WO2022161421A1 - 作为免疫调节剂的联苯类化合物及其制备方法和应用 - Google Patents

作为免疫调节剂的联苯类化合物及其制备方法和应用 Download PDF

Info

Publication number
WO2022161421A1
WO2022161421A1 PCT/CN2022/074183 CN2022074183W WO2022161421A1 WO 2022161421 A1 WO2022161421 A1 WO 2022161421A1 CN 2022074183 W CN2022074183 W CN 2022074183W WO 2022161421 A1 WO2022161421 A1 WO 2022161421A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
alkyl
formula
tetrahydro
imidazo
Prior art date
Application number
PCT/CN2022/074183
Other languages
English (en)
French (fr)
Inventor
杨千姣
山松
辛利军
潘德思
王晓亮
宋永连
黄卉云
魏奇
李志斌
鲁先平
Original Assignee
深圳微芯生物科技股份有限公司
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 深圳微芯生物科技股份有限公司 filed Critical 深圳微芯生物科技股份有限公司
Priority to CA3210314A priority Critical patent/CA3210314A1/en
Priority to AU2022214735A priority patent/AU2022214735A1/en
Priority to EP22745294.3A priority patent/EP4286383A1/en
Priority to CN202280008146.3A priority patent/CN116710445A/zh
Priority to KR1020237029267A priority patent/KR20230141812A/ko
Priority to JP2023546481A priority patent/JP2024505972A/ja
Priority to BR112023015451A priority patent/BR112023015451A2/pt
Publication of WO2022161421A1 publication Critical patent/WO2022161421A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/43Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • C07C211/44Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton having amino groups bound to only one six-membered aromatic ring
    • C07C211/49Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton having amino groups bound to only one six-membered aromatic ring having at least two amino groups bound to the carbon skeleton
    • C07C211/50Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton having amino groups bound to only one six-membered aromatic ring having at least two amino groups bound to the carbon skeleton with at least two amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton

Definitions

  • the invention belongs to the field of medicinal chemistry, in particular to biphenyl compounds as immunomodulators and a preparation method and application thereof.
  • Tumor immunotherapy is a new treatment method that inhibits or kills tumor cells by stimulating the body's immune system and enhancing its own anti-tumor immunity. This method has achieved a breakthrough after more than 100 years of efforts. In 2013, "Science” magazine listed tumor immunotherapy as the first of the top ten scientific breakthroughs of the year (Couzin-Frankel J., 2013, Science, 342: 1432-1433), which has become one of the most promising areas of anti-tumor treatment .
  • tumor cells Compared with normal cells, tumor cells have a variety of genetic and epigenetic changes.
  • the immune system can use the surface antigens produced by tumor cells to distinguish the two, thereby triggering an anti-tumor immune response.
  • T cell anti-tumor immunity after being activated by T cell receptor (TCR)-mediated antigen recognition signals, it comprehensively regulates T cell effects, including cytotoxic T lymphocytes, through co-stimulatory and co-suppressive signals.
  • TCR T cell receptor
  • Cytotoxic T-lymphocyte associated antigen 4 Cytotoxic T-lymphocyte associated antigen 4, CTLA4
  • PD-1 programmed death protein 1
  • T cell activation immunoglobulin inhibit V-domain (V-domain immunoglobulin suppressor of T-cell activation, VISTA), T cell immunoglobulin and mucin domain-containing-3 (TIM3)
  • TIM3 T cell immunoglobulin and mucin domain-containing-3
  • LAG3 lymphocyte activation gene 3
  • Inhibitory receptors such as inhibitory signals
  • activating receptors for stimulatory signals such as CD28, CD134 (OX40), Glucocorticoid-induced TNFR-related protein (GITR), CD137, CD27, HVEM, etc.
  • immune checkpoints are involved in maintaining immune tolerance to self-antigens and avoiding autoimmune diseases on the one hand; on the other hand, avoiding excessive activation of immune responses leading to tissue damage.
  • tumor cells In tumor cells, however, they can evade immune killing by suppressing T cell activation through immune checkpoints. Therefore, it is necessary to reactivate T cells to attack tumor cells by activating co-stimulatory signals (stepping on the "accelerator") and inhibiting co-inhibitory signals (releasing the "brake”), thereby realizing tumor immunotherapy.
  • PD-1 is expressed in activated T cells, B cells and myeloid cells. It belongs to the CD28 family and is a type 1 transmembrane glycoprotein on T cells, consisting of 288 amino acids.
  • the molecular structure of PD-1 is composed of an extracellular region with immunoglobulin IgV-like (amino acids 35-145), a transmembrane region, and a cytoplasmic tail region with the function of linking signal peptides. Play important functions (Cheng X., Veverka V., Radhakrishnan A., et al. 2013, J. Biol. Chem., 288:11771-11785).
  • PDC1 Programmed death protein ligand 1
  • T cells tumor cells
  • APCs antigen presenting cells
  • non-hematopoietic cells it is also a type 1 transmembrane glycoprotein, which consists of 290 amino acids.
  • T cell activation which is essential for maintaining immune tolerance in normal organisms, and when tumor cells neutralize virus infection, PD-1 on T cells is induced to be highly expressed , the expression of PD-L1 is up-regulated, resulting in the continuous activation of PD-1 signaling pathway and the inhibition of T cell proliferation, resulting in immune escape of tumor cells and pathogens (Fuller M.J., Callendret B., Zhu B., et al.2013, Proc.Natl .Acad.Sci.USA., 110:15001-15006; Dolan D.E., Gupta S., 2014, Cancer Control, 21:231-237; Chen L., Han X., 2015, J.Clin.Invest., 125 : 3384-3391; Postow M.A., Callahan M.K., Wolchok J.D., 2015, J.Clin.Oncol., 33:1974-1982
  • PD-L1 can interact with CD80 and inhibit the binding of PD-L1 and PD-1, as well as the ability to inhibit T cell activation. Therefore, blocking the immune activation caused by CD80/PD-L1 interaction may also promote the enhancement of T cell activity, thereby providing new therapeutic opportunities for immune-related diseases (Sugiura D., Maruhashi T., Okazakill-mi, et al. 2019, Science, 364:558-566).
  • small-molecule immunomodulators have certain advantages, including oral administration, more tissue permeability, and the ability to adjust pharmacological properties to minimize side effects.
  • small molecule inhibitors will have a lower price advantage and better medication compliance.
  • nivolumab T1/2 is 25.2 days, and the dosing frequency is once every two weeks; pembrolizumab T1/2 is 25 days, and the dosing frequency is Once every three weeks; atezolizumab T1/2 for 27 days and dosing frequency every three weeks.
  • the administration frequency of the above drugs is shorter than the drug half-life, indicating that the continuous exposure of such target drugs in vivo is the key to obtain ideal clinical efficacy.
  • the existing small molecule immunomodulators have low exposure in vivo and short duration of exposure, which will affect the clinical efficacy. Therefore, it is necessary to develop novel small-molecule PD-L1 immunomodulators with higher activity and better oral absorption characteristics, especially with sufficient in vivo exposure and sustained exposure time, and more targeting to tumor tissue to meet the unmet clinical requirements. Demand is significant.
  • One aspect of the present invention relates to a small molecule biphenyl compound capable of targeting PD-L1, or an isomer, pharmaceutically acceptable salt, precursor or metabolite thereof.
  • Another aspect of the present invention pertains to methods of making the compounds described herein.
  • Yet another aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of the present invention as an active ingredient, and the use of the compound or the pharmaceutical composition of the present invention in the manufacture of a medicament for the treatment and/or prevention of a disease associated with the target PD-L1 .
  • a small molecule PD-L1 inhibitor compared with antibodies, it needs to be optimized in terms of target binding strength and continuous exposure time in vivo, so as to narrow the possible gap between the two sides in terms of drug efficacy. Therefore, the inventors expect to develop new small molecule PD-L1 immunomodulators with higher activity and better oral absorption characteristics, especially with sufficient in vivo exposure and sustained exposure time and more targeting to tumor tissue. Address unmet clinical needs.
  • the present invention overcomes the shortcoming that the existing PD-1/PD-L1 antibody drugs all require injection and administration, and provides a novel small molecule immunomodulator with excellent orally absorbable characteristics, in particular, the compound of the present invention has It has an ideal in vivo exposure amount and continuous exposure time, and at the same time is targeted to tumor tissue, which can be enriched in tumor tissue and form a higher exposure concentration in tumor tissue, which is helpful for better anti-tumor activity in treatment. achieve better efficacy.
  • the present invention relates to a compound represented by formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite,
  • R 1 and R 2 are the same or different, selected from C 1 -C 6 alkyl, cyano, halogen;
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl, C 2 -C 6 alkynyl C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkoxy C 1 -C 6 alkyl, mono C 1 -C 6 alkylamino C 1 -C 6 alkyl, bis C 1 -C 6 alkyl amino C 1 -C 6 alkyl, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl, C 3 -C 14 cycloalkyl, 3- to 14-membered heterocycloalkyl, C 3 -C 14 cycloalkyl- C 1 -C 4 alkyl, 3- to 14-membered heterocycloalkyl-C 1 -C 4 alkyl;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl, wherein said C 1 -C 6 alkyl is optionally substituted with one or more substituents selected from hydroxyl, carboxyl and halogen;
  • X is selected from -O-, -S-;
  • R 5 -R 26 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms
  • R 1 and R 2 may be the same or different and are selected from methyl, cyano, halogen;
  • R 1 and R 2 may be the same or different and are selected from methyl, cyano, fluoro, chloro;
  • R 1 is selected from C 1 -C 6 alkyl, cyano, halogen
  • R 2 is selected from C 1 -C 6 alkyl, halogen
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, at least 1 hydrogen atom in these substituents is replaced by deuterium (D);
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl, at least 1 hydrogen atom in these substituents is replaced by deuterium (D);
  • R3 is selected from methyl, ethyl, -CHF2, and at least 1 hydrogen atom of these substituents is replaced by deuterium (D);
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • R is selected from hydrogen, methyl
  • X is selected from -O-;
  • R5 - R26 are selected from hydrogen atoms, preferably wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 5 -R 26 are all hydrogen atoms
  • R 5 -R 26 are hydrogen atoms, wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least one hydrogen atom is replaced by deuterium (D), the deuterated position may be on the corresponding aliphatic ring and aliphatic heterocycle. any location;
  • R 27 -R 30 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuterated position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • the present invention relates to compound shown in formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite,
  • R 1 and R 2 may be the same or different and are selected from methyl, methyl-d 3 (CD 3 ), cyano, fluorine, chlorine, bromine;
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl, C 2 -C 6 alkynyl C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkoxy C 1 -C 6 alkyl, mono C 1 -C 6 alkylamino C 1 -C 6 alkyl, bis C 1 -C 6 alkyl amino C 1 -C 6 alkyl, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl, C 3 -C 14 cycloalkyl, 3- to 14-membered heterocycloalkyl, C 3 -C 14 cycloalkyl- C 1 -C 4 alkyl, 3- to 14-membered heterocycloalkyl-C 1 -C 4 alkyl;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl, wherein said C 1 -C 6 alkyl is optionally substituted by one or more substituents selected from hydroxyl and halogen;
  • X is selected from -O-;
  • R 5 -R 26 are hydrogen atoms and at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and at least one hydrogen atom is replaced by deuterium (D), and the deuterium substitution position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • R 1 and R 2 may be the same or different and are selected from methyl, cyano, fluoro, chloro;
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, haloC 1 -C 6 alkyl;
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl;
  • R 3 is selected from methyl, ethyl, -CHF 2 ;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • R is selected from hydrogen, methyl
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuterated position can be any on the corresponding aliphatic ring and aliphatic heterocycle Location;
  • R 27 -R 30 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuteration position can be any position on the corresponding alicyclic and aliphatic heterocyclic rings;
  • the present invention relates to a compound represented by formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite,
  • R 1 and R 2 may be the same or different and are selected from methyl, methyl-d 3 (CD 3 ), cyano, fluorine, chlorine, bromine;
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl, C 2 -C 6 alkynyl C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkoxy C 1 -C 6 alkyl, mono C 1 -C 6 alkylamino C 1 -C 6 alkyl, bis C 1 -C 6 alkyl amino C 1 -C 6 alkyl, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl, C 3 -C 14 cycloalkyl, 3- to 14-membered heterocycloalkyl, C 3 -C 14 cycloalkyl- C 1 -C 4 alkyl, 3- to 14-membered heterocycloalkyl-C 1 -C 4 alkyl;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl, wherein said C 1 -C 6 alkyl is optionally substituted by one or more substituents selected from hydroxyl and halogen;
  • X is selected from -O-;
  • R 5 -R 26 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms and at least one hydrogen atom is substituted by deuterium (D), and the deuterium substitution position can be at any position on the corresponding alicyclic ring and aliphatic heterocyclic ring;
  • R 1 and R 2 may be the same or different and are selected from methyl, cyano, fluoro, chloro;
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, haloC 1 -C 6 alkyl;
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl;
  • R 3 is selected from methyl, ethyl, -CHF 2 ;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • R is selected from hydrogen, methyl
  • R5 - R26 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 5 -R 26 are selected from hydrogen atoms
  • R5 - R26 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • the present invention relates to a compound represented by formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite, wherein,
  • R 1 and R 2 may be the same or different and are selected from methyl, methyl-d 3 (CD 3 ), cyano, fluorine, chlorine;
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl-, C 2 -C 6 alkynyl C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkyl-, C 1 -C 6 alkoxy C 1 -C 6 alkyl-, mono C 1 -C 6 alkylamino C 1 -C 6 alkyl-, bis C 1 -C 6 alkyl Amino C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl-, C 3 -C 14 cycloalkyl, 3- to 14-membered heterocycloalkyl, these radicals At least one hydrogen atom in the group is replaced by deuterium (D);
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl, wherein said C 1 -C 6 alkyl is optionally substituted with halogen;
  • X is selected from -O-;
  • R 5 -R 26 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and at least one hydrogen atom is substituted by deuterium (D), and the deuterium substitution position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, at least 1 hydrogen atom in these substituents is replaced by deuterium (D);
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl, at least 1 hydrogen atom in these substituents is replaced by deuterium (D);
  • R3 is selected from methyl, ethyl, -CHF2, and at least 1 hydrogen atom of these substituents is replaced by deuterium (D);
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • R is selected from hydrogen, methyl
  • R5 - R26 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 5 -R 26 are selected from hydrogen atoms
  • R5 - R26 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least one hydrogen atom is replaced by deuterium (D), the deuterated position may be on the corresponding aliphatic ring and aliphatic heterocycle. any location;
  • R 27 -R 30 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuterated position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • the present invention relates to a compound represented by formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite, wherein,
  • R 1 and R 2 may be the same or different and are selected from methyl, methyl-d 3 (CD 3 ), cyano, fluorine, chlorine;
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl-, C 2 -C 6 alkynyl C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkyl-, C 1 -C 6 alkoxy C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl-;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl, wherein said C 1 -C 6 alkyl is optionally substituted with halogen;
  • X is selected from -O-;
  • R 5 -R 26 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and at least one hydrogen atom is replaced by deuterium (D), and the deuterium substitution position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, haloC 1 -C 6 alkyl;
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl;
  • R 3 is selected from methyl, ethyl, -CHF 2 ;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • R4 is selected from hydrogen
  • R 27 -R 30 are all selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuterated position may be on the corresponding aliphatic ring and aliphatic heterocycle. any location;
  • R 27 -R 30 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuterated position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • the present invention relates to a compound represented by formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite, wherein,
  • R 1 and R 2 may be the same or different and are selected from methyl, methyl-d 3 (CD 3 ), cyano, fluorine, chlorine;
  • R 3 is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl-, C 2 -C 6 alkynyl C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkyl-, C 1 -C 6 alkoxy C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl-;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl, wherein said C 1 -C 6 alkyl is optionally substituted with halogen;
  • X is selected from -O-;
  • R 5 -R 27 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 28 -R 30 are selected from hydrogen atoms and wherein at least one hydrogen atom is replaced by deuterium (D), and the deuterium substitution position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, haloC 1 -C 6 alkyl;
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl;
  • R 3 is selected from methyl, ethyl, -CHF 2 ;
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • R4 is selected from hydrogen
  • R 5 -R 26 are all selected from hydrogen atoms, or at least one is deuterium (D);
  • R 5 -R 26 are all selected from hydrogen atoms
  • At least one of R 5 -R 26 is deuterium (D);
  • the present invention relates to a compound represented by formula (I), its isomer, pharmaceutically acceptable salt, precursor or metabolite, wherein,
  • R 1 and R 2 may be identically or differently selected from methyl, methyl-d 3 (CD 3 ), cyano, fluorine, chlorine;
  • R 3 is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl-, C 2 -C 6 alkynyl C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkyl-, C 1 -C 6 alkoxy C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl-, at least 1 hydrogen in these groups Atoms are replaced by deuterium (D);
  • R 4 is selected from hydrogen
  • X is selected from -O-;
  • R 5 -R 27 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 28 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and at least one hydrogen atom is replaced by deuterium (D), and the deuterium substitution position can be at any position on the corresponding aliphatic ring and aliphatic heterocycle;
  • R 1 is selected from methyl, cyano, fluoro, chloro;
  • R 2 is selected from methyl, chloro
  • R 3 is selected from C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, at least 1 hydrogen atom in these substituents is replaced by deuterium (D);
  • R 3 is selected from C 1 -C 6 alkyl, fluoro C 1 -C 6 alkyl, at least 1 hydrogen atom in these substituents is replaced by deuterium (D);
  • R3 is selected from methyl, ethyl, -CHF2, and at least 1 hydrogen atom of these substituents is replaced by deuterium (D);
  • R5 - R26 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 5 -R 26 are selected from hydrogen atoms
  • R5 - R26 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R 27 -R 30 are selected from hydrogen atoms, or are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), and the deuterated position can be any on the corresponding aliphatic ring and aliphatic heterocycle Location;
  • R 27 -R 30 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms and wherein at least 1 hydrogen atom is replaced by deuterium (D), the deuterated position can be anywhere on the corresponding aliphatic and aliphatic heterocycles.
  • R 1 and R 2 are the same or different and are selected from methyl, cyano, halogen;
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkyl-, C 2 -C 6 alkynyl C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkyl-, C 1 -C 6 alkoxy C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl-, at least 1 of these groups hydrogen atoms are replaced by deuterium (D);
  • R 4 is selected from hydrogen, C 1 -C 6 alkyl
  • X is selected from -O-;
  • R 5 -R 26 are selected from hydrogen atoms
  • R 27 -R 30 are selected from hydrogen atoms.
  • R 1 and R 2 are the same or different and are selected from methyl, cyano, fluoro, chloro;
  • R 1 and R 2 are the same or different and are selected from methyl, fluoro, chloro;
  • R 1 and R 2 are the same or different and are selected from methyl, chloro.
  • R 3 is selected from hydrogen, C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, at least 1 hydrogen atom of these groups is replaced by deuterium (D);
  • R 3 is selected from C 1 -C 6 alkyl, wherein at least 1 hydrogen atom is replaced by deuterium (D);
  • R3 is selected from methyl, ethyl, propyl, butyl, pentyl, wherein at least 1 hydrogen atom is replaced by deuterium (D).
  • 1-15 hydrogen atoms in the R defined group are replaced by deuterium (D);
  • 1-9 hydrogen atoms in the R3 -defined group are replaced by deuterium (D).
  • R4 is selected from hydrogen, methyl, ethyl, propyl, butyl, pentyl;
  • R4 is selected from hydrogen.
  • the present invention relates to a compound represented by formula (I), preferably selected from the following compounds:
  • C1 - C6 alkyl specifically refers to the independently disclosed methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl and C6 alkyl, or the independently disclosed " C1 alkyl” -C 4 alkyl", or independently disclosed "C 1 -C 3 alkyl”.
  • halogen in the present invention is fluorine, chlorine, bromine or iodine, preferably fluorine or chlorine or bromine.
  • alkyl group in the present invention includes straight-chain or branched-chain alkyl groups.
  • the C 1 -C 6 alkyl group mentioned in the present invention refers to an alkyl group with 1-6 carbon atoms, preferably methyl, ethyl, n-propyl or isopropyl, n-butyl, isobutyl or tert-butyl.
  • the alkyl groups in the compounds of the present invention may be optionally substituted or unsubstituted, and the substituted substituents may include alkyl, halogen, alkoxy, haloalkyl, cyano, hydroxy, and the like.
  • Examples of alkyl groups of the present invention include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, and the like.
  • alkynyl in the present invention refers to a linear or branched monovalent hydrocarbon group of 2-12 carbon atoms, or 2-8 carbon atoms, or 2-6 carbon atoms, or 2-4 carbon atoms , wherein at least one position is unsaturated, that is, one CC is a sp triple bond, wherein the alkynyl group can be independently and optionally substituted by one or more substituents described in the present invention, wherein the alkynyl group can be C 2 - C6alkynyl , specific examples include, but are not limited to, ethynyl (-C ⁇ CH), propargyl ( -CH2C ⁇ CH ) and the like.
  • alkoxy group in the present invention refers to a group formed by connecting the above-mentioned alkyl group with an oxygen atom, wherein the oxygen atom has the ability to form bonds freely, such as "C 1 -C 6 alkoxy group", Specifically, such as methoxy, ethoxy, n-propoxy, n-butoxy, isopropoxy, tert-butoxy, cyclopropoxy and the like.
  • alkylamino group in the present invention refers to the group formed by connecting the above-mentioned alkyl group with an amino group, such as "C 1 -C 6 alkylamino group", specifically such as methylamino, ethylamino, dimethylamino , methyl isopropyl amino, etc.
  • halogenated C 1 -C 6 alkyl group and “halogenated C 1 -C 6 alkoxy group” in the present invention mean that one or more hydrogen atoms in the alkyl group and alkoxy group are replaced by halogen atoms , especially fluorine or chlorine atom substitution.
  • fluorine is preferred, eg -CF3 , -CHF2 , -CH2F , -CH2CH2F , -CH2CHF2 , -CH2CF3 , -OCF3 , -OCHF2 , -OCH 2 F, -OCH 2 CH 2 F, -OCH 2 CHF 2 or -OCH 2 CF 3 .
  • cycloalkyl in the present invention refers to a hydrocarbon monocyclic structure with a specified number of ring carbon atoms, which does not contain unsaturated bonds such as double bonds, including C 3 -C 14 cycloalkyl, C 3 -C 6 Cycloalkyl groups are, for example, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Cycloalkyl groups in the compounds of the present invention may be optionally substituted or unsubstituted, and the substituents may include alkyl groups, halogens, alkoxy groups, hydrocarbyl groups, hydroxyl groups, and the like.
  • heterocycle in the present invention refers to unsubstituted and substituted monocyclic or polycyclic non-aromatic, partially unsaturated or fully saturated ring systems containing one or more heteroatoms.
  • Preferred heteroatoms include N, O and S.
  • Monocyclic heterocycles include, but are not limited to, pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, dihydroimidazolyl, dihydrofuranyl, piperidinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like.
  • Polycyclic heterocyclic groups include spiro, bridged, and fused-ring heterocyclic groups, and the heterocyclic groups may be fused to aryl, heteroaryl or cycloalkyl rings.
  • Other similar compound names such as "C 2 -C 6 alkynyl C 1 -C 6 alkyl-", "C 3 -C 14 cycloalkyl-C 1 -C 4 alkyl-" can be understood with reference to the foregoing.
  • substituted means that one or more hydrogen atoms in a group are capable of being independently replaced with the corresponding number of substituents. Those skilled in the art can determine (either experimentally or theoretically) possible or impossible substitution positions without undue effort.
  • substituted refers to substituents including but not limited to: cyano, carboxyl, halogen, hydroxyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 1 -C 6 alkoxy, mono C 1 -C 6 alkylamino C 1 -C 6 alkyl-, bis C 1 -C 6 alkylamino C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkoxy C 1 -C 6 alkyl-, halogenated C 1 -C 6 alkoxy C 1 -C 6 alkyl-, C 3 -C 14 cycloalkyl, 3- to 14-membered heterocycloalkyl, C3-C14cycloalkyl- C1 -C4alkyl-, 3- to 14 -membered heterocycloalkyl- C1 - C4alkyl-, 3-
  • the "pharmaceutically acceptable salt” in the present invention refers to an acid addition salt prepared by reacting the compound of the present invention with a pharmaceutically acceptable acid, or a salt formed by the reaction of a compound with an acidic group and a basic compound.
  • the acid is preferably selected from inorganic acids (such as hydrochloric acid, sulfuric acid, phosphoric acid or hydrobromic acid, etc.), and organic acids (such as oxalic acid, maleic acid, fumaric acid, malic acid, tartaric acid, lysine, etc.) , histidine, citric acid or benzoic acid, etc.);
  • the basic compound is preferably selected from sodium hydroxide, potassium hydroxide, calcium hydroxide, sodium carbonate or potassium bicarbonate and the like.
  • the above-mentioned pharmaceutically acceptable salts are easy to separate, and can be purified by conventional separation methods, such as solvent extraction, dilution, recrystallization, column chromatography and preparative thin layer chromatography.
  • hydrogen atom as used herein generally means the isotopic form of hydrogen having a mass number of 1, ie 1 H.
  • Another aspect of the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the above-mentioned compound, or an isomer, pharmaceutically acceptable salt, precursor or metabolite thereof, as an active ingredient.
  • the compounds described in the present invention can optionally be used in combination with one or more other active ingredients, and the respective dosages and ratios can be adjusted by those skilled in the art according to specific conditions and specific conditions of patients, as well as clinical needs.
  • the compound of general formula (I) of the present invention can be prepared by those skilled in the art (experience or references).
  • Another aspect of the present invention also provides a process for the preparation of the above-mentioned compounds according to the present invention.
  • the following synthetic route describes the preparation method of the compound of formula (I) of the present invention.
  • the raw materials, reagents, catalysts, solvents, etc. used in the following synthetic schematic diagram can be prepared by methods well known to those of ordinary skill in the field of organic chemistry or can be obtained commercially.
  • All final derivatives of the present invention can be prepared by the methods described in the schematic diagrams or by analogous methods, which are well known to those of ordinary skill in the art of organic chemistry. All variables used in these diagrams are as defined in context.
  • the compounds of the general formula (I) and the related intermediates can be purified by common separation methods, such as extraction, recrystallization and silica gel column chromatography separation.
  • the 200-300 mesh silica gel and thin-layer chromatography silica gel plates used were produced by Qingdao Ocean Chemical Factory.
  • the chemical reagents used are commercial products of analytical or chemical purity of general reagents, and are used without further purification.
  • the present invention provides a kind of preparation method of compound shown in general formula (I), it comprises the steps:
  • the compound represented by the formula (I-c) and the compound represented by the formula (I-d') are subjected to a nucleophilic substitution reaction in the presence of a third base to obtain the compound represented by the formula (I-e);
  • the compound represented by the formula (I-e) and the compound represented by the formula (I-f') are subjected to a nucleophilic substitution reaction in the presence of a fourth base to obtain the compound represented by the formula (I);
  • the compound of formula (I') is used as a form of the final product (I).
  • the compound represented by the formula (I') can further undergo ester hydrolysis under basic conditions to obtain the compound represented by the formula (I"), and the compound represented by the formula (I") is also used as the final product (I).
  • R 4' has the same definition as R 4 except that it is not hydrogen, and R 4 is as defined above;
  • R 4" is hydrogen
  • M is selected from boronic esters or boronic acids, including but not limited to 4,4,5,5-tetramethyl-1,3,2-dioxolaborane, neopentyl glycol diboronate, 4,4, 4',4',5,5,5',5'-octamethyl-2,2'-bis(1,3,2-dioxaborane)B(OBu-n) 3 , B (OPr-i) 3 ; or,
  • M is selected from bromine, iodine, chlorine, fluorine, CF3SO3- (OTf);
  • W is selected from boronic acid esters or boronic acids, preferably from 4,4,5,5-tetramethyl-1,3,2-dioxolaborane, neopentyl glycol diboronate, 4,4, 4',4',5,5,5',5'-octamethyl-2,2'-bis(1,3,2-dioxaborane)B(OBu-n) 3 , B (OPr-i) 3 ; or,
  • W is selected from bromine, iodine, chlorine, fluorine, CF3SO3- ( OTf);
  • P 1 and P 2 are protecting groups, which may be the same or different, preferably selected from Boc (tert-butoxycarbonyl), Fmoc (9-fluorenemethoxycarbonyl), Cbz (N-benzyloxycarbonyl), methanesulfonyl, p- Tosyl, acetyl, methoxycarbonyl, ethoxycarbonyl, ((2-trimethylsilyl)ethoxy)methyl (SEM), tetrahydro-2H-pyran-2-yl (THP).
  • the first acid is preferably selected from trifluoroacetic acid (TFA), hydrochloric acid (HCl), acetic acid (HOAc), hydrobromic acid (HBr);
  • Described first base is preferably selected from piperidine, diethylamine;
  • the first solvent is preferably selected from dichloromethane (DCM), 1,2-dichloroethane, methanol (MeOH), ethanol (EtOH), 1,4-dioxane (1,4-dioxane) ), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF).
  • DCM dichloromethane
  • MeOH methanol
  • EtOH ethanol
  • 1,4-dioxane 1,4-dioxane
  • THF tetrahydrofuran
  • MeCN acetonitrile
  • DMF N,N'-dimethylformamide
  • the first catalyst is preferably selected from 1,1'-bis(dicyclohexylphosphino)ferrocene palladium dichloride (PdCl 2 (dcypf)), palladium acetate (Pd(OAc) 2 ), palladium dichloride (PdCl 2 ), tris(dibenzylideneacetone)dipalladium (Pd 2 (dba) 3 ), [1,1'-bis(diphenylphosphino)ferrocene]dichloro Palladium (PdCl 2 (dppf)), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium dichloromethane complex (PdCl 2 (dppf) ⁇ CH 2 Cl 2 ), Tetrakis(triphenylphosphine)palladium (Pd(PPh 3 ) 4 ), bis(tricyclohexylphosphine)palladium dichloride
  • the second base includes organic bases and inorganic bases, such as triethylamine (TEA), N,N-diisopropylethylamine (DIPEA), n-butyllithium, lithium diisopropylamide, bis-triethylamine Lithium methylsilylamide, potassium acetate (KOAc), sodium tert-butoxide (NaOBu-t), potassium tert-butoxide (KOBu-t), sodium hydride (NaH), potassium phosphate (K 3 PO 4 ), sodium carbonate (Na 2 CO 3 ), potassium carbonate (K 2 CO 3 ), lithium hydroxide (KOH), sodium hydroxide (NaOH);
  • TAA triethylamine
  • DIPEA N,N-diisopropylethylamine
  • n-butyllithium lithium diisopropylamide
  • bis-triethylamine Lithium methylsilylamide potassium acetate (KOAc),
  • the second solvent is preferably selected from 1,4-dioxane (1,4-dioxane), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF), and Mixed solvents of these solvents and water in different proportions.
  • the first reducing agent and the second reducing agent are preferably selected from sodium borohydride acetate, sodium borohydride, sodium cyanoborohydride;
  • the third solvent and the fifth solvent are preferably selected from dichloromethane (DCM), 1,2-dichloroethane, methanol (MeOH), ethanol (EtOH), 1,4-dioxane (1,4-dioxane) 4-dioxane), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF).
  • DCM dichloromethane
  • MeOH methanol
  • EtOH ethanol
  • 1,4-dioxane 1,4-dioxane
  • THF tetrahydrofuran
  • MeCN acetonitrile
  • DMF N,N'-dimethylformamide
  • the fourth solvent and the sixth solvent are preferably selected from dichloromethane (DCM), 1,2-dichloroethane, methanol (MeOH), ethanol (EtOH), 1,4-dichloroethane Oxane (1,4-dioxane), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF), N-methylpyrrolidone (NMP), pyridine (Py);
  • DCM dichloromethane
  • MeOH methanol
  • EtOH ethanol
  • 1,4-dichloroethane Oxane (1,4-dioxane
  • THF tetrahydrofuran
  • MeCN acetonitrile
  • DMF N,N'-dimethylformamide
  • NMP N-methylpyrrolidone
  • Py pyridine
  • the third base and the fourth base are preferably selected from triethylamine (TEA), N,N'-diisopropylethylamine (DIPEA), pyridine (Py), n-butyllithium, diisopropyl Lithium amide, lithium bistrimethylsilylamide, sodium tert-butoxide (NaOBu-t), potassium tert-butoxide (KOBu-t), sodium hydride (NaH), sodium carbonate (Na 2 CO 3 ), potassium carbonate (K 2 CO 3 ), Lithium Hydroxide (KOH), Sodium Hydroxide (NaOH).
  • the fifth base comprises preferably selected from lithium hydroxide (LiOH), lithium hydroxide (KOH), sodium hydroxide (NaOH).
  • the compound represented by formula (Ia) is subjected to appropriate acid, base or catalytic hydrogenolysis to remove the protecting group P 1 in a suitable solvent, and the obtained product is not separated and purified, and further reacts with the compound represented by formula (Ib) in a suitable solution.
  • the compound represented by the formula (Ic) is obtained by Suzuki reaction under a catalyst, and the appropriate solvent for the deprotection is preferably selected from dichloromethane (DCM), 1,2-dichloroethane , methanol (MeOH), ethanol (EtOH), 1,4-dioxane (1,4-dioxane), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF) ; Described acid is preferably selected from trifluoroacetic acid (TFA), hydrochloric acid (HCl), acetic acid (HOAc), hydrobromic acid (HBr); Described base is preferably selected from piperidine, diethylamine; Described
  • the catalyst is preferably selected from 1,1'-bis(dicyclohexylphosphino)ferrocene palladium dichloride (PdCl 2 (dcypf)), palladium dichloride
  • the compound represented by the formula (I-c) and the compound represented by the formula (I-d) undergo reductive amination reaction in the presence of a suitable solvent and a reducing agent to obtain the compound represented by the formula (I-e), and the reducing agent is preferably selected from sodium borohydride acetate.
  • the solvents include but are not limited to dichloromethane (DCM), 1,2-dichloroethane, methanol (MeOH), ethanol (EtOH), 1,4-dioxane Hexacyclo(1,4-dioxane), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF); or,
  • the compound represented by the formula (Ic) and the compound represented by the formula (I-d') undergo a nucleophilic substitution reaction in the presence of a suitable solvent and a base to obtain the compound represented by the formula (Ie), and the base is preferably selected from triethylamine.
  • the solvent includes but is not limited to dichloromethane (DCM), 1,2-dichloroethane, methanol (MeOH), ethanol (EtOH), 1,4-dioxane (1,4-dichloroethane) dioxane), tetrahydrofuran (THF), acetonitrile (MeCN), N,N'-dimethylformamide (DMF), N-methylpyrrolidone (NMP),
  • the compound represented by formula (Ie) is deprotected by appropriate acid, base or catalytic hydrogenolysis in a suitable solvent to remove the protective group P 2 , and the obtained product is not separated and purified, and is further combined with the compound represented by formula (If) in a suitable solvent and reduction.
  • Reductive amination reaction takes place in the presence of an agent to obtain a compound represented by formula (I'), and the acid is preferably selected from trifluoroacetic acid (TFA), hydrochloric acid (HCl), acetic acid (HOAc), and hydrobromic acid (HBr);
  • the base includes but is not limited to piperidine and diethylamine;
  • the reducing agent is preferably selected from sodium borohydride acetate, sodium borohydride, sodium cyanoborohydride;
  • the solvent includes but not limited to dichloromethane (DCM).
  • the compound represented by the formula (Ie) is deprotected by appropriate acid, base or catalytic hydrogenolysis in a suitable solvent to remove the protective group P 2 , and the obtained product is not separated and purified, and is further combined with the compound represented by the formula (I-f') in a suitable solution.
  • a nucleophilic substitution reaction occurs in the presence of a solvent and a base to obtain a compound represented by formula (I'), and the base is preferably selected from triethylamine (TEA), N,N'-diisopropylethylamine (DIPEA), Pyridine (Py), n-butyllithium, lithium diisopropylamide, lithium bistrimethylsilylamide, sodium tert-butoxide (NaOBu-t), potassium tert-butoxide (KOBu-t), sodium hydride (NaH ), sodium carbonate (Na 2 CO 3 ), potassium carbonate (K 2 CO 3 ), lithium hydroxide (KOH), sodium hydroxide (NaOH);
  • the solvents include but are not limited to dichloromethane (DCM), 1, 2-Dichloroethane, methanol (MeOH), ethanol (EtOH), 1,4-dioxane (1,4-dioxane), tetrahydro
  • the compound represented by the above formula (I') is used as a form of the final product (I).
  • the compound represented by the formula (I') can be further subjected to an ester hydrolysis reaction under alkaline conditions to obtain the compound represented by the formula (I"), and the compound represented by the formula (I") is also used as a kind of the final product (I).
  • the base is preferably selected from lithium hydroxide (LiOH), lithium hydroxide (KOH), sodium hydroxide (NaOH);
  • M is preferably selected from boronic acid esters or boronic acids, including but not limited to 4,4,5,5-tetramethyl-1,3,2-dioxolaborane, neopentyl glycol diboronate, 4, 4,4',4',5,5,5',5'-Octamethyl-2,2'-bis(1,3,2-dioxaborane)B(OBu-n) 3 , B(OPr-i) 3 ; or,
  • M is preferably selected from bromine, iodine, chlorine, fluorine, CF3SO3- (OTf);
  • W is preferably selected from boronic acid esters or boronic acids, including but not limited to 4,4,5,5-tetramethyl-1,3,2-dioxaborolane, neopentyl glycol diboronate, 4, 4,4',4',5,5,5',5'-Octamethyl-2,2'-bis(1,3,2-dioxaborane)B(OBu-n) 3 , B(OPr-i) 3 ; or,
  • W is preferably selected from bromine, iodine, chlorine, fluorine, CF3SO3- ( OTf);
  • P 1 and P 2 are protecting groups, which may be the same or different, preferably selected from Boc (tert-butoxycarbonyl), Fmoc (9-fluorenemethoxycarbonyl), Cbz (N-benzyloxycarbonyl), methanesulfonyl, p- Tosyl, acetyl, methoxycarbonyl, ethoxycarbonyl, ((2-trimethylsilyl)ethoxy)methyl (SEM), tetrahydro-2H-pyran-2-yl (THP).
