WO2022214102A1 - 作为kras g12d抑制剂的杂环化合物 - Google Patents

作为kras g12d抑制剂的杂环化合物 Download PDF

Info

Publication number
WO2022214102A1
WO2022214102A1 PCT/CN2022/086183 CN2022086183W WO2022214102A1 WO 2022214102 A1 WO2022214102 A1 WO 2022214102A1 CN 2022086183 W CN2022086183 W CN 2022086183W WO 2022214102 A1 WO2022214102 A1 WO 2022214102A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
group
formula
hydrogen
Prior art date
Application number
PCT/CN2022/086183
Other languages
English (en)
French (fr)
Inventor
张汉承
贾薇
蔡聪聪
Original Assignee
杭州英创医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 杭州英创医药科技有限公司 filed Critical 杭州英创医药科技有限公司
Priority to CN202280027547.3A priority Critical patent/CN117715915A/zh
Priority to CN202211215403.9A priority patent/CN116891487A/zh
Publication of WO2022214102A1 publication Critical patent/WO2022214102A1/zh
Priority to PCT/CN2023/087690 priority patent/WO2023198078A1/zh

Links

Classifications

    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the present invention relates to the field of medicinal chemistry; in particular, the present invention relates to a novel derivative containing a tricyclic heteroaryl group, its synthesis method and its use as a KRAS G12D inhibitor in the preparation of medicines for the treatment of tumors and other related application in disease.
  • KRAS Kerrsten Rat Sarcoma Viral Oncogene Homolog
  • HRAS Resten Rat Sarcoma Viral Oncogene Homolog
  • NRAS NRAS
  • KRAS KRAS protein plays an important role in the occurrence and development of cancer. 20%-30% of human tumors have RAS mutations, among which KRAS mutations have the highest frequency, which are more common in lung cancer, pancreatic cancer and colon cancer.
  • KRAS protein contains 188 amino acid residues and has multiple functional motifs, including nucleotide binding region, switch I region and switch II region, nucleotide exchange factor (GEF) binding region, GTPase activating protein (GAP) binding region regions and effector protein binding regions, etc.
  • GEF nucleotide exchange factor
  • GAP GTPase activating protein
  • KRAS is regulated by a variety of upstream signaling factors, such as receptor tyrosine kinases, integrins, G protein-coupled receptors, etc.
  • upstream signaling factors such as receptor tyrosine kinases, integrins, G protein-coupled receptors, etc.
  • KRAS exhibits an activated (on) or inactive (off) state through the transition of GTP or GDP binding states, respectively, and transmits signals received upstream to downstream effector proteins.
  • KRAS protein itself has relatively slow nucleotide exchange rate and weak GTP hydrolysis function. After receiving upstream stimulation signals, with the assistance of guanine nucleotide exchange factors such as SOS1 (Son of Sevenless 1), KRAS protein releases GDP and binds GTP. GTP-bound RAS protein is in an activated state, recruits downstream effector proteins, activates signaling pathways such as Raf-MEK-ERK and PI3K-AKT-mTOR, and promotes cell growth, proliferation, survival, metabolism, and angiogenesis.
  • SOS1 Non of Sevenless 1
  • GTPase activating protein GAP When the GTPase activating protein GAP binds to KRAS, it will significantly promote the GTP hydrolysis function of KRAS, GTP is hydrolyzed to GDP, the GDP-bound KRAS is transformed into an inactive state, and the molecular switch is turned off.
  • RAS When RAS is mutated, its intrinsic or GAP-induced GTPase activity is inhibited, RAS is always in a GTP-bound state, and the molecular switch is continuously opened, which in turn leads to continuous activation of various downstream signaling pathways, and abnormal cell proliferation and growth.
  • KRAS mutations In pancreatic cancer, up to about 90% of patients have KRAS mutations, of which about 40% point mutations are KRAS G12D mutations. In addition, about 13.3% of colorectal cancer, about 10% of rectal cancer, about 4% of non-small cell lung cancer and about 1.7% of small cell lung cancer patients have KRAS G12D mutation.
  • KRAS G12D Due to its important role in tumorigenesis and its high mutation frequency, KRAS G12D is a very attractive antitumor target. To date, there are no effective KRAS G12D inhibitor drugs on the market. Based on the above background, we developed a class of structurally novel KRAS G12D inhibitors.
  • the purpose of the present invention is to provide a new class of KRAS G12D inhibitors.
  • the first aspect of the present invention provides a compound of the following formula (I), or an optical isomer, a pharmaceutically acceptable salt, a prodrug, a deuterated derivative, a hydrate, or a solvate thereof. :
  • Ar is selected from aryl or heteroaryl; the aryl or heteroaryl is optionally substituted with one or more groups selected from the group consisting of halogen, C 1-4 alkyl, C 1-4 haloalkane base, hydroxyl, C 1-4 alkoxy, C 1-4 haloalkoxy, C 2-4 alkenyl, C 2-4 halo alkenyl, C 2-4 alkynyl, C 2-4 halo alkyne base, C 3-6 cycloalkyl, 3- to 6-membered heterocyclyl, C 3-6 cycloalkyl-O-, 3- to 6-membered heterocyclyl-O-, C 3-6 cycloalkane base C 2-4 alkynyl, 3- to 6-membered heterocyclyl C 2-4 alkynyl, C 1-4 haloalkyl C 2-4 alkynyl, NR 2 R 2 , CN, SR 2 , -OC( O)R k , -OP(
  • R 5 is selected from hydrogen or C 1-4 alkyl
  • R a is selected from hydrogen, halogen, or C 1-4 alkyl
  • A is selected from chemical bonds, -O-, -S-, -NR 6 -; wherein, R 6 is selected from hydrogen or C 1-4 alkyl;
  • R 1 is selected from C 3-8 cycloalkyl groups or 4- to 12-membered heterocyclic groups, wherein the heterocyclic groups include partially unsaturated or saturated monocyclic or polycyclic heterocyclic groups, and the polycyclic heterocyclic groups include spiro Ring, fused or bridged heterocyclyl; said cycloalkyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of halogen, C 1-4 alkyl, C 1- 4 haloalkyl, hydroxyl, C 1-4 alkoxy, C 1-4 haloalkoxy, -CH 2 OC(O)NR 9 R 10 , CN, SR 2 , C 2-4 alkenyl, C 2-4 Alkynyl, C 3-8 cycloalkyl, 3- to 8-membered heterocyclyl, NR h R h , -(CR 7 R 8 ) m -OR h , -(CR 7 R 8 ) m -NR
  • each of the above-mentioned alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl groups is optionally and each independently substituted with 1-3 substituents each independently selected from the group of Substituted: halogen, C 1-4 alkyl, C 1-4 haloalkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 3-8 cycloalkyl, 3- to 8-membered heterocyclyl, Aryl, heteroaryl, CN, NO2, ORh , SRh , NRhRh , C(O) Rt , C(O ) ORh , C(O ) NRhRh , NRhC ( O)R t , NR h S(O) 2 R t , or S(O) 2 R t , provided that the resulting chemical structure is stable and meaningful; wherein R t is a C 1-4 alkane group
  • the above-mentioned aryl group is an aromatic group containing 6-12 carbon atoms; the heteroaryl group is a 5- to 15-membered (preferably 5- to 12-membered) heteroaromatic group.
  • each R d is independently selected from hydrogen, C 1-4 alkyl, C 3-6 cycloalkyl; or two R d together with the same carbon atom to which it is attached forms C 3-6 cycloalkyl.
  • formula (I) is formula (II):
  • Ar is selected from aryl or heteroaryl; the aryl or heteroaryl is optionally substituted with one or more groups selected from the group consisting of halogen, C 1-4 alkyl, C 1-4 haloalkane base, hydroxyl, C 1-4 alkoxy, C 1-4 haloalkoxy, C 2-4 alkenyl, C 2-4 halo alkenyl, C 2-4 alkynyl, C 2-4 halo alkyne base, C 3-6 cycloalkyl, 3- to 6-membered heterocyclyl, C 3-6 cycloalkyl-O-, 3- to 6-membered heterocyclyl-O-, C 3-6 cycloalkane base C 2-4 alkynyl, 3- to 6-membered heterocyclyl C 2-4 alkynyl, C 1-4 haloalkyl C 2-4 alkynyl, NR 2 R 2 , CN, SR 2 , -OC( O)R k , -OP(
  • R 5 is selected from hydrogen or C 1-4 alkyl
  • R a is selected from hydrogen, halogen, or C 1-4 alkyl
  • A is selected from chemical bonds, -O-, -S-, -NR 6 -; wherein, R 6 is selected from hydrogen or C 1-4 alkyl;
  • R 1 is selected from C 3-8 cycloalkyl groups or 4- to 12-membered heterocyclic groups, wherein the heterocyclic groups include partially unsaturated or saturated monocyclic or polycyclic heterocyclic groups, and the polycyclic heterocyclic groups include spiro Ring, fused or bridged heterocyclyl; said cycloalkyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of halogen, C 1-4 alkyl, C 1- 4 haloalkyl, hydroxyl, C 1-4 alkoxy, C 1-4 haloalkoxy, -CH 2 OC(O)NR 9 R 10 , CN, SR 2 , C 2-4 alkenyl, C 2-4 Alkynyl, C 3-8 cycloalkyl, 3- to 8-membered heterocyclyl, NR h R h , -(CR 7 R 8 ) m -OR h , -(CR 7 R 8 ) m -NR
  • R 11 is selected from hydrogen, halogen, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy, C 1-4 haloalkoxy, C 2-4 alkynyl, C 3-6 ring Alkyl, or CN.
  • Ar is selected from the group consisting of phenyl and naphthyl.
  • Ar is selected from the following groups:
  • formula (I) is formula (IIIa) or formula (IIIb),
  • formula (I) is formula (IV),
  • Each R d is independently selected from hydrogen, C 1-4 alkyl, C 3-6 cycloalkyl; or two R d together form a C 3-6 cycloalkyl with the same carbon atom to which it is attached; and at least Two R d together with the same carbon atom to which they are attached form C 3-6 cycloalkyl;
  • Ar, A, B, R 1 , R 11 are as defined above.
  • formula (I) is formula (V),
  • R 1 is selected from C 3-8 cycloalkyl or 4- to 12-membered heterocyclyl
  • R 12 is selected from hydrogen, halogen, C 1-4 alkyl, C 1-4 haloalkyl, -CH 2 OC(O)NR 9 R 10 , C 2-4 alkenyl, C 2-4 alkynyl, C 3 -8 cycloalkyl, 3- to 8-membered heterocyclyl, CN, OR h , SR h , NR h Rh , -(CR 7 R 8 ) m -OR h , -(CR 7 R 8 ) m - NR h R h ;
  • n is selected from 0, 1, or 2;
  • Ar, X, Y, R, A , B, R7 , R8 , R9 , R10, Rh , and m are as defined above.
  • formula (I) is formula (VI),
  • R is selected from the group consisting of:
  • Ar is selected from the group consisting of:
  • R 14 and R 15 are each independently selected from halogen, C 1-4 alkyl, C 1-4 haloalkyl, hydroxy, C 1-4 alkoxy, C 1-4 haloalkoxy, C 2-4 alkenyl , C 2-4 haloalkenyl, C 2-4 alkynyl, C 2-4 haloalkynyl, C 3-6 cycloalkyl, 3- to 6-membered heterocyclyl, C 3-6 cycloalkane radical-O-, 3- to 6-membered heterocyclyl-O-, NR 2 R 2 , CN, SR 2 , -OC(O)R k , -OP(O)(OR m ) 2 , -O- CH(R n )-OP(O)(OR m ) 2 ; wherein each R 2 is each independently hydrogen or C 1-4 alkyl; R k is selected from C 1-12 alkyl, wherein alkyl is optionally Substituted with one or more
  • k is selected from 0, 1, or 2;
  • p is selected from 0, 1, 2, 3, 4, or 5;
  • q is selected from 0, 1, 2, 3, or 4;
  • formula (I) is formula (VII),
  • R 13 , R 14 , R 11 , k, p are as defined above.
  • formula (I) is formula (VIII),
  • R, A, B, R 1 , R 11 , R 5 , and Ra are as defined above; R 15 and q are as defined above.
  • formula (I) is formula (IX),
  • formula (I) is formula (X),
  • Structural Fragments Selected from the following groups:
  • Ar is selected from the group consisting of:
  • formula (I) is formula (XI),
  • R 11 is selected from hydrogen, halogen, C 1-4 alkyl
  • R 11' is selected from hydrogen, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkoxy, or CN;
  • Ar and R are as defined above.
  • formula (I) is formula (XII),
  • R 11 and R 11' are as defined above;
  • R 13 , R 14 , k, p are as defined above.
  • formula (I) is formula (XIII),
  • R 11 is selected from hydrogen, halogen, C 1-4 alkyl
  • X is selected from N or CR 11' ; wherein, R 11' is selected from hydrogen, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkoxy, or CN;
  • R 14 is selected from halogen, C 1-4 alkyl, C 1-4 haloalkyl, hydroxy, C 1-4 alkoxy, C 1-4 haloalkoxy, C 2-4 alkenyl, C 2-4 halo Alkenyl, C 2-4 alkynyl, C 2-4 haloalkynyl, C 3-6 cycloalkyl, 3- to 6-membered heterocyclyl, C 3-6 cycloalkyl-O-, 3 - to 6-membered heterocyclyl-O-, NR 2 R 2 , CN, SR 2 ; wherein each R 2 is each independently hydrogen or C 1-4 alkyl;
  • R 14' is selected from hydrogen, -OC(O)R k , -OP(O)(OR m ) 2 , or -O-CH(R n )-OP(O)(OR m ) 2 ; wherein R k is selected from C 1-12 alkyl, wherein alkyl is optionally substituted with one or more groups selected from the group consisting of hydroxy, C 1-4 alkoxy, NR 2 R 2 , C(O)OH, C(O)OC 1-2 alkyl, or CN; R m is selected from hydrogen or C 1-4 alkyl; R n is selected from hydrogen or C 1-4 alkyl;
  • R 17 is selected from hydrogen or -CO(O)-CH(R n )-OC(O)-U; wherein, R n is selected from hydrogen or C 1-4 alkyl; U is selected from C 1-18 alkyl;
  • k is selected from 0, 1, or 2;
  • p is selected from 0, 1, 2, 3, or 4.
  • Another aspect of the present invention provides a compound of the following formula (XIV), or an optical isomer, a pharmaceutically acceptable salt, a prodrug, a deuterated derivative, a hydrate, or a solvate thereof. :
  • R 11 is selected from hydrogen, halogen, C 1-4 alkyl
  • X is selected from N or CR 11' ; wherein, R 11' is selected from hydrogen, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkoxy, or CN;
  • R 18 is selected from halogen, C 1-4 alkyl, or C 1-4 alkoxy;
  • Ar and R are as defined above.
  • the compound of formula (I) is selected from the following group:
  • "*" represents a chiral center, which can optionally be R configuration or S configuration, or a mixture of R configuration and S configuration;
  • the carbon atom to which the bond is attached may optionally be in the R or S configuration, or optionally in cis or trans.
  • the second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the compound described in the first aspect of the present invention, or an optical isomer, pharmaceutically acceptable salt, prodrug, deuterated Derivatives, hydrates, solvates, and pharmaceutically acceptable carriers.
  • the third aspect of the present invention provides a compound according to the first aspect of the present invention, or an optical isomer, a pharmaceutically acceptable salt, a prodrug, a deuterated derivative, a hydrate, or a solvate thereof.
  • the use of it is used to prepare a pharmaceutical composition for the treatment of diseases, disorders or conditions related to KRAS G12D activity or expression.
  • the disease, disorder or condition is selected from the group consisting of pancreatic cancer, non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, colon cancer, colorectal cancer, thyroid cancer, embryonic cancer Rhabdomyosarcoma, granular cell tumor of the skin, melanoma, liver cancer, rectal cancer, bladder cancer, throat cancer, breast cancer, prostate cancer, glioblastoma, ovarian cancer, head and neck squamous cell carcinoma, cervical cancer, esophageal cancer, Kidney cancer, skin cancer, lymphoma, gastric cancer, acute myeloid leukemia, myelofibrosis, B-cell lymphoma, monocytic leukemia, polycythemia splenomegaly, eosinophilic leukemia syndrome multiple, bone marrow cancer, etc. Solid tumors and hematological tumors.
  • the inventors After long-term and in-depth research, the inventors have unexpectedly discovered a class of KRAS G12D inhibitors with novel structures, as well as their preparation methods and applications.
  • the compounds of the present invention can be applied to the treatment of various diseases related to the activity of the small GTPases. Based on the above findings, the inventors have completed the present invention.
  • each chiral carbon atom may optionally be in the R configuration or the S configuration, or a mixture of the R configuration and the S configuration.
  • alkyl refers to a straight chain (ie, unbranched) or branched saturated hydrocarbon group containing only carbon atoms, or a combination of straight and branched chain groups .
  • a carbon number limitation eg, C 1-10
  • C 1-8 alkyl refers to an alkyl group containing 1-8 carbon atoms, including methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, or similar groups.
  • alkenyl refers to a straight or branched, carbon chain group having at least one carbon-carbon double bond. Alkenyl groups can be substituted or unsubstituted. When an alkenyl group is preceded by a carbon number limitation (eg, C2-8 ), it means that the alkenyl group contains 2-8 carbon atoms.
  • C 2-8 alkenyl refers to an alkenyl group containing 2-8 carbon atoms, including vinyl, propenyl, 1,2-butenyl, 2,3-butenyl, butadienyl, or the like group.
  • alkynyl refers to an aliphatic hydrocarbon group having at least one carbon-carbon triple bond.
  • the alkynyl group can be straight or branched, or a combination thereof.
  • a carbon number limitation eg, C 2-8 alkynyl
  • C 2-8 alkynyl refers to straight or branched chain alkynyl groups having 2-8 carbon atoms, including ethynyl, propynyl, isopropynyl, butynyl, isobutynyl, sec-butynyl, tert-butynyl, or similar groups.
  • cycloalkyl refers to a monocyclic, bicyclic or polycyclic (fused, bridged or spiro) ring system group having saturated or partially saturated .
  • a cycloalkyl group is preceded by a carbon number limitation (eg C 3-10 ), it means that the cycloalkyl group contains 3-10 carbon atoms.
  • C 3-8 cycloalkyl refers to saturated or partially unsaturated monocyclic or bicycloalkyl having 3-8 carbon atoms, including cyclopropyl, cyclobutyl, cyclo pentyl, cycloheptyl, or similar groups.
