CN113045565A - Novel K-Ras G12C inhibitors - Google Patents

Novel K-Ras G12C inhibitors Download PDF

Info

Publication number
CN113045565A
CN113045565A CN201911386644.8A CN201911386644A CN113045565A CN 113045565 A CN113045565 A CN 113045565A CN 201911386644 A CN201911386644 A CN 201911386644A CN 113045565 A CN113045565 A CN 113045565A
Authority
CN
China
Prior art keywords
alkyl
substituted
halogen
compound
alkoxy
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.)
Pending
Application number
CN201911386644.8A
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 CN201911386644.8A priority Critical patent/CN113045565A/en
Priority to PCT/CN2020/139544 priority patent/WO2021129824A1/en
Priority to CN202080090785.XA priority patent/CN114901661A/en
Publication of CN113045565A publication Critical patent/CN113045565A/en
Pending 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
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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 various optical isomers, various crystal forms and pharmaceutically acceptable salts 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

Novel K-Ras G12C inhibitors
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a novel K-Ras G12C inhibitor, a preparation method thereof and a use method of the compound.
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; 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. The interaction of GTP enzyme activator proteins (GAPs) and Ras in cells greatly promotes the activity of Ras GTPase, so that the excessive activation of Ras proteins 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.
Mirati reports in patents WO2017/201161, WO2019/099524 and US2019/270743 that K-Ras G12C inhibitors with tetrahydropyridopyrimidine as a parent nucleus have the disadvantages of poor or unstable activity, such as poor stability of compound A (Example 7) in US 2019/270743. In addition, some compounds have low activity, for Example, compound B (Example 494) in WO2019/099524 has antiproliferative activity on NCI-H358 cells of more than 1 μ M.
Figure BDA0002342540580000021
Therefore, there is a need to research and discover K-Ras G12C inhibitors with high activity and good stability.
Disclosure of Invention
The invention aims to provide a compound with a structural general formula shown in formula (1), or optical isomers, crystal forms, pharmaceutically acceptable salts, hydrates or solvates thereof:
Figure BDA0002342540580000031
in formula (1):
y is a bond or C1-C6 alkylene;
R1is aryl or heteroaryl, which may be substituted with 1to 3 of the following groups: halogenA hydroxy group, an amino group, a C1-C3 alkyl group, a C2-C4 alkenyl group, a C3-C6 cycloalkyl group, a C1-C3 alkoxy group, a halogen-substituted C1-C3 alkyl group, or a halogen-substituted C1-C3 alkoxy group, and when substituted with a plurality of substituents, the substituents may be the same or different;
R2is aminoalkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, each of which may be substituted with 1to 3 of the following groups: H. halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy, C3-C6 cycloalkyl, - (C1-C3 alkyl) -cyano, - (C1-C3 alkyl) - (C1-C3 alkoxy), - (C1-C3 alkyl) - (C3-C6 cycloalkyl), deuterated C1-C3 alkyl, halogen-substituted C1-C3 alkyl, or halogen-substituted C1-C3 alkoxy, which when substituted with multiple substituents, may be the same or different.
Q is
Figure BDA0002342540580000032
Wherein R is3And R4Independently H, D, halogen or C1-C3 alkyl;
Figure BDA0002342540580000033
is a 4-7 membered saturated heterocycloalkyl, partially saturated heterocycloalkyl or heteroaryl group having at least one N atom, which may be substituted with 1-3 of the following groups: H. d, halogen,
Figure BDA0002342540580000034
CN、NMe2、NEt2SMe, C1-C3 alkyl, C3-C6 cycloalkyl, -C (O) O- (C1-C3 alkyl), -C (O) NH- (C1-C3 alkyl), -C (O) NMe2、-C(O)NEt2- (C1-C3 alkyl) -NMe2- (C1-C3 alkyl) -NEt2Halogen-substituted C1-C3 alkyl or cyano-substituted C1-C3 alkyl, which when substituted with a plurality of substituents may be the same or different;
R5is C3-C6 cycloalkyl, - (C1-C3 alkyl) -cyano, - (C1-C3 alkyl) -hydroxy, - (C1-C3 alkyl) - (C3-C6 cycloalkyl), - (C1-C3 alkane- (C1-C3 alkoxy), - (C1-C3 alkyl) - (halogen-substituted C1-C3 alkoxy), heterocycloalkyl, partially saturated heterocycloalkyl, heteroaryl, - (C1-C3 alkyl) -heterocycloalkyl, - (C1-C3 alkyl) -heteroaryl or- (C1-C3 alkyl) -NR6R7The heterocycloalkyl, partially saturated heterocycloalkyl, heteroaryl, - (C1-C3 alkyl) -heterocycloalkyl or- (C1-C3 alkyl) -heteroaryl may be substituted with 1to 3 of the following groups: halogen, OH,
Figure BDA0002342540580000041
CN、NMe2、NEt2-C (O) O- (C1-C3 alkyl), -C (O) NH- (C1-C3 alkyl), -C (O) NMe2、-C(O)NEt2C1-C3 alkyl, C3-C6 cycloalkyl, halogen-substituted C1-C3 alkyl or cyano-substituted C1-C3 alkyl, which when substituted with multiple substituents may be the same or different, wherein R is6And R7Independently H, C1-C3 alkyl, C3-C6 cycloalkyl, or halogen substituted C1-C3 alkyl.
In another preferred embodiment, wherein in the general formula (1), Y is a bond, -CH2-, -CH (Me) -or-CH2CH2-。
In another preferred embodiment, wherein in said general formula (1), R1Comprises the following steps:
Figure BDA0002342540580000042
Figure BDA0002342540580000043
