyl ester in place of (R)-4-(7-bromo-6-chloro-3-cyano-8-fluoro-2-(((3R,4R)-4-methoxy -1-Methylpyrrolidin-3-yl)oxy)quinolin-4-yl)-2-methylpiperazine-1-carboxylic acid tert-butyl ester.
  • Example 18 was carried out as described in Example 3, using cyclopropylboronic acid in step A instead of (3-((dimethylamino)methyl)phenyl)boronic acid.
  • Step A tert-Butyl 4-(7-Bromo-2,6-dichloro-3-cyano-8-fluoro-2-hydroxyquinolin-4-yl)piperazine-1-carboxylate
  • Step B to Step D 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8-fluoro -2-Hydroxyquinoline-3-carbonitrile
  • Example 19 The subsequent synthesis of Example 19 was carried out as described in the synthesis of Example 1, using 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoro-2-hydroxyquinoline-4- yl)piperazine-1-carboxylate tert-butyl ester in place of (R)-4-(7-bromo-6-chloro-3-cyano-8-fluoro-2-(((3R,4R)-4-methyl Oxy-1-methylpyrrolidin-3-yl)oxy)quinolin-4-yl)-2-methylpiperazine-1-carboxylate tert-butyl ester.
  • Step A 4-(7-Bromo-6-chloro-3-cyano-2-((diphenylmethylene)amino)-8-fluoroquinolin-4-yl)piperazine-1-carboxylic acid tert. Butyl ester
  • Step B 4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-2-((di Benzylidene)amino)-8-fluoroquinolin-4-yl)piperazine-1-carboxylate tert-butyl ester
  • Step C 7-(2-Amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-2-((diphenylmethylene)amino)-8-fluoro-4-( Piperazin-1-yl)quinoline-3-carbonitrile
  • Step D 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-2-((diphenyl Methylene)amino)-8-fluoroquinoline-3-carbonitrile
  • Step E 4-(4-Acryloylpiperazin-1-yl)-2-amino-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8- Fluoroquinoline-3-carbonitrile
  • Step A (S)-Benzyl 4-(7-Bromo-6-chloro-3-cyano-8-fluoroquinolin-4-yl)-2-(cyanomethyl)piperazine-1-carboxylate (21-1) and (S)-4-(7-bromo-6-chloro-3-cyano-8-fluoro-2-hydroxyquinolin-4-yl)-2-(cyanomethyl) ) benzyl piperazine-1-carboxylate (22-1)
  • Step B (2S)-4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-8 - Fluoroquinolin-4-yl)-2-(cyanomethyl)piperazine-1-carboxylate benzyl ester (21-2) and (2S)-4-(7-(2-((tert-butyl Oxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-8-fluoro-2-hydroxyquinolin-4-yl)-2-(cyano Methyl)piperazine-1-carboxylate benzyl ester (22-2)
  • reaction tube was sealed, and the reaction was heated to 115 °C by microwave for 2 h. Cooled to room temperature, diluted with water (60 mL), and extracted with EA (50 mL ⁇ 3).
  • Step A (4-(6-Chloro-3-cyano-4-((S)-3-(cyanomethyl)piperazin-1-yl)-8-fluoroquinolin-7-yl)- 7-Fluorobenzo[d]thiazol-2-yl)carbamate tert-butyl ester
  • Step B 7-(2-Amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-4-((S)-3-(cyanomethyl)piperazin-1-yl )-8-Fluoroquinoline-3-carbonitrile
  • Step C 4-((S)-4-Acryloyl-3-(cyanomethyl)piperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazole-4- yl)-6-chloro-8-fluoroquinoline-3-carbonitrile
  • Step A 7-(2-Amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-4-((S)-3-(cyanomethyl)piperazin-1-yl )-8-Fluoro-2-hydroxyquinoline-3-carbonitrile
  • Step B 4-((S)-4-Acryloyl-3-(cyanomethyl)piperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazole-4- yl)-6-chloro-8-fluoro-2-hydroxyquinoline-3-carbonitrile
  • Example 32 The synthesis of Example 32 was carried out as described in Example 17, using (S)-4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquinoline-4- yl)-2-(cyanomethyl)piperazine-1-carboxylate benzyl ester (Intermediate F) in place of 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoroquine Linn-4-yl)piperazine-1-carboxylate tert-butyl ester (Intermediate B2).
  • Example 33 The synthesis of Example 33 was carried out as described in Example 20, substituting (2-((tert-butoxycarbonyl)amino)benzo[d]thiazol-4-yl)boronic acid in step B for (2-((tert-butoxycarbonyl)amino)benzo[d]thiazol-4-yl)boronic acid Butoxycarbonyl)amino)-7-fluorobenzothiazol-4-yl)boronic acid.
  • Example 34 The synthesis of Example 34 was carried out as described in Example 20, using (2-((tert-butoxycarbonyl)amino)-5-fluorobenzo[d]thiazol-4-yl)boronic acid in step B instead of (2 -((tert-butoxycarbonyl)amino)-7-fluorobenzothiazol-4-yl)boronic acid.
  • Step A tert-Butyl 4-(7-Bromo-6-chloro-3-cyano-8-fluoro-2-(methylamino)quinolin-4-yl)piperazine-1-carboxylate
  • Step B to Step D 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-8-fluoro -2-(Methylamino)quinoline-3-carbonitrile
  • Example 35 The subsequent synthetic procedure of Example 35 was carried out as described in Example 19, using 4-(7-bromo-6-chloro-3-cyano-8-fluoro-2-(methylamino)quinoline in step B) -4-yl)piperazine-1-carboxylate tert-butyl ester in place of 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoro-2-hydroxyquinolin-4-yl)piperidine tert-butyl oxazine-1-carboxylate.
  • Example 36 The synthesis of Example 36 was as described in Example 35, using dimethylamine hydrochloride instead of methylamine hydrochloride in step A.
  • 19 F NMR (376 MHz, DMSO-d 6 ) ⁇ -112.51, -120.42.
  • LCMS (m/z): 554.5 (M+H).
  • Step A tert-butyl 4-(2-acetamido-7-bromo-6-chloro-3-cyano-8-fluoroquinolin-4-yl)piperazine-1-carboxylate
  • Step B to Step D N-(4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro- 3-cyano-8-fluoroquinolin-2-yl)acetamide
  • Example 37 The subsequent synthetic procedure of Example 37 was carried out as described in Example 19, using 4-(2-acetamido-7-bromo-6-chloro-3-cyano-8-fluoroquinolin-4-yl in step B) ) piperazine-1-carboxylate tert-butyl ester instead of 4-(7-bromo-2,6-dichloro-3-cyano-8-fluoro-2-hydroxyquinolin-4-yl)piperazine-1- tert-butyl carboxylate.
  • Step A 4-(7-Bromo-6-chloro-3-cyano-8-fluoro-2-((trimethylsilyl)ethynyl)quinolin-4-yl)piperazine-1-carboxylic acid tert-butyl ester
  • Step B 4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-8-fluoro- 2-((Trimethylsilyl)ethynyl)quinolin-4-yl)piperazine-1-carboxylate tert-butyl ester
  • Step C 4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-2-ethynyl -8-Fluoroquinolin-4-yl)piperazine-1-carboxylate tert-butyl ester
  • Steps D and E 4-(4-Acryloylpiperazin-1-yl)-7-(2-amino-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-2-ethynyl -8-Fluoroquinoline-3-carbonitrile
  • Step A 4-(7-(2-((tert-butoxycarbonyl)amino)-7-fluorobenzo[d]thiazol-4-yl)-6-chloro-3-cyano-8-methoxy Quinolin-4-yl)piperazine-1-carboxylate tert-butyl ester
  • Example 39 The steps of removing the protecting group and introducing the acryloyl chloride involved in the subsequent synthesis of Example 39 can basically be carried out by referring to the method described in Example 1.
  • Luminescent Cell Viability Assay kit evaluates and verifies the proliferation inhibitory activity of the compounds of the present invention on KRas G12C mutated NCI-H358 human non-small cell lung cancer cells.
  • a control group i.e., a culture medium control
  • 3D complete medium 10% FBS in RPMI1640 solution containing 1% methylcellulose
  • Control group ie, cell control
  • compound AMG510 or the following reference compounds were used as positive controls. Place the cell plate in a cell incubator overnight.
  • the end-point detection thaw the CellTiter-Glo reagent and move the cell plate to room temperature for 30 minutes, add 100 ⁇ L of CellTiter-Glo to each well of the cell plate, shake it on an orbital shaker for 5 minutes to fully lyse the cells, and place the cell plate on the cell plate.
  • the luminescence value of each well was scanned at full wavelength with a multi-function microplate reader (Molecular Devices, Spectramax M3 microplate reader).
  • Inhibition rate% [1-(Lum test drug -Lum culture medium control )/(Lum cell control -Lum culture medium control )] ⁇ 100%
  • IC50 values were calculated using GraphPad Prism 7.0 software, fitting the data using nonlinear S-curve regression, resulting in a dose-response curve.
  • the compounds of the present invention showed satisfactory anti-cell proliferation activity on KRas G12C-mutated NCI-H358 human non-small cell lung cancer cells.
  • the tested example compounds all showed anti-cell proliferative activity with IC 50 values generally ⁇ 1 ⁇ M, such as ⁇ 0.5 ⁇ M, ⁇ 0.1 ⁇ M, preferably ⁇ 50 nM, more preferably ⁇ 20 nM, most preferably ⁇ 10 nM, for example
  • Compounds of Examples 17, 20, 32, 33, 34, 37 all showed IC50 values of ⁇ 50 nM, and compounds of Examples 1, 2, 3, 5, 6, 8, 9, 10, 39 showed IC50 values of ⁇ 20 nM.
  • Specific data for some representative example compounds are shown in Table 1.
  • Example 1 94.7 0.015
  • Example 2 97.6 0.003
  • Example 3 97.7 0.009
  • Example 4 94.8 0.053
  • Example 5 94.9 0.007
  • Example 6 95.0 0.017
  • Example 7 90.0 0.26
  • Example 8 94.0 0.018
  • Example 9 98.8 0.0086
  • Example 10 96.1 0.018 Reference Compound A 96.9 0.011 Reference Compound B / 0.35
  • Example 2 Rat pharmacokinetic properties of the compounds of the present invention
  • ⁇ Test material Male SD rats, age: 6-8 weeks, body weight 220-250g, purchased from Zhaoyan (Suzhou) New Drug Research Center Co., Ltd.; Tolbutamide (Aladdin, Item No. H1401054) ; Sulfobutyl ⁇ -cyclodextrin (Captisol, Shandong Binzhou Zhiyuan Bio, Cat. No. 20191013); Propylene Glycol (15) Stearate (Solutol, Meilun Bio, Cat. No. S0206A); DMSO (Vetec, Cat. No. WXBD0293V); Acetonitrile (Sigma-Aldrich, Cat. No. WXBD1744V); methanol (Sigma-Aldrich, Cat. No. WXBD2831V).
  • Example analysis ⁇ Establish a compound LC-MS/MS analysis method.
  • Standard curve preparation Pipet 20 ⁇ L of 1mg/mL DMSO stock solution for each compound, transfer it to 900 ⁇ L of 50% methanol working solution, and dilute step by step to obtain a concentration of 20000, 10000, 5000, 1000, 500, 100, 50, 20 , 10ng/mL standard curve working solution, then draw 5 ⁇ L standard curve working solution and mix with 45 ⁇ L rat blank plasma to obtain a standard concentration of 2000, 1000, 500, 100, 50, 10, 5, 2, 1ng/mL Curve, used to quantify unknown samples.
  • Sample pretreatment 50 ⁇ L unknown plasma sample and standard curve sample, add 250 ⁇ L acetonitrile containing internal standard as a precipitant, precipitate plasma protein, extract the test compound in the plasma, centrifuge at low temperature for 20 minutes, take the supernatant, mix the supernatant with 0.1% formic acid aqueous solution was mixed, and 5 ⁇ L was injected to analyze the blood concentration of the drug.
  • Compound AMG510 Prepared as described in Lanman B et al, J. Med. Chem. 2020, 63, 52-65.
  • Example 9 has excellent pharmacokinetic properties, as shown in Table 3 for details.
  • Example 3 Antitumor activity of compounds of the present invention in human non-small cell lung cancer NCI-H358 xenograft mouse model
  • This experiment evaluated and verified the proliferation inhibitory activity of the compounds of the present invention in the human non-small cell lung cancer NCI-H358 xenograft mouse model.
  • the NCI-H358 cell line carries the KRAS G12C mutation, which was provided by Kangyuan Bochuang Biotechnology (Beijing) Co., Ltd. (from ATCC, product number CRL-5807).
  • mice 6-8 week old female NPSG mice were subcutaneously inoculated on the right shoulder with 5 ⁇ 10 6 NCI-H358 cells (containing 50% Matrigel) in an inoculation volume of 0.1 mL.
  • NCI-H358 cells containing 50% Matrigel
  • Dosing was started immediately after grouping, and the day of dosing was regarded as day 0.
  • Dosing was given once a day by gavage at a dose of 10 mg/kg or a solvent control (10% cyclodextrin in 50 mM citrate buffer pH 5.0). During the experiment, tumor volume and body weight were measured twice a week.
  • Average tumor inhibition rate TGI% [(C mean -C 0 mean )-(T mean -T 0 mean )]/(C mean -C 0 mean )*100%, where T is the administration group tumor volume, T 0 is the initial tumor volume of the administration group, C is the tumor volume of the control group, and C 0 is the initial tumor volume of the control group.
  • Embodiment 4 the compound of the present invention to cytochrome P450 inhibition test
  • ⁇ Test materials Human liver microsomes (Corning company, product number 452161); reduced nicotinamide adenine dinucleotide phosphate (NADPH, MCE company, product number HY-F0003/CS-4998); phenacetin, double Chlorophenolic acid, ⁇ -naphthoflavone, omeprazole and ketoconazole were purchased from TCI; S-mephenytoin and testosterone were purchased from CAYMAN; midazolam was purchased from Bioreclamation IVT; quinidine was purchased from Damas -beta; sulfaphenazole was purchased from MCE; bufurolol was purchased from TRC.
  • K-buffer 100 mM potassium phosphate buffer (K-buffer) was prepared with potassium dihydrogen phosphate and dipotassium hydrogen phosphate, and the pH was adjusted to 7.4.
  • To make up 400X test compound and reference inhibitor Dissolve 8 ⁇ L of a 10 mM stock of test compound in 12 ⁇ L of acetonitrile.
  • 4 ⁇ NADPH potassium phosphate solution 66.7mg NADPH was added to 10mL 0.1M K-buffer, pH 7.4.
  • Preparation of 4x substrate potassium phosphate solution prepare different substrates according to the concentration requirements with 10mL of 0.1M K-buffer to prepare the solution required for 4x concentration determination.
  • HMM human liver microsomes
  • Example 5 The proliferation inhibitory effect of the compounds of the present invention on a series of KRas mutant cells
  • the CellTiter-Glo (CTG) kit of Promega Company was used to evaluate the antiproliferative activity of the representative compounds of the present invention on 12 KRas mutant tumor cell lines.
  • ⁇ Test materials RPMI1640 medium (Hyclone, product number SH30809.01); fetal bovine serum (FBS) (Gibco, product number 10099-141); phosphate buffered saline PBS (Solarbio, product number P1020-500); DMSO (Sigma, Cat. No. D8418-1L); Assay Kit CTG (Promega, Cat. No. G7573); 96-well cell culture plate (Thermo, Cat. No. 165305); Shaker (QILINBEIER, Cat. No. QB-9001); Cell incubator (Thermo Scientific, Cat. No. Model 3100 Series); microscope (OLYMPUS, cat. no.
  • CKX41SF multi-plate reader
  • BMG LABTECH cat. no. Plus
  • biological safety cabinet Thermo, Cat. No. Model 1300 Series A2.
  • Various cell lines used in the following experiments were purchased from Kangyuan Bochuang Biotechnology (Beijing) Co., Ltd.
  • Each of the above cell lines was cultured in the indicated complete medium at 37°C, 5% CO 2 , and cells in logarithmic growth phase were harvested and counted using a platelet counter. Cell viability was detected by trypan blue exclusion method to ensure cell viability was above 90%. Cell density was adjusted using complete medium and then seeded in 96-well cell culture plates at 90 ⁇ L per well for a total of 3000 cells. Cells in 96-well plates were cultured at 37°C, 5% CO2 .
  • Inhibition rate% [1-(Lum test drug -Lum culture medium control )/(Lum cell control -Lum culture medium control )] ⁇ 100%
  • Example 6 Inhibitory effect of the compounds of the present invention on the proliferation of KRas G12C inhibitor-resistant model cells
  • the CellTiter-Glo (CTG) kit provided by Promega was used to evaluate the effect of the test compounds on the cell proliferation of 5 KRas cell lines (with acquired resistance to the KRas G12C inhibitor Adagrasib), and the reference compounds A and AMG510 served as control compounds.
  • Example 5 The experimental materials and instruments used are the same as those listed in Example 5 above.
  • the various cell lines used in this experiment were from Kangyuan Bochuang Biotechnology (Beijing) Co., Ltd.
  • Example 7 Using the same method and conditions as Example 5, the cell growth inhibitory activity of the test compound was investigated, and the results are shown in Table 7.
  • Example 7 In vivo pharmacodynamic study of the compounds of the present invention on human pancreatic cancer Mia PaCa-2 cells subcutaneously xenografted tumor BALB/c nude mice model
  • ⁇ Test material BALB/c nude mice, 6-8 weeks old, female, Zhejiang Weitong Lihua Laboratory Animal Technology Co., Ltd. Human pancreatic cancer Mia PaCa-2 cells (ATCC, Cat. No. CRL-1420). Matrigel (Matrigel, Corning, Cat. No. 356234).
  • TGI (%) [1-(the average tumor volume at the end of the treatment group-the average tumor volume at the beginning of the treatment group)/(solvent control) The average tumor volume at the end of treatment in the control group - the average tumor volume in the solvent control group at the beginning of treatment)] ⁇ 100%.
  • Example 9 Group Tumor volume (mm 3 ) (day 21) TGI(%) solvent control 1202 / Example 9 (30mpk) 162 97.7% Example 9 (10mpk) 246 89.8% Example 9 (3mpk) 665 50.4% AMG510 (10mpk) 432 72.3% .

