[go: up one dir, main page]

CN120603829A - Heteroaromatic ring compound and its preparation method and use - Google Patents

Heteroaromatic ring compound and its preparation method and use

Info

Publication number
CN120603829A
CN120603829A CN202480009595.9A CN202480009595A CN120603829A CN 120603829 A CN120603829 A CN 120603829A CN 202480009595 A CN202480009595 A CN 202480009595A CN 120603829 A CN120603829 A CN 120603829A
Authority
CN
China
Prior art keywords
optionally substituted
alkyl
methyl
cycloalkyl
preparation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202480009595.9A
Other languages
Chinese (zh)
Inventor
党群
刘卫敏
王琦琳
刘辉
胡冬梅
贾丽敏
李帅
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Yiweikang Pharmaceutical Co ltd
Original Assignee
Shanghai Yiweikang Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Yiweikang Pharmaceutical Co ltd filed Critical Shanghai Yiweikang Pharmaceutical Co ltd
Publication of CN120603829A publication Critical patent/CN120603829A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • C07D209/88Carbazoles; Hydrogenated carbazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

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

Abstract

杂芳环类化合物及其制备方法和用途。具体地,涉及式(I)的化合物,及其立体异构体、几何异构体、互变异构体、药用盐、前药、水合物、溶剂化物或同位素标记的类似物。还涉及所述化合物的制备方法和用途。 Heteroaromatic ring compounds, preparation methods, and uses thereof. Specifically, the present invention relates to compounds of formula (I), and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates, or isotope-labeled analogs thereof. The present invention also relates to preparation methods and uses of the compounds.

