CN113045570A - Spiro-containing quinazoline compounds - Google Patents

Spiro-containing quinazoline compounds Download PDF

Info

Publication number
CN113045570A
CN113045570A CN201911386239.6A CN201911386239A CN113045570A CN 113045570 A CN113045570 A CN 113045570A CN 201911386239 A CN201911386239 A CN 201911386239A CN 113045570 A CN113045570 A CN 113045570A
Authority
CN
China
Prior art keywords
alkyl
compound
ras
general formula
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
CN201911386239.6A
Other languages
Chinese (zh)
Inventor
谢雨礼
樊后兴
曹刚
钱立晖
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wigen Biomedicine Technology Shanghai Co Ltd
Original Assignee
Wigen Biomedicine Technology Shanghai Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wigen Biomedicine Technology Shanghai Co Ltd filed Critical Wigen Biomedicine Technology Shanghai Co Ltd
Priority to CN201911386239.6A priority Critical patent/CN113045570A/en
Priority to KR1020227023548A priority patent/KR20220122662A/en
Priority to AU2020414943A priority patent/AU2020414943A1/en
Priority to US17/767,034 priority patent/US20230002382A1/en
Priority to MX2022006609A priority patent/MX2022006609A/en
Priority to PCT/CN2020/139530 priority patent/WO2021129820A1/en
Priority to CN202080090796.8A priority patent/CN114929704A/en
Priority to JP2022539444A priority patent/JP2023508482A/en
Priority to CA3162106A priority patent/CA3162106A1/en
Priority to BR112022010267A priority patent/BR112022010267A2/en
Priority to EP20905322.2A priority patent/EP4083042A1/en
Publication of CN113045570A publication Critical patent/CN113045570A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The invention relates to a compound shown in a formula (1) and a preparation method thereof, and application of the compound shown in the formula (1) and each isomer, each crystal form and pharmaceutically acceptable salt thereof as an irreversible inhibitor of G12C mutant K-Ras protein in preparation of drugs for resisting Ras related diseases such as tumors and the like.