Abstract

可用作KRas抑制剂的具有式(I)结构的化合物、包含这类化合物的药物组合物、制备这类化合物的方法以及这些化合物在治疗癌症中的用途。

Description

用于癌症治疗的KRas抑制剂 技术领域
本发明涉及药物化学领域。更具体地,本发明涉及一类可用作KRas抑制剂的具有新结构的化合物、包含这类化合物的药物组合物、制备这类化合物的方法以及这些化合物在治疗癌症中的用途。
背景技术
Ras,即大鼠肉瘤致癌基因同系物,代表一组密切相关的单体球形蛋白,属于GTP酶蛋白家族。具体而言,在正常生理条件下,Ras接受生长因子和各种其他细胞外信号而被激活,负责调节细胞生长、存活、迁移和分化等功能。Ras的这些调节功能是通过GDP结合状态和GTP结合状态之间的转换即“分子开关”来进行(Alamgeer等人,Current Opin Pharmacol.2013,13:394-401)。与GDP结合的Ras是非活性形式,处于休眠或关闭状态,此时信号系统关闭,当其暴露于一些促生刺激时会被活化,例如其可以被鸟嘌呤核苷酸交换因子(GEF)诱导而释放GDP并与GTP结合,结果是Ras被由此“开启”,从而转化为Ras活性形式,其募集并活化各类下游效应子,进行信号传递,能够将细胞表面的信号传送至细胞质中,从而控制众多关键的细胞过程如分化、存活和增殖(Zhi Tan等人,Mini-Reviews in Medicinal Chemistry,2016,16,345-357)。
Ras具有GTP酶活性,其可以裂解GTP的末端磷酸而将其转化为GDP,即将其自身转化为非活性状态。但是Ras的内源性GTP酶活性非常低,将GTP-Ras转化为GDP-Ras需要外源性蛋白GAP(GTP酶激活蛋白)。GAP与Ras相互作用并促进GTP向GDP的转化。因此,任何影响Ras与GAP相互作用或者影响GTP向GDP转化的Ras基因突变,都会导致Ras长时间处于活化状态,由此向细胞持续传达生长和分裂的信号,刺激细胞不断增殖,最终导致肿瘤形成和发展。
在人类肿瘤相关的基因中,存在三种遍在表达的Ras基因H-RAS、K-RAS和N-RAS,其分别编码高度同源的、约21KDa的HRas、NRas、KRas蛋白。1982年,研究人员首次发现Ras在癌细胞系中突变活化(Chang,E.H.等人,Proceedings of the National Academy of Sciences of the United States of America,1982,79(16),4848-4852)。随后在不同癌症类型中进行的大型基因组测序研究揭示,Ras蛋白在超过30%的癌症类型中发生突变,尤其在胰腺癌(>90%)、结肠癌(45%)和肺癌(35%)中的突变率最高。转基因和基因工程小鼠模型也已经揭示,突变的Ras蛋白足以驱动并引发多种类型的癌症,且Ras致癌基因对于多种癌症类型的肿瘤的维持和进展也是至关重要的,例如在Ras突变癌症细胞系和癌症动物模型中,已经显示RNA干预能够减缓肿瘤的生长。这些研究使得Ras肿瘤蛋白成为药学领域中广为接受的非常有吸引力的抗癌药物靶点。
研究表明,Ras突变最常见于KRas,约85%的Ras突变驱动的癌症中可以观察到KRas突变;绝大部分Ras突变发生在密码子G12、G13和Q61上,其中约80%的KRas突变又发生于密码子12的甘氨酸处,例如G12C突变、G12D突变、G13D突变等。KRas突变常见于胰腺癌、肺腺癌、结直肠癌、胆囊癌、甲状腺癌和胆管癌,也可见于25%的非小细胞肺癌患者中(McCormick,F.等人,Clinical Cancer Research 21(8),1797-1801,2015)。因此,KRas突变蛋白已经成为Ras药物靶点研究中最重要的分支,对于其抑制剂的开发也被视为抗癌/肿瘤药物开发中非常具有前景的研发方向。
但是,过去三十多年针对Ras的药物研发显示,由于Ras蛋白表面光滑,缺少明显的用于结合小分子抑制剂的沟状或口袋装结构,而且其对鸟嘌呤底物的亲和力非常高(皮摩尔级),使得其小分子抑制剂的开发陷入了难以解决的困境,由此Ras在业内长久以来被认为是“不可成药的”靶点。尽管如此,靶向Ras突变蛋白的持续努力已经取得了一些令人鼓舞的成果,一系列Ras抑制剂被开发出来,它们通过多种途径对Ras、尤其KRas突变进行抑制,包括直接靶向Ras、抑制Ras的表达水平、破坏Ras蛋白的定位、靶向合成致死组分、靶向Ras-GEF相互作用、靶向Ras和效应子的相互作用以及靶向Ras的二聚化(Zhi Tan等人,Mini-Reviews in Medicinal Chemistry,2016,16,345-357)。
已经开发的Ras抑制剂,例如针对最常见的突变蛋白KRas-G12C的抑制剂,包括变构共价抑制剂,例如6H05系列、喹唑啉系列、ARS系列以及四氢吡啶并嘧啶系列,还有正向结合共价抑制剂,这些抑制剂被综述在文献(Duan Ni等人,Pharmacology&Therapeutics,https://doi.org/10.1016/j.pharmthera.2019.06.007)中。若干专利文献中也述及了各种结构类型的KRas抑制剂,例如CN10256421、US2019/0144444A1以及WO2019/110751A1等。
但是,上述KRas抑制剂仍然存在正待解决的问题,其“成药性”仍然不令人满意。例如,很多KRas依赖性癌症对于这类靶向治疗剂容易产生耐药性、存在副作用如脱靶效应、产生化学活性的代谢物、代谢稳定性差,或者产生免疫原性的共价加合物(John P.O’Bryan等人,Pharmacological Research 139(2019)503-511;Duan Ni等人,Pharmacology&Therapeutics,https://doi.org/10.1016/j.pharmthera.2019.06.007)。因此,临床上仍然需要更多可供选择的KRas抑制剂,这些抑制剂期望具有与已有抑制剂相当或改善的KRas抑制活性、改善的“成药性”、更好的安全性如较少的药物相互作用或代谢性质、改善的药代动力学性质,和/或针对不同的患者群体或者特定的肿瘤类型具有更高的选择性。
本发明提供了具有KRas突变蛋白抑制活性的新结构抑制剂化合物。这些本发明化合物、尤其是本发明优选的化合物因具有改进的结构模式,相比现有技术已有的KRas突变蛋白抑制剂,具有以下技术效果:
(1)保留了相当的或增强的、甚至显著增强的KRas突变蛋白以及相关癌细胞增殖抑制活性;
(2)不同的生物活性谱而用于不同的疾病类型或患者群体;
(3)具有改善的代谢稳定性,从而带来更好的药动学性质;
(4)具有改善的物理化学性质,从而具有良好的成药性和安全性,比如更容易在体内吸收等。
发明简述
本发明人通过研究发现,本文所定义的式(I)的化合物、其异构体或它们药学上可接受的盐或溶剂合物是有效的Ras突变、尤其KRas突变蛋白抑制剂,能够抑制细胞中的Ras突变、尤其KRas活性,可用于治疗或预防Ras突变、尤其KRas突变蛋白介导的或得益于Ras突变、尤其KRas突变蛋白抑制的疾病或病症,尤其是可通过抑制Ras突变、尤其KRas突变蛋白来抑制异常的细胞增殖、从而治疗或预防肿瘤或癌症。
本发明第一方面提供式(I)的化合物、其异构体或它们药学上可接受的盐或溶剂合物,
Figure PCTCN2021128977-appb-000001
其中,
A选自C-R a或N,其中R a选自卤素、CN、硝基、C 3-6环烷基或任选被卤素取代的C 1-6烷基;
R 1、R 2和R 3各自独立地选自H、卤素或C 1-6烷基,其中所述烷基任选被独立地选自卤素、-N(R c) 2、-OR c或3-8元杂环烷基的基团取代;
R b在每次出现时独立地选自H、卤素、CN或任选被卤素或CN取代的C 1-6烷基;
X选自键、-O-或-NH-;
R 4选自H、卤素、C 1-6烷基、-C 0-6烷基-C 6-10芳基、-C 0-6烷基-C 3-8环烷基、-C 0-6烷基-C 3-8环烯基、-C 0-6烷基-3-8元杂环烷基、-C 0-6烷基-3-8元杂环烯基、-C 0-6烷基-5-10元杂芳基,其中所述烷基、芳基、环烷基、环烯基、杂环烷基、杂环烯基和杂芳基任选被独立地选自以下的一个或多个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-SR c、-(CR cR c) 0-6-(CO) 0- 1-OR c、-(CR cR c) 0-6-(CO) 0-1-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-(CO) 0-1-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环 状基团上的原子及相邻的原子一起形成4-7元含氮环状基团;
R c在每次出现时独立地选自H或任选被卤素取代的C 1-6烷基,或者其中连接在同一个碳原子或氮原子上的两个R c各自独立地与它们所连接的碳原子或氮原子一起形成3-6元环状基团;
E选自卤素、-O-R d或-N(R d) 2-,其中R d各自独立地为H或任选被卤素取代的C 1-6烷基;
R 5
Figure PCTCN2021128977-appb-000002
m为0或1;
Figure PCTCN2021128977-appb-000003
表示芳族环;
B和D各自独立地选自N或C;
Z、G和Y各自独立地选自C、N、O或S;
R 6、R 7和R 8各自独立地选自H、卤素和任选被卤素取代的C 1-6烷基;
条件是,B和D不同时为N;且Z、G、Y、D、B中至多三个不为C。
本发明进一步提供式(II)化合物、其异构体、药学上可接受的盐或溶剂合物。
Figure PCTCN2021128977-appb-000004
其中R 1、R 2、R 3、R 4、R 6、R 7、R 8、A、R b各自具有上文对式(I)化合物给出的含义。
本发明第二方面提供了包含本发明式(I)或式(II)的化合物、其异构体或它们药学上可接受的盐或溶剂合物的药物组合物。
本发明第三方面提供了用作药物的式(I)或式(II)的化合物、其异构体或它们药学上可接 受的盐或溶剂合物。
本发明第四方面提供了用于治疗和/或预防由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变介导的疾病的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物。
本发明第五方面提供了本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物或包含其的药物组合物在制备用于治疗和/或预防由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变介导的疾病的药物中的用途。
本发明第六方面提供了治疗和/或预防由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变介导的疾病的方法,包括向有需要的对象施用治疗有效量的本发明式(I)或式(II)的化合物、其异构体或它们药学上可接受的盐或溶剂合物或包含其的药物组合物。
本发明第七方面提供了用于制备本发明式(I)或式(II)的化合物、其异构体或它们药学上可接受的盐或溶剂合物的方法。
本发明第八方面提供了药物组合,其包含本发明式(I)或式(II)的化合物、其异构体或它们药学上可接受的盐或溶剂合物和一种或多种其他药物活性剂。
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,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“Ras突变”或“Ras突变蛋白”是指其中一个或多个密码子发生突变的Ras基因所编码和表达的蛋白,典型地包括但不限于Ras的密码子12位的甘氨酸、密码子13位的甘氨酸或密码子61位的谷氨酰胺发生突变的Ras蛋白,例如突变的HRas、NRas或KRas。这些残基位于Ras的活性位点,其突变可损害Ras的固有的或GAP-催化的GTP酶活性,导致与GTP结合的Ras持续存在。
对本发明的目的而言,“Ras突变”或“Ras突变蛋白”可互换使用,且一般地是指突变的HRas、NRas或KRas,例如但不限于HRas-G12C(密码子G12处甘氨酸向半胱氨酸的突变)、NRas-G12C、KRas-G12C、KRas-G12D(密码子G12处甘氨酸向天冬氨酸的突变)、KRas-G13D(密码子G13处甘氨酸向天冬氨酸的突变);特别地是指KRas突变蛋白,更特别地是指KRas-G12C突变蛋白、KRas-G12D突变蛋白、KRas-G13D突变蛋白,最特别地是指KRas- G12C突变蛋白。
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式(I)化合物、其异构体或它们药学上可接受的盐或溶剂合物,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Ras突变介导的疾病、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Ras突变介导的疾病、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式(I)化合物、其异构体或它们药学上可接受的盐或溶剂合物,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KRas活性)降低量约、至多约或至少约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。就本发明式(I)化合物而言,相对于Ras蛋白的一个或多个密码子发生的各类突变,其具有选择性抑制KRas、HRas或NRas蛋白的G12或G13突变的能力,例如G12C突变、G12D突变和G13D突变,优选选择性抑制KRas蛋白的G12C突变的能力。例如,与对另一种特定Ras突变相比,本发明对特定Ras突变具有至少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Ras突变的活性相比,对特定Ras突变(例如KRas-G12C)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“Ras突变介导的疾病”是指Ras突变对所述疾病的发生和发展起到促进作用,或抑制Ras突变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,“Ras突变介导的疾病”优选指的是KRas突变介导的疾病,最优选KRas-G12C介导的疾病,更进一步优选癌症或肿瘤。
本文所用的术语“癌症”或“肿瘤”是指异常的细胞生长和增殖,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。对本发明的各个方面而言,所述癌症或肿瘤包括但不限于肺腺癌、肺癌、骨癌、胰腺癌、皮肤癌、头颈癌、皮肤或眼内黑素瘤、子宫癌、卵巢癌、直肠癌、肛门区域癌、胃癌、结肠癌、乳腺癌、输卵管癌、子宫内膜癌、子宫颈癌、阴道癌、外阴癌、霍奇金病、食道癌、小肠癌、内分泌系统癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、尿道癌、阴茎癌、前列腺癌、慢性或急性白血病、淋巴细胞性淋巴瘤、膀胱 癌、肾脏或输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊柱肿瘤、脑干神经胶质瘤或垂体腺瘤。
对于本发明的各个方面,优选地,所述癌症或肿瘤与Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变相关,包括但不限于上述肿瘤类型以及其优选范围。本发明特别优选的肿瘤包括肺癌、肺腺癌、结肠癌、直肠癌、胰腺癌、子宫内膜癌、胆管癌、白血病和卵巢癌。
本文所用的术语“受试者”、“个体”或“患者”是指脊椎动物。在某些实施方案中,脊椎动物为哺乳动物。哺乳动物包括但不限于农场动物(如牛)、运动动物、宠物(如豚鼠、猫、狗、兔子和马)、灵长类动物、小鼠和大鼠。在某些实施方案中,哺乳动物是人类。
本文所用的术语“治疗有效量”是指通常足以对需要治疗的癌症或肿瘤患者产生有益治疗效果的量或剂量。本领域技术人员可以通过常规方法、结合常规影响因素来确定本发明中活性成分的有效量或剂量。
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,例如这些活性剂已知调节其他生物活性通路,或者调节本发明化合物所涉及生物活性通路中的不同组分,或甚至是与本发明化合物的生物靶点相重叠。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,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。
一方面,可以与本发明化合物组合使用的其他活性剂包括但不限于化疗剂、治疗性抗体和放疗,例如烷化剂、抗代谢物、细胞周期抑制剂、有丝分裂抑制剂、拓扑异构酶抑制剂、抗激素类药物、血管生成抑制剂或细胞毒性剂。
本文所用的术语“药学上可接受的”意指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“药学上可接受的盐”是指保留了母体化合物的生物学有效性和性质并且在生物学或其它方面不是不可取的那些盐,包括酸加成盐和碱加成盐。“药学上可接受的酸加成盐”可由具有游离碱的化合物与无机酸或有机酸形成,无机酸例如盐酸、氢溴酸、硫酸、硝酸、碳酸、磷酸等,有机酸可以选自脂族、脂环族、芳香族、芳脂族、杂环类、羧酸类和磺酸类有机酸,如甲酸、乙酸、丙酸、乙醇酸、葡萄糖酸、乳酸、丙酮酸、草酸、苹果酸、马来酸、丙二酸、琥珀酸、富马酸、酒石酸、柠檬酸、天冬氨酸、抗坏血酸、谷氨酸、邻氨基苯甲酸、苯甲酸、肉桂酸、扁桃酸、双羟萘酸、苯乙酸、甲磺酸、乙磺酸、苯磺酸、对甲苯磺酸、水杨酸等。“药学上可接受的碱加成盐”包括衍生自无机碱如钠、钾、锂、铵、钙、镁、铁、锌、铜、锰、铝的盐等的那些,以及衍生自药学上可接受有机无毒碱的盐,包括但不限 于伯胺、仲胺和叔胺、取代铵,包括天然存在的取代胺、环状胺和碱性离子交换树脂,如氨、异丙胺、三甲胺、二乙胺、三乙胺、三丙胺、乙醇胺、二乙醇胺、2-二甲氨基乙醇、2-二乙氨基乙醇、氨丁三醇、二环己胺、赖氨酸、精氨酸、组氨酸、咖啡因、普鲁卡因、海巴明、胆碱、甜菜碱、乙二胺、葡糖胺、甲基葡糖胺、三乙醇胺、可可碱、嘌呤、哌嗪、哌啶、N-乙基哌啶、聚胺树脂等。
本文所用的术语“异构体”是指化合物在结构上可能存在的任何立体异构体、对映体混合物、包括外消旋物、非对映异构体混合物、几何异构体、阻旋异构体和/或互变异构体。所述异构体立体化学的确定和分离方法为本领域技术人员所熟知(S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-Hill Book Company,New York;和Eliel,E.和Wilen,S.,“Stereochemistry of Organic Compounds”,John Wiley&Sons,Inc.,New York,1994),故本发明涵盖上文所定义式(I)化合物的所有可能的异构体形式,及其药学可接受的盐或溶剂合物。
本文化合物结构式或结构片段中使用的
Figure PCTCN2021128977-appb-000005
Figure PCTCN2021128977-appb-000006
表示立体中心即手性中心的绝对构型,相应地在本发明所提供的化合物或中间体的命名中以R和S表示关于该手性中心的绝对构型,该绝对构型的确定是本领域技术人员所熟知的。
本文所涉及结构片段中使用的
Figure PCTCN2021128977-appb-000007
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“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。
本文所使用的术语“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,从而形成本发明化合物或其药学上可接受的盐的同位素变化形式,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例如 2H、 3H、 13C、 14C、 15N、 17O、 18O、 31P、 32P、 35S、 18F和 36Cl。应当理解,本发明化合物及其药学上可接受的盐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。例如其中掺入了放射性同位素(例如 3H或 14C)的那些本发明化合物及其药学上可接受的盐的某些同位素变化形式,可用于药物和/或底物组织分布研究。氚代即 3H和碳-14即 14C同位素由于易于制备和可检测性,因此是特别优选的。此外,用同位素如氘即 2H取代可以提供由于更高的代谢稳定性而产生的某些治疗优势,例如增加的体内半衰期或降低的剂量要求,因此在某些情况下是优选的。另外,可以制备被正电子发射同位素(例如 11C、 18F、 15O和 13N)取代的本发明化合物,它们可以用于正电子断层扫描 (PET)研究用于底物受体占有率检测。
本文所用的术语“代谢物”意指特定化合物经由体内代谢生成的产物。这类产物可例如源自所施用化合物的氧化、还原、水解、酰胺化、脱酰胺化、酯化、去酯化、酶促剪切等。代谢物产物的鉴定和分析以本领域技术人员熟知的方式进行。
本文所用的术语“前药”是指可以在生理条件下或通过溶剂分解转化为本文所述的生物活性化合物、例如式(I)化合物的化合物。因此,术语“前药”是指药学上可接受的生物活性化合物的前体。在一些方面,前药在施用于受试者时是无活性的,但是例如通过水解在体内转化为活性化合物。前药化合物通常在哺乳动物生物体中提供溶解性、组织相容性或延迟释放的优点(参见例如Bundgard,H.,Design of Prodrugs(1985),第7-9,第21-24页(Elsevier,Amsterdam)。前药的讨论可见于Higuchi,T.等人的ACS Symposium Series,第14卷,以及“药物设计中的生物可逆性载体”,爱德华·B·罗氏(Edward B.Roche),美国Pharmaceutical Association&Pergamon Press,1987,将其全部内容引入本文作为参考。术语“前药”还意指包括任何共价键合的载体,当将这种前药给予哺乳动物受试者时,它们在体内释放活性化合物。如本文所述的活性化合物的前药通常是通过修饰存在于活性化合物中的官能团而制备,使得该修饰物可以在常规操作中或在体内裂解成母体活性化合物。前药包括这样的化合物,其中羟基、氨基或巯基键合至当将该前药施用于哺乳动物时裂解形成游离羟基、游离氨基或游离巯基的任何基团。前药的实例包括但不限于羟基官能团的乙酸酯、甲酸酯和苯甲酸酯衍生物,或活性化合物中胺官能团的乙酰胺、甲酰胺和苯甲酰胺衍生物。在一些实施方案中,前药包括含磷酸盐/酯的前药、含硼酸酯的前药、含硫代磷酸盐/酯的前药、含硫酸盐/酯的前药、含肽的前药、D-氨基酸-修饰的前药、糖基化的前药、含β-内酰胺的前药、含任选取代的苯氧基乙酰胺的前药或含任选取代的苯基乙酰胺的前药以及5-氟胞嘧啶和5-氟尿苷前药。
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,适合于人使用,且具有足够的纯度和足够低的毒性,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“卤素”或“卤代”意指F、Cl、Br或I。此外,术语“被卤素取代的”基团旨在包括单卤代或多卤代基团,其中一个或多个相同或不同的卤素取代基团中的一个或多个氢。
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,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。
如本文中所使用的术语“烷硫基”指-S-烷基,其中所述烷基如以上对于“烷基”所定义。具体地,该术语包括基团-S-C 1-6烷基,更具体的-S-C 1-3烷基。烷硫基的代表性实例包括但不限于甲硫基、乙硫基、丙硫基(包括正丙硫基、异丙硫基)、丁硫基(包括正丁硫基、异丁硫基、叔丁硫基)、戊硫基(包括正戊硫基、异戊硫基、新戊硫基)、己硫基(包括正己硫基、异己硫基)等。特定的烷硫基具有1至3个碳原子。
如本文中所使用的术语“卤素取代的C 1-C 6烷基”指上文所述的C 1-C 6烷基,其中一个或多个(例如1、2、3、4或5个)氢原子被卤素代替。本领域技术人员应当理解,当卤素取代基多于一个时,卤素可以相同也可以不同,并且可以位于相同或不同的C原子上。“卤素取代的C 1-C 6烷基”的实例有例如-CH 2F、-CHF 2、-CF 3、-CCl 3、-C 2F 5、-C 2Cl 5、-CH 2CF 3、-CH 2Cl、-CH 2CH 2CF 3或-CF(CF 3) 2等。
如本文中所使用的术语“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3至10个,3至8个,3至7个,3至6个,5至6个碳原子。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,诸如双环[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“环烯基”意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不饱和烃环结构,包含至少一个(例如1、2、或3个)碳碳双键。环烯基可具有3至12个碳原子(即C 3-C 12环烯基),例如3至10个,3至8个,3至7个,3至6个,5至6个碳原子。适合的环烯基的实例包括但不限于单环结构,如环丙烯基、环丁烯基、环戊烯基、环戊二烯基、环己烯基、环己二烯基、环庚烯基、环庚二烯基、环庚三烯基或环辛烯基。
本文所用的术语“杂环烷基”意指包括一或多个(例如1、2、3或4个)独立地选自O、N及S的杂原子及指定环原子数的单环、稠合多环、螺环或桥接多环非芳族饱和环结构,或其N-氧化物,或其S-氧化物或S-二氧化物。杂环烷基可具有3至12个环成员(可称为3-12元杂环烷基),例如3至10个环成员,3至8个环成员,3至7个环成员,4至7个环成员、4至6个环成员、5至6个环成员。杂环烷基通常含有至多4个(例如1个、2个、3个或4个)杂原子。适合的杂环烷基的实例包括但不限于氮杂环丁烷基、氧杂环丁烷基、硫杂环丁基、吡咯烷基(例如1-吡咯烷基、2-吡咯烷基及3-吡咯烷基)、四氢呋喃基(例如1-四氢呋喃基、2-四氢呋喃基及3-四氢呋喃基)、四氢噻吩基(例如1-四氢噻吩基、2-四氢噻吩基及3-四氢 噻吩基)、哌啶基(例如1-哌啶基、2-哌啶基、3-哌啶基及4-哌啶基)、四氢吡喃基(例如4-四氢吡喃基)、四氢噻喃基(例如4-四氢噻喃基)、吗啉基(例如吗啉代)、硫吗啉基、二噁烷基、哌嗪基或氮杂环庚烷基、二氮杂环庚烷基例如1,4-二氮杂环庚基、3,6-二氮杂-双环[3.1.1]庚基或3-氮杂-双环[3.2.1]辛基。杂环烷基中与化合物其余部分连接的原子可以是碳原子,也可以是杂原子,只要化学上可行即可。
优选的杂环烷基例如
Figure PCTCN2021128977-appb-000008
Figure PCTCN2021128977-appb-000009
Figure PCTCN2021128977-appb-000010
应当理解具有不对称中心的结构涵盖其外消旋的和/或单一的对映异构形式,例如
Figure PCTCN2021128977-appb-000011
可代表
Figure PCTCN2021128977-appb-000012
和/或
Figure PCTCN2021128977-appb-000013
本文所用的术语“杂环烯基”意指包含至少一个(例如1、2或3个)双键的本文所定义的“杂环烷基”。适合的杂环烯基的实例包括但不限于:
Figure PCTCN2021128977-appb-000014
其中各W选自CH 2、NH、O及S;各Y选自NH、O、C(=O)、SO 2及S;且各Z选自N及CH,条件是各个环中至少包含一个选自N、O或S的原子;例如吡咯啉基(例如1-吡咯啉基、2-吡咯烷基、3-吡咯啉基、4-吡咯啉基或5-吡咯啉基)、二氢呋喃基(例如1-二氢呋喃基、2-二氢呋喃基、3-二氢呋喃基、4-二氢呋喃基或5-二氢呋喃基)、二氢噻吩基(例如1-二氢噻吩基、2-二氢噻吩基、3-二氢噻吩基或4-二氢噻吩基)、四氢吡啶基(例如1-、2-、3-、4-、5-或6-四氢吡啶基)、四氢吡喃基(例如4-四氢吡喃基)或四氢噻喃基(例如4-四氢噻喃基)。
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“杂芳基”意指包括一或多个(例如1、2、3或4个)独立地选自O、N及 S的杂原子及指定环原子数的单环或稠合多环芳族环结构,或其N-氧化物,或其S-氧化物或S-二氧化物。具体地,该芳族环结构可具有5至10个环成员。杂芳基可为例如5-6元单环、或由稠合的两个6元环、稠合的两个5元环、稠合的6元环和5元环、或稠合的5元环和4元环形成的稠合双环结构。杂芳基环通常将含有至多4个杂原子、更通常至多3个杂原子、更通常至多2个、例如单个独立地选自O、N及S的杂原子,其中N和S可以是氧化状态如N氧化物、S=O或S(O) 2。在一个实施方案中,杂芳基环含有至少一个环氮原子、至少一个环硫原子或至少一个环氧原子。例如,杂芳基可以是包含1、2、3或4个独立地选自N、O或S的杂原子的稠合环,例如苯并呋喃、苯并噻吩、吲哚、苯并咪唑、吲唑、苯并三唑、吡咯并[2,3-b]吡啶、吡咯并[2,3-c]吡啶、吡咯并[3,2-c]吡啶、吡咯并[3,2-b]吡啶、咪唑并[4,5-b]吡啶、咪唑并[4,5-c]吡啶、吡唑并[4,3-d]吡啶、吡唑并[4,3-c]吡啶、吡唑并[3,4-c]吡啶、吡唑并[3,4-b]吡啶、异吲哚、嘌呤、中氮茚、咪唑并[1,2-a]吡啶、咪唑并[1,5-a]吡啶、吡唑并[1,5-a]哒嗪、吡咯并[1,2-b]嘧啶、咪唑并[1,2-c]嘧啶、5H-吡咯并[3,2-b]吡嗪、1H-吡唑并[4,3-b]吡嗪、1H-吡唑并[3,4-d]嘧啶、7H-吡咯并[2,3-d]嘧啶、喹啉、异喹啉、噌啉、喹唑啉、喹喔啉、酞嗪、1,6-萘啶、1,7-萘啶、1,8-萘啶、1,5-萘啶、2,6-萘啶、2,7-萘啶、吡啶并[3,2-d]嘧啶、吡啶并[4,3-d]嘧啶、吡啶并[3,4-d]嘧啶、吡啶并[2,3-d]嘧啶、吡啶并[2,3-b]吡嗪、吡啶并[3,4-b]吡嗪、嘧啶并[5,4-d]嘧啶、吡嗪并[2,3-b]吡嗪和嘧啶并[4,5-d]嘧啶。例如,杂芳基可以是包含1或2个独立地选自N、O或S的杂原子的5-6元杂芳基。适合的5元单环杂芳基的实例包括但不限于吡咯基、呋喃基、噻吩基、咪唑基、呋咱基、噁唑基、噁二唑基、噁三唑基、异噁唑基、噻唑基、异噻唑基、吡唑基、三唑基及四唑基;适合的6元单环杂芳基的实例包括但不限于吡啶基、吡嗪基、哒嗪基、嘧啶基及三嗪基。杂芳基中与化合物其余部分连接的原子可以是碳原子,也可以是杂原子,只要化学上可行即可。
取代基被描述为“任选取代的”意指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或更多个,或其中可衍生的任何范围)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。在一个实施方案中,任选取代的基团被1个取代基取代。在另一个实施方案中,任选取代的基团被2个取代基取代。在另一个实施方案中,任选取代的基团被3个取代基取代。在另一个实施方案中,任选取代的基团被4个取代基取代。
有机合成领域普通技术人员均理解,稳定的化学可行的杂环,无论芳族还是非芳族,其中最大杂原子数或所含杂原子的类型由环大小、不饱和度及杂原子的价数决定。一般而言,杂环可具有1至4个杂原子,前提是杂环或杂芳环在化学可行及稳定。
本文所用的术语“羟基”是指-OH基团。
本文所用的术语“巯基”是指-SH基团。
本文所用的术语“硝基”是指-NO 2基团。
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,或其中可衍生的任何范围)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。在一个实施方案中,任选取代的基团具有1个取代基。在另一个实施方案中,任选取代的基团具有2个取代基。在另一个实施方案中,任选取代的基团具有3个取代基。在另一个实施方案中,任选取代的基团具有4个取代基。在另一个实施方案中,任选取代的基团具有5个取代基。
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,C 1-6包括C 1-2、C 1-3、C 1-4、C 2-6、C 3-6等。同理,本发明化合物定义中的n元至n+m元表示环原子数为n至n+m个,例如3-12元环包括3元环、4元环、、5元环、6元环、12元环等,也包括n至n+m元的任何一个范围,例如3-12元环包括3-6元环、3-8元环、3-9元环、4-7元环、4-5元环、5-6元环、5-7元环、5-8元环、5-9元环、6-7元环、6-8元环和6-10元环等。
如在本说明书和随后的权利要求书中所使用的,词语“包含”和该词语的变体如“包括”和“含有”,意指“包括但不限于”,并且不意图排除例如其他添加剂、成分、整数或步骤。当将要素描述为包括多个成分、步骤或条件时,应理解的是,该要素也可以被描述为包括该多个成分、步骤或条件的任何组合,或“由多个或组合的成分、步骤或条件组成”或“基本上由多个或组合的成分、步骤或条件组成”。
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、药盒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,不包括其任何盐、水合物或溶剂化物,除非说明书中指出该剂量基于盐、水合物或溶剂化物的重量。
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优选实施方式中定义的式(I)或式(II)化合物、包括其异构体,包括阻转异构体、对映体混合物、特别是外消旋体、非对映异构体混合物、几何异构体、互变异构体、溶剂化物、代谢物、同位素变体、盐(例如药学上可接受的盐)和前药。
因此,式I化合物的上述各类异构体和衍生物由此均涵盖在本发明范围内,其各自的含义、制备及具体示例如上文“定义”部分所定义,或为本领域技术所熟知。在一些实施方案中,代谢物、同位素变体或前药以及其任意组合被酌情排除在外。然而,优选地为式(I)或式(II)化合物和/或其药学上可接受的盐的基本上纯的对映体(对映异构体纯)或非对映异构体。
本发明还涵盖式(I)或式(II)化合物的N-氧化物,只要这些化合物含有碱性氮原子,例如存在于含氮杂环中的氮原子。本发明的某些化合物可以多晶型或无定形形式存在,故也落入本发明的范围内。
一方面,本发明提供了式(I)的化合物、其异构体或它们药学上可接受的盐或溶剂合物,
Figure PCTCN2021128977-appb-000015
其中,
A选自C-R a或N,其中R a选自卤素、CN、硝基、C 3-6环烷基或任选被卤素取代的C 1-6烷基;
R 1、R 2和R 3各自独立地选自H、卤素或C 1-6烷基,其中所述烷基任选被独立地选自卤素、-N(R c) 2、-OR c或3-8元杂环烷基的基团取代;
R b在每次出现时独立地选自H、卤素、CN或任选被卤素或CN取代的C 1-6烷基;
X选自键、-O-或-NH-;
R 4选自H、卤素、C 1-6烷基、-C 0-6烷基-C 6-10芳基、-C 0-6烷基-C 3-8环烷基、-C 0-6烷基-C 3-8环烯基、-C 0-6烷基-3-8元杂环烷基、-C 0-6烷基-3-8元杂环烯基、-C 0-6烷基-5-10元杂芳基,其中所述烷基、芳基、环烷基、环烯基、杂环烷基、杂环烯基和杂芳基任选被独立地选自以下的一个或多个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-SR c、-(CR cR c) 0-6-(CO) 0- 1-OR c、-(CR cR c) 0-6-(CO) 0-1-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-(CO) 0-1-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环;
R c在每次出现时独立地选自H或任选被卤素取代的C 1-6烷基,或者其中连接在同一个碳原子或氮原子上的两个R c各自独立地与它们所连接的碳原子或氮原子一起形成3-6元环状基团;
E选自卤素、-O-R d或-N(R d) 2-,其中R d各自独立地为H或任选被卤素取代的C 1-6烷基;
R 5
Figure PCTCN2021128977-appb-000016
m为0或1;
Figure PCTCN2021128977-appb-000017
表示芳族环;
B和D各自独立地选自N或C;
Z、G和Y各自独立地选自C、N、O或S;
R 6、R 7和R 8各自独立地选自H、卤素和任选被卤素取代的C 1-6烷基;
条件是,B和D不同时为N;且Z、G、Y、D、B中至多三个不为C;
或其异构体、药学上可接受的盐或溶剂合物。
在一种式(I)化合物的实施方式中,A为N。
在一种式(I)化合物的实施方式中,A为C-R a,其中R a选自卤素。
在一种式(I)化合物的实施方式中,A为C-R a,其中R a为F或Cl,优选Cl。
在一种式(I)化合物的实施方式中,R 1选自H或卤素。
在一种式(I)化合物的实施方式中,R 1为H或F。
在一种式(I)化合物的实施方式中,R 2和R 3均为H。
在一种式(I)化合物的实施方式中,R b为H。
在一种式(I)化合物的实施方式中,R b不为H,且存在1个R b或存在2个R b
在一种式(I)化合物的实施方式中,存在一个R b且R b为C 1-6烷基。
在具体的实施方式中,存在一个R b且R b为C 1-6烷基、优选C 1-3烷基;在更具体的实施方式中,存在一个R b且R b为-CH 3;在更具体的实施方式中,存在一个R b且R b
Figure PCTCN2021128977-appb-000018
在更具体的实施方式中,存在一个R b且R b
Figure PCTCN2021128977-appb-000019
且在哌嗪环上的连接方式为
Figure PCTCN2021128977-appb-000020
在 更具体的实施方式中,存在一个R b且R b
Figure PCTCN2021128977-appb-000021
且在哌嗪环上的连接方式为
Figure PCTCN2021128977-appb-000022
在一种式(I)化合物的实施方式中,存在一个R b且R b为CN;在具体的实施方式中,存在一个R b且R b为CN,且连接于酰胺氮邻位的碳原子上。
在一种式(I)化合物的实施方式中,存在一个R b且R b为C 1-6烷基、优选C 1-3烷基,被CN取代;在具体的实施方式中,存在一个R b且R b为氰基甲基;在更具体的实施方式中,存在一个R b且R b为氰基甲基,且在哌嗪环上的连接方式为
Figure PCTCN2021128977-appb-000023
在一种式(I)化合物的实施方式中,存在两个R b且R b为C 1-6烷基、优选C 1-3烷基;在具体的实施方式中,存在两个R b且R b为-CH 3;在更具体的实施方式中,存在两个R b且R b为-CH 3,且在哌嗪环上的连接方式为
Figure PCTCN2021128977-appb-000024
在一种式(I)化合物的实施方式中,X为键。
在一种式(I)化合物的实施方式中,X为-O-或-NH-。
在一种式(I)化合物的实施方式中,X为-O-。
在一种式(I)化合物的实施方式中,X为-NH-。