  • “Spirocycloalkyl” refers to a bicyclic or polycyclic group in which a single carbon atom (called a spiro atom) is shared between the monocyclic rings, these may contain one or more double bonds, but none of the rings have fully conjugated pi electrons system.
  • “Fused cycloalkyl” refers to an all-carbobicyclic or polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more rings may contain one or more bicyclic bonds, but none of the rings have a fully conjugated pi electron system.
  • “Bridged cycloalkyl” refers to an all-carbon polycyclic group in which any two rings share two non-directly attached carbon atoms, these may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system .
  • the atoms contained in the cycloalkyl group are all carbon atoms.
  • the following are some examples of cycloalkyl groups, and the present invention is not limited to the following cycloalkyl groups.
  • Aryl refers to an all-carbon monocyclic or fused polycyclic (ie, rings that share adjacent pairs of carbon atoms) groups having a conjugated pi-electron system, such as phenyl and naphthyl.
  • the aryl ring can be fused to other cyclic groups (including saturated and unsaturated rings), but cannot contain heteroatoms such as nitrogen, oxygen, or sulfur, and the point of attachment to the parent must be in a conjugated pi-electron system on the carbon atom of the ring.
  • Aryl groups can be substituted or unsubstituted.
  • Heteroaryl refers to an aromatic monocyclic or polycyclic group containing one to more heteroatoms (optionally selected from nitrogen, oxygen and sulfur), or a heterocyclic group containing one to more heteroatoms A polycyclic group formed by condensing nitrogen, oxygen and sulfur) with an aryl group, and the attachment site is located on the aryl group. Heteroaryl groups can be optionally substituted or unsubstituted. The following are some examples of heteroaryl groups, and the present invention is not limited to the following heteroaryl groups.
  • Heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent wherein one or more ring atoms are selected from nitrogen, oxygen or sulfur and the remaining ring atoms are carbon.
  • monocyclic heterocyclyl groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl.
  • Polycyclic heterocyclyl refers to heterocyclyl including spiro, fused and bridged rings.
  • “Spirocyclic heterocyclyl” refers to a polycyclic heterocyclic group in which each ring in the system shares one atom (called a spiro atom) with other rings in the system, wherein one or more ring atoms are selected from nitrogen, oxygen or sulfur, the remaining ring atoms are carbon.
  • “Fused-ring heterocyclyl” refers to a polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, one or more rings may contain one or more double bonds, but no A ring has a fully conjugated pi electron system and one or more ring atoms are selected from nitrogen, oxygen or sulfur, and the remaining ring atoms are carbon.
  • “Bridged heterocyclyl” refers to a polycyclic heterocyclic group in which any two rings share two atoms that are not directly connected, these may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system , and one or more ring atoms are selected from nitrogen, oxygen or sulfur, and the remaining ring atoms are carbon. If both saturated and aromatic rings are present in the heterocyclyl group (for example, the saturated and aromatic rings are fused together), the point of attachment to the parent must be on the saturated ring. Note: When the point of attachment to the parent is on an aromatic ring, it is called a heteroaryl group, not a heterocyclyl group. The following are some examples of heterocyclic groups, and the present invention is not limited to the following heterocyclic groups.
  • halogen refers to F, Cl, Br, and I.
  • substituted refers to the replacement of one or more hydrogen atoms on a specified group with a specified substituent. Particular substituents are those described correspondingly in the preceding paragraphs, or the substituents appearing in the various examples. Unless otherwise specified, an optionally substituted group may have at any substitutable position of the group a substituent selected from a specified group, which may be the same or different at each position.
  • a cyclic substituent such as a heterocyclyl
  • substituents contemplated by the present invention are those that are stable or chemically achievable.
  • the substituents are for example (but not limited to): C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 3-8 cycloalkyl, 3- to 12-membered heterocyclyl , aryl, heteroaryl, halogen, hydroxyl, carboxyl (-COOH), C 1-8 aldehyde group, C 2-10 acyl group, C 2-10 ester group, amino group.
  • a pharmaceutically acceptable salt refers to a salt suitable for contact with the tissue of a subject (eg, a human) without undue side effects.
  • a pharmaceutically acceptable salt of a compound of the present invention includes a salt of a compound of the present invention that has an acidic group (eg, potassium, sodium, magnesium, calcium) or has a basic salts of compounds of the invention (eg, sulfates, hydrochlorides, phosphates, nitrates, carbonates).
  • the present invention provides a class of compounds of formula (I), or their deuterated derivatives, their salts, isomers (enantiomers or diastereomers, if present), hydrates , the use of a pharmaceutically acceptable carrier or excipient for inhibiting KRAS G12D.
  • the compounds of the present invention are useful as a KRAS G12D inhibitor.
  • the present invention is a single inhibitor of KRAS G12D, and achieves the purpose of preventing, relieving or curing diseases by regulating the activity of KRAS G12D.
  • Indicated diseases include pancreatic cancer, non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, colon cancer, colorectal cancer, thyroid cancer, embryonal rhabdomyosarcoma, granular cell tumor of the skin, melanoma, liver cancer, rectal cancer Cancer, bladder cancer, throat cancer, breast cancer, prostate cancer, glioblastoma, ovarian cancer, head and neck squamous cell carcinoma, cervical cancer, esophageal cancer, kidney cancer, skin cancer, lymphoma, stomach cancer, acute myeloid leukemia , myelofibrosis, B-cell lymphoma, monocytic leukemia, polycythemia splenomegaly, multiple eosinophilic leukemia syndrome
  • compositions may be combined with pharmaceutically acceptable excipients or The carriers are formulated together and the resulting compositions can be administered in vivo to mammals, such as men, women and animals, for the treatment of conditions, symptoms and diseases.
  • the compositions may be: tablets, pills, suspensions, solutions, emulsions, capsules, aerosols, sterile injectable solutions. Sterile powder, etc.
  • the pharmaceutically acceptable excipients include microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, calcium hydrogen phosphate, mannitol, hydroxypropyl-beta-cyclodextrin, beta-cyclodextrin (increase), glycine, disintegrants (such as starch, croscarmellose sodium, complex silicates and high molecular polyethylene glycol), granulation binders (such as polyvinylpyrrolidone, sucrose, gelatin and gum arabic) and lubricants such as magnesium stearate, glycerin and talc.
  • disintegrants such as starch, croscarmellose sodium, complex silicates and high molecular polyethylene glycol
  • granulation binders such as polyvinylpyrrolidone, sucrose, gelatin and gum arabic
  • lubricants such as magnesium stearate, glycerin and talc.
  • the pharmaceutical composition is in a dosage form suitable for oral administration, including but not limited to tablets, solutions, suspensions, capsules, granules, powders.
  • the amount of a compound or pharmaceutical composition of the present invention administered to a patient is not fixed, but is usually administered in a pharmaceutically effective amount. Meanwhile, the amount of the compound actually administered can be determined by the physician according to the actual situation, including the condition to be treated, the route of administration selected, the actual compound administered, the individual condition of the patient, and the like.
  • the dosage of a compound of the present invention will depend upon the particular use of the treatment, the mode of administration, the patient's condition, and the judgment of the physician.
  • the ratio or concentration of a compound of the present invention in a pharmaceutical composition depends on a variety of factors, including dosage, physicochemical properties, route of administration, and the like.
  • Boc tert-butoxycarbonyl
  • Catacxium-A-Pd-G3 [n-butylbis(1-adamantyl)phosphine](2-amino-1,1'-biphenyl-2-yl)palladium(II) methanesulfonate
  • DIPEA or DIEA N,N-diisopropylethylamine
  • DIBAL-H diisobutylaluminum hydride
  • HATU N,N,N',N'-tetramethyl-O-(7-azabenzotriazol-1-yl)hexafluorophosphate urea
  • LiHMDS Lithium Bistrimethylsilylamide
  • Ph phenyl
  • TBAF Tetra-n-butylammonium fluoride
  • TIPS triisopropylsilyl
  • TsOH p-toluenesulfonic acid
  • the compound formulas (I-A), (I-B) and (I-C) of the present invention can be prepared by the following methods:
  • the target compound with a single chiral configuration can use the R or S isomer of compound (VIIa) in the first step of the reaction, and finally the target compound with chirality of R or S can be obtained; it can also be resolved by chirality.
  • each group is as described above.
  • the reagents and conditions of each step can be selected from conventional reagents or conditions in the art for such preparation methods. After the structure of the compound of the present invention is disclosed, the above selection can be performed by those skilled in the art according to the knowledge in the art.
  • compositions and methods of administration are provided.
  • the compound of the present invention Since the compound of the present invention has excellent inhibitory activity against KRAS G12D, the compound of the present invention and its various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and containing the compound of the present invention are the main activities
  • the pharmaceutical composition of the ingredients can be used for the treatment, prevention and alleviation of diseases related to KRAS G12D activity or expression.
  • the pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof and a pharmacologically acceptable excipient or carrier within a safe and effective amount.
  • the "safe and effective amount” refers to: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention per dose, more preferably 5-200 mg of the compound of the present invention per dose.
  • the "one dose” is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to one or more compatible solid or liquid filler or gelling substances which are suitable for human use and which must be of sufficient purity and sufficiently low toxicity. "Compatibility” as used herein means that the components of the composition can be admixed with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds.
  • Examples of pharmaceutically acceptable carrier moieties include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid) , magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (such as ), wetting agents (such as sodium lauryl sulfate), colorants, flavors, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
  • the mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration .
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with (a) fillers or compatibilizers, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders such as, for example, hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, For example, glycerol; (d) disintegrants, such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) Absorption accelerators such as quaternary amine compounds; (g) wetting agents such as cetyl alcohol and glyceryl monostea
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared using coatings and shell materials, 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 in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be employed are polymeric substances and waxes. 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.
  • liquid dosage forms may contain inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances, and the like.
  • inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylform
  • compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
  • 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 such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • suspending agents such as 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 non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • Dosage forms for topical administration of the compounds of this 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 that may be required if necessary.
  • the compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) in need of treatment, and the dose is the effective dose considered pharmaceutically, for a 60kg body weight, the daily dose is
  • the administration dose is usually 1 to 2000 mg, preferably 5 to 500 mg.
  • the specific dosage should also take into account the route of administration, the patient's health and other factors, which are all within the skill of the skilled physician.
  • KRAS G12D inhibitor preparation and application thereof, which can inhibit the activity of KRAS G12D at a very low concentration.
  • a pharmaceutical composition for treating diseases related to KRAS G12D activity is provided.
  • Compound 1a was synthesized according to the method in patent WO2021041671.
  • Compound 1a 60 mg, 0.24 mmol
  • N,N-diisopropylethylamine (215 mg, 1.66 mmol) were dissolved in dichloromethane (3 mL), and the mixture was placed at -40 °C and stirred while adding compound 1b ( 50 mg, 0.24 mmol).
  • the reaction mixture was stirred at -40°C for 1 hour. After the reaction is completed, add water.
  • the mixture was extracted with dichloromethane (3 x 15 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
  • Compound 1f was synthesized according to the method in patent WO2021041671.
  • Compound 1e 42 mg, 0.08 mmol
  • compound 1f 43 mg, 0.13 mmol
  • [n-butylbis(1-adamantyl)phosphine](2-amino-1,1'-biphenyl-2 -base) palladium (II) 5.5 mg, 0.008 mmol
  • Tetrahydrofuran 1.5 mL
  • aqueous potassium phosphate 1.5 M, 0.15 mL
  • Compound 3g was synthesized according to the method in patent WO2021041671.
  • Compound 3f 34 mg, 0.061 mmol
  • [n-butylbis(1-adamantyl)phosphine](2-amino-1,1'-biphenyl-2-yl)palladium(II) 5 mg , 0.0061 mmol
  • compound 3g 32 mg, 0.086 mmol
  • potassium phosphate 1.5 M, 0.5 mL was added and heated to 60 °C for 3 hours under nitrogen protection.
  • reaction solution was quenched with saturated aqueous ammonium chloride solution (5 mL), then concentrated hydrochloric acid (3 mL) was added, stirred at room temperature for 1 hour, added with water (30 mL), extracted three times with ethyl acetate (30 mL ⁇ 3), and the combined organic layers were It was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained crude product was separated and purified by silica gel column chromatography to obtain compound 4c (188 mg, 18%) as a pale yellow oil.
  • Compound 4g was synthesized according to the method in patent WO2021041671.
  • Compound 4f (29mg, 0.05mmol), compound 4g (33mg, 0.10mmol), potassium phosphate (32mg, 0.15mmol), [n-butylbis(1-adamantyl)phosphine](2-amino- 1,1'-Biphenyl-2-yl)palladium(II) (4 mg, 0.005 mmol) was dissolved in a mixed solution of 1,4-dioxane/water (0.5 mL/0.1 mL). The reaction mixture was heated and stirred at 90°C for 2 hours under nitrogen atmosphere.
  • Compound 12a was prepared according to the method in patent WO2021041671.
  • Compound 13a was prepared according to the method in patent WO2021041671.
  • Compound 14a was prepared according to the method in patent WO2021041671.
  • Embodiment 20 the preparation of compound 20, compound 20S, compound 20R
  • Compound 20-P1 and Compound 20-P2 one is compound 20S and the other is compound 20R.
  • Compound activity using AGS (human gastric cancer cell line, KRAS G12D mutation), GP2D (human colon cancer cell line, KRAS G12D mutation), HPAC (human pancreatic cancer cell line, KRAS G12D mutation) and AsPC1 (human pancreatic cancer cell line, KRAS G12D mutation) G12D mutation) 3D cell proliferation inhibition expression, using Cell Titer-Glo (CTG) method.
  • Compounds were dissolved in DMSO to make 10 mM stock solutions, and the compounds were diluted accordingly in culture medium. Remove the cell supernatant medium, rinse once with DPBS, add 2 mL of trypsin, and leave it for a while at 37°C.
  • Ave_H represents the average reading of the DMSO well
  • Sample represents the average reading of the compound well
  • Ave_L represents the average reading of the positive control group at 10 ⁇ M.
  • the log(inhibitor) vs.response-Variable slope of the analysis software GraphPad Prism 5 was used to fit the dose-response curve to obtain the IC of each compound on cell proliferation inhibition. 50 value or inhibition rate.
  • the activities of some representative compounds are shown in Table 1.