Description

Spiro-containing quinazoline compounds
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a quinazoline compound containing a spiro ring, a preparation method thereof and application of the compound as a K-Ras G12C inhibitor in preparation of antitumor drugs.
Background
Ras protein family is an important signal transduction transmission molecule in cells, and plays an important role in growth and development. Analysis and studies of a large number of in vitro tumor cells, animal models, and human tumor samples indicate that overactivation of Ras family proteins is an early event in human tumor development and is one of the important causes for the development and progression of a variety of cancers. Targeting and inhibition of Ras protein activity is therefore an important tool for the treatment of related tumors.
Ras proteins exist in two forms, which bind to GDP in an unactivated resting state; and when cells receive signals such as growth factor stimulation, Ras proteins bind to GTP and are activated. Activated Ras proteins recruit a variety of signal-transfer proteins, promoting phosphorylation of downstream signaling molecules such as ERK, S6, thereby activating the Ras signaling pathway, regulating cell growth, survival, migration and differentiation. The Ras protein itself GTPase enzyme activity can be the hydrolysis of GTP back to GDP. And the interaction of GTP enzyme activator proteins (GAPs) and Ras in cells greatly promotes the activity of Ras GTPase, so that the over-activation of Ras protein is prevented.
Mutations in the K-Ras, H-Ras and N-Ras proteins in the Ras protein family are one of the common genetic mutations in many tumors, and are the major factors that lead to the overactivation of Ras proteins in tumors. Compared with wild Ras protein, these mutations result in unregulated Ras protein activity, stable binding of GTP, and sustained activation, thereby promoting growth, migration, and differentiation of tumor cells. Among these, K-Ras protein mutations are the most common, accounting for 85% of all Ras mutations, while N-Ras (12%) and H-Ras (3%) are relatively rare. K-Ras mutations are very common in a variety of cancers: including pancreatic (95%), colorectal (45%), and lung (25%), among others, while relatively rare (< 2%) in breast, ovarian, and brain cancers. The K-Ras mutation site is mainly concentrated at position G12, with mutations at G12C being most common. For example, in non-small cell lung cancer (NSCLC), K-Ras G12C accounts for 50% of all K-Ras mutations, followed by G12V and G12D. Genomics research shows that K-Ras mutation in non-small cell lung cancer does not coexist with EGFR, ALK, ROS1, RET and BRAF mutation, but coexists with STK11, KEAP1, TP53 and other mutations, and suggests that the K-Ras mutation and the STK11, KEAP1, TP53 mutation and other synergistic effects are possibly involved in malignant transformation, proliferation and invasion of cells. In addition to tumors, abnormal activation of Ras protein is also involved in non-neoplastic diseases including diabetes, neurodegenerative diseases, and the like, and thus it can be seen that small molecule compounds targeting Ras protein can benefit a large number of cancer patients carrying specific genetic variations and non-cancer patients with overactivation of the Ras pathway.
Since forty years of discovery of Ras mutations in tumors, although we have made a more thorough understanding of the pathogenesis of the Ras pathway, no clinically effective therapeutic approach to targeting Ras proteins has been available for a large number of patients carrying Ras protein mutations and overactivation of the Ras pathway. Therefore, the development of a high-activity small-molecule inhibitor aiming at the Ras protein, in particular to the K-Ras G12C protein with high mutation frequency has important clinical significance.
The K-Ras G12C mutant protein is used as a leading drug target, and the current research is not many, and only a few compounds enter clinical research stages, such as AMG510 of Amgen company and MRTX849 of Mirati company. Cell in 2018 reported a covalent inhibitor ARS-1620 targeting the K-Ras G12C mutation [ Cell,2018,172:578-589 ]. A spiro compound with K-Ras G12C activity and anti-tumor activity in mice is reported in WO2018/143315, and the general formula A and the structure of a representative compound B (example 35 in the patent) are as follows (please refer to the patent for the definition of each symbol in the formula):
Figure BDA0002342540200000021
currently, there is an urgent need for the research and discovery of compounds having good K-Ras G12C activity and superior pharmacokinetic properties.
Disclosure of Invention
The invention aims to provide a compound with a structural general formula shown as a formula (1), or each isomer, each crystal form, pharmaceutically acceptable salt, hydrate or solvate thereof:
Figure BDA0002342540200000031
in formula (1):
m is an integer of 1 or 2;
n is an integer of 1 or 2;
R1is H, halogen, C1-C3 alkyl, C2-C4 alkenyl, C2-C4 alkynyl or C3-C6 cycloalkyl;
R2is C1-C3 alkoxy, halogeno C1-C3 alkoxy or-NRaRbWherein R isaAnd RbIndependently H, C1-C3 alkyl, halo-C1-C3 alkyl, or RaAnd RbA heterocycloalkyl group having 4 to 7 members for the N atom, which may be substituted by 1 to 3 halogen atoms;
R3is composed of
Figure BDA0002342540200000032
Figure BDA0002342540200000033
Figure BDA0002342540200000034
Wherein R iscIs H or F, RdIs H, F, Cl or Me, ReIs H, F, Cl or Me, RfIs F, NH2Me or cyclopropyl, Rx1、Rx2、Rx3、Rx4、Rx5、Rx6And Rx7Independently H, F, Cl, OH, OMe, NH2、CF3C1-C3 alkyl or C3-C6 cycloalkyl;
R4is H, halogen, CN, C1-C3 alkyl, haloC 1-C3 alkyl or heteroaryl; and
when R is3Is composed of
Figure BDA0002342540200000041
And R is4When is H, R5Comprises the following steps:
Figure BDA0002342540200000042
Figure BDA0002342540200000043
wherein n is1、n2、n3、m1、m2And m3Independently is an integer of 1 or 2, RgIs C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3) alkoxy- (C2-C3) alkyl-, (halogenated C1-C3) alkoxy- (C2-C3) alkyl-, (C3-C6) cycloalkyl- (C1-C3) alkyl-, heterocycloalkyl- (C1-C3) alkyl-, halogenated C1-C3 alkyl or cyano-substituted C1-C3 alkyl, RhIs composed of
Figure BDA0002342540200000044
Figure BDA0002342540200000045
When R is3Is composed of
Figure BDA0002342540200000051
And R is4Halogen, CN, C1-C3 alkyl, halogenated C1-C3 alkyl or heteroaryl; or, when R is3Is composed of
Figure BDA0002342540200000052
Figure BDA0002342540200000053
Figure BDA0002342540200000054
When R is5Comprises the following steps:
Figure BDA0002342540200000055
Figure BDA0002342540200000056
Figure BDA0002342540200000057
wherein n is1、n2、n3、m1、m2And m3Independently is an integer of 1 or 2, RgIs C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3) alkoxy- (C2-C3) alkyl-, (halogenated C1-C3) alkoxy- (C2-C3) alkyl-, (C3-C6) cycloalkyl- (C1-C3) alkyl-, heterocycloalkyl- (C1-C3)1-C3) alkyl-, halogeno-C1-C3 alkyl or cyano-substituted C1-C3 alkyl, RhIs composed of
Figure BDA0002342540200000061
Figure BDA0002342540200000062
Figure BDA0002342540200000063
RiIs H, halogen, methyl or cyano.
In another preferred embodiment, wherein in said general formula (1), R1H, F, Cl, Me, Et, vinyl, isopropyl, ethynyl or cyclopropyl.
In another preferred embodiment, wherein in said general formula (1), R2Is CH3CH2O-、CF3CH2O-、CHF2CH2O-、
Figure BDA0002342540200000064
In another preferred embodiment, wherein in said general formula (1), R3Is composed of
Figure BDA0002342540200000065
Figure BDA0002342540200000066
Figure BDA0002342540200000071
In another preferred embodiment, wherein in said general formula (1), R4Is H, F, CN, Me, CF3
Figure BDA0002342540200000072
Figure BDA0002342540200000073
In another preferred embodiment, wherein in said general formula (1), when R is3Is composed of
Figure BDA0002342540200000074
Figure BDA0002342540200000075
And R is4When is H, R5Comprises the following steps:
Figure BDA0002342540200000076
