WO2022242725A1 - 一类新型蛋白降解剂及其应用 - Google Patents

一类新型蛋白降解剂及其应用 Download PDF

Info

Publication number
WO2022242725A1
WO2022242725A1 PCT/CN2022/093956 CN2022093956W WO2022242725A1 WO 2022242725 A1 WO2022242725 A1 WO 2022242725A1 CN 2022093956 W CN2022093956 W CN 2022093956W WO 2022242725 A1 WO2022242725 A1 WO 2022242725A1
Authority
WO
WIPO (PCT)
Prior art keywords
haloalkyl
alkyl
compound
cancer
alkynyl
Prior art date
Application number
PCT/CN2022/093956
Other languages
English (en)
French (fr)
Inventor
张玉华
马欣
野国中
王兆伏
陈金聚
赵存良
陈永峰
孔莹
Original Assignee
和径医药科技(上海)有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 和径医药科技(上海)有限公司 filed Critical 和径医药科技(上海)有限公司
Priority to CN202280016739.4A priority Critical patent/CN116940581A/zh
Publication of WO2022242725A1 publication Critical patent/WO2022242725A1/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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • 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
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system

Definitions

  • the invention belongs to the field of medicinal chemistry and synthesis. It specifically relates to a new class of PROTAC molecules targeting ALK, ROS1, and EGFR proteins and their mutant proteins, their preparation methods and applications, and pharmaceutical compositions containing the compounds.
  • the ubiquitin-proteasome pathway is a common endogenous protein degradation method, in which the protein to be degraded is first modified by ubiquitination, and then decomposed into smaller polypeptides, amino acids and reusable ubiquitin by the proteasome.
  • PROTAC proteolysis tarrgeting chimeras
  • PROTAC molecules can generally be divided into three parts, one end is a small molecule fragment (war head) that binds to a specific target protein, the other end is an E3 ligase ligand (E3 ligase ligand) with ubiquitination function, and the two ends are A linker that is connected together.
  • PROTAC molecules utilize the cell's protein ubiquitination degradation pathway to selectively degrade target proteins of interest.
  • the PROTAC molecule can bind to the target protein and the E3 ligase at the same time, which promotes the ubiquitination of the target protein, and then is released by the proteasome. identified and degraded.
  • Lung cancer is a major disease that threatens human health, and the mortality rate of lung cancer ranks first among all malignant tumors.
  • EGFR Epidermal Growth Factor Receptor, epidermal growth factor receptor
  • ALK anaplastic lymphoma kinase, anaplastic lymphoma kinase
  • ROS1 ROS proto-oncogene 1 receptor tyrosine kinase, c-ros Sarcoma oncogenic factor-receptor tyrosine kinase activating mutation cases accounted for about 30%, 8% and 2% respectively. Therefore, the development of new PROTAC molecules that can be resistant to existing EGFR inhibitors, ALK inhibitors, and ROS1 inhibitors has great research value and potential practical value.
  • PROTACs In the field of PROTACs, the published PROTAC molecules based on Brigatinib or similar structures are mainly studied as ALK degradation agents (such as WO2020249048, WO2019113071, WO2019042444, WO2017204445, etc.), but there is no public disclosure of EGFR C797S Pharmacological data related to the mutation. Similarly, other published Alectinib-based ALK degraders have not publicly disclosed relevant pharmacological data for EGFR C797S mutation (such as WO2020069106, WO2019114770, WO2019196812, etc.).
  • PROTAC molecules capable of degrading EGFR, ALK or ROS1 proteins and muteins thereof.
  • the present invention provides a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate, hydrate, polymorph forms, prodrugs or isotopic variants, and mixtures thereof:
  • K is selected from the following structures:
  • L 1 is a chemical bond, C(R) 2 or SO 2 ;
  • L 2 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2.
  • L 3 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2.
  • L 4 is selected from a chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L is selected from chemical bond, O, S, NR*, C(R) 2 or SO 2 ;
  • R 1 is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl or 3 to 7 membered heterocyclyl;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • L4 and L5 can together form -C ⁇ C- only when K is K1, K2 or K4.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, and optionally a pharmaceutically acceptable excipient.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient, which also comprises other therapeutic agents.
  • kits comprising a compound of the invention, and other therapeutic agents, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the present invention provides the use of a compound of the present invention in the manufacture of a medicament for the treatment and/or prevention of cancer.
  • the invention provides a method of treating and/or preventing cancer in a subject comprising administering to said subject a compound of the invention or a composition of the invention.
  • the invention provides a compound of the invention or a composition of the invention for use in the treatment and/or prevention of cancer.
  • the cancer is selected from lung cancer; lymphoma; inflammatory myofibroblastic tumor; colorectal cancer; glioma; astroglioblastoma; ovarian cancer; bone marrow cancer; transplant-related cancer ; neutropenia; leukemia; Wengwerichter's syndrome; bronchial cancer; prostate cancer; breast cancer; thyroid cancer; pancreatic cancer; neuroblastoma; extramedullary plasmacytoma; plasmacytoma; gastric cancer; Gastrointestinal stromal tumor; esophageal cancer; colorectal adenocarcinoma; esophageal squamous cell carcinoma; liver cancer; renal cell carcinoma; bladder cancer; endometrial cancer; melanoma; brain cancer; oral cancer; sarcoma; resistance to targeted drugs or tumors or diseases that depend on ALK, ROS1 or EGFR or any of their mutant proteins.
  • said cancer is selected from small cell lung cancer; non-small cell lung cancer; diffuse large B-cell lymphoma; non-Hodgkin's lymphoma; Positive lymphoma; primary lymphoma; B-cell lymphoma; recurrent B-cell non-Hodgkin's lymphoma; recurrent diffuse large B-cell lymphoma; recurrent mediastinal (thymic) large B-cell lymphoma; primary Mediastinal (thymic) large B-cell lymphoma; relapsed transformed non-Hodgkin's lymphoma; refractory B-cell non-Hodgkin's lymphoma; refractory diffuse large B-cell lymphoma; refractory primary Mediastinal (thymus) large B-cell lymphoma; refractory transformed non-Hodgkin's lymphoma; multiple myeloma; myelodysplastic syndrome (MDS); previously treated myelody
  • said cancer is selected from the group consisting of anaplastic lymphoma kinase (ALK) mutation-positive non-small cell lung cancer (NSCLC); ROS1-positive non-small cell lung cancer; EGFR-mutated non-small cell lung cancer; lung adenocarcinoma ; Lung cancer resistant to EGFR, ROS1 or ALK-targeted drugs; Lymphoma resistant to ALK-targeted drugs; or the following tumors, cancers or diseases that depend on ALK, ROS1 or EGFR or any mutant protein thereof: lung cancer, Lymphoma, inflammatory myofibroblastic tumor, colorectal cancer, glioma, astroglioblastoma, ovarian cancer, leukemia, breast cancer, thyroid cancer, neuroblastoma, extramedullary plasmacytoma, plasmacytoma Cell tumor, esophageal squamous cell carcinoma, renal cell carcinoma, bronchial carcinoma, prostate cancer, breast cancer, thyroid cancer,
  • ALK
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1 -2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 and C 5 -6 alkyl.
  • C 1-6 alkyl means a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms. In some embodiments, C 1-4 alkyl is preferred. Examples of C 1-6 alkyl groups include: methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), t-butyl Base (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl ( C 5 ), 3-methyl-2-butyl (C 5 ), tert-amyl (C 5 ) and n-hexyl (C 6 ).
  • C 1-6 alkyl also includes heteroalkyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced by heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus) instead.
  • An alkyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 2-6 alkenyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C 2-4 alkenyl is preferred. Examples of C 2-6 alkenyl include: ethenyl (C 2 ), 1-propenyl (C 3 ), 2-propenyl (C 3 ), 1-butenyl (C 4 ), 2-butenyl (C 4 ), butadienyl (C 4 ), pentenyl (C 5 ), pentenyl (C 5 ), hexenyl (C 6 ), and the like.
  • C alkenyl also includes heteroalkenyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced by heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus) instead.
  • An alkenyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 2-6 alkynyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C 2-4 alkynyl is preferred. Examples of C 2-6 alkynyl include, but are not limited to: ethynyl (C 2 ), 1-propynyl (C 3 ), 2-propynyl (C 3 ), 1-butynyl (C 4 ), 2-butynyl (C 4 ), pentynyl (C 5 ), hexynyl (C 6 ), and the like.
  • C2-6alkynyl also includes heteroalkynyl groups in which one or more (e.g., 1, 2 , 3 , or 4) carbon atoms are replaced by heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus) instead.
  • An alkynyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • Halo or "halogen” refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
  • C 1-6 haloalkyl refers to the above-mentioned “C 1-6 alkyl", which is substituted with one or more halogen groups.
  • C 1-4 haloalkyl is particularly preferred, more preferably C 1-2 haloalkyl.
  • Exemplary haloalkyl groups include, but are not limited to: -CF 3 , -CH 2 F, -CHF 2 , -CHFCH 2 F, -CH 2 CHF 2 , -CF 2 CF 3 , -CCl 3 , -CH 2 Cl , -CHCl 2 , 2,2,2-trifluoro-1,1-dimethyl-ethyl, and the like.
  • a haloalkyl group can be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 3-10 cycloalkyl refers to a non-aromatic cyclic hydrocarbon group having 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, C 3-7 cycloalkyl and C 3-6 cycloalkyl are particularly preferred, more preferably C 5-6 cycloalkyl. Cycloalkyl also includes ring systems wherein the aforementioned cycloalkyl ring is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring, and in such cases the number of carbons continues to indicate The number of carbons in the cycloalkyl system.
  • cycloalkyl groups include, but are not limited to: cyclopropyl (C 3 ), cyclopropenyl (C 3 ), cyclobutyl (C 4 ), cyclobutenyl (C 4 ), cyclopentyl ( C 5 ), cyclopentenyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ), cycloheptyl (C 7 ), cycloheptene Cycloheptadienyl (C 7 ), Cycloheptadienyl (C 7 ), Cycloheptatrienyl (C 7 ), and the like. Cycloalkyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • 3-12 membered heterocyclyl refers to a 3 to 12 membered non-aromatic ring system group having ring carbon atoms and 1 to 5 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, Sulfur, boron, phosphorus and silicon.
  • the point of attachment may be a carbon or nitrogen atom, as valence permits.
  • 4-12 membered heterocyclyl which is a 4 to 12 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms
  • 3-10 membered Heterocyclyl which is a 3 to 10 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms
  • a 3-7 membered heterocyclyl group having ring carbon atoms and 3 to 7 membered non-aromatic ring systems with 1 to 4 ring heteroatoms
  • preferably 3-6 membered heterocyclyl which is a 3 to 6 membered nonaromatic ring system with ring carbon atoms and 1 to 3 ring heteroatoms
  • 4-8 membered heterocyclic groups which are 4 to 8 membered non-aromatic ring systems having ring carbon atoms and 1 to 3 ring heteroatoms; more preferably 5-6 membered heterocyclic groups
  • Heterocyclyl also includes ring systems wherein the aforementioned heterocyclyl ring is fused to one or more cycloalkyl groups, wherein the point of attachment is on the cycloalkyl ring, or wherein the aforementioned heterocyclyl ring is fused to one or more aryl or Heteroaryl-fused ring systems wherein the point of attachment is on the heterocyclyl ring; and in such cases, the number of ring members continues to indicate the number of ring members in the heterocyclyl ring system.
  • Exemplary 3-membered heterocyclyl groups containing one heteroatom include, but are not limited to, aziridine, oxirane, thiorenyl.
  • Exemplary 4-membered heterocyclyl groups containing one heteroatom include, but are not limited to, azetidinyl, oxetanyl, and thietanyl.
  • Exemplary 5-membered heterocyclic groups containing one heteroatom include, but are not limited to: tetrahydrofuryl, dihydrofuryl, tetrahydrothiophenyl, dihydrothiophenyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2, 5-diketone.
  • Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, but are not limited to, dioxolanyl, oxasulfuranyl, disulfuranyl, and oxasulfuranyl Oxazolidin-2-ones.
  • Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to, triazolinyl, oxadiazolinyl, and thiadiazolinyl.
  • Exemplary 6-membered heterocyclyl groups containing one heteroatom include, but are not limited to, piperidinyl, tetrahydropyranyl, dihydropyridyl, and thianyl.
  • Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, but are not limited to, piperazinyl, morpholinyl, dithianyl, dioxanyl.
  • Exemplary 6-membered heterocyclyl groups containing three heteroatoms include, but are not limited to, triazinanyl.
  • Exemplary 7-membered heterocyclyl groups containing one heteroatom include, but are not limited to, azepanyl, oxepanyl, and thiepanyl.
  • Exemplary 5-membered heterocyclyls (also referred to herein as 5,6 -bicyclic heterocyclyls) fused to C aryl rings include, but are not limited to: indolinyl, isoindolinyl , dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinone, and the like.
  • Exemplary 6 -membered heterocyclyls (also referred to herein as 6,6-bicyclic heterocyclyls) fused to a C aryl ring include, but are not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, and many more.
  • a heterocyclyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 6-10 aryl means a monocyclic or polycyclic (e.g., bicyclic) 4n+2 aromatic ring system (e.g., having shared 6 or 10 ⁇ electrons) groups.
  • an aryl group has six ring carbon atoms ("C aryl”; eg, phenyl).
  • an aryl group has ten ring carbon atoms ("C 10 aryl”; eg, naphthyl, eg, 1-naphthyl and 2-naphthyl).
  • Aryl also includes ring systems wherein the aforementioned aryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on said aryl ring, in which case the number of carbon atoms continues to indicate The number of carbon atoms in the aryl ring system.
  • An aryl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • 5-14 membered heteroaryl means a 5-14 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms (e.g., having 6, 10 or 14 ⁇ electrons), wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur.
  • the point of attachment can be a carbon or nitrogen atom, as valence permits.
  • Heteroaryl bicyclic ring systems can include one or more heteroatoms in one or both rings.
  • Heteroaryl also includes ring systems wherein the aforementioned heteroaryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on said heteroaryl ring, in which case the carbon atoms Numbers continue to indicate the number of carbon atoms in the heteroaryl ring system.
  • 5-10 membered heteroaryl is preferred, which is a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms.
  • 5-6 membered heteroaryl is particularly preferred, which is a 5-6 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms .
  • Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyrrolyl, furyl, and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, oxadiazolyl (eg, 1,2,4-oxadiazolyl), and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyridyl.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to, pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively.
  • Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to, azepanyl, oxepanyl, and thiepanyl.
  • Exemplary 5,6-bicyclic heteroaryls include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuryl , Benzisofuryl, Benzimidazolyl, Benzoxazolyl, Benzisoxazolyl, Benzoxadiazolyl, Benzthiazolyl, Benzisothiazolyl, Benzthiadiazolyl, Indenazinyl and Purinyl.
  • Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to, naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, multiplinyl, quinoxalinyl, phthalazinyl, and quinazolinyl .
  • a heteroaryl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 6-10 arylene group and “5-14 membered heteroarylene group” represent the above-mentioned “C 6-10 aryl group” and “5-14 membered heteroaryl group”, respectively, wherein another hydrogen is removed to form divalent groups, and may be substituted or unsubstituted.
  • Preferred are C 6-10 arylene and 5-10 membered heteroarylene.
  • Representative C6-10 arylene groups include 1,2-phenylene, 1,3-phenylene, 1,4-phenylene, 1,2-naphthylene, 1,3-naphthylene , 1,4-naphthylene, 1,5-naphthylene, 1,6-naphthylene, 1,7-naphthylene, 1,8-naphthylene, 2,3-naphthylene, 2 ,5-naphthylene, 2,6-naphthylene, 2,7-naphthylene.
  • Representative 5-10 membered heteroarylene groups include 1,2-pyrrolylene, 1,3-pyrrolylene, 2,3-pyrrolylene, 2,4-pyrrolylene, 2,5-pyrrolylene 3,4-pyrrolene, 2,3-furyl, 2,4-furyl, 2,5-furyl, 3,4-furyl, 2,3-thienylene, 2,4-thienylene, 2,5-thienylene, 3,4-thienylene, 1,2-imidazolyl, 1,4-imidazolyl, 1,5-imidazolyl, 2, 4-Imidazolylene, 2,5-Imidazolylene, 4,5-Imidazolylene, 1,3-Pyrazolylene, 1,4-Pyrazolylene, 1,5-Pyrazolylene, 3 ,4-pyrazolylene, 3,5-pyrazolylene, 4,5-pyrazolylene, 2,4-oxazolylene, 2,5-oxazolylene, 4,5-oxazolylene Azolyl, 3,
  • Alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, etc. are defined herein as optionally substituted groups.
  • Each of R aa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two R aa groups are combined to form a heterocyclyl or Heteroaryl rings, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently replaced by 0, 1, 2, 3, 4 or 5 R groups group replacement;
  • Each of Rcc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two Rcc groups are combined to form a heterocycle radical or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently replaced by 0, 1, 2, 3, 4 or 5 R dd group substitution;
  • cancer includes, but is not limited to, the following cancers: breast, ovary, cervix, prostate, testis, esophagus, stomach, skin, lung, bone, colon, pancreas, thyroid, biliary tract, buccal and pharynx (mouth), lips, Cancers of the tongue, oral cavity, pharynx, small intestine, colorectum, large intestine, rectum, brain and central nervous system, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, adenocarcinoma, Adenoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder cancer, liver cancer, kidney cancer, bone marrow disorders, lymphoid disorders, Hodgkin's disease, hair cell carcinoma, and leukemia.
  • cancers include, but are not limited to, HER2-positive metastatic breast cancer, HER2-overexpressed metastatic gastric adenocarcinoma or gastroesophageal junction adenocarcinoma, non-small cell lung cancer with sensitive mutations in the epidermal growth factor receptor (EGFR) gene, non-small cell lung cancer containing Locally advanced or metastatic non-small cell lung cancer with squamous histology, metastatic advanced breast cancer, castration-resistant prostate cancer with disease progression during or after platinum chemotherapy.
  • EGFR epidermal growth factor receptor
  • treating refers to reversing, alleviating, inhibiting the progression of the disorder or condition to which the term applies, or one or more symptoms of such disorder or condition.
  • the noun “treat” as used herein refers to the action of the verb treat, which is as just defined.
  • the term "pharmaceutically acceptable salt” refers to those carboxylate salts, amino acid addition salts of the compounds of the present invention, which are suitable for use in contact with patient tissues within the scope of sound medical judgment without undue toxicity, Irritation, allergic effects, etc., commensurate with a reasonable benefit/risk ratio, are valid for their intended use, including, where possible, zwitterionic forms of the compounds of the invention.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, for example alkali and alkaline earth metal hydroxides or organic amines.
  • metals used as cations are sodium, potassium, magnesium, calcium and the like.
  • suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine and procaine.
  • Base addition salts of acidic compounds may be prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt, in conventional manner.
  • the free acid may be regenerated by contacting the salt form with the acid and isolating the free acid in a conventional manner.
  • the free acid forms differ somewhat from their respective salt forms in certain physical properties, such as solubility in polar solvents, but the salts are nevertheless equivalent to their respective free acids for the purposes of the present invention.
  • Salts may be sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, and the like prepared from inorganic acids. Salts can also be prepared from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like. Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, and the like.
  • Pharmaceutically acceptable salts may include cations based on alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, etc., as well as non-toxic ammonium, quaternary ammonium and amine cations, including but not limited to ammonium, tetramethyl Ammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, etc.
  • Salts of amino acids are also contemplated, such as arginine salts, gluconate salts, galacturonate salts, etc. (see for example Berge S.M. et al., "Pharmaceutical Salts," J.Pharm.Sci., 1977; 66:1- 19, which is incorporated by reference).
  • Subjects for administration include, but are not limited to: human (i.e., male or female of any age group, e.g., pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young Adult, middle-aged adult or older adult)) and/or non-human animals, e.g., mammals, e.g., primates (e.g., cynomolgus monkeys, rhesus monkeys), cows, pigs, horses, sheep , goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "human", “patient” and “subject” are used interchangeably herein.
  • treating includes an effect on a subject suffering from a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows down the disease, disorder or the development of a disease, disorder or condition ("therapeutic treatment”) and also includes effects that occur before a subject begins to suffer from a particular disease, disorder or condition (“prophylactic treatment").
  • an "effective amount" of a compound refers to an amount sufficient to elicit a desired biological response.
  • an effective amount of a compound of the invention may vary depending on factors such as, for example, the biological target, the pharmacokinetics of the compound, the disease being treated, the mode of administration, and the condition of the subject. Age Health conditions and symptoms.
  • An effective amount includes a therapeutically effective amount and a prophylactically effective amount.
  • a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder or condition, or to induce one or more symptoms associated with the disease, disorder or condition. Amount to delay or minimize.
  • a therapeutically effective amount of a compound refers to that amount of the therapeutic agent, alone or in combination with other therapies, which provides a therapeutic benefit in the treatment of a disease, disorder or condition.
  • the term "therapeutically effective amount” can include an amount that improves overall therapy, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
  • a prophylactically effective amount of a compound is an amount sufficient to prevent a disease, disorder or condition, or to prevent one or more symptoms associated with a disease, disorder or condition, or to prevent a disease , the amount of recurrence of the disorder or condition.
  • a prophylactically effective amount of a compound refers to that amount of a therapeutic agent, alone or in combination with other agents, which provides a prophylactic benefit in the prevention of a disease, disorder or condition.
  • the term “prophylactically effective amount” may include amounts that improve overall prophylaxis, or that enhance the prophylactic effect of other prophylactic agents.
  • Combination and related terms refer to the simultaneous or sequential administration of a compound of the invention and another therapeutic agent.
  • the compounds of the invention may be administered with the other therapeutic agent simultaneously or sequentially in separate unit dosage forms, or together with the other therapeutic agent in a single unit dosage form.
  • the compound of the present invention refers to the following formula (I) compound (including sub-general formula, such as formula (II), (III-1), (V-2) etc.), its pharmaceutically acceptable Salts, enantiomers, diastereoisomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof.
  • the present invention relates to a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate, hydrate, or pharmaceutically acceptable salt thereof.
  • a compound of general formula (I) or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate, hydrate, or pharmaceutically acceptable salt thereof.
  • K is selected from the following structures:
  • L 1 is a chemical bond, C(R) 2 or SO 2 ;
  • L 2 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2.
  • L 3 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2.
  • L 4 is selected from a chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L is selected from chemical bond, O, S, NR*, C(R) 2 or SO 2 ;
  • R 1 is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl or 3 to 7 membered heterocyclyl;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • L4 and L5 can together form -C ⁇ C- only when K is K1, K2 or K4.
  • K is K1; In another specific embodiment, K is K2; In another specific embodiment, K is K3; In another specific embodiment, K is K4; In another specific embodiment, K is K4; In a specific embodiment, K is K5.
  • E is E1; In another specific embodiment, E is E2; In another specific embodiment, E is E3; In another specific embodiment, E is E4; In another specific embodiment, E is E4; In a specific embodiment, E is E5.
  • L 1 is a chemical bond; in another specific embodiment, L 1 is C(R) 2 ; in another specific embodiment, L 1 is SO 2 .
  • L 1 is selected from a chemical bond, CH 2 or SO 2 ; in another more specific embodiment, L 1 is CH 2 .
  • L is a chemical bond; in another specific embodiment, L is O ; in another specific embodiment, L is S ; in another specific embodiment, L is NR *; in another specific embodiment, L 2 is C(R') 2 ; in another specific embodiment, L 2 is C(R') 2 C(R') 2 ; in another specific embodiment In, L 2 is C(R') 2 C(R') 2 C(R') 2 ; in another specific embodiment, L 2 is C(R') 2 C(R') 2 C(R ') 2 C(R') 2 ; in another specific embodiment, L 2 is C 6-10 arylene; in another specific embodiment, L 2 is 5 to 10 membered heteroarylene.
  • L 2 is selected from a chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R ') 2 , C(R') 2 C(R') 2 C(R') 2 , C 6-10 arylene or 5 to 10 membered heteroarylene; in another more
  • L 2 is selected from a chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ; in another more specific embodiment, L 2 is selected from a chemical bond, C(R') 2 or C (R') 2 C(R') 2 ; in another more specific embodiment, L 2 is selected from the group consisting of bond, O, S, NR*, CH 2
  • L is a chemical bond; in another specific embodiment, L is O; in another specific embodiment, L is S; in another specific embodiment, L is NR *; in another specific embodiment, L 3 is C(R') 2 ; in another specific embodiment, L 3 is C(R') 2 C(R') 2 ; in another specific embodiment In, L 3 is C(R') 2 C(R') 2 C(R') 2 ; in another specific embodiment, L 3 is C(R') 2 C(R') 2 C(R ') 2 C(R') 2 ; in another specific embodiment, L 3 is C 6-10 arylene; in another specific embodiment, L 3 is 5 to 10 membered heteroarylene.
  • L 3 is selected from the group consisting of bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C( R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ; in another more specific embodiment, L 3 is selected from Chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ; in another more specific embodiment, L 3 is selected from a chemical bond, C(R') 2 or C(R') 2 C(R') 2 ; In another more specific embodiment, L 3 is selected from a chemical bond, O, S, NR*, CH 2 , CH 2 CH 2 or 1,4-triazolylene; in
  • L 4 is a chemical bond; in another specific embodiment, L 4 is C(R') 2 ; in another specific embodiment, L 4 is C(R') 2 C(R ') 2 ; in another specific embodiment, L 4 is C(R') 2 C(R') 2 C(R') 2 ; in another specific embodiment, L 4 is C(R') 2 C(R') 2 C(R') 2 .
  • L 4 is selected from the group consisting of a bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R ') 2 or C(R') 2 C(R') 2 C(R') 2 ; in another more specific embodiment, L 4 is selected from a chemical bond, C(R') 2 or C(R') 2 C(R') 2 ; in another more specific embodiment, L 4 is selected from a bond, CH 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 or CH 2 CH 2 CH 2 CH 2 ; in another more specific embodiment, L 4 is selected from a bond, CH 2 or CH 2 CH 2 ; in another more specific embodiment, L 4 is selected from a bond or CH 2 .
  • L is a chemical bond; in another specific embodiment, L is O ; in another specific embodiment, L is S ; in another specific embodiment, L is NR *; in another specific embodiment, L5 is C(R) 2 ; in another specific embodiment, L5 is SO2 .
  • L is selected from O, S, NR*, C(R) 2 or SO 2 ; in another more specific embodiment, L is selected from bond, O, S, NR * or C(R) 2 ; in another more specific embodiment, L 5 is selected from O, S, NR* or C(R) 2 ; in another more specific embodiment, L 5 is selected from O , S or NR*; in another more specific embodiment, L5 is selected from O or C(R) 2 ; in another more specific embodiment, L5 is selected from chemical bond, O, S, NR* , CH 2 or SO 2 ; in another more specific embodiment, L 5 is selected from O, S, NH, CH 2 or SO 2 ; in another more specific embodiment, L 5 is selected from O, S , NH or CH 2 ; in another more specific embodiment, L 5 is selected from O, S or NH; in another more specific embodiment, L 5 is selected from O or CH 2 ; in another more specific embodiment In one embodiment, L 5 is selected from O or S; in another more specific embodiment, L 5
  • R is H; in another specific embodiment, R is C 1-6 alkyl; in another specific embodiment, R is C 1-6 haloalkyl ; in another In one specific embodiment, R 1 is C 2-6 alkenyl; in another specific embodiment, R 1 is C 2-6 alkynyl; in another specific embodiment, R 1 is C 3-7 ring Alkyl; In another specific embodiment, R 1 is 3 to 7 membered heterocyclyl. In another specific embodiment, R 1 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl or 3 to 7 membered heterocyclyl.
  • any technical solution or any combination thereof in any of the above specific embodiments may be combined with any technical solution or any combination thereof in other specific embodiments.
  • any technical solution of K or any combination thereof can be combined with any technical solution of E, L 1 -L 5 or any combination thereof.
  • the proviso is that L4 and L5 can together form -C ⁇ C- only when K is K1, K2 or K4.
  • the present invention intends to include the combination of all these technical solutions, which are not listed one by one due to space limitation.
  • the present invention provides a compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate, Hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • L 1 is C(R) 2 ;
  • L 5 is a chemical bond, O, S, NR* or C(R) 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 5 is O, S, NH or CH 2 ;
  • the present invention provides the above-mentioned compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is C(R) 2 ;
  • L 5 is O, S or NR*
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 5 is O, S or NH