Description

Heteroaromatic compounds, their preparation and use Technical Field
The application relates to the field of pharmaceutical chemistry, in particular to a heteroaromatic compound, a preparation method and application thereof.
Background
Cancer is one of the major fatal diseases in humans. Cancer cells are hyperproliferative, often accompanied by chromosomal instability (chromosomal instability), because with rapid division of cancer cells, even the entire chromosome may be replicated, mutated, or lost entirely. This instability is mainly related to cancer invasion, the more unstable the chromosomes, the more likely the DNA fragments from these chromosomes will eventually reach places where they would not otherwise exist, such as floating in the cytoplasm outside the nucleus. The cells recognize these free DNA fragments as evidence of viral invasion, triggering an autoimmune response inside the cells, triggering inflammation, recruiting immune cells to the tumor site to initiate the attack.
The cGAS-STING pathway is an early warning system that is intracellular in this process, and when DNA from the chromosome of a virus or cancer cell enters the cytoplasm, cGAS binds to it, forming the second messenger cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which acts as a warning signal to activate the STING pathway in the cell, producing type I interferon IFNs and other costimulatory molecules that trigger an immune response. This is an important anticancer innate immune pathway that initiates immune attack against virus-infected or cancerous cells. Although cancer cells theoretically cause killing of immune cells due to DNA fragments scattered in the cytoplasm, in practice cancer cells can still efficiently escape from monitoring the immune system, and metastasis and spread occur. In 2020, scientists at the Stonekette cancer research center have discovered a mechanism to elucidate this phenomenon because there is a molecule outside the cancer cells that breaks down the cGAMP signal before it reaches the immune cells. This cell surface scissors-like protein is the outer nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP 1), which is the primary hydrolase known to cGAMP. When cGAMP appears extracellularly, ENPP1 will rapidly hydrolyze cGAMP to AMP and GMP using its atpase activity, preventing the signal from activating STING pathway and thus failing to activate immune cells, AMP will further release adenosine, which is an immunosuppressive molecule. Thus, ENPP1 inhibitors would act as "double arrow" in immunomodulation.
At present, several companies are developing ENPP1 small molecule inhibition drugs for treating cancers, and foreign research institutions mainly comprise Mavupharma (purchased AbbVie in 2019), stingray Therapeutics and Riboscience, LLC. Wherein the study phase was first Riboscience, RBS2418 of LLC, company, was recruited at stage I beginning at month 3 of 2022, and the remaining company compounds were all in the preclinical study phase.
RBS2418 is Riboscience, an llc product under investigation, the structure of which has not been disclosed and activity data for this compound has not been found. There are two patent publications (WO 2020210649 and WO 2020140001) about ENPP1 small molecule inhibitors of this company, the structures of which are quinoline and phosphoric acid compounds (scheme 1), and the in vitro enzyme activity IC50 of some compounds is below 10 nM. At present, the clinical phase I recruitment is carried out, a climbing experiment of single drug and combined use with PD-1 antibody Perbrolizumab is planned, from 100mg BID to 800mg BID, oral administration is carried out, and drug effect and safety evaluation of single drug and combined use are carried out after dosage is determined. Phase I clinical is expected to be completed in 2024.
The Stingray Therapeutics company has 6 patent publications about ENPP1 small molecule inhibition up to the present, and has two structural types, namely quinoline compounds, the structural difference of which with Riboscience company compounds is mainly at the tail sulfamide part, thioether compounds, the structural difference of which with other in-vitro molecules is relatively large, and the in-vitro enzyme activity IC50 of most of the compounds is above 100 nM.
MV-626 is an ENPP1 inhibitor developed by MavuPharma, and the structure is not disclosed. In 7 months 2019, abbVie announced purchase MavuPharma, and further obtained its ENPP1 inhibitor MV-626 under investigation. From the analysis of this company's published patent, the structure is mainly a change of quinoline ring, bicyclic or tricyclic, and the tail is sulfamide. The compound has the inhibition activity on ENPP1 in nM level, has good PK property, and has the oral utilization rate of 100% in rats and mice.
Although some small molecules of ENPP1 inhibitors have been disclosed, no ENPP1 inhibitors are developed and marketed at present, so that development of new compounds with marketing potential, better drug effect and drug generation result is still urgent.
Disclosure of Invention
The invention designs a series of compounds with new structures shown in the general formula, and discovers that the compounds with the structures have excellent effects and actions and have positive significance in the development of ENPP1 inhibitors.
The invention aims to provide a compound with a brand new structure as an ENPP1 inhibitor, a preparation method thereof and application thereof in treating diseases mediated by ENPP 1.
The present invention provides compounds of formula (I), and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates, or isotopically-labeled analogs thereof,
Wherein the method comprises the steps of
L is selected from -(NRL3)SO2N(RL1)(RL2)、-SO2RL1、-C(O)ORL1、-C(O)N(RL1)(RL2)、-C(O)NHORL1、-P(O)(ORL1)(ORL2)、-P(O)(NHRL1)(ORL2)、-P(O)(ORL1)(RL2)、-P(O)(NHRL1)(NHRL2)、-P(O)(NHRL1)(RL2)、-P(O)(RL1)(RL2)、-P(O)(SRL1)(SRL2)、-P(O)(NHRL1)(SRL2)、-P(O)(SRL1)(RL2) or-B (OH) 2, wherein R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, or optionally substituted aryl, or R L1 and R L2 are joined to form an optionally substituted cyclic structure;
A is selected from optionally substituted alkylene, optionally substituted cycloalkylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, optionally substituted cycloalkylene, optionally substituted heterocyclylene alkylene, optionally substituted arylene alkylene, or optionally substituted heteroarylene;
Z is selected from- (chemical bond )、-O-、-S-、-CO-、-SO-、-SO2-、-C(RZ1)(RZ2)-、-N(RZ1)-、-CH2N(RZ1)-、-N(RZ1)CH2-、-CON(RZ1)-、-N(RZ1)CO-、-SON(RZ1)-、-SO2N(RZ1)-、-N(RZ1)SO-、-N(RZ1)SO2- or Wherein R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
R 1 is selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl 、-OR3、-SR3、-NR3R4、-COOR3、-CONR3R4、-NR3COR4、-COR3、-OCOR3、-SONR3R4、-SO2NR3R4、-NR3SOR4、-NR3SO2R4, or-Si (R 3)(R4)(R5), wherein R 3、R4 or R 5 are independently selected from hydrogen, deuterium, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
n=0, 1, 2 or 3;
X 1、X2、X3 and X 4 are independently selected from N or C (R X), wherein R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl 、-ORa、-SRa、-NRaRb、-COORa、-CONRaRb、-NRbCORa、-CORa、-OCORa、-SONRaRb、-SO2NRaRb、-NRbSORa、-NRbSO2Ra, or-Si (R a)(Rb)(Rc), wherein R a、Rb and R c are independently selected from hydrogen, deuterium, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or X 1 is joined with X 2 to form an optionally substituted cyclic structure, or X 2 is joined with X 3 to form an optionally substituted cyclic structure, or X 3 is joined with X 4 to form an optionally substituted cyclic structure, or X 1 and Z are joined to form an optionally substituted cyclic structure.
The present invention also provides a method of treating a disease mediated by ENPP1, comprising administering to a patient in need thereof a compound of formula (I).
The invention also provides the use of a compound of formula (I) in the manufacture of a medicament for the treatment of a disease mediated by ENPP 1.
Detailed Description
Definition of the definition
As used herein, the term "alkyl", alone or in combination, refers to a straight or branched saturated hydrocarbon chain, including C 1-6 alkyl groups having 1 to 6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 4-methylpentyl, neopentyl, n-hexyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1, 2-trimethylpropyl, and the like.
As used herein, the term "alkenyl", alone or in combination, refers to a straight or branched unsaturated hydrocarbon chain having at least one carbon-carbon double bond, for example, a C 2-6 -alkenyl group having 2 to 6 carbon atoms, such as vinyl, allyl, 1-butenyl, 2-butenyl, isobutenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 1-methyl-2-butenyl, 2, 3-dimethyl-2-butenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, and the like.
As used herein, the term "alkynyl", alone or in combination, refers to a straight or branched unsaturated hydrocarbon chain having at least one carbon-carbon triple bond, for example a C 2-6 -alkynyl group having 2 to 6 carbon atoms, such as ethynyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1-butynyl, 1-hexynyl, 2-hexynyl, 3-hexenyl, and the like.
As used herein, the term "cycloalkyl", alone or in combination, refers to a saturated non-aromatic cyclic hydrocarbon group, for example a C 3-6 -cycloalkyl group having 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
As used herein, the term "heterocyclyl", alone or in combination, refers to a non-aromatic cyclic group containing one or more (e.g., 1,2, 3, or 4) heteroatoms independently selected from O, S and N, such as a 4-6 membered heterocyclyl having 4 to 6 ring atoms or a 4-10 membered heterocyclyl having 4 to 10 ring atoms. Examples of heterocyclyl groups include pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, morpholinyl, indolinyl, pyranyl, tetrahydropyran, thiomorpholinyl, tetrahydrothienyl, isoxazolidinyl, isothiazolidinyl, oxazolidinyl, thiazolidinyl, azepanyl, benzoazepine, and the like.
As used herein, the term "aryl", alone or in combination, refers to a monocyclic or polycyclic aromatic ring, e.g., a C 6-10 aryl group having 6 to 10 carbon atoms, such as phenyl, naphthyl, indenyl, and the like.
As used herein, the term "heteroaryl", alone or in combination, refers to a monovalent aromatic heterocyclic monocyclic or multicyclic ring system comprising one or more (e.g., 1,2, 3, or 4) heteroatoms selected from N, O and S, the remaining ring atoms being carbon, e.g., a C 5-6 heteroaryl group having 5 to 6 ring atoms, a C 5-10 heteroaryl group having 5 to 10 ring atoms, and a C 5-12 heteroaryl group having 5 to 12 ring atoms. Examples of heteroaryl groups include pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, triazinyl, azaBasic, diazaA group, isoxazolyl, benzofuranyl, isothiazolyl, benzothienyl, indolyl, isoindolyl isobenzofuranyl, benzimidazolyl, benzoxazolyl, and benzisoxazolyl, benzothiazolyl, and benzisothiazolyl, benzoxadiazolyl, benzothiadiazolyl, and benzotriazole group, purine group, quinoline group benzotriazole group, purine group quinolinyl radical.
As used herein, the term "halogen" refers to fluorine, chlorine, bromine or iodine.
As used herein, the term "hydroxy" refers to an-OH group.
As used herein, the term "mercapto" refers to a-SH group.
As used herein, the term "amino" refers to the-NH 2 group.
As used herein, the term "cyano" refers to a —cn group.
As used herein, the term "carbonyl" refers to a —cho group.
As used herein, the term "nitro" refers to a-NO 2 group.
As used herein, the term "stereoisomer" refers to a compound composed of the same atoms bonded through the same bonds but having different three-dimensional structures, which are not interchangeable. "stereoisomers" include enantiomers, diastereomers, and other stereoisomeric forms as defined by absolute stereochemistry. The present invention is intended to include all such possible isomers, as well as their racemic and optically pure forms.
As used herein, the term "geometric isomer" refers to an isomeric form that is attributed to a carbon-carbon double bond or other geometric asymmetric center.
As used herein, the term "tautomer" refers to an isomeric form resulting from the exchange of a single bond with an adjacent double bond along with concomitant proton migration.
As used herein, the term "pharmaceutically acceptable salt" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety into its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, salts with inorganic acids such as hydrochloric, hydrobromic, phosphoric and sulfuric acids, and salts with organic carboxylic and sulfonic acids such as acetic, trifluoroacetic, propionic, maleic, fumaric, malic, citric, tartaric, lactic, benzoic, methanesulfonic, ethanesulfonic, benzenesulfonic, toluenesulfonic, naphthalenesulfonic and naphthalenedisulfonic acids, or salts with conventional bases such as alkali metal salts (e.g., sodium and potassium salts), alkaline earth metal salts (e.g., calcium and magnesium salts), ammonium salts derived from ammonia and organic amines (e.g., diethylamine, triethylamine, ethyldiisopropylamine, procaine, dibenzylamine, N-methylmorpholine, dihydroabietylamine (dihydroabiethylamine), methylpiperidine, L-arginine, creatine, choline, L-lysine, ethylenediamine, N-dibenzylethylenediamine (benzathine), ethanolamine, meglumine and tromethamine).
As used herein, the term "prodrug" refers to a compound that undergoes bioconversion prior to exhibiting its pharmacological effect. Prodrugs can be prepared using well known methods, such as those described in Burgers pharmaceutical chemistry and drug development (1995) 172-178,949-982 (Manfred E.Wolff).
As used herein, the term "solvate" refers to a complex formed by coordination with a solvent molecule. When the solvent molecule is water, the solvate is a hydrate.
The compounds as described herein may be isotopically labeled, i.e., one or more atoms in which are replaced by atoms having different atomic masses or mass numbers. Such isotopically-labeled compounds are considered to be within the scope of the present invention. Examples of isotopes that can be incorporated into compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as, but not limited to 2H、3H、11C、13C、14C、13N、15N、15O、17O、18O、31P、32P、35S、18F、36Cl、123I and 125 I, respectively.
In one aspect, the present invention provides compounds of formula (I), and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates, or isotopically-labeled analogs thereof,
Wherein the method comprises the steps of
L is selected from -(NRL3)SO2N(RL1)(RL2)、-SO2RL1、-C(O)ORL1、-C(O)N(RL1)(RL2)、-C(O)NHORL1、-P(O)(ORL1)(ORL2)、-P(O)(NHRL1)(ORL2)、-P(O)(ORL1)(RL2)、-P(O)(NHRL1)(NHRL2)、-P(O)(NHRL1)(RL2)、-P(O)(RL1)(RL2)、-P(O)(SRL1)(SRL2)、-P(O)(NHRL1)(SRL2)、-P(O)(SRL1)(RL2) or-B (OH) 2, wherein R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, or optionally substituted aryl, or R L1 and R L2 are joined to form an optionally substituted cyclic structure;
A is selected from optionally substituted alkylene, optionally substituted cycloalkylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, optionally substituted cycloalkylene, optionally substituted heterocyclylene alkylene, optionally substituted arylene alkylene, or optionally substituted heteroarylene;
Z is selected from- (chemical bond )、-O-、-S-、-CO-、-SO-、-SO2-、-C(RZ1)(RZ2)-、-N(RZ1)-、-CH2N(RZ1)-、-N(RZ1)CH2-、-CON(RZ1)-、-N(RZ1)CO-、-SON(RZ1)-、-SO2N(RZ1)-、-N(RZ1)SO-、-N(RZ1)SO2- or Wherein R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
R 1 is selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl 、-OR3、-SR3、-NR3R4、-COOR3、-CONR3R4、-NR3COR4、-COR3、-OCOR3、-SONR3R4、-SO2NR3R4、-NR3SOR4、-NR3SO2R4, or-Si (R 3)(R4)(R5), wherein R 3、R4 or R 5 are independently selected from hydrogen, deuterium, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
n=0, 1, 2 or 3;
X 1、X2、X3 and X 4 are independently selected from N or C (R X), wherein R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl 、-ORa、-SRa、-NRaRb、-COORa、-CONRaRb、-NRbCORa、-CORa、-OCORa、-SONRaRb、-SO2NRaRb、-NRbSORa、-NRbSO2Ra, or-Si (R a)(Rb)(Rc), wherein R a、Rb and R c are independently selected from hydrogen, deuterium, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or X 1 is joined with X 2 to form an optionally substituted cyclic structure, or X 2 is joined with X 3 to form an optionally substituted cyclic structure, or X 3 is joined with X 4 to form an optionally substituted cyclic structure, or X 1 and Z are joined to form an optionally substituted cyclic structure.
In some embodiments, R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, or optionally substituted C 6-10 aryl.
In some embodiments, R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, and aryl, and alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, and aryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, alkyl (e.g., C 1-6 alkyl), alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), heterocyclyl (e.g., 4-6 membered heterocyclyl), -OC (O) -alkyl (e.g., -OC (O) -C 1-6 alkyl), -C (O) O-alkyl (e.g., -C (O) O-C 1-6 alkyl), -OC (O) O-alkyl (e.g., -OC (O) O-C 1-6 alkyl), phenyl, or halophenyl (e.g., chlorophenyl or fluorophenyl).
In some embodiments, L is selected from :-(NRL3)SO2N(RL1)(RL2)、-SO2RL1、-C(O)ORL1、-C(O)NHORL1、-P(O)(ORL1)(ORL2)、-P(O)(NHRL1)(ORL2)、-P(O)(NHRL1)(NHRL2) or-B (OH) 2.
In some embodiments, R L1、RL2 and R L3 are independently selected from hydrogen, amino, optionally substituted alkyl (e.g., C 1-6 alkyl), and optionally substituted aryl (e.g., C 6-10 aryl).
In some embodiments, R L1、RL2 and R L3 are independently selected from hydrogen, unsubstituted alkyl and phenyl, and alkyl substituted with one or more substituents independently selected from: -OC (O) -C 1-6 alkyl, -C (O) O-C 1-6 alkyl, -OC (O) O-C 1-6 alkyl, phenyl or halophenyl.
In some embodiments, L is selected from :-P(O)(OH)2、-C(O)OH、-C(O)NHOH、-B(OH)2、-SO2NH2、-NHSO2NH2、-P(O)(OCH2OC(O)C(CH3)3)2、-P(O)(OCH2OC(O)OCH(CH3)2)2、-P(O)(NHCH(CH3)C(O)OCH(CH3)2)2、-P(O)(OC6H5)(NHCH(CH3)C(O)OCH(CH3)2).
In some embodiments, R L1 and R L2 are linked to form an optionally substituted 5-or 6-membered cyclic structure.
In some embodiments, R L1 and R L2 are joined to form a cyclic structure that is substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted phenyl (e.g., halophenyl), or optionally substituted 5-6 membered heteroaryl.
In some embodiments, R L1 and R L2 are joined to form a cyclic structure substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, unsubstituted C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 4-6 membered heterocyclyl, phenyl, or 5-6 membered heteroaryl, or C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 4-6 membered heterocyclyl, phenyl, or 5-6 membered heteroaryl substituted with a substituent independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl, or alkoxy.
In some embodiments, L is-P (O) (OR L1)(ORL2),RL1 and R L2 are independently optionally substituted alkyl (e.g., C 1-3 alkyl such as methyl OR ethyl), and R L1 and R L2 are joined to form an optionally substituted 5-OR 6-membered cyclic structure, L is-P (O) (OR L1)(ORL2),RL1 is ethyl and R L2 is methyl substituted with phenyl OR halophenyl (e.g., chlorophenyl OR fluorophenyl), and R L1 and R L2 are joined to form a 6-membered ring structure substituted with phenyl OR halophenyl (e.g., chlorophenyl OR fluorophenyl).
In some embodiments, a is selected from optionally substituted C 1-6 alkylene, optionally substituted C 3-6 cycloalkylene, optionally substituted 4-10 membered heterocyclylene, optionally substituted C 6-10 arylene, optionally substituted 5-10 membered heteroarylene, optionally substituted C 3-6 cycloalkylene C 1-3 alkylene, optionally substituted 4-10 membered heterocyclylene C 1-3 alkylene, optionally substituted C 6-10 arylene C 1-3 alkylene, or optionally substituted 5-10 membered heteroarylene C 1-3 alkylene.
In some embodiments, A is selected from unsubstituted alkylene, cycloalkylene, heterocyclylene, arylene, heteroarylene, cycloalkylene, heterocyclylene alkylene, arylene alkylene, or heteroarylalkylene, and alkylene, cycloalkylene, heterocyclylene, arylene, heteroarylene, cycloalkylene, heterocyclylene alkylene, arylene, arylalkylene, or heteroarylalkylene substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, acetyl, alkyl (e.g., C 1-6 alkyl), alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), heterocyclyl (e.g., 4-6 membered heterocyclyl), O-alkyl (e.g., -OC 1-6 alkyl), NH-alkyl (e.g., -NHC 1-6 alkyl), S-alkyl (e.g., -SC 1-6 alkyl), O-cycloalkyl (e.g., -OC 1-6 cycloalkyl), NH-cycloalkyl (e.g., -NHC 1-6), or SC-cycloalkyl (e.g., SC 1-6 cycloalkyl).
In some embodiments, the substituent is located at the alkylene, cycloalkylene, heterocyclylene, arylene, or heteroarylene moiety of a. In some embodiments, the substituent is on the cycloalkylene, heterocycloalkylene, arylalkylene, or heteroarylene moiety (e.g., ortho or meta with respect to the alkylene moiety).
In some embodiments, a is selected from optionally substituted C 1-4 alkylene (e.g., methylene, ethylene, propylene, butylene), optionally substituted arylene (e.g., phenylene), optionally substituted cycloalkylene C 1-4 alkylene, optionally substituted arylene C 1-4 alkylene, or optionally substituted heteroarylene C 1-4 alkylene.
In some embodiments, a is selected from methylene, ethylene, propylene, butylene, phenylene C 1-2 alkylene (e.g., phenylene methylene or phenylene ethylene), cyclohexylene C 1-2 alkylene (e.g., cyclohexylene methylene), pyridylene C 1-2 alkylene (e.g., pyridylene methylene).
In some embodiments, a is selected from phenylene C 1-2 alkylene (e.g., phenylene methylene or phenylene ethylene) substituted with halogen (e.g., fluorine or chlorine) or-O-alkyl (e.g., -OC 1-6 alkyl, e.g., methoxy). In some embodiments, a is selected from the group consisting of fluoro-phenylene-methylene, chloro-phenylene-methylene, methoxy-phenylene-methylene.
In some embodiments, R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, or optionally substituted 4-6 membered heterocyclyl.
In some embodiments, R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, or heterocyclyl, and alkyl, alkenyl, alkynyl, cycloalkyl, or heterocyclyl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, alkyl (e.g., C 1-6 alkyl), alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), or heterocyclyl (e.g., 4-6 membered heterocyclyl).
In some embodiments, Z is selected from- (chemical bond) or-C (R Z1)(RZ2) -.
In some embodiments, R Z1 and R Z2 are independently selected from hydrogen and optionally substituted alkyl (e.g., C 1-6 alkyl, such as methyl).
In some embodiments, Z is selected from- (bond), -CH 2 -, or-CH (CH 3) -.
In specific embodiments, Z is-CH (CH 3) -, which has the (R) or (S) configuration.
In some embodiments, R 1 is selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted C 6-10 aryl 、-OR3、-SR3、-NR3R4、-COOR3、-CONR3R4、-NR3COR4、-COR3、-OCOR3、-SONR3R4、-SO2NR3R4、-NR3SOR4、-NR3SO2R4, or-Si (R 3)(R4)(R5).
In some embodiments, R 1 is selected from unsubstituted alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl, and alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxyl, optionally substituted alkyl, optionally substituted cycloalkyl, or optionally substituted alkoxy.
In some embodiments, R 1 is selected from alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, and alkyl, cycloalkyl, or alkoxy substituted with substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl, or alkoxy.
In some embodiments, R 1 is selected from alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl substituted with one or more substituents independently selected from deuterium, fluoro, chloro, bromo, iodo, hydroxy, mercapto, amino, cyano, nitro, alkyl, cycloalkyl, alkoxy, alkylamino, carboxy, haloalkyl, haloalkoxy.
In some embodiments, R 3、R4 and R 5 are independently selected from hydrogen, deuterium, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted C 6-10 aryl, or optionally substituted 5-6 membered heteroaryl.
In some embodiments, R 3、R4 and R 5 are independently selected from hydrogen, deuterium, unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, and alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxyl, optionally substituted alkyl, optionally substituted cycloalkyl, or optionally substituted alkoxy.
In some embodiments, R 3、R4 and R 5 are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, and alkyl, cycloalkyl, or alkoxy substituted with a substituent independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl, or alkoxy.
In some embodiments, R 3、R4 and R 5 are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl substituted with one or more substituents independently selected from deuterium, fluorine, chlorine, bromine, iodine, hydroxyl, mercapto, amino, cyano, nitro, alkyl, cycloalkyl, alkoxy, alkylamino, carboxyl, haloalkyl, haloalkoxy.
In some embodiments, R 1 is selected from deuterium, halogen, hydroxy, amino, cyano, alkyl, alkenyl, cycloalkyl, OR aryl (e.g., phenyl) that is unsubstituted OR substituted with halogen, OR-OR 3. In some embodiments, R 3 is independently selected from alkyl (e.g., C 1-6 alkyl) that is unsubstituted or substituted with halo.
In some embodiments, R 1 is selected from fluoro, chloro, bromo, iodo, cyano, methyl, t-butyl, methoxy.
In some embodiments, n is 0, 1, or 2.
In some embodiments, R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 cycloalkyl, Optionally substituted 4-6 membered heterocyclyl 、-ORa、-SRa、-NRaRb、-COORa、-CONRaRb、-NRbCORa、-CORa、-OCORa、-SONRaRb、-SO2NRaRb、-NRbSORa、-NRbSO2Ra or-Si (R a)(Rb)(Rc), wherein R a、Rb and R c are independently selected from hydrogen, Deuterium, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, An optionally substituted 4-6 membered heterocyclyl, an optionally substituted C 6-10 aryl or an optionally substituted 5-6 membered heteroaryl, and the substituents in said optional substitution in R X are selected from one or more of hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, alkyl (e.g. C 1-6 alkyl), alkyl, Alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), heterocyclyl (e.g., 4-6 membered heterocyclyl), aryl (e.g., C 6-10 aryl), or heteroaryl (e.g., 5-6 membered heteroaryl).
In some embodiments, R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, -OR a、-SRa, and-NR aRb, wherein R a and R b are independently selected from hydrogen, deuterium, and optionally substituted alkyl.
In some embodiments, R X is independently selected from unsubstituted alkyl (e.g., C 1-6 alkyl) or alkyl substituted with deuterium or halogen (e.g., C 1-6 alkyl).
In some embodiments, R a and R b are independently selected from unsubstituted alkyl (e.g., C 1-6 alkyl) or alkyl substituted with deuterium or halogen (e.g., C 1-6 alkyl).
In some embodiments, R X is independently selected from hydrogen, deuterium, halogen, hydroxy, amino, cyano, alkyl (e.g., C 1-4 alkyl such as methyl, ethyl, propyl, isopropyl, t-butyl), haloalkyl, deuterated alkyl, -OR a、-SRa, OR-NR aRb, wherein R a and R b are independently selected from hydrogen, deuterium, alkyl (e.g., C 1-4 alkyl such as methyl OR ethyl), deuterated alkyl, OR haloalkyl.
In some embodiments, R X is independently selected from hydrogen, deuterium, fluorine, chlorine, bromine, iodine, hydroxyl, amino, cyano, methyl, ethyl, isopropyl, tert-butyl, trifluoromethyl, deuterated alkyl (e.g., 1D-methyl, 2D-methyl, and 3D-methyl), methoxy, ethoxy, methylthio, methylamino, or dimethylamino.
In some embodiments, X 1、X2、X3 and X 4 are independently C (R X).
In some embodiments, X 1、X2、X3 and X 4 are CH.
In some embodiments, one (e.g., X 1、X2、X3 or X 4) or both (e.g., X 1 and X 3) of X 1、X2、X3 and X 4 are independently C (R X), wherein R X is as defined above (excluding hydrogen), and the remainder are CH.
In some embodiments, one (e.g., X 1、X2、X3 or X 4) or two (e.g., X 1 and X 3;X2 and X 4) of X 1、X2、X3 and X 4 are independently N, the remainder are independently C (R X), wherein R X is as defined above.
In some embodiments, one of X 1、X2、X3 and X 4 is N, the remainder are CH.
In some embodiments, X 1 and X 2 are joined to form an optionally substituted 5-or 6-membered cyclic structure, or X 2 and X 3 are joined to form an optionally substituted 5-or 6-membered cyclic structure, or X 3 and X 4 are joined to form an optionally substituted 5-or 6-membered cyclic structure, or X 1 and Z are joined to form an optionally substituted 5-or 6-membered cyclic structure.
In some embodiments, the cyclic structure is substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted aryl, or optionally substituted 5-6 membered heteroaryl.
In some embodiments, the cyclic structure is substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, unsubstituted C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 4-6 membered heterocyclyl, aryl, or 5-6 membered heteroaryl, or C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 4-6 membered heterocyclyl, aryl, or 5-6 membered heteroaryl substituted with a substituent independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl, or alkoxy.
In some embodiments, the compound of formula (I), and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates, or isotopically-labeled analogs thereof, are selected from the group consisting of:
in another aspect, the present invention provides a pharmaceutical composition comprising the compound of formula (I) of the present invention and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates or isotopically-labeled analogues thereof as an active ingredient. The pharmaceutical composition can be used for preventing and/or treating diseases mediated by ENPP 1.
In some embodiments, the pharmaceutical compositions of the invention may further comprise additional agents (such as small molecules, polypeptides, nucleic acids, antibodies, antibody-coupled agents) for the prevention and/or treatment of cancer or tumor. The compound of formula (I) of the present invention, or a stereoisomer, a geometric isomer, a tautomer, a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isotopically labeled analog thereof, may provide enhanced anticancer effects when administered in combination with another drug for preventing and/or treating cancer or tumor, or a pharmaceutically acceptable salt thereof.
In some embodiments, representative examples of drugs for treating cancer or tumors include, but are not limited to: cell signaling inhibitors, chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozotocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin C, ixabepilone, tamoxifen, flutamide, gonadorelin analog, megestrol, prednisone, dexamethasone, methylprednisolone, thalidomide, fludarifenacin, tretinomycin, tretinoin, mitomycin, valproic, tamoxifen, and other drugs interferon alpha, calcium folinate, sirolimus lipidated, everolimus, afatinib, alisertib, amuvatinib, apatinib, axitinib, bortezomib, bosutinib, britinib, cabotinib, ceritinib, crenolanib, crizotinib, dabrafenib, dacatinib, daratinib, dacatinib, multi-vitamin tinib, erlotinib, foretinib, ganetespib, gefitinib, ibrutinib, enotinib, icotinib, imatinib, iniparib, lapatinib, lenvatinib (lenvatinib), linifanib, linsitinib, mosaic tinib, momelotinib, mo Tisha, lenatinib, nilatinib, nilapatinib (niraparib), oprozomib, olaparib, pictilisib, ponatinib, quinitinib (quizartinib), regtinib, rigosertib, lu Kapa panib, ruxolitinib, celecoxib, saridegib, sorafenib, sunitinib, tiratinib, tivantinib, tizanib, tofacitinib, trimetinib, vandetanib, valipanib, vemurafenib, vemoji, volasertib, alemtuzumab, bevacizumab, belantuo Shan Kangwei statin, katussah mab, cetuximab, denoxib mab, gemtuzumab, ipilimumab, nituzumab, ofatuzumab, panitumumab, rituximab, tositumomab, trastuzumab, PI3K inhibitors, CSF1R inhibitors, A2A and/or A2B receptor antagonists, IDO inhibitors, anti-PD-1 antibodies, anti-PD-L1 antibodies, LAG3 antibodies, TIM-3 antibodies, tit antibodies, CD47 antibodies, CLAUDIN 18.2.2 antibodies, anti-CTLA-4 antibodies, or any combination thereof.
In some embodiments, the pharmaceutical compositions of the invention may further comprise an additional Antibody-coupled drug (anti-Drug Conjugates, ADC) for the prevention and/or treatment of cancer or tumor. The compound of formula (I) of the present invention, or a stereoisomer, a geometric isomer, a tautomer, a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate or an isotopically labeled analog thereof, may provide enhanced anticancer effects when administered in combination with another antibody-conjugated drug for preventing and/or treating cancer or tumor.
In some embodiments, representative examples of antibody-conjugated drugs for preventing and/or treating cancer or tumors may include, but are not limited to, enmeflozumab, veltuzumab, oxtuzumab, vidipine, goli Sha Tuozhu mab, de Lu Tikang-trastuzumab, or any combination thereof.
In some embodiments, the pharmaceutical compositions of the present invention may further comprise additional therapeutic agents useful for the treatment of inflammatory, autoimmune, and immune-mediated diseases. The compounds of formula (I) of the present invention, or pharmaceutically acceptable salts, prodrugs, hydrates, solvates or isotopically-labeled analogs thereof, may provide enhanced therapeutic effects when administered in combination with additional therapeutic agents for the treatment of inflammatory, autoimmune and immune-mediated diseases.
In some embodiments, representative examples of therapeutic agents for the treatment of inflammatory, autoimmune, and immune-mediated diseases may include, but are not limited to, steroidal drugs (e.g., prednisone, hydropannisone, methyl hydropaniture, cortisone, hydroxy cortisone, betamethasone, dexamethasone, and the like), methotrexate, leflunomide, anti-tnfα agents (e.g., etanercept, infliximab, ada Li Shan anti, and the like), calcineurin inhibitors (e.g., tacrolimus, pimecrolimus, and the like), antihistamines (e.g., diphenhydramine, hydroxyzine, loratadine, ebastine, ketotifen, cetirizine, levocetirizine, fexofenadine, and the like), or any combination thereof.
In a further aspect, the present invention provides a method of preventing and/or treating a disease mediated by ENPP1, comprising administering to a patient in need thereof a compound of formula (I) or a pharmaceutical composition of the present invention.
In a further aspect, the present invention provides the use of a compound of formula (I) or a pharmaceutical composition of the invention in the manufacture of a medicament for the prevention and/or treatment of a disease mediated by ENPP 1.
In some embodiments, the disease mediated by ENPP1 comprises cancer, a tumor, an inflammatory disease, an autoimmune disease, a neurodegenerative disease, an attention-related disease, or an immune-mediated disease.
In some embodiments, the disease mediated by ENPP1 comprises cancer, a tumor, an inflammatory disease, an autoimmune disease, or an immune-mediated disease.
In some embodiments, the disease mediated by ENPP1 is selected from cancer or tumor. Representative examples of cancers and tumors include, but are not limited to, skin cancer, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, lung cancer, bone cancer, brain cancer, neuroblastoma, rectal cancer, colon cancer, familial adenomatous polyposis, hereditary non-polyposis colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, hypopharynx cancer, tongue cancer, salivary gland cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, renal parenchymal cancer, cervical cancer, uterine body cancer, endometrial cancer, choriocarcinoma, testicular cancer, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors hodgkin's lymphoma, non-hodgkin's lymphoma, burkitt's lymphoma, leukemia, acute Lymphoblastic Leukemia (ALL), chronic Lymphocytic Leukemia (CLL), acute Myelogenous Leukemia (AML), chronic Myelogenous Leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gall bladder carcinoma, bronchogenic carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, multiple myeloma, basal cell carcinoma, teratoma, retinoblastoma, choriocarcinoma, seminoma, rhabdomyosarcoma, craniopharyngeal tubular carcinoma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, ewing's sarcoma, and plasmacytoma.
In some embodiments, the diseases mediated by ENPP1 are selected from inflammatory diseases, autoimmune diseases, and immune-mediated diseases, representative examples of which include, but are not limited to, arthritis, rheumatoid arthritis, spinal arthritis, gouty arthritis, osteoarthritis, juvenile arthritis, other arthritic conditions, lupus, systemic Lupus Erythematosus (SLE), skin-related diseases, psoriasis, eczema, dermatitis, allergic dermatitis, pain, lung diseases, pulmonary inflammation, adult Respiratory Distress Syndrome (ARDS), pulmonary sarcoidosis, chronic pulmonary inflammatory diseases, chronic Obstructive Pulmonary Disease (COPD), cardiovascular diseases, atherosclerosis, myocardial infarction, congestive heart failure, myocardial ischemia reperfusion injury, inflammatory bowel disease, crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, sjogren's syndrome, autoimmune thyroid diseases, urticaria (rubella), multiple sclerosis, scleroderma, organ transplant rejection, xenograft, idiopathic thrombocytopenic purpura (p), parkinson's disease, alzheimer's disease, allergic diseases, diabetes, allergic diseases, sinusitis, allergic diseases, and sinus diseases.
Examples
The invention is illustrated hereinafter by the following non-limiting examples.
The starting materials in the examples of the present application are known and available from commercial suppliers or may be synthesized according to methods known in the art.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) and/or liquid chromatography (LC-MS) and/or liquid chromatography (HPLC). NMR was performed using Bruker AVANCE NEO MHz, LC-MS was performed using LCMS WATERS ACQUITY UPLC H-Class PLUS and/or SQD2, and HPLC was performed using WATERS ACQUITYUPLC and/or Agilent 1260.
Example 1 preparation of (4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 (4-methylbenzyl) preparation of di-tert-butyl phosphonate:
Di-tert-butyl phosphonate (20.0 g,108mmol,1.0 eq) was dissolved in tetrahydrofuran (200 mL), sodium hydride (0.516 g,129mmol,1.2 eq) was added at 0deg.C and the reaction stirred for half an hour, then 1-bromomethyl-4-methylbenzene (16.4 g,119mmol,1.1 eq) was added and the temperature was raised to 70deg.C for 16 hours. Water (100 mL) was added to the reaction solution to quench the reaction, and the reaction solution was extracted three times with ethyl acetate (100 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (20.0 g, yield: 62.1%) as a brown solid. LCMS (ESI) [2m+h ] + = 597.8.
Step 2 preparation of di-tert-butyl (4-bromomethyl) benzyl) phosphonate:
Di-tert-butyl (4-methylbenzyl) phosphonate (20.0 g,67.0mmol,1.0 eq) was dissolved in carbon tetrachloride (200 mL), and N-bromosuccinimide (NBS) (13.1 g,73.7mmol,1.1 eq) and dibenzoyl peroxide (BPO) (800 mg,3.40mmol,0.05 eq) were then added to the mixture and reacted at 78℃for 3 hours with stirring. The reaction solution was concentrated under reduced pressure, and after re-diluted with ethyl acetate, the reaction was quenched with water (100 mL) and extracted three times with ethyl acetate (100 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (20.0 g (crude product, yield: 79.1%) as a yellow liquid .1HNMR(400MHz,DMSO-d6)δ7.38(d,J=8.0Hz,2H),7.26(dd,J=8.0,2.0Hz,2H),4.67(s,2H),3.26(d,J=21.6Hz,2H),1.21(s,18H).
Step 3 preparation of di-tert-butyl 4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
3, 6-Dichlorocarbazole (100 mg,0.42mmol,1 eq.) was added to N, N-dimethylformamide (2 mL), sodium hydride (60% by mass, 34mg,0.84mmol,2 eq.) was added, and after stirring at 0℃for half an hour, di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (175 mg,0.42mmol,1 eq.) was added and the reaction mixture stirred at room temperature for 16 hours. The reaction solution was extracted three times with ethyl acetate (20 ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=100% -1/1) to give the title compound (53 mg, yield 23.5%) as a white solid LCMS (ESI) [ m+1] + =532.1.
Step 4 preparation of (4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonate (53 mg,0.1mmol,1 eq.) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (2.28 mg,0.02mmol,0.2 eq.) was added and the reaction stirred at room temperature for 1 hour. Filtering to obtain white solid, (4- ((3, 6-dichloro-9H-carbazole-9-yl) methyl) benzyl) phosphonic acid (32mg,0.08mmol,76.5%).LCMS(ESI)[M+1]+=420.1;1H NMR(400MHz,DMSO-d6)δ10.5(br,1H),8.37(d,J=2.0Hz,2H),7.70(d,J=8.8Hz,2H),7.49(dd,J=8.8,2.1Hz,2H),7.21-6.98(m,4H),5.64(s,2H),2.86(d,J=21.4Hz,2H).
Example 2
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3, 6-dimethoxycarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 2.LCMS(ESI)[M+1]+=412.2;1H NMR(400MHz,CD3OD)δ7.59(d,J=2.4Hz,2H),7.32(d,J=7.2Hz,2H),7.27(d,J=8.0Hz,2H),7.01-6.98(m,4H),5.43(s,2H),3.89(s,6H),2.80(d,J=20.0Hz,2H).
Example 3
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3, 6-dimethylcarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 3.LCMS(ESI)[M+1]+=380.2;1H NMR(400MHz,DMSO-d6)δ7.89(s,2H),7.45(d,J=8.3Hz,2H),7.21(d,J=8.2Hz,2H),7.05(dd,J=33.6,7.1Hz,4H),5.52(s,2H),2.83(s,2H),2.45(s,6H).
Example 4
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3, 6-dibromocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 4.LCMS(ESI)[M+1]+=506.9;1H NMR(400MHz,DMSO-d6)δ8.51(d,J=2.1Hz,2H),7.66(dd,J=8.7,2.9Hz,2H),7.60(ddd,J=8.7,3.2,1.9Hz,2H),7.13(dd,J=8.2,2.5Hz,2H),7.08-7.01(m,2H),5.64(s,2H),2.93-2.78(m,2H).
Example 5
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3, 6-diiodocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 5.LCMS(ESI)[M+1]+=603.1;1H NMR(400MHz,DMSO-d6)δ8.64(d,J=1.7Hz,2H),7.73(dd,J=8.6,1.8Hz,2H),7.53(d,J=8.6Hz,2H),7.12(dd,J=8.2,2.4Hz,2H),7.04(d,J=7.9Hz,2H),5.61(s,2H),2.86(d,J=21.4Hz,2H).
Example 6
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 2, 7-dimethylcarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 6.LCMS(ESI)[M+1]+=380.2;1H NMR(400MHz,DMSO-d6)δ7.97(d,J=7.6Hz,2H),7.36(s,2H),7.15-7.08(m,2H),7.01(t,J=8.4Hz,4H),5.53(s,2H),2.84(d,J=21.2Hz,2H),2.45(s,6H).
Example 7
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3-chlorocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 7.LCMS(ESI)[M+1]+=386.1;1H NMR(400MHz,DMSO-d6)δ8.38-8.15(m,2H),7.66(t,J=7.6Hz,2H),7.52-7.38(m,2H),7.23(t,J=7.2Hz,1H),7.14-7.04(m,4H),5.63(s,2H),2.85(d,J=21.2Hz,2H).
Example 8
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3-methoxycarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 8.LCMS(ESI)[M+1]+=382.1;1H NMR(400MHz,DMSO-d6)δ8.15(d,J=7.6Hz,1H),7.75(d,J=2.4Hz,1H),7.59(d,J=8.4Hz,1H),7.54(d,J=8.8Hz,1H),7.40(t,J=7.2Hz,1H),7.21-7.00(m,6H),5.58(d,J=2.0Hz,2H),3.86(s,3H),2.86(d,J=21.2Hz,2H).
Example 9
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 2, 7-dimethoxycarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 9.LCMS(ESI)[M+1]+=412.1;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.6Hz,2H),7.48(dd,J=8.9,2.4Hz,2H),7.18-6.93(m,6H),5.50(s,2H),3.83(d,J=2.4Hz,6H),2.85(d,J=21.2Hz,2H).
Example 10
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3, 6-di-tert-butylcarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 10.LCMS(ESI)[M+1]+=463.3;1HNMR(400MHz,DMSO-d6)δ8.19(s,2H),7.47(d,J=2.8Hz,4H),7.08(q,J=8.2Hz,4H),5.51(s,2H),2.82(d,J=21.3Hz,2H),1.39(s,18H).
Example 11
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3, 6-dicyanocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 11.LCMS(ESI)[M+1]+=402.3;1H NMR(400MHz,DMSO-d6)δ8.87(s,2H),7.95(t,J=1.9Hz,4H),7.12(q,J=7.8Hz,4H),5.77(s,2H),2.87(d,J=21.4Hz,2H).
Example 12
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 2-fluorocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 12.LCMS(ESI)[M+1]+=370.1;1H NMR(400MHz,CD3OD)δ8.07-8.03(m,2H),7.46(d,J=8.4Hz,1H),7.39-7.36(m,1H),7.24-7.16(m,4H),7.05(d,J=7.6Hz,2H),6.95-6.91(m,1H),5.51(s,2H),2.91(d,J=20.8Hz,2H).
Example 13
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3-fluorocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 13.LCMS(ESI)[M+1]+=370.1;1HNMR(400MHz,CD3OD)δ8.07(d,J=7.6Hz,1H),7.79(dd,J=9.2,2.4Hz,1H),7.48-7.30(m,3H),7.22-7.13(m,4H),7.03(d,J=8.0Hz,2H),5.53(s,2H),2.89(d,J=20.8Hz,2H).
Example 14
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 9H-pyrido [2,3-b ] indole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 14.LCMS(ESI)[M+1]+=353.1;1H NMR(400MHz,CD3OD)δ8.49(dd,J=7.6,2.0Hz,1H),8.43(dd,J=5.2,2.0Hz,1H),8.13(d,J=8.0Hz,1H),7.47(d,J=8.0Hz,1H),7.46-7.42(m,1H),7.27-7.25(m,2H),7.24-7.22(m,2H),7.11(d,J=7.6Hz,2H),5.67(s,2H),2.87(d,J=20.6Hz,2H).
Example 15
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 9H-pyrido [3,4-b ] indole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 15.LCMS(ESI)[M+1]+=353.1;1H NMR(400MHz,CD3OD)δ8.85(s,1H),8.32(d,J=5.6Hz,1H),8.23(d,J=8.0Hz,1H),8.13(d,J=5.6Hz,1H),7.65(d,J=8.0Hz,1H),7.60(t,J=8.0Hz,1H),7.32-7.31(m,1H),7.31-7.29(m,2H),7.04(d,J=8.0Hz,2H),5.64(s,2H),2.81(d,J=20.0Hz,2H).
Example 16
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 5H-pyrido [4,3-b ] indole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 16.LCMS(ESI)[M+1]+=353.1;1H NMR(400MHz,CD3OD)δ9.68(s,1H),8.59(dd,J=7.2,1.6Hz,1H),8.29(d,J=8.0Hz,1H),7.85(d,J=6.8Hz,1H),7.72-7.66(m,2H),7.50-7.46(m,1H),7.42(dd,J=8.0,2.0Hz,2H),7.37(d,J=8.0Hz,2H),5.72(s,2H),2.97(d,J=20.8Hz,2H).
Example 17
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 5H-pyrido [3,2-b ] indole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 17.LCMS(ESI)[M+1]+=353.1;1H NMR(400MHz,CD3OD)δ8.43(dd,J=4.8,1.2Hz,1H),8.33(d,J=8.0Hz,1H),7.99(dd,J=8.4,1.2Hz,1H),7.62(d,J=8.4Hz,1H),7.59-7.54(m,1H),7.45(dd,J=8.4,4.8Hz,1H),7.31(t,J=7.2Hz,1H),7.23(dd,J=8.0,2.4Hz,2H),7.07(d,J=8.0Hz,2H),5.60(s,2H),2.90(d,J=21.2Hz,2H).
Example 18
Similarly, the preparation of example 1 was carried out according to steps 3-4 of the preparation method of example 1 using commercially available 3-bromocarbazole and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate as starting materials 18.LCMS(ESI)[M+1]+=428.1;1H NMR(400MHz,CD3OD)δ8.22(d,J=1.7Hz,1H),8.08(d,J=7.8Hz,1H),7.55-7.33(m,4H),7.28(dd,J=8.2,1.9Hz,2H),7.20(t,J=7.0Hz,1H),7.00(d,J=8.0Hz,2H),5.51(s,2H),2.81(d,J=19.8Hz,1H).
Example 19 preparation of (4- ((3- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 (preparation of diethyl 4- (bromomethyl) benzyl) phosphonate:
1, 4-bis (bromomethyl) benzene (6 g,22.73mmol,1 eq.) was dissolved in 1, 4-dioxane solution (10 mL) followed by the addition of triethyl phosphite (15.11 g,90.92mmol,4 eq.). The reaction solution was subjected to microwave reaction in a microwave tube at 100℃for 1 hour. The reaction solution was concentrated at 60℃with an oil pump, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/V=10/1)) to give the title compound (2.3 g, yield: 31.51%) as a colorless oil. LCMS (ESI) [ m+h ] + = 321.2.
Step 2 preparation of 4- (3-bromo-9H-carbazole-9-methyl) benzylphosphonic acid diethyl ester:
3-bromocarbazole (350 mg,1.42mmol,1 eq.) was dissolved in ultra-dry tetrahydrofuran (10 mL), followed by the addition of diethyl (4- (bromomethyl) benzyl) phosphonate (913 mg,2.84mmol,2 eq.) and sodium hydride (454 mg,11.38mmol,8 eq.). The reaction was stirred at room temperature for 1 hour under argon protection and monitored by LCMS for completion. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by flash chromatography (silica gel, 100% ethyl acetate) to give the title compound (350 mg, yield: 50.6%) as a yellow solid. LCMS (ESI) [ m+h ] + = 486.0.
Step 3 preparation of diethyl 4- (3- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -9H-carbazole-9-methyl) benzyl) phosphonate:
Diethyl 4- (3-bromo-9H-carbazole-9-methyl) benzylphosphonate (340 mg,0.7mmol,1 eq) was dissolved in1, 4-dioxane (8 mL), pinacol diboronate (266 mg,1.05mmol,1.5 eq) and potassium acetate (137 mg,1.4mmol,2 eq) were added, and finally 1, 1-bis (diphenylphosphine) ferrocene palladium dichloride (103 mg,0.14mmol,0.2 eq) was added. The reaction was allowed to react for 1 hour at 110℃under argon. LCMS monitored completion of the reaction. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (15 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (340 mg, yield: 91%) as a pale yellow solid. LCMS (ESI) [ m+h ] + = 533.4.
Step 4 preparation of diethyl (4- ((3-trifluoromethyl-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
Diethyl 4- (3- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -9H-carbazole-9-methyl) benzyl) phosphonate (40 mg,0.07mmol,1 eq.) was dissolved in N, N-Dimethylformamide (DMF) (4 mL) and cesium carbonate (49 mg,0.15mmol,2 eq.) and trifluoromethyl (1, 10-diazaphenanthrene) copper (I) (35 mg,0.11mmol,1.5 eq.) were added. After stirring the reaction at 50 ℃ for 2 hours under argon, the reaction was monitored to be complete by LCMS. Saturated aqueous ammonium chloride (10 mL) was added and the mixture was extracted with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude title compound (20 mg, yield: 56.1%) as a yellow solid. LCMS (ESI) [ m+na ] + =498.2.
Step 5 preparation of (4- ((3- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((3-trifluoromethyl-9H-carbazol-9-yl) methyl) benzyl) phosphonate (20 mg,0.04mmol,1 eq) was dissolved in anhydrous dichloromethane (2 mL) and trimethylbromosilane (0.21 mL,1.37mmol,32.6 eq) was added. The reaction was heated to 50 ℃ and stirred for 1 hour. LCMS monitored completion of the reaction. Water (2 mL) was added to the reaction, and the organic and aqueous phases were separated. The organic phase was dried over anhydrous sodium sulfate, filtered, dried by spin-drying, dissolved in tetrahydrofuran, and prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (1.55 mg) as a white solid.
Example 20 preparation of (4- ((2- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((2-bromo-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
2-bromo-9H-carbazole (300 mg,1.22mmol,1 eq) was dissolved in anhydrous tetrahydrofuran (2 mL), sodium hydride (495mg, 4.88mmol,4 eq) was added, and after stirring at room temperature under nitrogen for 0.5 hours, diethyl (4- (bromomethyl) benzyl) phosphonate (783 mg,2.44mmol,2 eq) was added and reacted at room temperature under nitrogen for 1 hour. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was purified using a preparative plate (PE: EA (V/v=3/1)) to give the title compound (200 mg, yield: 33.7%) as a yellow solid. LCMS (ESI) [ m+h ] + =508.1.
Step 2 preparation of diethyl (4- ((2- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((2-bromo-9H-carbazol-9-yl) methyl) benzyl) phosphonate (150 mg,0.31mmol,1 eq) was dissolved in1, 4-dioxane (2 mL), pinacol ester (117 mg,0.46mmol,1.5 eq) and potassium acetate (61 mg,0.62mmol,2 eq) were added, and finally 1, 1-bis (diphenylphosphine) ferrocene palladium dichloride (45 mg,0.06mmol,0.2 eq) was added. The reaction solution was subjected to a microwave reaction at 110℃for 1 hour under nitrogen protection. After cooling to room temperature, a saturated aqueous ammonium chloride solution (10 mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (15 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was purified by passing through a silica gel preparation plate (100% ethyl acetate) to give the title compound (120 mg, yield: 54.7%) as a pale yellow solid. LCMS (ESI) [ m+h ] + =534.2.
Step 3 (4- ((2- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid diethyl ester:
Diethyl (4- ((2- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (80 mg,0.15mmol,1 eq.) was dissolved in N, N-dimethylformamide (1 mL), cesium carbonate (98 mg,0.3mmol,2 eq.) and copper (I) trifluoromethyl (1, 10-diazaphenanthrene) (70 mg,0.22mmol,1.5 eq.) were added and reacted under argon for 16 hours at 50℃saturated aqueous ammonium chloride (10 mL) was added and the mixture was extracted three times with ethyl acetate (10 mL. Times 3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude title compound (45 mg, yield: 50.5%) as a yellow solid. LCMS (ESI) [ m+h ] + = 476.1.
Step 4 preparation of (4- ((2- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (35 mg,0.07mmol,1 eq) was dissolved in anhydrous dichloromethane (1 mL) and trimethylbromosilane (56.35 mg,0.37mmol,5 eq) was added and the reaction heated to 50deg.C and stirred for 2 hours. The reaction solution was concentrated at low temperature, and the crude product was purified by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (2.62 mg, yield: 8.5%) as a white solid .LCMS(ESI):[M-H]-=418.2;1H NMR(400MHz,CD3OD)δ8.27(d,J=8.0Hz,1H),8.19(d,J=8.0Hz,1H),7.79(s,1H),7.55(d,J=8.0Hz,1H),7.51-7.46(m,2H),7.29-7.27(m,1H),7.25-7.22(m,2H),7.05(d,J=8.0Hz,2H),5.63(s,2H),2.94(d,J=20.9Hz,2H).