Figure BDA0002342540200000077
Figure BDA0002342540200000081
Figure BDA0002342540200000091
Figure BDA0002342540200000101
in another preferred embodiment, wherein in said general formula (1), when R is3Is composed of
Figure BDA0002342540200000102
Figure BDA0002342540200000103
And R is4Is F, CN, Me, CF3
Figure BDA0002342540200000104
Figure BDA0002342540200000105
When the current is over; or, when R is3Is composed of
Figure BDA0002342540200000106
Figure BDA0002342540200000107
Figure BDA0002342540200000111
Figure BDA0002342540200000112
When R is5Comprises the following steps:
Figure BDA0002342540200000113
Figure BDA0002342540200000114
Figure BDA0002342540200000121
Figure BDA0002342540200000131
in various embodiments, representative compounds of the invention have one of the following structures:
Figure BDA0002342540200000151
Figure BDA0002342540200000161
Figure BDA0002342540200000181
Figure BDA0002342540200000191
Figure BDA0002342540200000201
Figure BDA0002342540200000211
Figure BDA0002342540200000221
Figure BDA0002342540200000231
Figure BDA0002342540200000241
Figure BDA0002342540200000251
Figure BDA0002342540200000261
another object of the present invention is to provide a pharmaceutical composition comprising a pharmaceutically acceptable excipient or carrier and, as an active ingredient, a compound of the general formula (1) of the present invention, or its isomers, pharmaceutically acceptable inorganic or organic salts.
A further object of the present invention provides the use of the above-mentioned compounds of the present invention, or their respective isomers, pharmaceutically acceptable inorganic or organic salts, for the preparation of a medicament for the treatment of RAS related diseases.
Through synthesis and careful study of various novel compounds having K-RAS G12C inhibitory effect, the inventors found that when R is in the compound of formula (1)5When the compound is spiro, the compound has strong K-RAS G12C inhibition activity and the pharmacokinetics of the compoundThe properties are greatly improved, and the in vivo activity of the compound is enhanced. On the other hand, the present inventors have found that the 2-position (R) of acrylamide4Substituent) is substituted with a smaller volume of F atom, the compound also has excellent K-RAS G12C inhibitory activity and pharmacokinetic properties.
It is to be understood that both the foregoing general description and the following detailed description of the present invention are exemplary and explanatory and are intended to provide further explanation of the invention as claimed.
Synthesis of Compounds
The following specifically describes the production process of the compound of the general formula (1) of the present invention, but these specific processes do not set any limit to the present invention.
The compounds of formula (1) described above may be synthesized using standard synthetic techniques or known techniques in combination with the methods described herein. In addition, the solvents, temperatures and other reaction conditions mentioned herein may vary. The starting materials for the synthesis of the compounds may be obtained synthetically or from commercial sources. The compounds described herein and other related compounds having various substituents can be synthesized using well-known techniques and starting materials, including those found in March, ADVANCED ORGANIC CHEMISTRY 4thEd., (Wiley 1992); carey and Sundberg, ADVANCED ORGANIC CHEMISTRY 4thEd, Vols.A and B (Plenum 2000, 2001), Green and Wuts, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS 3rdThe method in ed., (Wiley 1999). The general method of compound preparation may be varied by the use of appropriate reagents and conditions for introducing different groups into the formulae provided herein.
In one aspect, the compounds described herein are according to methods well known in the art. However, the conditions of the method, such as reactants, solvent, base, amount of the compound used, reaction temperature, time required for the reaction, and the like, are not limited to the following explanation. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains. In one aspect, the present invention also provides a method for preparing the compound represented by the general formula (1), which comprises the following general reaction scheme 1:
general reaction scheme 1
Figure BDA0002342540200000281
Embodiments of the compounds of formula (1) can be prepared according to general reaction scheme 1, wherein m, n, R1、R2、R3、R4And R5As defined hereinbefore, PG represents a protecting group, X represents a boronic acid, boronic ester or trifluoroborate, T represents H, F, Cl or I, R1aRepresents C1-C3 alkyl, C2-C4 alkenyl, C2-C4 alkynyl or C3-C6 cycloalkyl. As shown in general reaction scheme 1, Compound A1 (synthesized with reference to WO 2018/143315) and Compound A2 are reacted under basic conditions to produce Compound A3, Compound A3 and R5H reacts under the alkaline condition to generate a compound A4, a compound A4 and R3H reacts under strongly basic conditions to form compound A5, and when T ═ I, compound A5 and R1Coupling reaction of-X to give compound A6, compound A6 and R3Coupling reaction of-X again gives compound a7, whereas when T ═ H, F or Cl, compound a5 is directed to R3And (4) carrying out coupling reaction again on the-X to obtain a compound A7, removing a protecting group from the compound A7 to obtain a compound A8, and obtaining a target compound A10 from the compound A8 and the compound A9.
Further forms of the compounds
"pharmaceutically acceptable" as used herein refers to a substance, such as a carrier or diluent, which does not diminish the biological activity or properties of the compound and which is relatively non-toxic, e.g., by being administered to an individual without causing unwanted biological effects or interacting in a deleterious manner with any of the components it contains.
The term "pharmaceutically acceptable salt" refers to a form of a compound that does not cause significant irritation to the organism to which it is administered and does not abrogate the biological activity and properties of the compound. In certain particular aspects, pharmaceutically acceptable salts are obtained by reacting a compound of formula (1) with an acid, such as an inorganic acid, e.g., hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid, nitric acid, phosphoric acid, etc., an organic acid, e.g., formic acid, acetic acid, propionic acid, oxalic acid, trifluoroacetic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, etc., and an acidic amino acid, e.g., aspartic acid, glutamic acid, etc.
References to pharmaceutically acceptable salts are understood to include solvent addition forms or crystalline forms, especially solvates or polymorphs. Solvates contain either stoichiometric or non-stoichiometric amounts of solvent and are selectively formed during crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is ethanol. Solvates of the compounds of formula (1) are conveniently prepared or formed as described herein. For example, the hydrate of the compound of formula (1) is conveniently prepared by recrystallization from a mixed solvent of water/organic solvent, using an organic solvent including, but not limited to, tetrahydrofuran, acetone, ethanol or methanol. In addition, the compounds mentioned herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to unsolvated forms for purposes of the compounds and methods provided herein.
In other embodiments, the compound of formula (1) is prepared in different forms, including, but not limited to, amorphous, pulverized, and nano-sized forms. In addition, the compound of formula (1) includes crystalline forms, and may also be polymorphic forms. Polymorphs include different lattice arrangements of the same elemental composition of a compound. Polymorphs typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal forms, optical and electrical properties, stability and solubility. Different factors such as recrystallization solvent, crystallization rate and storage temperature may cause a single crystal form to dominate.