  • the present invention provides the above-mentioned compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is C(R) 2 ;
  • L 5 is O or S
  • R is selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl;
  • R' is selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl;
  • R" is selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl;
  • the present invention provides the above-mentioned compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds or CH 2 ;
  • the present invention provides a compound of general formula (III) or (III-1), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemic forms, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • L 1 is a chemical bond, C(R) 2 or SO 2 ;
  • L 2 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2.
  • L 3 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2.
  • L 4 is selected from a chemical bond, C(R') 2 or C(R') 2 C(R') 2 ;
  • L is selected from chemical bond, O, S, NR*, C(R) 2 or SO 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (III) or (III-1), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate Helicoids, solvates, hydrates, polymorphs, prodrugs or isotope variants, and mixtures thereof, wherein,
  • L 1 is a chemical bond, CH 2 or SO 2 ;
  • L 2 is selected from chemical bond, O, S, NR*, CH 2 , CH 2 CH 2 , 1,3-phenylene, 1,4-phenylene, 1,4-triazolylene, 3,5- Pyridinylene or 2,4-pyrimidinylene;
  • L 3 is selected from chemical bond, O, S, NR*, CH 2 , CH 2 CH 2 or 1,4-triazolylene;
  • L 4 is selected from a chemical bond, CH 2 or CH 2 CH 2 ;
  • L is selected from a chemical bond, O, S, NR*, CH 2 or SO 2 ;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (III) or (III-1), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate Helicoids, solvates, hydrates, polymorphs, prodrugs or isotope variants, and mixtures thereof, wherein,
  • L 1 is C(R) 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R' ) 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 5 is S
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (III) or (III-1), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate Helicoids, solvates, hydrates, polymorphs, prodrugs or isotope variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 , L 3 and L 4 are independently selected from a chemical bond, CH 2 or CH 2 CH 2 ;
  • L 5 is S
  • the present invention provides the above-mentioned compound of general formula (III) or (III-1), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate Helicoids, solvates, hydrates, polymorphs, prodrugs or isotope variants, and mixtures thereof, wherein,
  • L 1 is C(R) 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R' ) 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 5 is S
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (III) or (III-1), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate Helicoids, solvates, hydrates, polymorphs, prodrugs or isotope variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 , L 3 and L 4 are independently selected from a chemical bond, CH 2 or CH 2 CH 2 ;
  • L 5 is S
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate, Hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • L 1 is C(R) 2 ;
  • L 2 is selected from chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 , C 6-10 arylene or 5 to 10 membered heteroarylene;
  • L 3 and L 4 are independently selected from a chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 5 is O, S, NR*, C(R) 2 or SO 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 is selected from a chemical bond, CH 2 , CH 2 CH 2 or 1,3-phenylene;
  • L 3 and L 4 are independently selected from a chemical bond, CH 2 or CH 2 CH 2 ;
  • L 5 is O, S, NH, CH 2 or SO 2 ;
  • R* is selected from H, C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention provides the above-mentioned compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is C(R) 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R' ) 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 5 is O or C(R) 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 , L 3 and L 4 are independently selected from a chemical bond, CH 2 or CH 2 CH 2 ;
  • L 5 is O or CH 2 ;
  • R* is selected from H, C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention provides a compound of general formula (V), (V-1) or (V-2), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer thereof conformers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • L 1 is C(R) 2 ;
  • L 5 is O, S, NR* or C(R) 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R' ) 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the compound of general formula (V), (V-1) or (V-2), or a pharmaceutically acceptable salt, enantiomer, diastereomer thereof Isomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is C(R) 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R' ) 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 5 is C(R) 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl or C 1-6 haloalkyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention provides the compound of general formula (V), (V-1) or (V-2), or a pharmaceutically acceptable salt, enantiomer, diastereomer thereof Isomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 , L 3 and L 4 are independently selected from a chemical bond, CH 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 or CH 2 CH 2 CH 2 CH 2 ;
  • L5 is CH2 .
  • the present invention provides the above-mentioned compound of general formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • L 1 is C(R) 2 ;
  • L 2 , L 3 and L 4 are independently selected from a chemical bond, C(R') 2 or C(R') 2 C(R') 2 ;
  • L 5 is C(R) 2 ;
  • R 1 is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl or 3 to 7 membered heterocyclyl;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 , L 3 and L 4 are independently selected from chemical bonds or CH 2 ;
  • L 5 is CH 2 ;
  • R 1 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl or 3 to 7 membered heterocyclyl.
  • the present invention provides the above-mentioned compound of general formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • L 1 is C(R) 2 ;
  • L 2 and L 4 are independently selected from a chemical bond, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 3 is selected from chemical bond, O, S, NR*, C(R') 2 , C(R') 2 C(R') 2 , C(R') 2 C(R') 2 C(R') 2 or C(R') 2 C(R') 2 C(R') 2 C(R') 2 ;
  • L 5 is C(R) 2 ;
  • R is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R' is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R" is selected from H, D, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R* is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the above-mentioned compound of general formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate thereof , hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L 1 is CH 2 ;
  • L 2 and L 4 are independently selected from a chemical bond, CH 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 or CH 2 CH 2 CH 2 CH 2 ;
  • L 3 is selected from a chemical bond, O, CH 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 or CH 2 CH 2 CH 2 CH 2 ;
  • L 5 is CH 2 ;
  • the present invention provides a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereoisomer, racemate, solvate, Hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, said compounds being selected from the group consisting of:
  • the compounds of the present invention may include one or more asymmetric centers, and thus may exist in various stereoisomeric forms, eg, enantiomeric and/or diastereomeric forms.
  • the compounds of the invention may be individual enantiomers, diastereoisomers or geometric isomers (eg cis and trans isomers), or may be in the form of a mixture of stereoisomers, Racemic mixtures and mixtures enriched in one or more stereoisomers are included.
  • Isomers can be separated from mixtures by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and formation and crystallization of chiral salts; or preferred isomers can be obtained by prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • organic compounds may form complexes with solvents in which they react or from which they are precipitated or crystallized. These complexes are known as "solvates”. When the solvent is water, the complex is called a "hydrate”. The invention covers all solvates of the compounds of the invention.
  • solvate refers to a form of a compound, or a salt thereof, which is associated with a solvent, usually formed by a solvolysis reaction. This physical association may include hydrogen bonding.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, diethyl ether, and the like.
  • the compounds described herein can be prepared, for example, in crystalline forms, and can be solvated.
  • Representative solvates include hydrates, ethanolates and methanolates.
  • hydrate refers to a compound that combines with water. Generally, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined.
  • a hydrate of a compound can be represented, for example, by the general formula R.x H 2 O, where R is the compound, and x is a number greater than zero.
  • a given compound may form more than one hydrate type, including, for example, monohydrates (x is 1), lower hydrates (x is a number greater than 0 and less than 1, for example, hemihydrates (R 0.5 H2 O)) and polyhydrates (x is a number greater than 1, eg, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • the compounds of the invention may be in amorphous or crystalline form (polymorphs). Furthermore, the compounds of the invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • polymorph refers to a crystalline form of a compound (or a salt, hydrate or solvate thereof) in a particular crystal packing arrangement. All polymorphs have the same elemental composition.
  • the invention also includes isotopically labeled compounds (isotopic variants) which are identical to those described in formula (I), but with one or more atoms represented by atoms having an atomic mass or mass number different from the atomic mass or mass number normally found in nature replaced.
  • isotopes that may be incorporated into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl.
  • the compounds of the present invention their prodrugs and pharmaceutically acceptable salts of the compounds or the prodrugs containing the above-mentioned isotopes and/or other isotopes of other atoms are within the scope of the present invention.
  • Certain isotopically-labeled compounds of the invention eg, those incorporating radioactive isotopes (eg, 3H and14C ), are useful in drug and/or substrate tissue distribution assays. Tritium, ie3H , and carbon- 14 , ie14C isotopes are particularly preferred because of their ease of preparation and detection.
  • isotope-labeled compound of formula (I) of the present invention and its prodrug can generally be prepared in this way.
  • prodrugs are also included within the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted in vivo to its active form having a medical effect, for example by hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, per intro This article serves as a reference.
  • a prodrug is any covalently bonded compound of the invention which, when administered to a patient, releases the parent compound in vivo.
  • Prodrugs are generally prepared by modifying functional groups in such a way that the modification can be cleaved by routine manipulation or in vivo to yield the parent compound.
  • Prodrugs include, for example, compounds of the invention wherein a hydroxy, amino, or thiol group is bonded to any group that, when administered to a patient, cleaves to form the hydroxy, amino, or thiol group.
  • representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide and benzoate/amide derivatives of the hydroxy, sulfhydryl and amino functional groups of the compounds of formula (I).
  • esters such as methyl ester, ethyl ester and the like can be used.
  • the esters themselves may be reactive and/or hydrolyzable under human in vivo conditions.
  • Suitable pharmaceutically acceptable in vivo hydrolyzable ester groups include those which break down readily in the human body to release the parent acid or a salt thereof.
  • the present invention also provides a pharmaceutical preparation comprising a therapeutically effective amount of a compound of formula (I) or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent or excipient thereof. All of these forms are included in the present invention.
  • the invention provides pharmaceutical compositions comprising a compound of the invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of a compound of the invention.
  • the pharmaceutical composition comprises a therapeutically effective amount of a compound of the invention.
  • the pharmaceutical composition comprises a prophylactically effective amount of a compound of the invention.
  • kits eg, pharmaceutical packs.
  • kits can include a compound of the invention, another therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispersible packs or other suitable container).
  • first and second containers e.g., vials, ampoules, bottles, syringes, and/or dispersible packs or other suitable container.
  • provided kits can also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent.
  • a compound of the invention and other therapeutic agent provided in a first container and a second container are combined to form a unit dosage form.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration , intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
  • the compounds provided herein are administered to a subject at risk of developing the condition, typically on the advice and supervision of a physician, at dosage levels as described above.
  • Subjects at risk of developing a particular condition generally include those with a family history of the condition, or those determined by genetic testing or screening to be particularly susceptible to developing the condition.
  • Chronic administration refers to administering a compound or a pharmaceutical composition thereof for a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue administration indefinitely, For example, the rest of the subject's life.
  • chronic administration is intended to provide a constant level of the compound in the blood over an extended period of time, eg, within the therapeutic window.
  • compositions of the present invention may be used to further deliver the pharmaceutical compositions of the present invention.
  • Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders.
  • a typical regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides from about 0.01 to about 20 mg/kg of the compound of the invention, with preferred doses each providing from about 0.1 to about 10 mg/kg, especially about 1 to about 5 mg/kg.
  • the transdermal dose is generally selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, preferably about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • Injection dosage levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour from about 1 to about 120 hours, especially 24 to 96 hours.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more may also be given in order to achieve adequate steady state levels.
  • the maximum total dose should not exceed approximately 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous carriers as well as buffering, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • the solid form may comprise, for example, any of the following components, or compounds of similar nature: binders, such as microcrystalline cellulose, tragacanth, or gelatin; excipients, such as starch or lactose, disintegrants, For example, alginic acid, Primogel, or corn starch; lubricants, for example, magnesium stearate; glidants, for example, colloidal silicon dioxide; sweeteners, for example, sucrose or saccharin; or flavoring agents, for example, peppermint, water Methyl sylate or orange flavoring.
  • binders such as microcrystalline cellulose, tragacanth, or gelatin
  • excipients such as starch or lactose, disintegrants, For example, alginic acid, Primogel, or corn starch
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound is typically a minor component, often about 0.05 to 10% by weight, the remainder being injectable excipients and the like.
  • Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient.
  • transdermal administration can be achieved using patches of the reservoir or porous membrane type, or various solid matrices.
  • compositions for oral administration, injection or topical administration are representative only. Other materials and processing techniques, etc. are described in Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, Section 8, which is incorporated herein by reference.
  • the compounds of the invention may also be administered in sustained release form, or from a sustained release delivery system.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention also relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation comprises water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are ⁇ -, ⁇ - and ⁇ -cyclodextrins composed of 6, 7 and 8 ⁇ -1,4-linked glucose units, respectively.
  • Treatment as defined herein may be applied as a sole therapy, or may include conventional surgery or radiotherapy or chemotherapy in addition to the compounds of the invention. Accordingly, the compounds of the present invention may also be used in combination with existing therapeutic agents for the treatment of cancer.
  • Such chemotherapy and the compound of the present invention may be administered simultaneously, sequentially, or separately, and may contain one or more of the following types of antineoplastic agents.
  • the structures of the compounds of the present invention are determined by nuclear magnetic resonance (NMR) or/and liquid chromatography-mass chromatography (LC-MS). NMR chemical shifts ( ⁇ ) are given in parts per million (ppm).
  • the determination of NMR is to use Bruker Avance III HD 500MHz nuclear magnetic instrument, and the determination solvent is deuterated dimethyl sulfoxide (DMSO-d6), deuterated methanol (CD 3 OD) and deuterated chloroform (CDCl 3 ), and the internal standard is four Methylsilane (TMS).
  • the thin-layer chromatography silica gel plate uses Liangchen-LCGY silica gel plate (20 ⁇ 20cm, acrylic acid 0.4-0.5mm). mm.
  • the medium-pressure preparative chromatography adopts Biotage (Isolera One automated flash purification system (200-400nm)).
  • the starting materials in the examples of the present invention are known and commercially available, or can be synthesized using or following methods known in the art.
  • H in K-H is amino hydrogen
  • R is a sulfonate group (such as mesylate group) or a halogen (such as Br and I).
  • KH is reacted with RL 1 -L 2 -L 3 -L 4 -L 5 -E under the condition of amide coupling to form the compound KL 1 -L 2 -L 3 -L 4 -L 5 -E of the present invention.
  • some compounds of the present invention can also be obtained by reducing, alkylating, etc. (eg hydrogenation, methylation) of other compounds of the present invention.
  • Amines and bromides undergo nucleophilic substitution in basic systems.
  • Pd/CaCO 3 catalyzed alkyne reduction, quinoline poisoning.
  • SH, OH, primary or secondary amines and halides undergo nucleophilic substitution in basic systems.
  • Thioether is oxidized under the action of m-CPBA.
  • Step 1 Preparation of 3-(4-(3-hydroxyprop-1-yn-1-yl)-1-oxoisoindol-2-yl)piperidine-2,6-dione
  • the second step 3-(2-(2,6-dicarbonylpiperidin-3-yl)-1-carbonylisoindolin-4-yl)prop-2-yn-1-yl methanesulfonate preparation of
  • the third step 3-(4-(3-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)prop-1-yn-1-yl)-1-carbonylisoindolin-2-yl)piper
  • the first step 3-(4-(4-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)but-1-yn-1-yl)-1-carbonylisoindolin-2-yl)piper
  • 3-(4-Bromo-1-carbonylisoindolin-2-yl)piperidine-2,6-dione (2.0 g, 6.2 mmol) was added to a 100 mL reaction flask, followed by bistriphenyl Phosphine palladium dichloride (0.43g, 0.62mmol), cuprous iodide (0.24g, 1.3mmol), then add DMF (25mL), TEA (2.5mL), and 2-(propane-2 -Alkyne-1-oxy)tetrahydro-2H-pyran (1.73g, 12.4mmol), then heated to 90°C and stirred for 16 hours.
  • the second step the preparation of 3-(4-(3-hydroxypropyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • Step 3 Preparation of 3-(2-(2,6-dicarbonylpiperidin-3-yl)-1-carbonylisoindolin-4-yl)propyl methanesulfonate
  • the fourth step 3-(4-(3-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)propyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-di
  • ketones 3-(4-(3-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)propyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-di
  • Step 1 Preparation of 3-(4-(6-hydroxyhex-1-yn-1-yl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step the preparation of 3-(4-(6-hydroxyhexyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • Step 3 Preparation of 6-(2-(2,6-dicarbonylpiperidin-3-yl)-1-carbonylisoindolin-4-yl)hexyl methanesulfonate
  • the fourth step 3-(4-(6-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)hexyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione preparation of
  • Step 1 Preparation of 7-(2-(2,6-dicarbonylpiperidin-3-yl)-1-carbonylisoindolin-4-yl)heptyl methanesulfonate
  • the second step 3-(4-(7-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)heptyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-di
  • ketones 3-(4-(7-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)heptyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-di
  • Step 1 Preparation of 3-(4-((4-(hydroxymethyl)phenyl)ethynyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step the preparation of 3-(4-((4-(bromomethyl)phenyl)ethynyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the third step 3-(4-((4-((4-((4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2 -yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)methyl)phenyl)ethynyl)-1-carbonylisoindolin-2-yl)
  • the first step (Z)-3-(4-(5-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidine -2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)pent-1-en-1-yl)-1-carbonylisoindoline-2
  • Step 1 Preparation of 3-(4-((bromoethyl)amino)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • Lenalidomide and 1,2-dibromoethane are raw materials, with reference to the method for the first step of Example 10 to prepare 3-(4-((bromoethyl) amino)-1-carbonylisoindoline- 2-yl)piperidine-2,6-dione, MS m/z (ESI): 366.04[M+H] + .
  • the second step 3-(4-((2-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2- Base) amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)ethyl)(methyl)amino)-1-carbonylisoindolin-2-yl)piper
  • Step 1 Preparation of 3-(6-(5-hydroxypent-1-yn-1-yl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step 5-(2-(2,6-dicarbonylpiperidin-3-yl)-3-carbonylisoindolin-5-yl)pent-4-yn-1-yl methanesulfonate preparation of
  • the third step 3-(6-(5-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)pent-1-yn-1-yl)-1-carbonylisoindolin-2-yl)piper
  • the third step 3-(4-((4-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2- Base) amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)butyl)amino)-1-carbonylisoindolin-2-yl)piperidine-2, Preparation of 6-diketones
  • the first step (2S,4S)-1-((S)-2-(10-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) Phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)decanoylamino)-3,3-dimethylbutyryl)
  • reaction solution was left to stand and separated, the aqueous phase was extracted with dichloromethane (50mL x 3), the combined organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Ethyl acetate (50 mL) was added to the residue, stirred for 2 minutes, and a solid formed.
  • Step 1 Preparation of 3-(4-(2-chloroethoxy)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step the preparation of 3-(4-(2-iodoethoxy)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the third step 3-(4-(2-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl )amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)ethoxy)-1-carbonylisoindolin-2-yl)piperidin-2,6- Preparation of diketones
  • the first step 3-(4-((4-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2- Base) amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)butyl)thio)-1-carbonylisoindolin-2-yl)piperidin-2 ,6-Diketone, Preparation of Hydrochloride Salt
  • the first step 3-(4-((5-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2- Base) amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)pentyl)thio)-1-carbonylisoindolin-2-yl)piperidin-2 , Preparation of 6-diketone
  • reaction solution was cooled to room temperature, poured into ethyl acetate (100 mL), solids were precipitated, filtered, and the solids were collected.
  • the filtrate was washed with water (30mL), extracted with ethyl acetate (50mL x 5).
  • the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
  • the first step (Z)-3-(4-(3-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidine -2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)prop-1-en-1-yl)-1-carbonylisoindoline-2
  • Step 1 Preparation of 3-(4-((4-bromobutyl)thio)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step the preparation of 3-(4-((4-bromobutyl)sulfonyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the third step 3-(4-((4-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2- Base) amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)butyl)sulfonyl)-1-carbonylisoindolin-2-yl)piperidin-2 , Preparation of 6-diketone
  • the first step (Z)-3-(4-(4-(4-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidine -2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)but-1-en-1-yl)-1-carbonylisoindoline-2
  • the first step 8-(4-(4-(4-(4-(4-((2-(2,6-dicarbonylpiperidin-3-yl)-1-carbonylisoindolin-4-yl)oxo) Butyl)piperazin-1-yl)piperidin-1-yl)-9-ethyl-6,6-dimethyl-11-carbonyl-6,11-dihydro-5H-benzo[b]carba
  • reaction solution was raised to 82°C and stirred for 1.5 hours. After the reaction was completed, the reaction solution was cooled to room temperature, and purified by high performance liquid chromatography (0.05% HCl, water/acetonitrile) to obtain the title compound 8-(4-(4-(4-(4-((2-(2,6-dicarbonyl Piperidin-3-yl)-1-carbonylisoindolin-4-yl)oxo)butyl)piperazin-1-yl)piperidin-1-yl)-9-ethyl-6,6 -Dimethyl-11-carbonyl-6,11-dihydro-5H-benzo[b]carbazole-3-carbonitrile (white solid, 12.9 mg, yield 31%, hydrochloride).
  • the first step (2-((2-((4-(4-(4-(4-(4-(but-3-en-1-ylsulfonyl)piperazin-1-yl)piperidin-1-yl)-2 Preparation of -methoxyphenyl)amino)-5-chloropyrimidin-4-yl)amino)phenyl)dimethylphosphine by oxidation
  • the second step (E)-3-(4-(4-((4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino) Pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)sulfonyl)but-1-en-1-yl)-1-carbonyl isodihydro Preparation of indol-2-yl)piperidine-2,6-dione
  • Step 1 Preparation of 3-(4-((5-bromopentyl)oxo)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step 3-(4-((5-((1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl) Amino)-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)pentyl)oxo)-1-carbonylisoindolin-2-yl)piperidine-2,6- Preparation of diketones
  • reaction solution was warmed up to 85°C and stirred for 16 hours. After the reaction was completed, the reaction solution was cooled to room temperature, and purified by high performance liquid chromatography (0.05% HCl, water/acetonitrile) to obtain the title compound 3-(4-((5-((1-(4-((5-chloro- 4-((2-(Dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)pentyl) Oxo)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione (white solid, 3.0 mg, yield 6%, hydrochloride).
  • the first step -(4-(4-((1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino) Preparation of -3-methoxyphenyl)piperidin-4-yl)(methyl)amino)butoxy)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • reaction solution was warmed up to 90°C and stirred for 18 hours. After the reaction was completed, the reaction solution was cooled to room temperature, filtered, and the filtrate was purified by high performance liquid chromatography (hydrochloric acid system, water/acetonitrile) to obtain the title compound 3-(4-(4-((1-(4-((5-chloro -4-((2-(Dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)butoxy yl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione (white solid, 10mg, yield: 21%, hydrochloride)
  • the first step 3-(4-(6-((1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl) amino Preparation of )-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)hexyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • reaction solution was warmed up to 85°C and stirred for 3 hours. After the reaction was completed, the reaction solution was cooled to room temperature, and purified by high performance liquid chromatography (0.05% HCl, water/acetonitrile) to obtain the title compound 3-(4-(6-((1-(4-((5-chloro-4 -((2-(Dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)hexyl)-1 -Carbonylisoindolin-2-yl)piperidine-2,6-dione (white solid, 4.0 mg, yield 9.5%, hydrochloride).
  • the first step 3-(4-((4-(4-(1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidine-2- Base) amino)-3-methoxyphenyl)piperidin-4-yl)piperazin-1-yl)butyl)(methyl)amino)-1-carbonylisoindolin-2-yl)piper
  • reaction solution was stirred for 4 hours under a hydrogen balloon system. After the reaction was completed, the reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (hydrochloric acid system, water/acetonitrile) to obtain the title compound 3-(4-((4-(4-(1-(4- ((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)piperazine- 1-yl)butyl)(methyl)amino)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione (white solid, 2mg, yield: 4%, hydrochloric acid Salt).
  • the first step 3-(4-(7-((1-(4-((5-chloro-4-((2-(dimethylphosphoryl) phenyl) amino) pyrimidin-2-yl) amino Preparation of )-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)heptyl)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • reaction solution was warmed up to 70°C and stirred for 16 hours. After the reaction was completed, the reaction solution was cooled to room temperature, filtered, and the filtrate was purified by preparative liquid chromatography (hydrochloric acid system, water/acetonitrile) to obtain the title compound 3-(4-(7-((1-(4-((5-chloro -4-((2-(Dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)heptyl )-1-carbonylisoindolin-2-yl)piperidine-2,6-dione (yellow solid, 7.0 mg, yield: 42%, hydrochloride).
  • Step 1 Preparation of 3-(4-((6-bromohexyl)oxo)-1-carbonylisoindolin-2-yl)piperidine-2,6-dione
  • the second step 3-(4-((6-((1-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl) Amino)-3-methoxyphenyl)piperidin-4-yl)(methyl)amino)hexyl)oxo)-1-carbonylisoindolin-2-yl)piperidine-2,6-di
  • the IC50 of the compound of the present invention is determined by CellTiter GLO reagent of Promega Company. Proceed as follows:
  • Tumor cell lines were cultured in the indicated medium in an incubator at 37 °C, 5% CO2 . Passage regularly, take the cells in the logarithmic growth phase for plating, and the cell density is 2000-4000 cells/well.
  • the compound of the present invention was serially diluted and then added to the cells.
  • the negative control was DMSO, and the blank control was culture fluid without cells.
  • the microplate of the cultured cells was put back into the incubator to continue culturing for 72 hours; the detection reagent was added to the cell culture medium according to the instructions of the CellTiter GLO reagent, and the luminescent signal was detected on the EnVision microplate reader (PerkinElmer) to measure the cell activity.
  • the growth inhibition of the compound of the present invention on cells is drawn by Prism Graphpad software and the IC50 of the compound of the present invention is counted.
  • embodiment compound shown in the present invention is in the inhibitory test of BaF3 (EGFR Del19/T790M/C797S), BaF3 (EGFR L858R/T790M/C797S) and BaF3 (CD74-ROS1-G2032R) mutant cell proliferation activity has a good inhibitory effect.