在一种式(I)化合物的实施方式中,R 4选自H、卤素、C 1-6烷基、-C 0-3烷基-苯基、-C 0-3烷基-C 3-6环烷基、-C 0-3烷基-C 3-6环烯基、-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基、-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂环烯基和-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,其中所述烷基、苯基、环烷基、环烯基、杂环烷基、杂环烯基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-SR c、-(CR cR c) 0- 6-OR c、-(CR cR c) 0-6-N(R c) 2,其中连接于苯基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环。
在一种式(I)化合物的实施方式中,R 4选自H、卤素、C 1-6烷基、-C 0-3烷基-苯基、-C 0-3烷基-C 3-6环烷基、-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基和-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,其中所述烷基、苯基、环烷基、杂环烷基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-SR c、-(CR cR c) 0-6-OR c、-(CR cR c) 0-6-N(R c) 2,其中连接于苯基、环烷基、杂环烷基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环。
在一种式(I)化合物的实施方式中,R 4选自H、卤素、C 1-6烷基、苯基、C 3-6环烷基、包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基和包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,其中所述烷基、苯基、环烷基、杂环烷基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c、-(CR cR c) 0-6-N(R c) 2,其中连接于苯基、环烷基、杂环烷基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环。
在一种式(I)化合物的实施方式中,-X-R 4为H。
在一种式(I)化合物的实施方式中,-X-R 4不为H。
在一种式(I)化合物的实施方式中,-X-R 4为-OH。
在一种式(I)化合物的实施方式中,-X-R 4为-NH 2
在一种式(I)化合物的实施方式中,-X-R 4为卤素;在具体的实施方案中,-X-R 4为Cl或F。
在一种式(I)化合物的实施方式中,R 4为C 1-6烷基,任选被1、2或3个独立地选自卤素、C 1-6烷基、羟基、C 1-6烷氧基和-(CR cR c) 0-6-N(R c) 2的基团取代。在具体的实施方式中,示例性R 4包括但不限于-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3、-C(CH 3)(CH 3)(CH 3)、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CH 2CF 3、-CH(CF 3)CH 3、-CH 2CH(CF 3)CH 3、-CH 2CH 2CH 2CF 3、-C(CF 3)(CH 3)(CH 3)、-CH 2OH、-CH 2CH 2OH、-CH 2CH 2CH 2OH、-CH 2CH(OH)CH 3、-CH 2CH(CH 2OH)CH 3、-CH 2CH 2CH 2CH 2OH、-C(CH 2OH)(CH 3)(CH 3)、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、-CH 2CH 2CH 2OCH 3、-CH 2CH(OCH 3)CH 3、-CH 2CH(CH 2OCH 3)CH 3、-CH 2CH 2CH 2CH 2OCH 3、-NH 2、-NHCH 3、-NHCH 2CH 3、-N(CH 3) 2、-N(CH 3)(CH 2CH 3)、-N(CH 2CH 3) 2、-CH 2NH 2、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CH 2NHCH 2CH 3、-CH 2N(CH 3)(CH 2CH 3)、-CH 2N(CH 2CH 3) 2、-CH 2CH 2NH 2、- CH 2CH 2NHCH 3、-CH 2CH 2N(CH 3) 2、-CH(CH 3)N(CH 3) 2、CH 2CH 2NHCH 2CH 3、-CH 2CH 2N(CH 2CH 3) 2、-CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2NHCH 3、-CH 2CH 2CH 2N(CH 3) 2、-CH 2CH(NH 2)CH 3、-CH 2CH(CH 2NH 2)CH 3、-CH 2CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2CH 2NHCH 3和-CH 2CH 2CH 2CH 2N(CH 3) 2
在一种优选的实施方式中,R 4为C 1-6烷基,任选被1、2或3个独立地选自卤素、C 1-6烷基、C 1-6烷氧基的基团取代;更优选地,R 4选自-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2OCH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3或-CF 3
在一种式(I)化合物的实施方式中,-X-R 4为C 1-6烷基,其中C 1-6烷基具有以上对作为R 4的C 1-6烷基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-O-C 1-6烷基,其中C 1-6烷基具有以上对作为R 4的C 1-6烷基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-NH-C 1-6烷基,其中C 1-6烷基具有以上对作为R 4的C 1-6烷基给出的实施方式、优选或更优选实施方式的含义。
在一种式(I)化合物的实施方式中,R 4为-C 0-3烷基-苯基、优选苯基,所述苯基任选被1、2或3个独立地选自卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2的基团取代。在具体的实施方式中,示例性取代基包括但不限于:F、Cl、Br、I、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3、-C(CH 3)(CH 3)(CH 3)、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CH 2CF 3、-CH(CF 3)CH 3、-CH 2CH(CF 3)CH 3、-CH 2CH 2CH 2CF 3、-C(CF 3)(CH 3)(CH 3)、-CH 2OH、-CH 2CH 2OH、-CH 2CH 2CH 2OH、-CH 2CH(OH)CH 3、-CH 2CH(CH 2OH)CH 3、-CH 2CH 2CH 2CH 2OH、-C(CH 2OH)(CH 3)(CH 3)、-OH、-OCH 3、-OCH 2CH 3、-OCH 2CH 2CH 3、-OCH(CH 3)(CH 3)、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、-CH 2CH 2CH 2OCH 3、-CH 2CH(OCH 3)CH 3、-CH 2CH(CH 2OCH 3)CH 3、-CH 2CH 2CH 2CH 2OCH 3、-NH 2、-NHCH 3、-NHCH 2CH 3、-N(CH 3) 2、-N(CH 3)(CH 2CH 3)、-N(CH 2CH 3) 2、-CH 2NH 2、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CH 2NHCH 2CH 3、-CH 2N(CH 3)(CH 2CH 3)、-CH 2N(CH 2CH 3) 2、-CH 2CH 2NH 2、-CH 2CH 2NHCH 3、-CH 2CH 2N(CH 3) 2、-CH(CH 3)N(CH 3) 2、CH 2CH 2NHCH 2CH 3、-CH 2CH 2N(CH 2CH 3) 2、-CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2NHCH 3、-CH 2CH 2CH 2N(CH 3) 2、-CH 2CH(NH 2)CH 3、-CH 2CH(CH 2NH 2)CH 3、-CH 2CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2CH 2NHCH 3和-CH 2CH 2CH 2CH 2N(CH 3) 2
在一种优选的实施方式中,R 4为苯基,被1、2或3个独立地选自卤素、任选被卤素取代的C 1-6烷基和-(CR cR c) 0-6-N(R c) 2的基团取代,示例性取代基包括但不限于F、Cl、Br、I、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3、-C(CH 3)(CH 3)(CH 3)、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CH 2CF 3、-CH(CF 3)CH 3、- CH 2CH(CF 3)CH 3、-CH 2CH 2CH 2CF 3、-C(CF 3)(CH 3)(CH 3)、-NH 2、-NHCH 3、-NHCH 2CH 3、-N(CH 3) 2、-N(CH 3)(CH 2CH 3)、-N(CH 2CH 3) 2、-CH 2NH 2、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CH 2NHCH 2CH 3、-CH 2N(CH 3)(CH 2CH 3)、-CH 2N(CH 2CH 3) 2、-CH 2CH 2NH 2、-CH 2CH 2NHCH 3、-CH 2CH 2N(CH 3) 2、-CH(CH 3)N(CH 3) 2、CH 2CH 2NHCH 2CH 3、-CH 2CH 2N(CH 2CH 3) 2、-CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2NHCH 3、-CH 2CH 2CH 2N(CH 3) 2、-CH 2CH(NH 2)CH 3、-CH 2CH(CH 2NH 2)CH 3、-CH 2CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2CH 2NHCH 3和-CH 2CH 2CH 2CH 2N(CH 3) 2;更优选地,R 4选自
Figure PCTCN2021128977-appb-000025
在一种式(I)化合物的实施方式中,R 4为-C 0-3烷基-苯基、优选苯基,被-(CR cR c) 0-6-N(R c) 2取代,其中R c各自独立地为H或C 1-6烷基,且其中N上的R c之一与其所连接的苯环上的原子及相邻的原子一起形成4-7元含氮杂环。在具体的实施方式中,R 4选自
Figure PCTCN2021128977-appb-000026
Figure PCTCN2021128977-appb-000027
Figure PCTCN2021128977-appb-000028
其中各个R c独立地选自H、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3和-C(CH 3)(CH 3)(CH 3)。
在一种优选的实施方式中,R 4
Figure PCTCN2021128977-appb-000029
在一种式(I)化合物的实施方式中,-X-R 4为-C 0-6烷基-苯基、优选-C 0-3烷基-苯基、更优选苯基,所述苯基具有以上对作为R 4的苯基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-O-C 0-6烷基-苯基、优选-O-C 0-3烷基-苯基、更优选-O-苯基,其中苯基具有以上对作为R 4的苯基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-NH-C 0-6烷基-苯基、优选C 0-3烷基-苯基、更优选-NH-苯基,其中苯基具有以上对作为R 4的苯基给出的实施方式、优选或更优选实施方式 的含义。
在一种式(I)化合物的实施方式中,R 4为-C 0-3烷基-C 3-6环烷基、优选-C 3-6环烷基,所述环烷基任选被1、2或3个独立地选自卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2的基团取代。在具体的实施方式中,示例性取代基包括但不限于:F、Cl、Br、I、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3、-C(CH 3)(CH 3)(CH 3)、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CH 2CF 3、-CH(CF 3)CH 3、-CH 2CH(CF 3)CH 3、-CH 2CH 2CH 2CF 3、-C(CF 3)(CH 3)(CH 3)、-CH 2OH、-CH 2CH 2OH、-CH 2CH 2CH 2OH、-CH 2CH(OH)CH 3、-CH 2CH(CH 2OH)CH 3、-CH 2CH 2CH 2CH 2OH、-C(CH 2OH)(CH 3)(CH 3)、-OH、-OCH 3、-OCH 2CH 3、-OCH 2CH 2CH 3、-OCH(CH 3)(CH 3)、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、-CH 2CH 2CH 2OCH 3、-CH 2CH(OCH 3)CH 3、-CH 2CH(CH 2OCH 3)CH 3、-CH 2CH 2CH 2CH 2OCH 3、-NH 2、-NHCH 3、-NHCH 2CH 3、-N(CH 3) 2、-N(CH 3)(CH 2CH 3)、-N(CH 2CH 3) 2、-CH 2NH 2、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CH 2NHCH 2CH 3、-CH 2N(CH 3)(CH 2CH 3)、-CH 2N(CH 2CH 3) 2、-CH 2CH 2NH 2、-CH 2CH 2NHCH 3、-CH 2CH 2N(CH 3) 2、-CH(CH 3)N(CH 3) 2、CH 2CH 2NHCH 2CH 3、-CH 2CH 2N(CH 2CH 3) 2、-CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2NHCH 3、-CH 2CH 2CH 2N(CH 3) 2、-CH 2CH(NH 2)CH 3、-CH 2CH(CH 2NH 2)CH 3、-CH 2CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2CH 2NHCH 3和-CH 2CH 2CH 2CH 2N(CH 3) 2
在一种优选的实施方式中,R 4选自环丙基、环丁基、环戊基和环己基;更优选环丙基。
在一种式(I)化合物的实施方式中,-X-R 4为-C 0-6烷基-C 3-6环烷基、优选-C 0-3烷基-C 3-6环烷基、更优选C 3-6环烷基,所述环烷基具有以上对作为R 4中的环烷基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-O-C 0-6烷基-C 3-6环烷基、优选-O-C 0-3烷基-C 3-6环烷基、更优选-O-C 3-6环烷基,所述环烷基具有以上对作为R 4中的环烷基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-NH-C 0-6烷基-C 3-6环烷基、优选-NH-C 0-3烷基-C 3-6环烷基、更优选-NH-C 3-6环烷基,所述环烷基具有以上对作为R 4中的环烷基给出的实施方式、优选或更优选实施方式的含义。
在一种式(I)化合物的实施方式中,R 4为-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基,优选包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基,其中所述杂环烷基任选被1、2或3个独立地选自卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2的基团取代。在具体的实施方式中,示例性取代基包括但不限于:F、Cl、Br、I、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3、-C(CH 3)(CH 3)(CH 3)、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CH 2CF 3、- CH(CF 3)CH 3、-CH 2CH(CF 3)CH 3、-CH 2CH 2CH 2CF 3、-C(CF 3)(CH 3)(CH 3)、-CH 2OH、-CH 2CH 2OH、-CH 2CH 2CH 2OH、-CH 2CH(OH)CH 3、-CH 2CH(CH 2OH)CH 3、-CH 2CH 2CH 2CH 2OH、-C(CH 2OH)(CH 3)(CH 3)、-OH、-OCH 3、-OCH 2CH 3、-OCH 2CH 2CH 3、-OCH(CH 3)(CH 3)、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、-CH 2CH 2CH 2OCH 3、-CH 2CH(OCH 3)CH 3、-CH 2CH(CH 2OCH 3)CH 3、-CH 2CH 2CH 2CH 2OCH 3、-NH 2、-NHCH 3、-NHCH 2CH 3、-N(CH 3) 2、-N(CH 3)(CH 2CH 3)、-N(CH 2CH 3) 2、-CH 2NH 2、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CH 2NHCH 2CH 3、-CH 2N(CH 3)(CH 2CH 3)、-CH 2N(CH 2CH 3) 2、-CH 2CH 2NH 2、-CH 2CH 2NHCH 3、-CH 2CH 2N(CH 3) 2、-CH(CH 3)N(CH 3) 2、CH 2CH 2NHCH 2CH 3、-CH 2CH 2N(CH 2CH 3) 2、-CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2NHCH 3、-CH 2CH 2CH 2N(CH 3) 2、-CH 2CH(NH 2)CH 3、-CH 2CH(CH 2NH 2)CH 3、-CH 2CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2CH 2NHCH 3和-CH 2CH 2CH 2CH 2N(CH 3) 2
在一种式(I)化合物的实施方式中,R 4中的4-6元杂环烷基选自氮杂环丁烷基、氧杂环丁烷基、硫杂环丁基、吡咯烷基(例如1-吡咯烷基、2-吡咯烷基及3-吡咯烷基)、四氢呋喃基(例如1-四氢呋喃基、2-四氢呋喃基及3-四氢呋喃基)、四氢噻吩基(例如1-四氢噻吩基、2-四氢噻吩基及3-四氢噻吩基)、哌啶基(例如1-哌啶基、2-哌啶基、3-哌啶基及4-哌啶基)、四氢吡喃基(例如4-四氢吡喃基)、四氢噻喃基(例如4-四氢噻喃基)、吗啉基(例如吗啉代)、硫吗啉基、二噁烷基或哌嗪基。
在一种优选的实施方式中,R 4选自
Figure PCTCN2021128977-appb-000030
Figure PCTCN2021128977-appb-000031
最优选R 4选自
Figure PCTCN2021128977-appb-000032
在一种式(I)化合物的实施方式中,-X-R 4为-C 0-6烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基、优选-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基、更优选包含1、2或3个独立地选自N、O或S的杂原子的4-6元 杂环烷基,所述杂环烷基具有以上对作为R 4的杂环烷基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-O-C 0-6烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基、优选-O-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂环烷基、更优选包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基,所述杂环烷基具有以上对作为R 4中的杂环烷基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-NH-C 0-6烷基-包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基、优选-NH-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂环烷基、更优选包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基,所述杂环烷基具有以上对作为R 4中的杂环烷基给出的实施方式、优选或更优选实施方式的含义。
在一种式(I)化合物的实施方式中,R 4为-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,优选包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,其中所述杂芳基任选被1、2或3个独立地选自卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2的基团取代。在具体的实施方式中,示例性取代基包括但不限于:F、Cl、Br、I、-CH 3、-CH 2CH 3、-CH 2CH 2CH 3、-CH(CH 3)CH 3、-CH 2CH(CH 3)CH 3、-CH 2CH 2CH 2CH 3、-C(CH 3)(CH 3)(CH 3)、-CH 2F、-CHF 2、-CF 3、-CH 2CF 3、-CH 2CH 2CF 3、-CH(CF 3)CH 3、-CH 2CH(CF 3)CH 3、-CH 2CH 2CH 2CF 3、-C(CF 3)(CH 3)(CH 3)、-CH 2OH、-CH 2CH 2OH、-CH 2CH 2CH 2OH、-CH 2CH(OH)CH 3、-CH 2CH(CH 2OH)CH 3、-CH 2CH 2CH 2CH 2OH、-C(CH 2OH)(CH 3)(CH 3)、-OH、-OCH 3、-OCH 2CH 3、-OCH 2CH 2CH 3、-OCH(CH 3)(CH 3)、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、-CH 2CH 2CH 2OCH 3、-CH 2CH(OCH 3)CH 3、-CH 2CH(CH 2OCH 3)CH 3、-CH 2CH 2CH 2CH 2OCH 3、-NH 2、-NHCH 3、-NHCH 2CH 3、-N(CH 3) 2、-N(CH 3)(CH 2CH 3)、-N(CH 2CH 3) 2、-CH 2NH 2、-CH 2NHCH 3、-CH 2N(CH 3) 2、-CH 2NHCH 2CH 3、-CH 2N(CH 3)(CH 2CH 3)、-CH 2N(CH 2CH 3) 2、-CH 2CH 2NH 2、-CH 2CH 2NHCH 3、-CH 2CH 2N(CH 3) 2、-CH(CH 3)N(CH 3) 2、CH 2CH 2NHCH 2CH 3、-CH 2CH 2N(CH 2CH 3) 2、-CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2NHCH 3、-CH 2CH 2CH 2N(CH 3) 2、-CH 2CH(NH 2)CH 3、-CH 2CH(CH 2NH 2)CH 3、-CH 2CH 2CH 2CH 2NH 2、-CH 2CH 2CH 2CH 2NHCH 3和-CH 2CH 2CH 2CH 2N(CH 3) 2
在一种式(I)化合物的实施方式中,R 4中的5-6元杂芳基选自吡咯基、呋喃基、噻吩基、咪唑基、呋咱基、噁唑基、噁二唑基、噁三唑基、异噁唑基、噻唑基、异噻唑基、吡唑基、三唑基及四唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基及三嗪基。
在一种优选的实施方式中,R 4选自
Figure PCTCN2021128977-appb-000033
Figure PCTCN2021128977-appb-000034
最优选地,R 4选自
Figure PCTCN2021128977-appb-000035
在一种式(I)化合物的实施方式中,-X-R 4为-C 0-6烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基、优选-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基、更优选包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,所述杂芳基具有以上对作为R 4中的杂芳基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-O-C 0-6烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基、优选-O-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基、更优选包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,所述杂芳基具有以上对作为R 4中的杂芳基给出的实施方式、优选或更优选实施方式的含义。在一种式(I)化合物的实施方式中,-X-R 4为-NH-C 0-6烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基、优选-NH-C 0-3烷基-包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基、更优选包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,所述杂芳基具有以上对作为R 4的杂芳基给出的实施方式、优选或更优选实施方式的含义。
在一种式(I)化合物的实施方式中,作为R 4取代基的-(CR cR c) 0-6-SR c、-(CR cR c) 0-6-(CO) 0-1-OR c、-(CR cR c) 0-6-(CO) 0-1-N(R c) 2各自优选为-(CR cR c) 0-6-SR c、-(CR cR c) 0-6-OR c、-(CR cR c) 0-6-N(R c) 2;更优选-(CR cR c) 0-3-SR c、-(CR cR c) 0-3-OR c、-(CR cR c) 0-3-N(R c) 2
在一种式(I)化合物的实施方式中,E为卤素,优选为F。
在一种式(I)化合物的实施方式中,E是-O-R d,R d为任选被卤素取代的C 1-6烷基,优选C 1- 6烷基,例如但不限于甲基、乙基、丙基、异丙基、三氟甲基、二氟甲基、三氟乙基等。
在一种式(I)化合物的实施方式中,R 5
Figure PCTCN2021128977-appb-000036
其中Z、G、Y、B、D至多两个不为C。示例性的R 5包括但不限于
Figure PCTCN2021128977-appb-000037
Figure PCTCN2021128977-appb-000038
在一种优选的实施方式中,R 5
Figure PCTCN2021128977-appb-000039
其中Z、Y、B、D至多两个不为C。示例性的R 5包括但不限于
Figure PCTCN2021128977-appb-000040
Figure PCTCN2021128977-appb-000041
在更优选的实施方式中,R 5
Figure PCTCN2021128977-appb-000042
Figure PCTCN2021128977-appb-000043
最优选R 5
Figure PCTCN2021128977-appb-000044
在一种优选的实施方式中,R 5
Figure PCTCN2021128977-appb-000045
更优选
Figure PCTCN2021128977-appb-000046
在一种式(I)化合物的实施方式中,R 6、R 7和R 8中至少一个是卤素,优选F。在更优选的实施方式中,R 6为F,且R 7和R 8为H。
在一种式(I)化合物的实施方式中,R 6、R 7和R 8中至少一个是卤素取代的C 1-6烷基,优选-CF 3
在一种式(I)化合物的实施方式中,R c为H或C 1-6烷基。
本发明还提供式(II)化合物、其异构体、药学上可接受的盐或溶剂合物。
Figure PCTCN2021128977-appb-000047
其中R 1、R 2、R 3、R 4、R 6、R 7、R 8、A、E、R b各自具有上文对式(I)化合物给出的含义。
在优选的式(II)化合物实施方式中,
A选自C-R a或N,其中R a选自卤素;
R 1、R 2和R 3各自独立地选自H、卤素或C 1-6烷基;
R b在每次出现时独立地选自H、卤素、CN或任选被卤素或CN取代的C 1-6烷基;
X选自键、-O-或-NH-;
R 4选自H、卤素、C 1-6烷基、-C 0-3烷基-C 6-10芳基、-C 0-3烷基-C 3-8环烷基、-C 0-3烷基-C 3-8环烯基、-C 0-3烷基-包含1、2或3个独立的选自N、O或S的杂原子的3-8元杂环烷基、-C 0- 3烷基-包含1、2或3个独立的选自N、O或S的杂原子的3-8元杂环烯基、-C 0-3烷基-包含1、2或3个独立的选自N、O或S的杂原子的5-10元杂芳基,其中所述烷基、芳基、环烷基、环烯基、杂环烷基、杂环烯基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c、-(CR cR c) 0-6-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环;
R c在每次出现时独立地选自H或任选被卤素取代的C 1-6烷基;
E选自卤素、-O-R d或-N(R d) 2-,其中R d各自独立地为H或任选被卤素取代的C 1-6烷基;
R 6、R 7和R 8各自独立地选自H、卤素和任选被卤素取代的C 1-6烷基;
或其异构体、药学上可接受的盐或溶剂合物。
在一种优选的式(II)化合物的实施方式中,A为C-R a,其中R a选自F或Cl,优选Cl。
在一种优选的式(II)化合物的实施方式中,R b为H。
在一种优选的式(II)化合物的实施方式中,R b不为H,且存在1个R b或存在2个R b
在一种优选的式(II)化合物的实施方式中,-X-R 4为H。
在一种优选的式(II)化合物的实施方式中,-X-R 4不为H。
在一种优选的式(II)化合物的实施方式中,R 6、R 7和R 8中至少一个是卤素,优选F;更优选R 6为F,且R 7和R 8为H。
在一种优选的式(II)化合物的实施方式中,E是卤素,优选F。
在一种优选的式(II)化合物的实施方式中,E是-O-R d,R d为任选被卤素取代的C 1-6烷基,优选C 1-6烷基,例如但不限于甲基、乙基、丙基、异丙基三氟甲基、二氟甲基、三氟乙基等。
在一种优选的式(II)化合物的实施方式中,R 4选自H、卤素、C 1-6烷基、苯基、C 3-6环烷基、包含1、2或3个独立的选自N、O或S的杂原子的4-6元杂环烷基和包含1、2或3个独立的选自N、O或S的杂原子的5-6元杂芳基,其中所述烷基、芳基、环烷基、杂环烷基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环 状基团上的原子及相邻的原子一起形成4-7元含氮杂环。
在一种优选的式(II)化合物的实施方式中,R 4选自卤素,优选F或Cl。
在一种优选的式(II)化合物的实施方式中,R 4选自C 1-6烷基,例如甲基、乙基、丙基、异丙基等,任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2
在一种优选的式(II)化合物的实施方式中,R 4选自苯基,任选被卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2取代。
在一种优选的式(II)化合物的实施方式中,R 4选自C 3-6环烷基,任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2
在一种优选的式(II)化合物的实施方式中,R 4选自包含1、2或3个独立的选自N、O或S的杂原子的4-6元杂环烷基,任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2
在一种优选的式(II)化合物的实施方式中,R 4选自包含1、2或3个独立的选自N、O或S的杂原子的5-6元杂芳基,任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2
在上述各个实施方式中,其中连接于芳基、环烷基、杂环烷基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环。
在一种优选的式(II)化合物的实施方式中,R c在每次出现时独立地选自H或C 1-6烷基。
本发明还提供这样的式(II)化合物的实施方式,其中R 1、R 2、R 3、R 4、R 6、R 7、R 8、A、E、R b各自具有上文对式(I)化合物给出的实施方式、优选、更优选或最优选实施方式中的含义。
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式或各个具体实施方式,还涵盖上述各个实施方式或具体实施方式的任何组合或亚组合构成的实施方式,也涵盖以上任何优选或例举的任何组合所构成的实施方式。
本发明的化合物的具体实施方式包括以下具体化合物或其异构体、药学上可接受的盐或溶剂合物,
Figure PCTCN2021128977-appb-000048
Figure PCTCN2021128977-appb-000049
Figure PCTCN2021128977-appb-000050
本文以上所定义的化合物及其各种具体实施方式是Ras突变、尤其是KRas突变的抑制剂,所述突变包括密码子G12、G13和Q61上的突变,例如G12C突变、G12D突变、G13D突变。本发明的化合物、尤其是本文上下文具体示例的化合物在细胞测定法中显示对Ras突变、尤其是KRas G12C突变细胞具有增殖抑制活性,如下文活性实施例部分所示。故本发明化合物可用于治疗或预防由Ras突变、优选KRas突变、最优选KRas G12C突变介导的疾病,例如可通过抑制Ras突变、优选KRas突变、最优选KRas G12C突变来治疗的疾病或病症,或Ras突变、优选KRas突变、最优选KRas G12C突变活性在其中扮演角色或牵涉的疾病或病症,尤其是通过抑制Ras突变、优选KRas突变、最优选KRas G12C突变来治疗或预防肿瘤或癌症。
除显示KRas G12C突变抑制活性外,一些本发明的化合物还显示对KRas G12D突变的抑制活性,另一些本发明化合物显示对KRas G13D突变的抑制活性。
除显示Ras突变、优选KRas突变抑制活性外,本文所定义的化合物及其各种具体实施方式、尤其是实施例化合物,因具有改进的结构模式,相比现有技术已有的KRas突变蛋白抑制剂,保留了相当的或增强的、甚至显著增强的KRas突变蛋白以及相关癌细胞增殖抑制活性;具有不同的生物活性谱而可用于新的适应症;具有改进的代谢稳定性,从而带来更好的药动学性质;并有改善的物理化学性质,从而具有良好的成药性,比如更容易在体内吸收等。
基于以上,本发明还提供以下各个方面的技术方案。
一方面,本发明提供了用作药物的本发明化合物、其异构体或它们药学上可接受的盐或溶剂合物。
另一方面,本发明提供用于治疗和/或预防由Ras突变、优选KRas突变介导的疾病的本发明化合物、其异构体或它们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,本发明提供药物组合物,其包含以上定义的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物,以及可药用载体、稀释剂或赋形剂。本发明的药物组合物可用于治疗或预防由Ras突变、尤其KRas突变介导的疾病,例如KRas G12C、KRas G12D或KRas G13D突变介导的疾病,例如肿瘤或癌症。
上述本发明药物组合物,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。
本发明药物组合物的给药和施用均符合良好的医学实践。在此背景下需要考虑的因素包括所治疗特定障碍、所治疗的特定哺乳动物、个体患者的临床情况、障碍的起因、药剂递送位置、施用方法、施用安排以及医生从业者熟知的其它因素。本发明药物组合物的最佳剂量水平和给药频率将通过药学领域所要求的临床试验确定。通常,例如,口服施用的日剂量范围在约0.001mg至约100mg每kg患者体重范围间,常为0.01mg至约50mg每kg体重,例如0.1至10mg每kg体重,优选约0.01至约35mg每kg体重,以单剂量或分剂量服用。对于70kg的人类受试者,适合的剂量范围为约0.07至约7000mg/天,优选约0.7至约2500mg/天。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。
本发明的组合物可采取任意合适方式施用,包括口服、局部(包括颊和舌下)、直肠、阴道、透皮、胃肠外、皮下、腹膜内、肺内、皮内、鞘内、吸入和硬膜外和鼻内,和如需局部治疗,也可采取病灶内施用。胃肠外输注包括肌肉、静脉内、动脉内、腹膜内或皮下施用。在一些实施方案中,采用口服施用。
本发明的组合物可以以任意便捷的施用形式给药,例如片剂、粉末、胶囊、锭剂、颗粒、溶液、分散剂、混悬剂、糖浆、喷雾、栓剂、凝胶、乳剂、贴剂等。所述组合物可含有药物制剂中的常规组分,例如稀释剂(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、其它已知添加剂以及其它活性剂。合适的载体和赋形剂为本领域技术人员熟知并详述于例如Ansel,Howard C.,等,Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems.Philadelphia:Lippincott,Williams&Wilkins,2004中。
治疗方法和用途
如上所述,本发明的式(I)或式(II)化合物及其各种具体实施方式的化合物、尤其是实施例中具体制备和表征的化合物,显示出对Ras突变、尤其是KRas突变、例如KRas G12C、KRas G12D或KRas G13D突变的抑制作用。
因此,另一方面,本发明提供了一种抑制细胞中Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变的方法,包括使细胞与本发 明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物相接触以抑制细胞中Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变的活性。