In another aspect, the compounds of formula (1) exist with axial chirality and/or chiral centers and thus occur as racemates, racemic mixtures, single enantiomers, diastereomeric compounds and single diastereomers. Each such axial chirality will independently give rise to two optical isomers and all possible optical isomers and diastereomeric mixtures and pure or partially pure compounds are included within the scope of the invention. The present invention is meant to include all such isomeric forms of these compounds.
Term(s) for
Unless otherwise defined, terms used in this application, including the specification and claims, are defined as follows. It must be noted that, in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Conventional methods of mass spectrometry, nuclear magnetism, HPLC, protein chemistry, biochemistry, recombinant DNA technology and pharmacology are used, if not otherwise stated. In this application, "or" and "means" and/or "are used unless otherwise stated.
Unless otherwise specified, "alkyl" refers to a saturated aliphatic hydrocarbon group, including straight and branched chain groups of 1 to 6 carbon atoms. Lower alkyl groups having 1 to 4 carbon atoms are preferred, such as methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl, tert-butyl. As used herein, "alkyl" includes unsubstituted and substituted alkyl groups, especially alkyl groups substituted with one or more halogens. Preferred alkyl groups are selected from CH3、CH3CH2、CF3、CHF2、CF3CH、iPr、nPr、iBu、nBu ortBu。
Unless otherwise specified, "alkenyl" refers to an unsaturated aliphatic hydrocarbon group containing a carbon-carbon double bond, including straight and branched chain groups of 1 to 6 carbon atoms. Lower alkenyl having 1 to 4 carbon atoms, such as vinyl, 1-propenyl, 1-butenyl or 2-methylpropenyl, is preferred.
Unless otherwise specified, "alkynyl" refers to unsaturated aliphatic hydrocarbon groups containing carbon-carbon triple bonds, including straight and branched chain groups of 1 to 6 carbon atoms. Lower alkenyl having 1 to 4 carbon atoms, such as ethynyl, 1-propynyl or 1-butynyl, is preferred.
Unless otherwise specified, "cycloalkyl" refers to a 3-to 6-membered all-carbon monocyclic aliphatic hydrocarbon group in which one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. For example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexane, cyclohexadiene and the like.
Unless otherwise specified, "alkoxy" refers to an alkyl group bonded to the rest of the molecule through an ether oxygen atom. Representative of alkoxy groups are alkoxy groups having 1 to 6 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy and tert-butoxy. As used herein, "alkoxy" includes unsubstituted and substituted alkoxy groups, especially alkoxy groups substituted with one or more halogens. Preferred alkoxy groups are selected from OCH3、OCF3、CHF2O、CF3CH2O、i-PrO、n-PrO、i-BuO、n-BuO ort-BuO。
Unless otherwise specified, "heteroaryl" refers to an aromatic group containing one or more heteroatoms (O, S or N), the heteroaryl being monocyclic or polycyclic, e.g., a monocyclic heteroaryl ring fused to one or more carbocyclic aromatic groups or other monocyclic heterocyclyl groups. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, indolyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, benzopyridyl, and pyrrolopyrimidinyl.
Unless otherwise specified, "heterocycloalkyl" refers to a saturated or partially unsaturated ring system radical containing one or more heteroatoms (O, S or N), where the nitrogen and sulfur atoms are optionally oxidized and the nitrogen atoms are optionally quaternized as ring atoms. Unless otherwise indicated, the ring system of "heterocycloalkyl" may be a monocyclic, bicyclic, spiro or polycyclic ring system. "heterocycloalkyl" can be attached to the rest of the molecule through more than one ring carbon or heteroatom. Examples of "heterocycloalkyl" include, but are not limited to, pyrrolidine, piperidine, N-methylpiperidine, tetrahydroimidazole, pyrazolidine, butyrolactam, valerolactam, imidazolidinone, hydantoin, dioxolane, phthalimide, piperidine, pyrimidine-2, 4(1H,3H) -dione, 1, 4-dioxane, morpholine, thiomorpholine-S-oxide, thiomorpholine-S, S-oxide, piperazine, pyran, pyridone, 3-pyrroline, thiopyran, pyrone, tetrahydrofuran, tetrahydrothiophene, quinuclidine, 2-azaspiro [3.3] heptane and the like.
Unless otherwise specified, "halogen" (or halo) refers to fluorine, chlorine, bromine or iodine. The term "halo" (or "halogen substituted") appearing before the radical name indicates that the radical is partially or fully halogenated, that is, substituted in any combination by F, Cl, Br or I, preferably by F or Cl.
Specific pharmaceutical and medical terms
The term "acceptable", as used herein, means that a prescribed component or active ingredient does not unduly adversely affect the health of the general therapeutic target.
The terms "treat," "treatment process," or "therapy" as used herein include alleviating, inhibiting, or ameliorating a symptom or condition of a disease; inhibiting the generation of complications; ameliorating or preventing underlying metabolic syndrome; inhibiting the development of a disease or condition, such as controlling the development of a disease or condition; alleviating the disease or symptoms; regression of the disease or symptoms; alleviating a complication caused by the disease or symptom, or preventing or treating a symptom caused by the disease or symptom. As used herein, a compound or pharmaceutical composition, when administered, can ameliorate a disease, symptom, or condition, particularly severity, delay onset, slow progression, or reduce duration of a condition. Whether fixed or temporary, continuous or intermittent, may be attributed to or associated with administration.
"active ingredient" means a compound represented by the general formula (1), and a pharmaceutically acceptable inorganic or organic salt of the compound of the general formula (1). The compounds of the present invention may contain one or more asymmetric centers (axial chirality) and thus occur as racemates, racemic mixtures, single enantiomers, diastereomeric compounds and individual diastereomers. Asymmetric centers that may be present depend on the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and all possible optical isomers and diastereomeric mixtures and pure or partially pure compounds are included within the scope of the invention. The present invention is meant to include all such isomeric forms of these compounds.
The terms "compound", "composition", "medicament" or "drug" are used interchangeably herein and refer to a compound or composition that, when administered to an individual (human or animal), is capable of inducing a desired pharmaceutical and/or physiological response through local and/or systemic action.
The term "administering" or "administration" as used herein refers to the direct administration of the compound or composition, or the administration of a prodrug (produg), derivative (derivative), or analog (analog) of the active compound, and the like.
Although numerical ranges and parameters setting forth the broad scope of the invention are approximate, the values set forth in the specific examples are presented as precisely as possible. Any numerical value, however, inherently contains certain standard deviations found in their respective testing measurements. As used herein, "about" generally means that the actual value is within plus or minus 10%, 5%, 1%, or 0.5% of a particular value or range. Alternatively, the term "about" means that the actual value falls within the acceptable standard error of the mean, as considered by those skilled in the art. Except in the experimental examples, or where otherwise expressly indicated, it is to be understood that all ranges, amounts, values and percentages herein used (e.g., to describe amounts of materials, length of time, temperature, operating conditions, quantitative ratios, and the like) are to be modified by the word "about". Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, these numerical parameters are to be understood as meaning both the number of significant digits indicated and the number resulting from applying ordinary rounding techniques.