Example 21 preparation of (4- ((4- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 preparation of diethyl (4- ((4-bromo-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
4-bromo-9H-carbazole (500 mg,2.04mmol,1 eq) was dissolved in ultra-dry tetrahydrofuran (10 mL), followed by the addition of diethyl (4- (bromomethyl) benzyl) phosphonate (653 mg,4.08mmol,2 eq) and sodium hydride (653 mg,16.3mmol,8 eq). After the reaction solution was stirred at room temperature for 1 hour under the protection of argon, a saturated aqueous ammonium chloride solution (10 mL) was added to the reaction mixture, and the mixture was extracted three times with ethyl acetate (10 ml×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by flash chromatography (silica gel, 100% ethyl acetate) to give the title compound (500 mg, yield: 50.6%) as a yellow solid. LCMS (ESI) [ m+h ] + = 486.0.
Step 2 preparation of diethyl (4- ((4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((4-bromo-9H-carbazol-9-yl) methyl) benzyl) phosphonate (500 mg,1.03mmol,1 eq) was dissolved in1, 4-dioxane (10 mL), pinacol ester (390 mg,1.55mmol,1.5 eq) and potassium acetate (202 mg,2.06mmol,2 eq) were added, and finally 1, 1-bis (diphenylphosphine) ferrocene palladium dichloride (150 mg,0.21mmol,0.2 eq) was added. The reaction was stirred at 110℃for 1 hour under argon. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (15 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (500 mg, yield: 91.1%) as a pale yellow solid. LCMS (ESI) [ m+h ] + =534.2.
Step 3 preparation of diethyl (4- ((4- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate
Diethyl (4- ((4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (160 mg,0.28mmol,1 eq.) was dissolved in N, N-dimethylformamide (4 mL) followed by cesium carbonate (195 mg,0.60mmol,2 eq.) and copper (I) trifluoromethyl (1, 10-diazaphenanthrene) (141 mg,0.44mmol,1.5 eq.) were added. The reaction was stirred at 50℃for 16 hours under argon, saturated aqueous ammonium chloride (10 mL) was added to the reaction, and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude title compound (50 mg, yield: 37.6%) as a yellow solid. LCMS (ESI) [ m+h ] + =476.2.
Step 4 preparation of (4- ((4- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((4- (trifluoromethyl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (35 mg,0.07mmol,1 eq.) was dissolved in dichloromethane (1 mL) and trimethylbromosilane (54 mg,0.35mmol,5 eq.) was added and the reaction heated to 50deg.C and stirred for 2 hours. The reaction solution was concentrated at low temperature, and the crude product was purified by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (6.24 mg, yield: 21.3%) as a white solid .LCMS(ESI):[M-H]-=418.2;1H NMR(400MHz,CD3OD)δ8.27(d,J=8.0Hz,1H),7.79(d,J=6.8Hz,1H),7.59(d,J=8.4Hz,1H),7.54-7.48(m,3H),7.28-7.21(m,3H),7.04(d,J=8.0Hz,2H),5.65(s,2H),2.93(d,J=21.2Hz,2H).
Example 22 preparation of (4- ((2, 4, 6-trifluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 preparation of 2-bromo-N- (3, 5-difluorophenyl) -4-fluoroaniline:
After 1,3, 5-trifluorobenzene (2.00 g,15.1mmol,1 eq.) was dissolved in dimethyl sulfoxide (10 mL), 2-bromo-4-fluoroaniline (3.45 g,18.2mmol,1.2 eq.) and potassium tert-butoxide (1.70 g,15.1mmol,1 eq.) were added and reacted at room temperature under argon for 2 hours. The reaction solution was quenched with saturated ammonium chloride (10 mL), extracted three times with ethyl acetate (10 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give a crude product which was purified by flash chromatography (silica gel, 100% petroleum ether) to give the title compound (700 mg, yield: 15.3%). LCMS (ESI) [ M-H ] - = 299.9.
Step 2, preparation of 2,4, 6-trifluoro-9H-carbazole:
2-bromo-N- (3, 5-difluorophenyl) -4-fluoroaniline (500 mg,1,66mmol,1 eq.) was dissolved in N-methylpyrrolidone (10 mL), and tert-butanol (477 mg,4.97mmol,3 eq.), cuprous iodide (63 mg,0.33mmol,0.2 eq.) and palladium acetate (37 mg,0.17mmol,0.1 eq.) were added. The reaction system was reacted for 2 hours at 150 ℃ under argon protection. The reaction solution was quenched with saturated ammonium chloride (10 mL), extracted three times with ethyl acetate (10 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give crude product, which was purified by silica gel preparation plate (dichloromethane: petroleum ether (V/v=2/1) to give the title compound (300 mg, yield: 81.9%). LCMS (ESI): M-H - =220.0.
Step 3 preparation of diethyl (4- ((2, 4, 6-trifluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
2,4, 6-trifluoro-9H-carbazole (20 mg,0.09mmol,1 eq) was dissolved in tetrahydrofuran (2 mL), followed by addition of sodium hydride (14 mg,0.36mmol,4 eq) and stirring at room temperature under nitrogen for 0.5H, followed by addition of diethyl (4- (bromomethyl) benzyl) phosphonate (57 mg,0.18mmol,2 eq). The reaction was stirred for 1 hour. The reaction solution was quenched with saturated ammonium chloride (5 mL), extracted three times with ethyl acetate (5 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (10 mg, 24.1% yield). LCMS (ESI) [ m+h ] + =462.1.
Step 4 preparation of (4- ((2, 4, 6-trifluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
(4- ((2, 4, 6-trifluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid (10 mg,0.02mmol,1 eq.) was dissolved in dichloromethane (1 mL) followed by the addition of trimethylbromosilane (16 mg,0.11mmol,5 eq.). The reaction system was stirred at 50 ℃ for 2 hours. The reaction solution was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (2.11 mg, yield) 24.0%).1H NMR(400MHz,CD3OD)δ7.74(dd,J=7.2,1.6Hz,1H),7.50(dd,J=8.8,4.0Hz,1H),7.25(dd,J=8.0,4.0Hz,2H),7.20(td,J=9.2,2.4Hz,1H),7.11(dd,J=9.6,2.0Hz,1H),7.03(d,J=7.6Hz,2H),6.76(td,J=10.0,2.0Hz,1H),5.52(s,2H),2.89(d,J=20.8Hz,2H).
Example 23
Similarly, the preparation of example 22 was carried out according to steps 1-4 of the preparation method of example 22 using commercially available 2-bromoaniline as starting material 23.LCMS(ESI)[M+1]+=388.1;1H NMR(400MHz,CD3OD)δ8.08(d,J=7.8Hz,1H),7.51(d,J=8.4Hz,1H),7.45-7.40(m,1H),7.31-7.28(m,1H),7.27-7.21(m,2H),7.08(dd,J=8.4,2.0Hz,1H),7.04(d,J=8.0Hz,2H),6.74(td,J=10.0,2.0Hz,1H),5.52(s,2H),2.90(d,J=19.2Hz,2H).
Example 24
Similarly, the preparation of example 22 was carried out according to steps 1-4 of the preparation method of example 22 using commercially available 2-bromo-5-fluoroaniline as starting material 24.LCMS(ESI)[M+1]+=406.1;1H NMR(400MHz,CD3OD)δ8.04(dd,J=8.4,6.6Hz,1H),7.28-7.20(m,3H),7.12(dd,J=9.6,2.8Hz,1H),7.05-6.99(m,3H),6.81-6.50(m,1H),5.50(s,2H),2.90(d,J=20.8Hz,2H).
EXAMPLE 25 preparation of (4- ((1-amino-3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 preparation of 1-nitro-3, 6-dichloro-9H-carbazole
3, 6-Dichloro-9H-carbazole (300 mg,1.27mmol,1 eq.) was dissolved in a mixed solvent of 1, 4-dioxane (3 mL) and acetic acid (3 mL), sodium nitrite (175 mg,2.54mmol,2 eq.) was slowly added thereto, and finally the reaction solution was stirred at 110℃for 4 hours. The reaction solution was cooled and poured into ice water, stirred slowly for 10 minutes, and filtered. After washing the cake with water, the cake was collected and dried to give the objective compound (320 mg, yield 90%).
Step 2 preparation of di-tert-butyl (4- ((1-nitro-3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
1-nitro-3, 6-dichloro-9H-carbazole (100 mg,0.36mmol,1 eq.) was dissolved in N, N-dimethylformamide (5 mL), and potassium carbonate (200 mg,1.42mmol,4 eq.) and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (134 mg,0.36mmol,1.0 eq.) were added and the reaction mixture reacted at room temperature for 16 hours. The reaction solution was extracted three times with ethyl acetate (20 ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=100% -1/1) to give the title compound (70 mg, 34% yield) as a yellow oily liquid LCMS (ESI) [ m+1-56-56] + = 465.0.
Step 3 preparation of di-tert-butyl 4- ((1-amino-3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
Di-tert-butyl (4- ((1-nitro-3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonate (70 mg,0.12mmol,1 eq) was dissolved in tetrahydrofuran (5 mL) and zinc powder (39 mg,0.6mmol,5 eq) and acetic acid (5 mL) were added separately thereto and the reaction mixture stirred at room temperature for 2 hours. The reaction solution was filtered, and the filtrate was concentrated to give the title compound (64 mg, yield 96%). LCMS (ESI) [ m+1-56-56] + = 435.0.
Step 4 preparation of (4- ((1-amino-3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
di-tert-butyl (4- ((1-amino-3, 6-dichloro-9H-carbazol-9-yl) methyl) benzyl) phosphonate (64 mg,0.12mmol,1 eq) was dissolved in DCM (5 mL) and trifluoroacetic acid (0.5 mL) was added and reacted at room temperature for 2 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L aqueous ammonia solution, acetonitrile) to give the title compound (8 mg, yield 15.6%) as a white solid .LCMS(ESI)[M+1]+=435.1;1H NMR(400MHz,DMSO-d6)δ8.18(d,J=2.0Hz,1H),7.68-7.55(m,2H),7.36(dd,J=8.8,2.0Hz,1H),7.04(d,J=7.2Hz,2H),6.90(d,J=7.6Hz,2H),6.81(d,J=2.0Hz,1H),5.75(s,2H),5.30(s,2H),2.72(d,J=21.2Hz,2H).
Example 26 preparation of (3- (3, 6-dichloro-9H-carbazol-9-yl) propyl) phosphonic acid:
Step 1 (preparation of diethyl 3- (3, 6-dichloro-9H-carbazol-9-yl) propyl) phosphonate:
3, 6-dichloro-9H-carbazole (150 mg,0.64mmol,1 eq) was dissolved in N, N-dimethylformamide (4 mL), cooled to 0℃and sodium hydride (60% by mass, 27.9mg,0.7mmol,1.1 eq) was added and stirred at 0℃for 30 minutes, diethyl (3-bromopropyl) phosphonate (181 mg,0.7mmol,1.1 eq) was added dropwise. Stirring was continued at 0℃for 1 hour after the completion of the dropwise addition. Quench with water (2 mL), extract three times with ethyl acetate (5 mL. Times.3) and combine the organic phases. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo. The crude product was purified by flash chromatography (silica gel, methanol: dichloromethane (V/v=60/1)) to give the title compound (240 mg, yield 90.52%) as an off-white solid. LCMS (ESI) [ m+1] + = 414.1.
Step 2 preparation of (3- (3, 6-dichloro-9H-carbazol-9-yl) propyl) phosphonic acid:
Diethyl (3- (3, 6-dichloro-9H-carbazol-9-yl) propyl) phosphonate (240 mg,0.58mmol,1 eq.) was dissolved in acetonitrile (8 mL) and trimethylbromosilane (444 mg,2.9mmol,5.0 eq.) was added and stirred at 50℃for 16H. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (130.0 mg, yield 62.58%) as a white solid .LCMS(ESI)[M+1]+=358.0;1H NMR(400MHz,DMSO-d6)δ8.34(d,J=2.1Hz,2H),7.72(d,J=8.8Hz,2H),7.50(dd,J=8.7,2.1Hz,2H),4.49(t,J=7.0Hz,2H),2.02-1.86(m,2H),1.59-1.42(m,2H).
Example 27 preparation of (4- (3, 6-dichloro-9H-carbazol-9-yl) butyl) phosphonic acid:
Step 1 preparation of diethyl 4- (3, 6-dichloro-9H-carbazol-9-yl) butyl) phosphonate:
3, 6-dichloro-9H-carbazole (50 mg,0.21mmol,1 eq.) was dissolved in N, N-dimethylformamide (2 mL), cooled to 0deg.C, sodium hydride (60% by mass, 9mg,0.23mmol,1.1 eq.) was added, and stirred at 0deg.C for 30 minutes, diethyl (4-bromobutyl) phosphonate (64 mg,0.23mmol,1.1 eq.) was added dropwise. After the completion of the dropwise addition, the mixture was slowly warmed to room temperature and stirred for 1 hour. Quench with water (2 mL), extract three times with ethyl acetate (5 mL. Times.3) and combine the organic phases. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo. The crude product was purified by flash chromatography (silica gel, methanol: dichloromethane (V/v=60/1)) to give the title compound (86.0 mg, yield 94.82%) as a pale yellow oil. LCMS (ESI) [ m+1] + = 428.10.
Step 2 preparation of (4- (3, 6-dichloro-9H-carbazol-9-yl) butyl) phosphonic acid:
Diethyl (4- (3, 6-dichloro-9H-carbazol-9-yl) butyl) phosphonate (80 mg,0.19mmol,1 eq.) was dissolved in acetonitrile (2 mL) and trimethylbromosilane (143 mg,0.93mmol,5 eq.) was added and stirred at 50℃for 16 hours. The reaction solution was concentrated, and the crude product was slurried with acetonitrile, acetonitrile/methanol, suction filtered, and dried to give the objective compound (41.0 mg, yield 58.98%) as a white solid .LCMS(ESI)[M+1]+=372.0;1H NMR(400MHz,DMSO-d6)δ8.33(d,J=2.1Hz,2H),7.69(d,J=8.8Hz,2H),7.49(dd,J=8.7,2.2Hz,2H),4.41(t,J=7.1Hz,2H),1.87-1.76(m,2H),1.58-1.42(m,4H).
EXAMPLE 28 preparation of ((4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) cyclohexyl) methyl) phosphonic acid:
step 1 preparation of diethyl ((4- (bromomethyl) cyclohexyl) methyl) phosphonate:
1, 4-bis (bromomethyl) cyclohexane (600 mg,2.22mmol,1 eq) and triethyl phosphite (184 mg,1.11mmol,0.5 eq) were dissolved in N, N-dimethylformamide (2.5 mL), heated to 150℃and reacted under microwaves for 3 min. Cooled to 0 ℃, quenched with water, the reaction was extracted three times with ethyl acetate (20 ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=100% -1/3) to give the title compound (100.0 mg, 13.75%) as a pale yellow oil LCMS (ESI) [ m+1] + =327.1.
Step 2 preparation of diethyl ((4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) cyclohexyl) methyl) phosphonate:
3, 6-dichloro-9H-carbazole (72.16 mg,0.31mmol,1 eq) was dissolved in N, N-dimethylformamide (2 mL), cooled to 0℃and sodium hydride (60% by mass, 8.07mg,0.34mmol,1.1 eq) was added and stirring continued for 30min, diethyl ((4- (bromomethyl) cyclohexyl) methyl) phosphonate (100 mg,0.31mmol,1 eq) was added and the reaction mixture stirred at room temperature for 16H. The reaction solution was quenched with water, extracted three times with ethyl acetate (20 ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, dichloromethane: methanol (V/v=0% -1/10)) to give the objective compound (35.0 mg, yield 23.41%) as a pale yellow oil. LCMS (ESI) [ m+1] + =482.1.
Step 3 preparation of ((4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) cyclohexyl) methyl) phosphonic acid:
Diethyl ((4- ((3, 6-dichloro-9H-carbazol-9-yl) methyl) cyclohexyl) methyl) phosphonate (35 mg,0.07mmol,1 eq) was dissolved in acetonitrile (2 mL), trimethylbromosilane (53.58 mg,0.35mmol,5 eq) was added and stirred at 50℃for 16 hours and 80℃for a further 16 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L formic acid solution, acetonitrile) to give the title compound (1.9 mg, yield 6.37%) as a white solid .LCMS(ESI)[M+1]+=426.2;1H NMR(400MHz,DMSO-d6)δ8.33(d,J=2.1Hz,2H),7.68(d,J=8.8Hz,2H),7.48(dd,J=8.8,2.2Hz,2H),4.24(d,J=7.3Hz,2H),3.36(d,J=6.1Hz,2H),1.95-1.64(m,3H),1.60-1.44(m,3H),1.22-1.05(m,2H),0.89(qd,J=11.1,9.4,2.5Hz,2H).
EXAMPLE 29 preparation of 3- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 (3-methylbenzyl) preparation of di-tert-butyl phosphonate:
Di-tert-butyl phosphonite (4.6 g,23.8mmol,1.1 eq.) is dissolved in tetrahydrofuran (40 mL), sodium hydride (60% mass, 951mg,23.8mmol,1.1 eq.) is added at 0deg.C and the reaction stirred for half an hour, 1-bromomethyl-3-methylbenzene (4.0 g,21.6mmol,1.0 eq.) is added and the reaction is allowed to warm to 70deg.C overnight. LCMS monitored the end of the reaction, quenched by the addition of water (30 mL) to the reaction solution and extracted three times with ethyl acetate (30 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and dried over silica gel. Purification by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) afforded the title compound (4.0 g, yield: 62.0%) as a brown solid .1H NMR(400MHz,DMSO-d6)δ7.16(t,J=7.6Hz,1H),7.06-7.00(m,3H),2.95(d,J=21.2Hz,2H),2.27(s,3H),1.36(s,18H).
Step 2 preparation of di-tert-butyl (3-bromomethyl) benzyl) phosphonate:
Di-tert-butyl (3-methylbenzyl) phosphonate (2.0 g,6.70mmol,1.0 eq.) was dissolved in carbon tetrachloride (20 mL), and N-bromosuccinimide (1.3 g,7.37mmol,1.1 eq.) and dibenzoyl peroxide (BPO) (80 mg,0.340mmol,0.05 eq.) were added to the mixture and reacted at 78℃for 3 hours with stirring. LCMS monitored the end of the reaction, water (50 mL) was added to the reaction and extracted three times with ethyl acetate (50 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (1.0 g, yield: 39.5%) as a brown oily liquid .1H NMR(400MHz,DMSO-d6)δ7.42-7.27(m,3H),7.20-7.16(m,1H),4.68(s,2H),3.01(d,J=21.2Hz,2H),1.37(s,18H).
Step 3 preparation of di-tert-butyl (3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
3, 6-dimethoxy-9H-carbazole (181 mg,0.800mmol,1.5 eq) was dissolved in tetrahydrofuran (2 mL), and sodium hydride (60% by mass, 32mg,0.800mmol,1.5 eq) was added at 0 ℃. Di-tert-butyl (3-bromomethyl) benzyl) phosphonate (200 mg,0.530mmol,1.0 eq.) was added and allowed to stir at room temperature for 2 hours. Water (10 mL) was added to the reaction solution to quench, and extracted three times with ethyl acetate (200 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and dried over silica gel. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) afforded the title compound (200 mg, yield: 72.0%) as a yellow solid. LCMS (ESI) [ M-2 tBu+3H]+ = 412.2.
Step 4 preparation of 3- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phosphonate (180 mg,0.340mmol,1.0 eq) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added to the mixture and stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to obtain a brown oily crude product. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (86.05 mg, yield: 60.8%) as a white solid .LCMS(ESI):[M+H]+=412.2;1H NMR(400MHz,DMSO-d6)δ7.72(d,J=2.8Hz,2H),7.39(d,J=8.8Hz,2H),7.22(s,1H),7.08(d,J=7.2Hz,1H),7.01-6.97(m,3H),6.56(d,J=7.9Hz,1H),5.44(s,2H),3.83(s,6H),2.66(d,J=20.0Hz,2H).
EXAMPLE 30 preparation of 4- (3, 6-dimethoxy-9H-amino-oxazol-9-yl) benzyl) phosphonic acid:
Step 1 (preparation of ethyl 4- (3, 6-dimethoxy-9H-carbazol-9-yl) benzyl) phosphonate:
3, 6-dimethoxy-9H-carbazole (200 mg,0.88mmol,1.0 eq) and diethyl (4-iodobenzyl) phosphonate (268 mg,1.3mmol,1.5 eq) were dissolved in dimethyl sulfoxide (3 mL), and 1, 10-phenanthroline (16 mg,0.09mmol,0.1 eq), cesium carbonate (514 mg,1.8mmol,2.0 eq) and cuprous iodide (17 mg,0.09mmol,0.1 eq) were added and the reaction was continued under 150℃inert gas for 16 hours. The reaction solution was concentrated under reduced pressure to give a crude product as a brown oil. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (120 mg, yield: 32.1%) as a white solid. LCMS (ESI) [ m+2mecn+h ] + = 408.3.
Step 2 preparation of 4- (3, 6-dimethoxy-9H-amino-oxazol-9-yl) benzyl) phosphonic acid:
Ethyl (4- (3, 6-dimethoxy-9H-carbazol-9-yl) benzyl) phosphonate (120 mg,0.28mmol,1.0 eq) was dissolved in acetonitrile (2 mL), and to this mixture was added trimethylbromosilane (648 mg,4.2mmol,15 eq) and reacted for 1 hour with stirring at 50 ℃. The reaction mixture was quenched with methanol and concentrated under reduced pressure to give crude product as brown oil. Preparation by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (76.54 mg, yield: 68.3%) as a white solid .LCMS(ESI):[M+H]+=398.0;1H NMR(400MHz,DMSO-d6)δ7.77(d,J=2.4Hz,2H),7.46(d,J=8.0Hz,2H),7.37(d,J=8.0Hz,2H),7.16(d,J=8.8Hz,2H),6.93(dd,J=8.8,2.4Hz,2H),3.85(s,6H),2.87(d,J=20.0Hz,2H).
Example 31 preparation of 4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenylboronic acid:
Step 1 preparation of 9- (4-bromobenzyl) -3, 6-dimethoxy-9H-carbazole:
3, 6-dimethoxy-9H-carbazole (500 mg,2.2mmol,1.0 eq) was added to N, N-dimethylformamide (6 mL), sodium hydride (60% by mass 132mg,5.5mmol,2.5 eq) was slowly added at 0deg.C, and the reaction was continued for 30min, 4-bromobenzyl bromide (1.1 g,4.4mmol,2.0 eq) was added, and the reaction was continued at room temperature for 3H. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) afforded the title compound (760 mg, yield: 87.2%) as a white solid. LCMS (ESI) [ m+h ] + = 395.1.
Step 2 preparation of 3, 6-dimethoxy-9- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzyl) -9H-carbazole:
9- (4-bromobenzyl) -3, 6-dimethoxy-9H-carbazole (200 mg,0.5mmol,1.0 eq.) was dissolved in dioxane (2 mL), and pinacol ester of bisboronic acid (192 mg,0.8mmol,1.5 eq.), potassium acetate (149 mg,1.5mmol,3.0 eq.) and 1,1' -bis (diphenylphosphine) ferrocene-palladium (II) dichloride dichloromethane complex (39 mg,0.1mmol,0.1 eq.) were added and reacted for 2 hours with stirring at 80 ℃. The reaction mixture was quenched with water (15 mL), extracted three times with ethyl acetate (25 mL. Times.3), the organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) afforded the title compound (200 mg, yield: 89.3%) as a white solid. LCMS (ESI) [ m+h ] + = 443.3.
Step 3 preparation of 4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenylboronic acid:
3, 6-dimethoxy-9- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzyl) -9H-carbazole (90 mg,0.2mmol,1.0 eq) was dissolved in water and tetrahydrofuran (1 mL), sodium periodate (129 mg,0.6mmol,3.0 eq) was added, and the reaction was stirred at room temperature for 1 hour, and then aqueous hydrochloric acid (1M, 0.4mL,0.4mmol,2.0 eq) was added to the reaction solution. The reaction was monitored by LCMS for end, quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL x 3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the crude product. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (46.1 mg, yield: 63.8%) as a pink solid .LCMS(ESI):[M+H]+=362.2;1HNMR(400MHz,DMSO-d6)δ7.93(s,2H),7.73(d,J=8.4Hz,2H),7.64(d,J=8.0Hz,2H),7.44(d,J=8.8Hz,2H),7.11-6.98(m,4H),5.56(s,2H),3.83(s,6H).
Example 32 preparation of (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenethyl) phosphonic acid:
Step 1 (E) - (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) styryl) phosphonic acid diethyl ester preparation:
9- (4-bromobenzyl) -3, 6-dimethoxy-9H-carbazole (350 mg,0.88mmol,1.0 eq) was dissolved in N, N-dimethylformamide (5 mL), and diethyl vinylphosphonate (725 mg,4.4mmol,5.0 eq), potassium carbonate (244 mg,1.7mmol,2.0 eq), palladium acetate (20 mg,0.09mmol,0.1 eq) and triphenylphosphine (93 mg,0.35mmol,0.4 eq) were added and the reaction was continued under 100℃inert gas for 16H. The reaction was monitored by LCMS, saturated aqueous sodium chloride (30 mL) was added to the reaction solution, and extracted three times with ethyl acetate (30 ml×3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and dried over silica gel. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) afforded the title compound (120 mg, yield: 28.4%) as a yellow oily liquid. LCMS (ESI) [ m+h+mecn ] + = 521.4.
Step 2 preparation of diethyl (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenethyl) phosphonate:
Diethyl (E) - (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) styryl) phosphonate (120 mg,0.25mmol,1.0 eq) was dissolved in methanol (2 mL), then palladium on carbon (50 mg) was added to the mixture, and after three replacements with hydrogen gas, the reaction was stirred at room temperature for 2 hours. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure and dried by spin-drying to give the title compound (100 mg, yield: 83.3%) as a colorless oil. LCMS (ESI) [ m+h ] + = 481.7.
Step 3 preparation of (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenethyl) phosphonic acid:
Diethyl (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenethyl) phosphonate (100 mg,0.21mmol,1.0 eq) was dissolved in acetonitrile (2 mL), trimethylbromosilane (477 mg,3.1mmol,15 eq) was added and the reaction stirred at 50℃for 16 hours. The reaction mixture was quenched with methanol and concentrated under reduced pressure to give crude product as brown oil. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (47.5 mg, yield: 53.9%) as a white solid .LCMS(ESI):[M+H]+=426.2;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.4Hz,2H),7.45(d,J=8.8Hz,2H),7.06-6.97(m,6H),5.49(s,2H),3.84(s,6H),2.67-2.61(m,2H),1.68-1.54(m,2H).
Example 33 preparation of 4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) phosphonic acid:
step 1, preparing diethyl p-tolylphosphonate:
1-iodo-4-methylbenzene (5.0 g,22.9mmol,1.0 eq.) and tetrakis (triphenylphosphine) palladium (270 mg,2.29mmol,0.1 eq.) were dissolved in toluene (50 mL), and triethyl phosphite (5.7 g,34.4mmol,1.5 eq.) and triethylamine (7.0 g,68.8mmol,3.0 eq.) were added and the reaction stirred at 85℃for 16 hours. Water (50 mL) was added to the reaction solution, and ethyl acetate (50 mL. Times.3) was extracted three times. The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and dried over silica gel. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) afforded the title compound (1.5 g, yield: 28.7%) as a brown solid. LCMS (ESI) [ m+h ] + = 229.1.
Step 2 preparation of diethyl (4- (dibromomethyl) phenyl) phosphonate
Diethyl p-tolylphosphonate (1.5 g,6.57mmol,1.0 eq.) was dissolved in carbon tetrachloride (20 mL), N-bromosuccinimide (2.3 g,13.1mmol,2.0 eq.) and dibenzoyl peroxide (80 mg,0.329mmol,0.05 eq.) were added and the reaction stirred at 78℃for 3 hours. Water (50 mL) was added to the reaction solution, and extracted three times with ethyl acetate (50 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. Reversed phase HPLC preparation (C18, 10mmol/L formic acid, acetonitrile) afforded the title compound (270 mg, yield: 10.6%) as a brown oily liquid. LCMS (ESI) [ m+h ] + = 386.9.
Step 3 preparation of diethyl ((4-bromomethyl) phenyl) phosphonate:
Diethyl (4- (dibromomethyl) phenyl) phosphonate (270 mg,0.699mmol,1.0 eq) was dissolved in acetonitrile (3 mL), diethyl phosphinate (97 mg,0.699mmol,1.0 eq) and N, N-diisopropylethylamine (181 mg,1.40mmol,2.0 eq) were added in this order and the reaction stirred at 0 ℃ for 2 hours. Aqueous sodium bicarbonate (10 mL) was added to the reaction solution, quenched, and extracted three times with ethyl acetate (10 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) to give the title compound (170 mg, yield: 79.1%) as a colorless oily liquid. LCMS (ESI) [ m+h ] + =307.0.
Step 4 preparation of diethyl 4- ((3, 6-dimethoxy-9H-carbazole-9-methyl) phenyl) phosphonate:
3, 6-dimethoxy-9H-carbazole (126 mg,0.553mmol,1.0 eq) was dissolved in tetrahydrofuran (2 mL), sodium hydride (mass fraction 60%,44mg,1.11mmol,2.0 eq) was added at 0℃and reacted for half an hour with stirring, diethyl ((4-bromomethyl) phenyl) phosphonate (170 mg,0.553mmol,1.0 eq) was added, and the mixture was stirred at room temperature for 2 hours. Aqueous ammonium chloride (10 mL) was added to the reaction mixture to quench, and ethyl acetate (20 mL. Times.3) was extracted three times. The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) afforded the title compound (100 mg, yield: 39.8%) as a colorless oily liquid. LCMS (ESI) [ m+h ] + =454.2.
Step 5 preparation of 4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) phosphonic acid:
Diethyl (4- ((3, 6-dimethoxy-9H-carbazole-9-methyl) phenyl) phosphonate (100 mg,0.220mmol,1.0 eq) was dissolved in acetonitrile (1 mL), then trimethylbromosilane (0.51 mL,3.31mmol,15 eq) was added to this mixture and reacted for 1 hour under stirring at 50 ℃ C., the reaction was quenched with methanol, concentrated under reduced pressure to give crude product as a brown oil which was prepared (C18, 10mmol/L ammonia, acetonitrile) by reverse phase HPLC to give the title compound (45.68 mg, yield: 52.3%) as a white solid .LCMS(ESI):[M+H]+=398.0;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.4Hz,2H),7.46-7.42(m,4H),7.03-6.94(m,4H),5.51(s,2H),3.84(s,6H).
EXAMPLE 34 preparation of 2- (4- (3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) acetic acid:
Step 1 preparation of methyl 2- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) acetate:
9- (4-bromobenzyl) -3, 6-dimethoxy-9H-carbazole (500 mg,1.26mmol,1.0 eq) was dissolved in ultra-dry toluene (5 mL), to which were then added methyl acetoacetate (220 mg,1.9mmol,1.5 eq), potassium phosphate (803 mg,3.8mmol,3.0 eq) and palladium acetate (28.4 mg,0.13mmol,0.1 eq) and finally t-Buxphos (2-di-tert-butyl-2 ',4',6' -triisopropylbiphenyl) (107 mg,0.25mmol,0.2 eq) were added and the reaction was stirred at 120℃for 16 hours. The reaction solution was concentrated, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=7/3)) to give the title compound (480 mg, yield: 97.8%) as a yellow solid. LCMS (ESI) [ m+h ] + =390.3.
Step 2 preparation of 2- (4- (3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) acetic acid:
Methyl 2- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) acetate (250 mg,0.64mmol,1.0 eq.) was dissolved in water and methanol (1:1, 3 mL), and lithium hydroxide (46 mg,1.9mmol,3.0 eq.) was added to the mixture, and the reaction was stirred at room temperature for 2 hours. The reaction was monitored by LCMS for end, and the reaction was concentrated under reduced pressure. Purification by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (160 mg, yield: 66.6%) as a white solid .LCMS(ESI):[M+H]+=376.2;1H NMR(400MHz,CD3OD)δ7.61(d,J=2.4Hz,2H),7.30(d,J=8.8Hz,2H),7.17(d,J=8.0Hz,2H),7.06-6.95(m,4H),5.47(s,2H),3.89(s,6H),3.45(s,2H).
Example 35 preparation of (2- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) ethylboronic acid
Step 1 preparation of 9- ((4-vinylphenyl) methyl) -3, 6-dimethoxy-9H-carbazole:
3, 6-dimethoxy-9H-carbazole (100 mg,0.44mmol,1.0 eq.) was dissolved in N, N-dimethylformamide (2 mL), sodium hydride (35 mg,0.88mmol,2.0 eq.) was added, and the reaction solution was reacted at 0℃for 0.5 hours. 4-chloromethylstyrene (153 mg,0.88mmol,2.0 eq.) was added and the reaction was continued at room temperature for 2 hours. The reaction solution was quenched with water (50 mL) and then extracted three times with ethyl acetate (50 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) to give the title compound (120 mg, yield: 79.4%). LCMS (ESI) [ m+h ] + = 344.2.
Step 2 preparation of 3, 6-dimethoxy-9- (4- (2- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) ethyl) phenyl) methyl) -9H-carbazole:
9- ((4-vinylphenyl) methyl) -3, 6-dimethoxy-9H-carbazole (120 mg,0.35mmol,1.0 eq.) was dissolved in chloroform (2 mL), and then pinacol borane (90 mg,0.7mmol,2.0 eq.) and calcium chloride (117 mg,1.1mmol,3.0 eq.) were added to this mixture. The reaction solution was stirred at 110 ℃ for 16 hours. The reaction solution was filtered, a saturated aqueous sodium chloride solution (50 mL) was added to the filtrate, and extracted three times with ethyl acetate (50 ml×3). The organic phase was collected, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) to give the title compound (80 mg, yield: 48.7%). LCMS (ESI) [ m+h ] + =472.3.
Step 3 preparation of (2- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) ethylboronic acid:
3, 6-dimethoxy-9- (4- (2- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) ethyl) phenyl) methyl) -9H-carbazole (80 mg,0.17mmol,1.0 eq) was dissolved in tetrahydrofuran/H 2 O (V/v=5/1, 2 ml), then lithium hydroxide (8 mg,0.34mmol,2 eq) was added to the mixture and the reaction stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure to obtain a crude white solid. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (37.42 mg, yield) :56.7%).LCMS(ESI):[M+H]+=390.2;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.4Hz,2H),7.52-7.40(m,4H),7.11-6.94(m,6H),5.50(s,2H),3.85(s,6H),2.55-2.51(m,2H),0.82(t,J=8.4Hz,2H).
EXAMPLE 36 preparation of 3- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) propanoic acid
Step 1 preparation of (E) -methyl 3- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenylacrylate:
9- ((4-bromophenyl) methyl) -3, 6-dimethoxy-9H-carbazole (350 mg,0.88mmol,1.0 eq) was dissolved in N, N-dimethylformamide (5 mL), and methyl acrylate (380 mg,4.4mmol,5.0 eq), potassium carbonate (244 mg,1.7mmol,2.0 eq), palladium acetate (20 mg,0.09mmol,0.1 eq) and triphenylphosphine (93 mg,0.35mmol,0.4 eq) were added and the reaction was continued under 100℃argon atmosphere for 16 hours. Saturated aqueous sodium chloride (30 mL) was added to the reaction solution, and extracted three times with ethyl acetate (30 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) to give the title compound (120 mg, yield: 33.9%) as a yellow oily liquid. LCMS (ESI) [ m+h ] + =402.1.
Step 2 preparation of methyl 3- ((3, 6-dimethoxy-9H-carbazole-9-methyl) phenyl) propanoate:
Methyl (E) -3- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenylacrylate (120 mg,0.30mmol,1.0 eq.) was dissolved in methanol (2 mL), then palladium on carbon (50 mg) was added to the mixture, and the reaction was stirred at room temperature for 2 hours after three times of hydrogen substitution.
Step 3 preparation of 3- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) phenyl) propanoic acid:
Methyl 3- ((3, 6-dimethoxy-9H-carbazole-9-methyl) phenyl) propionate (100 mg,0.25mmol,1.0 eq.) was dissolved in tetrahydrofuran/H 2 O (V/v=5/1, 2 ml), then lithium hydroxide (32 mg,0.75mmol,3 eq.) was added to this mixture and the reaction stirred at room temperature for 16 hours. The reaction solution was neutralized with a dilute hydrochloric acid solution, concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (10.69 mg, yield: 11.1%) as a white solid .LCMS(ESI):[M+H]+=390.2;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=1.2Hz,2H),7.47(d,J=8.8Hz,2H),7.09(d,J=8.0Hz,2H),7.06-6.96(m,4H),5.51(s,2H),3.85(s,6H),2.70(t,J=7.6Hz,2H),2.36(t,J=7.6Hz,2H).
EXAMPLE 37 preparation of (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) boronic acid
Step 1 preparation of 9- (4- (bromomethyl) benzyl) -3, 6-dimethoxy-9H-carbazole:
3, 6-dimethoxy-9H-carbazole (200 mg,2.2mmol,1.0 eq) was added to N, N-dimethylformamide (6 mL), sodium hydride (220 mg,5.5mmol,2.5 eq) was slowly added at 0deg.C and reacted for 30 minutes with stirring, then 1, 4-bis (bromomethyl) benzene (460 mg,1.76mmol,2.0 eq) was added, and the reaction was continued at room temperature for 3 hours. The reaction was quenched by the addition of water (15 mL) and extracted three times with ethyl acetate (25 mLx 3). The organic phase was collected, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=4/1)) to give the title compound as a white solid.
Step 2 preparation of 3, 6-dimethoxy-9- (4- ((4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) methyl) benzyl) -9H-carbazole:
9- (4- (bromomethyl) benzyl) -3, 6-dimethoxy-9H-carbazole (150 mg,0.37mmol,1.0 eq.) was dissolved in N, N-dimethylformamide (3 mL), to which was then added pinacol biborate (186 mg,1.7mmol,2.0 eq.), cuprous iodide (7 mg,0.04mmol,0.1 eq.) and lithium methoxide (42 mg,1.1mmol,3.0 eq.) and finally triphenylphosphine (12 mg,0.04mmol,0.12 eq.) were added and stirred at 35℃for 18 hours. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) to give the title compound (130 mg, yield: 79.1%) as a white solid.
Step 3 preparation of (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) boronic acid:
3, 6-dimethoxy-9- (4- ((4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) methyl) benzyl) -9H-carbazole (120 mg,0.26mmol,1.0 eq) was dissolved in tetrahydrofuran (2 mL), and lithium hydroxide (19 mg,0.78mmol,3.0 eq) was added to the mixture, and the reaction was stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (23.06 mg, yield: 23.6%) as a white solid .LCMS(ESI):[M+H]+=376.2;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.0Hz,2H),7.59(s,2H),7.47(d,J=8.8Hz,2H),7.03-6.93(m,6H),5.48(s,2H),3.84(s,6H),1.99(s,2H).
EXAMPLE 38 preparation of (6- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-3-yl) methyl) phosphonic acid
Step 1 preparation of 5- ((diethoxyphosphonyl) methyl) picolinate:
Methyl 5-bromomethylpyridine-2-carboxylate (3.5 g,15.2mmol,1.0 eq) was dissolved in 1, 4-dichloro hexacyclic ring (35 mL), and triethyl phosphite (2.5 g,15.2mmol,1.0 eq) was added to the mixture and stirred at 80 ℃ for 2 hours. The reaction solution was concentrated under reduced pressure and dried by spin. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=20/1)) to give the title compound (3.0 g, yield: 68.6%) as a yellow oil. LCMS (ESI) [ m+h ] + = 288.1.
Step 2 preparation of diethyl 6- (hydroxymethyl) pyridin-3-yl) methyl phosphonate:
5- ((diethoxyphosphonyl) methyl) picolinate (2.7 g,9.40mmol,1.0 eq.) was added to tetrahydrofuran (27 mL) at 0deg.C, lithium borohydride (204 mg,9.40mmol,1.0 eq.) was added slowly and the reaction was continued at room temperature for three hours. Water (50 mL) was added to the reaction solution, and extracted three times with ethyl acetate (50 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (1.5 g, yield: 61.6%) as a yellow oil. LCMS (ESI) [ m+h ] + =260.1.
Step 3 preparation of diethyl 6- (chloromethyl) pyridin-3-ylmethyl) phosphonate:
Diethyl (6- (hydroxymethyl) pyridin-3-yl) methyl) phosphonate (1.3 g,5.01mmol,1.0 eq) was added to dichloromethane (13 mL) at 0 ℃, thionyl chloride (894 mg,7.51mmol,1.5 eq) was slowly added and the reaction was continued at room temperature for one hour. The reaction solution was concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (1.0 g, yield: 71.9%) as a yellow oil. LCMS (ESI) [ m+h ] + =278.1.
Step 4 preparation of diethyl (6- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-3-yl) methyl) phosphonate:
3, 6-dimethoxy-9H-carbazole (200 mg,0.88mmol,1.0 eq) was added to N, N-dimethylformamide (2 mL), the reaction mixture was cooled to zero degree, sodium hydride (53 mg,1.32mmol,1.5 eq) was slowly added, and after half an hour of reaction, diethyl (6- (chloromethyl) pyridin-3-ylmethyl) phosphonate (367 mg,1.32mmol,0.1 eq) was added and the reaction mixture was continued at room temperature for one hour. LCMS monitored the completion of the reaction, water (50 mL) was added to the reaction solution and extracted three times with ethyl acetate (50 ml×3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/2)) to give the title compound (150 mg, yield: 36.5%) as a yellow solid. LCMS (ESI) [ m+h ] + = 469.1.
Step 5 preparation of (6- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
Diethyl (6- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-3-yl) methyl) phosphonate (100 mg,0.21mmol,1.0 eq) was dissolved in dichloromethane (2 mL) and then trimethylbromosilane (490 mg,3.15mmol,15 eq) was added to this mixture and the reaction stirred at 50 ℃ for 0.5 hours. The reaction solution was filtered, quenched with methanol, and concentrated under reduced pressure. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (27.82 mg, yield: 31.6%) as a white solid .LCMS(ESI):[M+H]+=413.2;1H NMR(400MHz,DMSO-d6)δ8.27(s,1H),7.72(d,J=2.4Hz,2H),7.43-7.38(m,3H),6.99(dd,J=8.8,2.4Hz,2H),6.65(d,J=7.6Hz,1H),5.52(s,2H),3.84(s,6H),2.60(d,J=20.4Hz,2H).
EXAMPLE 39 preparation of ((5- (3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-2-yl) methyl) phosphonic acid:
Step 1 preparation of methyl 6- (chloromethyl) nicotinate:
Methyl 6- (hydroxymethyl) nicotinate (4.0 g,23.9mmol,1.0 eq) was added to dichloromethane (40 mL), the reaction solution was cooled to 0 ℃, thionyl chloride (4.27 g,35.9mmol,1.5 eq) was slowly added and the reaction was continued at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure and dried by spin. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=20/1)) to give the title compound (3.5 g, yield: 77.7%) as a yellow oil. LCMS (ESI) [ m+h ] + = 186.0.
Step 2 preparation of 6- ((diethoxyphosphonic) methyl) nicotinic acid methyl ester:
diethyl phosphonite (2.6 g,18.9mmol,1.0 eq) was dissolved in tetrahydrofuran (35 mL), the reaction mixture was cooled to 0 ℃, sodium hydride (1.14 g,28.4mmol,1.5 eq) was slowly added, after half an hour of reaction, methyl 6- (chloromethyl) nicotinate (3.5 g,18.9mmol,1.5 eq) was added and stirring was continued at room temperature for two hours. The reaction solution was concentrated under reduced pressure and dried by spin-drying, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/3)) to give the title compound (3.0 g, yield: 55.3%) as a yellow oil. LCMS (ESI) [ m+h ] + = 288.1.
Step 3 preparation of diethyl 5- (hydroxymethyl) pyridin-2-yl) methyl phosphonate:
Methyl 6- ((diethoxyphosphonate) methyl) nicotinate (800 mg,2.79mmol,1.0 eq.) was added to tetrahydrofuran (8 mL), the reaction solution was cooled to zero, lithium aluminum hydride (127 mg,3.35mmol,1.2 eq.) was slowly added and the reaction was continued at room temperature for two hours. Water (50 mL) was added to the reaction solution, and extracted three times with ethyl acetate (50 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (180 mg, yield: 24.5%) as a yellow oil. LCMS (ESI) [ m+h ] + =260.1.
Step 4 preparation of diethyl 5- (chloromethyl) pyridin-2-yl) methyl phosphonate:
Diethyl (6- (hydroxymethyl) pyridin-3-yl) methyl) phosphonate (150 mg,0.58mmol,1.0 eq) was added to dichloromethane (13 mL) and thionyl chloride (103 mg,0.87mmol,1.5 eq) was slowly added and the reaction was continued at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure and dried by spin. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (110 mg, yield: 68.5%) as a yellow oil. LCMS (ESI) [ m+h ] + =278.1.
Step 5 preparation of diethyl (5- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-2-yl) methyl) phosphonate:
3, 6-dimethoxy-9H-carbazole (90 mg,0.39mmol,1.0 eq) was added to N, N-dimethylformamide (2 mL), the reaction mixture was cooled to 0℃and sodium hydride (24 mg,0.59mmol,1.5 eq) was slowly added, after half an hour of reaction, diethyl (5- (chloromethyl) pyridin-2-yl) methyl) phosphonate (110 mg,0.39mmol,1.0 eq) was added and the reaction mixture was continued at room temperature for 1 hour. Water (10 mL) was added to the reaction solution, and extracted three times with ethyl acetate (10 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/2)) to give the title compound (80 mg, yield: 43.1%) as a yellow solid. LCMS (ESI) [ m+h ] + = 469.2.
Step 6 preparation of ((5- (3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-2-yl) methyl) phosphonic acid:
diethyl (5- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) pyridin-2-yl) methyl) phosphonate (80 mg,0.17mmol,1.0 eq) was dissolved in dichloromethane (1 mL), and then trimethylbromosilane (390 mg,2.55mmol,15 eq) was added to this mixture and reacted for 0.5 hours with stirring at 50 ℃. The reaction solution was filtered, quenched with methanol, and concentrated under reduced pressure. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the product (39.15 mg, yield: 55.8%) as an off-white solid .LCMS(ESI):[M+H]+=413.2;1H NMR(400MHz,DMSO-d6)δ8.22(s,1H),7.72(d,J=2.4Hz,2H),7.50(d,J=9.2Hz,2H),7.29(dd,J=8.0,2.0Hz,1H),7.19(d,J=8.0Hz,1H),7.02(dd,J=8.8,2.4Hz,2H),5.51(s,2H),3.84(s,6H),2.85(d,J=20.8Hz,2H).
EXAMPLE 40 preparation of((4- ((3-ethyl-9H-carbazol-9-yl) methyl) phenyl) methyl) phosphonic acid:
step 1 preparation of di-tert-butyl ((4- ((3-bromo-9H-carbazol-9-yl) methyl) phenyl) methyl) phosphonate:
3-bromocarbazole (300 mg,1.22mmol,1 eq.) was dissolved in tetrahydrofuran (25 mL), then sodium hydride (244 mg,6.1mmol,5 eq.) was added, and di-tert-butyl ((4- (bromomethyl) phenyl) methyl) phosphonate (1.38 g,3.66mmol,3 eq.) was added to the mixture and the reaction was stirred at room temperature under argon for 1 hour. The reaction mixture was quenched with methanol (5 mL), diluted with saturated ammonium chloride solution (5 mL), extracted three times with ethyl acetate (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (400 mg, yield: 60.5%). LCMS (ESI) [ M-2tbu+3h ] + =430.0.
Step 2 preparation of di-tert-butyl ((4- ((3-vinyl-9H-carbazol-9-yl) methyl) phenyl) methyl) phosphonate:
Di-tert-butyl ((4- ((3-bromo-9H-carbazol-9-yl) methyl) phenyl) phosphonate (200 mg,0.37mmol,1 eq) was dissolved in1, 4-dioxane (5 mL) and water (1 mL), potassium ethylene trifluoroborate (148 mg,1.11mmol,3 eq), potassium carbonate (153 mg,1.11mmol,3 eq) and Pd (dppf) Cl 2 ((1, 1' -bis (diphenylphosphino) ferrocene) palladium dichloride) (54 mg,0.07mmol,0.2 eq) were added and reacted at 110℃for 1 hour. The reaction solution was diluted with ethyl acetate (20 mL), washed with saturated ammonium chloride (30 mL. Times.3), and the organic phase was dried, filtered, and concentrated under reduced pressure to give the title compound (150 mg, yield: 83.1%). LCMS (ESI) [ M-2tbu+3h ] + = 378.0.
Step 3 preparation of di-tert-butyl ((4- ((3-ethyl-9H-carbazol-9-yl) methyl) phenyl) methyl) phosphonate:
Di-tert-butyl ((4- ((3-vinyl-9H-carbazol-9-yl) methyl) phenyl) phosphonate (70 mg,0.14mmol,1 eq.) was dissolved in anhydrous methanol (5 mL), palladium on carbon (30 mg,43 wt%) was added and reacted at room temperature under hydrogen atmosphere for 1 hour. The reaction solution was filtered and concentrated to give the title compound (70 mg, yield: 99.6%). LCMS (ESI) [ M-2tbu+3h ] + = 380.2.
Step 4 preparation of ((4- ((3-ethyl-9H-carbazol-9-yl) methyl) phenyl) methyl) phosphonic acid:
Di-tert-butyl ((4- ((3-ethyl-9H-carbazol-9-yl) methyl) phenyl) methyl) phosphonate (70 mg,0.14mmol,1 eq.) was dissolved in dichloromethane (5 ml) and trifluoroacetic acid (1 mL,8.77mmol,62.6 eq.) was added and the reaction stirred at room temperature for 1 hour. The reaction solution was quenched with water, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (2.58 mg, yield) :7.1%).LCMS(ESI):[M+H]+=380.1;1H NMR(400MHz,CD3OD)δ8.05(d,J=8.0Hz,1H),7.89(s,1H),7.42(d,J=8.0Hz,1H),7.37-7.33(m,2H),7.28-7.23(m,3H),7.17-7.13(m,1H),7.02(d,J=8.0Hz,2H),5.67-5.63(m,2H),2.82-2.77(m,4H),1.32(t,J=7.6Hz,3H).
Example 41 preparation of (4- ((3-isopropyl-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 preparation of (4- ((3- (propyl-1-en-2-yl) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid diethyl ester:
The pinacol isopropenylborate (173 mg,1.03mmol,2 eq.) and diethyl (4- ((3-bromo-9H-carbazol-9-yl) methyl) benzyl) phosphonate (250 mg,0.51mmol,1 eq.) were dissolved in water (1 mL) and 1, 4-dioxane (5 mL), pd (dppf) Cl 2 ([ 1,1' -bis (diphenylphosphine) ferrocene ] palladium dichloride) (75 mg,0.1mmol,0.2 eq.) and potassium carbonate (213 mg,1.54mmol,3 eq.) were added and the reaction was stirred at 100℃for 1 hour, the reaction was diluted with saturated ammonium chloride (10 mL), ethyl acetate was extracted (30 mL. Times.3), the organic phase was collected, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound (280 mg, yield: 82.9%). LCMS (ESI) [ m+h ] + = 448.1.
Step 2 preparation of diethyl (4- ((3-isopropyl-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((3- (propyl-1-en-2-yl) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (50 mg,0.11mmol,1 eq) was dissolved in methanol (5 mL), then palladium on carbon (25 mg,50 wt%) was added to the mixture and the reaction was stirred at room temperature under hydrogen atmosphere for 1 hour. The reaction solution was filtered, and concentrated under reduced pressure to give the title compound (40 mg, yield: 80.1%). LCMS (ESI) [ m+na ] + = 472.1.
Step 3 preparation of (4- ((3-isopropyl-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((3-isopropyl-9H-carbazol-9-yl) methyl) benzyl) phosphonate (50 mg,0.11mmol,1 eq) was dissolved in dichloromethane (3 mL), and trimethylbromosilane (0.17 mL,1.1mmol,10 eq) was then added to this mixture and the reaction stirred at 50 ℃ for 0.5 hours. The reaction solution was filtered, quenched with methanol, concentrated under reduced pressure, and the crude product was purified by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (6.05 mg, yield: 14.0%) as a white solid .LCMS(ESI):[M-H]-=392.1;1H NMR(400MHz,CD3OD)δ8.07(d,J=7.6Hz,1H),7.93(d,J=1.2Hz,1H),7.43(d,J=8.4Hz,1H),7.38-7.35(m,2H),7.31-7.26(m,3H),7.17(t,J=6.8Hz,1H),7.01(d,J=8.0Hz,2H),5.50(s,2H),3.11-3.04(m,1H),2.81(d,J=19.6Hz,2H),1.34(d,J=6.8Hz,6H).
Example 42 preparation of (4- ((2- (methoxy-d 3) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 preparation of 2- (methoxy-d 3) -9H-carbazole:
Sodium (420 mg,18.3mmol,10 eq.) is dissolved in 2mL deuterated methanol and stirred at room temperature for 0.5H, then a solution of 2-bromo-9H-carbazole (450 mg,1.83mmol,1 eq.) and cuprous iodide (100 mg,0.53mmol,0.29 eq.) in N, N-dimethylformamide (3 mL) is added, the reaction is stirred under argon for 12H at 120℃and quenched with saturated ammonium chloride, extracted three times with ethyl acetate (10 mL. Times.3) and the organic phases are combined. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (257 mg, yield: 70.19%) as a yellow solid. LCMS (ESI) [ m+h ] + =201.1.
Step 2 preparation of diethyl (4- ((2- (methoxy-d 3) -9H-carbazol-9-yl) methyl) benzyl) phosphonate:
2- (methoxy-d 3) -9H-carbazole (255 mg,1.28mmol,1 eq) was dissolved in tetrahydrofuran (10 mL), and diethyl (4- (bromomethyl) benzyl) phosphonate (82 mg,2.56mmol,2 eq) and sodium hydride (256 mg,6.4mmol,5 eq) were added. After stirring at room temperature for 1 hour, the reaction solution was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (10 ml×3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (137 mg, yield: 24.3%) as a yellow oil. LCMS (ESI) [ m+h ] + = 441.2.
Step 3 preparation of (4- ((2- (methoxy-d 3) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((2- (methoxy-d 3) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (137 mg,0.31mmol,1 eq) was dissolved in dichloromethane (5 mL) and trimethylbromosilane (1 mL,6.5mmol,21 eq) was added and the reaction stirred under argon for 1 hour at 50 ℃. The reaction solution was purified by reverse phase preparation (C18, 10 mmol/ammonia, acetonitrile) to give the title compound (23.22 mg, yield: 19.42%) as a white solid .LCMS(ESI):[M+H]+=385.1;1H NMR(400MHz,CD3OD)δ7.95(dd,J=11.8,8.0Hz,2H),7.39(d,J=8.0Hz,1H),7.29(t,J=7.2Hz,1H),7.23-7.21(m,2H),7.14(t,J=7.2Hz,1H),7.04(d,J=8.0Hz,2H),6.95(d,J=2.0Hz,1H),6.80(dd,J=8.4,2.4Hz,1H),5.49(s,2H),2.89(d,J=20.8Hz,2H).
Example 43 preparation of (4- ((2, 3-difluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Step 1, preparation of 2, 3-difluoro-9H-carbazole:
Nitrophenyl benzene (200 mg,1.87mmol,1 eq.) was dissolved in anhydrous acetonitrile (5 mL), cesium fluoride (1.4 g,9.35mmol,5 eq.) was added, and after stirring under argon for 10 min, 4, 5-difluoro-2- (trimethylsilyl) phenol triflate (624 mg,1.87mmol,1 eq.) was added. And (3) reacting for 72 hours at room temperature under the protection of argon. The reaction solution was concentrated under reduced pressure to give a crude product, which was purified by a silica gel preparation plate (petroleum ether: dichloromethane (V/v=1/2)) to give the title compound (80 mg, yield: 21.1%). LCMS (ESI) [ M-H ] - =202.0.
Step 2 preparation of diethyl (4- ((2, 3-difluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
After 2, 3-difluoro-9H-carbazole (40 mg,0.2mmol,1 eq) was dissolved in tetrahydrofuran (1 mL), sodium hydride (32 mg,0.79mmol,4 eq) was added, stirred at room temperature for 0.5 hour under nitrogen protection, diethyl (4- (bromomethyl) benzyl) phosphonate (126 mg,0.39mmol,2 eq) was added, stirring was continued for 1 hour, the reaction solution was quenched with saturated ammonium chloride, ethyl acetate (10 mL. Times.3) was added for extraction three times, and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (65 mg, yield: 73.3%). LCMS (ESI) [ m+h ] + = 444.1.
Step 3 preparation of (4- ((2, 3-difluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((2, 3-difluoro-9H-carbazol-9-yl) methyl) benzyl) phosphonate (60 mg,0.14mmol,1 eq.) was dissolved in dichloromethane (1 mL), trimethylbromosilane (104 mg,0.68mmol,5 eq.) was added and the reaction mixture was reacted at 50℃for 2 hours and concentrated under reduced pressure, the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (4.70 mg, yield: 9.0%) as a white solid .LCMS(ESI):[M-H]-=386.1;1H NMR(400MHz,CD3OD)δ8.05(d,J=7.6Hz,1H),7.96(dd,J=10.8,8.0Hz,1H),7.49(d,J=8.0Hz,1H),7.43-7.39(m,1H),7.36(dd,J=11.6,2.8Hz,1H),7.23-7.19(m,3H),7.04(d,J=7.6Hz,2H),5.51(s,2H),2.92(d,J=20.8Hz,2H).
EXAMPLE 44 preparation of (4- ((3- (methylthio) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 3- (methylthio) -9H-carbazole:
3-bromo-9H-carbazole (500 mg,2.0mmol,1 eq) was dissolved in DMSO (10 mL), then sodium thiosulfate pentahydrate (1 g,4.1mmol,2 eq), dimethyl carbonate (915 mg,10.2mmol,5 eq), P (t-Bu) 3.HBF4 (tri-tert-butylphosphine tetrafluoroborate) (118 mg,0.4mmol,0.2 eq), potassium tert-butoxide (455 mg,4.1mmol,2 eq), tetrabutylammonium bromide (2 g,6.1mmol,3 eq) and Pd (acac) 2 (bis (acetylacetonato) palladium (II)) (6 mg,0.02mmol,0.01 eq) were added to this mixture, and the reaction was stirred at 120℃for 16 hours under argon. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (320 mg, yield: 73.8%). LCMS (ESI) [ M-H ] - =212.0.
Step 2 preparation of diethyl (4- ((3- (methylthio) -9H-carbazol-9-yl) methyl) benzyl) phosphonate:
3- (methylthio) -9H-carbazole (300 mg,1.41mmol,1 eq) was dissolved in tetrahydrofuran (10 mL), sodium hydride (450 mg,11.25mmol,8 eq) was added, and the reaction was stirred at room temperature for 0.5 hour under argon protection, and diethyl (4- (bromomethyl) benzyl) phosphonate (2.3 g,7.03mmol,5 eq) was added, and the reaction was stirred at room temperature for 1 hour under argon protection. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (500 mg, yield: 78.4%). LCMS (ESI) [ m+h ] + =454.1.