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式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物、或包含本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物的药物组合物。
另一方面,本发明提供了用于治疗和/或预防由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变介导的疾病的方法,包括向有需要的对象施用治疗有效量的本发明式(I)或式(II)的化合物、其异构体或它们药学上可接受的盐或溶剂合物、或包含本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物的药物组合物。
另一方面,本发明提供了本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物、或包含本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物的药物组合物的用途,用于抑制细胞中Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变,或用于抑制哺乳动物中异常细胞生长,或用于治疗和/或预防由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D、最优选KRas G12C突变介导的疾病。
另一方面,本发明提供了本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物、或包含本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物的药物组合物在制备用于治疗和/或预防由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D突变、最优选KRas G12C突变介导的疾病的药物中的用途。
对上述本发明提供的各个方法和用途技术方案而言,所述异常细胞生长或由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D、最优选KRas G12C突变介导的疾病尤其指的是癌症或肿瘤。示例性的所述癌症或肿瘤包括但不限于肺癌、肺腺癌、骨癌、胰腺癌、皮肤癌、头颈癌、皮肤或眼内黑素瘤、子宫癌、卵巢癌、直肠癌、肛门区域癌、胃癌、结肠癌、乳腺癌、输卵管癌、子宫内膜癌、子宫颈癌、阴道癌、外阴癌、霍奇金病、食道癌、小肠癌、内分泌系统癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、尿道癌、阴茎癌、前列腺癌、慢性或急性白血病、淋巴细胞性淋巴瘤、膀胱癌、肾脏或输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊柱肿瘤、脑干神经胶质瘤或垂体腺瘤。
对上述本发明提供的各个方法和用途技术方案而言,所述异常细胞生长或由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D介导的疾病优选选自肺腺癌、肺癌、结肠癌、直肠癌、胰腺癌、胆管癌、子宫内膜癌、卵巢癌、白血病;最优选选自肺腺 癌、结肠癌、直肠癌、胰腺癌、胆管癌。
因此,在该方面的优选实施方案中,本发明提供了用于通过抑制KRas-G12C突变而治疗或预防癌症或肿瘤的上述各项方法和用途技术方案。在更进一步优选的实施方案中,本发明提供了通过抑制KRas-G12C突变而治疗或预防肺腺癌、结肠癌、直肠癌、胰腺癌和胆管癌的上述各项方法和用途技术方案。
药物组合
本发明的化合物可以作为唯一的活性成分进行施用,也可以与另外的药物或疗法组合进行施用。
因此,另一方面,本发明提供了药物组合,其包含本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物以及其他活性剂,或由二者组成。该药物组合用于抑制哺乳动物中异常细胞生长,或用于治疗和/或预防由Ras突变、优选KRas突变介导的疾病。
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,例如这些活性剂可以是已知调节其他生物活性通路的化合物,或者可以是调节本发明化合物所涉及生物活性通路中的不同组分的化合物,或甚至是与本发明化合物的生物靶点相重叠的化合物。
在一个具体的实施方案中,可以与本发明化合物组合使用的其他活性剂包括但不限于化疗剂、治疗性抗体和放疗,例如烷化剂、抗代谢物、细胞周期抑制剂、有丝分裂抑制剂、拓扑异构酶抑制剂、抗激素类药物、血管生成抑制剂、细胞毒性剂。
与本发明组合使用的其他活性剂可以与本发明的化合物通过相同或不同的施用途径同时、分别或依次地进行施用。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,例如组合产品,优选为药盒形式,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。它们可以由相同或不同的制造商制备和/或配制。而且,本发明的化合物和其他活性剂可以(i)在将组合产品发送给医师之前(例如在包含本发明的化合物和另外的药物的药盒的情形中);(ii)在临施用前由医师自身(或在医师指导下);(iii)由患者自身、例如在本发明的化合物和其他活性剂的依次施用期间一起加入组合治疗中。
本发明的化合物还可以与抗肿瘤疗法组合,所述抗肿瘤疗法包括但不限于手术、辐射治疗、移植(例如干细胞移植、骨髓移植)、肿瘤免疫疗法和化疗等。
因此,另一方面,本发明还提供了药盒,其包含两种或多种单独的药物组合物,其中至少一种包含本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物,以及分别容纳所述组合物的装置,如容器、分装瓶或分立的箔包装,例如用于包装片剂、胶囊等的泡罩包装。本发明的药盒特别适用于施用不同的剂型,如口服剂型和胃肠外剂型,或者 适合于以不同的剂量间隔施用不同的组合物。
对于上述本发明的药物组合物、药物组合或药盒的技术方案而言,其中所涉及的异常细胞生长或由Ras突变、尤其KRas突变、优选KRas G12C、KRas G12D或KRas G13D、最优选KRas G12C突变介导的疾病如上文对于本发明方法和用途所定义。
对于上述本发明化合物、药物组合物、方法、用途、药物组合及药盒而言,优选上文所述式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物,更优选式(II)具体实施方案中所定义的化合物以及上文所列的具体化合物,即化合物1-25。
当本文描述药物或其药学上可接受的盐的剂量时,应理解,该剂量基于游离碱的重量,不包括其任何水合物或溶剂化物,除非说明书中指出该剂量基于盐、水合物或溶剂化物的重量。
附图说明
附图1显示实施例9化合物在人非小细胞肺癌NCI-H358异种移植小鼠模型中的抗肿瘤活性以及对体重的影响。
附图2显示实施例9化合物在人胰腺癌Mia PaCa-2细胞皮下异种移植肿瘤BALB/c裸小鼠模型中的抗肿瘤活性及对体重的影响。
本发明化合物的制备方法
另一方面,本发明还提供了本发明所定义化合物的制备方法。
本发明的式(I)或式(II)化合物、其异构体或它们药学上可接受的盐或溶剂合物可以通过多种方法、包括下文给出的方法、实施例中给出的方法或与之类似的方法制备。下文举例说明了合成本发明化合物的通用合成方案。
对于各通用合成方案的各个反应步骤而言,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。用于合成本发明化合物的方法步骤可以在本身已知的反应条件(包括具体提及的那些条件)下、在不存在或通常在存在溶剂或稀释剂(包括例如对所用试剂而言是惰性的且可溶解所用试剂的溶剂或稀释剂)的情况下、在不存在或存在催化剂、缩合剂或中和剂(例如离子交换剂,如阳离子交换剂,例如H +形式)的情况下、根据反应和/或反应物的性质在降低的、正常的或升高的温度(例如约-100℃至约190℃,包括例如约-78℃至约150℃,例如约0℃至约125℃、室温、-20至40℃或回流温度)下、在大气压力下或在密闭容器中、当适宜时在加压下、和/或在惰性气氛例如氩气或氮气气氛下进行。
根据所用化合物的反应性的不同,上述反应通常将在室温至所用溶剂的沸腾温度之间的温度进行。
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。
除非在方法的描述中另有说明,否则适用于任何特定反应的那些溶剂的溶剂包括:具体提及的那些溶剂,或者例如水;酯类,如低级链烷酸低级烷基酯,例如乙酸乙酯;醚类,如脂肪族醚,例如乙醚,或环状醚,例如四氢呋喃或二氧六环;液体芳族烃类,如苯或甲苯;醇类,如甲醇、乙醇或1-或2-丙醇;腈类,如乙腈;卤化烃类,如二氯甲烷或氯仿;酰胺类,如二甲基甲酰胺、N-甲基吡咯烷-2-酮或二甲基乙酰胺;碱类,如杂环氮碱类,例如吡啶或三乙胺;羧酸酐类,如低级链烷酸酸酐,例如乙酸酐;环状、直链或支链烃类,如环己烷、己烷或异戊烷;或这些溶剂的混合物,例如水溶液。该类溶剂混合物也可用于后处理,例如通过色谱法或分配进行的后处理。
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。如果中间体和终产物以固体形式获得,则纯化也可以通过重结晶或陈化来进行。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。
反应混合物以常规方式后处理,例如通过与水混合,分离各相,并在适当时通过色谱法纯化粗产物来进行。
本领域技术人员能认识到式(I)或式(II)化合物中是否存在立体中心。在反应的所有阶段,所形成的异构体的混合物可被分离成单个异构体,例如非对映异构体或对映异构体,或者分离成任何所需的异构体混合物,例如外消旋物或非对映异构体的混合物,参见例如E.L.Eliel,S.H.Wilen和L.N.Mander的“Stereochemistry of Organic Compounds”(Wiley-Interscience,1994)。
在某些特定情况下,可能有必要使用适当的保护基团保护特定的反应基团以避免与其他反应性基团的副反应,所述基团可能存在于式(I)或式(II)化合物中并且可能竞争或干扰反应。仅作为实例,如果式(I)或式(II)化合物中的一个或多个基团是或包含基团C(O)OH、NH 2或OH且该基团具有比期望反应位置相似或甚至更强反应性,则在期望的反应发生之前保护这些基团是有利的。在这些情况下,可能有必要进行额外的脱保护步骤以在在期望的反应完成之后除去这些保护基。适合的保护基和采用这样的适合保护基保护和脱保护不同取代基的方法是本领域技术人员众所周知的;其实例可以见于T.Greene和P.Wuts,Protective Groups in Organic Synthesis(第3版),John Wiley&Sons,NY(1999)中。
本发明还涉及如下制备方法:其中将可在下文所述各制备方法和流程中的任何步骤以中间体形式获得的化合物,用作起始材料并且进行剩余的方法步骤,或者其中起始材料在反应条件下原位形成或以衍生物的形式例如以被保护的形式或盐形式使用,或者可按照本发明的方法获得的化合物在所述方法条件下生成并且被进一步原位处理。
合成方案I:
本发明的化合物可以根据以下示例性方案制备或类似地制备,其中如果没有另外说明,则变量如上文所定义。
Figure PCTCN2021128977-appb-000051
本发明通过合成方案I制备式(I)或式(II)化合物。在步骤A中通过芳香族化合物的氯代反应得到化合物2。之后通过步骤B的甲酯化反应得到化合物3,再经过步骤C,氨酸缩合得到化合物4。化合物4在碱性条件下环化得到化合物5,再通过氯代反应得到中间体Int A。关键中间体Int A与带有单一保护基团的哌嗪或者哌嗪衍生物在碱性条件下通过亲核取代反应得到中间体Int B。后者通过芳香亲核取代反应(当X=O)或者金属催化偶联反应(当X=直接相连)得到化合物6。在步骤H中,化合物6通过催化偶联反应引入带有保护基的R 5基团得到化合物7。化合物7脱保护得到化合物8,后者与酰氯或者羧酸经酰化反应得到所示通式I化合物。
该合成方案1中所涉及的氯代、酯化、缩合、环化、亲核取代、催化偶联以及酰化反应的典型反应条件及所用试剂均是本领域熟知的,属于本领域技术人员的常规经验范围,或者可以有本领域技术人员基于本领域此类反应的典型条件、基于所用原料和目标产物的特征而做出适当改变而确定。
合成方案II:
其中-X-R 4为H的本发明化合物可按照下列示例性通用方案合成。
Figure PCTCN2021128977-appb-000052
在合成方案II中,中间体Int B通过催化还原脱氯得到中间体Int C,后者经过与方案I中相同的反应步骤H、I、J得到其中-X-R 4为H的本发明化合物。
合成方案III:
其中A为N的本发明化合物可按照下列示例性通用方案合成。
Figure PCTCN2021128977-appb-000053
在该合成方案中,化合物11发生碘代反应得到化合物12,后者通过金属催化的插羰反应得到化合物13。从化合物13出发,经过与方案I中相同的步骤C~J得到其中A为N的本发明化合物。
合成方案IV:
其中A为N且-X-R 4为H的本发明化合物可按照下列示例性通用方案合成。
Figure PCTCN2021128977-appb-000054
在该合成方案中,从中间体Int E出发,与方案II中方法相同,合成其中A为N且-X-R 4为H的本发明化合物。
以上合成方案II、III和IV中,
Figure PCTCN2021128977-appb-000055
代表哌嗪环,其中所涉及的各个反应的典型反应条件及所用试剂均是本领域熟知的,属于本领域技术人员的常规经验范围,或者可以由本领域技术人员基于本领域此类反应的典型条件、基于所用原料和目标产物的特征而做出适当改变而确定。
合成实施例
下面结合实施例对本发明作进一步的说明。需要说明的是,下述实施例是示例性的,不应视为对本发明保护范围的限制。
本文在对实施方案和随后的具体实施例的描述中,使用了以下缩写:
Boc(叔丁氧基羰基);n-BuLi(正丁基锂);t-BuOK(叔丁醇钾);CDCl 3(氘代氯仿);DCM(二氯甲烷);DIEA(N,N-二异丙基乙胺);DME(乙二醇二甲醚);DMF(N,N-二甲基甲酰胺);DMSO(二甲亚砜);DMSO-d 6(六氘代二甲亚砜);EA(乙酸乙酯);EDTA-K2(乙二胺四乙酸二钾盐);EtOH(乙醇);FCC(快速柱层析);g(克);h(小时);HATU(2-(7-氮杂苯并三氮唑)-N,N,N’,N’-四甲基脲六氟磷酸酯);HCl(氯化氢);HLM(人肝微粒体);H 2O(水);H 2SO 4(硫酸);IV(静脉给药);K 2CO 3(碳酸钾);KOH(氢氧化钾);LCMS(液质联机);LC-MS/MS(液谱-质谱-质谱联机);MeCN(乙腈);MeOH(甲醇);Methanol-d 4(四氘代甲醇);mg(毫克);MHz(兆赫兹);min(分钟);mL(毫升);mmol(毫摩尔);m/z(质荷比);N 2(氮气);NaCl(氯化钠);NaH(氢化钠);NaHCO 3(碳酸氢钠);NaOH(氢氧化钠);Na 2SO 3(亚硫酸钠);Na 2SO 4(硫酸钠);NCCH 2CO 2H(2-氰基乙酸);NCS(氯代丁二酰亚胺);NH 4Cl(氯化铵);NIS(碘代丁二酰亚胺);NMI(N-甲基咪唑);NMP(N-甲基吡咯烷酮);NMR(核磁共振);Pd/C(钯碳);Pd 2(dba) 3(三(二亚苄基丙酮)二钯); Pd(dppf)Cl 2(1,1'-双二苯基膦二茂铁二氯化钯);Pd(PPh 3) 4(四三苯基膦钯);PE(石油醚);PEG(聚乙二醇);PO(口服给药);POCl 3(三氯氧磷);r.t.(室温);SiO 2(硅胶);TCFH(N,N,N',N'-四甲基氯甲脒六氟磷酸盐);TEA(三乙胺);TFA(三氟乙酸);THF(四氢呋喃);TLC(薄层色谱);TsOH(对甲苯磺酸);TsOH·H 2O(对甲苯磺酸一水合物);μL(微升);μM(微摩尔浓度);μmol(微摩尔);v/v(液体体积比例);w/w(质量比例);Xantphos(4,5-双二苯基膦-9,9-二甲基氧杂蒽)。
在如下实施例中,给出了所合成目标化合物的名称及其结构。名称与结构之间出现任何偏差并非有意,在这种情况下,结构为决定性的。
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。除非另外说明,否则百分比和份数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。
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。
在下列实施例中, 1H-NMR谱是用Bruker(400MHz)记录,化学位移以相对于氘代溶剂峰(CDCl 3:δ=7.26ppm;CD 3OD:δ=3.31ppm;DMSO-d 6:δ=2.50ppm)的δ(ppm)表示;质谱是用Aglient 1100液相色谱+Aglient G6100质谱LCMS液质联用仪记录。
中间体a的合成
Figure PCTCN2021128977-appb-000056
(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸
Figure PCTCN2021128977-appb-000057
步骤A:N-((2-溴-5-氟苯基)氨基甲磺酰)苯甲酰胺
在置有磁子的圆底烧瓶中,将苯甲酰异硫氰酸酯(28g,210mmol)溶于THF(300mL)中。冰浴冷却后,在搅拌下,将2-溴-5-氟苯胺(40g,210mmol)的THF(100mL)溶液逐滴加入体系中, 滴加完成后继续搅拌反应1h。LCMS检测反应完成,减压浓缩后得到粗产品直接投下一步。LCMS(m/z):353.1(M+H)。
步骤B:1-(2-溴-5-氟苯基)硫脲
室温搅拌下,将NaOH(16.8g,420mmol)的水(150mL)溶液加入到N-((2-溴-5-氟苯基)氨基甲磺酰)苯甲酰胺(步骤A所得粗品)的THF(300mL)溶液中,加热至80℃反应16h。减压浓缩后,用EA(200mL)稀释,水(150mL)洗涤,分离水相和有机相,水相再用EA(100mL×2)萃取两次。合并有机相,用饱和氯化钠水溶液洗涤,用无水硫酸钠干燥。过滤,滤液减压浓缩后得到粗品,加入200mL石油醚打浆,过滤,得到白色固体1-(2-溴-5-氟苯基)硫脲(40g,收率77%)。LCMS(m/z):249.0,251.0(M+H)。
步骤C:4-溴-7-氟苯并[d]噻唑-2-胺氢溴酸盐
搅拌下,将液溴(23g,141mmol)的氯仿(70mL)溶液缓慢滴加至冰浴冷却的1-(2-溴-5-氟苯基)硫脲(35g,141mmol)的氯仿(300mL)溶液中。滴加完成后,移除冰浴,将体系加热至70℃反应48h。反应完成,减压浓缩,加入EA(100mL)打浆,过滤,得到白色固体4-溴-7-氟苯并[d]噻唑-2-胺氢溴酸盐(30.5g,收率66%)。LCMS(m/z):247.0,249.0(M+H)。
步骤D:(4-溴-7-氟苯并[d]噻唑-2-基)氨基甲酸叔丁基酯
室温条件下,在4-溴-7-氟苯并[d]噻唑-2-胺氢溴酸盐(30g,93mmol)和4-二甲氨基吡啶(14g,112mmol)的DCM(300mL)溶液中缓慢加入二碳酸二叔丁酯(61g,280mmol)的DCM(60mL)溶液。在室温下反应3h,LCMS检测反应完成。反应液经减压浓缩后,用EA(200mL)稀释,水(150mL)洗涤,分离水相和有机相,水相再用EA(200mL×2)萃取两次。合并有机相,用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩,加入石油醚和乙酸乙酯(20:1)打浆,过滤,得到白色固体(4-溴-7-氟苯并[d]噻唑-2-基)氨基甲酸叔丁基酯(18g,收率56%)。 1H NMR(400MHz,Chloroform-d)δ9.29(s,1H),7.54(dd,J=8.6,4.8Hz,1H),6.94–6.81(m,1H),1.52(s,9H).LCMS(m/z):291.0,293.0(M+H)。
步骤E:(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸
氮气保护搅拌下,将NaH(60%w/w,3g,75.6mmol)加入冰浴冷却的(4-溴-7-氟苯并[d]噻唑-2-基)氨基甲酸叔丁基酯(17.5g,50.4mmol)的THF(200mL)溶液中,在此温度下继续搅拌30min后,转移至-68℃干冰/乙醇浴中。之后缓慢加入正丁基锂(30.2mL,75.6mmol,2.5M/THF),搅拌10min后,加入硼酸三甲酯(156g,151mmol),继续搅拌反应30min。用饱和氯化铵水溶液(50mL)淬灭,用EA(200mL)稀释,水(150mL)洗涤,分离水相和有机相,水相再用EA(100mL×2)萃取两次。合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩后得到粗品。将所得粗品加入石油醚打浆,过滤,得到白色固体(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸(10.7g,收率68%)。 1H NMR(400MHz,DMSO-d 6)δ12.25(s,1H),8.50(s,2H),7.84(dd,J=8.0,6.4Hz,1H),7.22–7.13(m,1H),1.54(s,9H).LCMS(m/z):313.2(M+H)。
中间体Int A的合成
Figure PCTCN2021128977-appb-000058
7-溴-2,4,6-三氯-8-氟喹啉-3-碳腈
Figure PCTCN2021128977-appb-000059
步骤A:2-氨基-4-溴-5-氯-3-氟苯甲酸
在室温下将NCS(13.7g,102.6mmol)分批加入到溶解有2-氨基-4-溴-3-氟苯甲酸(20g,85.5mmol)的DMF(140mL)中,之后将反应液加热至75℃,并在此温度下搅拌20h。反应液冷却至室温后倒入到700mL冰水混合物中,析出沉淀。通过过滤收集固体,并分别用水(500mL)和石油醚(150mL)洗涤。所得产品真空干燥后得到浅黄色固体2-氨基-4-溴-5-氯-3-氟苯甲酸(20g,收率87%)。LCMS(m/z):268.0(M+H)。
步骤B:2-氨基-4-溴-5-氯-3-氟苯甲酸甲酯
将化合物2-氨基-4-溴-5-氯-3-氟苯甲酸(20g,74.5mmol)分散到甲醇(150mL)中。冰水浴冷却,搅拌下,将氯化亚砜(50mL)滴加到反应液中。滴加完毕后将反应混合物置于65℃反应20h。TLC监测反应完毕后,将溶剂浓缩至干燥后得到目标产物2-氨基-4-溴-5-氯-3-氟苯甲酸甲酯(15.8g,收率75%)。LCMS(m/z):282.0(M+H)。
步骤C:4-溴-5-氯-2-(2-氰基乙酰氨基)-3-氟苯甲酸甲酯
室温下,将化合物2-氨基-4-溴-5-氯-3-氟苯甲酸甲酯(15.8g,55.9mmol)、TCFH(18.8g,67.1mmol)、氰基乙酸(5.2g,61.3mmol)溶于乙腈(100mL)中。搅拌下加入N-甲基咪唑(6.7mL,83.9mmol)。反应混合物加热至60℃下搅拌20h。TLC监测反应完毕,将反应液冷至室温后倒入水(500mL)中,析出白色沉淀。通过过滤将固体收集,并分别用水(500mL)和乙酸乙酯/石油醚(1:4,150mL)洗涤,真空干燥后得到白色固体4-溴-5-氯-2-(2-氰基乙酰氨基)-3-氟苯甲酸甲酯(15g,收率76%)。LCMS(m/z):349.0(M+H)。
步骤D:7-溴-6-氯-8-氟-2,4-二羟基喹啉-3-碳腈
将化合物4-溴-5-氯-2-(2-氰基乙酰氨基)-3-氟苯甲酸甲酯(6g,17.2mmol)分散于THF(120mL)中。冰浴搅拌下分批加入叔丁醇钾(3.7g,32.6mmol)。添加完成后,反应混合物在冰浴下搅拌1h。LCMS监测反应完毕,减压蒸馏除去溶剂后,在体系中加入水(200mL),用1N盐酸调节混合液pH=7,过滤并收集固体。所得固体分别用水(100mL)和石油醚(150mL)洗涤,真空干燥后得到白色固体7-溴-6-氯-8-氟-2,4-二羟基喹啉-3-碳腈(5.4g,收率99%)。LCMS(m/z):317.0(M+H)。
步骤E:7-溴-2,4,6-三氯-8-氟喹啉-3-碳腈
在室温搅拌下,将化合物7-溴-6-氯-8-氟-2,4-二羟基喹啉-3-碳腈(5.4g,17mmol)分散到三氯氧磷(100mL)中。加热体系至120℃搅拌反应24h。冷却至室温后,真空浓缩除去大部分三氯氧磷,再加入乙腈(20mL)。将产物的乙腈溶液缓缓倒入至水(250mL)中,析出黄色固体。过滤并收集固体,分别用水(200mL)和石油醚(100mL)洗涤,真空干燥后得到黄色固体7-溴-2,4,6-三氯-8-氟喹啉-3-碳腈(5.4g,收率90%)。LCMS(m/z):353.0(M+H)。
中间体Int B1的合成
Figure PCTCN2021128977-appb-000060
(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯
Figure PCTCN2021128977-appb-000061
在室温搅拌下将DIEA(7.3mL,44mmol)和(R)-2-甲基哌嗪-1-羧酸叔丁基酯(4.4g,22mmol)依次加入到7-溴-2,4,6-三氯-8-氟喹啉-3-碳腈(中间体Int A,7.8g,22mmol)的THF(100mL)溶液中,所得混合物在室温下继续搅拌1h。反应完成后,用旋转蒸发仪浓缩样品,所得粗品用PE/THF混合溶剂(PE/THF=10:1,500mL)洗涤,得到黄色固体(R)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(9.0g,收率79%)。LCMS(m/z):519.2(M+H)。
中间体Int B2的合成
Figure PCTCN2021128977-appb-000062
4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
中间体Int B2的合成参照中间体Int B1中所述进行,用哌嗪-1-羧酸叔丁酯代替(R)-2-甲基哌嗪-1-羧酸叔丁基酯。LCMS(m/z):503.2(M+H)。
中间体Int C1的合成
Figure PCTCN2021128977-appb-000063
(R)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯
Figure PCTCN2021128977-appb-000064
在置有磁子的圆底烧瓶中加入(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体Int B1,206mg,0.4mmol),Pd(PPh 3) 4(23mg,20μmol),甲酸(55mg,1.2mmol),三乙胺(80.5mg,0.8mmol),氮气抽换三次后,加入DMF(4mL),反应体系加热到50℃,搅拌6h。反应完成后,用旋转蒸发仪浓缩样品,所得粗品用FCC(SiO 2,EA/PE=0-50%)纯化,得到(R)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(140mg,收率73%)。LCMS(m/z):485.3(M+H)。
中间体Int C2的合成
Figure PCTCN2021128977-appb-000065
4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
中间体Int C2的合成参照中间体Int C1中所述进行,用4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体B2)代替(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体B1)。LCMS(m/z):459.2,471.2(M+H)。
中间体Int D的合成
Figure PCTCN2021128977-appb-000066
2,4,7-三氯-8-氟-1,6-萘啶-3-碳腈
Figure PCTCN2021128977-appb-000067
步骤A:2-氯-3-氟-5-碘吡啶-4-胺
室温下圆底烧瓶中将化合物2-氯-3-氟吡啶-4-胺(3g,20.47mmol)和NIS(6.91g,30.71mmol)溶于30mL乙腈中,搅拌下,加入TsOH·H 2O(400mg,2mmol)。将反应液加热到70℃反应16h。TLC监测反应完毕后,将冷却至室温的反应液倒入100mL饱和食盐水中,用乙酸乙酯萃取(25mL×3),有机相合并后用饱和Na 2SO 3水溶液和饱和食盐水洗涤,无水Na 2SO 4干燥,将溶剂浓缩后得到淡黄色固体2-氯-3-氟-5-碘吡啶-4-胺(5.4g,收率97%)。LCMS(m/z):273.0(M+H)。
步骤B:4-氨基-6-氯-5-氟烟酸乙酯
将化合物2-氯-3-氟-5-碘吡啶-4-胺(5.4g,19.82mmol),Pd(dppf)Cl 2(695mg,0.991mmol)和三乙胺(555mg,49.6mmol)分散于无水乙醇中,在一氧化碳气氛下加热至90℃反应40h。反应完毕后,将反应液用硅藻土过滤,滤液浓缩后经FCC(SiO 2,EA/DCM=0-100%)纯化,得到黄色固体4-氨基-6-氯-5-氟烟酸乙酯(3.5g,收率81%)。LCMS(m/z):219.0(M+H)。
步骤C:6-氯-4-(2-氰基乙酰氨基)-5-氟烟酸乙酯
将氰基乙酸(778mg,9.15mmol)和三氟乙酸酐(1.44g,6.86mmol)溶于乙腈(5mL)中,在55℃下搅拌2h后,将反应液以及4-氨基-6-氯-5-氟烟酸乙酯(1.0g,4.57mmol)置于20mL的微波管中,在105℃下微波辐射2h。反应完毕后,将反应液倒入25mL水中,加入乙酸乙酯(15mL),有机相分别用水、饱和碳酸氢钠水溶液以及饱和食盐水洗涤,无水硫酸钠干燥,溶剂浓缩后 经FCC(SiO 2,THF/DCM=0-40%)纯化,得到淡黄色固体6-氯-4-(2-氰基乙酰氨基)-5-氟烟酸乙酯(550mg,收率42%)。LCMS(m/z):286.0(M+H)。
步骤D:7-氯-8-氟-2,4-二羟基-1,6-萘啶-3-碳腈
Figure PCTCN2021128977-appb-000068
将6-氯-4-(2-氰基乙酰氨基)-5-氟烟酸乙酯(300mg,1.05mmol)溶于THF(10mL)中,并在冰浴下搅拌2min,再将叔丁醇钾(236mg,2.10mmol)缓慢加入反应液中。在冰浴自然恢复室温的条件下,搅拌1h。LCMS检测反应结束后,把反应液滴加到搅拌的饱和NH 4Cl(50mL)溶液中,再用1N盐酸调节混合液pH=7,在这期间,白色絮状沉淀产生,过滤,收集固体,所得固体分别用水(10mL)和石油醚(10mL)洗涤,真空干燥后得到白色固体7-氯-8-氟-2,4-二羟基-1,6-萘啶-3-碳腈(200mg,收率79%)。LCMS(m/z):240.1(M+H)。
步骤E:2,4,7-三氯-8-氟-1,6-萘啶-3-碳腈
Figure PCTCN2021128977-appb-000069
将7-氯-8-氟-2,4-二羟基-1,6-萘啶-3-碳腈(200mg,0.83mmol)溶于POCl 3(15mL)中,加热反应液到120℃并搅拌过夜。TLC监测反应完成,冷却至室温,真空浓缩除去大部分三氯氧磷,再加入乙腈(1mL)。将产物的乙腈溶液倒入饱和NaHCO 3水溶液(30mL)中,有黄色固体生产,过滤,收集固体,所得固体分别用水(10mL)和石油醚(10mL)洗涤,真空干燥后得到黄色固体2,4,7-三氯-8-氟-1,6-萘啶-3-碳腈(190mg,收率82%)。
中间体Int E1的合成
Figure PCTCN2021128977-appb-000070
4-(2,7-二氯-3-氰基-8-氟-1,6-萘啶-4-基)哌嗪-1-羧酸叔丁酯
中间体Int E1的合成参照中间体Int B1中所述进行,用2,4,7-三氯-8-氟-1,6-萘啶-3-碳腈(中间体Int D)代替7-溴-2,4,6-三氯-8-氟喹啉-3-碳腈(中间体Int A),并用哌嗪-1-羧酸叔丁酯代替(R)-2-甲基哌嗪-1-羧酸叔丁基酯。LCMS(m/z):426.3(M+H)。
中间体Int F的合成
Figure PCTCN2021128977-appb-000071
(S)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯
中间体Int F的合成参照中间体Int B1中所述进行,用(S)-2(氰基甲基)哌嗪-1-羧酸苄基酯酸盐代替(R)-2-甲基哌嗪-1-羧酸叔丁基酯。LCMS(m/z):576.3,578.3(M+H)。
实施例1
Figure PCTCN2021128977-appb-000072
4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-3-碳腈
Figure PCTCN2021128977-appb-000073
步骤A:(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯
在室温搅拌下,将NaH(60%w/w,9.3mg,0.23mmol)加入到(3R,4R)-4-甲氧基-1-甲基吡咯烷-3-醇(27.8mg,0.21mmol)的THF(2mL)溶液中,所得混合物在室温继续搅拌10min。一次性加入(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁酯(中间体B1,110mg,0.21mmol),并在室温搅拌1h,LCMS监测中间体B1有剩余。再次加入预先搅拌5min的NaH(60%w/w,5mg,0.12mmol),(3R,4R)-4-甲氧基-1-甲基吡咯烷-3-醇(10mg,0.78mmol)的THF(1mL)溶液。所得混合物继续搅拌30min,LCMS监测反应完成,用饱和NH 4Cl水溶液 (2mL)淬灭反应,加入EA(30mL)稀释,所得有机相用饱和NaCl洗涤(20mL×3),无水Na 2SO 4干燥,过滤,浓缩得到粗品(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁酯(130mg,粗品),直接投入下一步。LCMS(m/z):612.4(M+H)。
步骤B:(2R)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯
室温下,在置有磁子的微波反应管中加入(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(170mg,0.28mmol),(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸(112mg,0.36mmol),Pd(dppf)Cl 2(24mg,0.03mmol)和K 3PO 4(104mg,0.49mmol)的1,4-二氧六环/H 2O(v/v=3:1,1.6mL)溶液。体系鼓氮气10min后,将反应管密封,微波加热至100℃反应1h。冷却至室温,用EA(20mL)稀释,饱和NaCl水溶液(20mL×3)洗涤,无水Na 2SO 4干燥,减压浓缩后用FCC(SiO 2,MeOH/DCM=0-20%)初步纯化得到(2R)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(150mg,62%纯度),直接用于下一步反应。LCMS(m/z):800.6(M+H)。
步骤C:7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-4-((R)-3-甲基哌嗪-1-基)喹啉-3-碳腈
室温搅拌下,将TFA(1mL)滴加到含有(2R)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(150mg,62%纯度)的DCM(2mL)溶液中,所得混合物在室温继续搅拌30min,LCMS监测反应完成。减压浓缩除去溶剂TFA,加入少量DCM再次减压浓缩,重复三次除去残余TFA,所得粗品无需纯化直接投入到下一步。
步骤D:4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-3-碳腈
将步骤C所得7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-4-((R)-3-甲基哌嗪-1-基)喹啉-3-碳腈粗品溶于DCM(2mL),用冰水浴冷却,加入DIEA(103uL,0.62mmol),搅拌5min后缓慢滴加丙烯酰氯(10uL,0.12mmol)的DCM(0.1mL)溶液。滴加完成后在冰水浴下继续搅拌20min,LCMS监测反应完成,体系中加入H 2O(5mL)及EA(30mL),分液除去水相,有机相用饱和NaCl水溶液(20mL×3)洗涤,无水Na 2SO 4干燥,浓缩后用制备高效液相色谱纯化粗品,得到白色固体4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-3-碳腈(15.2mg,四步总收率7.5%)。 1H NMR(400MHz,Methanol-d 4)δ8.12–7.99(m,1H),7.33–7.22(m,1H),7.07–6.97(m,1H),6.87(dd,J=16.7,10.7Hz,1H),6.30(d,J=16.8Hz,1H),5.83(d,J=10.7Hz,1H),5.65(s,1H),4.24–4.20(m,1H),4.09–3.89(m,2H),3.83–3.77(m,1H),3.62–3.48(m,5H),3.43–3.38(m,2H),3.26–3.18(m,2H),3.09–3.03(m,1H),2.98–2.92(m, 1H),2.60(s,3H),1.46(d,J=6.8Hz,3H). 19F NMR(376MHz,Methanol-d 4)δ-114.94,-120.48.LCMS(m/z):654.5(M+H)。
实施例2
Figure PCTCN2021128977-appb-000074
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-3-碳腈
实施例2的合成参照实施例1中所述进行,在步骤A中使用4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体B2)代替(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体B1)。 1H NMR(400MHz,DMSO-d 6)δ8.02–7.87(m,3H),7.24(dt,J=8.4,5.1Hz,1H),7.07(t,J=8.8Hz,1H),6.90(dd,J=16.7,10.5Hz,1H),6.19(dd,J=16.7,2.4Hz,1H),5.75(dd,J=10.4,2.4Hz,1H),5.39–5.31(m,1H),4.06–3.96(m,1H),3.94–3.81(m,4H),3.75–3.61(m,4H),3.32(s,3H),3.08–3.00(m,1H),2.92(dd,J=11.0,6.2Hz,1H),2.73–2.64(m,1H),2.33(dt,J=9.3,4.3Hz,1H),2.26–2.21(m,3H). 19F NMR(376MHz,DMSO-d 6)δ-112.27(d,J=4.4Hz),-119.59(d,J=22.4Hz).LCMS(m/z):640.5(M+H)。
实施例3
Figure PCTCN2021128977-appb-000075