Unless defined otherwise herein, the scientific and technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. Furthermore, as used herein, the singular tense of a noun, unless otherwise conflicting with context, encompasses the plural form of that noun; the use of plural nouns also covers the singular form of such nouns.
Therapeutic uses
The invention provides methods of treating diseases, including but not limited to conditions (e.g., cancer) involving G12C K-Ras, G12C H-Ras and/or G12C N-Ras mutations, using the compounds or pharmaceutical compositions of the invention.
In some embodiments, there is provided a method for treating cancer, the method comprising administering to an individual in need thereof an effective amount of any of the aforementioned pharmaceutical compositions that protect a compound of structural formula (1). In some embodiments, the cancer is mediated by K-Ras, H-Ras and/or G12C N-Ras mutations. In other embodiments, the cancer is lung cancer, pancreatic cancer, colon cancer, MYH-related polyposis, or colorectal cancer.
Route of administration
The compound and the pharmaceutically acceptable salt thereof can be prepared into various preparations, wherein the preparation comprises the compound or the pharmaceutically acceptable salt thereof in a safe and effective amount range and a pharmaceutically acceptable excipient or carrier. Wherein "safe, effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. The safe and effective amount of the compound is determined according to the age, condition, course of treatment and other specific conditions of a treated subject.
"pharmaceutically acceptable excipient or carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of being blended with the compounds of the present invention and with each other without significantly diminishing the efficacy of the compounds. Examples of pharmacologically acceptable excipients or carrier moieties are cellulose and derivatives thereofSubstances (such as sodium carboxymethylcellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, pulvis Talci, solid lubricant (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, oleum Sesami, peanut oil, oleum Olivarum, etc.), polyalcohol (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifier (such as Tween, etc.), and its preparation method
Figure BDA0002342540200000331
) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
When the compounds of the present invention are administered, they may be administered orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously), topically.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds. When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 1 to 2000mg, preferably 50 to 1000 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
The features mentioned above with reference to the invention, or the features mentioned with reference to the embodiments, can be combined arbitrarily. All the features disclosed in this specification may be combined in any combination, and each feature disclosed in this specification may be replaced by alternative features serving the same, equivalent or similar purpose. Thus, unless expressly stated otherwise, the features disclosed are merely generic examples of equivalent or similar features.
The various specific aspects, features and advantages of the compounds, methods and pharmaceutical compositions described above are set forth in detail in the following description, which makes the present invention clear. It should be understood herein that the detailed description and examples, while indicating specific embodiments, are given by way of illustration only. After reading the description of the invention, one skilled in the art can make various changes or modifications to the invention, and such equivalents fall within the scope of the invention as defined in the application.
In all of the embodiments described herein, the first,1H-NMR was recorded using a Vian Mercury 400 NMR spectrometer with chemical shifts expressed in delta (ppm); the silica gel used for separation is not illustrated to be 200-300 meshes, and the proportions of the eluents are volume ratios.
The invention employs the following abbreviations: CD (compact disc)3OD represents deuterated methanol; MeCN represents acetonitrile; DCM represents dichloromethane; DIPEA stands for diisopropylethylamine; dioxane represents 1, 4-Dioxane; DMF represents dimethylformamide; k3PO4Represents potassium phosphate; min represents min; MS represents mass spectrum; NaH represents sodium hydride; NMR stands for nuclear magnetic resonance; pd2(dba)3Represents tris (dibenzylideneacetone) dipalladium; pd (dppf) Cl2Represents 1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride; TFA (CF)3COOH) represents trifluoroacetic acid; TLC for thin layer chromatography; THF represents tetrahydrofuran; xantphos stands for 4, 5-bis (diphenylphosphino) -9, 9-dimethylxanthene.
The specific implementation mode is as follows:
example Synthesis of 7- (6-cyclopropyl-8-ethoxy-2- ((1- (2-methoxyethyl) piperidin-4-yl) oxy) -7- (5-methyl-1H-indazol-4-yl) quinazolin-4-yl) -2, 7-diazaspiro [3.5] non-2-yl) -2-fluoroprop-2-en-1-one (Compound 1)
Figure BDA0002342540200000361
Step 1: synthesis of Compounds 1-3
Suspending compound 1-1(5.5g,13.1mmol) in dioxane (80mL), adding DIPEA (10.1g,78.6mmol) under ice bath, adding 1-2(3.0g,13.1mmol), stirring for 30min, returning to room temperature, stirring for 1h, detecting by TLC, reacting completely, adding water, extracting with EA, drying and concentrating organic phase, and pulping EA to obtain yellow solid 1-3(4.5g, yield 56%).
1H NMR(400MHz,DMSO-d6)δ:8.26(d,J=1.5Hz,1H),3.79(s,4H),3.65(s,4H),1.86(t,J=5.3Hz,4H),1.39(s,9H);MS(ESI):MS(ESI):611.2[M+1]+.
Step 2: synthesis of Compounds 1-4
The compound 1-3(4.5g,7.4mmol) was dissolved in a mixed solution of DMF (40mL) and THF (40mL), 1- (2-methoxyethyl) -4-hydroxypiperidine (2.4g,14.8mmol), DABCO (0.2g,1.5mmol) were added, the mixture was stirred at room temperature overnight, the reaction was completed, water was added, EA was extracted, the organic phase was dried and concentrated, and column chromatography was performed to obtain the compound 1-3(4.1g, yield 76%).
1H NMR(400MHz,DMSO-d6)δ:8.14(s,1H),4.99(ddd,J=11.7,8.5,3.6Hz,1H),3.66(s,9H),3.44(t,J=5.8Hz,2H),3.24(s,3H),3.17(d,J=5.2Hz,1H),2.77(dt,J=9.5,8.8Hz,2H),2.26(t,J=9.8Hz,2H),2.00(d,J=12.0Hz,2H),1.84(d,J=4.3Hz,4H),1.74–1.61(m,2H),1.39(s,9H);MS(ESI):734.2[M+1]+.
And step 3: synthesis of Compounds 1-5
Trifluoroethanol (0.9g,8.4mmol) was dissolved in anhydrous DMF (10mL), NaH was added under ice bath, and stirred at room temperature for 5min to give sodium trifluoroethoxide. Compound 1-4(4.1g,5.6mmol) was dissolved in anhydrous THF (40mL) and tris was added as prepared aboveStirring overnight at room temperature for reaction, extracting with water and EA, concentrating organic phase, and performing column chromatography to obtain compound 1-5(4.5g, yield 99%), MS (ESI) 814.2[ M +1 ]]+
And 4, step 4: synthesis of Compounds 1-6
To a single neck flask, add compound 1-5(4.1g,5.5mmol), cyclopropylboronic acid (0.5g,6.1mmol), Pd (dppf) Cl2(0.9g,1.1mmol),K3PO4(0.4g,1.7mmol), MeCN (40mL), dioxane (40mL) and H were added sequentially2O (16.5mL), stirring at 100 deg.C for 5h under nitrogen protection, reacting completely, and performing column chromatography to obtain compound 1-6(2.5g, yield 62%), MS (ESI):728.3[ M +1 ]]+
And 5: synthesis of Compounds 1-7
