CN114853814A - Plasmin inhibitor, preparation method and application thereof - Google Patents

Plasmin inhibitor, preparation method and application thereof Download PDF

Info

Publication number
CN114853814A
CN114853814A CN202210060581.2A CN202210060581A CN114853814A CN 114853814 A CN114853814 A CN 114853814A CN 202210060581 A CN202210060581 A CN 202210060581A CN 114853814 A CN114853814 A CN 114853814A
Authority
CN
China
Prior art keywords
substituted
unsubstituted
alkyl
tert
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
CN202210060581.2A
Other languages
Chinese (zh)
Inventor
阳安乐
纪森
王志
王浩
张德伟
王宵
沈欢
向杰
鲜嘉陵
胡晓
张晓东
唐军
苏忠海
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.)
Scinnohub Pharmaceutical Co Ltd
Original Assignee
Scinnohub 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 Scinnohub Pharmaceutical Co Ltd filed Critical Scinnohub Pharmaceutical Co Ltd
Publication of CN114853814A publication Critical patent/CN114853814A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)]
    • C07F9/3804Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)] not used, see subgroups
    • C07F9/3834Aromatic acids (P-C aromatic linkage)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/60Quinoline or hydrogenated quinoline ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/6552Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a six-membered ring
    • C07F9/65522Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a six-membered ring condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms
    • C07F9/655363Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms the sulfur atom being part of a six-membered ring
    • C07F9/655372Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms the sulfur atom being part of a six-membered ring condensed with carbocyclic rings or carbocyclic ring systems

Landscapes

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

Abstract

The invention relates to a plasmin inhibitor, a preparation method thereof and application thereof in the field of pharmacy. The invention also provides a method for preparing the compound, a composition containing the compound, application of the compound serving as a plasmin inhibitor and pharmaceutical application of the compound.