Landscapes

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

Abstract

一类新型蛋白降解剂及其应用。涉及通式(I)的化合物,以及含有所述化合物的药物组合物,其用于抑制并诱导降解ALK、ROS1以及EGFR蛋白,可用于治疗与ALK、ROS1以及EGFR蛋白高度表达相关的肿瘤,例如肺癌、乳腺癌、前列腺癌等。还涉及上述化合物的制备和用途。

Description

一类新型蛋白降解剂及其应用
本申请要求申请日为2021年5月19日的中国专利申请2021105464549的优先权。本申请引用上述中国专利申请的全文。
技术领域
本发明属于药物化学与合成领域。具体涉及一类新型的靶向ALK、ROS1以及EGFR蛋白及其突变蛋白的PROTAC分子,及其制备方法和应用,以及包含该类化合物的药物组合物。
背景技术
泛素-蛋白酶体途径是较普遍的一种内源蛋白降解方式,其中需要降解的蛋白先被泛素化修饰,然后被蛋白酶体分解为较小的多肽、氨基酸以及可以重复使用的泛素。
PROTAC( proteolysis  targeting  chimeras),即蛋白降解靶向嵌合体,是近年来新兴的热门研究领域。PROTAC分子一般可分为三个部分,一端为与特定靶蛋白结合的小分子片段(war head),另一端为具有泛素化功能的E3连接酶的配体(E3 ligase ligand),以及将两者连接起来的连接体(linker)。PROTAC分子利用了细胞的蛋白泛素化降解途径,可选择性地降解目标靶蛋白。具体而言,由于PROTAC分子的两端分别为靶蛋白与E3连接酶的配体片段,所以PROTAC分子可同时与靶蛋白和E3连接酶结合,促进了靶蛋白的泛素化,进而被蛋白酶体识别并加以降解。
肺癌是威胁人类健康的重大疾病,肺癌死亡率己占所有恶性肿瘤首位。非小细胞肺癌患者中,伴有EGFR(Epidermal Growth Factor Receptor,表皮生长因子受体)、ALK(anaplastic lymphoma kinase,间变性淋巴瘤激酶)和ROS1(ROS proto-oncogene 1 receptor tyrosine kinase,c-ros肉瘤致癌因子-受体酪氨酸激酶)激活性突变的病例,占比分别为30%、8%和2%左右。因此,开发能够对现有EGFR抑制剂、ALK抑制剂、ROS1抑制剂耐药的新型PROTAC分子具有巨大的研究价值和潜在的实用价值。
在PROTAC领域,已公开的基于布加替尼(Brigatinib)或其类似结构的PROTAC分子,主要是作为ALK降解剂进行研究(例如WO2020249048、WO2019113071、WO2019042444、WO2017204445等),而没有公开披露针对EGFR C797S突变的相关药理数据。同样的,其他已公开的基于艾乐替尼(Alectinib)的ALK降解剂也没有公开披露针对EGFR C797S突变的相关药理数据(如WO2020069106、WO2019114770、WO2019196812等)。
因此,本领域对于能够降解EGFR、ALK或ROS1蛋白及其突变蛋白的PROTAC分子存在需求。
发明内容
在一个方面,本发明提供通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水 合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000001
其中,
K选自以下结构:
Figure PCTCN2022093956-appb-000002
E选自以下结构:
Figure PCTCN2022093956-appb-000003
L 1为化学键、C(R) 2或SO 2
L 2选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 4选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5选自化学键、O、S、NR*、C(R) 2或SO 2
或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
或者L 4和L 5一起形成顺式或反式的-CR=CR-,或-C≡C-;
其中Z为C=O或C(R”) 2
R 1选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-7环烷基或3至7元杂环基;
R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
条件是,仅当K为K1、K2或K4时,L 4和L 5可以一起形成-C≡C-。
在另一个方面,本发明提供了一种药物组合物,所述药物组合物含有本发明化合物,和任选地药学上可接受的赋形剂。
在另一个方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物,其还含有其它治疗剂。
在另一个方面,本发明提供了包含本发明化合物,和其它治疗剂以及药学上可接受的载剂、佐剂或媒剂的试剂盒。
在另一个方面,本发明提供了本发明化合物在制备用于治疗和/或预防癌症的药物中的用途。
在另一个方面,本发明提供了在受试者中治疗和/或预防癌症的方法,包括向所述受试者给药本发明化合物或本发明组合物。
在另一个方面,本发明提供了本发明化合物或本发明组合物,其用于治疗和/或预防癌症。
在具体实施方案中,所述癌症选自肺癌;淋巴瘤;炎症性肌纤维母细胞瘤;结直肠癌;脑胶质瘤;星形胶质母细胞瘤;卵巢癌;骨髓癌症;移植相关的癌症;中性粒细胞减少症;白血病;翁韦里希特综合症;支气管癌;前列腺癌;乳腺癌;甲状腺癌;胰腺癌;神经母细胞瘤;髓外浆细胞瘤;浆细胞瘤;胃癌;胃肠道间质瘤;食道癌;大肠腺癌;食管鳞状细胞癌;肝癌;肾细胞癌;膀胱癌;子宫内膜癌;黑色素瘤;脑癌;口腔癌;肉瘤;对靶向药物耐药的肿瘤;或依赖ALK、ROS1或EGFR或其任意突变蛋白的肿瘤或疾病。
在另一具体实施方案中,所述癌症选自小细胞肺癌;非小细胞肺癌;弥漫性大B细胞淋巴瘤;非霍奇金淋巴瘤;间变性淋巴瘤;间变性大细胞淋巴瘤;CD20阳性淋巴瘤;原发性淋巴瘤;B细胞淋巴瘤;复发性B细胞非霍奇金淋巴瘤;复发性弥漫性大B细胞淋巴瘤;复发性纵隔(胸腺)大B细胞淋巴瘤;原发性纵隔(胸腺)大B细胞淋巴瘤;复发性转化非霍奇金淋巴瘤;难治性B细胞非霍奇金淋巴瘤;难治性弥漫性大B细胞淋巴瘤;难治性原发性纵隔(胸腺)大B细胞淋巴瘤;难治性转化的非霍奇金淋巴瘤;多发性骨髓瘤;骨髓增生异常综合征(MDS);既往治疗的骨髓增生异常综合征;浆细胞骨髓瘤;阴燃骨髓瘤;闷烧多发性骨髓瘤;骨髓纤维化;急性髓细胞白血病(AML);与白血病相关的贫血;慢性粒细胞白血病;B细胞慢性淋巴细胞白血病;翁韦里希特综合症;支气管癌;前列腺癌;三阴性乳腺癌;偶发性乳腺癌;考登病患者;甲状腺癌;胰腺癌;神经母细胞瘤;髓外浆细胞瘤;浆细胞瘤;胃癌;胃肠道间质瘤;食道癌;大肠腺癌;食管鳞状细胞癌;肝癌;肾细胞癌;膀胱癌;子宫内膜癌;黑色素瘤;脑癌;口腔癌;横纹肌肉瘤;各种脂肪源性肿瘤;尤文肉瘤/原始神经外胚层瘤 (Ewing/PNETs);平滑肌肉瘤;对EGFR、ROS1或ALK靶向药物耐药的肿瘤。
在另一具体实施方案中,所述癌症选自间变性淋巴瘤激酶(ALK)突变阳性的非小细胞肺癌(NSCLC);ROS1阳性非小细胞肺癌;EGFR突变的非小细胞肺癌;肺腺癌;对EGFR、ROS1或ALK靶向药物耐药的肺癌;对ALK靶向药物耐药的淋巴瘤;或依赖选自ALK、ROS1或EGFR或其任意突变蛋白的以下肿瘤、癌症或疾病:肺癌、淋巴瘤、炎症性肌纤维母细胞瘤、结直肠癌、脑胶质瘤、星形胶质母细胞瘤、卵巢癌、白血病、乳腺癌、甲状腺癌、神经母细胞瘤、髓外浆细胞瘤、浆细胞瘤、食管鳞状细胞癌、肾细胞癌、支气管癌、前列腺癌、乳腺癌、甲状腺癌、胰腺癌、神经母细胞瘤、髓外浆细胞瘤、浆细胞瘤、胃癌、胃肠道间质瘤、食道癌、大肠腺癌、食管鳞状细胞癌、肝癌、肾细胞癌、膀胱癌、子宫内膜癌、黑色素瘤、脑癌、口腔癌或肉瘤。
由随后的具体实施方案、实施例和权利要求,本发明的其它目的和优点将对于本领域技术人员显而易见。
定义
化学定义
下面更详细地描述具体官能团和化学术语的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C 1-6烷基”包括C 1、C 2、C 3、C 4、C 5、C 6、C 1-6、C 1-5、C 1-4、C 1-3、C 1-2、C 2-6、C 2-5、C 2-4、C 2-3、C 3-6、C 3-5、C 3-4、C 4-6、C 4-5和C 5-6烷基。
“C 1-6烷基”是指具有1至6个碳原子的直链或支链饱和烃基团。在一些实施方案中,C 1-4烷基是优选的。C 1-6烷基的例子包括:甲基(C 1)、乙基(C 2)、正丙基(C 3)、异丙基(C 3)、正丁基(C 4)、叔丁基(C 4)、仲丁基(C 4)、异丁基(C 4)、正戊基(C 5)、3-戊基(C 5)、戊基(C 5)、新戊基(C 5)、3-甲基-2-丁基(C 5)、叔戊基(C 5)和正己基(C 6)。术语“C 1-6烷基”还包括杂烷基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。常规烷基缩写包括:Me(-CH 3)、Et(-CH 2CH 3)、iPr(-CH(CH 3) 2)、nPr(-CH 2CH 2CH 3)、n-Bu(-CH 2CH 2CH 2CH 3)或i-Bu(-CH 2CH(CH 3) 2)。
“C 2-6烯基”是指具有2至6个碳原子和至少一个碳碳双键的直链或支链烃基团。在一些实施方案中,C 2-4烯基是优选的。C 2-6烯基的例子包括:乙烯基(C 2)、1-丙烯基(C 3)、2-丙烯基(C 3)、1-丁烯基(C 4)、2-丁烯基(C 4)、丁二烯基(C 4)、戊烯基(C 5)、戊二烯基(C 5)、己烯基(C 6),等等。术语“C 2-6烯基”还包括杂烯基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烯基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 2-6炔基”是指具有2至6个碳原子、至少一个碳-碳叁键以及任选地一个或多个碳-碳双键的直链或支链烃基团。在一些实施方案中,C 2-4炔基是优选的。C 2-6炔基的例子包括但不限于:乙炔基(C 2)、1-丙炔基(C 3)、2-丙炔基(C 3)、1-丁炔基(C 4)、2-丁炔基(C 4),戊炔基(C 5)、己炔基(C 6),等等。术语“C 2- 6炔基”还包括杂炔基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、 硅、磷)替代。炔基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“卤代”或“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。
因此,“C 1-6卤代烷基”是指上述“C 1-6烷基”,其被一个或多个卤素基团取代。在一些实施方案中,C 1-4卤代烷基是特别优选的,更优选C 1-2卤代烷基。示例性的所述卤代烷基包括但不限于:-CF 3、-CH 2F、-CHF 2、-CHFCH 2F、-CH 2CHF 2、-CF 2CF 3、-CCl 3、-CH 2Cl、-CHCl 2、2,2,2-三氟-1,1-二甲基-乙基,等等。卤代烷基基团可以在任何可用的连接点上被取代,例如,1至5个取代基、1至3个取代基或1个取代基。
“C 3-10环烷基”是指具有3至10个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C 3-7环烷基和C 3-6环烷基是特别优选的,更优选C 5-6环烷基。环烷基还包括其中上述环烷基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基(C 3)、环丙烯基(C 3)、环丁基(C 4)、环丁烯基(C 4)、环戊基(C 5)、环戊烯基(C 5)、环己基(C 6)、环己烯基(C 6)、环已二烯基(C 6)、环庚基(C 7)、环庚烯基(C 7)、环庚二烯基(C 7)、环庚三烯基(C 7),等等。环烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“3-12元杂环基”是指具有环碳原子和1至5个环杂原子的3至12元非芳香环系的基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,优选4-12元杂环基,其为具有环碳原子和1至5个环杂原子的4至12元非芳香环系;在一些实施方案中,优选3-10元杂环基,其为具有环碳原子和1至5个环杂原子的3至10元非芳香环系;在一些实施方案中,优选3-7元杂环基,其为具有环碳原子和1至4个环杂原子的3至7元非芳香环系;优选3-6元杂环基,其为具有环碳原子和1至3个环杂原子的3至6元非芳香环系;优选4-8元杂环基,其为具有环碳原子和1至3个环杂原子的4至8元非芳香环系;更优选5-6元杂环基,其为具有环碳原子和1至3个环杂原子的5至6元非芳香环系。杂环基还包括其中上述杂环基环与一个或多个环烷基稠合的环体系,其中连接点在环烷基环上,或其中上述杂环基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在杂环基环上;且在这样的情况下,环成员的数目继续表示在杂环基环体系中环成员的数目。示例性的包含一个杂原子的3元杂环基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基(thiorenyl)。示例性的含有一个杂原子的4元杂环基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环基包括但不限于:二氧杂环戊烷基、氧硫杂环戊烷基(oxasulfuranyl)、二硫杂环戊烷基(disulfuranyl)和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环基包括但不限于:三唑啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环基包括但不限于:哌啶基、四氢吡喃基、二氢吡啶基和硫杂环己烷基(thianyl)。示例性的包含两个杂原子的6元杂环基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性 的包含三个杂原子的6元杂环基包括但不限于:六氢三嗪基(triazinanyl)。示例性的含有一个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的与C 6芳基环稠合的5元杂环基(在本文中也称作5,6-双环杂环基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与C 6芳基环稠合的6元杂环基(本文还指的是6,6-双环杂环基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。杂环基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 6-10芳基”是指具有6-10个环碳原子和零个杂原子的单环或多环的(例如,双环)4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“C 6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C 10芳基”;例如,萘基,例如,1-萘基和2-萘基)。芳基还包括其中上述芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述芳基环上,在这种情况下,碳原子的数目继续表示所述芳基环系统中的碳原子数目。芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“5-14元杂芳基”是指具有环碳原子和1-4个环杂原子的5-14元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6、10或14个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。杂芳基双环系统在一个或两个环中可以包括一个或多个杂原子。杂芳基还包括其中上述杂芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述杂芳基环上,在这种情况下,碳原子的数目继续表示所述杂芳基环系统中的碳原子数目。在一些实施方案中,5-10元杂芳基是优选的,其为具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系。在另一些实施方案中,5-6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环或双环的4n+2芳族环体系。示例性的含有一个杂原子的5元杂芳基包括但不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:三唑基、噁二唑基(例如,1,2,4-噁二唑基)和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括但不限于:吡啶基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并三唑基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。杂芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 6-10亚芳基”和“5-14元亚杂芳基”分别表示上述“C 6-10芳基”和“5-14元杂芳基”,其中另一个氢被 除去而形成的二价基团,并且可以是取代或未取代的。优选C 6-10亚芳基和5-10元亚杂芳基。代表性的C 6-10亚芳基包括1,2-亚苯基、1,3-亚苯基、1,4-亚苯基、1,2-亚萘基、1,3-亚萘基、1,4-亚萘基、1,5-亚萘基、1,6-亚萘基、1,7-亚萘基、1,8-亚萘基、2,3-亚萘基、2,5-亚萘基、2,6-亚萘基、2,7-亚萘基。代表性的5-10元亚杂芳基包括1,2-亚吡咯基、1,3-亚吡咯基、2,3-亚吡咯基、2,4-亚吡咯基、2,5-亚吡咯基、3,4-亚吡咯基、2,3-亚呋喃基、2,4-亚呋喃基、2,5-亚呋喃基、3,4-亚呋喃基、2,3-亚噻吩基、2,4-亚噻吩基、2,5-亚噻吩基、3,4-亚噻吩基、1,2-亚咪唑基、1,4-亚咪唑基、1,5-亚咪唑基、2,4-亚咪唑基、2,5-亚咪唑基、4,5-亚咪唑基、1,3-亚吡唑基、1,4-亚吡唑基、1,5-亚吡唑基、3,4-亚吡唑基、3,5-亚吡唑基、4,5-亚吡唑基、2,4-亚噁唑基、2,5-亚噁唑基、4,5-亚噁唑基、3,4-亚异噁唑基、3,5-亚异噁唑基、4,5-亚异噁唑基、2,4-亚噻唑基、2,5-亚噻唑基、4,5-亚噻唑基、3,4-亚异噻唑基、3,5-亚异噻唑基、4,5-亚异噻唑基、1,4-亚三唑基、1,5-亚三唑基、4,5-亚三唑基、1,2,4-噁二唑-3,5-亚基、1,2,3-噁二唑-4,5-亚基、1,2,4-噻二唑-3,5-亚基、1,2,3-噻二唑-4,5-亚基、2,3-亚吡啶基、2,4-亚吡啶基、2,5-亚吡啶基、2,6-亚吡啶基、3,4-亚吡啶基、3,5-亚吡啶基、3,4-亚哒嗪基、3,5-亚哒嗪基、3,6-亚哒嗪基、4,5-亚哒嗪基、2,4-亚嘧啶基、2,5-亚嘧啶基、4,5-亚嘧啶基、4,6-亚嘧啶基、2,3-亚吡嗪基、2,5-亚吡嗪基、2,6-亚吡嗪基、1,2,3-三嗪基-4,5-亚基、1,2,3-三嗪基-4,6-亚基、1,2,4-三嗪基-3,5-亚基、1,2,4-三嗪基-3,6-亚基、1,2,5-三嗪基-3,4-亚基、1,2,5-三嗪基-3,6-亚基、1,2,5-三嗪基-4,6-亚基、1,3,5-三嗪基-2,4-亚基、1,2,3,4-四嗪基-5,6-亚基、1,2,3,5-四嗪基-4,6-亚基、1,2,4,6-四嗪基-3,5-亚基等。
本文定义的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基等为任选取代的基团。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR aa、-ON(R bb) 2、-N(R bb) 2、-N(R bb) 3 +X -、-N(OR cc)R bb、-SH、-SR aa、-SSR cc、-C(=O)R aa、-CO 2H、-CHO、-C(OR cc) 2、-CO 2R aa、-OC(=O)R aa、-OCO 2R aa、-C(=O)N(R bb) 2、-OC(=O)N(R bb) 2、-NR bbC(=O)R aa、-NR bbCO 2R aa、-NR bbC(=O)N(R bb) 2、-C(=NR bb)R aa、-C(=NR bb)OR aa、-OC(=NR bb)R aa、-OC(=NR bb)OR aa、-C(=NR bb)N(R bb) 2、-OC(=NR bb)N(R bb) 2、-NR bbC(=NR bb)N(R bb) 2、-C(=O)NR bbSO 2R aa、-NR bbSO 2R aa、-SO 2N(R bb) 2、-SO 2R aa、-SO 2OR aa、-OSO 2R aa、-S(=O)R aa、-OS(=O)R aa、-Si(R aa) 3、-OSi(R aa) 3、-C(=S)N(R bb) 2、-C(=O)SR aa、-C(=S)SR aa、-SC(=S)SR aa、-SC(=O)SR aa、-OC(=O)SR aa、-SC(=O)OR aa、-SC(=O)R aa、-P(=O) 2R aa、-OP(=O) 2R aa、-P(=O)(R aa) 2、-OP(=O)(R aa) 2、-OP(=O)(OR cc) 2、-P(=O) 2N(R bb) 2、-OP(=O) 2N(R bb) 2、-P(=O)(NR bb) 2、-OP(=O)(NR bb) 2、-NR bbP(=O)(OR cc) 2、-NR bbP(=O)(NR bb) 2、-P(R cc) 2、-P(R cc) 3、-OP(R cc) 2、-OP(R cc) 3、-B(R aa) 2、-B(OR cc) 2、-BR aa(OR cc)、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(R bb) 2、=NNR bbC(=O)R aa、=NNR bbC(=O)OR aa、=NNR bbS(=O) 2R aa、=NR bb或=NOR cc取代;
R aa的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R aa基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R bb的每个独立地选自:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R bb基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R cc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R cc基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R dd的每个独立地选自:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR ee、-ON(R ff) 2、-N(R ff) 2、-N(R ff) 3 +X -、-N(OR ee)R ff、-SH、-SR ee、-SSR ee、-C(=O)R ee、-CO 2H、-CO 2R ee、-OC(=O)R ee、-OCO 2R ee、-C(=O)N(R ff) 2、-OC(=O)N(R ff) 2、-NR ffC(=O)R ee、-NR ffCO 2R ee、-NR ffC(=O)N(R ff) 2、-C(=NR ff)OR ee、-OC(=NR ff)R ee、-OC(=NR ff)OR ee、-C(=NR ff)N(R ff) 2、-OC(=NR ff)N(R ff) 2、-NR ffC(=NR ff)N(R ff) 2、-NR ffSO 2R ee、-SO 2N(R ff) 2、-SO 2R ee、-SO 2OR ee、-OSO 2R ee、-S(=O)R ee、-Si(R ee) 3、-OSi(R ee) 3、-C(=S)N(R ff) 2、-C(=O)SR ee、-C(=S)SR ee、-SC(=S)SR ee、-P(=O) 2R ee、-P(=O)(R ee) 2、-OP(=O)(R ee) 2、-OP(=O)(OR ee) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代,或者两个偕R dd取代基可结合以形成=O或=S;
其它定义
术语“癌症”包括但不限于下列癌症:乳腺、卵巢、子宫颈、前列腺、睾丸、食道、胃、皮肤、肺、骨、结肠、胰腺、甲状腺、胆道、颊腔与咽(口)、唇、舌、口腔、咽、小肠、结肠直肠、大肠、直肠、脑与中枢神经系统的癌症、成胶质细胞瘤、神经母细胞瘤、角化棘皮瘤、表皮样癌、大细胞癌、腺癌、腺瘤、滤泡癌、未分化的癌、乳头状癌、精原细胞瘤、黑色素瘤、肉瘤、膀胱癌、肝癌、肾癌、骨髓障碍、淋巴障碍、霍奇金氏病、毛细胞癌和白血病。更具体地,癌症包括但不限于HER2阳性的转移性乳腺癌、HER2过度表达的转移性胃腺癌或胃食管交界腺癌、表皮生长因子受体(EGFR)基因敏感突变的非小细胞肺癌、含铂化疗期间或化疗后疾病进展的局部晚期或转移性鳞状组织学类型非小细胞肺癌、转移性晚期乳腺癌、去势抵抗性前列腺癌。
本文所用的术语“治疗”涉及逆转、减轻、抑制该术语适用的障碍或病症的进展或者这类障碍或病症的一种或多种症状。本文所用的名词“治疗”涉及动词治疗的动作,后者是如刚才所定义的。
本文所用的术语“药学上可接受的盐”表示本发明化合物的那些羧酸盐、氨基酸加成盐,它们在可靠的医学判断范围内适用于与患者组织接触,不会产生不恰当的毒性、刺激作用、变态反应等,与合理的益处/风险比相称,就它们的预期应用而言是有效的,包括(可能的话)本发明化合物的两性离子形式。
药学上可接受的碱加成盐是与金属或胺生成的,例如碱金属与碱土金属氢氧化物或有机胺。用作 阳离子的金属的实例有钠、钾、镁、钙等。适合的胺的实例有N,N'-二苄基乙二胺、氯普鲁卡因、胆碱、二乙醇胺、乙二胺、N-甲基葡糖胺和普鲁卡因。
酸性化合物的碱加成盐可以这样制备,按照常规方式使游离酸形式与足量所需的碱接触,生成盐。按照常规方式使盐形式与酸接触,再分离游离酸,可以使游离酸再生。游离酸形式在某些物理性质上多少不同于它们各自的盐形式,例如在极性溶剂中的溶解度,但是出于本发明的目的,盐还是等价于它们各自的游离酸。
盐可以是从无机酸制备的硫酸盐、焦硫酸盐、硫酸氢盐、亚硫酸盐、亚硫酸氢盐、硝酸盐、磷酸盐、磷酸一氢盐、磷酸二氢盐、偏磷酸盐等。盐也可以是从有机酸制备的,例如脂肪族一元与二元羧酸、苯基取代的烷酸、羟基烷酸、烷二酸、芳香族酸、脂肪族与芳香族磺酸等。代表性盐包括乙酸盐、丙酸盐、辛酸盐、异丁酸盐、草酸盐、丙二酸盐、琥珀酸盐等。药学上可接受的盐可以包括基于碱金属与碱土金属的阳离子,例如钠、锂、钾、钙、镁等,以及无毒的铵、季铵和胺阳离子,包括但不限于铵、四甲基铵、四乙基铵、甲胺、二甲胺、三甲胺、三乙胺、乙胺等。还涵盖氨基酸的盐,例如精氨酸盐、葡糖酸盐、半乳糖醛酸盐等(例如参见Berge S.M.et al.,"Pharmaceutical Salts,”J.Pharm.Sci.,1977;66:1-19,引入此作为参考)。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。本文可互换使用术语“人”、“患者”和“受试者”。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括在受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药代动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗有效量和预防有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗益处的量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化的量。化合物的治疗有效量是指单独使用或与其它疗法联用时,治疗剂的量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其它治疗剂的治疗效果的量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的量,或足以预防与疾病、障碍或病症有关的一或多种症状的量,或防止疾病、障碍或病症复发的量。化合物的预防有效量是指单独使用或与其它药剂联用时,治疗剂的量,它在预防疾病、障碍或病症的过程中 提供预防益处。术语“预防有效量”可以包括改善总体预防的量,或增强其它预防药剂的预防效果的量。
“组合”以及相关术语是指同时或依次给药本发明化合物和其它治疗剂。例如,本发明化合物可以与其它治疗剂以分开的单位剂型同时或依次给药,或与其它治疗剂一起在单一单位剂型中同时给药。
具体实施方案
本文中,“本发明化合物”指的是以下的式(I)化合物(包括子通式,例如式(II)、(III-1)、(V-2)等)、其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物。
本文中,化合物使用标准的命名法命名。具有非对称中心的化合物,应该明白(除非另有说明)所有的光学异构体及其混合物均包含在内。此外,除非另有规定,本发明所包括的所有异构体化合物与碳碳双键可能以Z和E的形式出现。在不同的互变异构形式存在的化合物,一个所述化合物并不局限于任何特定的互变异构体,而是旨在涵盖所有的互变异构形式。
在一个实施方案中,本发明涉及通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000004
其中,
K选自以下结构:
Figure PCTCN2022093956-appb-000005
E选自以下结构:
Figure PCTCN2022093956-appb-000006
Figure PCTCN2022093956-appb-000007
L 1为化学键、C(R) 2或SO 2
L 2选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 4选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5选自化学键、O、S、NR*、C(R) 2或SO 2
或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
或者L 4和L 5一起形成顺式或反式的-CR=CR-,或-C≡C-;
其中Z为C=O或C(R”) 2
R 1选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-7环烷基或3至7元杂环基;
R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
条件是,仅当K为K1、K2或K4时,L 4和L 5可以一起形成-C≡C-。
K
在一个具体实施方案中,K为K1;在另一个具体实施方案中,K为K2;在另一个具体实施方案中,K为K3;在另一个具体实施方案中,K为K4;在另一个具体实施方案中,K为K5。
E
在一个具体实施方案中,E为E1;在另一个具体实施方案中,E为E2;在另一个具体实施方案中,E为E3;在另一个具体实施方案中,E为E4;在另一个具体实施方案中,E为E5。
L 1
在一个具体实施方案中,L 1为化学键;在另一个具体实施方案中,L 1为C(R) 2;在另一个具体实施方案中,L 1为SO 2
在一个更具体的实施方案中,L 1选自化学键、CH 2或SO 2;在另一个更具体的实施方案中,L 1为CH 2
L 2
在一个具体实施方案中,L 2为化学键;在另一个具体实施方案中,L 2为O;在另一个具体实施方案中,L 2为S;在另一个具体实施方案中,L 2为NR*;在另一个具体实施方案中,L 2为C(R’) 2;在 另一个具体实施方案中,L 2为C(R’) 2C(R’) 2;在另一个具体实施方案中,L 2为C(R’) 2C(R’) 2C(R’) 2;在另一个具体实施方案中,L 2为C(R’) 2C(R’) 2C(R’) 2C(R’) 2;在另一个具体实施方案中,L 2为C 6-10亚芳基;在另一个具体实施方案中,L 2为5至10元亚杂芳基。
在一个更具体的实施方案中,L 2选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;在另一个更具体的实施方案中,L 2选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 2选自化学键、C(R’) 2或C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 2选自化学键、O、S、NR*、CH 2、CH 2CH 2、1,3-亚苯基、1,4-亚苯基、1,4-亚三唑基、3,5-亚吡啶基或2,4-亚嘧啶基;在另一个更具体的实施方案中,L 2选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;在另一个更具体的实施方案中,L 2选自化学键、CH 2、CH 2CH 2或1,3-亚苯基;在另一个更具体的实施方案中,L 2选自化学键、CH 2或CH 2CH 2;在另一个更具体的实施方案中,L 2选自化学键或CH 2
L 3
在一个具体实施方案中,L 3为化学键;在另一个具体实施方案中,L 3为O;在另一个具体实施方案中,L 3为S;在另一个具体实施方案中,L 3为NR*;在另一个具体实施方案中,L 3为C(R’) 2;在另一个具体实施方案中,L 3为C(R’) 2C(R’) 2;在另一个具体实施方案中,L 3为C(R’) 2C(R’) 2C(R’) 2;在另一个具体实施方案中,L 3为C(R’) 2C(R’) 2C(R’) 2C(R’) 2;在另一个具体实施方案中,L 3为C 6-10亚芳基;在另一个具体实施方案中,L 3为5至10元亚杂芳基。
在一个更具体的实施方案中,L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 3选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 3选自化学键、C(R’) 2或C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 3选自化学键、O、S、NR*、CH 2、CH 2CH 2或1,4-亚三唑基;在另一个更具体的实施方案中,L 3选自化学键、O、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;在另一个更具体的实施方案中,L 3选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;在另一个更具体的实施方案中,L 3选自化学键、CH 2或CH 2CH 2;在另一个更具体的实施方案中,L 3选自化学键或CH 2
L 4
在一个具体实施方案中,L 4为化学键;在另一个具体实施方案中,L 4为C(R’) 2;在另一个具体实施方案中,L 4为C(R’) 2C(R’) 2;在另一个具体实施方案中,L 4为C(R’) 2C(R’) 2C(R’) 2;在另一个具体实施方案中,L 4为C(R’) 2C(R’) 2C(R’) 2C(R’) 2
在一个更具体的实施方案中,L 4选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 4选自化学键、C(R’) 2或C(R’) 2C(R’) 2;在另一个更具体的实施方案中,L 4选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;在另一个更具体的实施方案中,L 4选自化学键、CH 2或CH 2CH 2;在另一个更具体的实施方案中,L 4选自化学键或CH 2
L 5
在一个具体实施方案中,L 5为化学键;在另一个具体实施方案中,L 5为O;在另一个具体实施方案中,L 5为S;在另一个具体实施方案中,L 5为NR*;在另一个具体实施方案中,L 5为C(R) 2;在另一个具体实施方案中,L 5为SO 2
在一个更具体的实施方案中,L 5选自O、S、NR*、C(R) 2或SO 2;在另一个更具体的实施方案中,L 5选自化学键、O、S、NR*或C(R) 2;在另一个更具体的实施方案中,L 5选自O、S、NR*或C(R) 2;在另一个更具体的实施方案中,L 5选自O、S或NR*;在另一个更具体的实施方案中,L 5选自O或C(R) 2;在另一个更具体的实施方案中,L 5选自化学键、O、S、NR*、CH 2或SO 2;在另一个更具体的实施方案中,L 5选自O、S、NH、CH 2或SO 2;在另一个更具体的实施方案中,L 5选自O、S、NH或CH 2;在另一个更具体的实施方案中,L 5选自O、S或NH;在另一个更具体的实施方案中,L 5选自O或CH 2;在另一个更具体的实施方案中,L 5选自O或S;在另一个更具体的实施方案中,L 5为CH 2
L 2和L 3
在一个具体实施方案中,L 2和L 3一起形成顺式的-CR=CR-;在另一个具体实施方案中,L 2和L 3一起形成反式的-CR=CR-;在另一个具体实施方案中,L 2和L 3一起形成-C≡C-。
在一个更具体的实施方案中,L 2和L 3一起形成反式的-CR=CR-,或-C≡C-;在另一个更具体的实施方案中,L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-。
L 4和L 5
在一个具体实施方案中,L 4和L 5一起形成顺式的-CR=CR-;在一个具体实施方案中,L 4和L 5一起形成反式的-CR=CR-;在一个具体实施方案中,L 4和L 5一起形成-C≡C-。
在一个更具体的实施方案中,L 4和L 5一起形成顺式或反式的-CR=CR-;在另一个更具体的实施方案中,L 4和L 5一起形成顺式的-CR=CR-;在另一个更具体的实施方案中,L 4和L 5一起形成顺式或反式的-CH=CH-,或-C≡C-;在另一个更具体的实施方案中,L 4和L 5一起形成顺式或反式的-CH=CH-;在另一个更具体的实施方案中,L 4和L 5一起形成顺式的-CH=CH-。
Z
在一个具体实施方案中,Z为C=O;在另一个具体实施方案中,Z为C(R”) 2;在另一个具体实施方案中,Z为CH 2
R 1
在一个具体实施方案中,R 1为H;在另一个具体实施方案中,R 1为C 1-6烷基;在另一个具体实施方案中,R 1为C 1-6卤代烷基;在另一个具体实施方案中,R 1为C 2-6烯基;在另一个具体实施方案中,R 1为C 2-6炔基;在另一个具体实施方案中,R 1为C 3-7环烷基;在另一个具体实施方案中,R 1为3至7元杂环基。在另一个具体实施方案中,R 1选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基或3至7元杂环基。