Landscapes

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

Abstract

本发明提供了作为KRAS G12D抑制剂的化合物,具体地,本发明提供了一种如下式(I)所示结构的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物。所述的化合物可以用于治疗或预防与KRAS G12D的活性或表达量相关的疾病或病症。

Description

作为KRAS G12D抑制剂的杂环化合物 技术领域
本发明涉及药物化学领域;具体地说,本发明涉及一类新型含有三环杂芳基的衍生物,其合成方法及其作为一种KRAS G12D抑制剂在制备药物用于治疗肿瘤等相关多种疾病中的应用。
背景技术
KRAS(Kirsten Rat Sarcoma Viral Oncogene Homolog)是一种小GTP酶,属于RAS家族成员。目前已知的RAS基因有三种,HRAS、NRAS和KRAS。RAS蛋白在癌症发生发展中起着重要的作用,20%-30%的人类肿瘤中都存在RAS突变,其中KRAS突变频率最高,多见于肺癌、胰腺癌和结肠癌等。
KRAS蛋白包含188个氨基酸残基,有多个功能基序,包括核苷酸结合区、转换I区和转换II区、核苷酸交换因子(GEF)结合区域、GTP酶激活蛋白(GAP)结合区域和效应蛋白结合区域等。正常情况下,KRAS的活性受多种上游信号因子的调控,如受体酪氨酸激酶、整合素、G蛋白偶联受体等。KRAS作为细胞中的一种分子开关,通过GTP或GDP结合状态的转换,分别表现出激活(开)或失活(关)的状态,将上游接收到的信号传递给下游的效应蛋白。KRAS蛋白本身有比较慢的核苷酸交换速率和较弱的GTP水解功能。接收上游刺激信号后,在SOS1(Son of Sevenless 1)等鸟嘌呤核苷酸交换因子的辅助下,KRAS蛋白释放GDP,结合GTP。GTP结合的RAS蛋白处于激活状态,招募下游效应蛋白,激活Raf-MEK-ERK和PI3K-AKT-mTOR等信号通路,促进细胞的生长、增殖、存活、代谢以及血管生成等。GTP酶激活蛋白GAP和KRAS结合后,会显著促进KRAS的GTP水解功能,GTP水解为GDP,GDP结合状态的KRAS转变为失活状态,分子开关关闭。当RAS发生突变时,其固有或GAP诱导的GTP酶活性受到抑制,RAS一直处于GTP结合状态,分子开关持续开放,进而导致下游的多种信号通路处于持续性激活状态,细胞异常增殖和生长。
在胰腺癌中,高达90%左右的患者都有KRAS突变,其中约40%的点突变为KRAS G12D突变。另外,约13.3%的结直肠癌,10%左右的直肠癌,4%左右的非小细胞肺癌和1.7%左右的小细胞肺癌患者中都有KRAS G12D突变。
由于在肿瘤生成中发挥的重要作用和其高发的突变频率,KRAS G12D成为一个非常有吸引力的抗肿瘤靶点。目前为止,市场上还没有任何有效的KRAS G12D抑制剂药物。基于以上背景,我们开发了一类结构新颖的KRAS G12D抑制剂。
发明内容
本发明的目的是提供一类新型的KRAS G12D抑制剂。
本发明的第一方面,提供了一种如下式(I)所示结构的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物:
Figure PCTCN2022086183-appb-000001
式(I)中:
Ar选自芳基或杂芳基;所述的芳基或杂芳基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、C 3-6环烷基C 2-4炔基、3-至6-元杂环基C 2-4炔基、C 1-4卤代烷基C 2-4炔基、NR 2R 2、CN、SR 2、-OC(O)R k、-O-P(O)(OR m) 2、-O-CH(R n)-O-P(O)(OR m) 2;其中,各个R 2各自独立为氢或C 1-4烷基;R k选自C 1-12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢或C 1-4烷基;R n选自氢或C 1-4烷基;上面所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
R选自5-12元杂环基,包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、=O、羟基、CN、-CO(O)-CH(R n)-O-C(O)-U;其中,R n选自氢或C 1-4烷基;U选自C 1-18烷基;
Z选自化学键、-O-、-S-、-NR 5-、-C(R a) 2-、-C≡C-、-CR a=CR a-、-N=、-CR a=;其中,R 5选自氢或C 1-4烷基;R a选自氢、卤素、或C 1-4烷基;
A选自化学键、-O-、-S-、-NR 6-;其中,R 6选自氢或C 1-4烷基;
B选自-(CR 7R 8) m-、-(CR 7R 8) m-T-(CR 7R 8) m-;其中,T选自-C≡C-、-CR a=CR a-、C 3-6环烷基、3-至6-元杂环基;其中,所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、CN;R 7和R 8各自独立地选自下组:氢、卤素、或C 1-4烷基;或R 7和R 8与其相连接的碳原子一起形成C 3- 6环烷基;R a选自氢、卤素、或C 1-4烷基;各个m各自独立地选自0、1、2、或3;
R 1选自C 3-8环烷基或4-至12-元杂环基,其中,杂环基包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、-CH 2OC(O)NR 9R 10、CN、SR 2、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、NR hR h、-(CR 7R 8) m-OR h、-(CR 7R 8) m-NR hR h、或=M取代;其中,R 9和R 10各自独立地选自氢或C 1-4烷基,或R 9和R 10与其连接的氮原子一起形成4-至-8元杂环基,此杂环基含有1或2个N原子以及0或1个选自O、S的杂原子;各个R h各自独立为氢、C 1-4烷基、或C 1-4卤代烷基;M选自O或CR 3R 4;其中R 2、R 3、R 4、R 7、R 8、和m的定义如上所述;X和Y各自独立地选自选自N或CR 11;其中,R 11选自氢、卤素、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4炔基、C 3-6环烷基、或CN;
前提条件是,当R 1不具有=M取代,且M选自CR 3R 4时,结构片段
Figure PCTCN2022086183-appb-000002
不为
Figure PCTCN2022086183-appb-000003
其中,各个上述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选地且各自独立地被1-3个各自独立地选自下组的取代基取代:卤素、C 1-4烷基、C 1-4卤代烷基、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、芳基、杂芳基、CN、NO 2、OR h、SR h、NR hR h、C(O)R t、C(O)OR h、C(O)NR hR h、NR hC(O)R t、NR hS(O) 2R t、或S(O) 2R t,前提条件是所形成的化学结构是稳定的和有意义的;其中,R t为C 1-4烷基、C 2-4烯基、C 2-4炔基、C 3-8环烷基、4-至8-元杂环基、芳基、或杂芳基;各个R h各自独立为氢、C 1-4烷基、或C 1-4卤代烷基;或两个R h与其连接的氮原子一起形成3-至-8元杂环基,此杂环基含有1或2个N原子以及0或1个选自O、S的杂原子;
除非特别说明,上述的芳基为含有6-12个碳原子的芳香基团;杂芳基为5-至15-元(优选为5-至12-元)杂芳香基团。
在另一优选例中,当R 1不具有=M取代,且M选自CR 3R 4时,结构片段
Figure PCTCN2022086183-appb-000004
Figure PCTCN2022086183-appb-000005
其中,各个R d各自独立地选自氢、C 1-4烷基、C 3-6环烷基;或二个R d与其相连接的同一个碳原子一起形成C 3-6环烷基。
在另一优选例中,当R 1不具有=M取代,且M选自CR 3R 4时,结构片段
Figure PCTCN2022086183-appb-000006
Figure PCTCN2022086183-appb-000007
在另一优选例中,当R 1不具有=M取代,且M选自CR 3R 4时,结构片段
Figure PCTCN2022086183-appb-000008
Figure PCTCN2022086183-appb-000009
在另一优选例中,式(I)为式(II):
Figure PCTCN2022086183-appb-000010
式(II)中:
Ar选自芳基或杂芳基;所述的芳基或杂芳基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤 代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、C 3-6环烷基C 2-4炔基、3-至6-元杂环基C 2-4炔基、C 1-4卤代烷基C 2-4炔基、NR 2R 2、CN、SR 2、-OC(O)R k、-O-P(O)(OR m) 2、-O-CH(R n)-O-P(O)(OR m) 2;其中,各个R 2各自独立为氢或C 1-4烷基;R k选自C 1-12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢、C 1-4烷基;R n选自氢或C 1-4烷基;上面所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
R选自5-12元杂环基,包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、=O、羟基、CN、-CO(O)-CH(R n)-O-C(O)-U;其中,R n选自氢或C 1-4烷基;U选自C 1-18烷基;
Z选自化学键、-O-、-S-、-NR 5-、-C(R a) 2-、-C≡C-、-CR a=CR a-、-N=、-CR a=;其中,R 5选自氢或C 1-4烷基;R a选自氢、卤素、或C 1-4烷基;
A选自化学键、-O-、-S-、-NR 6-;其中,R 6选自氢或C 1-4烷基;
B选自-(CR 7R 8) m-、-(CR 7R 8) m-T-(CR 7R 8) m-;其中,T选自-C≡C-、-CR a=CR a-、C 3-6环烷基、3-至6-元杂环基;其中,所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、CN;R 7和R 8各自独立地选自下组:氢、卤素、或C 1-4烷基;或R 7和R 8与其相连接的碳原子一起形成C 3- 6环烷基;R a选自氢、卤素、或C 1-4烷基;各个m各自独立地选自0、1、2、或3;
R 1选自C 3-8环烷基或4-至12-元杂环基,其中,杂环基包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、-CH 2OC(O)NR 9R 10、CN、SR 2、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、NR hR h、-(CR 7R 8) m-OR h、-(CR 7R 8) m-NR hR h、或=M取代;其中,R 9和R 10各自独立地选自氢或C 1-4烷基,或R 9和R 10与其连接的氮原子一起形成4-至-8元杂环基,此杂环基含有1或2个N原子以及0或1个选自O、S的杂原子;各个R h各自独立为氢、C 1-4烷基、或C 1-4卤代烷基;M选自O或CR 3R 4;其中R 2、R 3、R 4、R 7、R 8、和m的定义如上所述;
R 11选自氢、卤素、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4炔基、C 3-6环烷基、或CN。
在另一优选例中,式(I)或式(II)中结构片段
Figure PCTCN2022086183-appb-000011
选自下组基团:
Figure PCTCN2022086183-appb-000012
Figure PCTCN2022086183-appb-000013
表示上述结构片段与式(I)或式(II)中其它结构的连接位点;
“*”表示手性中心;
上述基团任选地被0、1或2个R 13取代,其中R 13选自卤素、C 1-4烷基、=O、羟基、或CN。
在另一优选例中,Ar选自下组:苯基、萘基。
在另一优选例中,Ar选自下组基团:
Figure PCTCN2022086183-appb-000014
Figure PCTCN2022086183-appb-000015
Figure PCTCN2022086183-appb-000016
表示上述结构片段与式(I)或式(II)中其它结构的连接位点;
“*”表示手性中心。
在另一优选例中,式(I)或式(II)中结构片段
Figure PCTCN2022086183-appb-000017
选自下组的基团:
Figure PCTCN2022086183-appb-000018
“*”表示手性中心;
Figure PCTCN2022086183-appb-000019
表示上述结构片段与式(I)或式(II)中其它结构的连接位点。
在另一优选例中,式(I)为式(IIIa)或式(IIIb),
Figure PCTCN2022086183-appb-000020
“*”表示手性中心;
其中Ar、A、B、R 1、R 11的定义如上文中所述。
在另一优选例中,式(I)为式(IV),
Figure PCTCN2022086183-appb-000021
各个R d各自独立地选自氢、C 1-4烷基、C 3-6环烷基;或二个R d与其相连接的同一个碳原子一起形成C 3-6环烷基;且至少二个R d与其相连接的同一个碳原子一起形成C 3-6环烷基;
Ar、A、B、R 1、R 11的定义如上文中所述。
在另一优选例中,式(I)为式(V),
Figure PCTCN2022086183-appb-000022
R 1选自C 3-8环烷基或4-至12-元杂环基;
R 12选自氢、卤素、C 1-4烷基、C 1-4卤代烷基、-CH 2OC(O)NR 9R 10、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、CN、OR h、SR h、NR hR h、-(CR 7R 8) m-OR h、-(CR 7R 8) m-NR hR h
n选自0、1、或2;
Ar、X、Y、R、A、B、R 7、R 8、R 9、R 10、R h、和m的定义如上文中所述。
在另一优选例中,式(I)为式(VI),
Figure PCTCN2022086183-appb-000023
R选自下组基团:
Figure PCTCN2022086183-appb-000024
Ar选自下组基团:
Figure PCTCN2022086183-appb-000025
“*”表示手性中心;
Figure PCTCN2022086183-appb-000026
表示R基团与式(VI)化合物其它结构的连接位点;
Figure PCTCN2022086183-appb-000027
表示Ar基团与式(VI)化合物其它结构的连接位点;
R 13选自卤素、C 1-4烷基、=O、羟基、或CN;
R 14和R 15各自独立地选自卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、NR 2R 2、CN、SR 2、-OC(O)R k、-O-P(O)(OR m) 2、-O-CH(R n)-O-P(O)(OR m) 2;其中,各个R 2各自独立为氢或C 1-4烷基;R k选自C 1-12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢或C 1-4烷基;R n选自氢或C 1-4烷基;上面所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
k选自0、1、或2;
p选自0、1、2、3、4、或5;
q选自0、1、2、3、或4;
R 11的定义如上文所述;R d的定义如上文所述。
在另一优选例中,式(I)为式(VII),
Figure PCTCN2022086183-appb-000028
“*”表示手性中心;
R 13、R 14、R 11、k、p的定义如上文中所述。
在另一优选例中,式(I)为式(VIII),
Figure PCTCN2022086183-appb-000029
E选自选自化学键、-O-、-S-、-NR 5-、-C≡C-、或-CR a=CR a-;
R 16选自C 3-6环烷基或3-至6-元杂环基;所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
R、A、B、R 1、R 11、R 5、和R a的定义如上文中所述;R 15和q的定义如上文中所述。
在另一优选例中,式(I)为式(IX),
Figure PCTCN2022086183-appb-000030
其中Ar、A、B、R 1、R 11的定义如上文中所述。
在另一优选例中,式(I)为式(X),
Figure PCTCN2022086183-appb-000031
其中Ar、A、B、R 1、R 11的定义如上文中所述。
在另一优选例中,式(IX)或式(X)中
结构片段
Figure PCTCN2022086183-appb-000032
选自下组基团:
Figure PCTCN2022086183-appb-000033
Ar选自下组基团:
Figure PCTCN2022086183-appb-000034
“*”表示手性中心;
Figure PCTCN2022086183-appb-000035
表示结构片段
Figure PCTCN2022086183-appb-000036
或Ar与式(IX)或式(X)中其它结构的连接位点。在另一优选例中,式(I)为式(XI),
Figure PCTCN2022086183-appb-000037
“*”表示手性中心;
R 11选自氢、卤素、C 1-4烷基;
R 11’选自氢、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷氧基、或CN;
Ar和R的定义如上文中所述。
在另一优选例中,式(I)为式(XII),
Figure PCTCN2022086183-appb-000038
“*”表示手性中心;
R 11和R 11’的定义如上文中所述;
R 13、R 14、k、p的定义如上文中所述。
在另一优选例中,式(I)为式(XIII),
Figure PCTCN2022086183-appb-000039
“*”表示手性中心;
R 11选自氢、卤素、C 1-4烷基;
X选自N或CR 11’;其中,R 11’选自氢、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷氧基、或CN;
R 13选自卤素、C 1-4烷基、=O、羟基、或CN;
R 14选自卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、NR 2R 2、CN、SR 2;其中,各个R 2各自独立为氢或C 1-4烷基;
R 14’选自氢、-OC(O)R k、-O-P(O)(OR m) 2、或-O-CH(R n)-O-P(O)(OR m) 2;其中,R k选自C 1- 12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢或C 1-4烷基;R n选自氢或C 1-4烷基;
R 17选自氢或-CO(O)-CH(R n)-O-C(O)-U;其中,R n选自氢或C 1-4烷基;U选自C 1-18烷基;
k选自0、1、或2;
p选自0、1、2、3、或4。
本发明的另一方面,提供了一种如下式(XIV)所示结构的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物:
Figure PCTCN2022086183-appb-000040
式(XIV)中:
“*”表示手性中心;
R 11选自氢、卤素、C 1-4烷基;
X选自N或CR 11’;其中,R 11’选自氢、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷氧基、或CN;
R 18选自卤素、C 1-4烷基、或C 1-4烷氧基;
Ar和R的定义如上文中所述。
在另一优选例中,所述的式(I)化合物选自下组:
Figure PCTCN2022086183-appb-000041
Figure PCTCN2022086183-appb-000042
Figure PCTCN2022086183-appb-000043
Figure PCTCN2022086183-appb-000044
Figure PCTCN2022086183-appb-000045
Figure PCTCN2022086183-appb-000046
Figure PCTCN2022086183-appb-000047
Figure PCTCN2022086183-appb-000048
Figure PCTCN2022086183-appb-000049
Figure PCTCN2022086183-appb-000050
Figure PCTCN2022086183-appb-000051
Figure PCTCN2022086183-appb-000052
上述结构式中“*”表示手性中心,可以任选地为R构型或S构型,或R构型和S构型的混合物;用
Figure PCTCN2022086183-appb-000053
键连接的碳原子可以任选地为R构型或S构型,或任选地为顺式或反式。
本发明的第二方面,提供了一种药物组合物,所述药物组合物包含本发明第一方面所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,以及药学上可接受的载体。
本发明的第三方面,提供了一种如本发明第一方面所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物的用途,其用于制备治疗与KRAS G12D活性或表达量相关的疾病,病症或病状的药物组合物。
在另一优选例中,所述疾病,病症或病状选自下组:胰腺癌、非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、结肠癌、结直肠癌、甲状腺癌、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、黑色素瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征多发性、骨髓癌等各种实体瘤和血液瘤。
具体实施方式
本发明人经过长期而深入的研究,意外地发现了一类结构新颖的KRAS G12D抑制剂,以及它们的制备方法和应用。本发明化合物可以应用于与所述小GTP酶的活性相关的各种疾病的治疗。基于上述发现,发明人完成了本发明。
术语
除特别说明之处,本文中提到的“或”具有与“和/或”相同的意义(指“或”以及“和”)。
除特别说明之处,本发明的所有化合物之中,各手性碳原子(手性中心)可以任选地为R构型或S构型,或R构型和S构型的混合物。
如本文所用,在单独或作为其他取代基一部分时,术语“烷基”指只含碳原子的直链(即,无支链)或支链饱和烃基,或直链和支链组合的基团。当烷基前具有碳原子数限定(如C 1-10)时,指所述的烷基含有1-10个碳原子。例如,C 1-8烷基指含有1-8个碳原子的烷基,包括甲基、乙基、丙基、异丙基、丁基、异丁基、仲丁基、叔丁基、或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“烯基”是指直链或支链,具有至少一个碳-碳双键的碳链基团。烯基可以是取代的或未取代的。当烯基前具有碳原子数限定 (如C 2-8)时,指所述的烯基含有2-8个碳原子。例如,C 2-8烯基指含有2-8个碳原子烯基,包括乙烯基、丙烯基、1,2-丁烯基、2,3-丁烯基、丁二烯基、或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“炔基”是指具有至少一个碳-碳三键的脂肪族碳氢基团。所述的炔基可以是直链或支链的,或其组合。当炔基前具有碳原子数限定(如C 2-8炔基)时,指所述的炔基含有2-8个碳原子。例如,术语“C 2-8炔基”指具有2-8个碳原子的直链或支链炔基,包括乙炔基、丙炔基、异丙炔基、丁炔基、异丁炔基、仲丁炔基、叔丁炔基、或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“环烷基”指具有饱和的或部分饱和的单元环,二环或多环(稠环、桥环或螺环)环系基团。当某个环烷基前具有碳原子数限定(如C 3-10)时,指所述的环烷基含有3-10个碳原子。在一些优选实施例中,术语“C 3-8环烷基”指具有3-8个碳原子的饱和或部分不饱和的单环或二环烷基,包括环丙基、环丁基、环戊基、环庚基、或类似基团。“螺环烷基”指单环之间共用一个碳原子(称螺原子)的二环或多环基团,这些可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。“稠环烷基”指系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳二环或多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。“桥环烷基”指任意两个环共用两个不直接连接的碳原子的全碳多环基团,这些可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。所述环烷基所含原子全部为碳原子。如下是环烷基的一些例子,本发明并不仅局限下述的环烷基。
Figure PCTCN2022086183-appb-000054
除非有相反陈述,否则下列用在说明书和权利要求书中的术语具有下述含义。“芳基”指具有共轭的π电子体系的全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,例如苯基和萘基。所述芳基环可以稠合于其它环状基团(包括饱和和不饱和环),但不能含有杂原子如氮,氧,或硫,同时连接母体的点必须在具有共轭的π电子体系的环上的碳原子上。芳基可以是取代的或未取代的。如下是芳基的一些例子,本发明并不仅局限下述的芳基。