Description

Plasmin inhibitor, preparation method and application thereof
Technical Field
The invention relates to the field of medicinal chemistry, in particular to a plasmin inhibitor, a preparation method thereof and application thereof in the field of pharmacy.
Background
Plasmin is a proteolytic enzyme that degrades fibrin. When tissue damage causes vascular rupture, a hemostatic mechanism is triggered: vasoconstriction, platelet embolization, initiation of the clotting process, and final formation of stable fibrin. At the same time, the fibrinolytic system is activated due to fibrin deposition, and this system is responsible for fibrin formation and cleavageMaintains balance, and plays a role in maintaining the smoothness of blood vessels and remodeling damaged tissues in the process of repairing damaged blood vessel walls (Tengborn L,
Figure RE-GDA0003551338940000011
M,Berntorp E.Thromb Res.2015Feb;135(2):231-42)。
the fibrinolytic system includes plasminogen, tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA). Plasminogen binds to lysine residues on the surface of fibrin and is converted to plasmin by an activator (i.e., tPA) released from endothelial cells. Fibrinolysis inhibition can be used to treat bleeding. The use of antifibrinolytics can reduce blood loss in cardiac surgery, trauma, orthopedic surgery, solid organ transplantation, obstetrics and gynecology, neurosurgery and non-surgical diseases (Ng W, JerathA,
Figure RE-GDA0003551338940000012
m. analesthesol Intensive ther.2015; 47(4):339-50). In the early 1950 s, lysine amino acids were found to inhibit plasminogen activation, but were too weak to be used for the treatment of fibrinolytic hemorrhagic disease. In 1953, Shosuke Okamoto et al showed several thiol and amino carbonic acids to have plasma protein-resistant effects, and found that synthetic derivative of lysine, epsilon-aminocaproic acid (EACA), has a strong inhibitory effect on plasminogen. EACA has been widely used clinically, but requires larger doses in addition to mild gastrointestinal side effects such as nausea. In 1962, 4-amino-methyl-cyclohexane-carbonic acid (AMCHA) was found, which contains two stereoisomers, and further studies showed that its trans form (trans-4-aminomethylcyclohexanecarboxylic acid, i.e. tranexamic acid, TXA) has anti-fibrinolytic capacity, about 10 times higher activity than EACA, and proved to have stronger tolerance (Tengborn L,
Figure RE-GDA0003551338940000013
M,Berntorp E.Thromb Res.2015 Feb;135(2):231-42)。
tranexamic acid is a synthetic lysine derivative and antifibrinolytic agent that forms reversible complexes with plasminogen. By combining with plasminogen, the interaction between plasminogen and plasmin heavy chain and fibrin lysine residue is blocked, so as to prevent the combination of plasminogen and fibrin surface and further delay fibrinolysis. Tranexamic acid has been approved for the treatment of severe menstrual bleeding and various surgical hemorrhagic diseases, and is currently the most clinically used hemostatic drug. However, a large number of literature reports show that the tranexamic acid is easy to cause gastrointestinal adverse reactions such as nausea, vomiting, diarrhea and dyspepsia after being orally taken, and the dosage of the tranexamic acid is large, so that patients may have epilepsy and other complications after being taken.
Other similar hemostatic drugs, such as aminocaproic acid, have the problems of rapid excretion in human body, weak hemostatic effect, short action duration, more toxic reaction and the like, and can form thrombus when the dosage is excessive, thereby limiting the application to patients with thrombosis tendency or thrombotic vascular disease history and renal insufficiency. The aminomethylbenzoic acid mechanism has 4-5 times stronger action with aminocaproic acid. Has obvious effect on common chronic bleeding, but has no hemostatic effect on wound bleeding and cancer bleeding. In addition, too large an amount may also promote thrombosis. Aprotinin, a commonly used hemostatic drug in heart bypass surgery, was withdrawn from the market by the FDA in 2008 because it can induce renal failure, myocardial infarction, heart failure, and the like.
Other mechanisms of hemostatic drugs, such as carbachol acting on blood vessels, can induce epilepsy after repeated use; the hemostatic thrombin for promoting the blood coagulation process can be only applied to gastrointestinal bleeding or local bleeding.
In view of the clinical limitations of available hemostatic drugs, more or less defects in terms of dosage, clinical indications, etc., and the problems of large dosage, more adverse reactions, easy occurrence of epilepsy and other complications of the existing similar drugs, a new hemostatic drug is needed to be developed to better meet the clinical requirements.
Disclosure of Invention
It is an object of the present invention to overcome the above-mentioned drawbacks of the prior art and to provide a new compound with blood clotting and hemostatic activities.
Specifically, the invention provides compounds shown in the following formula I, and pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof:
Figure RE-GDA0003551338940000021
wherein X is selected from CH 2 、NR 3 O, S; y is selected from CH and N; in certain specific embodiments, X is NR 3 Y is N;
R 1 selected from hydrogen, NRaRb, hydroxyl, substituted or unsubstituted aryl, substituted or unsubstituted alkyl, substituted or unsubstituted aromatic heterocyclic group; wherein Ra and Rb are independently selected from hydrogen, alkyl and halogenated alkyl;
R 2 selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted lipoheterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted arylheterocyclyl;
R 3 selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted lipoheterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted arylheterocyclyl, - (CH) 2 ) mNRcRd, where m ═ 1,2,3,4, Rc, Rd are each independently selected from hydrogen, alkyl, haloalkyl;
in certain specific embodiments, R is as defined herein 1 Selected from hydrogen, NRaRb, hydroxyl, substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted C 1 -C 4 Alkyl, substituted or unsubstituted 6-10 membered aromatic heterocyclic group; wherein Ra and Rb are independently selected from hydrogen, alkyl and halogenated alkyl;
in certain specific embodiments, R is as defined herein 2 Selected from hydrogen, substituted or unsubstituted C 1 -C 4 Alkyl radical, C 1 -C 4 Haloalkyl, substituted or unsubstituted C 3 -C 6 Cycloalkyl, substituted or unsubstituted 4-to 8-membered aliphatic heterocyclic group, substituted or unsubstituted 6-to 10-membered aromatic hydrocarbon groupA substituted or unsubstituted 6-10 membered aromatic heterocyclic group;
in certain specific embodiments, R is as defined herein 3 Selected from hydrogen, substituted or unsubstituted C 1 -C 4 Alkyl radical, C 1 -C 4 Haloalkyl, substituted or unsubstituted C 3 -C 6 Cycloalkyl, substituted or unsubstituted 4-8 membered aliphatic heterocyclic group, substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted 6-10 membered aromatic heterocyclic group;
in certain specific embodiments, the alkyl groups of the present invention are selected from methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl; said alkyl group being optionally substituted with one or more groups selected from halogen, amino, hydroxy, cycloalkyl, lipoheterocyclyl;
in certain specific embodiments, the cycloalkyl groups of the present invention are selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl;
in certain specific embodiments, the lipoheterocyclyl groups of the present invention are selected from the group consisting of oxetanyl, pyrrolidinyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothienyl, piperidinyl, morpholinyl;
in certain specific embodiments, the aryl groups of the present invention are selected from phenyl, naphthyl;
in certain specific embodiments, the aromatic heterocyclic group of the present invention is selected from the group consisting of pyridyl, pyrimidinyl, pyridazinyl, imidazolyl, pyrazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, 1,2, 4-oxadiazolyl;
in certain specific embodiments, R is as defined herein 1 Selected from the group consisting of hydroxy, said R 2 Selected from hydrogen;
in certain specific embodiments, X is selected from CH 2 、NR 3 O, S, wherein R is 3 Selected from hydrogen, -Q- (R) Q ) n And Q is selected from C 1 -C 4 Alkyl radical, C 3 -C 6 Cycloalkyl, 4-6 membered saturated aliphatic heterocyclic group, said n is 0 or 1, said R Q Selected from methyl, phenyl, tetrahydropyranyl, morpholinyl, piperidinyl, methylPiperidyl, o-chlorophenyl, carboxyl, hydroxyl, dimethylamino and halogen; further, Q is preferably selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, cyclopropylalkyl, cyclobutylalkyl, cyclopentylalkyl, piperidinyl, tetrahydropyranyl, morpholinyl, cyclopropoxy;
in certain specific embodiments, X is preferably selected from CH 2 、NR 3 O, S, wherein R is 3 Selected from the group consisting of hydrogen, tetrahydropyranyl, tetrahydropyranylmethyl, methyl, ethyl, n-propyl, isopropyl, t-butyl, trifluoromethyl, trifluoroethyl, benzyl, phenethyl, phenyl, piperidinemethyl, methylpiperidinylmethyl, hydroxyethyl, hydroxymethyl, carboxymethyl, carboxyethyl, piperidinyl, methylpiperidinyl, piperidinylmethyl, cyclopropyl, cyclopentyl, cyclobutyl, dimethylaminopropyl, dimethylaminoethyl, epoxypropyl, o-chlorophenylethyl;
more preferably from CH 2 、NR 3 O, S, wherein R is 3 Selected from the group consisting of hydrogen, methyl, ethyl, isopropyl, t-butyl, tetrahydropyranyl, tetrahydropyranylmethyl, trifluoromethyl, trifluoroethyl, benzyl, phenethyl, phenyl, piperidinylmethyl, methylpiperidinylmethyl, hydroxyethyl, hydroxymethyl, carboxymethyl, carboxyethyl, piperidinyl;
further preferably selected from CH 2 、NR 3 O, S, wherein R is 3 Selected from hydrogen, tetrahydropyranyl, tetrahydropyranylmethyl, methyl, ethyl, tert-butyl, carboxymethyl;
particularly preferably O, NR 3 Wherein said R is 3 Selected from hydrogen, tetrahydropyranyl, tetrahydropyranylmethyl.
In certain embodiments, Y of the present invention is preferably selected from N;
in certain embodiments, the tetrahydropyranylmethyl group of the present invention may be tetrahydropyran-4-ylmethyl, tetrahydropyran-2-ylmethyl, tetrahydropyran-3-ylmethyl; in certain embodiments, the tetrahydropyranylmethyl group of the present invention is preferably tetrahydropyran-4 ylmethyl;
in certain embodiments, the tetrahydropyranyl group of the present invention may be tetrahydropyran-4-yl, tetrahydropyran-2-yl, tetrahydropyran-3-yl; in certain embodiments, the tetrahydropyranyl group of the present invention is preferably tetrahydropyran-4 yl;
in certain embodiments, the piperidinyl groups of the present invention may be piperidin-4-yl, piperidin-2-yl, piperidin-3-yl; in certain embodiments, the piperidinyl groups of the present invention are preferably piperidin-4-yl;
in certain embodiments, the piperidinylmethyl group of the present invention can be piperidin-4-ylmethyl, piperidin-2-ylmethyl, piperidin-3-ylmethyl; in certain embodiments, the piperidinylmethyl group of the present invention is preferably piperidin-4-ylmethyl;
in certain embodiments, methylpiperidinylmethyl or methylpiperidinyl as described herein means that the methyl group is substituted at any position on the piperidine ring of the piperidinylmethyl or piperidinyl group; in certain embodiments, the methylpiperidinylmethyl or, preferably, N-methylpiperidinylmethyl, the methylpiperidinyl is, preferably, N-methylpiperidinyl;
in certain embodiments, the phenylethyl group of the present invention can be a 2-phenylethyl group, a 1-phenylethyl group; in certain embodiments, the phenethyl groups of the present invention are preferably 1-phenylethyl;
in certain embodiments, the trifluoroethyl group of the present invention can be a 2,2, 2-trifluoroethyl group, a 1,1, 2-trifluoroethyl group; in certain embodiments, the trifluoroethyl group of the present invention is preferably a 2,2, 2-trifluoroethyl group;
in certain specific embodiments, the compounds of the present invention have the following structure:
Figure RE-GDA0003551338940000041
Figure RE-GDA0003551338940000051
another object of the present invention is to provide a pharmaceutical composition, comprising at least one of the aforementioned compounds, or pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, and at least one pharmaceutically acceptable excipient.
Another object of the present invention is to provide a use of the aforementioned compound or a pharmaceutically acceptable salt, hydrate, isomer, prodrug and mixture thereof, or a pharmaceutical composition for preparing a medicament. The medicine has blood coagulation and hemostasis therapeutic activities, and can be used for treating abnormal hemorrhage caused by hyperfibrinolysis, surgical operation and postoperative hemorrhage, etc.
It is another object of certain of the present invention to provide a method of treating and/or ameliorating bleeding diseases or conditions comprising administering to a subject in need thereof one or more of the foregoing pharmaceutical compositions or a compound of formula i or a pharmaceutically acceptable salt, hydrate, isomer, prodrug or mixture thereof.