wherein R isaAnd RbIndependently H, halogen, hydroxyl, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl or halogen substituted C1-C3 alkoxy.
In another preferred embodiment, wherein in said general formula (1), R2Comprises the following steps:
Figure BDA0002342540580000044
Figure BDA0002342540580000045
wherein n is 1,2 or3,RcAnd RdIndependently H, halogen, CN, OH, C1-C3 alkyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl or halogen substituted C1-C3 alkoxy, ReIs C1-C3 alkyl, C3-C6 cycloalkyl, deuterated C1-C3 alkyl, - (C1-C3 alkyl) -cyano, - (C1-C3 alkyl) - (C1-C3 alkoxy), - (C1-C3 alkyl) - (C3-C6 cycloalkyl), deuterated C1-C3 alkyl, halogen-substituted C1-C3 alkyl or halogen-substituted C1-C3 alkoxy, and when substituted by a plurality of substituents, the substituents may be the same or different.
In another preferred embodiment, wherein in said general formula (1), Q is
Figure BDA0002342540580000051
Wherein R is3And R4Independently is H or D;
Figure BDA0002342540580000052
is composed of
Figure BDA0002342540580000053
Figure BDA0002342540580000054
Figure BDA0002342540580000061
Figure BDA0002342540580000071
In another preferred embodiment, wherein in said general formula (1), Q is
Figure BDA0002342540580000072
Wherein R is5Is composed of
Figure BDA0002342540580000073
Figure BDA0002342540580000081
In various embodiments, representative compounds of the invention have one of the following structures:
Figure BDA0002342540580000091
Figure BDA0002342540580000101
Figure BDA0002342540580000111
Figure BDA0002342540580000121
Figure BDA0002342540580000131
Figure BDA0002342540580000141
Figure BDA0002342540580000151
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 each of its optical isomers, a pharmaceutically acceptable inorganic or organic salt.
Still another object of the present invention is to provide the use of the above-mentioned compound of the present invention, or each optical isomer, pharmaceutically acceptable inorganic or organic salt thereof, for the preparation of a medicament for treating RAS-related diseases.
Through synthesis and careful study of various novel compounds with K-RAS G12C inhibiting effect, the inventors found that in the compounds of formula (1), when Q is enamine structure
Figure BDA0002342540580000152
In which
Figure BDA0002342540580000153
When the compound is heteroaryl or substituted heteroaryl, the K-RAS G12C inhibitory activity of the compound is greatly improved, and the compound has good stability, while the compound in which the aryl group is connected by a carbon-carbon single bond and acrylamide in the patent WO2019/099524 has poor activity, such as that the antiproliferative activity of the compound B on NCI-H358 cells is more than 1 mu M; in addition to this, the present invention is,
Figure BDA0002342540580000154
when the compound is heterocyclic alkyl or lacks electron-withdrawing groups such as CO beside an N atom, the stability of the compound is poor. On the other hand, the present inventors have found 2 (R) of acrylamide5Group) with a bulky or substituted heteroaryl group, the compounds also have good K-RAS G12C inhibitory activity and compound stability, whereas the 2-position of acrylamide in patent US2019/270743 with a smaller group has poor compound stability, such as compound a listed above.
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, e.g.,but are not limited to, Aldrich Chemical co. (Milwaukee, Wis.) or Sigma Chemical co. (st. 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 CHEMISTRY4thEd., (Wiley 1992); carey and Sundberg, ADVANCED ORGANIC CHEMISTRY 4thEd, Vols.A and B (Plenum2000, 2001), Green and Wuts, PROTECTIVE GROUPSIN 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 is prepared by the following general reaction scheme 1, scheme 2 or scheme 3:
general reaction scheme 1
Figure BDA0002342540580000171
Embodiments of the compounds of formula (1) may be prepared according to general reaction scheme 1 (method A), wherein R1、R2Y and Q are as defined above, PG and PG1Represents different protecting groups of amine groups, and X represents Br, I, trifluoromethanesulfonate, boric acid, borate or potassium fluoroborate. As shown in general reaction scheme 1, the raw material A1 (synthesized in reference patent WO 2017/201161) undergoes oxidation reaction to obtain compound A2, compound A2 and compound R2-Y-OH under basic conditionsThe reaction is carried out to obtain a compound A3, the compound A3 removes the protecting group PG (e.g., Boc) to obtain a compound A4, a compound A4 and R1Coupling with-X to give compound A5, Compound A5 deprotecting the protecting group PG1(e.g., Cbz) to give compound A6, and condensation reaction of compound A6 with Q-COOH gave the title compound A7.
General reaction scheme 2
Figure BDA0002342540580000181
Embodiments of the compounds of formula (1) may be prepared according to general reaction scheme 2 (method B), wherein R1、R2Y and R5W represents Br or I, K represents boric acid, boric acid ester or potassium fluoroborate, R is as defined above5In (2) directly bonded to K is a carbon atom. As shown in general reaction scheme 2, condensation reaction of intermediate A6 and compound B1 gives compound B2, compound B2 and R5The Suzuki coupling reaction of-K gives the end product B3.
General reaction scheme 3
Figure BDA0002342540580000182