Step 3 preparation of (4- ((3- (methylthio) -9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((3- (methylthio) -9H-carbazol-9-yl) methyl) benzyl) phosphonate (110 mg,0.24mmol,1 eq) was dissolved in dichloromethane (2 mL) and trimethylbromosilane (0.4 mL,2.61mmol,10 eq) was added and reacted at 50℃for 1 hour. The reaction solution was concentrated at low temperature, dissolved with 2mL of methanol, and purified by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (11 mg, yield) :12.2%).LCMS(ESI):[M+H]-=396.1;1H NMR(400MHz,CD3OD)δ8.11-8.08(m,2H),7.47-7.38(m,4H),7.23-7.17(m,3H),7.03(d,J=8.0Hz,2H),5.52(s,2H),2.88(d,J=20.8Hz,1H),2.53(s,3H).
EXAMPLE 45 preparation of((6- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
Step 1 preparation of diethyl ((6- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate
5H-pyrido [3,2-b ] indole (200 mg,1.19mmol,1.0 eq.) was added to tetrahydrofuran (5 mL), the reaction mixture was cooled to 0℃and sodium hydride (72 mg,1.78mmol,1.5 eq.) was slowly added and after half an hour of reaction at 0℃diethyl ((6- (chloromethyl) pyridin-3-yl) methyl) phosphonate (264 mg,1.31mmol,1.1 eq.) was added and the reaction was continued at room temperature for 1 hour. The reaction mixture was quenched with water (50 mL), and extracted three times with ethyl acetate (50 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated and added to silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/2)) to give the title compound (200 mg, yield: 41.1%) as a yellow solid. LCMS (ESI) [ m+h ] + =410.2.
Step 2 preparation of((6- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
diethyl ((6- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (200 mg,0.49mmol,1.0 eq) was dissolved in dichloromethane (2 mL) and trimethylbromosilane (1.12 g,7.33mmol,15 eq) was added and the reaction stirred at 50℃for 1 hour. The reaction solution was filtered, quenched with methanol, and concentrated under reduced pressure. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (125.3 mg, yield: 72.4%) as an off-white solid .LCMS(ESI):[M+H]+=354.0;1H NMR(400MHz,DMSO-d6,)δ8.46(dd,J=4.4,1.2Hz,1H),8.25(s,1H),8.21(d,J=7.6Hz,1H),8.02(dd,J=8.4,1.2Hz,1H),7.66(d,J=8.4Hz,1H),7.53-7.43(m,2H),7.38(dd,J=8.4,4.4Hz,1H),7.26(t,J=7.2Hz,1H),6.93(d,J=8.0Hz,1H),5.65(s,2H),2.63(d,J=20.8Hz,2H).
Example 46 preparation of (4- ((3-amino-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
step 1 preparation of diethyl (4- ((3-nitro-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
3-nitro-9H-carbazole (500 mg,2.36mmol,1 eq.) was dissolved in tetrahydrofuran (10 mL) and sodium hydride (756 mg,18.9mmol,8 eq.) was added. The reaction mixture was stirred at room temperature under argon for 0.5 hours, diethyl (4- (bromomethyl) benzyl) phosphonate (3.8 g,11.8mmol,5 eq.) was added and reacted at room temperature for 1 hour. To the reaction mixture was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (900 mg, yield: 84.3%). LCMS (ESI) [ m+h ] + =453.2.
Step 2 preparation of diethyl (4- ((3-amino-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((3-nitro-9H-carbazol-9-yl) methyl) benzyl) phosphonate (500 mg,1.11mmol,1 eq.) was dissolved in anhydrous methanol (10 mL), palladium on carbon (50 mg,10 wt%) was added, hydrogen protected, and the reaction was stirred at room temperature for 1 hour. The reaction solution was filtered and concentrated under reduced pressure to give the title compound (400 mg, yield: 85.7%) as LCMS (ESI): M+H ] + = 423.2.
Step 3 preparation of (4- ((3-amino-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((3-amino-9H-carbazol-9-yl) methyl) benzyl) phosphonate (100 mg,0.24mmol,1 eq) was dissolved in dichloromethane (2 mL) and trimethylbromosilane (0.5 mL,3.27mmol,14 eq) was added and reacted at 50℃for 1 hour. The reaction solution was quenched with 2mL of methanol and purified by reverse phase HPLC preparation (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (13.5 mg, yield) :15.6%).LCMS(ESI):[M+H]+=367.0;1H NMR(400MHz,CD3OD)δ7.97(d,J=8.0Hz,1H),7.51(d,J=2.0Hz,1H),7.37-7.33(m,2H),7.31-7.23(m,3H),7.09(t,J=6.8Hz,1H),7.00(d,J=8.0Hz,2H),6.94(dd,J=8.8,2.4Hz,1H),5.89(s,2H),2.81(d,J=20.4Hz,2H).
EXAMPLE 47 preparation of((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphorus-based) bis (oxo)) bis (methylene) bis (2, 2-dimethylpropionate):
Step 1 preparation of(((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphorus-based) bis (oxo)) bis (methylene) bis (2, 2-dimethylpropionate):
(4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid (95 mg,0.23mmol,1.0 eq) was dissolved in acetonitrile (2 mL), and then iodomethyl 2, 2-dimethylpropionate (278 mg,1.15mmol,5 eq) and N, N-diisopropylethylamine (149 mg,1.15mmol,5 eq) were added to the mixture, and the reaction was stirred at 60℃for 16 hours. The reaction solution was concentrated under reduced pressure. Purification of the crude product by reverse phase chromatography (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (46.8 mg, yield: 31.8%) as a white solid .LCMS(ESI):[M+NH4]+=657.7;1H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.4Hz,2H),7.45(d,J=8.8Hz,2H),7.17-7.09(m,2H),7.06-6.97(m,4H),5.64-5.40(m,6H),3.85(s,6H),3.26(d,J=20.4Hz,2H),1.07(s,18H).
Example 48 preparation of((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphorus-based) bis (oxo)) bis (methylene) diisopropylbis (carbonate):
step 1 preparation of((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphorus-based) bis (oxo)) bis (methylene) diisopropylbis (carbonate):
(4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid (200 mg,0.49mmol,1.0 eq.) was dissolved in acetonitrile (2 mL), and to this mixture was added iodomethylpropan-2-ylcarbonate (598 mg,2.45mmol,5.0 eq.) and N, N-diisopropylethylamine (317 mg,2.45mmol,5.0 eq.) and reacted under stirring at 60℃for 16 hours. The reaction solution was concentrated under reduced pressure. Preparation of crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (100 mg, yield: 31.7%) as a brown oil .LCMS(ESI):[M+NH4]+=661.2;1H NMR(400MHz,DMSO-d6)δ7.74(d,J=2.4Hz,2H),7.46(d,J=8.8Hz,2H),7.11(dd,J=8.4,2.4Hz,2H),7.04-7.00(m,4H),5.54-5.45(m,6H),4.80-4.74(m,2H),3.85(s,6H),3.28(d,J=22.0Hz,2H),1.21-1.19(m,12H).
EXAMPLE 49 preparation of diisopropyl 2,2'- ((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphoryl) bis (azanediyl)) (2S, 2' S) -dipropionate:
step 1 preparation of (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonochloride:
(4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid (1.3 g,3.16mmol,1.0 eq.) was dissolved in dichloromethane (15 mL), then oxalyl chloride (803 mg,6.32mmol,2.0 eq.) and N, N-dimethylformamide (23.1 mg,0.32mmol,0.1 eq.) were added and the reaction stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure and dried to give the title compound (1.3 g, yield: 92.9%) as an off-white solid. LCMS (ESI) [ m+h ] + =440.2.
Step 2 preparation of 2,2'- ((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphorus group) bis (azanediyl)) (2 s, 2's) -diisopropyldipropionate:
(4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonochloride (100 mg,0.22mmol,1.0 eq.) was dissolved in dichloromethane (2 mL), and L-alanine isopropyl ester (87.8 mg,7.33mmol,3 eq.) and N, N-diisopropylethylamine (114 mg,1.12mmol,5 eq.) were added and the reaction stirred at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure and dried by spin-drying, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (9.8 mg, yield: 6.89%) as a white solid .LCMS(ESI):[M+H]+=638.4;1H NMR(400MHz,DMSO-d6,ppm)δ7.73(d,J=2.4Hz,2H),7.46(d,J=8.8Hz,2H),7.13(dd,J=8.0,2.0Hz,2H),7.02(d,J=8.8Hz,2H),7.00(d,J=8.8Hz,2H),5.52(s,2H),4.90-4.72(m,2H),4.43-4.33(m,1H),4.30-4.19(m,1H),3.84(s,6H),3.70-3.56(m,1H),3.54-3.51(m,1H),2.99-2.84(m,2H),1.16-1.12(m,15H),1.02-1.00(m,3H).
Example 50 preparation of 4- (3-chlorophenyl) -2- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) -1,3, 2-dioxaphosphorinane 2-oxide:
step 1 preparation of 1- (3-chlorophenyl) propane-1, 3-diol:
ethyl 3- (3-chlorophenyl) -3-oxopropionate (2.0 g,8.8mmol,1.0 eq.) was added to methanol (20 mL) followed by slow addition of sodium borohydride (1.7 g,44.0mmol,5.0 eq.) and the reaction was continued at 65℃for 2 hours. The reaction solution was concentrated under reduced pressure to obtain a crude product as a transparent oil. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) gave the title compound (1.4 g, yield: 85.4%) as a colorless oil .1H NMR(400MHz,DMSO-d6,ppm)δ7.36-7.25(m,4H),5.28(brs,1H),4.69-4.66(m,1H),4.58-4.39(brs,1H),3.54-3.39(m,2H),1.76-1.67(m,2H).
Step 2 preparation of 4- (3-chlorophenyl) -2- (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) -1,3, 2-dioxaphosphorinane 2-oxide:
(4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonochloride (80 mg,0.18mmol,1.0 eq) was dissolved in dichloromethane (2 mL), and 1- (3-chlorophenyl) propane-1, 3-diol (66.6 mg,0.36mmol,2 eq) and N, N-diisopropylethylamine (115 mg,0.89mmol,5 eq) were added to the mixture and the reaction stirred at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure and dried by spin-drying, and the product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (3.56 mg, yield: 3.52%) as a white solid .LCMS(ESI):[M+H]+=562.2;1H NMR(400MHz,DMSO-d6,ppm)δ7.73(d,J=2.4Hz,2H),7.44(d,J=8.8Hz,2H),7.36-7.07(m,6H),7.01-6.97(m,4H),5.50(s,2H),4.63-4.50(m,1H),3.86-3.82(m,8H),3.68-3.52(m,2H),2.83-2.75(m,2H).
EXAMPLE 51 preparation of (((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) (phenoxy) phosphoryl) -L-alanine isopropyl ester:
Step 1 preparation of diphenyl 4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonate:
(4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonodichloride (1.00 g,2.23mmol,1.0 eq.) was dissolved in dichloromethane (10 mL) and N, N-diisopropylethylamine (1.44 g,11.2mmol,5.0 eq.) and phenol (1.05 g,11.2mmol,5.0 eq.) were added sequentially. The reaction was continued at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (500 mg, yield: 39.8%) as a white solid. LCMS (ESI) [ m+h ] + = 564.4.
Step 2 preparation of (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonic acid hydroxyphenyl ester:
Diphenyl (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonate (500 mg,0.887mmol,1.0 eq) was dissolved in methanol (5 mL) and water (5 mL), sodium hydroxide (710 mg,17.7mmol,20.0 eq) was added and the reaction was continued at 50 ℃ for 16 hours. The reaction solution was concentrated under reduced pressure and dried by spin. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (150 mg, yield: 34.7%) as a white oil. LCMS (ESI) [ m+h ] + =488.3.
Step 3 preparation of phenyl 4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) chlorophosphonate:
Phenyl (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) phosphonate (150 mg,0.308mmol,1.0 eq.) was added to dichloromethane (2 mL), oxalyl chloride (399mg, 3.08mmol,10.0 eq.) was added, and finally three drops of N, N-dimethylformamide were added. The reaction was continued at room temperature for 1 hour. After quenching with methanol, LCMS was used to monitor the completion of the reaction, and the reaction mixture was concentrated under reduced pressure to give the title compound (100 mg, yield: 64.1%) as a white solid. LCMS (ESI) [ M-cl+meo+h ] + =502.2.
Step 4 preparation of (((4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) benzyl) (phenoxy) phosphoryl) -L-alanine isopropyl ester:
Phenyl (4- ((3, 6-dimethoxy-9H-carbazol-9-yl) methyl) chlorophosphonate (100 mg,0.20mmol,1.0 eq.) was dissolved in dichloromethane (2 mL), N-diisopropylethylamine (128 g,0.99mmol,5.0 eq.) was added, L-alanine isopropyl ester (130 mg,0.99mmol,5.0 eq.) was added, the reaction was reacted at room temperature for 2 hours, the reaction was concentrated under reduced pressure to give crude product, and reversed phase HPLC was used to prepare (C18, 10mmol/L formic acid, acetonitrile) the title compound (4.99 mg, yield: 4.15%) was obtained as a white solid .LCMS(ESI):[M+H]+=601.2;1H NMR(400MHz,CDCl3,ppm)δ7.55(d,J=2.2Hz,2H),7.32-7.27(m,1H),7.26-7.21(m,5H),7.13-7.01(m,7H),5.44(s,2H),4.95-4.84(m,1H),4.01-3.86(m,7H),3.33-3.01(m,3H),1.20-1.03(m,9H).
Example 52 preparation of (4- (4- (dimethylamino) -7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 4, 6-difluoro-3-nitro-1H-indole-2-carboxylic acid:
Concentrated nitric acid (1 mL) was added to acetic anhydride (9 mL) under ice bath, and 4, 6-difluoro-1H-indole-2-carboxylic acid (5 g,25.4mmol,1 eq.) was added and the reaction was allowed to react for two hours under ice bath until a solid appeared. After completion of the reaction by LCMS, the reaction solution was filtered to give the title compound (5.8 g, yield: 94.4%). LCMS (ESI) [ M-H ] - =241.2.
Step 2 preparation of 4, 6-difluoro-3-nitro-1H-indole-2-carboxylic acid methyl ester:
4, 6-difluoro-3-nitro-1H-indole-2-carboxylic acid (2 g,8.26mmol,1 eq.) was added to methanol (50 mL) followed by thionyl chloride (2 mL,16.8mmol,2.04 eq.). The reaction was continued for 1 hour after the reaction was warmed to 80 ℃. LCMS monitored completion of the reaction, and the reaction mixture was concentrated under reduced pressure, diluted with ethyl acetate, neutralized with saturated aqueous sodium bicarbonate, extracted three times with ethyl acetate (20 ml×3), and concentrated under reduced pressure to give the title compound (600 mg, yield: 28.3%). LCMS (ESI) [ M-H ] - =255.2.
Step 3 preparation of 4, 6-difluoro-3-nitro-1H-indole-2-carboxamide:
Methyl 4, 6-difluoro-3-nitro-1H-indole-2-carboxylate (2 g,7.81mmol,1 eq.) was added to 1, 4-dioxane (24 mL), and aqueous ammonia (24 mL,0.68mol,87.7 eq.) was added and the reaction stirred at 100℃for 16 hours. LCMS monitored completion of the reaction, concentrated under reduced pressure, diluted with ethyl acetate, washed three times with aqueous ammonium chloride and the organic phase concentrated to give the title compound (1.4 g, yield: 74.3%). LCMS (ESI) [ M-H ] - =240.1.
Step 4 preparation of 3-amino-4, 6-difluoro-1H-indole-2-carboxamide:
4, 6-difluoro-3-nitro-1H-indole-2-carboxamide (250 mg,1.04mmol,1 eq.) was dissolved in water (5 mL) and ethanol (5 mL), followed by the addition of iron powder (289 mg,5.18mmol,5 eq.) and ammonium chloride solid (554 mg,10.4mmol,10 eq.). The reaction was heated to 80 ℃ and stirred for 1 hour. LCMS monitored completion of the reaction, the reaction was cooled, concentrated under reduced pressure, sonicated with dichloromethane for ten minutes, filtered, and the filtrate extracted three times with dichloromethane and concentrated to give the title compound (150 mg, yield: 68.5%), LCMS (ESI): [ m+h ] + =212.1.
Step 5 preparation of 7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-4-ol:
3-amino-4, 6-difluoro-1H-indole-2-carboxamide (400 mg,1.89mmol,1 eq.) was dissolved in dichloromethane (7 mL) and triethylamine (575 mg,5.68mmol,3 eq.) and trifluoroacetic anhydride (1.19 g,5.68mmol,3 eq.) were added and reacted for 2 hours at room temperature. The reaction solution was diluted with saturated aqueous sodium hydrogencarbonate solution (30 mL), the mixture was extracted with ethyl acetate (30 ml×3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by thin layer preparative plate chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) to give the title compound (200 mg, yield: 36.6%) as a brown solid. LCMS (ESI) [ M-H ] - = 288.0.
Step 6 preparation of 4-chloro-7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indole:
7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-4-ol (200 mg,0.69mmol,1 eq.) is dissolved in acetonitrile (3 mL) and N, N-diisopropylethylamine (447 mg,3.46mmol,5 eq.) and phosphorus oxychloride (530 mg,3.46mmol,5 eq.) are added in sequence and reacted at 120℃for 2 hours. After completion of the LCMS monitoring reaction, the reaction was diluted with saturated aqueous sodium bicarbonate (30 mL) and extracted three times with ethyl acetate (30 ml×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was the title compound (100 mg, yield: 47.0%) as a brown solid. LCMS (ESI) [ M-H ] - = 306.0.
Step 7 preparation of 7, 9-difluoro-N, N-dimethyl-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-4-amine
4-Chloro-7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indole (100 mg,0.33mmol,1 eq.) was dissolved in acetonitrile (3 mL) and dimethylamine (1 mL) followed by N, N-diisopropylethylamine (0.5 mL) was added. The reaction was heated to 100 ℃ under argon and stirred for 1 hour, LCMS monitored reaction complete. To the reaction solution was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated in vacuo to give the title compound (50 mg, yield: 48.6%) as a yellow liquid. LCMS (ESI) [ m+h ] + = 317.0.
Step 8 preparation of diethyl (4- ((4- (dimethylamino) -7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
7, 9-Difluoro-N, N-dimethyl-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-4-amine (20 mg,0.06mmol,1 eq.) is dissolved in tetrahydrofuran (2 mL) and di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (24 mg,0.06mmol,1 eq.) and sodium hydride (15.2 mg,0.38mmol,6 eq.) are added. The reaction was stirred at 25℃for 1 hour under argon, and the reaction was monitored by LCMS to completion. To the reaction was added saturated aqueous ammonium chloride (10 mL), and the mixture was extracted three times with ethyl acetate (10 mL. Times.3). The organic phases were combined, and after drying over anhydrous sodium sulfate, the title compound (20 mg, yield: 51.6%) was obtained after concentration in vacuo as a yellow liquid. LCMS (ESI) [ m+h ] + =613.1.
Step 9 preparation of (4- (4- (dimethylamino) -7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Di-tert-butyl (4- (4- (dimethylamino) -7, 9-difluoro-2- (trifluoromethyl) -5H-pyrimido [5,4-b ] indol-5-yl) methyl) phosphonate (61 mg,0.1mmol,1 eq.) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (49 mg,0.5mmol,5 eq.) was added. The reaction was stirred at room temperature under argon for 0.5 hours. LCMS monitored completion of the reaction. Water (2 mL) was added to the reaction, and the aqueous phase was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) and purified to give the title compound (1.8 mg,0.01mmol, 3.64% yield) as a white solid .LCMS(ESI):[M+H]+=501.1;1H NMR(400MHz,CD3OD)δ7.30(d,J=6.4Hz,2H),7.07(d,J=7.6Hz,2H),7.03-7.01(m,1H),6.67-6.62(m,1H),5.32(s,2H),3.10(s,6H),2.94(d,J=20.4Hz,2H).
EXAMPLE 53 preparation of (4- ((7, 9-difluoro-4-hydroxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
1-Chloro-5, 7-difluoro-9H-pyrimido [3,4-b ] indole (110 mg,0.46mmol,1 eq.) was dissolved in N, N-dimethylformamide (15 mL), diethyl (4- (bromomethyl) benzyl) phosphonate (221 mg,0.69mmol,1.5 eq.) and potassium carbonate (64 mg,0.46mmol,1 eq.) were added, after stirring at 70℃for 1 hour, the reaction mixture was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (10 mL. Times 3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (130 mg, yield: 59.0%) as a yellow oil. LCMS (ESI) [ m+h ] + = 480.0.
Step 2 preparation of (4- ((7, 9-difluoro-4-hydroxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (110 mg,0.23mmol,1 eq) was dissolved in dichloromethane (5 mL) and trimethylbromosilane (1 mL,6.5mmol,28 eq) was added and the reaction stirred under argon at 50℃for 1 hour. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10 mmol/ammonia water, acetonitrile) to give the title compound (50.87 mg, yield: 56.5%) as a white solid .LCMS(ESI):[M+H]+=406.1;1H NMR(400MHz,CD3OD)δ8.16(s,1H),7.28-7.26(m,2H),7.08(d,J=8.0Hz,2H),7.00(d,J=10.0Hz,1H),6.65(t,J=8.0Hz,1H),5.94(s,2H),2.80(d,J=19.6Hz,2H).
EXAMPLE 54 preparation of 4- (4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-4-ol
3-Amino-4, 6-difluoro-1H-indole-2-carboxamide (1.5 g,7.1mmol,1 eq.) was dissolved in acetonitrile (15 mL) and hydrochloric acid (15 mL,4 mol/l 1, 4-dioxane solution) was added and reacted for 1 hour at 120℃with microwaves. LCMS checked the completion of the reaction, diluted with ethyl acetate, neutralized with sodium bicarbonate, extracted three times with ethyl acetate (50 ml×3) and concentrated to give the title compound (1.3 g, yield: 77.8%). LCMS (ESI) [ m+h ] + =236.0.
Step 2 preparation of 4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole
7, 9-Difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-4-ol (200 mg,0.85mmol,1 eq.) was added to phenylphosphoryl dichloride (15 mL), the reaction was warmed to 180℃and stirred for 1 hour. The reaction solution was diluted with ethyl acetate (20 mL), neutralized with sodium hydrogencarbonate, extracted three times with ethyl acetate (30 mL. Times.3), and concentrated to give the title compound (150 mg, yield: 69.5%). LCMS (ESI) [ m+h ] + =254.0.
Step 3 preparation of di-tert-butyl (4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (180 mg,0.71mmol,1.0 eq.) was added to N, N-dimethylformamide (3 mL), followed by di-tert-butyl (4-bromomethyl) phosphonate (402 mg,1.06mmol,1.5 eq.) and potassium carbonate (393 mg,2.84mmol,4.0 eq.) and the reaction was continued at 80℃for 1 hour. The reaction mixture was quenched with water (10 mL) and extracted three times with ethyl acetate (20 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (155 mg, yield: 39.7%) as a white solid. LCMS (ESI) [ m+na ] + = 572.1.
Step 4 preparation of 4- (4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phosphonate (50 mg,0.09mmol,1.0 eq.) was dissolved in dichloromethane (1 mL) and trifluoroacetic acid (0.5 mL,5.1mmol,56 eq.) was added and the reaction stirred at room temperature for 12 hours. The reaction solution was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (39.4 mg, yield: 71.0%) as a white solid .LCMS(ESI):[M+H]+=437.9;1H NMR(400MHz,DMSO-d6)δ7.56(d,J=10.0Hz,1H),7.21(t,J=8.4Hz,1H),7.08(d,J=7.6Hz,2H),6.94(d,J=7.6Hz,2H),5.84(s,2H),2.73(s,3H),2.69(d,J=19.2Hz,2H).
EXAMPLE 55 preparation of((4- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phenyl) methyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-5H-pyrimido [5,4-b ] indole:
4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (800 mg,3.34mmol,1 eq.) was dissolved in methanol (10 mL), palladium on carbon (36 mg,4.5 wt%) was added and stirred at room temperature under a hydrogen atmosphere for 1 hour. The reaction solution was filtered, and concentrated under reduced pressure to give the title compound (300 mg, yield: 43.8%). LCMS (ESI) [ m+h ] + =206.1.
Step 2 preparation of diethyl ((4- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phenyl) methyl) phosphonate:
7, 9-difluoro-5H-pyrimido [5,4-b ] indole (200 mg,0.97mmol,1 eq.) was dissolved in N, N-dimethylformamide (15 mL), diethyl ((4- (bromomethyl) phenyl) methyl) phosphonate (626 mg,1.95mmol,2 eq.) and potassium carbonate (404 mg,2.92mmol,3 eq.) were added and reacted for 1 hour at 80℃diluted with saturated ammonium chloride water and ethyl acetate (30 mL. Times.3) extracted three times. The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (250 mg, yield: 57.6%). LCMS (ESI) [ m+h ] + =446.1.
Step 3 preparation of ((4- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phenyl) methyl) phosphonic acid:
((4- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phenyl) methyl) phosphonate diethyl ester (100 mg,0.22mmol,1 eq) was dissolved in dichloromethane (6 mL) and trimethylbromosilane (2 mL,13.06mmol,29.68 eq) was added and stirred at 50℃for 1 hour. The reaction solution was concentrated under reduced pressure, and the crude product was purified by reverse phase preparation (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (22.23mg,0.06mmol,25.96%).LCMS(ESI):[M+H]+=390.0;1H NMR(400MHz,D2O)δ8.81-8.73(m,2H),7.08-7.05(m,2H),6.96-6.90(m,2H),6.78-6.70(m,2H),5.41-5.13(m,2H),2.66-2.56(m,2H).
EXAMPLE 56 preparation of 4- ((4, 8-dichloro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 5-chloro-3-nitro-1H-indole-2-carboxylic acid methyl ester:
Concentrated nitric acid (3 mL) was added to acetic anhydride (27 mL) at 0deg.C, and 5-chloro-1H-indole-2-carboxylic acid methyl ester (20 g,102.3mmol,1 eq.) was added and the reaction solution reacted at room temperature for two hours. The reaction solution was filtered, and the solid phase was washed with water and dried to give the title compound (20 g, yield: 81.3%). LCMS (ESI) [ M-H ] - =239.1.
Step 2 preparation of 5-chloro-3-nitro-1H-indole-2-carboxamide:
Methyl 5-chloro-3-nitro-1H-indole-2-carboxylate (2.8 g,10.42mmol,1 eq.) was dissolved in 1, 4-dioxane (8 mL), aqueous ammonia (8 mL,228mmol,22 eq.) was added, reacted for 1 hour at 100℃with microwaves, saturated aqueous ammonium chloride (20 mL) was added, and extracted three times with ethyl acetate (20 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude title compound (2 g, yield: 80.1%) as a yellow solid. LCMS (ESI) [ M-H ] - =238.0.
Step 3 preparation of 3-amino-5-chloro-1H-indole-2-carboxamide:
To a solution of 5-chloro-3-nitro-1H-indole-2-carboxamide (2 g,8.35mmol,1 eq.) in ethanol (20 mL) and water (2 mL) was added iron powder (2.33 g,41.8mmol,5 eq.) and ammonium chloride (2.23 g,41.8mmol,5 eq.). The reaction was stirred at 90 ℃ for 16 hours under a hydrogen atmosphere. Filtration, concentration under reduced pressure, addition of saturated aqueous sodium hydrogencarbonate (30 mL) and extraction with ethyl acetate (20 mL. Times.3). The organic phase was collected, dried and concentrated to give a crude title compound (1.8 g, crude yield: 90%) as a brown solid. LCMS (ESI) [ m+h ] + =210.1.
Step 4 preparation of 8-chloro-5H-pyrimido [5,4-b ] indol-4-ol:
3-amino-5-chloro-1H-indole-2-carboxamide (500 mg,2.39mmol,1 eq.) and formamidine acetate (99.95 mg,0.96mmol,2 eq.) were dissolved in absolute ethanol (3 mL) and the reaction was refluxed for 16 hours. The reaction was cooled to room temperature and filtered to give the title compound (300 mg,1.37mmol, 57.27%) as a grey solid. LCMS (ESI) [ m+h ] + =220.0.
Step 5 preparation of 4, 8-dichloro-5H-pyrimido [5,4-b ] indole:
8-chloro-5H-pyrimido [5,4-b ] indol-4-ol (400 mg,1.28mmol,1.0 eq.) is added to phenylphosphonic dichloride (4 mL) and reacted at 180℃for 1 hour. The reaction mixture was quenched with water (10 mL) and extracted three times with ethyl acetate (20 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, dried over silica gel, and purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) to give the title compound (150 mg, yield: 34.6%) as a white solid. LCMS (ESI) [ m+h ] + =238.0.
Preparation of di-tert-butyl ((4, 8-dichloro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
4, 8-dichloro-5H-pyrimido [5,4-b ] indole (150 mg,0.630mmol,1.0 eq.) was added to N, N-dimethylformamide (2 mL), followed by di-tert-butyl ((4-bromomethyl) benzyl) phosphonate (238 mg,0.630mmol,1.0 eq.) and potassium carbonate (261 mg,1.89mmol,3.0 eq.) and reacted at 80℃for 1 hour. The reaction mixture was quenched with water (10 mL) and extracted three times with ethyl acetate (20 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. Flash chromatography purification (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) afforded the title compound (200 mg, yield: 59.4%) as a white solid. LCMS (ESI) [ m+na ] + =556.1.
Step 7 preparation of 4- ((4, 8-dichloro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4, 8-dichloro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phosphonate (100 mg,0.190mmol,1.0 eq.) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (0.5 mL,5.1mmol,56 eq.) was added to the mixture, the reaction was stirred at room temperature for 2 hours, and the reaction solution was concentrated under reduced pressure. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (32.48 mg, yield: 42.1%) as a white solid .LCMS(ESI):[M+H]+=422.0;1HNMR(400MHz,DMSO-d6)δ8.95(s,1H),8.34(s,1H),7.81-7.72(m,2H),7.11(d,J=8.4Hz,2H),6.98(d,J=8.0Hz,2H),5.92(s,2H),2.65(d,J=20.4Hz,2H).
Example 57 preparation of (4- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (300 mg,1.18mmol,1 eq.) was dissolved in N, N-dimethylformamide (10 mL), and anhydrous potassium carbonate (490 mg,3.54mmol,3 eq.) was added and reacted at 70℃for 2 hours. The reaction solution was diluted with a saturated ammonium chloride solution, extracted three times with ethyl acetate (30 mL. Times.3), filtered, and concentrated to give the title compound (500 mg, yield: 85.8%). LCMS (ESI) [ m+h ] + =494.2.
Step 2 preparation of diethyl (4- ((7, 9-difluoro-2-methyl-3, 4-dihydro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (350 mg,0.71mmol,1 eq) was dissolved in anhydrous methanol (10 mL), palladium on carbon (200 mg,57 wt%) was added, hydrogen was replaced three times, and the reaction was continued at ambient temperature for 2 hours. The reaction solution was filtered and concentrated to give the crude title compound (300 mg, yield: 91.6%). LCMS (ESI) [ m+h ] + =462.1.
Step 3 preparation of diethyl (4- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((7, 9-difluoro-2-methyl-3, 4-dihydro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (300 mg,0.65mmol,1 eq) was dissolved in dichloromethane (5 mL), manganese dioxide (113 mg,1.3mmol,2 eq) was added and reacted for 2 hours at ambient temperature. The reaction solution was concentrated by filtration to give the crude title compound (200 mg, yield: 67.0%). LCMS (ESI) [ m+h ] + =460.0.
Step 4 preparation of (4- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.44mmol,1 eq) was dissolved in dichloromethane (6 mL) and trimethylbromosilane (1 mL,6.53mmol,14.8 eq) was added and reacted at 50 ℃ for 1 hour. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (27.3 mg, yield) :15.9%).LCMS(ESI):[M+H]+=404.0;1H NMR(400MHz,D2O)δ8.69(s,1H),7.14-7.09(m,2H),6.99-6.94(m,3H),6.78-6.73(m,1H),5.33(s,2H),2.71-2.64(m,5H).
EXAMPLE 58 preparation of (4- ((8-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 (preparation of 5-methoxy-2- (4-methoxybenzyl) amino) benzonitrile:
2-fluoro-5-methoxybenzonitrile (16 g,0.105mol,1.0 eq.) was dissolved in dimethyl sulfoxide (150 mL), then 4-methoxybenzylamine (15.9 g,0.116mol,1.2 eq.) and potassium carbonate (16.0 g,0.116mol,1.2 eq.) were added and heated to 100℃under argon and stirred for 16 hours. After completion of the reaction, the mixture was cooled, filtered, and the filtrate was diluted with water (300 mL), and the mixture was extracted three times with ethyl acetate (300 ml×3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated to give the crude title compound (22 g,0.82mol, 77.7% yield) as a yellow solid. LCMS (ESI) [ m+h ] + = 269.1.
Step 2 preparation of 3-amino-5-methoxy-1- (4-methoxybenzyl) -1H-indole-2-carboxylic acid ethyl ester:
(5-methoxy-2- (4-methoxybenzyl) amino) benzonitrile (4 g,0.01 mol,1.0 eq) and ethyl bromoacetate (14.7 g,0.088mol,8.0 eq) were dissolved in anhydrous N, N-dimethylformamide (80 mL), then potassium t-butoxide (4.9 g,0.044mol,4 eq) was added, the reaction was stirred at room temperature under argon for 1 hour, and then potassium t-butoxide (4.9 g,0.044mol,4 eq) was added over one hour, and the reaction remained light brown. The mixture was stirred at room temperature for 16 hours, and the filtrate was diluted with saturated aqueous ammonium chloride (100 mL) and extracted three times with ethyl acetate (100 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=6/1)) to give the title compound (1.0 g,0.0025mol, yield 25.1%) as a yellow oily liquid. LCMS (ESI) [ m+h ] + = 355.0.
Step 3 preparation of 5-methoxy-1- (4-methoxybenzyl) -3- (3- (2, 2-trichloroacetyl) ureido) -1H-indole-2-carboxylic acid ethyl ester:
3-amino-5-methoxy-1- (4-methoxybenzyl) -1H-indole-2-carboxylic acid ethyl ester (4.5 g,0.013mol,1.0 eq.) was dissolved in anhydrous tetrahydrofuran (40 mL), and trichloroacetyl isocyanate (2.4 g,0.013mol,1 eq.) was added and the reaction stirred at room temperature under argon for 10 minutes. The reaction solution was concentrated directly under reduced pressure to give the crude title compound (5.8 g,0.01mmol, yield 82.4%) as a yellow solid. LCMS (ESI) [ M-H ] - =540.2.
Step 4 preparation of 8-methoxy-5- (4-methoxybenzyl) -5H-pyrimido [5,4-b ] indole-2, 4-diol:
Ethyl 5-methoxy-1- (4-methoxybenzyl) -3- (3- (2, 2-trichloroacetyl) ureido) -1H-indole-2-carboxylate (5.8 g,0.01 mol,1.0 eq.) was dissolved in methanol (60 mL), then sodium hydroxide (2.6 g,0.066mol,6 eq.) was added and the reaction solution was heated to 90℃under argon and stirred for 1 hour. Reaction completion was monitored by LCMS. The reaction solution was cooled to room temperature, and the mixture was adjusted to pH 2 with 1 mol/liter of aqueous hydrochloric acid to give a suspension, which was filtered, and the solid was dried to give the objective compound (2.0 g,0.008mol, yield 48.5%) as a yellow solid. LCMS (ESI) [ m+h ] + = 352.2.
Step 5 preparation of 2, 4-dichloro-8-methoxy-5H-pyrimido [5,4-b ] indole
8-Methoxy-5- (4-methoxybenzyl) -5H-pyrimido [5,4-b ] indole-2, 4-diol (500 mg,1.42mmol,1 eq.) was dissolved in phenylphosphonic dichloride (10 mL) and the reaction was heated to 180deg.C under argon and stirred for 2 hours. The reaction solution was cooled to room temperature, then placed in an ice bath, slowly added dropwise with saturated aqueous sodium bicarbonate solution for quenching, and the pH was adjusted to 6-7. The mixture was extracted three times with ethyl acetate (300 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=4/1)) to give the title compound (250 mg, yield: 65.5%) as a pale yellow solid. LCMS (ESI) [ m+h ] + = 268.0.
Step 6 preparation of 8-methoxy-4, 5-dihydro-3H-pyrimido [5,4-b ] indole
2, 4-Dichloro-8-methoxy-5H-pyrimido [5,4-b ] indole (100 mg,0.37mmol,1 eq.) was dissolved in methanol (2 mL) and palladium on carbon (50 wt%, 50 mg) was then added to the mixture, the hydrogen was purged three times, and the reaction was carried out at 25℃for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound (74 mg, yield: 99.4%) as a yellow solid. LCMS (ESI) [ m+h ] + =202.1.
Step 7 preparation of 8-methoxy-5H-pyrimido [5,4-b ] indole
8-Methoxy-4, 5-dihydro-3H-pyrimido [5,4-b ] indole (74 mg,0.37mmol,1 eq.) was dissolved in dichloromethane (2 mL) and manganese dioxide (64 mg,0.74mmol,2 eq.) was added to the mixture and the nitrogen sparged three times at 25℃for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound (70 mg, yield: 95.0%) as a yellow solid. LCMS (ESI) [ m+h ] + =200.0.
Step 8 preparation of diethyl (4- ((8-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
8-Methoxy-5H-pyrimido [5,4-b ] indole (70 mg,0.35mmol,1 eq.) was dissolved in tetrahydrofuran (2 mL), then sodium hydride (56.4 mg,1.41mmol,4.0 eq.) was added to the mixture and reacted for 0.5 hours with stirring, then diethyl (4- (bromomethyl) benzyl) phosphonate (169 mg,0.53mmol,1.5 eq.) was added to the mixture and reacted for 2 hours at 25 ℃. The reaction solution was diluted with saturated aqueous ammonium chloride (20 mL), the mixture was extracted with ethyl acetate (20 ml×3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by thin layer preparative plate chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (90 mg, yield: 58.5%) as a yellow solid. LCMS (ESI) [ m+h ] + =440.1.
Step 9 preparation of (4- ((8-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((8-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (85 mg,0.19mmol,1 eq) was dissolved in acetonitrile (1 mL), trimethylbromosilane (2915 mg,1.90mmol,10 eq) was added, the reaction was heated to 50℃and the reaction stirred for 2 hours. After completion of the reaction, water (1 mL) was added to the reaction mixture, and the aqueous phase was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) and purified to give the title compound (35 mg, yield: 51.8%) as a yellow solid .LCMS(ESI):[M+H]+=384.0;1H NMR(400MHz,CD3OD)δ9.09(s,1H),8.98(s,1H),7.85(d,J=2.4Hz,1H),7.66(d,J=11.2Hz,1H),7.37(dd,J=8.8,2.4Hz,1H),7.28(d,J=8.0,2.0Hz,2H),7.09(d,J=8.0Hz,2H),5.65(s,2H),3.93(s,3H),2.88(d,J=20.4Hz,2H).
EXAMPLE 59 preparation of (4- ((4- (dimethylamino) -7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-N, 2-trimethyl-5H-pyrimido [5,4-b ] indol-4-amine:
4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (400 mg,1.6mmol,1.0 eq.) was added to ultra-dry acetonitrile (5 mL) and N, N-diisopropylethylamine (611 mg,4.7mmol,3.0 eq.) and dimethylamine (142 mg,3.2mmol,2.0 eq.) were slowly added and the reaction was continued at 90℃for 1 hour. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, dichloromethane: methanol (V/v=19/1)) to give the title compound (195 mg, yield: 46.0%) as a yellow solid. LCMS (ESI) [ m+h ] + = 263.1.
Preparation of di-tert-butyl 4- ((4- (dimethylamino) -7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
7, 9-difluoro-N, N, 2-trimethyl-5H-pyrimido [5,4-b ] indol-4-amine (195 mg,0.74mmol,1.0 eq.) is dissolved in tetrahydrofuran (3 mL) and sodium hydride (35.6 mg,0.89mmol,1.2 eq.) is added at 0deg.C and allowed to react for 30 minutes at room temperature. Di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (560 mg,1.5mmol,2.0 eq.) was added, stirred at room temperature for 2 hours, the reaction quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (144 mg, yield: 37.0%) as a white solid. LCMS (ESI) [ m+h ] + = 559.4.
Step 3 preparation of (4- ((4- (dimethylamino) -7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((4- (dimethylamino) -7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (250 mg,0.45mmol,1.0 eq.) was dissolved in methanol (3 mL) and a dioxane solution of hydrogen chloride (0.33 mL,4 mol/liter) was added and stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (57.71 mg, yield: 28.7%) as a white solid .LCMS(ESI):[M+H]+=447.2;1H NMR(400MHz,DMSO-d6)δ7.43(d,J=9.6Hz,1H),7.01-6.95(m,3H),6.79(d,J=7.6Hz,2H),5.50(s,2H),2.96(s,6H),2.59(s,3H),2.53(d,J=12.4Hz,2H).
Example 60 preparation of (4- ((4- (dimethylamino) -7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-N, N-dimethyl-5H-pyrimido [5,4-b ] indol-4-amine:
4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (300 mg,1.3mmol,1.0 eq.) was added to ultra-dry acetonitrile (5 mL) and N, N-diisopropylethylamine (481mg, 3.8mmol,3.0 eq.) and dimethylamine (113 mg,2.5mmol,2.0 eq.) were slowly added and the reaction was continued for 16 hours at 90 ℃. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, concentrated, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (300 mg, yield: 96.7%) as a yellow solid. LCMS (ESI) [ m+h ] + =249.1.
Step 2 preparation of di-tert-butyl 4- ((4- (dimethylamino) -7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
7, 9-difluoro-N, N-dimethyl-5H-pyrimido [5,4-b ] indol-4-amine (300 mg,1.2mmol,1.0 eq.) is dissolved in tetrahydrofuran (5 mL) and sodium hydride (96 mg,2.4mmol,2.0 eq.) is added thereto at 0℃and reacted for 30 minutes at 0 ℃. Di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (684 mg,1.8mmol,2.0 eq.) was added and the reaction stirred at room temperature for 2 hours. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, and concentrated. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (300 mg, yield: 45.5%) as a white solid. LCMS (ESI) [ m+h ] + = 545.5.
Step 3 preparation of (4- ((4- (dimethylamino) -7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((4- (dimethylamino) -7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.4mmol,1.0 eq) was dissolved in methanol (3 mL), and then a dioxane solution of hydrogen chloride (0.3 mL,4 mol/l) was added to the mixture and the reaction was stirred at room temperature for 16 hours. The reaction solution was quenched with methanol, concentrated under reduced pressure to give a crude white product, which was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (86.14 mg, yield: 53.9%) as a white solid .LCMS(ESI):[M+H]+=433.2;1H NMR(400MHz,DMSO-d6)δ8.62(s,1H),7.57(dd,J=9.8,1.4Hz,1H),7.10(td,J=10.2,1.5Hz,1H),7.04(dd,J=8.0,1.7Hz,2H),6.88(d,J=7.9Hz,2H),5.64(s,2H),3.03(s,6H),2.78(d,J=21.4Hz,2H);19F NMR(376.5MHz,DMSO-d6)δ-107.89,-112.77.
Example 61 preparation of (4- ((7, 9-difluoro-4-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-4-methoxy-5H-pyrimido [5,4-b ] indole:
4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (500 mg,2.1mmol,1.0 eq.) was added to methanol (5 mL) and sodium methoxide (226 mg,4.2mmol,2.0 eq.) was slowly added and the reaction continued at 70℃for 1 hour. The reaction was cooled, quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, concentrated, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (380 mg, yield: 77.4%) as a yellow solid. LCMS (ESI) [ m+h ] + =236.0.
Step 2 preparation of di-tert-butyl 4- ((7, 9-difluoro-4-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
7, 9-difluoro-4-methoxy-5H-pyrimido [5,4-b ] indole (380 mg,1.6mmol,1.0 eq.) was dissolved in tetrahydrofuran (5 mL), sodium hydride (128 mg,3.2mmol,2.0 eq.) was added at 0deg.C, reacted for 30 minutes at 0deg.C, di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (910 mg,2.4mmol,1.5 eq.) was added and stirred at room temperature for 2 hours. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, concentrated, and dried over silica gel. Flash chromatography purification (silica gel, dichloromethane: methanol (V/v=10/1)) afforded the title compound (160 mg, yield: 18.6%) as a white solid. LCMS (ESI) [ m+h ] + =532.2.
Step 3 preparation of (4- ((7, 9-difluoro-4-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Di-tert-butyl (4- ((7, 9-difluoro-4-methoxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (160 mg,0.3mmol,1.0 eq) was dissolved in methanol (3 mL), and then a dioxane solution of hydrogen chloride (1.5 mL,1 mol/l) was added to the mixture and the reaction was stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure to give a crude product, which was prepared by reversed phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (91.9 mg, yield: 73.1%) as a white solid .LCMS(ESI):[M+H]+=420.0;1H NMR(400MHz,DMSO-d6,ppm)δ8.64(s,1H),7.49(d,J=9.2Hz,1H),7.09-7.01(m,5H),5.66(s,2H),4.10(s,3H),2.61(d,J=20.8Hz,2H).
EXAMPLE 62 preparation of((6- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid
Step 1 preparation of diethyl ((6- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate:
4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (500 mg,2.1mmol,1.0 eq.) was dissolved in tetrahydrofuran (5 mL) and sodium hydride (168 mg,4.2mmol,2.0 eq.) was added at 0deg.C and reacted for 30min at 0deg.C. Diethyl ((6- (chloromethyl) pyridin-3-yl) methyl) phosphonate (580 mg,2.1mmol,1.0 eq.) was added and the reaction stirred at room temperature for 2 hours. The reaction mixture was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, concentrated, and dried over silica gel. Flash chromatography purification (silica gel, dichloromethane: methanol (V/v=10/1)) afforded the title compound (600 mg, yield: 59.7%) as a yellow solid. LCMS (ESI) [ m+h ] + =480.9.
Step 2 preparation of ((6- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
Diethyl ((6- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (20 mg,0.04mmol,1.0 eq) was dissolved in dichloromethane (2 mL), trimethylbromosilane (96 mg,0.63mmol,15 eq) and bis (trimethylsilyl) amine (101 mg,0.63mmol,15 eq) were added and stirred at 50℃for 1 hour. The reaction mixture was quenched with methanol and concentrated under reduced pressure to give crude product as a brown oil. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (12.72 mg, yield: 72.0%) as a white solid .LCMS(ESI):[M+H]+=425.0;1H NMR(400MHz,DMSO-d6,ppm)δ8.88(s,1H),8.10(s,1H),7.62(d,J=8.4Hz,1H),7.54(d,J=8.0Hz,1H),7.23(t,J=9.2Hz,1H),7.13(d,J=8.0Hz,1H),5.96(s,2H),2.61(d,J=20.8Hz,2H).
EXAMPLE 63 preparation of((6- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid
Step 1((6- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid diethyl ester:
4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (457 mg,1.8mmol,1.0 eq.) was dissolved in N, N-dimethylformamide (5 mL), diethyl ((6- (chloromethyl) pyridin-3-yl) methyl) phosphonate (500 mg,1.8mmol,1.0 eq.) and potassium carbonate (748 mg,5.4mmol,3.0 eq.) were added in sequence and reacted for 1 hour at 80 ℃. The reaction solution was cooled to room temperature, quenched with water (15 mL), and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, concentrated, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=95/5)) to give the title compound (200 mg, yield: 22.8%) as a white solid. LCMS (ESI) [ m+h ] + = 495.1.
Step 2 preparation of ((6- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
Diethyl ((6- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (60 mg,0.12mmol,1.0 eq.) was dissolved in dichloromethane (2 mL) and trimethylbromosilane (274 mg,1.8mmol,15 eq.) and bis (trimethylsilyl) amine (254 mg,1.8mmol,15 eq.) were added in sequence and reacted for 1 hour with stirring at 50 ℃. The reaction solution was quenched with methanol and concentrated under reduced pressure to give a crude product as a brown oil. Preparation by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (39.29 mg, yield: 74.6%) as a white solid .LCMS(ESI):[M+H]+=439.0;1H NMR(400MHz,DMSO-d6,ppm)δ8.11(s,1H),7.63-7.46(m,2H),7.19(t,J=10.0Hz,1H),7.11(d,J=8.0Hz,1H),5.91(s,2H),2.72(s,3H),2.68(d,J=20.8Hz,2H).
EXAMPLE 64 preparation of((6- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid
Step 1 preparation of diethyl ((6- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate:
Diethyl ((6- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (130 mg,0.26mmol,1.0 eq) was dissolved in ethanol (2 mL), palladium on carbon (100 mg,76 wt%) and potassium carbonate (182 mg,1.3mmol,5.0 eq) were added, and the reaction was stirred under hydrogen atmosphere for 2 hours after three hydrogen substitutions. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure and dried by spin-drying to give the title compound (110 mg, yield: 91.9%) as a white solid. LCMS (ESI) [ m+h ] + =461.3.
Step 2 preparation of((6- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
Diethyl ((6- ((7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (120 mg,0.26mmol,1.0 eq) was dissolved in dichloromethane (2 mL), and trimethylbromosilane (600 mg,3.9mmol,15 eq) was added and stirred at 50 ℃ for 1 hour. The reaction solution was quenched with methanol and concentrated under reduced pressure to give a crude product as a brown oil. Preparation by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (36.29 mg, yield: 34.5%) as a white solid .LCMS(ESI):[M+H]+=405.0;1H NMR(400MHz,DMSO-d6,ppm)δ9.13(s,1H),8.16(s,1H),7.56(dd,J=10.0,2.0Hz,2H),7.19(d,J=8.0Hz,1H),7.09(t,J=10.0Hz,1H),5.66(s,2H),2.74(s,3H),2.53(d,J=20.8Hz,2H).
EXAMPLE 65 preparation of((6- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid
Step 1 preparation of diethyl ((6- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate:
Diethyl ((6- ((4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (200 mg,0.42mmol,1.0 eq) was dissolved in ethanol (3 mL), then palladium on carbon (100 mg,50 wt%) and potassium carbonate (288 mg,2.1mmol,5.0 eq) were added to this solution, and the reaction was stirred at room temperature under a hydrogen atmosphere for 2 hours after three hydrogen substitutions. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure and dried by spin-drying to give the title compound (150 mg, yield: 81.1%) as a colorless oil. LCMS (ESI) [ m+h ] + = 447.1.
Step 2 preparation of ((6- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonic acid:
Diethyl ((6- ((7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) pyridin-3-yl) methyl) phosphonate (150 mg,0.34mmol,1.0 eq) was dissolved in dichloromethane (2 mL) and trimethylbromosilane (772 mg,5.1mmol,15 eq) was added and the reaction stirred at 50℃for 1 hour. The reaction solution was quenched with methanol and concentrated under reduced pressure to give a crude product as a brown oil. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (56.91 mg, yield: 42.9%) as a white solid .LCMS(ESI):[M+H]+=391.0;1H NMR(400MHz,DMSO-d6,ppm)δ9.26(s,1H),9.07(s,1H),8.15(s,1H),7.60(dd,J=10.0,1.6Hz,1H),7.52(d,J=8.0Hz,1H),7.27(d,J=8.0Hz,1H),7.17-7.12(m,1H),5.72(s,2H),2.58(d,J=20.8Hz,2H);
19F NMR(376.5MHz,DMSO-d6,ppm)δ-105.51,-111.38。
EXAMPLE 66 preparation of (4- ((7, 9-difluoro-4-hydroxy-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
4-Chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (180 mg,0.7mmol,1 eq.) was dissolved in N, N-dimethylformamide (5 mL), diethyl (4- (bromomethyl) benzyl) phosphonate (4576 mg,1.4mmol,2 eq.) and potassium carbonate (196 mg,1.4mmol,2 eq.) were added, and after stirring for 1 hour under argon at 70℃the reaction solution was quenched with saturated ammonium chloride solution, extracted three times with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (145 mg, yield: 41.4%) as a yellow liquid. LCMS (ESI) [ m+h ] + =494.0.
Step 2 preparation of (4- ((7, 9-difluoro-4-hydroxy-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (145 mg,0.29mmol,1 eq) was dissolved in dichloromethane (5 mL), trimethylbromosilane (1 mL,6.4mmol,22 eq) was added and stirred for 1 hour at 50 ℃ under argon. The reaction solution was purified by reverse phase preparation (C18, 10 mmol/ammonia, acetonitrile) to give the title compound (4.38 mg, yield: 15.9%) as a white solid .LCMS(ESI):[M+H]+=420.1;1H NMR(400MHz,CD3OD)δ7.27(d,J=6.4Hz,2H),7.06(d,J=7.6Hz,2H),6.93(d,J=9.6Hz,1H),6.61(t,J=10.4Hz,1H),5.91(s,2H),2.80(d,J=19.6Hz,2H),2.47(s,3H).
EXAMPLE 67 preparation of (4- ((4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 5, 6-difluoro-1H-indole-2-carboxylic acid methyl ester
5, 6-Difluoro-1H-indole-2-carboxylic acid (10 g,40.31mmol,1 eq.) was dissolved in methanol (100 mL) and thionyl chloride (9.59 g,80.62mmol,2 eq.) was added dropwise. The reaction was continued for 3 hours after the reaction was warmed to 60 ℃. The reaction solution was concentrated, neutralized with saturated aqueous sodium hydrogencarbonate, suction filtered, water washed, and dried under vacuum to give the title compound (6.0 g, yield) :70.49%).1H NMR(400MHz,DMSO-d6)δ12.14(s,1H),7.68(dd,J=11.1,8.1Hz,1H),7.36(ddd,J=11.0,7.0,0.9Hz,1H),7.16(dd,J=2.2,0.9Hz,1H),3.88(s,3H).
Step 2 preparation of 5, 6-difluoro-3-nitro-1H-indole-2-carboxylic acid methyl ester
Concentrated nitric acid (8 mL) was added dropwise to acetic anhydride (72 mL) in an ice bath, stirring was continued for 15 minutes after the addition was completed, 5, 6-difluoro-1H-indole-2-carboxylic acid methyl ester (5.1 g,24.15mmol,1 eq.) was dissolved in acetic anhydride (10 mL), and the nitric acid/acetic anhydride mixed solution prepared above was added dropwise in an ice bath. Stirring was continued at this temperature for 3 hours after the addition was completed. The reaction mixture was poured into ice water to precipitate a large amount of solid, which was suction-filtered, washed with water and dried to give the crude title compound (6.4 g, yield 100%) as a yellow solid .1H NMR(400MHz,DMSO-d6)δ13.61(s,1H),7.95(dd,J=10.7,7.8Hz,1H),7.66(dd,J=10.3,6.9Hz,1H),3.98(s,3H).
Step 3 preparation of 5, 6-difluoro-3-nitro-1H-indole-2-carboxamide:
Methyl 5, 6-difluoro-3-nitro-1H-indole-2-carboxylate (6.4 g,24.98mmol,1 eq.) was dissolved in a mixed solution of ammonia/dioxane (60.0 mL/60.0 mL) and reacted at 115℃for 3 hours in a closed jar. The reaction mixture was concentrated under reduced pressure and dried to give the title compound (6.0 g, yield 99.6%).1H NMR(400MHz,DMSO-d6)δ13.32(s,1H),8.33(s,1H),8.06(s,1H),7.94(dd,J=11.0,8.0Hz,1H),7.55(dd,J=10.5,6.9Hz,1H).
Step 4 preparation of 3-amino-5, 6-difluoro-1H-indole-2-carboxamide
5, 6-Difluoro-3-nitro-1H-indole-2-carboxamide (6.0 g,24.88mmol,1 eq.) was dissolved in methanol (120 mL), pd/C (600 mg,10 wt%) was added thereto under nitrogen atmosphere, and the reaction mixture was stirred at room temperature under hydrogen atmosphere (1 atm) for 16 hours. The reaction mixture was diluted with methanol (500 mL) and filtered through silica gel, and the filtrate was dried by spin-drying to give the crude title compound (5.5 g, yield > 100%). LCMS (ESI) [ m+h ] + =212.1.
Step 5 preparation of 7, 8-difluoro-5H-pyrimido [5,4-b ] indol-4-ol:
3-amino-5, 6-difluoro-1H-indole-2-carboxamide (2 g,9.47mmol,1 eq.) was dissolved in ethanol (20 mL) and formamidine acetate (2 g,18.9mmol,2 eq.) was added and the reaction mixture reacted at 90℃for 2 hours. The reaction was cooled to room temperature, filtered, and the filter cake was rinsed with ethanol, and the solid was collected and dried to give the title compound (1.6 g, 76.4% yield). LCMS (ESI) [ m+h ] + =222.
Step 6 preparation of 4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indole
7, 8-Difluoro-5H-pyrimido [5,4-b ] indol-4-ol (1.6 g,7.23mmol,1 eq.) was added to phenylphosphoryl dichloride (20 mL), the reaction was warmed to 180℃and stirred for 4 hours. The reaction solution was diluted with ethyl acetate (20 mL), neutralized with sodium hydrogencarbonate, extracted three times with ethyl acetate (30 mL. Times.3), and concentrated to give the title compound (300 mg, yield: 62.7%). LCMS (ESI) [ m+h ] + =240.0.
Step 7 preparation of di-tert-4- ((4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