Figure PCTCN2022086183-appb-000055
“杂芳基”指包含一个到多个杂原子(任选自氮、氧和硫)的具有芳香性的单环或多环基团,或者包含杂环基(含一个到多个杂原子任选自氮、氧和硫)与芳基稠合形成的多环基团,且连接位点位于芳基上。杂芳基可以是任选取代的或未取代的。如下是杂芳基的一些例子,本发明并不仅局限下述的杂芳基。
Figure PCTCN2022086183-appb-000056
“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其中一个或多个环原子选自氮、氧或硫,其余环原子为碳。单环杂环基的非限制性实施例包含吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基。多环杂环基指包括螺环、稠环和桥环的杂环基。“螺环杂环基”指系统中的每个环与体系中的其他环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子选自氮、氧或硫,其余环原子为碳。“稠环杂环基”指系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,而且其中一个或多个环原子选自氮、氧或硫,其余环原子为碳。“桥环杂环基”指任意两个环共用两个不直接连接的原子的多环杂环基团,这些可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,而且其中一个或多个环原子选自氮、氧或硫,其余环原子为碳。如果杂环基里同时有饱和环和芳环存在(比如说饱和环和芳环稠合在一起),连接到母体的点一定是在饱和的环上。注:当连接到母体的点在芳环上时,称为杂芳基,不称为杂环基。如下是杂环基的一些例子,本发明并不仅局限下述的杂环基。
Figure PCTCN2022086183-appb-000057
如本文所用,在单独或作为其他取代基一部分时,术语“卤素”指F、Cl、Br和I。
如本文所用,术语“取代”(在有或无“任意地”修饰时)指特定的基团上的一个或多个氢原子被特定的取代基所取代。特定的取代基为在前文中相应描述的取代基,或各实施例中所出现的取代基。除非特别说明,某个任意取代的基团可以在该基团的任何可取代的位点上具有一个选自特定组的取代基,所述的取代基在各个位置上可以是相同或不同的。环状取代基,例 如杂环基,可以与另一个环相连,例如环烷基,从而形成螺二环系,即两个环具有一个共用碳原子。本领域技术人员应理解,本发明所预期的取代基的组合是那些稳定的或化学上可实现的组合。所述取代基例如(但并不限于):C 1-8烷基、C 2-8烯基、C 2-8炔基、C 3-8环烷基、3-至12-元杂环基,芳基、杂芳基、卤素、羟基、羧基(-COOH)、C 1-8醛基、C 2-10酰基、C 2-10酯基、氨基。
为了方便以及符合常规理解,术语“任意取代”或“任选取代”只适用于能够被取代基所取代的位点,而不包括那些化学上不能实现的取代。
如本文所用,除非特别说明,术语“药学上可接受的盐”指适合与对象(例如,人)的组织接触,而不会产生不适度的副作用的盐。在一些实施例中,本发明的某一化合物的药学上可接受的盐包括具有酸性基团的本发明的化合物的盐(例如,钾盐,钠盐,镁盐,钙盐)或具有碱性基团的本发明的化合物的盐(例如,硫酸盐,盐酸盐,磷酸盐,硝酸盐,碳酸盐)。
用途:
本发明提供了一类式(I)化合物,或它们的氘代衍生物、它们的盐、异构体(对映异构体或非对映异构体,如果存在的情况下)、水合物、可药用载体或赋形剂用于抑制KRAS G12D的用途。
本发明化合物可用作一种KRAS G12D抑制剂。
本发明是KRAS G12D的单一抑制剂,通过调节KRAS G12D的活性达到预防、缓解或治愈疾病的目的。所指疾病包括胰腺癌、非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、结肠癌、结直肠癌、甲状腺癌、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、黑色素瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征多发性、骨髓癌等各种实体瘤和血液瘤。
可将本发明化合物及其氘代衍生物,以及药学上可接受的盐或其异构体(如果存在的情况下)或其水合物和/或组合物与药学上可接受的赋形剂或载体配制在一起,得到的组合物可在体内给予哺乳动物,例如男人、妇女和动物,用于治疗病症、症状和疾病。组合物可以是:片剂、丸剂、混悬剂、溶液剂、乳剂、胶囊、气雾剂、无菌注射液。无菌粉末等。一些实施例中,药学上可接受的赋形剂包括微晶纤维素、乳糖、柠檬酸钠、碳酸钙、磷酸氢钙、甘露醇、羟丙基-β-环糊精、β-环糊精(增加)、甘氨酸、崩解剂(如淀粉、交联羧甲基纤维素钠、复合硅酸盐和高分子聚乙二醇),造粒粘合剂(如聚乙烯吡咯烷酮、蔗糖、明胶和阿拉伯胶)和润滑剂(如硬脂酸镁、甘油和滑石粉)。在优选的实施方式中,所述药物组合物是适于口服的剂型,包括但不限于片剂、溶液剂、混悬液、胶囊剂、颗粒剂、粉剂。向患者施用本发明化合物或药物组合物的量不固定,通常按药用有效量给药。同时,实际给予的化合物的量可由医师根据实际情况决定,包括治疗的病症、选择的给药途径、给予的实际化合物、患者的个体情况等。本发明化合物的剂量取决于治疗的具体用途、给药方式、患者状态、医师判断。本发明化合物在药物组合物中的比例或浓度取决于多种因素,包括剂量、理化性质、给药途径等。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。
化合物的通用合成方法
缩写及化学试剂
Ac=乙酰基
Boc=叔丁氧羰基
t-BuOK=叔丁醇钾
Catacxium-A-Pd-G3=甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)
m-CPBA=3-氯过氧苯甲酸
DAST=二乙胺基三氟化硫
DCM=二氯甲烷
1,4-dioxane=1,4-二氧六环
DIPEA or DIEA=N,N-二异丙基乙胺
DIBAL-H=二异丁基氢化铝
DMAP=4-二甲基氨基吡啶
DMF=N,N-二甲基甲酰胺
DMSO=二甲基亚砜
EtOAc or EA=乙酸乙酯
HATU=N,N,N′,N′-四甲基-O-(7-氮杂苯并三唑-1-基)六氟磷酸脲)
LiHMDS=双三甲基硅基胺基锂
Me=甲基
NMP=N-甲基吡咯烷酮
MOM=甲氧基甲基
Ph=苯基
Py=吡啶
i-PrOH=异丙醇
TBAF=四正丁基氟化铵
TIPS=三异丙基硅基
TEA=三乙胺
Tf=三氟甲磺酰基
TFA=三氟乙酸
THF=四氢呋喃
TMS=三甲基硅基
TsOH=对甲苯磺酸
本发明化合物式(I-A)、(I-B)、(I-C)可以通过以下方法制备得到:
反应式1:
Figure PCTCN2022086183-appb-000058
化合物(Ia)在惰性溶剂中于碱性条件下经取代反应得到中间体(Ib),中间体(Ib)在惰性溶剂中于碱性条件下经取代反应得到中间体(Ic),接下来中间体(Ic)与化合物(I-A-3)反应得到中间体(Id),再经过一步或者两步脱保护反应得到目标化合物。
上述反应式中Z和R的定义与上文中的表述相同。
反应式2:
Figure PCTCN2022086183-appb-000059
化合物(Ia)在惰性溶剂中与化合物(I-B-1)反应得到中间体(If),中间体(If)在惰性溶剂中于碱性条件下经取代反应得到中间体(Ig),中间体(Ig)选择性还原环外双键的到中间体(Ih),中间体(Ih)在惰性溶剂中与化合物(I-B-2)反应得到中间体(Ii),最后经过一步或者两步脱保护反应得到目标化合物。
反应式3:
Figure PCTCN2022086183-appb-000060
化合物(Ia)在惰性溶剂中于碱性条件下经取代反应得到中间体(Ik),中间体(Ik)在碱性条件下经取代反应得到中间体(Il),中间体(Il)在惰性溶剂中与化合物(I-C-2)反应得到中间体(Im),再经过脱保护反应得到目标化合物。
上述反应式中R b和R c的定义与上文中Ar上的取代基表述相同。
反应式4:
Figure PCTCN2022086183-appb-000061
化合物(VIIa)与化合物(VIIb)在惰性溶剂中于碱性条件下反应得到化合物(VIIc),再经还原反应得到化合物(VIId)。化合物(Ia)与化合物(VIIe)经取代反应得到中间体(VIIf),中间体(VIIf)与化合物(VIId)在碱性条件下经取代反应得到中间体(VIIg),中间体(VIIg)在惰性溶剂中与化合物(VIIh)反应得到中间体(VIIi),再经过脱保护反应得到目标化合物。单一手性构型的目标化合物可在第一步反应中使用化合物(VIIa)的R或者S异构体,最终会得到手性为R或者S的目标化合物;也可通过手性拆分的方法拆分中间体(VIIi)然后脱去保护基团,得到手性为R或者S的目标化合物;还可以通过手性拆分化合物(VII-A)外消旋体的方法得到手性为R或者S的目标化合物。
上述反应式中R 13、R 14、k、和p的定义与权力要求9中的表述相同。
上述各式中,各基团的定义如上文中所述。各步骤的试剂和条件可以选用本领域进行该类制备方法常规的试剂或条件,在本发明的化合物结构公开后,上述选择可以由本领域技术人员根据本领域知识进行。
药物组合物和施用方法
由于本发明化合物具有优异的对KRAS G12D的抑制活性,因此本发明化合物及其各种晶型,药学上可接受的无机或有机盐,水合物或溶剂合物,以及含有本发明化合物为主要活性成分的药物组合物可用于治疗、预防以及缓解与KRAS G12D活性或表达量相关的疾病。
本发明的药物组合物包含安全有效量范围内的本发明化合物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有5-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如
Figure PCTCN2022086183-appb-000062
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨 醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。
本发明化合物可以单独给药,或者与其他药学上可接受的化合物联合给药。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选5~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
本发明的主要优点包括:
1.提供了一种如式I所示的化合物。
2.提供了一种结构新颖的KRAS G12D抑制剂,及其制备和应用,所述的抑制剂在极低浓度下即可抑制KRAS G12D的活性。
3.提供了一类治疗与KRAS G12D活性相关疾病的药物组合物。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
实施例
实施例1:化合物1的制备
Figure PCTCN2022086183-appb-000063
化合物1a参照专利WO2021041671中的方法合成。将化合物1a(60mg,0.24mmol)和N,N-二异丙基乙胺(215mg,1.66mmol)溶于二氯甲烷(3mL)中,混合液置于-40℃下边搅拌边加入化合物1b(50mg,0.24mmol)。反应混合液在-40℃下搅拌1小时。待反应完毕,加水。混合液经二氯甲烷萃取(3×15mL)。合并的有机相经 饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:乙酸乙酯=5:1)得到黄色固体产物1c(90mg,收率88%)。MS m/z 428.3[M+H] +
化合物1d通过商业购买得到。将化合物1c(60mg,0.14mmol)、化合物1d(34mg,0.21mmol)和N,N-二异丙基乙胺(54mg,0.42mmol)依次溶于1,4-二氧六环(1.5mL)中,反应混合液90℃下搅拌过夜。待反应完毕后,反应混合液减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物1e(42mg,收率54%)。MS m/z 551.5[M+H] +
化合物1f参照专利WO2021041671中的方法合成。将化合物1e(42mg,0.08mmol)、化合物1f(43mg,0.13mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(5.5mg,0.008mmol)加入反应瓶中,置换氮气3次。然后加入四氢呋喃(1.5mL)和磷酸钾水溶液(1.5M,0.15mL)。反应混合液在氮气保护下60℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物1g(33mg,收率60%)。MS m/z 721.8[M+H] +
将化合物1g(29mg,0.04mmol)溶于二氯甲烷(3mL)中,然后加入三氟乙酸(0.5mL),反应混合液室温下搅拌2小时。待反应完毕,反应液减压浓缩。所得粗品再溶于甲醇,滴加适量氨水调节pH至7左右。混合液再减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=15:1,2%氨水)得到淡黄色固体产物1(13.6mg,收率59%)。 1H NMR(500MHz,CD 3OD)δ9.13(s,1H),7.60(d,J=7.8Hz,1H),7.42-7.37(m,1H),7.34(t,J=2.2Hz,1H),7.18-7.15(m,1H),6.94-6.88(m,1H),5.65-5.49(m,1H),4.75-4.66(m,2H),4.39-4.34(m,2H),4.20(s,2H),4.05-3.83(m,3H),3.57-3.43(m,3H),2.76-2.65(m,1H),2.65-2.54(m,1H),2.47-2.41(m,1H),2.38-2.29(m,2H),2.22-2.14(m,1H),1.09-1.02(m,4H)ppm。MS m/z 577.6[M+H] +
实施例2:化合物2的制备
Figure PCTCN2022086183-appb-000064
化合物2a和化合物1f参照专利WO2021041671中的方法合成。将化合物2a(22mg,0.04mmol)、化合物1f(27mg,0.08mmol)、甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(3mg,0.004mmol)和磷酸钾水溶液(1.5M,0.08 mL)依次溶于1,4-二氧六环(1mL)中,反应液经氮气鼓泡1分钟。反应混合液于密封管中100℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色固体产物2b(27mg,收率94%)。MS m/z 721.9[M+H] +
将化合物2b(25mg,0.03mmol)溶于二氯甲烷(2mL)中,然后加入85%间氯过氧苯甲酸(8mg,0.04mmol)。反应混合液室温下搅拌1小时。待反应完毕,反应液减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=8:1)得到淡黄色固体产物2c(11mg,收率43%)。MS m/z 737.9[M+H] +
将化合物2c(10mg,0.01mmol)溶于乙腈(1.5mL)中,然后加入4M氯化氢的1,4-二氧六环溶液(0.2mL)。待反应完毕,反应液减压浓缩。所得粗品再溶于甲醇,滴加适量氨水调节pH至7左右。混合液再减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1,2%氨水)得到淡黄色固体产物2(5.3mg,收率66%)。 1H NMR(500MHz,CD 3OD)δ9.07(s,1H),7.60(d,J=8.0Hz,1H),7.42-7.36(m,1H),7.33(t,J=2.2Hz,1H),7.16(d,J=1.5Hz,1H),6.91(dd,J=13.1,7.6Hz,1H),5.72-5.53(m,1H),5.00(d,J=11.7Hz,1H),4.80-4.72(m,2H),4.53(d,J=11.8Hz,1H),4.19-3.99(m,3H),3.93(s,2H),3.84-3.79(m,2H),3.75-3.68(m,1H),3.13-3.00(m,1H),2.50-2.35(m,2H),2.29-2.21(m,3H),2.01-1.91(m,4H)ppm。MS m/z 593.7[M+H] +
实施例3:化合物3的制备
Figure PCTCN2022086183-appb-000065
将化合物3a(10g,70mmol)溶于二氯甲烷(30mL)和乙腈(30ml)的混合溶液中。反应液冷却至0℃,依次加入N,N-二异丙基乙胺(9.50g,84mmol)和溴甲基甲基醚(8.75g,70mmol),继续搅拌2小时。待原料大部分转化后,减压浓缩除去 大部分溶剂,加入乙酸乙酯,加入1N HCl溶液调节体系至弱酸性。有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩后用硅胶柱色谱纯化(石油醚:乙酸乙酯=75:25)得到化合物3b(600mg,收率4.6%)。 1H NMR(500MHz,CD 3OD)δ6.91(t,J=1.6Hz,1H),6.72(d,J=1.6Hz,2H),4.97(s,2H),3.34(s,3H)ppm。
将化合物3b(640mg,3.4mmol)溶于二氯甲烷(10mL),依次加入溶有吡啶(200mg)的二氯甲烷溶液(1mL)和化合物3c(1.24g,3.4mmol),室温下反应2小时。待原料大部分转化后,加入1N HCl溶液调节体系至弱酸性,再加入少量饱和亚硫酸氢钠溶液洗涤,水相用二氯甲烷萃取。合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩后用硅胶柱色谱纯化(石油醚:乙酸乙酯=85:15)得到化合物3d(420mg,45%)。 1H NMR(500MHz,CDCl 3)δ7.16(d,J=2.8Hz,1H),7.06(d,J=2.8Hz,1H),5.58(brs,1H),5.08(s,2H),3.47(s,3H)ppm。
将化合物3d(250mg,0.93mmol)溶于DMF(2mL),依次加入碳酸铯(630mg,1.9mmol)和溴代环丁烷(630mg,4.7mmol),80℃下反应16小时。加水淬灭反应,水相用乙酸乙酯萃取,合并后的有机相用无水硫酸钠干燥,浓缩后用硅胶柱色谱纯化(石油醚:乙酸乙酯=90:10)得到化合物3e(220mg)。 1H NMR(500MHz,CDCl 3)δ7.16(d,J=2.9Hz,1H),7.04(d,J=2.9Hz,1H),5.10(s,2H),4.53-4.47(m,1H),3.47(s,3H),2.43-2.22(m,4H),1.73(m,1H),1.44(m,1H)ppm。
将化合物3e(15mg,0.046mmol)、Pd(dppf)Cl 2·CH 2Cl 2(4mg,0.005mmol)、双联频哪醇硼酸酯(24mg,0.093mmol)、乙酸钾(15mg,0.138mmol)溶于1,4-二氧六环(1mL),在氮气保护下加热至100℃反应4小时。待原料大部分转化后,加水淬灭反应,水相用乙酸乙酯萃取2次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩后用硅胶柱色谱纯化(石油醚:乙酸乙酯=95:5)得到化合物3f(5mg,45%yield)。 1H NMR(500MHz,CDCl 3)δ7.09(d,J=3.0Hz,1H),6.92(d,J=3.0Hz,1H),5.12(s,2H),4.13–4.04(m,1H),3.47(s,3H),2.03-1.93(m,4H),1.35-1.15(m,14H)ppm。
化合物3g参照专利WO2021041671中的方法合成。将化合物3f(34mg,0.061mmol)、甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(5mg,0.0061mmol)、化合物3g(32mg,0.086mmol)在氮气保护下溶于四氢呋喃(2mL),加入磷酸钾(1.5M,0.5mL)在氮气保护下加热至60℃反应3小时。待原料大部分转化后,加水淬灭反应,水相用乙酸乙酯萃取2次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩后用硅胶柱色谱纯化(二氯甲烷:甲醇=95:5)得到化合物3h(31mg,收率37%)。MS m/z 757.5[M+H] +
将化合物3h(15mg,0.02mmol)溶于乙腈(1mL),加入氯化氢的1,4-二氧六环溶液(6M,1mL),于室温下搅拌30分钟。待原料全部转化后,减压旋干溶剂,加入2滴氨水调节pH至碱性,用二氯甲烷稀释,无水硫酸钠干燥后,用制备薄层色谱纯化得到目标产物化合物3(7mg,收率57%)。 1H NMR(500MHz,CD 3OD)δ9.05(s,1H),6.97(d,J=2.9Hz,1H),6.79(d,J=2.9Hz,1H),5.38-5.27(m,1H),4.60(t,J=12.0Hz,2H),4.29(dd,J=34.1,10.6Hz,2H),4.16-4.06(m,1H),3.74-3.66(m,3H),3.35(d,J=14.1Hz,1H),3.30-3.22(m,3H),3.04(td,J=9.7,5.8Hz,1H),2.40-2.13(m,3H),2.07–2.00(m,2H),1.88(m,4H),1.80-1.69(m,3H),1.41(m,2H),1.29-1.19(m,2H)ppm。
实施例4:化合物4、4S、4R的制备
Figure PCTCN2022086183-appb-000066
化合物4b采用文献(European Journal of Organic Chemistry,2015,vol.2015,#4,p.871-875)中的方法合成。将叔丁醇钾(851mg,7.58mmol)溶于DMF(10mL)中,氮气氛围下冷却至-50℃,滴加化合物4a(890mg,4.21mmol,外消旋体)和化合物4b(733mg,3.79mmol)的DMF(5mL)溶液,反应液缓慢升温至室温搅拌0.5小时。将反应液以饱和氯化铵水溶液(5mL)淬灭,再加入浓盐酸(3mL),室温搅拌1小时,加水(30mL),以乙酸乙酯(30mL×3)萃取三次,合并的有机层以饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经硅胶柱层析分离纯化得淡黄色油状物化合物4c(188mg,18%)。 1H NMR(500MHz,CDCl 3)δ4.39-4.33(m,1H),4.23(q,J=7.1Hz,2H),3.78-3.72(m,1H),3.16-3.11(m,1H),2.85-2.76(m,1H),2.66-2.60(m,1H),2.53-2.46(m,1H),2.43-2.36(m,1H),2.19-2.11(m,1H),1.29(t,J=7.1Hz,3H)。MS m/z 246.3[M+H] +
将氢化铝锂(58mg,0.77mmol)溶于四氢呋喃(1mL)中,冰浴、氮气氛围下加入化合物4c(188mg,0.77mmol)的四氢呋喃(2mL)溶液。反应液65℃加热搅拌3小时。反应液冰浴下依次加入水(0.06mL),15%氢氧化钠(0.06mL),水(0.18mL),室温搅拌15分钟,再以无水硫酸钠干燥,过滤,滤液减压浓缩,经硅胶柱层析分离纯化得淡黄色油状物化合物4d(38mg,26%)。
化合物4e参照专利WO2021041671中的方法合成。将化合物4e(80mg,0.19mmol),化合物4d(38mg,0.20mmol)溶于二氧六环(1mL)中,再加入二异丙基乙基胺(72mg,0.56mmol),反应液于氮气氛围下90℃加热搅拌24小时。将反应液减压浓缩,经硅胶柱层析分离纯化得类白色固体化合物4f(33mg,30%)。MS m/z 581.5,583.4[M+H] +
化合物4g参照专利WO2021041671中的方法合成。将化合物4f(29mg, 0.05mmol),化合物4g(33mg,0.10mmol),磷酸钾(32mg,0.15mmol),甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(4mg,0.005mmol)溶于1,4-二氧六环/水(0.5mL/0.1mL)混合溶液中。反应混合物氮气氛围下90℃加热搅拌2小时。将反应液经硅藻土过滤,滤液减压浓缩,经硅胶柱层析分离纯化得白色固体化合物4h(27mg,72%)。MS m/z 751.5[M+H] +
将化合物4h(27mg,0.36mmol)溶于乙腈(0.5mL)中,滴加4M氯化氢的1,4-二氧六环(0.2mL),反应液室温搅拌1小时。将反应液减压浓缩,以氨水调节pH至7~8,再减压浓缩,所得粗品经制备型薄层板分离纯化得白色固体化合物4(13mg,60%,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.04(s,1H),7.59(d,J=8.3Hz,1H),7.41-7.36(m,1H),7.33(brs,1H),7.16(d,J=1.7Hz,1H),6.91(dd,J=13.0,7.6Hz,1H),4.71-4.64(m,2H),4.35(dd,J=43.8,10.8Hz,2H),3.87-3.72(m,5H),3.46(d,J=14.1Hz,1H),3.20-3.14(m,1H),2.81(d,J=15.8Hz,1H),2.76-2.71(m,1H),2.53(d,J=16.0Hz,1H),2.18-2.09(m,1H),2.04-1.97(m,1H),1.96-1.83(m,6H)ppm。 19F NMR(471MHz,CD 3OD)δ-92.24–-93.42(m),-115.68–-115.86(m),-140.74(d,J=2.7Hz)ppm。MS m/z 607.7[M+H] +。化合物4(外消旋体)用手性柱拆分得到化合物4S和4R。
实施例5:化合物5的制备
Figure PCTCN2022086183-appb-000067
化合物5d的合成参照化合物1g的合成方法。