To a single-necked flask, compound 1-6(2.5g,3.4mmol), 5-methyl-1H-indazole-4-boronic acid (0.9g,5.1mmol), Pd were added2(dba)3(0.3g,0.4mmol),Xatphos(0.3g,0.7mmol),K3PO4(2.2g,10.2mmol), dioxane (40mL) and H were added2O (4mL), stirring overnight at 120 ℃ under the protection of nitrogen, reacting completely, and performing column chromatography to obtain compound 1-7(1g, yield 38%), MS (ESI):780.4[ M +1 ]]+
Step 6: synthesis of Compounds 1-8
Dissolving compound 1-7(1g,1.3mmol) in DCM (15mL), adding TFA (5mL), stirring at room temperature for 2h, reacting completely, concentrating, basifying with saturated sodium carbonate, EA extracting, drying and concentrating to obtain compound 1-8(0.9g, yield 99%), MS (ESI):680.4[ M + 1%)]+
And 7: synthesis of Compound 1
Compound 1-8(150mg,0.2mmol), 2-fluoroacrylic acid (20mg,0.22mmol) were dissolved in DCM (15mL), DIPEA (52mg,0.4mmol), HATU (114mg,0.3mmol) were added under ice bath and stirred overnight, the reaction was completed, the reaction solution was washed with saturated brine, the organic phase was dried and concentrated, and column chromatography gave compound 1(30mg, yield 20%).
1H NMR(400MHz,CD3OD)δ:7.47-7.37(m,2H),7.30(d,J=8.6Hz,1H),7.24(s,1H),5.52(d,J=3.4Hz,0.5H),5.40(d,J=3.4Hz,0.5H),5.15(d,J=3.4Hz,0.5H),5.11(d,J=3.4Hz,0.5H),4.54(dq,J=17.7,8.8Hz,1H),4.23-4.12(m,3H),3.83(s,2H),3.73-3.59(m,4H),3.54(t,J=5.2Hz,2H),3.28(s,3H),3.10(dd,J=10.1,6.2Hz,2H),2.88(s,2H),2.85-2.72(m,2H),2.13(d,J=26.2Hz,5H),1.97(dd,J=11.9,6.8Hz,6H),1.36(dt,J=14.0,6.6Hz,1H),1.27-1.17(m,5H);MS(ESI):752.4[M+1]+
Two chiral isomers of the compound 1 can be obtained by adopting a chiral column separation and purification method:
Figure BDA0002342540200000381
examples 2-155 Synthesis of Compounds 2-155
Using different starting materials, the objective compounds 2 to 155 were obtained according to a similar synthesis method as in example 1.
TABLE 1
Figure BDA0002342540200000382
Figure BDA0002342540200000391
Figure BDA0002342540200000401
Figure BDA0002342540200000411
Figure BDA0002342540200000421
Figure BDA0002342540200000431
Figure BDA0002342540200000441
Figure BDA0002342540200000451
Figure BDA0002342540200000461
Figure BDA0002342540200000471
Figure BDA0002342540200000481
Figure BDA0002342540200000491
Figure BDA0002342540200000501
Figure BDA0002342540200000511
Figure BDA0002342540200000521
Figure BDA0002342540200000531
Figure BDA0002342540200000541
Figure BDA0002342540200000551
Figure BDA0002342540200000561
Figure BDA0002342540200000571
Figure BDA0002342540200000581
Figure BDA0002342540200000591
Figure BDA0002342540200000601
Figure BDA0002342540200000611
Figure BDA0002342540200000621
Figure BDA0002342540200000631
Figure BDA0002342540200000641
Figure BDA0002342540200000651
Figure BDA0002342540200000661
Figure BDA0002342540200000671
Figure BDA0002342540200000681
Figure BDA0002342540200000691
Figure BDA0002342540200000701
EXAMPLE 156 detection of the amount of pERK and ERK proteins in H358 cells by Compounds
H358 cells were seeded in a 24-well plate, after one day of growth, a test compound (1. mu.M concentration) was added, after 24 hours of compound action, after cell lysis, cell lysates were transferred to a 96-well ELISA plate, levels of pERK and ERK in the lysates were determined using an ELISA kit (abcam 176660), the ratio of pERK to ERK was calculated and compared to DMSO group, the percentage of inhibition of pERK activity by the compound was calculated, and the results are shown in Table 2 below.
TABLE 2 inhibitory Activity of Compounds of the invention against H358 intracellular pERK levels
Figure BDA0002342540200000702
Figure BDA0002342540200000711
Figure BDA0002342540200000721
+ represents an inhibition rate of less than or equal to 50%
+ indicates an inhibition of 50% to 90%
And +++ indicates an inhibition of greater than 90%.
Antiproliferative activity of the compounds of example 157 on H358 cells
2500H 358 cells were plated in ultra low adsorption 96-well plates (corning,7007) and after one day of growth, a gradient dilution compound (5. mu.M maximum, 5-fold dilution, five total doses) was added, and after three days of compound addition, Cell Titer Glow (Promega, G9681) was added to evaluate pellet growth and calculate IC50The values, results are given in table 3 below.
TABLE 3 antiproliferative activity of the compounds of the invention on H358 cells
Figure BDA0002342540200000722
Figure BDA0002342540200000731
Figure BDA0002342540200000741
+ represents a compound of IC50Greater than 1 μ M
+ represents the IC of the compound50Is 0.3 to 1 μ M
+ + + + + denotes IC of the Compound50Less than 0.3. mu.M.
As can be seen from the data in tables 2 and 3, the antiproliferative activity of the compounds of the invention on H358 cells was mostly less than 0.3. mu.M, R5Is a spiro substituent or 2 position (R) of acrylamide4Substituent) with a smaller volume of F atom, the compounds all have strong K-RAS G12C inhibitory activity.
Example 158 in vivo pharmacokinetic evaluation in mice
The compound was administered intravenously at 2mg/kg and orally at 10mg/kg (0.5% CMC-Na suspension), with 15 ICR mice per group and males per mouse, 3 discrete time points per mouse were collected, 3 mice per time point. Sampling time points were pre-dose, 5min, 15min, 30min, 1h,3h, 5h, 8h, 12h and 24h post-dose, and the mice were bled at each time point for about 80 μ L from the orbital or heart blood. All whole blood samples were collected in EDTA K2The plasma was separated after centrifugation at 4 ℃ for 10min (1500-. And (3) measuring the concentration of the compound in the plasma by using a liquid chromatography-tandem mass spectrometry method, and solving corresponding pharmacokinetic parameters according to a plasma concentration-time curve.
TABLE 4 Compound mouse pharmacokinetic parameters
Figure BDA0002342540200000751
NA indicates that data is not available
As can be seen from the above table, the compounds all have good oral absorption characteristics, half-life (t)1/2) Maximum blood concentration (C)max) Area under the time curve (AUC)0-t) And oral bioavailability metabolic parameters, etc. were superior to control drug B (example 35 in WO 2018/143315). The good oral absorption property has important significance in the aspects of improving the drug effect of the drug, reducing the dosage, saving the cost and the like.
Example 159 evaluation of antitumor Activity in mice
Human pancreatic cancer Mia PaCa-2 cells were treated with 1640 containing 10% fetal bovine serum at 37 ℃ with 5% CO2And (4) performing conventional culture in an incubator, and collecting cells after passage until the cells reach the required amount. Will be 1 × 107Injecting Mia PaCa-2 cells into the left north of each nude mouse until the tumor grows to 150mm3Thereafter, animals were randomized into groups to begin dosing. Are respectively 1) solvent pairGroup, 8; 2) compound 1, compound 2, compound 5, compound 31 and compound 131 groups, 8 per group. The solvent control group was gavaged twice daily with 0.5% CMC-Na; compound 1, compound 2, compound 5, compound 31 and compound 131 groups were gavaged once daily with a 0.5% CMC-Na suspension. Tumor volumes were measured two and four weeks per week, mouse body weights were measured, and nude mice were sacrificed on day 21 of dosing, and the test results are shown in table 5 below.
TABLE 5 Experimental treatment of human pancreatic cancer Mia PaCa-2 nude mouse transplantable tumors with Compounds
Compound (I) Dosage (mg/kg) Dosing regimens Anti-tumor effect
1 10 qd*21 39% withdrawal
2 10 qd*21 23% withdrawal
5 10 qd*21 30% withdrawal
31 10 qd*21 32% retraction
131 10 qd*21 37% retraction
B 10 qd*21 25% withdrawal
As can be seen from the data in the above table, the compound of the present invention has a strong in vivo antitumor activity, and the compound 1, 5, 31 and 131 have stronger in vivo activity than the control drug B, because the compound of the present invention can regress the tumor after being continuously administered for 21 days at a dose of 10 mg/kg/day.