After 4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indole (100 mg,0,42mmol,1 eq.) was dissolved in acetonitrile (10 mL), di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (315 mg,0.83mmol,2 eq.) and N, N-diisopropylethylamine (1.50 mL,11.61mmol,27.64 eq.) were added and stirred at 100℃for 2 hours. The reaction solution was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (20 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (200 mg, yield: 88.9%). LCMS (ESI) [ m+h ] + = 424.0.
Step 8 preparation of (4- ((4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-ester of (4- ((4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.37mmol,1 eq.) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (2.00 mL,17.54mmol,47.41 eq.) was added and reacted at 50 ℃ for 2 hours. The reaction solution was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (38 mg, yield: 24.3%) as a white solid .LCMS(ESI):[M+H]+=424.0;1H NMR(400MHz,CD3OD)δ8.83(s,1H),8.24(t,J=8.0Hz,1H),7.66(dd,J=12.0,8.0Hz,1H),7.22(dd,J=8.0,2.4Hz,2H),7.02(d,J=8.0Hz,2H),6.01(s,2H),3.04(d,J=20.4Hz,2H).
EXAMPLE 68 preparation of (4- ((4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-4-ol:
3-amino-5, 6-difluoro-1H-indole-2-carboxamide (500 mg,2.37mmol,1 eq.) was dissolved in acetonitrile (5 mL), hydrogen chloride (2.5 mL,4 mol/L dioxane solution) was added, the vessel was closed, and the temperature was raised to 120℃and the reaction was stirred for 2 hours. The reaction solution was filtered, and the solid was dried to give the title compound (500 mg, yield: 89.8%). LCMS (ESI) [ m+h ] + =236.1.
Step 2 preparation of 4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole:
7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-4-ol (500 mg,2.26mmol,1 eq.) was dissolved in phenylphosphoryl dichloride (10 mL). The reaction was carried out at 180℃for 1 hour. The reaction solution was cooled, neutralized with saturated sodium bicarbonate, extracted three times with ethyl acetate (20 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (500 mg, yield: 92.3%). LCMS (ESI) [ m+h ] + = 253.9.
Step 3 preparation of di-tert-4- ((4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (200 mg,0.79mmol,1 eq.) was dissolved in N, N-dimethylformamide (10 mL), di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (892 mg,2.37mmol,3 eq.) and potassium carbonate (327 mg,2.37mmol,3 eq.) were added and the reaction stirred at 70℃for 1 hour. The reaction solution was diluted with saturated ammonium chloride, ethyl acetate (10 mL. Times.3) was added thereto, and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (200 mg, yield: 46.1%)
LCMS(ESI):[M+H]+=438.0。
Step 4 preparation of (4- ((4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
di-tert-butyl (4- ((4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (100 mg,0.18mmol,1 eq) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (0.5 mL,4.38mmol,24.4 eq) was added and the reaction stirred at room temperature for 1 hour. The reaction solution was quenched with methanol, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (15 mg, yield) :16.7%).LCMS(ESI):[M+H]+=436.0;1H NMR(400MHz,CD3OD)δ8.22-8.18(m,1H),7.62(dd,J=11.2,6.4Hz,1H),7.26-7.24(m,2H),6.96(d,J=8.0Hz,2H),5.92(s,2H),2.92(d,J=21.0Hz,2H),2.80(s,3H).
Example 69 preparation of (4- ((7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of di-tert-butyl (4- ((7, 8-difluoro-3, 4-dihydro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
Di-tert-butyl (4- ((4-chloro-7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.37mmol,1 eq.) was dissolved in anhydrous methanol (20 mL), palladium on carbon (40 mg,20% (weight percent)) was added, hydrogen was replaced three times, and the reaction was stirred at room temperature for 2 hours. The reaction solution was filtered to remove palladium on carbon, and the filtrate was concentrated in vacuo to give the title compound (150 mg, yield: 81.1%). LCMS (ESI) [ m+h ] + =504.2.
Step 2 preparation of di-tert-butyl 4- ((7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
Di-tert-butyl (4- ((7, 8-difluoro-3, 4-dihydro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (150 mg,0.28mmol,1 eq) was dissolved in dichloromethane (20 mL), manganese dioxide (97 mg,1.12mmol,4 eq) was added and the reaction stirred at room temperature for 2 hours. Manganese dioxide solid was removed by filtration, and the filtrate was concentrated under reduced pressure to give the title compound (120 mg,0.24mmol, 85.49%). LCMS (ESI) [ m+h ] + =502.2.
Step 3 preparation of (4- ((7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((7, 8-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phosphonate (120 mg,0.24mmol,1 eq.) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL,17.5mmol,73 eq.) was added and reacted for 2 hours at 50 ℃. The reaction solution was concentrated and prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (27 mg, yield: 28.9%) as a white solid .LCMS(ESI):[M+H]+=390.0;1H NMR(400MHz,CD3OD)δ9.14(s,1H),9.04(s,1H),8.26-8.19(m,1H),7.76-7.67(m,1H),7.30-7.27(m,2H),7.14-7.07(m,2H),5.67(s,2H),2.90(d,J=20.9Hz,2H).
Example 70 preparation of (4- ((7, 9-difluoro-4-methoxy-2-methyl-5-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-4-methoxy-2-methyl-5-pyrimido [5,4-b ] indole:
4-chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (300 mg,1.18mmol,1 eq.) was dissolved in anhydrous methanol (5 mL) and sodium methoxide (96 mg,1.77mmol,1.5 eq.) was added and reacted at 70℃for 1 hour under argon. The reaction solution was quenched with saturated ammonium chloride (10 mL), extracted three times with ethyl acetate (10 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (200 mg, yield: 67.9%) as a yellow solid. LCMS (ESI) [ m+h ] + =250.0.
Step 2 preparation of diethyl (4- ((7, 9-difluoro-4-methoxy-2-methyl-5-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
7, 9-difluoro-4-methoxy-2-methyl-5-pyrimido [5,4-b ] indole (100 mg,0.4mmol,1 eq.) was dissolved in N, N-dimethylformamide (5 mL) and potassium carbonate (111 mg,0.8mmol,2 eq.) and diethyl (4- (bromomethyl) phosphonate (515 mg,1.61mmol,4 eq.) were added and reacted at 70℃for 2 hours under argon. The reaction solution was quenched with saturated ammonium chloride (10 mL), extracted three times with ethyl acetate (10 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (180 mg, yield: 91.7%) as a yellow solid. LCMS (ESI) [ m+h ] + = 490.2.
Step 3 preparation of (4- ((7, 9-difluoro-4-methoxy-2-methyl-5-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((7, 9-difluoro-4-methoxy-2-methyl-5-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (100 mg,0.2mmol,1 eq) was dissolved in dichloromethane (2 mL) and HMDS (hexamethyldisilazane) (989 mg,6.13mmol,30 eq) and trimethylbromosilane (1 mL) were added and stirred at 50 ℃ for 16 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (3.26 mg, yield: 3.8%) as a white solid .LCMS(ESI):[M-H]-=432.1;1H NMR(400MHz,CD3OD)δ7.30(dd,J=8.4,1.6Hz,2H),7.22(dd,J=9.6,1.6Hz,1H),7.01(d,J=8.0Hz,2H),6.84(t,J=10.4Hz,1H),5.71(s,2H),4.18(s,3H),2.81(d,J=19.6Hz,2H),2.72(s,3H).
Example 71 preparation of (4- ((7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((7, 8-difluoro-2-methyl-3, 4-dihydro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((4-chloro-7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (300 mg,0.61mmol,1 eq) was dissolved in methanol (10 mL), palladium on carbon (130 mg,43% (weight percent)) was added and stirred under hydrogen at room temperature for 1 hour. The organic phase was filtered and concentrated to give the title compound (200 mg, yield: 71.1%). LCMS (ESI) [ m+h ] + = 462.3.
Step 2 preparation of diethyl (4- ((7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((7, 8-difluoro-2-methyl-3, 4-dihydro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.43mmol,1 eq) was dissolved in dichloromethane (5 mL), manganese dioxide (75 mg,0.87mmol,2 eq) was added and stirred at room temperature for 1 hour. The reaction solution was filtered, washed with methylene chloride, and the organic phase was concentrated to give the title compound (150 mg, yield: 75.9%). LCMS (ESI) [ m+h ] + =460.1.
Step 3 preparation of (4- ((7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((7, 8-difluoro-2-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (90 mg,0.2mmol,1 eq) was dissolved in dichloromethane (2 mL), then trimethylbromosilane (0.15 mL,0.98mmol,5 eq) was added and the reaction warmed to 50 ℃ and stirred for 1 hour. The reaction solution was quenched with methanol, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (6 mg, yield) :7.6%).LCMS(ESI):[M+H]+=404.0;1H NMR(400MHz,CD3OD)δ9.01(s,1H),8.20(dd,J=10.0,8.0Hz,1H),7.67(dd,J=10.0,8.0Hz,1H),7.27(dd,J=8.0,2.0Hz,2H),7.10(d,J=8.0Hz,2H),5.63(s,2H),2.92(d,J=20.8Hz,2H),2.84(s,3H).
Example 72 preparation of (4- ((7, 9-difluoro-2, 4-dimethyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-2, 4-dimethyl-5H-pyrimido [5,4-b ] indole
4-Chloro-7, 9-difluoro-2-methyl-5H-pyrimido [5,4-b ] indole (200 mg,0.79mmol,1 eq.) and methylboronic acid (47 mg,0.79mmol,1 eq.) were dissolved in1, 4-dioxane (3 mL), 1-bis (diphenylphosphino) ferrocene palladium dichloride (58 mg,0.08mmol,0.1 eq.) and cesium carbonate (771 mg,2.37mmol,3 eq.) were added and the reaction solution heated to 120℃and stirred for 2 hours. The reaction solution was quenched with saturated aqueous ammonium chloride (80 mL), the mixture was extracted with ethyl acetate (80 ml×3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by thin layer preparative plate chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (180 mg, yield: 97.9%) as a brown solid. LCMS (ESI) [ m+h ] + = 234.0.
Step 2 preparation of diethyl (4- ((7, 9-difluoro-2, 4-dimethyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
7, 9-Difluoro-2, 4-dimethyl-5H-pyrimido [5,4-b ] indole (150 mg,0.64mmol,1 eq.) was dissolved in tetrahydrofuran (2 mL), sodium hydride (77 mg,1.92mmol,3 eq.) was added and the reaction was stirred for 0.5 hours, then diethyl (4- (bromomethyl) benzyl) phosphonate (308 mg,0.96mmol,1.5 eq.) was added to the mixture and the reaction was stirred for 2 hours. The reaction solution was diluted with saturated aqueous ammonium chloride (50 mL), extracted with ethyl acetate (50 ml×3), the organic phases were combined, washed with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by thin layer chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (100 mg, yield: 33.0%) as a brown solid. LCMS (ESI) [ m+h ] + =474.1.
Step 3 preparation of (4- ((7, 9-difluoro-2, 4-dimethyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((7, 9-difluoro-2, 4-dimethyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (100 mg,0.21mmol,1 eq) was dissolved in dichloromethane (2 mL), then trimethylbromosilane (647 mg,4.22mmol,20 eq) was added to this mixture and the reaction was heated to 50 ℃ and stirred for 2 hours. Water (1 mL) was added to the reaction mixture, and the aqueous phase was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) and purified to give the title compound (22 mg, yield: 25.0%) as a white solid .LCMS(ESI):[M+H]+=418.1;1H NMR(400MHz,CD3OD,ppm)δ7.27(dd,J=8.0,2.0Hz,2H),7.21(dd,J=9.6,1.6Hz,1H),6.92(td,J=10.0,2.0Hz,1H),6.87(d,J=8.0Hz,2H),5.79(s,2H),2.96(d,J=20.8Hz,2H),2.79(s,3H),2.78(s,3H).
EXAMPLE 73 preparation of (4- ((7, 9-difluoro-4-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-4-methyl-5H-pyrimido [5,4-b ] indole
4-Chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (300 mg,1.25mmol,1 eq.) and methylboronic acid (90 mg,1.50mmol,1.2 eq.) were dissolved in1, 4-dioxane (3 mL), and to this mixture 1, 1-bis (diphenylphosphino) ferrocene palladium dichloride (92 mg,0.13mmol,0.1 eq.) and cesium carbonate (1.22 g,3.75mmol,3 eq.) were added, the reaction solution was heated to 120℃and stirred for 2 hours. The reaction solution was diluted with saturated aqueous ammonium chloride (80 mL), the mixture was extracted with ethyl acetate (80 ml×3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by thin layer preparative plate chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (80 mg, yield: 29.2%) as a brown solid. LCMS (ESI) [ m+h ] + =220.0.
Step 2 preparation of diethyl (4- ((7, 9-difluoro-4-methyl-5-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
7, 9-Difluoro-4-methyl-5H-pyrimido [5,4-b ] indole (70 mg,0.32mmol,1 eq.) was dissolved in tetrahydrofuran (2 mL), sodium hydride (38 mg,0.96mmol,3 eq.) was added and the reaction was stirred for 0.5 hours, then diethyl (4- (bromomethyl) benzyl) phosphonate (123 mg,0.38mmol,1.2 eq.) was added to the mixture and the reaction was stirred for 2 hours. The reaction solution was diluted with saturated aqueous ammonium chloride (30 mL), extracted with ethyl acetate (30 ml×3), and the organic phases were combined, washed with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by thin layer preparative plate chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (60 mg, yield: 40.8%) as a brown solid. LCMS (ESI) [ m+h ] + =460.1.
Step 3 preparation of (4- ((7, 9-difluoro-4-methyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((7, 9-difluoro-4-methyl-5-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (60 mg,0.13mmol,1 eq) was dissolved in dichloromethane (1 mL), trimethylbromosilane (400 mg,2.61mmol,20 eq) was added and the reaction was heated to 50 ℃ and stirred for 2 hours. Water (1 mL) was added to the reaction mixture, and the aqueous phase was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) and purified to give the title compound (10 mg, yield: 19.0%) as a white solid .LCMS(ESI):[M+H]+=404.0;1H NMR(400MHz,CD3OD)δ8.88(s,1H),7.29-7.25(m,3H),6.97(td,J=10.0,2.0Hz,1H),6.88(d,J=7.9Hz,2H),5.83(s,2H),2.93(d,J=20.8Hz,2H),2.85(s,3H).
EXAMPLE 74 preparation of (4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-4-ol
3-Amino-4, 6-difluoro-1H-indole-2-carboxamide (20 g,94.7mmol,1 eq.) was dissolved in triethyl orthopropionate (200 mL), and the reaction was heated to 150℃and stirred for 16 hours. The reaction solution was directly filtered, and the filtered solid was dried to give the crude title compound (16.0 g, yield: 67.8%). LCMS (ESI) [ m+h ] + =250.0.
Step 2 preparation of 4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indole
2-Ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-4-ol (16 g,64.2mmol,1 eq.) is dissolved in phenylphosphoryl dichloride (100 mL), the reaction mixture is heated to 180℃and stirred for 5 hours. The reaction solution was quenched with saturated aqueous sodium bicarbonate (200 mL), the mixture was extracted three times with dichloromethane (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure and dried by rotation to give the crude title compound (14.0 g, yield: 81.5%). LCMS (ESI) [ m+h ] + = 268.0.
Step 3 preparation of di-tert-butyl 4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
After 4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (200 mg,0.75mmol,1 eq.) was dissolved in N, N-dimethylformamide (10 mL), di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (563 mg,1.50mmol,2 eq.) and potassium carbonate (310 mg,2.25mmol,3 eq.) were added and the reaction mixture was heated to reflux for 2 hours at 80℃and quenched with saturated ammonium chloride. The mixture was extracted three times with ethyl acetate (100 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (350 mg, yield: 82.9%). LCMS (ESI) [ m+h ] + = 564.1.
Step 4 preparation of (4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
di-tert-butyl (4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (150 mg,0.27mmol,1 eq.) was dissolved in dichloromethane (3 mL) and trifluoroacetic acid (1 mL,8.77mmol,33 eq.) was added and reacted for 2 hours at room temperature. The reaction solution was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia water, acetonitrile) to give the title compound (20 mg, yield: 16.4%) as a white solid .LCMS(ESI):[M+H]+=452.0;1H NMR(400MHz,CD3OD)δ7.29(d,J=8.4,2.0Hz,2H),7.20(d,J=9.6,2.0Hz,1H),7.01(d,J=7.6Hz,2H),6.83(dt,J=10.0,2.0Hz,1H),5.71(s,2H),3.00(q,J=7.6Hz,2H),2.81(d,J=20.0Hz,2H),1.41(t,J=7.6Hz,3H).
EXAMPLE 75 preparation of (4- ((4-chloro-7, 9-difluoro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-4-ol
3-Amino-4, 6-difluoro-1H-indole-2-carboxamide (5 g,23.7mmol,1 eq.) and 2, 6-dimethyl-3, 5-heptanedione (36 g,237mmol,10 eq.) were dissolved in ethanol (100 mL), phosphoric acid (10 mL) was slowly added dropwise, and the reaction was heated to 100℃and stirred for 3 days. The reaction solution was directly filtered, and the solid was dried to give the crude title compound (4.5 g, yield: 72.2%). LCMS (ESI) [ m+h ] + =264.0.
Step 2 preparation of 4-chloro-2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indole
2-Isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-4-ol (7 g,26.6mmol,1 eq.) was dissolved in phenylphosphoryl dichloride (70 mL), the reaction was heated to 180℃and stirred for 4 hours. The reaction solution was quenched with saturated aqueous sodium hydrogencarbonate (200 mL), extracted three times with ethyl acetate (200 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure and dried by rotation to give the crude title compound (6.0 g, yield: 81.1%). LCMS (ESI) [ m+h ] + = 282.0.
Step 3 preparation of di-tert-butyl (4- ((4-chloro-2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
After dissolving 4-chloro-2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (100 mg,0.35mmol,1 eq.) in N, N-dimethylformamide (10 mL), di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (263 mg,0.70mmol,2 eq.) and potassium carbonate (150 mg,1.06mmol,3 eq.) were added and stirred at 70℃for 2 hours. The reaction solution was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (50 mL. Times.3), and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (100 mg, yield: 48.8%). LCMS (ESI) [ m+h ] + =578.0.
Step 4 preparation of (4- ((4-chloro-7, 9-difluoro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Di-tert-butyl (4- ((4-chloro-2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (100 mg,0.17mmol,1 eq) was dissolved in dichloromethane (20 mL) and trifluoroacetic acid (2 mL) was added and the reaction was heated to 25℃and stirred for 2 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (6 mg, yield: 6.2%) as a white solid .LCMS(ESI):[M+H]+=466.0;1H NMR(400MHz,CD3OD)δ7.32(dd,J=8.4,1.6Hz,2H),7.26(dd,J=10.0,2.0Hz,1H),6.99-6.92(m,3H),5.89(s,2H),3.39-3.28(m,1H),2.82(d,J=19.6Hz,2H),1.41(d,J=6.8Hz,6H).
Example 76 preparation of (4- ((2-ethyl-7, 9-difluoro-4-hydroxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
4-Chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (200 mg,0.74mmol,1 eq.) was dissolved in N, N-dimethylformamide (10 mL), diethyl (4- (bromomethyl) benzyl) phosphonate (480 mg,1.49mmol,2 eq.) and potassium carbonate (306 mg,2.22mmol,3 eq.) were added and stirred at 80℃for 2 hours. The reaction solution was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (50 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated in vacuo to give the title compound (200 mg, yield: 52.7%). LCMS (ESI) [ m+h ] + = 508.0.
Step 2 preparation of (4- ((2-ethyl-7, 9-difluoro-4-hydroxy-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (100 mg,0.20mmol,1 eq) was dissolved in dichloromethane (20 mL) at room temperature and trimethylbromosilane (2 mL) was added and the reaction was heated to 50 ℃ and stirred for 4 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (48 mg, yield: 56.4%) as a white solid .LCMS(ESI):[M+H]+=434.0;1H NMR(400MHz,CD3OD)δ7.28(dd,J=8.4,2.0Hz,2H),7.07(d,J=8.0Hz,2H),6.93(dd,J=9.6,2.0Hz,1H),6.60(dt,J=10.0,2.0Hz,1H),5.92(s,2H),2.80(d,J=20.0Hz,2H),2.74(q,J=7.6Hz,2H),1.32(t,J=7.6Hz,3H).
Example 77 preparation of (4- ((7, 9-difluoro-4-hydroxy-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((7, 9-difluoro-4-chloro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
4-Chloro-2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (300 mg,1.07mmol,1 eq.) was dissolved in N, N-dimethylformamide (10 mL), diethyl (4- (bromomethyl) benzyl) phosphonate (683 mg,2.14mmol,2 eq.) and potassium carbonate (447 mg,3.21mmol,3 eq.) were added and stirred at 80℃for 2 hours. The reaction solution was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (50 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (360 mg, yield: 64.6%). LCMS (ESI) [ m+h ] + = 522.0.
Step 2 preparation of (4- ((7, 9-difluoro-4-hydroxy-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((7, 9-difluoro-4-chloro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.38mmol,1 eq) was dissolved in dichloromethane (20 mL), trimethylbromosilane (2 mL) was added and the reaction heated to 50 ℃ and stirred for 4 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (28 mg, yield: 16.3%) as a white solid .LCMS(ESI):[M+H]+=448.1;1H NMR(400MHz,CD3OD)δ7.27(d,J=6.8Hz,2H),7.06(d,J=7.6Hz,2H),6.93(d,J=9.2Hz,1H),6.60(t,J=10.0Hz,1H),5.92(s,2H),3.14-2.99(m,1H),2.80(d,J=19.6Hz,2H),1.33(d,J=7.2Hz,6H).
EXAMPLE 78 preparation of (4- ((2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((4-chloro-2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (180 mg,0.35mmol,1 eq) was dissolved in anhydrous methanol (5 mL), potassium carbonate (48 mg,0.35mmol,1 eq) and palladium on carbon (18 mg,10 wt%) were added, the reaction system was replaced with a hydrogen atmosphere, and the reaction mixture was stirred at room temperature for 1 hour. Dichloromethane (10 mL) was added to the reaction solution, which was filtered, and the filtrate was concentrated under reduced pressure to give the title compound (110 mg, yield: 66.4%). LCMS (ESI) [ m+h ] + =474.1.
Step 2 preparation of (4- ((2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2-ethyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (110 mg,0.23mmol,1 eq) was dissolved in dichloromethane (2 mL), trimethylbromosilane (0.5 mL,3.79mmol,16 eq) was added and the reaction was heated to 50 ℃ and stirred for 1 hour. Water (2 mL) was added to the reaction solution, and the mixture was filtered, and the solid was washed with tetrahydrofuran (2 mL), and the filtered and dried to give the title compound (37 mg, yield: 37.1%) as a white solid .LCMS(ESI):[M+H]+=418.0;1H NMR(400MHz,CD3OD)δ9.37(s,1H),7.47(dd,J=8.6,2.0Hz,1H),7.30(dd,J=8.4,2.4Hz,2H),7.22(d,J=8.0Hz,2H),7.13(td,J=10.0,2.0Hz,1H),5.77(s,2H),3.26(q,J=7.6Hz,2H),3.08(d,J=21.6Hz,2H),1.53(t,J=7.6Hz,3H).
Example 79 preparation of (4- ((7, 9-difluoro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((7, 9-difluoro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate
After dissolving diethyl (4- ((4-chloro-7, 9-difluoro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (150 mg,0.29mmol,1 eq) in tetrahydrofuran (10 mL), palladium on carbon (30 mg,20 wt%) and potassium carbonate (40 mg,0.29mmol,1 eq) were added and stirred at 50 ℃ for 2 hours. The reaction solution was filtered, and concentrated in vacuo to give the title compound (100 mg, yield: 71.0%). LCMS (ESI) [ m+h ] + = 488.0.
Step 2 preparation of (4- ((7, 9-difluoro-2-isopropyl-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2-isopropyl-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) phosphonate (100 mg,0.29mmol,1 eq) was dissolved in dichloromethane (20 mL), trimethylbromosilane (2 mL) was added, the reaction was heated to 50 ℃ and stirred for 4 hours, the reaction mixture was dried by spinning, and the crude product was slurried twice with tetrahydrofuran/water (V/v=1/1) and once with tetrahydrofuran to give the title compound (29 mg, yield: 23.2%) as a white solid .LCMS(ESI):[M+H]+=432.1;1H NMR(400MHz,CD3OD,ppm)δ8.99(s,1H),7.35-7.31(m,3H),7.08(d,J=8.0Hz,2H),6.89(td,J=10.0,2.0Hz,1H),5.59(s,2H),3.41-3.34(m,1H),2.82(d,J=19.6Hz,2H),1.41(d,J=7.2Hz,6H).
EXAMPLE 80 preparation of (4- ((8-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 8-chloro-5H-pyrido [3,2-b ] indole as starting material 80.LCMS(ESI)[M+1]+=387.1;1H NMR(400MHz,CD3OD)δ8.47(d,J=4.4,1.2Hz,1H),8.30(d,J=2.0Hz,1H),8.03(d,J=8.8,2.0Hz,1H),7.63(d,J=8.8Hz,1H),7.52-7.48(m,2H),7.24(d,J=8.0,2.0Hz,2H),7.06(d,J=8.0Hz,2H),5.61(s,2H),2.88(d,J=20.8Hz,2H).
EXAMPLE 81 preparation of (4- ((8-bromo-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 8-bromo-5H-pyrido [3,2-b ] indole as starting material 81.LCMS(ESI)[M+1]+=430.9;1H NMR(400MHz,CD3OD)δ8.48-8.45(m,2H),8.04(dd,J=8.4,1.2Hz,1H),7.66(dd,J=8.8,2.0Hz,1H),7.58(d,J=8.8Hz,1H),7.50(dd,J=8.4,4.8Hz,1H),7.23(dd,J=8.0,2.0Hz,2H),7.08(d,J=8.0Hz,2H),5.61(s,2H),2.94(d,J=21.2Hz,2H).
EXAMPLE 82 preparation of (4- ((4-chloro-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 1 was carried out following steps 3-4 of the preparation method of example 1 using commercially available 4-chloro-9H-pyrimido [4,5-b ] indole as starting material 82.LCMS(ESI)[M+1]+=388.0;1H NMR(400MHz,DMSO-d6)δ.89(s,1H),8.35(d,J=8.0Hz,1H),7.83(d,J=8.4Hz,1H),7.67(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.21(d,J=8.0Hz,2H),7.16-7.14(m,2H),5.72(s,2H),2.88(d,J=21.4Hz,2H).
EXAMPLE 83 preparation of (4- ((4-bromo-6-chloro-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 4, 6-dichloro-9H-pyrimido [4,5-b ] indole
4-Chloro-9H-pyrimido [4,5-b ] indole (200 mg,1.0mmol,1 eq.) was dissolved in a mixed solution of dichloromethane (10 mL) and acetic acid (10 mL), N-chlorosuccinimide (131 mg,1.0mmol,1 eq.) was added, and after stirring for 30 minutes under argon atmosphere at 60℃the reaction solution was quenched with saturated sodium bicarbonate solution, extracted three times with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (230 mg, yield: 98.6%) as a yellow liquid. LCMS (ESI) [ m+h ] + =237.9.
Step 2 preparation of diethyl (4- ((4, 6-dichloro-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonate
4, 6-Dichloro-9H-pyrimido [4,5-b ] indole (140 mg,0.6mmol,1 eq.) was dissolved in N, N-dimethylformamide (15 mL), diethyl (4- (bromomethyl) benzyl) phosphonate (378 mg,1.2mmol,2 eq.) and potassium carbonate (122 mg,0.9mmol,1.5 eq.) were added, the reaction was quenched with saturated ammonium chloride solution after stirring for 1 hour at 70℃under argon atmosphere, extracted three times with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (170 mg, yield: 60.4%) as a yellow liquid. LCMS (ESI) [ m+h ] + = 478.0.
Step 3 preparation of (4- ((4-bromo-6-chloro-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((4, 6-dichloro-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonate (90 mg,0.2mmol,1 eq) was dissolved in dichloromethane (5 mL) and trimethylbromosilane (1 mL,6.5mmol,33 eq) was added and stirred under argon atmosphere at 50℃for 1 hour. The reaction solution was purified by reverse phase HPLC (C18, 10 mmol/ammonia, acetonitrile) to give the title compound (7 mg, yield: 10.5%) as a white solid .LCMS(ESI):[M-H]+=463.9;1H NMR(400MHz,CD3OD)δ8.76(s,1H),8.48(d,J=2.4Hz,1H),7.67(d,J=8.8Hz,1H),7.60(dd,J=8.8,2.0Hz,1H),7.27-7.24(m,2H),7.18(d,J=8.0Hz,2H),5.69(s,2H),2.89(d,J=20.8Hz,2H).
EXAMPLE 84 preparation of (4- ((6-chloro-4-hydroxy-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of (4- ((6-chloro-4-hydroxy-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((4, 6-dichloro-9H-pyrimido [4,5-b ] indol-9-yl) methyl) benzyl) phosphonate (80 mg,0.2mmol,1 eq) was dissolved in dichloromethane (5 mL), trimethylbromosilane (53. Mu.L, 0.4mmol,2 eq) was added and stirred under argon atmosphere at 50℃for 1 hour. The reaction mixture was directly purified by reverse phase HPLC (C18, 10 mmol/ammonia, acetonitrile) to give the title compound (12.9 mg, yield: 17.7%) as a white solid .LCMS(ESI):[M+H]+=404.0;1H NMR(400MHz,CD3OD)δ8.21(s,1H),8.10(d,J=2.0Hz,1H),7.50(d,J=5.2Hz,1H),7.33(dd,J=8.4,2.0Hz,1H),7.26-7.22(m,2H),7.16(d,J=8.4Hz,2H),5.60(s,2H),2.91(d,J=21.0Hz,2H).
EXAMPLE 85 preparation of (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 2-chloro-6- (2-nitrophenyl) pyridine:
After (2-nitrophenyl) boric acid (2 g,12.0mmol,1 eq.) was dissolved in 1,4 dioxane (5 mL), 2, 6-dichloropyridine (1.7 g,12.0mmol,1 eq.) and potassium carbonate (3.3 g,24.0mmol,2 eq.) were added and reacted for 1 hour with microwaves at 120 ℃. The reaction mixture was quenched with saturated ammonium chloride (10 mL), and extracted three times with ethyl acetate (10 mL. Times.3). The organic phase was dried over anhydrous sodium sulfate, concentrated in vacuo to give a crude product, which was purified by flash chromatography (silica gel, petroleum ether: dichloromethane (V/v=1/1)) to give the title compound (200 mg, yield: 7.1%). LCMS (ESI) [ m+h ] + =235.0.
Step 2 preparation of 2-chloro-5H-pyrido [3,2-b ] indole:
2-chloro-6- (2-nitrophenyl) pyridine (400 mg,1.7mmol,1 eq.) was dissolved in triethyl phosphite (4 mL), raised to 180℃and reacted under nitrogen atmosphere for 10 hours, and the reaction solution was concentrated to give the title compound (200 mg, yield: 57.9%). LCMS (ESI) [ M-H ] - = 201.0.
Step 3 preparation of diethyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
After 2-chloro-5H-pyrido [3,2-b ] indole (40 mg,0.2mmol,1 eq.) was dissolved in tetrahydrofuran (1 mL), sodium hydride (32 mg,0.8mmol,4 eq.) was added, diethyl (4- (bromomethyl) benzyl) phosphonate (126..8 mg,0.4mmol,2 eq.) was added and reacted at room temperature for 2 hours. The reaction solution was quenched with saturated ammonium chloride (10 mL), extracted three times with ethyl acetate (10 ml×3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/2)) to give the title compound ((30 mg, yield: 68.6%). LCMS (ESI): m+h ] + = 443.0.
Step 4 preparation of (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (30 mg,0.07mmol,1 eq) was dissolved in dichloromethane (1 mL), trimethylbromosilane (103 mg,0.68mmol,10 eq) was added and the temperature was raised to 50℃for 1 hour. The reaction solution was concentrated under reduced pressure, and the crude product was prepared in reverse phase (C18, 10 mmol/ammonia water, acetonitrile) to give the title compound (2.57 mg, yield) :9.5%).LCMS(ESI):[M+H]+=387.0;1H NMR(400MHz,CD3OD)δ8.29(d,J=8.0Hz,1H),7.98(d,J=8.4Hz,1H),7.65(d,J=8.4Hz,1H),7.58-7.55(m,1H),7.42(d,J=8.8Hz,1H),7.32-7.29(m,2H),7.16(d,J=8.0Hz,1H),7.02(d,J=8.0Hz,2H),5.57(s,2H),2.82(d,J=19.8Hz,2H).
EXAMPLE 86 preparation of (4- ((2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 6-methyl-3-nitro-2-phenylpyridine
6-Chloro-3-nitro-2-phenylpyridine (450 mg,1.9mmol,1 eq), methylboronic acid (231 mg,3.8mmol,2 eq), 1-bis (diphenylphosphine) ferrocene palladium dichloride (418 mg,0.58mmol,0.3 eq) and cesium carbonate (1.88 g,5.7mmol,3 eq) were dissolved in a mixed solvent of dioxane (30 mL) and water (6 mL), and the mixture was heated to 80℃under argon atmosphere, and stirred for 2 hours. The reaction solution was cooled and then quenched with water, the mixture was extracted three times with methylene chloride (100 mL. Times.3), the organic phases were combined, washed with a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L pure water, acetonitrile) to give the title compound (300 mg, yield: 73.0%) as a white solid. LCMS (ESI) [ m+h ] + =215.0.
Step 2 preparation of 2-methyl-5H-pyrido [3,2-b ] indole
6-Methyl-3-nitro-2-phenylpyridine (300 mg,1.4mmol,1 eq.) was dissolved in o-dichlorobenzene (5 mL), triphenylphosphine (734.60 mg,2.8mmol,2 eq.) was added to the mixture, and the reaction was continued under 200℃inert gas for 2 hours. After the reaction solution was concentrated under reduced pressure, the crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (200 mg, yield: 61.8%) as a white solid. LCMS (ESI) [ m+h ] + =183.1.
Step 3 preparation of diethyl (4- ((2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
2-methyl-5H-pyrido [3,2-b ] indole (110 mg,0.6mmol,1.0 eq.) was dissolved in tetrahydrofuran (5 mL), and sodium hydride (29 mg,0.72mmol,1.2 eq.) was added to the reaction solution at 0deg.C. The reaction solution was stirred at 0℃for 20 minutes, and diethyl (4- (bromomethyl) benzyl) phosphonate (232 mg,0.73mmol,1.2 eq) was then added to the mixture. The reaction solution was stirred at room temperature for two hours. The reaction solution was quenched with water and extracted three times with ethyl acetate (100 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (110 mg, yield: 43.1%) as a yellow solid. LCMS (ESI) [ m+h ] + =423.1.
Step 4 preparation of (4- ((2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (110 mg,0.26mmol,1 eq) was dissolved in dichloromethane (20 mL) and trimethylbromosilane (2 mL) was added and the reaction was heated to 50℃and stirred for 2 hours. The reaction solution was concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (27.8 mg, yield: 29.1%) as a white solid .LCMS(ESI):[M+H]+=367.0;1H NMR(400MHz,CD3OD)δ8.37(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.62(d,J=8.4Hz,1H),7.58-7.54(m,1H),7.39(d,J=8.8Hz,1H),7.33-7.29(m,1H),7.23(dd,J=8.4,2.4Hz,2H),7.06(d,J=8.0Hz,2H),5.61(s,2H),2.91(d,J=20.8Hz,2H),2.74(s,3H).
Example 87 (4- ((2-methoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of di-tert-butyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
2-Chloro-5H-pyrido [3,2-b ] indole (1.0 g,5.0mmol,1.0 eq.) was dissolved in N, N-dimethylformamide (5 mL), sodium hydride (240 mg,6mmol,1.2 eq.) was added to the reaction solution at 0℃and the reaction stirred at 0℃for 30 minutes, to which mixture di-tert-butyl (4- (bromomethyl) benzyl) phosphonate (1.9 g,5mmol,1.0 eq.) was added. Stirring was continued for two hours, quenched with water and extracted three times with ethyl acetate (100 ml×3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) to give the title compound (1.0 g, yield: 41.7%) as a yellow solid. LCMS (ESI) [ m+na ] + =520.8.
Step 2 preparation of di-tert-4- ((2-methoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
Di-tert-butyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.4mmol,1.0 eq) was dissolved in toluene (1 mL) and methanol (1 mL), followed by cesium carbonate (393 mg,1.2mmol,3.0 eq), palladium acetate (9 mg,0.04mmol,0.1 eq) and 2-di-tert-butyl phosphino-2 ',4',6' -triisopropylbiphenyl (34 mg,0.08mmol,0.2 eq). The reaction was continued for 2 hours at 100℃under inert gas. Saturated aqueous sodium chloride (20 mL) was added to the reaction solution, and extracted three times with ethyl acetate (30 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=2/3)) to give the title compound (60 mg, yield: 30.3%) as a yellow oily liquid.
Step 3 (4- ((2-methoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((2-methoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) phosphonate (65 mg,0.13mmol,1.0 eq) was dissolved in methanol (75 mL), and a methanol solution of hydrogen chloride (14.2 mg,0.39mmol,3.0 eq) was added to the mixture, and the reaction was continued at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to obtain a crude product. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (28 mg yield: 56.3%) as a white solid .LCMS(ESI):[M+H]+=383.3;1H NMR(400MHz,DMSO-d6,ppm)δ8.09(d,J=8.4Hz,1H),7.99(d,J=8.8Hz,1H),7.63(d,J=8.4Hz,1H),7.43(t,J=7.6Hz,1H),7.20(t,J=7.2Hz,1H),7.06(d,J=6.4Hz,2H),6.97(d,J=8.0Hz,2H),6.86(d,J=8.8Hz,1H),5.55(s,2H),3.97(s,3H),2.62(d,J=20.8Hz,2H).
Example 88 preparation of (4- ((2- (methylamino) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of di-tert-butyl 4- ((2- (methylamino) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
Di-tert-butyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (500 mg,1.00mmol,1.0 eq.) was dissolved in toluene (5 mL) and methylamine hydrochloride (338 mg,5.01mmol,5.0 eq.), cesium carbonate (979 mg,3.01mmol,3.0 eq.), palladium acetate (17 mg,0.010mmol,0.1 eq.) and 2-di-tert-butyl phosphino-2 ',4',6' -triisopropylbiphenyl (85 mg,0.200mmol,0.2 eq.) were added. The reaction was continued for 2 hours under an inert gas at 110 ℃. Saturated aqueous sodium chloride (10 mL) was added to the reaction solution, and extracted three times with ethyl acetate (20 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, ethyl acetate (100%)) to give the title compound (75 mg, yield: 19.0%) as a yellow oily liquid. LCMS (ESI) [ m+h ] += 494.3.
Step 2 preparation of (4- ((2- (methylamino) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((2- (methylamino) -5H-pyrido [3,2-b ] indol-5-yl) methyl) phosphonate (75 mg,0.152mmol,1.0 eq) was dissolved in hydrogen chloride in methanol (2 mL,3 mol in methanol) and stirred at 50 ℃ C. For 4 hours, the reaction was concentrated under reduced pressure to give crude product as a brown oil which was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (15 mg, yield: 28.1%) as a white solid .LCMS(ESI):[M+H]+=382.3;1H NMR(400MHz,DMSO-d6,ppm)δ8.00(d,J=8.0Hz,1H),7.73(d,J=8.8Hz,1H),7.53(d,J=8.4Hz,1H),7.34(t,J=7.6Hz,1H),7.14-7.05(m,3H),6.98(d,J=8.0Hz,2H),6.59(d,J=8.8Hz,1H),6.25(brs,1H),5.48(s,2H),2.88(s,3H),2.72(d,J=21.1Hz,2H).
Example 89 preparation of (4- ((8-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 2- (3-fluorophenyl) -3-nitropyridine
2-Chloro-3-nitropyridine (2 g,9.9mmol,1 eq.) was dissolved in a mixed solution of 1, 4-dioxane (15 mL) and water (15 mL), and (3-fluorophenyl) boric acid (1.4 g,9.9mmol,1 eq.) and 1, 1-bis (diphenylphosphine) ferrocene palladium dichloride (0.7 g,0.9mmol,0.1 eq.) and potassium carbonate (2.0 g,14.8mmol,1.5 eq.) were added, and after stirring the reaction solution under argon for 1 hour at 120℃the reaction solution was quenched with saturated ammonium chloride solution, extracted three times with ethyl acetate (10 mL. Times.3) and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (2 g, yield: 93.1%) as a yellow liquid. LCMS (ESI) [ m+h ] + =219.0.
Step 2 preparation of 8-fluoro-5H-pyrido [3,2-b ] indole and 6-fluoro-5H-pyrido [3,2-b ] indole
2- (3-Fluorophenyl) -3-nitropyridine (2 g,9.2mmol,1 eq.) was dissolved in 1, 2-dichlorobenzene (40 mL), triphenylphosphine (4.8 g,18.3mmol,2 eq.) was added, after stirring the reaction solution under argon for 5 hours at 200℃the reaction solution was concentrated under reduced pressure and the crude product was purified by reverse phase HPLC preparation (C18, 10 mmol/ammonium bicarbonate, acetonitrile) to give the title compound 8-fluoro-5H-pyrido [3,2-B ] indole (A, 1g, yield: 58.6%) and 6-fluoro-5H-pyrido [3,2-B ] indole (B, 150mg, yield: 8.8%).
8-Fluoro-5H-pyrido [3,2-b ] indoles :LCMS(ESI):[M+H]+=187.0;1H NMR(400MHz,DMSO-d6,ppm)δ11.50(s,1H),8.47(d,J=4.0Hz,1H),7.92-7.88(m,2H),7.60-7.58(m,1H),7.43-7.37(m,2H).
6-Fluoro-5H-pyrido [3,2-b ] indoles :LCMS(ESI):[M+H]+=187.0;1H NMR(400MHz,DMSO-d6,ppm)δ11.91(s,1H),8.50(d,J=4.0Hz,1H),8.02-8.00(m,1H),7.93-7.90(m,1H),7.47-7.36(m,2H),7.24-7.22(m,1H).
Step 3 preparation of diethyl (4- ((8-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
8-Fluoro-5H-pyrido [3,2-b ] indole (60 mg,0.3mmol,1 eq) was dissolved in tetrahydrofuran (15 mL), sodium hydride (64 mg,1.6mmol,5 eq) and diethyl (4- (bromomethyl) benzyl) phosphonate (207 mg,0.6mmol,2 eq) were added, the reaction was stirred at room temperature under argon for 1 hour, then quenched with saturated ammonium chloride solution, extracted three times with ethyl acetate (10 mL. Times.3) and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (60 mg, yield: 43.7%) as a yellow liquid. LCMS (ESI) [ m+h ] + = 427.0.
Step 4 preparation of (4- ((8-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((8-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (60 mg,0.1mmol,1 eq) was dissolved in dichloromethane (5 mL) and trimethylbromosilane (1 mL,6.5mmol,46 eq) was added and the reaction stirred under argon at 50℃for 1 hour. The reaction solution was concentrated, and the crude product was purified by reverse phase HPLC (C18, 10 mmol/ammonia, acetonitrile) to give the title compound (3.07 mg, yield: 5.9%) as a white solid .LCMS(ESI):[M+H]+=371.0;1H NMR(400MHz,CD3OD)δ8.45(dd,J=4.8,1.2Hz,1H),8.03-7.98(m,2H),7.62(dd,J=9.2,4.0Hz,1H),7.48(dd,J=8.4,4.4Hz,1H),7.36-7.31(m,1H),7.25-7.22(m,2H),7.06(d,J=8.0Hz,2H),5.61(s,2H),2.88(d,J=20.8Hz,2H).
EXAMPLE 90 preparation of (4- ((6-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((6-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
6-Fluoro-5H-pyrido [3,2-b ] indole (50 mg,0.3mmol,1 eq) was dissolved in tetrahydrofuran (15 mL), sodium hydride (60 mg,1.5mmol,5 eq) and diethyl (4- (bromomethyl) benzyl) phosphonate (173 mg,0.5mmol,2 eq) were added, the reaction was stirred at room temperature under argon for 1 hour, after which the saturated ammonium chloride solution quenched, ethyl acetate (10 mL. Times.3) was added to extract three times, the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (60 mg, yield: 52.4%) as a yellow liquid. LCMS (ESI) [ m+h ] + = 427.1.
Step 2 preparation of (4- ((6-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((6-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (60 mg,0.1mmol,1 eq) was dissolved in dichloromethane (5 mL) and trimethylbromosilane (1 mL,6.5mmol,46 eq) was added and the reaction stirred under argon at 50℃for 1 hour. The reaction solution was concentrated, and the crude product was purified by reverse phase HPLC (C18, 10 mmol/ammonia, acetonitrile) to give the title compound (2.01 mg, yield: 3.9%) as a white solid .LCMS(ESI):[M+H]+=371.0;1H NMR(400MHz,CD3OD)δ8.48(d,J=4.8Hz,1H),8.14(d,J=8.8Hz,1H),8.06(d,J=8.8Hz,1H),7.51-7.48(m,1H),7.35-7.22(m,4H),7.05(d,J=7.6Hz,2H),5.74(s,2H),2.88(d,J=20.4Hz,2H).
EXAMPLE 91 preparation of (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 2- (2, 4-difluorophenyl) -3-nitropyridine:
2-chloro-3-nitropyridine (300 mg,1.89mmol,1 eq.) and (2, 4-difluorophenyl) boronic acid (598 mg,3.78mmol,2 eq.) are dissolved in a mixed solvent of toluene (4 mL), water (2 mL) and ethanol (2 mL), and palladium acetate (21 mg,0.09mmol,0.05 eq.) and triphenylphosphine (100 mg,0.38mmol,0.2 eq.) are added and reacted at 90℃for 1 hour. The reaction was quenched with saturated aqueous ammonium chloride, extracted three times with ethyl acetate (10 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (300 mg, yield: 67.1%). LCMS (ESI) [ m+h ] + =236.9.
Step 2 preparation of 7, 9-difluoro-5H-pyrido [3,2-b ] indole:
After 2- (2, 4-difluorophenyl) -3-nitropyridine (300 mg,1.27mmol,1 eq.) was dissolved in dichlorobenzene (5 mL), triphenylphosphine (666 mg,2.54mmol,2 eq.) was added, warmed to 200℃and stirred for 2 hours. The reaction solution was concentrated by cooling, and the crude product was purified by Prep-TLC Prep plate (silica gel, dichloromethane: methanol (V/v=10/1)) to give the title compound (200 mg, yield: 77.1%). LCMS (ESI) [ m+h ] + =205.1.
Step 3 preparation of diethyl (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- (bromomethyl) benzyl) phosphonate (944 mg,2.94mmol,3 eq) and 7, 9-difluoro-5H-pyrido [3,2-b ] indole (200 mg,0.98mmol,1 eq) were dissolved in N, N-dimethylformamide (10 mL), potassium carbonate (406 mg,2.94mmol,3 eq) was added and the temperature was raised to 70 ℃ for 2 hours. The reaction solution was cooled, diluted with saturated aqueous ammonium chloride, extracted with ethyl acetate (30 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated by filtration to give the crude title compound (230 mg, yield: 83.1%). LCMS (ESI) [ m+h ] + =445.1.
Step 4 preparation of (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (230 mg,0.52mmol,1 eq) was dissolved in dichloromethane (5 mL), trimethylbromosilane (1 mL,6.53mmol,12 eq) was added and stirred at 55℃for 1 hour, the reaction quenched with methanol, concentrated, and the crude product prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (21 mg, yield :10.4%).LCMS(ESI):[M+H]+=389.0;1HNMR(400MHz,CD3OD)δ8.48(dd,J=4.8,1.2Hz,1H),8.01(dd,J=8.4,1.2Hz,1H),7.47(dd,J=8.0,3.2Hz,1H),7.25(dd,J=7.6,2.0Hz,3H),7.07(d,J=8.0Hz,2H),6.88(dt,J=10.0,2.0Hz,1H),5.59(s,2H),2.92(d,J=20.8Hz,2H).
Example 92 preparation of (4- ((2-ethoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of di-tert-butyl 4- ((2-ethoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
Di-tert-butyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phosphonate (200 mg,0.40mmol,1.0 eq.) was dissolved in ethanol (2 mL) and 2-di-tert-butyl phosphino-2 ',4',6' -triisopropylbiphenyl (34 mg,0.08mmol,0.2 eq.), cesium carbonate (390 mg,1.2mmol,3.0 eq.) and palladium acetate (7 mg,0.04mmol,0.1 eq.) were added. The reaction was continued for 2 hours under the protection of inert gas at 100 ℃. Saturated aqueous sodium chloride (10 mL) was added to the reaction solution, and extracted three times with ethyl acetate (20 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) to give the title compound (70 mg, yield: 34.4%) as a yellow oily liquid. LCMS (ESI) [ m+h ] + = 509.3.
Step 2 preparation of (4- ((2-ethoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl (4- ((2-ethoxy-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (70 mg,0.14mmol,1.0 eq) was dissolved in methanol (1 mL) and a solution of hydrogen chloride in methanol (31 mg,0.84mmol,6.0 eq) was added. The reaction solution was stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to give a crude product as a brown oil. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (23.5 mg, yield: 42.3%) as a white solid .LCMS(ESI):[M+H]+=397.2,;1H NMR(400MHz,CD3OD)δ8.19(d,J=8.0Hz,1H),7.84(d,J=8.8Hz,1H),7.53(d,J=8.4Hz,1H),7.44(t,J=7.6Hz,1H),7.23-7.19(m,3H),7.04(d,J=8.0Hz,2H),6.84(d,J=8.8Hz,1H),5.54(s,2H),4.45(q,J=7.2Hz,2H),2.91(d,J=20.8Hz,2H),1.44(t,J=7.2Hz,3H).
Examples 93 and 94 preparation of (4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonic acid:
step 1 preparation of diethyl (4-Acetylbenzyl) phosphonate
1- (4- (Bromomethyl) phenyl) ethan-1-one (2.4 g,11.2mmol,1.0 eq) was added to triethyl phosphite (5 mL) and the reaction continued for 5 hours at 120 ℃. The reaction solution was concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1)) to give the title compound (2.5 g, yield: 83.3%) as a transparent oily liquid. LCMS (ESI) [ m+h ] + =271.2.
Step 2 preparation of diethyl (S) - (4- (1-hydroxyethyl) benzyl) phosphonate
2-Methyl-CBS-oxazaborole (693 mg,2.5mmol,0.5 eq.) was dissolved in dichloromethane (7 mL) and methyltetrahydrofuran (7 mL), to this solution was added borane-tetrahydrofuran complex (7.5 mL,7.5mmol,1.5 eq.) and diethyl (4-acetylbenzyl) phosphonate (1.35 g,5.0mmol,1 eq.) at-40℃and the reaction was continued for 2 hours at-20 ℃. The reaction solution was quenched with methanol (30 mL), concentrated, and the crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=20/1)) to give the title compound (600 mg, yield: 44.1%) as a clear oily liquid. LCMS (ESI) [2m+h ] + = 545.2.
Step 3 preparation of diethyl (4- (1-chloroethyl) benzyl) phosphonate
Diethyl (S) - (4- (1-hydroxyethyl) benzyl) phosphonate (1.0 g,3.6mmol,1.0 eq) was dissolved in dichloromethane (10 mL) and thionyl chloride (2.2 g,18.4mmol,5.0 eq) was added and the reaction stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure, and the crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (600 mg, yield: 47.1%) as a clear oily liquid. LCMS (ESI) [ m+h ] + = 290.9.
Step 4 preparation of diethyl (4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonate
3, 6-Dimethoxy-9H-carbazole (783 mg,3.5mmol,2.0 eq) was dissolved in N, N-dimethylformamide (8 mL), and sodium hydride (138 mg,3.5mmol,2.0 eq) was added to this solution at 0℃and reacted at 0℃for 30 minutes. Diethyl (4- (1-chloroethyl) benzyl) phosphonate (500 mg,1.7mmol,1.0 eq) was then added thereto and the reaction stirred at room temperature for 2 hours. The reaction was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (400 mg, yield: 48.2%) as a clear oily liquid. LCMS (ESI) [2m+h ] + = 963.2.
Step 5 chiral resolution of diethyl (4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonate to give isomer A and isomer B
Racemic compound diethyl (4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonate (100 mg) was resolved chiral by SFC (System: WATERS SFC 150; column: DAICEL CHIRALCE)Column size 250 x 25mm10 μm, mobile phase A supercritical CO 2, mobile phase B MeOH (+0.1% 7.0mol/l ammonia in MeOH) to give two single configuration compounds, isomer A50 mg, t R = 2.137min and isomer B50 mg, t R = 2.578min.
Step 6:R preparation of 4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonic acid and S- (4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonic acid:
Diethyl 4- (1- (3, 6-dimethoxy-9H-carbazol-9-yl) ethyl) benzyl) phosphonate (50 mg,0.1mmol,1.0 eq.) in step 5 was dissolved in dichloromethane (2 mL) respectively, and trimethylbromosilane (238 mg,1.5mmol,15 eq.) was added to the mixture and the reaction stirred at 50℃for 1 hour. The reaction solution was quenched with methanol and concentrated under reduced pressure to give crude product as colorless oil. The crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the compound of example 93 (or 94) (30 mg, yield: 68.2%) and the compound of example 94 (or 93) (25 mg, yield: 56.8%).
Example 93 (or 94) Compounds :LCMS(ESI):[M+H]+=426.2;1H NMR(400MHz,DMSO-d6,ppm)δ7.71(d,J=2.4Hz,2H),7.28(d,J=9.2Hz,2H),7.13(d,J=7.2Hz,2H),7.07(d,J=8.0Hz,2H),6.92(dd,J=8.8,2.4Hz,2H),6.03-6.01(m,1H),3.83(s,6H),2.67(d,J=20.8Hz,2H),1.83(d,J=6.8Hz,3H).
Example 94 (or 93) Compounds :LCMS(ESI):[M+H]+=426.2;1H NMR(400MHz,DMSO-d6,ppm)δ7.71(d,J=2.4Hz,2H),7.28(d,J=9.2Hz,2H),7.12(d,J=7.2Hz,2H),7.05(d,J=8.0Hz,2H),6.92(dd,J=8.8,2.4Hz,1H),6.03-6.01(m,1H),3.82(s,6H),2.61(d,J=20.8Hz,2H),1.83(d,J=6.8Hz,3H).
EXAMPLE 95 preparation of((4- (4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 7, 9-difluoro-5H-pyrimido [5,4-b ] indol-4-ol:
3-amino-4, 6-difluoro-1H-indole-2-carboxamide (2 g,9.47mmol,1 eq.) was dissolved in ethanol (20 mL) and formamidine acetate (2 g,18.9mmol,2 eq.) was added and the reaction heated to 100deg.C for 2 hours. LCMS monitored completion of the reaction, the reaction was cooled, concentrated under reduced pressure, the filter cake rinsed with ethanol, and the solid collected and dried to give the target compound (1.6 g, 76.4%) which was used directly in the next reaction. LCMS (ESI) [ M-H ] - =220.1.
Step 2 preparation of 4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole
7, 9-Difluoro-5H-pyrimido [5,4-b ] indol-4-ol (442 mg,2mmol,1 eq.) is added to phenylphosphoryl dichloride (8 mL), the reaction system is warmed to 180℃and stirred for one hour. The reaction solution was diluted with ethyl acetate (20 mL), neutralized with sodium hydrogencarbonate, extracted three times with ethyl acetate (30 mL. Times.3), and concentrated to give the title compound (300 mg, yield: 62.7%). LCMS (ESI) [ m+h ] + =240.0.
Step 3 preparation of di-tert-butyl ((4- (4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate:
4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indole (300 mg,1.3mmol,1.0 eq.) was added to N, N-dimethylformamide (3 mL), followed by di-tert-butyl (4-bromomethyl) phosphonate (218 mg,1.9mmol,1.5 eq.) and finally potassium carbonate (692 mg,5.0mmol,4.0 eq.) and the reaction was continued at 80℃for two hours. The reaction was quenched with water (20 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1)) to give the title compound (500 mg, yield: 74.6%) as a yellow solid. LCMS (ESI) [ m+na ] + =558.1.
Step 4 preparation of ((4- (4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Di-tert-butyl ((4- (4-chloro-7, 9-difluoro-5H-pyrimido [5,4-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.37mmol,1.0 eq) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (127 mg,1.1mmol,3.0 eq) was added to the mixture and the reaction stirred at room temperature for two hours. The reaction solution was adjusted to pH about 7 with ammonia water and concentrated under reduced pressure. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (42.95 mg, yield: 27.4%) as a white solid .LCMS(ESI):[M+H+MeCN]+=465.2,;1H NMR(400MHz,DMSO-d6)δ8.90(s,1H),7.58(dd,J=9.6,2.0Hz,1H),7.25(td,J=10.1,2.0Hz,1H),7.07(dd,J=8.0,1.6Hz,2H),6.94(d,J=8.0Hz,2H),5.85(s,2H),2.59(d,J=20.0Hz,2H).
Example 96 preparation of (4- ((2-cyano-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Step 1 preparation of diethyl (4- ((2-cyano-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
Diethyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (300 mg,0.68mmol,1.0 eq) was dissolved in N, N-dimethylformamide (3 mL), and to this mixture was then added zinc cyanide (239 mg,2.0mmol,3.0 eq), bis [2- (diphenylphosphino) cyclopent-2, 4-dien-1-yl ] iron (75 mg,0.14mmol,0.2 eq) and tetrakis (triphenylphosphine) palladium (157 mg,0.14mmol,0.2 eq). The reaction solution was stirred under microwave conditions at 110 ℃ for 1 hour. The reaction was monitored by LCMS for end. The reaction solution was quenched with water (15 mL) and extracted three times with ethyl acetate (25 mL. Times.3). The organic phase was collected, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated. The crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (250 mg, yield: 85.2%) as a yellow solid. LCMS (ESI) [ m+h ] += 434.0.
Step 2 preparation of (4- ((2-cyano-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((2-cyano-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (150 mg,0.35mmol,1.0 eq) was dissolved in dichloromethane (2 mL) and trimethylbromosilane (803 mg,5.3mmol,15 eq) was then added to this mixture. The reaction solution was stirred at 50 ℃ for 1 hour. LCMS monitored the end of the reaction. The reaction mixture was quenched with methanol and concentrated under reduced pressure to give crude product as a brown oil. Preparation of the crude product by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) gave the title compound (102.77 mg, yield: 77.8%) as a white solid .LCMS(ESI):[M+H]+=378.0;1H NMR(400MHz,DMSO-d6)δ8.28(d,J=7.6Hz,1H),8.23(d,J=8.4Hz,1H),7.98(d,J=8.4Hz,1H),7.77(d,J=8.4Hz,1H),7.63(t,J=7.6Hz,1H),7.37(t,J=7.6Hz,1H),7.08-7.01(m,4H),5.64(s,2H),2.64(d,J=15.6Hz,2H).