Definition of terms
As used herein, the following terms and phrases are intended to have the following meanings, unless otherwise indicated. A particular term or phrase, unless specifically defined, should not be considered as indefinite or unclear, but rather construed according to ordinary meaning. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
The term "pharmaceutically acceptable" as used herein, is intended to cover only those materials that are suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of acid in neat solution or in a suitable inert solvent.
The compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, as well as racemic and other mixtures thereof, all of which are within the scope of the present invention.
"alkyl" refers to a straight or branched chain-containing saturated aliphatic hydrocarbon group such as: c 1 -C 4 Alkyl refers to saturated aliphatic hydrocarbon groups containing 1 to 4 carbon atoms, including but not limited to methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, and the like, and their various isomers.
"cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent. For example, "C 3 -C 6 Cycloalkyl "refers to cycloalkyl groups containing 3 to 6 carbon atoms, typically C 3 -C 6 Cycloalkyl groups include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl and the like.
"Heterocyclyl" refers to a saturated, monocyclic hydrocarbon substituent in which one or more ring atoms are replaced with a heteroatom selected from N, O, S, the remaining ring atoms being carbon. For example: "3-8 membered heterocycloaliphatic" refers to a saturated cyclic hydrocarbon substituent containing 3-8 ring atoms, one or more of which is substituted with a heteroatom selected from N, O, S, with the remaining ring atoms being carbon. Specific examples include, but are not limited to: oxetanyl, pyrrolidinyl, tetrahydrofuranyl, morpholinyl and the like.
"Arylheterocyclyl" refers to an aromatic cyclic substituent in which one or more ring atoms are substituted with a heteroatom selected from N, O, S, the remaining ring atoms being carbon. For example: "5-6 membered aromatic heterocyclic ring" means an aromatic heterocyclic group containing 5 to 6 ring atoms, and specific examples include, but are not limited to, pyridyl, pyrimidinyl, pyridazinyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, isoxazolyl, 1,2, 4-oxadiazolyl.
"Lipocyclyl" means a saturated cyclic substituent in which one or more ring atoms are substituted with a heteroatom selected from N, O, S, for example "3-5 membered heterocyclyl" means a saturated or unsaturated cyclic substituent comprising 3 to 5 ring atoms in which one or more ring atoms are substituted with a heteroatom selected from N, O, S, specific examples including but not limited to: oxetanyl, azetidinyl, tetrahydrofuryl, pyrrolidinyl, thiazolyl, and the like.
"aryl" refers to aromatic ring groups, e.g., "6-10 membered aryl" refers to aromatic ring groups containing 6 to 10 carbon ring atoms, and examples of aryl moieties include phenyl, naphthyl, and the like.
"optionally" means that the subsequently described event or circumstance may, but need not, occur.
Abbreviations used in this disclosure are known to those skilled in the art and, unless otherwise indicated, are intended to have the meaning reported in the art. For example: DMF means N, N-dimethylformamide; THF means tetrahydrofuran; me is methyl.
Experiments prove that the compound has excellent blood coagulation and hemostasis activities, is obviously superior to the hemostatic drug tranexamic acid which is the most widely clinically applied at present, and has great clinical application value.
Abbreviations in the present invention have the following meanings:
Figure RE-GDA0003551338940000071
Figure RE-GDA0003551338940000081
Detailed Description
The following examples, which are intended to be illustrative only and not to be limiting as to the scope of the invention, illustrate the synthesis of the compounds and intermediates of the invention. Except for special descriptions, the raw materials and reagents involved in the invention can be obtained from commercial sources, and the specific sources do not influence the implementation of the technical scheme of the invention.
EXAMPLE 1 preparation of (4-amino-3, 4-dihydro-2H-pyrano [2,3-b ] pyridin-7-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000082
Step 1: preparation of (3- ((tert-butyldimethylsilyl) oxy) propylidene) -2-methylpropane-2-sulfinamide
3- ((tert-butyldimethylsilyl) oxy) propanal (5.6g) was dissolved in dichloromethane (50mL) and tert-butylsulfinamide (5.4g) and anhydrous copper sulfate (23.8g) were added and stirred at room temperature for 48 h. TLC showed complete consumption of starting material, filtered through celite, and purified by column chromatography to give the title compound (7.35 g).
Figure RE-GDA0003551338940000083
MS(ESI)m/z(M+H) + =292.1.
Step 2: preparation of 3- ((tert-butyldimethylsilyl) oxy) -1- (2, 6-dichloropyridin-3-yl) propyl) -2-methylpropane-2-sulfinamide
2, 6-dichloropyridine (3.74g) was weighed into a 250mL three-necked flask, dissolved in tetrahydrofuran (50mL), replaced with argon three times, cooled to-78 ℃, then lithium diisopropylamide (LDA,12.5mL) was added slowly over 10min, stirred for 30min, and then a solution of N- (3- ((tert-butyldimethylsilyl) oxy) propylene) -2-methylpropane-2-sulfinamide (7.35g) dissolved in 10mL tetrahydrofuran was added slowly dropwise with a syringe, and reacted at-78 ℃ for 2 h. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by column chromatography to afford the title compound (2.6 g).
Figure RE-GDA0003551338940000091
MS(ESI)m/z(M+H) + =438.9.
And step 3: preparation of 1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide
3- ((tert-Butyldimethylsilyl) oxy) -1- (2, 6-dichloropyridin-3-yl) propyl) -2-methylpropane-2-sulfinamide (2.6g) was weighed out and dissolved in tetrahydrofuran (30mL) and cooled to 0 ℃.1M tetrabutylammonium fluoride solution (17.8mL) was added and the reaction was carried out at 0 ℃ for 1 h. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, washed 2 times with saturated brine, dried over anhydrous sodium sulfate, and purified by column chromatography to afford the title compound (1.6 g).
Figure RE-GDA0003551338940000092
MS(ESI)m/z(M+H) + =325.0.
And 4, step 4: preparation of 7-chloro-3, 4-dihydro-2H-pyrano [2,3-b ] pyridin-4-yl) -2-methylpropan-2-sulfinamide
1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (200mg), potassium tert-butoxide (173mg) in tert-butanol (5mL) was weighed and reacted at 85 ℃ for 2 h. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and purified by column chromatography to afford the title compound (133 mg).
Figure RE-GDA0003551338940000093
MS(ESI)m/z(M+H) + =289.1.
And 5: preparation of diethyl 4- ((-tert-butylsulfinyl) amino) -3, 4-dihydro-2H-pyrano [2,3-b ] pyridin-7-yl) phosphonate
weighing-N- (7-chloro-3, 4-dihydro-2H-pyrano [2,3-b ] pyridin-4-yl) -2-methylpropane-2-sulfinamide (50mg) and dissolving in toluene (2mL), sequentially adding tris (dibenzylideneacetone) dipalladium (16mg), 1,1' -bis (diphenylphosphino) ferrocene (19mg), triethylamine (53mg), diethyl phosphite (48mg) and argon replacing for 3 times, and reacting at 120 ℃ for 2.5H. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (13 mg).
Figure RE-GDA0003551338940000101
MS(ESI)m/z(M+H) + =391.0.
Step 6: preparation of (4-amino-3, 4-dihydro-2H-pyrano [2,3-b ] pyridin-7-yl) phosphonic acid hydrochloride
Diethyl (4- ((tert-butylsulfinyl) amino) -3, 4-dihydro-2H-pyrano [2,3-b ] pyridin-7-yl) phosphonate (13mg) was weighed out, dissolved in concentrated hydrochloric acid (3mL) and reacted at 100 ℃ for 3H. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and purified by pre-HPLC to give the title compound (6.5 mg).
Figure RE-GDA0003551338940000102
MS(ESI)m/z(M+H) + =231.0.
1 H NMR(400MHz,Deuterium Oxide)δ8.34(dd,J=8.0,3.1Hz,1H),7.70(t,J=7.3Hz,1H), 4.78(s,1H),4.72(q,J=3.9,3.2Hz,2H),2.57(ddd,J=13.4,9.5,5.8Hz,1H),2.39(ddt,J=15.7, 10.8,4.7Hz,1H).
Example 2 preparation of (4-amino-3, 4-dihydro-2H-benzopyran-7-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000103
Step 1: preparation of N- (7-bromobenzopyran-4-alkylidene) -2-methylpropane-2-sulfinamide
7-bromo-4-dihydrochromone (2.5g) was dissolved in tetrahydrofuran (4mL), tert-butylsulfinamide (2.0g) and tetraethyl titanate (4mL) were added in sequence, the reaction was allowed to proceed overnight at 85 deg.C, and TLC indicated complete reaction of the starting materials. Quenched by addition of water, filtered, the filter cake washed with ethyl acetate, the filtrate backwashed 2 times with saturated brine (2X 15mL), the organic phases combined, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (2.1 g).
Figure RE-GDA0003551338940000111
MS(ESI)m/z(M+H) + =329.9.
Step 2: preparation of N- (7-bromobenzopyran-4-yl) -2-methylpropane-2-sulfinamide
Weighing N- (7-bromobenzopyran-4-alkylidene) -2-methylpropane-2-sulfinamide (1.4g) and dissolving the N- (7-bromobenzopyran-4-alkylidene) -2-methylpropane-2-sulfinamide in tetrahydrofuran (10mL), slowly adding 1.0M lithium tri-sec-butylborohydride (10.6mL) at the temperature of 0 ℃, slowly heating the system to room temperature for reaction for 3h, and adding ammonium chloride aqueous solution to quench after TLC shows that the raw materials are completely reacted. Extraction with ethyl acetate was performed 2 times, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (738 mg).
Figure RE-GDA0003551338940000112
MS(ESI)m/z(M+H) + =332.0.
And 3, step 3: preparation of diethyl (4- ((tert-butylsulfinyl) amino) chroman-7-yl) phosphonate
Weighing N- (7-bromobenzopyran-4-yl) -2-methylpropane-2-sulfinamide (0.32g) into a microwave reaction, sequentially adding N, N-diisopropylethylamine (0.21mL), diethyl phosphite (0.16mL), palladium acetate (8mg), dppf (21mg), replacing with argon for three times, adding 3mL of anhydrous N, N-dimethylformamide/ethylene glycol dimethyl ether (9:1, v/v), reacting at 115 ℃ for 25min in the microwave reactor, and displaying that the raw materials are completely reacted by TLC. After quenching with water, extraction with ethyl acetate was performed 2 times, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (233 mg).
Figure RE-GDA0003551338940000113
MS(ESI)m/z(M+H) + =390.0.
And 4, step 4: preparation of (4-amino-3, 4-dihydro-2H-benzopyran-7-yl) phosphonic acid hydrochloride
Diethyl (4- ((tert-butylsulfinyl) amino) chroman-7-yl) phosphonate (0.145g) was weighed out and dissolved in 12M concentrated hydrochloric acid (4mL), the system was warmed to 100 ℃ and reacted overnight, LC-MS showed complete reaction of the starting material, and after concentration under reduced pressure, the title compound (39mg) was purified by pre-HPLC.
Figure RE-GDA0003551338940000121
MS(ESI)m/z(M-H) - =227.9.
1 H NMR(400MHz,Deuterium Oxide)δ7.32(dd,J=7.9,4.4Hz,1H),7.20(dd,J=12.5,7.9Hz, 1H),7.11(d,J=14.7Hz,1H),4.52(d,J=5.6Hz,1H),4.17(tdt,J=12.1,9.1,3.6Hz,2H),2.29 (ddd,J=14.4,9.5,4.7Hz,1H),2.12-2.01(m,1H).
EXAMPLE 3 preparation of (4-amino-1, 2,3, 4-tetrahydroquinolin-7-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000122
Step 1: preparation of methyl 3- ((3-bromophenyl) amino) propionate
M-bromoaniline (9g), methyl acrylate (4.6g) and glacial acetic acid (297. mu.L) were placed in a sealed tube and reacted at 110 ℃ for 12 h. After TLC detection of the material consumption, diluted with ethyl acetate, washed three times with water, dried over anhydrous sodium sulfate and concentrated. The crude product was purified by column chromatography to give the title compound (10 g).
Figure RE-GDA0003551338940000131
MS(ESI)m/z(M+H) + =257.9.
Step 2: preparation of methyl 3- (N- (3-bromophenyl) -4-methylphenylsulfonamido) propionate
Methyl 3- ((3-bromophenyl) amino) propionate (10g) and 4-toluenesulfonyl chloride (8.1g) were dissolved in pyridine (20mL) and reacted at room temperature for 2 h. After the consumption of the starting material by TLC, the reaction mixture was concentrated to dryness, diluted with ethyl acetate, washed once with hydrochloric acid (1N) and saturated brine in this order, dried over anhydrous sodium sulfate and concentrated. The crude product was purified by column chromatography to give the title compound (15.6 g).
Figure RE-GDA0003551338940000132
MS(ESI)m/z(M+H) + =411.9.
And step 3: preparation of 3- (N- (3-bromophenyl) -4-methylphenylsulfonamido) propionic acid
Methyl 3- (N- (3-bromophenyl) -4-methylphenylsulfonamido) propionate (15.6g) was dissolved in dioxane/water/concentrated hydrochloric acid (180/60/20 mL) and reacted at 110 ℃ overnight. After completion of the reaction, the reaction solution was concentrated to dryness, diluted with water, extracted three times with ethyl acetate, then dried over anhydrous sodium sulfate and concentrated, and the crude product was purified by column chromatography to give the title compound (12 g).
Figure RE-GDA0003551338940000133
MS(ESI)m/z(M+H) + =397.9.
And 4, step 4: preparation of 7-bromo-2, 3-dihydroisoquinolin-4-one