  • the compound represented by the formula (Ia) can also be deprotected by removing the protecting group P 1 .
  • the compound represented by (Ic); the compound represented by the formula (Ic) first undergoes the above-mentioned reductive amination reaction with the compound represented by the formula (If) to obtain the compound represented by the formula (Ih); or the compound represented by the formula (Ic)
  • the compound represented by the formula (I-f') is first subjected to the above-mentioned nucleophilic substitution reaction to obtain the compound represented by the formula (Ih), and then the compound represented by the formula (Ih) is deprotected, and then the compound represented by the formula (Id)
  • the compound is subjected to the above-mentioned reductive amination reaction to obtain the compound represented by the formula (I'); or the compound represented by the formula (I-d') undergoes the above-mentioned affinity with the compound represented by the formula (I-d') after deprotecti
  • the compound represented by the above formula (I'-1) is used as a form of the final product (I).
  • the compound represented by the formula (I') can be further subjected to an ester hydrolysis reaction under alkaline conditions to obtain the compound represented by the formula (I"-1), and the compound represented by the formula (I"-1) is also used as the final product (I ) in a form;
  • the compound represented by the formula (Ia) can also be deprotected by removing the protecting group P 1 .
  • the compound represented by (Ic); the compound represented by the formula (Ic) first undergoes the aforementioned reductive amination reaction with the compound represented by the formula (If) to obtain the compound represented by the formula (Ih); or the compound represented by the formula (Ic)
  • the compound first undergoes the aforementioned nucleophilic substitution reaction with the compound represented by the formula (I-f') to obtain the compound represented by the formula (Ih), and then the compound represented by the formula (Ih) further undergoes the aforementioned ester hydrolysis reaction to obtain
  • the compound represented by the formula (Ii), and then the compound represented by the formula (Ii) is deprotected, and then the compound represented by the formula (Id) undergoes the above-mentioned reductive amination reaction to obtain the compound represented by the formula (I"'); Or After the compound shown in (Ii) is de
  • the present invention also provides the preparation method of the used formula (I-a), comprising the following steps:
  • Formula (I-a-1) and formula (I-a-2) are subjected to an aminolysis reaction under the action of the sixth base and the seventh solvent to obtain the compound represented by the formula (I-a).
  • the sixth base is preferably selected from potassium tert-butoxide (KOBu-t), sodium tert-butoxide (NaOBu-t), sodium hydride (NaH), n-butyllithium, sodium methoxide ( MeONa), sodium ethoxide (EtONa);
  • the seventh solvent is preferably selected from anhydrous tetrahydrofuran (THF), anhydrous 1,4-dioxane (1,4-dioxane), anhydrous acetonitrile (MeCN).
  • THF tetrahydrofuran
  • MeCN acetonitrile
  • the present invention also provides the preparation method of the used formula (I-b), comprising the following steps:
  • the preparation strategy of the compound represented by the formula (I-b) is the same as the preparation strategy of the above-mentioned formula (I-a).
  • the present invention also provides the preparation method of the used formula (I-a-1), comprising the following steps:
  • the seventh base is preferably selected from potassium tert-butoxide (KOBu-t), sodium tert-butoxide (NaOBu-t), sodium hydride (NaH), n-butyllithium, sodium methoxide ( MeONa), sodium ethoxide (EtONa);
  • the compound represented by the formula (Ia-1-1) and the compound represented by the formula (Ia-1-2) are reacted with a suitable base to obtain the compound represented by the formula (Ia-1-3).
  • said base is preferably selected from potassium tert-butoxide (KOBu-t), sodium tert-butoxide (NaOBu-t), sodium hydride (NaH), n-butyllithium, sodium methoxide (MeONa), sodium ethoxide (EtONa) ); then make formula (Ia-1-3) and CO 2 (dry ice) under the action of n-butyllithium to further undergo carbonylation reaction to obtain the compound shown in (Ia-1-4); Finally, make formula (Ia- 1-4) The activated form of acid chloride is reacted with methanol (MeOH), or the compound shown in (Ia-1) is obtained under the action of methanol (MeOH)/concentrated sulfuric acid.
  • KBu-t potassium tert-butoxid
  • the present invention also provides the preparation method of the used formula (I-a-1), comprising the following steps:
  • formula (I-a-1-6) is formed into a quaternary ammonium salt to obtain the compound shown in (I-a-1-8);
  • P 3 refers to a fragment that can form a quaternary ammonium salt in order to promote the next reduction reaction, including but not limited to benzyl (Bn), p-methoxybenzyl (PMB), p-methoxycarbonylbenzyl, allyl;
  • the eighth base is preferably selected from potassium carbonate (K 2 CO 3 ), potassium tert-butoxide (KOBu-t), sodium tert-butoxide (NaOBu-t), sodium hydride (NaH), n-Butyllithium, sodium methoxide (MeONa), sodium ethoxide (EtONa).
  • the third reducing agent is preferably selected from iron powder, zinc powder, tin dichloride (SnCl 2 ), sodium sulfide (Na 2 S), hydrazine hydrate.
  • the fourth reducing agent is preferably selected from benzyl (Bn), p-methoxybenzyl (PMB), p-methoxycarbonylbenzyl, allyl.
  • the compound represented by the formula (Ia-1-1) and the compound represented by the formula (Ia-1-2) are obtained under the action of a suitable base to obtain the compound represented by the formula (Ia-1-3), wherein the The base is preferably selected from potassium carbonate (K 2 CO 3 ), potassium tert-butoxide (KOBu-t), sodium tert-butoxide (NaOBu-t), sodium hydride (NaH), n-butyllithium, sodium methoxide (MeONa ), sodium ethoxide (EtONa); then formula (Ia-1-3) reduces nitro to amino group under the action of reducing agent to obtain compound shown in (Ia-1-4), wherein said reducing agent is preferably selected from From iron powder, zinc powder, tin dichloride (SnCl 2 ), sodium sulfide (Na 2 S), hydrazine hydrate.
  • a suitable base is preferably selected from potassium carbonate (K 2 CO 3 ), potassium tert-butoxid
  • (Ia-1-4) is subjected to cyclization reaction to obtain the compound shown in (Ia-1-6), and then a quaternary ammonium salt is formed to obtain the compound shown in formula (Ia-1-8), and then in the reducing agent
  • the compound represented by formula (Ia-1-9) is obtained under the action, wherein the reducing agent is preferably selected from benzyl (Bn), p-methoxybenzyl (PMB), p-methoxycarbonylbenzyl, alkene propyl group; further, remove P 3 to obtain the compound of formula (Ia-1-10), and then undergo P 1 protection, carbonylation reaction, and esterification to obtain the compound of formula (Ia-1).
  • the preparation strategy of the formula (I-b-1) used in the present invention is the same as the preparation strategy of the above-mentioned formula (I-a-1).
  • the present invention also provides an unpublished and reported intermediate I-2a-1(I-a-1), I-1d(I-d), I-2a-2(I-a used in the preparation of the compound represented by the general formula (I) -2), I-3a-2(I-a-2), I-4a-2(I-a-2), I-5a-2(I-a-2), these intermediates include but are not limited to:
  • the present invention provides the preparation method of the above-mentioned intermediate I-1d (I-d), comprising the following steps:
  • formula (I-1d-2) is obtained by deuterated methylation reaction with deuterated methylation reagent to obtain formula (I-1d-3);
  • the fifth reducing agent is preferably selected from lithium aluminum hydride (LAH), borane (BH 3 ), NaBH 4 /AlCl 3 ;
  • said methylating reagent is preferably selected from deuterated iodomethane (CD 3 I);
  • said second acid is preferably selected from trifluoroacetic acid (TFA), hydrochloric acid (HCl), acetic acid (HOAc), hydrobromic acid (HBr);
  • the present invention provides the preparation method of the above-mentioned unknown intermediate I-2a-2 (I-a-2), comprising the following steps:
  • the bromination reagent is preferably selected from N-bromosuccinimide (NBS), sodium bromide (NaBr), potassium bromide (KBr), sodium bromate (NaBrO 3 ), potassium bromate (KBrO 3 ) );
  • the second catalyst is preferably selected from 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (Xantphos), 1,1'-bis(dicyclohexylphosphino) dimethylene Iron palladium dichloride (PdCl 2 (dcypf)), palladium acetate (Pd(OAc) 2 ), palladium dichloride (PdCl 2 ), tris(dibenzylideneacetone)dipalladium (Pd 2 (dba) 3 ) , [1,1'-bis(diphenylphosphino)ferrocene] palladium dichloride (PdCl 2 (dppf)), [1,1'-bis(diphenylphosphino)ferrocene]dichloride Palladium dichloromethane complex (PdCl 2 (dppf) ⁇ CH 2 Cl 2 ), tetrakis(triphenylphosphine) palladium (Pd(
  • said third acid is preferably selected from trifluoroacetic acid (TFA), hydrochloric acid (HCl), acetic acid (HOAc), hydrobromic acid (HBr);
  • chlorination reagent is preferably selected from N-chlorosuccinimide (NCS);
  • the sixth reducing agent is preferably selected from iron powder, zinc powder, tin dichloride (SnCl 2 ), sodium sulfide (Na 2 S), and hydrazine hydrate.
  • the present invention provides the preparation method of the above-mentioned unknown intermediate I-3a-2 (I-a-2), comprising the following steps:
  • said fluorination reagent is preferably selected from potassium fluoride, cesium fluoride;
  • the seventh reducing agent is preferably selected from iron powder, zinc powder, tin dichloride (SnCl 2 ), sodium sulfide (Na 2 S), and hydrazine hydrate.
  • the present invention provides the preparation method of the above-mentioned unknown intermediate I-4a-2 (I-a-2), comprising the following steps:
  • the eighth reducing agent is preferably selected from iron powder, zinc powder, tin dichloride (SnCl 2 ), sodium sulfide (Na 2 S), hydrazine hydrate;
  • the third catalyst is preferably selected from 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (Xantphos), 1,1'-bis(dicyclohexylphosphino) dimethylene Iron palladium dichloride (PdCl 2 (dcypf)), palladium acetate (Pd(OAc) 2 ), palladium dichloride (PdCl 2 ), tris(dibenzylideneacetone)dipalladium (Pd 2 (dba) 3 ) , [1,1'-bis(diphenylphosphino)ferrocene] palladium dichloride (PdCl 2 (dppf)), [1,1'-bis(diphenylphosphino)ferrocene]dichloride Palladium dichloromethane complex (PdCl 2 (dppf) ⁇ CH 2 Cl 2 ), tetrakis(triphenylphosphine) palladium (Pd(
  • the fourth acid is preferably selected from trifluoroacetic acid (TFA), hydrochloric acid (HCl), acetic acid (HOAc), and hydrobromic acid (HBr).
  • TFA trifluoroacetic acid
  • HCl hydrochloric acid
  • HOAc acetic acid
  • HBr hydrobromic acid
  • the present invention provides the aforementioned compounds or their stereoisomers, pharmaceutically acceptable salts, precursors or metabolites or the aforementioned pharmaceutical compositions in preparation for treatment and/or prevention related to the target PD-L1 use in medicine for a disease, or
  • the disease associated with the target PD-L1 includes tumor, cancer, or other immune-related disease.
  • the present invention provides the aforementioned compounds or stereoisomers, pharmaceutically acceptable salts, precursors or metabolites thereof, or the aforementioned pharmaceutical compositions for use in the treatment and/or prevention of and target PD - L1-related disease, or
  • the disease associated with the target PD-L1 includes tumor, cancer, or other immune-related disease.
  • the present invention provides a method of treating and/or preventing a disease associated with the target PD-L1, comprising administering to a subject in need thereof a therapeutically and/or prophylactically effective amount of the aforementioned compound or a stereoisomer thereof Conforms, pharmaceutically acceptable salts, precursors or metabolites or pharmaceutical compositions of the foregoing.
  • the present invention also provides a method of inhibiting PD-L1 activity, comprising administering to a cell (eg, a mammalian cell) an effective amount of the foregoing compound or a stereoisomer, pharmaceutically acceptable salt, precursor or metabolite or the foregoing pharmaceutical composition.
  • a cell eg, a mammalian cell
  • the disease associated with targeting PD-L1 includes tumor, cancer, or other immune-related disease.
  • vertebrate refers to a mammal.
  • the mammals include bovines, equines, ovines, porcines, canines, felines, rodents, primates, such as humans, cats, dogs or pigs. Mammals include, but are not limited to, livestock (such as cattle), pets (such as cats, dogs, and horses), primates, mice, and rats. In certain embodiments, the mammal refers to a human.
  • the term "therapeutically effective amount” or “prophylactically effective amount” refers to an amount sufficient to treat or prevent a patient's disease but low enough to avoid serious side effects (at a reasonable benefit/risk ratio) within the scope of sound medical judgment.
  • a therapeutically effective amount of a compound will depend on the particular compound selected (eg, taking into account the potency, effectiveness and half-life of the compound), the route of administration selected, the disease being treated, the severity of the disease being treated, the severity of the patient being treated. Factors such as age, size, weight and physical ailment, medical history of the patient being treated, duration of treatment, nature of concurrent therapy, desired therapeutic effect, and the like vary, but can still be routinely determined by those skilled in the art.
  • the specific dosage and method of use of the compound or its stereoisomer, pharmaceutically acceptable salt, precursor or metabolite for different patients depends on many factors, including the patient's age, weight, sex, Natural health status, nutritional status, active strength of the drug, time of administration, metabolic rate, severity of the condition, and the subjective judgment of the treating physician. It is preferred here to use doses between 0.001 and 1000 mg/kg body weight/day.
  • the present invention provides a novel biphenyl-based small-molecule immunosuppressant with excellent orally absorbable characteristics for treating or preventing immune-related diseases.
  • these compounds or pharmaceutical compositions containing them as active ingredients can maximize the clinical efficacy on these diseases within a safe therapeutic window.
  • Figure 1 shows the tissue distribution of Compound I-1 (50 mg/kg, PO) 18 days after administration to hPD1/hPD-L1 mice.
  • FIG. 1 shows the tissue distribution of Compound 1-1, Example 17, Compound 14 (INCB086550) and Example 180 relative to plasma in mice.
  • Mass spectrometry conditions instrument Thermo MSQ Plus; ion source ESI (EA+EA-); cone voltage 30V; capillary voltage 3.00KV; source temperature 350°C;
  • Chromatographic conditions instrument Thermo U3000; detector DAD-3000 (RS) (diode array detector); chromatographic column Shimadzu Inertsil ODS-HL HP 3 ⁇ m 3.0 ⁇ 100mm; flow rate 0.4mL/min; column temperature 30°C; mobile phase CH 3OH/ H2O /HCOOH (75/ 25 /0.2).
  • the raw materials in the following examples can be obtained commercially unless otherwise specified, for example, they can be purchased from Shanghai Bide Pharmaceutical Technology Co., Ltd., Jiangsu Aikang Biomedical Research and Development Co., Ltd., Nanjing Yaoshi Technology Co., Ltd., Shanghai Shaoyuan Reagent Co., Ltd., Hechun Biotechnology (Shanghai) Co., Ltd.
  • I-2a-1-1 (10.00 g, 44.8 mmol, 1.0 eq), NaH (2.69 g, 112.0 mmol, 2.5 eq), THF (40 mL) were stirred at 0 °C for 0.5 h, and I-2a was added -1-2 (10.00 g, 69.0 mmol, 1.5 eq) continued to stir for 16 h.
  • the reaction solution was diluted with ethyl acetate (EtOAc, 60 mL), and washed once with saturated brine.
  • I-2a-1-9 (16.20g, 47.0mmol, 1.0eq) was dissolved in methanol (100mL), ammonium formate (29.64g, 470.0mmol, 10.0eq) and palladium hydroxide (20wt%, 3.24g) were added, The reaction solution was heated to 80°C for 4 h, cooled to room temperature, filtered with suction, and the filtrate was concentrated to obtain a transparent viscous oil I-2a-1-10, which was directly used in the next reaction. (14.50 g, the yield is 100%). LC-MS MS-ESI (m/z) 141.1 [M+H] + .
  • I-2a-1-12 (4.50g, 15.8mmol, 1.0eq) was dissolved in DCM (80mL), the reaction solution was cooled with an ice bath, DMF (0.1mL) was added, oxalyl chloride (4.01g, 31.6mmol, oxalyl chloride) was added dropwise. 2.0eq), after the dropwise addition was completed, continue stirring at room temperature for 30min, then cool to below zero in an ice-salt bath, add methanol (10mL) to the reaction solution, then add TEA (5mL), stir for 10min, dilute with water, separate the liquid, and concentrate the organic phase.
  • reaction solution was extracted with EtOAc (3000 mL x 2), and the organic phases were combined and washed once with saturated brine. The organic phase was dried over anhydrous Na 2 SO 4 and concentrated to give I-2a-2-2 as a tan solid. (36.50 g, 90.8% yield).
  • I-2a-2-2 (36.50g, 177.0mmol, 1.0eq), Xantphos (10.30g, 17.7mmol, 0.1eq), Cs2CO3 (115.00g, 354.0mmol, 2.0eq ), Pd2 ( dpa ) 3 (9.73 g, 10.6 mmol, 0.06 eq), commercially available I-2a-2-3 (24.90 g, 213.0 mmol, 1.2 eq) was dissolved in toluene (60 mL) and stirred at 100 °C for 16 h.
  • I-2a-2-5 (18.00 g, 127.0 mmol, 1.0 eq) was added to a solution of N-chlorosuccinimide (NCS, 16.9 g, 127.0 mmol, 1.0 eq) in DMF (50 mL), the The reaction solution was stirred at 75°C for 1 h. The completion of the reaction was monitored by TLC, the reaction was quenched with water, and then the reaction solution was extracted with EtOAc (100 mL), and the organic phases were combined and washed with saturated brine.
  • N-chlorosuccinimide N-chlorosuccinimide
  • tert-butyl nitrite (12.8 g, 124.0 mmol, 1.5 eq) was added to a solution of CuBr2 (22.2 g, 99.2 mmol, 1.2 eq) in acetonitrile (MeCN, 100 mL), then the mixture was warmed To 65°C, a solution of home-made I-2a-2-6 (14.5 g, 82.6 mmol, 1.0 eq) in MeCN (50 mL) was added, and the reaction solution was continued to stir at this temperature for 1 h.
  • the white solid I-14a was prepared from commercially available I-1a-1 (788.32 mg, 2.67 mmol, 1.0 eq) and previously prepared I-2a-2 (560.00 mg, 2.67 mmol, 1.0 eq) and KOBu-t (593.91 mg) , 5.53 mmol, 2.0 eq) was prepared following a similar procedure in Intermediate I-3a. (780.00 mg, 61.9% yield).
  • I-4a-2-3 (7.00g, 27.6mmol, 1.0eq), Xantphos (1.60g, 2.77mmol, 0.1eq), Cs2CO3 (18.00g, 55.3mmol, 2.0eq ), Pd2 ( dpa ) 3 (1.52 g, 1.7 mmol, 0.06 eq) and commercially available I-2a-2-3 were dissolved in toluene (60 mL) and stirred at 100 °C for 16 h. HPLC monitored the completion of the reaction, the reaction solution was cooled to 25° C., diluted with water, and then extracted with EtOAc (100 mL ⁇ 2), the organic phases were combined and washed once with saturated brine.
  • the white solid intermediate I-7b was obtained from the commercially available I-7b-2 (1.00 g, 4.20 mmol, 1.0 eq) and the previously prepared intermediate I-2a-1 (1.24 g, 4.20 mmol, 1.0 eq), KOBu -t (935.71 mg, 8.40 mmol, 2.0 eq) was prepared following a similar procedure as in Intermediate 1-3b. (1.00 g, 47.2% yield). LC-MS MS-ESI (m/z) 504.3 [M+H] + .
  • the white solid intermediate I-9b was obtained from the commercially available I-9b-2 (1.50 g, 6.44 mmol, 1.0 eq) and the previously prepared intermediate I-2a-1 (1.90 g, 4.44 mmol, 1.0 eq), KOBu -t (1.44 g, 12.88 mmol, 2.0 eq) was prepared following a similar procedure as in Intermediate 1-3b. (1.80 g, 56.0% yield). LC-MS MS-ESI (m/z) 500.3 [M+H] + .
  • White solid intermediate I-14b-2 is I-3a-2 (1.00 g, 5.2 mmol, 1.0 eq) prepared previously, pinacol biboronate (1.97 g, 7.77 mmol, 1.5 eq), Pd (dppf )Cl 2 ⁇ CH 2 Cl 2 (409.11 mg, 0.5 mmol, 0.1 eq), anhydrous potassium acetate (1.52 g, 30.2 mmol, 3.0 eq) Prepared following a similar procedure as in Intermediate I-3b-2. (880.00 mg, 70.5% yield). LC-MS MS-ESI (m/z) 241.2 [M+H] + .
  • I-1d-1 (300.0 g, 1.31 mol, 1.0 eq) was dissolved in THF (2.5 L), the reaction solution was cooled to 0 °C under nitrogen protection, and BH 3 -Me 2 S (1.57 mol, 157.0 mL, 1.2 eq). After dripping, the reaction solution was naturally heated to 20° C. and continued to be stirred for 16 h. TLC showed that the reaction was complete. The reaction solution was cooled to 0°C, and MeOH (500 mL) was added dropwise to quench the reaction. After dripping, the reaction solution was directly concentrated to dryness to obtain a colorless oily substance I-1d-2. (280.00 g, yield 99.4%).
  • I-1d-2 (20.00g, 93.3mmol, 1.0eq) was dissolved in DMF (150mL), silver oxide (Ag2O, 64.88g, 280.0mmol, 3.0eq) and deuterated iodomethane (CD 3 I, 50.00 were added) g, 344.9 mmol, 3.7 eq), the addition was completed, covered with a balloon to prevent CD 3 I from escaping, and stirred at 20° C. for 32 h. Gas phase monitoring raw material reaction has 30% remaining, which can be directly processed.
  • the reaction solution was filtered to remove insolubles, and water (150 mL) was added to the filtrate, extracted with methyl tert-butyl ether (MTBE, 300 mL ⁇ 2), washed with saturated brine (100 mL ⁇ 3), dried over anhydrous magnesium sulfate, filtered, and the filtrate was desolvated under reduced pressure.
  • MTBE methyl tert-butyl ether
  • saturated brine 100 mL ⁇ 3
  • I-2d-1 (300.00g, 1.31mol, 1.0eq) was dissolved in THF (2.5L), the reaction solution was cooled to 0°C under nitrogen protection, and the dimethyl sulfide complex of borane was added dropwise Compound (1.57mol, 157mL, 1.2eq). After dripping, the reaction solution was naturally heated to 20° C. and continued to be stirred for 16 h. TLC showed that the reaction was complete. The reaction solution was cooled to 0°C, and MeOH (500 mL) was added dropwise to quench the reaction. After dripping, the reaction solution was directly concentrated to dryness to obtain a colorless oily substance I-2d-2 (280.00 g, yield 99.4%).
  • I-2d-2 (5.00g, 23.33mmol, 1.0eq) was dissolved in N,N'-dimethylformamide (50mL), iodomethane (33.12g, 233.30mmol, 10.0eq) and Ag 2 were added sequentially O (16.22 g, 70.00 mmol, 3.0 eq).
  • the reaction solution was stirred at 20°C for 16h. TLC showed that a small amount of starting material remained, and a major spot was formed.
  • the reaction solution was poured into water (50 mL) and extracted with MTBE (50 mL ⁇ 3). The organic phases were combined, washed with saturated brine (30 mL ⁇ 3), and dried over anhydrous magnesium sulfate.
  • I-2d-3 (4.50 g, 19.71 mmol, 1.0 eq) was dissolved in Acetone (40 mL) and 2M hydrochloric acid (29.56 mL, 3.0 eq) was added. The reaction solution was stirred at 25°C for 4h. TLC showed that the reaction was complete. Concentrate to remove acetone, add water (30 mL), and extract with MTBE (20 mL ⁇ 3). The organic phases were combined, washed with saturated brine (30 mL), and dried over anhydrous magnesium sulfate.
  • the pale yellow solid I-2 is obtained from the intermediate 4-(2-(2-((3'-(5-(tert-butoxycarbonyl)-1-methyl-4,5,6,7-tetrahydro-1H -Imidazo[4,5-c]pyridine-2-carboxamido)-2-chloro-2'-fluoro-[1,1'-biphenyl]-3-yl)carbamoyl)-1- Methyl-1,4,6,7-tetrahydro-5H-imidazo[4,5-c]pyridin-5-yl)ethyl)bicyclo[2.2.1]heptane-1-carboxylic acid I-2i (400.00 mg, 0.48 mmol, 1.0 eq, the synthesis refers to the preparation method of intermediate I-1i), TFA (5 mL), TEA (1 mL), 2-(4-((methoxy-d 3 )methyl)bicyclo [2.2.1] Heptan-1-yl)acetaldehyde I-1d
  • the pale yellow solid I-3 is obtained from the intermediate 4-(2-(2-((3'-(5-(tert-butoxycarbonyl)-1-methyl-4,5,6,7-tetrahydro-1H -Imidazo[4,5-c]pyridine-2-carboxamido)-2'-chloro-2-methyl-[1,1'-biphenyl]-3-yl)carbamoyl)-1 -Methyl-1,4,6,7-tetrahydro-5H-imidazo[4,5-c]pyridin-5-yl)ethyl)bicyclo[2.2.1]heptane-1-carboxylic acid I- 3i (400.00 mg, 0.48 mmol, 1.0 eq, synthetic reference to the preparation method of intermediate I-1i), TFA (5 mL), TEA (1 mL), 2-(4-((methoxy-d 3 )methyl) Bicyclo[2.2.1]heptan-1-yl)acetaldehyde I-1d (133.2
  • the pale yellow solid I-4 is obtained from the intermediate 4-(2-(2-((3'-(5-(tert-butoxycarbonyl)-1-methyl-4,5,6,7-tetrahydro-1H -Imidazo[4,5-c]pyridine-2-carboxamido)-2-chloro-2'-methyl-[1,1'-biphenyl]-3-yl)carbamoyl)-1 -Methyl-1,4,6,7-tetrahydro-5H-imidazo[4,5-c]pyridin-5-yl)ethyl)bicyclo[2.2.1]heptane-1-carboxylic acid I- 4i (400.00 mg, 0.48 mmol, 1.0 eq, synthetic reference to the preparation of intermediate I-1i), TFA (5 mL), TEA (1 mL), 2-(4-((methoxy-d 3 )methyl) Bicyclo[2.2.1]heptan-1-yl)acetaldehyde I-1d (133.2 mg
  • the yellow intermediate I-7i was prepared from intermediate I-7h (600.00 mg, 0.71 mmol, 1.0 eq) and LiOH ⁇ H 2 O (314.77 mg, 7.10 mmol)) following a similar procedure as in intermediate I-2i . (450.00 mg, 75.9% yield).
  • White compound 1-7 is composed of intermediate 1-7i (225 mg, 0.27 mmol, 1.0 eq), TFA (5 mL), TEA (1 mL) and 2-(4-((methoxy-d 3 )methyl)bicycle [2.2.1] Heptane-1-yl)acetaldehyde I-1d (147.15 mg, 0.54 mmol, 2.0 eq), NaBH(OAc) 3 (290.88 mg, 1.35 mmol, 5.0 eq) according to compound 1-5 Prepared in similar steps. (120.00 mg, 49.1% yield). LC-MS MS-ESI (m/z) 904.5 [M+H] + .
  • Yellow solid intermediate I-9c is composed of I-2a (1.34g, 3.60mmol, 1.0eq), intermediate I-9b (1.80g, 3.60mmol, 1.0eq), PdCl 2 (dcypf) (271.40mg, 0.36mmol , 0.1 eq), anhydrous Na 2 CO 3 (1.72 g, 16.2 mmol, 4.5 eq) was prepared following a similar procedure in Intermediate I-1c. (1.40 g, 58.5% yield). LC-MS MS-ESI (m/z) 665.3 [M+H] + .
  • the yellow intermediate I-9h is composed of intermediate I-9c (700.00 mg, 1.05 mmol, 1.0 eq) and 4-(2-oxoethyl)bicyclo[2.2.1]heptane-1-carboxylate methyl ester I -1f (414.70 mg, 2.10 mmol, 2.0 eq), TEA (1 mL), NaBH(OAc) 3 (1.12 g, 5.3 mmol, 5.0 eq) Prepared following similar procedures in Intermediate 1-1h. (650.00 mg, 73.2% yield). LC-MS MS-ESI (m/z) 845.4 [M+H] + .
  • Yellow intermediate 1-9i was prepared from intermediate 1-9h (650.00 mg, 0.77 mmol, 1.0 eq) and LiOH ⁇ H 2 O (340.86 mg, 7.70 mmol) following a similar procedure as in intermediate 1-2i. (460.00 mg, 71.9% yield).
  • White compound 1-9 was prepared from intermediate 1-9i (230.00 mg, 0.28 mmol, 1.0 eq), TFA (5 mL), TEA (1 mL) and 2-(4-((methoxy-d 3 )methyl) Bicyclo[2.2.1]heptan-1-yl)acetaldehyde I-1d (153.35mg, 0.56mmol, 2.0eq), NaBH(OAc) 3 (310.28mg, 1.40mmol, 5.0eq) according to compound I-5 prepared by similar steps. (125.00 mg, 49.6% yield). LC-MS MS-ESI (m/z) 900.5 [M+H] + .
  • the white compound I-10 was obtained from intermediate I-9i (230.00 mg, 0.28 mmol, 1.0 eq), TFA (5 mL), TEA (1 mL) and 2-(4-((methoxy)methyl)bicyclo[2.2 .1] Heptan-1-yl)acetaldehyde I-2d (151.12 mg, 0.56 mmol, 2.0 eq), NaBH(OAc) 3 (310.28 mg, 1.40 mmol, 5.0 eq) following similar steps in compound 1-5 prepared. (124.00 mg, 49.3% yield). LC-MS MS-ESI (m/z) 897.5 [M+H] + .
  • the yellow intermediate I-14c is composed of intermediate I-14a (780.0 mg, 1.65 mmol, 1.0 eq), intermediate I-14b (766.78 mg, 1.52 mmol, 1.0 eq), PdCl 2 (dcypf) (114.62 mg, 0.15 mmol, 0.1 eq), anhydrous Na 2 CO 3 (724.00 mg, 6.8 mmol, 4.5 eq) was prepared following a similar procedure in Intermediate I-1c. (625.65 mg, 59.2% yield). LC-MS MS-ESI (m/z) 669.3 [M+H] + .
  • the yellow intermediate I-14h was obtained from intermediate I-14c (625.65 mg, 0.94 mmol, 1.0 eq) and commercially available 4-(2-oxoethyl)bicyclo[2.2.1]heptane-1-carboxylic acid Methyl ester 1-1f (370.80 mg, 1.88 mmol, 2.0 eq), TEA (1 mL), NaBH(OAc) 3 (996.7 mg, 4.7 mmol, 5.0 eq) were prepared following similar procedures as in intermediate 1-1h. (601.08 mg, 75.1% yield). LC-MS MS-ESI (m/z) 849.4 [M+H] + .
  • Yellow intermediate 1-14i was prepared from intermediate 1-9h (601.08 mg, 0.71 mmol, 1.0 eq) and LiOH ⁇ H 2 O (315.86 mg, 7.10 mmol) following a similar procedure as in intermediate 1-2i. (451.32 mg, 75.9% yield).
  • White compound 1-14 was prepared from intermediate 1-14i (225.00 mg, 0.27 mmol, 1.0 eq), TFA (5 mL), TEA (1 mL) and 2-(4-((methoxy-d 3 )methyl) Bicyclo[2.2.1]heptan-1-yl)acetaldehyde I-1d (147.15mg, 0.54mmol, 2.0eq), NaBH(OAc) 3 (290.88mg, 1.35mmol, 5.0eq) according to compound I-5 prepared by similar steps. (60.00 mg, 24.6% yield). LC-MS MS-ESI (m/z) 904.5 [M+H] + .
  • the white compound I-15 is composed of intermediate I-14i (225.00 mg, 0.27 mmol, 1.0 eq), TFA (5 mL), TEA (1 mL) and 2-(4-(methoxymethyl)bicyclo[2.2.1] Heptan-1-yl)acetaldehyde I-2d (143.05 mg, 0.54 mmol, 2 eq), NaBH(OAc) 3 (290.88 mg, 1.35 mmol, 5.0 eq) were prepared following similar procedures to compound 1-5. (60.00 mg, 25.0% yield). LC-MS MS-ESI (m/z) 901.5 [M+H] + .
  • the detection method is used for the evaluation of the in vitro biological activity of the compounds of the present invention, including the in vitro protein level binding inhibitory activity evaluation method and the cell level biological function activity evaluation method.
  • the purpose of this test is to comprehensively evaluate the inhibitory activities of different compounds on the binding of PD-1 and PD-L1 and CD80 and PD-L1 in liquid phase in vitro and the inhibition of T cell activation signals after the binding of PD-1 and PD-L1 in cell models. Block the impact.
  • HTRF Homogeneous Time-Resolved Fluorescence
  • His-tagged recombinant human PD-1 protein His-PD-1 protein, Cat#: 10377-H08H-50
  • recombinant human PD-L1-Fc fusion protein PD-L1-Fc fusion protein, Cat#: 10084 -H02H-100
  • Sino Biological Inc. anti-hFc-Eu 3+ antibody and anti-His-XL665 antibody were purchased from Cisbio Company
  • other related reagents such as dilution buffer (Diluent buffer 5, Cat#: 62DL5DDC), detection buffer (PPI-Europium detection buffer, Cat#: 61DB9RDF), etc. were purchased from Cisbio.
  • the fluorescence detection instrument Tecan (Spark 10M) was purchased from Swiss Tecan Company.
  • a control group is also set for the detection reaction, including a 0% inhibition positive control without the addition of the test compound, and a 100% inhibition negative control without the addition of PD-1 protein. Duplicate wells were used for all assays.
  • inhibition rate (%) [1-(fluorescence signal ratio of detection well-100% inhibition of negative control fluorescence signal ratio)]/(0% inhibition of positive control fluorescence signal ratio-100 % inhibition of negative control fluorescence signal ratio) ⁇ 100%.
  • the 50% inhibitory concentration (IC 50 ) was calculated after the inhibition rate of PD-1/PD-L1 binding was calculated for the test compounds with different concentration gradients respectively.
  • the IC50 data of the representative compounds of the present invention for inhibiting the binding of PD-1 and PD-L1 in vitro are shown in Table 2 below:
  • the compound of the present invention has a good activity of inhibiting PD-1/PD-L1 in vitro, and the compound of the general formula (I) of the present invention also has the ability to inhibit PD-1/PD-L1. active.
  • PD-L1 can also exert immunosuppressive activity by binding to CD80.
  • CD80 binding to PD-L1 or binding inhibition experiments can also be detected by homogeneous time-resolved fluorescence (HTRF).
  • HTRF homogeneous time-resolved fluorescence
  • anti-hFc-Eu 3+ antibody and anti-His-XL665 antibody were used to bind the hFC tag fused to PD-L1 and the His tag fused to CD80, respectively, and excited by 320nm wavelength laser, due to PD-L1 and The binding of CD80 enables energy transfer from Eu to XL665 fluorescein, which excites the latter to emit light.
  • an inhibitor of the interaction between PD-L1 and CD80 is added, the combination of the two is destroyed, so that Eu and XL665 are far away, energy cannot be transferred, and XL665 will not be excited.
  • His-tagged recombinant human CD80 protein (His-CD80 protein, Cat#: 10698-H08H-100), recombinant human PD-L1-Fc fusion protein (PD-L1-Fc fusion protein, Cat#: 10084-H02H-100 ) were purchased from Sino Biological Inc., anti-hFc-Eu 3+ antibody and anti-His-XL665 antibody were purchased from Cisbio Company, and other related reagents such as dilution buffer (Diluent buffer 5, Cat#: 62DL5DDC) ), detection buffer (PPI-Europium detection buffer, Cat#: 61DB9RDF), etc. were purchased from Cisbio Company.
  • the fluorescence detection instrument Tecan (Spark 10M) was purchased from Swiss Tecan Company.
  • the experimental process was carried out according to the procedure required by the instruction manual of the detection reagent (Invitrogen). The process is as follows:
  • test compound 5 ⁇ L of the test compound, 2.5 ⁇ L of His-CD80 protein and 2.5 ⁇ L of PD-1-Fc fusion protein solution were mixed and incubated at room temperature for 15 min; then 5 ⁇ L of anti-His-XL665 antibody and 5 ⁇ L of anti-His-XL665 antibody were added to the system. hFc-Eu 3+ antibody was incubated for 3h and then detected.
  • a control group is also set for the detection reaction, including a 0% inhibition positive control without the addition of the test compound, and a 100% inhibition negative control without the addition of CD80 protein. Duplicate wells were used for all assays.
  • inhibition rate (%) [1-(fluorescence signal ratio of detection well-100% inhibition of negative control fluorescence signal ratio)]/(0% inhibition of positive control fluorescence signal ratio-100 % inhibition of negative control fluorescence signal ratio) ⁇ 100%.
  • the 50% inhibitory concentration (IC 50 ) was calculated after calculating the CD80/PD-L1 binding inhibition rate of the test compounds with different concentration gradients.
  • the IC50 data of the representative compounds of the present invention for inhibiting the binding of CD80 and PD-L1 in vitro are shown in Table 3:
  • the compound of the present invention has a good inhibitory activity of CD80/PD-L1 in vitro, and the compound of the general formula (I) of the present invention also has an activity of inhibiting CD80/PD-L1.
  • PD-1 is mainly expressed on the surface of activated T cells, while its ligand PD-L1 is widely expressed.
  • antigen-presenting cells such as dendritic cells, macrophages, and B cells
  • many tumor cells can also suppress anti-tumor immune effects by upregulating the expression of PD-L1.
  • antigen-presenting cells also express PD-L1 ligand molecules, etc., which bind to PD-1 molecules on the surface of activated T cells, thereby inhibiting T cell activation and avoiding It can cause damage to surrounding normal tissues due to the excessive proliferation and activation of T cells.
  • CHO-PD-L1-CD3L cells stably expressing human PD-L1 molecule and anti-CD3 single-chain antibody (ScFv) and stable Jurkat-PD-1-NFAT cells expressing human PD-1 molecule and NFAT reporter gene.
  • the anti-CD3ScFv on the CHO cell surface binds to the membrane CD3 molecule of Jurkat cells, which will transmit an activation signal into Jurkat cells, but due to the presence of PD-L1 on the CHO cell surface and PD-1 molecules on the Jurkat cell surface at the same time Binding and inward transmission of inhibitory activation signals, so that the luciferase reporter gene can not be expressed.
  • CHO-PD-L1-CD3L cells expressing human PD-L1 molecule and anti-CD3 single-chain antibody (ScFv) and Jurkat-PD-1-NFAT cells stably expressing human PD-1 molecule and NFAT reporter gene were developed by Dr. Bo Chen (Connoya Biomedical Technology (Chengdu) Co., Ltd.) independently constructed and presented it.
  • Puromycin (Cat#540411) and Hygromycin B (Cat#V900372) for stable culture of transfected cells were purchased from Sigma, PMA (Cat#P1585) was purchased from Sigma Company, anti-human PD-L1 antibody (Cat#GMP-A066) Purchased from Novoprotein.
  • the luciferase substrate solution (Cat#E6485) and the luciferase-specific cell lysate 5 ⁇ (Cat#E1531) were purchased from Promega.
  • the fluorescence detection instrument Tecan (Spark 10M) was purchased from Tecan Company, Switzerland.
  • a control group is also set for the detection reaction, including a solvent control without the addition of the test compound and an anti-human PD-1 antibody added as a positive control in the experimental system. All tests use duplicate holes;
  • Inhibition rate of T cell activation signal (%) (original value of chemiluminescence in the detection well - solvent control)/(the highest measured in the detection well of the compound) Chemiluminescence raw value - solvent control) ⁇ 100%.
  • the 50% inhibitory concentration (EC 50 ) was further calculated after the inhibition rates of T cell activation signals were calculated for the test compounds with different concentration gradients.
  • the EC 50 data of the compounds of the present invention blocking PD-1 and PD-L1-mediated T cell activation inhibitory signals are shown in Table 4:
  • Table 4 EC 50 of the compounds of the present invention blocking PD-1/PD-L1-mediated inhibitory signal of T cell activation
  • the compound of the present invention has the activity of effectively blocking the inhibitory signal of T cell activation mediated by immune checkpoints at the cellular level, and the compound of the general formula (I) of the present invention also has the ability to block PD- 1/PD-L1-mediated T cell activation signaling inhibitory activity.
  • mice Twenty-four 6-8-week-old B-hPD-1/hPD-L1mice female mice were purchased from Biositu Jiangsu Gene Biotechnology Co., Ltd. Randomly divided into 4 groups, 6 mice in each group, the test compounds were formulated in a vehicle containing 5% DMSO, 60% PEG400 and 35% purified water, including I-1 and 3 control molecules Example 17, compound 14 (INCB086550) , and Example 180, the compound was orally administered at 50 mg/kg, once a day, and blood was collected alternately through the fundus venous plexus after administration on the 18th day, and the blood collection time points were 15min, 30min, 1h, 2h, 4h, 8h, 24h and 32h.
  • Example 17 is a compound disclosed by Incyte on page 64 of the patent WO2019/217821 document, and the inventors synthesized the compound with reference to the synthetic method therein as a reference molecule.
  • the chemical structure of Example 17 was confirmed by LC-MS MS-ESI(m/z) 911.4[M+H] + .
  • sodium hydroxide solution (Example 17) was added to the NMR sample.
  • the molar ratio to sodium hydroxide was 1 to 2), to which MeOD was subsequently added for structural confirmation.
  • Compound 14 is the compound disclosed in Table 2 of the patent CN110267953A by Incyte Company, and the inventor synthesized the compound with reference to the synthetic method therein, which was used as a reference molecule.
  • Compound 14 (INCB086550) is a small molecule PD-L1 inhibitor with the fastest clinical progress and is currently in phase 2 research.
  • Example 180 is an example disclosed in patent CN110267953A by Incyte Company, and the inventor synthesized this compound with reference to the synthetic method therein to serve as a reference molecule.
  • the test compounds were formulated in a vehicle containing 5% DMSO, 60% PEG400 and 35% purified water, including I-1 and 3 control molecules
  • the concentration of the representative compounds of the present invention in tumor tissue is significantly higher than that in plasma, that is, the concentration of tumor tissue at 4h and 24h is 4-6 times and 17-27 times that of plasma.
  • the tumor tissue concentration at 4h and 24h was 5-8 times and 7-30 times that of the control molecule, indicating that the compound of the present invention showed good tumor tissue targeting.
  • Compounds of the invention have unexpected enrichment and targeting effects on tumor tissue compared to control molecules.
  • the biphenyl compounds of the present invention have excellent PD-L1 inhibitory activity, and can be used as medicines for treating or preventing diseases related to this effect.