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(3-((二甲氨基)甲基)苯基)-8-氟喹啉-3-碳腈
Figure PCTCN2021128977-appb-000076
步骤A:4-(7-溴-6-氯-3-氰基-2-(3-((二甲氨基)甲基)苯基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
在微波管中加入化合物4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体B2,300mg,0.60mmol),(3-((二甲氨基)甲基)苯基)硼酸(108mg,0.60mmol),Pd(PPh 3) 4(35mg,0.03mmol),K 2CO 3(166mg,1.2mmol),乙腈(3mL)和水(1mL)。用氮气置换3次后,微波加热到40℃,反应1h。待冷却至室温,将反应液浓缩,粗品经FCC(SiO 2,EA/PE=0-60%)纯化得到黄色固体4-(7-溴-6-氯-3-氰基-2-(3-((二甲氨基)甲基)苯基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(260mg,收率72%)。LCMS(m/z):602.4(M+H)。
步骤B至步骤D:4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(3-((二甲氨基)甲基)苯基)-8-氟喹啉-3-碳腈
实施例3的后续合成参照实施例1合成所述进行,在步骤B使用4-(7-溴-6-氯-3-氰基-2-(3-((二甲氨基)甲基)苯基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯代替(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯。 1H NMR(400MHz,DMSO-d 6)δ8.11-8.10(m,1H),7.92(s,2H),7.81-7.70(m,2H),7.53-7.46(m,2H),7.31-7.27(m,1H),7.11-7.09(m,1H),6.91(dd,J=16.6,10.4Hz,1H),6.22-6.17(m,1H),5.77-5.74(m,1H),3.98-9.84(m,4H),3.83-3.70(m,4H),3.51-3.46(m,2H),2.24-2.13(m,6H).LCMS(m/z):645.5(M+H)。
实施例4
Figure PCTCN2021128977-appb-000077
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(1-(2-(二甲氨基)乙基)-1H-吡唑-5-基)-8-氟喹啉-3-碳腈
实施例4的合成参照实施例3中所述进行,在步骤A中使用(1-(2-(二甲氨基)乙基)-1H-吡唑-5-基)硼酸代替(3-((二甲氨基)甲基)苯基)硼酸。 1H NMR(400MHz,CDCl 3)δ7.96(s,1H),7.63(s,1H),7.40–7.30(m,1H),7.01–6.93(m,2H),6.68-6.59(m,1H),6.39(d,J=16.8Hz,1H),6.26(s,2H),5.80(d,J=10.6Hz,1H),4.66(t,J=7.5Hz,2H),4.04–3.71(m,8H),3.39(s,1H),2.98(s,1H),2.38(s,6H). 19F NMR(376MHz,CDCl 3)δ-111.07,-115.38.LCMS(m/z):648.5(M+H)。
实施例5
Figure PCTCN2021128977-appb-000078
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(3-(1-(二甲氨基)乙基)苯基)-8-氟喹啉-3-碳腈
实施例5的合成参照实施例3中所述进行,在步骤A中使用(3-(1-(二甲氨基)乙基)苯基)硼酸代替(3-((二甲氨基)甲基)苯基)硼酸。 1H NMR(400MHz,MeOH-d 4)δ8.16(d,J=1.6Hz,1H),7.87(s,1H),7.84–7.78(m,1H),7.61–7.52(m,2H),7.30(dd,J=8.5,5.4Hz,1H),7.09–6.99(m,1H),6.89(dd,J=16.8,10.6Hz,1H),6.31(dd,J=16.7,1.9Hz,1H),5.84(dd,J=10.6,1.9Hz,1H),4.12–4.00(m,4H),3.97–3.84(m,4H),2.30(s,6H),1.57–1.45(m,3H). 19F NMR(376MHz,MeOH-d 4)δ-114.82,-118.65(d,J=9.3Hz).LCMS(m/z):658.5(M+H)。
实施例6
Figure PCTCN2021128977-appb-000079
4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(3-((二甲氨基)甲基)苯基)-8-氟喹啉-3-碳腈
实施例6的合成参照实施例3中所述进行,在步骤A中使用(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体Int B1)代替4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体Int B2)。 1H NMR(400MHz,DMSO-d 6)δ8.14(d,J=3.3Hz,1H),7.92(d,J=19.9Hz,2H),7.82(s,1H),7.77(d,J=7.6Hz,1H),7.56–7.46(m,2H),7.33–7.27(m,1H),7.13–7.06(m,1H),6.94–6.84(m,1H),6.23–6.14(m,1H),5.78–5.72(m,1H),4.95–4.23(m,4H),4.08(t,J=12.7Hz,1H),4.00–3.90(m,1H),3.77(t,J=12.7Hz,1H),3.53(s,2H),2.21(s,6H),1.36–1.27(m,3H).LCMS(m/z):658.5(M+H)。
实施例7
Figure PCTCN2021128977-appb-000080
4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(1-(2-(二甲氨基)乙基)-1H-吡唑-5-基)-8-氟喹啉-3-碳腈
实施例6的合成参照实施例3中所述进行,在步骤A中使用(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体Int B1)代替4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体Int B2),并用(1-(2-(二甲氨基)乙基)-1H-吡唑-5-基)硼酸代替(3-((二甲氨基)甲基)苯基)硼酸。 1H NMR(400MHz,DMSO-d 6)δ8.14(s,1H),7.90(d,J=11.8Hz,2H),7.62(d,J=1.9Hz,1H),7.29(dd,J=8.6,5.3Hz,1H),7.09(t,J=8.8Hz,1H),6.94–6.83(m,2H),6.17(d,J=16.7Hz,1H),5.79–5.70(m,1H),4.62–4.32(m,4H),4.11–3.92(m,3H),3.81–3.66(m,2H),3.55(s,2H),2.10–1.83(m,6H),1.34–1.20(m,3H).LCMS(m/z):662.5,(M+H)。
实施例8
Figure PCTCN2021128977-appb-000081
4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
Figure PCTCN2021128977-appb-000082
步骤A:(2R)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯
在微波管中加入(R)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体C1,120mg,0.25mmol),(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸(232mg,0.75mmol),Pd(dppf)Cl 2(18.3mg,25μmol),K 3PO 4(211mg,1.0mmol),氮气抽换三次后,加入1,4-二氧六环/水(v/v=3:1,4ml),放入微波反应器,加热至90℃,反应1h。反应完成后,体系冷却至室温,用旋转蒸发仪旋干反应溶剂,粗品用FCC(SiO 2,EA/PE=0-80%)纯化后得到黄色固体(2R)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基 哌嗪-1-羧酸叔丁基酯(92mg,收率47%)。LCMS(m/z):671.5(M+H)。
步骤B:7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-4-((R)-3-甲基哌嗪-1-基)喹啉-3-碳腈
室温搅拌下,将TFA(2mL)滴加入(2R)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(92mg,0.14mmol)的DCM(1ml)溶液中。继续搅拌一小时,LCMS监测反应完成。用旋转蒸发仪旋干反应溶剂,加入少量DCM再次浓缩,重复三次除去TFA残余,得7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-4-((R)-3-甲基哌嗪-1-基)喹啉-3-碳腈粗品,直接用于下一步。LCMS(m/z):471.3(M+H)。
步骤C:4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
在置有磁子的圆底烧瓶中加入7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-4-((R)-3-甲基哌嗪-1-基)喹啉-3-碳腈(步骤B所得粗品),加入DCM(3ml)。冰水浴中冷却至0℃,加入DIPEA(2ml),随后逐滴加入丙烯酰氯(11mg,0.12mmol)的DCM(1.0mL)溶液。滴加完成后在冰水浴中继续搅拌30min。反应完成后,加入水(5mL)淬灭反应,用DCM萃取(10mL×3)。合并有机相,用饱和NaCl(10mL×3)洗涤,无水Na 2SO 4干燥。旋转蒸发仪浓缩后用制备高效液相色谱纯化得到白色固体4-((R)-4-丙烯酰-3-甲基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈(26mg,收率35%)。 1H NMR(400MHz,DMSO-d 6)δ8.88(s,1H),8.08(dd,J=4.3,1.4Hz,1H),7.92(s,2H),7.38–7.24(m,1H),7.12–7.04(m,1H),6.86(dd,J=16.6,10.5Hz,1H),6.17(dd,J=16.6,2.4Hz,1H),5.81–5.68(m,1H),4.47(d,J=125.4Hz,3H),3.94(t,J=12.8Hz,1H),3.87–3.64(m,2H),3.56–3.41(m,1H),1.49–1.19(m,3H).LCMS(m/z):525.3(M+H)。
实施例9
Figure PCTCN2021128977-appb-000083
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
实施例9的合成参照实施例8中所述进行,在步骤A中使用4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体Int C2)代替(R)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体Int C1)。 1H NMR(400MHz,Methanol-d 4)δ8.78(s,1H),8.10(d,J=1.8Hz,1H),7.26(dd,J=8.4,5.4Hz,1H),7.06–6.97(m,1H),6.87(dd,J=16.8,10.7Hz,1H),6.30(dd,J=16.8,1.9Hz,1H),5.83(dd,J=10.6,1.9Hz,1H),4.05–3.95(m,4H),3.90–3.78(m,4H). 19F NMR(376MHz,Methanol-d 4)δ-114.73,-119.40.LCMS(m/z):511.3(M+H)。
实施例10
Figure PCTCN2021128977-appb-000084
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(2-甲基-1,2,3,4-四氢异喹啉-5-基)喹啉-3-碳腈
Figure PCTCN2021128977-appb-000085
步骤A:4-(7-溴-6-氯-3-氰基-8-氟-2-(2-甲基-1,2,3,4-四氢异喹啉-5-基)喹啉-4-基)哌嗪-1-羧酸叔丁酯
在置有磁子的微波反应管中加入4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体B2,150mg,0.3mmol)、(2-甲基-1,2,3,4-四氢异喹啉-5-基)硼酸(243mg,0.9mmol)、K 3PO 4(189mg,0.9mmol)的CH 3CN/H 2O(v/v=3:1,2mL),通入氮气排气5min。加入Pd(dppf)Cl 2(22mg,0.03mmol),继续鼓氮气5分钟。微波加热到45℃反应4小时,冷却至室温。加入EA(10mL)和H 2O(5mL)分液,水相用EA(10mL×2)萃取。合并有机相,用饱和NaCl(30mL)洗涤,无水Na 2SO 4干燥,过滤,滤液减压浓缩后得到粗品,经FCC(SiO 2,EA/PE=0-100%)纯化得棕色固体4-(7-溴-6-氯-3-氰基-8-氟-2-(2-甲基-1,2,3,4-四氢异喹啉-5-基)喹啉-4-基)哌嗪-1-羧酸叔丁酯(80mg,收率44%)。LCMS(m/z):614.4(M+H)。
步骤B至步骤D:4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(2-甲基-1,2,3,4-四氢异喹啉-5-基)喹啉-3-碳腈
实施例10的后续合成参照实施例1合成所述进行,在步骤B使用4-(7-溴-6-氯-3-氰基-8-氟-2-(2-甲基-1,2,3,4-四氢异喹啉-5-基)喹啉-4-基)哌嗪-1-羧酸叔丁酯代替(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯。 1H NMR(400MHz,Methanol-d 4)δ8.15(s,1H),7.36–7.22(m,4H),7.01(t,J=8.8Hz,1H), 6.86(dd,J=16.8,10.6Hz,1H),6.29(dd,J=16.7,1.9Hz,1H),5.82(dd,J=10.5,1.9Hz,1H),4.57(s,2H),3.99(s,4H),3.85(s,4H),3.74(s,2H),2.84(d,J=5.7Hz,1H),2.73(t,J=5.9Hz,1H),2.45(s,3H).LCMS(m/z):654.5(M+H)。
实施例11
Figure PCTCN2021128977-appb-000086
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-1,6-萘啶-3-碳腈
实施例11的合成参照实施例1中所述进行,在步骤A中使用4-(2,7-二氯-3-氰基-8-氟-1,6-萘啶-4-基)哌嗪-1-羧酸叔丁酯(中间体E1)代替(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体B1)。 1H NMR(400MHz,DMSO-d 6)δ9.14(s,1H),7.95(s,2H),7.44(dd,J=8.5,5.8Hz,1H),7.13–7.04(m,1H),6.91(dd,J=16.6,10.4Hz,1H),6.19(dd,J=16.7,2.4Hz,1H),5.75(dd,J=10.4,2.4Hz,1H),5.47–5.39(m,1H),4.08–3.99(m,1H),3.89(s,2H),3.83(s,6H),3.34(s,3H),3.08(dd,J=10.0,6.4Hz,1H),2.98(dd,J=11.1,6.2Hz,1H),2.75(dd,J=11.3,2.8Hz,1H),2.40(dd,J=10.0,4.9Hz,1H),2.29(s,3H). 19F NMR(376MHz,DMSO-d 6)δ-111.71,-135.46.LCMS(m/z):607.5(M+H)。
实施例12
Figure PCTCN2021128977-appb-000087
4-((R)-4-丙烯醇-3-甲基苯丙胺-1-yl)-2'-氨基-6-氯-5',8-二氟-[7,8'-双喹啉]-3-碳氰
实施例12的合成参照实施例8中所述进行,在步骤A中使用(5-氟-2-((4-甲氧基苄基)氨基)喹啉-8-基)硼酸代替(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸。LCMS(m/z):519.1(M+H)。
实施例13
Figure PCTCN2021128977-appb-000088
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-甲氧基喹啉-3-碳腈
实施例13的合成参照实施例1中所述进行,在步骤A中使用4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体Int B2)代替(R)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯(中间体Int B1),并用甲醇钠代替(3R,4R)-4-甲氧基-1-甲基吡咯烷-3-醇的NaH混合物。 1H NMR(400MHz,DMSO-d 6)δ7.94(d,J=5.9Hz,3H),7.25(dd,J=8.4,5.6Hz,1H),7.08(t,J=8.8Hz,1H),6.91(dd,J=16.7,10.5Hz,1H),6.20(dd,J=16.7,2.2Hz,1H),5.76(dd,J=10.4,2.2Hz,1H),4.05(s,3H),3.87(d,J=15.7Hz,4H),3.68(s,4H). 19F NMR(376MHz,DMSO)δ-112.27,-119.27.LCMS(m/z):541.3(M+H)。
实施例14
Figure PCTCN2021128977-appb-000089
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(1-甲基-1H-吡唑-5-基)喹啉-3-碳腈
实施例14的合成参照实施例3中所述进行,在步骤A中使用(1-甲基-1H-吡唑-5-基)硼酸代替(3-((二甲氨基)甲基)苯基)硼酸。 1H NMR(400MHz,DMSO-d 6)δ8.12(s,1H),7.94(s,2H),7.61(s,1H),7.29(dd,J=8.3,5.7Hz,1H),7.10(t,J=8.8Hz,1H),7.00–6.86(m,2H),6.20(d,J=16.7Hz,1H),5.77(d,J=8.7Hz,1H),4.04(s,3H),3.90(d,J=14.7Hz,4H),3.79(s,4H). 19F NMR(376MHz,DMSO)δ-112.07,-118.03.LCMS(m/z):591.4(M+H)。
实施例15
Figure PCTCN2021128977-appb-000090
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(1,3-二甲基-1H-吡唑-4-基)-8-氟喹啉-3-碳腈
实施例15的合成参照实施例3中所述进行,在步骤A中使用(1,3-二甲基-1H-吡唑-4-基)硼酸代替(3-((二甲氨基)甲基)苯基)硼酸。 1H NMR(400MHz,DMSO-d 6)δ8.30(s,1H),8.05(d,J=1.5Hz,1H),7.94(s,2H),7.28(dd,J=8.5,5.6Hz,1H),7.09(t,J=8.8Hz,1H),6.93(dd,J=16.7,10.4Hz,1H),6.20(dd,J=16.6,2.4Hz,1H),5.77(dd,J=10.4,2.4Hz,1H),3.89(d,J=11.9Hz,7H),3.73(d,J=5.5Hz,4H),2.41(s,3H). 19F NMR(376MHz,DMSO)δ-112.23,-118.42.LCMS(m/z):605.4(M+H)。
实施例16
Figure PCTCN2021128977-appb-000091
7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-4-(4-(2-氟丙烯酰)哌嗪-1-基)喹啉-3-碳腈
Figure PCTCN2021128977-appb-000092
将化合物7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-4-(哌嗪-1-基)喹啉-3-碳腈(88mg,0.13mmol),化合物2-氟丙烯酸(13.3mg,0.15mmol)和HATU(77mg,0.20mmol)溶于DCM(5mL),室温搅拌下,将DIPEA(52mg,0.40mmol)滴加进反应液中。LCMS监测反应完毕后,将20mL水加入反应体系,分离并收集有机相,浓缩干燥后,通过制备高效液相色谱纯化得到白色固体7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-4-(4-(2-氟丙烯酰)哌嗪-1-基)喹啉-3-碳腈(9mg,收率13%)。 1H NMR(400MHz,DMSO-d 6)δ8.89(s,1H),8.06(s,1H),7.94(s,2H),7.29(dd,J=8.4,5.6Hz,1H),7.09(t,J=8.8Hz,1H),5.38–5.33(m,1H),5.32(dd,J=64,4.1Hz,1H),3.91-3.84(m 4H),3.79-3.74(m,4H). 19F NMR(376MHz,DMSO-d 6)δ-105.25,-112.15,-117.60.LCMS:529.3(M+H)。
实施例17
Figure PCTCN2021128977-appb-000093
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-甲基喹啉-3-碳腈
Figure PCTCN2021128977-appb-000094
步骤A:4-(7-溴-6-氯-3-氰基-8-氟-2-甲基喹啉-4-基)哌嗪-1-羧酸叔丁酯
向反应瓶中加入4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体B2,500mg,0.99mmol)、甲基硼酸(71mg,1.19mmol)、K 2CO 3(411mg,2.98mmol),DME(3mL),通入氮气排气3min。加入Pd(dppf)Cl 2(72mg,0.099mmol),继续鼓氮气3分钟后密闭瓶盖,90℃反应1小时,冷却至室温。加入EA(10mL)和H 2O(5mL)分液,水相用EA(20mL×2)萃取。合并有机相,用饱和NaCl(30mL)洗涤,无水Na 2SO 4干燥,过滤,滤液减压浓缩后得到粗品,经FCC(SiO 2,EA/PE=0-100%)纯化,得黄色固体4-(7-溴-6-氯-3-氰基-8-氟-2-甲基喹啉-4-基)哌嗪-1-羧酸叔丁酯(120mg,收率25%)。LCMS(m/z):483.3(M+H)。
步骤B至步骤D:4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-甲基喹啉-3-碳腈
实施例17的后续合成参照实施例1合成所述进行,在步骤B使用4-(7-溴-6-氯-3-氰基-8-氟-2-甲基喹啉-4-基)哌嗪-1-羧酸叔丁酯代替(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯。 1H NMR(400MHz,DMSO-d 6)δ8.04–8.01(m,1H),7.94(s,2H),7.27(dd,J=8.4,5.6Hz,1H),7.12–7.05(m,1H),6.92(dd,J=16.7,10.5Hz,1H),6.20(dd,J=16.6,2.4Hz,1H),5.76(dd,J=10.5,2.4Hz,1H),3.93–3.82(m,4H),3.69(s,4H),2.74(s,3H). 19F NMR(376MHz,DMSO-d 6)δ-112.20,-118.13.LCMS(m/z):525.4(M+H)。
实施例18
Figure PCTCN2021128977-appb-000095
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-环丙基-8-氟喹啉-3-碳腈
实施例18的合成参照实施例3中所述进行,在步骤A中使用环丙基硼酸代替(3-((二甲氨基)甲基)苯基)硼酸。1H NMR(400MHz,DMSO-d6)δ8.01(d,J=1.5Hz,1H),7.96(s,2H),7.27(dd,J=8.5,5.6Hz,1H),7.13–7.05(m,1H),6.94(dd,J=16.7,10.4Hz,1H),6.22(dd,J=16.6,2.4Hz,1H),5.78(dd,J=10.4,2.4Hz,1H),4.00–3.81(m,4H),3.79–3.65(m,4H),2.57–2.54(m,1H),1.25–1.12(m,4H).LCMS:551.5(M+H)。
实施例19
Figure PCTCN2021128977-appb-000096
4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-羟基喹啉-3-碳腈
Figure PCTCN2021128977-appb-000097
步骤A:4-(7-溴-2,6-二氯-3-氰基-8-氟-2-羟基喹啉-4-基)哌嗪-1-羧酸叔丁酯
在室温搅拌下,将KOH(44mg,0.78mmol)加入到4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(200mg,0.39mmol)的THF和H 2O(v/v=2:1,4mL)混合溶液中,所得混合物在80℃继续搅拌3h。LCMS监测反应完成,将反应液减压浓缩除去THF后,倒入饱和NH 4Cl水溶液(4mL)中,缓慢滴加1N稀盐酸调至pH=5-6,产物析出,过滤,所得滤饼分别用水和石油醚洗涤两次,真空干燥得4-(7-溴-2,6-二氯-3-氰基-8-氟-2-羟基喹啉-4-基)哌嗪-1-羧酸叔丁酯(190mg,粗品),直接投入下一步。LCMS(m/z):429.2,431.1(M-56)。
步骤B至步骤D:4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-羟基喹啉-3-碳腈
实施例19的后续合成参照实施例1合成所述进行,在步骤B使用4-(7-溴-2,6-二氯-3-氰基-8-氟-2-羟基喹啉-4-基)哌嗪-1-羧酸叔丁酯代替(R)-4-(7-溴-6-氯-3-氰基-8-氟-2-(((3R,4R)-4- 甲氧基-1-甲基吡咯烷-3-基)氧基)喹啉-4-基)-2-甲基哌嗪-1-羧酸叔丁基酯。 1H NMR(400MHz,DMSO-d 6)δ12.11(s,1H),7.98(s,2H),7.74(s,1H),7.22(dd,J=8.5,5.6Hz,1H),7.11–7.04(m1H),6.90(dd,J=16.6,10.4Hz,1H),6.19(dd,J=16.7,2.3Hz,1H),5.76(dd,J=10.4,2.3Hz,1H),3.85(d,J=19.0Hz,4H),3.65(s,4H). 19F NMR(376MHz,DMSO)δ-112.03,-123.05.LCMS(m/z):527.3(M+H)。
实施例20
Figure PCTCN2021128977-appb-000098
4-(4-丙烯酰哌嗪-1-基)-2-氨基-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
Figure PCTCN2021128977-appb-000099
步骤A:4-(7-溴-6-氯-3-氰基-2-((二苯亚甲基)氨基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
在微波管中加入4-(4-丙烯酰哌嗪-1-基)-2-氨基-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈(350mg,694μmol),二苯基甲烷亚胺(126mg,694μmol),Pd 2(dba) 3(64mg,69.4μmol),XantPhos(80mg,139μmol),Cs 2CO 3(679mg,2.08mmol)和dioxane(5mL),氮气置换1分钟后,加盖,微博加热至80℃,反应1h。反应完成后,体系冷却至室温,把反应液倒入水(50mL),用EA(50mL×3)萃取,收集萃取液,浓缩,进一步FCC(SiO 2,EA/PE=0-50%)纯化得到黄色固体4-(7-溴-6-氯-3-氰基-2-((二苯亚甲基)氨基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(280mg,收率62%)。LCMS(m/z):650.4(M+H)。
步骤B:4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-2-((二苯亚甲基)氨基)- 8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
室温下,在置有磁子的反应管中加入4-(7-溴-6-氯-3-氰基-2-((二苯亚甲基)氨基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(280mg,431μmol),(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸(135mg,431μmol),Pd(dppf)Cl 2(32mg,43μmol),Na 2CO 3(137mg,1.29mmol)和1,4-二氧六环/H 2O(v/v=4:1,8mL)溶液。体系鼓氮气1min后,将反应管密封,微波加热至115℃反应2h。冷却至室温,用水(60mL)稀释,EA(50mL×3)萃取,无水Na 2SO 4干燥,减压浓缩后得到4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-2-((二苯亚甲基)氨基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(310mg,收率86%).LCMS(m/z):836.6(M+H)。
步骤C:7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-((二苯基亚甲基)氨基)-8-氟-4-(哌嗪-1-基)喹啉-3-碳腈
室温条件下,把TFA:CH 2Cl 2=1:1(15mL)加入4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-2-((二苯亚甲基)氨基)-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(310mg,371μmol)中,搅拌半个小时,LCMS检测反应结束后,用EA(10mL)稀释,并倒入饱和NaHCO 3(20mL),用EA(30mL×3)萃取,无水Na 2SO 4干燥,收集萃取液,得到7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-((二苯基亚甲基)氨基)-8-氟-4-(哌嗪-1-基)喹啉-3-碳腈(140mg,收率59%)。LCMS(m/z):636.6(M+H)。
步骤D:4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-((二苯亚甲基)氨基)-8-氟喹啉-3-碳腈
在冰浴条件下,用注射器把丙烯酰氯(24mg,264μmol)在CH 2Cl 2(0.5mL)的稀释液滴加到7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-((二苯基亚甲基)氨基)-8-氟-4-(哌嗪-1-基)喹啉-3-碳腈(140mg,220μmol),DIPEA(85mg,660μmol)和CH 2Cl 2(5mL)的混合溶液中,搅拌10分钟。LCMS检测反应结束后,把反应液倒入水中(50mL),用CH 2Cl 2(30mL×3)萃取,收集萃取液,浓缩,得到黄色固体4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-((二苯亚甲基)氨基)-8-氟喹啉-3-碳腈(150mg,粗品)。LCMS(m/z):690.0(M+H)。
步骤E:4-(4-丙烯酰哌嗪-1-基)-2-氨基-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
室温下,把4-(4-丙烯酰哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-((二苯亚甲基)氨基)-8-氟喹啉-3-碳腈(150mg,217μmol)溶于30mL乙醇中,并加入1.5mL饱和柠檬酸水溶液,升温至60℃,搅拌过夜。LCMS检测到产物生成,浓缩反应液,然后在体系中加入水(30mL),用EA(50mL×3),浓缩得到的粗产品进一步通过制备高效液相色谱分离,得到4-(4-丙烯酰哌嗪-1-基)-2-氨基-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈(11mg,收率10%)。 1H NMR(400MHz,Chloroform-d)δ7.71(d,J=1.6Hz,1H),7.25–7.15(m,1H),7.05–6.91(m,1H),6.69–6.55(m,1H),6.38(dd,J=16.7,1.8Hz,1H),5.80(dd,J=10.6,1.8Hz,1H),5.68(s,2H),4.06–3.80(m,4H),3.76–3.56(m,4H). 19F NMR(376MHz,Chloroform-d)δ-111.62,-119.98.LCMS(m/z):526.4(M+H)。
化合物21-2和22-2的合成
Figure PCTCN2021128977-appb-000100
(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(21-2)和(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(22-2)
Figure PCTCN2021128977-appb-000101
步骤A:(S)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(21-1)和(S)-4-(7-溴-6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(22-1)
在置有磁子的圆底烧瓶中加入(S)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(Int F,500mg,866.2μmol),Pd(PPh 3) 4(100mg,87μmol),甲酸(159mg,3.46mmol),三乙胺(438mg,4.33mmol),氮气抽换三次后,加入DMF(15mL),反应体系加热到55℃,搅拌6h。反应完成后,冷却到室温,把反应液倒入150mL水中,黄色固体产生,过滤,并用PE(15mL)和H 2O(15mL)洗涤滤饼,收集固体,得到(S)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(21-1)和(S)-4-(7-溴-6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(22-1)的混合物(530mg)。LCMS(m/z):544.3(M+H)(21-1)和LCMS(m/z):558.3(M+H)(22-1)。
步骤B:(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(21-2)和(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)- 6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(22-2)
室温下,在置有磁子的反应管中加入(S)-4-(7-溴-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(21-1)和(S)-4-(7-溴-6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(22-1)的混合物(530mg,976.43μmol),(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸(335.24mg,1.07mmol),Pd(dppf)Cl 2(71.64mg,97.64μmol),Na 2CO 3(313mg,2.93mmol)和1,4-二氧六环/H 2O(4:1,10mL)溶液。体系鼓氮气1min后,将反应管密封,微波加热至115℃反应2h。冷却至室温,用水(60mL)稀释,EA(50mL×3)萃取。合并萃取液,无水Na 2SO 4干燥,减压浓缩后用FCC(SiO 2,EA/PE=0-60%)纯化,得到(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(21-2)(80mg,收率17%).LCMS(m/z):730.5(M+H);和(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(22-2)(380mg,收率78%)。LCMS(m/z):746.5(M+H),646.4(M-100+H)。
实施例21
Figure PCTCN2021128977-appb-000102
4-((S)-4-丙烯酰-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
Figure PCTCN2021128977-appb-000103
步骤A:(4-(6-氯-3-氰基-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-7-基)-7-氟苯并[d]噻唑-2-基)氨基甲酸叔丁酯
在室温条件下,把TMSI(110mg,548μmol)滴加到(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(80mg,110μmol)和MeCN(3mL)的混合溶液中,并在室温条件下搅拌反应5个小时,然后加入Et 3N(1mL)并继续搅拌10分钟。LCMS检测反应完成后,把反应液倒入H 2O(30mL),用EA(30mL×3)萃取,收集萃取液,得到(4-(6-氯-3-氰基-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-7-基)-7-氟苯并[d]噻唑-2-基)氨基甲酸叔丁酯(68mg,粗品)。LCMS(m/z):596.4(M+H)。