以上任一具体实施方案中的任一技术方案或其任意组合,可以与其它具体实施方案中的任一技术方案或其任意组合进行组合。例如,K的任一技术方案或其任意组合,可以与E、L 1-L 5等的任一技术 方案或其任意组合进行组合。条件是,仅当K为K1、K2或K4时,L 4和L 5可以一起形成-C≡C-。本发明旨在包括所有这些技术方案的组合,限于篇幅,不再一一列出。
在更具体的实施方案中,本发明提供了通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000008
其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2或C(R’) 2C(R’) 2;或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;或者L 4和L 5一起形成顺式或反式的-CR=CR-;
L 5为化学键、O、S、NR*或C(R) 2
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键或CH 2;或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;或者L 4和L 5一起形成顺式或反式的-CH=CH-;
L 5为O、S、NH或CH 2
Z为C=O或CH 2
在更具体的实施方案中,本发明提供了上述通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键或C(R’) 2;或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
L 5为O、S或NR*;
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键或CH 2;或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;
L 5为O、S或NH;
Z为C=O或CH 2
在更具体的实施方案中,本发明提供了上述通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键或C(R’) 2;或者L 2和L 3一起形成反式的-CR=CR-,或-C≡C-;
L 5为O或S;
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基;
R’选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基;
R”选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基;
在更具体的实施方案中,本发明提供了上述通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键或CH 2
L 5为O;
Z为C=O或CH 2
在更具体的实施方案中,本发明提供了通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000009
Figure PCTCN2022093956-appb-000010
其中,
L 1为化学键、C(R) 2或SO 2
L 2选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 4选自化学键、C(R’) 2或C(R’) 2C(R’) 2
L 5选自化学键、O、S、NR*、C(R) 2或SO 2
或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
或者L 4和L 5一起形成顺式或反式的-CR=CR-;
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为化学键、CH 2或SO 2
L 2选自化学键、O、S、NR*、CH 2、CH 2CH 2、1,3-亚苯基、1,4-亚苯基、1,4-亚三唑基、3,5-亚吡啶基或2,4-亚嘧啶基;
L 3选自化学键、O、S、NR*、CH 2、CH 2CH 2或1,4-亚三唑基;
L 4选自化学键、CH 2或CH 2CH 2
L 5选自化学键、O、S、NR*、CH 2或SO 2
或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;
或者L 4和L 5一起形成顺式或反式的-CH=CH-;
Z为C=O或CH 2
其中,R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的 混合物,其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5为S;
或者L 4和L 5一起形成顺式的-CR=CR-;
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键、CH 2或CH 2CH 2
L 5为S;
或者L 4和L 5一起形成顺式的-CH=CH-;
Z为C=O或CH 2
在更具体的实施方案中,本发明提供了上述通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5为S;
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键、CH 2或CH 2CH 2
L 5为S;
Z为C=O或CH 2
在更具体的实施方案中,本发明提供了通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000011
其中,
L 1为C(R) 2
L 2选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
L 3和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5为O、S、NR*、C(R) 2或SO 2
或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2选自化学键、CH 2、CH 2CH 2或1,3-亚苯基;
L 3和L 4独立地选自化学键、CH 2或CH 2CH 2
L 5为O、S、NH、CH 2或SO 2
或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;
Z为C=O或CH 2
其中,R*选自H、C 1-6烷基或C 1-6卤代烷基。
在更具体的实施方案中,本发明提供了上述通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5为O或C(R) 2
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键、CH 2或CH 2CH 2
L 5为O或CH 2
Z为C=O或CH 2
其中,R*选自H、C 1-6烷基或C 1-6卤代烷基。
在更具体的实施方案中,本发明提供了通式(V)、(V-1)或(V-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000012
其中,
L 1为C(R) 2
L 5为O、S、NR*或C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(V)、(V-1)或(V-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5为C(R) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基或C 1-6卤代烷基;
R’选自H、D、卤素、-CN、C 1-6烷基或C 1-6卤代烷基。
在更具体的实施方案中,本发明提供了上述通式(V)、(V-1)或(V-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2
L 5为CH 2
在更具体的实施方案中,本发明提供了上述通式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000013
其中,
L 1为C(R) 2
L 2、L 3和L 4独立地选自化学键、C(R’) 2或C(R’) 2C(R’) 2
L 5为C(R) 2
或者L 4和L 5一起形成顺式或反式的-CR=CR-,或-C≡C-;
Z为C=O或C(R”) 2
R 1选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-7环烷基或3至7元杂环基;
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2、L 3和L 4独立地选自化学键或CH 2
L 5为CH 2
或者L 4和L 5一起形成顺式或反式的-CH=CH-,或-C≡C-;
Z为C=O或CH 2
R 1选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基或3至7元杂环基。
在更具体的实施方案中,本发明提供了上述通式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2022093956-appb-000014
其中,
L 1为C(R) 2
L 2和L 4独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
L 5为C(R) 2
Z为C=O或C(R”) 2
其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
在更具体的实施方案中,本发明提供了上述通式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L 1为CH 2
L 2和L 4独立地选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2
L 3选自化学键、O、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2
L 5为CH 2
Z为C=O或CH 2
在更具体的实施方案中,本发明提供了通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,所述化合物选自以下:
Figure PCTCN2022093956-appb-000015
Figure PCTCN2022093956-appb-000016
Figure PCTCN2022093956-appb-000017
Figure PCTCN2022093956-appb-000018
Figure PCTCN2022093956-appb-000019
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理 缔合可包括氢键键合。常规溶剂包括水、甲醇、乙醇、乙酸、DMSO、THF、乙醚等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R·x H 2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R·0.5 H 2O))和多水合物(x为大于1的数,例如,二水合物(R·2 H 2O)和六水合物(R·6 H 2O))。
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元素组成。
本发明还包括同位素标记的化合物(同位素变体),它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如 3H和 14C)的那些可用于药物和/或底物组织分布测定。氚、即 3H和碳-14、即 14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即 2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(I)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(I)化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH) 的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
本发明还提供药物制剂,包含治疗有效量的式(I)化合物或其治疗学上可接受的盐和其药学上可接受的载体、稀释剂或赋形剂。所有这些形式都属于本发明。
药物组合物和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的本发明化合物。在一些实施方案中,所述药物组合物包含治疗有效量的本发明化合物。在一些实施方案中,所述药物组合物包含预防有效量的本发明化合物。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
给药
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂 量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington's Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材料的描述可在Remington's Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精。
联合用药
本文中定义的治疗可作为单独治疗应用,或除本发明化合物之外,可包括常规外科手术或放疗或化疗。因此,本发明化合物还可与用于治疗癌症的现有治疗药剂联合使用。
除了使用本发明化合物治疗以外,还涉及到常规的手术或放射疗法或化学疗法或免疫疗法。这种化学疗法与本发明化合物可以同时地、连续地、或分别地给药,并且可包含以下类型的抗肿瘤剂的一种或多种。
具体实施方式
后文所用缩写含义如下所示:
Figure PCTCN2022093956-appb-000020
Figure PCTCN2022093956-appb-000021
本发明的化合物结构是通过核磁共振(NMR)或/和液质联用色谱(LC-MS)来确定的。NMR化学位移(δ)以百万分之一(ppm)的单位给出。NMR的测定是用Bruker Avance III HD 500MHz核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代甲醇(CD 3OD)和氘代氯仿(CDCl 3),内标为四甲基硅烷(TMS)。
液质联用色谱LCMS的测定用Waters H-Class+SQD2质谱仪。HPLC的测定使用SHIMADZU LC-20A高压液相色谱仪(ACE Excel 2 C18 50×2.1mm Column)。样品制备分离是用Waters AutoPurificaiton高压液相色谱仪(C18 5μm 50×30mm Column)。
薄层层析硅胶板使用良臣-LCGY硅胶板(20×20cm,丙烯酸0.4-0.5mm),TLC采用的规格是0.15mm~0.20mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
柱层析一般使用山东青岛工厂硅胶(200~300目)为载体。
中压制备色谱采用的是Biotage(Isolera One automated flash purification system(200-400nm))。
本发明实施例中的起始原料是已知的并且可以在市场上买到,或者可以采用或按照本领域已知的方法来合成。
在无特殊说明的情况下,本发明的所有反应均在连续的磁力搅拌下,在干燥氮气或氩气氛下进行,溶剂为干燥溶剂,反应温度单位为摄氏度。
本发明化合物和中间体可以采用以下通用合成方法进行制备:
方案1:
Figure PCTCN2022093956-appb-000022
其中K-H中的H为氨基氢,R为磺酸酯基(例如甲磺酸酯基)或卤素(例如Br和I)。
将K-H与R-L 1-L 2-L 3-L 4-L 5-E在酰胺偶联的条件下进行反应,形成本发明化合物K-L 1-L 2-L 3-L 4-L 5-E。此外,本发明的一些化合物还可以通过对本发明其他化合物进行还原、烷基化等(例如氢化、甲基化)而获得。
例如,如下方案1-1至1-6:
方案1-1
Figure PCTCN2022093956-appb-000023
胺与磺酸酯在碱性体系下进行亲核取代。
方案1-2
Figure PCTCN2022093956-appb-000024
胺与溴化物在碱性体系下进行亲核取代。
方案1-3(还原反应)
Figure PCTCN2022093956-appb-000025
Pd/CaCO 3催化炔还原,喹啉毒化。
方案1-4
Figure PCTCN2022093956-appb-000026
Pd/C催化烯烃还原。
方案1-5(甲基化反应)
Figure PCTCN2022093956-appb-000027
方案1-6(偶联反应)
Figure PCTCN2022093956-appb-000028
中间体合成方案2:
Figure PCTCN2022093956-appb-000029
将3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮与炔在钯催化剂和CuI的催化下进行偶联反应得到相应的醇,该相应的醇再与MsCl进行亲核取代得到磺酸酯。
中间体合成方案3:
Figure PCTCN2022093956-appb-000030
SH、OH、伯胺或仲胺与卤化物在碱性体系下进行亲核取代。
中间体合成方案4:
Figure PCTCN2022093956-appb-000031
硫醚在m-CPBA作用下被氧化。
此外,本发明的一些化合物或其他中间体可以参照方案1中的具体方案或方案2-4进行制备。
实施例1
3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000032
第一步:3-(4-(3-羟基丙-1-炔-1-基)-1-氧代异吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000033
将化合物3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-炔-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮(500mg,1.3mmol)加入一个100mL的反应瓶中,随后加入二氯甲烷(20mL)和TFA(2mL)室温下搅拌3h。将反应液浓缩然后加入饱和碳酸氢钠(50mL),二氯甲烷(2*50mL)萃取,浓缩正相柱层析纯化分离(二氯甲烷:甲醇=10:1)得标题化合物3-(4-(3-羟基丙-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色固体,270mg,收率69%)。MS m/z(ESI):299.2.[M+H] +.
第二步:3-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丙-2-炔-1-基甲磺酸酯的制备
Figure PCTCN2022093956-appb-000034
将化合物3-(4-(3-羟基丙-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(270mg,0.91mmol)加入一个500mL的反应瓶中,随后加入DCM(250mL),然后依次加入TEA(275mg,2.7mmol),MsCl(207mg,1.8mmol),加完后室温搅拌2h。将反应混合物水洗(2*100mL),饱和食盐水洗(100mL),浓缩后打浆(石油醚:乙酸乙酯=1:1)得标题化合物3-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丙-2-炔-1-基甲磺酸酯(白色固体,0.21g,收率62%)。
MS m/z(ESI):377.5[M+H] +.
第三步:3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000035
将化合物3-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丙-2-炔-1-基甲磺酸酯(0.2g,0.53mmol),原料2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(0.3g,0.53mmol)加入一个25mL的反应瓶中,随后加入NMP(4mL),然后在氮气保护下加入碘化钠(0.158g,1.05mmol),以及DIPEA(0.2g,1.58mmol),加完后80℃搅拌16h。将反应混合物加入(100mL)乙酸乙酯,水洗(100mL),食盐水洗(100mL),有机相浓缩后先用正相柱层析(二氯甲烷:甲醇=10:1)初步纯化,再反相柱(乙腈/(水+0.05%HCl))纯化得到标题化合物3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐(白色固体,0.32g,收率71%,盐酸盐)。
1H NMR(800MHz,CD 3OD)δ8.21(s,2H),7.86(d,J=7.6Hz,1H),7.82(dd,J=7.6,0.6Hz,1H),7.77-7.69(m,1H),7.69-7.55(m,3H),7.49(t,J=7.4Hz,1H),7.37(s,1H),7.10(d,J=7.6Hz,1H),5.21(dd,J=13.4,5.2Hz,1H),4.70(dd,J=105.7,17.7Hz,2H),4.47(s,2H),3.95(d,J=12.4Hz,5H),3.84(d,J=39.4Hz,9H),3.61(d,J=49.5Hz,2H),2.93(ddd,J=17.8,13.7,5.4Hz,1H),2.79(ddd,J=17.6,4.4,2.3Hz,1H),2.64(qd,J=13.3,4.5Hz,1H),2.54(d,J=12.1Hz,2H),2.42(d,J=10.3Hz,2H),2.23-2.16(m,1H),1.87(d,J=13.5Hz,6H)。MS m/z(ESI):850.9[M+H] +
实施例2
3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000036
第一步:3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000037
参照实例1的方法,在本领域可理解的适当条件下,采用2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化和中间体4-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丁-3-炔-1-基甲磺酸酯制备得到目标化合物3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐(淡黄色固体,4.0mg,收率18%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.20(s,2H),7.80(d,J=6.9Hz,1H),7.76-7.70(m,2H),7.69-7.58(m,2H),7.51(dt,J=24.8,7.5Hz,2H),7.35(s,1H),7.08(d,J=8.2Hz,1H),5.21(dd,J=13.3,5.2Hz,1H),4.60(dd,J=49.4,17.6Hz,2H),3.89(d,J=62.8Hz,14H),3.63(dd,J=16.8,9.9Hz,4H),3.17(dd,J=8.9,4.8Hz,2H),2.99-2.86(m,1H),2.86-2.75(m,1H),2.65-2.56(m,1H),2.52(d,J=11.4Hz,2H),2.39(d,J=10.9Hz,2H),2.24-2.16(m,1H),1.87(d,J=13.6Hz,6H)。MS m/z(ESI):864.4[M+H] +.
实施例3
3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000038
第一步:3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-炔-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000039
将3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(2.0g,6.2mmol)加入一个100mL的反应瓶中,随后加入双三苯基膦二氯化钯(0.43g,0.62mmol),碘化亚铜(0.24g,1.3mmol),然后在氮气保护下加入DMF(25mL),TEA(2.5mL),以及2-(丙-2-炔-1-氧基)四氢-2H-吡喃(1.73g,12.4mmol),然后升温至90℃搅拌16小时。TLC检测反应结束后,将反应混合物浓缩,然后柱层析纯化(二氯甲烷:甲醇=20:1),得标题化合物3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-炔-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,1.40g,收率59.1%)。MS m/z(ESI):383.3[M+H] +.
第二步:3-(4-(3-羟基丙基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000040
将3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-炔-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮(1.0g,2.6mmol)加入一个500mL的反应瓶中,随后加入乙醇(150mL),湿钯碳(5%,0.5g),然后在氢气氛围下升温至50℃搅拌16h。LCMS检测反应结束后,将反应混合物过滤浓缩后柱层析纯化(二氯甲烷:甲醇=10:1),得标题化合物3-(4-(3-羟基丙基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,0.5g,收率63.2%)。MS m/z(ESI):303.5[M+H] +.
第三步:3-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丙基甲磺酸酯的制备
Figure PCTCN2022093956-appb-000041
将3-(4-(3-羟基丙基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(0.5g,1.66mmol)加入一个500mL的反应瓶中,随后加入二氯甲烷(220mL),然后在室温氮气保护下加入TEA(0.84g,8.3mmol),以及甲基磺酰氯(0.57g,4.98mmol),加完后室温搅拌0.5h。TLC检测反应结束后,将反应混合物水洗(2*150mL),饱和食盐水洗(150mL)。有机相浓缩后打浆(石油醚:乙酸乙酯=1:1),得标题化合物3-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丙基甲磺酸酯(白色固体,0.58g,收率92.2%)。
MS m/z(ESI):381.5[M+H] +.
第四步:3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000042
将3-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)丙基甲磺酸酯(0.26g,0.68mmol)、(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(0.3g,0.53mmol)加入一个25mL的反应瓶中,随后加入NMP(6mL),然后在氮气保护下加入碘化钠(0.12g,0.8mmol),以及DIPEA(0.2g,1.58mmol),加完后80℃搅拌16h。LCMS检测反应结束后,将反应混合物加入乙酸乙酯(100mL),然后水洗(50mL),食盐水洗(50mL)。有机相浓缩后先用正相柱层析(二氯甲烷:甲醇=10:1)初步纯化,然后用反相柱(乙腈/(水+0.05%HCl))进一步纯化,得标题化合物3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,0.27g,收率60%)。
1H NMR(500MHz,MeOD)δ8.24(s,1H),8.16(s,1H),7.77-7.67(m,2H),7.63(t,J=7.9Hz,1H),7.60-7.44(m,4H),7.19(s,1H),6.95(d,J=8.1Hz,1H),5.19(dd,J=13.3,5.2Hz,1H),4.58(dd,J=49.1, 17.1Hz,2H),3.99-3.56(m,14H),3.38(dd,J=20.4,12.4Hz,4H),2.98-2.75(m,4H),2.57(qd,J=13.3,4.6Hz,1H),2.44(d,J=11.9Hz,2H),2.31-2.16(m,5H),1.87(d,J=13.6Hz,6H).MS m/z(ESI):854.9[M+H] + .
实施例4
3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000043
第一步:3-(4-(6-羟基己-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000044
将3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(500mg,1.54mmol)加入一个50mL的反应单口瓶中,随后分别加入双三苯基膦二氯化钯(21.7mg,0.03mmol),碘化亚铜(14.7mg,0.08mmol),无水DMF(15mL),三乙胺(2.5mL)和己-5-炔-1-醇(304mg,3.09mmol),氩气置换3次,然后升温至80℃并搅拌12小时。反应结束后,将反应混合物过滤,浓缩,粗品经柱层析纯化(洗脱剂梯度:二氯甲烷:甲醇=20:1),旋干得标题化合物3-(4-(6-羟基己-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色固体,380mg,收率72%)。MS m/z(ESI):341.5[M+H] +
第二步:3-(4-(6-羟基己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000045
将3-(4-(6-羟基己-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(220mg,0.65mmol)加入到一个100mL的反应单口瓶中,随后加入乙醇(50mL),湿钯碳(5%,200mg),氢气(25psi)置换3次,然后升温至50℃并搅拌16h。反应结束后,将反应混合物过滤,浓缩干得标题化合物3-(4-(6-羟基己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(类白色固体,210mg,收率96%),粗品未进行进一步纯化,直接用于下一步反应。MS m/z(ESI):345.6[M+H] +
第三步:6-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)己基甲磺酸酯的制备
Figure PCTCN2022093956-appb-000046
将3-(4-(6-羟基己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(200mg,0.58mmol)加入一个100mL的反应瓶中,随后加入无水二氯甲烷(50mL)和三乙胺(293mg,2.89mmol),然后在冰水浴下缓慢滴加甲基磺酰氯(200mg,1.74mmol)的二氯甲烷溶液,滴毕,允许升温至室温并搅拌0.5h。随后向反应混合物中加入水(50mL)淬灭反应,二氯甲烷萃取(2*30mL),合并有机相,水洗(2*50mL),饱和食盐水洗(50mL),无水硫酸钠干燥,减压浓缩。向粗品中加入(石油醚:乙酸乙酯(V 1:V 2)=1:1)(20mL)打浆,抽滤得标题化合物6-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)己基甲磺酸酯(白色固体,200mg,收率82%)。
第四步:3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000047
将6-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)己基甲磺酸酯(20mg,0.05mmol),(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(27mg,0.05mmol)加入一个25mL的反应瓶中,随后加入碘化钠(7.1mg,0.05mmol),NMP(3mL)和DIPEA(18.4mg,0.15mmol),加完后氮气保护下缓慢升温至80℃并搅拌16h。应结束后,将反应混合物过滤,滤液经反相制备(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-50%),减压除去乙腈,冻干得标题化合物3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐(白色固体,10.2mg,收率24%,盐酸盐)。
1H NMR(500MHz,MeOD)δ8.24(s,1H),8.17(s,1H),7.76-7.69(m,1H),7.67-7.61(m,2H),7.58-7.51(m,1H),7.51-7.45(m,3H),7.23(s,1H),6.99(d,J=8.1Hz,1H),5.19(dd,J=13.3,5.2Hz,1H),4.52(q,J=17.0Hz,2H),4.00-3.56(m,14H),3.50-3.40(m,2H),3.29-3.23(m,2H),2.98-2.89(m,1H),2.84-2.73(m,3H),2.62-2.52(m,1H),2.47(d,J=12.3Hz,2H),2.34-2.24(d,J=11.1Hz,2H),2.22-2.16(m,1H),1.89(s,3H),1.86(s,3H),1.84-1.79(m,2H),1.78-1.70(m,2H),1.52-1.42(m,4H)。MS m/z(ESI):897.3[M+H] +.
实施例5
3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000048
第一步:7-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)庚基甲磺酸酯的制备
Figure PCTCN2022093956-appb-000049
将化合物7-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)庚-6-炔-1-基甲磺酸酯(100mg,0.23mmol)加入一个50mL的反应瓶中,随后加入乙醇(15mL),湿钯碳(30mg),然后在氢气氛围下升温至40℃搅拌2h。将反应混合物过滤,浓缩得标题化合物7-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)庚基甲磺酸酯(白色固体,98mg,收率97%,盐酸盐)。MS m/z(ESI):437.8.[M+H] +.
第二步:3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000050
参照方案2的方法,在本领域可理解的适当条件下,采用2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化和中间体7-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)庚基甲磺酸酯制备得到标题化合物3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐(白色固体,33.0mg,收率69%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.20(s,2H),7.73(ddd,J=13.8,7.7,1.3Hz,1H),7.64(dq,J=12.3,7.6Hz,3H),7.54-7.43(m,3H),7.36(s,1H),7.09(d,J=7.8Hz,1H),5.19(dd,J=13.3,5.2Hz,1H),4.51(q,J=16.9Hz,2H),4.00-3.57(m,16H),3.29-3.22(m,2H),2.98-2.86(m,1H),2.85-2.70(m,3H),2.57(ddd,J=26.7,13.4,4.7Hz,3H),2.39(d,J=11.8Hz,2H),2.19(dtd,J=12.8,5.3,2.3Hz,1H),1.87(d,J=13.6Hz,6H),1.73(dd,J=24.3,17.7Hz,4H),1.43(s,6H)。LCMS(MS m/z(ESI):910.4[M+H] +.
实施例6
3-(4-((4-((4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000051
第一步:3-(4-((4-(羟甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000052
将化合物3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(2.0g,6.2mmol),4-乙炔基苄醇(1.6g,12.4mmol),二(三苯基磷基)二氯化钯(0.43mg,0.62mmol),碘化亚铜(0.24mg,1.2mmol),三乙胺(8.2mL,61.9mmol)溶于无水DMF(15mL)中。反应液用氮气置换三次,升温至90℃,搅拌18小时。反应完成后,将反应液减压浓缩,加入二氯甲烷(30mL),搅拌2分钟,有淡黄色固体生成。将混合物过滤,滤饼用二氯甲烷(5mL x 3)洗涤,收集滤饼得到标题化合物3-(4-((4-(羟甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(灰白色固体,2.1g,收率90%)。
1H NMR(500MHz,DMSO-d 6)δ11.02(s,1H),7.81-7.77(m,2H),7.61-7.58(m,3H),7.40(d,J=5.0Hz,2H),5.32(t,J=5.0Hz,1H),5.18(dd,J=5.0,10.0Hz,1H),4.61-4.43(m,4H),2.97-2.90(m,1H),2.63-2.56(m,1H),2.54-2.51(m,1H),2.05-2.01(m,1H)。MS m/z(ESI):375.0[M+H] +.