Claims (11)

1. A compound with a structure shown as a general formula (1) or isomers, crystal forms, pharmaceutically acceptable salts, hydrates or solvates thereof:
Figure FDA0002342540190000011
in formula (1):
m is an integer of 1 or 2;
n is an integer of 1 or 2;
R1is H, halogen, C1-C3 alkyl, C2-C4 alkenyl, C2-C4 alkynyl or C3-C6 cycloalkyl;
R2is C1-C3 alkoxy, halogeno C1-C3 alkoxy or-NRaRbWherein R isaAnd RbIndependently H, C1-C3 alkyl, halo-C1-C3 alkyl, or RaAnd RbWith the N atom forming a 4-to 7-membered hetero ringCycloalkyl, said heterocycloalkyl being substituted by 1 to 3 halogen atoms;
R3is composed of
Figure FDA0002342540190000012
Figure FDA0002342540190000021
Figure FDA0002342540190000022
Wherein R iscIs H or F, RdIs H, F, Cl or Me, ReIs H, F, Cl or Me, RfIs F, NH2Me or cyclopropyl, Rx1、Rx2、Rx3、Rx4、Rx5、Rx6And Rx7Independently H, F, Cl, OH, OMe, NH2、CF3C1-C3 alkyl or C3-C6 cycloalkyl;
R4is H, halogen, CN, C1-C3 alkyl, haloC 1-C3 alkyl or heteroaryl; and
when R is3Is composed of
Figure FDA0002342540190000023
And R is4When is H, R5Comprises the following steps:
Figure FDA0002342540190000024
Figure FDA0002342540190000025
wherein n is1、n2、n3、m1、m2And m3Independently is an integer of 1 or 2, RgIs C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3) alkoxy- (C2-C3) alkyl-, (halogenated C1-C3) alkoxy- (C2-C3) alkyl-, (C3-C6) cycloalkyl- (C1-C3) alkyl-, heterocycloalkyl- (C1-C3) alkyl-, halogenated C1-C3 alkyl or cyano-substituted C1-C3 alkyl, RhIs composed of
Figure FDA0002342540190000031
Figure FDA0002342540190000032
When R is3Is composed of
Figure FDA0002342540190000033
And R is4Halogen, CN, C1-C3 alkyl, halogenated C1-C3 alkyl or heteroaryl; or, when R is3Is composed of
Figure FDA0002342540190000034
Figure FDA0002342540190000035
Figure FDA0002342540190000036
When R is5Comprises the following steps:
Figure FDA0002342540190000037
Figure FDA0002342540190000038
Figure FDA0002342540190000041
Figure FDA0002342540190000042
wherein n is1、n2、n3、m1、m2And m3Independently is an integer of 1 or 2, RgIs C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3) alkoxy- (C2-C3) alkyl-, (halogenated C1-C3) alkoxy- (C2-C3) alkyl-, (C3-C6) cycloalkyl- (C1-C3) alkyl-, heterocycloalkyl- (C1-C3) alkyl-, halogenated C1-C3 alkyl or cyano-substituted C1-C3 alkyl, RhIs composed of
Figure FDA0002342540190000043
Figure FDA0002342540190000044
Figure FDA0002342540190000045
RiIs H, halogen, methyl or cyano.
2. The compound according to claim 1, wherein in the general formula (1), R is1H, F, Cl, Me, Et, isopropyl, vinyl, ethynyl or cyclopropyl.
3. The compound according to claim 1-2, wherein in the general formula (1), R is2Is CH3O-、CH3CH2O-、CF3CH2O-、CHF2CH2O-、
Figure FDA0002342540190000046
Figure FDA0002342540190000047
4. A compound according to claims 1 to 3, wherein in the general formula (1), R is3Is composed of
Figure FDA0002342540190000051
Figure FDA0002342540190000052
5. The compound according to claim 1 to 4, wherein in the general formula (1), R is4Is H, F, CN, Me, CF3
Figure FDA0002342540190000053
6. The compound according to claim 1 to 5, wherein in the general formula (1), when R is3Is composed of
Figure FDA0002342540190000061
Figure FDA0002342540190000062
And R is4When is H, R5Comprises the following steps:
Figure FDA0002342540190000063
Figure FDA0002342540190000071
Figure FDA0002342540190000081
7. the compound according to claim 1 to 5, wherein in the general formula (1), when R is3Is composed of
Figure FDA0002342540190000082
Figure FDA0002342540190000083
And R is4Is F, CN, Me, CF3
Figure FDA0002342540190000084
Figure FDA0002342540190000085
When the current is over; or, when R is3Is composed of
Figure FDA0002342540190000091
Figure FDA0002342540190000092
Figure FDA0002342540190000093
When R is5Comprises the following steps:
Figure FDA0002342540190000094
Figure FDA0002342540190000095
Figure FDA0002342540190000101
Figure FDA0002342540190000111
Figure FDA0002342540190000121
8. the compound of claims 1-7, or a pharmaceutically acceptable salt thereof, wherein the compound has one of the following structures:
Figure FDA0002342540190000131
Figure FDA0002342540190000141
Figure FDA0002342540190000151
Figure FDA0002342540190000161
Figure FDA0002342540190000171
Figure FDA0002342540190000181
Figure FDA0002342540190000191
Figure FDA0002342540190000201
Figure FDA0002342540190000211
Figure FDA0002342540190000221
Figure FDA0002342540190000231
9. a pharmaceutical composition for the treatment, regulation and/or prevention of diseases associated with the K-Ras G12C mutant protein, comprising a pharmaceutically acceptable excipient or carrier and, as an active ingredient, a compound of any one of claims 1-8, or an isomer, pharmaceutically acceptable salt, hydrate or solvate thereof.
10. A use of the compound of any one of claims 1-8, or its isomers, crystal forms, pharmaceutically acceptable salts, hydrates or solvates, characterized by being used in a method for treating a disorder mediated by a K-Ras G12C mutation in an individual in need thereof.
11. The method of claim 10 wherein the condition is cancer, and the cancer is hematological cancer and solid tumors.
CN201911386239.6A 2019-12-27 2019-12-27 Spiro-containing quinazoline compounds Withdrawn CN113045570A (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
CN201911386239.6A CN113045570A (en) 2019-12-27 2019-12-27 Spiro-containing quinazoline compounds
KR1020227023548A KR20220122662A (en) 2019-12-27 2020-12-25 Quinazoline Compounds Containing Spiro Rings
AU2020414943A AU2020414943A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound
US17/767,034 US20230002382A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compounds
MX2022006609A MX2022006609A (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound.
PCT/CN2020/139530 WO2021129820A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound
CN202080090796.8A CN114929704A (en) 2019-12-27 2020-12-25 Spiro-containing quinazoline compounds
JP2022539444A JP2023508482A (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compounds
CA3162106A CA3162106A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound
BR112022010267A BR112022010267A2 (en) 2019-12-27 2020-12-25 COMPOUND, PHARMACEUTICAL COMPOSITION, AND USE OF COMPOUNDS
EP20905322.2A EP4083042A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911386239.6A CN113045570A (en) 2019-12-27 2019-12-27 Spiro-containing quinazoline compounds