Abstract

一种式(I)所示联苯类化合物、其制备方法及其应用,以及包含所述化合物作为活性成分的药物组合物。所述化合物是具有优异的可口服吸收特征的新型小分子免疫调节剂,可用于治疗和/或预防与免疫相关的多种疾病。

Description

作为免疫调节剂的联苯类化合物及其制备方法和应用
本申请要求于2021年2月1日提交到中国国家知识产权局的发明名称为“作为免疫调节剂的联苯类化合物及其制备方法和应用”的中国专利申请202110137428.0的优先权,其内容通过引用以整体并入本文。
技术领域
本发明属于药物化学领域,具体涉及作为免疫调节剂的联苯类化合物及其制备方法和应用。
背景技术
肿瘤免疫治疗是一种通过激发人体的免疫***,增强自身的抗肿瘤免疫力,从而抑制或杀死肿瘤细胞的新治疗方法。该方法经过百余年的努力取得了突破性进展。2013年,《Science》杂志将肿瘤免疫治疗列为年度十大科学突破之首(Couzin-Frankel J.,2013,Science,342:1432-1433),已成为最具前景的抗肿瘤治疗领域之一。
肿瘤细胞相比正常细胞,具有多种遗传学和表观遗传学的改变,免疫***可利用肿瘤细胞产生的表面抗原将二者区分,进而引发抗肿瘤免疫反应。在T细胞抗肿瘤免疫过程中,其被T细胞受体(T cell receptor,TCR)介导的抗原识别信号激活后,通过共刺激和共抑制信号综合调节T细胞效应,包括细胞毒性T淋巴细胞相关抗原4(Cytotoxic T-lymphocyte associated antigen 4,CTLA4)、程序性死亡受体1(Programmed death protein 1,PD-1)、T细胞活化的免疫球蛋白抑制V型结构域(V-domain immunoglobulin suppressor of T-cell activation,VISTA)、T细胞免疫球蛋白及黏蛋白结构域的分子3(T cell immunoglobulin and mucin domain–containing-3,TIM3)、淋巴细胞活化基因3(Lymphocyte activation gene 3,LAG3)等抑制信号的抑制性受体,及CD28、CD134(OX40)、糖皮质激素诱导的TNFR相关蛋白(Glucocorticoid-induced TNFR-related protein,GITR)、CD137、CD27、HVEM等刺激信号的活化性受体(Mellman I.,Coukos G.,Dranoff G.,2011,Nature,480:480-489)。在正常生理条件下,免疫检查点一方面参与维持自身抗原的免疫耐受,避免自身免疫性疾病;另一方面避免免疫反应过度激活导致组织损伤。然而,在肿瘤细胞中,其可通过免疫检查点抑制T细胞激活而逃避免疫杀伤。因此,需要通过激活共刺激信号(踩“油门”)并抑制共抑制信号(松“刹车”)而重新激活T细胞攻击肿瘤细胞,进而实现肿瘤免疫治疗。
PD-1表达于激活的T细胞、B细胞及骨髓细胞中,属于CD28家族,是T 细胞上的一种1型跨膜糖蛋白,由288个氨基酸组成。PD-1的分子结构由具有免疫球蛋白IgV样(氨基酸35-145)的胞外区、跨膜区、具有连接信号肽功能的胞质尾区构成,其上的胞外区与配体结合发挥重要功能(Cheng X.,Veverka V.,Radhakrishnan A.,et al.2013,J.Biol.Chem.,288:11771-11785)。程序性死亡配体1(Programmed death protein ligand 1,PD-L1)是PD-1的配体之一,属于B7家族,可持续性表达于多种肿瘤细胞、T细胞、抗原呈递细胞(APC)及多种非造血细胞中,也为1型跨膜糖蛋白,它由290个氨基酸组成。PD-1与PD-L1相互作用会抑制T细胞激活,这对于维持正常机体的免疫耐受至关重要,而在肿瘤细胞中和病毒感染时,T细胞上的PD-1被诱导性高表达,PD-L1的表达上调,导致PD-1信号通路持续激活而抑制T细胞增殖,造成肿瘤细胞和病原体的免疫逃逸(Fuller M.J.,Callendret B.,Zhu B.,et al.2013,Proc.Natl.Acad.Sci.USA.,110:15001-15006;Dolan D.E.,Gupta S.,2014,Cancer Control,21:231-237;Chen L.,Han X.,2015,J.Clin.Invest.,125:3384-3391;Postow M.A.,Callahan M.K.,Wolchok J.D.,2015,J.Clin.Oncol.,33:1974-1982)。近年上市的PD-1和PD-L1的多个抗体药物充分证明了阻断PD-1/PD-L1相互作用在肿瘤的免疫治疗和免疫相关的其他多种疾病中是一种非常有效的治疗手段。
研究发现,PD-L1能够与CD80发生相互作用并抑制PD-L1和PD-1结合,以及抑制T细胞激活的能力。因此,阻断CD80/PD-L1相互作用引起的免疫激活,也可能促进T细胞活性增强,进而为免疫相关的疾病提供了新的治疗机会(Sugiura D.,Maruhashi T.,Okazaki ll-mi,et al.2019,Science,364:558-566)。
至目前,靶向PD-1/PD-L1抗体药物取得了重要进展。然而,所有的抗体药物均须注射给药、具有多种ADMET问题、免疫***相关的严重副作用等。与抗体药物相比,小分子的免疫调节剂具有一定优势,包括可口服、更具组织渗透性、可通过药理学特性调整使得最大限度降低副作用等。另外,小分子抑制剂将具有更低的价格优势,更好的用药依从性。
PD-1/PD-L1抗体药物的临床研究表明,纳武利尤单抗T1/2为25.2天,给药频率为二周一次;帕博利珠单抗T1/2为25天,给药频率为三周一次;阿替利珠单抗T1/2为27天,给药频率为三周一次。上述药物的给药频率均短于药物半衰期,说明这类靶点药物在体内的持续暴露是获得理想临床疗效的关键。然而,已有的小分子免疫调节剂在体内暴露量偏低,持续暴露时间偏短,将影响临床疗效。因此,开发活性更高同时具有更优异口服吸收特征,特别是具有足够体内暴露量和持续暴露时间以及对肿瘤组织更具靶向性的新型小分子PD-L1免疫调节剂以满足尚未满足的临床需求具有重大意义。
发明内容
本发明的一个方面,涉及能够靶向PD-L1的一种小分子联苯类化合物,或其异构体、药学上可接受的盐、前体或代谢产物。
本发明另一方面涉及本文所述化合物的制备方法。
本发明的又一方面涉及包含本发明化合物作为活性成分的药物组合物,以及本发明化合物或药物组合物在制备用于治疗和/或预防与靶点PD-L1相关的疾病的药物中的用途。
作为小分子的PD-L1抑制剂,相比于抗体类,需要从靶点结合强度以及体内持续暴露时间上优化,缩小双方在药效上可能存在的差距。因此,发明人期望开发活性更高同时具有更优异口服吸收特征,特别是具有足够体内暴露量和持续暴露时间以及对肿瘤组织更具有靶向性的的新型小分子PD-L1的免疫调节剂以满足尚未满足的临床需求。
本发明克服了现有的PD-1/PD-L1抗体药物均需要注射给药的缺点,提供了一种具有优异的可口服吸收特征的新型小分子免疫调节剂,特别是本发明的化合物具有具有理想的体内暴露量和持续暴露时间,同时对肿瘤组织具有靶向性,可以在肿瘤组织富集并形成更高的肿瘤组织暴露浓度,有助于治疗中更好的发挥抗肿瘤活性,从而达到更优疗效。
本发明的上述目的是通过提供本发明的式(I)所示化合物、其异构体、药学上可接受的盐、前体或代谢产物来实现的。
为此,在第一方面,本发明涉及式(I)所示化合物,其异构体、药学上可接受的盐、前体或代谢产物,
Figure PCTCN2022074183-appb-000001
其中,
R 1和R 2相同或不同,选自C 1-C 6烷基、氰基、卤素;
R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基、C 2-C 6炔基C 1-C 6烷基、卤 代C 1-C 6烷基、C 1-C 6烷氧基C 1-C 6烷基、单C 1-C 6烷基氨基C 1-C 6烷基、双C 1-C 6烷基氨基C 1-C 6烷基、卤代C 1-C 6烷氧基C 1-C 6烷基、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基、3元至14元杂环烷基-C 1-C 4烷基;
R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被一个或多个选自羟基、羧基和卤素的取代基取代;
X选自-O-、-S-;
R 5-R 26选自氢原子;
R 27-R 30选自氢原子;
其中,上述R 1-R 30定义中各基团中的至少1个氢原子被氘(D)替代。
在一些实施方案中,R 1和R 2可相同或不同,选自甲基、氰基、卤素;
在一些实施方案中,R 1和R 2可相同或不同,选自甲基、氰基、氟、氯;
在一些实施方案中,R 1选自C 1-C 6烷基、氰基、卤素;
在一些实施方案中,R 2选自C 1-C 6烷基、卤素;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 4选自氢、C 1-C 6烷基;
在一些实施方案中,R 4选自氢、甲基;
在一些实施方案中,X选自-O-;
在一些实施方案中,R 5-R 26选自氢原子,优选地其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 5-R 26均为氢原子;
在一些实施方案中,R 5-R 26为氢原子,其中至少1个氢原子为氘(D)替代;
在一些实施方案中,R 27-R 30选自氢原子,或者选自氢原子且其中至少被1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 27-R 30选自氢原子;
在一些实施方案中,R 27-R 30选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在另一个方面,本发明涉及式(I)所示化合物,其异构体、药学上可接受 的盐、前体或代谢产物,
R 1和R 2可相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯、溴;
R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基、C 2-C 6炔基C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基C 1-C 6烷基、单C 1-C 6烷基氨基C 1-C 6烷基、双C 1-C 6烷基氨基C 1-C 6烷基、卤代C 1-C 6烷氧基C 1-C 6烷基、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基、3元至14元杂环烷基-C 1-C 4烷基;
R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被一个或多个选自羟基、卤素的取代基取代;
X选自-O-;
R 5-R 26为氢原子且至少1个氢原子被氘(D)替代;
R 27-R 30选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 1和R 2可相同或不同,选自甲基、氰基、氟、氯;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2
在一些实施方案中,R 4选自氢、C 1-C 6烷基;
在一些实施方案中,R 4选自氢、甲基;
在一些实施方案中,R 27-R 30选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 27-R 30选自氢原子;
在一些实施方案中,R 27-R 30选自氢原子且其中至少1个氢原子被氘(D)取替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在另一个方面,本发明涉及式(I)所示化合物,其异构体、药学上可接受的盐、前体或代谢产物,
R 1和R 2可相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯、溴;
R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基、C 2-C 6炔基C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基C 1-C 6烷基、单C 1-C 6烷基氨基C 1-C 6烷基、双C 1-C 6烷基氨基C 1-C 6烷基、卤代C 1-C 6烷氧基C 1-C 6烷基、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基、3元至14元杂环烷基-C 1-C 4烷基;
R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被一个或多个选自羟基、卤素的取代基取代;
X选自-O-;
R 5-R 26选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
R 27-R 30选自氢原子且其中至少被1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 1和R 2可相同或不同,选自甲基、氰基、氟、氯;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2
在一些实施方案中,R 4选自氢、C 1-C 6烷基;
在一些实施方案中,R 4选自氢、甲基;
在一些实施方案中,R 5-R 26选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 5-R 26选自氢原子;
在一些实施方案中,R 5-R 26选自氢原子且其中至少1个氢原子被氘(D)替代;
在一个优选的方面,本发明涉及式(I)所示的化合物,其异构体、药学上可接受的盐、前体或代谢产物,其中,
R 1和R 2可相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、单C 1-C 6烷基氨基C 1-C 6烷基-、双C 1-C 6烷基氨基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-、C 3-C 14环烷基、3元至14元杂环烷基,这些基团中的至少1个氢原子被氘(D)替代;
R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被卤素取代;
X选自-O-;
R 5-R 26选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
R 27-R 30选自氢原子,或者选自氢原子且其中至少被1个氢原子被氘(D)取代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基,这些取代基中的 至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 4选自氢、C 1-C 6烷基;
在一些实施方案中,R 4选自氢、甲基;
在一些实施方案中,R 5-R 26选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 5-R 26选自氢原子;
在一些实施方案中,R 5-R 26选自氢原子且其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 27-R 30选自氢原子,或者选自氢原子且其中至少被1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 27-R 30选自氢原子;
在一些实施方案中,R 27-R 30选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在另一个优选的方面,本发明涉及式(I)所示的化合物,其异构体、药学上可接受的盐、前体或代谢产物,其中,
R 1和R 2可相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-;
R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被卤素取代;
X选自-O-;
R 5-R 26选自氢原子且其中至少1个氢原子被氘(D)替代;
R 27-R 30选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2
在一些实施方案中,R 4选自氢、C 1-C 6烷基;
在一些实施方案中,R 4选自氢;
在一些实施方案中,R 27-R 30全部选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位 置;
在一些实施方案中,R 27-R 30选自氢原子;
在一些实施方案中,R 27-R 30选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在另一个优选的方面,本发明涉及式(I)所示的化合物,其异构体、药学上可接受的盐、前体或代谢产物,其中,
R 1和R 2可相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
R 3选自C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-;
R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被卤素取代;
X选自-O-;
R 5-R 27选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
R 28-R 30选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2
在一些实施方案中,R 4选自氢、C 1-C 6烷基;
在一些实施方案中,R 4选自氢;
在一些实施方案中,R 5-R 26全部选自氢原子,或者至少1个为氘(D);
在一些实施方案中,R 5-R 26全部选自氢原子;
在一些实施方案中,R 5-R 26至少1个为氘(D);
在一个尤其优选的方面,本发明涉及式(I)所示的化合物,其异构体、药学上可接受的盐、前体或代谢产物,其中,
R 1和R 2可相同或不同地选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
R 3选自C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-,这些基团中的至少1个氢原子被氘(D)替代;
R 4选自氢;
X选自-O-;
R 5-R 27选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
R 28-R 30选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替 代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 1选自甲基、氰基、氟、氯;
在一些实施方案中,R 2选自甲基、氯;
在一些实施方案中,R 3选自C 1-C 6烷基、卤代C 1-C 6烷基,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自C 1-C 6烷基、氟代C 1-C 6烷基,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自甲基、乙基、-CHF 2,这些取代基中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 5-R 26选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 5-R 26选自氢原子;
在一些实施方案中,R 5-R 26选自氢原子且其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 27-R 30选自氢原子,或者选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置;
在一些实施方案中,R 27-R 30选自氢原子;
在一些实施方案中,R 27-R 30选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置可以在相应脂肪环和脂肪杂环上的任意位置。
在一些实施方案中,R 1和R 2相同或不同,选自甲基、氰基、卤素;
R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-,这些基团中的至少1个氢原子被氘(D)替代;
R 4选自氢、C 1-C 6烷基;
X选自-O-;
R 5-R 26选自氢原子;
R 27-R 30选自氢原子。
在一些实施方案中,R 1和R 2相同或不同,选自甲基、氰基、氟、氯;
在一些实施方案中,R 1和R 2相同或不同,选自甲基、氟、氯;
在一些实施方案中,R 1和R 2相同或不同,选自甲基、氯。
在一些实施方案中,R 3选自氢、C 1-C 6烷基、卤代C 1-C 6烷基,这些基团中的至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自C 1-C 6烷基,其中至少1个氢原子被氘(D)替代;
在一些实施方案中,R 3选自甲基、乙基、丙基、丁基、戊基,其中至少1个氢原子被氘(D)替代。
在一些实施方案中,R 3定义基团中的1-15个氢原子被氘(D)替代;
在一些实施方案中,R 3定义基团中的1-9个氢原子被氘(D)替代。
在一些实施方案中,R 4选自氢、甲基、乙基、丙基、丁基、戊基;
在一些实施方案中,R 4选自氢。
本发明涉及式的(I)所示的化合物,优选选自以下化合物:
Figure PCTCN2022074183-appb-000002
Figure PCTCN2022074183-appb-000003
Figure PCTCN2022074183-appb-000004
Figure PCTCN2022074183-appb-000005
Figure PCTCN2022074183-appb-000006
Figure PCTCN2022074183-appb-000007
Figure PCTCN2022074183-appb-000008
Figure PCTCN2022074183-appb-000009
各术语定义
在本说明书的各部分,本发明公开化合物的取代基按照基团种类或范围公开。特别指出,本发明包括这些基团种类和范围的各个成员的每一个独立的次级组合。 例如,术语“C 1-C 6烷基”特别指独立公开的甲基、乙基、C 3烷基、C 4烷基、C 5烷基和C 6烷基,或者独立公开的“C 1-C 4烷基”,或者独立公开的“C 1-C 3烷基”。
本发明所述的“卤素”为氟、氯、溴或碘,优先为氟或氯或溴。
本发明所述的“烷基”,包括直链或支链的烷基。本发明中所述的C 1-C 6烷基,是指碳原子数为1-6的烷基,优选为甲基、乙基、正丙基或异丙基、正丁基、异丁基或叔丁基。本发明化合物中的烷基可以是任选取代或未取代的,取代的取代基可以包括烷基、卤素、烷氧基、卤代烷基、氰基、羟基等。本发明烷基的实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基等。
本发明所述的“烯基”表示2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子的直链或支链的一价烃基,其中至少一个位置为不饱和状态,即一个C-C为sp 2双键,其中烯基基团可以独立且任选地被一个或多个本发明所描述的取代基所取代,包括基团有“反”,“顺”或“E”,“Z”的定位,其中烯基可以为C 2-C 6烯基,具体的实例包括,但并不限于,乙烯基(-CH=CH 2)、烯丙基(-CH 2CH=CH 2)等等。
本发明所述的“炔基”表示2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子的直链或支链的一价烃基,其中至少一个位置为不饱和状态,即一个C-C为sp三键,其中炔基基团可以独立且任选地被一个或多个本发明所描述的取代基所取代,其中炔基可以为C 2-C 6炔基,具体的实例包括,但并不限于,乙炔基(-C≡CH)、炔丙基(-CH 2C≡CH)等等。
本发明所述的“烷氧基”,是指上述烷基与氧原子相连所形成的基团,其中,氧原子具有自由成键能力,如可以为“C 1-C 6烷氧基”,具体如甲氧基、乙氧基、正丙氧基、正丁氧基、异丙氧基、叔丁氧基、环丙氧基等。
本发明所述的“烷基氨基”,是指上述烷基与氨基相连所形成的基团,如可以为“C 1-C 6烷基氨基”,具体如甲氨基、乙氨基、二甲氨基、甲基异丙基氨基等。
本发明所述的“卤代C 1-C 6烷基”和“卤代C 1-C 6烷氧基”,是指所述烷基和烷氧基中一个或多个氢原子被卤素原子,特别是氟或氯原子取代。在一些实施方案中,优选氟代,例如-CF 3、-CHF 2、-CH 2F、-CH 2CH 2F、-CH 2CHF 2、-CH 2CF 3、-OCF 3、-OCHF 2、-OCH 2F、-OCH 2CH 2F、-OCH 2CHF 2或者-OCH 2CF 3
本发明所述的“环烷基”,是指具有指定环碳原子数目的烃基单环结构,其中不含有双键等不饱和键,包括C 3-C 14环烷基,C 3-C 6环烷基,例如为环丙基、环丁基、环戊基、环己基。本发明化合物中的环烷基可以是任选取代或未取代的,取代基可以包括烷基、卤素、烷氧基、烃基、羟基等。
本发明所述的“杂环”是指含有一个或多个杂原子的未取代和取代的单环或多环非芳香环系,部分不饱和或完全饱和的环体系。优选的杂原子包括N、O和 S。单环杂环包括但不限于,吡咯烷基、咪唑烷基、四氢呋喃基、二氢咪唑基、二氢呋喃基、哌啶基、吗啉基、硫代吗啉基、高哌嗪基等。多环杂环基包括螺环、桥环、稠环的杂环基,所述的杂环基环可以稠合于芳基、杂芳基或环烷基环。
从所有上述描述中,对本领域技术人员显而易见的是,其名称是复合名称的任意基团,例如“C 2-C 6烯基C 1-C 6烷基-”,应该指的是常规地从其衍生的部分例如从被C 2-C 6烯基取代的C 1-C 6烷基来构建,其中C 2-C 6烯基和C 1-C 6烷基如上文所定义。其他类似的复合名称如“C 2-C 6炔基C 1-C 6烷基-”、“C 3-C 14环烷基-C 1-C 4烷基-”可以参照前述内容进行理解。
除非另有具体定义,否则,本文所述的“取代的”指基团中的一个或多个氢原子能够独立地被相应数目的取代基取代。本领域的技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代位置。所述“取代的”所指的取代基是指包括但不限于:氰基、羧基、卤素、羟基、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、单C 1-C 6烷基氨基C 1-C 6烷基-、双C 1-C 6烷基氨基C 1-C 6烷基-、卤代C 1-C 6烷基、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧C 1-C 6烷基-、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基-、3元至14元杂环烷基-C 1-C 4烷基-。
本文所述的化合物中的立体异构体,当以化学名称特别指定为(R)-或(S)-异构体时,应分别理解为主要构型为(R)-异构体或(S)-异构体。任何不对称碳原子可以存在于(R)-、(S)-或(R、S)-构型中,优选以(R)-或(S)-构型存在。
本发明所述的“药学可接受的盐”是指本发明化合物与药学上可接受的酸进行反应制得的酸加成盐,或者其中具有酸性基团的化合物和碱性化合物反应生成的盐。其中,所述的酸较佳的选自无机酸(如盐酸、硫酸、磷酸或氢溴酸等),和有机酸(如草酸、马来酸、富马酸、苹果酸、酒石酸、赖氨酸、组氨酸、柠檬酸或苯甲酸等);所述的碱性化合物较佳的选自氢氧化钠、氢氧化钾、氢氧化钙、碳酸钠或碳酸氢钾等。上述药学上可接受的盐容易分离,可采用常规分离方法提纯,如溶剂萃取、稀释、重结晶、柱色谱和制备薄层色谱等。
除非特别指出,否则本文中所述的“氢原子”通常意指质量数为1的氢的同位素形式,即 1H。
本发明的另一方面还涉及药物组合物,所述药物组合物含有上述的化合物,或其异构体、药学上可接受的盐、前体或代谢产物作为活性成分。
本发明所述的化合物任选地可与其它一种或多种活性成分联合使用,其各自用量和比例可由本领域技术人员根据具体病症和患者具体情况以及临床需要等而进行调整。
本发明所述通式(I)的化合物,其异构体、药学上可接受的盐、前体或代 谢产物都能够被本领域的技术人员(经验或参考文献)制备得到。
当本发明所述通式(I)的化合物的结构式与中文名称不符时,以结构式为准。
因此,本发明的另一个方面还提供了上述根据本发明的化合物的制备方法。
下面的合成路线描述了本发明的式(I)化合物的制备方法,如下合成示意图中所用原料、试剂、催化剂、溶剂等均可通过有机化学领域普通技术人员熟知的方法制备或者可商购得到。本发明的全部最终衍生物都可都过示意图中所描述的方法或其类似方法制得,这些方法都是有机化学领域普通技术人员熟知的。这些示意图中应用的全部可变因素如上下文定义。
制备方法
下面各变量的定义如前所述,而新变量定义如本部分内容所述。另外,通式(I)所述的化合物及涉及的中间体均可通过常见的分离方法进行纯化,如萃取、重结晶及硅胶柱层析分离等。所用200-300目的硅胶和薄层层析硅胶板均由青岛海洋化工厂生产。所用化学试剂为一般试剂的分析纯或化学纯市售商品,使用时未经进一步纯化。
本发明提供一种通式(I)所示化合物的制备方法,其包括如下步骤:
Figure PCTCN2022074183-appb-000010
1)使式(I-a)所示化合物在第一溶剂中经第一酸、第一碱或者催化氢解脱去保护基P 1,所得产物未经分离纯化,进一步与式(I-b)所示化合物在第二溶 剂中并且在第一催化剂和第二碱存在下发生Suzuki反应得到式(I-c)所示化合物;
2)在第三溶剂中,使式(I-c)所示化合物和式(I-d)所示化合物在第一还原剂存在下发生还原胺化反应得到式(I-e)所示化合物;或者,
在第四溶剂中,使式(I-c)所示化合物与式(I-d')所示化合物在第三碱存下发生亲核取代反应得到式(I-e)所示化合物;
3)在第五溶剂中,使式(I-e)所示化合物和式(I-f)所示化合物在第二还原剂存在下发生还原胺化反应得到式(I)所示化合物;或者,
在第六溶剂中,使式(I-e)所示化合物与式(I-f')所示化合物在第四碱存下发生亲核取代反应得到式(I)所示化合物;
任选地,还包括4)在第五碱存在下,使式(I')所示化合物经过酯水解反应得到式(I”)所示化合物;
如上所述,式(I')所示化合物作为终产物(I)的一种形式。另外,式(I')所示化合物还可进一步在碱性条件下经过酯水解反应得到式(I”)所示化合物,所述式(I”)所示化合物也作为终产物(I)的一种形式;
其中:
X、R 1-R 30的定义如前所述;
R 4'除了不为氢以外,其定义与R 4相同,R 4的定义如前所述;
R 4”为氢;
M选自硼酸酯或硼酸,包括但不限于4,4,5,5-四甲基-1,3,2-二氧杂戊硼烷、联硼酸新戊二醇酯、4,4,4',4',5,5,5',5'-八甲基-2,2'-二(1,3,2-二氧杂环戊硼烷)B(OBu-n) 3、B(OPr-i) 3;或者,
M选自溴、碘、氯、氟、CF 3SO 3-(OTf);
W选自硼酸酯或硼酸,优选地选自4,4,5,5-四甲基-1,3,2-二氧杂戊硼烷、联硼酸新戊二醇酯、4,4,4',4',5,5,5',5'-八甲基-2,2'-二(1,3,2-二氧杂环戊硼烷)B(OBu-n) 3、B(OPr-i) 3;或者,
W选自溴、碘、氯、氟、CF 3SO 3-(OTf);
P 1和P 2是保护基,可相同或不同,优选地选自Boc(叔丁氧羰基)、Fmoc(9-芴甲氧羰基)、Cbz(N-苄氧羰基)、甲磺酰基、对甲苯磺酰基、乙酰基、甲氧羰基、乙氧羰基、((2-三甲基硅)乙氧)甲基(SEM)、四氢-2H-吡喃-2-基(THP)。
在一些实施方案中,所述的第一酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr);
所述的第一碱优选地选自哌啶、二乙胺;
所述的第一溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、 乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF)。
在一些实施方案中,所述第一催化剂优选地选自1,1'-双(二环己基膦基)二茂铁二氯化钯(PdCl 2(dcypf))、醋酸钯(Pd(OAc) 2)、二氯化钯(PdCl 2)、三(二亚苄基丙酮)二钯(Pd 2(dba) 3)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(PdCl 2(dppf))、[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(PdCl 2(dppf)·CH 2Cl 2)、四(三苯基膦)钯(Pd(PPh 3) 4)、双(三环己基膦)二氯化钯(PdCl 2(P(Cy) 3) 2)、2-二环己基膦-2',6'-二甲氧基联苯(SPhos);
所述第二碱包括有机碱和无机碱类,例如三乙胺(TEA)、N,N-二异丙基乙基胺(DIPEA)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、醋酸钾(KOAc)、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、磷酸钾(K 3PO 4)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH);
所述第二溶剂优选地选自1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF),以及这些溶剂与水以不同比例形成的混合溶剂。
在一些实施方案中,所述第一还原剂和第二还原剂优选地选自醋酸硼氢化钠、硼氢化钠、氰基硼氢化钠;
所述第三溶剂和第五溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF)。
在一些实施方案中,所述第四溶剂和第六溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF)、N-甲基吡咯烷酮(NMP)、吡啶(Py);
所述的第三碱和第四碱优选地选自三乙胺(TEA)、N,N'-二异丙基乙基胺(DIPEA)、吡啶(Py)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH)。
在一些实施方案中,所述第五碱包括优选地选自氢氧化锂(LiOH)、氢氧化锂(KOH)、氢氧化钠(NaOH)。
具体的,式(I-a)所示化合物在适当的溶剂下经适当的酸、碱或者催化氢解脱去保护基P 1,所得产物未经分离纯化,进一步与式(I-b)所示化合物在适当的碱性条件及溶剂下,并在催化剂下经Suzuki反应得到式(I-c)所示化合物,所述的脱保护的适当溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇 (MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF);所述的酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr);所述的碱优选地选自哌啶、二乙胺;所述催化剂优选地选自1,1'-双(二环己基膦基)二茂铁二氯化钯(PdCl 2(dcypf))、醋酸钯(Pd(OAc) 2)、二氯化钯(PdCl 2)、三(二亚苄基丙酮)二钯(Pd 2(dba) 3)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(PdCl 2(dppf))、[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(PdCl 2(dppf)·CH 2Cl 2)、四(三苯基膦)钯(Pd(PPh 3) 4)、双(三环己基膦)二氯化钯(PdCl 2(P(Cy) 3) 2)、2-二环己基膦-2',6'-二甲氧基联苯(SPhos);所述碱性条件的试剂包括有机碱和无机碱类,优选地选自三乙胺(TEA)、N,N-二异丙基乙基胺(DIPEA)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、醋酸钾(KOAc)、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、磷酸钾(K 3PO 4)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH);所述发生Suzuki反应的溶剂优选地选自1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF),以及这些溶剂与水以不同比例形成的混合溶剂;
式(I-c)所示化合物与式(I-d)所示化合物在适当的溶剂和还原剂存在下发生还原胺化反应得到式(I-e)所示化合物,所述还原剂优选地选自醋酸硼氢化钠、硼氢化钠、氰基硼氢化钠;所述溶剂包括但不限于二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF);或者,
式(I-c)所示化合物与式(I-d')所示化合物在适当的溶剂和碱存在下发生亲核取代反应得到式(I-e)所示化合物,所述碱优选地选自三乙胺(TEA)、N,N'-二异丙基乙基胺(DIPEA)、吡啶(Py)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH);所述溶剂包括但不限于二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF)、N-甲基吡咯烷酮(NMP)、吡啶(Py);
式(I-e)所示化合物在适当的溶剂下经适当的酸、碱或者催化氢解脱去保护基P 2,所得产物未经分离纯化,进一步与式(I-f)所示化合物在适当的溶剂和还原剂存在下发生还原胺化反应得到式(I')所示化合物,所述的酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr);所述的碱包括但不限于哌啶、二乙胺;所述还原剂优选地选自醋酸硼氢化钠、硼氢化钠、氰基硼氢化钠;所述溶剂包括但不限于二氯甲烷(DCM),1,2-二氯乙烷、甲醇 (MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF);或者,
式(I-e)所示化合物在适当的溶剂下经适当的酸、碱或者催化氢解脱去保护基P 2,所得产物未经分离纯化,进一步与式(I-f')所示化合物在适当的溶剂和碱存在下发生亲核取代反应得到式(I')所示化合物,所述碱优选地选自三乙胺(TEA)、N,N'-二异丙基乙基胺(DIPEA)、吡啶(Py)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH);所述溶剂包括但不限于二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF)、N-甲基吡咯烷酮(NMP)、吡啶(Py);
上述式(I')所示化合物作为终产物(I)的一种形式。另外,式(I')所示化合物还可进一步在碱性条件下经过酯水解反应得到式(I”)所示化合物,式(I”)所示化合物也作为终产物(I)的一种形式,所述的碱优选地选自氢氧化锂(LiOH)、氢氧化锂(KOH)、氢氧化钠(NaOH);
M优选地选自硼酸酯或硼酸,包括但不限于4,4,5,5-四甲基-1,3,2-二氧杂戊硼烷、联硼酸新戊二醇酯、4,4,4',4',5,5,5',5'-八甲基-2,2'-二(1,3,2-二氧杂环戊硼烷)B(OBu-n) 3、B(OPr-i) 3;或者,
M优选地选自溴、碘、氯、氟、CF 3SO 3-(OTf);
W优选地选自硼酸酯或硼酸,包括但不限于4,4,5,5-四甲基-1,3,2-二氧杂戊硼烷、联硼酸新戊二醇酯、4,4,4',4',5,5,5',5'-八甲基-2,2'-二(1,3,2-二氧杂环戊硼烷)B(OBu-n) 3、B(OPr-i) 3;或者,
W优选地选自溴、碘、氯、氟、CF 3SO 3-(OTf);
P 1和P 2是保护基,可相同或不同,优选地选自Boc(叔丁氧羰基)、Fmoc(9-芴甲氧羰基)、Cbz(N-苄氧羰基)、甲磺酰基、对甲苯磺酰基、乙酰基、甲氧羰基、乙氧羰基、((2-三甲基硅)乙氧)甲基(SEM)、四氢-2H-吡喃-2-基(THP)。
本发明的化合物制备方法还可通过式(I-a)所示化合物经脱去保护基P 1,所得产物未经分离纯化,进一步与式(I-b)所示化合物发生如前所述的Suzuki反应得到式(I-c)所示化合物;式(I-c)所示化合物先与式(I-f)所示化合物发生如上所述的还原胺化反应得到式(I-h)所示化合物;或者通过式(I-c)所示化合物先与式(I-f')所示化合物发生如上所述的亲核取代反应得到式(I-h)所示化合物,随后(I-h)所示化合物经脱保护后,再与式(I-d)所示化合物发生如上所述的还原胺化反应得到式(I')所示化合物;或者(I-h)所示化合物经脱保护后,再与式(I-d')所示化合物发生如上所述的亲核取代反应得到式(I'-1) 所示化合物,或者进一步发生如上所述的酯水解反应得到式(I”-1)所示化合物,该制备方法包括:
Figure PCTCN2022074183-appb-000011
其中,上述式(I'-1)所示化合物作为终产物(I)的一种形式。另外,式(I')所示化合物还可进一步在碱性条件下经过酯水解反应得到式(I”-1)所示 化合物,式(I”-1)所示化合物也作为终产物(I)的一种形式;
其中,各取代基的定义如前所述。
本发明的化合物制备方法还可通过式(I-a)所示化合物经脱去保护基P 1,所得产物未经分离纯化,进一步与式(I-b)所示化合物发生如前所述的Suzuki反应得到式(I-c)所示化合物;式(I-c)所示化合物先与式(I-f)所示化合物发生如前所述的还原胺化反应得到式(I-h)所示化合物;或者通过式(I-c)所示化合物先与式(I-f')所示化合物发生如前所述的亲核取代反应得到式(I-h)所示化合物,随后式(I-h)所示化合物进一步发生如上所述的酯水解反应得到式(I-i)所示化合物,随后(I-i)所示化合物经脱保护后,再与式(I-d)所示化合物发生如上所述的还原胺化反应得到式(I”')所示化合物;或者(I-i)所示化合物经脱保护后,再与式(I-d')所示化合物发生如上所述的亲核取代反应得到式(I”')所示化合物,该制备方法包括:
Figure PCTCN2022074183-appb-000012
其中,上述式(I”')所示化合物可以作为终产物(I);
其中,各取代基的定义如前所述。
在通式(I)所示化合物的制备过程中,本发明还提供了所用到的式(I-a)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000013
式(I-a-1)与式(I-a-2)在第六碱和第七溶剂作用下经过氨解反应得到式(I-a)所示化合物。
其中:各取代基的定义如前所述。
在一些实施方案中,所述的第六碱优选地选自叔丁醇钾(KOBu-t)、叔丁醇钠(NaOBu-t)、氢化钠(NaH)、正丁基锂、甲醇钠(MeONa)、乙醇钠(EtONa);
在一些实施方案中,所述第七溶剂优选地选自无水四氢呋喃(THF)、无水1,4-二氧六环(1,4-dioxane)、无水乙腈(MeCN)。
在通式(I)所示化合物的制备过程中,本发明还提供了所用到的式(I-b)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000014
式(I-b)所示化合物的制备策略与上述的式(I-a)的制备策略相同。
其中:各取代基的定义如前所述。
在通式(I-a)所示化合物的制备过程中,本发明还提供了所用到的式(I-a-1)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000015
1)使式(I-a-1-1)所示化合物与式(I-a-1-2)所示化合物在第七碱的作用下得到式(I-a-1-3)所示化合物;
在一些实施方案中,所述的第七碱优选地选自叔丁醇钾(KOBu-t)、叔丁醇钠(NaOBu-t)、氢化钠(NaH)、正丁基锂、甲醇钠(MeONa)、乙醇钠(EtONa);
2)使式(I-a-1-3)进一步经插羰反应得到(I-a-1-4)所示化合物;
3)使式(I-a-1-4)经过酯化反应得到(I-a-1)所示化合物;
其中:各取代基的定义如前所述。
更具体的,使式(I-a-1-1)所示化合物与式(I-a-1-2)所示化合物在适当碱的作用下得到式(I-a-1-3)所示化合物。其中所述的碱优选地选自叔丁醇钾(KOBu-t)、叔丁醇钠(NaOBu-t)、氢化钠(NaH)、正丁基锂、甲醇钠(MeONa)、乙醇钠(EtONa);随后使式(I-a-1-3)在正丁基锂的作用下与CO 2(干冰)进一步发生插羰反应得到(I-a-1-4)所示化合物;最后,使式(I-a-1-4)经过酰氯的活化形式与甲醇(MeOH)反应,或者在甲醇(MeOH)/浓硫酸作用下得到得到(I-a-1)所示化合物。
在通式(I-a)所示化合物的制备过程中,本发明还提供了所用到的式(I-a-1)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000016
1)使式(I-a-1-1)所示化合物与式(I-a-1-2)所示化合物在第八碱的作用下得到式(I-a-1-3)所示化合物;2)使式(I-a-1-3)进一步在第三还原剂将硝基还原为氨基得到(I-a-1-4)所示化合物;
3)使式(I-a-1-4)经成环反应得到(I-a-1-6)所示化合物;
4)为了进一步发生还原反应,将式(I-a-1-6)形成季铵盐得到(I-a-1-8)所示化合物;
5)使式(I-a-1-8)在第四还原剂作用下将其还原得到式(I-a-1-9)所示化合
物;
6)使式(I-a-1-9)经脱去P 3得到式(I-a-1-10)所示化合物;
7)使式(I-a-1-10)所示化合物再经进一步保护得到式(I-a-1-11)所示化 合物;
8)使式(I-a-1-11)进一步经插羰反应得到(I-a-1-12)所示化合物;
9)使式(I-a-1-12)经过酯化反应得到(I-a-1)所示化合物。
其中:P 3是指为了促进下一步还原反应,同时能形成季铵盐的片段,包括但不限于苄基(Bn)、对甲氧基苄基(PMB)、对甲氧基羰基苄基、烯丙基;
其他各取代基的定义如权利要求9或10所述。
在一个实施方案中,所述第八碱优选地选自碳酸钾(K 2CO 3)、叔丁醇钾(KOBu-t)、叔丁醇钠(NaOBu-t)、氢化钠(NaH)、正丁基锂、甲醇钠(MeONa)、乙醇钠(EtONa)。
在一个实施方案中,所述第三还原剂优选地选自铁粉、锌粉、二氯化锡(SnCl 2)、硫化钠(Na 2S)、水合肼。
在一个实施方案中,所述第四还原剂优选地选自苄基(Bn)、对甲氧基苄基(PMB)、对甲氧基羰基苄基、烯丙基。
更具体的,使式(I-a-1-1)所示化合物与式(I-a-1-2)所示化合物在适当碱的作用下得到式(I-a-1-3)所示化合物,其中所述的碱优选地选自碳酸钾(K 2CO 3)、叔丁醇钾(KOBu-t)、叔丁醇钠(NaOBu-t)、氢化钠(NaH)、正丁基锂、甲醇钠(MeONa)、乙醇钠(EtONa);随后式(I-a-1-3)在还原剂的作用下将硝基还原为氨基得到(I-a-1-4)所示化合物,其中所述的还原剂优选地选自铁粉、锌粉、二氯化锡(SnCl 2)、硫化钠(Na 2S)、水合肼。进一步地,将(I-a-1-4)经成环反应得到(I-a-1-6)所示化合物,随后形成季铵盐得到式(I-a-1-8)所示化合物,进而在还原剂的作用下得到式(I-a-1-9)所示化合物,其中所述的还原剂优选地选自苄基(Bn)、对甲氧基苄基(PMB)、对甲氧基羰基苄基、烯丙基;进一步地,脱去P 3得到式(I-a-1-10)所示化合物,随后经过P 1保护、插羰反应、酯化反应得到式(I-a-1)所示化合物。
在通式(I-b)所示化合物的制备过程中,本发明提供的所用到的式(I-b-1)的制备策略与上述的式(I-a-1)的制备策略相同。
本发明还提供一种制备通式(I)所示化合物中使用到的未公开报道的中间体I-2a-1(I-a-1)、I-1d(I-d)、I-2a-2(I-a-2)、I-3a-2(I-a-2)、I-4a-2(I-a-2)、I-5a-2(I-a-2),这些中间体包括但不限于:
Figure PCTCN2022074183-appb-000017
本发明提供了上述中间体I-1d(I-d)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000018
1)将式(I-1d-1)用第五还原剂还原得到式(I-1d-2);
2)将式(I-1d-2)用氘代甲基化试剂经氘代甲基化反应得到式(I-1d-3);
3)使式(I-1d-3)最后在第二酸性条件下经脱保护得到式(I-1d);
其中所述的第五还原剂优选地选自氢化铝锂(LAH)、硼烷(BH 3)、NaBH 4/AlCl 3
其中所述的甲基化试剂优选地选自氘代碘甲烷(CD 3I);
其中所述的第二酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr);
本发明提供了上述未知中间体I-2a-2(I-a-2)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000019
1)使式(I-2a-2-1)在溴代试剂的作用下经溴代反应得到式(I-2a-2-2);
2)使式(I-2a-2-2)在第二催化剂作用下经C-N偶联反应得到式(I-2a-2-4);
3)使式(I-2a-2-4)在第三酸性条件下经脱保护得到式(I-2a-2-5);
4)使式(I-2a-2-5)在氯代试剂的作用下经氯代反应得到式(I-2a-2-6);
5)使式(I-2a-2-6)经重氮化反应得到式(I-2a-2-7);
6)使式(I-2a-2-7)在第六还原剂作用下进行硝基还原得到(I-2a-2);
其中所述的溴代试剂优选地选自N-溴代丁二酰亚胺(NBS)、溴化钠(NaBr)、溴化钾(KBr)、溴酸钠(NaBrO 3)、溴酸钾(KBrO 3);
其中所述的第二催化剂优选地选自4,5-双二苯基膦-9,9-二甲基氧杂蒽(Xantphos)、1,1'-双(二环己基膦基)二茂铁二氯化钯(PdCl 2(dcypf))、醋酸钯(Pd(OAc) 2)、二氯化钯(PdCl 2)、三(二亚苄基丙酮)二钯(Pd 2(dba) 3)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(PdCl 2(dppf))、[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(PdCl 2(dppf)·CH 2Cl 2)、四(三苯基膦)钯(Pd(PPh 3) 4)、双(三环己基膦)二氯化钯(PdCl 2(P(Cy) 3) 2)、2-二环己基膦-2',6'-二甲氧基联苯(SPhos);
其中所述的第三酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr);
其中所述的氯代试剂优选地选自N-氯代丁二酰亚胺(NCS);
其中所述的第六还原剂优选地选自铁粉、锌粉、二氯化锡(SnCl 2)、硫化钠(Na 2S)、水合肼。