将化合物5d(19mg,0.026mmol)溶于乙腈(2mL)中,然后加入4M氯化氢的1,4-二氧六环溶液(0.2mL)。反应混合液室温下搅拌1小时。待反应完毕,反应液减压浓缩。所得粗品再溶于甲醇,滴加适量氨水调节pH至7左右。混合液再减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=15:1,2%氨水)得到淡黄色固体产物5(13.8mg,收率90%)。 1H NMR(500MHz,CD 3OD)δ8.94(s,1H),7.59(d,J=8.0Hz,1H),7.41-7.35(m,1H),7.32(t,J=2.1Hz,1H),7.14(d,J=2.3Hz,1H),6.94-6.87(m,1H),5.38-5.21(m,1H),4.68-4.55(m,2H),4.35-4.16(m,4H),3.29-3.14(m,4H),3.06-2.97(m,4H),2.37-1.85(m,10H)ppm。MS m/z 591.5[M+H] +
实施例6:化合物6的制备
Figure PCTCN2022086183-appb-000068
化合物6的合成参照化合物1的合成方法,得到黄色固体化合物6(9.32mg)。 1H NMR(500MHz,CD 3OD)δ9.18(s,1H),7.59(d,J=7.8Hz,1H),7.41-7.35(m,1H),7.32(t,J=2.2Hz,1H),7.14(d,J=2.4Hz,1H),6.93-6.88(m,1H),5.37-5.24(m,1H),4.48-4.44(m,1H),4.35-4.23(m,2H),3.87-3.80(m,2H),3.29-3.15(m,3H),3.05-2.96(m,1H),2.37-2.17(m,8H),2.14-2.05(m,3H),2.03-1.95(m,2H),1.93-1.83(m,1H)ppm。MS m/z 591.7[M+H] +
实施例7:化合物7的制备
Figure PCTCN2022086183-appb-000069
化合物7的合成参照化合物1的合成方法,得到白色固体化合物7(4.19mg)。 1H NMR(500MHz,CD 3OD)δ9.16(s,1H),7.59(d,J=8.3Hz,1H),7.42-7.36(m,1H),7.33(t,J=2.0Hz,1H),7.16(d,J=2.3Hz,1H),6.94-6.88(m,1H),5.41-5.24(m,1H),5.22-5.10(m,1H),4.32(dd,J=31.5,10.6Hz,2H),4.00-3.91(m,2H),3.46(s,3H),3.37-3.32(m,1H)3.28-3.20(m,2H),3.11-3.00(m,1H),2.68-2.56(m,2H),2.39-1.83(m,12H)ppm。MS m/z 605.6[M+H] +
实施例8:化合物8的制备
Figure PCTCN2022086183-appb-000070
化合物8c的合成参照化合物1e的合成方法。
将化合物8c(30mg,0.05mmol)、化合物1f(25mg,0.07mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(8mg,0.01mmol)和碳酸铯(52mg,0.16mmol)溶于1,4-二氧六环/水(1.2/0.4mL)混合溶液中,反应液经氮气鼓泡1分钟。反应混合液于密封管中90℃搅拌过夜。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=30:1)得到无色油状化合物8d(9mg,收率23%)。MS m/z 735.9[M+H] +
参照化合物5的合成方法,得到白色固体化合物8(2.91mg)。 1H NMR(500MHz,CD 3OD)δ9.16(s,1H),7.59(d,J=8.3Hz,1H),7.42-7.36(m,1H),7.33(t,J=2.1Hz,1H),7.16(d,J=2.4Hz,1H),6.95-6.89(m,1H),5.39-5.24(m,1H),4.87-4.78(m,2H),4.37-4.25(m,2H),3.89-3.77(m,2H),3.36-3.32(m,2H),3.29-3.20(m,3H),3.18-3.12(m,2H),3.06-3.00(m,1H),2.38-2.28(m,1H),2.27-2.07(m,6H),2.04-1.97(m,2H),1.95-1.85(m,1H)ppm。MS m/z 591.6[M+H] +
实施例9:化合物9的制备
Figure PCTCN2022086183-appb-000071
化合物9a参照专利WO2021041671中的方法合成。化合物9b的合成参照化合物 8d的合成方法。
将化合物9b(14mg,0.015mmol)溶于乙腈(2mL)中,然后加入4M氯化氢的1,4-二氧六环溶液(0.3mL)。反应混合液在0℃下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物9c(19mg,收率100%)。MS m/z 753.8[M+H] +
化合物9c(19mg,0.025mmol)溶于DMF(0.5mL)中,再加入氟化铯(38.2mg,0.25mmol),反应混合物室温搅拌2小时。反应混合物加水,经乙酸乙酯萃取(3×5mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物9(4.02mg)。 1H NMR(500MHz,CD 3OD)δ9.11(s,1H),7.82(d,J=7.6Hz,1H),7.50(d,J=6.1Hz,1H),7.41-7.37(m,1H),7.33(d,J=2.5Hz,1H),7.15(d,J=2.5Hz,1H),5.39-5.23(m,1H),4.85-4.75(m,2H),4.36-4.23(m,2H),3.88-3.77(m,2H),3.36-3.33(m,1H),3.29-3.20(m,4H),3.18-3.10(m,2H),3.07-2.99(m,2H),2.37-2.20(m,2H),2.16-1.96(m,7H),1.94-1.86(m,1H)ppm。MS m/z 597.6[M+H] +
实施例10:化合物10、10S、10R的制备
Figure PCTCN2022086183-appb-000072
将化合物4f(18mg,0.03mmol)、化合物9a(30mg,0.06mmol)、磷酸钾(19mg,0.09mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(2.1mg,0.003mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物10a(21mg,收率75%)。MS m/z 913.9[M+H] +
将化合物10a(21mg,0.02mmol)溶于乙腈(1mL)中,然后加入4M氯化氢的1,4-二氧六环溶液(0.3mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物10b(18mg,收率100%)。MS m/z 768.8[M+H] +
将化合物10b(18mg,0.02mmol)溶于DMF(0.5mL)中,再加入氟化铯(30mg,0.20mmol),反应混合物室温搅拌2小时。反应混合物加水,经乙酸乙酯萃取(3×5mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物10(5mg,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.00(s,1H),7.83-7.79(m,1H),7.50(dd,J=7.1,1.1Hz,1H),7.40-7.37(m,1H),7.32(d,J=2.5Hz,1H),7.15(d,J=2.5 Hz,1H),4.69-4.55(m,2H),4.38-4.25(m,2H),3.83-3.77(m,1H),3.76-3.63(m,4H),3.47-3.41(m,1H),3.19-3.12(m,1H),3.02(s,1H),2.82-2.77(m,1H),2.74-2.67(m,1H),2.55-2.48(m,1H),2.1-2.09(m,1H),2.04-1.96(m,1H),1.94-1.79(m,6H)ppm。MS m/z 613.7[M+H] +。化合物10(外消旋体)用手性柱拆分得到化合物10S和10R。
实施例11:化合物11、11S、11R的制备
Figure PCTCN2022086183-appb-000073
参照实施例4的方法,化合物11、11S、11R通过化合物4f与11a(参照专利WO2021041671中的方法合成)反应制备而得。
实施例12:化合物12、12S、12R的制备
Figure PCTCN2022086183-appb-000074
化合物12a参照专利WO2021041671中的方法制备。
将化合物4f(16mg,0.03mmol)、化合物12a(27mg,0.06mmol)、磷酸钾(19mg,0.09mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(2.1mg,0.003mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物12b(19mg,收率70%)。MS m/z 871.9[M+H] +
将化合物12b(19mg,0.02mmol)溶于乙腈(1mL)中,然后加入4M氯化氢的1,4-二氧六环溶液(0.3mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物12c(17mg,收率100%)。MS m/z 771.8[M+H] +
将上一步得到的化合物12c(17mg)溶于DMF(0.5mL)中,再加入氟化铯(33mg,0.22mmol),反应混合物室温搅拌2小时。反应混合物加水,经乙酸乙酯萃取(3×5mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=10:1)得到白色固体化合物12(5mg,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.02(s,1H),8.12-8.09(m,2H),7.68-7.62(m,2H),7.44(t,J=8.5Hz,1H),4.65-4.58(m,2H),4.38-4.25(m,2H),3.83-3.77(m,1H),3.75-3.63(m,4H),3.45-3.40(m,2H),3.17-3.10(m,1H),2.83-2.76(m,1H),2.71-2.67(m,1H),2.54-2.48(m,1H),2.14-2.09(m,1H),2.03-1.95(m,1H),1.94-1.84(m,4H),1.83-1.78(m,2H)ppm。MS m/z 615.6[M+H] +。化合物12(外消旋体)用手性柱拆分得到化合物12S和12R。
实施例13:化合物13、13S、13R的制备
Figure PCTCN2022086183-appb-000075
化合物13a参照专利WO2021041671中的方法制备。
将化合物4f(31mg,0.05mmol)、化合物13a(22mg,0.08mmol)、磷酸钾(32mg,0.15mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(3.5mg,0.005mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下60℃搅拌1小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物13b(5.2mg,收率14%)。MS m/z 691.7[M+H] +
将化合物13b(5.2mg,0.007mmol)溶于乙腈(1mL)中,然后加入4M氯化氢的1,4-二氧六环溶液(0.2mL)。反应混合液在室温下搅拌0.5小时。待反应完毕,反应液减压浓缩,经硅胶柱层析分离纯化(二氯甲烷:甲醇=15:1)得到类白色固体产物13(4.1mg,收率92%,外消旋体)。LCMS:MS m/z 591.6[M+H] +,纯度100%(254nm)。化合物13(外消旋体)用手性柱拆分得到化合物13S和13R。
实施例14:化合物14、14S、14R的制备
Figure PCTCN2022086183-appb-000076
化合物14a参照专利WO2021041671中的方法制备。
将化合物4f(16mg,0.03mmol)、化合物14a(15mg,0.05mmol)、磷酸钾(19mg,0.09mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(2.1mg,0.003mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物14b(12mg,收率60%)。MS m/z 719.8[M+H] +
将化合物14b(12mg,0.017mmol)溶于乙腈(1mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,0.3mL)。反应混合液在室温下搅拌0.5小时。待反应完毕,反应液减压浓缩,经硅胶柱层析分离纯化(二氯甲烷:甲醇=15:1)得到类白色固体产物14(7mg,收率70%,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.07(s,1H),8.05(dd,J=8.1,1.1Hz,1H),7.92(dd,J=9.0,6.0Hz,1H),7.57-7.52(m,1H),7.46(d,J=6.9Hz,1H),7.36(t,J=9.4Hz,1H),4.67-4.59(m,2H),4.39-4.27(m,2H),3.83-3.77(m,1H),3.76-3.66(m,4H),3.45-3.40(m,1H),3.17-3.12(m,1H),2.83-2.77(m,1H),2.73-2.67(m,1H),2.59-2.49(m,2H),2.30-2.21(m,1H),2.16-2.10(m,1H),2.04-1.96(m,1H),1.94-1.77(m,6H),0.82(t,J=7.4Hz,3H)ppm。MS m/z 619.7[M+H] +。化合物14(外消旋体)用手性柱拆分得到化合物14S和14R。
实施例15:化合物15的制备
Figure PCTCN2022086183-appb-000077
化合物15的合成参照化合物1的合成方法,得到白色固体化合物15(8.5mg)。 1H NMR(500MHz,CD 3OD)δ9.03(s,1H),7.59(d,J=8.4Hz,1H),7.44-7.34(m,1H),7.34-7.30(m,1H),7.18-7.14(m,1H),6.93-6.89(m,1H),5.39-5.24(m,1H),4.64-4.55(m,2H),4.34-4.25(m,2H),3.36-3.33(m,1H),3.29-3.22(m,3H),3.10-2.98(m,4H),2.40-2.12(m,3H),2.06-1.96(m,2H),1.95-1.86(m,1H),1.20(d,J=5.0Hz,6H)ppm。MS m/z 579.7[M+H] +
实施例16:化合物16的制备
Figure PCTCN2022086183-appb-000078
化合物16的合成参照化合物1的合成方法,得到白色固体化合物16(5.86mg)。 1H NMR(500MHz,CD 3OD)δ9.12(s,1H),7.59(d,J=8.2Hz,1H),7.42-7.35(m,1H),7.34-7.31(m,1H),7.17(s,1H),6.92(dd,J=13.1,7.6Hz,1H),5.40-5.20(m,1H),4.38-4.23(m,2H),3.35-3.32(m,3H),3.29-3.16(m,4H),3.10-3.00(m,3H),2.39-2.10(m,3H),2.05-1.96(m,2H),1.96-1.82(m,1H),1.23-1.17(m,2H),1.09-1.00(m,2H)ppm。MS m/z 577.6[M+H] +
实施例17:化合物17的制备
Figure PCTCN2022086183-appb-000079
化合物17的合成参照化合物1的合成方法,得到白色固体化合物17(9.96mg)。 1H NMR(500MHz,CD 3OD)δ9.07(s,1H),7.59(d,J=8.2Hz,1H),7.42-7.36(m,1H),7.34-7.31(m,1H),7.17(d,J=2.4Hz,1H),6.92(dd,J=13.1,7.6Hz,1H),5.43-5.23(m,1H),4.38-4.25(m,2H),4.07-4.01(m,2H),3.96-3.86(m,2H),3.38-3.33(m,1H),3.28-3.23(m,2H),3.17-3.12(m,2H),3.09-3.02(m,1H),2.42-2.11(m,3H),2.07-1.97(m,2H),1.96-1.86(m,1H),1.26(d,J=4.3Hz,6H)ppm。MS m/z 579.6[M+H] +
实施例18:化合物18、18S、18R的制备
Figure PCTCN2022086183-appb-000080
将化合物4f(16mg,0.03mmol)、化合物18a(24mg,0.06mmol)、磷酸钾(19mg,0.09mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(2.1mg,0.003mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物18b(15mg,收率64%)。MS m/z 853.9[M+H] +
将化合物18b(15mg,0.02mmol)溶于乙腈(1mL)中,然后加入氯化氢的1,4- 二氧六环溶液(4M,0.3mL)。反应混合液在室温下搅拌1小时。反应液减压浓缩得到黄色固体产物18c(13mg,收率100%)。MS m/z 753.7[M+H] +
将上一步得到化合物18c(13mg)溶于DMF(0.5mL)中,再加入氟化铯(26mg,0.17mmol),反应混合物室温搅拌2小时。反应混合物加水,经乙酸乙酯萃取(3×5mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物18(5mg,收率54%,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.02(s,1H),8.10(dd,J=8.3,1.2Hz,1H),8.08-8.04(dd,J=8.3,1.2Hz,1H),7.74(dd,J=7.2,1.2Hz,1H),7.69-7.65(m,1H),7.59(dd,J=7.1,1.2Hz,1H),7.55-7.50(m,1H),4.71-4.59(m,2H),4.41-4.35(m,1H),4.31-4.26(m,1H),3.85-3.73(m,5H),3.49-3.43(m,1H),3.20-3.15(m,1H),3.10(s,1H),2.83-2.78(m,1H),2.76-2.69(m,1H),2.56-2.49(m,1H),2.16-2.10(m,1H),2.04-1.97(m,1H),1.97-1.84(m,6H)。MS m/z 597.6[M+H] +。化合物18(外消旋体)用手性柱拆分得可到化合物18S和18R。
实施例19:化合物19、19S、19R的制备
Figure PCTCN2022086183-appb-000081
将化合物1a(150mg,0.59mmol)和二异丙基乙胺(384mg,2.97mmol)溶于二氯甲烷(3mL)中,置于-40℃下搅拌。然后缓慢滴加化合物19a(126mg,0.59mmol)的二氯甲烷(1mL)稀释液。反应混合液在-40℃下搅拌30分钟。加水稀释,混合液经二氯甲烷(3x10mL)萃取。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:乙酸乙酯=5:1)得到黄色固体产物19b(142mg,56%)。MS m/z 428.3[M+H]+。
将化合物19b(97mg,0.23mmol),化合物4d(43mg,0.23mmol)溶于二氧六环(1mL)中,再加入二异丙基乙基胺(89mg,0.69mmol),反应液于氮气氛围下90℃加热搅拌48小时。将反应液减压浓缩,混合物经硅胶柱层析分离纯化得白色固体化合物19c(40mg,30%)。MS m/z 581.5,583.4[M+H] +
将化合物19c(17mg,0.03mmol)、化合物9a(29mg,0.06mmol)、磷酸钾(19mg,0.09mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(2.1mg,0.003mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、 水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物19d(19mg,收率71%)。MS m/z 913.8[M+H] +
将化合物19d(19mg,0.02mmol)溶于乙腈(1mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,0.3mL)。反应混合液在室温下搅拌1小时。反应液减压浓缩得到黄色固体产物19e(16mg,收率100%)。MS m/z 768.8[M+H] +
将上一步得到化合物19e(16mg)溶于DMF(0.5mL)中,再加入氟化铯(30mg,0.20mmol),反应混合物室温搅拌2小时。反应混合物加水,经乙酸乙酯萃取(3×5mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物19(5mg,收率39%,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.04(s,1H),7.82(dd,J=8.0,0.8Hz,1H),7.51(dd,J=7.1,1.1Hz,1H),7.41-7.37(m,1H),7.32(d,J=2.5Hz,1H),7.16(d,J=2.5Hz,1H),5.19-5.05(m,2H),4.37-4.24(m,2H),3.82-3.79(m,1H),3.47-3.42(m,1H),3.28-3.19(m,2H),3.18-3.12(m,1H),3.08(s,1H),2.98-2.92(m,2H),2.84-2.78(m,1H),2.74-2.69(m,1H),2.56-2.48(m,1H),2.16-2.10(m,4H),2.05-1.94(m,2H),1.95-1.86(m,2H)。MS m/z 613.7[M+H] +。化合物19(外消旋体)用手性柱拆分得可到化合物19S和19R。
实施例20:化合物20、化合物20S、化合物20R的制备
Figure PCTCN2022086183-appb-000082
将化合物4f(307mg,0.53mmol)、化合物20a(406mg,0.79mmol,参照专利WO2021041671中的方法合成或购买而得)、磷酸钾(337mg,1.59mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(38mg,0.05mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(5mL)、水(1mL)。反应混合液在氮气保护下100℃搅拌2小时。冷却至室温,加水。混合液经乙酸乙酯萃取(3×30mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物20b(486mg,收率99%)。MS m/z 931.9[M+H] +
将化合物20b(486mg,0.52mmol)溶于乙腈(10mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,3mL)。反应混合液在室温下搅拌1小时。待反应完毕,反 应液减压浓缩得到黄色固体产物20c(411mg,收率100%)。MS m/z 787.7[M+H] +
将上一步得到的化合物20c(411mg,0.52mmol)溶于DMF(10mL)中,再加入氟化铯(790mg,5.20mmol),反应混合物室温搅拌过夜。反应混合物加水,经乙酸乙酯萃取。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物20(174mg,收率53%)。化合物20采用HPLC方法使用手性柱(CHIRALPAK AD-H,ADH0CE-XG136,0.46cm I.D.×25cm L;Hexane/EtOH/DEA=60/40/0.1(V/V/V);1.0ml/min)拆分得到第一个异构体为化合物20-P1(rt=4.93min,41mg)和第二个异构体化合物20-P2(rt=6.18min,43mg)。
化合物20-P1和化合物20-P2:其中一个为化合物20S,另一个为化合物20R。
化合物20-P1: 1H NMR(500MHz,CD 3OD)δ9.02(s,1H),7.86(dd,J=9.1,5.7Hz,1H),7.35(d,J=2.5Hz,1H),7.32(t,J=8.9Hz,1H),7.20(d,J=2.5Hz,1H),4.73-4.61(m,2H),4.42-4.25(m,2H),3.90-3.75(m,5H),3.47(d,J=14.0Hz,1H),3.36(s,1H),3.21-3.15(m,1H),2.84-2.71(m,2H),2.58-2.50(m,1H),2.18-2.08(m,1H),2.06-1.85(m,7H)ppm。MS m/z 631.2[M+H] +
化合物20-P2: 1H NMR(500MHz,CD 3OD)δ9.01(s,1H),7.85(dd,J=9.1,5.7Hz,1H),7.34(d,J=2.5Hz,1H),7.32(t,J=9.0Hz,1H),7.20(d,J=2.5Hz,1H),4.71-4.59(m,2H),4.43-4.23(m,2H),3.87-3.72(m,5H),3.45(d,J=14.0Hz,1H),3.35(s,1H),3.19-3.13(m,1H),2.85-2.69(m,2H),2.56-2.47(m,1H),2.16-2.07(m,1H),2.05-1.83(m,7H)ppm。MS m/z 631.2[M+H] +
实施例21:化合物21的制备