Publications (1)

Publication Number Publication Date
CN113045570A true CN113045570A (en) 2021-06-29

Family

ID=76507332

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911386239.6A Withdrawn CN113045570A (en) 2019-12-27 2019-12-27 Spiro-containing quinazoline compounds

Country Status (1)

Country Link
CN (1) CN113045570A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022083616A1 (en) * 2020-10-21 2022-04-28 贝达药业股份有限公司 Quinazoline compound and pharmaceutical composition thereof
CN115335379A (en) * 2020-03-25 2022-11-11 微境生物医药科技(上海)有限公司 Spiro-containing quinazoline compounds
WO2022247760A1 (en) * 2021-05-22 2022-12-01 上海科州药物研发有限公司 Heterocyclic compounds as kras inhibitor, and preparation therefor and use thereof in treatment

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115335379A (en) * 2020-03-25 2022-11-11 微境生物医药科技(上海)有限公司 Spiro-containing quinazoline compounds
WO2022083616A1 (en) * 2020-10-21 2022-04-28 贝达药业股份有限公司 Quinazoline compound and pharmaceutical composition thereof
WO2022247760A1 (en) * 2021-05-22 2022-12-01 上海科州药物研发有限公司 Heterocyclic compounds as kras inhibitor, and preparation therefor and use thereof in treatment

Similar Documents

Publication Publication Date Title
CN115335379B (en) Spirocyclic quinazoline compounds
WO2021129820A1 (en) Spiro ring-containing quinazoline compound
CN113767103B (en) Novel spirocyclic K-Ras G12C inhibitors
WO2021129824A1 (en) New-type k-ras g12c inhibitor
CN113544128B (en) KRAS-G12C inhibitors
EP4144732A1 (en) Benzothiazolyl biaryl compound, and preparation method and use
IL293962A (en) Kras mutant protein inhibitors
CA3213029A1 (en) Parp inhibitor containing piperazine structure, preparation method therefor and pharmaceutical use thereof
CN115315427B (en) HPK1 inhibitor and preparation method and application thereof
CN116390728B (en) Quinazoline derivative, preparation method and application thereof
CN113045570A (en) Spiro-containing quinazoline compounds
EP3428159A1 (en) Crystalline forms of mesylate salt of pyridinyl amino pyrimidine derivative, preparation methods therefor, and applications thereof
EP3418277B1 (en) Substituted amino six-membered nitric heterocyclic ring compound and preparation and use thereof
CN113754659A (en) Spiro-containing quinazoline compounds
CN111606890A (en) Acryloyl group-containing nuclear transport modulators and uses thereof
EP4159731A1 (en) New pyrazine compound
WO2023116895A1 (en) Polymorph of kras inhibitor, preparation method therefor, and use thereof
CN113880804A (en) Novel benzimidazole compounds
CN116783194A (en) Spirocyclic quinazoline derivatives
CN113943288A (en) 5, 6-dihydropyrazino [2,3-c ] isoquinoline compounds
TW202306955A (en) Naphthyridine derivative as ATR inhibitor and method for preparing same
CN116813646A (en) Substituted bridged ring inhibitors and preparation method and application thereof
CN117222649A (en) Pyrrolopyrimidine derivatives, process for their preparation and their use
CN117384162A (en) Selective HER2 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WW01 Invention patent application withdrawn after publication

Application publication date: 20210629

WW01 Invention patent application withdrawn after publication