本发明提供了上述未知中间体I-3a-2(I-a-2)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000020
1)使式(I-2a-2-7)在氟化试剂的作用下经氟化反应得到式(I-3a-2-1);
2)使式(I-3a-2-1)在第七还原剂的作用下将硝基还原得到(I-3a-2);
其中所述的氟化试剂优选地选自氟化钾、氟化铯;
其中所述的第七还原剂优选地选自铁粉、锌粉、二氯化锡(SnCl 2)、硫化钠(Na 2S)、水合肼。
本发明提供了上述未知中间体I-4a-2(I-a-2)的制备方法,包括以下步骤:
Figure PCTCN2022074183-appb-000021
1)使式(I-2a-2-2)在第八还原剂作用下将硝基还原得到(I-4a-2-1);
2)使式(I-4a-2-1)经重氮化反应得到式(I-4a-2-2);
3)使式(I-4a-2-2)经亲电取代反应得到式(I-4a-2-3);
4)使式(I-4a-2-3)在第三催化剂作用下经C-N偶联反应得到式(I-4a-2-4);
5)使式(I-4a-2-4)在第四酸性条件下经脱保护得到式(I-4a-2)。
其中所述的第八还原剂优选地选自铁粉、锌粉、二氯化锡(SnCl 2)、硫化钠(Na 2S)、水合肼;
其中所述的第三催化剂优选地选自4,5-双二苯基膦-9,9-二甲基氧杂蒽(Xantphos)、1,1'-双(二环己基膦基)二茂铁二氯化钯(PdCl 2(dcypf))、醋酸钯(Pd(OAc) 2)、二氯化钯(PdCl 2)、三(二亚苄基丙酮)二钯(Pd 2(dba) 3)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(PdCl 2(dppf))、[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(PdCl 2(dppf)·CH 2Cl 2)、四(三苯基膦)钯(Pd(PPh 3) 4)、双(三环己基膦)二氯化钯(PdCl 2(P(Cy) 3) 2)、2-二环己基膦-2',6'-二甲氧基联苯(SPhos);
其中所述的第四酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr)。
另外,本发明提供了前述的化合物或其立体异构体、药学上可接受的盐、前体或代谢产物或者前述的药物组合物在制备用于治疗和/或预防与靶点PD-L1相关的疾病的药物中的用途,或者
在制备用于抑制PD-L1活性的药物中的用途,或者
在制备作为PD-L1抑制剂的药物中的用途,或者
在制备作为靶向PD-L1信号通路的免疫调节剂的药物中的用途。
在一些实施方案中,所述与靶点PD-L1相关的疾病包括肿瘤、癌症或者其他免疫相关疾病。
在另一个方面,本发明提供了前述的化合物或其立体异构体、药学上可接受的盐、前体或代谢产物或者前述的药物组合物,其用于治疗和/或预防与靶点PD-L1相关的疾病,或者
用于抑制PD-L1活性,或者
作为PD-L1抑制剂,或者
作为靶向PD-L1信号通路的免疫调节剂。
在一些实施方案中,所述与靶点PD-L1相关的疾病包括肿瘤、癌症或者其他免疫相关疾病。
在另一个方面,本发明提供了治疗和/或预防与靶点PD-L1相关的疾病的方法,其包括给予有需要的受试者治疗和/或预防有效量的前述的化合物或其立体异构体、药学上可接受的盐、前体或代谢产物或者前述的药物组合物。
本发明还提供了抑制PD-L1活性的方法,其包括给予细胞(例如哺乳动物细胞)有效量的前述的化合物或其立体异构体、药学上可接受的盐、前体或代谢产物或者前述的药物组合物。
在一些实施方案中,所述与靶向PD-L1相关的疾病包括肿瘤、癌症或者免疫相关的其他疾病。
在本发明中,“受试者”指脊椎动物。在某些实施方案中,脊椎动物指哺乳动物。所述哺乳动物包括牛科动物、马科动物、羊科动物、猪科动物、犬科动物、猫科动物、啮齿类动物、灵长类动物,例如是人,猫,狗或猪。哺乳动物包括,但不限于,牲畜(诸如牛)、宠物(诸如猫、犬、和马)、灵长类动物、小鼠和大鼠。在某些实施方案中,哺乳动物指人。
本发明中,术语“治疗有效量”或“预防有效量”是指在合理的医学判断范围内,足以治疗或预防患者疾病但足够低地避免严重副作用(在合理的利益/风险比)的量。化合物的治疗有效量将根据所选择的具体化合物(例如考虑化合物的效力、有效性和半衰期)、所选择的给药途径、所治疗的疾病、所治疗的疾病的严重性、所治疗的患者的年龄、大小、体重和身体疾病、所治疗的患者的医疗史、治疗持续时间、并行疗法的性质、所需的治疗效果等因素发生变化,但仍可以由本领域技术人员常规确定。
另外需要指出,所述化合物或其立体异构体、药学上可接受的盐、前体或代谢产物针对不同患者的特定使用剂量和使用方法决定于诸多因素,包括患者的年龄,体重,性别,自然健康状况,营养状况,药物的活性强度,服用时间,代谢速率,病症的严重程度以及诊治医师的主观判断。这里优选使用剂量介于0.001-1000mg/kg体重/天。
本发明提供了一种新颖的具有优异可口服吸收特征的联苯类的小分子免疫抑制剂,用于治疗或预防与免疫相关的疾病。同时,这些化合物或者包含其作为活性成分的药物组合物等可在安全治疗窗口内能将对这些疾病的临床疗效达到最大化。
附图说明
图1表示hPD1/hPD-L1小鼠给药18天后化合物I-1的组织分布(50mg/kg,PO)。
图2表示化合物I-1、Example17、化合物14(INCB086550)及实施例180在小鼠体内相对血浆的组织分布。
具体实施方式
本发明中所提供的实施例和制备例进一步阐明并举例说明了本发明所述化合物及其制备方法。应当理解,下述制备例和实施例不以任何方式限制本发明的范围。
LC-MS分析方法:
质谱条件:仪器Thermo MSQ Plus;离子源ESI(EA+EA-);锥孔电压30V;毛细管电压3.00KV;源温度350℃;
色谱条件:仪器Thermo U3000;检测器DAD-3000(RS)(二极管阵列检测器);色谱柱 岛津Inertsil ODS-HL HP 3μm 3.0×100mm;流速0.4mL/min;柱温30℃;流动相CH 3OH/H 2O/HCOOH(75/25/0.2)。
HPLC分析方法(一):
仪器Thermo U3000;检测器VWD-3×00(RS)(紫外检测器);色谱柱 岛津Shim-pack GIST C8 5μm 4.6×250mm;流速1.0mL/min;柱温30℃;流动相CH 3OH/H 2O/TEA/HOAc(80/20/0.2/0.1)。
HPLC分析方法(二):
仪器Thermo U3000;检测器VWD-3×00(RS)(紫外检测器);色谱柱 岛津Shim-pack GIST C8 5μm 4.6×250mm;流速1.0mL/min;柱温30℃;流动相CH 3OH/pH4.5 HCOONH 4(85/15)。
1H-NMR分析方法:
1H-NMR在室温下采用BRUKER AVANCE-400MHz型核磁共振波谱仪在DMSO-d 6或CDCl 3等中以TMS为内标物测定,信号峰表示为s(单峰),d(双峰),t(三重峰),q(四重峰),m(多重峰),dd(双二重峰)。耦合常数(J)的单位为赫兹(Hz)。
关键缩略语:
1,4-dioxane 1,4-二氧六环;    DCM      二氯甲烷
MeOH        甲醇;            EtOAc    乙酸乙酯
TEA         三乙胺            DIPEA    N,N'-二异丙基乙基胺
TFA          三氟乙酸;         THF        四氢呋喃
NaHCO 3       碳酸氢钠           Na 2SO 4      硫酸钠
NaBH(OAc) 3   醋酸硼氢化钠;     LiOH·H 2O  水合氢氧化锂
PdCl 2(dcypf) 1,1'-双(二环己基膦基)二茂铁二氯化钯
TLC          薄层色谱法
按照上述说明方法,本发明制备了代表性化合物I-1—I-15(见表1)。
表1本发明所述的代表性化合物I-1—I-15
Figure PCTCN2022074183-appb-000022
下面结合具体实例进一步阐述本发明内容,但本发明的保护范围并不仅仅局限于这些实例。本发明所述的百分比除特别注明外,均为重量百分比。说明书中所描述的数值范围,如计量单位、反应条件、化合物物理状态或百分比,均是为了提供明白无误的书面参考。本领域技术人员在实施本发明时,使用在此范围之外或有别于单个数值的温度、浓度、数量、碳原子数等,仍然有可能得到预期结果。另外,以下实施例中的原料如无特别说明,均可商购获得,例如可以购自上海毕得医药科技有限公司、江苏艾康生物医药研发有限公司、南京药石科技股份有限公司、上海韶远试剂有限公司、赫淳生物科技(上海)有限公司。
中间体2-((3-溴-2-氯苯基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-1a的制备
Figure PCTCN2022074183-appb-000023
将I-1a-1(16.02g,54.24mmol,1.0eq)和I-1a-2(11.2g,54.24mmol,1.0eq)溶于无水THF(30mL)中,搅拌5min后,将KOBu-t(18.26g,162.72mmol,3.0eq)的无水THF(20mL)溶液加入其中,环境温度下搅拌1h。用饱和食盐水(200mL)淬灭反应,EtOAc(200mL)萃取2次。合并有机相,无水Na 2SO4干燥,粗品经硅胶柱(n-Hep/EtOAc(v/v)=3/1)分离后得到浅黄色固体I-1a。(23.70g,收率93.0%)。LC-MS MS-ESI(m/z)469.1[M+H] +
中间体2-((3-溴-2-氯苯基)氨甲酰基)-1-甲基-d 3-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-2a的制备
Figure PCTCN2022074183-appb-000024
将市售的I-2a-1-1(10.00g,44.8mmol,1.0eq),NaH(2.69g,112.0mmol,2.5eq),THF(40mL)在0℃下搅拌0.5h,加入I-2a-1-2(10.00g,69.0mmol,1.5eq)继续搅拌16h。反应液用乙酸乙酯(EtOAc,60mL)稀释,用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(DCM/MeOH(v/v)=98/2)分离后,得到淡黄色油状物I-2a-1-3。(7.00g,收率64.5%)。LC-MS MS-ESI(m/z)241.2[M+H] +
将自制的I-2a-1-3(7.00g,28.9mmol,1.0eq),正丁基锂(2.5M,13.9mL,1.2eq),无水THF(80mL)在-50℃以下搅拌1h,通入CO 2气体并同时继续搅拌1h。反应液升到室温,加入水(4mL),升温至70℃搅拌1h。反应液浓缩得到粗品,经柱层析(DCM/MeOH(v/v)=10/1)分离后,得到黄色固体I-2a-1-4。(6.50g,收率79.2%)。LC-MS MS-ESI(m/z)285.2[M+H] +
将自制的I-2a-1-4(2.00g,7.03mmol,1.0eq),草酰氯(1.07g,8.44mmol,1.2eq),催化量的DMF,DCM(10mL)在室温下搅拌1.5h,,加入MeOH(2mL)。 反应液浓缩得到粗品,经柱层析(DCM/MeOH(v/v)=98/2)分离后,得到黄色固体I-2a-1。(700.00mg,收率33.4%)。LC-MS MS-ESI(m/z)299.2[M+H] +
将自制的I-2a-1(700.00mg,2.35mmol,1.0eq),I-1a-2(485.21mg,2.35mmol,1.0eq),KOBu-t(659.18mg,5.88mmol,2.5eq),THF(40mL)在0℃下搅拌0.5h反应液用EtOAc(60mL)稀释,用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=5/1)分离后,得到白色固体I-2a。(300.00mg,收率27.0%)。LC-MS MS-ESI(m/z)472.1[M+H] +
中间体5-(叔丁基)2-甲基1-甲基-d 3-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-2,5-二羧酸酯I-2a-1的另种制备
Figure PCTCN2022074183-appb-000025
将市售的I-2a-1-1(15.85g,100.0mmol,1.0eq)溶解于异丙醇(150mL),加入K 2CO 3(41.46g,300.0mmol,3.0eq),冰浴冷却,分批加入氘代甲胺盐酸盐I-2a-1-2(10.57g,150.0mmol,1.5eq),在室温下机械搅拌2h。反应液加水(150mL)稀释,用DCM萃取(150mLx3),饱和NaHCO 3水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到黄色固体I-2a-1-3,直接用于下一步反应。(12.50g,收率80%)。LC-MS MS-ESI(m/z)157.1[M+H] +
将I-2a-1-3(9.50g,80.0mmol,1.0eq)溶解于甲醇(100mL)和甲酸(20mL)的混合溶液,室温下分批缓慢加入市售的还原铁粉(22.34g,400.0mmol,1.5eq),机械搅拌2h,抽滤,滤液旋干,得到浅褐色固体I-2a-1-4,直接用于下一步反应。(8.50g,收率84.2%)。LC-MS MS-ESI(m/z)127.1[M+H] +
将I-2a-1-4(8.50g,67.36mmol,1.0eq)悬浮于原甲酸三乙酯(100mL),加入甲酸(1mL),反应液加热至100℃反应3h,浓缩反应液,粗品经硅胶柱层 析(DCM/MeOH(v/v)=20/1)分离后,得到浅黄色固体I-2a-1-6。(6.40g,收率69.7%)。LC-MS MS-ESI(m/z)137.1[M+H] +
将I-2a-1-6(6.40g,47.0mmol,1.0eq)溶解于DCM(20mL),加入溴化苄(BnBr)(8.04g,47.0mmol,1.0eq),室温反应3h时,浓缩反应液,得到泡沫状固体I-2a-1-8,直接用于下一步反应。(15.50g,收率按100%计)。LC-MS MS-ESI(m/z)227.1[M+H] +
将I-2a-1-8(15.50g,47.0mmol,1.0eq)溶解于甲醇(50mL),缓慢加入NaBH 4(5.33g,141.0mmol,3.0eq),室温反应2h,加水稀释(100mL),DCM萃取(100mL),浓缩有机相,得到透明粘稠油状物I-2a-1-9,直接用于下一步反应。(16.20g,收率按100%计)。LC-MS MS-ESI(m/z)231.1[M+H] +
将I-2a-1-9(16.20g,47.0mmol,1.0eq)溶解于甲醇(100mL),加入甲酸铵(29.64g,470.0mmol,10.0eq)和氢氧化钯(20wt%,3.24g),反应液加热至80℃反应4h,冷却至室温,抽滤,滤液浓缩得到透明粘稠油状物I-2a-1-10,直接用于下一步反应。(14.50g,收率按100%计)。LC-MS MS-ESI(m/z)141.1[M+H] +
将I-2a-1-10(14.50g,47.0mmol,1.0eq)溶解于DCM(80mL),加入饱和NaHCO 3溶液(60mL),室温反应16h,分液,浓缩有机相,粗品经硅胶柱柱层析(DCM/MeOH(v/v)=20/1)分离后,得到透明粘稠油状物I-2a-1-11。(8.20g,收率72.6%)。LC-MS MS-ESI(m/z)241.2[M+H] +
将I-2a-1-11(8.20g,34.1mmol,1.0eq)溶解于THF(80mL),反应液用干冰/乙醇浴冷却至负七十度以下,滴加正丁基锂(2.5M,27.3mL,68.2mmol,2.0eq),滴加完毕,维持温度继续搅拌30min,向反应液通入CO 2,反应30min,滴加水淬灭(5mL),缓慢回至室温,浓缩有机相,粗品经硅胶柱层析(DCM/MeOH(v/v)=7/1)分离后,得到浅黄色固体I-2a-1-12。(4.50g,收率46.4%)。LC-MS MS-ESI(m/z)285.2[M+H] +
将I-2a-1-12(4.50g,15.8mmol,1.0eq)溶解于DCM(80mL),反应液用冰浴冷却,加入DMF(0.1mL),滴加草酰氯(4.01g,31.6mmol,2.0eq),滴加完毕,室温继续搅拌30min,再冰盐浴冷却至零度以下,向反应液加入甲醇(10mL),再加入TEA(5mL),搅拌10min,加水稀释,分液,浓缩有机相,粗品经硅胶柱层析(DCM/MeOH(v/v)=10/1)分离后,得到米白色固体I-2a-1。(3.60g,收率76.2%)。LC-MS MS-ESI(m/z)299.2[M+H] +
中间体3-溴-2-氯苯-4,5,6-d 3-胺I-2a-2的制备
Figure PCTCN2022074183-appb-000026
将市售的I-2a-2-1(25.00g,195.0mmol,1.0eq),NaBrO 3(29.40g,195.0mmol,1.0eq)加入到浓硫酸(100mL)和水(100mL)的混合溶剂中,并在40-45℃下搅拌,然后维持在该温度下缓慢加入K 2SO 4(241.00mg,1.39mmol,0.007eq),随后将该反应液继续在该温度下搅拌16h。HPLC监测反应完全,在20℃下,用Na 2CO 3将反应液的pH调到7-8。反应液用EtOAc萃取(3000mL x 2),合并有机相并用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到黄褐色固体I-2a-2-2。(36.50g,收率90.8%)。
将I-2a-2-2(36.50g,177.0mmol,1.0eq),Xantphos(10.30g,17.7mmol,0.1eq),Cs 2CO 3(115.00g,354.0mmol,2.0eq),Pd 2(dpa) 3(9.73g,10.6mmol,0.06eq),市售的I-2a-2-3(24.90g,213.0mmol,1.2eq)溶解于甲苯(60mL)中,并在100℃下搅拌16h。LC-MS监测反应完全,将反应液降温至25℃,并用水(1500mL)稀释,随后用EtOAc萃取(1500mL x 3),合并有机相并用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=100/1-90/10)分离后,得到黄色固体I-2a-2-4。(49.50g,粗品)。
将I-2a-2-4(49.50g,204.3mmol,1.0eq)溶解在DCM中,随后加入TFA(349.45g,3.06mol,15eq),该反应液在40℃下搅拌16h。TLC监测原料部分反应后,用水淬灭反应,而后用1M NaOH将反应液的pH调到9。反应液用EtOAc萃取(1000mL x 2),合并有机相并用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=100/1-92/8)分离后,得到黄色固体I-2a-2-5。(18.00g,收率61.9%)。
将I-2a-2-5(18.00g,127.0mmol,1.0eq)加入到N-氯代丁二酰亚胺(NCS,16.9g,127.0mmol,1.0eq)的DMF(50mL)溶液中,该反应液在75℃下搅拌1h。TLC监测反应完全,用水淬灭反应,随后反应液用EtOAc萃取(100mL),合并有机相并用饱和食盐水洗。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=100/1-90/1)分离后,得到黄色固体I-2a-2-6。(15.50g,收率69.7%)。
在25℃下,将亚硝酸叔丁酯(12.8g,124.0mmol,1.5eq)加入到CuBr 2(22.2g,99.2mmol,1.2eq)的乙腈(MeCN,100mL)溶液中,随后将该混合物升温至65℃,将自制的I-2a-2-6(14.5g,82.6mmol,1.0eq)的MeCN(50mL)溶液加入其中,该反应液继续在该温度下搅拌1h。TLC监测反应完全,将反应液降温至25℃,用NaHCO 3(500mL)水溶液淬灭反应,反应液用EtOAc萃取(500mL x 2),合并有机相并用硫代硫酸钠水溶液(300mL)和饱和食盐水(500mL)洗涤。有机相用无水MgSO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=100/1)分离后,得到白色固体I-2a-2-7。(9.30g,收率47.0%)。
将I-2a-2-7((9.88g,41.3mmol,1.0eq)溶解在乙醇(EtOH,90mL)和水(30mL)的混合溶剂中,向其中加入Fe粉(6.91g,124mmol,3.0eq)和HOAc(13.2mL,230mmol,5.6eq),该反应液在30℃下搅拌16h。TLC监测反应完全,用5M NaOH将反应液的pH调到7-8,反应液再用水(100mL)稀释,随后用EtOAc萃取(200mL),合并有机相并用饱和食盐水洗。有机相用无水Na 2SO 4干燥,浓缩得黄褐色油I-2a-2。(8.50g,收率97.8%)。 1H-NMR(400MHz,CDCl 3)δppm4.18(s,2H).
中间体2-((3-溴-2-氯苯基-4,5,6-d 3)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-3a的制备
Figure PCTCN2022074183-appb-000027
将前述制备的I-2a-2(1.05g,5.0mmol,1.0eq)和I-2a-1(1.49g,5.0mmol,1.0eq)溶解于无水THF(50mL),加入KOBu-t(1.12g,10.0mmol,2.0eq),在N 2保护下室温搅拌1h,用饱和食盐水(100mL)淬灭反应,EtOAc(80mL)萃取2次。合并有机相,无水Na 2SO 4干燥,粗品经硅胶柱层析(EtOAc/hexane(v/v)=1/3)分离后,得到白色固体I-3a。(950.00mg,收率39.9%)。LC-MS MS-ESI(m/z)475.1[M+H] +
中间体3-溴-2-氟苯-4,5,6-d 3-胺I-3a-2的制备
Figure PCTCN2022074183-appb-000028
将前述制备的I-2a-2-7(21.60g,89.8mmol,1.0eq)、氟化钾(10.40g,178.0mmol,2.0eq)和氟化铯(13.60g,89.8mmol,1.0eq)溶解于DMF(200mL)中,反应液在140℃下搅拌5h,LC-MS监测反应完全,随后将反应液冷却至25℃并用水(100mL)稀释,用EtOAc(150mL x 2)萃取,合并有机层,用饱和食盐水洗5次有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE)分离后,浓缩后得到的化合物再进一步经过制备HPLC(色谱柱型号:Welch Xtimate C18 10μm 250×50mm;流动相:水(千分之一的氨水+千分之一的碳酸氢胺)-乙腈;B%:25%-65%,23min)分离后,浓缩得到黄色液体I-3a-2-1。(14.50g,收率72.4%)。
将I-3a-2-1(13.00g,58.2mmol,1.0eq)溶解于EtOH(120mL)和水(40mL)的混合溶剂中,向其中加入铁粉(9.80g,174.0mmol,3.0eq)和HOAc(19.60g,325.0mmol,5.6eq),反应液在30℃下搅拌16h,LC-MS监测反应完全,抽滤,浓缩,随后将该混合物经柱层析(PE/EtOAc(v/v)=100/1-90/10)分离后,浓缩后得到的化合物再进一步经过制备HPLC(色谱柱型号:Phenomenex luna C18 10μm 250×80mm;流动相:水(千分之一的碳酸氢胺)-乙腈;B%:20%-60%,25min)分离后,浓缩得到黄色油状物I-3a-2。(5.1g,收率44.8%)。 1H-NMR(400MHz,DMSO)δppm 7.11(s,2H).
中间体2-((3-溴-2-氯苯基-4,5,6-d 3)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-14a的制备
Figure PCTCN2022074183-appb-000029
白色固体I-14a由市售的I-1a-1(788.32mg,2.67mmol,1.0eq)和前述制备的I-2a-2(560.00mg,2.67mmol,1.0eq)和KOBu-t(593.91mg,5.53mmol,2.0eq)按照中间体I-3a中的类似步骤制备而得。(780.00mg,收率61.9%)。LC-MS MS-ESI(m/z)472.1[M+H] +
中间体3-溴-2-甲基苯-4,5,6-d 3-胺I-4a-2的制备
Figure PCTCN2022074183-appb-000030
将前述制备的I-2a-2-2(11.00g,53.3mmol,1.0eq)溶解于EtOH(120mL)和水(90mL)的混合溶剂中,向其中加入铁粉(17.80g,320.0mmol,6.0eq)和NH 4Cl(17.10g,320.0mmol,6.0eq),反应液在60℃下搅拌16h,TLC监测反应完全,抽滤,浓缩,EtOAc洗涤(600mLx3),随后再浓缩得到黄色油状物I-4a-2-1。(45.0g,收率78.0%)。
将I-4a-2-1(19.00g,107.0mmol,1.0eq)、左旋樟脑磺酸(30.00g,129.0mmol,1.2eq)、亚硝酸叔丁酯(13.3g,129.0mmol,1.2eq)加入到TBAB(69.5g,215.0mmol,2.0eq)和CuBr 2(241.00mg,1.08mmol,0.01eq)的乙腈(MeCN,200mL)溶液中,该反应混合物在60℃下搅拌3h,HPLC监测反应完全,浓缩反应液,用NaHCO 3(500mL)水溶液调节pH为8,反应液用MTBE萃取(100mL x 3),合并有机相并用无水Na 2SO 4干燥,浓缩得到粗品,经减压蒸馏(146℃,油泵)后,得到黄色液体I-4a-2-2。(22.00g,收率42.4%)。
在一个三口瓶中,将I-4a-2-2(7.50g,31.2mmol,1.0eq)和CH 3I(4.88g,34.3mmol,1.1eq)溶解于无水THF中,氩气保护,将该反应液冷却到-78℃,并向其中缓慢加入LDA(2M,21.8mL),并维持在该温度下继续搅拌1h。HPLC检测反应得到71%的产物后,用中水淬灭反应,随后用EtOAc萃取(300mLx3),无水Na 2SO 4干燥,浓缩得红色液体I-4a-2-3(8.70g,粗品)。
将I-4a-2-3(7.00g,27.6mmol,1.0eq),Xantphos(1.60g,2.77mmol,0.1eq),Cs 2CO 3(18.00g,55.3mmol,2.0eq),Pd 2(dpa) 3(1.52g,1.7mmol,0.06eq)和市售的I-2a-2-3溶解于甲苯(60mL)中,并在100℃下搅拌16h。HPLC监测反应完全,将反应液降温至25℃,并用水稀释,随后用EtOAc萃取(100mL x 2),合并有机相并用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=98/1-2/1)分离后,得到黄色固体I-4a-2-4。(3.80g,47.4%)。
将I-4a-2-4(3.80g,13.1mmol,1.0eq)溶解在EtOAc(10mL)中,随后加入HCl/EtOAc(1.16g,13.1mmol,1.0eq),该反应液在40℃下搅拌24h。HPLC 监测原料反应完全,将反应液进行抽滤,滤饼用EtOA(10mL)洗涤,浓缩得到白色固体I-4a-2的盐酸盐。(2.77g,收率93.4%)。 1H-NMR(400MHz,CDCl 3)δppm 2.28(s,3H).
中间体2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-1b的制备
Figure PCTCN2022074183-appb-000031
将市售的I-1a-1(1.41g,4.77mmol,1.0eq)和市售的I-1b-2(1.21g,4.77mmol,1.0eq)溶于无水THF(30mL)中,搅拌5min后,将KOBu-t(1.61g,14.31mmol,3.0eq)的无水THF(20mL)溶液加入其中,环境温度下搅拌1h。用饱和食盐水(200mL)淬灭反应,EtOAc(200mL)萃取2次。合并有机相,无水Na 2SO4干燥,粗品经硅胶柱(n-Hep/EtOAc(v/v)=3/1)分离后得到白色固体I-1b。(2.23
g,收率90.5%)。LC-MS MS-ESI(m/z)517.2[M+H] +
中间体2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)氨甲酰基)-1-甲基-d 3-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-2b的制备
Figure PCTCN2022074183-appb-000032
将前述制备的I-2a-1(700.00mg,2.35mmol,1.0eq),I-1b-2(595.73mg,2.35mmol,1.0eq),KOBu-t(659.18mg,5.88mmol,2.5eq),THF(40mL)在0℃下搅拌0.5h反应液用EtOAc(60mL)稀释,用饱和食盐水洗1次。有机相用无水Na 2SO 4干燥,浓缩得到粗品,经柱层析(PE/EtOAc(v/v)=5/1)分离后,得到白色固体I-2b。(460.00mg,收率35.0%)。LC-MS MS-ESI(m/z)520.2[M+H] +
中间体2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基-4,5,6-d 3-胺I-3b-2的制备
Figure PCTCN2022074183-appb-000033
将前述制备的I-2a-2(2.09g,10.0mmol,1.0eq),联硼酸频那醇酯(3.81g,15.0mmol,1.5eq),Pd(dppf)Cl 2·CH 2Cl 2(408.32mg,0.5mmol,0.05eq),无水乙酸钾(2.94g,30.0mmol,3.0eq)和1,4-二氧六环(50mL)的混合液在N 2保护下加热到100℃并搅拌3h,冷却至环境温度。浓缩反应液,粗品经硅胶柱层析(EtOAc/hexane(v/v)=1/4)分离后,得到白色固体I-3b-2。(1.83g,收率71.3%)。LC-MS MS-ESI(m/z)257.2[M+H] +
中间体2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基-4,5,6-d 3)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-3b的制备
Figure PCTCN2022074183-appb-000034
将前述制备的I-3b-2(1.28g,5.0mmol,1.0eq)和I-2a-1(1.49g,5.0mmol,1.0eq)溶解于无水THF(50mL),加入KOBu-t(1.12g,10.0mmol,2.0eq),在N 2保护下室温搅拌1h,用饱和食盐水(100mL)淬灭反应,EtOAc(80mL)萃取2次。合并有机相,无水Na 2SO 4干燥,粗品经硅胶柱层析(EtOAc/hexane(v/v)=1/3)分离后,得到灰白色固体I-3b。(660.00mg,收率25.2%)。LC-MS MS-ESI(m/z)523.3[M+H] +
中间体2-((2-氟-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-7b的制备
Figure PCTCN2022074183-appb-000035
白色固体中间体I-7b是由市售的I-7b-2(1.00g,4.20mmol,1.0eq)和前述制备的中间体I-2a-1(1.24g,4.20mmol,1.0eq),KOBu-t(935.71mg,8.40mmol,2.0eq)按照中间体I-3b中的类似步骤制备而得。(1.00g,收率47.2%)。LC-MS MS-ESI(m/z)504.3[M+H] +
中间体1-(甲基-d 3)-2-((2-甲基-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-9b的制备
Figure PCTCN2022074183-appb-000036
白色固体中间体I-9b是由市售的I-9b-2(1.50g,6.44mmol,1.0eq)和前述制备的中间体I-2a-1(1.90g,4.44mmol,1.0eq),KOBu-t(1.44g,12.88mmol,2.0eq)按照中间体I-3b中的类似步骤制备而得。(1.80g,收率56.0%)。LC-MS MS-ESI(m/z)500.3[M+H] +
中间体2-氟-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基-4,5,6-d 3-胺I-14b-2的制备
Figure PCTCN2022074183-appb-000037
白色固体中间体I-14b-2是由前述制备的I-3a-2(1.00g,5.2mmol,1.0eq),联硼酸频那醇酯(1.97g,7.77mmol,1.5eq),Pd(dppf)Cl 2·CH 2Cl 2(409.11mg,0.5mmol,0.1eq),无水乙酸钾(1.52g,30.2mmol,3.0eq)按照中间体I-3b-2中的类似步骤制备而得。(880.00mg,收率70.5%)。LC-MS MS-ESI(m/z)241.2[M+H] +
中间体2-((2-氟-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基-4,5,6-d 3)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-14b的制备
Figure PCTCN2022074183-appb-000038
Figure PCTCN2022074183-appb-000039
将市售的I-1d-1(300.0g,1.31mol,1.0eq)溶于THF(2.5L)中,在氮气保护下将反应液降温至0℃,滴加BH 3-Me 2S(1.57mol,157.0mL,1.2eq)。滴毕,反 应液自然升温至20℃,并继续搅拌16h。TLC显示反应结束。将反应液降温至0℃,滴加MeOH(500mL)淬灭反应。滴毕,反应液直接浓缩干得无色油状物I-1d-2。(280.00g,收率99.4%)。 1H-NMR(400MHz,CDCl 3)δppm 4.48(t,J=5.5Hz,1H),3.82(s,2H),3.32(s,6H),1.81(d,2H),1.32-1.58(m,10H).
将I-1d-2(20.00g,93.3mmol,1.0eq)溶于DMF(150mL)中,加入氧化银(Ag2O,64.88g,280.0mmol,3.0eq)和氘代碘甲烷(CD 3I,50.00g,344.9mmol,3.7eq),加毕,用气球套住,防止CD 3I逸出,20℃搅拌32h。气相监测原料反应有30%剩余,直接处理。反应液过滤除去不溶物,滤液加入水(150mL),甲基叔丁基醚(MTBE,300mLx2)萃取,饱和食盐水(100mLx3)洗涤,无水硫酸镁干燥,过滤,滤液减压脱溶,粗品经硅胶柱柱层析(n-Hep/EtOAc(v/v)=100/1~30/1)分离得到黄色油状物I-1d-3。(14.00g,收率64.8%)。 1H-NMR(400MHz,CDCl 3)δppm 4.47(t,J=5.5Hz,1H),3.42(s,2H),3.31(s,6H),1.81(d,J=5.5Hz,2H),1.22-1.60(m,10H)。
将I-1d-3(14.00g,60.5mmol,1.0eq)溶于丙酮(Acetone,50mL)中,加入3M盐酸(60.52mL,3.0eq)。反应液在20℃搅拌16h。TLC显示反应结束。浓缩除去Acetone,再加入水后用MTBE(100mLx3)萃取。有机相合并,饱和食盐水(50mL)洗涤,无水硫酸镁干燥。粗品经硅胶柱柱层析(n-Hep/EtOAc(v/v)=100/1~30/1)分离得无色液体I-1d。(7.47g,66.6%)。 1H-NMR(400MHz,CDCl 3)δppm 9.81(t,1H),3.43(s,2H),2.57(d,2H),1.35-1.65(m,10H).
中间体2-(4-((甲氧基)甲基)双环[2.2.1]庚烷-1-基)乙醛I-2d的制备
Figure PCTCN2022074183-appb-000040
将市售的I-2d-1(300.00g,1.31mol,1.0eq)溶于THF(2.5L)中,在氮气保护下将反应液降温至0℃,滴加硼烷的二甲硫醚络合物(1.57mol,157mL,1.2eq)。滴毕,反应液自然升温至20℃,并继续搅拌16h。TLC显示反应结束。将反应液降温至0℃,滴加MeOH(500mL)淬灭反应。滴毕,反应液直接浓缩干得无色油状物I-2d-2(280.00g,收率99.4%)。 1H-NMR(400MHz,CDCl 3)δppm 4.48(t,J=5.5Hz,1H),3.82(s,2H),3.32(s,6H),1.81(d,2H),1.32-1.58(m,10H).
将I-2d-2(5.00g,23.33mmol,1.0eq)溶于N,N'-二甲基甲酰胺(50mL)中,依次加入碘甲烷(33.12g,233.30mmol,10.0eq)和Ag 2O(16.22g,70.00mmol,3.0 eq)。反应液在20℃搅拌16h。TLC显示有少量原料剩余,有主点生成。反应液倒入水(50mL)中,MTBE(50mLx3)萃取。有机相合并,饱和食盐水(30mLx3)洗涤,无水硫酸镁干燥。粗品经硅胶柱柱层析(n-Hep/EtOAc(v/v)=100/1~10/1)分离后,得无色油状物I-2d-3(4.50g,收率84.5%)。 1H-NMR(400MHz,CDCl 3)δppm 4.47(t,J=5.5Hz,1H),3.37(s,3H),3.42(s,2H),3.30(s,6H),1.82(d,J=5.5Hz,2H),1.22-1.60(m,10H).
将I-2d-3(4.50g,19.71mmol,1.0eq)溶于Acetone(40mL)中,加入2M盐酸(29.56mL,3.0eq)。反应液在25℃搅拌4h。TLC显示反应结束。浓缩除去丙酮,再加入水(30mL),MTBE(20mLx3)萃取。有机相合并,饱和食盐水(30mL)洗涤,无水硫酸镁干燥。粗品经硅胶柱柱层析(n-Hep/EtOAc(v/v)=100/1~10/1)分离后,得无色液体I-2d(2.31g,收率64.3%)。 1H-NMR(400MHz,CDCl 3)δppm 9.81(t,1H),3.43(s,2H),3.41(s,3H),2.56(d,2H),1.39-1.68(m,10H).
实施例1
4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-1
Figure PCTCN2022074183-appb-000041
Figure PCTCN2022074183-appb-000042
中间体:2-((2,2'-二氯-3'-(1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-1c的制备
将2-((3-溴-2-氯苯基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-1a(530.0mg,1.13mmol,1.0eq)溶于DCM(10mL)中,加入TFA(10mL),在环境温度下搅拌1h。浓缩反应液,将残余物溶解在1,4-二氧六环(10mL)中,加入2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基) 苯基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-1b(583.08mg,1.13mmol,1.0eq),PdCl 2(dcypf)(83.05mg,0.11mmol,0.1eq),无水Na 2CO 3(359.34mg,3.39mmol,3.0eq)和水(5mL),所得混合液用微波加热到110℃并反应1h,冷却至环境温度。用水(100mL)淬灭反应,用DCM(100mL)萃取3次,合并有机相,浓缩。粗品经硅胶柱柱层析(DCM/MeOH(v/v)=10/1)分离后,得到黄色固体I-1c。(363.0mg,收率47.3%)。LC-MS MS-ESI(m/z)679.2[M+H] +
中间体:2-((2,2'-二氯-3'-(5-(2-(4-(甲氧基羰基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-1h的制备
将I-1c(2.40g,3.53mmol,1.0eq)溶于DCM(60mL)中,加入TEA(3mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(1.04g,5.29mmol,1.5eq),所得混合液在环境温度下搅拌1h后,加入NaBH(OAc) 3(4.49g,21.18mmol,6.0eq)并继续搅拌16h。反应液用饱和NaHCO 3溶液淬灭,用DCM/MeOH(10/1,300mL萃取3次)。合并有机相,用无水Na 2SO 4干燥,浓缩。粗品经硅胶柱柱层析(DCM/MeOH(v/v)=20/1)分离后,得到黄色固体I-1h。(2.76g,收率92.1%)。LC-MS MS-ESI(m/z)859.3[M+H] +
中间体:4-(2-(2-((3'-(5-(叔丁氧羰基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2,2'-二氯-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-1i的制备
将中间体I-1h(2.67g,3.11mmol,1.0eq)溶于THF(300mL)中,一次性加入氢氧化锂一水合物(LiOH·H 2O,2.61g,62.20mmol,20.0eq)的水(300mL)溶液,所得混合液在环境温度下搅拌16h。浓缩除去THF,用1M稀盐酸调pH至5-6。过滤收集固体,烘干得到浅黄色固体I-1i。(1.84g,收率69.9%)。LC-MS MS-ESI(m/z)845.3[M+H] +
化合物:4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-1的制备
将中间体I-1i(1.84g,2.17mmol,1.0eq)溶于DCM(30mL)中,加入TFA(30mL),所得溶液在环境温度下搅拌1h。浓缩反应液,将残余物溶解在DCM(60mL)中,再次浓缩,得到的黄色固体直接用于下一阶段。将得到的三氟乙酸盐溶于DCM(60mL)中,加入TEA(3mL)和2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(602.23mg,3.25mmol,1.5eq)。所得混合液在环境温度下搅 拌1h后,加入NaBH(OAc) 3(2.76g,13.02mmol,6.0eq),继续搅拌16h。反应液用饱和NaHCO 3溶液淬灭,用DCM/MeOH(10/1,300mL萃取3次)萃取3次。合并有机相,用无水Na 2SO 4干燥,浓缩。粗品经硅胶柱柱层析(DCM/MeOH(v/v)=15/1)分离后,得到类白色固体I-1。(1.24g,收率62.4%)。LC-MS MS-ESI(m/z)914.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 12.00(s,1H),9.90(s,2H),8.37(d,J=8.2Hz,2H),7.49(t,J=7.9Hz,2H),7.14(d,J=7.5Hz,2H),3.90(s,6H),3.46-3.43(m,4H),3.32(s,2H),2.80(s,4H),2.68(s,4H),2.58(s,4H),1.88-1.81(m,2H),1.73-1.69(m,4H),1.57-1.21(m,16H),1.12(s,2H).
实施例2
4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-2
Figure PCTCN2022074183-appb-000043
化合物:4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-2的制备
淡黄色固体I-2是由中间体4-(2-(2-((3'-(5-(叔丁氧羰基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-氯-2'-氟-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-2i(400.00mg,0.48mmol,1.0eq,合成参考中间体I-1i的制备方法),TFA(5mL),TEA(1mL),2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(133.20mg, 0.72mmol,1.5eq)和NaBH(OAc) 3(610.56mg,2.88mmol,6.0eq)按照化合物I-1中的类似步骤制备而得。(95.00mg,收率22.0%)。LC-MS MS-ESI(m/z)898.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.93(s,1H),9.78(s,1H),8.34(d,J=8.3Hz,1H),8.07(t,J=7.3Hz,1H),7.48(t,J=7.9Hz,1H),7.31(t,J=7.9Hz,1H),7.21(d,J=7.5Hz,1H),7.16(t,J=6.7Hz,1H),3.89(s,3H),3.87(s,3H),3.41-3.39(m,4H),3.32(s,2H),2.78-2.71(m,4H),2.68-2.60(m,4H),2.56-2.51(m,4H),1.90-1.81(m,2H),1.72-1.69(m,4H),1.59-1.19(m,16H),1.11(s,2H).
实施例3
4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-3
Figure PCTCN2022074183-appb-000044
化合物:4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-3的制备
淡黄色固体I-3是由中间体4-(2-(2-((3'-(5-(叔丁氧羰基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-氯-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-3i(400.00mg,0.48mmol,1.0eq,合成参考中间体I-1i的制备方法),TFA(5mL),TEA(1mL),2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(133.2 mg,0.72mmol,1.5eq)和NaBH(OAc) 3(610.56mg,2.88mmol,6.0eq)按照化合物I-1中的类似步骤制备而得。(71.00mg,收率16.5%)。LC-MS MS-ESI(m/z)894.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.90(s,1H),9.73(s,1H),8.34(d,J=7.1Hz,1H),7.73(d,J=7.9Hz,1H),7.45(t,J=7.9Hz,1H),7.29(t,J=7.8Hz,1H),7.06(d,J=6.5Hz,1H),7.00(d,J=7.5Hz,1H),3.89(s,3H),3.86(s,3H),3.40(d,J=3.8Hz,4H),3.32(s,2H),2.79-2.70(m,4H),2.68-2.61(m,4H),2.56-2.50(m,4H),1.98(s,3H),1.91-1.80(m,2H),1.73-1.67(m,4H),1.58-1.17(m,16H),1.11(s,2H).
实施例4
4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-4
Figure PCTCN2022074183-appb-000045
化合物:4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-4的制备
淡黄色固体I-4是由中间体4-(2-(2-((3'-(5-(叔丁氧羰基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-氯-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-4i(400.00mg,0.48mmol,1.0eq,合成参考中间体I-1i的制备方法),TFA(5mL),TEA(1mL),2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(133.2 mg,0.72mmol,1.5eq)和NaBH(OAc) 3(610.56mg,2.88mmol,6.0eq)按照化合物I-1中的类似步骤制备而得。(64.00mg,收率14.9%)。LC-MS MS-ESI(m/z)894.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.90(s,1H),9.72(s,1H),8.35(dd,J=8.2,1.4Hz,1H),7.73(d,J=7.8Hz,1H),7.45(t,J=7.9Hz,1H),7.29(t,J=7.8Hz,1H),7.06(dd,J=7.6,1.5Hz,1H),7.00(d,J=7.6Hz,1H),3.89(s,3H),3.86(s,3H),3.40(s,4H),3.32(s,2H),2.78-2.70(m,4H),2.69-2.60(m,4H),2.56-2.50(m,4H),1.98(s,3H),1.91-1.80(m,2H),1.71-1.67(m,4H),1.58-1.19(m,16H),1.12(s,2H).
实施例5
4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-5
Figure PCTCN2022074183-appb-000046
中间体:2-((2,2'-二氯-3'-(1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-2c的制备
将2-((3-溴-2-氯苯基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-2a(1.10g,2.33mmol,1.0eq)溶于DCM(10mL)中,加入TFA(10mL),在环境温度下搅拌30min。浓缩反应液,将残余物溶解在1,4-二氧六环(10mL)中,加入2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁 酯I-2b(950.00mg,1.82mmol,1.0eq),PdCl 2(dcypf)(137.51mg,0.18mmol,0.1eq),无水Na 2CO 3(866.40mg,8.19mmol,4.5eq)和水(5mL)按照中间体I-1c中的类似步骤制备而得白色固体I-2c。(620.00mg,收率49.7%)。LC-MS MS-ESI(m/z)685.4[M+H] +
中间体:2-((2,2'-二氯-3'-(5-(2-(4-(甲氧基羰基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-2h的制备
将I-2c(620.00mg,0.89mmol,1.0eq)溶于DCM(20mL)中,加入TEA(1mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(350.80mg,1.78mmol,2.0eq),所得混合液在环境温度下搅拌30min后,加入NaBH(OAc) 3(954.00mg,4.50mmol,5.0eq)按照中间体I-1h中的类似步骤制备而得白色固体I-2h。(490.00mg,收率63.6%)。LC-MS MS-ESI(m/z)865.4[M+H] +
中间体:4-(2-(2-((3'-(5-(叔丁氧羰基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2,2'-二氯-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-2i的制备
称量LiOH·H 2O(239.40mg,5.70mmol),溶解于水(20mL)。将中间体I-2h(490.00mg,0.57mmol,1.0eq)溶于THF(20mL)中,再加入上述氢氧化锂溶液,室温搅拌过夜。浓缩除去THF,用1M稀盐酸调pH至5-6,EtOAc(30mL)萃取2次。合并有机相,无水Na 2SO 4干燥,浓缩,得到中间体I-2i。(300.00mg,收率57.2%)。LC-MS MS-ESI(m/z)920.6[M+H] +
化合物:4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-5的制备
将中间体I-2i(200.00mg,0.22mmol,1.0eq)溶于DCM(5mL)中,加入TFA(5mL),所得溶液在环境温度下搅拌30min。浓缩反应液,将残余物溶解在DCM中,再次浓缩,得到的黄色固体直接用于下一阶段。将得到的三氟乙酸盐溶于DCM中,加入TEA(1mL)和2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(119.90mg,0.44mmol,2.0eq)。所得混合液在环境温度下搅拌1h后,加入NaBH(OAc) 3(237.01mg,1.10mmol,5.0eq),继续搅拌16h。反应液用水淬灭,分离有机相,用无水Na 2SO 4干燥,浓缩。粗品经制备(DCM/MeOH(v/v)=10/1)分离后,得到I-5。(105.00mg,收率51.8%)。LC-MS MS-ESI(m/z)920.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.89(s,2H),8.38(d,J=7.5Hz,2H),7.48(t,J=7.9Hz,2H),7.14(dd,J=7.6Hz,1.4Hz,2H),3.46-3.38(m,4H), 3.32(s,2H),2.77-2.71(m,4H),2.69-2.59(m,4H),2.56-2.51(m,4H),1.91-1.80(m,2H),1.76-1.64(m,4H),1.57-1.18(m,16H),1.12(s,2H).
实施例6
4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-6
Figure PCTCN2022074183-appb-000047
化合物:4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-6的制备
将中间体I-2i(100.00mg,0.11mmol,1.0eq)溶解于DCM(5mL),一次性加入TFA(5mL),室温下搅拌30min,旋干反应液得到浅黄透明油状物;将浅黄色透明油状物溶解于DCM,加入TEA(1mL)和2-(4-((甲氧基)甲基)双环[2.2.1]庚烷-1-基)乙醛I-2d(60.00mg,0.22mmol,2eq),室温搅拌30min;加入NaBH(OAc) 3(166.5mg,0.55mmol,5.0eq)按照化合物I-5中的类似步骤制备而得化合物I-6。(50.00mg,收率49.5%)。LC-MS MS-ESI(m/z)917.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.89(s,2H),8.38(d,J=7.8Hz,2H),7.48(t,J=7.9Hz,2H),7.14(d,J=6.5Hz,2H),3.38-3.43(m,4H),3.32(s,2H),3.23(s,3H),2.79-2.70(m,4H),2.69-2.59(m,4H),2.56-2.51(m,4H),1.91-1.80(m,2H),1.76-1.64(m,4H),1.57-1.20(m,16H),1.11(s,2H).
实施例7
4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-7
Figure PCTCN2022074183-appb-000048
中间体:2-((2'-氯-2-氟-3'-(1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-7c的制备
将2-((3-溴-2-氯苯基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-2a(718.7mg,1.52mmol,1.0eq)溶于DCM(10mL)中,加入TFA(10mL),在环境温度下搅拌30min。浓缩反应液,将残余物溶解在1,4-二氧六环(10mL)中,加入自制的2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-7b(766.54mg,1.52mmol,1.0eq),PdCl 2(dcypf)(114.59mg,0.152mmol,0.1eq),无水Na 2CO 3(722.00mg,6.80mmol,4.5eq)按照中间体I-1c中的类似步骤制备而得I-7c。(625.00mg,收率59.2%)。LC-MS MS-ESI(m/z)669.3[M+H] +
中间体:2-((2'-氯-2-氟-3'-(5-(2-(4-(甲氧基羰基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-7h的制备
将I-7c(625.00mg,0.94mmol,1.0eq)溶于DCM(20mL)中,加入TEA(1mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(370.50mg,1.88mmol, 2.0eq),所得混合液在环境温度下搅拌30min后,加入NaBH(OAc) 3(996.00mg,4.7mmol,5.0eq)按照中间体I-1h中的类似步骤制备而得黄色固体I-7h。(600.00mg,收率75.1%)。LC-MS MS-ESI(m/z)849.4[M+H] +
中间体:4-(2-(2-((3'-(5-(叔丁氧羰基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-氯-2'-氟-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-7i的制备
黄色中间体I-7i是由中间体I-7h(600.00mg,0.71mmol,1.0eq)和LiOH·H 2O(314.77mg,7.10mmol))按照中间体I-2i中的类似步骤制备而得。(450.00mg,收率75.9%)。LC-MS MS-ESI(m/z)835.4[M+H] +
化合物:4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-7的制备
白色化合物I-7是由中间体I-7i(225mg,0.27mmol,1.0eq),TFA(5mL),TEA(1mL)和2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(147.15mg,0.54mmol,2.0eq),NaBH(OAc) 3(290.88mg,1.35mmol,5.0eq)按照化合物I-5中的类似步骤制备而得。(120.00mg,收率49.1%)。LC-MS MS-ESI(m/z)904.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.94(s,1H),9.79(s,1H),8.34(d,J=8.0Hz,1H),8.06(t,J=7.4Hz,1H),7.49(t,J=7.9Hz,1H),7.31(t,J=7.9Hz,1H),7.21(d,J=7.5Hz,1H),7.16(t,J=6.8Hz,1H),3.45-3.36(m,4H),3.32(s,2H),2.77-2.71(m,4H),2.69-2.59(m,4H),2.57-2.52(m,4H),1.91-1.80(m,2H),1.76-1.64(m,4H),1.58-1.19(m,16H),1.12(s,2H).
实施例8
4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-8
Figure PCTCN2022074183-appb-000049
化合物:4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-8的制备
白色化合物I-8是由中间体I-7i(225.00mg,0.27mmol,1.0eq),TFA(5mL),TEA(1mL)和2-(4-((甲氧基)甲基)双环[2.2.1]庚烷-1-基)乙醛I-2d(143.05mg,0.54mmol,2.0eq),NaBH(OAc) 3(290.88mg,1.35mmol,5.0eq)按照化合物I-5中的类似步骤制备而得。(120.00mg,收率50.1%)。LC-MS MS-ESI(m/z)901.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.93(s,1H),9.79(s,1H),8.34(d,J=8.2Hz,1H),8.06(t,J=7.8Hz,1H),7.49(t,J=8.0Hz,1H),7.31(t,J=7.7Hz,1H),7.21(d,J=7.3Hz,1H),7.16(t,J=6.8Hz,1H),3.44-3.38(m,4H),3.33(s,2H),3.23(s,3H),2.77-2.71(m,4H),2.69-2.61(m,4H),2.56-2.51(m,4H),1.91-1.80(m,2H),1.76-1.64(m,4H),1.61-1.20(m,16H),1.12(s,2H).
实施例9
4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-9
Figure PCTCN2022074183-appb-000050
中间体:2-((2'-氯-2-甲基-3'-(1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-9c的制备
黄色固体中间体I-9c是由I-2a(1.34g,3.60mmol,1.0eq),中间体I-9b(1.80g,3.60mmol,1.0eq),PdCl 2(dcypf)(271.40mg,0.36mmol,0.1eq),无水Na 2CO 3(1.72g,16.2mmol,4.5eq)按照中间体I-1c中的类似步骤制备而得。(1.40g,收率58.5%)。LC-MS MS-ESI(m/z)665.3[M+H] +
中间体:2-((2'-氯-3'-(5-(2-(4-(甲氧基羰基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-9h的制备
黄色中间体I-9h是由中间体I-9c(700.00mg,1.05mmol,1.0eq)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(414.70mg,2.10mmol,2.0eq),TEA(1mL),NaBH(OAc) 3(1.12g,5.3mmol,5.0eq)按照中间体I-1h中的类似步骤制备而得。(650.00mg,收率73.2%)。LC-MS MS-ESI(m/z)845.4[M+H] +
中间体:4-(2-(2-((3'-(5-(叔丁氧羰基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-氯-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-9i的制备
黄色中间体I-9i是由中间体I-9h(650.00mg,0.77mmol,1.0eq)和LiOH·H 2O(340.86mg,7.70mmol)按照中间体I-2i中的类似步骤制备而得。(460.00mg,收率71.9%)。LC-MS MS-ESI(m/z)831.4[M+H] +
化合物:4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-9的制备
白色化合物I-9是由中间体I-9i(230.00mg,0.28mmol,1.0eq),TFA(5mL),TEA(1mL)和2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(153.35mg,0.56mmol,2.0eq),NaBH(OAc) 3(310.28mg,1.40mmol,5.0eq)按照化合物I-5中的类似步骤制备而得。(125.00mg,收率49.6%)。LC-MS MS-ESI(m/z)900.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.90(s,1H),9.72(s,1H),8.34(dd,J=8.2,1.3Hz,1H),7.73(d,J=7.9Hz,1H),7.45(t,J=7.9Hz,1H),7.29(t,J=7.8Hz,1H),7.06(dd,J=7.6,1.4Hz,1H),7.00(d,J=7.4Hz,1H),3.42-3.38(m,4H),3.32(s,2H),2.77-2.70(m,4H),2.68-2.60(m,4H),2.55-2.50(m,4H),1.98(s,3H),1.91-1.80(m,2H),1.76-1.65(m,4H),1.57-1.21(m,16H),1.12(s,2H).
实施例10
4-(2-(2-((2-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-10
Figure PCTCN2022074183-appb-000051
白色化合物I-10是由中间体I-9i(230.00mg,0.28mmol,1.0eq),TFA(5mL),TEA(1mL)和2-(4-((甲氧基)甲基)双环[2.2.1]庚烷-1-基)乙醛I-2d(151.12mg,0.56mmol,2.0eq),NaBH(OAc) 3(310.28mg,1.40mmol,5.0eq)按照化合物I-5中的类似步骤制备而得。(124.00mg,收率49.3%)。LC-MS MS-ESI(m/z)897.5 [M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.90(s,1H),9.73(s,1H),8.34(d,J=6.9Hz,1H),7.72(d,J=8.2Hz,1H),7.45(t,J=7.9Hz,1H),7.30(t,J=8.0Hz,1H),7.06(dd,J=7.6,1.4Hz,1H),7.00(d,J=7.7Hz,1H),3.44-3.37(m,4H),3.33(s,2H),3.24(s,3H),2.79-2.70(m,4H),2.69-2.60(m,4H),2.56-2.51(m,3H),1.98(s,3H),1.91-1.80(m,2H),1.74-1.65(m,4H),1.57-1.21(m,16H),1.12(s,2H).
实施例11
4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-11
Figure PCTCN2022074183-appb-000052
中间体:2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-11e的制备
将中间体I-9c(350.00mg,0.53mmol,1.0eq)和2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(194.60mg,1.05mmol,2.0eq)溶解于DCM(20mL),加入TEA(1mL),室温搅拌30min;加入NaBH(OAc) 3(560.00mg,2.7mmol,5.0eq),室温反应过夜。加水(20mL)淬灭,分离有机相,干燥旋干,粗品经硅胶柱柱层析(DCM/MeOH(v/v)=20/1)分离后,得到中间体I-9e。(325.00mg,收率74.2%)。LC-MS MS-ESI(m/z)834.5[M+H] +
中间体:4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙 基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸甲酯I'-11的制备
将中间体I-9e(325.00mg,0.39mmol,1.0eq)溶解于DCM(5mL),一次性加入TFA(5mL),室温下搅拌30min,旋干反应液得到浅黄透明油状物;将浅黄色透明油状物溶解于DCM,加入TEA(1mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(212.55mg,0.77mmol,2.0eq),室温搅拌30min;加入NaBH(OAc) 3(420.14mg,1.95mmol,5.0eq),室温反应过夜。加水(20mL)淬灭,分离有机相,干燥旋干,粗品经制备TLC(DCM/MeOH(v/v)=10/1)分离后,得到中间体I'-11。(210.00mg,收率58.9%)。LC-MS MS-ESI(m/z)914.5[M+H] +
化合物:4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-11的制备
称量LiOH·H 2O(95.82mg,2.3mmol),溶解于水(10mL);将中间体I'-11(210.00mg,0.23mmol,1.0eq)溶解于THF(10mL),再加入上述LiOH·H 2O溶液,室温搅拌过夜;旋干THF,水相调节pH至弱酸性,EtOAc(30mL)萃取2次。合并有机相,无水Na 2SO 4干燥,浓缩,粗品经制备TLC(DCM/MeOH(v/v)=10/1)分离后,得到化合物I-11。(122.00mg,收率58.9%)。LC-MS MS-ESI(m/z)900.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.90(s,1H),9.73(s,1H),8.34(d,J=8.3Hz,1H),7.73(d,J=8.1Hz,1H),7.45(t,J=8.0Hz,1H),7.30(t,J=8.0Hz,1H),7.06(d,J=6.5Hz,1H),7.00(d,J=7.4Hz,1H),3.43-3.37(m,4H),3.32(s,2H),2.79-2.70(m,4H),2.70-2.61(m,4H),2.56-2.51(m,4H),1.98(s,3H),1.92-1.79(m,2H),1.77-1.65(m,4H),1.59-1.20(m,16H),1.12(s,2H).
实施例12
4-(2-(2-((2'-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-12
Figure PCTCN2022074183-appb-000053
中间体:2-((2'-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-12e的制备
将中间体I-9c(350.00mg,0.53mmol,1.0eq)和2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙醛I-2d(191.12mg,1.05mmol,2.0eq)溶解于DCM(20mL),加入TEA(1mL),室温搅拌30min;加入NaBH(OAc) 3(560.00mg,2.7mmol,5.0eq),室温反应过夜。加水(20mL)淬灭,分离有机相,干燥旋干,粗品经硅胶柱柱层析(DCM/MeOH(v/v)=20/1)分离后,得到中间体I-12e。(324.3mg,收率72.6%)。LC-MS MS-ESI(m/z)831.5[M+H] +
中间体:4-(2-(2-((2'-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸甲酯I'-12的制备
将中间体I-9e(324.03mg,0.39mmol,1.0eq)溶解于DCM(5mL),一次性加入TFA(5mL),室温下搅拌30min,旋干反应液得到浅黄透明油状物;将浅黄色透明油状物溶解于DCM,加入TEA(1mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(212.55mg,0.77mmol,2.0eq),室温搅拌30min;加入NaBH(OAc) 3(420.14mg,1.95mmol,5.0eq),室温反应过夜。加水(20mL)淬灭,分离有机相,干燥旋干,粗品经制备TLC(DCM/MeOH(v/v)=10/1)分离后,得到中间体I'-12。(208.00mg,收率58.5%)。LC-MS MS-ESI(m/z)911.5[M+H] +
化合物:4-(2-(2-((2'-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-12的制备
称量LiOH·H 2O(100.85mg,2.43mmol),溶解于水(10mL);将中间体I'-12(215.00mg,0.24mmol,1.0eq)溶解于THF(10mL),再加入上述LiOH·H 2O溶液,室温搅拌过夜;旋干THF,水相调节pH至弱酸性,EtOAc(30mL)萃取2次。合并有机相,无水Na 2SO 4干燥,浓缩,粗品经制备TLC(DCM/MeOH(v/v)=10/1)分离后,得到化合物I-12。(10.00mg,收率4.64%)。LC-MS MS-ESI(m/z)897.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.98(s,1H),9.72(s,1H),8.33(d,J=8.2Hz,1H),7.72(d,J=7.5Hz,1H),7.45(t,J=7.9Hz,1H),7.30(t,J=7.8Hz,1H),7.06(d,J=7.6Hz,1H),7.00(d,J=7.4Hz,1H),3.44-3.38(m,4H),3.32(s,2H),3.24(s,3H),2.78-2.70(m,4H),2.68-2.62(m,4H),2.55-2.51(m,4H),1.98(s,3H),1.91-1.79(m,2H),1.75-1.64(m,4H),1.56-1.19(m,16H),1.11(s,2H).
实施例13
4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-13
Figure PCTCN2022074183-appb-000054
中间体:2-((2,2'-二氯-3'-(1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢 -5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-13c的制备
将2-((3-溴-2-氯苯基-4,5,6-d 3)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-3a(950.00mg,1.99mmol,1.0eq)溶于DCM(10mL)中,加入TFA(10mL),在环境温度下搅拌30min。浓缩反应液,将残余物溶解在1,4-二氧六环(10mL)中,加入2-((2-氯-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基-4,5,6-d 3)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-3b(660.00mg,1.26mmol,1.0eq),PdCl 2(dcypf)(95.20mg,0.13mmol,0.1eq),无水Na 2CO 3(400.60mg,3.78mmol,3.0eq)和水(5mL),所得混合液用微波加热到110℃并反应3h,冷却至环境温度。浓缩反应液,粗品经硅胶柱柱层析(DCM/MeOH(v/v)=10/1)分离后,得到白色固体I-13c。(420.00mg,收率48.2%)。LC-MS MS-ESI(m/z)691.4[M+H] +
中间体:2-((2,2'-二氯-3'-(5-(2-(4-(甲氧基羰基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-13h的制备
将I-13c(420.00mg,0.60mmol,1.0eq)溶于DCM(20mL)中,加入TEA(1mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(130.10mg,0.66mmol,1.1eq),所得混合液在环境温度下搅拌30min后,加入NaBH(OAc) 3(636.00mg,3.0mmol,5.0eq),室温反应过夜。加水(20mL)淬灭,分离有机相,干燥旋干,粗品经硅胶柱柱层析(DCM/MeOH(v/v)=20/1)分离后,得到黄色固体I-13h。(390.00mg,收率73.7%)。LC-MS MS-ESI(m/z)871.5[M+H] +
中间体:4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸甲酯I'-13的制备
将中间体I-13h(390.00mg,0.44mmol,1.0eq)溶解于DCM(5mL),一次性加入TFA(5mL),室温下搅拌30min,旋干反应液得到浅黄透明油状物;将浅黄色透明油状物溶解于DCM/MeOH(v/v,10/1),加入TEA(1mL)和4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(99.45mg,0.53mmol,1.2eq),室温搅拌30min;加入NaBH(OAc) 3(474.13mg,2.23mmol,5.0eq),室温反应过夜。加水(20mL)淬灭,分离有机相,干燥旋干,粗品经制备TLC(DCM/MeOH(v/v)=20/1)分离后,得到中间体I'-13。(140.00mg,收率33.2%)。LC-MS MS-ESI(m/z)940.6[M+H] +
化合物:4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙 基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-13的制备
称量LiOH·H 2O(210.00mg,5.00mmol),溶解于水(50mL),配制成溶液;将中间体I'-13(140.00mg,0.14mmol,1.0eq)溶解于THF(20mL),再加入上述LiOH·H 2O的溶液(20mL),室温搅拌过夜,浓缩THF,水相调节pH至弱酸性,析出白色固体,抽滤,烘干得到I-13。(84.00mg,收率60.9%)。LC-MS MS-ESI(m/z)926.6[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.89(s,2H),3.42-3.30(m,6H),2.79-2.70(m,4H),2.69-2.62(m,4H),2.55-2.51(m,4H),1.90-1.79(m,2H),1.75-1.66(m,4H),1.56-1.43(m,8H),1.40(s,2H),1.36-1.22(m,6H),1.11(s,2H).
实施例14
4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-14
Figure PCTCN2022074183-appb-000055
中间体:2-((2'-氯-2-氟-3'-(1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-14c的制备
黄色中间体I-14c是由中间体I-14a(780.0mg,1.65mmol,1.0eq),中间体I-14b(766.78mg,1.52mmol,1.0eq),PdCl 2(dcypf)(114.62mg,0.15mmol,0.1eq),无水Na 2CO 3(724.00mg,6.8mmol,4.5eq)按照中间体I-1c中的类似步骤制备 而得。(625.65mg,收率59.2%)。LC-MS MS-ESI(m/z)669.3[M+H] +
中间体:2-((2'-氯-2-氟-3'-(5-(2-(4-(甲氧基羰基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-甲酸叔丁酯I-14h
黄色中间体I-14h是由中间体I-14c(625.65mg,0.94mmol,1.0eq)和市售的4-(2-氧代乙基)双环[2.2.1]庚烷-1-羧酸甲酯I-1f(370.80mg,1.88mmol,2.0eq),TEA(1mL),NaBH(OAc) 3(996.7mg,4.7mmol,5.0eq)按照中间体I-1h中的类似步骤制备而得。(601.08mg,收率75.1%)。LC-MS MS-ESI(m/z)849.4[M+H] +。中间体:4-(2-(2-((3'-(5-(叔丁氧羰基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-氯-2'-氟-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-14i
黄色中间体I-14i由中间体I-9h(601.08mg,0.71mmol,1.0eq)和LiOH·H 2O(315.86mg,7.10mmol)按照中间体I-2i中的类似步骤制备而得。(451.32mg,收率75.9%)。LC-MS MS-ESI(m/z)835.4[M+H] +
化合物:4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-14的制备
白色化合物I-14是由中间体I-14i(225.00mg,0.27mmol,1.0eq),TFA(5mL),TEA(1mL)和2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙醛I-1d(147.15mg,0.54mmol,2.0eq),NaBH(OAc) 3(290.88mg,1.35mmol,5.0eq)按照化合物I-5中的类似步骤制备而得。(60.00mg,收率24.6%)。LC-MS MS-ESI(m/z)904.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.93(s,1H),9.78(s,1H),3.89(s,3H),3.87(s,3H),3.42-3.38(m,4H),3.32(s,2H),2.78-2.70(m,4H),2.69-2.61(m,4H),2.57-2.51(m,4H),1.91-1.80(m,2H),1.76-1.65(m,4H),1.56-1.21(m,16H),1.11(s,2H).
实施例15
4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-15
Figure PCTCN2022074183-appb-000056
化合物:4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸I-15的制备
白色化合物I-15是由中间体I-14i(225.00mg,0.27mmol,1.0eq),TFA(5mL),TEA(1mL)和2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙醛I-2d(143.05mg,0.54mmol,2eq),NaBH(OAc) 3(290.88mg,1.35mmol,5.0eq)按照化合物I-5中的类似步骤制备而得。(60.00mg,收率25.0%)。LC-MS MS-ESI(m/z)901.5[M+H] +1H-NMR(400MHz,DMSO-d 6)δppm 9.94(s,1H),9.79(s,1H),3.90(s,3H),3.87(s,3H),3.43-3.38(m,4H),3.33(s,2H),3.24(s,3H),2.78-2.70(m,4H),2.69-2.60(m,4H),2.57-2.51(m,4H),1.91-1.81(m,2H),1.77-1.65(m,4H),1.58-1.22(m,16H),1.12(s,2H).
体外生物学评价
本检测方法用于本发明所述化合物的体外生物学活性评价,包括体外蛋白水平结合抑制活性评价方法和细胞水平生物学功能活性评价方法。
本检测的目的在于综合评价不同化合物对体外液相PD-1和PD-L1及CD80和PD-L1结合的抑制活性和对细胞模型上PD-1和PD-L1结合后抑制T细胞活化信号的阻断影响。
实施例A 体外PD-1和PD-L1结合的抑制活性评价
实验主要原理
均相时间分辨荧光法(HTRF):本方法应用了融合表达hFc标签的重组人PD-L1蛋白及融合表达His标签的重组人PD-1蛋白,二者为可相互作用的配体和受体。当分别使用含Eu元素鳌合标记物的anti-hFc抗体和含XL665荧光素标 记的anti-His抗体与上述两个对应标签结合,且通过320nm波长激光激发后,由于配体-受体结合使得能量能够从Eu元素转移到XL665荧光素上,激发后者发出665nm波长的发射光。而当加入PD-L1与PD-1相互作用的抑制剂时,配体与受体的结合被破坏,使得Eu和XL665距离较远,能量不能转移,XL665不会被激发。
实验材料与设备
带His标签的重组人PD-1蛋白(His-PD-1蛋白,Cat#:10377-H08H-50)、重组人PD-L1-Fc融合蛋白(PD-L1-Fc融合蛋白,Cat#:10084-H02H-100)购自义翘神州公司(Sino Biological Inc.),anti-hFc-Eu 3+抗体、anti-His-XL665抗体购自Cisbio公司,其它相关试剂如稀释缓冲液(Diluent buffer 5,Cat#:62DL5DDC)、检测缓冲液(PPI-Europium detection buffer,Cat#:61DB9RDF)等均从Cisbio公司购买。荧光检测仪器Tecan(Spark 10M)从瑞士Tecan公司购买。
实验主要过程
实验过程按照检测试剂使用说明书要求的流程进行。流程如下:
(1)实验准备:用稀释缓冲液将测试化合物稀释成不同浓度梯度(在20μL最终反应体系中最高终浓度为10μM),将His-PD-1蛋白稀释成800nM(在20μL最终反应体系中终浓度100nM),PD-L1-Fc融合蛋白稀释成16nM(终浓度为2nM);用检测缓冲液分别按试剂要求将anti-His-XL665抗体和anti-hFc-Eu 3+抗体稀释20倍和100倍。
(2)先将5μL测试化合物、2.5μL PD-L1-Fc融合蛋白和2.5μL His-PD-1蛋白溶液混匀后,室温孵育15min;随后向该体系加入5μL anti-His-XL665抗体和5μL anti-hFc-Eu 3+抗体,继续孵育3h后检测。
(3)检测反应同时设置有对照组,包括未添加测试化合物的0%抑制阳性对照、未添加PD-1蛋白的100%抑制阴性对照。所有检测采用复孔。
(4)使用荧光检测仪Tecan(Spark 10M)检测每孔的荧光信号,激发波长为320nm,检测的发射波长分别为620nm和665nm。PD-1和PD-L1相互结合的强度参照荧光信号比值Em665/Em620。
(5)测试化合物的结合抑制率计算公式:抑制率(%)=[1–(检测孔荧光信号比值–100%抑制阴性对照荧光信号比值)]/(0%抑制阳性对照荧光信号比值–100%抑制阴性对照荧光信号比值)×100%。不同浓度梯度的测试化合物分别计算PD-1/PD-L1结合抑制率后,计算50%抑制浓度(IC 50)。本发明代表性化合物体外抑制PD-1和PD-L1结合的IC 50数据见下表2:
表2本发明代表性化合物抑制体外PD-1/PD-L1结合的IC 50数据
Figure PCTCN2022074183-appb-000057
由上述结果可见,本发明所述的化合物具有很好的抑制体外PD-1/PD-L1的活性,本发明所述的通式(I)的化合物同样具有抑制PD-1/PD-L1的活性。
实施例B 体外CD80和PD-L1结合的抑制活性评价
实验主要原理
除PD-1外,PD-L1也可通过与CD80结合,发挥免疫抑制活性。同样地,体外CD80与PD-L1结合或结合抑制实验也可通过均相时间分辨荧光法(HTRF)进行检测。当使用anti-hFc-Eu 3+抗体和anti-His-XL665抗体分别与融合表达于PD-L1的hFC标签及融合表达于CD80的His标签结合,且320nm波长激光激发后,由于PD-L1与CD80的结合使得能量可以从Eu元素转移到XL665荧光素上,激发后者发光。当加入PD-L1与CD80相互作用的抑制剂时,二者的结合被破坏,使得Eu和XL665距离较远,能量不能转移,XL665不会被激发。
实验材料与设备
带His标签的重组人CD80蛋白(His-CD80蛋白,Cat#:10698-H08H-100)、重组人PD-L1-Fc融合蛋白(PD-L1-Fc融合蛋白,Cat#:10084-H02H-100)购自义翘神州公司(Sino Biological Inc.),anti-hFc-Eu 3+抗体、anti-His-XL665抗体购自Cisbio公司,其它相关试剂如稀释缓冲液(Diluent buffer 5,Cat#:62DL5DDC)、检测缓冲液(PPI-Europium detection buffer,Cat#:61DB9RDF)等均从Cisbio公司购买。荧光检测仪器Tecan(Spark 10M)从瑞士Tecan公司购买。
实验主要过程
实验过程按照检测试剂使用说明书要求的流程进行(Invitrogen)。流程如下:
(1)实验准备:用稀释缓冲液将测试化合物稀释成不同浓度梯度(在20μL最终反应体系中最高终浓度为10μM),将His-CD80蛋白稀释成800nM(在20μL最终反应体系中终浓度100nM),PD-L1-Fc融合蛋白稀释成16nM(终浓度为2nM);用检测缓冲液分别按试剂要求将anti-His-XL665抗体和anti-hFc-Eu 3+抗体稀释20倍和100倍。
(2)先将5μL测试化合物、2.5μL His-CD80蛋白和2.5μL PD-1-Fc融合蛋 白溶液混匀后,室温孵育15min;随后向该体系加入5μL anti-His-XL665抗体和5μL anti-hFc-Eu 3+抗体,继续孵育3h后检测。
(3)检测反应同时设置有对照组,包括未添加测试化合物的0%抑制阳性对照、未添加CD80蛋白的100%抑制阴性对照。所有检测采用复孔。
(4)使用荧光检测仪Tecan(Spark 10M)检测每孔的荧光信号,激发波长为320nm,检测的发射波长分别为620nm和665nm。CD80/PD-L1相互结合的强度参照荧光信号比值Em665/Em620。
(5)测试化合物的结合抑制率计算公式:抑制率(%)=[1–(检测孔荧光信号比值–100%抑制阴性对照荧光信号比值)]/(0%抑制阳性对照荧光信号比值–100%抑制阴性对照荧光信号比值)×100%。不同浓度梯度的测试化合物分别计算CD80/PD-L1结合抑制率后,计算50%抑制浓度(IC 50)。本发明代表性化合物体外抑制CD80和PD-L1结合的IC 50数据见表3:
表3本发明代表性化合物抑制体外CD80/PD-L1结合的IC 50数据
Figure PCTCN2022074183-appb-000058
由上述结果可见,本发明所述的化合物具有很好的抑制体外CD80/PD-L1的活性,本发明所述的通式(I)的化合物同样具有抑制CD80/PD-L1的活性。
实施例C 细胞水平免疫检查点PD-1和PD-L1介导的抑制T细胞活化信号的评价
作为免疫检查点分子,PD-1主要表达于活化的T细胞表面,而其配体PD-L1则表达广泛。除了抗原递呈细胞如树突状细胞、巨噬细胞、B细胞外,许多肿瘤细胞也可通过上调PD-L1的表达来实现抑制抗肿瘤免疫效应。对于正常免疫应答而言,抗原递呈细胞除了通过免疫共刺激分子激活T细胞外,还表达PD-L1配体分子等,与活化T细胞表面PD-1分子结合,从而抑制T细胞活化,避免了T细胞过度增殖活化而造成对周围正常组织的损伤。
实验主要原理
为了检测PD-1和PD-L1相互作用对免疫反应中T细胞活化信号的影响,构建稳定表达人PD-L1分子和抗CD3单链抗体(ScFv)的CHO-PD-L1-CD3L细胞以及稳定表达人PD-1分子和NFAT报告基因的Jurkat-PD-1-NFAT细胞。当两 种细胞共同孵育时,CHO细胞表面抗CD3ScFv和Jurkat细胞的膜CD3分子结合,会向Jurkat细胞内传递活化信号,但由于同时有CHO细胞表面PD-L1和Jurkat细胞表面的PD-1分子结合而向内传递抑制活化信号,使荧光素酶报告基因不能表达。当加入免疫检查点抗体或小分子抑制剂后,PD-1与PD-L1的结合被阻断,CD3ScFv抗体与CD3交联介导T细胞活化信号所激活的NFAT通路不再受到抑制信号的影响,下游荧光素酶报告基因开始表达。通过加入催化底物可检测到与报告基因活化呈正比的化学发光信号。
实验材料与设备
表达人PD-L1分子和抗CD3单链抗体(ScFv)的CHO-PD-L1-CD3L细胞以及稳定表达人PD-1分子和NFAT报告基因的Jurkat-PD-1-NFAT细胞均由陈博博士(康诺亚生物医药科技(成都)有限公司)自主构建并赠送。用于转染细胞稳定培养的Puromycin(Cat#540411)和Hygromycin B(Cat#V900372)购自Sigma,PMA(Cat#P1585)购自Sigma公司,抗人PD-L1抗体(Cat#GMP-A066)购自Novoprotein公司。荧光素酶的底物液(Cat#E6485)和荧光素酶专用细胞裂解液5×(Cat#E1531)均购自Promega公司。荧光检测仪器Tecan(Spark 10M)购自瑞士Tecan公司。
实验主要过程
(1)实验前一天,在96孔细胞培养板接种100μL的CHO-PD-L1-CD3L细胞(约4×10 4个/孔),培养基为含有10%FBS、8μg/mL Puromycin和200μg/mL Hygromycin B的DMEM/F12,37℃培养过夜;
(2)使用0.1%PBST先将测试化合物稀释成不同的浓度梯度,加入96孔板中,预先孵育30min;并使用含有10%FBS、8μg/mL Puromycin和200μg/mL Hygromycin B的RPMI 1640完全培养基将Jurkat-PD-1-NFAT细胞计数调整为2×10 5个/mL,同时加入100ng/mL的PMA(使用DMSO配制储存液浓度为10mg/mL)用于放大T细胞活化信号;向96孔板中每孔加入上述100μL的Jurkat-PD-1-NFAT细胞,进行共培养;
(3)检测反应同时设置有对照组,包括未添加测试化合物的溶剂对照和加入抗人PD-1抗体作为实验体系的阳性对照。所有检测采用复孔;
(4)继续在37℃孵育6h后,直接加入40μL的5×细胞裂解液,混匀后室温放置10min以完全裂解细胞;取50μL裂解后的细胞溶液转入荧光检测板,并加入30μL荧光素酶底物液,立即在荧光检测仪上选择化学发光检测程序进行测定。
(5)测试化合物的细胞水平T细胞活化信号的抑制率计算公式:T细胞活化信号的抑制率(%)=(检测孔化学发光原始数值–溶剂对照)/(化合物检测孔中 测得的最高化学发光原始数值–溶剂对照)×100%。不同浓度梯度的测试化合物分别计算T细胞活化信号的抑制率后,进一步计算50%抑制浓度(EC 50)。本发明化合物阻断PD-1和PD-L1介导的T细胞活化抑制信号的EC 50数据见表4:
表4本发明化合物阻断PD-1/PD-L1介导的T细胞活化抑制信号的EC 50
Figure PCTCN2022074183-appb-000059
由上述结果可见,本发明所述的化合物具有在细胞水平有效阻断免疫检查点介导的T细胞活化抑制信号的活性,本发明所述的通式(I)的化合物同样具有阻断PD-1/PD-L1介导的T细胞活化信号抑制活性。
实施例D 药代动力学试验
取24只6-8周龄B-hPD-1/hPD-L1mice雌小鼠,购于百奥赛图江苏基因生物技术有限公司。随机分组4组,每组6只,将受试化合物配制于含5%DMSO、60%PEG400和35%纯化水的溶媒中,包括I-1和3个对照分子Example 17、化合物14(INCB086550)、和实施例180,口服给予化合物50mg/kg,1次/天,于第18天给药后通过眼底静脉丛交替采血,取血时间点为15min,30min,1h,2h,4h,8h,24h和32h。收集血液约0.1mL于离心管中(肝素钠抗凝),5000rpm离心5min,分离血浆,于-20℃冻存待测。血浆样品处理后,利用液质联用仪(LC-MS/MS)测定血浆中化合物浓度。运用Phoenix WinNonlin 7.0计算药代动力学参数。数据总结见表5。
表5不同化合物重复给药后在人源化小鼠体内的药代参数特征
Figure PCTCN2022074183-appb-000060
aExample 17是Incyte公司在专利WO2019/217821文件中的第64页公开的化合物,发明人参考其中的合成方法合成了该化合物,以用于作对照分子。Example 17的化学结构经过了LC-MS MS-ESI(m/z)911.4[M+H] +确认,同时因 Example 17的溶解度不佳,核磁样品中加入了助溶解的氢氧化钠溶液(Example17与氢氧化钠的摩尔比为1比2),随后向其中加入MeOD用于结构确证。 1H-NMR(400MHz,MeOD)δppm 8.45(dd,J=8.3,1.2Hz,2H),7.43(t,J=8.0Hz,2H),7.12-7.07(m,2H),3.95(d,J=14.4Hz,6H),3.55(s,4H),2.96-2.84(m,4H),2.72-2.82(m,4H),2.70-2.62(m,4H),2.00-1.88(m,4H),1.87-1.78(m,4H),1.66-1.37(m,16H).的结构确认,该化合物的结构式如下:
Figure PCTCN2022074183-appb-000061
b化合物14(INCB086550)是Incyte公司在专利CN110267953A中的表2中公开的化合物,发明人参考其中的合成方法合成了该化合物,以用于作对照分子。化合物14(INCB086550)是临床进展最快的一种小分子PD-L1抑制剂,目前处于2期研究中。
化合物14(INCB086550)化学结构经过了LC-MS MS-ESI(m/z)694.2[M+H] +1H-NMR(400MHz,DMSO)δppm 9.31(s,1H),8.84(s,1H),8.48(d,J=8.0Hz,1H),8.17(s,1H),8.13(d,J=7.7Hz,1H),8.09-8.03(m,2H),7.83(s,1H),7.54(t,J=7.7Hz,1H),7.43(d,J=7.4Hz,1H),7.34(t,J=7.9Hz,1H),7.18(d,J=5.8Hz,1H),6.91(d,J=7.4Hz,1H),4.76(s,1H),4.26-4.18(m,1H),3.85-3.65(m,4H),2.84-2.69(m,3H),2.69-2.60(m,2H),2.58-2.46(m,3H),2.45(s,3H),2.38(dd,J=9.6,3.6Hz,1H),2.08(s,3H),2.05-1.95(m,2H),1.94-1.83(m,1H),1.63-1.52(m,1H).的结构确认,该化合物的结构式如下:
Figure PCTCN2022074183-appb-000062
c实施例180是Incyte公司在专利CN110267953A中公开的实施例,发明人参考其中的合成方法合成了该化合物,以用于作对照分子。实施例180化学结构经过了LC-MS MS-ESI(m/z)775.0[M+H] +1H-NMR(400MHz,DMSO)δppm9.90(s,2H),8.38(dd,J=7.6,2.7Hz,2H),7.49(t,J=8.0Hz,2H),7.14(d,J=7.7Hz,2H),3.90(s,6H),3.57(t,J=6.0Hz,2H),3.51-3.46(m,4H),2.85-2.77(m,4H),2.70-2.64(m,4H),2.64-2.59(m,4H),1.90-1.76(m,2H),1.68-1.55(m,2H),1.54-1.22(m,6H).的结构确认,该化合物的结构式如下:
Figure PCTCN2022074183-appb-000063
由上述结果可见,本发明所述的代表性化合物在50mg/kg重复给药后,小鼠体内血浆暴露量(AUC (0-t))、体内平均滞留时间(MRT)和半衰期(T 1/2)均显著高于3个对照分子。重复给药能更好体现临床治疗条件下的药代特征,相比对照分子,本发明所述化合物在重复给药后体内暴露量和持续暴露时间(AUC/T 1/2)均有预料不到的提高,有助于临床治疗中更好地发挥抗肿瘤活性。
实施例E 肿瘤组织分布试验
取16只B-hPD-1/hPD-L1mice人源化雌小鼠,6-8周龄,购于百奥赛图江苏基因生物技术有限公司,适应1周后,皮下接种MC38-PD-L1细胞,2×10 6个/部位,待肿瘤生长到约200m 3后,分4组,每组4只。将受试化合物配制于含5%DMSO、60%PEG400和35%纯化水的溶媒中,包括I-1和3个对照分子Example 17、化合物14(INCB086550)、和实施例180,口服给予化合物50mg/kg,1次/天,于第18天给药4h、24h后取血和肿瘤组织。称取一定量的组织,加磷 酸盐缓冲盐(PBS)溶液匀浆,经样品处理后,进LC-MS/MS分析,测定血浆和组织中化合物浓度。实验结果见表6、图1和图2。
表6不同化合物重复给药后在人源化小鼠体内肿瘤组织分布
Figure PCTCN2022074183-appb-000064
由上述结果可见,在重复给药后,本发明所述的代表性化合物在肿瘤组织中的浓度明显高于血浆,即4h和24h的肿瘤组织浓度是血浆的4-6倍和17-27倍;同时在相同给药条件下,4h和24h的肿瘤组织浓度是对照分子的5-8倍和7-30倍,说明本发明化合物表现出良好的肿瘤组织靶向性。与对照分子相比,本发明化合物在肿瘤组织上具有预料不到的富集和靶向效应。
产业上的可应用性
本发明的联苯类化合物具有优异的PD-L1抑制活性,可成为治疗或预防与该作用相关的疾病的药物。
以上所述仅是本发明的优选实施方式,应当指出,对于本技术领域的普通技术人员来说,在不脱离本发明原理的前提下,还可以做出若干改进和润饰,这些改进和润饰也应视为本发明的保护范围。

Claims (22)

  1. 一种式(I)的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    Figure PCTCN2022074183-appb-100001
    其中,
    R 1和R 2相同或不同,选自C 1-C 6烷基、氰基、卤素;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基、C 2-C 6炔基C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基C 1-C 6烷基、单C 1-C 6烷基氨基C 1-C 6烷基、双C 1-C 6烷基氨基C 1-C 6烷基、卤代C 1-C 6烷氧基C 1-C 6烷基、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基、3元至14元杂环烷基-C 1-C 4烷基;
    R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被一个或多个选自羟基、羧基和卤素的取代基取代;
    X选自-O-、-S-;
    R 5-R 26选自氢原子;
    R 27-R 30选自氢原子;
    其中,上述R 1-R 30定义中各基团中的至少1个氢原子被氘(D)替代。
  2. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,
    R 1和R 2相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯、溴;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、单C 1-C 6烷基氨基C 1-C 6烷基-、双C 1-C 6烷基氨基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基-、3元至14元杂环烷基-C 1-C 4烷基-;
    R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被一个或多个选自羟基和卤素的取代基取代;
    X选自-O-;
    R 5-R 26选自氢原子,其中至少1个氢原子被氘(D)替代;
    R 27-R 30选自氢原子,优选地其中至少1个氢原子被氘(D)替代,氘代位置在相应脂肪环和脂肪杂环上的任意位置。
  3. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,
    R 1和R 2相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯、溴;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、单C 1-C 6烷基氨基C 1-C 6烷基-、双C 1-C 6烷基氨基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-、C 3-C 14环烷基、3元至14元杂环烷基、C 3-C 14环烷基-C 1-C 4烷基-、3元至14元杂环烷基-C 1-C 4烷基-;
    R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被一个或多个选自羟基和卤素的取代基取代;
    X选自-O-;
    R 5-R 26选自氢原子,优选地其中至少1个氢原子被氘(D)替代;
    R 27-R 30选自氢原子,其中至少1个氢原子被氘(D)替代,氘代位置在相应脂肪环和脂肪杂环上的任意位置。
  4. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,
    R 1和R 2相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、单C 1-C 6烷基氨基C 1-C 6烷基-、双C 1-C 6烷基氨基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-、C 3-C 14环烷基、3元至14元杂环烷基,这些基团中的至少1个氢原子被氘(D)替代;
    R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被卤素取代;
    X选自-O-;
    R 5-R 26选自氢原子,优选地其中至少1个氢原子被氘(D)替代;
    R 27-R 30选自氢原子,优选地其中至少1个氢原子被氘(D)取代,氘代位置在相应脂肪环和脂肪杂环上的任意位置。
  5. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或 代谢产物,
    其中,
    R 1和R 2相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-;
    R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被卤素取代;
    X选自-O-;
    R 5-R 26为氢原子且其中至少1个氢原子被氘(D)替代;
    R 27-R 30选自氢原子,优选地其中至少1个氢原子被氘(D)替代,氘代位置在相应脂肪环和脂肪杂环上的任意位置。
  6. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,
    R 1和R 2相同或不同,选自甲基、甲基-d 3、氰基、氟、氯;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-;
    R 4选自氢、C 1-C 6烷基,其中所述的C 1-C 6烷基任选地被卤素取代;
    X选自-O-;
    R 5-R 26选自氢原子,优选地其中至少1个氢原子被氘(D)替代;
    R 27-R 30选自氢原子且其中至少1个氢原子被氘(D)替代,氘代位置在相应脂肪环和脂肪杂环上的任意位置。
  7. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,
    R 1和R 2相同或不同,选自甲基、甲基-d 3(CD 3)、氰基、氟、氯;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-,这些基团中的至少1个氢原子被氘(D)替代;
    R 4选自氢;
    X选自-O-;
    R 5-R 26选自氢原子,优选地其中至少1个氢原子被氘(D)替代;
    R 27-R 30选自氢原子,优选地其中至少1个氢原子被氘(D)替代,氘代位置在相应脂肪环和脂肪杂环上的任意位置。
  8. 根据权利要求1所述的化合物、其立体异构体、药学上可接受的盐、前体或 代谢产物,
    其中,R 1和R 2相同或不同,选自甲基、氰基、卤素;
    R 3选自氢、C 1-C 6烷基、C 2-C 6烯基C 1-C 6烷基-、C 2-C 6炔基C 1-C 6烷基-、卤代C 1-C 6烷基-、C 1-C 6烷氧基C 1-C 6烷基-、卤代C 1-C 6烷氧基C 1-C 6烷基-,这些基团中的至少1个氢原子被氘(D)替代;
    R 4选自氢、C 1-C 6烷基;
    X选自-O-;
    R 5-R 26选自氢原子;
    R 27-R 30选自氢原子。
  9. 根据权利要求8所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,R 1和R 2相同或不同,选自甲基、氰基、氟、氯;
    优选的,R 1和R 2相同或不同,选自甲基、氟、氯;
    优选的,R 1和R 2相同或不同,选自甲基、氯。
  10. 根据权利要求8所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,R 3选自氢、C 1-C 6烷基、卤代C 1-C 6烷基,这些基团中的至少1个氢原子被氘(D)替代;
    优选的,R 3选自C 1-C 6烷基,C 1-C 6烷基中的至少1个氢原子被氘(D)替代;
    优选的,R 3选自甲基、乙基、丙基、丁基、戊基,这些基团中的至少1个氢原子被氘(D)取代。
  11. 根据权利要求10所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,R 3定义的基团中的1-15个氢原子被氘(D)替代,优选1-9个氢原子被氘(D)替代。
  12. 根据权利要求8所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,
    其中,R 4选自氢、甲基、乙基、丙基、丁基、戊基;
    优选的,R 4选自氢。
  13. 根据权利要求1-12任一项所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,其中所述化合物选自:
    4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰 基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2'-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2,2'-二氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5, 5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2'-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-甲基-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-甲基-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2'-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2-(甲基-d 3)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-(甲基-d 3)-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-2'-(甲基-d 3)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-(甲基-d 3)-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-((甲氧基-d 3)甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸;
    4-(2-(2-((2-氯-2'-氟-3'-(5-(2-(4-(甲氧甲基)双环[2.2.1]庚烷-1-基)乙基)-1-甲基-4,5,6,7-四氢-1H-咪唑并[4,5-c]吡啶-2-甲酰胺基)-[1,1'-联苯基]-3-基-4,4',5,5',6,6'-d 6)氨甲酰基)-1-甲基-1,4,6,7-四氢-5H-咪唑并[4,5-c]吡啶-5-基)乙基)双环[2.2.1]庚烷-1-羧酸。
  14. 权利要求1-13任一项所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物的制备方法,包括以下步骤:
    Figure PCTCN2022074183-appb-100002
    1)使式(I-a)所示化合物在第一溶剂中经第一酸、第一碱或者催化氢解脱去保护基P 1,所得产物未经分离纯化,进一步与式(I-b)所示化合物在第二溶剂中并且在第一催化剂和第二碱存在下发生Suzuki反应得到式(I-c)所示化合 物;
    2)在第三溶剂中,使式(I-c)所示化合物和式(I-d)所示化合物在第一还原剂存在下发生还原胺化反应得到式(I-e)所示化合物;或者,
    在第四溶剂中,使式(I-c)所示化合物与式(I-d')所示化合物在第三碱存下发生亲核取代反应得到式(I-e)所示化合物;
    3)在第五溶剂中,使式(I-e)所示化合物和式(I-f)所示化合物在第二还原剂存在下发生还原胺化反应得到作为式(I)所示化合物的式(I')所示化合物;或者,
    在第六溶剂中,使式(I-e)所示化合物与式(I-f')所示化合物在第四碱存下发生亲核取代反应得到作为式(I)所示化合物的式(I')所示化合物;
    任选地,还包括
    4)在第五碱存在下,使式(I')所示化合物经过酯水解反应得到式(I”)所示化合物;
    其中:
    X、R 1-R 30的定义如权利要求1-13任一项所述;
    R 4'除了不为氢以外,其定义与R 4相同;
    R 4”为氢;
    M选自硼酸酯或硼酸,优选地选自4,4,5,5-四甲基-1,3,2-二氧杂戊硼烷、联硼酸新戊二醇酯、4,4,4',4',5,5,5',5'-八甲基-2,2'-二(1,3,2-二氧杂环戊硼烷)B(OBu-n) 3、B(OPr-i) 3;或者,
    M选自溴、碘、氯、氟、CF 3SO 3-(OTf);
    W选自硼酸酯或硼酸,优选地选自4,4,5,5-四甲基-1,3,2-二氧杂戊硼烷、联硼酸新戊二醇酯、4,4,4',4',5,5,5',5'-八甲基-2,2'-二(1,3,2-二氧杂环戊硼烷)B(OBu-n) 3、B(OPr-i) 3;或者,
    W选自溴、碘、氯、氟、CF 3SO 3-(OTf);
    P 1和P 2是保护基,可相同或不同,优选地选自Boc(叔丁氧羰基)、Fmoc(9-芴甲氧羰基)、Cbz(N-苄氧羰基)、甲磺酰基、对甲苯磺酰基、乙酰基、甲氧羰基、乙氧羰基、((2-三甲基硅)乙氧)甲基(SEM)、四氢-2H-吡喃-2-基(THP)。
  15. 根据权利要求14所述的制备方法,其中,
    所述的第一酸优选地选自三氟乙酸(TFA)、盐酸(HCl)、醋酸(HOAc)、氢溴酸(HBr);
    所述的第一碱优选地选自哌啶、二乙胺;
    所述的第一溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、 N,N'-二甲基甲酰胺(DMF);
    所述第一催化剂优选地选自1,1'-双(二环己基膦基)二茂铁二氯化钯(PdCl 2(dcypf))、醋酸钯(Pd(OAc) 2)、二氯化钯(PdCl 2)、三(二亚苄基丙酮)二钯(Pd 2(dba) 3)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(PdCl 2(dppf))、[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(PdCl 2(dppf)·CH 2Cl 2)、四(三苯基膦)钯(Pd(PPh 3) 4)、双(三环己基膦)二氯化钯(PdCl 2(P(Cy) 3) 2)、2-二环己基膦-2',6'-二甲氧基联苯(SPhos);
    所述第二碱优选地选自有机碱和无机碱类,例如三乙胺(TEA)、N,N-二异丙基乙基胺(DIPEA)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、醋酸钾(KOAc)、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、磷酸钾(K 3PO 4)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH);
    所述第二溶剂优选地选自1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF),以及这些溶剂与水以不同比例形成的混合溶剂;
    第一还原剂和第二还原剂优选地选自醋酸硼氢化钠、硼氢化钠、氰基硼氢化钠;
    所述第三溶剂和第五溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF);
    所述第四溶剂和第六溶剂优选地选自二氯甲烷(DCM),1,2-二氯乙烷、甲醇(MeOH)、乙醇(EtOH)、1,4-二氧六环(1,4-dioxane)、四氢呋喃(THF)、乙腈(MeCN)、N,N'-二甲基甲酰胺(DMF)、N-甲基吡咯烷酮(NMP)、吡啶(Py);
    所述的第三碱和第四碱优选地选自三乙胺(TEA)、N,N'-二异丙基乙基胺(DIPEA)、吡啶(Py)、正丁基锂、二异丙基氨基锂、双三甲基硅基氨基锂、叔丁醇钠(NaOBu-t)、叔丁醇钾(KOBu-t)、氢化钠(NaH)、碳酸钠(Na 2CO 3)、碳酸钾(K 2CO 3)、氢氧化锂(KOH)、氢氧化钠(NaOH);
    所述第五碱优选地选自氢氧化锂(LiOH)、氢氧化锂(KOH)、氢氧化钠(NaOH)。
  16. 权利要求1-13任一项所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物的制备方法,包括以下步骤:
    Figure PCTCN2022074183-appb-100003
    使式(I-a)所示化合物经脱去保护基P 1,所得产物未经分离纯化进一步与式(I-b)所示化合物发生Suzuki反应得到式(I-c)所示化合物;使式(I-c)所示化合物先与式(I-f)所示化合物发生还原胺化反应得到式(I-h)所示化合物;或者通过使式(I-c)所示化合物先与式(I-f')所示化合物发生亲核取代反应得到式(I-h)所示化合物,随后使(I-h)所示化合物经脱 保护后再与式(I-d)所示化合物发生还原胺化反应得到作为终产物(I)的式(I'-1)所示化合物;或者使(I-h)所示化合物经脱保护后再与式(I-d')所示化合物发生亲核取代反应得到作为终产物(I)的式(I'-1)所示化合物,任选地,式(I'-1)所示化合物进一步发生酯水解反应得到式(I”-1)所示化合物;
    其中,各步骤所述的脱保护反应、Suzuki反应、还原胺化反应、亲核取代反应、酯水解反应如权利要求14或15所述;
    其中,各取代基的定义如权利要求14或15所述。
  17. 权利要求1-13任一项所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物的制备方法,包括以下步骤:
    Figure PCTCN2022074183-appb-100004
    使式(I-a)所示化合物经脱去保护基P 1,所得产物未经分离纯化进一步与 式(I-b)所示化合物发生Suzuki反应得到式(I-c)所示化合物;使式(I-c)所示化合物先与式(I-f)所示化合物发生还原胺化反应得到式(I-h)所示化合物;或者通过使式(I-c)所示化合物先与式(I-f')所示化合物发生亲核取代反应得到式(I-h)所示化合物,随后使式(I-h)所示化合物进一步发生酯水解反应得到式(I-i)所示化合物,随后使(I-i)所示化合物经脱保护后再与式(I-d)所示化合物发生如上所述的原胺化反应得到作为终产物(I)的式(I”')所示化合物;或者使(I-i)所示化合物经脱保护后再与式(I-d')所示化合物发生亲核取代反应得到作为终产物(I)的式(I”')所示化合物;
    优选地,各步骤所述的脱保护反应、Suzuki反应、还原胺化反应、亲核取代反应、酯水解反应的条件如权利要求14或15所述;
    其中,各取代基的定义如权利要求14或15所述。
  18. 化合物,选自,
    Figure PCTCN2022074183-appb-100005
  19. 根据权利要求18所述的化合物在制备式(I)所示化合物中的应用。
  20. 一种药物组合物,其包含权利要求1-13任一项所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物,以及任选的可药用载体。
  21. 权利要求1-13任一项所述的化合物、其立体异构体、药学上可接受的盐、前体或代谢产物或者权利要求20所述的药物组合物在制备用于治疗和/或预防与靶点PD-L1相关的疾病的药物中的用途,或者
    在制备用于抑制PD-L1活性的药物中的用途,或者
    在制备作为PD-L1抑制剂的药物中的用途,或者
    在制备作为靶向PD-L1信号通路的免疫调节剂的药物中的用途。
  22. 根据权利要求21所述的用途,其中,所述与靶向PD-L1相关的疾病选自肿瘤、癌症或者其他免疫相关疾病。
PCT/CN2022/074183 2021-02-01 2022-01-27 作为免疫调节剂的联苯类化合物及其制备方法和应用 WO2022161421A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA3210314A CA3210314A1 (en) 2021-02-01 2022-01-27 Biphenyl compound as immunomodulator, preparation method therefor and use thereof
AU2022214735A AU2022214735A1 (en) 2021-02-01 2022-01-27 Biphenyl compound as immunomodulator, preparation method therefor and use thereof
EP22745294.3A EP4286383A1 (en) 2021-02-01 2022-01-27 Biphenyl compound as immunomodulator, preparation method therefor and use thereof
CN202280008146.3A CN116710445A (zh) 2021-02-01 2022-01-27 作为免疫调节剂的联苯类化合物及其制备方法和应用
KR1020237029267A KR20230141812A (ko) 2021-02-01 2022-01-27 면역 조절제로서의 바이페닐 화합물, 이의 제조방법 및 용도
JP2023546481A JP2024505972A (ja) 2021-02-01 2022-01-27 免疫調節剤としてのビフェニル系化合物とその製造方法及び応用
BR112023015451A BR112023015451A2 (pt) 2021-02-01 2022-01-27 Composto de bifenila como imunomodulador, método de preparação para o mesmo e uso do mesmo