Figure PCTCN2022086183-appb-000083
将化合物19b(150mg,0.35mmol)和化合物1d(84mg,0.53mmol)溶于1,4-二氧六环(2mL)中,再加入N,N-二异丙基乙胺(137mg,1.05mmol),反应液于氮气氛围下90℃加热搅拌18小时。将反应液减压浓缩,经硅胶柱层析分离纯化得白色固体化合物21b(120mg,62%)。MS m/z 551.2,553.2[M+H] +
将化合物21b(38mg,0.07mmol)、化合物20a(53mg,0.10mmol)、磷酸钾(44mg,0.21mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(4.2mg,0.007mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。反应液冷却至室 温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物21c(60mg,收率96%)。MS m/z 901.5[M+H] +
将化合物21c(60mg,0.07mmol)溶于乙腈(1mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,0.3mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物21d(50mg,收率100%)。MS m/z 757.5[M+H] +
将化合物21d(52mg,0.07mmol)溶于N,N-二甲基甲酰胺(1mL)中,再加入氟化铯(96mg,0.7mmol)。反应混合液在室温下搅拌12小时。反应液过滤,滤液减压浓缩,所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到淡黄色固体产物21(18mg,收率48%)。 1H NMR(500MHz,CD 3OD)δ9.04(s,1H),7.86(dd,J=9.2,5.7Hz,1H),7.35(d,J=2.5Hz,1H),7.32(t,J=8.9Hz,1H),7.21(d,J=2.5Hz,1H),5.31(d,J=54.1Hz,1H),5.12(d,J=25.4Hz,2H),4.37-4.17(m,2H),3.41(d,J=7.2Hz,1H),3.27-3.24(m,2H),3.23-3.20(m,2H),3.20-3.17(m,1H),3.06-3.00(m,1H),2.97-2.91(m,2H),2.37-2.21(m,2H),2.15-2.09(m,5H),2.05-1.96(m,2H),1.93-1.86(m,1H)ppm。MS m/z 601.1[M+H] +
实施例22:化合物22的制备
Figure PCTCN2022086183-appb-000084
将化合物1e(25mg,0.05mmol)、化合物20a(35mg,0.07mmol)、磷酸钾(32mg,0.15mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(3mg,0.005mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。反应液冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物22a(21mg,收率51%)。MS m/z 902.5[M+H] +
将化合物22a(22mg,0.02mmol)溶于乙腈(1mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,0.1mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物22b(19mg,收率100%)。MS m/z 757.5[M+H] +
将化合物21b(20mg,0.02mmol)溶于N,N-二甲基甲酰胺(1mL)中,再加入氟化铯(38mg,0.2mmol)。反应混合液在室温下搅拌12小时。反应液过滤,滤液减压浓缩,所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到淡黄色固体产物22(8mg,收率53%)。MS m/z 601.2[M+H] +1H NMR(500MHz,CD 3OD)δ8.98(s,1H),7.87-7.84(m,1H),7.35(d,J=2.5Hz,1H),7.32(t,J=9.0Hz,1H),7.21(d,J=2.5 Hz,1H),5.31(d,J=53.9Hz,1H),4.35-4.18(m,2H),4.14-4.10(m,2H),3.98(s,2H),3.38(d,J=7.3Hz,1H),3.30-3.23(m,2H),3.13(t,J=5.1Hz,2H),3.06-2.99(m,1H),2.36-2.21(m,2H),2.17-2.10(m,1H),2.05-1.96(m,2H),1.95-1.85(m,1H),0.70(s,4H)ppm。
实施例23:化合物23的制备
Figure PCTCN2022086183-appb-000085
将化合物19c(135mg,0.23mmol)、化合物20a(179mg,0.35mmol)、磷酸钾(146mg,0.69mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(17mg,0.02mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(2.5mL)、水(0.5mL)。反应混合液在氮气保护下100℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×20mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物23a(120mg,收率55%)。MS m/z 931.4[M+H] +
将化合物23a(120mg,0.13mmol)溶于乙腈(4mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,1.0mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物23b(101mg,收率100%)。MS m/z 787.7[M+H] +
将上一步得到的化合物23b(101mg,0.13mmol)溶于N,N-二甲基甲酰胺(2mL)中,再加入氟化铯(195mg,1.28mmol),反应混合物室温搅拌过夜。反应混合物加水,经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物23(48mg,收率59%,外消旋体)。 1H NMR(500MHz,CD 3OD)δ9.05(s,1H),7.86(dd,J=9.0,5.7Hz,1H),7.35(d,J=2.5Hz,1H),7.32(t,J=9.0Hz,1H),7.22(d,J=2.5Hz,1H),5.19-5.06(m,2H),4.42-4.25(m,2H),3.88-3.80(m,1H),3.50-3.43(m,1H),3.41(s,1H),3.30-3.22(m,2H),3.21-3.15(m,1H),3.04-2.95(m,2H),2.84-2.78(m,1H),2.77-2.70(m,1H),2.58-2.49(m,1H),2.20-2.07(m,4H),2.07-1.86(m,4H)ppm。MS m/z 631.1[M+H] +。化合物23(外消旋体)用手性柱拆分得可到化合物23S和23R。
实施例24:化合物24的制备
Figure PCTCN2022086183-appb-000086
将化合物4d(100mg,0.53mmol),化合物1c(204mg,0.48mmol)溶于1,4-二氧六环(1.5mL)中,再加入N,N-二异丙基乙胺(190mg,1.47mmol),反应液于氮气氛围下90℃加热搅拌48小时。将反应液减压浓缩,经硅胶柱层析分离纯化得白色固体化合物24a(127mg,41%)。 1H NMR(500MHz,CD 3OD)δ8.84(s,1H),4.33(dd,J=42.2,10.9Hz,2H),4.14-4.09(m,2H),3.95(s,2H),3.89-3.82(m,1H),3.77-3.72(m,2H),3.54-3.48(m,1H),3.24-3.17(m,1H),2.83-2.74(m,2H),2.58-2.51(m,1H),2.19-2.09(m,1H),2.07-1.99(m,1H),1.97-1.89(m,2H),1.48(s,9H),1.06-1.02(m,2H),0.93-0.89(m,2H)ppm。MS m/z 581.5,583.4[M+H] +
将化合物24a(127mg,0.22mmol)、化合物20a(168mg,0.33mmol)、磷酸钾(140mg,0.66mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(16mg,0.02mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(2.5mL)、水(0.5mL)。反应混合液在氮气保护下100℃搅拌2小时。待反应完毕,冷却至室温,加水。混合液经乙酸乙酯萃取(3×20mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物24b(117mg,收率57%)。MS m/z 931.4[M+H] +
将化合物24b(117mg,0.12mmol)溶于乙腈(4mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,1mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物24c(99mg,收率100%)。MS m/z 787.6[M+H] +
将上一步得到的化合物24c(99mg,0.12mmol)溶于N,N-二甲基甲酰胺(2mL)中,再加入氟化铯(191mg,1.26mmol),反应混合物室温搅拌过夜。反应混合物加水,经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物24(57mg,收率72%,外消旋体)。 1H NMR(500MHz,CD 3OD)δ 8.98(s,1H),7.85(dd,J=9.0,5.5Hz,1H),7.35(d,J=2.5Hz,1H),7.32(t,J=9.0Hz,1H),7.22(d,J=2.5Hz,1H),4.39-4.24(m,2H),4.16-4.07(m,2H),3.98(s,2H),3.86-3.78(m,1H),3.50-3.43(m,1H),3.39(s,1H),3.21-3.09(m,3H),2.84-2.77(m,1H),2.77-2.69(m,1H),2.57-2.48(m,1H),2.17-2.07(m,1H),2.06-1.96(m,1H),1.96-1.86(m,2H),0.75-0.65(m,4H)ppm。MS m/z 631.1[M+H] +。化合物24(外消旋体)用手性柱拆分得可到化合物24S和24R。
实施例25:化合物25的制备
Figure PCTCN2022086183-appb-000087
将化合物20(34mg,0.05mmol),N,N-二异丙基乙胺(19mg,0.15mmol)溶于二氯甲烷(2mL)中,冷却至-40℃,滴加化合物25a(13mg,0.05mmol)的二氯甲烷溶液(1mL)。反应液室温搅拌0.5小时。反应液加水,经二氯甲烷萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=20:1)得到白色固体化合物25(12mg,收率26%)。 1H NMR(500MHz,CD 3OD)δ9.05(s,1H),7.86(dd,J=9.0,5.7Hz,1H),7.36(d,J=2.5Hz,1H),7.32(t,J=9.0Hz,1H),7.20(d,J=2.5Hz,1H),5.83(s,2H),4.76-4.63(m,2H),4.59-4.41(m,4H),4.08-4.01(m,1H),3.84-3.65(m,3H),3.42-3.35(m,1H),3.34(s,1H),3.00-2.94(m,1H),2.93-2.87(m,1H),2.69-2.62(m,1H),2.39(t,J=7.3Hz,2H),2.26-2.19(m,1H),2.14-1.96(m,5H),1.89-1.83(m,2H),1.67-1.59(m,2H),1.38-1.26(m,12H),0.91-0.85(m,3H)。MS m/z 859.3[M+H] +。化合物25(外消旋体)经手性拆分可得到化合物25S和化合物25R。
实施例26:化合物26的制备
Figure PCTCN2022086183-appb-000088
将化合物25(6mg,0.007mmol),N,N-二异丙基乙胺(3mg,0.02mmol)溶于二氯甲烷(1mL)中,冷却至-40℃,滴加葵酰氯(2mg,0.01mmol)的二氯甲烷溶液(1mL)。反应液室温搅拌0.5小时,加水,经二氯甲烷萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经制备型薄层板分离纯化(二氯甲烷:甲醇=20:1)得到白色固体化合物26(4mg,收率56%)。 1H NMR(500MHz,CD 3OD)δ9.06(s,1H),8.09(dd,J=9.0,5.7Hz,1H),7.88(d,J=2.4Hz,1H),7.48(t,J=9.0Hz,1H),7.44(d,J=2.4Hz,1H),5.83(s,2H),4.74-4.63(m,2H),4.58-4.47(m,3H),4.47-4.41(m,1H),4.09-4.01(m,1H),3.84-3.73(m,2H),3.72-3.65(m,1H),3.45(s,1H),3.42-3.37(m,1H),2.99-2.93(m,1H),2.93-2.86(m,1H),2.69-2.62(m,3H),2.39(t,J=7.3Hz,2H),2.26-2.20(m,1H),2.14-2.07(m,1H),2.06-1.97(m,4H),1.89-1.83(m,2H),1.80-1.73(m,2H),1.66-1.59(m,2H),1.48-1.41(m,2H),1.38-1.23(m,22H),0.92-0.83(m,6H)。MS m/z 1013.4[M+H] +。化合物26(外消旋体)经手性拆分可得到化合物26S和化合物26R。
实施例27:化合物27的制备
Figure PCTCN2022086183-appb-000089
将化合物20b(100mg,0.11mmol)溶于二氯甲烷(3mL)中,滴加氯化氢的1,4-二氧六环溶液(4M,0.1mL)。反应液室温搅拌2小时。将反应液减压浓缩,所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=20:1)得化合物27a(67mg,收率71%)。MS m/z 887.1[M+H] +
将化合物27a(13mg,0.015mmol)、N,N-二异丙基乙胺(6mg,0.05mmol)溶于二氯甲烷(2mL)中,冷却至-40℃滴加葵酰氯(4mg,0.02mmol)。反应液室温搅拌0.5小时,加水,经二氯甲烷萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩,得到黄色固体化合物27b(16mg,收率100%)。MS m/z 1041.4[M+H] +
将化合物27b(16mg,0.015mmol)溶于乙腈(2mL)中,滴加氯化氢的1,4-二氧六环溶液(4M,0.5mL)。反应液室温搅拌3小时,减压浓缩得到黄色固体产物27c(14mg,收率100%)。MS m/z 941.4[M+H] +
将上一步得到的化合物27c(14mg,0.015mmol)溶于N,N-二甲基甲酰胺(1mL)中,再加入氟化铯(23mg,0.15mmol),反应混合物室温搅拌过夜。反应混合物加水,经乙酸乙酯萃取。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化 合物27(3mg,收率26%)。MS m/z 785.3[M+H] +1H NMR(500MHz,CD 3OD)δ9.04(s,1H),8.09(dd,J=9.0,5.7Hz,1H),7.88(d,J=2.4Hz,1H),7.48(t,J=9.0Hz,1H),7.44(d,J=2.4Hz,1H),4.75-4.65(m,2H),4.45-4.39(m,1H),4.34-4.27(m,1H),3.94-3.74(m,5H),3.47(t,J=12.5Hz,2H),3.19(dd,J=11.0,6.9Hz,1H),2.84-2.79(m,1H),2.76(d,J=8.4Hz,1H),2.65(t,J=7.4Hz,2H),2.58-2.52(m,1H),2.18-2.10(m,1H),2.09-1.85(m,7H),1.80-1.73(m,2H),1.45-1.23(m,12H),0.91-0.86(m,3H)。化合物27(外消旋体)经手性拆分可得到化合物27S和化合物27R。
实施例28:化合物28的制备
Figure PCTCN2022086183-appb-000090
将化合物27a(11mg,0.012mmol)、N,N-二异丙基乙胺(6mg,0.05mmol)溶于二氯甲烷(2mL)中,冷却至-40℃滴加乙酰氯(2mg,0.02mmol)。反应液室温搅拌0.5小时,加水,经二氯甲烷萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩,得到黄色固体化合物28a(11mg,收率100%)。MS m/z 929.6[M+H] +
将化合物28a(11mg,0.012mmol)溶于乙腈(2mL)中,滴加氯化氢的1,4-二氧六环溶液(4M,0.5mL)。反应液室温搅拌3小时,减压浓缩得到黄色固体产物28b(10mg,收率100%)。MS m/z 829.5[M+H] +
将上一步得到的化合物28b(10mg,0.012mmol)溶于N,N-二甲基甲酰胺(1mL)中,再加入氟化铯(18mg,0.12mmol),反应混合物室温搅拌过夜。反应混合物加水,经乙酸乙酯萃取。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物28(2mg,收率25%)。MS m/z 673.1[M+H] +。化合物28(外消旋体)经手性拆分可得到化合物28S和化合物28R。
实施例29:化合物29的制备
Figure PCTCN2022086183-appb-000091
将化合物27a(39mg,0.04mmol)溶于N,N-二甲基甲酰胺(1mL)中,再加入氟化铯(67mg,0.44mmol),反应混合物室温搅拌过夜。反应混合物加水,经乙酸乙酯萃取。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=10:1)得到黄色固体化合物29a(23mg,收率73%)。MS m/z 731.3[M+H] +
将化合物29a(10mg,0.014mmol)、N,N-二异丙基乙胺(6mg,0.05mmol)溶于乙腈(2mL)中,冰浴下滴加三氯氧磷(3mg,0.02mmol)。反应液冰浴搅拌10分钟,再加入饱和碳酸氢钠水溶液(0.5mL),室温搅拌1小时。反应混合物加水,经乙酸乙酯萃取。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩得到黄色固体化合物29b(10mg,收率91%)。MS m/z 811.3[M+H] +
将化合物29b(10mg,0.012mmol)溶于乙腈(2mL)中,滴加氯化氢的1,4-二氧六环溶液(4M,0.5mL)。反应液室温搅拌3小时,减压浓缩。所得粗品经制备型薄层板(二氯甲烷:甲醇=10:1)分离纯化得到黄色固体化合物29(4mg,收率45%)。MS m/z 711.1[M+H] +。将化合物29(4mg,0.005mmol)溶于乙腈(1mL),加入氢氧化钠(0.4mg,0.01mmol)水溶液(0.5mL)。反应混合物经冻干得黄色固体化合物29的二钠盐29-Na(4mg,收率100%)。MS m/z 711.1[M+H] +。化合物29(外消旋体)经手性拆分可得到化合物29S和化合物29R。
实施例30:化合物30的制备
Figure PCTCN2022086183-appb-000092
将化合物30a(100mg,0.20mmol,采用专利WO2022002102中的方法合成),化合物4d(56mg,0.30mmol),碳酸铯(193mg,0.60mmol)三乙烯二胺(22mg,0.20mmol)溶于N,N-二甲基甲酰胺(0.5mL),四氢呋喃(0.5mL)的混合溶液中,室温搅拌3小时。反应液加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=30:1)得到黄色油状产物30b(70mg,收率54%)。MS m/z 658.0&660.0[M+H] +
将化合物30b(35mg,0.05mmol)、化合物20a(41mg,0.08mmol)、磷酸钾(34mg,0.15mmol)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(3mg,0.005mmol)加入反应瓶中,置换氮气3次。然后加入1,4-二氧六环(0.5mL)、水(0.1mL)。反应混合液在氮气保护下100℃搅拌2小时。反应液冷却至室温,加水。混合液经乙酸乙酯萃取(3×10mL)。合并的有机相经饱和食盐水洗涤、无水硫酸钠干燥、过滤后减压浓缩。所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到黄色油状产物30c(30mg,收率56%)。MS m/z 964.5&965.5[M+H] +
将化合物30c(30mg,0.03mmol)溶于乙腈(1mL)中,然后加入氯化氢的1,4-二氧六环溶液(4M,0.1mL)。反应混合液在室温下搅拌1小时。待反应完毕,反应液减压浓缩得到黄色固体产物30d(26mg,收率100%)。MS m/z 820.3&822.3[M+H] +
将化合物30d(26mg,0.03mmol)溶于N,N-二甲基甲酰胺(1mL)中,再加入氟化铯(45mg,0.3mmol)。反应混合液在室温下搅拌12小时。反应液过滤,滤液减压浓缩,所得粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=25:1)得到淡黄色固体产物30(4mg,收率20%)。 1H NMR(500MHz,MeOD-d 4)δ7.84(dd,J=9.2,5.9Hz,1H),7.82(br,J=4.8Hz,1H),7.34-7.27(m,2H),7.03(d,J=2.1Hz,1H),4.52(dd,J= 34.3,13.0Hz,2H),4.39-4.32(m,1H),4.30-4.24(m,1H),3.92-3.80(m,3H),3.77-3.63(m,2H),3.47(d,J=14.1Hz,1H),3.26(s,1H),3.22-3.15(m,1H),2.81(d,J=15.7Hz,1H),2.78-2.70(m,1H),2.53(d,J=16.2Hz,1H),2.17-2.10(m,1H),2.05-1.89(m,7H)。MS m/z 664.1&666.1[M+H] +。化合物30(外消旋体)经手性拆分可得到化合物30S和化合物30R。
实施例31:3D细胞增殖抑制实验
化合物活性用AGS(人胃癌细胞系,KRAS G12D突变)、GP2D(人结肠癌细胞系,KRAS G12D突变)、HPAC(人胰腺癌细胞系,KRAS G12D突变)和AsPC1(人胰腺癌细胞系,KRAS G12D突变)3D细胞增殖抑制表示,采用Cell Titer-Glo(CTG)方法。将化合物用DMSO溶解,配置成10mM的母液,再用培养基将化合物进行相应的稀释。去掉细胞上层培养基,用DPBS冲洗一次,加入2mL的胰酶,37℃条件下放置一会,待细胞从培养皿上脱离后,加入5mL新鲜的培养基,混悬细胞,1000rpm,室温离心5分钟。去掉上层培养基,重新用5mL新鲜的培养基混悬细胞,使用Countess TM II数细胞个数。使用Echo550加入200nL的化合物到Ultra-low attachment 384孔板中,每孔再加入40μL的细胞。在37℃,5%的CO2条件下培养七天后,加入CTG,使用Envision记录信号。根据以下公式计算抑制率:Inhibition%=(Ave_H-Sample)/(Ave_H-Ave_L)x 100%。其中Ave_H代表DMSO孔平均读值,Sample代表化合物孔的平均读值,Ave_L代表10μM的阳性对照组平均读值。以浓度的log值作为X轴,百分比抑制率为Y轴,采用分析软件GraphPad Prism 5的log(inhibitor)vs.response-Variable slope拟合量效曲线,从而得出各个化合物对细胞增殖抑制的IC 50值或抑制率。部分代表性化合物的活性如表1所示。
表1细胞3D增殖抑制活性
Figure PCTCN2022086183-appb-000093
实施例32:KRAS G12D&SOS1结合实验
用100%DMSO将阳性药和待测化合物(10mM储液)3倍梯度稀释,转移2μL稀释后 的化合物到18μL的稀释液中,混合均匀。然后取2μL稀释后的化合物到384孔板中,每个样品重复两次。在1000rpm条件下离心384孔板。每孔加入4μL KRAS G12D&GTP混合液,再加入4μL SOS1,孵育15分钟。然后加入10μL的Tag-Tb和Tag-XL665,在4℃条件下孵育3小时。使用BMG读取HTRF(665nM和615nM数值)信号。以浓度的log值作为X轴,665nM/615nM作为Y轴,采用Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*HillSlope))拟合量效曲线,从而得出各个化合物对酶活性的IC 50值。部分代表性化合物的活性如表2所示。
表2 KRAS G12D&SOS1结合抑制活性
化合物 IC 50(nM)
10 <15
20 <15
21 <15
22 <15
28 <30
30 <60
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (23)

  1. 一种如下式(I)所示结构的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物:
    Figure PCTCN2022086183-appb-100001
    式(I)中:
    Ar选自芳基或杂芳基;所述的芳基或杂芳基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、C 3-6环烷基C 2-4炔基、3-至6-元杂环基C 2-4炔基、C 1-4卤代烷基C 2-4炔基、NR 2R 2、CN、SR 2、-OC(O)R k、-O-P(O)(OR m) 2、-O-CH(R n)-O-P(O)(OR m) 2;其中,各个R 2各自独立为氢或C 1-4烷基;R k选自C 1-12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢或C 1-4烷基;R n选自氢或C 1-4烷基;上面所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
    R选自5-12元杂环基,包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、=O、羟基、CN、-CO(O)-CH(R n)-O-C(O)-U;其中,R n选自氢或C 1-4烷基;U选自C 1-18烷基;
    Z选自化学键、-O-、-S-、-NR 5-、-C(R a) 2-、-C≡C-、-CR a=CR a-、-N=、-CR a=;其中,R 5选自氢或C 1-4烷基;R a选自氢、卤素、或C 1-4烷基;
    A选自化学键、-O-、-S-、-NR 6-;其中,R 6选自氢或C 1-4烷基;
    B选自-(CR 7R 8) m-、-(CR 7R 8) m-T-(CR 7R 8) m-;其中,T选自-C≡C-、-CR a=CR a-、C 3-6环烷基、3-至6-元杂环基;其中,所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、CN;R 7和R 8各自独立地选自下组:氢、卤素、或C 1-4烷基;或R 7和R 8与其相连接的碳原子一起形成C 3- 6环烷基;R a选自氢、卤素、或C 1-4烷基;各个m各自独立地选自0、1、2、或3;
    R 1选自C 3-8环烷基或4-至12-元杂环基,其中,杂环基包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、-CH 2OC(O)NR 9R 10、CN、SR 2、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、NR hR h、-(CR 7R 8) m-OR h、-(CR 7R 8) m-NR hR h、或=M取代;其中,R 9和R 10各自独立地选自氢或C 1-4烷基,或R 9和R 10与其连接的氮原子一起形成4-至-8元杂环基,此杂环基含有1或2个N原子以及0或1个选自O、S的杂原子;各个R h各自独立为氢、C 1-4烷 基、或C 1-4卤代烷基;M选自O或CR 3R 4;其中R 2、R 3、R 4、R 7、R 8、和m的定义如上所述;X和Y各自独立地选自选自N或CR 11;其中,R 11选自氢、卤素、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4炔基、C 3-6环烷基、或CN;
    前提条件是,当R 1不具有=M取代,且M选自CR 3R 4时,结构片段
    Figure PCTCN2022086183-appb-100002
    不为
    Figure PCTCN2022086183-appb-100003
    其中,各个上述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选地且各自独立地被1-3个各自独立地选自下组的取代基取代:卤素、C 1-4烷基、C 1-4卤代烷基、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、芳基、杂芳基、CN、NO 2、OR h、SR h、NR hR h、C(O)R t、C(O)OR h、C(O)NR hR h、NR hC(O)R t、NR hS(O) 2R t、或S(O) 2R t,前提条件是所形成的化学结构是稳定的和有意义的;其中,R t为C 1-4烷基、C 2-4烯基、C 2-4炔基、C 3-8环烷基、4-至8-元杂环基、芳基、或杂芳基;各个R h各自独立为氢、C 1-4烷基、或C 1-4卤代烷基;或两个R h与其连接的氮原子一起形成3-至-8元杂环基,此杂环基含有1或2个N原子以及0或1个选自O、S的杂原子;
    除非特别说明,上述的芳基为含有6-12个碳原子的芳香基团;杂芳基为5-至15-元(优选为5-至12-元)杂芳香基团。
  2. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(II):
    Figure PCTCN2022086183-appb-100004
    式(II)中:
    Ar选自芳基或杂芳基;所述的芳基或杂芳基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、C 3-6环烷基C 2-4炔基、3-至6-元杂环基C 2-4炔基、C 1-4卤代烷基C 2-4炔基、NR 2R 2、CN、SR 2、-OC(O)R k、-O-P(O)(OR m) 2、-O-CH(R n)-O-P(O)(OR m) 2;其中,各个R 2各自独立为氢或C 1-4烷基;R k选自C 1-12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢、C 1-4烷基;R n选自氢或C 1-4烷基;上面所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
    R选自5-12元杂环基,包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的杂环基任选地被一个或多个选自下组的基团取代: 卤素、C 1-4烷基、=O、羟基、CN、-CO(O)-CH(R n)-O-C(O)-U;其中,R n选自氢或C 1-4烷基;U选自C 1-18烷基;
    Z选自化学键、-O-、-S-、-NR 5-、-C(R a) 2-、-C≡C-、-CR a=CR a-、-N=、-CR a=;其中,R 5选自氢或C 1-4烷基;R a选自氢、卤素、或C 1-4烷基;
    A选自化学键、-O-、-S-、-NR 6-;其中,R 6选自氢或C 1-4烷基;
    B选自-(CR 7R 8) m-、-(CR 7R 8) m-T-(CR 7R 8) m-;其中,T选自-C≡C-、-CR a=CR a-、C 3-6环烷基、3-至6-元杂环基;其中,所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、CN;R 7和R 8各自独立地选自下组:氢、卤素、或C 1-4烷基;或R 7和R 8与其相连接的碳原子一起形成C 3- 6环烷基;R a选自氢、卤素、或C 1-4烷基;各个m各自独立地选自0、1、2、或3;
    R 1选自C 3-8环烷基或4-至12-元杂环基,其中,杂环基包括部分不饱和或饱和的单环或多环杂环基,多环杂环基包括螺环、稠环或桥环杂环基;所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、-CH 2OC(O)NR 9R 10、CN、SR 2、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、NR hR h、-(CR 7R 8) m-OR h、-(CR 7R 8) m-NR hR h、或=M取代;其中,R 9和R 10各自独立地选自氢或C 1-4烷基,或R 9和R 10与其连接的氮原子一起形成4-至-8元杂环基,此杂环基含有1或2个N原子以及0或1个选自O、S的杂原子;各个R h各自独立为氢、C 1-4烷基、或C 1-4卤代烷基;M选自O或CR 3R 4;其中R 2、R 3、R 4、R 7、R 8、和m的定义如上所述;
    R 11选自氢、卤素、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4炔基、C 3-6环烷基、或CN。
  3. 如权利要求1或2任一所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)或式(II)中结构片段
    Figure PCTCN2022086183-appb-100005
    选自下组基团:
    Figure PCTCN2022086183-appb-100006
    Figure PCTCN2022086183-appb-100007
    表示上述结构片段与式(I)或式(II)中其它结构的连接位点;
    “*”表示手性中心;
    上述基团任选地被0、1或2个R 13取代,其中R 13选自卤素、C 1-4烷基、=O、羟基、或CN。
  4. 如权利要求1-3任一所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,Ar选自下组基团:
    Figure PCTCN2022086183-appb-100008
    Figure PCTCN2022086183-appb-100009
    表示上述结构片段与式(I)或式(II)中其它结构的连接位点;
    “*”表示手性中心。
  5. 如权利要求1-4任一所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)或式(II)中结构片段
    Figure PCTCN2022086183-appb-100010
    选自下组的基团:
    Figure PCTCN2022086183-appb-100011
    “*”表示手性中心;
    Figure PCTCN2022086183-appb-100012
    表示上述结构片段与式(I)或式(II)中其它结构的连接位点。
  6. 如权利要求1-5任一所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(IIIa)或式(IIIb),
    Figure PCTCN2022086183-appb-100013
    “*”表示手性中心;
    其中Ar、A、B、R 1、R 11的定义如权利要求1所述。
  7. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(IV),
    Figure PCTCN2022086183-appb-100014
    各个R d各自独立地选自氢、C 1-4烷基、C 3-6环烷基;或二个R d与其相连接的同一个碳原子一起形成C 3-6环烷基;且至少二个R d与其相连接的同一个碳原子一起形成C 3-6环烷基;
    Ar、A、B、R 1、R 11的定义如权利要求1所述。
  8. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(V),
    Figure PCTCN2022086183-appb-100015
    R 1选自C 3-8环烷基或4-至12-元杂环基;
    R 12选自氢、卤素、C 1-4烷基、C 1-4卤代烷基、-CH 2OC(O)NR 9R 10、C 2-4烯基、C 2-4炔基、C 3-8环烷基、3-至8-元杂环基、CN、OR h、SR h、NR hR h、-(CR 7R 8) m-OR h、-(CR 7R 8) m-NR hR h
    n选自0、1、或2;
    Ar、X、Y、R、A、B、R 7、R 8、R 9、R 10、R h、和m的定义如权利要求1所述。
  9. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(VI),
    Figure PCTCN2022086183-appb-100016
    R选自下组基团:
    Figure PCTCN2022086183-appb-100017
    Ar选自下组基团:
    Figure PCTCN2022086183-appb-100018
    “*”表示手性中心;
    Figure PCTCN2022086183-appb-100019
    表示R基团与式(VI)化合物其它结构的连接位点;
    Figure PCTCN2022086183-appb-100020
    表示Ar基团与式(VI)化合物其它结构的连接位点;
    R 13选自卤素、C 1-4烷基、=O、羟基、或CN;
    R 14和R 15各自独立地选自卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、NR 2R 2、CN、SR 2、-OC(O)R k、-O-P(O)(OR m) 2、-O-CH(R n)-O-P(O)(OR m) 2;其中,各个R 2各自独立为氢或C 1-4烷基;R k选自C 1-12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢或C 1-4烷基;R n选自氢或C 1-4烷基;上面所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
    k选自0、1、或2;
    p选自0、1、2、3、4、或5;
    q选自0、1、2、3、或4;
    R 11的定义如权利要求1所述;R d的定义如权利要求7所述。
  10. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(VII),
    Figure PCTCN2022086183-appb-100021
    “*”表示手性中心;
    R 13、R 14、R 11、k、p的定义如权利要求9所述。
  11. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(VIII),
    Figure PCTCN2022086183-appb-100022
    E选自选自化学键、-O-、-S-、-NR 5-、-C≡C-、或-CR a=CR a-;
    R 16选自C 3-6环烷基或3-至6-元杂环基;所述的环烷基或杂环基任选地被一个或多个选自下组的基团取代:卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4炔基、CN、或=M取代;其中M选自O或CR 3R 4;其中R 3和R 4各自独立地选自下组:氢、氟、或C 1-4烷基;
    R、A、B、R 1、R 11、R 5、和R a的定义如权利要求1所述;R 15和q的定义如权利要求9所述。
  12. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(IX),
    Figure PCTCN2022086183-appb-100023
    其中Ar、A、B、R 1、R 11的定义如权利要求1所述。
  13. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(X),
    Figure PCTCN2022086183-appb-100024
    其中Ar、A、B、R 1、R 11的定义如权利要求1所述。
  14. 如权利要求12和13任一所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(IX)或式(X)中
    结构片段
    Figure PCTCN2022086183-appb-100025
    选自下组基团:
    Figure PCTCN2022086183-appb-100026
    Ar选自下组基团:
    Figure PCTCN2022086183-appb-100027
    “*”表示手性中心;
    Figure PCTCN2022086183-appb-100028
    表示结构片段
    Figure PCTCN2022086183-appb-100029
    或Ar与式(IX)或式(X)中其它结构的连接位点。
  15. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(XI),
    Figure PCTCN2022086183-appb-100030
    “*”表示手性中心;
    R 11选自氢、卤素、C 1-4烷基;
    R 11’选自氢、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷氧基、或CN;
    Ar和R的定义如权利要求9所述。
  16. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(XII),
    Figure PCTCN2022086183-appb-100031
    “*”表示手性中心;
    R 11和R 11’的定义如权利要求15所述;
    R 13、R 14、k、p的定义如权利要求9所述。
  17. 如权利要求1所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,式(I)为式(XIII),
    Figure PCTCN2022086183-appb-100032
    “*”表示手性中心;
    R 11选自氢、卤素、C 1-4烷基;
    X选自N或CR 11’;其中,R 11’选自氢、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷氧基、或CN;
    R 13选自卤素、C 1-4烷基、=O、羟基、或CN;
    R 14选自卤素、C 1-4烷基、C 1-4卤代烷基、羟基、C 1-4烷氧基、C 1-4卤代烷氧基、C 2-4烯基、C 2-4卤代烯基、C 2-4炔基、C 2-4卤代炔基、C 3-6环烷基、3-至6-元杂环基、C 3-6环烷基-O-、3-至6-元杂环基-O-、NR 2R 2、CN、SR 2;其中,各个R 2各自独立为氢或C 1-4烷基;
    R 14’选自氢、-OC(O)R k、-O-P(O)(OR m) 2、或-O-CH(R n)-O-P(O)(OR m) 2;其中,R k选自C 1- 12烷基,其中烷基任选地被一个或多个选自下组的基团取代:羟基、C 1-4烷氧基、NR 2R 2、C(O)OH、C(O)OC 1-2烷基、或CN;R m选自氢或C 1-4烷基;R n选自氢或C 1-4烷基;
    R 17选自氢或-CO(O)-CH(R n)-O-C(O)-U;其中,R n选自氢或C 1-4烷基;U选自C 1-18烷基;
    k选自0、1、或2;
    p选自0、1、2、3、或4。
  18. 一种如下式(XIV)所示结构的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物:
    Figure PCTCN2022086183-appb-100033
    式(XIV)中:
    “*”表示手性中心;
    R 11选自氢、卤素、C 1-4烷基;
    X选自N或CR 11’;其中,R 11’选自氢、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷氧基、或CN;
    R 18选自卤素、C 1-4烷基、或C 1-4烷氧基;
    Ar和R的定义如权利要求9所述。
  19. 如权利要求1所述的化合物,其特征在于,所述的式(I)化合物选自下组:
    Figure PCTCN2022086183-appb-100034
    Figure PCTCN2022086183-appb-100035
    Figure PCTCN2022086183-appb-100036
    Figure PCTCN2022086183-appb-100037
    Figure PCTCN2022086183-appb-100038
    Figure PCTCN2022086183-appb-100039
    Figure PCTCN2022086183-appb-100040
    Figure PCTCN2022086183-appb-100041
    Figure PCTCN2022086183-appb-100042
    Figure PCTCN2022086183-appb-100043
    Figure PCTCN2022086183-appb-100044
    Figure PCTCN2022086183-appb-100045
    上述结构式中“*”表示手性中心,可以任选地为R构型或S构型,或R构型和S构型的混合物;用
    Figure PCTCN2022086183-appb-100046
    键连接的碳原子可以任选地为R构型或S构型,或任选地为顺式或反式。
  20. 如权利要求1-19任一所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,其特征在于,所述的药学上可接受的盐选自下组:钾盐、钠盐、镁盐、钙盐、硫酸盐、盐酸盐、磷酸盐、磺酸盐,或碳酸盐。
  21. 一种药物组合物,其特征在于,包含权利要求1至20中任一项所述的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物,以及药学上可接受的载体。
  22. 一种权利要求1至20中任一项的化合物,或其光学异构体,药学上可接受的盐,前药,氘代衍生物,水合物,溶剂合物的用途,其特征在于,用于制备治疗与KRAS G12D活性或表达量相关的疾病,病症或病状的药物组合物。
  23. 如权利要求22所述的用途,其特征在于,所述疾病,病症或病状选自下组:胰腺癌、非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、结肠癌、结直肠癌、甲状腺癌、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、黑色素瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征多发性、骨髓癌等各种实体瘤和血液瘤。
PCT/CN2022/086183 2021-04-09 2022-04-11 作为kras g12d抑制剂的杂环化合物 WO2022214102A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202280027547.3A CN117715915A (zh) 2021-04-09 2022-04-11 作为kras g12d抑制剂的杂环化合物
CN202211215403.9A CN116891487A (zh) 2021-04-09 2022-09-30 作为kras g12d抑制剂的杂环化合物
PCT/CN2023/087690 WO2023198078A1 (zh) 2022-04-11 2023-04-11 作为kras g12d抑制剂的多环化合物

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
CN202110384952.8 2021-04-09
CN202110384952 2021-04-09
CN202110414411.5 2021-04-16
CN202110414411 2021-04-16
CN202110768789.5 2021-07-07
CN202110768789 2021-07-07
CN202110932970.5 2021-08-13
CN202110932970 2021-08-13
CN202111193190.X 2021-10-13
CN202111193190 2021-10-13
CN202111460701.X 2021-12-02
CN202111460701 2021-12-02

Publications (1)

Publication Number Publication Date
WO2022214102A1 true WO2022214102A1 (zh) 2022-10-13

Family

ID=83545199

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/086183 WO2022214102A1 (zh) 2021-04-09 2022-04-11 作为kras g12d抑制剂的杂环化合物

Country Status (2)

Country Link
CN (2) CN117715915A (zh)
WO (1) WO2022214102A1 (zh)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023020521A1 (en) * 2021-08-18 2023-02-23 Jacobio Pharmaceuticals Co., Ltd. Pyridine fused pyrimidine derivatives and use thereof
WO2023061294A1 (zh) * 2021-10-13 2023-04-20 再鼎医药(上海)有限公司 含氮杂环类衍生物调节剂、其制备方法及应用
WO2023072188A1 (zh) * 2021-10-29 2023-05-04 贝达药业股份有限公司 Kras g12d抑制剂及其在医药上的应用
WO2023098426A1 (zh) * 2021-12-02 2023-06-08 上海和誉生物医药科技有限公司 一种7-(萘-1-基)吡啶并[4,3-d]嘧啶衍生物及其制备和应用
WO2023103523A1 (zh) * 2021-12-09 2023-06-15 苏州浦合医药科技有限公司 取代的双环杂芳基化合物作为kras g12d抑制剂
WO2023114733A1 (en) * 2021-12-13 2023-06-22 Quanta Therapeutics, Inc. Kras modulators and uses thereof
WO2023134465A1 (zh) * 2022-01-11 2023-07-20 上海艾力斯医药科技股份有限公司 一种含氮杂环化合物、其制备方法、中间体及应用
WO2023138583A1 (zh) * 2022-01-21 2023-07-27 上海湃隆生物科技有限公司 杂环类化合物、药物组合物及其应用
WO2023179703A1 (en) * 2022-03-24 2023-09-28 Beigene , Ltd. Heterocyclic compounds, compositions thereof, and methods of treatment therewith
WO2023198078A1 (zh) * 2022-04-11 2023-10-19 杭州英创医药科技有限公司 作为kras g12d抑制剂的多环化合物
US11912723B2 (en) 2022-02-09 2024-02-27 Quanta Therapeutics, Inc. KRAS modulators and uses thereof
WO2024041589A1 (zh) * 2022-08-25 2024-02-29 上海艾力斯医药科技股份有限公司 含氮杂环化合物、其制备方法、其中间体及其应用
WO2024051852A1 (zh) * 2022-09-09 2024-03-14 上海翰森生物医药科技有限公司 含嘧啶多环类生物抑制剂、其制备方法和应用
WO2024083246A1 (en) * 2022-10-21 2024-04-25 Ascentage Pharma (Suzhou) Co., Ltd. Kras inhibitors

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102911172A (zh) * 2011-08-04 2013-02-06 上海恒瑞医药有限公司 杂芳基并嘧啶类衍生物、其制备方法和用途
WO2020146613A1 (en) * 2019-01-10 2020-07-16 Mirati Therapeutics, Inc. Kras g12c inhibitors
CN112105419A (zh) * 2018-11-09 2020-12-18 豪夫迈·罗氏有限公司 稠环化合物
WO2021041671A1 (en) * 2019-08-29 2021-03-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
WO2021257736A1 (en) * 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
WO2021259331A1 (zh) * 2020-06-24 2021-12-30 南京明德新药研发有限公司 八元含n杂环类化合物
WO2021260111A1 (en) * 2020-06-25 2021-12-30 Tolremo Therapeutics Ag Combination of a cbp/p300 bromodomain inhibitor and a kras inhibitor for the treatment of cancer
WO2022002102A1 (en) * 2020-06-30 2022-01-06 InventisBio Co., Ltd. Quinazoline compounds, preparation methods and uses thereof
WO2022015375A1 (en) * 2020-07-16 2022-01-20 Mirati Therapeutics, Inc. Kras g12d inhibitors
WO2022031952A2 (en) * 2020-08-07 2022-02-10 City Of Hope Treatments for cancers having kras mutations
WO2022047260A1 (en) * 2020-08-28 2022-03-03 Kumquat Biosciences Inc. Heterocyclic compounds and uses thereof
WO2022042630A1 (en) * 2020-08-26 2022-03-03 InventisBio Co., Ltd. Heteroaryl compounds, preparation methods and uses thereof
WO2022051569A1 (en) * 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 3-piperidinyl-pyrrolo[2,3-b]pyridines and related compounds and their use in treating medical conditions
WO2022060583A1 (en) * 2020-09-03 2022-03-24 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
WO2022061251A1 (en) * 2020-09-18 2022-03-24 Plexxikon Inc. Compounds and methods for kras modulation and indications therefor
WO2022068921A1 (zh) * 2020-09-30 2022-04-07 上海医药集团股份有限公司 一种喹唑啉类化合物及其应用

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102911172A (zh) * 2011-08-04 2013-02-06 上海恒瑞医药有限公司 杂芳基并嘧啶类衍生物、其制备方法和用途
CN112105419A (zh) * 2018-11-09 2020-12-18 豪夫迈·罗氏有限公司 稠环化合物
WO2020146613A1 (en) * 2019-01-10 2020-07-16 Mirati Therapeutics, Inc. Kras g12c inhibitors
WO2021041671A1 (en) * 2019-08-29 2021-03-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
WO2021257736A1 (en) * 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
WO2021259331A1 (zh) * 2020-06-24 2021-12-30 南京明德新药研发有限公司 八元含n杂环类化合物
WO2021260111A1 (en) * 2020-06-25 2021-12-30 Tolremo Therapeutics Ag Combination of a cbp/p300 bromodomain inhibitor and a kras inhibitor for the treatment of cancer
WO2022002102A1 (en) * 2020-06-30 2022-01-06 InventisBio Co., Ltd. Quinazoline compounds, preparation methods and uses thereof
WO2022015375A1 (en) * 2020-07-16 2022-01-20 Mirati Therapeutics, Inc. Kras g12d inhibitors
WO2022031952A2 (en) * 2020-08-07 2022-02-10 City Of Hope Treatments for cancers having kras mutations
WO2022042630A1 (en) * 2020-08-26 2022-03-03 InventisBio Co., Ltd. Heteroaryl compounds, preparation methods and uses thereof
WO2022047260A1 (en) * 2020-08-28 2022-03-03 Kumquat Biosciences Inc. Heterocyclic compounds and uses thereof
WO2022060583A1 (en) * 2020-09-03 2022-03-24 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
WO2022051569A1 (en) * 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 3-piperidinyl-pyrrolo[2,3-b]pyridines and related compounds and their use in treating medical conditions
WO2022061251A1 (en) * 2020-09-18 2022-03-24 Plexxikon Inc. Compounds and methods for kras modulation and indications therefor
WO2022068921A1 (zh) * 2020-09-30 2022-04-07 上海医药集团股份有限公司 一种喹唑啉类化合物及其应用

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023020521A1 (en) * 2021-08-18 2023-02-23 Jacobio Pharmaceuticals Co., Ltd. Pyridine fused pyrimidine derivatives and use thereof
WO2023061294A1 (zh) * 2021-10-13 2023-04-20 再鼎医药(上海)有限公司 含氮杂环类衍生物调节剂、其制备方法及应用
WO2023072188A1 (zh) * 2021-10-29 2023-05-04 贝达药业股份有限公司 Kras g12d抑制剂及其在医药上的应用
WO2023098426A1 (zh) * 2021-12-02 2023-06-08 上海和誉生物医药科技有限公司 一种7-(萘-1-基)吡啶并[4,3-d]嘧啶衍生物及其制备和应用
WO2023103523A1 (zh) * 2021-12-09 2023-06-15 苏州浦合医药科技有限公司 取代的双环杂芳基化合物作为kras g12d抑制剂
WO2023114733A1 (en) * 2021-12-13 2023-06-22 Quanta Therapeutics, Inc. Kras modulators and uses thereof
WO2023134465A1 (zh) * 2022-01-11 2023-07-20 上海艾力斯医药科技股份有限公司 一种含氮杂环化合物、其制备方法、中间体及应用
WO2023138583A1 (zh) * 2022-01-21 2023-07-27 上海湃隆生物科技有限公司 杂环类化合物、药物组合物及其应用
US11912723B2 (en) 2022-02-09 2024-02-27 Quanta Therapeutics, Inc. KRAS modulators and uses thereof
WO2023179703A1 (en) * 2022-03-24 2023-09-28 Beigene , Ltd. Heterocyclic compounds, compositions thereof, and methods of treatment therewith
WO2023198078A1 (zh) * 2022-04-11 2023-10-19 杭州英创医药科技有限公司 作为kras g12d抑制剂的多环化合物
WO2024041589A1 (zh) * 2022-08-25 2024-02-29 上海艾力斯医药科技股份有限公司 含氮杂环化合物、其制备方法、其中间体及其应用
WO2024051852A1 (zh) * 2022-09-09 2024-03-14 上海翰森生物医药科技有限公司 含嘧啶多环类生物抑制剂、其制备方法和应用
WO2024083246A1 (en) * 2022-10-21 2024-04-25 Ascentage Pharma (Suzhou) Co., Ltd. Kras inhibitors

Also Published As

Publication number Publication date
CN117715915A (zh) 2024-03-15
CN116891487A (zh) 2023-10-17

Similar Documents

Publication Publication Date Title
WO2022214102A1 (zh) 作为kras g12d抑制剂的杂环化合物
CN113544128B (zh) Kras-g12c抑制剂
WO2022171184A1 (zh) 作为sos1抑制剂的杂环化合物
CN113767103B (zh) 新型螺环类K-Ras G12C抑制剂
JP7039802B2 (ja) Rho-関連プロテインキナーゼ阻害剤、rho-関連プロテインキナーゼ阻害剤を含む医薬組成物、当該医薬組成物の調製方法及び使用
CN106687446B (zh) 作为t790m/wt-egfr的选择性和不可逆的激酶抑制剂的5-氨基-4-氨甲酰基-吡唑化合物及其用途
CN115836072A (zh) Kras g12c蛋白的抑制剂和其用途
CN113164761A (zh) α-1抗胰蛋白酶的调节剂
CA2813607C (en) Substituted pyridazine carboxamide compounds
CN113748114A (zh) 一种喹唑啉化合物及其在医药上的应用
WO2022268230A1 (zh) 作为kif18a抑制剂的化合物
WO2017202390A1 (zh) 作为fgfr4抑制剂的杂环化合物
WO2019057123A1 (zh) 作为ido抑制剂和/或ido-hdac双重抑制剂的多环化合物
EP3428159A1 (en) Crystalline forms of mesylate salt of pyridinyl amino pyrimidine derivative, preparation methods therefor, and applications thereof
WO2020042995A1 (zh) 一种高活性sting蛋白激动剂化合物
WO2021208918A1 (zh) 作为egfr抑制剂的三环化合物
WO2022002077A1 (zh) 一种芳基并芳杂环衍生物及其制备方法和用途
WO2023098832A1 (zh) 一类作为小gtp酶kras突变抑制剂的吡啶并嘧啶类衍生物
CN111212830A (zh) N-(取代磺酰基)苯甲酰胺类衍生物及其制备方法和医药用途
WO2022228576A1 (zh) 一种靶向蛋白调节剂的化合物及其应用
WO2023041055A1 (zh) Kif18a抑制剂
CN113874354A (zh) 吡啶酮类衍生物、其制备方法及其在医药上的应用
WO2019179515A1 (zh) 受体抑制剂、包含其的药物组合物及其用途
WO2022188889A1 (zh) 作为parp7抑制剂的化合物
EP4174068A1 (en) Novel compound, and pharmaceutical composition for preventing or treating resistant cancer, comprising same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22784168

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280027547.3

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22784168

Country of ref document: EP

Kind code of ref document: A1