CN111606904A - Azaindole compound and application thereof - Google Patents

Azaindole compound and application thereof Download PDF

Info

Publication number
CN111606904A
CN111606904A CN202010265070.5A CN202010265070A CN111606904A CN 111606904 A CN111606904 A CN 111606904A CN 202010265070 A CN202010265070 A CN 202010265070A CN 111606904 A CN111606904 A CN 111606904A
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
stereoisomer
substituted
acceptable salt
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.)
Granted
Application number
CN202010265070.5A
Other languages
Chinese (zh)
Other versions
CN111606904B (en
Inventor
余细勇
蓝辉耀
胡文辉
杨忠金
武楠楠
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.)
Guangzhou Medical University New Drug Manufacturing Co ltd
Guangzhou Medical University
Original Assignee
Guangzhou Medical University New Drug Manufacturing Co ltd
Guangzhou Medical University
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 Guangzhou Medical University New Drug Manufacturing Co ltd, Guangzhou Medical University filed Critical Guangzhou Medical University New Drug Manufacturing Co ltd
Priority to CN202010265070.5A priority Critical patent/CN111606904B/en
Publication of CN111606904A publication Critical patent/CN111606904A/en
Application granted granted Critical
Publication of CN111606904B publication Critical patent/CN111606904B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides an azaindole compound with a structure shown in a formula (I) and application thereof. The azaindole compound can effectively inhibit SMAD 3-phosphorylation, has good antitumor activity in a rat LCC-induced tumor model, and compared with the compound SIS3, part of the azaindole compound provided by the invention has remarkably improved water solubility. The azaindole compound is hopeful to become an effective medicament for preventing and treating tumors.
Figure DDA0002440957960000011