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110137428 2021-02-01
CN202110137428.0 2021-02-01

Publications (1)

Publication Number Publication Date
WO2022161421A1 true WO2022161421A1 (zh) 2022-08-04

Family

ID=82653005

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/074183 WO2022161421A1 (zh) 2021-02-01 2022-01-27 作为免疫调节剂的联苯类化合物及其制备方法和应用

Country Status (9)

Country Link
EP (1) EP4286383A1 (zh)
JP (1) JP2024505972A (zh)
KR (1) KR20230141812A (zh)
CN (1) CN116710445A (zh)
AU (1) AU2022214735A1 (zh)
BR (1) BR112023015451A2 (zh)
CA (1) CA3210314A1 (zh)
TW (1) TWI814226B (zh)
WO (1) WO2022161421A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116903595A (zh) * 2023-06-01 2023-10-20 遵义医科大学珠海校区 一种嘧啶二酮类化合物及其制备方法和应用
WO2023207933A1 (zh) * 2022-04-26 2023-11-02 深圳微芯生物科技股份有限公司 一种作为免疫调节剂的联苯类化合物的晶型及其制备方法
WO2023207937A1 (zh) * 2022-04-26 2023-11-02 深圳微芯生物科技股份有限公司 一种作为免疫调节剂的联苯类化合物的盐型、晶型及其制备方法
EP4190782A4 (en) * 2020-10-29 2024-01-03 Shenzhen Chipscreen Biosciences Co Ltd BIPHENYL COMPOUND USED AS AN IMMUNOMODULATOR, PREPARATION METHOD AND APPLICATION THEREOF

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018106850A1 (en) * 2016-12-07 2018-06-14 Auspex Pharmaceuticals, Inc. Deuterated aminopyridine compounds
CN109020958A (zh) * 2018-08-28 2018-12-18 湖南华腾制药有限公司 一种氘代色氨酸羟化酶抑制剂
CN110267953A (zh) 2016-12-22 2019-09-20 因赛特公司 四氢咪唑并[4,5-c]吡啶衍生物作为pd-l1内在化诱导剂
WO2019217821A1 (en) 2018-05-11 2019-11-14 Incyte Corporation Tetrahydro-imidazo[4,5-c]pyridine derivatives as pd-l1 immunomodulators

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018106850A1 (en) * 2016-12-07 2018-06-14 Auspex Pharmaceuticals, Inc. Deuterated aminopyridine compounds
CN110267953A (zh) 2016-12-22 2019-09-20 因赛特公司 四氢咪唑并[4,5-c]吡啶衍生物作为pd-l1内在化诱导剂
WO2019217821A1 (en) 2018-05-11 2019-11-14 Incyte Corporation Tetrahydro-imidazo[4,5-c]pyridine derivatives as pd-l1 immunomodulators
CN109020958A (zh) * 2018-08-28 2018-12-18 湖南华腾制药有限公司 一种氘代色氨酸羟化酶抑制剂

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CHEN L.HAN X., J. CLIN. INVEST., vol. 125, 2015, pages 3384 - 3391
CHENG X.VEVERKA VRADHAKRISHNAN A. ET AL., J. BIOL. CHEM., vol. 288, 2013, pages 11771 - 11785
COUZIN-FRANKEL J., SCIENCE, vol. 342, 2013, pages 1432 - 1433
DOLAN D.E.GUPTA S., CANCER CONTROL, vol. 21, 2014, pages 231 - 237
FULLER M.J.CALLENDRET B.ZHU B. ET AL., PROC. NATL USA, vol. 110, 2013, pages 15001 - 15006
MELLMAN I.COUKOS G.DRANOFF G., NATURE, vol. 480, 2011, pages 480 - 489
POSTOW M.A.CALLAHAN M.K.WOLCHOK J.D., J. CLIN. ONCOL., vol. 33, 2015, pages 1974 - 1982
SUGIURA D.MARUHASHI T.OKAZAKI 11-MI ET AL., SCIENCE, vol. 364, 2019, pages 558 - 566

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4190782A4 (en) * 2020-10-29 2024-01-03 Shenzhen Chipscreen Biosciences Co Ltd BIPHENYL COMPOUND USED AS AN IMMUNOMODULATOR, PREPARATION METHOD AND APPLICATION THEREOF
WO2023207933A1 (zh) * 2022-04-26 2023-11-02 深圳微芯生物科技股份有限公司 一种作为免疫调节剂的联苯类化合物的晶型及其制备方法
WO2023207937A1 (zh) * 2022-04-26 2023-11-02 深圳微芯生物科技股份有限公司 一种作为免疫调节剂的联苯类化合物的盐型、晶型及其制备方法
CN116903595A (zh) * 2023-06-01 2023-10-20 遵义医科大学珠海校区 一种嘧啶二酮类化合物及其制备方法和应用
CN116903595B (zh) * 2023-06-01 2024-02-13 遵义医科大学珠海校区 一种嘧啶二酮类化合物及其制备方法和应用

Also Published As

Publication number Publication date
JP2024505972A (ja) 2024-02-08
KR20230141812A (ko) 2023-10-10
BR112023015451A2 (pt) 2023-10-10
EP4286383A1 (en) 2023-12-06
CA3210314A1 (en) 2022-08-04
AU2022214735A1 (en) 2023-08-31
TWI814226B (zh) 2023-09-01
CN116710445A (zh) 2023-09-05
TW202237602A (zh) 2022-10-01

Similar Documents

Publication Publication Date Title
WO2022161421A1 (zh) 作为免疫调节剂的联苯类化合物及其制备方法和应用
CN107074851B (zh) 作为αvβ6整联蛋白拮抗剂用于治疗例如纤维变性疾病的萘啶衍生物
JP6484253B2 (ja) Toll様受容体7アゴニストとしての化合物および組成物
WO2022089511A1 (zh) 作为免疫调节剂的联苯类化合物及其制备方法和应用
US11667663B2 (en) Cyclic dinucleotides as anticancer agents
TWI685483B (zh) 作為lsd1抑制劑之環丙胺
TW201946626A (zh) 作為免疫調節劑之雜環化合物
KR20140044791A (ko) Ror감마t 억제제
CN115298165A (zh) 作为HIF-2α抑制剂的四氢化萘和四氢喹啉化合物
JP2021530442A (ja) 縮合三環系複素環化合物およびその治療上の使用
TW202007687A (zh) 經進一步取代之***并喹噁啉衍生物
CN115477660A (zh) 作为免疫调节剂的联苯类化合物及其制备方法和应用
US20230234970A1 (en) Immunosuppressant, and preparation method therefor and use thereof
CN111635373A (zh) 多环磺酰胺类RORγ调节剂
CN114249726A (zh) 联苯类化合物、其制备方法及应用
WO2023274396A1 (zh) 苯并氮杂环类化合物及其在药物中的应用
WO2018171611A1 (zh) 6-吡唑-[1,2,4]***并[4,3-a]吡啶-3-酰胺类衍生物、其制备方法及其在医药上的应用
WO2017008681A1 (zh) 酰胺类衍生物、其制备方法及其在医药上的用途
CN111747927B (zh) 作为免疫调节剂的化合物及其应用
CN112759541B (zh) 类吲哚衍生物及其用途
WO2023133271A1 (en) Bicyclic phthalazin-1(2h)-one derivatives and related uses
WO2023051635A1 (zh) 一种稠环化合物、其制备方法及其应用
WO2022237781A1 (zh) 酰胺衍生物及其应用

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: 22745294

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280008146.3

Country of ref document: CN

ENP Entry into the national phase

Ref document number: 3210314

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2023546481

Country of ref document: JP

Ref document number: MX/A/2023/009052

Country of ref document: MX

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023015451

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20237029267

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2022214735

Country of ref document: AU

Date of ref document: 20220127

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2023122609

Country of ref document: RU

Ref document number: 2022745294

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022745294

Country of ref document: EP

Effective date: 20230901

WWE Wipo information: entry into national phase

Ref document number: 11202305813W

Country of ref document: SG

ENP Entry into the national phase

Ref document number: 112023015451

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20230801