3- (N- (3-bromophenyl) -4-methylphenylsulfonamido) propionic acid (12g) was dissolved in anhydrous dichloromethane (100mL), and thionyl chloride (10.8mL) and 2-3 drops of N, N-dimethylformamide were added to conduct a reaction at 40 ℃ under reflux for 2 hours. After the consumption of the starting material was detected by LC-MS, the reaction was concentrated to dryness and diluted with anhydrous dichloromethane (50 mL). Then, aluminum chloride (8g) was dissolved in anhydrous dichloromethane, and the above solution was dropwise added to the aluminum chloride solution in an ice bath. After the addition was complete, the reaction was carried out overnight at room temperature. After TLC detection of the material consumption, the reaction was quenched with ice-water, neutralized with sodium hydroxide solution, extracted three times with ethyl acetate, dried over anhydrous sodium sulfate and concentrated, and the crude product was purified by column chromatography to give the title compound (3.5 g).
Figure RE-GDA0003551338940000134
MS(ESI)m/z(M+H) + =225.9.
And 5: preparation of 7-bromo-4-oxo-3, 4-dihydroquinoline-1 (2H) -carboxylic acid tert-butyl ester
7-bromo-2, 3-dihydroisoquinolin-4-one (762mg) was dissolved in 20mL of anhydrous dichloromethane, and 4-dimethylaminopyridine (830mg) and di-tert-butyl dicarbonate (873mg) were added to the solution in an ice bath to react at room temperature for 4 hours. After TLC detection of the starting material consumption, the reaction was concentrated to dryness and the crude product was purified by column chromatography to give the title compound (795 mg).
Figure RE-GDA0003551338940000141
MS(ESI)m/z(M+H) + =326.0.
Step 6: preparation of tert-butyl 7-bromo-4- ((tert-butylsulfinyl) imino) -3, 4-dihydroquinoline-1 (2H) -carboxylate
7-bromo-4-oxo-3, 4-dihydroquinoline-1 (2H) -carboxylic acid tert-butyl ester (795mg) and tert-butylsulfinamide (443mg) were dissolved in 2-methyltetrahydrofuran (10mL), 1mL of tetraethyl titanate was added, and the mixture was transferred to 80 ℃ and reacted overnight. After TLC detection of the end of consumption of starting material, it was diluted with ethyl acetate, added 1mL of water, filtered, concentrated and the crude product was purified by column chromatography to give the title compound (907 mg).
Figure RE-GDA0003551338940000142
MS(ESI)m/z(M+H) + =429.1
And 7: preparation of tert-butyl 7-bromo-4- (1, 1-dimethylethylenesulfonamido) -3, 4-dihydroquinoline-1 (2H) -carboxylic acid salt
Tert-butyl 7-bromo-4- ((tert-butylsulfinyl) imino) -3, 4-dihydroquinoline-1 (2H) -carboxylic acid salt (907mg) was dissolved in 10mL of anhydrous tetrahydrofuran, and lithium tri-sec-butylborohydride (6.3mL) was slowly added in an ice bath, followed by reaction at room temperature for 2H. After TLC detection of the starting material consumption, the reaction was concentrated to dryness and the crude product was purified by column chromatography to give the title compound (380 mg).
Figure RE-GDA0003551338940000143
MS(ESI)m/z(M+Na) + =453.0.
And 8: preparation of tert-butyl 7- (diethoxyphosphoryl) -4- ((R) -1, 1-dimethylethylenesulfonamido) -3, 4-dihydroquinoline-1 (2H) -carboxylate
Tert-butyl 7-bromo-4- ((R) -1, 1-dimethylethylsulphinylideneamino) -3, 4-dihydroquinoline-1 (2H) -carboxylate (300mg), diethyl phosphite (193mg), tris (dibenzylideneacetone) dipalladium (64mg), 1' -bis (diphenylphosphino) ferrocene (dppf,77.6mg) and triethylamine (141mg) were dissolved in 10mL of toluene, transferred to 125 ℃ under nitrogen and reacted overnight. After TLC detection of complete consumption of starting material, it was diluted with ethyl acetate, filtered and concentrated, and the crude product was purified by pre-TLC to give the title compound (96 mg).
Figure RE-GDA0003551338940000151
MS(ESI)m/z(M+Na) + =511.0.
And step 9: preparation of (4-amino-1, 2,3, 4-tetrahydroquinolin-7-yl) phosphonic acid hydrochloride
Tert-butyl 7- (diethoxyphosphoryl) -4- (1, 1-dimethylethylenesulfonamido) -3, 4-dihydroquinoline-1 (2H) -carboxylic acid salt (96mg) was added to concentrated hydrochloric acid (20mL) and stirred at 100 ℃ overnight. After complete consumption of starting material by LC-MS assay, it was concentrated under reduced pressure and purified by pre-HPLC to give the title compound (22.6 mg).
Figure RE-GDA0003551338940000152
MS(ESI)m/z(M-16) + =211.9.
1 H NMR(400MHz,Deuterium Oxide)δ7.22(dd,J=7.7,3.8Hz,1H),7.05(td,J=15.3,13.0,9.6 Hz,2H),4.45(d,J=4.3Hz,1H),3.24(dt,J=7.3,3.3Hz,2H),2.16-1.94(m,2H).
EXAMPLE 4 preparation of (5-amino-8- (2-hydroxyethyl) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000153
Step 1: preparation of 3- ((-tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (200mg) was weighed out and dissolved in dichloromethane (4mL), and placed at 0 ℃ to react with triethylamine (0.17mL) and methanesulfonyl chloride (48. mu.L) sequentially added thereto at 0 ℃ for 1.5 h. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (240 mg).
Figure RE-GDA0003551338940000161
MS(ESI)m/z(M+H) + =402.9
Step 2: preparation of N- (7-chloro-1- (2-hydroxyethyl) -1,2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
3- ((tert-Butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (240mg) was weighed, dissolved in N, N-dimethylformamide (5mL), and then ethanolamine (109mg) and triethylamine (0.25mL) were added thereto to react at 70 ℃ for 1.5 hours. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, washed 2 times with saturated brine, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (127 mg).
Figure RE-GDA0003551338940000162
MS(ESI)m/z(M+H) + =332.0.
And step 3: preparation of N- (1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -7-chloro-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
N- (7-chloro-1- (2-hydroxyethyl) -1,2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide (110mg, N) was weighed out and dissolved in dichloromethane (4mL) and cooled to 0 ℃. Imidazole (45mg), tert-butyldimethylsilyl chloride (100mg) were added thereto, and the mixture was warmed to room temperature to react for 2 hours. TLC indicated complete consumption of starting material, quenched with water, extracted 3 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (139 mg).
Figure RE-GDA0003551338940000163
MS(ESI)m/z(M+H) + =446.2
And 4, step 4: preparation of diethyl (8- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -5- ((tert-butylsulfinyl) amino) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate
N- (1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -7-chloro-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide ((139mg) was weighed out and dissolved in toluene (4mL), tris (dibenzylideneacetone) dipalladium (29mg), 1,1' -bis (diphenylphosphino) ferrocene (35mg), triethylamine (95mg), diethyl phosphite (87mg) were added in this order and replaced with argon 3 times, and reaction was carried out at 120 ℃ for 3 h.TLC, showing complete consumption of the starting material, quenched with water, extracted with ethyl acetate 2 times, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (32 mg).
Figure RE-GDA0003551338940000171
MS(ESI)m/z(M+H) + =548.2
And 5: preparation of (5-amino-8- (2-hydroxyethyl) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonic acid hydrochloride
Diethyl (8- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -5- ((tert-butylsulfinyl) amino) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate (32mg) was weighed out and dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ for 3 h. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and purified by pre-HPLC to give the title compound (5.23 mg).
Figure RE-GDA0003551338940000172
MS(ESI)m/z(M+H) + =274.0
1 H NMR(400MHz,Deuterium Oxide)δ7.87(dd,J=7.4,2.9Hz,1H),7.09(t,J=8.1Hz,1H), 4.63(t,J=5.2Hz,1H),3.87(t,J=4.4Hz,2H),3.75-3.57(m,4H),2.29(ddt,J=14.2,9.5,4.8Hz, 1H),2.19(dq,J=15.0,5.4Hz,1H).
EXAMPLE 5 preparation of (5-amino-8-benzyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000173
Step 1: preparation of 3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (250mg) was weighed out and dissolved in dichloromethane (4mL), placed at 0 ℃ and triethylamine (0.21mL) and methanesulfonyl chloride (60. mu.L) were added in that order to react at 0 ℃ for 1.5 h. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (300 mg).
Figure RE-GDA0003551338940000181
MS(ESI)m/z(M+H) + =402.9
Step 2: preparation of N- (1-benzyl-7-chloro-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (300mg) was weighed, dissolved in N, N-dimethylformamide (6mL), and reacted at 70 ℃ for 2 hours with benzylamine (160mg) and triethylamine (0.30 mL). TLC showed complete consumption of the starting material, quenched with water, extracted 2 times with ethyl acetate, washed 2 times with saturated brine, dried over anhydrous sodium sulfate and purified by column chromatography to give the title compound (356 mg).
Figure RE-GDA0003551338940000182
MS(ESI)m/z(M+H) + =378.0
And step 3: preparation of diethyl (8-benzyl-5- ((tert-butylsulfinyl) amino) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate
N- (1-benzyl-7-chloro-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide ((180mg) was weighed and dissolved in toluene (2mL), tris (dibenzylideneacetone) dipalladium (44mg), 1,1' -bis (diphenylphosphino) ferrocene (53mg), triethylamine (0.2mL), diethyl phosphite (132mg) was added in this order, and after replacement with argon gas for 3 times, the reaction was carried out at 120 ℃ for 9 h.TLC showing that the starting material was consumed, water was added for quenching, ethyl acetate was extracted 2 times, dried over anhydrous sodium sulfate, and purified by Prep-TLC to obtain the title compound (32 mg).
Figure RE-GDA0003551338940000183
MS(ESI)m/z(M+H) + =480.1
And 4, step 4: preparation of (5-amino-8-benzyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonic acid hydrochloride
Diethyl (8-benzyl-5- ((tert-butylsulfinyl) amino) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate (27mg) was weighed out, dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ for 3 h. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and purified by pre-HPLC to give the title compound (3.44 mg).
Figure RE-GDA0003551338940000191
MS(ESI)m/z(M+H) + =320.2
1 H NMR(400MHz,Deuterium Oxide)δ7.91(dd,J=6.9,2.7Hz,1H),7.33(q,J=7.5,7.0Hz, 5H),7.09(t,J=8.2Hz,1H),4.95–4.80(m,2H),4.67(s,1H),3.84–3.72(m,2H),2.35(tt,J=9.5, 4.8Hz,1H),2.23(dq,J=15.1,5.4Hz,1H).
EXAMPLE 6 preparation of (5-amino-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000192
Step 1: preparation of 3- ((-tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (270mg) was weighed out and dissolved in dichloromethane (5mL), and placed at 0 ℃ to react with triethylamine (0.21mL) and methanesulfonyl chloride (64. mu.L) for 1.5h at 0 ℃. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (330 mg).
Figure RE-GDA0003551338940000193
MS(ESI)m/z(M+H) + =402.9
Step 2: preparation of N- (7-chloro-1- (4-methoxybenzyl) -1,2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (330mg) was weighed, dissolved in N, N-dimethylformamide (6mL), and p-methoxybenzylamine (338mg) and triethylamine (0.34mL) were added to react at 70 ℃ for 3 hours. TLC showed complete consumption of the starting material, quenched with water, extracted 2 times with ethyl acetate, washed 2 times with saturated brine, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (336 mg).
Figure RE-GDA0003551338940000201
MS(ESI)m/z(M+H) + =408.1
And step 3: preparation of diethyl (5- ((tert-butylsulfinyl) amino) -8- (4-methoxybenzyl) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate
N- (7-chloro-1- (4-methoxybenzyl) -1,2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide (336mg) was weighed out and dissolved in toluene (6mL), and tris (dibenzylideneacetone) dipalladium (76mg), 1,1' -bis (diphenylphosphino) ferrocene (92 mg), triethylamine (0.35mL), diethyl phosphite (0.21mL), argon-substituted 3 times, and reacted at 120 ℃ overnight. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (110 mg).
Figure RE-GDA0003551338940000202
MS(ESI)m/z(M+H) + =510.2
And 4, step 4: preparation of (5-amino-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonic acid hydrochloride
Diethyl (5- ((tert-butylsulfinyl) amino) -8- (4-methoxybenzyl) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate (65mg) was weighed out and dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ overnight. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and purified by pre-HPLC to give the title compound (17.5 mg).
Figure RE-GDA0003551338940000203
MS(ESI)m/z(M+H) + =230.1
1 H NMR(400MHz,Deuterium Oxide)δ7.87(dd,J=7.4,2.8Hz,1H),7.02(dd,J=9.0,7.3Hz, 1H),4.63(t,J=5.0Hz,1H),3.59(dt,J=13.9,5.2Hz,1H),3.45(ddd,J=13.9,9.2,4.8Hz,1H), 2.15(dq,J=10.3,5.0Hz,2H).
EXAMPLE 7 preparation of (5-amino-5, 6,7, 8-tetrahydronaphthalen-2-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000211
Step 1: preparation of N- (6-bromo-3, 4-dihydronaphthalen-1 (2H) -alkylidene) -2-methylpropane-2-sulfinamide
6-bromo-3, 4-dihydronaphthalen-1 (2H) -one (300mg,1.33mmol), 2-methylpropane-2-sulfinamide (242mg) was dissolved in 2-methylfuran, added to tetraethoxytitanate (1mL) and reacted at 85 ℃ for 4H under nitrogen protection, and TLC monitored for completion of the reaction. After cooling to room temperature, the reaction mixture was dropped into a mixed solution of ethyl acetate and saturated brine, suction-filtered through celite, and the filtrate was concentrated and then column-purified to obtain the title compound (399 mg).
Figure RE-GDA0003551338940000212
MS(ESI)m/z(M+H) + =328.0/330.0.
Step 2: preparation of N- (6-bromo-1, 2,3, 4-tetrahydronaphthalen-1-yl) -2-methylpropane-2-sulfinamide
N- (6-bromo-3, 4-dihydronaphthalene-1 (2H) -alkylene) -2-methylpropane-2-sulfinamide (399mg) was added dropwise to a lithium tri-sec-butylborohydride solution (3.7mL) at-15 ℃ under nitrogen protection, the reaction was carried out for 1H at the temperature, and the completion of the reaction was monitored by TLC. Quenched with saturated ammonium chloride solution, extracted with ethyl acetate, dried over anhydrous sodium sulfate of the organic phase, concentrated and purified by column to give the title compound (316 mg).
Figure RE-GDA0003551338940000213
MS(ESI)m/z(M+H) + =330.0/332.0.
And step 3: preparation of (5- ((tert-butylsulfinyl) amino) diethyl) -5,6,7, 8-tetrahydronaphthalen-2-yl) phosphonate
N- (6-bromo-1, 2,3, 4-tetrahydronaphthalen-1-yl) -2-methylpropane-2-sulfinamide (158mg) was weighed and dissolved in toluene (5mL), and diethyl phosphite (133mg), triethylamine (200uL), tris (dibenzylideneacetone) dipalladium (44mg), 1,1' -bis (diphenylphosphino) ferrocene (54mg) were added in this order, and then replaced with argon for 3 times, followed by reaction at 120 ℃ for 2 hours. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (112 mg).
Figure RE-GDA0003551338940000221
MS(ESI)m/z(M+H) + =388.1.
And 4, step 4: preparation of (5-amino-5, 6,7, 8-tetrahydronaphthalen-2-yl) phosphonic acid hydrochloride
(5- ((tert-butylsulfinyl) amino) diethyl) -5,6,7, 8-tetrahydronaphthalen-2-yl) phosphonate (112mg) was weighed out and dissolved in concentrated hydrochloric acid (10mL) and reacted at 100 ℃ overnight. LCMS showed complete consumption of starting material, concentrated under reduced pressure and isolated by pre-HPLC to afford the title compound (30 mg).
Figure RE-GDA0003551338940000222
MS(ESI)m/z(M-H) - =226.0.
1 H NMR(400MHz,Deuterium Oxide)δ7.47(t,J=11.2Hz,2H),7.32(dd,J=7.8,3.4Hz, 1H),4.50(t,J=5.4Hz,1H),2.77(dtd,J=24.6,17.2,6.5Hz,2H),2.08(dt,J=13.5,6.9Hz,1H), 1.92(dq,J=14.2,5.2Hz,1H),1.79(p,J=6.7Hz,2H).
EXAMPLE 8 preparation of (5-amino-5, 6,7, 8-tetrahydroquinolin-2-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000223
Step 1: preparation of N- (2-chloro-7, 8-dihydroquinolin-5 (6H) -alkylidene) -2-methylpropane-2-sulfinamide
2-chloro-7, 8-dihydroquinolin-5 (6H) -one (300mg) and 2-methylpropane-2-sulfinamide (300mg) were dissolved in 2-methylfuran (10mL), added to tetraethoxytitanate (1mL), and reacted at 85 ℃ for 4H under nitrogen protection, and the completion of the reaction was monitored by TLC. After cooling to room temperature, the reaction mixture was dropped into a mixed solution of ethyl acetate and saturated brine, suction-filtered through celite, and the filtrate was concentrated and then column-purified to obtain the title compound (433 mg).
Figure RE-GDA0003551338940000231
MS(ESI)m/z(M+H) + =285.9.
Step 2: preparation of N- ((S) -2-chloro-5, 6,7, 8-tetrahydroquinolin-5-yl) -2-methylpropane-2-sulfinamide
N- (2-chloro-7, 8-dihydroquinolin-5 (6H) -alkylene) -2-methylpropane-2-sulfinamide (433mg) was added dropwise to a lithium tri-sec-butylborohydride solution (4.5mL) at-15 ℃ under nitrogen protection, the reaction was carried out at that temperature for 1H, and the completion of the reaction was monitored by TLC. Quenched with saturated ammonium chloride solution, extracted with ethyl acetate, dried over anhydrous sodium sulfate of the organic phase, concentrated and purified on a column to give the title compound (276 mg).
Figure RE-GDA0003551338940000232
MS(ESI)m/z(M+H) + =287.0.
And step 3: preparation of (5- ((tert-butylsulfinyl) amino) diethyl) -5,6,7, 8-tetrahydroquinolin-2-yl) phosphonate
N- (2-chloro-5, 6,7, 8-tetrahydroquinolin-5-yl) -2-methylpropane-2-sulfinamide (100mg) was weighed and dissolved in toluene (5mL), and diethyl phosphite (97mg), triethylamine (98. mu.L), tris (dibenzylideneacetone) dipalladium (32mg), 1,1' -bis (diphenylphosphino) ferrocene (39mg) were added in this order, and then replaced with argon for 3 times, followed by reaction at 120 ℃ for 2 hours. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to afford the title compound (68mg, 50% yield).
Figure RE-GDA0003551338940000233
MS(ESI)m/z(M+H) + =389.2.
And 4, step 4: preparation of (5-amino-5, 6,7, 8-tetrahydroquinolin-2-yl) phosphonic acid hydrochloride
(5- ((tert-butylsulfinyl) amino) diethyl) -5,6,7, 8-tetrahydroquinolin-2-yl) phosphonate (68mg) was weighed out and dissolved in concentrated hydrochloric acid (5mL) and reacted at 100 ℃ overnight. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and isolated by pre-HPLC to give the title compound (25 mg).
Figure RE-GDA0003551338940000241
MS(ESI)m/z(M+H) + =229.0.
1 H NMR(400MHz,Deuterium Oxide)δ8.49(dd,J=8.0,2.5Hz,1H),8.02(t,J=7.8Hz,1H), 4.75(d,J=4.6Hz,1H),3.15(tdd,J=19.1,12.9,5.9Hz,2H),2.19(tt,J=12.2,6.2Hz,1H),2.00 (ddq,J=11.3,7.9,4.9,4.1Hz,3H).
EXAMPLE 9 preparation of (4-Aminothiopyran-7-yl) phosphonic acid hydrochloride
Figure RE-GDA0003551338940000242
Step 1: preparation of 2- ((3-bromophenyl) thio) propionic acid
After M-bromophenylthiol (0.27mL) was dissolved in 5mL of 2M aqueous sodium hydroxide solution and reacted at 100 ℃ for 2 hours, the temperature of the reaction system was lowered to 60 ℃, 3-chloropropionic acid (302mg) was added and reacted at 60 ℃ overnight, and the completion of the reaction was monitored by TLC. After cooling to room temperature, the pH of the system was adjusted to 2 with 3M aqueous hydrochloric acid in an ice bath, a large amount of solid precipitated, which was filtered off, the filter cake was washed three times with water, the filter cake was taken out and dried under reduced pressure to give the title compound (500 mg).
Figure RE-GDA0003551338940000243
MS(ESI)m/z(M-H) - =258.8/260.8.
Step 2: preparation of 7-bromothiochroman-4-ones
2- ((3-bromophenyl) thio) propionic acid (500mg) was dissolved in cold concentrated sulfuric acid (10mL), stirred for half an hour in an ice water bath, then moved to room temperature and stirred for 3h, and the reaction was monitored by TLC for completion. The system was slowly poured into ice, extracted three times with ethyl acetate, the organic phases were combined, washed successively with saturated sodium bicarbonate solution, saturated brine once, dried over anhydrous sodium sulfate, concentrated and column-purified to give the title compound (320 mg).
Figure RE-GDA0003551338940000251
MS(ESI)m/z(M+H) + =242.9/244.8.
1 H NMR(400MHz,Chloroform-d)δ7.98-7.96(d,J=8.5Hz,1H),7.48-7.47(d,J=1.9Hz, 1H),7.33-7.30(dd,J=8.5,1.9Hz,1H),3.28-3.25(m,2H),3.01-2.96(m,2H).
And step 3: preparation of N- (7-bromothiopyran-4-alkylidene) -2-methylpropane-2-sulfinamide
7-Bromothiochroman-4-one (320mg), 2-methylpropane-2-sulfinamide (240mg) was dissolved in tetraethyl titanate (3mL), reacted at 100 ℃ for 1h under nitrogen protection, and the completion of the reaction was monitored by TLC. After cooling to room temperature, the reaction mixture was dropped into a mixed solution of ethyl acetate and saturated brine, suction-filtered through celite, and the filtrate was concentrated and then column-purified to give the title compound (320 mg).
Figure RE-GDA0003551338940000252
MS(ESI)m/z(M+H) + =345.9/348.0.
And 4, step 4: preparation of N- (7-bromothiochroman-4-yl) -2-methylpropane-2-sulfinamide
N- (7-bromothiopyran-4-alkylidene) -2-methylpropane-2-sulfinamide (320mg) was taken, under the protection of nitrogen, lithium tri-sec-butylborohydride solution (2.8mL) was added dropwise at-15 ℃ and reacted at that temperature for 1h, and the completion of the reaction was monitored by TLC. Quenched with saturated ammonium chloride solution, extracted with ethyl acetate, dried over anhydrous sodium sulfate of the organic phase, concentrated and purified by column to give the title compound (260 mg).
Figure RE-GDA0003551338940000253
MS(ESI)m/z(M+H) + =369.8/371.9.
1 H NMR(400MHz,Chloroform-d)δ7.29(d,J=2.7Hz,1H),7.23-7.15(m,2H),4.53-4.49 (ddd,J=9.1,5.9,3.6Hz,1H),3.49-3.47(d,J=8.7Hz,1H),3.22-3.15(ddd,J=12.9,10.5,3.6Hz, 1H),3.06-3.00(ddd,J=12.8,6.4,3.9Hz,1H),2.48-2.32(m,2H),1.25(s,9H).
And 5: preparation of diethyl (4- ((tert-butylsulfinyl) amino) thiopyran-7-yl) phosphonate
N- (7-Bromothiochroman-4-yl) -2-methylpropane-2-sulfinamide (50mg) was weighed and dissolved in toluene (3mL), and diethyl phosphite (39mg), triethylamine (58. mu.L), tris (dibenzylideneacetone) dipalladium (13mg), 1,1' -bis (diphenylphosphino) ferrocene (16mg) were added in this order, and the mixture was substituted with argon for 3 times, followed by reaction at 120 ℃ for 2 hours. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (50 mg).
Figure RE-GDA0003551338940000261
MS(ESI)m/z(M+H) + =406.1.
And 6: preparation of (4-aminothiopyran-7-yl) phosphonic acid hydrochloride
Diethyl (4- ((tert-butylsulfinyl) amino) thiopyran-7-yl) phosphonate (50mg) was weighed out and dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ overnight. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and isolated by pre-HPLC to afford the title compound (4.1 mg).
Figure RE-GDA0003551338940000262
MS(ESI)m/z(M-H) - =243.9.
1 H NMR(400MHz,Deuterium Oxide)δ7.44-7.41(d,J=13.2Hz,1H),7.39-7.27(m,2H), 4.60-4.58(t,J=4.3Hz,1H),3.12-2.97(m,2H),2.43-2.34(m,1H),2.28-2.20(m,1H).
EXAMPLE 10 preparation of (4-amino-3, 4-dihydro-2H-thiopyrano [2,3-b ] pyridin-7-yl) phosphonate hydrochloride
Figure RE-GDA0003551338940000263
Step 1: preparation of 3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (360mg) was weighed out and dissolved in dichloromethane (6mL), and placed at 0 ℃ to react with triethylamine (0.46mL) and methanesulfonyl chloride (86. mu.L) for 1.5h at 0 ℃. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (440 mg).
Figure RE-GDA0003551338940000271
MS(ESI)m/z(M+H) + =402.9.
Step 2: preparation of (-) -3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl) ethanol thioester
3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (200mg) was weighed out, dissolved in N, N-dimethylformamide (4mL), and reacted with potassium thioacetate (57mg) at 40 ℃ for 2 hours. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, washed 2 times with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (220 mg).
Figure RE-GDA0003551338940000272
MS(ESI)m/z 0(M+H) + =383..
And step 3: preparation of N- (7-chloro-3, 4-dihydro-2H-thiopyrano [2,3-b ] pyridin-4-yl) -2-methylpropane-2-sulfinamide
(3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl) ethanethiolate (220mg) was weighed out and dissolved in tetrahydrofuran (4mL), and 4mL of a 2M aqueous solution of sodium hydroxide was added to the solution to conduct a reaction at 60 ℃ for 2 hours. TLC showed complete consumption of starting material, quenched with water, extracted 3 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (108 mg).
Figure RE-GDA0003551338940000273
MS(ESI)m/z(M+H) + =305.0.
And 4, step 4: preparation of diethyl (4- ((tert-butylsulfinyl) amino) -3, 4-dihydro-2H-thiopyrano [2,3-b ] pyridin-7-yl) phosphonate
N- (7-chloro-3, 4-dihydro-2H-thiopyrano [2,3-b ] pyridin-4-yl) -2-methylpropane-2-sulfinamide (108mg) was weighed and dissolved in toluene (3mL), and tris (dibenzylideneacetone) dipalladium (33mg), 1,1' -bis (diphenylphosphino) ferrocene (40mg), triethylamine (148. mu.L), diethyl phosphite (92. mu.L) were added in this order, and then replaced with argon gas for 6 hours at 120 ℃. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (45 mg).
Figure RE-GDA0003551338940000281
MS(ESI)m/z(M+H) + =407.1.
And 5: preparation of (4-amino-3, 4-dihydro-2H-thiopyrano [2,3-b ] pyridin-7-yl) phosphonate hydrochloride
Diethyl (4- ((tert-butylsulfinyl) amino) -3, 4-dihydro-2H-thiopyrano [2,3-b ] pyridin-7-yl) phosphonate (45mg) was weighed out, dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ overnight. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and isolated by pre-HPLC to give the title compound (5.49 mg).
Figure RE-GDA0003551338940000282
MS(ESI)m/z(M+H) + =247.0.
1 H NMR(400MHz,Deuterium Oxide)δ7.87(dd,J=7.4,2.8Hz,1H),7.02(dd,J=9.0,7.3Hz, 1H),4.63(t,J=5.0Hz,1H),3.59(dt,J=13.9,5.2Hz,1H),3.45(ddd,J=13.9,9.2,4.8Hz,1H), 2.15(dq,J=10.3,5.0Hz,2H).
EXAMPLE 11 preparation of (5-amino-8-methyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate hydrochloride
Figure RE-GDA0003551338940000283
Step 1: preparation of 3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (360mg) was weighed out and dissolved in dichloromethane (6mL), and placed at 0 ℃ to react with triethylamine (0.46mL) and methanesulfonyl chloride (86. mu.L) for 1.5h at 0 ℃. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (440 mg).
Figure RE-GDA0003551338940000291
MS(ESI)m/z(M+H) + =402.9.
Step 2: preparation of N- (7-chloro-1-methyl-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
3- ((tert-Butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (220mg) was weighed into a sealed tube, dissolved by adding tetrahydrofuran (5mL), and reacted with a 2M tetrahydrofuran solution of methylamine (1.64mL) and triethylamine (0.23mL) at 100 ℃ overnight. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (110 mg).
Figure RE-GDA0003551338940000292
MS(ESI)m/z(M+H) + =302.1.
And step 3: preparation of diethyl (5- ((tert-butylsulfinyl) amino) -8-methyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate
Weighing N- (7-chloro-1-methyl-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide (110mg) and dissolving in toluene (3mL), sequentially adding tris (dibenzylideneacetone) dipalladium (34mg,0.037mmol), 1,1' -bis (diphenylphosphino) ferrocene (41 mg), triethylamine (0.15mL), diethyl phosphite (94 μ L), replacing with argon for 3 times, and reacting at 120 ℃ for 6 h. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (73 mg).
Figure RE-GDA0003551338940000293
MS(ESI)m/z 404.1(M+H) + .
And 4, step 4: preparation of (5-amino-8-methyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate hydrochloride
Diethyl (5- ((tert-butylsulfinyl) amino) -8-methyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate (73mg) was weighed out and dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ for 8 h. LCMS indicated complete consumption of material, concentration under reduced pressure and isolation by pre-HPLC afforded the title compound (26.7 mg).
Figure RE-GDA0003551338940000301
MS(ESI)m/z(M+H) + =244.0.
1 H NMR(400MHz,Deuterium Oxide)δ7.82(dd,J=7.4,2.9Hz,1H),7.06(dd,J=9.3,7.2Hz, 1H),4.62(t,J=5.1Hz,1H),3.66(dt,J=8.5,5.2Hz,2H),3.21(s,3H),2.31–2.11(m,2H).
EXAMPLE 12 preparation of (5-amino-8-methyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate hydrochloride
Figure RE-GDA0003551338940000302
Step 1: preparation of 3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (500mg) was weighed out and dissolved in dichloromethane (8 mL), and placed at 0 ℃ to react with triethylamine (0.64mL) and methanesulfonyl chloride (119. mu.L) for 1.5h at 0 ℃. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound (600 mg).
Figure RE-GDA0003551338940000303
MS(ESI)m/z(M+H) + =402.9
Step 2: preparation of N- (7-chloro-1-isopropyl-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (200mg) was weighed into a sealed tube, dissolved by adding tetrahydrofuran (4mL), and reacted at 120 ℃ overnight with isopropylamine (127. mu.L) and triethylamine (0.20 mL). TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (34 mg).
Figure RE-GDA0003551338940000311
MS(ESI)m/z(M+H) + =330.1.
And step 3: preparation of diethyl (5-amino-8-isopropyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate
N- (7-chloro-1-isopropyl-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide (34mg) was weighed and dissolved in toluene (1.5mL), and tris (dibenzylideneacetone) dipalladium (10mg), 1,1' -bis (diphenylphosphino) ferrocene (12mg), triethylamine (43. mu.L), diethyl phosphite (40. mu.L) were added in this order, and then replaced with argon gas for 3 times, and reacted at 120 ℃ overnight. TLC indicated complete consumption of starting material, concentrated under reduced pressure in vacuo and purified by Prep-TLC to give the title compound (18 mg).
Figure RE-GDA0003551338940000312
MS(ESI)m/z(M+H) + =328.1.
And 4, step 4: preparation of (5-amino-8-methyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate hydrochloride
Diethyl (5-amino-8-isopropyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate (18mg) was weighed out and dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ overnight. LCMS indicated complete consumption of starting material, concentrated under reduced pressure and isolated by pre-HPLC to give the title compound (5.2 mg).
Figure RE-GDA0003551338940000313
MS(ESI)m/z(M+H) + =272.1.
1 H NMR(400MHz,Deuterium Oxide)δ7.77(dd,J=7.4,2.8Hz,1H),6.96(t,J=7.5Hz,1H), 4.57(s,1H),4.43(p,J=6.6Hz,1H),3.63(dt,J=13.8,5.1Hz,1H),3.45(ddd,J=14.1,8.4,5.9 Hz,1H),2.16(q,J=5.2,4.3Hz,2H),1.24(dd,J=8.7,6.6Hz,6H).
Example 13
Figure RE-GDA0003551338940000321
Step 1: preparation of 3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate
N- (1- (2, 6-dichloropyridin-3-yl) -3-hydroxypropyl) -2-methylpropane-2-sulfinamide (300mg) was weighed out and dissolved in dichloromethane (5mL), and placed at 0 ℃ to react with triethylamine (0.38mL) and methanesulfonyl chloride (71. mu.L) for 1.5h at 0 ℃. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (372 mg).
Figure RE-GDA0003551338940000322
MS(ESI)m/z 402.9(M+H) +
Step 2: preparation of N- (7-chloro-1-isobutyl-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide
3- ((tert-butylsulfinyl) amino) -3- (2, 6-dichloropyridin-3-yl) propyl methanesulfonate (186mg) was weighed into a sealed tube, and N, N-dimethylformamide (4mL) was added thereto for dissolution, and isobutylamine (0.14mL) and triethylamine (0.19mL) were added thereto and reacted at 70 ℃ for 3 hours. TLC showed complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound (70 mg).
Figure RE-GDA0003551338940000323
MS(ESI)m/z(M+H) + =344.1.
And step 3: preparation of diethyl (5- ((tert-butylsulfinyl) amino) -8-isobutyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate
Weighing N- (7-chloro-1-isobutyl-1, 2,3, 4-tetrahydro-1, 8-naphthyridin-4-yl) -2-methylpropane-2-sulfinamide (70mg) and dissolving in toluene (2mL), sequentially adding tris (dibenzylideneacetone) dipalladium (19mg), 1,1' -bis (diphenylphosphino) ferrocene (23mg), triethylamine (85 muL), diethyl phosphite (52 muL) and replacing for 3 times with argon, and reacting at 120 ℃ for 10 h. TLC indicated complete consumption of starting material, quenched with water, extracted 2 times with ethyl acetate, dried over anhydrous sodium sulfate, and purified by Prep-TLC to give the title compound (20 mg).
Figure RE-GDA0003551338940000331
MS(ESI)m/z(M+H) + =446.1.
And 4, step 4: preparation of (5-amino-8-isobutyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate hydrochloride
Diethyl (5- ((tert-butylsulfinyl) amino) -8-isobutyl-5, 6,7, 8-tetrahydro-1, 8-naphthyridin-2-yl) phosphonate (20mg) was weighed out and dissolved in concentrated hydrochloric acid (4mL) and reacted at 100 ℃ overnight. LCMS indicated complete consumption of material, concentration under reduced pressure and isolation of the title compound by pre-HPLC (12 mg).
Figure RE-GDA0003551338940000332
MS(ESI)m/z(M+H) + =286.1
1 H NMR(400MHz,Deuterium Oxide)δ7.82(dd,J=7.5,2.7Hz,1H),6.99(t,J=7.6Hz,1H), 4.62(t,J=5.1Hz,1H),3.65(tdd,J=17.9,10.1,4.6Hz,2H),3.49(dd,J=15.2,7.5Hz,1H),3.34 (dd,J=15.3,7.9Hz,1H),2.32–2.04(m,3H),0.91(dd,J=6.7,1.6Hz,6H).
The following compounds of the examples were prepared using conventional commercially available starting materials and reagents, according to the preparation methods of the preceding examples in combination with conventional means of separation and purification in the art:
Figure RE-GDA0003551338940000333
Figure RE-GDA0003551338940000341
Figure RE-GDA0003551338940000351
Figure RE-GDA0003551338940000361
Figure RE-GDA0003551338940000371
example 33
Figure RE-GDA0003551338940000372
Step 1: preparation of 7-bromo-4-oxo-2, 3-dihydroisoquinoline-1 (2H) -carboxylic acid tert-butyl ester
7-bromo-2, 3-dihydroisoquinolin-4-one (762mg) was dissolved in 20mL of anhydrous dichloromethane, and 4-dimethylaminopyridine (830mg) and di-tert-butyl dicarbonate (873mg) were added to the solution in an ice bath to react at room temperature for 4 hours. After TLC detection of the starting material consumption, the reaction was concentrated to dryness and the crude product was purified by column chromatography to give the title compound (795 mg).
Figure RE-GDA0003551338940000381
MS(ESI)m/z(M+H) + =326.0.
Step 2: preparation of tert-butyl 7-bromo-4- ((tert-butylsulfinyl) imino) -3, 4-dihydroisoquinoline-1 (2H) -carboxylate
7-bromo-4-oxo-3, 4-dihydroisoquinoline-1 (2H) -carboxylic acid tert-butyl ester (795mg) and tert-butylsulfinamide (443mg) were dissolved in 2-methyltetrahydrofuran (10mL), and 1mL of tetraethyl titanate was added and the mixture was transferred to 80 ℃ for reaction overnight. After TLC detection of the end of consumption of starting material, it was diluted with ethyl acetate, added 1mL of water, filtered, concentrated and the crude product was purified by column chromatography to give the title compound (907 mg).
Figure RE-GDA0003551338940000382
MS(ESI)m/z(M+H) + =429.1
And step 3: preparation of tert-butyl 7-bromo-4- (1, 1-dimethylethylenesulfonamido) -3, 4-dihydroisoquinoline-1 (2H) -carboxylate
Tert-butyl 7-bromo-4- ((tert-butylsulfinyl) imino) -3, 4-dihydroisoquinoline-1 (2H) -carboxylate (907mg) was dissolved in 10mL of anhydrous tetrahydrofuran, and lithium tri-sec-butylborohydride (6.3mL) was slowly added in an ice bath, followed by reaction at room temperature for 2H. After TLC detection of the starting material consumption, the reaction was concentrated to dryness and the crude product was purified by column chromatography to give the title compound (380 mg).
Figure RE-GDA0003551338940000383
MS(ESI)m/z(M+Na) + =453.0.
And 4, step 4: preparation of tert-butyl 7- (diethoxyphosphoryl) -4- ((R) -1, 1-dimethylethylenesulfonamido) -3, 4-dihydroisoquinoline-1 (2H) -carboxylate
Tert-butyl 7-bromo-4- ((R) -1, 1-dimethylethylenesulfonamido) -3, 4-dihydroisoquinoline-1 (2H) -carboxylate (300mg), diethyl phosphite (193mg), tris (dibenzylideneacetone) dipalladium (64mg), 1' -bis (diphenylphosphino) ferrocene (dppf,77.6mg), and triethylamine (141mg) were dissolved in 10mL of toluene, transferred to 125 ℃ under nitrogen protection, and reacted overnight. After TLC detection of complete consumption of starting material, it was diluted with ethyl acetate, filtered and concentrated, and the crude product was purified by pre-TLC to give the title compound (96 mg).
Figure RE-GDA0003551338940000384
MS(ESI)m/z(M+Na) + =511.0.
And 5: preparation of (4-amino-1, 2,3, 4-tetrahydroquinolin-7-yl) phosphonic acid hydrochloride
Tert-butyl 7- (diethoxyphosphoryl) -4- (1, 1-dimethylethylenesulfonamido) -3, 4-dihydroisoquinoline-1 (2H) -carboxylate (96mg) was added to concentrated hydrochloric acid (20mL), and stirred at 100 ℃ overnight. After complete consumption of starting material by LC-MS assay, it was concentrated under reduced pressure and purified by pre-HPLC to give the title compound (22.6 mg).
Figure RE-GDA0003551338940000391
MS(ESI)m/z(M-16) + =211.9。
1 H NMR(400MHz,Deuterium Oxide):δ7.74–7.63(m,1H),7.58–7.45(m,2H),4.96(t,J=5.8Hz,1H),4.58–4.33(m,2H),3.88(dd,J=13.8,5.8Hz,1H),3.66(dd,J=13.8,5.9Hz,1H).
Biological assay
Test example 1: plasma clot degradation experiments
1. Purpose of experiment
The inhibition of human plasma clot degradation by the compounds of the invention was determined.
2. Experimental materials and instruments
Figure RE-GDA0003551338940000392
3. Experimental procedure
3.1 collecting fresh healthy human blood, using 0.109M trisodium citrate as anticoagulant, mixing 1 part anticoagulant and 9 parts blood, centrifuging at 2000Xg for 20 minutes at room temperature, collecting supernatant (blood plasma), subpackaging and storing at-80 ℃ for later use.
3.2 day of experiment, plasma was thawed in a 37 ℃ water bath and all reagents except tPA were pre-warmed at 37 ℃.
3.3 Add 12.5. mu.L, 80mM CaCl to 96-well plates 2 (HEPES buffer, pH 7.4), and then 25. mu.L of a solution diluted with physiological saline was added theretoFor test compounds of different concentrations, equal volumes of saline were added to the negative control wells.
3.4 mu.L of pre-warmed plasma was mixed with 12.5. mu.L of tPA at 4nM (HEPES buffer, pH 7.4), immediately added to a 96-well plate, absorbance was measured at 405nM, and the reading was taken every 2 minutes for 15 hours continuously.
3.5 the absorption value changes with time, and rises first and then falls, and the time corresponding to the median of the absorption value in the descending section and the time corresponding to the median of the absorption value in the ascending section are the plasma Clot degradation time (Clot lysis time). Calculating the plasma clot degradation time relative to the plasma clot degradation time in the wells with different concentrations of compound by taking the plasma clot degradation time of the negative control wells as a reference to obtain the inhibition rate:
inhibition% Clot lysis time Compound pore Clot lysis time )100%
3.6 fitting dose-response curves
The log values of the compound concentrations were taken as the X-axis and the percent inhibition as the Y-axis, and the dose-effect curves were fitted using the analysis software GraphPad Prism 5 log (inhibitor) vs. response-Variable slope (Variable slope), to derive the IC of each compound on cell activity 50 The value is obtained.
Calculating the formula: y ═ min + (max-min)/(1+10^ ((Logicc) 50 -X)×Hillslope))。
Inhibition of plasma clot degradation by Compounds of the invention the IC of the compounds of the examples of the invention was calculated by the assay described above 50 The values are all greatly lower than the currently most widely used hemostatic drug tranexamic acid (IC under the same test conditions) 50 4.75. mu.M).
IC of Compounds in examples of the invention 50 ratio value (IC) 50 ratio ═ Compound IC of the invention 50 Value/tranexamic acid IC 50 Values) are as follows:
example numbering IC 50 ratio(uM) Example numbering IC 50 ratio(uM)
1 0.06 18 /
2 0.11 19 0.11
3 0.64 20 0.49
4 0.51 21 /
5 0.39 22 0.95
6 0.22 23 0.15
7 0.54 24 0.73
8 0.48 25 0.73
9 0.62 26 /
10 0.37 27 0.33
11 0.53 28 /
12 0.82 29 /
13 0.28 30 0.97
14 0.67 31 /
15 0.24 32 0.3
16 0.42 33 2.19
17 /
Note: "/" indicates no detection.
Test data show that the compound can effectively inhibit the degradation of plasma clot, has excellent blood coagulation and hemostatic activities, has an effective dose far lower than that of the most frequently used hemostatic medicament in clinic at present, can effectively avoid adverse reactions and complications caused by high-dose medicament administration, and has excellent medicament prospect.

Claims (10)

1. A compound having the structure of formula I, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof:
Figure FDA0003478102200000011
wherein X is selected from CH 2 、NR 3 O, S; y is selected from CH and N;
R 1 selected from hydrogen, NRaRb, hydroxy, substituted or unsubstitutedSubstituted or unsubstituted alkyl, substituted or unsubstituted aromatic heterocyclic group; wherein Ra and Rb are independently selected from hydrogen, alkyl and halogenated alkyl;
R 2 selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted lipoheterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted arylheterocyclyl;
R 3 selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted lipoheterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted arylheterocyclyl, - (CH) 2 ) mNrcRd, wherein m ═ 1,2,3,4, Rc, Rd are each independently selected from hydrogen, alkyl, haloalkyl.
2. The compound of claim 1, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, wherein R is 1 Selected from hydrogen, NRaRb, hydroxyl, substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted C 1 -C 4 Alkyl, substituted or unsubstituted 6-10 membered aromatic heterocyclic group; wherein Ra and Rb are independently selected from hydrogen, alkyl and halogenated alkyl.
3. The compound of claim 1 or 2, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, wherein R is 2 Selected from hydrogen, substituted or unsubstituted C 1 -C 4 Alkyl radical, C 1 -C 4 Haloalkyl, substituted or unsubstituted C 3 -C 6 Cycloalkyl, substituted or unsubstituted 4-8 membered aliphatic heterocyclic group, substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted 6-10 membered aromatic heterocyclic group.
4. The compound of any one of claims 1-3, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, wherein R is 3 Selected from hydrogen, substituted or unsubstituted C 1 -C 4 Alkyl radical, C 1 -C 4 Haloalkyl, substituted or unsubstituted C 3 -C 6 Cycloalkyl, substituted or unsubstituted 4-8 membered aliphatic heterocyclic group, substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted 6-10 membered aromatic heterocyclic group.
5. The compound according to any one of claims 1 to 4, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, wherein the alkyl group is selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl; said alkyl group being optionally substituted with one or more groups selected from halogen, amino, hydroxy, cycloalkyl, lipoheterocyclyl; the cycloalkyl is selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl; the lipid heterocyclic group is selected from oxetanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothienyl, piperidinyl, morpholinyl; the aryl is selected from phenyl and naphthyl; the aromatic heterocyclic group is selected from pyridyl, pyrimidyl, pyridazinyl, imidazolyl, pyrazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl and 1,2, 4-oxadiazolyl.
6. The compound according to any one of claims 1-4, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, wherein X is selected from CH 2 、NR 3 O, S, wherein R is 3 Selected from hydrogen, -Q- (R) Q ) n And Q is selected from C 1 -C 4 Alkyl radical, C 3 -C 6 Cycloalkyl, 4-6 membered saturated aliphatic heterocyclic group, said n is 0 or 1, said R Q Selected from methyl, phenyl, tetrahydropyranyl, morpholinyl, piperidinyl, methylpiperidinyl, o-chlorophenyl, carboxy, hydroxy, dimethylamino, halogen;
preferably, Q is selected from Q, preferably from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, cyclopropylalkyl, cyclobutylalkyl, cyclopentylalkyl, piperidinyl, tetrahydropyranyl, morpholinyl, cyclopropoxy.
7. The compound of any one of claims 1-6, pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, wherein the compound of formula I has the structure:
Figure FDA0003478102200000021
Figure FDA0003478102200000031
8. a pharmaceutical composition comprising at least one compound of any one of claims 1-7, or pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, and at least one pharmaceutically acceptable excipient.
9. Use of a compound of any one of claims 1-8, or pharmaceutically acceptable salts, hydrates, isomers, prodrugs and mixtures thereof, or a pharmaceutical composition, for the manufacture of a medicament.
10. Use according to claim 9, characterized in that the medicament has the therapeutic activity of blood coagulation, hemostasis; can be used for treating abnormal hemorrhage due to hyperfibrinolysis, and hemorrhage after surgery and operation.
CN202210060581.2A 2021-02-03 2022-01-19 Plasmin inhibitor, preparation method and application thereof Pending CN114853814A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110151840 2021-02-03
CN2021101518408 2021-02-03

Publications (1)

Publication Number Publication Date
CN114853814A true CN114853814A (en) 2022-08-05

Family

ID=82627959

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210060581.2A Pending CN114853814A (en) 2021-02-03 2022-01-19 Plasmin inhibitor, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114853814A (en)

Similar Documents

Publication Publication Date Title
CA3124898C (en) Heterocyclic compound, intermediate, preparation method therefor and application thereof
WO2020156323A1 (en) Immunomodulators, compositions and methods thereof
TW202122388A (en) Rip1 inhibitory compounds and methods for making and using the same
JP2018108997A (en) Kynurenine-3-monooxygenase inhibitor, pharmaceutical composition thereof, and method of application therefor
JP7395730B2 (en) Heterocyclic RIP1 inhibitory compounds
CN102325754B (en) Cyclic compound having hetero atom
CN113874015B (en) Thienopyridine inhibitors of RIPK2
CA3131337C (en) Substituted heterocyclic amide compound and preparation method therefor and pharmaceutical use thereof
CA3166455A1 (en) Heterocyclic compounds useful for modulating baf complexes
CA2973202A1 (en) Factor xia inhibitors
WO2023232069A1 (en) Azaquinolinone derivative, preparation method therefor and use thereof
ES2912499T3 (en) Tetrahydropyran-based thiodisaccharide mimetics as galectin-3 inhibitors
CN112292376A (en) Quaternary lactam compound and pharmaceutical application thereof
CN106928252B (en) A kind of compound and the preparation method and application thereof inhibiting ROCK
TWI803177B (en) Plasmin inhibitor, preparation method and application thereof
CN114853814A (en) Plasmin inhibitor, preparation method and application thereof
TWI810773B (en) Plasmin inhibitor, preparation method and application thereof
EP1346994A1 (en) Cholesterol biosynthesis inhibitors containing as the active ingredient tricyclic spiro compounds
KR20220000854A (en) Novel compounds, method for preparing the same and pharmaceutical composition for preventing and treating cancer comprising the same
CN101175756A (en) Tricyclic spiro compound comprising acyl group bound to nitrogen atom in the ring
CN115703776A (en) Plasmin inhibitor, preparation method and application thereof
WO2020243049A1 (en) Factor xi activation inhibitors
CN113135930B (en) Furopyridone imidazole compound, preparation method and application thereof
CN112778273B (en) Cyclic ketopyridone compounds and preparation method and application thereof
CA3218932A1 (en) 2,8-dihydropyrazolo[3,4-b]indole derivatives for use in the treatment of cancer

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