Description

Azaindole compound and application thereof
Technical Field
The invention relates to the technical field of pharmaceutical chemistry, and particularly relates to an azaindole compound and application thereof.
Background
Tumor growth and metastasis are the leading cause of death in cancer patients. Transforming growth factor beta (TGF-beta) exerts oncogenic effects during the invasive and metastatic steps of tumors, inhibition of TGF-beta may cause side effects due to its pleiotropic and complex functions, while SMAD3, the downstream mediator of TGF-beta signaling, contains DNA binding domains and binds directly to promoters in target genes to regulate transcription, but SMAD2 and SMAD4 do not. In addition, SMAD3 has the ability to modulate the expression of key target proteins in tumor immunotherapy, e.g., SMAD3 can up-regulate the expression of programmed death receptor 1(PD-1) on T cells, but SMAD2 cannot up-regulate the expression of PD-1; silencing of the target protein SMAD3 can inhibit the growth and metastasis of tumor cells by enhancing the anti-tumor activity of NK cells and neutrophils in the tumor microenvironment. Therefore, the development of potent and low-toxicity drugs to selectively inhibit SMAD3 protein is a potential anti-tumor strategy.
Small molecule compounds that selectively target inhibition of SMAD3 are still very rare, and therefore, it is of great interest to find more types of selective inhibitors of SMAD3 protein.
Disclosure of Invention
Based on the above, the invention provides a novel azaindole compound which can selectively inhibit SMAD 3-phosphorylation but not inhibit SMAD2, and can improve the effect by improving the proliferation of NK cells in a tumor microenvironment, and the compound has the potential to become a small molecular drug for tumor immunotherapy.
The technical scheme is as follows:
an azaindole compound having a structure shown in formula (I) or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a prodrug molecule thereof:
Figure BDA0002440957940000011
wherein:
d is selected from: C. n; and when D is N, R2Is absent;
a and B are respectively and independently selected from: n, CR3
R1Selected from: H. c1-C6Alkyl radical, C6-C10Aryl radical, C3-C8Cycloalkyl, halogen, CN;
R2selected from: H. c1-C6An alkyl group;
each R3Each independently selected from: H. c1-C6Alkyl, halogen, CN;
R4selected from:
Figure BDA0002440957940000021
R5each R5aAnd R5bEach independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8A cycloalkyl group;
each R6Each independently selected from:
Figure BDA0002440957940000022
R7selected from: H. c1-C6Alkyl, one or more R13Substituted C6-C10Aryl, one or more R13Substituted 6-10 membered heteroaryl;
each R8Each independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8Cycloalkyl, hydroxy, C1-C6Alkoxy radical, C1-C6Alkoxy-substituted C1-C6An alkoxy group;
each R9Each independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8Cycloalkyl, hydroxy, C1-C6Alkoxy radical, C1-C6Alkoxy-substituted C1-C6An alkoxy group;
R10selected from: H. c1-C6An alkyl group;
R11selected from: hydroxy, C1-C6Alkoxy, -N (R)14)(R15);
R12Selected from: H. c1-C6Alkyl, one or more R13Substituted C6-C10Aryl, one or more R13Substituted 6-10 membered heteroaryl;
each R13Each independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8Cycloalkyl, hydroxy, C1-C6Alkoxy radical, C1-C6Alkoxy-substituted C1-C6An alkoxy group;
R14and R15Each independently selected from: H. c1-C6Alkyl, amino substituted C1-C6Alkyl radical、C1-C6Alkylamino substituted C1-C6An alkyl group;
n is selected from: 1.2, 3 and 4;
m is selected from: 0.1, 2,3, 4;
p is selected from: an integer between 0 and 7;
q is selected from: 1.2, 3 and 4;
y is selected from: 1.2, 3,4, 5;
and, when D is C, A is N, B is CR3,R1Is phenyl, R2Is composed of
Figure BDA0002440957940000031
When R is8Is other than C1-C6An alkoxy group.
In some of these embodiments, D is C, A is N, B is CR4
In some of these embodiments, the azaindoles have the structure of formula (II):
Figure BDA0002440957940000032
in some of these embodiments, R5Is H.
In some of these embodiments, n is selected from: 1. 2; m is selected from: 1 or 2.
In some of these embodiments, R7Selected from: one or more R13Substituted phenyl, one or more R13A substituted six-membered nitrogen-containing heteroaryl.
In some of these embodiments, each R13Each independently selected from: H. c1-C6Alkyl radical, C1-C6An alkoxy group.
In some of these embodiments, R7Selected from:
Figure BDA0002440957940000033
in some of these embodiments, the azaindoles have the structure of formula (III):
Figure BDA0002440957940000034
in some of these embodiments, the azaindoles have the structure of formula (IV):
Figure BDA0002440957940000041
in some of these embodiments, each R8Each independently selected from: H. hydroxy, C1-C3Alkoxy radical, C1-C3Alkoxy-substituted C1-C3An alkoxy group.
In some of these embodiments, each R8Each independently selected from: hydroxyl, methoxy substituted ethoxy.
In some of these embodiments, R8Are all hydroxyl groups.
In some of these embodiments, the azaindoles have the structure of formula (V):
Figure BDA0002440957940000042
in some of these embodiments, R9Selected from: H. halogen, hydroxy;
R10selected from: H. c1-C3An alkyl group;
R11selected from: c1-C6An alkoxy group.
In some of these embodiments, the azaindoles have the structure of formula (VI):
Figure BDA0002440957940000043
in some of these embodiments, R12Selected from: one or more R13Substituted C6-C10A heteroaryl group; each R13Each independently selected from: H. c1-C6An alkyl group.
In some of these embodiments, R12Selected from:
Figure BDA0002440957940000051
in some of these embodiments, R5aAnd R5bEach independently selected from: H. a halogen.
In some of these embodiments, R5bIs H, R5aIs F.
In some of these embodiments, R1Selected from: H. c1-C3Alkyl radical, C6-C10Aryl radical, C3-C6A cycloalkyl group.
In some of these embodiments, R1Selected from: H. phenyl, cyclopropyl.
In some of these embodiments, R2Selected from: c1-C3An alkyl group.
In some of these embodiments, R3Is H.
The invention also provides application of the azaindole compound or pharmaceutically acceptable salt thereof or stereoisomer thereof or prodrug molecule thereof.
The specific technical scheme is as follows:
the azaindole compound or the pharmaceutically acceptable salt thereof or the stereoisomer thereof or the prodrug molecule thereof is applied to the preparation of the SMAD3 inhibitor.
The azaindole compound or the pharmaceutically acceptable salt thereof or the stereoisomer thereof or the prodrug molecule thereof can be applied to the preparation of drugs for preventing and/or treating tumors.
In some of these embodiments, the tumor is: lung cancer, melanoma, breast cancer, and liver cancer.
The invention also provides a medicinal composition for preventing and/or treating tumors.
The specific technical scheme is as follows:
a pharmaceutical composition for preventing and/or treating tumors comprises an active ingredient and a pharmaceutically acceptable carrier, wherein the active ingredient comprises the azaindole compound or the pharmaceutically acceptable salt thereof or the stereoisomer thereof or prodrug molecules thereof.
The research of the invention discovers a series of novel azaindole compounds which can effectively and selectively inhibit SMAD 3-phosphorylation but not inhibit SMAD2, have good anti-tumor activity in a rat LCC-induced tumor model, and can improve the anti-tumor effect by improving the proliferation of NK cells in a tumor microenvironment.
In addition, the water solubility of the drug compound is a large reason for influencing the development of the drug preparation, and if the poor water solubility of the compound can cause low bioavailability, unstable metabolism (drug discovery. today 2004,9, 1020-. The azaindole compound or the salt thereof has better water solubility, and the solubility of HCl salt of part of compounds in water is more than 20mg/mL, even reaches 94mg/mL or higher, which is favorable for improving the bioavailability of the compounds and is favorable for the later preparation development. The azaindole compound is hopeful to become an effective medicament for preventing and treating tumors.
Drawings
Figure 1 is a graph of the results of selective inhibition of TGF- β dependent phosphorylation of SMAD3 by compound 16 d.
FIG. 2 is a graph showing the results of Compound 16d in effectively inhibiting the growth of mouse homologous Lewis Lung Carcinoma (LLC).
Fig. 3 is a graph showing the results of compound 16d in inhibiting cancer progression by enhancing NK cell aggregation in LLC model mice.
Detailed Description
In the compounds of the present invention, when any variable (e.g., R, etc.) occurs more than one time in any constituent, its definition in each occurrence is independent of its definition in every other occurrence. Also, combinations of substituents and variables are permissible only if such combinations result in stable compounds. The line drawn from a substituent into the ring system indicates that the indicated bond can be attached to any ring atom that can be substituted. If the ring system is polycyclic, it means that such a bond is only attached to any suitable carbon atom of the adjacent ring. It is to be understood that substituents and substitution patterns on the compounds of the present invention may be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by those skilled in the art and by the methods set forth below from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these groups may be on the same carbon atom or on different carbon atoms, so long as the structure is stable.
The term "alkyl" as used herein is intended to include both branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, "C1-C6Alkyl radical "middle" C1-C6The definition of "includes groups having 1,2,3,4, 5 or 6 carbon atoms in a linear or branched arrangement. For example, "C1-C6Alkyl "specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, pentyl, hexyl.
The term "cycloalkyl" refers to a monocyclic saturated aliphatic hydrocarbon group having the specified number of carbon atoms. For example, "cycloalkyl" includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
The term "alkoxy" refers to a group having the structure-O-alkyl, such as-OCH3、-OCH2CH3、-OCH2CH2CH3、-O-CH2CH(CH3)2、-OCH2CH2CH2CH3、-O-CH(CH3)2And the like.
The term "heteroaryl" refers to an aromatic ring containing 1 or more heteroatoms selected from O, N or S, and heteroaryl groups within the scope of the present invention include, but are not limited to: quinolyl, pyrazolyl, pyrrolyl, thienyl, furyl, pyridyl, pyrimidinyl, pyrazinyl, triazolyl, imidazolyl, oxazolyl, isoxazolyl, pyridazinyl; "heteroaryl" is also understood to include any N-oxide derivative of a nitrogen-containing heteroaryl group.
The term "substituted" refers to the replacement of a hydrogen radical in a particular structure with a radical of a specified substituent.
As understood by those skilled in the art, "halo" or "halo" as used herein means chloro, fluoro, bromo, and iodo.
Unless otherwise defined, alkyl, cycloalkyl, aryl, heteroaryl, and heterocycloalkyl substituents may be unsubstituted or substituted. E.g. C1-C6Alkyl groups may be substituted with one, two or three substituents selected from OH, halogen, alkoxy, dialkylamino or heterocyclyl, e.g. morpholinyl, piperidinyl and the like.
The invention includes the free forms of the compounds of formulae I-VI, as well as pharmaceutically acceptable salts and stereoisomers thereof. Some specific exemplary compounds herein are protonated salts of amine-based compounds. The term "free form" refers to the amine compound in a non-salt form. Included pharmaceutically acceptable salts include not only exemplary salts of the particular compounds described herein, but also all typical pharmaceutically acceptable salts of the free forms of the compounds of formulas I-VI. The free form of a particular salt of the compound may be isolated using techniques known in the art. For example, the free form can be regenerated by treating the salt with a dilute aqueous solution of a suitable base, such as a dilute aqueous NaOH solution, a dilute aqueous potassium carbonate solution, dilute aqueous ammonia, and a dilute aqueous sodium bicarbonate solution. The free forms differ somewhat from their respective salt forms in certain physical properties, such as solubility in polar solvents, but for the purposes of the invention such acid and base salts are otherwise pharmaceutically equivalent to their respective free forms.