第二步:3-(4-((4-(溴甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000053
将化合物3-(4-((4-(羟甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮((1.5g,4.0mmol)溶于无水二氯甲烷(150mL)中,室温下向反应液中滴加三溴化磷(1.1g,4.0mmol)。反应液搅拌2小时。反应完成后,加水(50mL)淬灭。反应液静置分层,水相用二氯甲烷(50mL x 3)萃取,合并的有机相用无水硫酸钠干燥,过滤,减压浓缩。向剩余物加入乙酸乙酯(50mL),搅拌2分钟,有固体生成。将混合物过滤,滤饼用二氯甲烷(5mL x 3)洗涤,收集滤饼得标题化合物3-(4-((4-(溴甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,1.3g,收率74%)。
1H NMR(500MHz,DMSO-d 6)δ11.03(s,1H),7.83-7.79(m,2H),7.64-7.60(m,3H),7.54(d,J=5.0Hz,2H),5.18(dd,J=5.0,10.0Hz,1H),4.76(s,2H),4.62-4.44(m,2H),3.03-2.89(m,1H),2.59-2.67(m,1H),2.53-2.53(m,1H),2.06-2.01(m,1H)。MS m/z(ESI):437[M+H] +.
第三步:3-(4-((4-((4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000054
将化合物3-(4-((4-(溴甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(1.1g,2.5mmol),化合物2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(1.4g,2.5mmol)溶于无水N-甲基吡咯烷酮(15mL)中,室温下向反应液中加入三乙胺(0.6g,5.0mmol)。反应室温液搅拌2小时。反应完成后,将反应液倒入水中(100mL),搅拌10分钟。有白色固体析出。将混合物过滤,滤饼用水(10mL x 3)洗涤,收集滤饼,溶于二氯甲烷/甲醇溶液中。溶液用无水硫酸钠干燥,过滤,减压浓缩。剩余物用柱层析纯化(二氯甲烷/甲醇=10/1)得淡黄色粗品。然后再反向柱层析纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-60%)得到标题化合物3-(4-((4-((4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)甲基)苯基)乙炔基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐(白色固体,1.5g,收率74%,盐酸盐) 1H NMR(500MHz,CD 3OD)δ8.22(s,1H),8.17(s,1H),7.83(d,J=10.0Hz,1H),7.79(d,J=10.0Hz,1H),7.76-7.69(m,5H),7.64(t,J=10.0Hz,1H),7.59(t,J=10.0Hz,1H),7.56-7.46(m,2H),7.25(s,1H),7.00(d,J=5.0Hz,1H),5.20(dd,J=5.0,10.0Hz,1H),4.63(q,J=15.0Hz,2H),4.54(s,2H),3.96(s,2H),3.94(s,3H),3.88-3.65(m,9H),3.55-3.41(m,2H),2.97-2.89(m,1H),2.82-2.77(m,1H),2.62-2.51(m,1H),2.48-2.41(m,2H),2.34-2.25(m,2H),2.23-2.19(m,1H),1.88(s,3H),1.86(s,3H).
MS m/z(ESI):926.0[M+H] +.
实施例7
(Z)-3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000055
第一步:(Z)-3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000056
将化合物3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(700mg,0.79mmol)加入一个250mL的反应瓶中,随后加入乙醇(150mL),5%Pd/CaCO 3(300mg)以及喹啉(1滴),然后在氢气氛围下室温搅拌2.5h。将反应混合物过滤,减压浓缩,粗品先经正相柱层析(洗脱剂梯度:二氯甲烷/甲醇=0-7%),最后经反向柱纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-50%),减压除去乙腈,冻干得到标题化合物(Z)-3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,460mg,盐酸盐,收率65%,盐酸盐)。 1H NMR(500MHz,MeOD)δ8.19(s,2H),7.76-7.70(m,2H),7.68-7.62(m,1H),7.61-7.54(m,3H),7.51-7.45(m,1H),7.33(s,1H),7.07(d,J=8.4Hz,1H),6.61(d,J=11.5Hz,1H),5.98-5.90(m,1H),5.19(dd,J=13.4,5.1Hz,1H),4.45(dd,J=41.2,17.3Hz,2H),4.00-3.54(m,18H),3.26-3.20(m,2H),2.97-2.87(m,1H),2.83-2.75(m,1H),2.59-2.48(m,3H),2.42-2.33(m,4H),2.22-2.16(m,1H),2.02-1.92(m,2H),1.88(s,3H),1.85(s,3H)。MS(ESI)m/z:880.7[M+H] +
实施例8
3-(4-((2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000057
第一步:3-(4-((溴乙基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000058
以来那度胺和1,2-二溴乙烷为原料,参照实施例10的第一步的方法制备3-(4-((溴乙基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,MS m/z(ESI):366.04[M+H] +
第二步:3-(4-((2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000059
将化合物3-(4-((2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(20mg,23.4umol)溶于2mL乙腈和2mL水中,再加入甲醛水溶液(37%,20.1mg,234umol),室温下搅拌2小时。然后再加入氰基 硼氢化钠(29.4mg,469umol),室温下搅拌16小时。反应结束后,将反应液过滤后浓缩,然后再反向柱层析纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-60%)得到标题化合物3-(4-((2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐。(白色固体,7.2mg,收率:35%,盐酸盐)
1H NMR(500MHz,CD 3OD)δ8.26-8.24(m,1H),8.11(s,1H),7.70(ddd,J=14.0,7.7,1.5Hz,1H),7.65-7.51(m,3H),7.44(ddd,J=12.5,6.8,3.0Hz,3H),6.98(s,1H),6.79(d,J=8.7Hz,1H),5.20(dd,J=13.3,5.2Hz,1H),4.79-4.63(m,2H),3.97-3.88(m,5H),3.80-3.42(m,12H),3.18(s,2H),3.04(s,3H),2.93(ddd,J=18.5,13.7,5.4Hz,1H),2.80(ddd,J=17.6,4.6,2.3Hz,1H),2.67(qd,J=13.2,4.6Hz,1H),2.42-2.29(m,3H),2.21(dtd,J=12.8,5.3,2.3Hz,1H),2.09(t,J=12.3Hz,2H),1.88(d,J=13.6Hz,6H).
MS m/z(ESI):869.8[M+H] +.
实施例9
3-(6-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000060
第一步:3-(6-(5-羟基戊-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000061
参照实例3的方法,在本领域可理解的适当条件下,采用戊-4-炔-1-醇和中间体3-(6-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮制备得到标题化合物3-(6-(5-羟基戊-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色固体,180mg,收率89%)。MS m/z(ESI):327.4.[M+H] +.
第二步:5-(2-(2,6-二羰基哌啶-3-基)-3-羰基异二氢吲哚-5-基)戊-4-炔-1-基甲磺酸酯的制备
Figure PCTCN2022093956-appb-000062
参照方案1的方法,在本领域可理解的适当条件下,采用中间体3-(6-(5-羟基戊-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮制备得到标题化合物5-(2-(2,6-二羰基哌啶-3-基)-3-羰基异二氢吲哚-5-基)戊-4-炔-1-基甲磺酸酯(白色固体,170mg,收率76%)。MS m/z(ESI):405.5[M+H] +.
第三步:3-(6-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000063
参照方案1的方法,在本领域可理解的适当条件下,采用采用2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化和中间体5-(2-(2,6-二羰基哌啶-3-基)-3-羰基异二氢吲哚-5-基)戊-4-炔-1-基甲磺酸酯制备得到目标化合物3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,30.0mg,收率65%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.20(d,J=16.3Hz,2H),7.82(s,1H),7.80-7.61(m,4H),7.57(d,J=8.0Hz,1H),7.50(t,J=7.1Hz,1H),7.40(s,1H),7.12(d,J=8.5Hz,1H),5.15(dd,J=13.3,5.2Hz,1H),4.51(q,J=17.5Hz,2H),4.05-3.60(m,16H),3.54-3.45(m,2H),2.90(ddd,J=18.8,13.6,5.4Hz,1H),2.78(ddd,J=17.6,4.5,2.3Hz,1H),2.69(t,J=6.8Hz,2H),2.60-2.36(m,5H),2.23-2.08(m,3H),1.87(d,J=13.6Hz,6H).MS m/z(ESI):878.7[M+H] +.
实施例10
3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000064
第一步:(E)-3-(4-((4-溴丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000065
将化合物3-(4-氨基-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(200mg,0.77mmol),反式-1,4-二溴丁二烯(165mg,0.77mmol)溶于无水N-甲基吡咯烷酮(5mL)中,室温下向反应液中加入二异丙基乙基胺(199mg,1.54mmol),搅拌18小时。反应完成后,反应液用水(10mL)稀释,乙酸乙酯(50mL x 3)萃取。合并的有机相用无水硫酸钠干燥,过滤,减压浓缩。剩余物用柱层析纯化(二氯甲烷/甲醇=10/1)得到标题化合物(E)-3-(4-((4-溴丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色 固体,130mg,收率43%,盐酸盐)。 1H NMR(500MHz,DMSO-d 6)δ11.01(s,1H),7.28(t,J=5.0Hz,1H),6.96(d,J=5.0Hz,1H),6.72(d,J=5.0Hz,1H),5.93-5.88(m,2H),5.10-5.13(m,1H),4.28-4.14(m,4H),3.87-3.84(m,2H),2.96-2.89(m,1H),2.66-2.63(m,1H),2.33-2.30(m,1H),2.06-2.02(m,2H)。
MS m/z(ESI):392.0[M+H] +.
第二步:(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000066
将化合物(E)-3-(4-((4-溴丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(13mg,35umol),化合物2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(20mg,35umol)溶于无水N-甲基吡咯烷酮(2mL)中,室温下向反应液中加入二异丙基乙基胺(18mg,0.14mmol)。反应液在室温下搅拌18小时。反应完成后,反应液通过反向柱层析纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-60%)得到标题化合物(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,3mg,收率10%)。
1H NMR(500MHz,CD 3OD)δ7.82-7.73(m,2H),7.73-7.71(m,1H),7.70-7.63(m,2H),7.45-7.42(m,1H),7.39(s,1H),7.23-7.20(m,2H),7.02-6.95(m,2H),6.28-6.24(m,1H),5.89-5.82(m,1H),5.16(dd,J=5.0,10.0Hz,1H),4.44-4.35(m,2H),4.05(s,2H),3.77(s,3H),3.69-3.61(m,4H),3.62-3.51(m,10H),2.92-2.89(m,1H),2.82-2.78(m,1H),2.54-2.48(m,3H),2.32-2.19(m,3H),1.89(s,3H),1.86(s,3H)。
MS m/z(ESI):881.0[M+H] +.
第三步:3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000067
将化合物(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(11mg,12umol)溶于乙醇(3mL)中,室温下向反应液中加入林德拉钯催化剂(5mg),反应体系用氢气置换三次。反应液在氢气氛下(1atm)搅拌4小时。反应完成后,反应液过滤,滤液减压浓缩,然后再反向柱层析纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-60%)得到标题化合物3-(4-((4-(4-(1-(4-((5-氯-4- ((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮。(白色固体,0.4mg,收率4%,盐酸盐)MS m/z(ESI):884.2[M+H] +.
实施例11
(2S,4S)-1-((S)-2-(10-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)癸酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺
Figure PCTCN2022093956-appb-000068
第一步:(2S,4S)-1-((S)-2-(10-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)癸酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺的制备
Figure PCTCN2022093956-appb-000069
将化合物(2S,4S)-1-((S)-2-(10-溴癸酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺(23.3mg,35.1umol),化合物2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(20mg,35.1umol)溶于无水N-甲基吡咯烷酮(2mL)中,室温下向反应液中加入碘化钠(5.3mg,35.1umol),二异丙基乙基胺(13.6mg,105umol)。反应液升温至70℃,搅拌16小时。反应完成后,反应液降至室温,过滤,然后再反向柱层析纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-60%)得到标题化合物(2S,4S)-1-((S)-2-(10-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)癸酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺,盐酸盐(白色固体,17.8mg,收率44%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.87(s,1H),8.33(dd,J=8.4,4.5Hz,1H),7.66(d,J=8.8Hz,1H),7.60(ddd,J=14.1,7.8,1.6Hz,1H),7.54-7.38(m,5H),7.29-7.22(m,1H),6.66(d,J=2.6Hz,1H),6.45(dd,J=8.8,2.6Hz,1H),4.65(s,H),4.60-4.47(m,4H),4.35(d,J=15.4Hz,1H),3.90(d,J=11.0Hz,1H),3.80(dd,J=10.9,3.9Hz,1H),3.69(d,J=12.1Hz,2H),2.78-2.65(m,6H),2.47(s,6H),2.35-2.16(m,4H),2.13-1.99(m,4H),1.83(d,J=13.5Hz,7H),1.73-1.50(m,7H),1.33(s,13H),1.03(s,10H)。
MS m/z(ESI):1153.2[M+H] +.
实施例12
(E)-3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000070
第一步:(E)-3-(4-(5-羟基戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000071
将3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(200mg,619umol),3-乙烯基-1-丙醇(107mg,1.24mmol),三二亚苄基丙酮二钯(113mg,124umol),三叔丁基膦四氟硼酸盐(35.9mg,124umol),N-甲基二环己基胺(484mg,2.48mmol)溶于无水二氧六环(10mL)中。反应液用氮气置换三次,升温至55℃,搅拌16小时。反应完成后,将反应液过滤后加入乙酸乙酯和水萃取,合并有机相浓缩,柱层析得到标题化合物(E)-3-(4-(5-羟基戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(浅灰色产品,300mg,crude)。
1H NMR(500MHz,DMSO-d 6)δ11.01(d,J=7.5Hz,1H),7.72(dd,J=7.8,1.1Hz,1H),7.64-7.54(m,1H),7.53-7.42(m,1H),6.49(d,J=16.1Hz,1H),6.40(d,J=16.0Hz,1H),5.14(ddd,J=13.3,9.7,5.9Hz,1H),4.61-4.43(m,2H),3.52-3.42(m,2H),3.18(d,J=5.3Hz,1H),2.94(s,1H),2.67-2.56(m,1H),2.47(d,J=13.1Hz,1H),2.36-2.22(m,2H),2.08-1.97(m,1H),1.63(dq,J=8.2,6.5Hz,2H)。
MS m/z(ESI):329.4[M+H] +.
第二步:(E)-5-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)戊-4-烯-1-基甲磺酸酯的制备
Figure PCTCN2022093956-appb-000072
将(E)-3-(4-(5-羟基戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(50.0mg,153umol)和三乙胺(61.6mg,609umol)溶于无水二氯甲烷(50mL)中,0℃向反应液中滴加甲磺酰氯(34.9mg,305umol)。反应液搅拌2小时。反应完成后,加水(50mL)淬灭。反应液静置分层,水相用二氯甲烷(50mL x 3)萃取,合并的有机相用无水硫酸钠干燥,过滤,减压浓缩。向剩余物加入乙酸乙酯(50mL),搅拌2分钟,有固体生成。将混合物过滤,滤饼用乙酸乙酯(5mL x 3)洗涤,收集滤饼得标题化合物(E)-5-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)戊-4-烯-1-基甲磺酸酯(灰白色产品,82.5mg,crude)。MS m/z(ESI):407.6[M+H]
第三步:(E)-3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000073
将(E)-5-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)戊-4-烯-1-基甲磺酸酯(14.3mg,35.1umol),2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(20.0mg,35.1umol)溶于无水N-甲基吡咯烷酮(2mL)中,室温下向反应液中加入二异丙基乙基胺(13.6mg,105umol)和碘化钠(5.3mg,35.1umol)。加热至70℃反应16小时。反应结束后,过滤,用制备液相色谱纯化得到标题化合物(E)-3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色固体,18.0mg,收率58%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.26(s,1H),8.12(s,1H),7.72-7.65(m,5H),7.62-7.45(m,3H),7.35-7.16(m,1H),6.64(d,J=15.8Hz,1H),6.38(dd,J=14.7,7.9Hz,1H),5.20(dd,J=13.2,5.1Hz,1H),4.70-4.52(m,2H),3.99-3.56(m,16H),3.46-3.35(m,2H),3.01-2.90(m,1H),2.87-2.75(m,1H),2.70-2.53(m,5H),2.44(q,J=7.0Hz,2H),2.20(dd,J=11.9,5.8Hz,1H),2.09(t,J=8.2Hz,2H),1.87(d,J=13.5Hz,6H).MS m/z(ESI):881.2[M+H].
实施例13
3-(4-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000074
第一步:3-(4-(2-氯乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000075
将3-(4-羟基-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(50mg,0.19mmol),1-溴-2-氯乙烷(43mg,0.23mmol),无水碳酸钾(53mg,0.38mmol)和NMP(5mL)一起加入一个50mL的反应单口瓶中,随后缓慢升温至45℃并搅拌2小时。反应结束后,向反应混合物中加入水(20mL),乙酸乙酯萃取(2*30mL),合并有机相,水洗(2*20mL),饱和食盐水(20mL),无水硫酸钠干燥,减压旋干,粗品经柱层析纯化(洗脱剂梯度:二氯甲烷:甲醇=20:1),旋干得标题化合物3-(4-(2-溴乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮和3-(4-(2-氯乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色固体,60mg,收率96%)。MS m/z(ESI):323.4[M+H] +
第二步:3-(4-(2-碘乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000076
将混合物3-(4-(2-溴乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮和3-(4-(2-氯乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(60mg,0.18mmol)加入到一个50mL的反应单口瓶中,随后加入丙酮(20mL)和碘化钠(122mg,0.90mmol),然后升温至回流状态并搅拌16h。将反应混合物减压浓缩,随后加入水(20mL)和二氯甲烷萃取(2*30mL),合并有机相,水洗(20mL),饱和食盐水(20mL),无水硫酸钠干燥,减压旋干得标题化合物3-(4-(2-碘乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(黄色固体,40mg,收率60%),粗品未进行进一步纯化,直接用于下一步反应。
MS m/z(ESI):415.3[M+H] +
第三步:3-(4-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000077
将3-(4-(2-碘乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(10mg,0.02mmol),(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(7.3mg,0.02mmol),碘化钠(3.3mg,0.02mmol),DIPEA(1滴)和无水NMP(2mL)一起加入一个10mL的反应瓶中,随后缓慢加热至80℃并搅拌12h。将反应混合物过滤,滤液经反向制备分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-50%),减压除去乙腈,冻干得到标题化合物3-(4-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)乙氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,3.9mg,收率26%,盐酸盐)。
1H NMR(500MHz,MeOD)δ8.23(s,1H),8.16(s,1H),7.71(dd,J=13.8,7.8Hz,1H),7.63(t,J=7.7Hz,1H),7.54(t,J=7.8Hz,2H),7.49-7.44(m,2H),7.30(d,J=8.1Hz,1H),7.21(s,1H),6.97(d,J=8.7Hz,1H),5.17(dd,J=13.3,5.1Hz,1H),4.67(d,J=17.5Hz,1H),4.63-4.58(m,2H),4.53(d,J=17.6Hz,1H),3.98-3.69(m,16H),3.51-3.37(m,2H),2.97-2.88(m,1H),2.83-2.75(m,1H),2.61-2.52(m,1H),2.46(d,J=11.5Hz,2H),2.33-2.23(m,2H),2.22-2.15(m,1H),1.88(s,3H),1.85(s,3H)。
MS m/z(ESI):856.8[M+H] +.
实施例14
3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000078
第一步:3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮,盐酸盐的制备
Figure PCTCN2022093956-appb-000079
3-(4-((4-溴丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(28.1mg,68.4umol),(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(30mg,52.6umol)溶于无水N-甲基吡咯烷酮(2mL)中,室温下向反应液中加入碘化钠(7.9mg,52.6umol),二异丙基乙基胺(34.0mg,263umol)。反应液升温至70℃,搅拌16小时。反应完成后,反应液降至室温,过滤,滤液用制备液相色谱纯化(盐酸体系,水/乙腈)得标题化合物3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,22.0mg,收率,47%,盐酸盐)
1H NMR(500MHz,CD 3OD)δ8.21(s,1H),8.18(s,1H),7.81-7.63(m,5H),7.58-7.52(m,3H),7.20(d,J=8.7Hz,1H),5.19(dd,J=13.3,5.2Hz,1H),4.60-4.41(m,2H),4.01-3.65(m,18H),3.18-3.12(m,2H),2.92(ddd,J=18.4,13.4,5.3Hz,1H),2.79(d,J=17.8Hz,1H),2.69-2.46(m,5H),2.20(d,J=11.1Hz,1H),2.01(d,J=25.7Hz,2H),1.87(d,J=13.5Hz,6H),1.73(s,2H).MS m/z(ESI):900.7[M+H] +.
实施例15
3-(4-((5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000080
第一步:3-(4-((5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000081
将(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(500mg,0.88mmol),3-(4-((5-溴戊基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(447mg,1.05mmol)溶于无水NMP(5mL)中,室温下向反应液中加入DIEA(227mg,1.76mmol)。反应液升温至80℃,搅拌18小时。反应完成后,反应液降至室温,倒入乙酸乙酯中(100mL),有固体析出,过滤,收集固体。滤液用水(30mL)洗涤,乙酸乙酯(50mL x 5)萃取。合并的有机相用无水硫酸钠干燥,过滤,减压浓缩。剩余物与滤饼合并,用柱层析纯化(二氯甲烷/甲醇=10/1)得淡黄固体(570mg)。该固体用高效液相色谱纯化(盐酸体系,水/乙腈)得到标题化合物3-(4-((5-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)戊基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮。(灰白色固体,460mg,收率:57%,盐酸盐)
1H NMR(500MHz,CD 3OD)δ8.25(s,1H),8.21(s,1H),7.78-7.65(m,4H),7.62-7.55(m,2H),7.53-7.47(m,1H),7.29(s,1H),7.09-7.03(m,1H),5.21(dd,J=5.0,15.0Hz,1H),4.55-4.40(m,2H),4.00(s,2H),3.97(s,3H),3.95-3.60(m,9H),3.53-3.45(m,2H),3.33-3.28(m,2H),3.21-3.06(m,2H),3.03-2.90(m,1H),2.85-2.81(m,1H),2.60-2.57(m,1H),2.46-2.56(m,2H),2.34-2.42(m,2H),2.21-2.26(m,1H),1.92(s,3H),1.89(s,3H),1.89-1.82(m,2H),1.76-1.72(m,2H),1.62-1.58(m,2H).MS m/z(ESI):914.0[M+H] +.
实施例16
(Z)-3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000082
第一步:(Z)-3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000083
将3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(200mg,0.24mmol)加入一个100mL的反应瓶中,随后加入EtOH(30mL),接着依次加入Pd/CaCO 3(50mg)以及喹啉(1滴),然后在氢气球氛围下室温搅拌16h。将反应混合物过滤,正相柱层析(二氯甲烷/甲醇=0~9%)得目标化合物(淡黄色固体,105mg,收率34.8%),然后再反向柱层析纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%~60%)得到标题化合物(Z)-3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2- 基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,84mg,42%,盐酸盐)
1H NMR(500MHz,MeOD)δ8.32(s,1H),8.13(s,1H),7.84(d,J=7.4Hz,1H),7.72(d,J=14.5Hz,1H),7.67-7.54(m,3H),7.45(s,2H),7.10(d,J=11.0Hz,1H),7.02(s,1H),6.84(s,1H),6.15(d,J=11.5Hz,1H),5.22(dd,J=13.5,4.8Hz,1H),4.52(dd,J=46.5,17.5Hz,2H),4.09(s,2H),3.99-3.88(m,5H),3.76-3.42(m,8H),3.25-3.15(m,3H),3.03-2.91(m,1H),2.84(t,J=19.4Hz,1H),2.55(d,J=13.5Hz,1H),2.32(d,J=12.2Hz,2H),2.22(s,1H),2.07(d,J=17.6Hz,2H),1.90(d,J=13.6Hz,6H).
MS m/z(ESI):852.35[M+H] +.
实施例17
3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)磺酰)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000084
第一步:3-(4-((4-溴丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000085
将3-(4-巯基-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(500mg,1.81mmol)加入一个25mL的反应瓶中,依次加入DMF(10mL),1,4-二溴丁烷(783mg,3.62mmol),碳酸钾(750mg,5.43mmol),加完后室温搅拌1h,反应完成后,加入水淬灭,乙酸乙酯(30mL x 2)萃取,有机相用水洗(30mL x 2),饱和食盐水洗(30mL),无水硫酸钠干燥,过滤浓缩后正相柱层析(二氯甲烷/甲醇=0~5%)得到标题化合物3-(4-((4-溴丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡红色固体,450mg,收率60.4%)。MS m/z(ESI):411.23[M+H] +.
第二步:3-(4-((4-溴丁基)磺酰)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000086
将3-(4-((4-溴丁基)硫代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(50mg,0.12mmol)加入一个25mL的反应瓶中,依次加入DCM(10mL),m-CPBA(84mg,0.48mmol),加完后氮气环境下室温搅拌16h,反应完成后,加入饱和硫代硫酸钠淬灭,二氯甲烷(10mL x 2)萃取,有机相用水(10mL x 2),饱和食盐水洗(10mL),无水硫酸钠干燥,过滤浓缩后正相柱层析(二氯甲烷/甲醇=0~5%)得到3-(4-((4-溴丁基)磺酰)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,40mg,收率74.2%)。MS m/z(ESI):443.2[M+H] + .
第三步:3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)磺酰)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000087
参照实例1的方法,制备得到标题化合物3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)磺酰)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,14.0mg,收率17.1%,盐酸盐)。
1H NMR(500MHz,MeOD)δ8.33-8.10(m,4H),7.85(t,J=7.7Hz,1H),7.72(dd,J=14.0,7.7Hz,1H),7.62(t,J=7.8Hz,1H),7.47(dd,J=18.5,11.0Hz,2H),7.16(s,1H),6.94(s,1H),5.20(dd,J=13.3,5.2Hz,1H),4.94-4.86(m,2H),4.01-3.89(m,6H),3.85-3.57(m,8H),3.47-3.34(m,4H),3.27(d,J=8.0Hz,2H),2.98-2.86(m,1H),2.86-2.75(m,1H),2.66-2.54(m,1H),2.42(d,J=10.5Hz,2H),2.23(dd,J=8.9,3.8Hz,3H),1.97(dd,J=13.1,5.3Hz,2H),1.88-1.75(m,8H).MS m/z(ESI):932.84[M+H] +.
实施例18
(Z)-3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000088
第一步:(Z)-3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000089
将3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-炔-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(20mg,0.023mmol)加入一个50mL的反 应瓶中,随后加入EtOH(10mL),接着依次加入Pd/CaCO 3(20mg)以及喹啉(1滴),然后在氢气球氛围下室温搅拌2h。将反应混合物过滤,prep-HPLC纯化分离(洗脱剂(v/v):乙腈/(水+0.05%HCl)=10%-50%)得到标题化合物(Z)-3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,8.0mg,收率:40.0%,盐酸盐)。 1H NMR(500MHz,MeOD)δ8.26(s,1H),8.14(s,1H),7.84-7.56(m,5H),7.47(dd,J=26.6,18.5Hz,2H),7.11(s,1H),6.90(s,1H),6.75(d,J=11.5Hz,1H),5.90(dt,J=11.6,7.2Hz,1H),5.19(dd,J=13.5,5.1Hz,1H),4.47(dd,J=43.7,17.3Hz,2H),4.07-3.32(m,18H),2.96-2.89(m,1H),2.84-2.78(m,3H),2.58-2.50(m,1H),2.37(d,J=11.6Hz,2H),2.25-2.06(m,3H),1.87(d,J=13.6Hz,6H).MS m/z(ESI):866.48[M+H] +.
实施例19
8-(4-(4-(4-((2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)氧代)丁基)哌嗪-1-基)哌啶-1-基)-9-乙基-6,6-二甲基-11-羰基-6,11-二氢-5H-苯并[b]咔唑-3-甲腈
Figure PCTCN2022093956-appb-000090
第一步:8-(4-(4-(4-((2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)氧代)丁基)哌嗪-1-基)哌啶-1-基)-9-乙基-6,6-二甲基-11-羰基-6,11-二氢-5H-苯并[b]咔唑-3-甲腈的制备
Figure PCTCN2022093956-appb-000091
将化合物9-乙基-6,6-二甲基-11-羰基-8-(4-(哌嗪-1-基)哌啶-1-基)-6,11-二氢-5H-苯并[b]咔唑-3-甲腈(25mg,52umol),化合物3-(4-(4-溴丁氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(25mg,63umol)溶于无水N-甲基吡咯烷酮(3mL)中,室温下向反应液中加入二异丙基乙基胺(80uL,480umol),碘化钾(2mg,12umol)。反应液升温至82℃,搅拌1.5小时。反应完成后,反应液降至室温,用高效液相色谱纯化(0.05%HCl,水/乙腈)得标题化合物8-(4-(4-(4-((2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)氧代)丁基)哌嗪-1-基)哌啶-1-基)-9-乙基-6,6-二甲基-11-羰基-6,11-二氢-5H-苯并[b]咔唑-3-甲腈(白色固体,12.9mg,收率31%,盐酸盐)。
1H NMR(500MHz,DMSO-d 6)δ12.86(s,1H),11.36(s,1H),10.99(s,1H),8.32(d,J=5.0Hz,1H),8.07(s,1H),8.01(s,1H),7.61(d,J=5.0Hz,1H),7.51(t,J=7.5Hz,1H),7.37(s,1H),7.34(d,J=5.0Hz,1H),7.27(d,J=10.0Hz,1H),5.13(dd,J=5.0,10.0Hz,1H),4.45(d,J=15.0Hz,1H),4.28(d,J=15.0Hz,1H),4.20-4.18(m,2H),3.86-3.862(m,4H),3.65-3.49(m,4H),3.24-3.35(m,5H),2.84-2.97(m,1H), 2.82(t,J=7.5Hz,2H),2.73(q,J=10.0Hz,2H),2.59-2.64(m,1H),2.48-2.44(m,1H),2.26-2.24(m,2H),2.02-1.94(m,7H),1.86-1.80(m,6H),1.29(t,J=7.5Hz,3H).MS m/z(ESI):796.7[M+H] +.
实施例20
(E)-3-(4-(4-((4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)磺酰)丁-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000092
第一步:(2-((2-((4-(4-(4-(丁-3-烯-1-基磺酰)哌嗪-1-基)哌啶-1-基)-2-甲氧苯基)氨基)-5-氯嘧啶-4-基)氨基)苯基)二甲基膦氧化的制备
Figure PCTCN2022093956-appb-000093
将(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(100mg,0.175mmol)加入一个25mL的反应瓶中,依次加入DCM(11mL),TEA(53mg,0.525mmol),然后滴加丁-3-烯-1-磺酰氯化(41mg,0.265mmol),加完后室温搅拌20min,反应完成后,加入水淬灭,二氯甲烷(20mL x 2)萃取,有机相用无水硫酸钠干燥,浓缩后正相柱层析(二氯甲烷/甲醇=0~6%)得到标题化合物(2-((2-((4-(4-(4-(丁-3-烯-1-基磺酰)哌嗪-1-基)哌啶-1-基)-2-甲氧苯基)氨基)-5-氯嘧啶-4-基)氨基)苯基)二甲基膦氧化(白色固体,85mg,收率70.4%)。
MS m/z(ESI):688.44[M+H] +.
第二步:(E)-3-(4-(4-((4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)磺酰)丁-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000094
将(2-((2-((4-(4-(4-(丁-3-烯-1-基磺酰)哌嗪-1-基)哌啶-1-基)-2-甲氧苯基)氨基)-5-氯嘧啶-4-基)氨基)苯基)二甲基膦氧化(40mg,0.058mmol)加入一个10mL的反应瓶中,随后加入3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(28mg,0.087mmol),溶于dioxane(3mL),接着依次加入Cy 2NMe(34mg,0.174mmol),(t-Bu) 3PBF 4 -(3.4mg,0.012mmol)以及Pd 2(dba) 3(5.3mg,0.006mmol),然后在氮气球氛围下100℃搅拌6h。将反应混合物过滤,prep-HPLC纯化分离(洗脱剂(v/v):乙腈/(水 +0.05%HCl)=10%-50%)得到标题化合物(E)-3-(4-(4-((4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)磺酰)丁-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色固体,20.0mg,收率37.0%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.21(s,2H),7.79-7.46(m,7H),7.35(d,J=29.1Hz,1H),7.07(t,J=15.1Hz,1H),6.70(t,J=17.8Hz,1H),6.50-6.30(m,1H),5.22(dt,J=13.4,4.7Hz,1H),4.60(ddd,J=29.2,22.8,14.6Hz,2H),4.07-3.39(m,16H),3.31-3.12(m,2H),3.02-2.75(m,4H),2.59(ddd,J=26.8,13.5,4.6Hz,1H),2.46(s,2H),2.38-2.18(m,3H),1.90(d,J=13.6Hz,6H).MS m/z(ESI):930.5[M+H] +.
实施例21
3-(4-((5-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)戊基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000095
第一步:3-(4-((5-溴戊基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000096
将化合物3-(4-羟基-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(65mg,0.25mmol)溶于乙腈(5mL),依次加入1,5-二溴戊烷(86mg,0.375mmol),碳酸钾(70mg,0.5mmol),加完后搅拌回流18小时,加入水淬灭,乙酸乙酯萃取,有机相用水洗一遍,饱和食盐水洗一遍,饱和硫酸钠干燥,浓缩后正相柱层析(二氯甲烷/甲醇=0-5%)得标题化合物3-(4-((5-溴戊基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(灰色固体,24mg,收率24%)。MS m/z(ESI):411.1[M+H] +.
第二步:3-(4-((5-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)戊基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000097
将化合物3-(4-((5-溴戊基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(24mg,0.06mmol),化合物(2-((5-氯-2-((2-甲氧基-4-(4-(甲基氨基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(30 mg,0.06mmol)溶于无水N-甲基吡咯烷酮(3mL)中,室温下向反应液中加入二异丙基乙基胺(23mg,0.18mmol),碘化钠(9mg,0.06mmol)。反应液升温至85℃,搅拌16小时。反应完成后,反应液降至室温,用高效液相色谱纯化(0.05%HCl,水/乙腈)得标题化合物3-(4-((5-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)戊基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,3.0mg,收率6%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.22(s,1H),8.01(s,1H),7.64-7.59(m,1H),7.52(t,J=10.0Hz,1H),7.41-7.35(m,3H),7.29(d,J=10.0Hz,1H),7.14(d,J=10.0Hz,1H),6.99-6.97(m,1H),6.79-6.77(m,1H),5.06(dd,J=5.0,10.0Hz,1H),4.40-4.30(m,2H),4.13(d,J=5.0Hz,2H),3.80(s,3H),3.63-3.56(m,1H),3.36-3.31(m,1H),3.32-3.27(m,3H),3.10-3.07(m,2H),2.82(s,3H),2.66-2.64(m,1H),2.47-2.39(m,1H),2.20-2.04(m,5H),1.86-1.83(m,2H),1.79(s,3H),1.77(s,3H),1.58-1.53(m,2H),1.48-1.44(m,1H)。
MS m/z(ESI):843.6[M+H] +.
实施例22
3-(4-(4-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)丁氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000098
第一步:-(4-(4-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)丁氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000099
将(2-((5-氯-2-((2-甲氧基-4-(4-(甲基氨基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(30mg,58.25umol),3-(4-(4-溴丁氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(28mg,69.90umol)溶于无水NMP(2mL)中,室温下向反应液中加入DIEA(23mg,174.76umol),碘化钠(8mg,58.25umol)。反应液升温至90℃,搅拌18小时。反应完成后,反应液降至室温,过滤,滤液用高效液相色谱纯化(盐酸体系,水/乙腈)得到标题化合物3-(4-(4-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)丁氧基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,10mg,收率:21%,盐酸盐)
1H NMR(500MHz,CD 3OD)δ8.22(s,1H),8.01(s,1H),7.62-7.58(m,1H),7.53-7.49(m,1H),7.41(t,J=10.0Hz,1H),7.33-7.29(m,3H),7.15(d,J=10.0Hz,1H),6.92-6.87(m,1H),6.72-6.67(m,1H),5.06(dd,J=5.0,10.0Hz,1H),4.45-4.33(m,2H),4.16(d,J=5.0Hz,2H),3.85-3.78(m,5H),3.62-3.49(m, 1H),3.36-3.31(m,1H),3.17-3.05(m,4H),2.82(s,3H),2.80-2.76(m,1H),2.67-2.62(m,1H),2.44-2.38(m,1H),2.21-2.15(m,2H),2.08-1.88(m,6H),1.80(s,3H),1.77(s,3H).MS m/z(ESI):829.0[M+H] +.
实施例23
3-(4-(6-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000100
第一步:3-(4-(6-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000101
将化合物6-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)己基甲磺酸酯(21mg,0.05mmol),化合物(2-((5-氯-2-((2-甲氧基-4-(4-(甲基氨基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(25mg,0.05mmol)溶于无水N-甲基吡咯烷酮(3mL)中,室温下向反应液中加入二异丙基乙基胺(20mg,0.15mmol),碘化钠(7.5mg,0.05mmol)。反应液升温至85℃,搅拌3小时。反应完成后,反应液降至室温,用高效液相色谱纯化(0.05%HCl,水/乙腈)得标题化合物3-(4-(6-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)己基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,4.0mg,收率9.5%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.26(s,1H),8.17(s,1H),7.76(dd,J=5.0,10.0Hz,1H),7.72-7.66(m,3H),7.54-7.48(m,4H),7.24(d,J=10.0Hz,1H),5.06(dd,J=5.0,15.0Hz,1H),4.60-4.50(m,2H),4.00(s,3H),3.96-3.92(m,1H),3.88-3.83(m,4H),3.37-3.32(m,1H),3.20-3.15(m,1H),2.94(s,3H),2.83-2.82(m,3H),2.79-2.76(m,2H),2.64-2.49(m,5H),2.25-2.21(m,1H),1.91(s,3H),1.88(s,3H),1.85-1.80(m,1H),1.74-1.72(m,2H),1.53-1.50(m,4H).MS m/z(ESI):842.1[M+H] +.
实施例24
(E)-3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000102
第一步:(E)-3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-烯-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000103
将化合物3-(4-溴-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(160mg,0.5mmol)溶解于DMF(3mL),在室温氮气保护下依次加入2-丙烯氧基吡喃(284mg,2.0mmol),三乙胺(150mg,1.5mmol),三(邻甲基苯基)磷(15mg,0.05mmol)和醋酸钯(5.6mg,0.025mmol),加热至100℃,氮气下搅拌16小时。反应完毕冷却室温。加入乙酸乙酯和水分层,有机层用食盐水洗涤,减压浓缩后用正相色谱柱纯化(二氯甲烷:甲醇=20:1)得标题化合物(E)-3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-烯-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,56mg,收率30%)。MS m/z(ESI):385.5[M+H] +.
第二步:(E)-3-(4-(3-羟基丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000104
将化合物(E)-3-(1-羰基-4-(3-((四氢-2H-吡喃-2-基)氧代)丙-1-烯-1-基)异二氢吲哚-2-基)哌啶-2,6-二酮溶于甲醇(3mL),加入对甲苯磺酸。室温搅拌4小时。抽滤,滤液用反相高效液相色谱柱分离(0.05%HCl,水/乙腈)标题化合物(E)-3-(4-(3-羟基丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,12mg,收率26.5%)。
MS m/z:301.4[M+H] +.
第三步:(E)-3-(4-(3-溴丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000105
将化合物(E)-3-(4-(3-羟基丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(12mg,0.04mmol)溶解于DCM(3mL),加入三乙胺(12mg,0.12mmol),然后加入三溴化磷(9mg,0.08mmol),加完 后室温搅拌3小时。溶剂旋干,加入乙酸乙酯和饱和碳酸氢钠后分层,有机层用无水硫酸钠干燥,过滤旋干得标题化合物(E)-3-(4-(3-溴丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(黄色油状物,18mg,crude),直接用于下一步。MS m/z(ESI):365.2[M+H] +
第四步:(E)-3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000106
将化合物(E)-3-(4-(3-溴丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(18mg,0.05mmol),化合物(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(27mg,0.05mmol)溶于无水N-甲基吡咯烷酮(3mL)中,室温下向反应液中加入二异丙基乙基胺(20mg,0.15mmol),碘化钠(7.5mg,0.05mmol)。反应液升温至85℃,搅拌3小时。反应完成后,反应液降至室温z,用高效液相色谱纯化(0.05%HCl,水/乙腈)标题化合物(E)-3-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丙-1-烯-1-基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固,4.0mg,收率9.4%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.22-8.25(m,1H),7.93(s,1H),7.65(dd,J=5.0,15.0Hz,2H),7.56(d,J=10.0Hz,1H),7.53-7.45(m,1H),7.44-7.40(m,2H),7.19-7.15(m,1H),6.64(d,J=10.0Hz,1H),6.57(s,1H),6.36(d,J=10.0Hz,1H),6.32-6.25(m,1H),5.09(dd,J=5.0,15.0Hz,1H),4.53-4.44(m,5H),3.75(s,3H),3.63-3.60(m,1H),2.50-2.72(m,8H),2.14-2.05(m,1H),1.97-1.89(m,2H),1.75(s,3H),1.72(s,3H),1.67-1.55(m,2H),1.25-1.19(m,4H)。MS m/z(ESI):852.5[M+H] +.
实施例25
(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000107
第一步:(E)-3-(4-((4-溴丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000108
将(E)-3-(4-((4-溴丁-2-烯-1-基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(100mg,0.25mmol),乙酸(1滴)溶于乙腈(8mL)中,随后加入甲醛水溶液(0.5mL),加完反应液变澄清。反应液在室温下搅拌18小时后,加入氰基硼氢化钠(80mg,1.25mmol)。反应继续搅拌1小时。反应液用水(10mL)洗涤,二氯甲烷(20mL x 3)萃取。合并的有机相用无水硫酸钠干燥,过滤,减压浓缩。剩余物用柱层析纯化(二氯甲烷/甲醇=10/1)得到标题化合物(E)-3-(4-((4-溴丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(淡黄色油状物,60mg,收率:60%)
1H NMR(500MHz,DMSO-d 6)δ10.96(s,1H),7.36(t,J=5.0Hz,1H),7.16(d,J=5.0Hz,1H),6.99(d,J=5.0Hz,1H),6.05-6.02(m,3H),5.12-5.06(m,1H),4.51-4.33(m,2H),4.15-4.12(m,1H),3.84-3.82(m,1H),3.27(s,3H),2.94-2.89(m,1H),2.63-2.60(m,1H),2.58-2.54(m,1H),2.36-2.33(m,1H),2.05-1.95(m,1H).MS m/z(ESI):406[M+H] +.
第二步:(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000109
将(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(100mg,0.17mmol)和(E)-3-(4-((4-溴丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(85mg,0,21mmol),溶于无水NMP(3mL)中,向反应液中加入DIEA(43mg,0.34mmol)。反应液在室温下搅拌18小时。反应完成后,反应液用高效液相色谱纯化(盐酸体系,水/乙腈)得到标题化合物(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,65mg,收率:43%,盐酸盐)
1H NMR(500MHz,Methanol-d 4)δ8.51-8.48(m,1H),8.20(s,1H),7.83(d,J=10.0Hz,1H),7.77(dd,J=5.0,10.0Hz,1H),7.68(t,J=10.0Hz,1H),7.59(t,J=10.0Hz,1H),7.49(d,J=5.0Hz,1H),7.46-7.40(m,1H),7.30(d,J=10.0Hz,1H),6.83(d,J=2.5Hz,1H),6.62(dd,J=2.5,10.0Hz,1H),5.96-5.90(m,1H),5.85-5.79(m,1H),5.31(dd,J=5.0,15.0Hz,1H),4.75-4.65(m,3H),4.06-3.97(m,5H),3.90-3.85(m,2H),3.31-3.21(m,2H),3.13(s,3H),3.09-3.04(m,1H),2.94-2.81(m,6H),2.75-2.57(m,6H),2.36-2.33(m,1H),2.18-2.11(m,2H),2.02(s,3H),1.99(s,3H),1.88-1.77(m,2H).MS m/z(ESI):895.0[M+H] +.
实施例26
3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000110
第一步:3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000111
将(E)-3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁-2-烯-1-基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(50mg,56umol)溶于乙醇(10mL)中,室温下向反应液中加入林德拉钯催化剂(20mg),反应体系用氢气置换三次。反应液在氢气球体系下搅拌4小时。反应完成后,反应液过滤,滤液减压浓缩,剩余物用高效液相色谱纯化(盐酸体系,水/乙腈)得到标题化合物3-(4-((4-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)丁基)(甲基)氨基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,2mg,收率:4%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.22-8.25(m,1H),7.93(s,1H),7.56(d,J=5.0Hz,1H),7.50(dd,J=5.0,15.0Hz,1H),7.42(t,J=10.0Hz,1H),7.32(t,J=10.0Hz,1H),7.23(d,J=10.0Hz,1H),7.11-7.18(m,1H),7.03(d,J=10.0Hz,1H),6.57(d,J=2.5Hz,1H),6.36(dd,J=2.5,10.0Hz,1H),5.06(dd,J=5.0,15.0Hz,1H),4.49-4.46(m,2H),4.43(d,J=10.0Hz,2H),3.75(s,3H),3.62-3.57(m,2H),3.21-3.15(m,3H),2.84(s,3H),2.82-2.78(m,1H),2.71-2.65(m,1H),2.62-2.55(m,5H),2.47-2.39(m,4H),2.29-2.25(m,3H),2.11-2.05(m,1H),1.94-1.88(m,2H),1.75(s,3H),1.73(s,3H),1.57-1.43(m,5H)。
MS m/z(ESI):897.0[M+H] +.
实施例27
3-(4-(7-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000112
第一步:3-(4-(7-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000113
将(2-((5-氯-2-((2-甲氧基-4-(4-(甲基氨基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(10mg,19.4umol)和7-(2-(2,6-二羰基哌啶-3-基)-1-羰基异二氢吲哚-4-基)庚基甲磺酸酯(8.5mg,19.4umol),溶于无水NMP(2mL)中,室温下向反应液中加入碘化钠(2.9mg,19.4umol),DIEA(7.5mg,58.2umol)。反应液升温至70℃,搅拌16小时。反应完成后,反应液降至室温,过滤,滤液用制备液相色谱纯化(盐酸体系,水/乙腈)得到标题化合物3-(4-(7-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)庚基)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(黄色固体,7.0mg,收率:42%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ8.25-8.00(m,2H),7.62(ddd,J=13.9,7.7,1.5Hz,1H),7.54(dt,J=8.6,4.4Hz,2H),7.48-7.33(m,4H),7.13(s,1H),6.90(d,J=8.6Hz,1H),5.12-5.06(m,1H),4.41(q,J=16.9Hz,2H),3.84-3.75(m,5H),3.67(d,J=17.6Hz,1H),3.39(s,2H),3.03(td,J=12.9,12.1,5.7Hz,1H),2.87-2.76(m,4H),2.72-2.61(m,3H),2.46(dtd,J=17.5,13.4,4.0Hz,1H),2.27-2.06(m,5H),1.78(d,J=13.5Hz,6H),1.74-1.56(m,5H),1.34(d,J=4.5Hz,6H).MS m/z(ESI):855.3[M+H] +.
实施例28
3-(4-((6-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)己基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮
Figure PCTCN2022093956-appb-000114
第一步:3-(4-((6-溴己基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000115
将3-(4-羟基-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(130mg,0.5mmol)溶于乙腈(20mL),加入1,6-二溴己烷(183mg,0.75mmol)和碳酸钾(152mg,1.1mmol),加热至80℃,反应16h。将反应混合物浓缩然后柱层析纯化(乙酸乙酯:石油醚=2:3)得标题化合物3-(4-((6-溴己基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,50mg,收率,23.6%)。MS m/z(ESI):425.1[M+H] +.
第二步:3-(4-((6-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)己基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮的制备
Figure PCTCN2022093956-appb-000116
将化合物3-(4-((6-溴己基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(35mg,0.083mmol)和化合物(2-((5-氯-2-((2-甲氧基-4-(4-(甲基氨基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(42mg,0.083mmol)溶于无水N-甲基吡咯烷酮(3mL)中,室温下向反应液中加入二异丙基乙基胺(27uL,174.76mmol),升温至90℃,搅拌18hrs。反应完成后,反应液降至室温,过滤,滤液用反相高效液相色谱纯化(0.05%HCl,水/乙腈)得标题化合物3-(4-((6-((1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)(甲基)氨基)己基)氧代)-1-羰基异二氢吲哚-2-基)哌啶-2,6-二酮(白色固体,8.5mg,收率,12%,盐酸盐)。 1H NMR(500MHz,CD 3OD)δ8.16(s,1H),8.01(brs,1H),7.69-7.59(m,3H),7.53(s,1H),7.44-7.37(m,2H),7.27(d,J=5.0Hz,1H),7.21(d,J=5.0Hz,1H),7.12(d,J=10.0Hz,1H),5.06(dd,J=5.0,10.0Hz,1H),4.40-4.30(m,2H),4.08(d,J=7.5Hz,2H),3.95-3.86(m,6H),3.75-3.73(m,2H),3.34-3.32(m,1H),3.12-3.06(m,1H),2.84(s,3H),2.82-2.16(m,1H),2.70-2.66(m,1H),2.53-2.37(m,5H),2.11-2.04(m,1H),1.82-1.80(m,4H),1.79(s,3H),1.76(s,3H),1.55-1.50(m,2H),1.45-1.42(m,2H).MS m/z(ESI):857.3[M+H] +.
实施例29
(2S,4R)-1-((S)-2-(11-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)十一烷酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺
Figure PCTCN2022093956-appb-000117
第一步:(2S,4R)-1-((S)-2-(11-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)十一烷酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺的制备
Figure PCTCN2022093956-appb-000118
将化合物(2S,4R)-1-((S)-2-(11-溴十一烷酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺(24mg,0.035mmol),化合物(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基膦氧化(20mg,0.032mmol)溶于无水N-甲基吡咯烷酮(3mL)中,室温下向反应液中加入二异丙基乙基胺(13mg,0.105mmol)。反应液升温至80℃,搅拌18小时。反应完成后,反应液过滤,滤液用反相高效液相色谱纯化(0.05%HCl,水/乙腈)得标题化合物2S,4R)-1-((S)-2-(11-(4-(1-(4-((5-氯-4-((2-(二甲基磷基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧苯基)哌啶-4-基)哌嗪-1-基)十一烷酰氨基)-3,3-二甲基丁酰)-4-羟基-N-(4-(4-甲硫基唑-5-基)苯甲基)吡咯烷-2-甲酰胺(白色固体,12mg,收率32%,盐酸盐)。
1H NMR(500MHz,CD 3OD)δ10.07(s,1H),8.30(s,1H),8.13(s,1H),7.81-7.72(m,3H),7.66(s,1H),7.61-7.55(m,3H),7.33(d,J=5.0Hz,1H),4.65(s,1H),4.60-4.41(m,4H),4.02-3.82(m,18H),2.68-2.65(m,6H),2.37-2.24(m,3H),2.12-2.08(m,1H),1.91(s,3H),1.88(s,3H),1.64-1.63(m,2H),1.44-1.36(m,4H),1.04(s,9H)。MS m/z(ESI):1166.4[M+H] +.
以下结合测试例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。
生物学测试
实验细胞:
Figure PCTCN2022093956-appb-000119
Figure PCTCN2022093956-appb-000120
化合物半数抑制浓度(IC50)测定:
本发明化合物IC50采用Promega公司的CellTiter GLO试剂来进行测定。步骤如下:
将肿瘤细胞系在指定培养基中在37℃,5%CO 2的培养箱中进行培养。定期传代,取处于对数生长期的细胞用于铺板,细胞密度为2000-4000个细胞/每孔。第二天,将本发明化合物进行系列稀释后加入细胞中,阴性对照为DMSO,空白对照为不含细胞的培养液。将培养细胞的微孔板放回培养箱中继续培养72小时;按照CellTiter GLO试剂说明书将检测试剂加入细胞培养液中,在EnVision酶标仪(PerkinElmer)上检测发光信号,进行细胞活性测定。
本发明化合物对细胞的生长抑制通过Prism Graphpad软件进行绘制并统计本发明化合物IC50。
具体数据如下表所示:
Figure PCTCN2022093956-appb-000121
Figure PCTCN2022093956-appb-000122
实验结论:
通过以上数据得出,本发明所示的实施例化合物在BaF3(EGFR Del19/T790M/C797S)、BaF3(EGFR L858R/T790M/C797S)和BaF3(CD74-ROS1-G2032R)突变细胞增殖活性的抑制试验中具有良好的抑制作用。
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。

Claims (44)

  1. 通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100001
    其中,
    K选自以下结构:
    Figure PCTCN2022093956-appb-100002
    E选自以下结构:
    Figure PCTCN2022093956-appb-100003
    L 1为化学键、C(R) 2或SO 2
    L 2选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
    L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
    L 4选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5选自化学键、O、S、NR*、C(R) 2或SO 2
    或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
    或者L 4和L 5一起形成顺式或反式的-CR=CR-,或-C≡C-;
    其中Z为C=O或C(R”) 2
    R 1选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-7环烷基或3至7元杂环基;
    R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    条件是,仅当K为K1、K2或K4时,L 4和L 5一起形成-C≡C-。
  2. 权利要求1的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 1为化学键、CH 2或SO 2;优选为C(R) 2;优选为L 1为CH 2
  3. 权利要求1或2的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 2选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;优选为化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;优选为化学键、C(R’) 2或C(R’) 2C(R’) 2;优选为化学键、O、S、NR*、CH 2、CH 2CH 2、1,3-亚苯基、1,4-亚苯基、1,4-亚三唑基、3,5-亚吡啶基或2,4-亚嘧啶基;优选为化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;优选为化学键、CH 2、CH 2CH 2或1,3-亚苯基;优选为化学键、CH 2或CH 2CH 2;优选为化学键或CH 2
  4. 权利要求1-3中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;优选为化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;优选为化学键、C(R’) 2或C(R’) 2C(R’) 2;优选为化学键、O、S、NR*、CH 2、CH 2CH 2或1,4-亚三唑基;优选为化学键、O、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;优选为化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;优选为化学键、CH 2或CH 2CH 2;优选为化学键或CH 2
  5. 权利要求1-4中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 4选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2;优选为化学键、C(R’) 2或C(R’) 2C(R’) 2;优选为化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2;优选为化学键、CH 2或CH 2CH 2;优选为化学键或CH 2
  6. 权利要求1-5中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 5选自O、S、NR*、C(R) 2或SO 2;优选为化学键、O、S、NR*或C(R) 2;优选为O、S、NR*或C(R) 2;优选为O、S或NR*;优选为O或C(R) 2;优选为C(R) 2;优选为化学键、O、S、NR*、CH 2或SO 2;优选为O、S、NH、CH 2或SO 2;优选为O、S、NH或CH 2;优选为O、S或NH;优选为O或CH 2;优选为O或S;优选为CH 2;优选为O;优选为S。
  7. 权利要求1-6中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 2和L 3一起形成反式的-CR=CR-,或-C≡C-;优选地,L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-。
  8. 权利要求1-7中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,L 4和L 5一起形成顺式或反式的-CR=CR-;优选地,L 4和L 5一起形成顺式的-CR=CR-;优选地,L 4和L 5一起形成顺式或反式的-CH=CH-,或-C≡C-;优选地,L 4和L 5一起形成顺式或反式的-CH=CH-;优选地,L 4和L 5一起形成顺式的-CH=CH-。
  9. 权利要求1-8中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,Z为C=O或CH 2;优选CH 2
  10. 权利要求1-9中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R 1选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基或3至7元杂环基。
  11. 权利要求1-10中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基。
  12. 权利要求1-11中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R’选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基。
  13. 权利要求1-12中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R”选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基。
  14. 通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100004
    其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2或C(R’) 2C(R’) 2;或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;或者L 4和L 5一起形成顺式或反式的-CR=CR-;
    L 5为化学键、O、S、NR*或C(R) 2
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  15. 权利要求14的通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键或CH 2;或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;或者L 4和L 5一起形成顺式或反式的-CH=CH-;
    L 5为O、S、NH或CH 2
    Z为C=O或CH 2
  16. 权利要求14的通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键或C(R’) 2;或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
    L 5为O、S或NR*;
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  17. 权利要求16的通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键或CH 2;或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;
    L 5为O、S或NH;
    Z为C=O或CH 2
  18. 权利要求14的通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键或C(R’) 2;或者L 2和L 3一起形成反式的-CR=CR-,或-C≡C-;
    L 5为O或S;
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基;
    R’选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基;
    R”选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基。
  19. 权利要求18的通式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键或CH 2
    L 5为O;
    Z为C=O或CH 2
  20. 通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100005
    其中,
    L 1为化学键、C(R) 2或SO 2
    L 2选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、 C 6-10亚芳基或5至10元亚杂芳基;
    L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
    L 4选自化学键、C(R’) 2或C(R’) 2C(R’) 2
    L 5选自化学键、O、S、NR*、C(R) 2或SO 2
    或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
    或者L 4和L 5一起形成顺式或反式的-CR=CR-;
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  21. 权利要求20的通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为化学键、CH 2或SO 2
    L 2选自化学键、O、S、NR*、CH 2、CH 2CH 2、1,3-亚苯基、1,4-亚苯基、1,4-亚三唑基、3,5-亚吡啶基或2,4-亚嘧啶基;
    L 3选自化学键、O、S、NR*、CH 2、CH 2CH 2或1,4-亚三唑基;
    L 4选自化学键、CH 2或CH 2CH 2
    L 5选自化学键、O、S、NR*、CH 2或SO 2
    或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;
    或者L 4和L 5一起形成顺式或反式的-CH=CH-;
    Z为C=O或CH 2
    其中,R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  22. 权利要求20的通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5为S;
    或者L 4和L 5一起形成顺式的-CR=CR-;
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  23. 权利要求22的通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键、CH 2或CH 2CH 2
    L 5为S;
    或者L 4和L 5一起形成顺式的-CH=CH-;
    Z为C=O或CH 2
  24. 权利要求20的通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5为S;
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  25. 权利要求24的通式(III)或(III-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键、CH 2或CH 2CH 2
    L 5为S;
    Z为C=O或CH 2
  26. 通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100006
    其中,
    L 1为C(R) 2
    L 2选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2C(R’) 2、C 6-10亚芳基或5至10元亚杂芳基;
    L 3和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5为O、S、NR*、C(R) 2或SO 2
    或者L 2和L 3一起形成顺式或反式的-CR=CR-,或-C≡C-;
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  27. 权利要求26的通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2选自化学键、CH 2、CH 2CH 2或1,3-亚苯基;
    L 3和L 4各自独立地选自化学键、CH 2或CH 2CH 2
    L 5为O、S、NH、CH 2或SO 2
    或者L 2和L 3一起形成顺式或反式的-CH=CH-,或-C≡C-;
    Z为C=O或CH 2
    其中,R*选自H、C 1-6烷基或C 1-6卤代烷基。
  28. 权利要求26的通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5为O或C(R) 2
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  29. 权利要求28的通式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键、CH 2或CH 2CH 2
    L 5为O或CH 2
    Z为C=O或CH 2
    其中,R*选自H、C 1-6烷基或C 1-6卤代烷基。
  30. 通式(V)、(V-1)或(V-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100007
    其中,
    L 1为C(R) 2
    L 5为O、S、NR*或C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  31. 权利要求30的通式(V)、(V-1)或(V-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5为C(R) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基或C 1-6卤代烷基;
    R’选自H、D、卤素、-CN、C 1-6烷基或C 1-6卤代烷基。
  32. 权利要求30的通式(V)、(V-1)或(V-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2
    L 5为CH 2
  33. 通式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100008
    其中,
    L 1为C(R) 2
    L 2、L 3和L 4各自独立地选自化学键、C(R’) 2或C(R’) 2C(R’) 2
    L 5为C(R) 2
    或者L 4和L 5一起形成顺式或反式的-CR=CR-,或-C≡C-;
    Z为C=O或C(R”) 2
    R 1选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-7环烷基或3至7元杂环基;
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  34. 权利要求33的通式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2、L 3和L 4各自独立地选自化学键或CH 2
    L 5为CH 2
    或者L 4和L 5一起形成顺式或反式的-CH=CH-,或-C≡C-;
    Z为C=O或CH 2
    R 1选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基或3至7元杂环基。
  35. 通式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2022093956-appb-100009
    其中,
    L 1为C(R) 2
    L 2和L 4各自独立地选自化学键、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 3选自化学键、O、S、NR*、C(R’) 2、C(R’) 2C(R’) 2、C(R’) 2C(R’) 2C(R’) 2或C(R’) 2C(R’) 2C(R’) 2C(R’) 2
    L 5为C(R) 2
    Z为C=O或C(R”) 2
    其中,R选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R’选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R”选自H、D、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R*选自H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基。
  36. 权利要求35的通式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    L 1为CH 2
    L 2和L 4各自独立地选自化学键、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2
    L 3选自化学键、O、CH 2、CH 2CH 2、CH 2CH 2CH 2或CH 2CH 2CH 2CH 2
    L 5为CH 2
    Z为C=O或CH 2
  37. 权利要求1的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,所述化合物选自以下:
    Figure PCTCN2022093956-appb-100010
    Figure PCTCN2022093956-appb-100011
    Figure PCTCN2022093956-appb-100012
    Figure PCTCN2022093956-appb-100013
    Figure PCTCN2022093956-appb-100014
    Figure PCTCN2022093956-appb-100015
  38. 药物组合物,其含有权利要求1-37中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
  39. 权利要求1-37中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体在制备用于治疗和/或预防癌症的药物中的用途。
  40. 一种在受试者中治疗和/或预防癌症的方法,所述方法包括向所述受试者给药权利要求1-37中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求38的药物组合物。
  41. 权利要求1-37中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求38的药物组合物,其用于治疗和/或预防癌症。
  42. 权利要求39的用途或权利要求40的方法或权利要求41的化合物或组合物的用途,其中所述癌症选自肺癌;淋巴瘤;炎症性肌纤维母细胞瘤;结直肠癌;脑胶质瘤;星形胶质母细胞瘤;卵巢癌;骨髓癌症;移植相关的癌症;中性粒细胞减少症;白血病;翁韦里希特综合症;支气管癌;前列腺癌;乳腺癌;甲状腺癌;胰腺癌;神经母细胞瘤;髓外浆细胞瘤;浆细胞瘤;胃癌;胃肠道间质瘤; 食道癌;大肠腺癌;食管鳞状细胞癌;肝癌;肾细胞癌;膀胱癌;子宫内膜癌;黑色素瘤;脑癌;口腔癌;肉瘤;对靶向药物耐药的肿瘤;或依赖ALK、ROS1或EGFR或其任意突变蛋白的肿瘤或疾病。
  43. 权利要求39的用途或权利要求40的方法或权利要求41的化合物或组合物的用途,其中所述癌症选自小细胞肺癌;非小细胞肺癌;弥漫性大B细胞淋巴瘤;非霍奇金淋巴瘤;间变性淋巴瘤;间变性大细胞淋巴瘤;CD20阳性淋巴瘤;原发性淋巴瘤;B细胞淋巴瘤;复发性B细胞非霍奇金淋巴瘤;复发性弥漫性大B细胞淋巴瘤;复发性纵隔(胸腺)大B细胞淋巴瘤;原发性纵隔(胸腺)大B细胞淋巴瘤;复发性转化非霍奇金淋巴瘤;难治性B细胞非霍奇金淋巴瘤;难治性弥漫性大B细胞淋巴瘤;难治性原发性纵隔(胸腺)大B细胞淋巴瘤;难治性转化的非霍奇金淋巴瘤;多发性骨髓瘤;骨髓增生异常综合征(MDS);既往治疗的骨髓增生异常综合征;浆细胞骨髓瘤;阴燃骨髓瘤;闷烧多发性骨髓瘤;骨髓纤维化;急性髓细胞白血病(AML);与白血病相关的贫血;慢性粒细胞白血病;B细胞慢性淋巴细胞白血病;翁韦里希特综合症;支气管癌;前列腺癌;三阴性乳腺癌;偶发性乳腺癌;考登病患者;甲状腺癌;胰腺癌;神经母细胞瘤;髓外浆细胞瘤;浆细胞瘤;胃癌;胃肠道间质瘤;食道癌;大肠腺癌;食管鳞状细胞癌;肝癌;肾细胞癌;膀胱癌;子宫内膜癌;黑色素瘤;脑癌;口腔癌;横纹肌肉瘤;各种脂肪源性肿瘤;尤文肉瘤/原始神经外胚层瘤(Ewing/PNETs);平滑肌肉瘤;对EGFR、ROS1或ALK靶向药物耐药的肿瘤。
  44. 权利要求39的用途或权利要求40的方法或权利要求41的化合物或组合物的用途,其中所述癌症选自间变性淋巴瘤激酶(ALK)突变阳性的非小细胞肺癌(NSCLC);ROS1阳性非小细胞肺癌;EGFR突变的非小细胞肺癌;肺腺癌;对EGFR、ROS1或ALK靶向药物耐药的肺癌;对ALK靶向药物耐药的淋巴瘤;或依赖选自ALK、ROS1或EGFR或其任意突变蛋白的以下肿瘤、癌症或疾病:肺癌、淋巴瘤、炎症性肌纤维母细胞瘤、结直肠癌、脑胶质瘤、星形胶质母细胞瘤、卵巢癌、白血病、乳腺癌、甲状腺癌、神经母细胞瘤、髓外浆细胞瘤、浆细胞瘤、食管鳞状细胞癌、肾细胞癌、支气管癌、前列腺癌、乳腺癌、甲状腺癌、胰腺癌、神经母细胞瘤、髓外浆细胞瘤、浆细胞瘤、胃癌、胃肠道间质瘤、食道癌、大肠腺癌、食管鳞状细胞癌、肝癌、肾细胞癌、膀胱癌、子宫内膜癌、黑色素瘤、脑癌、口腔癌或肉瘤。
PCT/CN2022/093956 2021-05-19 2022-05-19 一类新型蛋白降解剂及其应用 WO2022242725A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280016739.4A CN116940581A (zh) 2021-05-19 2022-05-19 一类新型蛋白降解剂及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110546454.9 2021-05-19
CN202110546454 2021-05-19

Publications (1)

Publication Number Publication Date
WO2022242725A1 true WO2022242725A1 (zh) 2022-11-24

Family

ID=84140833

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/093956 WO2022242725A1 (zh) 2021-05-19 2022-05-19 一类新型蛋白降解剂及其应用

Country Status (2)

Country Link
CN (1) CN116940581A (zh)
WO (1) WO2022242725A1 (zh)

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017204445A2 (ko) * 2016-05-24 2017-11-30 한국화학연구원 Alk 단백질의 분해를 유도하는 약학적 조성물 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물
CN109422733A (zh) * 2017-09-03 2019-03-05 上海美志医药科技有限公司 一类抑制并降解酪氨酸蛋白激酶alk的化合物
WO2019113071A1 (en) * 2017-12-05 2019-06-13 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating alk-mediated cancer
CN109912655A (zh) * 2017-12-13 2019-06-21 上海科技大学 Alk蛋白降解剂及其抗肿瘤应用
CN110357889A (zh) * 2018-04-09 2019-10-22 上海科技大学 蛋白降解靶向化合物、其抗肿瘤应用、其中间体及中间体应用
CN110684015A (zh) * 2018-07-06 2020-01-14 四川大学 靶向alk的protac及其应用
WO2020069106A1 (en) * 2018-09-27 2020-04-02 Dana-Farber Cancer Institute, Inc. Degraders that target alk and therapeutic uses thereof
CN112079866A (zh) * 2019-06-12 2020-12-15 上海科技大学 Alk蛋白调节剂及其抗肿瘤应用
CN112341436A (zh) * 2020-11-20 2021-02-09 中国药科大学 基于靶向抑制和降解alk的苯并咔唑类蛋白水解靶向嵌合分子、制备方法及用途
KR20210016103A (ko) * 2019-07-31 2021-02-15 주식회사 온코빅스 Alk 단백질의 분해를 유도하는 신규 화합물 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학 조성물
WO2021036922A1 (zh) * 2019-08-23 2021-03-04 北京泰德制药股份有限公司 抑制并诱导降解egfr和alk的化合物
WO2021173677A1 (en) * 2020-02-25 2021-09-02 Dana-Farber Cancer Institute, Inc. Potent and selective degraders of alk
CN114262321A (zh) * 2021-12-07 2022-04-01 中国药科大学 一种双稳态光调控小分子蛋白降解剂、制备方法及应用

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017204445A2 (ko) * 2016-05-24 2017-11-30 한국화학연구원 Alk 단백질의 분해를 유도하는 약학적 조성물 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물
CN109422733A (zh) * 2017-09-03 2019-03-05 上海美志医药科技有限公司 一类抑制并降解酪氨酸蛋白激酶alk的化合物
WO2019113071A1 (en) * 2017-12-05 2019-06-13 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating alk-mediated cancer
CN109912655A (zh) * 2017-12-13 2019-06-21 上海科技大学 Alk蛋白降解剂及其抗肿瘤应用
CN110357889A (zh) * 2018-04-09 2019-10-22 上海科技大学 蛋白降解靶向化合物、其抗肿瘤应用、其中间体及中间体应用
CN110684015A (zh) * 2018-07-06 2020-01-14 四川大学 靶向alk的protac及其应用
WO2020069106A1 (en) * 2018-09-27 2020-04-02 Dana-Farber Cancer Institute, Inc. Degraders that target alk and therapeutic uses thereof
CN112079866A (zh) * 2019-06-12 2020-12-15 上海科技大学 Alk蛋白调节剂及其抗肿瘤应用
KR20210016103A (ko) * 2019-07-31 2021-02-15 주식회사 온코빅스 Alk 단백질의 분해를 유도하는 신규 화합물 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학 조성물
WO2021036922A1 (zh) * 2019-08-23 2021-03-04 北京泰德制药股份有限公司 抑制并诱导降解egfr和alk的化合物
WO2021173677A1 (en) * 2020-02-25 2021-09-02 Dana-Farber Cancer Institute, Inc. Potent and selective degraders of alk
CN112341436A (zh) * 2020-11-20 2021-02-09 中国药科大学 基于靶向抑制和降解alk的苯并咔唑类蛋白水解靶向嵌合分子、制备方法及用途
CN114262321A (zh) * 2021-12-07 2022-04-01 中国药科大学 一种双稳态光调控小分子蛋白降解剂、制备方法及应用

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CHELSEA E. POWELL, YANG GAO, LI TAN, KATHERINE A. DONOVAN, RADOSłAW P. NOWAK, AMANDA LOEHR, MAGDA BAHCALL, ERIC S. FISCHER, P: "Chemically Induced Degradation of Anaplastic Lymphoma Kinase (ALK)", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 61, no. 9, 10 May 2018 (2018-05-10), US , pages 4249 - 4255, XP055628330, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.7b01655 *
CHENGWEI ZHANG, HAN XIAO-RAN, YANG XIAOBAO, JIANG BIAO, LIU JING, XIONG YUE, JIN JIAN: "Proteolysis Targeting Chimeras (PROTACs) of Anaplastic Lymphoma Kinase (ALK)", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 151, 1 May 2018 (2018-05-01), AMSTERDAM, NL , pages 304 - 314, XP055581975, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2018.03.071 *
CHUNG HYO KANG, DONG HO LEE, CHONG OCK LEE, JAE DU HA, CHI HOON PARK, JONG YEON HWANG: "Induced protein degradation of anaplastic lymphoma kinase (ALK) by proteolysis targeting chimera (PROTAC)", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ELSEVIER, AMSTERDAM NL, vol. 505, no. 2, 28 September 2018 (2018-09-28), Amsterdam NL , pages 542 - 547, XP055765181, ISSN: 0006-291X, DOI: 10.1016/j.bbrc.2018.09.169 *
PHULL MANJINDER SINGH; JADAV SURENDER SINGH; GUNDLA RAMBABU; MAINKAR PRATHAMA S.: "A perspective on medicinal chemistry approaches towards adenomatous polyposis coli and Wnt signal based colorectal cancer inhibitors", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 212, 3 January 2021 (2021-01-03), AMSTERDAM, NL , XP086494847, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2020.113149 *
REN CHAOWEI, SUN NING, LIU HAIXIA, KONG YING, SUN RENHONG, QIU XING, CHEN JINJU, LI YAN, ZHANG JIANSHUI, ZHOU YUEDONG, ZHONG HUI, : "Discovery of a Brigatinib Degrader SIAIS164018 with Destroying Metastasis-Related Oncoproteins and a Reshuffling Kinome Profile", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 64, no. 13, 8 July 2021 (2021-07-08), US , pages 9152 - 9165, XP055981658, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.1c00373 *
REN CHAOWEI; SUN NING; KONG YING; QU XIAOJUAN; LIU HAIXIA; ZHONG HUI; SONG XIAOLING; YANG XIAOBAO; JIANG BIAO: "Structure-based discovery of SIAIS001 as an oral bioavailability ALK degrader constructed from Alectinib", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 217, 11 March 2021 (2021-03-11), AMSTERDAM, NL , XP086577045, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2021.113335 *
SUN NING; REN CHAOWEI; KONG YING; ZHONG HUI; CHEN JINJU; LI YAN; ZHANG JIANSHUI; ZHOU YUEDONG; QIU XING; LIN HAIFAN; SONG XIAOLING: "Development of a Brigatinib degrader (SIAIS117) as a potential treatment for ALK positive cancer resistance", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 193, 29 February 2020 (2020-02-29), AMSTERDAM, NL , XP086118072, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2020.112190 *
XIE SHAOWEN, SUN YUAN, LIU YULIN, LI XINNAN, LI XINUO, ZHONG WENYI, ZHAN FEIYAN, ZHU JINGJIE, YAO HONG, YANG DONG-HUA, CHEN ZHE-SH: "Development of Alectinib-Based PROTACs as Novel Potent Degraders of Anaplastic Lymphoma Kinase (ALK)", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 64, no. 13, 8 July 2021 (2021-07-08), US , pages 9120 - 9140, XP093005046, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.1c00270 *
ZHANG HUALIN, XIE RULIANG, AI-FURAS HAWAA, LI YUPENG, WU QINGXIA, LI JIAN, XU FANG, XU TIANFENG: "Design, Synthesis, and Biological Evaluation of Novel EGFR PROTACs Targeting Del19/T790M/C797S Mutation", ACS MEDICINAL CHEMISTRY LETTERS, AMERICAN CHEMICAL SOCIETY, US, vol. 13, no. 2, 10 February 2022 (2022-02-10), US , pages 278 - 283, XP093005050, ISSN: 1948-5875, DOI: 10.1021/acsmedchemlett.1c00645 *

Also Published As

Publication number Publication date
CN116940581A (zh) 2023-10-24

Similar Documents

Publication Publication Date Title
TWI765908B (zh) 苯並咪唑類化合物激酶抑制劑及其製備方法和應用
CN111372925B (zh) 作为c-MET/AXL抑制剂的尿嘧啶类化合物
ES2426482T3 (es) Inhibidor de IGF-1R
CN112552294B (zh) 含哌嗪杂环类衍生物抑制剂、其制备方法和应用
CN111153901A (zh) 一类含氮稠杂环类shp2抑制剂化合物、制备方法和用途
WO2020259432A1 (zh) Kras-g12c抑制剂
WO2021219090A1 (zh) 喹喔啉二酮衍生物作为kras g12c突变蛋白的不可逆抑制剂
WO2021027911A1 (zh) 新型螺环类K-Ras G12C抑制剂
CN112142735A (zh) 一类稠和氰基吡啶类化合物、制备方法和用途
WO2019134539A1 (zh) 二氢吡唑酮并嘧啶类化合物及其制备方法和用途
WO2012019427A1 (zh) 酞嗪酮类衍生物、其制备方法及其在医药上的应用
EA019309B1 (ru) Модуляторы ampk (амф-активируемой протеинкиназы)
CN110698481A (zh) 杂芳基吡啶酮和氮杂-吡啶酮酰胺化合物
WO2015081813A1 (zh) 具有alk抑制活性的化合物及其制备与用途
JP7384536B2 (ja) キナゾリン化合物並びにその調製方法、使用及び医薬組成物
WO2023134266A1 (zh) 2-哌啶基或2-吡唑基取代的嘧啶化合物作为egfr抑制剂
CN114436976A (zh) 一种新型喹唑啉类衍生物及其制备和应用
CN113527299A (zh) 一类含氮稠环类化合物、制备方法和用途
WO2020238776A1 (zh) 取代的稠合双环类衍生物、其制备方法及其在医药上的应用
CN113045569B (zh) 用作ret激酶抑制剂的化合物及其应用
CN115028644A (zh) Sos1抑制剂杂环化合物
CN114437116A (zh) 杂环化合物及其制备方法、药物组合物和应用
WO2018028491A1 (zh) 吲哚胺2,3-双加氧酶抑制剂及其在药学中的用途
JP2022538901A (ja) ピラゾロン縮合ピリミジン化合物、その製造方法及び使用
CN109111439B (zh) 一种酰胺类化合物及包含该化合物的组合物及其用途

Legal Events

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

Ref document number: 22804045

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280016739.4

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: 22804045

Country of ref document: EP

Kind code of ref document: A1