Example 97 preparation of (4- ((2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((2- (prop-1-en-2-yl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((2-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.45mmol,1 eq.) isopropenylboronic acid pinacol ester (152 mg,0.90mmol,2 eq.), 1-bis (diphenylphosphine) ferrocene palladium dichloride (66 mg,0.09mmol,0.2 eq.) and cesium carbonate (441 mg,1.34mmol,3 eq.) are dissolved in a mixed solvent of dioxane (10 mL) and water (2 mL). The reaction was heated to 100 ℃ under argon and stirred for 2 hours. The reaction solution was cooled to room temperature and quenched with an aqueous solution. The mixture was extracted three times with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (180 mg, yield: 88.9%) as an oil. LCMS (ESI) [ m+h ] += 449.1.
Step 2 preparation of diethyl (4- ((2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((2- (prop-1-en-2-yl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (180 mg,0.40mmol,1 eq) was dissolved in methanol (30 mL), palladium on carbon (90 mg,10 wt%) was added and replaced three times with hydrogen, and the reaction was heated to 40 ℃ and stirred for 3 hours. The reaction solution was filtered, the filter cake was washed three times with methanol, the filtrates were combined, and the filtrate was concentrated under reduced pressure to give the crude title compound ((150 mg, yield: 83.4%) as an oil, LCMS (ESI): [ m+h ] += 451.1.
Step 3 preparation of (4- ((2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (150 mg,0.33mmol,1 eq) was dissolved in dichloromethane (30 mL) and trimethylbromosilane (1.0 mL) was added and the reaction was heated to 50deg.C and stirred for 2 hours. The reaction solution was dried by spin-drying, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (63 mg, yield: 47.9%) as a white solid .LCMS(ESI):[M+H]+=395.1;1H NMR(400MHz,Methanol-d4)δ8.40(d,J=8.0Hz,1H),7.89(d,J=8.8Hz,1H),7.58(d,J=8.0Hz,1H),7.52-7.48(m,1H),7.35(d,J=8.8Hz,1H),7.31-7.24(m,3H),7.03-7.00(m,2H),5.53(s,2H),3.29-3.20(m,1H),2.81(d,J=19.6Hz,2H),1.40(d,J=7.0Hz,6H).
Example 98 preparation of (4- ((2- (methylsulfanyl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 6- (methylthio) -3-nitro-2-phenylpyridine
6-Chloro-3-nitro-2-phenylpyridine (600 mg,2.6mmol,1 eq.) was dissolved in methanol (8 mL), a propylene glycol solution of methyl mercaptan (1.2 g,10 wt%, 1 eq.) and potassium carbonate (600 mg,4.3mmol,1.7 eq.) were added, the reaction was stirred under argon for 3 hours at 80℃and then cooled, quenched with saturated ammonium chloride solution (10 mL), extracted three times with ethyl acetate (10 mL. Times.3) and the organic phases were combined and concentrated under reduced pressure to give the title compound (350 mg, yield: 55.5%) after purification by preparative plate (PE: EA (V/V=1/1)) as yellow liquid. LCMS (ESI) [ m+h ] + = 247.0.
Step 2 preparation of 2- (methylthio) -5H-pyrido [3,2-b ] indole
6- (Methylthio) -3-nitro-2-phenylpyridine (350 mg,1.4mmol,1 eq.) was dissolved in 1, 2-dichlorobenzene (5 mL), triphenylphosphine (373 mg,1.4mmol,1 eq.) was added, and after stirring the reaction solution under argon for 2 hours at 200℃the reaction solution was concentrated under reduced pressure and purified by preparative plate (PE: EA (V/V=3/1)) to give the title compound (40 mg, yield: 13.1%) as a yellow liquid. LCMS (ESI) [ m+h ] + =215.0.
Step 3 preparation of diethyl (4- ((2- (methylsulfanyl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
2- (Methylthio) -5H-pyrido [3,2-b ] indole (40 mg,0.2mmol,1 eq) was dissolved in tetrahydrofuran (5 mL), sodium hydride (40 mg, 60% by weight, 1.0mmol,5 eq) was added, and after stirring at room temperature for half an hour, diethyl (4- (bromomethyl) benzyl) phosphonate (120 mg,0.4mmol,2 eq) was added, and after stirring at room temperature under argon for further 1 hour, the reaction was quenched with saturated ammonium chloride solution, extracted three times with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (60 mg, yield: 70.7%) as a yellow liquid. LCMS (ESI) [ m+h ] + = 455.1.
Step 4 preparation of (4- ((2- (methylthio) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2- (methylsulfanyl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (60 mg,0.1mmol,1 eq) was dissolved in dichloromethane (5 mL) and trimethylbromosilane (1.0 mL) was added and the reaction stirred under argon for 1 hour at 40 ℃. LCMS monitored reaction completion and purification by reverse phase HPLC (C18, 10 mmol/ammonia, acetonitrile) afforded the title compound (3.04 mg, yield: 5.8%) as a white solid .LCMS(ESI):[M+H]+=399.0;1H NMR(400MHz,Methanol-d4)δ8.30(d,J=8.0Hz,1H),7.81(d,J=8.8Hz,1H),7.57-7.50(m,2H),7.32-7.21(m,4H),7.05(d,J=8.0Hz,2H),5.55(s,2H),2.93(d,J=21.2Hz,2H),2.67(s,3H).
EXAMPLE 99 preparation of (4- ((2-chloro-7, 9-difluoro-5H-pyridin [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 6-chloro-2- (2, 4-difluorophenyl) -3-nitropyridine
2-Bromo-6-chloro-3-nitropyridine (4.20 g,17.69mmol,1 eq), (2, 4-difluorophenyl) boronic acid (2.51 g,15.92mmol,0.9 eq), tetrakis (triphenylphosphine) palladium (2.04 g,1.77mmol,0.1 eq) and potassium carbonate (7.33 g,53.07mmol,3 eq) were dissolved in a mixed solvent of dioxane (30 mL) and water (6 mL). The reaction was heated to 100 ℃ under argon and stirred for 2 hours. After LCMS monitored the reaction was complete, the reaction was cooled to room temperature and quenched with aqueous solution. The mixture was extracted with dichloromethane (500 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) to give the title compound (2.90 g, yield: 60.6%) as a white solid. LCMS (ESI) [ m+h ] + =271.1.
Step 2 preparation of 2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indole:
6-chloro-2- (2, 4-difluorophenyl) -3-nitropyridine (2.9 g,10.72mmol,1 eq.) was dissolved in o-dichlorobenzene (10 mL) and triphenylphosphine (5.62 g,21.44mmol,2 eq.) was added to the mixture and the reaction was continued for 2 hours under nitrogen at 200 ℃. The reaction was monitored by LCMS for end. After the reaction solution was concentrated under reduced pressure, the crude product was purified by flash chromatography (silica gel, methylene chloride: methanol (V/v=10/1)) to give the title compound (1.35 g, yield: 52.8%) as a white solid. LCMS (ESI) [ m+h ] + =239.0.
Step 3 preparation of diethyl (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indole (700 mg,2.93mmol,1 eq.) was dissolved in tetrahydrofuran (30 mL) and sodium hydride (224 mg,60 wt%, 5.86mmol,2 eq.) was then added to this mixture at 0 ℃. The reaction mixture was stirred at 0℃for 30 minutes, and diethyl (4- (bromomethyl) benzyl) phosphonate (1.88 g,5.86mmol,2 eq.) was then added to the mixture. The reaction mixture was stirred at room temperature for 2 hours. The reaction was monitored by LCMS for end, the reaction was quenched with water and extracted with ethyl acetate (200 ml×3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (585 mg, yield: 41.7%) as a yellow solid. LCMS (ESI) [ m+h ] + =479.1.
Step 4 preparation of (4- ((2-chloro-7, 9-difluoro-5H-pyridin [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.42mmol,1 eq) was dissolved in dichloromethane (20 mL), trimethylbromosilane (2 mL) was added and the reaction was heated to 40 ℃ and stirred for 2 hours. Reaction completion was monitored by LCMS. The reaction solution was dried by spin-drying, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (65 mg, yield: 36.6%) as a white solid .LCMS(ESI)[M+H]+=423.1;1H NMR(400MHz,Methanol-d4)δ7.99(d,J=8.8Hz,1H),7.43(d,J=8.8Hz,1H),7.32(dd,J=8.4,2.0Hz,2H),7.26(dd,J=9.6,2.0Hz,1H),7.02(d,J=8.0Hz,2H),6.88-6.81(m,1H),5.53(s,2H),2.82(d,J=20.0Hz,2H).
Example 100 preparation of (4- ((7, 9-difluoro-2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((7, 9-difluoro-2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (50 mg,0.10mmol,1 eq.), methylboronic acid (13 mg,0.20mmol,2 eq.), tetrakis (triphenylphosphine) palladium (20 mg,0.02mmol,0.2 eq.) and cesium carbonate (102 mg,0.30mmol,3 eq.) were dissolved in a mixed solvent of dioxane (10 mL) and water (2 mL). The reaction was heated to 100 ℃ under argon and stirred for 2 hours. After LCMS monitored the reaction was complete, the reaction was cooled to room temperature and quenched with aqueous solution. The mixture was extracted three times with ethyl acetate (10 ml×3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (crude 60 mg) as an oil. LCMS (ESI) [ m+h ] + = 459.1.
Step 2 preparation of (4- ((7, 9-difluoro-2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((7, 9-difluoro-2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (60 mg,0.13mmol,1 eq) was dissolved in dichloromethane (5 mL), trimethylbromosilane (1 mL) was added and the reaction stirred under argon for 2 hours at 40 ℃. LCMS monitored completion of the reaction and purification by reverse phase preparation (C18, 10 mmol/ammonia, acetonitrile) afforded the title compound (6.20 mg, yield: 11.8%) as a white solid .LCMS(ESI):[M+H]+=402.9;1H NMR(400MHz,Methanol-d4)δ7.87(d,J=8.4Hz,1H),7.32(d,J=8.4Hz,1H),7.25-7.23(m,2H),7.19(dd,J=9.6,1.6Hz,1H),7.05(d,J=7.9Hz,2H),6.86-6.80(m,1H),5.54(s,2H),2.93(d,J=21.2Hz,2H),2.70(s,3H).
Example 101 preparation of (4- ((7, 9-difluoro-2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((7, 9-difluoro-2- (prop-1-en-2-yl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (350 mg,0.73mmol,1 eq.) pinacol isopropenylborate (245 mg,1.46mmol,2 eq.), 1-bis (diphenylphosphine) ferrocene palladium dichloride (107 mg,0.15mmol,0.2 eq.) and cesium carbonate (714 mg,2.19mmol,3 eq.) were dissolved in a mixed solvent of dioxane (15 mL) and water (3 mL). The reaction was heated to 100 ℃ under argon, stirred for 1 hour, and after completion of LCMS monitoring the reaction, the reaction solution was cooled to room temperature and quenched with aqueous solution. The mixture was extracted three times with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (300 mg, yield: 84.8%) as an oil. LCMS (ESI) [ m+h ] + =485.1.
Step 2 preparation of diethyl (4- ((7, 9-difluoro-2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((7, 9-difluoro-2- (prop-1-en-2-yl) -5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (250 mg,0.52mmol,1 eq) was dissolved in methanol (40 mL), palladium on carbon (150 mg,10 wt%) was added, hydrogen was replaced three times, and the reaction was heated to 40 ℃ and stirred for 3 hours. Reaction completion was monitored by LCMS. The reaction solution was filtered, and the residue was washed three times with methanol, and the filtrates were combined and concentrated under reduced pressure to give the crude title compound (200 mg, yield: 78.8%) as an oil. LCMS (ESI) [ m+h ] + = 487.1.
Step 3 preparation of (4- ((7, 9-difluoro-2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((7, 9-difluoro-2-isopropyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (200 mg,0.41mmol,1 eq) was dissolved in dichloromethane (30 mL), and trimethylbromosilane (627 mg,4.10mmol,10 eq) was added and the reaction heated to 40 ℃ and stirred for 1 hour. The completion of the reaction was monitored by LCMS, the reaction mixture was dried by spin-drying, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (117 mg, yield: 66.1%) as a white solid .LCMS(ESI):[M+H]+=431.1;1H NMR(400MHz,Methanol-d4)δ7.89(d,J=8.4Hz,1H),7.35(d,J=8.4Hz,1H),7.32(dd,J=8.0,2.0Hz,2H),7.18(dd,J=9.6,2.0Hz,1H),7.01(d,J=9.6Hz,2H),6.82-6.76(m,1H),5.50(s,2H),3.31-3.21(m,1H),2.82(d,J=19.6Hz,2H),1.37(d,J=7.2Hz,6H).
EXAMPLE 102 preparation of (4- ((7, 9-difluoro-2-methylamino-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of diethyl (4- ((7, 9-difluoro-2-methylamino-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate
Diethyl (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (100 mg,0.21mmol,1 eq.) solution of methylamine in tetrahydrofuran (2.1 mL,4.20mmol, 2M), palladium acetate (14 mg,0.06mmol,0.3 eq.), 2-di-tert-butyl phosphino-2 ',4',6' -triisopropylbiphenyl (60 mg,0.06mmol,0.3 eq.) and cesium carbonate (204 mg,0.63mmol,3 eq.) are dissolved in a mixed solvent of dioxane (10 mL) and water (2 mL). The reaction was heated to 100 ℃ under argon and stirred for 18 hours. After LCMS monitored the reaction was complete, the reaction was cooled to room temperature and quenched with aqueous solution. The mixture was extracted three times with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (20 mg, yield: 20.2%) as an oil. LCMS (ESI) [ m+h ] + =474.1.
Step 2 preparation of (4- ((7, 9-difluoro-2-methylamino-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Diethyl (4- ((7, 9-difluoro-2-methylamino-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (20 mg,0.04mmol,1 eq) was dissolved in dichloromethane (5 mL), and trimethylbromosilane (62 mg,0.40mmol,10 eq) was added and the reaction heated to 40 ℃ and stirred for 2 hours. The completion of the reaction was monitored by LCMS, the reaction mixture was dried by spin-drying, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) to give the title compound (1.36 mg, yield: 6.3%) as a white solid .LCMS(ESI):[M+H]+=418.1;1H NMR(400MHz,DMSO-d6)δ7.72(d,J=8.8Hz,1H),7.33(d,J=10.0Hz,1H),7.08(d,J=7.2Hz,2H),6.98(d,J=8.0Hz,2H),6.85(t,J=10.4Hz,1H),6.58(d,J=8.8Hz,1H),6.36(s,1H),5.46(s,2H),2.84(s,3H),2.69(d,J=20.0Hz,2H).
Example 103 preparation of (4- ((7, 8-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Step 1 preparation of 2- (3, 4-difluorophenyl) -3-nitropyridine:
2-chloro-3-nitropyridine (1.0 g,6.31mmol,1 eq.) and (3, 4-difluorophenyl) boronic acid (1 g,6.94mmol,1.1 eq.) were dissolved in a mixed solvent of 1,4 dioxane (8 mL) and water (2 mL), followed by the addition of potassium carbonate (2.6 g,18.93mmol,3 eq.) and 1, 1-bis (diphenylphosphine) ferrocene palladium dichloride (463 mg,0.63mmol,0.1 eq.) under nitrogen protection, and warmed to 120℃for 2h. After completion of the reaction, LCMS was monitored, quenched with saturated aqueous ammonium chloride, extracted three times with ethyl acetate (10 mL. Times.3), and the combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (1.3 g, yield: 87.2%). LCMS (ESI) [ m+h ] + =237.0.
Step 2 preparation of 7, 8-difluoro-5H-pyrido [3,2-b ] indole and 6, 7-difluoro-5H-pyrido [3,2-b ] indole:
After 2- (3, 4-difluorophenyl) -3-nitropyridine (1.0 g,4.23mmol,1 eq.) was dissolved in dichlorobenzene (10 mL), triphenylphosphine (2.2 g,8.47mmol,2 eq.) was added, warmed to 200℃and stirred for 5h. After completion of the reaction, LCMS was monitored, quenched with saturated aqueous ammonium chloride, extracted three times with ethyl acetate (10 mL. Times.3), the combined organic phases dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was prepared by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) to give the title compound A (200 mg, yield: 23.1%) and the title compound B (15 mg, yield: 1.7%).
7, 8-Difluoro-5H-pyrido [3,2-b ] indoles :LCMS(ESI):[M+H]+=205.0;1H NMR(400MHz,DMSO-d6)δ11.72(s,1H),8.47(d,J=4.4Hz,1H),8.13-8.11(m,1H),7.92(d,J=8.0Hz,1H),7.63-7.60(m,1H),7.42-7.39(m,1H).
6, 7-Difluoro-5H-pyrido [3,2-b ] indoles :LCMS(ESI):[M+H]+=205.0;1H NMR(400MHz,DMSO-d6)δ12.22(s,1H),8.47(dd,J=4.4,1.2Hz,1H),7.99-7.57(m,1H),7.92(dd,J=8.0,1.2Hz,1H),7.45(dd,J=8.4,2.4Hz,1H),7.28-7.23(m,1H).
Step 3 preparation of diethyl (4- ((7, 8-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate:
After 7, 8-difluoro-5H-pyrido [3,2-b ] indole (100 mg,0.49mmol,1 eq.) was dissolved in tetrahydrofuran (2 mL), sodium hydride (78 mg,60 wt%, 1.96mmol,4 eq.) was added, the reaction mixture was stirred at room temperature for 0.5H under nitrogen protection, diethyl (4- (bromomethyl) benzyl) phosphonate (315 mg,0.98mmol,2 eq.) was added and stirring was continued for 1H, after LCMS monitoring the reaction was completed, the reaction mixture was quenched with saturated ammonium chloride, extracted three times with ethyl acetate (10 mL. Times.3) and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give the title compound (80 mg, yield: 36.7%). LCMS (ESI) [ m+h ] + =445.1.
Step 4 preparation of (4- ((7, 8-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid:
Diethyl (4- ((7, 8-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonate (70 mg,0.16mmol,1 eq) was dissolved in dichloromethane (1 mL), trimethylbromosilane (245 mg,1.6mmol,10 eq) was added and the temperature was raised to 40℃for 2 hours. The reaction was monitored by LCMS, quenched with methanol, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L ammonia, acetonitrile) to give the title compound (7.41 mg, yield :11.9%).LCMS(ESI)[M+H]+=389.0;1H NMR(400MHz,Methanol-d4)δ8.45(dd,J=4.8,1.2Hz,1H),8.14(dd,J=10.4,8.0Hz,1H),8.03(d,J=7.2Hz,1H),7.57(dd,J=11.2,6.4Hz,1H),7.46(dd,J=8.4,4.8Hz,1H),7.31-7.29(m,2H),7.03(d,J=8.0Hz,2H),5.56(s,2H),2.84(d,J=20.0Hz,2H).
EXAMPLE 104 preparation of (4- ((6, 7-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 89 was carried out following steps 3-4 of the preparation method of example 89 using synthetic 6, 7-difluoro-5H-pyrido [3,2-b ] indole as starting material 104.LCMS(ESI)[M+1]+=389.0;1H NMR(400MHz,Methanol-d4)δ8.47(d,J=3.2Hz,1H),8.09-8.06(m,2H),7.48(dd,J=8.4,2.4Hz,1H),7.31(dd,J=8.0,2.4Hz,2H),7.22-7.16(m,1H),7.02(d,J=8.0Hz,2H),5.70(s,2H),2.82(d,J=20.0Hz,2H).
Example 105 preparation of (4- ((7, 9-difluoro-3-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 89 was carried out according to steps 1-4 of the preparation method of example 89 using commercially available 2-chloro-5-methyl-3-nitropyridine and 2, 4-difluorophenylboronic acid as starting materials 105.LCMS(ESI)[M+1]+=403.1;1H NMR(400MHz,Methanol-d4)δ8.32(s,1H),7.81(s,1H),7.25(dd,J=8.0,2.0Hz,2H),7.17(dd,J=9.6,2.0Hz,1H),7.05(d,J=8.0Hz,2H),6.86-6.80(m,1H),5.52(s,2H),2.90(d,J=20.8Hz,2H),2.52(s,3H).
Example 106 preparation of (4- ((7, 9-difluoro-4-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 89 was carried out according to steps 1-4 of the preparation method of example 89 using commercially available 2-chloro-4-methyl-3-nitropyridine and 2, 4-difluorophenylboronic acid as starting materials 106.LCMS(ESI)[M+1]+=403.1;1H NMR(400MHz,Methanol-d4)δ8.33(d,J=4.8Hz,1H),7.32(d,J=8.0Hz,2H),7.22(d,J=4.8Hz,1H),7.10(d,J=9.6Hz,1H),6.87-6.82(m,1H),6.79(d,J=8.0Hz,2H),5.76(s,2H),2.83(d,J=20.0Hz,2H),2.70(s,3H).
Example 107 preparation of (4- ((7, 9-difluoro-3-fluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphonic acid
Similarly, the preparation of example 89 was carried out according to steps 1-4 of the preparation method of example 89 using commercially available 2-chloro-5-fluoro-3-nitropyridine and 2, 4-difluorophenylboronic acid as starting materials 107.LCMS(ESI)[M+1]+=407.0;1HNMR(400MHz,Methanol-d4)δ8.38(s,1H),7.87(d,J=9.6Hz,1H),7.39-7.20(m,3H),7.07-7.05(m,2H),6.89-6.87(m,1H),5.54(s,2H),2.82(d,J=20.0Hz,2H).
EXAMPLE 108 preparation of 2- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Step 1 preparation of methyl 2- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) acetate
7, 9-Difluoro-5H-pyrido [3,2-b ] indole (210 mg,1.03mmol,1 eq.) and methyl 2- (4- (bromomethyl) phenyl) acetate (500 mg,2.06mmol,2 eq.) were dissolved in acetonitrile (10 mL) and cesium carbonate (675mg, 2.06mmol,2 eq.) was added and the reaction heated to 70℃and stirred for 3 hours. Reaction completion was monitored by LCMS. The reaction solution was concentrated, then diluted with water (50 mL), the mixture was extracted three times with methylene chloride (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure and dried by spin-drying to give the crude title compound (200 mg, yield: 53.0%). LCMS (ESI) [ m+h ] + = 367.1.
Step 2 preparation of 2- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Methyl 2- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) acetate (350 mg,0.96mmol,1 eq.) was dissolved in tetrahydrofuran: water: methanol (V/V/V=1:1:1, 12 mL), followed by hydroxylamine hydrochloride (2.0 g,28.8mmol,30 eq.) and potassium hydroxide (1.62 g,28.8mmol,30 eq.). The reaction was stirred at room temperature for 5 hours. The completion of the reaction was monitored by LCMS, the reaction mixture was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) to give the title compound (33.38 mg, yield: 9.5%) as a white solid .LCMS(ESI):[M+H]+=368.1;1H NMR(400MHz,DMSO-d6)δ10.58(s,1H),8.75(s,1H),8.53(d,J=3.6Hz,1H),8.08(d,J=7.2Hz,1H),7.56(d,J=8.0Hz,1H),7.46(dd,J=8.4,4.4Hz,1H),7.17-7.08(m,5H),5.67(s,2H),3.20(s,2H).
EXAMPLE 109 preparation of 2- (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Similarly, the preparation of example 108 was carried out following steps 1-2 of the preparation method of example 108 using commercially available 5-pyrido [3,2-b ] indole and methyl 2- (4- (bromomethyl) phenyl) acetate as starting materials 109.LCMS(ESI)[M+1]+=332.2;1H NMR(400MHz,DMSO-d6)δ10.56(s,1H),8.72(s,1H),8.49(d,J=4.4Hz,1H),8.25-8.22(m,1H),8.07(d,J=8.4Hz,1H),7.71(d,J=8.8Hz,1H),7.57-7.53(m,1H),7.44(dd,J=8.4,4.8Hz,1H),7.31-7.28(m,1H),7.16-7.10(m,4H),5.67(s,2H),3.19(s,2H).
EXAMPLE 110 preparation of 2- (4- ((8-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Similarly, the preparation of example 108 was carried out following steps 1-2 of the preparation method of example 108 using commercially available 8-chloro-5H-pyrido [3,2-b ] indole and methyl 2- (4- (bromomethyl) phenyl) acetate as starting materials 110.LCMS(ESI)[M+1]+=366.2;1H NMR(400MHz,DMSO-d6)δ10.57(s,1H),8.59(d,J=8.0Hz,1H),8.24(d,J=8.0Hz,2H),7.81(d,J=8.0Hz,1H),7.63-7.59(m,2H),7.16-7.09(m,4H),5.71(s,2H),3.19(s,2H).
EXAMPLE 111 preparation of 5- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) -N-hydroxypentanamide
Similarly, the preparation of example 108 was carried out following steps 1-2 of the preparation method of example 108 using commercially available methyl 5-bromopentanoate and the intermediate 7, 9-difluoro-5H-pyrido [3,2-b ] indole as starting materials 111.LCMS(ESI)[M+1]+=320.1;1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.52(d,J=4.4Hz,1H),8.10(d,J=8.0Hz,1H),7.57-7.53(m,1H),7.48(dd,J=8.4,4.8Hz,1H),7.10-7.04(m,1H),4.43(t,J=7.2Hz,2H),1.96(t,J=7.6Hz,2H),1.76-1.69(m,2H),1.54-1.46(m,2H).
EXAMPLE 112 preparation of 4- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) -N-hydroxybutyramide
Similarly, the preparation of example 108 was carried out following steps 1-2 of the preparation method of example 108 using commercially available methyl 4-bromobutyrate and the intermediate 7, 9-difluoro-5H-pyrido [3,2-b ] indole as starting materials 112.LCMS(ESI)[M+1]+=306.1;1H NMR(400MHz,DMSO-d6)δ10.35(s,1H),8.71(s,1H),8.52(d,J=4.4Hz,1H),8.09(d,J=8.4Hz,1H),7.54(dd,J=10.0,2.0Hz,1H),7.49(dd,J=8.4,4.4Hz,1H),7.11-7.05(m,1H),4.42(t,J=6.4Hz,2H),2.03-1.98(m,4H).
EXAMPLE 113 preparation of 4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) -N-hydroxybenzoamide
In analogy to the preparation of example 108 following steps 1-2 of the preparation method of example 108 using commercially available ethyl 4- (bromomethyl) benzoate and the intermediate 7, 9-difluoro-5H-pyrido [3,2-b ] indole as starting materials 113.LCMS(ESI)[M+1]+=354.1;1H NMR(400MHz,DMSO-d6)δ11.12(s,1H),8.98(s,1H),8.54(d,J=4.0Hz,1H),8.09(d,J=8.0Hz,1H),7.65(d,J=8.4Hz,2H),7.57(d,J=8.4Hz,1H),7.48(dd,J=8.4,4.4Hz,1H),7.23(d,J=8.0Hz,2H),7.15-7.10(m,1H),5.76(s,2H).
EXAMPLE 114 preparation of 2- (4- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) phenyl) -N-hydroxyacetamide
Step 1 preparation of methyl 2- (4- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) phenyl) acetate:
7, 9-difluoro-5H-pyrido [3,2-b ] indole (400 mg,1.96mmol,1.0 eq.) methyl 2- (4-bromophenyl) acetate (897 mg,3.92mmol,2 eq.), nitrosation ketone (38 mg,0.40mmol,0.2 eq.) and cesium carbonate (1.18 g,5.88mmol,3 eq.) were dissolved in N, N-dimethylformamide (10 mL). The reaction solution was stirred at 120 ℃ for 16 hours. The reaction was monitored by LCMS for end, the reaction quenched with water and extracted three times with ethyl acetate (100 mL x 3). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (350 mg, yield: 50.7%) as a yellow solid. LCMS (ESI) [ m+h ] + = 353.1.
Step 2 preparation of 2- (4- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) phenyl) -N-hydroxyacetamide
Methyl 2- (4- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) phenyl) acetate (70 mg,0.20mmol,1 eq.) is dissolved in tetrahydrofuran: water: methanol (3:3:3) 9mL, followed by hydroxylamine hydrochloride (276 mg,3.97mmol,20 eq.) and potassium hydroxide (223 mg,3.97mmol,20 eq.) are added. The reaction was heated to 60 ℃ for 10 hours. Reaction completion was monitored by LCMS. The reaction solution was dried by spin-drying, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) to give the title compound (4 mg, yield: 5.7%) as a white solid .LCMS(ESI):[M+H]+=354.1;1H NMR(400MHz,DMSO-d6)δ10.76(s,1H),8.92(s,1H),8.61(d,J=3.6Hz,1H),7.76(d,J=8.4Hz,1H),7.63-7.57(m,4H),7.47(dd,J=8.4,4.4Hz,1H),7.24-7.18(m,1H),7.05(dd,J=9.2,1.6Hz,1H),3.46(s,2H).
EXAMPLE 115 preparation of 3- (7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) -N-hydroxypropionamide
Similarly, the preparation of example 108 was carried out following steps 1-2 of the preparation method of example 108 using commercially available methyl 3-bromopropionate and the intermediate 7, 9-difluoro-5H-pyrido [3,2-b ] indole as starting materials 115.LCMS(ESI)[M+1]+=291.1;1H NMR(400MHz,DMSO-d6)δ10.34(s,1H),8.51(d,J=4.4Hz,1H),8.05(d,J=8.0Hz,1H),7.49-7.47(m,2H),7.10-7.06(m,1H),4.63(t,J=6.4Hz,2H),2.52(s,2H).
EXAMPLE 116 preparation of 3- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxypropionamide
In analogy to the preparation of example 108 following steps 1-2 of the preparation method of example 108 using commercially available methyl 3- (4- (bromomethyl) phenyl) propionate and the intermediate 7, 9-difluoro-5H-pyrido [3,2-b ] indole as starting materials 116.LCMS(ESI)[M+1]+=382.2;1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),8.54(dd,J=4.8,1.6Hz,1H),8.12(dd,J=8.4,1.2Hz,1H),7.58(dd,J=10.0,2.0Hz,1H),7.48(dd,J=8.4,4.8Hz,1H),7.13-7.08(m,5H),5.66(s,2H),2.73(t,J=7.2Hz,2H),2.18(t,J=8.0Hz,2H).
EXAMPLE 117 preparation of 2- (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Similarly, the preparation of example 108 was carried out following steps 1-2 of the preparation method of example 108, using commercially available methyl 2- (4- (bromomethyl) phenyl) acetate and the intermediate 2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indole as starting materials 117.LCMS(ESI)[M+1]+=402.1;1H NMR(400MHz,DMSO-d6)δ10.58(s,1H),8.75(d,J=1.6Hz,1H),8.19(d,J=8.8Hz,1H),7.60(d,J=8.0Hz,1H),7.56(d,J=8.8Hz,1H),7.17-7.12(m,5H),5.69(s,2H),3.20(s,2H).
EXAMPLE 118 preparation of 2- (4- ((7, 9-difluoro-2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Step 1 preparation of 2- (4- ((7, 9-difluoro-2-methyl-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Methyl 2- (4- ((2-chloro-7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) acetate (12 mg,0.03mmol,1 eq), 2,4, 6-trimethyl-1, 3, 5-trioxytriborane (8 mg,0.06mmol,2 eq), 1-bis (diphenylphosphine) ferrocene palladium dichloride (4.4 mg,0.01mmol,0.2 eq) and cesium carbonate (29 mg,0.09mmol,3 eq) were dissolved in a mixed solvent of dioxane (2.5 mL) and water (0.5 mL). The reaction was heated to 120 ℃ under argon and stirred with microwaves for 1 hour. The completion of the reaction was monitored by LCMS, the reaction mixture was concentrated under reduced pressure, and the crude product was prepared by reverse phase HPLC (C18, 10mmol/L formic acid, acetonitrile) to give the title compound (4.86 mg, yield: 11.4%) as a white solid .LCMS(ESI):[M+H]+=382.1;1H NMR(400MHz,DMSO-d6)δ10.58(s,1H),8.75(s,1H),7.97(d,J=8.8Hz,1H),7.51(d,J=10.0Hz,1H),7.33(d,J=8.4Hz,1H),7.16-7.08(m,4H),7.04(d,J=10.4Hz,1H),5.63(s,2H),3.20(s,2H),2.63(s,3H).
EXAMPLE 119 preparation of 2- (4- ((7, 9-dichloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) -N-hydroxyacetamide
Similarly, the preparation of example was carried out in accordance with step 1-2 of the preparation method of example 89 and then in accordance with step 1-2 of the preparation method of example 108 using commercially available 2-bromo-3-nitropyridine and 2, 4-dichlorobenzoboric acid as starting materials 119.LCMS(ESI)[M+1]+=399.9;1H NMR(400MHz,DMSO-d6,ppm)δ10.59(s,1H),8.61(s,1H),8.14(d,J=8.8Hz,1H),7.93(s,1H),7.55-7.52(m,1H),7.43(s,1H),7.17(d,J=8.4Hz,2H),7.09(d,J=8.0Hz,2H),5.73(s,2H),3.21(s,2H).
EXAMPLE 120 preparation of 4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) benzenesulfonamide
Preparation of 4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) benzenesulfonamide:
5H-pyrido [3,2-b ] indole (100 mg,0.59mmol,1.0 eq.) was added to N-N dimethylformamide (1 mL), then sodium hydride (35 mg,0.89mmol,1.5 eq.) was added thereto at 0℃and the resulting mixture was reacted at room temperature for 30 minutes after three substitutions with argon. 4- (bromomethyl) benzenesulfonamide (149 mg,0.59mmol,1.0 eq.) was then added portionwise thereto. The reaction was continued at room temperature for 16 hours. LCMS monitored the completion of the reaction, quenched the reaction by adding water (50 mL) to the reaction solution and extracted three times with ethyl acetate (50 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated. Preparation of the crude product by reverse phase HPLC (C18, 0.1% formic acid, acetonitrile) gave the title compound 4 (44 mg, yield: 22%) as a white solid .LCMS(ESI):[M+H]+=338.2;1H NMR(400MHz,DMSO-d6,ppm)δ8.53(d,J=4.4Hz,1H),8.27(d,J=7.6Hz,1H),8.14-8.09(m,1H),7.72(t,J=7.2Hz,3H),7.57(t,J=7.2Hz,1H),7.50-7.46(m,1H),7.35-7.26(m,5H),5.82(s,2H).
EXAMPLE 121 preparation of (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) methanesulfonamide
Step 1 preparation of 5- (4- (bromomethyl) benzyl) -7, 9-difluoro-5H-pyrido [3,2-b ] indole
7, 9-Difluoropyrido [3,2-b ] indole (1.0 g,4.9mmol,1.0 eq.) was added to tetrahydrofuran (10 mL), then sodium hydride (254 mg,7.35mmol,1.5 eq.) was added thereto at 0℃and replaced three times with argon, and the resulting mixture was reacted at room temperature for 30 minutes. 1, 4-bis (bromomethyl) benzene (2.59 g,4.9mmol,2.0 eq.) was then added portionwise thereto. The reaction was continued at room temperature for 2 hours. LCMS monitored the completion of the reaction, quenched by the addition of water (100 mL) to the reaction solution and extracted three times with ethyl acetate (150 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=3/1) to give the title compound (280 mg, yield: 14.7%) as a white solid LCMS (ESI): [ m+h ] + =387.0.
Step 2:S- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) thioacetate preparation:
5- (4- (bromomethyl) benzyl) -7, 9-difluoro-5H-pyrido [3,2-b ] indole (280 mg,0.72mmol,1.0 eq.) was dissolved in tetrahydrofuran (3 mL), then thioacetic acid (110 mg,1.44mmol,2.0 eq.) and diazabicyclo (219 mg,1.44mmol,2.0 eq.) were added thereto, and the reaction was continued at room temperature for 2 hours. LCMS monitored the completion of the reaction, quenched the reaction by adding water (50 mL) to the reaction solution and extracted three times with ethyl acetate (50 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=1/1) to give the title compound (150 mg, yield: 54.5%) as a white solid LCMS (ESI): [ m+h ] + = 383.2.
Step 3 preparation of (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) methanesulfonamide:
N-chlorosuccinimide (70 mg,0.52mmol,2.0 eq.) was added portionwise to a solution of hydrochloric acid (2M, 1 mL) in acetonitrile (1 mL) at 0 ℃. The resulting mixture was stirred at 0 ℃ for 5 minutes. Then, to the mixture was added dropwise a solution of S- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) thioacetate (100 mg,0.26mmol,1.0 eq.) in acetonitrile (1 mL) at 0℃and the reaction solution was stirred for 10 minutes. Then, the reaction solution was slowly dropped into ammonia water (10 mL), and the reaction was continued at room temperature for 10 minutes. LCMS monitored reaction completion and crude product was prepared by reverse phase HPLC (C18, 0.1% formic acid, acetonitrile) to give the title compound (8.2 mg, yield: 7.7%) as a white solid .LCMS(ESI):[M+H]+=388.2;1H NMR(400MHz,DMSO-d6,ppm)δ8.55-8.52(m,1H),8.12(dd,J=8.4,1.2Hz,1H),7.59(dd,J=10.0,2.0Hz,1H),7.51-7.45(m,1H),7.28(d,J=8.0Hz,2H),7.20(d,J=8.0Hz,2H),7.14-7.07(m,1H),6.79(s,2H),5.71(s,2H),4.19(s,2H).
EXAMPLE 122 preparation of (4- ((8-chloro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) methanesulfonamide
Similarly, the preparation of example 121 was carried out following steps 1-3 of the preparation method of example 121 using commercially available 8-chloropyrido [3,2-b ] indole and 1, 4-bis (bromomethyl) benzene as starting materials 122.LCMS(ESI)[M+1]+=386.0;1H NMR(400MHz,DMSO-d6,ppm)δ8.53(d,J=4.8Hz,1H),8.21(s,1H),8.15(d,J=8.4Hz,1H),7.80(d,J=8.8Hz,1H),7.62-7.55(m,1H),7.53-7.47(m,1H),7.26(d,J=8.0Hz,2H),7.18(d,J=8.0Hz,2H),6.78(s,2H),5.73(s,2H),4.18(s,2H).
Example 123 preparation of (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) -2-fluorobenzyl) phosphonic acid
Step 1 (preparation of di-tert-butyl 2-fluoro-4-methylbenzyl) phosphonate:
Di-tert-butyl phosphite (2.3 g,11.8mmol,1.2 eq.) was added to ultra-dry tetrahydrofuran (25 mL), sodium hydride (788 mg,19.7mmol,2.0 eq.) was added to the reaction solution at 0℃and the reaction was continued for 30 minutes, to which was added 2-fluoro-4-methyl bromobenzyl (2.0 g,9.85mmol,1.0 eq.). The reaction was continued at 70℃for 16 hours. LCMS monitored the completion of the reaction, quenched the reaction by the addition of water (50 mL) and extracted three times with ethyl acetate (100 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=9/1)) to give the title compound (2.7 g, yield: 86.6%) as a clear oily liquid. LCMS (ESI) [2m+h ] + = 633.1.
Step 2 preparation of di-tert-butyl 4- (bromomethyl) -2-fluorobenzyl) phosphonate:
Di-tert-butyl (2-fluoro-4-methylbenzyl) phosphonate (1.0 g,3.16mmol,1.0 eq.) was added to carbon tetrachloride (10 mL), followed by N-bromosuccinimide (319 mg,3.48mmol,1.1 eq.) and dibenzoyl peroxide (77 mg,0.32mmol,0.1 eq.) added sequentially thereto. The reaction was continued at 78 ℃ for 3 hours. LCMS monitored completion of the reaction, the reaction was concentrated by filtration, then quenched with water (50 mL) and extracted three times with ethyl acetate (100 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=5/1)) to give the title compound (500 mg, yield: 40.0%) as a transparent oil .1H NMR(400MHz,DMSO-d6,ppm)δ7.44-7.37(m,1H),7.34-7.29(m,1H),7.25-7.24(m,1H),4.68(s,2H),3.05(d,J=21.2Hz,2H),1.37(s,18H).
Step 3 preparation of di-tert-butyl 4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) -2-fluorobenzyl) phosphonate:
5H-pyrido [3,2-B ] indole (200 mg,1.2mmol,1.0 eq.) was added to ultra-dry tetrahydrofuran (5 mL), sodium hydride (71 mg,1.78mmol,1.5 eq.) was added to the reaction solution at 0deg.C, the reaction was continued for 30 minutes, and di-tert-butyl (4- (bromomethyl) -2-fluorobenzyl) phosphonate (470 mg,1.2mmol,1.0 eq.) was added thereto. The reaction was continued at room temperature for 16 hours. LCMS monitored the completion of the reaction, quenched the reaction by the addition of water (20 mL) and extracted three times with ethyl acetate (50 mL x 3). The organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated and dried over silica gel. The crude product was purified by flash chromatography (silica gel, petroleum ether: ethyl acetate (V/v=9/1)) to give the title compound (350 mg, yield: 61.0%) as a white solid. LCMS (ESI) [ m+h ] + =483.0.
Step 4 preparation of (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) -2-fluorobenzyl) phosphonic acid:
Di-tert-butyl (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) -2-fluorobenzyl) phosphonate (100 mg,0.20mmol,1.0 eq.) was dissolved in methanol (2 mL) and to this mixture was added a solution of hydrogen chloride in methanol (4M, 0.6mmol,3.0 eq.). The reaction mixture was stirred at room temperature for 16 hours. The reaction was monitored by LCMS for end. The reaction solution was concentrated. The crude product was prepared by reverse phase HPLC (C18, 0.1% ammonia, acetonitrile) to give the title compound (21.01 mg, yield: 27%) as a white solid .LCMS(ESI):[M+H]+=370.9,tR=1.071min.1H NMR(400MHz,DMSO-d6,ppm)δ8.49(d,J=4.0Hz,1H),8.23(d,J=7.6Hz,1H),8.09(d,J=8.0Hz,1H),7.73(d,J=8.4Hz,1H),7.55(t,J=7.6Hz,1H),7.47-7.40(m,1H),7.30(t,J=7.2Hz,1H),7.21-7.20(m,1H),6.94(d,J=10.0Hz,1H),6.87(d,J=7.2Hz,1H),5.67(s,2H),2.80(d,J=20.0Hz,2H).
Example 124 preparation of (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) -2-chlorobenzyl) phosphonic acid
Similarly, the preparation of example 123 was carried out according to steps 1-4 of the preparation method of example 123 using commercially available 2-chloro-4-methylbenzyl bromide and di-tert-butyl phosphite as starting materials 124.LCMS(ESI)[M+1]+=386.8;1H NMR(400MHz,DMSO-d6,ppm)δ8.49(d,J=4.4Hz,1H),8.23(d,J=7.7Hz,1H),8.08(d,J=8.0Hz,1H),7.72(d,J=8.4Hz,1H),7.55-7.54(m,1H),7.42(d,J=8.4Hz,1H),7.31(d,J=7.6Hz,2H),7.18(s,1H),6.99(d,J=7.2Hz,1H),5.65(s,2H),2.80(d,J=20.0Hz,2H).
Example 125 preparation of (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) -2-methoxybenzyl) phosphonic acid
Similarly, the preparation of example 123 was carried out according to steps 1-4 of the preparation method of example 123 using commercially available 2-methoxy-4-methyl bromobenzyl and di-tert-butyl phosphite as starting materials 125.LCMS(ESI)[M+1]+=382.9;11H NMR(400MHz,DMSO-d6,ppm)δ8.48(d,J=4.4Hz,1H),8.22(d,J=7.8Hz,1H),8.08(d,J=8.4Hz,1H),7.73(d,J=8.4Hz,1H),7.54(t,J=7.6Hz,1H),7.42(dd,J=8.0,4.4Hz,1H),7.28(t,J=7.4Hz,1H),7.15-7.11(m,1H),6.90(s,1H),6.49(d,J=7.6Hz,1H),5.60(s,2H),3.62(s,3H),2.74(d,J=20.0Hz,2H).
EXAMPLE 126 preparation of isopropyl (((4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) (phenoxy) phosphoryl) -L-alaninate
Similarly, the preparation of example was carried out in accordance with steps 1-4 of the preparation method of example 51, using example 17 as starting material 126.LCMS(ESI)[M+1]+=542.4;1H NMR(400MHz,DMSO-d6,ppm)δ8.55(d,J=4.4Hz,1H),8.28(d,J=7.6Hz,1H),8.22(d,J=8.0Hz,1H),7.76(dd,J=8.4,4.4Hz,1H),7.61-7.57(m,1H),7.53(dd,J=8.0,4.8Hz,1H),7.33(t,J=7.6Hz,1H),7.33-7.22(m,4H),7.15-7.00(m,5H),5.71(s,2H),5.54-5.46(m,1H),4.79-4.71(m,1H),3.77-3.62(m,1H),3.29-3.17(m,2H),1.08-1.00(m,8H),0.88(d,J=7.2Hz,1H).
EXAMPLE 127 preparation of 2- (4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-oxide
Similarly, the example was prepared according to steps 1-2 of the preparation method of example 50 using example 17 as starting material 127.LCMS(ESI):[M+H]+=503.3;1H NMR(400MHz,DMSO-d6,ppm)δ8.53(d,J=4.8Hz,1H),8.26(d,J=7.6Hz,1H),8.20-8.13(m,1H),7.73(d,J=8.0Hz,1H),7.58-7.52(m,1H),7.48-7.28(m,5H),7.22-7.15(m,5H),5.72(s,2H),5.54(d,J=12.0Hz,1H),4.50-4.40(m,1H),4.21-4.14(m,1H),3.47(s,2H),3.31(s,2H).
EXAMPLE 128 preparation of((4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphoryl) bis (oxy) bis (methylene) bis (2, 2-dimethylpropionate)
Similarly, the example was prepared according to steps 1-2 of the preparation method of example 47 using the example 17 as starting material 128.LCMS(ESI):[M+H]+=581.3;1H NMR(400MHz,DMSO-d6,ppm)δ8.49(d,J=4.4Hz,1H),8.24(d,J=7.6Hz,1H),8.13-8.01(m,1H),7.72(d,J=8.4Hz,1H),7.61-7.50(m,1H),7.44(dd,J=8.4,4.4Hz,1H),7.34-7.24(m,1H),7.18-7.06(m,4H),5.68(s,2H),5.58-5.42(m,4H),2.04-1.96(m,2H),1.04(s,18H).
EXAMPLE 129 preparation of diisopropyl 2,2'- ((4- ((5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) phosphorus radical) bis (azanediyl)) (2S, 2' S) -dipropionate
Similarly, the example was prepared according to steps 1-2 of the preparation method of example 49 using the example 17 as starting material 129.LCMS(ESI):[M+H]+=579.5;1H NMR(400MHz,DMSO-d6,ppm)δ8.59(d,J=4.8Hz,1H),8.30(d,J=7.6Hz,2H),7.78(d,J=8.4Hz,1H),7.63-7.58(m,2H),7.35(t,J=7.6Hz,1H),7.17-7.10(m,4H),5.72(s,2H),4.82-4.78(m,2H),4.41-4.28(m,2H),3.71-3.69(m,1H),3.56-3.52(m,1H),3.02-2.85(m,2H),1.16-1.12(m,15H),0.99(d,J=7.2Hz,3H).
EXAMPLE 130 preparation of N- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) sulfamide
Step 1 preparation of 4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl carbamic acid tert-butyl ester
The compound 7, 9-difluoro-5H-pyrido [3,2-b ] indole (400 mg,1.9mmol,1.0 eq.) was added to N, N-dimethylformamide (20 mL), tert-butyl 4- (bromomethyl) benzylcarbamate (1.8 g,5.9mmol,3.0 eq.) and potassium carbonate (812 mg,5.9mmol,3.0 eq.) were added. The reaction mixture was reacted at 70 ℃ for 3 hours. LCMS monitored completion of the reaction, the reaction was diluted with ethyl acetate (100 mL) and then washed with saturated brine (50 ml×3), dried and the organic phase concentrated. The crude product was purified by flash chromatography (silica gel, petroleum ether: tetrahydrofuran=2:1) to give the title compound (430 mg, yield: 51.8%) as a yellow solid. LCMS (ESI) [ m+h ] + =424.1.
Step 2 preparation of (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) methylamine hydrochloride
Tert-butyl 4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl carbamate (400 mg,0.9mmol,1.0 eq) is dissolved in dichloromethane (4 mL), 4M dioxane (4 mL) of hydrochloric acid is added, the reaction is stirred for 4 hours at 20℃after completion of the LCMS monitoring reaction, the reaction system is suction filtered, the filter cake is collected, and the crude product is slurried with methyl tert-butyl ether (5 mL) to give the title compound (430 mg, crude) as a pale yellow solid. LCMS (ESI) [ m+h ] + = 324.1.
Preparation of tert-butyl 3:N- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) sulfamoyl carbamate
The compound (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) phenyl) methylamine hydrochloride (260 mg,0.8mmol,1.0 eq) was added to dry dichloromethane (5.2 mL), nitrogen was replaced, triethylamine (405 mg,4.01mmol,5 eq) was added at 0 ℃, stirred for 10 minutes, and tert-butyl N- (chlorosulfonyl) carbamate (173 mg,0.8mmol,1.0 eq) was added dropwise at 0 ℃. The reaction mixture was reacted at 20 ℃ for 14 hours, LCMS monitored reaction completion. The system was diluted with dichloromethane (10 mL), then washed with saturated brine (5 mL. Times.3), dried and the organic phase was dried by spinning. The crude product was purified by flash chromatography (silica gel, petroleum ether: tetrahydrofuran=1:1) to give the title compound (210 mg,0.4mmol, yield: 52.1%) as a yellow solid. LCMS (ESI) [ m+h ] + =503.2.
Step 4 preparation of N- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) sulfamide
Tert-butyl N- (4- ((7, 9-difluoro-5H-pyrido [3,2-b ] indol-5-yl) methyl) benzyl) sulfamoyl carbamate (210 mg,0.42mmol,1.0 eq) was dissolved in dichloromethane (2.1 mL) and 4M dioxane hydrochloride (2.1 mL) was added and the reaction mixture reacted at 20℃for 4 hours. LCMS monitored the reaction. After completion of the LCMS detection reaction, the reaction mixture was concentrated and the crude product was purified by Prep-HPLC (YMC TRIART PREP C-S, 250X 50mm,10 μm,0.1% aqueous HCl, acetonitrile) to give the title compound (24.79 mg,0.06mmol, yield: 14.7%) as a pale yellow solid .LCMS(ESI)[M+H]+=403.2;1H NMR(400MHz,DMSO-d6)δ8.53(d,J=4.8Hz,1H),8.19(d,J=8.0Hz,1H),7.58(dd,J=2.0Hz,8.0Hz,1H),7.52(dd,J=2.4Hz,3.6Hz,1H),7.22(d,J=8.0Hz,2H),7.13(d,J=8.4Hz,2H),7.12-7.06(m,1H),5.68(s,2H),3.96(s,2H).
Example 131
In analogy to the preparation of example 130 following steps 1-4 of the preparation method of example 130 using 7, 9-difluoro-5H-pyrido [3,2-b ] indole and tert-butyl N- [4- (bromomethyl) phenyl ] carbamate as starting materials 131.LCMS(ESI):[M+H]+=389.2;1H NMR(400MHz,DMSO-d6)δ9.48(s,1H),8.57(d,J=4.8Hz,1H),8.25(d,J=8.0Hz,1H),7.64(dd,J=1.6Hz,10.0Hz,1H),7.57(dd,J=2.4Hz,3.6Hz,1H),7.16(d,J=8.8Hz,2H),7.05(d,J=8.4Hz,2H),7.03-6.95(m,1H),5.65(s,2H).
Example 132
In analogy to the preparation of example 130 following steps 1-4 of the preparation method of example 130 using 7, 9-difluoro-5H-pyrimido [5,4-b ] indole and tert-butyl N- [4- (bromomethyl) benzyl ] carbamate as starting materials 132.LCMS(ESI):[M+H]+=404.2;1H NMR(400MHz,DMSO-d6)δ9.37(s,1H),9.31(s,1H),7.69(d,J=8.8Hz,1H),7.30-7.16(m,5H),7.10-6.82(m,1H),6.72-6.30(m,1H),5.76(s,2H),4.01(s,2H).
Example 133
In analogy to the preparation of example 130 following steps 1-4 of the preparation method of example 130 using 7, 9-difluoro-5H-pyrimido [5,4-b ] indole and tert-butyl N- [4- (bromomethyl) phenyl ] carbamate as starting materials 133.LCMS(ESI):[M+H]+=390.2;1H NMR(400MHz,DMSO-d6)δ9.51(br,1H),9.43(s,1H),9.18(s,1H),7.73(d,J=8.0Hz,1H),7.30-7.22(m,3H),7.08(d,J=8.4Hz,2H),6.80-6.40(m,2H),5.72(s,2H).
Example 134
Similarly, the preparation of example 130 was carried out following steps 1-4 of the preparation method of example 130 using 7, 9-difluoro-5H-pyrido [3,2-b ] indole and tert-butyl N- (4-bromobutyl) carbamate as starting materials 134.LCMS(ESI):[M+H]+=355.0;1H NMR(400MHz,DMSO-d6)δ8.63(d,J=4.4Hz,1H),8.37(m,J=8.0Hz,1H),7.73-7.65(m,2H),7.20(t,J=9.6Hz,1H),4.51(t,J=11.2Hz,2H),2.91(t,J=6.8Hz,2H),1.88-1.80(m,2H),1.58-1.48(m,2H).
Example 135
Similarly, the preparation of example 130 was carried out following steps 1-4 of the preparation method of example 130 using 7, 9-difluoro-5H-pyrido [3,2-b ] indole and tert-butyl N- (4-bromopropyl) carbamate as starting materials 135.LCMS(ESI):[M+H]+=341.2;1H NMR(400MHz,DMSO-d6)δ8.62(d,J=4.8Hz,1H),8.44(d,J=8.4Hz,1H),7.76-7.65(m,2H),7.20(t,J=10.4Hz,1H),4.53(t,J=7.6Hz,2H),2.96(t,J=6.8Hz,2H),1.97(t,J=7.2Hz,2H).
Biological experiments:
1. determination of EnPP1 Activity inhibitory Activity of Compounds by AMP-Glo method
Test principle under the catalysis of extracellular nucleotide pyrophosphatase/phosphodiesterase I (ENPP 1), cGAMP can be hydrolyzed into AMP and GMP, and the AMP-Glo TM assay (AMP-Glo TM detection kit) is a homogeneous method for detecting AMP by using bioluminescence principle, and can be used for detecting ENPP1 enzymatic activity reaction producing AMP.
Test methods 1-fold enzyme reaction buffer (50mM Tris7.5;150mM NaCl;0.5mM Cacl2;1. Mu.M Zncl 2) was prepared. Test compounds were dissolved in DMSO and diluted to 100 times the final concentration, transferred to assay plates using Echo550, and incubated for 30 minutes at room temperature with 20nM ENPP1 (Biovendor) enzyme washes, respectively. 40. Mu.M substrate 2,3-cGAMP was added to the assay plate, reacted at room temperature for 15 minutes, 5. Mu.L of AMP-Glo TM reagent I was added, the enzymatic reaction of AMP production was stopped and AMP was converted to ADP, reacted at room temperature for 120 minutes, 10. Mu.L of AMP detection reagent was added, left at room temperature for 30 minutes, and the plate was subjected to luminescence data reading on an Envison instrument. After raw data of different concentrations of the compounds are obtained, the data are subjected to standardized treatment according to a formula inh% = (Max-Sample)/(Max-Min) ×100% to obtain enzyme activity inhibition rate inh% (wherein Max is a luminescence value of an enzyme-containing positive hole, min is a luminescence value of a non-enzyme-containing negative hole, sample is a luminescence value of a compound-treated Sample hole), inhibition rate inh% (Y) corresponding to each concentration (X) is input into EXCEL, and half inhibition concentration IC 50 value of each compound is calculated by using XLfit plugin according to a built-in four-parameter fitting formula y=bottom+ (Top-Bottom)/(1+ (IC 50/X) × HillSlope).
TABLE 1 EnPP1 Activity inhibitory Activity
The results show that the series of compounds have strong ENPP1 enzyme inhibition activity.
2. Determination of Compound inhibitory Activity against cellular level ENPP1 Using Elisa method
The principle of the test is that extracellular nucleotide pyrophosphatase/phosphodiesterase I (ENPP 1) in cells is utilized to hydrolyze intracellular cGAMP into AMP and GMP, and a kit CGAMP ELISA kit (Cayman) is used to detect the content of the cGAMP, so that the inhibition condition of the compound on the activity of the cell level ENPP1 is obtained.
Test methods MDA-MB 231 cells (ATCC, cat. HTB-26) were resuscitated in Leibovitz's L (10% FBS,1% PS) and inoculated into reaction plates (cell number 28000 cells/well) at 100 uL/well, and incubated overnight in a 37℃100% air incubator. Test compounds were dissolved and diluted to 0.1% of final concentration in DMSO, transferred to reaction plates 5 uL/well, and co-incubated with 1mg/ml dsDNA, 37 ℃ and incubated in a 100% air incubator for 48 hours. Supernatants were collected for CGAMP ELISA assays, microplates read OD450 on an Envison instrument, and data analyzed. After obtaining the original data of different concentrations of the compound, converting the cGAMP concentration (pg/mL) according to a cGAMP standard curve, and performing standardization treatment on the data according to a formula activation% = (Sample-ave_l)/(ave_h-ave_l) to obtain the activation efficiency activation% of each concentration point on the cGAMP, wherein H is ave_positive control, represents the OD value of a positive well containing a high concentration ENPP1 inhibitor, L is ave_dmso control, represents a well without the ENPP1 inhibitor, and Sample is the luminescence value of a compound treatment Sample well. The activation rate activation% (Y) corresponding to each concentration (X) was entered in EXCEL and the half activation concentration EC50 value for each compound was calculated using XLfit plug-in according to the built-in four-parameter fitting formula y=bottom+ (Top-Bottom)/(1+10 ((log EC 50-X) × HillSlope)).
TABLE 2 cellular level ENPP1 inhibitory Activity
Test example 3 pharmacokinetic assay for determining the concentration of Compounds in mice Using LC-MS/MS
Test principle LC-MS/MS was used to determine the drug concentration of the drug of interest in plasma at different time points and to investigate the pharmacokinetic profile of the drug in mice after single intraperitoneal injection of 25 mpk.
Test methods appropriate amounts of the compounds were precisely weighed and dissolved using 10% DMSO (Sigma-Aldrich, SHBN, 5059), 10% Solutol HS15 (BASF, 30744347G 0) and 80% physiological saline (Zhengjiangdubangyaoye, 2110080102) solvents to give a 2.5mg/mL concentration of the drug delivery formulation. Male CD-1 mice (available from Shanghai Ji Hui laboratory animal feeding Co.) were dosed by intraperitoneal injection at 25mpk doses of 9 mice, cross-sampled at 0.083,0.25hr, 0.5hr, 1hr, 2hr, 4hr, 8hr and 24hr (N=3/time point), about 110. Mu.L of whole blood was collected at each time point (K2 EDTA anticoagulated), placed on wet ice, 100. Mu.L of whole blood was added to a tube containing 10. Mu.L of stabilizer AEBSF/NaF (100 mM/40mg/mL in H 2 O), the sample was immediately centrifuged at 2000g for 5 min to give plasma, 40. Mu.L of plasma was added to a tube containing 4. Mu.L of stabilizer PMSF (100 mM), the sample was first frozen in dry ice, and then stored in a refrigerator below-60 ℃ for a long period until the sample was analyzed, and the whole sample treatment process was carried out under wet ice. Plasma concentration measurements were performed using the Triple-quad MS system (6500+, SCIEX). Standard curve and quality control preparation, namely diluting and preparing working solution by DMSO, and adding the 3 mu L standard curve and quality control working solution into 57 mu L blank plasma (containing stabilizing agent). Sample preparation to 20. Mu.L of plasma sample 150. Mu.L of internal standard solution (Diclofenac, 60 ng/mL) was added, mixed well for 10 min, centrifuged at 5800rpm for 10 min, and 90. Mu.L of supernatant was transferred to a new plate for sample analysis. Chromatographic conditions the respective mobile phase composition, elution gradient conditions, flow rate and retention time were optimized according to the sample, the column was Waters ACQUITY UPLC HSS T (2.1X105 mm,1.8 μm) and the sample volume was 1. Mu.L. And the mass spectrum adopts an electrospray ion source (Turbo spray), and a multi-channel reaction monitoring (MRM) mode is selected for secondary mass spectrum analysis under a positive ion detection mode. According to the drug concentration-time data, the pharmacokinetic parameters including the peak concentration Cmax, the peak time Tmax, the area under the drug-time curve AUC and the elimination half-life t1/2 are calculated according to a non-atrioventricular model by using WinNonlin 8.2 software. The AUC was calculated as a linear trapezoidal method (linear up log down).
TABLE 2 preferred pharmacokinetic parameters of Compounds
The test results show that the compound of the invention has good in vivo pharmacokinetics and has the potential of patent medicine.