Pharmaceutically acceptable salts of the invention can be synthesized from compounds of the invention containing a basic or acidic moiety by conventional chemical methods. In general, salts of basic compounds are prepared by ion exchange chromatography or by reaction of the free base with a stoichiometric amount or excess of an inorganic or organic acid in the form of the desired salt in an appropriate solvent or combination of solvents. Similarly, salts of acidic compounds are formed by reaction with suitable inorganic or organic bases.
Thus, pharmaceutically acceptable salts of the compounds of the present invention include the conventional non-toxic salts of the compounds of the present invention formed by the reaction of a basic compound of the present invention and an inorganic or organic acid. For example, conventional non-toxic salts include those derived from inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, and the like, as well as those prepared from organic acids such as acetic acid, propionic acid, succinic acid, glycolic acid, stearic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, pamoic acid, maleic acid, hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, sulfanilic acid, 2-acetoxy-monobenzoic acid, fumaric acid, toluenesulfonic acid, methanesulfonic acid, ethane disulfonic acid, oxalic acid, isethionic acid, trifluoroacetic acid, and the like.
If a compound of the invention is acidic, an appropriate "pharmaceutically acceptable salt" refers to a salt prepared by a pharmaceutically acceptable non-toxic base including inorganic and organic bases. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc, and the like. Particularly preferred are ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases including salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins such as arginine, betaine, caffeine, choline, N' -dibenzylethylenediamine, diethylamine, 2-dimethylaminoethanol, aminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, histidine, hydroxycobalamin, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, piperdine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
Berg et al, "Pharmaceutical Salts," j.pharm.sci.' 1977: 66: 1-19 describe in more detail the preparation of the pharmaceutically acceptable salts described above and other typical pharmaceutically acceptable salts.
Since acidic moieties such as carboxyl groups deprotonated in a compound under physiological conditions may be anionic and such charge may then be balanced out by a protonated or alkylated basic moiety such as a quaternary nitrogen atom bearing a cation internally, it should be noted that the compounds of the present invention are potential internal salts or zwitterions.
In one embodiment, the present application provides a method of treating hyperproliferative diseases or conditions, such as tumors, in humans and other mammals, using compounds having the formulas I-VI and pharmaceutically acceptable salts thereof.
In one embodiment, the compounds of the present application and pharmaceutically acceptable salts thereof may be used to treat or control non-small cell lung cancer, lymphoma, esophageal cancer, ovarian cancer, melanoma, cervical cancer, urothelial cancer, pancreatic cancer, breast cancer, liver cancer, gastric cancer, bile duct cancer, leukemia, melanoma, colon cancer, rectal cancer, endometrial cancer, or brain glioma.
Drug metabolites and prodrugs: metabolites of the compounds and pharmaceutically acceptable salts thereof to which this application relates, and prodrugs that can be converted in vivo to the structures of the compounds and pharmaceutically acceptable salts thereof to which this application relates, are also included in the claims of this application.
Combined medication: the compounds of formulae I-VI may be combined with other agents known to treat or ameliorate similar conditions. When administered in combination, the mode of administration and dosage of the original drug is maintained, while the compounds of formulae I-VI are administered simultaneously or subsequently. When the compounds of formulae I-VI are administered concurrently with one or more other drugs, it is preferred to use pharmaceutical compositions containing both one or more known drugs and the compounds of formulae I-VI. Pharmaceutical combinations also include administration of the compounds of formulae I-VI in overlapping time periods with one or more other known agents. When the compounds of formulae I-VI are administered in combination with one or more other drugs, the dosages of the compounds of formulae I-VI or the known drugs may be lower than the dosages when they are administered alone.
Drugs or active ingredients that may be used in combination with the compounds of formulae I-VI include, but are not limited to:
estrogen receptor modulators, androgen receptor modulators, retinal-like receptor modulators, cytotoxins/cytostatics, antiproliferatives, protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protein kinase inhibitors, reverse transcriptase inhibitors, angiogenesis inhibitors, cell proliferation and survival signal inhibitors, drugs that interfere with cell cycle checkpoints and apoptosis inducers, cytotoxic drugs, tyrosine protein inhibitors, EGFR inhibitors, VEGFR inhibitors, serine/threonine protein inhibitors, Bcr-Abl inhibitors, c-Kit inhibitors, Met inhibitors, Raf inhibitors, MEK inhibitors, MMP inhibitors, topoisomerase inhibitors, histidine deacetylase inhibitors, proteasome inhibitors, CDK inhibitors, Bcl-2 family protein inhibitors, MDM2 family protein inhibitors, inhibitors of apoptosis, inhibitors of tumor growth, and the like, IAP family protein inhibitors, STAT family protein inhibitors, PI3K inhibitors, AKT inhibitors, integrin blockers, interferon-alpha, interleukin-12, COX-2 inhibitors, p53, p53 activators, VEGF antibodies, EGF antibodies, and the like.
In one embodiment, the pharmaceutical or active ingredients that may be used in combination with the compounds of formulas I-VI include, but are not limited to: aldesleukin, alendronic acid, interferon, atrazine, allopurinol sodium, palonosetron hydrochloride, hexamethylmelamine, aminoglutethimide, amifostine, amrubicin, ambrolidine, anastrozole, dolasetron, aranesp, arglabin, arsenic trioxide, anoxin, 5-azacytidine, azathioprine, bacillus calmette or tide bacillus calmette, betadine, betamethasone acetate, betamethasone sodium phosphate preparation, bexarotene, bleomycin sulfate, bromouroxime, bortezomib, busulfan, calcitonin, alezomab injection, capecitabine, carboplatin, custard, cefesone, simon, daunorubicin, phenylbutyric acid azone, mustard, cladribine, clodronate, cyclophosphamide, alexanide, dacarbazine, actinomycin, dexamethasone, estramustin phosphate, estramustine, dexamethasone, estradiol phosphate, estradiol valerate phosphate, valproate, doxylamine, and mixtures thereof, Dinil interleukin 2, dibume, deslorelin, delazoxan, diethylstilbestrol, tolbutan, docetaxel, doxifluridine, doxorubicin, dronabinol, azulene-166-chitosan complex, eligard, labyrinase, epirubicin hydrochloride, aprepitant, epirubicin, alfafurtine, erythropoietin, eptaplatin, levamisole, estradiol formulations, 17-beta-estradiol, estramustine sodium phosphate, ethinylestradiol, amifostine, hydroxyphosphoric acid, pirimiphoside, etoposide, favuzole, tamoxifen formulations, filgrastim, phenastidine, floxuridine, fluconazole, fludarabine, 5-fluorodeoxyuridine monophosphate, 5-fluorouracil, flumetmesterone, flunomide, fulvestrant, 1-beta-D-arabinofuranosylcytisidine-5 '-stearoyl-5' -stearoyl phosphate, flutamide, fluvastatin, Fotemustine, fulvestrant, gamma globulin, gemcitabine, gemtuzumab ozogamicin, imatinib mesylate, carmustine wafer capsule, goserelin, glanesilong hydrochloride, histrelin, and meclizine, hydrocortisone, erythro-hydroxynonyladenine, hydroxyurea, temin bemomab, idarubicin, ifosfamide, interferon alpha 2A, interferon alpha 2B, interferon alpha nl, interferon alpha n3, interferon beta, interferon gamma la, interleukin 2, intron A, iressa, irinotecan, kateride, lentinan sulfate, letrozole, leucovorin, leuprolide, leuprorelin, levamisole acetate, levamisole, calcium levofolinate, sodium levothyroxine preparation, Lomustine, lonidamine, dronabinol, mechlorethamine, mecobalamin, medroxyprogesterone acetate, megestrol acetate, melphalan, esterified estrogen, 6-ryopurine, mesna, methotrexate, methyl aminoacetonate, miltefosine, milbemycin, mitomycin C, mitotane, mitoxantrone, trilostane, doxorubicin citrate liposome, nedaplatin, pegylated filgrastim, omprex interleukin, neupogen, nilutamide, tamoxifen, NSC-631570, recombinant human interleukin 1-beta, octreotide, ondansetron hydrochloride, hydrocortisone oral solution, oxaliplatin, paclitaxel, prednisone sodium phosphate formulation, pemetrexed, domethacin, pentostatin, streptolysin formulation, pilocarpine hydrochloride, bordeauxins, plicamycin, pemetrexen, phenomycin, and prednisone, Spiprantelone, prednisone, pemetrexed, procarbazine, recombinant human erythropoietin, raltitrexed, ribi, rhenium-186 etidronate, merosal, dygulin-A, romopeptide, pilocarpine hydrochloride tablet, octreotide, samustine, semustine, Sizopyran, sobuzosin, Succinum methylprednisolone, Pafoscarnet, Stemonaccid, streptozocin, strontium chloride-89, levothyroxine sodium, tamoxifen, tamsulosin, tasolomide, Tastolactone, Tetharidin, Tecetithiozine, temozolomide, teniposide, testosterone propionate, megestrol, thioguanine, thiotepa, thyrotropine, Teluzole, topotecan, toremifene, tositumomab, Suzuzumab, Ottoepirubicin, Vavea tablet, methotrexate, trimetrexamine, triptorelin, trexate, troglib, troglitazone, trexate, triptorelin pamoate, eufordine, uridine, valrubicin, vesnarinone, vinblastine, vincristine, vinblastine amide, vinorelbine, vilulizine, dexpropinimine, neat stastine ester, pindoline, paclitaxel protein stabilizing formulations, acolbifene, interferon r-lb, affinitak, aminopterin, azoxifene, aspristil, atamestane, atrasentan, BAY43-9006, avastin, CCI-779, CDC-501, Celopabrol, cetuximab, clinacatto, cyproterone acetate, decitabine, DN-101, doxorubicin-MTC, dSLIM, dutasteride, eocharin, eflornithine, irinotecan, fenretinide, histamine dihydrochloride, histrelin hydrogel implants, bamine-DOTMP, holmium-phosphate, ibandron, gamma-gamma, interferon-containing, loxapine, PEG-L-1582, hematoporphyrin-L-2, and leupeptin, Lancet, lasofoxifene, libra, lonafamib, mirtaxifene, minodronate, MS-209, liposomal MTP-PE, MX-6, nafarelin, nemorubicin, neovastat, nolatrexed, Olimersen, onco-TCS, osidem, paclitaxel polyglutamate, sodium pamoate, PN-401, QS-21, quasicil, R-1549, raloxifene, ranpirnase, 13-cis-tretinoin, satraplatin, seocalcitol, T-138067, tarceva, docosahexanoic acid paclitaxel, thymosin alpha l, gazofuralin, tipifarnib, tirapazamine, TLK-286, toremifene, trans MID-7R, vastada, pravastatin, vatalan, valatib, vinoporfin, vinpocetine, Z-100 and lypocetine or combinations thereof.
The invention is further described in the following examples, which are not intended to limit the scope of the invention.
Example 1: synthesis of Compounds 5a-5j
Figure BDA0002440957940000101
Synthesis of intermediate 4 a:
Figure BDA0002440957940000111
NaH (192mg,4.8mmol) was placed in a reaction flask, nitrogen was replaced, 7mL of anhydrous DMF was added for dissolution, diethyl phosphonoacetate was added under ice-water bath conditions, stirring was carried out for 0.5 hour, a solution of Compound 3a (636mg, 3.973mmol) in 7.5mL of DMF was added dropwise, and stirring was carried out at room temperature for 8 hours. Sampling a sample, adding water to quench the reaction after the reaction is basically finished, stirring for 2 hours, extracting with ethyl acetate for three times, washing with saturated salt water, drying, filtering, concentrating, and purifying by a silica gel column to obtain a yellow solid compound n1(520mg, 60.5%); compound n1(520mg,2.4mmol) was dissolved in THF/MeOH/H2To the O (5mL/8mL/8mL) mixed solution was added NaOH (288mg, 7.2mmol), and the mixture was stirred at room temperature for 4 hours. Sampling the sample point plate, and basically finishing the reaction. HCl (1M) was added to adjust pH 5.0 and filtered to give intermediate 4a (413mg, 85%).
Synthesis of intermediate 4 b:
Figure BDA0002440957940000112
to a solution of compound 3b (300mg, 1.5mmol) in pyridine (10mL) was added malonic acid n2(312mg, 3mmol) and piperidine (1 mL). The mixture was stirred at 110 ℃ for 6 hours. The mixture was then cooled to room temperature and treated with NaHCO3And (4) acidifying the aqueous solution. pH was adjusted to 5 with HCl (1M) and extracted with EA. Washing with saturated brine, anhydrous MgSO4Drying, filtration and concentration gave the crude product 4b as a yellow solid which was used in the next step without further purification (188mg, crude).
Synthesis of intermediate 4 c:
Figure BDA0002440957940000113
the compound 3c (360mg,1.52mmol) and ethyl fluoroacetate (177mg,1.67mmol) were placed in a reaction flask, purged with nitrogen, dissolved with anhydrous DCM, and TiCl was added dropwise4(1M in CH2Cl21.82mL,1.82mmol) was stirred at room temperature for 0.5 h, then TEA (0.42mL,3.02mmol) was added and stirred at room temperature for 2 h. After the reaction was completed, the sample was taken out, EA was extracted three times, and the organic phases were combined, dried, filtered, concentrated, and purified by silica gel column chromatography to obtain compound n3(220mg, 47%).
1H NMR(400MHz,CDCl3)8.46–8.36(m,2H),7.61-7.51(m,3H),7.47–7.38(m,2H),7.22(dd,J=8.0,4.8Hz,1H),6.96(d,J=40.1Hz,1H),4.29(q,J=7.1Hz,2H),3.79(s,3H),1.32(t,J=7.1Hz,3H);13C NMR(100MHz,CDCl3)161.9(d,J=33.5Hz),148.7,144.1(d,J=2.2Hz),143.9,143.8(d,J=257.7Hz),130.8(d,J=18.0Hz),130.7,129.6,129.5,128.8,118.4,117.3(d,J=3.5Hz),111.5(d,J=9.3Hz),104.9(d,J=2.5Hz),61.4,30.1,14.3.
Compound n3(220mg, 0.71mmol) and LiOH. H2O (85mg,2.02mmol) was placed in a reaction flask, THF and water (10mL and 10mL) were added. After stirring at room temperature for 3 hours, a sample was taken and the reaction was essentially complete, HCl acidified to pH6, concentrated in vacuo, EA extracted three times, the organic phases combined and concentrated to give intermediate 4c as an off-white solid (150mg, 71%).
1H NMR(400MHz,DMSO-d6)8.36(d,J=4.4Hz,1H),8.21(d,J=7.8Hz,1H),7.66–7.47(m,5H),7.24(dd,J=7.9,4.6Hz,1H),6.76(d,J=40.5Hz,1H),3.66(s,3H);13C NMR(100MHz,DMSO-d6)162.8(d,J=33.9Hz),148.6,144.4(d,J=255.7Hz),144.3(d,J=2.0Hz),144.2,131.2,130.3(d,J=18.0Hz),130.1,129.6,129.3,117.9,117.8(d,J=3.3Hz),110.7(d,J=9.8Hz),104.1(d,J=2.4Hz),30.2.
Synthesis of intermediate 4 d:
Figure BDA0002440957940000121
a solution of compound 3c (300mg, 1.27mmol), tert-butyl 2-cyanoacetate (198mg, 1.40mmol), piperidine (36mg, 0.42mmol), acetic acid (174mg, 2.90mmol) and toluene (10ml) was placed in a reaction flask and stirred for 15 hours. The sample point plate reaction was substantially complete. With EA (80mL) and saturated NaHCO3Extraction with aqueous solution (30mL), drying, filtration, concentration, and purification by silica gel column chromatography gave compound n4(410mg, 90%) as a white solid.
1H NMR(400MHz,CDCl3)8.82(d,J=8.0Hz,1H),8.47(d,J=4.3Hz,1H),8.08(s,1H),7.63–7.55(m,3H),7.45–7.38(m,2H),7.34(dd,J=8.1,4.7Hz,1H),3.83(s,3H),1.53(s,9H).13C NMR(100MHz,CDCl3)162.9,150.1,149.2,148.8,144.6,131.9,130.8,130.3,128.9,128.5,118.3,117.6,108.1,98.1,82.6,77.4,77.0,76.7,30.5,28.1.
Compound n4(390mg, 1.08mmol) was placed in a reaction flask and 10mL CH was added2Cl2Dissolve, add 3ml of LTFA, and stir at room temperature for 12 hours. The sample point plate reaction was substantially complete. Concentration gave 4d as a yellow solid (311mg, 95%).
1HNMR(400MHz,DMSO-d6)8.70(d,J=7.7Hz,1H),8.51(d,J=4.2Hz,1H),7.98(s,1H),7.71–7.57(m,5H),7.43(dd,J=8.1,4.7Hz,1H),3.78(s,3H).13C NMR(100MHz,DMSO-d6)165.1,151.0,149.2,149.0,145.1,131.5,130.9,129.3,128.4,118.8,118.5,117.2,107.4,96.7,30.8.
Synthesis of intermediate 4 e:
Figure BDA0002440957940000131
intermediate 4e (8.03g, 85%) was prepared starting from compound 3c (8.01g,33.90mmol) according to the procedure for the synthesis of compound 4 a.
1H NMR(400MHz,DMSO-d6)8.45–8.31(m,2H),7.68–7.55(m,5H),7.48(d,J=16.0Hz,1H),7.32(dd,J=7.9,4.8Hz,1H),6.40(d,J=16.0Hz,1H),3.69(s,3H).13C NMR(100MHz,DMSO-d6)168.8,148.8,145.7,144.4,137.7,131.3,130.2,129.6,129.4,129.3,118.3,117.9,114.6,108.0,30.1.HRMS(ESI)calcd for C17H14N2O2(M+H)+279.1128,found279.1128.
Synthesis of intermediate 4 f:
Figure BDA0002440957940000132
intermediate 4f (747mg, 55%) was prepared starting from compound 3d (940.4mg,4.0mmoL) according to the procedure for the synthesis of compound 4 a.1H NMR(400MHz,DMSO-d6)7.94(d,J=7.8Hz,1H),7.65–7.55(m,4H),7.53–7.43(m,3H),7.31(dt,J=24.9,7.5Hz,2H),6.33(d,J=16.0Hz,1H),3.62(s,3H).
Synthesis of intermediate 4 g:
Figure BDA0002440957940000133
starting from compound 3e (300mg, 1.27mmol), intermediate 4g (200mg, crude) was prepared according to the synthetic procedure for compound 4 b.
Synthesis of Compound 5a
Figure BDA0002440957940000141
To a solution of compound 4a (126mg,0.623mmol) and TEA (0.3mL) in DMF (3mL) was added HATU (237mg,0.52mmol), the mixture was stirred at 0 ℃ for 0.5 hour, and 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (120mg,0.623mmol) was added. After stirring at room temperature for 5 hours, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give compound 5a as a white solid (117mg, 50%).
1H NMR(400MHz,CDCl3)8.39(dd,J=4.7,1.4Hz,1H),8.18(d,J=7.4Hz,1H),7.88(d,J=15.3Hz,1H),7.47(s,1H),7.19(dd,J=7.9,4.7Hz,1H),6.89(d,J=15.4Hz,1H),6.66(d,J=7.8Hz,2H),4.79(s,2H),3.89(s,5H),3.86(s,6H),2.87(t,J=28.1Hz,2H).13CNMR(100MHz,CDCl3)166.7,148.8,147.9,143.9,135.8,132.3,128.5,127.3,125.8,124.4,118.5,116.8,112.9,111.3,111.1,109.5,56.1,47.2,44.6,43.8,40.2,31.5.HRMS(ESI)calcd for C22H23N3O3(M+H)+378.1812found 378.1804.
Synthesis of Compound 5b
Figure BDA0002440957940000142
To a solution of compound 4b (200mg,0.826mmol) and 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (140mg, 0.60mmol) in DMF (15mL) was added DIPEA (1mL) and HATU (380mg, 1 mmol). Stirred at room temperature for 16 hours. Sampling a sample point plate, basically finishing the reaction, adding water for dilution, and performing EA extraction. The combined organic phase was washed with saturated brine and then with anhydrous MgSO4Drying, filtration, concentration and purification by silica gel column chromatography gave compound 5b (90mg, 26%) as a yellow solid product.
1H NMR(400MHz,CDCl3)8.36(dd,J=4.7,1.4Hz,1H),8.27(d,J=15.4Hz,1H),8.14(d,J=7.2Hz,1H),7.22-7.14(m,1H),6.88(d,J=15.4Hz,1H),6.75-6.63(m,2H),4.82(s,2H),4.05–3.77(m,11H),3.00-2.80(m,2H),2.01–1.87(m,1H),1.35–1.22(m,2H),0.97–0.85(m,2H).13C NMR(100MHz,CDCl3)167.0,148.3,144.6,143.1,135.7,127.8,127.3,126.1,125.9,124.4,118.4,116.8,112.9,112.7,111.5,111.2,109.5,109.2,56.0,47.2,44.5,43.8,40.1,28.3,6.7,6.3.HRMS(ESI)calcd for C25H27N3O3(M+H)+418.2125,found418.2128.
Synthesis of Compound 5c
Figure BDA0002440957940000151
To a solution of compound 4c (130mg,0.44mmol) and 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (111mg,0.48mmol) in DMF (8mL) were added DIPEA (0.23mL,1.32mmol) and HATU (200mg,0.53 mmol). Stirred at room temperature for 1 hour. Sampling a sample point plate, basically finishing the reaction, adding water for dilution, and performing EA extraction. The combined organic phase was washed with saturated brine and then with anhydrous MgSO4Drying, filtration, concentration and purification by silica gel column chromatography gave the product compound 5c (165mg, 79%) as a white solid.
1H NMR(400MHz,CDCl3)8.43(d,J=4.7Hz,1H),8.37(d,J=7.2Hz,1H),7.58–7.47(m,3H),7.46–7.39(m,2H),7.23(dd,J=7.9,4.8Hz,1H),6.78–6.55(m,3H),4.77–4.61(m,2H),3.91–3.73(m,11H),2.89–2.81(m,2H).13C NMR(100MHz,CDCl3)162.9,162.6,149.6,148.6,147.8,147.8,147.0,143.6,143.1,130.7,130.6,130.4,129.9,129.3,128.8,126.3,124.7,118.4,116.9,111.44,109.7,109.6,109.1,104.9,67.1,56.0,30.0.HRMS(ESI)calcd for C28H26FN3O3(M+H)+472.2031,found 472.2034.
Synthesis of compound 5 d:
Figure BDA0002440957940000152
to compound 4d (182mg,0.6mmol) in CH2Cl2To the solution (5mL) was added oxalyl chloride (228mg,1.8mmol), followed by dropwise addition of DMF and stirring at room temperature for 2 hours. Vacuum concentration, the residue dissolved in THF (10mL), 0 degrees C conditions, adding 6, 7-two methoxy-1, 2,3, 4-four hydrogen isoquinoline hydrochloride (138mg,0.6mmol) and TEA (0.4mL,3mmol) in THF (20mL) solution, at room temperature stirring for 1 hours, EA extraction, saturated saline water washing, drying, concentration, silica gel column chromatography purification to obtain light yellow solid compound 5d (160mg, 56%).
1H NMR(400MHz,CDCl3)8.77(d,J=7.9Hz,1H),8.49(d,J=3.7Hz,1H),7.87(s,1H),7.64–7.54(m,3H),7.49–7.39(m,2H),7.35(dd,J=8.0,4.7Hz,1H),6.65(d,J=2.5Hz,2H),4.97–4.52(m,2H),3.97–3.79(m,11H),3.02-2.81(m,2H).13C NMR(100MHz,CDCl3)163.74,147.86,147.82,147.16,146.87,146.80,143.30,130.55,129.78,129.19,127.94,127.60,125.07,123.30,117.65,116.85,116.52,110.42,108.06,107.10,98.82,76.25,54.98,54.95,29.46,28.66,27.12.HRMS(ESI)calcd for C29H26N4O3(M+H)+479.2078,found479.2079.
Synthesis of compound SIS 3:
Figure BDA0002440957940000161
to a solution of compound 4e (556mg,2mmol) in THF (20mL) was added oxalyl chloride (508mg,4mmol), and the mixture was stirred at room temperature for 2 hours. Vacuum concentration, the residue dissolved in THF (10mL), 0 degrees C conditions, adding 6, 7-two methoxy-1, 2,3, 4-four hydrogen isoquinoline hydrochloride (460mg,2mmol) and TEA (606mg,6mmol) in THF (20mL) solution, at room temperature for 1 hours, EA extraction, saturated saline water washing, drying, concentration, silica gel column chromatography purification to obtain yellow solid compound SIS3(500mg, 55%).
1H NMR(400MHz,CDCl3)8.44(d,J=4.7Hz,1H),8.27(s,1H),7.77(d,J=15.4Hz,1H),7.59–7.49(m,3H),7.45(d,J=7.4Hz,2H),7.28–7.26(m,2H),6.87(d,J=15.4Hz,1H),6.64(s,2H),4.74–4.70(m,2H),3.87–3.78(m,11H),2.84(d,J=20.7Hz,2H).13C NMR(100MHz,CDCl3)166.8,148.9,147.8,144.4,143.6,136.0,130.7,129.9,129.4,128.8,128.3,126.1,118.5,117.1,113.6,111.2,109.5,109.1,56.0,47.1,44.4,43.7,40.0,29.9,29.2,28.2.HRMS(ESI)calcd for C28H27N3O3(M+H)+454.2125,found 454.2126.
Synthesis of compound 5 f:
Figure BDA0002440957940000171
compound SIS3(220mg, 0.48mmol) and Pd/C (22mg, 10% w/w) were placed in a reaction flask, replaced with hydrogen, dissolved with MeOH (20mL), and stirred at room temperature overnight. The resulting mixture was filtered and the filtrate was evaporated to give the crude product which was purified by silica gel column chromatography to give compound 5f (120mg, 54%) as a white solid.
1H NMR(400MHz,CDCl3)8.34(d,J=4.3Hz,1H),7.95(d,J=7.7Hz,1H),7.53–7.40(m,5H),7.11–7.03(m,1H),6.59(d,J=8.5Hz,1H),6.55(s,0.5H),6.33(s,0.5H),4.59(s,1H),4.29(s,1H),3.85–3.82(m,6H),3.75(t,J=5.9Hz,1H),3.70–3.66(m,3H),3.39(t,J=5.8Hz,1H),3.14–3.07(m,2H),2.71–2.53(m,4H).13C NMR(100MHz,CDCl3)171.3,171.2,148.1,147.9,147.8,147.6,147.6,142.8,138.2,138.2,131.3,130.4,128.7,128.6,128.5,126.9,126.9,125.7,125.4,124.1,120.2,115.5,115.4,111.5,111.1,110.4,110.3,109.4,108.7,56.0,46.9,43.9,43.2,39.6,35.2,34.9,29.5,28.8,27.9,20.4,20.3.HRMS(ESI)calcd for C28H29N3O3(M+H)+456.2282,found456.2288.
Synthesis of 5g of Compound:
Figure BDA0002440957940000172
compound 4e (200mg,0.72mmol) and oxalyl chloride (254mg, 2.00mmol) were placed in a reaction flask, followed by the addition of 10mL of THF. Stirred at room temperature for 1 hour. Concentration under reduced pressure gave an intermediate, which was dissolved in 1,2,3, 4-tetrahydro-6, 7-isoquinolinediol hydrobromide (176mg,0.72mmol) and DIPEA (213mg, 1.65mmol) plus 10mL of THF. Stir at room temperature for 1.5 hours. The sample was taken and the plate reaction was essentially complete, EA was extracted three times, the organic phases were combined, dried, filtered, concentrated and purified by silica gel column chromatography to give 5g (130mg, 42%).
1H NMR(400MHz,DMSO-d6)8.79-8.74(m,1H),8.60(dd,J=7.6Hz,1H),8.43(d,J=4.0Hz,1H),7.67–7.59(m,3H),7.56–7.54(m,2H),7.48(d,J=15.4Hz,1H),7.33(s,1H),7.05(d,J=15.4Hz,1H),6.64–6.52(m,2H),4.58(d,J=86.5Hz,2H),3.83–3.69(m,5H),2.64(d,J=48.7Hz,2H).13C NMR(100MHz,DMSO-d6)165.94,148.69,144.59,144.34,144.23,144.09,135.14,131.22,129.88,129.53,129.22,125.38,124.59,124.23,117.85,115.62,114.18,114.01,113.70,108.75,55.38,44.25,30.01,28.99.HRMS(ESI)calcd forC26H23N3O3(M+H)+426.1812,found 426.1816.
Synthesis of compound 5 h:
Figure BDA0002440957940000181
compound 4e (200mg,0.72mmol) and oxalyl chloride (254mg, 2.0mmol) were placed in a reaction flask, and 10mL of THF was added. Stirred at room temperature for 1 hour. Vacuum concentrating to obtain intermediate, adding 6, 7-bis (2-methoxyethoxy) -1,2,3, 4-tetrahydroisoquinoline hydrochloride (210mg,0.66mmol) and DIPEA (213mg, 1.65mmol), and dissolving with 10ml of THF. Stir at room temperature for 1.5 hours. The sample was taken and the plate reaction was essentially complete, EA was extracted three times, the organic phases were combined, dried, filtered, concentrated, and purified by silica gel column chromatography to give a yellow solid compound 5h (120mg, 29%).
1H NMR(400MHz,CDCl3)8.43(d,J=4.0Hz,1H),8.24(s,1H),7.76(d,J=15.4Hz,1H),7.56–7.50(m,3H),7.45–7.43(m,2H),7.25(s,1H),6.85(d,J=15.4Hz,1H),6.71(d,J=3.4Hz,2H),5.29(s,1H),4.69(d,J=12.4Hz,2H),4.22–4.04(m,4H),3.90–3.71(m,9H),3.45(s,6H),2.81(d,J=19.7Hz,2H).13C NMR(100MHz,CDCl3)166.7,148.8,148.1,147.7,144.4,143.5,135.9,130.7,129.9,129.4,128.8,128.3,127.1,118.6,117.1,115.2,113.6,113.2,109.1,77.3,71.1,69.0,59.2,53.5,47.1,44.3,43.6,40.0,29.9,29.2,28.1.HRMS(ESI)calcd for C32H35N3O5(M+H)+542.2649,found 542.2654.
Synthesis of compound 5 i:
Figure BDA0002440957940000191
to a solution of compound 4f (120mg,0.432mmol) and TEA (0.24mL) in DMF (3mL) was added HATU (197mg,0.518mmol), stirred at 0 ℃ for 0.5 h, and 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (99mg,0.432mmol) was added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give the corresponding compound 5i (52mg, 27%) as a white solid.
1H NMR(400MHz,CDCl3)7.98(s,1H),7.79(d,J=15.4Hz,1H),7.61–7.27(m,10H),6.91(d,J=15.4Hz,1H),6.63(s,2H),4.72(s,2H),3.85(s,6H),3.62(s,3H),2.85(s,2H).13C NMR(100MHz,CDCl3)167.3,147.8,144.5,138.0,136.8,131.0,130.5,129.1,128.8,125.8,122.9,121.4,120.4,113.0,112.7,111.6,111.3,110.9,110.2,109.6,109.1,56.1,56.1,44.5,43.8,31.2,29.3.HRMS(ESI)calcd for C29H28N2O3(M+H)+453.2173,found453.2169.
Synthesis of compound 5 j:
Figure BDA0002440957940000192
to a solution of compound 4g (200mg,0.72mmol) and 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (165mg, 0.72mmol) in anhydrous DMF (3mL) were added DIPEA (1mL) and HATU (380mg, 1mmol), stirred for 16 hours, diluted with water, extracted with EA, washed with saturated brine, anhydrous MgSO 24Drying, filtration, concentration and purification by silica gel column chromatography gave the product 5j (90mg, 27%) as a yellow solid.
1H NMR(400MHz,DMSO-d6)9.48(s,1H),8.65–8.47(m,2H),7.74–7.67(m,3H),7.64–7.56(m,2H),7.46(d,J=15.6Hz,1H),7.15(d,J=15.6Hz,1H),6.83–6.74(m,2H),4.78(s,1H),4.60(s,1H),3.86(s,3H),3.78–3.67(m,8H),2.81(s,1H),2.70(s,1H).13C NMR(100MHz,CDCl3)162.5,148.3,148.0,147.9,147.8,133.4,130.8,130.4,129.4,127.9,126.9,125.3,124.1,122.8,116.0,111.6,111.5,111.2,109.4,109.2,108.8,56.0,47.1,44.6,32.5,29.1.HRMS(ESI)calcd for C28H27N3O3(M+H)+454.2125,found 454.2129.
Example 2 Synthesis of Compound 9
Figure BDA0002440957940000201
Compound 6(208mg, 1mmol) and AlCl3(667mg, 5mmol) was placed in a reaction flask, purged with nitrogen, dissolved with 20ml of LPCM, and stirred at room temperature for 1 hour. Methyl 2-chloro-2-oxoacetate (122mg, 1mmol) was added, stirred at 0 ℃ for 0.5 hour, then warmed to room temperature and heated under N2Stirring was carried out overnight under protective conditions. Sampling point plate, basically finishing reaction, extractingExtracting with DCM for three times, mixing organic phases, drying, filtering, concentrating, and purifying with silica gel column chromatography to obtain yellow solid compound 7(270mg, 92%);1H NMR(400MHz,CDCl3)8.71(d,J=7.8Hz,1H),8.49(d,J=4.7Hz,1H),7.63–7.53(m,3H),7.48(d,J=5.7Hz,2H),7.35(dd,J=7.8,4.8Hz,1H),3.74(s,3H),3.24(s,3H).13CNMR(100MHz,CDCl3)177.5,162.6,149.1,144.0,137.7,132.1,119.8,119.2,112.7,52.9.
placing compound 7(270mg, 0.92mmol) and LiOH (66mg, 2.76mmol) in a reaction flask, adding MeOH (10mL) and water (5mL) to dissolve, stirring overnight at room temperature, sampling the dot plate, substantially completing the reaction, concentrating the mixture under vacuum, extracting EA (2 × 30mL), combining the organic layers, drying, filtering, concentrating, purifying by silica gel column chromatography to obtain compound 8(230mg, 89%) as a white solid;1H NMR(400MHz,CDCl3)8.71(d,J=7.6Hz,1H),8.40(d,J=4.3Hz,1H),7.65–7.48(m,5H),7.39–7.31(m,1H),3.69(s,3H).13C NMR(100MHz,CDCl3)182.0,165.8,150.5,147.8,144.7,131.4,130.4,129.1,128.5,119.7,119.5,109.2,30.1.
compound 8(230mg, 0.82mmol), 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (228mg, 0.98mmol), DIPEA (222mg,1.72mmol) and HATU (373mg,0.98mmol) were placed in a reaction flask, dissolved in DMF (5mL), and stirred at room temperature for 8 hours. Sampling the sample point plate, and basically finishing the reaction. Vacuum concentrating, EA extracting, drying, filtering, concentrating, and purifying with silica gel column chromatography to obtain white solid compound 9(134mg, 36%);1H NMR(400MHz,CDCl3)8.77(dd,J=18.0,7.7Hz,1H),8.48(d,J=4.4Hz,1H),7.50–7.42(m,3H),7.39–7.32(m,1H),7.30–7.22(m,2H),6.56(d,J=5.0Hz,1H),6.45(s,1H),4.37(s,1H),4.01(s,1H),3.84(dd,J=19.4,15.6Hz,6H),3.65(d,J=26.2Hz,3H),3.45(t,J=5.3Hz,1H),3.30(t,J=6.0Hz,1H),2.73(t,J=5.2Hz,1H),2.39(t,J=5.8Hz,1H).13C NMR(100MHz,CDCl3)187.3,165.8,149.3,148.1,147.9,147.8,144.8,131.2,130.2,128.4,126.5,125.5,123.7,123.5,119.4,111.4,110.4,109.2,108.8,56.03,55.98,55.95,46.9,43.5,42.5,38.7,29.7,28.5,27.1.HRMS(ESI)calcd for C27H25N3O4(M+H)+456.1918,found 456.1921.
EXAMPLE 3 Synthesis of Compound 12
Figure BDA0002440957940000211
NaH (112mg,2.8mmol) was placed in a reaction flask, nitrogen was replaced, 5mL of anhydrous DMF was added and dissolved, methyl phosphonoacetate diethyl ester (555mg,2.2mmol) was added under ice-water bath conditions, stirring was carried out for 0.5 hour, a DMF solution of compound 10(451mg,2.0mmol) was added dropwise, and stirring was carried out at room temperature for 8 hours. After the reaction was completed, the reaction was quenched with water, stirred for 2 hours, extracted three times with ethyl acetate, washed with saturated brine, dried, filtered, concentrated, and purified on silica gel column to give a yellow intermediate n7(542mg, 84%).
Intermediate n7(542mg,1.68mmol) was dissolved in CH2Cl2(9mL) and TFA (2mL) were combined and stirred at room temperature overnight, and the plate was sampled and the reaction was essentially complete to give compound 11(550mg, crude).
To a solution of compound 11(150mg,0.565mmol) and TEA (0.5mL,3.6mmol) in DMF (3.5mL) was added HATU (258mg,0.678mmol), the mixture was stirred at 0 ℃ for 0.5 hour, and 6, 7-dimethoxy-1, 2,3, 4-tetrahydroisoquinoline hydrochloride (130mg,0.565mmol) was added. Stirring at room temperature for 5 hours, adding ice water to dilute after the reaction is basically finished, performing suction filtration, and purifying by a silica gel column to obtain a compound 12(234mg, 94%);1H NMR(400MHz,MeOH)9.39–9.26(m,1H),8.89(d,J=3.5Hz,1H),7.65(t,J=10.6Hz,3H),7.52(t,J=6.0Hz,4H),7.14(dd,J=15.6,9.7Hz,1H),6.64–6.48(m,2H),4.54(d,J=16.8Hz,2H),3.71(dd,J=7.6,3.6Hz,1H),3.65(d,J=7.7Hz,6H),3.62–3.55(m,1H),2.67(d,J=5.1Hz,2H).13C NMR(100MHz,MeOH)164.9,164.9,156.8,148.1,148.0,147.8,144.8,138.7,138.5,136.2,136.1,130.79,130.75,129.3,129.2,127.2,127.2,126.7,126.2,124.7,124.5,124.2,122.7,122.6,117.8,113.6,111.6,111.5,109.6,109.4,55.2,55.10,55.05,44.3,43.6,40.6,28.5,27.4.HRMS(ESI)calcdfor C26H24N4O3(M+H)+441.1921,found 441.1921.
example 4 Synthesis of Compounds 13a-13h, Compounds 14a-14b
Figure BDA0002440957940000221
Synthesis of compound 13 a:
Figure BDA0002440957940000222
to a solution of compound 4e (63mg,0.227mmol) and TEA (0.095mL) in DMF (0.915mL) was added HATU (104mg,0.273mmol), stirred at 0 ℃ for 0.5 h and L-phenylalanine methyl ester hydrochloride (49mg,0.227mmol) was added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give compound 13a as a white solid (59mg, 59%).
1H NMR(400MHz,CDCl3)8.40(dd,J=4.8,1.5Hz,1H),8.19(dd,J=7.9,1.5Hz,1H),7.68(d,J=15.6Hz,1H),7.56–7.49(m,3H),7.44–7.39(m,2H),7.30–7.26(m,2H),7.25–7.17(m,2H),7.11(dd,J=7.9,1.5Hz,2H),6.39(d,J=15.6Hz,1H),6.07(d,J=7.7Hz,1H),5.01(dt,J=7.7,5.7Hz,1H),3.74(s,3H),3.73(s,3H),3.17(t,J=6.1Hz,2H).13C NMR(100MHz,CDCl3)172.4,166.5,149.0,145.2,143.7,136.1,135.3,130.8,129.6,129.5,129.4,128.9,128.7,128.6,127.1,118.4,117.3,115.8,108.6,53.4,52.4,38.1,30.0.HRMS(ESI)calcd for C27H25N3O3(M+H)+440.1969,found 440.1965.
Synthesis of compound 13 b:
Figure BDA0002440957940000231
to a solution of compound 4e (120mg,0.432mmol) and TEA (0.18mL) in DMF (1.8mL) was added HATU (164mg,0.432mmol), stirred at 0 ℃ for 0.5 h and D-phenylalanine methyl ester hydrochloride (93mg,0.432mmol) was added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give compound 13b as a white solid (85mg, 44%).
1H NMR(400MHz,CDCl3)8.40(dd,J=4.8,1.4Hz,1H),8.20(dd,J=7.9,1.1Hz,1H),7.66(d,J=15.6Hz,1H),7.56–7.46(m,3H),7.40(ddd,J=6.3,4.4,2.7Hz,2H),7.23–7.17(m,2H),7.12–7.07(m,2H),6.37(d,J=15.6Hz,1H),4.99(dt,J=7.7,5.7Hz,1H),3.73(s,3H),3.72(s,3H),3.22–3.10(m,2H).13C NMR(100MHz,CDCl3)172.4,166.5,148.9,145.2,143.6,136.1,135.3,130.8,129.6,129.6,129.4,128.9,128.7,128.6,127.1,118.5,117.3,115.9,108.6,53.4,52.4,38.1,30.0.HRMS(ESI)calcd for C27H25N3O3(M+H)+440.1969,found 440.1966.
Synthesis of compound 13 c:
Figure BDA0002440957940000232
to a solution of compound 4e (120mg,0.432mmol) and TEA (0.18mL) in DMF (1.8mL) was added HATU (164mg,0.432mmol), stirred at 0 ℃ for 0.5 h and L-tyrosine methyl ester (84mg, 0.432mmol) was added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give compound 13c as a white solid (141mg, 72%).
1H NMR(400MHz,CDCl3)8.36(dd,J=4.8,1.4Hz,1H),8.15(dd,J=8.0,1.4Hz,1H),7.65(d,J=15.6Hz,1H),7.49–7.43(m,3H),7.38–7.33(m,2H),7.15(dd,J=7.9,4.8Hz,1H),6.90(d,J=8.5Hz,2H),6.68(d,J=8.5Hz,2H),6.39(d,J=15.6Hz,1H),6.16(d,J=7.8Hz,1H),4.95(dt,J=7.7,5.7Hz,1H),3.70(s,3H),3.69(s,3H),3.06(qd,J=14.0,5.7Hz,2H).13C NMR(100MHz,CDCl3)171.5,165.7,154.7,147.7,144.3,142.5,134.4,129.6,129.3,128.5,128.3,127.8,126.0,117.5,116.3,114.7,114.6,107.4,52.5,51.4,36.2,29.0.HRMS(ESI)calcd for C27H25N3O4(M+H)+456.1918,found 456.1914.
Synthesis of compound 13 d:
Figure BDA0002440957940000241
the compound 13a (55mg,0.125mmol) and LiOH. H2O (78mg,1.859mmol) was placed in a reaction flask, and H was added2The O/MeOH (2mL/2mL) mixed solution was dissolved and stirred at room temperature for 4 hours. Compound 13d (49mg, 92%) was collected as a white solid by filtration after adjusting the pH to 5 with HCl (1M).
HNMR(400MHz,CDCl3)8.44–7.79(m,3H),7.50(d,J=15.6Hz,1H),7.38(s,3H),7.09(t,J=29.1Hz,9H),6.43(d,J=6.8Hz,1H),6.27(d,J=15.5Hz,1H),4.89(d,J=4.8Hz,1H),3.56(s,3H),3.11(d,J=32.7Hz,2H).13C NMR(100MHz,CDCl3)174.2,167.4,148.6,145.3,143.4,136.2,135.6,130.7,129.6,129.4,129.3,128.9,128.8,128.6,127.0,118.5,117.3,115.5,108.5,53.9,37.4,30.0.HRMS(ESI)calcd for C26H23N3O3(M+H)+426.1812,found 426.1810.
Synthesis of compound 13 e:
Figure BDA0002440957940000242
compound 13b (70mg,0.159mmol) and LiOH. H2O (52mg,1.239mmol) was placed in a reaction flask and H was added2The O/MeOH (1mL/1mL) mixed solution was dissolved and stirred at room temperature for 4 hours. Compound 13e (66mg, 98%) was collected as a white solid by filtration after adjusting the pH to 5 with HCl (1M).
1H NMR(400MHz,CDCl3)8.39(dd,J=4.7,1.2Hz,1H),8.15(d,J=7.2Hz,1H),7.65(d,J=15.6Hz,1H),7.57–7.47(m,3H),7.39(dd,J=7.1,2.2Hz,2H),7.26–7.15(m,6H),6.35(d,J=15.6Hz,1H),6.12(d,J=3.4Hz,1H),4.92(d,J=5.7Hz,1H),3.72(s,3H),3.23(ddd,J=20.1,14.0,5.6Hz,2H).13C NMR(100MHz,CDCl3)167.3,148.3,145.3,143.2,136.1,135.4,130.7,129.5,129.4,129.2,129.1,128.8,128.5,127.0,118.6,117.2,115.5,108.5,67.9,37.4,30.1.HRMS(ESI)calcd for C26H23N3O3(M+H)+426.1812,found426.1808.
Synthesis of compound 13 f:
Figure BDA0002440957940000251
compound 13c (42mg,0.092mmol) and LiOH (16mg,0.691mmol) were placed in a reaction flask, THF/H was added2The O (2mL/2mL) mixed solution was dissolved and stirred at room temperature for 4 hours. Compound 13f (40mg, 98%) was collected as a white solid by filtration after adjusting the pH to 5 with HCl (1M).
1H NMR(400MHz,MeOH)8.38(dd,J=8.0,1.4Hz,1H),8.30(d,J=1.5Hz,1H),7.62–7.36(m,7H),7.24(dd,J=7.9,4.8Hz,1H),7.02(d,J=8.4Hz,2H),6.64(m,J=15.1,12.1Hz,3H),4.58–4.50(m,1H),3.66(s,3H),3.02(ddd,J=20.7,13.8,6.0Hz,2H).13C NMR(100MHz,MeOH)177.2,167.4,155.5,148.5,144.7,143.0,133.0,130.7,130.2,129.6,129.2,129.1,129.0,128.6,118.6,117.5,117.1,114.6,108.7,56.4,37.3,28.9.HRMS(ESI)calcd for C26H23N3O4(M+H)+442.1761,found 442.1763.
Synthesis of Compound 13 g:
Figure BDA0002440957940000252
to a solution of compound 4e (100mg,0.360mmol) and TEA (0.15mL) in DMF (1.5mL) was added HATU (137mg,0.360mmol), stirred at 0 ℃ for 0.5 h and 4-chloro-D-phenylalanine methyl ester hydrochloride (90mg, 0.360mmol) was added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was almost completed, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified with silica gel to obtain 13g (164mg, 96%) of a white solid compound.
1H NMR(400MHz,CDCl3)8.40(dd,J=4.8,1.5Hz,1H),8.20(dd,J=7.9,1.5Hz,1H),7.66(d,J=15.6Hz,1H),7.56–7.46(m,3H),7.41(ddd,J=6.3,4.4,2.6Hz,2H),7.24–7.18(m,3H),7.08–6.96(m,2H),6.37(d,J=15.6Hz,1H),6.00(d,J=7.4Hz,1H),4.97(dd,J=13.1,5.6Hz,1H),3.73(s,3H),3.72(s,3H),3.14(qd,J=13.9,5.6Hz,2H).13C NMR(100MHz,CDCl3)172.2,166.5,148.9,145.3,143.7,135.5,134.6,133.1,130.8,130.8,129.6,129.6,128.9,128.8,128.7,118.4,117.4,115.6,108.5,53.3,52.5,37.5,30.0.HRMS(ESI)calcd for C27H24ClN3O3(M+H)+474.1579,found 474.1577.
Synthesis of compound 13 h:
Figure BDA0002440957940000261
to a solution of compound 4e (93mg,0.334mmol) and TEA (0.23mL) in DMF (2.5mL) was added HATU (127mg,0.334mmol), stirred at 0 ℃ for 0.5 h, and n15(78mg,0.334mmol) was added. Stirring was carried out at room temperature for 5 hours, a sample was taken, the reaction was substantially completed, diluted with ice water, suction-filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified with silica gel column to obtain a white solid compound (13 h, 45mg, 27%).
1H NMR(400MHz,MeOH)8.97(dd,J=8.0,1.2Hz,1H),8.56(dd,J=5.8,1.2Hz,1H),7.71(dd,J=8.0,5.8Hz,1H),7.68–7.61(m,3H),7.54(ddd,J=5.5,2.9,1.6Hz,2H),7.46(d,J=16.0Hz,1H),7.32–7.16(m,6H),6.90(d,J=16.0Hz,1H),4.54(dd,J=8.5,6.7Hz,1H),3.82(s,3H),3.63(ddd,J=15.1,7.1,4.8Hz,1H),3.41–3.32(m,2H),3.27–3.12(m,3H),3.03(dd,J=13.8,8.5Hz,1H),2.87(d,J=0.6Hz,6H).13C NMR(100MHz,MeOH)174.9,169.0,148.3,143.5,138.2,137.1,133.4,133.3,132.1,131.7,130.3,129.7,129.2,128.0,123.9,120.6,118.4,111.6,58.4,57.3,44.0,43.9,38.5,35.8,31.9.HRMS(ESI)calcd for C30H33N5O2(M+H)+496.2707,found 496.2706.
Synthesis of compound 14 a:
Figure BDA0002440957940000271
to a solution of compound 4e (102.5mg,0.369mmol) and TEA (0.155mL) in DMF (2mL) was added HATU (140mg,0.369mmol), stirred at 0 ℃ for 0.5 h and 2- (1-piperazinyl) pyrimidine (60.51mg, 0.367mmol) added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give compound 14a as a white solid (52mg, 33%).
1H NMR(400MHz,CDCl3)8.40(dd,J=4.7,1.4Hz,1H),8.31(d,J=4.8Hz,2H),8.19(dd,J=7.9,1.4Hz,1H),7.76(d,J=15.3Hz,1H),7.58–7.34(m,5H),7.22(dd,J=7.8,4.7Hz,1H),6.77(d,J=15.4Hz,1H),6.51(t,J=4.8Hz,1H),3.86(s,4H),3.73(s,7H).13CNMR(100MHz,CDCl3)166.7,161.6,157.9,148.9,144.6,143.7,136.5,130.8,129.9,129.5,128.9,128.3,118.6,117.3,112.9,110.5,109.1,43.8,29.9,29.4.HRMS(ES)calcdfor C25H24N6O(M+H)+425.2084,found 425.2078.
Synthesis of compound 14 b:
Figure BDA0002440957940000272
to a solution of compound 4e (120mg,0.432mmol) and TEA (0.18mL) in DMF (1.8mL) was added HATU (164mg,0.432mmol), stirred at 0 ℃ for 0.5 h, and 1- (4-pyridyl) piperazine (70.5mg, 0.443mmol) was added. After stirring at room temperature for 5 hours, a sample was taken, the reaction was substantially complete, diluted with ice water, filtered, dissolved in ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and purified on silica gel to give compound 14b as a white solid (163mg, 89%).
1H NMR(400MHz,DMSO-d6)8.58(dd,J=7.9,1.4Hz,1H),8.44(dd,J=4.7,1.4Hz,1H),8.26(d,J=7.2Hz,2H),7.66–7.59(m,3H),7.57–7.54(m,2H),7.51(d,J=15.4Hz,1H),7.33(dd,J=7.9,4.7Hz,1H),7.09(d,J=7.4Hz,2H),7.04(d,J=15.4Hz,1H),3.71(s,3H),3.70(s,8H).13C NMR(100MHz,DMSO-d6)166.1,156.4,148.8,145.1,144.3,143.2,135.8,131.3,130.0,129.8,129.6,129.3,117.9,117.9,113.3,108.8,108.2,49.3,30.1,27.4.HRMS(ESI)calcd for C26H25N5O(M+H)+424.2132,found 424.2128.
EXAMPLE 5 Synthesis of Compounds 16a-16d
Figure BDA0002440957940000281
Synthesis of intermediate 15 a:
to a solution of 3c (4.3g, 18.2mmol) in THF (30mL) was added LDA (18.2mL, 36.4mmol) in THF dropwise at 0 deg.C. After warming to room temperature and stirring for 1 hour, 1-acetylpyrrolidin-2-one (9.25g, 72.9mmol) was added and stirring was carried out at 45 ℃ for 48 hours. After the reaction of the sample point plate is basically finished, saturated NaHCO is added3And (4) quenching. Addition of NH4Diluted Cl (3mL) and water (50mL), extracted with EA, washed with saturated brine (50mL), anhydrous Na2SO4Drying, concentrating, and purifying with silica gel column chromatography to obtain white solid compound 15a (2.3mg, 42%);1H NMR(400MHz,CDCl3)8.42(dd,J=4.7,1.4Hz,1H),8.03(dd,J=7.9,1.4Hz,1H),7.61–7.37(m,6H),7.18(dd,J=7.9,4.7Hz,1H),6.86(s,1H),3.82(s,3H),3.32(t,J=6.6Hz,2H),2.47–2.27(m,2H).13C NMR(100MHz,CDCl3)172.9,148.4,143.4,141.3,131.0,130.6,128.9,128.8,128.0,123.1,119.7,116.5,108.5,39.7,29.9,26.9.
synthesis of compound 16 a:
Figure BDA0002440957940000282
to a solution of compound 15a (450mg, 1.5mmoL) in THF (30mL) was added NaH (80mg, 2mmoL) in portions and stirred at 0 deg.C for 0.5 h. 4- (bromomethyl) -1, 2-dimethoxybenzene (416mg, 1.8mmol) was added thereto, and the mixture was stirred at room temperature for 5 hours, after completion of the reaction on the sample plate, EA was extracted, and the extract was washed with saturated brine and anhydrous Na2SO4Drying, concentration and purification by silica gel column chromatography gave compound 16a (210mg, 31%) as a pale yellow solid.
1H NMR(400MHz,CDCl3)8.40(d,J=4.7Hz,1H),8.02(d,J=7.9Hz,1H),7.55(t,J=2.7Hz,1H),7.53–7.40(m,5H),7.15(dd,J=7.7,4.8Hz,1H),6.82–6.77(m,3H),4.47(s,2H),3.86(s,6H),3.79(s,3H),3.12(t,J=6.6Hz,2H),2.27–2.18(m,2H).13C NMR(100MHz,CDCl3)169.3,149.2,148.5,148.4,143.4,141.1,131.1,130.6,129.7,129.2,128.9,128.8,128.0,122.9,120.8,119.7,116.4,111.5,111.0,108.5,56.0,55.9,47.1,43.9,29.9,24.6.HRMS(ESI)calcd for C28H27N3O3(M+H)+454.2125,found 454.2127.
Synthesis of compound 16 b:
Figure BDA0002440957940000291
starting from compound 3c (300mg, 1.27mmol) and 1-acetylpiperidin-2-one (716mg,5.08mmol), compound 16b (60mg, 13%) was prepared as a pale yellow solid according to the synthetic method and procedure for compound 16 a.
1H NMR(400MHz,CDCl3)8.36(d,J=0.4Hz,1H),7.94(s,1H),7.57-7.52(m,6H),7.20(s,1H),6.93-6.78(m,3H),4.52(s,2H),3.75(d,J=0.8Hz,9H),3.27(s,2H),2.42(s,2H),1.66(s,2H).13C NMR(100MHz,CDCl3)165.3,149.2,148.4,143.3,140.8,128.2,120.6,116.3,111.5,111.0,108.6,56.0,55.9,50.8,47.2,30.0,27.4,23.1.HRMS(ESI)calcd for C29H29N3O3(M+H)+468.2282,found 468.2285.
Synthesis of compound 16 c:
Figure BDA0002440957940000292
starting from compound 15a (100mg, 0.330mmol) and 2-chloromethyl-3, 5-dimethyl-4-methoxypyridine (50mg,0.224mmol), compound 16c (19mg, 12%) was prepared as a pale yellow solid, referring to the synthetic method and procedure for compound 16 a.
1H NMR(400MHz,MeOH)8.62(d,J=7.7Hz,1H),8.51(d,J=5.4Hz,1H),8.43(s,1H),7.65–7.53(m,6H),7.42(s,1H),4.81(s,2H),4.14(s,3H),3.86(s,3H),3.44(t,J=5.4Hz,2H),2.58(s,2H),2.47(s,3H),2.38(s,3H).13C NMR(100MHz,MeOH)171.6,170.8,149.0,143.9,142.6,140.6,136.6,134.4,131.1,130.6,129.9,129.2,129.1,129.0,123.1,122.2,116.4,109.7,60.9,45.3,43.6,30.4,24.7,13.2,10.2.HRMS(ESI)calcd forC28H28N4O2(M+H)+453.2285,found 453.2281.
Synthesis of compound 16 d:
Figure BDA0002440957940000301
starting from compound 15a (500mg, 1.65mmol) and 2-methanesulfonyloxymethyl-3, 5, 6-trimethylpyrazine n16(891mg,3.869mmol), compound 16d (76mg, 11%) was prepared as a pale yellow solid, referring to the synthetic method and procedure for compound 16 a.
1H NMR(400MHz,CDCl3)8.41(dd,J=4.7,1.5Hz,1H),8.03(dd,J=7.9,1.5Hz,1H),7.56(t,J=2.7Hz,1H),7.54–7.41(m,5H),7.17(dd,J=7.9,4.7Hz,1H),4.68(s,2H),3.81(s,3H),3.21(t,J=6.6Hz,2H),2.54(s,3H),2.50(s,3H),2.47(s,3H),2.26(td,J=6.6,2.7Hz,2H).;13C NMR(100MHz,CDCl3)169.3,150.3,148.9,148.5,145.8,143.4,141.1,131.1,130.6,129.2,128.9,128.8,128.0,123.2,119.7,116.5,108.5,46.5,44.6,29.9,24.8,21.6,21.5,20.7;HRMS(ESI)calcd for C27H27N5O(M+H)+438.2288,found438.2295.
Example 6: pharmacological action of azaindole compound or salt thereof
Luciferase reporter gene screening experiments, the experimental steps were as follows: a) an adenovirus carrying the SMAD3 reporter gene is used for infecting A549 cells for 24 h. b) Cells were pretreated with blank or test compound (5. mu.M and 10. mu.M) for 2h, and then stimulated with 0.1ng/mL TGF-. beta.for 24 h. c) The cells are lysed, luciferase substrate is added to the cell lysate, and luciferase activity is tested to obtain a level of inhibition of phosphorylation of SMAD3 by comparing the luciferase activity of the test compound to that of the blank. The results are shown in Table 1.
TABLE 1 inhibitory Activity of azaindoles on SMAD3 phosphorylation (inhibition%, μ M)
Figure BDA0002440957940000311
The results of the activity test show that: the compound of the invention shows inhibitory activity on test cells, and can effectively inhibit the phosphorylation of SMAD3 protein.
Example 7: pharmaceutical Properties of azaindoles or salts thereof
Compound SIS3, compound 16d, compound 13h, compound 5b or their HCl salts (prepared by dissolving compound in 4 equivalents of HCl in methanol, followed by spin-drying the solvent and vacuum drying) were added to pure water or phosphate buffer (50mM, pH 7.4), respectively, at 25 ℃. After shaking and centrifugation, the supernatant was taken to measure the concentration of compound SIS3, compound 16d, compound 13h, or compound 5b in the solution to calculate the corresponding solubility. The detection method comprises the following steps: HPLC (Agilent 1260Infinity) is adopted, the chromatographic column is Shimadzu-GL Wondasil C18-WR, the mobile phase is acetonitrile-water-TFA (68/32/1 ‰), the detection wavelength is 254nm, and the sample injection amount is 10 μ L.
The results are shown in table 2: the azaindole compound has better water solubility, the water solubility of the compound 16d, the compound 13h and the compound 5b is far better than that of the existing SMAD3 small molecule inhibitor compound SIS3, and the solubility of the hydrochloride of the compound 16d, the compound 13h and the compound 5b in water is 667 times, 163 times and 723 times of the solubility of the compound SIS3 & HCl in water respectively.
Table 2 solubility test results
Figure BDA0002440957940000321
Example 8: compound 16d selectively inhibits activation of SMAD3
A549 cells were pretreated for 2h without or with drugs (1. mu.M and 5. mu.M), then stimulated with 1ng/mL TGF-beta for 30 min, lysed, and subjected to Western blot analysis, incubated with either the phospho-SMAD3 or the phospho-SMAD2 antibody and the corresponding secondary antibody (LI-COR IRDYe 800 label) for signal detection, and after washing the membrane with strip buffer (25mM glycine-HCl, pH2, 1% SDS) for 30 min, then re-detected with either SMAD3 or SMAD2 primary antibody, the results are shown in FIG. 1: compound 16d selectively inhibited activation of SMAD3 protein, but not SMAD2 protein.
Example 9: anticancer Activity of Water-soluble Compound 16d in LLC transplantation tumor model in B1 mouse
C57BL/6 mice subcutaneous inoculation 2 × 106LLC cells (CRL-1642, ATCC). When the tumor size reaches 50mm3At this time, mice were randomly divided into 4 groups of 6 mice each and received compound 16d (0, 1.25, 2.5 or 5.0 μ g/g/day, injection in saline, i.p.) for 20 days. Tumor volumes were measured every 5 days and tumor weights were measured at the end of the experiment, and the results are shown in FIG. 2, where treatment of water-soluble compound 16d in mice significantly reduced the volume and weight of LLC tumors in a dose-dependent manner.
Next, the mechanism by which compound 16d attenuated lung cancer progression in mice was investigated by pathological sections of the mice. As shown in FIG. 3, the mice treated with compound 16d increased the number of tumor-infiltrating NK cells in a dose-dependent manner, and the 16-day treatment at a dose of 2.5 or 5.0. mu.g/g significantly increased the accumulation of NK cells compared to control mice, indicating that there was an enhanced anti-tumor immune response in mice treated with compound 16 d.
These results indicate that compound 16d can significantly inhibit cancer progression by enhancing NK cell-mediated anti-cancer immunity in LLC model mice.
The technical features of the above-mentioned embodiments can be arbitrarily combined, and for the sake of brevity, all possible combinations of the technical features in the following embodiments are not described, however, as long as there is no contradiction between the combinations of the technical features, the combinations should be considered as the scope of the present description.
The above-mentioned embodiments only express several embodiments of the present invention, and the description thereof is more specific and detailed, but not construed as limiting the scope of the present invention. It should be noted that, for a person skilled in the art, several variations and modifications can be made without departing from the inventive concept, which falls within the scope of the present invention. Therefore, the protection scope of the present patent shall be subject to the appended claims.

Claims (18)

1. An azaindole compound having a structure shown in formula (I) or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a prodrug molecule thereof:
Figure FDA0002440957930000011
wherein:
d is selected from: C. n; and when D is N, R2Is absent;
a and B are respectively and independently selected from: n, CR3
R1Selected from: H. c1-C6Alkyl radical, C6-C10Aryl radical, C3-C8Cycloalkyl, halogen, CN;
R2selected from: H. c1-C6An alkyl group;
each R3Each independently selected from: H. c1-C6Alkyl, halogen, CN;
R4selected from:
Figure FDA0002440957930000012
R5each R5aAnd R5bEach independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8A cycloalkyl group;
each R6Each independently selected from:
Figure FDA0002440957930000013
R7selected from: H. c1-C6Alkyl, one or more R13Substituted C6-C10Aryl, one or more R13Substituted 6-10 membered heteroaryl;
each R8Each independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8Cycloalkyl, hydroxy, C1-C6Alkoxy radical, C1-C6Alkoxy-substituted C1-C6An alkoxy group;
each R9Each independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8Cycloalkyl, hydroxy, C1-C6Alkoxy radical, C1-C6Alkoxy-substituted C1-C6An alkoxy group;
R10selected from: H. c1-C6An alkyl group;
R11selected from: hydroxy, C1-C6Alkoxy, -N (R)14)(R15);
R12Selected from: H. c1-C6Alkyl, one or more R13Substituted C6-C10Aryl, one or more R13Substituted 6-10 membered heteroaryl;
each R13Each independently selected from: H. halogen, CN, C1-C6Alkyl radical, C3-C8Cycloalkyl, hydroxy, C1-C6Alkoxy radical, C1-C6Alkoxy-substituted C1-C6An alkoxy group;
R14and R15Each independently selected from: H. c1-C6Alkyl, amino substituted C1-C6Alkyl radical, C1-C6Alkylamino substituted C1-C6An alkyl group;
n is selected from: 1.2, 3 and 4;
m is selected from: 0.1, 2,3, 4;
p is selected from: an integer between 0 and 7;
q is selected from: 1.2, 3 and 4;
y is selected from: 1.2, 3,4, 5;
and, when D is C, A is N, B is CR3,R1Is phenyl, R2Is composed of
Figure FDA0002440957930000021
When R is8Is other than C1-C6An alkoxy group.
2. The azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof according to claim 1, wherein D is C, A is N, and B is CR4
3. An azaindole compound or a pharmaceutically acceptable salt or stereoisomer or prodrug molecule thereof according to claim 1, wherein the azaindole compound has a structure represented by formula (II), (IV), (V) or (VI):
Figure FDA0002440957930000022
Figure FDA0002440957930000031
4. the azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof of claim 3, wherein R is5Is H.
5. The azaindole compound or a pharmaceutically acceptable salt or stereoisomer or prodrug molecule thereof of claim 3, wherein n is selected from: 1. 2; m is selected from: 1 or 2.
6. The azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof of claim 3, wherein R is7Selected from: one or more R13Substituted phenyl, one or more R13A substituted six-membered nitrogen-containing heteroaryl.
7. According to claimThe azaindole compound of claim 6, or a pharmaceutically acceptable salt or stereoisomer or prodrug molecule thereof, wherein each R is13Each independently selected from: H. c1-C6Alkyl radical, C1-C6An alkoxy group.
8. The azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof of claim 6, wherein R is7Selected from:
Figure FDA0002440957930000032
9. the azaindole compound or a pharmaceutically acceptable salt or stereoisomer or prodrug molecule thereof of claim 3, wherein each R is8Each independently selected from: H. hydroxy, C1-C3Alkoxy radical, C1-C3Alkoxy-substituted C1-C3An alkoxy group; more preferably, each R8Each independently selected from: hydroxy, methoxy-substituted ethoxy; more preferably, R8Are all hydroxyl groups.
10. The azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof of claim 3, wherein R is9Selected from: H. halogen, hydroxy;
R10selected from: H. c1-C3An alkyl group;
R11selected from: c1-C6An alkoxy group.
11. The azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof of claim 3, wherein R is12Selected from: one or more R13Substituted C6-C10A heteroaryl group; each one ofR13Each independently selected from: H. c1-C6An alkyl group;
more preferably, R12Selected from:
Figure FDA0002440957930000041
12. azaindoles or their pharmaceutically acceptable salts or their stereoisomers or their prodrug molecules as claimed in any one of claims 1-11 wherein R is5aAnd R5bEach independently selected from: H. halogen; and/or the presence of a catalyst in the reaction mixture,
R1selected from: H. c1-C3Alkyl radical, C6-C10Aryl radical, C3-C6A cycloalkyl group; and/or the presence of a catalyst in the reaction mixture,
R2selected from: c1-C3An alkyl group.
13. The azaindole compound or the pharmaceutically acceptable salt or the stereoisomer or prodrug molecule thereof of claim 12, wherein R is5bIs H, R5aIs F; and/or the presence of a catalyst in the reaction mixture,
R1selected from: H. phenyl, cyclopropyl; and/or the presence of a catalyst in the reaction mixture,
R3is H.
14. The azaindole compound or a pharmaceutically acceptable salt or stereoisomer or prodrug molecule thereof according to claim 1, selected from the group consisting of:
Figure FDA0002440957930000042
Figure FDA0002440957930000051
Figure FDA0002440957930000061
15. use of an azaindole of any of claims 1-14, or a pharmaceutically acceptable salt or stereoisomer thereof, or a prodrug molecule thereof, for the preparation of an SMAD3 inhibitor.
16. Use of azaindoles of any one of claims 1-14, or a pharmaceutically acceptable salt or stereoisomer thereof, or a prodrug molecule thereof, in the manufacture of a medicament for the prevention and/or treatment of tumors.
17. The use of claim 16, wherein the tumor is: lung cancer, melanoma, breast cancer, and liver cancer.
18. A pharmaceutical composition for preventing and/or treating tumor, comprising an active ingredient and a pharmaceutically acceptable carrier, wherein the active ingredient comprises the azaindole compound or the pharmaceutically acceptable salt thereof or the stereoisomer thereof or the prodrug molecule thereof according to any one of claims 1 to 14.
CN202010265070.5A 2020-04-07 2020-04-07 Azaindole compound and application thereof Active CN111606904B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010265070.5A CN111606904B (en) 2020-04-07 2020-04-07 Azaindole compound and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010265070.5A CN111606904B (en) 2020-04-07 2020-04-07 Azaindole compound and application thereof

Publications (2)

Publication Number Publication Date
CN111606904A true CN111606904A (en) 2020-09-01
CN111606904B CN111606904B (en) 2021-10-15

Family

ID=72197642

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010265070.5A Active CN111606904B (en) 2020-04-07 2020-04-07 Azaindole compound and application thereof

Country Status (1)

Country Link
CN (1) CN111606904B (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112794851A (en) * 2021-01-26 2021-05-14 复旦大学 3- (pyridine-3-yl) -7-azaindole derivative PI3K delta inhibitor and preparation method and application thereof
CN114280308A (en) * 2021-11-16 2022-04-05 中国科学院合肥物质科学研究院 Application of Smad3 protein as novel anti-cisplatin drug-resistant target of ovarian cancer
US11426412B2 (en) 2017-10-18 2022-08-30 Jubilant Epipad LLC Imidazo-pyridine compounds as PAD inhibitors
US11459338B2 (en) 2017-11-24 2022-10-04 Jubilant Episcribe Llc Heterocyclic compounds as PRMT5 inhibitors
US11529341B2 (en) 2018-03-13 2022-12-20 Jubilant Prodel LLC Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
US11629135B2 (en) 2017-11-06 2023-04-18 Jubilant Prodell Llc Pyrimidine derivatives as inhibitors of PD1/PD-L1 activation
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008022467A1 (en) * 2006-08-25 2008-02-28 Mark Lautens 2-substituted azain doles and 2 substituted thienopyrroles, their precursors and novel processes for the preparation thereof
CN103435612A (en) * 2013-08-30 2013-12-11 四川好医生药业集团有限公司 Compound for treating diabetes mellitus

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008022467A1 (en) * 2006-08-25 2008-02-28 Mark Lautens 2-substituted azain doles and 2 substituted thienopyrroles, their precursors and novel processes for the preparation thereof
CN103435612A (en) * 2013-08-30 2013-12-11 四川好医生药业集团有限公司 Compound for treating diabetes mellitus

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors
US11426412B2 (en) 2017-10-18 2022-08-30 Jubilant Epipad LLC Imidazo-pyridine compounds as PAD inhibitors
US11629135B2 (en) 2017-11-06 2023-04-18 Jubilant Prodell Llc Pyrimidine derivatives as inhibitors of PD1/PD-L1 activation
US11459338B2 (en) 2017-11-24 2022-10-04 Jubilant Episcribe Llc Heterocyclic compounds as PRMT5 inhibitors
US11529341B2 (en) 2018-03-13 2022-12-20 Jubilant Prodel LLC Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
CN112794851A (en) * 2021-01-26 2021-05-14 复旦大学 3- (pyridine-3-yl) -7-azaindole derivative PI3K delta inhibitor and preparation method and application thereof
CN114280308A (en) * 2021-11-16 2022-04-05 中国科学院合肥物质科学研究院 Application of Smad3 protein as novel anti-cisplatin drug-resistant target of ovarian cancer

Also Published As

Publication number Publication date
CN111606904B (en) 2021-10-15

Similar Documents

Publication Publication Date Title
CN111606904B (en) Azaindole compound and application thereof
CN105601573B (en) 2-aminopyrimidine compound and pharmaceutical composition and application thereof
AU2016309833B2 (en) Pharmaceutical composition and application replacing quinolone derivative, pharmaceutical acceptable salt, or stereoisomer
CN103012399B (en) 7-oxopyridinopyrimidine compound as well as medicinal composition and application thereof
CN113166110B (en) 2-aminopyrimidine compound and application thereof
CN110305161A (en) 2- amino-metadiazine compound and its application
CN111989329B (en) Crystal form of mono-mesylate of deuterated 3- (4, 5-substituted aminopyrimidine) phenyl compound and preparation method thereof
CN112351971B (en) Quinoline or quinazoline compound and application thereof
EP4056558A1 (en) Indazole compound, pharmaceutical composition of same, and applications thereof
CN114539263B (en) Nitrogen-containing heterocyclic compound, and pharmaceutical composition and application thereof
CN112538038A (en) 2-indolone ERR alpha inverse agonist and pharmaceutical composition and application thereof
CN112851667B (en) Nitrogen-containing heterocyclic ring compound and medicinal composition and application thereof
CN112313213B (en) 3-amino pyrazole compound and application thereof
CN111875583A (en) Triazole derivative and preparation method and application thereof
KR20150105302A (en) Cyclic sulfonamide containing derivatives as inhibitors of hedgehog signaling pathway
WO2023246903A1 (en) Selenium-containing heterocyclic compound, pharmaceutical composition thereof and use thereof
CN117917402A (en) Isoindolylamide compound, and pharmaceutical composition and application thereof
EP4334305A1 (en) Wdr5 inhibitors and modulators
CN115772167A (en) Compound containing urea structure and application thereof
CN110461827A (en) Quinoxaline and pyrido-pyrazine derivatives as PI3K- beta inhibitor
NZ711522A (en) 6-(5-hydroxy-1h-pyrazol-1-yl)nicotinamide derivatives and their use as phd inhibitors

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
GR01 Patent grant
GR01 Patent grant
CB03 Change of inventor or designer information

Inventor after: Yu Xiyong

Inventor after: Hu Wenhui

Inventor after: Yang Zhongjin

Inventor after: Wu Nannan

Inventor before: Yu Xiyong

Inventor before: Lan Huiyao

Inventor before: Hu Wenhui

Inventor before: Yang Zhongjin

Inventor before: Wu Nannan

CB03 Change of inventor or designer information