步骤B:7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-3-碳 腈
在室温条件下,把TFA:DCM(v/v=1:1,5mL)加到(4-(6-氯-3-氰基-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-7-基)-7-氟苯并[d]噻唑-2-基)氨基甲酸叔丁酯(68mg,粗品)中,并在室温条件下搅拌反应半个小时,LCMS检测反应完成后,直接浓缩除去大部分的反应液,然后把反应液用EA(5mL)稀释,并倒入饱和NaHCO 3(20mL),用EA(30mL×3)萃取,无水Na 2SO 4干燥,收集萃取液,得到7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-3-碳腈(60mg,粗品)。LCMS(m/z):496.3(M+H)。
步骤C:4-((S)-4-丙烯酰-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈
在冰浴条件下,用注射器把丙烯酰氯(11mg,121μmol)在DCM(0.5mL)的稀释液滴加到7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-3-碳腈(60mg,粗品),饱和NaHCO 3(1.5mL)和DCM(3mL)的混合溶液中,搅拌10分钟。LCMS检测反应结束后,把反应液倒入水中(30mL),用DCM(20mL×2)萃取,收集萃取液,浓缩,得到的粗产品进一步通过制备高效液相色谱分离后,得到白色固体4-((S)-4-丙烯酰-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-碳腈(7mg,三步总收率10%)。 1H NMR(400MHz,Methanol-d 4)δ8.77(s,1H),8.07(d,J=1.6Hz,1H),7.26–7.11(m,1H),7.00–6.88(m,1H),6.82(s,1H),6.26(dd,J=16.7,1.9Hz,1H),5.80(dd,J=10.7,1.8Hz,1H),5.24(d,J=24.8Hz,1H),4.21(s,1H),4.10–3.71(m,4H),3.43(d,J=12.4Hz,1H),3.15(s,1H),3.01–2.86(m,1H). 19F NMR(376MHz,Methanol-d 4)δ-114.73,-118.76.LCMS(m/z):550.4(M+H)。
实施例22
Figure PCTCN2021128977-appb-000104
4-((S)-4-丙烯酰-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-羟基喹啉-3-碳腈
Figure PCTCN2021128977-appb-000105
步骤A:7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟-2-羟基喹啉 -3-碳腈
在室温条件下,把TMSI(510mg,2.55mmol)滴加到(2S)-4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-羟基喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(380mg,510μmol)和MeCN(10mL)的混合溶液中,并在室温条件下搅拌反应5个小时,然后加入Et 3N(3mL)并继续搅拌10分钟。LCMS检测反应完成后,把反应液倒入H 2O(30mL),用EA(50mL×2)萃取,收集萃取液,得到7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟-2-羟基喹啉-3-碳腈黄色固体(200mg,收率76%)。LCMS(m/z):512.3(M+H)。
步骤B:4-((S)-4-丙烯酰-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-羟基喹啉-3-碳腈
在冰浴条件下,用注射器把丙烯酰氯(38mg,430μmol)在CH 2Cl 2(0.5mL)的稀释液滴加到7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-4-((S)-3-(氰基甲基)哌嗪-1-基)-8-氟喹啉-3-碳腈(200mg,391μmol),DIPEA(151mg,1.17mmol)和DCM(5mL)的混合溶液中,搅拌10分钟。LCMS检测反应结束后,把反应液倒入水中(50mL),用DCM(30mL×3)萃取,收集萃取液,浓缩,得到的粗产品进一步通过制备高效液相色谱分离后,得到黄色固体4-((S)-4-丙烯酰-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-羟基喹啉-3-碳腈(52mg,收率24%)。 1H NMR(400MHz,Methanol-d 4)δ7.83(s,1H),7.27–7.16(m,1H),7.04–6.95(m,1H),6.88(s,1H),6.32(dd,J=16.6,1.9Hz,1H),5.86(dd,J=10.6,1.9Hz,1H),5.48–5.12(m,1H),5.02(s,1H),4.09–3.80(m,4H),3.53–3.39(m,2H),3.05–2.87(m,1H). 19F NMR(376MHz,DMSO-d 6)δ-112.18,-123.14.LCMS(m/z):566.4(M+H)。
实施例23-31
按照与上述类似的方法制备以下化合物:
Figure PCTCN2021128977-appb-000106
Figure PCTCN2021128977-appb-000107
实施例32
Figure PCTCN2021128977-appb-000108
4-((S)-4-丙烯酰基-3-(氰基甲基)哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-甲基喹啉-3-腈
实施例32的合成参照实施例17中所述进行,在步骤A中使用(S)-4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄基酯(中间体F)代替4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(中间体B2)。LCMS(m/z):564.2(M+H)。
实施例33
Figure PCTCN2021128977-appb-000109
4-(4-丙烯酰基哌嗪-1-基)-2-氨基-7-(2-氨基苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-腈
实施例33的合成参照实施例20中所述进行,在步骤B中使用(2-((叔丁氧羰基)氨基)苯并[d]噻唑-4-基)硼酸代替(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸。 1H NMR(400MHz,DMSO-d 6)δ7.80–7.69(m,2H),7.63(s,2H),7.19–7.01(m,4H),6.91(dd,J=16.6,10.5Hz,1H),6.19(dd,J=16.6,2.4Hz,1H),5.75(dd,J=10.3,2.4Hz,1H),3.97–3.76(m,4H),3.66–3.49(m,4H).LCMS(m/z):508.4(M+H)。
实施例34
Figure PCTCN2021128977-appb-000110
4-(4-丙烯酰基哌嗪-1-基)-2-氨基-7-(2-氨基-5-氟苯并[d]噻唑-4-基)-6-氯-8-氟喹啉-3-腈
实施例34的合成参照实施例20中所述进行,在步骤B中使用(2-((叔丁氧羰基)氨基)-5-氟苯并[d]噻唑-4-基)硼酸代替(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸。 1H NMR(400MHz,DMSO-d 6)δ8.00–7.67(m,4H),7.24–7.08(m,2H),7.08–6.99(m,1H),6.91(dd,J=16.6,10.5Hz,1H),6.19(d,J=16.6Hz,1H),5.76(d,J=10.8Hz,1H),3.99–3.73(m,4H),3.73–3.46(m,4H).LCMS(m/z):526.4(M+H)。
实施例35
Figure PCTCN2021128977-appb-000111
4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(甲基氨基)喹啉-3-甲腈
Figure PCTCN2021128977-appb-000112
步骤A:4-(7-溴-6-氯-3-氰基-8-氟-2-(甲氨基)喹啉-4-基)哌嗪-1-羧酸叔丁酯
在室温条件下,将NaH(143mg,3.6mmol)加入到甲胺盐酸盐(121mg,1.8mmol)和THF(8mL)的混合溶液中并搅拌10分钟,然后把4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(300mg,0.60mmol)加入混合液中并在室温下反应2小时。LCMS检测产物生成后,把反应液加入到H 2O(30mL)中,并用EA(50mL×3)萃取。合并有机相用饱和食盐水洗涤,无水Na 2SO 4干燥,得到黄色固体4-(7-溴-6-氯-3-氰基-8-氟-2-(甲氨基)喹啉-4-基)哌嗪-1-羧酸叔丁酯(200mg,收率67%)。LCMS(m/z):499.6(M+H)。
步骤B至步骤D:4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-氟-2-(甲基氨基)喹啉-3-甲腈
实施例35的后续合成步骤参照实施例19中所述进行,在步骤B中使用4-(7-溴-6-氯-3-氰基-8-氟-2-(甲基氨基)喹啉-4-基)哌嗪-1-羧酸叔丁酯代替4-(7-溴-2,6-二氯-3-氰基-8-氟-2-羟基喹啉-4-基)哌嗪-1-羧酸叔丁酯。LCMS(m/z):540.4(M+H)。
实施例36
Figure PCTCN2021128977-appb-000113
4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-(二甲基氨基)-8-氟喹啉-3-腈
实施例36的合成参照实施例35所述,在步骤A中使用二甲胺盐酸盐代替甲胺盐酸盐。 1H NMR(400MHz,DMSO-d 6)δ7.91(s,2H),7.80(s,1H),7.23-7.20(m,1H),7.08–7.03(m,1H),6.94–6.87(m,1H),6.19(d,J=1.6Hz,1H),5.76(d,J=1.2Hz,1H),3.87(d,J=1.6Hz,4H),3.64(s,4H),3.16(s,6H). 19F NMR(376MHz,DMSO-d 6)δ-112.51,-120.42.LCMS(m/z):554.5(M+H)。
实施例37
Figure PCTCN2021128977-appb-000114
N-(4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-2-基)乙酰胺
Figure PCTCN2021128977-appb-000115
步骤A:4-(2-乙酰氨基-7-溴-6-氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
在室温条件下,向置有磁子的圆底烧瓶中加入4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(600mg,1.2mmol),乙酰胺(77mg,1.3mmol),Cs 2CO 3(1.16g,3.6mmol)和二氧六环(10mL)。置换氮气后,将Pd 2(dba) 3(109mg,0.12mmol),Xantphos(65mg,0.12mmol)加入体系,进一步置换氮气后加热至90℃,搅拌反应过夜。LCMS监测反应完成,将反应液倒入水中,乙酸乙酯萃取。合并有机相用无水硫酸钠干燥、减压浓缩得粗品经FCC(SiO 2,EA/PE=0-100%)纯化得到4-(2-乙酰氨基-7-溴-6-氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(205mg,收率33%)。LCMS(m/z):526.1(M+H)。
步骤B至步骤D:N-(4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-2-基)乙酰胺
实施例37的后续合成步骤参照实施例19所述进行,在步骤B中使用4-(2-乙酰氨基-7-溴-6-氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯代替4-(7-溴-2,6-二氯-3-氰基-8-氟-2-羟基喹啉-4-基)哌嗪-1-羧酸叔丁酯。 1H NMR(400MHz,DMSO-d 6)δ11.01(s,1H),8.03(d,J=1.5Hz,1H),7.97–7.90(m,2H),7.27(dd,J=8.4,5.6Hz,1H),7.13–7.04(m,1H),6.91(dd,J=16.6,10.4Hz,1H),6.19(dd,J=16.7,2.4Hz,1H),5.76(dd,J=10.4,2.4Hz,1H),3.97–3.81(m,4H),3.74– 3.60(m,4H),2.13(s,3H). 19F NMR(376MHz,DMSO-d 6)δ-112.16,-118.37.LCMS(m/z):568.1(M+H)。
实施例38
Figure PCTCN2021128977-appb-000116
4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-乙炔基-8-氟喹啉-3-腈
Figure PCTCN2021128977-appb-000117
步骤A:4-(7-溴-6-氯-3-氰基-8-氟-2-((三甲基硅基)乙炔基)喹啉-4-基)哌嗪-1-羧酸叔丁酯
室温条件下,把4-(7-溴-2,6-二氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(500mg,0.99mmol)和乙炔基三甲基硅烷(292mg,3.0mmol),Pd(PPh 3) 4(115mg,0.099mmol) ,CuI(38mg,0.20mmol),TEA(301mg,3.0mmol)加入到DMF(10mL)中,氮气置换三次,并在70℃反应16小时。检测产物生成后,把反应液加入到H 2O(100mL)中,并用EA(100mL×2)萃取,饱和食盐水洗涤,无水Na 2SO 4干燥,浓缩后经FCC(SiO 2,EA/PE=0-50%)得到黄色固体4-(7-溴-6-氯-3-氰基-8-氟-2-((三甲基硅基)乙炔基)喹啉-4-基)哌嗪-1-羧酸叔丁酯(350mg,收率62%)。LCMS(m/z):566.0(M+H)。
步骤B:4-(7-(2-((叔丁氧基羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-((三甲基甲硅烷基)乙炔基)喹啉-4-基)哌嗪-1-羧酸叔丁酯
室温条件下,在置有磁子的反应瓶中加入4-(7-溴-6-氯-3-氰基-8-氟-2-((三甲基硅基)乙炔基)喹啉-4-基)哌嗪-1-羧酸叔丁酯(210mg,0.37mmol),(2-((叔丁氧羰基)氨基)-7-氟苯并噻唑-4-基)硼酸(139mg,0.45mmol),K 2CO 3(154mg,1.1mmol)和二氧六环/水(v/v=3:1,5mL)。体系置换氮气后加入Pd(dppf)Cl 2(27mg,0.037mmol),再次置换氮气,升温至90℃搅拌反应1小时。冷至室温,将反应液倒入水中,乙酸乙酯萃取。合并有机相用无水硫酸钠干燥、减压浓缩得 粗品经FCC(SiO 2,EA/PE=0-100%)纯化得到4-(7-(2-((叔丁氧基羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-((三甲基甲硅烷基)乙炔基)喹啉-4-基)哌嗪-1-羧酸叔丁酯(90mg,收率32%)。LCMS(m/z):753.2(M+H)。
步骤C:4-(7-(2-((叔丁氧基羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-2-乙炔基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯
将4-(7-(2-((叔丁氧基羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟-2-((三甲基甲硅烷基)乙炔基)喹啉-4-基)哌嗪-1-羧酸叔丁酯(90mg,0.12mmol),K 2CO 3(33mg,0.24mmol)和乙腈(2mL)的混合物在室温下搅拌反应1h。LCMS监测反应完成,体系倒入水中,EA萃取。合并有机相用盐水洗涤,无水硫酸钠干燥,过滤浓缩得4-(7-(2-((叔丁氧基羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-2-乙炔基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(70mg,粗品),无需纯化,直接用于下一步反应。LCMS(m/z):681.2(M+H)。
步骤D和E:4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-2-乙炔基-8-氟喹啉-3-腈
实施例38后续合成所涉及保护基脱除及烯丙酰氯引入步骤基本上可参照实施例1中所述方法进行。LCMS(m/z):535.1(M+H)。
实施例39
Figure PCTCN2021128977-appb-000118
4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-甲氧基喹啉-3-腈
Figure PCTCN2021128977-appb-000119
步骤A:4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-甲氧基喹啉-4-基)哌嗪-1-羧酸叔丁酯
在置有磁子的反应瓶中加入4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-氟喹啉-4-基)哌嗪-1-羧酸叔丁酯(70mg,0.11mmol),甲醇钠(20mg,0.37mmol),
Figure PCTCN2021128977-appb-000120
分子筛(100mg)和二氧六环(2mL),旋紧瓶盖,加热至110℃搅拌8h。反应完成后,加入EA(20mL)稀释反应体系,依次用水(15mL)和饱和食盐水(15mL)洗涤,无水硫酸钠干燥。过滤,减压浓 缩得粗品经FCC(SiO 2,EA/PE=0-80%)纯化得4-(7-(2-((叔丁氧羰基)氨基)-7-氟苯并[d]噻唑-4-基)-6-氯-3-氰基-8-甲氧基喹啉-4-基)哌嗪-1-羧酸叔丁酯(42mg,收率59%)。LCMS(m/z):669.1(M+H)。
步骤B和C:4-(4-丙烯酰基哌嗪-1-基)-7-(2-氨基-7-氟苯并[d]噻唑-4-基)-6-氯-8-甲氧基喹啉-3-腈
实施例39后续合成所涉及保护基脱除及烯丙酰氯引入步骤基本上可参照实施例1中所述方法进行。 1H NMR(400MHz,DMSO-d 6)δ8.86(s,1H),7.96(s,1H),7.89–7.82(m,2H),7.16(dd,J=8.4,5.7Hz,1H),7.07–7.01(m,1H),6.91(dd,J=16.7,10.5Hz,1H),6.19(dd,J=16.7,2.4Hz,1H),5.75(dd,J=10.4,2.4Hz,1H),3.94–3.82(m,4H),3.78(s,3H),3.70–3.64(m,4H). 19F NMR(376MHz,DMSO-d 6)δ-113.34.LCMS(m/z):523.1(M+H)。
活性实施例
实施例1:本发明化合物对KRas G12C突变细胞的增殖抑制效果
本实验使用Promega公司的
Figure PCTCN2021128977-appb-000121
Luminescent Cell Viability Assay试剂盒,评估并验证了本发明化合物对KRas G12C突变的NCI-H358人非小细胞肺癌细胞的增殖抑制活性。
【试验材料】:NCI-H358细胞系(中国医学科学院基础医学研究所细胞资源中心,资源编号:3111C0001CCC000470),96孔透明平底黑壁细胞培养板(Greiner Bio one公司,货号#655096),RPMI-1640培养基(GE公司,货号#SH30809.01),胎牛血清FBS(Thermo Fisher公司,货号#10099-141),
Figure PCTCN2021128977-appb-000122
Luminescent Cell Viability Assay试剂盒(Promega公司,货号#G7573),PBS(Solarbio公司,货号#P1020),胰酶(Thermo Fisher,货号#25200072),DMSO(Sigma公司,货号#D2650),甲基纤维素Methylcellulose(SIGMA,货号#9004-67-5)。
【实验步骤】:向96孔细胞培养板中加入180μL细胞悬液(含1%甲基纤维素的10%FBS的RPMI1640溶液),使细胞培养板中细胞密度为1500个活细胞/孔。设置不含细胞、不含化合物、仅含3D完全培养基(含1%甲基纤维素的10%FBS的RPMI1640溶液)的对照组(即培养液对照),并设置不含化合物、含细胞的对照组(即细胞对照)。在测定过程中,使用化合物AMG510或以下参比化合物为阳性对照。将细胞板置于细胞培养箱中培养过夜。配制10倍药物溶液(含1%DMSO的10%FBS的RPMI1640溶液),浓度为10μM,在接种有细胞的96孔板中每孔加入20μL药物溶液,使每孔中化合物终浓度为:1μM,每个化合物设置三个复孔,DMSO含量为0.1%,以同样方式配制并添加化合物AMG510或以下参比化合物的溶液至阳性对照孔中。细胞板置于细胞培养箱中继续培养120h。终点检测时,融化CellTiter-Glo试剂并将细胞板移至室温平衡30min,加100μL的CellTiter-Glo至细胞板每个孔中,在定轨摇床上振动5min使细胞充分裂解,将细胞板放置于室温20min以稳定冷光信号,用多功能酶标仪(Molecular Devices公司,Spectramax M3酶标仪)全波长扫描各孔的冷光值。
【受试样品】实施例1-39的化合物,以及参比化合物A
Figure PCTCN2021128977-appb-000123
(按照WO2020/081282A1所述方法制备和表征)和参比化合物B
Figure PCTCN2021128977-appb-000124
(参照WO2015054572所述方法制备和表征)。
【数据分析】使用下列公式和GraphPad Prism 7.0软件计算各化合物作用下的细胞抑制率:
抑制率%=【1-(Lum 待测药-Lum 培养液对照)/(Lum 细胞对照-Lum 培养液对照)】×100%
IC 50值使用GraphPad Prism 7.0软件,利用非线性S曲线回归拟合数据,得出剂量-效应曲线,并由此计算。
【实验结果】本发明化合物对KRas G12C突变的NCI-H358人非小细胞肺癌细胞显示了令人满意的抗细胞增殖活性。具体地,所试验的实施例化合物均显示具有抗细胞增殖活性,IC 50值一般地<1μM,例如<0.5μM,<0.1μM,优选<50nM,更优选<20nM,最优选<10nM,例如实施例17、20、32、33、34、37化合物均显示<50nM的IC 50值,实施例1、2、3、5、6、8、9、10、39化合物显示<20nM的IC 50值。一些代表性实施例化合物的具体数据显示于表1。
表1.代表性实施例化合物对NCI-H358细胞增殖的抑制活性(1μM浓度抑制率及IC 50)
化合物 抑制率%(1μM) IC 50(μM)
实施例1 94.7 0.015
实施例2 97.6 0.003
实施例3 97.7 0.009
实施例4 94.8 0.053
实施例5 94.9 0.007
实施例6 95.0 0.017
实施例7 90.0 0.26
实施例8 94.0 0.018
实施例9 98.8 0.0086
实施例10 96.1 0.018
参比化合物A 96.9 0.011
参比化合物B / 0.35
实施例2:本发明化合物的大鼠药代动力学特性
2.1通过大鼠盒式给药(Cassette)药代动力学实验评价了本发明部分化合物的药代动力学特征。
【试验材料】:雄性SD大鼠,周龄:6-8周,体重220-250g,购自昭衍(苏州)新药研究中心有限公司;甲苯磺丁脲(Tolbutamide)(阿拉丁,货号H1401054);磺丁基β环糊精(Captisol,山东滨州智源生物,货号20191013);丙二醇(15)硬脂酸酯(Solutol,美仑生物,货号S0206A);DMSO(Vetec公司,货号WXBD0293V);乙腈(Sigma-Aldrich,货号WXBD1744V);甲醇(Sigma-Aldrich,货号WXBD2831V)。
【实验步骤】:将化合物组合配制到5%DMSO/10%Solutol/85%(20%Captisol)的溶剂中,最终每个化合物的浓度为1mg/mL,将药物制剂按照1mL/kg的注射体积尾静脉注射给SD大鼠,分别在5min,15min,30min,1h,2h,4h,8h,24h从颈外静脉穿刺采血,低温离心20分钟,收集血浆,-80℃保存待测。
【样品分析】:建立化合物LC-MS/MS分析方法。
标准曲线配制:将每个化合物吸取20μL 1mg/mL DMSO储备液,转移至900μL 50%甲醇工作液中,逐级稀释,得到一条浓度为20000,10000,5000,1000,500,100,50,20,10ng/mL的标准曲线工作液,再吸取5μL标准曲线工作液与45μL大鼠空白血浆混合,得到一条浓度为2000、1000、500、100、50、10、5、2、1ng/mL的标准曲线,用于定量未知样品。
样品前处理:50μL未知血浆样品及标准曲线样品,加入250μL含有内标的乙腈作为沉淀剂,沉淀血浆蛋白,萃取血浆中的待测化合物,低温离心20分钟,取上清液,将上清液与0.1%甲酸的水溶液混合,吸取5μL进样分析药物血药浓度。
【数据处理】:用质谱分析软件绘制标准曲线,定量未知样品,根据未知样品各时间点药物浓度用Winnonlin 8.2计算药物动力学参数。
【实验结果】:实验结果显示,在盒式给药药代动力学评价中,本发明化合物显示良好或甚至改进的药代动力学性质。
表2.
实施例 T 1/2(h) AUC 0-t(ng·hr/mL) Cl(mL/min/kg)
1 2.87 657 20.9
9 0.67 1047 15.8
10 2.66 592 27.4
13 1.37 1946 8.58
14 1.58 1096 15.1
15 1.03 717 23.3
16 3.5 2787 5.93
参比化合物A 1.00 623 27.1
AMG510 0.49 231 71.8
化合物AMG510:
Figure PCTCN2021128977-appb-000125
按照Lanman B等人,J.Med.Chem.2020,63,52-65所述方法制备。
2.2本发明代表性化合物的药代动力学性质
【试验材料】同上2.1。
【实验步骤】:按照与上述2.1中类似的方法对大鼠静脉注射给药,配制化合物浓度3mg/mL,药物配方、给药及样品采集参照2.1所述进行。口服给药组,将药物配成3mg/mL的0.5%甲基纤维素MC-400cp(阿拉丁,M112866)混悬液,10mL/kg灌胃给药。给药后分别在0.25、0.5、1、2、4、6、8、24小时从颈外静脉穿刺采血,低温离心20分钟,收集血浆,-80℃保存待测。
后续样品分析及数据处理方法与2.1所述方法类似,参照进行。
【实验结果】:结果显示实施例9具有优异的药代动力学性质,具体见表3。
表3
Figure PCTCN2021128977-appb-000126
实施例3:本发明化合物在人非小细胞肺癌NCI-H358异种移植小鼠模型中的抗肿瘤活性
本实验在人非小细胞肺癌NCI-H358异种移植小鼠模型中评估并验证了本发明化合物的增殖抑制活性。
【试验材料】:NCI-H358细胞系携带KRAS G12C突变,由康源博创生物技术(北京)有限公司提供(来源于ATCC,货号CRL-5807)。NPSG小鼠,雌性,由北京菲诺克生物技术有限公司提供。
【实验步骤】:将6~8周龄雌性NPSG小鼠于右侧肩部皮下接种5×10 6个NCI-H358细胞(含50%基质胶),接种体积为0.1mL。当肿瘤生长至平均体积160~220mm 3时,根据肿瘤大小和 小鼠体重随机分组给药。分组后随即开始给药,给药当天视为第0天。每天给药一次,灌胃给药,剂量为10mg/kg或溶剂对照(含10%环糊精pH5.0的50mM柠檬酸盐缓冲液)。实验期间,每周2次测量肿瘤体积和体重。肿瘤体积计算公式为V=D×d×d/2,其中D为肿瘤长径,d为肿瘤短径。平均肿瘤抑制率TGI%=[(C 平均值-C 0平均值)-(T 平均值-T 0平均值)]/(C 平均值-C 0平均值)*100%,其中T为给药组肿瘤体积,T 0为给药组初始肿瘤体积,C为对照组肿瘤体积,C 0为对照组初始肿瘤体积。
【实验结果】:
实验结果由表4显示,代表性本发明化合物能够显著抑制NCI-H358肿瘤生长。在同等剂量条件下,与对照化合物AMG510相比,本发明化合物显示出可比或更优的抑瘤效果。同时,各组小鼠体重均无明显改变。
表4
组别 肿瘤体积(mm 3)(第14天) TGI(%)
溶剂对照组 1170 /
实施例1 478.4 71
实施例2 497.3 69
实施例8 519.2 67
AMG510 525.1 66
此外,采用与上述相同的材料和方法,还另外考察了实施例9化合物在不同剂量下在上述非小细胞肺癌小鼠模型中的肿瘤抑制活性,以及对体重的影响,结果如附图1所示。可见,代表性实施例化合物显示优异的肿瘤抑制活性,且对体重没有明显的副作用。
实施例4:本发明化合物对细胞色素P450抑制试验
本实验评价发明化合物对细胞色素P450的抑制作用。
【试验材料】:人肝微粒体(Corning公司,货号452161);还原型烟酰胺腺嘌呤二核苷酸磷酸(NADPH,MCE公司,货号HY-F0003/CS-4998);非那西丁,双氯酚酸,α-萘黄酮,奥美拉唑和酮康唑均购自TCI公司;S-美芬妥英和睾酮购自CAYMAN公司;咪达唑仑购自Bioreclamation IVT;奎尼丁购自Damas-beta;磺胺苯吡唑购自MCE;丁呋洛尔购自TRC。
【实验步骤】:
配制0.1M磷酸钾缓冲液(K-buffer):用磷酸二氢钾和磷酸氢二钾配制100mM磷酸钾缓冲液(K-buffer),调节pH到7.4。
配制400×受试化合物及参比抑制剂:将8μL的10mM受试化合物储备液溶于12μL乙腈中。配置CYP1A2,CYP2C9和CYP2D6抑制剂的混合溶液:将12μL 1mMα-萘黄酮,10μL 40mM磺胺苯吡唑,10μL 10mM奎尼丁和8μL DMSO溶液混合。配置CYP3A4和CYP2C19 的抑制剂溶液:将8μL的DMSO溶液溶于12μL乙腈中。
配制4×NADPH磷酸钾溶液:66.7mg NADPH加入到10mL 0.1M K-buffer,pH7.4。配制4×底物磷酸钾溶液:将不同底物按照浓度要求用10mL 0.1M K-buffer配制成4倍浓度测定所需溶液。
配制0.2mg/mL人肝微粒体(HLM)溶液:将10μL 20mg/mL的人肝微粒体加入到990μL K-buffer中,冰浴保存待用。
将600μL的0.2mg/mL HLM加入到96孔板中,加入3μL 400倍受试化合物溶液;将200μL的0.2mg/mL HLM加入到96孔板中,加入1μL稀释后的阳性对照抑制剂溶液。分装30μL化合物与人的肝微粒体的混合溶液到96孔板中,然后再加入15μL的底物溶液。将上述获得的溶液和配制好的NADPH溶液于37℃预热5min。将15μL预热的NADPH溶液加入反应板,混匀,开始反应。37℃孵育反应板。3A4反应5分钟;1A2,2C9,2D6反应10分钟;2C19反应45分钟。反应结束时,加入120μL含内标的乙腈终止反应。样品涡旋振荡10min,采用5594g离心15分钟,制备样品送至LC-MS/MS分析。
【实验结果】:
实验结果显示(表5),在测试浓度下,与AMG510及参比化合物A相比,代表性本发明化合物对于药物代谢关键CYP亚型没有显著抑制作用,表现出更好的药物-药物相互作用安全性。
表5
Figure PCTCN2021128977-appb-000127
实施例5:本发明化合物对一系列KRas突变细胞的增殖抑制效果
本实验采用Promega公司的CellTiter-Glo(CTG)试剂盒,评估本发明代表性化合物对12株KRas突变肿瘤细胞株的抗增殖活性。
【试验材料】:RPMI1640培养基(Hyclone,货号SH30809.01);胎牛血清(FBS)(Gibco,货号10099-141);磷酸盐缓冲盐水PBS(Solarbio,货号P1020-500);DMSO(Sigma,货号D8418-1L);检测试剂盒CTG(Promega,货号G7573);96孔细胞培养板(Thermo,货号165305);振板仪(QILINBEIER,货号QB-9001);细胞培养箱(Thermo Scientific,货号Model 3100 Series);显微 镜(OLYMPUS,货号CKX41SF);多功能酶标仪(BMG LABTECH,货号
Figure PCTCN2021128977-appb-000128
Plus);生物安全柜(Thermo,货号Model 1300 Series A2)。以下实验中所使用的各种细胞系均购自康源博创生物科技(北京)有限公司。
【实验方法】:
细胞系 突变类型 组织来源 生长特点 完全培养基
H23 KRas-G12C 贴壁 RPMI-1640+10%FBS
H1373 KRas-G12C 贴壁 RPMI-1640+10%FBS
MIA-Paca-2 KRas-G12C 胰腺 贴壁 DMEM+10%FBS
SW837 KRas-G12C 结直肠 贴壁 RPMI-1640+10%FBS
Kyse-410 KRas-G12C 食管 贴壁 RPMI-1640+10%FBS
HCT116 KRas-G13D 结直肠 贴壁 RPMI-1640+10%FBS
T84 KRas-G13D 结直肠 贴壁 DMEM+10%FBS
A549 KRasG12S 贴壁 DMEM+10%FBS
将上述各个细胞株于37℃,5%CO 2条件下分别培养于所示完全培养基中,收获处于对数生长期的细胞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。使用完全培养基调整细胞密度,随后接种于96孔细胞培养板,每孔接种90μL,共3000个细胞。将96孔板中的细胞置于37℃、5%CO 2条件下培养。
配制10倍供试化合物培养基溶液,检测最高浓度为10μM,9个浓度,3.16倍稀释,然后转移每个稀释浓度的化合物溶液各10μL至96孔细胞板的相应实验孔中,每个浓度设置三个复孔。将已加药的96孔板中的细胞置于37℃、5%CO 2条件下继续培养72小时,之后进行CTG分析。
融化CTG试剂并平衡细胞板至室温30分钟,每孔加入100μLCTG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,读取冷光值,收集数据。使用GraphPad Prism 7.0软件分析数据,利用非线性S曲线回归拟合数据,得出剂量-效应曲线,并按照本领域技术人员熟知的常规方法计算得到相对和绝对IC 50值,以及最大抑制率%。
抑制率%=【1-(Lum 待测药-Lum 培养液对照)/(Lum 细胞对照-Lum 培养液对照)】×100%
【实验结果】:
表6.
Figure PCTCN2021128977-appb-000129
Figure PCTCN2021128977-appb-000130
表6实验结果显示,本发明代表性化合物在一系列KRas肿瘤细胞中显示出优于参比化合物A的抗增殖活性,并且具有良好的选择性。
实施例6:本发明化合物对KRas G12C抑制剂耐药性模型细胞增殖抑制效果
获得性耐药是影响KRas G12C抑制剂治疗效果的关键因素之一,本实验通过在KRas G12C抑制剂Adagrasib的获得性耐药模型(Engl J Med 2021;384:2382-93.)中进行测量,评估本发明化合物在已知耐药模型中的潜在应用价值。
本实验采用Promega公司提供的CellTiter-Glo(CTG)试剂盒,评估供试化合物对5株KRas细胞株(对KRas G12C抑制剂Adagrasib具有获得性耐药性)的细胞增殖的影响,以参比化合物A及AMG510作为对照化合物。
【试验材料】:所使用的实验材料和仪器设备同上文实施例5所罗列。本实验中所使用的各种细胞系均来自康源博创生物科技(北京)有限公司。
【实验方法】:
细胞系 细胞系类型 生长特点 完全培养基
BaF3-KRas-G12C-Y96C 小鼠原B细胞 悬浮 RPMI-1640+10%FBS
BaF3-KRas-G12C-Y96D 小鼠原B细胞 悬浮 RPMI-1640+10%FBS
BaF3-KRas-G12C-R68S 小鼠原B细胞 悬浮 RPMI-1640+10%FBS
BaF3-KRas-G12C-H95Q 小鼠原B细胞 悬浮 RPMI-1640+10%FBS
BaF3-KRas-G12C-H95D 小鼠原B细胞 悬浮 RPMI-1640+10%FBS
采用与实施例5相同的方法和条件,考察供试化合物对细胞生长抑制活性,结果见表7。
表7.
Figure PCTCN2021128977-appb-000131
Figure PCTCN2021128977-appb-000132
结果显示,本发明代表性化合物在上述耐药细胞模型中显示出优于参比化合物A及AMG510的抑制活性。
实施例7:本发明化合物对人胰腺癌Mia PaCa-2细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学研究
评价了本发明代表性化合物在人胰腺癌Mia PaCa-2细胞皮下异种移植肿瘤模型中的体内药效。
【试验材料】:BALB/c裸小鼠,6-8周龄,雌性,浙江维通利华实验动物技术有限公司。人胰腺癌Mia PaCa-2细胞(ATCC,货号:CRL-1420)。基质胶(Matrigel,Corning公司,货号356234)。
【实验方法】:将0.2mL(5×10 6个)Mia PaCa-2细胞(加基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到约137mm 3时随机分为各个实验组(溶剂对照组;AMG51010mpk;实施例9化合物3mpk;实施例9化合物10mpk;实施例9化合物30mpk),每组8只小鼠,并开始给药,每天灌胃给药一次,溶媒为0.5%甲基纤维素(400cp)水溶液。实验期间,每周2次测量肿瘤体积和体重。
肿瘤体积按照以下公式计算:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。
供试化合物的抑瘤疗效TGI(%)按照以下公式计算:TGI(%)=【1-(处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)】×100%。
【实验结果】:实验结果参见表8及附图2,其显示,代表性实施例化合物在人胰腺癌Mia PaCa-2上体现靶点相关的优异肿瘤抑制活性,活性优于对照化合物AMG510,且各组动物体重均无明显变化。
表8.
组别 肿瘤体积(mm 3)(第21天) TGI(%)
溶剂对照组 1202 /
实施例9(30mpk) 162 97.7%
实施例9(10mpk) 246 89.8%
实施例9(3mpk) 665 50.4%
AMG510(10mpk) 432 72.3%

Claims (24)

  1. 式(I)的化合物,
    Figure PCTCN2021128977-appb-100001
    其中,
    A选自C-R a或N,其中R a选自卤素、CN、硝基、C 3-6环烷基或任选被卤素取代的C 1-6烷基;
    R 1、R 2和R 3各自独立地选自H、卤素或C 1-6烷基,其中所述烷基任选被独立地选自卤素、-N(R c) 2、-OR c或3-8元杂环烷基的基团取代;
    R b在每次出现时独立地选自H、卤素、CN或任选被卤素或CN取代的C 1-6烷基;
    X选自键、-O-或-NH-;
    R 4选自H、卤素、C 1-6烷基、-C 0-6烷基-C 6-10芳基、-C 0-6烷基-C 3-8环烷基、-C 0-6烷基-C 3-8环烯基、-C 0-6烷基-3-8元杂环烷基、-C 0-6烷基-3-8元杂环烯基、-C 0-6烷基-5-10元杂芳基,其中所述烷基、芳基、环烷基、环烯基、杂环烷基、杂环烯基和杂芳基任选被独立地选自以下的一个或多个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-SR c、-(CR cR c) 0-6-(CO) 0- 1-OR c、-(CR cR c) 0-6-(CO) 0-1-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-(CO) 0-1-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环;
    R c在每次出现时独立地选自H或任选被卤素取代的C 1-6烷基,或者其中连接在同一个碳原子或氮原子上的两个R c各自独立地与它们所连接的碳原子或氮原子一起形成3-6元环状基团;
    E选自卤素、-O-R d或-N(R d) 2-,其中R d各自独立地为H或任选被卤素取代的C 1-6烷基;
    R 5
    Figure PCTCN2021128977-appb-100002
    m为0或1;
    Figure PCTCN2021128977-appb-100003
    表示芳族环;
    B和D各自独立地选自N或C;
    Z、G和Y各自独立地选自C、N、O或S;
    R 6、R 7和R 8各自独立地选自H、卤素和任选被卤素取代的C 1-6烷基;
    条件是,B和D不同时为N;且Z、G、Y、D、B中至多三个不为C;
    或其异构体、药学上可接受的盐或溶剂合物。
  2. 根据权利要求1的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中A为N或C-R a,其中R a选自卤素,优选Cl。
  3. 根据权利要求1或2的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 1选自H或卤素,优选H或F。
  4. 根据权利要求1-3任一项的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R b为H;或存在一个R b且R b为CN或任选被CN取代的C 1-6烷基;或存在两个R b且R b为C 1-6烷基。
  5. 根据权利要求1-4任一项的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 4选自H、卤素、C 1-6烷基、苯基、C 3-6环烷基、包含1、2或3个独立地选自N、O或S的杂原子的4-6元杂环烷基和包含1、2或3个独立地选自N、O或S的杂原子的5-6元杂芳基,其中所述烷基、苯基、环烷基、杂环烷基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2,其中R c在每次出现时独立地选自H或C 1-6烷基。
  6. 根据权利要求1-5任一项的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中E为卤素,优选为F,或E为-O-R d,R d为任选被卤素取代的C 1-6烷基。
  7. 根据权利要求1-6任一项的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 5
    Figure PCTCN2021128977-appb-100004
    其中Z、Y、B、D至多两个不为C,选自
    Figure PCTCN2021128977-appb-100005
    Figure PCTCN2021128977-appb-100006
    Figure PCTCN2021128977-appb-100007
    优选R 5
    Figure PCTCN2021128977-appb-100008
  8. 根据权利要求7的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 6、R 7和R 8中至少一个是卤素,优选F。
  9. 根据权利要求7的式(I)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 5选自
    Figure PCTCN2021128977-appb-100009
    优选R 5
    Figure PCTCN2021128977-appb-100010
  10. 式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,
    Figure PCTCN2021128977-appb-100011
    其中:
    A选自C-R a或N,其中R a选自卤素;
    R 1、R 2和R 3各自独立地选自H、卤素或C 1-6烷基;
    R b在每次出现时独立地选自H、卤素、CN或任选被卤素或CN取代的C 1-6烷基;
    X选自键、-O-或-NH-;
    R 4选自H、卤素、C 1-6烷基、-C 0-3烷基-C 6-10芳基、-C 0-3烷基-C 3-8环烷基、-C 0-3烷基-C 3-8环烯基、-C 0-3烷基-包含1、2或3个独立的选自N、O或S的杂原子的3-8元杂环烷基、-C 0- 3烷基-包含1、2或3个独立的选自N、O或S的杂原子的3-8元杂环烯基、-C 0-3烷基-包含1、2或3个独立的选自N、O或S的杂原子的5-10元杂芳基,其中所述烷基、芳基、环烷基、环烯基、杂环烷基、杂环烯基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c、-(CR cR c) 0-6-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环;
    R c在每次出现时独立地选自H或任选被卤素取代的C 1-6烷基;
    E选自卤素、-O-R d或-N(R d) 2-,其中R d各自独立地为H或任选被卤素取代的C 1-6烷基;
    R 6、R 7和R 8各自独立地选自H、卤素和任选被卤素取代的C 1-6烷基;
    或其异构体、药学上可接受的盐或溶剂合物。
  11. 根据权利要求10的式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,其中A为C-R a,其中R a为卤素,优选Cl。
  12. 根据权利要求10的式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 6、R 7和R 8中至少一个是卤素,优选F。
  13. 根据权利要求12的式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 6为F,且R 7和R 8为H。
  14. 根据权利要求10的式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,其中E是卤素,优选F。
  15. 根据权利要求10的式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,其中E是-O-R d,R d为任选被卤素取代的C 1-6烷基。
  16. 根据权利要求10-15任一项的式(II)化合物或其异构体、药学上可接受的盐或溶剂合物,其中R 4选自H、卤素、C 1-6烷基、苯基、C 3-6环烷基、包含1、2或3个独立的选自N、O或S的杂原子的4-6元杂环烷基和包含1、2或3个独立的选自N、O或S的杂原子的5-6元杂芳基,其中所述烷基、芳基、环烷基、杂环烷基和杂芳基任选被独立地选自以下的1、2或3个基团取代:卤素、任选被卤素取代的C 1-6烷基、-(CR cR c) 0-6-OR c和-(CR cR c) 0-6-N(R c) 2,其中连接于芳基、环烷基、环烯基、杂环烷基、杂环烯基或杂芳基上的-(CR cR c) 0-6-N(R c) 2任选通过其中N上连接的基团、连同与其所连接的环状基团上的原子及相邻的原子一起形成4-7元含氮杂环。
  17. 化合物,选自
    Figure PCTCN2021128977-appb-100012
    Figure PCTCN2021128977-appb-100013
    Figure PCTCN2021128977-appb-100014
    或其异构体、药学上可接受的盐或溶剂合物。
  18. 药物组合物,包含根据权利要求1-17任一项的化合物、其异构体或它们药学上可接受的盐或溶剂合物,以及药学上可接受的赋形剂。
  19. 根据权利要求1-17任一项的化合物、其异构体或它们药学上可接受的盐或溶剂合物,用作药物,优选用于治疗和/或预防由KRas突变、优选KRas G12C突变介导的疾病。
  20. 治疗和/或预防由Ras突变、优选KRas G12C介导的疾病的方法,包括向有需要的对象施用治疗有效量的根据权利要求1-17任一项的化合物、其异构体或它们药学上可接受的盐或溶剂合物或根据权利要求18的药物组合物。
  21. 根据权利要求1-17任一项的化合物、其异构体或它们药学上可接受的盐或溶剂合物或根据权利要求18的药物组合物用于预防或治疗由KRas突变、优选KRas G12C突变介导的疾病的用途。
  22. 根据权利要求1-17任一项的化合物、其异构体或它们药学上可接受的盐或溶剂合物或根据权利要求18的药物组合物在制备用于预防或治疗由KRas突变、优选KRas G12C突变介导的疾病的用药物中的用途。
  23. 根据权利要求21或22的用途,其中由KRas突变、优选KRas G12C突变介导的疾病选自肺癌、肺腺癌、骨癌、胰腺癌、皮肤癌、头颈癌、皮肤或眼内黑素瘤、子宫癌、卵巢癌、直肠癌、肛门区域癌、胃癌、结肠癌、乳腺癌、输卵管癌、子宫内膜癌、子宫颈癌、阴道癌、外阴癌、霍奇金病、食道癌、小肠癌、内分泌系统癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、尿道癌、阴茎癌、前列腺癌、慢性或急性白血病、淋巴细胞性淋巴瘤、膀胱癌、肾脏或输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊柱肿瘤、脑干神经胶质瘤或垂体腺瘤。
  24. 根据权利要求23的用途,其中由KRas突变、优选KRas G12C突变介导的疾病选自肺腺癌、肺癌、结肠癌、直肠癌、胰腺癌、胆管癌、子宫内膜癌、卵巢癌、白血病;最优选选自肺腺癌、结肠癌、直肠癌、胰腺癌、胆管癌。
PCT/CN2021/128977 2020-11-06 2021-11-05 用于癌症治疗的KRas抑制剂 WO2022095960A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US18/035,341 US20230406849A1 (en) 2020-11-06 2021-11-05 Kras inhibitors for treatment of cancers
EP21888652.1A EP4242207A1 (en) 2020-11-06 2021-11-05 Kras inhibitors for treatment of cancers
JP2023526934A JP2023547522A (ja) 2020-11-06 2021-11-05 がんの治療のためのKRas阻害剤
CN202180074854.2A CN116867779A (zh) 2020-11-06 2021-11-05 用于癌症治疗的KRas抑制剂

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202011229677 2020-11-06
CN202011229677.4 2020-11-06
CN202111280310.XA CN114920738A (zh) 2020-11-06 2021-11-01 用于癌症治疗的KRas抑制剂
CN202111280310.X 2021-11-01

Publications (1)

Publication Number Publication Date
WO2022095960A1 true WO2022095960A1 (zh) 2022-05-12

Family

ID=81457512

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/128977 WO2022095960A1 (zh) 2020-11-06 2021-11-05 用于癌症治疗的KRas抑制剂

Country Status (6)

Country Link
US (1) US20230406849A1 (zh)
EP (1) EP4242207A1 (zh)
JP (1) JP2023547522A (zh)
CN (1) CN116867779A (zh)
TW (1) TW202233188A (zh)
WO (1) WO2022095960A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022232332A1 (en) * 2021-04-29 2022-11-03 Amgen Inc. 2-aminobenzothiazole compounds and methods of use thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106488910A (zh) * 2013-10-10 2017-03-08 亚瑞克西斯制药公司 Kras g12c的抑制剂
CN107849022A (zh) * 2015-04-10 2018-03-27 亚瑞克西斯制药公司 取代的喹唑啉化合物和其使用方法
CN108779097A (zh) * 2015-11-16 2018-11-09 亚瑞克西斯制药公司 包含取代的杂环基的2-取代的喹唑啉化合物及其使用方法
US20190144444A1 (en) 2017-11-15 2019-05-16 Mirati Therapeutics, Inc. Kras g12c inhibitors
WO2019110751A1 (en) 2017-12-08 2019-06-13 Astrazeneca Ab Tetracyclic compounds as inhibitors of g12c mutant ras protein, for use as anti-cancer agents
CN110869357A (zh) * 2017-05-25 2020-03-06 亚瑞克西斯制药公司 化合物及其用于治疗癌症的使用方法
CN110869358A (zh) * 2017-05-25 2020-03-06 亚瑞克西斯制药公司 Kras的共价抑制剂
WO2020081282A1 (en) 2018-10-15 2020-04-23 Eli Lilly And Company Kras g12c inhibitors
CN112142735A (zh) * 2020-04-09 2020-12-29 上海凌达生物医药有限公司 一类稠和氰基吡啶类化合物、制备方法和用途
CN113563323A (zh) * 2020-04-29 2021-10-29 上海凌达生物医药有限公司 一类苯并噻唑基联芳基类化合物、制备方法和用途

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106488910A (zh) * 2013-10-10 2017-03-08 亚瑞克西斯制药公司 Kras g12c的抑制剂
CN107849022A (zh) * 2015-04-10 2018-03-27 亚瑞克西斯制药公司 取代的喹唑啉化合物和其使用方法
CN108779097A (zh) * 2015-11-16 2018-11-09 亚瑞克西斯制药公司 包含取代的杂环基的2-取代的喹唑啉化合物及其使用方法
CN110869357A (zh) * 2017-05-25 2020-03-06 亚瑞克西斯制药公司 化合物及其用于治疗癌症的使用方法
CN110869358A (zh) * 2017-05-25 2020-03-06 亚瑞克西斯制药公司 Kras的共价抑制剂
US20190144444A1 (en) 2017-11-15 2019-05-16 Mirati Therapeutics, Inc. Kras g12c inhibitors
WO2019110751A1 (en) 2017-12-08 2019-06-13 Astrazeneca Ab Tetracyclic compounds as inhibitors of g12c mutant ras protein, for use as anti-cancer agents
WO2020081282A1 (en) 2018-10-15 2020-04-23 Eli Lilly And Company Kras g12c inhibitors
CN112142735A (zh) * 2020-04-09 2020-12-29 上海凌达生物医药有限公司 一类稠和氰基吡啶类化合物、制备方法和用途
CN113563323A (zh) * 2020-04-29 2021-10-29 上海凌达生物医药有限公司 一类苯并噻唑基联芳基类化合物、制备方法和用途

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
"McGraw-Hill Dictionary of Chemical Terms", 1984, MCGRAW-HILL BOOK COMPANY
ALAMGEER ET AL., CURRENT OPIN PHARMACOL, vol. 13, 2013, pages 394 - 401
ANSEL, HOWARD C. ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, WILLIAMS& WILKINS
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
CHANG, E.H. ET AL., PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 79, no. 16, 1982, pages 4848 - 4852
DUAN NI ET AL., PHARMACOLOGY & THERAPEUTICS, Retrieved from the Internet <URL:https://doi.org/10.1016/j.pharmthera.2019.06.007>
E.L. ELIELS. H. WILENL.N. MANDER'S: "Stereochemistry of Organic Compounds", 1994, WILEY-INTERSCIENCE
EDWARD B. ROCHE: "Bioreversible Carriers in Drug Design", 1987, PHARMACEUTICAL ASSOCIATION
ENGL J MED, vol. 384, 2021, pages 2382 - 93
HIGUCHI, T. ET AL., ACS SYMPOSIUM, vol. 14
JOHN P. O'BRYAN ET AL., PHARMACOLOGICAL RESEARCH, vol. 139, 2019, pages 503 - 511
LANMAN B., J. MED. CHEM., vol. 63, 2020, pages 52 - 65
MCCORMICK, F. ET AL., CLINICAL CANCER RESEARCH, vol. 21, no. 8, 2015, pages 1797 - 1801
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
ZHI TAN ET AL., MINI-REVIEWS IN MEDICINAL CHEMISTRY, vol. 16, 2016, pages 345 - 357

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022232332A1 (en) * 2021-04-29 2022-11-03 Amgen Inc. 2-aminobenzothiazole compounds and methods of use thereof

Also Published As

Publication number Publication date
US20230406849A1 (en) 2023-12-21
TW202233188A (zh) 2022-09-01
EP4242207A1 (en) 2023-09-13
CN116867779A (zh) 2023-10-10
JP2023547522A (ja) 2023-11-10

Similar Documents

Publication Publication Date Title
JP7311228B2 (ja) Rho-関連プロテインキナーゼ阻害剤、それを含む医薬組成物並びにその調製方法及び使用
CN113637005A (zh) 用于癌症治疗的kras抑制剂
CN114269735B (zh) 二氢或四氢喹唑啉类化合物及其中间体、制备方法和应用
TWI409268B (zh) 醫藥化合物
US9890168B2 (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
IL293962A (en) Mutant kras protein inhibitors
TWI697490B (zh) 用於作為shp2抑制劑之新穎雜環衍生物
JP2008507534A (ja) フラノピリジン誘導体および使用方法
WO2017148440A1 (zh) 作为flt3抑制剂的蝶啶酮衍生物及应用
JP2007502820A (ja) N−置換ピラゾリル−アミジル−ベンズイミダゾリルc−Kit阻害剤
CN110650959B (zh) 治疗化合物和组合物及其使用方法
CN113195471B (zh) 多取代吡啶酮类衍生物及其在医药上的应用
WO2014090692A1 (en) Novel bi-ring phenyl-pyridines/pyrazines for the treatment of cancer
CN110678467A (zh) 治疗化合物和组合物及其使用方法
JP2007502822A (ja) c−Kit阻害剤としてのN3−置換イミダゾピリジン誘導体
JP7434249B2 (ja) 複素環化合物
TW201917129A (zh) 含吡唑基的三并環類衍生物、其製備方法和應用
WO2020125759A1 (zh) 作为wnt信号通路抑制剂的化合物及其医学应用
WO2022095960A1 (zh) 用于癌症治疗的KRas抑制剂
US20220017520A1 (en) Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine
CN112585138A (zh) 可用于治疗癌症的作为ErbB调节剂的4-取代的吡咯并[2,3-b]吡啶
WO2022089406A1 (zh) 含氮稠杂环类化合物及其制备方法和应用
CN111587250A (zh) 作为jak抑制剂的吡唑并嘧啶化合物
WO2022037694A1 (zh) 一类五元并六元杂环化合物及其作为蛋白受体激酶抑制剂的用途
CN114920738A (zh) 用于癌症治疗的KRas抑制剂

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: 21888652

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2023526934

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 202180074854.2

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021888652

Country of ref document: EP

Effective date: 20230606