Claims (39)

  1. A compound of formula (I), and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates or isotopically-labeled analogues thereof,
    Wherein the method comprises the steps of
    L is selected from -(NRL3)SO2N(RL1)(RL2)、-SO2RL1、-C(O)ORL1、-C(O)N(RL1)(RL2)、-C(O)NHORL1、-P(O)(ORL1)(ORL2)、-P(O)(NHRL1)(ORL2)、-P(O)(ORL1)(RL2)、-P(O)(NHRL1)(NHRL2)、-P(O)(NHRL1)(RL2)、-P(O)(RL1)(RL2)、-P(O)(SRL1)(SRL2)、-P(O)(NHRL1)(SRL2)、-P(O)(SRL1)(RL2) or-B (OH) 2, wherein R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, or optionally substituted aryl, or R L1 and R L2 are joined to form an optionally substituted cyclic structure;
    A is selected from optionally substituted alkylene, optionally substituted cycloalkylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, optionally substituted cycloalkylene, optionally substituted heterocyclylene alkylene, optionally substituted arylene alkylene, or optionally substituted heteroarylene;
    Z is selected from- (chemical bond )、-O-、-S-、-CO-、-SO-、-SO2-、-C(RZ1)(RZ2)-、-N(RZ1)-、-CH2N(RZ1)-、-N(RZ1)CH2-、-CON(RZ1)-、-N(RZ1)CO-、-SON(RZ1)-、-SO2N(RZ1)-、-N(RZ1)SO-、-N(RZ1)SO2- or Wherein R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
    R 1 is selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl 、-OR3、-SR3、-NR3R4、-COOR3、-CONR3R4、-NR3COR4、-COR3、-OCOR3、-SONR3R4、-SO2NR3R4、-NR3SOR4、-NR3SO2R4, or-Si (R 3)(R4)(R5), wherein R 3、R4 or R 5 are independently selected from hydrogen, deuterium, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
    n=0, 1, 2 or 3;
    X 1、X2、X3 and X 4 are independently selected from N or C (R X), wherein R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl 、-ORa、-SRa、-NRaRb、-COORa、-CONRaRb、-NRbCORa、-CORa、-OCORa、-SONRaRb、-SO2NRaRb、-NRbSORa、-NRbSO2Ra, or-Si (R a)(Rb)(Rc), wherein R a、Rb and R c are independently selected from hydrogen, deuterium, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or X 1 is joined with X 2 to form an optionally substituted cyclic structure, or X 2 is joined with X 3 to form an optionally substituted cyclic structure, or X 3 is joined with X 4 to form an optionally substituted cyclic structure, or X 1 and Z are joined to form an optionally substituted cyclic structure.
  2. The compound of claim 1, wherein R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, or optionally substituted C 6-10 aryl.
  3. The compound of claim 1, wherein R L1、RL2 and R L3 are independently selected from hydrogen, deuterium, halogen, amino, cyano, unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, and aryl, and alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, and aryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, alkyl (e.g., C 1-6 alkyl), alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), heterocyclyl (e.g., 4-6 membered heterocyclyl), OC (O) -alkyl (e.g., OC (O) -C 1-6 alkyl), C (O) O-alkyl (e.g., C (O) O-C 1-6 alkyl), OC (O) O-alkyl (e.g., OC (O) O-C 1-6 alkyl), phenyl, or halophenyl (e.g., chlorophenyl or fluorophenyl).
  4. The compound of claim 1, wherein L is selected from :-(NRL3)SO2N(RL1)(RL2)、-SO2RL1、-C(O)ORL1、-C(O)NHORL1、-P(O)(ORL1)(ORL2)、-P(O)(NHRL1)(ORL2)、-P(O)(NHRL1)(NHRL2) or-B (OH) 2.
  5. The compound of claim 1, wherein R L1、RL2 and R L3 are independently selected from hydrogen, amino, optionally substituted alkyl (e.g., C 1-6 alkyl), and optionally substituted aryl (e.g., C 6-10 aryl);
    Preferably, R L1、RL2 and R L3 are independently selected from hydrogen, unsubstituted alkyl and phenyl, and alkyl substituted with one or more substituents independently selected from: -OC (O) -C 1-6 alkyl, -C (O) O-C 1-6 alkyl, -OC (O) O-C 1-6 alkyl, phenyl or halophenyl.
  6. The compound of claim 1, wherein L is selected from :-P(O)(OH)2、-C(O)OH、-C(O)NHOH、-B(OH)2、-SO2NH2、-NHSO2NH2、-P(O)(OCH2OC(O)C(CH3)3)2、-P(O)(OCH2OC(O)OCH(CH3)2)2、-P(O)(NHCH(CH3)C(O)OCH(CH3)2)2、-P(O)(OC6H5)(NHCH(CH3)C(O)OCH(CH3)2).
  7. The compound of claim 1, wherein R L1 and R L2 are linked to form an optionally substituted 5-or 6-membered cyclic structure;
    Preferably, R L1 and R L2 are joined to form a cyclic structure substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted phenyl (e.g., halophenyl), or optionally substituted 5-6 membered heteroaryl;
    More preferably, R L1 and R L2 are joined to form a cyclic structure substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, unsubstituted C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 4-6 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, or C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 4-6 membered heterocyclyl, phenyl or 5-6 membered heteroaryl substituted with substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl or alkoxy.
  8. The compound of claim 1, wherein L is-P (O) (OR L1)(ORL2),RL1 and R L2 are independently optionally substituted alkyl (e.g., C 1-3 alkyl such as methyl OR ethyl), and R L1 and R L2 are joined to form an optionally substituted 5-OR 6-membered cyclic structure;
    Preferably, L is-P (O) (OR L1)(ORL2),RL1 is ethyl and R L2 is methyl substituted with phenyl OR halophenyl (e.g., chlorophenyl OR fluorophenyl), and R L1 and R L2 are joined to form a 6-membered ring structure substituted with phenyl OR halophenyl (e.g., chlorophenyl OR fluorophenyl).
  9. The compound of claim 1, wherein a is selected from optionally substituted C 1-6 alkylene, optionally substituted C 3-6 cycloalkylene, optionally substituted 4-10 membered heterocyclylene, optionally substituted C 6-10 arylene, optionally substituted 5-10 membered heteroarylene, optionally substituted C 3-6 cycloalkylene C 1-3 alkylene, optionally substituted 4-10 membered heterocyclylene C 1-3 alkylene, optionally substituted C 6-10 arylene C 1-3 alkylene, or optionally substituted 5-10 membered heteroarylene C 1-3 alkylene.
  10. The compound of claim 1, wherein A is selected from the group consisting of unsubstituted alkylene, cycloalkylene, heterocyclylene, arylene, heteroarylene, cycloalkylalkylene, heterocyclylene alkylene, arylene alkylene, or heteroarylalkylene, and alkylene, cycloalkylene, heterocyclylene, arylene, heteroarylene, cycloalkylene, heterocyclylene alkylene, arylene alkylene, or heteroarylalkylene substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, acetyl, alkyl (e.g., C 1-6 alkyl), alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), heterocyclyl (e.g., 4-6 membered heterocyclyl), O-alkyl (e.g., -OC 1-6 alkyl), NH-alkyl (e.g., -NHC 1-6 alkyl), S-alkyl (e.g., -SC 1-6 alkyl), O-cycloalkyl (e.g., -OC 1-6 cycloalkyl), NH-C35cycloalkyl (e.g., -NHC 1-6 cycloalkyl), or cycloalkyl-35.
  11. The compound of claim 1, wherein a is selected from optionally substituted C 1-4 alkylene (e.g., methylene, ethylene, propylene, butylene), optionally substituted arylene (e.g., phenylene), optionally substituted cycloalkylene C 1-4 alkylene, optionally substituted arylene C 1-4 alkylene, or optionally substituted heteroarylene C 1-4 alkylene;
    Preferably, A is selected from methylene, ethylene, propylene, butylene, phenylene C 1-2 alkylene (e.g., phenylene methylene or phenylene ethylene), cyclohexylene C 1-2 alkylene (e.g., cyclohexylene methylene), pyridylene C 1-2 alkylene (e.g., pyridylmethylene), or
    Preferably, A is selected from phenylene C 1-2 alkylene (e.g., phenylene methylene or phenylene ethylene) substituted with halogen (e.g., fluorine or chlorine) or-O-alkyl (e.g., -OC 1-6 alkyl, e.g., methoxy), more preferably, A is selected from fluorophenylene-methylene, chlorophenylmethylene, methoxyphenylene-methylene.
  12. The compound of claim 1, wherein R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, or optionally substituted 4-6 membered heterocyclyl.
  13. The compound of claim 1, wherein R Z1 and R Z2 are independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, or heterocyclyl, and alkyl, alkenyl, alkynyl, cycloalkyl, or heterocyclyl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, alkyl (e.g., C 1-6 alkyl), alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), or heterocyclyl (e.g., 4-6 membered heterocyclyl).
  14. The compound of claim 1, wherein Z is selected from- (bond) or-C (R Z1)(RZ2) -; preferably, R Z1 and R Z2 are independently selected from hydrogen and optionally substituted alkyl (e.g., C 1-6 alkyl such as methyl), more preferably Z is selected from- (a bond), -CH 2 -, or-CH (CH 3) -.
  15. The compound of claim 1, wherein Z is-CH (CH 3) -, having the (R) or (S) configuration.
  16. The compound of claim 1, wherein R 1 is selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted C 6-10 aryl 、-OR3、-SR3、-NR3R4、-COOR3、-CONR3R4、-NR3COR4、-COR3、-OCOR3、-SONR3R4、-SO2NR3R4、-NR3SOR4、-NR3SO2R4, or-Si (R 3)(R4)(R5).
  17. The compound of claim 1, wherein R 1 is selected from unsubstituted alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl, and alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxyl, optionally substituted alkyl, optionally substituted cycloalkyl, or optionally substituted alkoxy.
  18. The compound of claim 1, wherein R 1 is selected from alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, and alkyl, cycloalkyl, or alkoxy substituted with substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl, or alkoxy;
    Preferably, R 1 is selected from the group consisting of alkyl, alkenyl, cycloalkyl, heterocyclyl, or aryl substituted with one or more substituents independently selected from deuterium, fluoro, chloro, bromo, iodo, hydroxy, mercapto, amino, cyano, nitro, alkyl, cycloalkyl, alkoxy, alkylamino, carboxy, haloalkyl, haloalkoxy.
  19. The compound of claim 1, wherein R 3、R4 and R 5 are independently selected from hydrogen, deuterium, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted C 6-10 aryl, or optionally substituted 5-6 membered heteroaryl;
    Preferably, R 3、R4 and R 5 are independently selected from hydrogen, deuterium, unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, and alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxyl, optionally substituted alkyl, optionally substituted cycloalkyl or optionally substituted alkoxy.
  20. The compound of claim 1, wherein R 3、R4 and R 5 are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl substituted with one or more substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, and alkyl, cycloalkyl, or alkoxy substituted with substituents independently selected from deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, carboxy, alkyl, cycloalkyl, or alkoxy;
    Preferably, R 3、R4 and R 5 are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl substituted with one or more substituents independently selected from deuterium, fluoro, chloro, bromo, iodo, hydroxy, mercapto, amino, cyano, nitro, alkyl, cycloalkyl, alkoxy, alkylamino, carboxy, haloalkyl, haloalkoxy.
  21. The compound of claim 1, wherein R 1 is selected from deuterium, halogen, hydroxy, amino, cyano, alkyl, alkenyl, cycloalkyl OR aryl (e.g., phenyl) unsubstituted OR substituted with halogen, OR-OR 3, preferably R 3 is independently selected from alkyl (e.g., C 1-6 alkyl) unsubstituted OR substituted with halogen;
    Preferably, R 1 is selected from fluorine, chlorine, bromine, iodine, cyano, methyl, tert-butyl, methoxy.
  22. The compound of claim 1, wherein n is 0, 1 or 2.
  23. The compound of claim 1, wherein R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 cycloalkyl, Optionally substituted 4-6 membered heterocyclyl 、-ORa、-SRa、-NRaRb、-COORa、-CONRaRb、-NRbCORa、-CORa、-OCORa、-SONRaRb、-SO2NRaRb、-NRbSORa、-NRbSO2Ra or-Si (R a)(Rb)(Rc), wherein R a、Rb and R c are independently selected from hydrogen, Deuterium, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, An optionally substituted 4-6 membered heterocyclyl, an optionally substituted C 6-10 aryl or an optionally substituted 5-6 membered heteroaryl, and the substituents in said optional substitution in R X are selected from one or more of hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, alkyl (e.g. C 1-6 alkyl), alkyl, Alkenyl (e.g., C 2-6 alkenyl), alkynyl (e.g., C 2-6 alkynyl), cycloalkyl (e.g., C 3-6 cycloalkyl), heterocyclyl (e.g., 4-6 membered heterocyclyl), aryl (e.g., C 6-10 aryl), or heteroaryl (e.g., 5-6 membered heteroaryl).
  24. The compound of claim 1, wherein R X is independently selected from hydrogen, deuterium, halogen, hydroxy, mercapto, amino, cyano, nitro, optionally substituted alkyl, -OR a、-SRa, and-NR aRb, wherein R a and R b are independently selected from hydrogen, deuterium, and optionally substituted alkyl;
    Preferably, R X is independently selected from unsubstituted alkyl (e.g., C 1-6 alkyl) or alkyl substituted with deuterium or halogen (e.g., C 1-6 alkyl).
  25. The compound of claim 1, wherein R a and R b are independently selected from unsubstituted alkyl (e.g., C 1-6 alkyl) or alkyl substituted with deuterium or halogen (e.g., C 1-6 alkyl).
  26. The compound of claim 1, wherein R X is independently selected from hydrogen, deuterium, halogen, hydroxy, amino, cyano, alkyl (e.g., C 1-4 alkyl such as methyl, ethyl, propyl, isopropyl, t-butyl), haloalkyl, deuterated alkyl, -OR a、-SRa, OR-NR aRb, wherein R a and R b are independently selected from hydrogen, deuterium, alkyl (e.g., C 1-4 alkyl such as methyl OR ethyl), deuterated alkyl, OR haloalkyl;
    Preferably, R X is independently selected from hydrogen, deuterium, fluorine, chlorine, bromine, iodine, hydroxyl, amino, cyano, methyl, ethyl, isopropyl, tert-butyl, trifluoromethyl, deuteroalkyl, methoxy, ethoxy, methylthio, methylamino, or dimethylamino.
  27. The compound of claim 1, wherein X 1、X2、X3 and X 4 are independently C (R X), preferably X 1、X2、X3 and X 4 are CH.
  28. The compound of claim 1, wherein one (e.g., X 1、X2、X3 or X 4) or two (e.g., X 1 and X 3) of X 1、X2、X3 and X 4 are independently C (R X), wherein R X is as defined in claim 1 but does not include hydrogen and the remainder are CH, or
    One (e.g., X 1、X2、X3 or X 4) or two (e.g., X 1 and X 3;X2 and X 4) of X 1、X2、X3 and X 4 are independently N, the remainder are independently C (R X), wherein R X is as defined in claim 1.
  29. The compound of claim 1, wherein one of X 1、X2、X3 and X 4 is N, the remainder are CH.
  30. The compound of claim 1, wherein the compound is selected from the group consisting of:
  31. A pharmaceutical composition comprising as active ingredient a compound of formula (I) according to any one of claims 1 to 30 and stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates or isotopically labeled analogues thereof.
  32. The pharmaceutical composition according to claim 31, further comprising an additional drug (such as a small molecule, polypeptide, nucleic acid, antibody-coupled drug) for the prevention and/or treatment of cancer or tumor, and/or an additional therapeutic agent for the treatment of inflammatory, autoimmune and immune-mediated diseases.
  33. The pharmaceutical composition of claim 32, wherein the additional agents for preventing and/or treating cancer or tumor include, but are not limited to: cell signaling inhibitors, chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozotocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin C, ixabepilone, tamoxifen, flutamide, gonadorelin analog, megestrol, prednisone, dexamethasone, methylprednisolone, thalidomide, interferon alpha calcium folinate, sirolimus lipidated, everolimus, afatinib, alisertib, amuvatinib, apatinib, axitinib, bortezomib, bosutinib, britinib, cabatinib, ceridinib, crenolanib, crizotinib, dabrafenib, dacatinib, danatinib, dasatinib, multi-vitamin tinib, erlotinib, foretinib, ganetespib, gefitinib, ibrutinib, enotinib, imatinib, inipanib, lapatinib, lenvatinib, linifanib, linsitinib, masatinib, momelotinib, mo Tisha, lenatinib, nilatinib, oprozomib, olatinib, pazopanib, pictilisib, ponatinib, quinitinib, regafil, rigosertib, lu Kapa, ruatinib, ceritinib, calitinib, saridegib, soratinib, lenatinib, tiratinib, tivantinib, tivozanib, tofacitinib, trimetanib, vandetanib, valicarb, vemurafenib, volasertib, alemtuzumab, bevacizumab, belantol Shan Kangwei statin, katuzumab, cetuximab, denomumab, gemtuzumab, ipilimumab, nituzumab, ofatuzumab, panitumumab, rituximab, tositumomab, trastuzumab, PI3K inhibitor, CSF1R inhibitor, A2A and/or A2B receptor antagonist, IDO inhibitor, anti-PD-1 antibody, anti-PD-L1 antibody, g3 antibody, TIM-3 antibody, TIGIT antibody, CD47 antibody, CLAUDIN 18.2.2 antibody, anti-CTLA-4 antibody, or any combination thereof.
  34. The pharmaceutical composition of claim 32, wherein the antibody conjugated drug comprises, but is not limited to, enmeflozumab, veltuzumab, oxepituzumab, vidipine, golimumab Sha Tuozhu, de Lu Tikang-trastuzumab, or any combination thereof.
  35. The pharmaceutical composition of claim 32, wherein the additional therapeutic agent for the treatment of inflammatory, autoimmune, and immune-mediated diseases is selected from the group consisting of steroidal drugs (e.g., prednisone, hydropannisone, methyl hydropaniture, cortisone, hydroxy cortisone, betamethasone, dexamethasone, and the like), methotrexate, leflunomide, anti-tnfα agents (e.g., etanercept, infliximab, ada Li Shan anti, and the like), calcineurin inhibitors (e.g., tacrolimus, pimecrolimus, and the like), antihistamines (e.g., diphenhydramine, hydroxyzine, loratadine, ebastine, ketotifen, cetirizine, levocetirizine, fexofenadine, and the like), or any combination thereof.
  36. Use of a compound of formula (I) according to any one of claims 1-30, as well as stereoisomers, geometric isomers, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates or isotopically-labeled analogues thereof or of a pharmaceutical composition according to any one of claims 31-35, for the preparation of a medicament for the prevention and/or treatment of a disease mediated by ENPP 1.
  37. The use of claim 36, wherein the ENPP 1-mediated disease is selected from cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease, or immune-mediated disease.
  38. The use of claim 37, wherein the cancers and tumors comprise: skin cancer, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, lung cancer, bone cancer, brain cancer, neuroblastoma, rectal cancer, colon cancer, familial adenomatous polyposis, hereditary non-polyposis colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, hypopharynx cancer, tongue cancer, salivary gland cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, renal parenchymal cancer, cervical cancer, uterine body cancer, endometrial cancer, choriocarcinoma, testicular cancer, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors hodgkin's lymphoma, non-hodgkin's lymphoma, burkitt's lymphoma, leukemia, acute Lymphoblastic Leukemia (ALL), chronic Lymphocytic Leukemia (CLL), acute Myelogenous Leukemia (AML), chronic Myelogenous Leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gall bladder carcinoma, bronchogenic carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, multiple myeloma, basal cell carcinoma, teratoma, retinoblastoma, choriocarcinoma, seminoma, rhabdomyosarcoma, craniopharyngeal tubular carcinoma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, ewing's sarcoma, and plasmacytoma.
  39. The use of claim 37, wherein the inflammatory, autoimmune, and immune-mediated diseases include arthritis, rheumatoid arthritis, spinal arthritis, gouty arthritis, osteoarthritis, juvenile arthritis, other arthritic conditions, lupus, systemic Lupus Erythematosus (SLE), skin-related diseases, psoriasis, eczema, dermatitis, allergic dermatitis, pain, pulmonary diseases, pulmonary inflammation, adult Respiratory Distress Syndrome (ARDS), pulmonary sarcoidosis, chronic pulmonary inflammatory diseases, chronic Obstructive Pulmonary Disease (COPD), cardiovascular diseases, atherosclerosis, myocardial infarction, congestive heart failure, myocardial ischemia reperfusion injury, inflammatory bowel disease, crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, sjogren's syndrome, autoimmune thyroid diseases, urticaria (rubella), multiple sclerosis, scleroderma, organ transplant rejection, xenografts, idiopathic Thrombocytopenic Purpura (ITP), parkinson's disease, alzheimer's disease, pelvic inflammatory disease, diabetes-related diseases, allergies, allergic diseases, sinus-sinus conditions, and allergic diseases.
CN202480009595.9A 2023-03-31 2024-03-29 Heteroaromatic ring compound and its preparation method and use Pending CN120603829A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202310334779X 2023-03-31
CN202310334779 2023-03-31
PCT/CN2024/084813 WO2024199444A1 (en) 2023-03-31 2024-03-29 Heteroaromatic ring compound, preparation method therefor, and use thereof