Embodiments of the compounds of formula (1) may be prepared according to general reaction scheme 3 (method C), wherein R1、R2Y and R5W represents Br or I, R, as defined hereinbefore5Directly attached to H in (b) is a nitrogen atom. Intermediate B2 and Compound R as shown in general reaction scheme 25And carrying out Ullmann coupling reaction on the-H to obtain a final product C1.
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) have one or more stereogenic centers and thus occur as racemates, racemic mixtures, single enantiomers, diastereomeric compounds and single 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.
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.
"alkyl" refers to a saturated aliphatic hydrocarbon group, including straight and branched chain groups of 1to 6 carbon atoms. Lower alkyl groups having 1to 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。
"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.
"alkoxy" means a linkage to the rest of the molecule through an ether oxygen atomA partial alkyl group. Representative of alkoxy groups are alkoxy groups having 1to 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。
"aryl" refers to a group having at least one aromatic ring structure, i.e., a carbocyclic aryl group having a conjugated pi-electron system, such as a benzene ring and a naphthalene ring.
"heteroaryl" refers to an aromatic group containing one or more heteroatoms (O, S or N), the heteroaryl group being monocyclic or polycyclic, e.g., a monocyclic heteroaryl ring fused to one or more carbocyclic aromatic groups or other monocyclic heterocyclic 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.
"heterocycloalkyl" refers to a saturated or partially unsaturated ring system radical containing one or more heteroatoms (O, S or N) in which 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.
"halo" (or halo) means fluoro, chloro, bromo, or iodo. The term "halogen substituted" (or "halo") as appearing before the radical name means that the radical is partially or fully halo, that is, substituted in any combination by F, Cl, Br or I, preferably by F or Cl.
The term "bond" or "single bond" refers to a chemical bond between two atoms or between two moieties when the atoms connected by the bond are considered part of a larger structure. In one aspect, when a group described herein is a bond, the absence of a reference group allows for the formation of a bond between the remaining defined groups.
The term "membered ring" includes any cyclic structure. The term "element" is intended to mean the number of backbone atoms constituting a ring. Thus, for example, cyclohexyl, pyridyl, pyranyl, thiopyranyl are six-membered rings and cyclopentyl, pyrrolyl, furanyl and thienyl are five-membered rings.
The term "fragment" refers to a specific part or functional group of a molecule. Chemical moieties are generally considered to be chemical entities contained in or attached to a molecule.
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 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 the treatment of cancer, the method comprising administering to an individual in need thereof an effective amount of a pharmaceutical composition of any of the foregoing compounds of protective structure (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 pharmaceutically acceptable excipients or carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate etc.), gelatin, talc, solid lubricants (e.g. stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g. soybean oil, sesame oil, peanut oil, olive oil etc.), polyols (e.g. propylene glycol, glycerol, mannitol, sorbitol etc.), emulsifiers (e.g. propylene glycol, glycerol, mannitol, sorbitol etc.)
Figure BDA0002342540580000231
) 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 1to 2000mg, preferably 50 to 100 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: ar represents argon; BFMO represents N1, N2-bis (furan-2-ylmethyl) oxyalkanamide; CDCl3Represents deuterated chloroform; CH (CH)3CN represents acetonitrile; cs2CO3Represents cesium carbonate; CuI represents cuprous iodide; DCM represents dichloromethane; DIPEA stands for diisopropylethylamine; dioxane represents 1, 4-Dioxane; DMF represents dimethylformamide; DMSO represents divaricate knotweed herb; EA represents ethyl acetate; EtOH stands for ethanol; h represents hour; NaOH represents sodium hydroxide; KOAc represents potassium acetate; LC-MS stands for liquid-mass spectrometry; m-CPBA represents m-chloroperoxybenzoic acid; MeOH represents methanol; min represents min; MS represents mass spectrum; na (Na)2SO4Represents sodium sulfate; NMR stands for nuclear magnetic resonance; pd2(dba)3Represents tris (dibenzylideneacetone) dipalladium; pd (dppf) Cl2Represents 1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride; PE represents petroleum ether; ruphos stands for 2-bicyclohexylphosphine-2 ',6' -diisopropyloxybiphenyl;t-BuOK for potassium tert-butoxide; T3P represents 1-propylphosphoric anhydride; TFA (CF)3COOH) represents trifluoroacetic acid; THF represents tetrahydrofuran; toluene stands for Toluene.
The specific implementation mode is as follows:
preparation example Synthesis of- ((S) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile (A-6a)
Figure BDA0002342540580000261
(S) -4- (4- ((benzyloxy) carbonyl) -3- (cyanomethyl) piperazin-1-yl) -2- (methylsulfonyl) -5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester (A-2)
A-1(72.5g,134.6mmol) and DCM (725mL) were added to a 1L single-neck flask, cooled to 0-5 ℃ in an ice-salt bath under the protection of Ar, and m-CPBA (81.3g,471.1mmol) was added and the reaction was continued for 2 hours. After the completion of the reaction was monitored by LC-MS, the mixture was filtered, the filter cake was washed with DCM (50 mL. times.3), a saturated sodium bicarbonate solution (200mL) and a saturated sodium thiosulfate solution (300mL) were added to the filtrate, stirred at room temperature for 30min, the organic phase was separated, washed successively with a saturated sodium thiosulfate solution (200 mL. times.2) and a saturated sodium chloride solution (200mL), anhydrous Na2SO4Drying, filtering, and concentrating the filtrate to obtain crude extractProduct A-2(80g, 100% yield), ESI-MS M/z 571.3[ M + H ]]+
4- ((S) -4- ((benzyloxy) carbonyl) -3- (cyanomethyl) piperazin-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidine-7 (6H) -carboxylic acid tert-butyl ester (A-3a)
A-2(80g,134.6mmol), THF (600mL) and (S) - (1-methylpyrrolidin-2-yl) methanol (31g,269.2mmol) were added to a 1L single-neck flask, cooled to 0-5 ℃ in an ice-salt bath under Ar protection, then sodium tert-butoxide (32.3g,336.5mmol) was added and the reaction was continued for 2 h. After TLC and LC-MS to monitor the reaction was complete, the system was quenched with EA (300mL), saturated ammonium chloride solution (200mL), stirred, separated, the aqueous phase was extracted with EA (200mL), the organic phases were combined and washed with saturated sodium chloride solution (200 mL. times.2), concentrated, and the residue was purified by column chromatography (DCM/MeOH. 50/1to 30/1) to give product A-3a as a yellowish brown oil (60.5g, 74% yield), ESI-MS M/z:606.3[ M + H/M ])]+
(S) -benzyl 2- (cyanomethyl) -4- (2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazine-1-carboxylate (A-4a)
Adding A-3a (60.5g,99.88mmol) and DCM (150mL) into a 1L single-neck bottle, cooling the mixed solution to 0-5 ℃ by using an ice salt bath under the protection of Ar, then adding TFA (228g,2mol), stirring and reacting for 2h at r.t., monitoring the reaction by LC-MS, concentrating the system, adding DCM (300mL) into the residue, adjusting the pH to 8-9 by using a saturated sodium carbonate solution, stirring, separating liquid, extracting the aqueous phase by using DCM (200mL 5), combining the organic phases, and using anhydrous Na for the organic phase2SO4Drying, filtering, concentrating the filtrate to obtain yellow foamy solid product A-4a (54.2g, yield 107%), ESI-MS M/z 506.3[ M + H ]]+
(S) -benzyl 4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -2- (cyanomethyl) piperazine-1-carboxylate (A-5a)
A1L single-neck bottle is charged with A-4a (45.5g,90.1mmol), 1-bromo-8-chloronaphthalene (32.6g,135mmol), cesium carbonate (73.3g,225mmol), Ruphos (8.4g,18mmol) and toluene (800mL), and after replacement protection of the mixed solution with Ar, Pd is added2(dba)3(16.5g,18mmol), heating to 90 ℃ under the protection of Ar, and stirring for reactionAnd 6 h. After the completion of the reaction was monitored by LC-MS, the system was filtered while hot, the filter cake was washed with DCM (100mL), the filtrate was concentrated and purified by column chromatography (DCM/MeOH 50/1to 20/1) to give the product A-5a as a yellowish brown foamy solid (36.6g, yield: 61%), ESI-MS M/z:666.4[ M + H ]: 666.4]+
2- ((S) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile (A-6a)
A-5a (40.9g,61.39mmol), THF (100mL), MeOH (600mL), and 10% Pd/C (5g, wet% ═ 50%), H, were added to a 1L single-neck flask2After three times of replacement, the mixture is stirred vigorously at room temperature and normal pressure for reaction for 20 hours. After the LC-MS monitoring reaction is completed, the system is added with ammonia methanol solution (20mL,7M), stirred for 15min and then filtered by diatomite, the filtrate is concentrated to dryness to obtain a yellow-brown foamy solid product A-6a (31.3g, yield: 95%), ESI-MS M/z:532.3[ M + H ], (yield: 95%) (]+
Preparation examples 2-10 Synthesis of intermediates A-6b to A-6j
Different raw materials are utilized, and a synthesis method of the intermediate A-6a is adopted to obtain the target intermediates A-6b to A-6 j.
TABLE 1
Figure BDA0002342540580000281
Figure BDA0002342540580000291
Preparation example Synthesis of- ((S) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile (A-6a)
Figure BDA0002342540580000301
2- ((S) -1- (2-bromoacryloyl) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile (B-2a)
A-6a (10.64g,20.0mmol) and 2-bromoacrylic acid (B-1, 9.06g,60mmol), DIPEA (25.8g,200mmol) and DMF (200mL) are added into a 100mL single-neck bottle, and T is slowly dropped at room temperature under the protection of mixed liquid Ar3P (50.9g, 50% in DMF,80mmol) was added dropwise, the mixture was stirred at room temperature for 1H and quenched with water (200mL), EA (200 mL. times.2) was extracted, the organic phases were combined, concentrated, and the residue was purified by column chromatography (DCM/MeOH 50/1to 20/1 to give a light brown solid (5.45g, 41% yield), ESI-MS M/z 664.2/666.2[ M + H ] M + H]+
Preparation examples 12-20 Synthesis of intermediates B-2B to B-2j
Different raw materials are utilized, and the target intermediates B-2B to B-2j are obtained by adopting a synthesis method of the intermediate B-2 a.
TABLE 2
Figure BDA0002342540580000302
Figure BDA0002342540580000311
Figure BDA0002342540580000321
Example Synthesis of- ((S) -1- ((E) -3- (1H-1,2, 4-triazol-1-yl) acryloyl) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile (Compound 1)
Figure BDA0002342540580000322
Compound 1 was prepared according to method a as described below:
2- ((S) -1- ((E) -3- (1H-1,2, 4-triazol-1-yl) acryloyl) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
A100 mL single-neck flask was charged with A-6a (369mg,0.693mmol), (E) -3- (1H-1,2, 4-triazol-1-yl) acrylic acid (145mg,1.04mmol), DIPEA (269mg,2.08mmol) and DMF (10mL), and the mixture was added dropwise with T under Ar at room temperature3P (662mg, 50% in DMF,1.04mmol), stirred at RT for 1h and quenched with water, EA (10 mL. times.2) extracted, the organic phases combined, concentrated and the residue pre-TLC (DCM/MeOH/NH)3.H2O-500/20/1) to give compound 1(205mg, 45% yield) as a pale yellow solid.
1H NMR(400MHz,CDCl3)δ:8.32(s,1H),8.08(d,J=14.0Hz,2H),7.73(d,J=8.1Hz,1H),7.60(t,J=7.2Hz,1H),7.50(d,J=7.5Hz,1H),7.41(dd,J=11.5,7.9Hz,1H),7.31(t,J=7.8Hz,1H),7.24-7.16(m,1H),7.12(d,J=9.6Hz,1H),5.14(s,1H),4.52-4.34(m,2H),4.15(dt,J=35.4,22.5Hz,4H),3.90-3.76(m,1H),3.57(s,1H),3.45(d,J=13.9Hz,1H),3.13(td,J=42.7,39.9,21.0Hz,5H),2.86-2.69(m,2H),2.55(dd,J=28.8,9.7Hz,4H),2.33(s,1H),2.06(s,1H),1.77(s,4H);ESI-MS m/z:653.4[M+H]+.
Example 2-example 85 Synthesis of Compound 2-Compound 85
Compound 2-compound 85 can be obtained by using intermediates a-6a to a-6j as starting materials and using synthesis method a, similar to the synthesis method of compound 1.
TABLE 3
Figure BDA0002342540580000331
Figure BDA0002342540580000341
Figure BDA0002342540580000351
Figure BDA0002342540580000361
Figure BDA0002342540580000371
Figure BDA0002342540580000381
Figure BDA0002342540580000391
Figure BDA0002342540580000401
Figure BDA0002342540580000411
Figure BDA0002342540580000421
Figure BDA0002342540580000431
Figure BDA0002342540580000441
Figure BDA0002342540580000451
Figure BDA0002342540580000461
Figure BDA0002342540580000471
Figure BDA0002342540580000481
Figure BDA0002342540580000491
Figure BDA0002342540580000501
Figure BDA0002342540580000511
Figure BDA0002342540580000521
Example Synthesis of 862- ((S) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -1- (2-phenylpropenoyl) piperazin-2-yl) acetonitrile (Compound 86)
Figure BDA0002342540580000522
Compound 86 was prepared according to method B as described below:
2- ((S) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -1- (2-phenylpropenoyl) piperazin-2-yl) acetonitrile
A50 mL single neck flask was charged with B-2a (50mg,0.075mmol), phenylboronic acid (18mg,0.15mmol), KOAc (19mg,0.192mmol), Dioxane (5mL) and H2O (1mL), Ar replacement protected, Pd (dppf) Cl2(10mg), rapidly heating the mixed solution to reflux under the protection of Ar, stirring for reacting for 20min, adding water (5mL) and EA (10mL) into the system, stirring, separating the solution,the organic phase was concentrated and the residue was pre-TLC (DCM/MeOH/NH)3.H2O-500/20/1) to give the product (8mg, 16% yield).
1H NMR(400MHz,CDCl3)δ:7.75(dt,J=8.1,1.6Hz,1H),7.61-7.52(m,1H),7.45-7.31(m,7H),7.22(dt,J=7.5,1.5Hz,1H),7.17-7.02(m,1H),6.03(s,1H),5.82(s,1H),5.04(m,1H),4.44-4.24(m,2H),4.15-4.05(m,2H),4.00-3.71(m,3H),3.52(d,J=11.3Hz,2H),3.43-3.33(m,1H),3.26-2.76(m,7H),2.63(q,J=6.5,5.9Hz,1H),2.54(d,J=14.1Hz,1H),2.42(s,3H),2.22(dt,J=9.8,7.3Hz,1H),2.03(d,J=11.9Hz,1H),1.89-1.75(m,2H);ESI-MS m/z:663.2[M+H]+.
EXAMPLE 87 EXAMPLE 127 Compound 87 Synthesis of Compound 127
Using intermediates B-2a to B-2j as starting materials, compound 87-compound 127 can be obtained by synthesis method B, which is similar to the synthesis method of compound 86.
TABLE 4
Figure BDA0002342540580000531
Figure BDA0002342540580000541
Figure BDA0002342540580000551
Figure BDA0002342540580000561
Figure BDA0002342540580000571
Figure BDA0002342540580000581
Figure BDA0002342540580000591
Figure BDA0002342540580000601
Figure BDA0002342540580000611
Figure BDA0002342540580000621
Example Synthesis of 1282- ((S) -1- (2- (1H-imidazol-1-yl) acryloyl) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile (Compound 128)
Figure BDA0002342540580000622
Compound 128 was prepared according to procedure C as described below:
2- ((S) -1- (2- (1H-imidazol-1-yl) acryloyl) -4- (7- (8-chloronaphthalen-1-yl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
A10 mL single-necked flask was charged with B-2a (25mg,0.038mmol), imidazole (4mg,0.056mmol), CuI (2mg,0.008mmol), Cs2CO3(33mg,0.098mmol), DMSO (2mL) and BFMO (2mg,0.008mmol), the mixture was heated to 80 ℃ under Ar protection and stirred for 1 h. After MS monitoring that the basic reaction of the raw materials is complete, EA (10mL) and H are added to the mixture2O (10mL), stirred, separated, the organic phase concentrated and the residue subjected to pre-TLC (DCM/MeOH/NH)3.H2O-500/20/1) to give compound 128(5mg, 20% yield).
1H NMR(400MHz,DMSO-d6)δ:8.22(s,1H),8.10-8.03(m,1H),7.93(d,J=7.6Hz,1H),7.87(s,1H),7.78-7.72(m,1H),7.57(dd,J=18.4,7.7Hz,2H),7.49-7.42(m,1H),7.38(s,2H),7.10(s,1H),5.33(t,J=4.7Hz,1H),5.03(s,2H),4.66(s,1H),4.42(s,1H),4.34-4.12(m,4H),4.06(s,3H),3.22-3.00(m,5H),2.94(s,3H),2.69-2.66(m,1H),2.37-2.32(m,3H),2.05-1.90(m,3H);ESI-MS m/z:652.3[M+H]+.
EXAMPLE 129 Synthesis of example 148 Compound 129 Compound 148
Compound 129-compound 148 can be obtained using intermediates B-2a to B-2j as starting materials, using synthesis method C, analogous to the synthesis method of compound 128.
TABLE 5
Figure BDA0002342540580000631
Figure BDA0002342540580000641
Figure BDA0002342540580000651
Figure BDA0002342540580000661
Figure BDA0002342540580000671
Figure BDA0002342540580000681
EXAMPLE 149 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 action of the compound, after lysis of the cells, 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 6 below.
TABLE 6 inhibitory Activity of Compounds of the invention against pERK levels in H358 cells
Figure BDA0002342540580000682
Figure BDA0002342540580000691
+ 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%.
EXAMPLE 150 antiproliferative Activity of Compounds 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 7 below.
TABLE 7 antiproliferative activity of the compounds of the invention on H358 cells
Figure BDA0002342540580000701
Figure BDA0002342540580000711
+ represents a compound of IC50Greater than 1 μ M
+ represents the IC of the compound50Is 0.3 to 1. mu.M
+ + + + + denotes IC of the Compound50Less than 0.3. mu.M.
As is apparent from the activity data in tables 7 and 8 above, in the compound of the general formula (1), when Q is an enamine structure
Figure BDA0002342540580000721
In which
Figure BDA0002342540580000722
In the case of heteroaryl or substituted heteroaryl, the K-RAS G12C inhibitory activity of the compound is greatly improved, while the compound in which the heteroaryl is connected with acrylamide through a carbon-carbon single bond in the patent WO2019/099524 has poor activity, such as the antiproliferative activity of the compound B on NCI-H358 cells is more than 1 mu M. The carbon-nitrogen single bond is presumed to have stronger dipole moment than the carbon-carbon single bond, and the nitrogen atom loses partial electrons, which is favorable for stabilizing the intermediate state of the cysteine of the K-RAS protein after the acrylamide addition, so that the compound has strong activity. On the other hand, when the compound of the present invention has a bulky heteroaryl or substituted heteroaryl substituent at the 2-position of acrylamide, such as imidazolyl or pyrimidinyl, the compound also has excellent K-RAS G12C inhibitory activity, presumably because the N atom on the heteroaryl group facilitates binding to K-RAS protein.
Stability testing of the Compounds of example 151
Accurately weighing two parts (2-3 mg each) of the test compound, storing one part at the temperature of-20 ℃ in a refrigerator, placing one part in a room temperature, dissolving the sample in a 50mL volumetric flask after 72 hours, adding a proper amount of methanol, adding water to a constant volume to reach a scale, and shaking up to obtain the solution to be tested. The HPLC was used to measure the area of the peak of the liquid phase for both solutions and to determine the area of the peak of the liquid phase per unit concentration of the sample placed at room temperature divided by the area of the peak of the unit concentration of the sample stored at-20 ℃ in the refrigerator, the percentage obtained representing the residual amount of the compound after 72 hours of room temperature placement.
TABLE 8 stability of the Compounds after 72 hours at Room temperature
Figure BDA0002342540580000723
Figure BDA0002342540580000731
As shown by the stability data of Compound A in Table 9, the poor stability of the compound is presumed to be caused by intermolecular polymerization of the basic nitrogen atom in the molecule and the double bond of acrylamide. Compared with the contrast medicament A, the compound of the invention carries out the substitution of a group with larger volume on acrylamide, so that the stability of the compound of the invention is greatly improved, which has very important significance for the process synthesis, quality control and pharmacy of the compound.
Example 152 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 × 107A Mia PaCa-2 cell was injected into the left dorsal side of each nude mouse, and the tumor was allowed to grow to 150mm3Thereafter, animals were randomized into groups to begin dosing. 1) solvent control group, 8 respectively; 2) compound 1, compound 5, compound 70, compound 92, compound 97 and compound 121 groups, 8 per group. The solvent control group was intragastrically administered with 0.5% CMC-Na once a day; compound 1, compound 5, compound 70, compound 92, compound 97 and compound 121 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 9 below.
TABLE 9 Experimental therapeutic Effect of Compounds on human pancreatic cancer Mia PaCa-2 nude mouse transplantable tumors
Compound (I) Dosage (mg/kg) Dosing regimens Anti-tumor effect
1 10 qd*21 67% inhibition
5 10 qd*21 63% inhibition
70 10 qd*21 77% inhibition
92 10 qd*21 72% inhibition
121 10 qd*21 Inhibition of 76%
A 10 qd*21 58% inhibition
Compared with a contrast medicament A, the compound has stronger in-vitro activity and compound stability, and meanwhile, an in-vivo anti-tumor test also shows that the compound has better in-vivo activity, so that the compound has better drug effect and drug forming property in clinical application.

Claims (10)

1. A compound with a structure shown as a general formula (1) or each optical isomer, each crystal form, pharmaceutically acceptable salt, hydrate or solvate thereof:
Figure RE-FDA0002424850450000011
in formula (1):
y is a bond or C1-C6 alkylene;
R1is aryl or heteroaryl, which may be substituted with 1to 3 of the following groups: halogen, hydroxy, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen-substituted C1-C3 alkyl or halogen-substituted C1-C3 alkoxy, and when substituted with a plurality of substituents, the substituents may be the same or different;
R2is aminoalkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, each of which may be substituted with 1to 3 of the following groups: H. halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy, C3-C6 cycloalkyl, - (C1-C3 alkyl) -cyano, - (C1-C3 alkyl) - (C1-C3 alkoxy), - (C1-C3 alkyl) - (C3-C6 cycloalkyl), deuterated C1-C3 alkyl, halogen-substituted C1-C3 alkyl, or halogen-substituted C1-C3 alkoxy, which when substituted with multiple substituents, may be the same or different.
Q is
Figure RE-FDA0002424850450000012
Wherein R is3And R4Independently H, D, halogen or C1-C3 alkyl;
Figure RE-FDA0002424850450000013
is a 4-7 membered saturated heterocycloalkyl, partially saturated heterocycloalkyl or heteroaryl group having at least one N atom, which may be substituted with 1-3 of the following groups: H. d, halogen,
Figure RE-FDA0002424850450000014
CN、NMe2、NEt2SMe, C1-C3 alkyl, C3-C6 cycloalkyl, -C (O) O- (C1-C3 alkyl), -C (O) NH- (C1-C3 alkyl), -C (O) NMe2、-C(O)NEt2- (C1-C3 alkyl) -NMe2- (C1-C3 alkyl) -NEt2Halogen-substituted C1-C3 alkyl or cyano-substituted C1-C3 alkyl, which when substituted with a plurality of substituents may be the same or different;
R5is C3-C6 cycloalkyl, - (C1-C3 alkyl) -cyano, - (C1-C3 alkyl) -hydroxy, - (C1-C3 alkyl) - (C3-C6 cycloalkyl), - (C1-C3 alkyl) - (C1-C3 alkoxy), - (C1-C3 alkyl) - (halogen substituted C1-C3 alkoxy), heterocycloalkyl, partially saturated heterocycloalkyl, heteroaryl, - (C1-C3 alkyl) -heterocycloalkyl, - (C1-C3 alkyl) -heteroaryl or- (C1-C3 alkyl) -NR6R7The heterocycloalkyl, partially saturated heterocycloalkyl, heteroaryl, - (C1-C3 alkyl) -heterocycloalkyl or- (C1-C3 alkyl) -heteroaryl may be substituted with 1to 3 of the following groups: halogen, OH,
Figure RE-FDA0002424850450000021
CN、NMe2、NEt2-C (O) O- (C1-C3 alkyl), -C (O) NH- (C1-C3 alkyl), -C (O) NMe2、-C(O)NEt2C1-C3 alkyl, C3-C6 cycloalkyl, halogen-substituted C1-C3 alkyl or cyano-substituted C1-C3 alkyl, which when substituted with multiple substituents may be the same or different, wherein R is6And R7Independently H, C1-C3 alkyl, C3-C6 cycloalkyl, or halogen substituted C1-C3 alkyl.
2. The compound according to claim 1, wherein in the general formula (1), Y is a bond, -CH2-, -CH (Me) -or-CH2CH2-。
3. The compound according to claim 1-2, wherein in the general formula (1), R is1Comprises the following steps:
Figure RE-FDA0002424850450000022
Figure RE-FDA0002424850450000023
Figure RE-FDA0002424850450000024
wherein R isaAnd RbIndependently H, halogen, hydroxyl, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl or halogen substituted C1-C3 alkoxy.
4. A compound according to claims 1to 3, wherein in the general formula (1), R is2Comprises the following steps:
Figure RE-FDA0002424850450000025
Figure RE-FDA0002424850450000031
wherein n is 1,2 or 3, RcAnd RdIndependently H, halogen, CN, OH, C1-C3 alkyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl or halogen substituted C1-C3 alkoxy, ReIs C1-C3 alkyl, C3-C6 cycloalkyl, deuterated C1-C3 alkyl, - (C1-C3 alkyl) -cyano, - (C1-C3 alkyl) - (C1-C3 alkoxy), - (C1-C3 alkyl) - (C3-C6 cycloalkyl), deuterated C1-C3 alkyl, halogen-substituted C1-C3 alkyl or halogen-substituted C1-C3 alkoxy, and when substituted by a plurality of substituents, the substituents may be the same or different.
5. As claimed in claims 1-4Wherein in the general formula (1), Q is
Figure RE-FDA0002424850450000032
Wherein R is3And R4Independently is H or D;
Figure RE-FDA0002424850450000033
is composed of
Figure RE-FDA0002424850450000034
Figure RE-FDA0002424850450000035
Figure RE-FDA0002424850450000041
Figure RE-FDA0002424850450000051
6. The compound according to claim 1to 4, wherein in the general formula (1), Q is
Figure RE-FDA0002424850450000052
Wherein R is5Is composed of
Figure RE-FDA0002424850450000053
Figure RE-FDA0002424850450000061
7. The compound of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein the compound has one of the following structures:
Figure RE-FDA0002424850450000062
Figure RE-FDA0002424850450000071
Figure RE-FDA0002424850450000081
Figure RE-FDA0002424850450000091
Figure RE-FDA0002424850450000101
Figure RE-FDA0002424850450000111
Figure RE-FDA0002424850450000121
8. a pharmaceutical composition for the treatment, regulation and/or prevention of diseases associated with the K-Ras G12C mutant protein, which comprises a pharmaceutically acceptable excipient or carrier, and as an active ingredient, a compound according to any one of claims 1to 7, or each optical isomer, pharmaceutically acceptable salt, hydrate or solvate thereof.
9. Use of a compound according to any one of claims 1to 7, or each optical isomer, each crystalline form, a pharmaceutically acceptable salt, hydrate or solvate thereof, for the treatment of a disorder mediated by the K-Ras G12C mutation in a subject in need thereof.
10. The method of claim 9 wherein the condition is cancer, and the cancer is hematological cancer and solid tumors.
CN201911386644.8A 2019-12-27 2019-12-27 Novel K-Ras G12C inhibitors Pending CN113045565A (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN201911386644.8A CN113045565A (en) 2019-12-27 2019-12-27 Novel K-Ras G12C inhibitors
PCT/CN2020/139544 WO2021129824A1 (en) 2019-12-27 2020-12-25 New-type k-ras g12c inhibitor
CN202080090785.XA CN114901661A (en) 2019-12-27 2020-12-25 Novel K-Ras G12C inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911386644.8A CN113045565A (en) 2019-12-27 2019-12-27 Novel K-Ras G12C inhibitors

Publications (1)

Publication Number Publication Date
CN113045565A true CN113045565A (en) 2021-06-29

Family

ID=76507334

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201911386644.8A Pending CN113045565A (en) 2019-12-27 2019-12-27 Novel K-Ras G12C inhibitors
CN202080090785.XA Pending CN114901661A (en) 2019-12-27 2020-12-25 Novel K-Ras G12C inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202080090785.XA Pending CN114901661A (en) 2019-12-27 2020-12-25 Novel K-Ras G12C inhibitors

Country Status (2)

Country Link
CN (2) CN113045565A (en)
WO (1) WO2021129824A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11932633B2 (en) 2018-05-07 2024-03-19 Mirati Therapeutics, Inc. KRas G12C inhibitors
EP3908283A4 (en) 2019-01-10 2022-10-12 Mirati Therapeutics, Inc. Kras g12c inhibitors
EP4021444A4 (en) 2019-08-29 2023-01-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
CA3152025A1 (en) 2019-09-24 2021-04-01 David BRIERE Combination therapies
AU2020372881A1 (en) 2019-10-28 2022-06-09 Merck Sharp & Dohme Llc Small molecule inhibitors of KRAS G12C mutant
US11702418B2 (en) 2019-12-20 2023-07-18 Mirati Therapeutics, Inc. SOS1 inhibitors
KR20230081726A (en) 2020-09-03 2023-06-07 레볼루션 메디슨즈, 인크. Use of SOS1 inhibitors to treat malignancies with SHP2 mutations
CN117683049A (en) 2020-09-15 2024-03-12 锐新医药公司 Indole derivatives as RAS inhibitors for the treatment of cancer
US20230106174A1 (en) 2021-05-05 2023-04-06 Revolution Medicines, Inc. Ras inhibitors
KR20240017811A (en) 2021-05-05 2024-02-08 레볼루션 메디슨즈, 인크. RAS inhibitors for the treatment of cancer
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
CN117677398A (en) 2021-07-27 2024-03-08 东丽株式会社 Medicine for treating and/or preventing cancer
AR127308A1 (en) 2021-10-08 2024-01-10 Revolution Medicines Inc RAS INHIBITORS
TW202334138A (en) * 2021-11-05 2023-09-01 美商新領域醫藥公司 Methods for treatment of cancer
WO2023114954A1 (en) 2021-12-17 2023-06-22 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023150394A1 (en) * 2022-02-07 2023-08-10 Frontier Medicines Corporation Methods for treatment of cancer
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240189A1 (en) * 2022-06-10 2023-12-14 Bristol-Myers Squibb Company Tetrahydropyrido 3,4-d pyrimidine derivatives as kras inhibitors
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2904393A1 (en) * 2013-03-15 2014-09-25 Araxes Pharma Llc Covalent inhibitors of kras g12c
AU2017266911B2 (en) * 2016-05-18 2021-09-02 Array Biopharma, Inc. KRas G12C inhibitors
US10280172B2 (en) * 2016-09-29 2019-05-07 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10647715B2 (en) * 2017-11-15 2020-05-12 Mirati Therapeutics, Inc. KRas G12C inhibitors
UA125802C2 (en) * 2017-11-15 2022-06-08 Міраті Терапьютікс, Інк. Kras g12c inhibitors
AR114164A1 (en) * 2017-12-21 2020-07-29 Boehringer Ingelheim Int PYRIDOPYRIMIDINONES SUBSTITUTED WITH BENZYLAMINE AND DERIVATIVES AS INHIBITORS OF SOS1
BR112020020307A2 (en) * 2018-04-04 2021-01-12 Arvinas Operations, Inc. PROTEOLYSIS MODULATORS AND ASSOCIATED METHODS OF USE
CN110256421A (en) * 2019-06-26 2019-09-20 微境生物医药科技(上海)有限公司 KRAS-G12C inhibitor

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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

Also Published As

Publication number Publication date
WO2021129824A1 (en) 2021-07-01
CN114901661A (en) 2022-08-12

Similar Documents

Publication Publication Date Title
CN113045565A (en) Novel K-Ras G12C inhibitors
CN115335379B (en) Spirocyclic quinazoline compounds
CN113767103B (en) Novel spirocyclic K-Ras G12C inhibitors
EP4083042A1 (en) Spiro ring-containing quinazoline compound
CN113544128B (en) KRAS-G12C inhibitors
IL293962A (en) Kras mutant protein inhibitors
CN115315427B (en) HPK1 inhibitor and preparation method and application thereof
CN116390728B (en) Quinazoline derivative, preparation method and application thereof
WO2022199586A1 (en) Pyrimidopyridine inhibitor, preparation method therefor, and use thereof
CN113045570A (en) Spiro-containing quinazoline compounds
WO2021254384A1 (en) Novel pyrido[2,3-d]pyrimidine-7(8h)-one derivative
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
CN113754659A (en) Spiro-containing quinazoline compounds
WO2022174765A1 (en) Fused ring compound as wee-1 inhibitor
WO2022171088A1 (en) Pyrazolo[3,4-d]pyrimidin-3-one derivative
CN115515959A (en) Tricyclic heterocycles as TEAD binders
EP4159731A1 (en) New pyrazine compound
CN113880804A (en) Novel benzimidazole compounds
TW202306955A (en) Naphthyridine derivative as ATR inhibitor and method for preparing same
WO2022228511A1 (en) Fused ring compound as wee-1 inhibitor, and preparation method therefor and use thereof
CN116783194A (en) Spirocyclic quinazoline derivatives
WO2022171126A1 (en) Fused ring compound used as wee-1 inhibitor
WO2022262857A1 (en) Arylphosphine oxide compounds
WO2022012593A1 (en) 5,6-dihydropyrazino[2,3-c]isoquinoline compound
CN117658915A (en) Tri-ring compound and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210629

WD01 Invention patent application deemed withdrawn after publication