Publications (1)

Publication Number Publication Date
CN120603829A true CN120603829A (en) 2025-09-05

Family

ID=92903373

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202480009595.9A Pending CN120603829A (en) 2023-03-31 2024-03-29 Heteroaromatic ring compound and its preparation method and use

Country Status (2)

Country Link
CN (1) CN120603829A (en)
WO (1) WO2024199444A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2483496A1 (en) * 2002-04-26 2003-11-06 Warner-Lambert Company Llc Inhibitors of checkpoint kinases (wee1 and chk1)
WO2007081755A2 (en) * 2006-01-09 2007-07-19 Metabasis Therapeutics, Inc. Indole-benzimidazole and indazole inhibitors of tyrosine phosphatases
FR2928645A1 (en) * 2008-03-14 2009-09-18 Sanofi Aventis Sa NOVEL CARBAZOLE DERIVATIVES INHIBITORS OF HSP90, COMPOSITIONS CONTAINING SAME AND USE THEREOF
WO2011011186A2 (en) * 2009-07-22 2011-01-27 The Board Of Trustees Of The University Of Illinois Hdac inhibitors and therapeutic methods using the same
SK288508B6 (en) * 2013-10-15 2017-10-03 Ústav Experimentálnej Farmakológie A Toxikológie Sav Use of 5-carboxymethyl-3-mercapto-1,2,4-triazino-[5,6-b]indoles and pharmaceutical preparation containing them
KR20230171634A (en) * 2022-06-14 2023-12-21 주식회사 레고켐 바이오사이언스 Ectonucleotide pyrophosphatase-phosphodiesterase-1 inhibitors and pharmaceutical compositions comprising the same

Also Published As

Publication number Publication date
WO2024199444A1 (en) 2024-10-03

Similar Documents

Publication Publication Date Title
AU2018352142B2 (en) Imidazo-pyridine compounds as PAD inhibitors
ES2670416T3 (en) Imidazo [4,5-c] quinolin-2-one compounds and their use in the treatment of cancer
ES2853924T3 (en) Derivatives of 8- [6- [3- (amino) propoxy] -3-pyridyl] 1-isopropylimidazo [4,5-c] quinolin-2-one as selective modulators of mutated ataxia telangiectasia kinase (atm) for cancer treatment
CN104011052B (en) Compound
ES2738123T3 (en) Heterocyclic sulfur-containing derivative that has β-secretase inhibitory activity
TW202003511A (en) Heterocyclic compounds as immunomodulators
KR20220061958A (en) Heterobicyclic amides as inhibitors of CD38
PT2719699E (en) Certain triazolopyrazines, compositions thereof and methods of use therefor
ES2686747T3 (en) 4,5,6,7-tetrahydro-pyrazolo [1,5-a] pyrimidine derivatives and substituted 2,3-dihydro-1H-imidazo [1,2-b] pyrazole derivatives as ROS1 inhibitors
AU2023237198A1 (en) Inhibitors of KEAP1-Nrf2 protein-protein interaction
TW201731841A (en) 6-aryl or 6-fluorenyl-tetra-morpholin-4-yl-pyridyl-2-ketone
TW202446371A (en) 2-azabicyclo[2.2.1]heptane kras inhibitors
AU2014295101A1 (en) New isoindoline or isoquinoline compounds, a process for their preparation and pharmaceutical compositions containing them
TWI644916B (en) Piperazine dipyridine compound
ES3042412T3 (en) Azaquinolines as inhibitors of cd38
ES3019235T3 (en) Heterocyclic compounds capable of activating sting
JPWO2019189555A1 (en) Heterocyclic compound
CN116023368A (en) CRBN immunomodulator
AU2012363557A1 (en) Novel imidazopyridine derivatives as a tyrosine kinase inhibitor
CN112912142B (en) 5-Azaindazole derivatives as adenosine receptor antagonists
CN119384420A (en) New PAR-2 inhibitors
ES2902346T3 (en) Azaindolylpyridone and diazaindolylpyridone compounds
KR20160019492A (en) Prodrug derivatives of substituted triazolopyridines
CN120603829A (en) Heteroaromatic ring compound and its preparation method and use
JP4599164B2 (en) Pyridindrone derivatives substituted at the 3-position by a heterocyclic group, their production and their therapeutic use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination