CN114437038A - Pyridazine alkyne compound and application thereof - Google Patents

Pyridazine alkyne compound and application thereof Download PDF

Info

Publication number
CN114437038A
CN114437038A CN202111305068.7A CN202111305068A CN114437038A CN 114437038 A CN114437038 A CN 114437038A CN 202111305068 A CN202111305068 A CN 202111305068A CN 114437038 A CN114437038 A CN 114437038A
Authority
CN
China
Prior art keywords
substituted
alkyl
unsubstituted
membered
compound
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
CN202111305068.7A
Other languages
Chinese (zh)
Inventor
张学军
常少华
王洪强
万海星
井真中
李群
陈浩民
田华
杨俊�
李莉娥
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.)
Humanwell Healthcare Group Co ltd
Wuhan Humanwell Innovative Drug Research and Development Center Ltd Co
Original Assignee
Humanwell Healthcare Group Co ltd
Wuhan Humanwell Innovative Drug Research and Development Center Ltd Co
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 Humanwell Healthcare Group Co ltd, Wuhan Humanwell Innovative Drug Research and Development Center Ltd Co filed Critical Humanwell Healthcare Group Co ltd
Publication of CN114437038A publication Critical patent/CN114437038A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The invention provides a novel compound capable of effectively inhibiting CD73 activity, which is a compound shown in formula I, a tautomer, a stereoisomer, a hydrate, a solvate, a pharmaceutically acceptable salt or a prodrug thereof, a preparation method thereof and application thereof in preparing medicines.

Description

Pyridazine alkyne compound and application thereof
PRIORITY INFORMATION
The present application claims priority and benefit from the patent application No. 202011224592.7 filed on 5.11.2020 to the chinese national intellectual property office and is hereby incorporated by reference in its entirety.
Technical Field
The invention belongs to the field of medical chemistry, particularly relates to a pyridazine alkyne compound, and more particularly relates to a pyridazine alkyne compound, a preparation method thereof, and application thereof in preparing medicines.
Background
CD73, also known as extracellular 5' -nucleotidohydrolase, an exonuclease belonging to the metal phosphatase superfamily, is a peripheral glycoprotein, which is anchored in the plasma membrane via Glycosylphosphatidylinositol (GPI), has a molecular weight of 70KD, and is encoded by the NT5E gene. CD73 is widely expressed on the cell surface of various tissues including brain, lung, heart, spleen, lymph nodes, kidney, colon, vascular endothelium and bone marrow; various immune cells are also expressed, including macrophages, neutrophils, myeloid suppressor cells (MDSCs), Dendritic Cells (DCs), natural killer cells (NK), and regulatory T cells (Treg) (Soleimani A et al, Biochimie,2020,176: 21-30.); a variety of tumor cells also have high expression of CD73, such as: melanoma, breast, pancreatic, ovarian, colon, and prostate cancer, among others (Gao Z et al, Biomed Res Int,2014,2014: 460654.). CD73 is also present in soluble form (sCD73) in biological fluids including serum and retains holoenzyme activity.
CD73 exerts a physiological, pathological role primarily by hydrolyzing AMP (adenosine monophosphate) to produce extracellular Adenosine (ADO) via binding to 4G protein-coupled receptors (GPCRs): a1 adenosine receptor (A1AR), A2A adenosine receptor (A2AR), A2B adenosine receptor (A2BR) and A3 adenosine receptor (A3AR), of which A2AR (Linden J et al, Annu. Rev. Immunol.,2019,37: 325-347) play a major role. Adenosine Receptors (ARs) are not only expressed in tumor cells, but also on the cell surface of immune cells and vascular endothelial cells infiltrating in the tumor microenvironment, and ADO, by binding to the receptors, produces a variety of immunosuppressive and tumor-promoting effects.
CD73 is closely associated with tumor growth, angiogenesis and metastasis. Under normal physiological conditions, extracellular ADO levels were between 20 and 300nM, but in the tumor microenvironment, were elevated to maintenance of micromolar levels (30-100 μ M), whereas high extracellular ADO concentrations were primarily affected by CD73 hydrolysis of AMP production. Studies have shown increased levels of soluble CD73(sCD73) in the plasma of cancer patients compared to healthy humans (Klemens M R et al, biochem. biophysis. res. commun.,1990,172: 1371-7.). In gastrointestinal stromal tumors, tumor-infiltrating NK cells express higher levels of CD73, while loss of A2AR signaling in NK cells may improve CD73+Tumor metastasis and enhanced anti-tumor immune responses (Young A et al, Cancer cell.2016; 30(3): 391-403.). CD73 is up-regulated in pancreatic ductal carcinoma (PDAC) compared to normal pancreatic tissue and is associated with tumor size, metastasis and poor prognosis (Harvey Jerry B et al, Front Immunol,2020,11: 508). In preclinical studies by the ORIC company, the CD73 selective inhibitor ORIC-533 significantly reduced ADO concentration in the tumor microenvironment while reducing tumor volume. The results of the studies show that the expression of CD73 is up-regulated in various tumors, and the inhibition of CD73 can possibly reduce the ADO concentration, thereby inhibiting the growth and metastasis of the tumors.
In addition to its single use, CD73 inhibitors can block tumor growth by relieving immunosuppression, and can be used in combination with other targeted therapies and/or immunotherapy, radiation therapy to increase anti-tumor effects. In several tumor models in mice, anti-CD 73 in combination with anti-PD-1/L1 (programmed death receptor 1/ligand 1) and/or anti-CTLA-4 (cytotoxic T lymphocyte-associated protein 4) antibodies was more effective than treatment with anti-PD-L1 and/or anti-CTLA-4 antibodies alone (Allard B et al, clin. cancer res.,2013,19: 5626-35.); the level of CD73 was found to be upregulated in melanoma patients receiving anti-PD-1 antibody immunotherapy, whereas a unique population of CD 73-highly expressed macrophages persists in glioblastoma patients following anti-PD-1 therapy, and CD73 deficiency enhances the efficacy of anti-PD-1 and anti-CTLA-4 in the mouse glioblastoma model (Goswami S et al, nat. med.,2020,26: 39-46.); radiotherapy causes destruction of a part of tumor cells, releases a large amount of ATP intracellularly to the outside of the cells, and converts it to adenosine under the action of CD73 on the surface of tumor cells or in the form of free ATP, resulting in immunosuppressive effects, which is considered to be one of the causes of poor prognosis after radiotherapy in a part of patients, and thus a synergistic effect may be generated by the combination of a CD73 inhibitor and radiotherapy (Wennerberg E et al, Cancer Immunol Res,2020,8: 465-478.).
Some of the anti-CD 73 mabs (MEDI9447, BMS986179, SRF373/NZV930, CPI-006/CPX-006, TJ004309) and selective small molecule inhibitors (LY3475070, AB680) have now entered clinical stage, with encouraging early outcome (NCT 54027141) in part of clinical trials, and CD73 inhibition may be a promising approach to treat tumors.
Disclosure of Invention
The invention aims to provide a novel CD73 inhibitor which can be used for preparing a medicament for treating tumor-related diseases.
In a first aspect of the present invention, the present invention provides a compound, which is a compound represented by formula I, a tautomer, a stereoisomer, a hydrate, a solvate, a pharmaceutically acceptable salt, or a prodrug thereof:
Figure BDA0003339874850000021
wherein the content of the first and second substances,
m is 0,1, 2,3 or 4;
Figure BDA0003339874850000022
in R1Independently selected from hydrogen, halogen, hydroxy, cyano, amino, unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-; the quilt RaSubstituted C1-C6Alkyl, or said RaSubstituted C1-C6In alkyl-O-, the radical is represented by RaThe substitution can be one or more, and R isaEach independently is the following substituent: halogen, hydroxy, cyano, amino, C1-C6Alkyl radical, C1-C6alkyl-O-, -COOH, -C (═ O) NH2(ii) a When the number of the substituents is plural, the substituents may be the same or different; when m is not 0 or 1, R1Independently are the same or different; n is 0,1, 2 or 3;
R2selected from hydrogen, unsubstituted or substituted by RbSubstituted C1-C6Alkyl, unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl, unsubstituted or substituted by RbSubstituted 5-8 membered aryl, unsubstituted or substituted by RbSubstituted 5-8 membered heteroaryl, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, or, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkenyl; the quilt RbSubstituted C1-C6Alkyl, said by RbSubstituted C3-C6Cycloalkyl radicals, said being RbSubstituted 5-8 membered aryl, said substituted RbSubstituted 5-8 membered heteroaryl, said substituted by RbSubstituted 4-8 membered heterocycloalkyl, or said substituted RbIn the substituted 4-8 membered heterocycloalkenyl group, the group represented by RbThe substitution may be one or more, and R isbEach independently is the following substituent: halogen, hydroxy, cyano, amino, carboxy, C3-C6Cycloalkyl radical, C1-C6Alkyl, C substituted by 1-5 identical or different halogens1-C6Alkyl, or, C1-C6alkyl-O-; when the number of the substituents is plural, the substituents may be the same or different;
said unsubstituted or substituted by RbIn the substituted 5-8-membered heteroaryl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocyclic alkyl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocycloalkenyl, the heteroatoms are selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3.
In a preferred embodiment of the present invention, the compound of formula I, tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug thereof is:
Figure BDA0003339874850000031
wherein the content of the first and second substances,
m is 0,1, 2,3 or 4;
Figure BDA0003339874850000032
in R1Independently selected from hydrogen, halogen, hydroxy, cyano, amino, unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-; the quilt RaSubstituted C1-C6Alkyl, or said RaSubstituted C1-C6In alkyl-O-, the substituents each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C1-C6Alkyl radical, C1-C6alkyl-O-, -COOH, -C (═ O) NH2(ii) a When the substituent is plural, the substituents are the same or different; when m is not 0 or 1, R1Independently are the same or different;
n is 0,1, 2 or 3;
R2selected from hydrogen, unsubstituted or substituted by RbSubstituted C1-C6Alkyl, unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl, unsubstituted or substituted by RbSubstituted 5-8 membered aryl, unsubstituted or substituted by RbSubstituted 5-8 membered heteroaryl, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, or, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkenyl; the quilt RbSubstituted C1-C6Alkyl, said by RbSubstituted C3-C6Cycloalkyl radicals, said being RbSubstituted 5-8 membered aryl, said substituted RbSubstituted 5-8 membered heteroaryl, said substituted by RbSubstituted 4-8 membered heterocycloalkyl, or said substituted RbIn substituted 4-8 membered heterocycloalkenyl, the substitutions each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C3-C6Cycloalkyl radical, C1-C6Alkyl, C substituted by 1-5 identical or different halogens1-C6Alkyl, or, C1-C6alkyl-O-; when the number of the substituents is plural, the substituents may be the same or different;
said unsubstituted or substituted by RbIn the substituted 5-8-membered heteroaryl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocyclic alkyl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocycloalkenyl, the heteroatoms are selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3.
In a preferred embodiment of the invention, when R is2Is unsubstituted or substituted by RbSubstituted C1-C6When alkyl, said C1-C6Alkyl is C1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl or isobutyl.
In a preferred embodiment of the invention, when R is2Is not takenIs substituted or substituted by RbSubstituted C1-C6When it is alkyl, the substituent RbThe number of (a) is 1 to 3, preferably 1.
In a preferred embodiment of the invention, when R is2Is unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl radical, said C3-C6Cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, preferably cyclopropyl or cyclobutyl.
In a preferred embodiment of the invention, when R is2Is unsubstituted or substituted by RbWhen substituted 5-8 membered aryl, said 5-8 membered aryl is independently phenyl or naphthyl, preferably phenyl.
In a preferred embodiment of the invention, when R is2Is unsubstituted or substituted by RbWhen substituted 5-8 membered heteroaryl, the 5-8 membered heteroaryl is independently pyrrole, pyrazole, triazole, furan, oxazole, thiophene, thiazole, pyridine, pyrazine or pyrimidine, preferably pyrazole, furan, thiophene, pyridine.
In a preferred embodiment of the invention, when R is2Is unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, said 4-8 membered heterocycloalkyl is independently azetidine, oxetane, tetrahydropyrrolyl, tetrahydrofuranyl, hexahydropyran or tetrahydro-2H-thiopyran 1, 1-dioxide, preferably azetidine or oxetane.
In a preferred embodiment of the invention, when R is2Is unsubstituted or substituted by RbWhen substituted with a 4-8 membered heterocycloalkenyl group, the 4-8 membered heterocycloalkenyl group is independently a dihydropyridinyl group, a tetrahydropyridinyl group, a tetrahydropyrimidinyl group, a pyrrolinyl group, an imidazolinyl group, a pyrazolinyl group, a dihydroimidazolyl group, a dihydropyrazolyl group, a dihydrooxazolyl group, a dihydrooxadiazolyl group, a dihydrothiazolyl group, a dihydroisothiazolyl group, a dihydrothienyl group, a dihydropyrrolyl group, a3, 4-dihydro-2H-pyranyl group, a dihydrofuranyl group, a dihydropyrazinyl group, a dihydropyrimidyl group or a fluorodihydrofuranyl group, preferably a1, 2,3, 4-tetrahydropyridinyl group, a1, 2-dihydropyridinyl group, a1, 4-dihydropyridinyl group, a1, 2,3, 6-tetrahydropyridinyl group, a3, 4-dihydro-2H-pyranyl group or a dihydrofuranyl groupA furyl group.
In a preferred embodiment of the invention, RbIs a hydroxyl group.
In a preferred embodiment of the invention, when R isbIs C1-C6When alkyl, said C1-C6Alkyl is C1-C4Alkyl, preferably methyl, ethyl, n-propyl or isopropyl.
In a preferred embodiment of the invention, when R isbWhen the halogen is F, Cl, Br, I, preferably F or Cl.
In a preferred embodiment of the present invention,
Figure BDA0003339874850000041
is composed of
Figure BDA0003339874850000042
Preferably is
Figure BDA0003339874850000043
In a preferred embodiment of the invention, when R is1When halogen is used, the halogen is F, Cl, Br or I, preferably F or Cl.
In a preferred embodiment of the invention, when R is1When it is halogen, m is 0,1 or 2.
In a preferred embodiment of the invention, when R is1Is unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-said C1-C6Alkyl is independently C1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl or isobutyl.
In a preferred embodiment of the invention, when R is1Is unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6When alkyl-O-, m is 1 or 2, preferably m is 1.
In bookIn a preferred embodiment of the invention, when R is1Is as a quilt RaSubstituted C1-C6Alkyl, or, by RaSubstituted C1-C6In the case of alkyl-O-, the number of said substitution is independently 1 to 3, preferably 2.
In a preferred embodiment of the invention, when R is1Is as a quilt RaSubstituted C1-C6Alkyl, or, by RaSubstituted C1-C6When alkyl-O-, the substituents are each independently C1-C6Alkyl, or C1-C6alkyl-O-, C as described for said substitution1-C6Alkyl is independently C1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl or isobutyl.
In a preferred embodiment of the invention, when R isaWhen halogen is used, the halogen is F, Cl, Br or I, preferably F or Cl.
In a preferred embodiment of the present invention, when
Figure BDA0003339874850000044
Is composed of
Figure BDA0003339874850000045
When is in use, the
Figure BDA0003339874850000046
Is composed of
Figure BDA0003339874850000047
Figure BDA0003339874850000048
In a preferred embodiment of the present invention, when
Figure BDA0003339874850000049
Is composed of
Figure BDA00033398748500000410
When is in use, the
Figure BDA00033398748500000411
Is composed of
Figure BDA00033398748500000412
In a preferred embodiment of the present invention, when
Figure BDA00033398748500000413
Is composed of
Figure BDA00033398748500000414
When is in use, the
Figure BDA00033398748500000415
Is composed of
Figure BDA00033398748500000416
Figure BDA0003339874850000051
In a preferred embodiment of the present invention, when
Figure BDA0003339874850000052
Is composed of
Figure BDA0003339874850000053
When is in use, the
Figure BDA0003339874850000054
Is composed of
Figure BDA0003339874850000055
In a preferred embodiment of the present invention,
Figure BDA0003339874850000056
is composed of
Figure BDA0003339874850000057
In a preferred embodiment of the invention, R2Is composed of
Figure BDA0003339874850000058
In a preferred embodiment of the present invention,
Figure BDA0003339874850000059
is composed of
Figure BDA00033398748500000510
In a preferred embodiment of the present invention, the compound represented by formula I, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug is:
Figure BDA00033398748500000511
wherein the content of the first and second substances,
R1independently selected from hydrogen, halogen, hydroxy, cyano, amino, unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-; the quilt RaSubstituted C1-C6Alkyl, or said RaSubstituted C1-C6In alkyl-O-, the substituents each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C1-C6Alkyl radical, C1-C6alkyl-O-, -COOH, -C (═ O) NH2(ii) a When the number of the substituents is plural, the substituents may be the same or different;
R2selected from hydrogen, unsubstituted or substituted by RbSubstituted C1-C6Alkyl, unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl, unsubstituted or substituted by RbSubstituted 5-8 membered aryl, unsubstituted or substituted by RbSubstituted 5-8 membered heteroaryl, unsubstituted or substituted by RbSubstituted 4-8 membered heterocyclic ringsAlkyl, or, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkenyl; the quilt RbSubstituted C1-C6Alkyl, said by RbSubstituted C3-C6Cycloalkyl radicals, said being RbSubstituted 5-8 membered aryl, said substituted by RbSubstituted 5-8 membered heteroaryl, said substituted by RbSubstituted 4-8 membered heterocycloalkyl, or said substituted RbIn substituted 4-8 membered heterocycloalkenyl, the substitutions each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C3-C6Cycloalkyl radical, C1-C6Alkyl, C substituted by 1-5 identical or different halogens1-C6Alkyl, or, C1-C6alkyl-O-; when the number of the substituents is plural, the substituents may be the same or different;
said unsubstituted or substituted by RbIn the substituted 5-8-membered heteroaryl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocyclic alkyl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocycloalkenyl, the heteroatoms are selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3.
In a preferred embodiment of the present invention, the compound represented by formula I, the hydrate, the solvate, the pharmaceutically acceptable salt or the prodrug thereof is
Figure BDA0003339874850000061
Wherein R is1And R2Having the definitions as previously described.
In a preferred embodiment of the invention, R1Selected from difluoromethyl, trifluoromethyl, dichloromethyl, trichloromethyl or isopropyl; r2Selected from methyl, ethyl or cyclopropyl.
In a preferred embodiment of the present invention, the compound according to formula I, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug is selected from any one of the following compounds:
Figure BDA0003339874850000062
in a second aspect of the present invention, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of the above compound, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug, and a pharmaceutically acceptable excipient.
According to a specific embodiment of the present invention, the pharmaceutical composition of the present invention may include a therapeutically effective amount of the above-mentioned compound, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug, and a pharmaceutically acceptable carrier, diluent or excipient mixed to prepare a pharmaceutical preparation suitable for oral or parenteral administration. Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, and oral routes. The formulations may be administered by any route, for example by infusion or bolus injection, by a route of absorption through epithelial or cutaneous mucosa (e.g. oral mucosa or rectum, etc.). Administration may be systemic or local. Examples of the formulation for oral administration include solid or liquid dosage forms, specifically, tablets, pills, granules, powders, capsules, syrups, emulsions, suspensions and the like. The formulations may be prepared by methods known in the art and include carriers, diluents or excipients conventionally used in the art of pharmaceutical formulation.
In a third aspect of the invention, the invention provides the use of the above compound, or a tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug thereof, pharmaceutical composition in combination with PD-1 antibody, PD-L1 antibody, CTLA-4 antibody or PD-1 inhibitor, PD-L1 inhibitor, CTLA-4 inhibitor for the manufacture of a medicament for the treatment of CD 73-associated diseases, which medicament is useful for the treatment of cancer. These cancers include, for example, bladder cancer, breast cancer, cholangiocarcinoma, rectal cancer, colon cancer, stomach cancer, gallbladder cancer, glioblastoma, head and neck cancer, liver cancer, lung cancer, lymphoma, medulloblastoma, melanoma, ovarian cancer, pancreatic cancer, prostate cancer, or renal cancer.
In a fourth aspect of the present invention, the present invention provides the use of the above compound, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug, or the above pharmaceutical composition in the preparation of a medicament for treating diseases related to CD 73.
According to a specific embodiment of the invention, the compound or the tautomer, the stereoisomer, the hydrate, the solvate, the pharmaceutically acceptable salt or the prodrug thereof or the pharmaceutical composition is used for preparing a medicament for treating CD73 related diseases, and the medicament can be used for treating cancers. These cancers include, for example, bladder cancer, breast cancer, cholangiocarcinoma, colorectal cancer, colon cancer, gastric cancer, gallbladder cancer, glioblastoma, head and neck cancer, liver cancer, lung cancer, lymphoma, medulloblastoma, melanoma, ovarian cancer, pancreatic cancer, prostate cancer, or renal cancer.
Terms and definitions
Unless otherwise indicated, the terms and definitions used in the present application, including in the specification and claims of the present application, are as follows.
It will be understood by those skilled in the art that, according to the convention used in the art, in the structural formulae of the present application,
Figure BDA0003339874850000071
for delineating chemical bonds, which are the points at which moieties or substituents are attached to a core structure or a backbone structure.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, 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 pharmaceutically acceptable salts of non-toxic acids or bases, including salts of inorganic acids and bases, organic acids and bases.
In addition to pharmaceutically acceptable salts, other salts are also contemplated by the present invention. They may serve as intermediates in the purification of the compounds or in the preparation of other pharmaceutically acceptable salts or may be used in the identification, characterization or purification of the compounds of the invention.
The term "pharmaceutical composition" denotes a mixture of one or more compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof with other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate the administration of the compound to an organism.
The term "adjuvant" refers to a pharmaceutically acceptable inert ingredient. Examples of classes of the term "excipient" include, without limitation, binders, disintegrants, lubricants, glidants, stabilizers, fillers, diluents, and the like. Excipients enhance the handling characteristics of the pharmaceutical formulation, i.e., make the formulation more amenable to direct compression by increasing flowability and/or cohesiveness.
The term "prodrug" refers to a compound of the invention that can be converted to a biologically active compound under physiological conditions or by solvolysis. Prodrugs of the invention are prepared by modifying functional groups in the compounds, which modifications may be routinely made or removed in vivo to give the parent compound. Prodrugs include compounds of the present invention wherein a hydroxy or amino group is attached to any group that, when administered to a mammalian subject, cleaves to form a free hydroxy or a free amino group, respectively.
The term "stereoisomer" refers to isomers resulting from the different arrangement of atoms in a molecule, including cis-trans isomers, enantiomers, diastereomers, and conformers.
Depending on the choice of starting materials and process, the compounds according to the invention may be present as one of the possible isomers or as a mixture thereof, for example as pure optical isomers, or as a mixture of isomers, for example as racemic and diastereomeric mixtures, depending on the number of asymmetric carbon atoms. When describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule with respect to the chiral center (or centers) in the molecule. The prefixes D and L or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or L indicates that the compound is left-handed. Compounds prefixed with (+) or D are dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of each other. A particular stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often referred to as a mixture of enantiomers. A 50:50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process. Many geometric isomers of olefins, C ═ N double bonds, and the like, may also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. When compounds described herein contain olefinic double bonds, such double bonds include both E and Z geometric isomers, unless otherwise specified. If the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may be in the cis or trans (cis-or trans-) configuration.
When bonds to chiral carbons in the formulae of the present invention are depicted as straight lines, it is to be understood that both the (R) and (S) configurations of the chiral carbons and their enantiomerically pure compounds and mixtures resulting therefrom are included within the scope of this formula. The enantiomers or enantiomerically pure compounds herein are illustrated by Maehr, J.chem.Ed.1985, 62: 114-120. Unless otherwise indicated, the absolute configuration of a stereocenter is indicated by wedge bonds and dashed bonds.
Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral preparations, or resolved using conventional techniques. The compounds of the present invention containing asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Resolution of racemic mixtures of compounds can be carried out by any of a number of methods known in the art. Exemplary methods include fractional recrystallization using chiral resolving acids, which are optically active salt-forming organic acids. Suitable resolving agents for use in the fractional recrystallization process are, for example, the D and L forms of optically active acids, such as tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or various optically active camphorsulfonic acids, such as β -camphorsulfonic acid. Other resolving agents suitable for fractional crystallization processes include stereoisomerically pure forms of α -methyl-benzylamine (e.g., S and R forms or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1, 2-diaminocyclohexane, and the like. Resolution of the racemic mixture can also be carried out by elution on a chromatographic column packed with an optically active resolving agent (e.g. dinitrobenzoylphenylglycine). The method can be performed by High Performance Liquid Chromatography (HPLC) or Supercritical Fluid Chromatography (SFC). The choice of the particular method and the conditions of elution, the choice of the chromatography column can be selected by the person skilled in the art according to the structure of the compound and the results of the test. Further, any enantiomer or diastereomer of the compounds described herein may also be obtained by stereoorganic synthesis using optically pure starting materials or reagents of known configuration.
The term "tautomer" refers to an isomer of a functional group resulting from the rapid movement of an atom in two positions in a molecule. The compounds of the invention may exhibit tautomerism. Tautomeric compounds may exist in two or more interconvertible species. Prototropic tautomers result from the migration of a covalently bonded hydrogen atom between two atoms. Tautomers generally exist in equilibrium, and attempts to isolate a single tautomer often result in a mixture whose physicochemical properties are consistent with the mixture of compounds. The position of equilibrium depends on the chemical properties within the molecule. For example, in many aliphatic aldehydes and ketones such as acetaldehyde, the keto form predominates; whereas in phenol the enol type predominates. The present invention encompasses all tautomeric forms of the compounds.
The compounds of the invention may be present in one or more of the constituent partsThe atoms of the compounds contain unnatural proportions of atomic isotopes. For example, the compounds may be labelled with radioisotopes, such as deuterium (g) ((R))2H) Tritium (A)3H) Iodine-125 (125I) Or C-14(14C) In that respect All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
The term "effective amount" or "therapeutically effective amount" with respect to a drug or pharmacologically active agent refers to a sufficient amount of the drug or agent that is non-toxic but achieves the desired effect. For oral dosage forms of the invention, an "effective amount" of one active agent in a composition is the amount required to achieve the desired effect when combined with another active agent in the composition. The determination of an effective amount varies from person to person, depending on the age and general condition of the recipient and also on the particular active substance, and an appropriate effective amount in an individual case can be determined by a person skilled in the art according to routine tests.
The terms "active ingredient," "therapeutic agent," "active substance," or "active agent" refer to a chemical entity that is effective in treating a target disorder, disease, or condition.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent, including deuterium and hydrogen variants, so long as the valency of the particular atom is normal and the substituted compound is stable. When the substituent is a keto group (i.e., ═ O), it means that two hydrogen atoms are substituted. The keto substitution does not occur on the aromatic group. The term "optionally substituted" means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemical realizability.
The term "C1-C6Alkyl "is understood to mean a straight-chain or branched, saturated monovalent hydrocarbon radical having 1,2,3,4, 5 or 6 carbon atoms. Alkyl is, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-ethylpropyl, 1, 2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 2, 3-dimethylbutyl, 1, 3-dimethylbutyl or 1, 2-dimethylbutyl and the like or isomers thereof. In particular, the radicals have 1,2 or 3 carbon atoms ("C)1-C3Alkyl groups) such as methyl, ethyl, n-propyl or isopropyl.
The term "C1-C6alkyl-O- "is understood to mean that the alkyl radical is bonded to the rest of the molecule via an oxygen atom, where" C "is1-C6Alkyl "has the above definition. Such as methyl-O-, ethyl-O-.
The term "C3-C6Cycloalkyl "is understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring having 3 to 6 carbon atoms, including fused or bridged polycyclic ring systems. Such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
The term "4-8 membered heterocyclyl" or "4-8 membered heterocycloalkyl" is understood to mean a saturated, unsaturated or partially saturated monocyclic, bicyclic or tricyclic ring having 4 to 8 atoms, wherein 1,2,3,4 or 5 ring atoms are selected from N, O and S, which may be connected through carbon or nitrogen, unless otherwise indicated, wherein-CH is2-The group is optionally replaced by-C (O) -; and wherein unless otherwise stated to the contrary, the ring nitrogen atom or the ring sulfur atom is optionally oxidized to form an N-oxide or S-oxide or the ring nitrogen atom is optionally quaternized; wherein-NH in the ring is optionally substituted with acetyl, formyl, methyl or methanesulfonyl; and the ring is optionally substituted with one or more halogens. It is understood that when the total number of S and O atoms in the heterocyclic group exceeds 1, these heteroatoms are not adjacent to each other. If the heterocyclyl is bicyclic or tricyclic, at least one ring may optionally be a heteroaromatic ring or an aromatic ring, provided that at least one ring is non-heteroaromatic. If the heterocyclic group is monocyclic, it is not necessarily aromatic. Examples of heterocyclyl groups include, but are not limited to, piperidinyl, N-acetylpiperidinyl, N-methylpiperidinyl, N-formylpiperazinyl, N-methylsulfonylpiperazinyl, homopiperazinyl, piperazinyl, azetidinyl, piperidinyl, piperazinyl, and the like,Oxetanyl, morpholinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, indolinyl, tetrahydropyranyl, dihydro-2H-pyranyl, tetrahydrofuranyl, tetrahydrothiopyranyl-1-oxide, tetrahydrothiopyranyl-1, 1-dioxide, 1H-pyridin-2-one, and 2, 5-dioxoimidazolidinyl.
The term "4-8 membered heterocycloalkenyl" is to be understood as a non-aromatic mono-or polycyclic group containing 4 to 8 ring atoms, preferably 5 to 6 ring atoms, wherein the 4-8 membered heterocycloalkenyl comprises 1 to 3 heteroatoms selected from N, O, S and P and contains at least one carbon-carbon double bond or carbon-nitrogen double bond. An aza, oxa or thia comprised in the group name means that at least one nitrogen, oxygen or sulfur atom respectively is a ring atom. The nitrogen or sulfur atom of the 4-8 membered heterocycloalkenyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S-dioxide. Preferred 4-8 membered heterocycloalkenyl groups include, but are not limited to, 1,2,3, 4-tetrahydropyridyl, 1, 2-dihydropyridyl, 1, 4-dihydropyridyl, 1,2,3, 6-tetrahydropyridyl, 1,4,5, 6-tetrahydropyrimidinyl, 2-pyrrolinyl, 3-pyrrolinyl, 2-imidazolinyl, 2-pyrazolinyl, dihydroimidazolyl, dihydrooxazolyl, dihydrooxadiazolyl, dihydrothiazolyl, 3, 4-dihydro-2H-pyranyl, dihydrofuranyl, fluorodihydrofuranyl, oxides thereof, and the like. "4-8 membered heterocycloalkenyl" may also include two available hydrogen atoms on the same carbon atom of the ring simultaneously substituted with a single group ═ O (i.e., to form a carbonyl group).
The term "5-to 8-membered aryl" is to be understood as meaning a mono-, bi-or tricyclic hydrocarbon ring having a monovalent or partial aromaticity of 5 to 8 carbon atoms, in particular a ring having 6 carbon atoms ("C6Aryl "), such as phenyl; when the 5-to 8-membered aryl group is substituted, it may be mono-or poly-substituted. And, the substitution site thereof is not limited, and may be, for example, ortho-, para-or meta-substitution.
The term "5-8 membered heteroaryl" is to be understood as a monovalent monocyclic, bicyclic or tricyclic aromatic ring group having 5-8 ring atoms, in particular 5 or 6 carbon atoms, and comprising 1-5 heteroatoms independently selected from N, O and S. Preferably 1 to 3 and independently selected from N, O and S, and further may be benzo-fused in each case. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl and the like; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and the like.
The term "halo" or "halogen" is fluorine, chlorine, bromine and iodine.
In addition, it should be noted that, unless otherwise explicitly indicated, the recitation of "… … independently" as used herein is to be understood in a broad sense to mean that each individual species so described is independent of the other and may be the same or different specific groups. In more detail, the expression "… … independently" can mean that the specific options expressed between the same symbols do not affect each other in different groups, or that the specific options expressed between the same symbols do not affect each other in the same groups.
Advantageous effects
According to the embodiment of the invention, the CD73 inhibitor which has a novel structure, excellent pharmacokinetic property and good drug effect or drug success rate is provided, and can be used for effectively treating CD73 related diseases and symptoms.
The compound of the invention has good inhibition effect on CD73 enzyme and good in vitro drug effect. In addition, mouse experiment results show that the compound of the invention has excellent pharmacokinetic properties and good drug forming property.
In addition, the compound of the invention has obvious effect of inhibiting the growth of CT-26 colorectal cancer and E.G7-OVAT cell lymphoma by being used alone or being used together with PD-1/L1 antibody.
Additional aspects and advantages of the invention will be set forth in part in the description which follows and, in part, will be obvious from the description, or may be learned by practice of the invention.
Detailed Description
The scheme of the invention will be explained with reference to the examples. It will be appreciated by those skilled in the art that the following examples are illustrative of the invention only and should not be taken as limiting the scope of the invention. The examples, where specific techniques or conditions are not indicated, are to be construed according to the techniques or conditions described in the literature in the art or according to the product specifications. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
Unless otherwise specified, the compounds of the present invention are structurally defined by Nuclear Magnetic Resonance (NMR) and/or Mass Spectrometry (MS). NMR shift in units of 10-6(ppm). Solvents for NMR measurement were deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and an internal standard was Tetramethylsilane (TMS).
Abbreviations of the present invention are defined as follows:
m: molar concentration, e.g. 1M hydrochloric acid for 1mol/L hydrochloric acid solution
DCM: methylene dichloride
DMP: dess-martin oxidizer
DMF N, N-dimethylformamide
DAST: diethylaminosulfur trifluoride
DMSO, DMSO: dimethyl sulfoxide
A dioxane: 1, 4-dioxane
TEA: triethylamine
THF: tetrahydrofuran (THF)
TEMPO: 2,2,6, 6-tetramethylpiperidine oxide
LC-MS: liquid chromatography-mass spectrometry
IC50: the median inhibitory concentration is the concentration at which half of the maximal inhibitory effect is achieved.
Comparative example 1: preparation of Positive control Compound 1
5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (control Compound 1)
Figure BDA0003339874850000101
Reference is made to the process of patent WO2019168744 Al.
1H NMR(400MHz,DMSO-d6)δ11.51(s,2H),8.27(s,1H),7.77(s,1H),6.17-5.85(m,1H),2.69(s,3H),2.32-2.29(m,1H),1.77-1.70(m,1H),1.32-1.18(m,2H).
LC-MS,M/Z(ESI):295.0[M+H]+
The "control Compound 1" mentioned below refers to the compound described in comparative example 1.
Comparative example 2: preparation of Positive control Compound 2
5- (5- ((1S,2R) -2-isopropylcyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (control Compound 2)
Figure BDA0003339874850000111
Reference is made to the process of patent WO2019168744 Al.
1H NMR(400MHz,CD3OD)δ8.65(s,1H),8.22(s,1H),2.89(s,3H),2.07-2.10(m,1H),1.33-1.45(m,4H),1.10(d,6H).
LC-MS,M/Z(ESI):287.0[M+H]+
The "control Compound 2" mentioned below refers to the compound described in comparative example 2.
Preparation 1: preparation of intermediate A
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (intermediate A)
Figure BDA0003339874850000112
The synthetic route for intermediate a is shown below:
Figure BDA0003339874850000113
the first step is as follows: synthesis of ethyl (1S,2S) -2- ((benzyloxy) methyl) cyclopropane-1-carboxylate (A-3)
Figure BDA0003339874850000121
Sodium hydride (58.5g, 1.46mol, content 60%) was suspended in toluene (3000mL) under nitrogen, triethyl phosphonoacetate (327.7g, 1.46mol) was added dropwise at 0 ℃ and stirred at 25 ℃ for 1 hour after completion of the addition, and then (S) - (+) -glycidyl benzyl ether (200g, 1.22mol) was added to the reaction mixture and the temperature was raised to 130 ℃ for reaction for 12 hours. The reaction mixture was diluted with water (5000mL), then extracted with ethyl acetate (2000 mL. times.2), the organic layers were combined, the organic layer was washed with saturated brine (2000mL), dried over sodium sulfate, and concentrated to give the crude product. Separation and purification with silica gel column (petroleum ether: ethyl acetate (V/V) ═ 50:1-10:1, gradient elution) gave ethyl (1S,2S) -2- ((benzyloxy) methyl) cyclopropane-1-carboxylate (a-3) as a red oil (180g, 63% yield).
1H NMR(400MHz,CDCl3)δ7.18-7.25(m,5H),4.42(s,2H),4.00-4.04(m,2H),3.33-3.37(m,1H),3.24-3.28(m,1H),1.62-1.66(m,1H),1.46-1.49(m,1H),1.06-1.16(m,4H),0.75-0.78(m,1H).
The second step is that: synthesis of ethyl (1S,2S) -2- (hydroxymethyl) cyclopropane-1-carboxylate (A-4)
Figure BDA0003339874850000122
Ethyl (1S,2S) -2- ((benzyloxy) methyl) cyclopropane-1-carboxylate (A-3) (120g, 512.2mmol) was dissolved in ethanol (1200mL), and palladium on carbon (30.0g, content 10%) was added under nitrogen protection, followed by substitution 3 times with hydrogen gas and reaction at 50Psi pressure for 24 hours. Cooled to room temperature, filtered through celite to remove palladium on carbon, the filter cake was washed 3 times with ethanol, and the filtrate was concentrated to give (1S,2S) -ethyl 2- (hydroxymethyl) cyclopropane-1-carboxylate (a-4) as a yellow oil (65.0g, 85% yield).
The third step: synthesis of ethyl (1S,2S) -2-formylcyclopropane-1-carboxylate (A-5)
Figure BDA0003339874850000123
Ethyl (1S,2S) -2- (hydroxymethyl) cyclopropane-1-carboxylate (A-4) (100g, 693.6mmol) was dissolved in dichloromethane (1500mL), and dess-martin oxidant (353.0g, 832.4mmol) was added slowly at 0 ℃ and then reacted at 25 ℃ for 12 hours. After completion of the reaction, the reaction solution was poured into an aqueous sodium carbonate solution (500mL) and an aqueous sodium sulfite solution (500mL), followed by extraction with methylene chloride (2000 mL. times.2), and the organic phases were combined, washed with a saturated saline solution (500mL), dried over sodium sulfate, filtered, and concentrated to give ethyl (1S,2S) -2-formylcyclopropane-1-carboxylate (A-5) as a yellow oil (67.0g, yield 68%).
1H NMR(400MHz,CDCl3)δ9.31(d,1H),4.18(q,2H),2.40-2.46(m,1H),2.24-2.28(m,1H),1.59-1.64(m,1H),1.50-1.54(m,1H),1.35-1.20(m,3H).
The fourth step: synthesis of ethyl (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylate (A-6)
Figure BDA0003339874850000124
Ethyl (1S,2S) -2-formylcyclopropane-1-carboxylate (A-5) (95g, 668.3mmol) was dissolved in methylene chloride (1200mL), and diethylaminosulfur trifluoride (237.0g, 194mL, 1.47mol) was added dropwise at 0 ℃ and the reaction was stirred at 25 ℃ for 2 hours. The reaction mixture was quenched with saturated aqueous sodium bicarbonate (1000mL) and then extracted with dichloromethane (500mL × 2), the organic layers were combined, the organic phase was washed with saturated brine (1000mL), dried over sodium sulfate, and concentrated to give ethyl (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylate (a-6) (95g, crude) as a yellow oil which was used directly in the next step.
1H NMR(400MHz,CDCl3)δ5.62-5.91(m,1H),4.16(q,2H),1.88-1.96(m,2H),1.26-1.30(m,4H),1.12-1.16(m,1H).
The fifth step: synthesis of (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylic acid (A-7)
Figure BDA0003339874850000131
Ethyl (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylate (A-6) (95.0g, 578.7mmol) was dissolved in methanol (500mL) and water (100mL), followed by addition of sodium hydroxide (69.5g, 1.74mmol) and reaction at 25 ℃ for 12 hours. After completion of the reaction, the reaction mixture was concentrated, water (500mL) was added, extraction was performed with methyl t-butyl ether (500mL × 2), the aqueous phase was collected, the pH of the aqueous phase was adjusted to 3 with 1M hydrochloric acid, followed by extraction with ethyl acetate (500mL × 3), the organic phases were combined, washed with saturated brine (500mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylic acid (a-7) (47.0g, yield 60%) as a yellow oil.
1H NMR(400MHz,CDCl3)δ9.52(br.s,1H),5.65-5.94(m,1H),1.89-1.94(m,2H),1.34-1.37(m,1H),1.23-1.27(m,1H).
And a sixth step: synthesis of 3, 6-dichloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazine (A-9)
Figure BDA0003339874850000132
3, 6-dichloropyridazine (6.60g, 44.3mmol) and (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylic acid (6.03g, 44.3mmol) were dissolved in water (150mL), concentrated sulfuric acid (6.74mL) was added, and the temperature was raised to 70 ℃ under nitrogen. Then, an aqueous solution of silver nitrate (4.20g, 24.7mmol, 7.5mL) was added rapidly, and then an aqueous solution of ammonium persulfate (30.3g, 132.9mmol, 75mL) was added slowly dropwise, and the reaction was continued at 70 ℃ for 2 hours. The reaction solution was adjusted to pH 9 with aqueous ammonia, followed by extraction with ethyl acetate (500 mL. times.2), and the organic layers were combined, and the organic phase was washed with saturated brine (500mL), dried over sodium sulfate, and concentrated to give a crude product. Purification by silica gel column separation (petroleum ether: ethyl acetate (V/V) ═ 20:1-3:1, gradient elution) afforded 3, 6-dichloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazine (a-9) as a yellow oil (5.00g, 46.4% yield).
1H NMR(400MHz,CDCl3)δ7.10(s,1H),5.79-6.08(m,1H),2.39-2.45(m,1H),1.68-1.75(m,1H),1.51-1.54(m,1H),1.22-1.25(m,1H).
LC-MS,M/Z(ESI):239.1[M+H]+
The seventh step: synthesis of 3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (A)
Figure BDA0003339874850000133
3, 6-dichloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazine (5.00g, 20.9mmol) and 2, 4-dimethoxypyrimidine-5-boronic acid (3.85g, 20.9mmol) were dissolved in 1, 4-dioxane (50mL) and water (10mL), sodium carbonate (6.65g, 62.7mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium (1.53g, 2.09mmol) were added under nitrogen, and the mixture was heated to 70 ℃ for 1 hour. The reaction mixture was diluted with water (50mL) and extracted with ethyl acetate (100 mL. times.3), the organic phases were combined, the organic phase was washed with saturated brine (100mL), dried over sodium sulfate and concentrated to give the crude product. Separation and purification on silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-2:1, gradient elution) gave 3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (a) as a yellow oil (4.5g, 48% yield).
LC-MS,M/Z(ESI):343.1[M+H]+
Preparation 2: preparation of intermediate B
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (intermediate B)
Figure BDA0003339874850000141
The synthetic route of intermediate B is shown below:
Figure BDA0003339874850000142
the first step is as follows: synthesis of tert-butyl (1S,2S) -2- ((benzyloxy) methyl) cyclopropane-1-carboxylate (B-3)
Figure BDA0003339874850000143
Sodium hydride (14.6g, 365.4mmol, content 60%) was suspended in toluene (500mL) under nitrogen, followed by dropwise addition of tert-butyl diethylphosphonoacetate (92.2g, 365.4mmol), stirring at 25 ℃ for 30 minutes after completion of the addition, and then (S) - (+) -glycidyl benzyl ether (50.0g, 304.5mmol) was added to the reaction mixture, and the temperature was raised to 130 ℃ for reaction for 8 hours. The reaction mixture was diluted with water (100mL) and extracted with ethyl acetate (100 mL. times.2), the organic layers were combined, the organic phase was washed with saturated brine (50mL), dried over sodium sulfate, and concentrated to give the crude product. Separation and purification by silica gel column (petroleum ether: ethyl acetate (V/V) ═ 50:1-10:1, gradient elution) gave the compound tert-butyl (1S,2S) -2- ((benzyloxy) methyl) cyclopropane-1-carboxylate (B-3) as a yellow oil (55g, yield 68.8%).
The second step is that: synthesis of tert-butyl (1S,2S) -2- (hydroxymethyl) cyclopropane-1-carboxylate (B-4)
Figure BDA0003339874850000144
Tert-butyl (1S,2S) -2- ((benzyloxy) methyl) cyclopropane-1-carboxylate (B-3) (55g, 209.6mmol) was dissolved in ethanol (500mL), palladium on carbon (20.0g, content 10%) was added under nitrogen protection, followed by substitution 3 times with hydrogen gas and reaction at 50 ℃ for 24 hours under a pressure of 50 Psi. After cooling to room temperature, palladium on carbon was removed by filtration through celite, the cake was washed 3 times with ethanol, and the filtrate was concentrated to give (1S,2S) -tert-butyl 2- (hydroxymethyl) cyclopropane-1-carboxylate (B-4) (36.0g, yield 99.7%) as a yellow oil.
1H NMR(400MHz,CDCl3)δ3.50–3.63(m,2H),1.67–1.72(m,1H),1.47(s,9H),1.38(t,1H),1.14–1.89(m,1H),0.78-0.84(m,1H)。
The third step: synthesis of tert-butyl (1S,2S) -2- (fluoromethyl) cyclopropane-1-carboxylate (B-5)
Figure BDA0003339874850000151
Tert-butyl (1S,2S) -2- (hydroxymethyl) cyclopropane-1-carboxylate (B-4) (2.5g, 14.5mmol) was dissolved in methylene chloride (25mL), diethylaminosulfur trifluoride (4.68g, 3.84mL, 29.0mmol) was added dropwise at 0 ℃ and the reaction was stirred at 0 ℃ for 1 hour. The reaction mixture was quenched with saturated aqueous sodium bicarbonate (100mL) and then extracted with dichloromethane (100 mL. times.2), the organic layers were combined, the organic phase was washed with saturated brine (50mL), dried over sodium sulfate, and concentrated to give the crude product. Separation and purification by silica gel column (petroleum ether: ethyl acetate (V/V) ═ 50:1-10:1, gradient elution) gave (1S,2S) -2- (fluoromethyl) cyclopropane-1-carboxylic acid tert-butyl ester (B-5) as a yellow oil (2.0g, yield 79%).
1H NMR(400MHz,CDCl3)δ4.19-4.42(m,2H),1.75–1.82(m,1H),1.55-1.59(m,1H),1.46(s,9H),1.18–1.23(m,1H),0.83-0.88(m,1H)。
The fourth step: synthesis of (1S,2S) -2- (fluoromethyl) cyclopropane-1-carboxylic acid (B-6)
Figure BDA0003339874850000152
Tert-butyl (1S,2S) -2- (fluoromethyl) cyclopropane-1-carboxylate (B-5) (2.0g, 11.5mmol) was dissolved in a solution of hydrogen chloride (4M) in 1, 4-dioxane (10mL), and the mixture was stirred at 20 ℃ for 1 hour. The reaction was concentrated to give (1S,2S) -2- (fluoromethyl) cyclopropane-1-carboxylic acid (B-6) (1.3g, 95% yield) as a yellow oil.
1H NMR(400MHz,CDCl3)δ4.16-4.52(m,2H),1.88-1.94(m,1H),1.67-1.71(m,1H),1.34–1.37(m,1H),1.01-1.05(m,1H)。
The fifth step: synthesis of 3, 6-dichloro-4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (B-8)
Figure BDA0003339874850000153
3, 6-dichloropyridazine (630mg, 4.23mmol) and (1S,2S) -2- (fluoromethyl) cyclopropane-1-carboxylic acid (B-6) (500mg, 4.23mmol) were dissolved in water, concentrated sulfuric acid (0.5mL) was added, and the temperature was raised to 70 ℃ under nitrogen. Then, an aqueous solution of silver nitrate (359.6mg, 2.12mmol, 5mL) was added rapidly, and then an aqueous solution of ammonium persulfate (2.90g, 12.7mmol, 10mL) was added slowly dropwise, and the reaction was continued at 70 ℃ for 1 hour. The reaction solution was adjusted to pH 9 with aqueous ammonia, followed by extraction with ethyl acetate (100 mL. times.2), and the organic layers were combined, and the organic phase was washed with saturated brine (50mL), dried over sodium sulfate, and concentrated to give a crude product. Purification by silica gel column separation (petroleum ether: ethyl acetate (V/V) ═ 10:1-3:1, gradient elution) gave 3, 6-dichloro-4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (B-8) as a yellow oil (500mg, yield 26%).
And a sixth step: synthesis of 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (B)
Figure BDA0003339874850000154
3, 6-dichloro-4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (300mg, 1.36mmol) and 2, 4-dimethoxypyrimidine-5-boronic acid were dissolved in 1, 4-dioxane (10mL) and water (2mL), sodium carbonate (359.6mg, 3.39mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium (99.3mg, 135.7. mu. mol) were added under nitrogen protection, and the mixture was heated to 70 ℃ for reaction for 2 hours. The reaction mixture was diluted with water (50mL) and extracted with ethyl acetate (50 mL. times.2), the organic layers were combined, the organic phase was washed with saturated brine (50mL), dried over sodium sulfate, and concentrated to give the crude product. The crude product was isolated by column on silica gel (petroleum ether: ethyl acetate (V/V) ═ 5:1-1:1, gradient elution) to give 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (B) as a yellow solid (150mg, 34% yield).
1H NMR(400MHz,CDCl3)δ9.06(s,1H),7.55(s,1H),4.45-4.58(m,2H),4.10(s,3H),4.08(s,3H),2.24-2.29(m,1H),1.65-1.69(m,1H),1.31-1.35(m,1H),1.19-1.23(m,1H)。
LC-MS,M/Z(ESI):324.9[M+H]+
Example 1: preparation of target Compound 1
5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (prop-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 1)
Figure BDA0003339874850000161
The synthetic route for the target compound 1 is shown below:
Figure BDA0003339874850000162
the first step is as follows: synthesis of 4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (prop-1-yn-1-yl) pyridazine (1B)
Figure BDA0003339874850000163
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (500mg, 1.46mmol), tributyl (prop-1-yn-1-yl) stannane (960.3mg, 2.92mmol), 1, 1-bis (diphenylphosphino) ferrocene palladium chloride (102.4mg, 0.146mmol) were dissolved in 1, 4-dioxane (10mL) under nitrogen, and then reacted at 80 ℃ for 3 hours under nitrogen. After completion of the reaction, the reaction mixture was concentrated to give a crude product. Separation and purification on silica gel column (petroleum ether: ethyl acetate (V/V) ═ 5:1-1:1, gradient elution) gave 4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (prop-1-yn-1-yl) pyridazine (1B) as a yellow oil (600mg, 98% yield).
LC-MS,M/Z(ESI):347.0[M+H]+
The second step is that: 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (prop-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (1)
Figure BDA0003339874850000171
4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (prop-1-yn-1-yl) pyridazine (600mg, 1.73mmol) was dissolved in aqueous hydrochloric acid (1M, 10mL) and reacted at 70 ℃ for 10 hours. The reaction was concentrated and then separated by reverse phase high performance liquid chromatography (column: Waters Xbridge150 × 25mM × 5 μm; mobile phase: a ═ water + ammonium bicarbonate (10mM), B ═ acetonitrile; gradient: 12% -48% B, 10 min) to give 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (prop-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (1) (80mg, yield 14.5%) as a yellow solid.
1H NMR(400MHz,CD3OD)δ8.43(s,1H),8.01(s,1H),5.76-6.05(m,1H),2.54-2.60(m,1H),2.22(s,3H),1.86-1.88(m,1H),1.42-1.46(m,1H),1.31-1.33(m,1H).
LC-MS,M/Z(ESI):318.9[M+H]+
Example 2: preparation of target Compound 2
5- (6- (but-1-yn-1-yl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (title compound 2)
Figure BDA0003339874850000172
The synthetic route for the target compound 2 is shown below:
Figure BDA0003339874850000173
the first step is as follows: synthesis of 3- (but-1-yn-1-yl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (2B)
Figure BDA0003339874850000174
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200mg, 0.583mmol) was dissolved in N, N-dimethylformamide (5mL), cuprous iodide (11.1mg, 58.4. mu. mol), triethylamine (236.2mg,2.33mmol) and bis (triphenylphosphine) palladium dichloride (40.9mg, 58.4. mu. mol) were added under nitrogen, followed by butyl-1-yne (15psi) and allowed to warm to 80 ℃ for 2 hours. The reaction mixture was diluted with water (25mL) and extracted with ethyl acetate (25 mL. times.2), the organic layers were combined, the organic phase was washed with saturated brine (25mL), dried over sodium sulfate, and concentrated to give the crude product. Separation and purification on silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-1:1, gradient elution) gave 3- (but-1-yn-1-yl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (2B) as a yellow oil (200mg, 95%).
LC-MS,M/Z(ESI):361.1[M+H]+
The second step is that: synthesis of 5- (6- (but-1-yn-1-yl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (2)
Figure BDA0003339874850000181
3- (cyclopropylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200mg, 0.555mmol) was dissolved in aqueous hydrochloric acid (1M, 5mL) and reacted at 50 ℃ for 0.5 hour. The reaction was concentrated and then separated by reverse phase high performance liquid chromatography (column: 3 Phenomenex Luna C1875 × 30mm × 3 μm; mobile phase: a ═ water +0.05 vol% HCl (36.5%), B ═ acetonitrile; gradient: 25% -45% B, 8 min) to give 5- (6- (but-1-yn-1-yl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (2) (70.7mg, 38% yield) as a yellow solid.
1H NMR(400MHz,CD3OD)δ8.49(s,1H),8.08(s,1H),5.78-6.08(m,1H),2.59-2.65(m,3H),1.88-1.95(m,1H),1.46-1.52(m,1H),1.37-1.43(m,1H),1.32(t,3H).
LC-MS,M/Z(ESI):332.9[M+H]+
Example 3: preparation of target Compound 3
5- (6- (cyclopropylethynyl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 3)
Figure BDA0003339874850000182
The synthetic route for the target compound 3 is shown below:
Figure BDA0003339874850000183
the first step is as follows: synthesis of 3- (cyclopropylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (3B)
Figure BDA0003339874850000191
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200mg, 583.5. mu. mol) and cyclopropylacetylene were dissolved in N, N-dimethylformamide (5mL), cuprous iodide (11.1mg, 58.4. mu. mol), triethylamine (236.2mg,2.33mmol) and bis (triphenylphosphine) palladium dichloride (40.9mg, 58.4. mu. mol) were added under nitrogen protection, and the reaction was warmed to 80 ℃ for 2 hours. The reaction mixture was diluted with water (25mL) and extracted with ethyl acetate (25 mL. times.2), the organic layers were combined, the organic phase was washed with saturated brine (25mL), dried over sodium sulfate, and concentrated to give the crude product. Separation and purification on silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-1:1, gradient elution) gave 3- (cyclopropylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (3B) as a yellow oil (200mg, 92% yield).
LC-MS,M/Z(ESI):373.2[M+H]+
The second step is that: synthesis of 5- (6- (cyclopropylethynyl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (3)
Figure BDA0003339874850000192
3- (cyclopropylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200mg, 537.1. mu. mol) was dissolved in aqueous hydrochloric acid (1M, 10mL) and reacted at 75 ℃ for 2 hours. The reaction was concentrated and then separated by reverse phase high performance liquid chromatography using (column: Phenomenex Gemini-NX C1875 × 30mM × 3 μm; mobile phase: a ═ water + ammonium bicarbonate (10mM), B ═ acetonitrile; gradient: 15% -45% B, 8 min) to give 5- (6- (cyclopropylethynyl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (3) (50mg, 27% yield) as a yellow solid.
1H NMR(400MHz,CD3OD)δ8.43(s,1H),7.99(s,1H),5.78-6.07(m,1H),2.50-2.55(m,1H),1.81-1.87(m,1H),1.62-1.69(m,1H),1.41-1.46(m,1H),1.32-1.36(m,1H),1.15-1.01(m,2H),0.98-0.85(m,2H).
LC-MS,M/Z(ESI):345.2[M+H]+
Example 4: preparation of target Compound 4
5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (3-hydroxy-3-methylbut-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 4)
Figure BDA0003339874850000193
The synthetic route for the target compound 4 is shown below:
Figure BDA0003339874850000201
the first step is as follows: synthesis of 4- (4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-yl) -2-methyl-3-yn-2-ol (4B)
Figure BDA0003339874850000202
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200.0mg, 583.6. mu. mol) and 2-methylbut-3-yn-2-ol (73.6mg, 875.3. mu. mol) were dissolved in N, N-dimethylformamide (5.00mL) under a nitrogen atmosphere, and further added with cuprous iodide (11.1mg, 58.4. mu. mol), triethylamine (236.2mg,2.33mmol) and bis (triphenylphosphine) palladium (40.9mg, 58.4. mu. mol), followed by reaction at 80 ℃ for 2 hours. After completion of the reaction, the reaction mixture was added to water (10mL), extracted with ethyl acetate (20 mL. times.2), and the organic phase was washed twice with saturated brine (50mL), after which the organic phase was dried over anhydrous sodium sulfate and concentrated to give 4- (4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-yl) -2-methyl-3-yn-2-ol (4B) as a yellow oil (140.0mg, yield 68.6%).
LC-MS,M/Z(ESI):391.2[M+H]+
The second step is that: synthesis of 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (3-hydroxy-3-methylbut-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target product 4)
Figure BDA0003339874850000203
4- (4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-yl) -2-methyl-3-yn-2-ol (140.0mg, 358.6. mu. mol) was dissolved in hydrochloric acid (1M,3.59mL) and reacted at 50 ℃ for 12 hours. After completion of the reaction, the reaction was directly lyophilized to give a crude product, which was isolated by reverse phase high performance liquid chromatography using (column: 3. Phenomenex Luna C1875X 30mm X3. mu.m; mobile phase: A. water +0.05 vol% HCl (36.5%), B. acetonitrile; gradient: 16% -36% B, 7 min) to give 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (3-hydroxy-3-methylbut-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (4) (17.5mg, yield 13.2%) as a yellow solid.
1H NMR(400MHz,CDCl3)δ8.46(s,1H),8.04(s,1H),5.82-6.11(m,1H),2.57-2.62(m,1H),1.64-1.92(m,1H),1.64(s,6H),1.47-1.51(m,1H),1.35-1.37(m,1H).
LC-MS,M/Z(ESI):363.2[M+H]+
Example 5: preparation of target Compound 5
5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (3-methylbut-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 5)
Figure BDA0003339874850000211
The synthetic route for the target compound 5 is shown below:
Figure BDA0003339874850000212
the first step is as follows: synthesis of 4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (3-methylbut-1-yn-1-yl) pyridazine (5B)
Figure BDA0003339874850000213
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (253.2mg, 583.6. mu. mol) and 3-methylbut-1-yne (59.6mg, 875.3. mu. mol) were dissolved in N, N-dimethylformamide (5.00mL) under a nitrogen atmosphere, to which cuprous iodide (11.1mg, 58.4. mu. mol), triethylamine (236.2mg,2.33mmol) and bis (triphenylphosphine) palladium dichloride (40.9mg, 58.4. mu. mol) were added, followed by reaction at 80 ℃ for 2 hours. After completion of the reaction, the reaction mixture was added to water (10mL), extracted with ethyl acetate (30 mL. times.2), and the organic phase was washed twice with saturated brine (50mL), after which the organic phase was dried over anhydrous sodium sulfate and concentrated to give 4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (3-methylbut-1-yn-1-yl) pyridazine (5B) as a yellow oily compound (180.0mg, 76.6% yield).
LC-MS,M/Z(ESI):375.3[M+H]+
The second step is that: synthesis of 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (3-methylbut-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target Compound 5)
Figure BDA0003339874850000214
4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (3-methylbut-1-yn-1-yl) pyridazine (80.0mg, 213.7. mu. mol) was dissolved in hydrochloric acid (1M,2.14mL) and reacted at 40 ℃ for 12 hours. After completion of the reaction, the reaction was directly lyophilized to give 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (3-methylbut-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (5) (40.5mg, yield 50.4%) as a yellow solid.
1H NMR(400MHz,CDCl3)δ8.67(s,1H),8.23(s,1H),5.84-6.13(m,1H),2.99-3.03(m,1H),2.69-2.71(m,1H),2.03-2.05(m,1H),1.56-1.59(m,2H),1.35-1.37(m,6H)。
LC-MS,M/Z(ESI):347.2[M+H]+
Example 6: preparation of target Compound 6
5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- ((1-methyl-1H-pyrazol-4-yl) ethynyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 6)
Figure BDA0003339874850000221
The synthetic route for the target compound 6 is shown below:
Figure BDA0003339874850000222
the first step is as follows: synthesis of 4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- ((1-methyl-1H-pyrazol-4-yl) ethynyl) pyridazine (6B)
Figure BDA0003339874850000223
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200.0mg, 583.6. mu. mol) and 4-ethynyl-1-methyl-pyrazole (154.8mg,1.46mmol) were dissolved in N, N-dimethylformamide (5.00mL) under a nitrogen blanket, to which were added cuprous iodide (22.2mg, 116.7. mu. mol), triethylamine (236.2mg,2.33mmol) and bis (triphenylphosphine) palladium dichloride (81.9mg, 116.7. mu. mol), followed by reaction at 80 ℃ for 2 hours. After completion of the reaction, the reaction mixture was added to water (10mL), extracted with ethyl acetate (30mL × 2), and the organic phase was washed twice with saturated brine (50mL), after which the organic phase was dried over anhydrous sodium sulfate and concentrated to give 4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- ((1-methyl-1H-pyrazol-4-yl) ethynyl) pyridazine (6B) as a yellow oil (160.0mg, yield 66.5%).
LC-MS,M/Z(ESI):413.2[M+H]+
The second step is that: synthesis of 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- ((1-methyl-1H-pyrazol-4-yl) ethynyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target Compound 6)
Figure BDA0003339874850000224
4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- ((1-methyl-1H-pyrazol-4-yl) ethynyl) pyridazine (160.0mg, 450.9. mu. mol) was dissolved in hydrochloric acid (1M,3.88mL) and then reacted at 50 ℃ for 12 hours. After completion of the reaction, the reaction was directly lyophilized to give a crude product, which was isolated by reverse phase high performance liquid chromatography using (column: 3. Phenomenex Luna C1875X 30mm X3 μm; mobile phase: A. RTM. +0.05 vol% HCl (36.5%), B. RTM.; acetonitrile; gradient: 18% -38% B, 7 min.) to afford 5- (5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- ((1-methyl-1H-pyrazol-4-yl) ethynyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (6) (66.8mg, 43.4% yield) as a yellow solid.
1H NMR(400MHz,DMSO-d6)δ11.65(s,1H),11.56(s,1H),8.40(d,1H),8.23(s,1H),7.96(s,1H),7.82(s,1H),5.91-6.21(m,1H),3.90(s,3H),2.50-2.56(m,1H),1.89-1.93(m,1H),1.43-1.46(m,1H),1.31-1.33(m,1H).
LC-MS,M/Z(ESI):385.2[M+H]+
Example 7: preparation of target Compound 7
5- (6- (Cyclobutylethynyl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 7)
Figure BDA0003339874850000231
The synthetic route for the target compound 7 is shown below:
Figure BDA0003339874850000232
the first step is as follows: synthesis of tributyl (cyclobutylethynyl) stannane (7A)
Figure BDA0003339874850000233
N-butyllithium (2.5M,7.03mL) was added dropwise to tetrahydrofuran (5mL) at-5 ℃ and the temperature was kept below 10 ℃. 6-chloro-1-hexyne (1g,8.58mmol) was added dropwise to the system at about 5 ℃ and stirred for 2 hours. Then, tri-n-butyltin chloride (3.07g,9.43mmol) was added dropwise to the system, and the reaction was carried out for 0.5 hour. The reaction was quenched with potassium fluoride solution (50mL), then extracted with ethyl acetate (50mL × 3), the organic layers were combined, dried over sodium sulfate, and concentrated to give tributyl (cyclobutylethynyl) stannane (7A) as a yellow oil (3g, 94.7% yield).
The second step is that: synthesis of 3- (cyclobutylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (7B)
Figure BDA0003339874850000241
3-chloro-4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (200mg, 583.5. mu. mol) and tributyl (cyclobutylethynyl) stannane (7A) (323.1mg, 875.3. mu. mol) were dissolved in 1, 4-dioxane (10mL), bis (triphenylphosphine) palladium dichloride (40.9mg, 58.4. mu. mol) was added under nitrogen protection, and the temperature was raised to 80 ℃ for reaction for 3 hours. And (4) spin-drying the reaction system to obtain a crude product. Purification by silica gel column separation (petroleum ether: ethyl acetate (V/V) ═ 5:1-1:1, gradient elution) gave 3- (cyclobutylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (7B) as a yellow oil (200mg, 88.7% yield).
LC-MS,M/Z(ESI):387.2[M+H]+
The third step: synthesis of 5- (6- (cyclobutylethynyl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (7)
Figure BDA0003339874850000242
3- (Cyclobutylethynyl) -4- ((1S,2S) -2- (difluoromethyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (3) (200mg, 435.1. mu. mol) was dissolved in aqueous hydrochloric acid (1M, 4.35mL) and reacted at 50 ℃ for 12 hours. The reaction was spun dry and the crude product was separated by two reverse phase high performance liquid chromatography steps (column: 3 Phenomenex Luna C1875 x 30mm x 3 μm; mobile phase: a ═ water +0.05 vol% HCl (36.5%), B ═ acetonitrile; gradient: 29% -49% B, 7.5 min) and (column: Phenomenex Gemini-NX C1875 x 30mm x 3 μm; mobile phase: a ═ water + ammonium bicarbonate (10mmol), B ═ acetonitrile; gradient: 24% -44% B, 8 min) to give the yellow solid compound 5- (6- (cyclobutylethynyl) -5- ((1S,2S) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (7) (10.3mg, 6.55% yield).
1H NMR(400MHz,CD3OD)δ8.47(s,1H),8.02(s,1H),5.94(td,1H),3.39-3.48(m,1H),2.55-2.60(m,1H),2.40-2.48(m,2H),2.27-2.37(m,2H),1.99-2.12(m,2H),1.83-1.89(m,1H),1.42-1.47(m,1H),1.33-1.38(m,1H).
LC-MS,M/Z(ESI):359.1[M+H]+
Example 8: preparation of target Compound 8
5- (6- (cyclopropylethynyl) -5- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target compound 8)
Figure BDA0003339874850000243
The synthetic route for the target compound 8 is shown below:
Figure BDA0003339874850000251
the first step is as follows: synthesis of 3- (cyclopropylethynyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (8B)
Figure BDA0003339874850000252
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (120.0mg, 369.5. mu. mol) and ethynylcyclopropane (24.4mg, 369.5. mu. mol) were dissolved in N, N-dimethylformamide (5.00mL) under a nitrogen atmosphere, and copper iodide (7.04mg, 36.9. mu. mol), triethylamine (149.6mg,1.48mmol) and bis (triphenylphosphine) palladium dichloride (25.9mg, 36.9. mu. mol) were added thereto, followed by reaction at 80 ℃ for 2 hours. After completion of the reaction, the reaction mixture was added to water (10mL), extracted with ethyl acetate (20mL × 2), and the organic phase was washed twice with saturated brine (50mL), after which the organic phase was dried over anhydrous sodium sulfate and concentrated to give 3- (cyclopropylethynyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (8B) as a yellow oil (90.0mg, 39.2% yield).
LC-MS,M/Z(ESI):355.2[M+H]+
The second step is that: synthesis of 5- (6- (cyclopropylethynyl) -5- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target Compound 8)
Figure BDA0003339874850000253
3- (cyclopropylethynyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazine (90.0mg, 253.9. mu. mol) was dissolved in hydrochloric acid (1M,2.54mL) and reacted at 50 ℃ for 12 hours. After completion of the reaction, the reaction was directly lyophilized to give a crude product, which was isolated by reverse phase high performance liquid chromatography using (column: 3. Phenomenex Luna C1875X 30mm X3 μm; mobile phase: A. RTM. water +0.05 vol% HCl (36.5%), B. RTM. acetonitrile; gradient: 21% -41% B, 7 min) to give 5- (6- (cyclopropylethynyl) -5- ((1S,2S) -2- (fluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (8) (21.4mg, 23.9% yield) as a yellow solid.
1H NMR(400MHz,CDCl3)δ8.61(s,1H),8.17(s,1H),4.32-4.86(m,2H),2.43-2.46(m,1H),1.73-1.89(m,1H),1.70-1.72(m,1H),1.09-1.12(m,2H),0.99-1.02(m,2H),0.98-0.99(m,2H).
LC-MS,M/Z(ESI):327.3[M+H]+
Example 9: preparation of target Compound 9
5- (6- (cyclopropylethynyl) -5- ((1R,2R) -2- (difluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (title compound 9)
Figure BDA0003339874850000261
The synthetic route for the target compound 9 is shown below:
Figure BDA0003339874850000262
the first step is as follows: synthesis of 3- (2-cyclopropylethynyl) -4- [ (1R,2R) -2- (difluoromethyl) cyclopropyl ] -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (9A)
Figure BDA0003339874850000263
3-chloro-4- [ (1R,2R) -2- (difluoromethyl) cyclopropyl ] -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (250mg, 729. mu. mol) and ethynylcyclopropane (120mg,1.82mmol) were dissolved in N, N-dimethylacetamide (4.00mL), and then dichlorobis (triphenylphosphine) palladium (II) (102mg, 145. mu. mol), cuprous iodide (27.7mg, 145. mu. mol) and triethylamine (295mg,2.92mmol) were added to the reaction solution and reacted at 80 ℃ for 2 hours under nitrogen atmosphere. After completion of the reaction, the reaction mixture was poured into water (5mL), extracted with ethyl acetate (20mL × 2), washed with brine (20mL × 2), dried over anhydrous sodium sulfate, filtered, concentrated, and then separated and purified by a silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-1:1) to give the compound 3- (2-cyclopropylethynyl) -4- [ (1R,2R) -2- (difluoromethyl) cyclopropyl ] -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (9A) (260mg, crude product) as a yellow oily compound.
LC-MS,M/Z(ESI):373.2[M+H]+
The second step is that: 5- [6- (2-Cyclopropylethynyl) -5- [ (1R,2R) -2- (difluoromethyl) cyclopropyl ] pyridazin-3-yl ] -1H-pyrimidine-2, 4-dione (target compound 9)
Figure BDA0003339874850000264
3- (2-Cyclopropylethynyl) -4- [ (1R,2R) -2- (difluoromethyl) cyclopropyl ] -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine (260mg, 451. mu. mol) was dissolved in 1M aqueous hydrochloric acid (3mL) and reacted at 50 ℃ for 12 hours. After the completion of the reaction, the reaction solution was concentrated to give a product, which was isolated by high performance liquid chromatography (column: Phenomenex luna C18150 × 40mm × 15 μm; mobile phase: a ═ water + 0.05% by volume hydrochloric acid (36.5%), B ═ acetonitrile; gradient: 25% -45%, 6.5 minutes) to give 5- [6- (2-cyclopropylethynyl) -5- [ (1R,2R) -2- (difluoromethyl) cyclopropyl ] pyridazin-3-yl ] -1H-pyrimidine-2, 4-dione (9) (41.0mg, yield 17.2%) as a yellow solid.
1H NMR(400MHz,DMSO_d6):δ11.59-11.61(m,1H),11.53(s,1H),8.33-8.35(m,1H),7.88(s,1H),5.89-6.19(m,1H),2.42-2.44(m,1H),1.81-1.85(m,1H),1.69-1.70(m,1H),1.39-1.40(m,1H),1.21-1.26(m,1H),0.99-1.01(m,2H),0.87-0.89(m,2H).
LC-MS,M/Z(ESI):345.1[M+H]+
Example 10: preparation of target Compound 10
5- [6- (2-Cyclopropylethynyl) -5- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazin-3-yl ] -1H-pyrimidine-2, 4-dione (target compound 10)
Figure BDA0003339874850000271
The synthetic route for the target compound 10 is shown below:
Figure BDA0003339874850000272
the first step is as follows: synthesis of (1S,2S) -2- (ethoxycarbonyl) cyclopropanecarboxylic acid (10A)
Figure BDA0003339874850000273
Ethyl (1S,2S) -2- (hydroxymethyl) cyclopropanecarboxylate (5.0g, 34.7mmol) was dissolved in acetonitrile (50mL), and 2,2,6, 6-tetramethylpiperidine oxide (436.3mg,2.8mmol), sodium dihydrogen phosphate (6.66g,55.5mmol), and disodium hydrogen phosphate (7.88g,55.5mmol) were added in that order at 25 ℃. Then, a sodium hypochlorite solution (0.5mL) and sodium chlorite (6.27g,69.4mmol) were dissolved in 25mL of water, and slowly added dropwise to the reaction system at 0 ℃ followed by stirring at 25 ℃ for 12 hours. The reaction system was diluted with water (100mL), followed by extraction with ethyl acetate (100 mL. times.2), and the organic layers were combined, added with saturated aqueous sodium carbonate (100mL), and stirred for 10 minutes. The organic phase was separated, the aqueous phase was adjusted to pH 2-3 with 6M hydrochloric acid solution, followed by extraction with ethyl acetate (100 mL. times.2), the organic phases were combined, the organic phase was washed with saturated brine (100mL), dried over anhydrous sodium sulfate, and concentrated to give (1S,2S) -2- (ethoxycarbonyl) cyclopropanecarboxylic acid (10A) (4.8g, 87.5% yield) as a colorless oil.
1H NMR(400MHz,CDCl3)δ10.34(br.s,1H),4.14(q,2H),2.11-2.22(m,2H),1.43-1.50(m,2H),1.25(t,3H)。
The second step is that: synthesis of ethyl (1S,2S) -2- (trifluoromethyl) cyclopropanecarboxylate (10B)
Figure BDA0003339874850000281
(1S,2S) -2- (ethoxycarbonyl) cyclopropanecarboxylic acid (3.0g,19.0mmol) was charged into an autoclave, sulfur tetrafluoride (9.0g,83.3mmol) was added at-78 deg.C, and then the reaction system was heated to 70 deg.C in the autoclave for 16 hours. Dichloromethane (20mL) was added to the reaction, and the organic phase was washed with saturated aqueous sodium bicarbonate (500mL), dried over anhydrous sodium sulfate, and concentrated to give ethyl (1S,2S) -2- (trifluoromethyl) cyclopropanecarboxylate (10B) as a yellow oil (1.17g, 33.9% yield).
1H NMR(400MHz,CDCl3)δ4.17-4.19(m,2H),2.10-2.20(m,1H),2.00-2.05(m,1H),1.20-1.40(m,5H).
The third step: synthesis of (1S,2S) -2- (trifluoromethyl) cyclopropanecarboxylic acid (10C)
Figure BDA0003339874850000282
Ethyl (1S,2S) -2- (trifluoromethyl) cyclopropanecarboxylate (1.1g,6.0mmol) was dissolved in tetrahydrofuran (10mL) and water (5mL), followed by addition of lithium hydroxide monohydrate (634mg,15.1mmol) and reaction at 80 ℃ for 6 hours. After completion of the reaction, water (20mL) was added, and extraction was performed with dichloromethane (30mL × 2), and the aqueous phase was collected, adjusted to pH 3 with 6M hydrochloric acid, and then extracted with dichloromethane (30mL × 3), and the organic phases were combined, washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give (1S,2S) -2- (trifluoromethyl) cyclopropanecarboxylic acid (10C) as a brown oil (500mg, 53.7% yield).
1H NMR(400MHz,CDCl3)δ9.80(br.s,1H),2.20-2.23(m,1H),2.04-2.06(m,1H),1.27-1.44(m,2H)。
The fourth step: synthesis of 3, 6-dichloro-4- ((1S,2S) -2- (trifluoromethyl) cyclopropyl) pyridazine (10D)
Figure BDA0003339874850000283
3, 6-dichloropyridazine (450mg,3.02mmol) and (1S,2S) -2- (trifluoromethyl) cyclopropanecarboxylic acid (465mg, 3.02mmol) were dissolved in water (15mL) and concentrated sulfuric acid (0.5mL) was added and the temperature was raised to 70 ℃ under nitrogen. Then, an aqueous solution of silver nitrate (257mg,1.51mmol,1.5mL) was added rapidly, and then an aqueous solution of ammonium persulfate (2.07g,9.06mmol,5mL) was added dropwise slowly, and the reaction was continued at 70 ℃ for 1 hour. The reaction solution was adjusted to pH 9 with aqueous ammonia, followed by extraction with ethyl acetate (40 mL. times.2), and the organic layers were combined, and the organic phase was washed with saturated brine (50mL), dried over sodium sulfate, and concentrated to give a crude product. Separation was then carried out by reverse phase high performance liquid chromatography using (column: Phenomenex luna C18150 × 40mm × 15 μm; mobile phase: a ═ water +0.1 vol% TFA, B ═ acetonitrile; gradient: 35% -65% B, 10 min) to give 3, 6-dichloro-4- ((1S,2S) -2- (trifluoromethyl) cyclopropyl) pyridazine (10D) as a yellow oil (350mg, 43.8% yield).
LC-MS,M/Z(ESI):256.9[M+H]+
The fifth step: synthesis of 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (trifluoromethyl) cyclopropyl) pyridazine (10E)
Figure BDA0003339874850000291
3, 6-dichloro-4- ((1S,2S) -2- (trifluoromethyl) cyclopropyl) pyridazine (350mg,1.32mmol) and 2, 4-dimethoxypyrimidine-5-boronic acid (343mg,1.32mmol) were dissolved in dioxane (5mL) and water (1mL), sodium carbonate (420mg, 3.96mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium (97mg, 132. mu. mol) were added under nitrogen, and the mixture was heated to 50 ℃ for reaction for 12 hours. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (20 mL. times.2), the organic phases were combined, the organic phase was washed with saturated brine (50mL), dried over sodium sulfate and concentrated to give the crude product. Separation and purification on silica gel column (petroleum ether: ethyl acetate (V/V) ═ 50:1-3:1, gradient elution) gave 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2S) -2- (trifluoromethyl) cyclopropyl) pyridazine (10E) as a yellow oil (300mg, 44% yield).
LC-MS,M/Z(ESI):361.0[M+H]+
And a sixth step: synthesis of 3- (2-cyclopropylethynyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazine (10F)
Figure BDA0003339874850000292
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazine (500mg,1.39mmol) and ethynylcyclopropane (229mg,3.47mmol) were dissolved in N, N-dimethylacetamide (5.00mL), and then dichlorobis (triphenylphosphine) palladium (II) (194mg,277umol), cuprous iodide (52.8mg, 277. mu. mol) and triethylamine (561mg,5.54mmol) were added to the reaction solution and reacted at 80 ℃ for 2 hours under nitrogen. After completion of the reaction, the reaction mixture was poured into water (10mL), extracted with ethyl acetate (20mL × 2), washed with brine (20mL × 2), dried over anhydrous sodium sulfate, filtered, concentrated, and then separated and purified by a silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-1:1) to give the compound 3- (2-cyclopropylethynyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazine (10F) as a yellow oily compound (503mg, crude product).
LC-MS,M/Z(ESI):391.1[M+H]+
The seventh step: 5- [6- (2-Cyclopropylethynyl) -5- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazin-3-yl ] -1H-pyrimidine-2, 4-dione (target compound 10)
Figure BDA0003339874850000293
3- (2-Cyclopropylethynyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazine (503mg, 503. mu. mol) was dissolved in 1M aqueous sulfuric acid (5mL) and reacted at 50 ℃ for 12 hours. After the completion of the reaction, the reaction solution was concentrated to give a product, which was then separated by high performance liquid chromatography (column: Phenomenex luna C18150 × 40mm × 15 μm; mobile phase: a ═ water + 0.05% by volume hydrochloric acid (36.5%), B ═ acetonitrile; gradient: 30% -50%, 12 minutes) to give 5- [6- (2-cyclopropylethynyl) -5- [ (1S,2S) -2- (trifluoromethyl) cyclopropyl ] pyridazin-3-yl ] -1H-pyrimidine-2, 4-dione (10) (29.0mg, yield 15.2%) as a yellow solid.
1H NMR(400MHz,DMSO_d6):δ8.38(s,1H),7.96(s,1H),2.70-2.72(m,1H),2.32-2.34(m,1H),1.66-1.70(m,1H),1.49-1.51(m,2H),1.00-1.03(m,2H),0.82-0.84(m,2H).
LC-MS,M/Z(ESI):363.1[M+H]+
Example 11: preparation of target Compound 11
5- (5- ((1S,2R) -2-isopropylcyclopropyl) -6- (prop-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (target Compound 11)
Figure BDA0003339874850000301
The synthetic route for the target compound 11 is shown below:
Figure BDA0003339874850000302
the first step is as follows: synthesis of (S) -2-chloro-3-methylbutan-1-ol (11B)
Figure BDA0003339874850000303
(S) -2-chloro-3-methylbutyric acid (30.0g,0.22mmol) was dissolved in tetrahydrofuran (300mL), lithium aluminum hydride (9.17g,0.24mmol) was slowly added at 0-10 ℃ and after the addition was completed, the mixture was stirred at 25 ℃ for 1 hour, and then the temperature was raised to 50 ℃ for reaction for 1 hour. After completion of the reaction, the reaction mixture was cooled to 0-10 ℃ and water (9mL), a 15% aqueous solution of sodium hydroxide (9mL) and water (27mL) were added in this order. Then filtered through celite, the filter cake was washed with tetrahydrofuran (100 mL. times.3), and the filtrate was concentrated to give the crude product. Separation and purification by silica gel column (petroleum ether: ethyl acetate (V/V) ═ 5:1-2:1) gave (S) -2-chloro-3-methylbutan-1-ol (11B) as a yellow oil (8.4g, 31% yield).
1H NMR(400MHz,CDCl3)δ3.91-3.94(m,1H),3.74-3.82(m,2H),2.06-2.09(m,1H),1.99-2.05(m,1H),1.04(dd,6H).
The second step is that: synthesis of (R) -2-isopropyloxirane (11C)
Figure BDA0003339874850000304
Potassium hydroxide (52.1g,0.93mol) was dissolved in water (50mL), cooled to 0-5 ℃ and (S) -2-chloro-3-methylbutan-1-ol (25.0g,0.20mol) was added dropwise and reacted at 25 ℃ for 1 hour. After completion of the reaction, the reaction mixture was distilled at 25 ℃ and the fractions were collected by cooling with a dry ice ethanol bath to give (R) -2-isopropyloxirane (11C) as a yellow oil (16.0g, 91.1% yield).
1H NMR(400MHz,CDCl3)δ2.71-2.74(m,2H),2.52-2.54(m,1H),1.47-1.53(m,1H),1.04(d,3H),0.97(d,3H).
The third step: synthesis of ethyl (1S,2R) -2-isopropylcyclopropane-1-carboxylate (11D)
Figure BDA0003339874850000311
Triethyl phosphonoacetate (14.3g,63.8mmol) was dissolved in 1, 4-dioxane (20mL), n-butyllithium (2.5M,30.2mL) was added dropwise at 0 deg.C, and after completion of the addition, the reaction was stirred at 25 deg.C for 0.5 h, and then the reaction was transferred to a stuffer tank, followed by addition of a solution of (R) -2-isopropyloxirane (5.00g, 58.1mmol) in 1, 4-dioxane (10 mL). The closed tank is screwed down, and the temperature is raised to 145 ℃ for reaction for 12 hours. After completion of the reaction, the reaction system was cooled, followed by addition of water (100mL), extraction with methyl t-butyl ether (100 mL. times.2), drying of the organic phase over anhydrous sodium sulfate, filtration and concentration to give ethyl (1S,2R) -2-isopropylcyclopropane-1-carboxylate (11D) (7.0g, yield 80.8%) as a yellow oil.
1H NMR(400MHz,CDCl3)δ4.09-4.13(m,2H),1.36-1.39(m,1H),1.19-1.27(m,5H),1.09-1.13(m,1H),0.96-0.99(m,6H),0.69-0.75(m,1H).
The fourth step: synthesis of (1S,2R) -2-isopropylcyclopropane-1-carboxylic acid (11E)
Figure BDA0003339874850000312
Ethyl (1S,2R) -2-isopropylcyclopropane-1-carboxylate (7.00g,44.8mmol) was dissolved in 1, 4-dioxane (60mL) and water (60mL), followed by addition of sodium hydroxide (17.9g,448.1mmol) and reaction at 100 ℃ for 7 hours. After completion of the reaction, extraction was performed with methyl t-butyl ether (100mL × 2), the aqueous phase was collected, the pH of the aqueous phase was adjusted to 1-2 with concentrated hydrochloric acid, followed by extraction with methyl t-butyl ether (100mL × 2), the organic phases were combined, washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give (1S,2R) -2-isopropylcyclopropane-1-carboxylic acid (11E) (6.40g, crude product) as a yellow oil to be used in the next step as it is.
1H NMR(400MHz,CDCl3)δ1.56-1.60(m,1H),1.45-1.50(m,1H),1.37-1.40(m,1H),1.21-1.27(m,1H),1.60-1.85(m,6H),0.97-1.02(m,1H).
The fifth step: synthesis of 3, 6-dichloro-4- ((1S,2R) -2-isopropylcyclopropyl) pyridazine (11F)
Figure BDA0003339874850000313
3, 6-dichloropyridazine (6.27g,42.1mmol) and (1S,2S) -2- (difluoromethyl) cyclopropane-1-carboxylic acid (5.39g,42.1mmol) were dissolved in water (100mL), concentrated sulfuric acid (5.39mL) was added, and the temperature was raised to 70 ℃ under nitrogen. Then, an aqueous solution of silver nitrate (3.57g,21.0mmol,25mL) was added rapidly, and then an aqueous solution of ammonium persulfate (28.8g,126.2mmol,50mL) was added slowly dropwise, and the reaction was continued at 70 ℃ for 1 hour. After completion of the reaction, the reaction solution was adjusted to pH 9 with aqueous ammonia, followed by extraction with ethyl acetate (200 mL. times.2), and the organic layers were combined, washed with saturated brine (100mL), dried over anhydrous sodium sulfate, and concentrated to give a crude product. Separation and purification by silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-3:1) gave 3, 6-dichloro-4- ((1S,2R) -2-isopropylcyclopropyl) pyridazine (11F) as a yellow oil (3.20g, yield 32.9%).
LC-MS,M/Z(ESI):231.0[M+H]+
And a sixth step: synthesis of 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2R) -2-isopropylcyclopropyl) pyridazine (11G)
Figure BDA0003339874850000321
3, 6-dichloro-4- ((1S,2R) -2-isopropylcyclopropyl) pyridazine (2.10g,9.09mmol) and 2, 4-dimethoxypyrimidine-5-boronic acid (1.67g,9.09mmol) were dissolved in 1, 4-dioxane (10mL) and water (3mL), sodium carbonate (2.89g,27.3mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium (664.8mg,0.91mmol) were added under nitrogen, and the mixture was heated to 100 ℃ for reaction for 2 hours. The reaction mixture was diluted with water (100mL), followed by extraction with ethyl acetate (150 mL. times.2), the organic phases were combined, the organic phase was washed with saturated brine (100mL), dried over anhydrous sodium sulfate, and concentrated to give the crude product. Separation and purification by silica gel column (petroleum ether: ethyl acetate (V/V) ═ 10:1-2:1) gave 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2R) -2-isopropylcyclopropyl) pyridazine (11G) as a yellow oil (2.00G, yield 65.7%).
LC-MS,M/Z(ESI):335.1[M+H]+
The seventh step: synthesis of 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2R) -2-isopropylcyclopropyl) -3- (prop-1-yn-1-yl) pyridazine (11H)
Figure BDA0003339874850000322
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- [ (1S,2R) -2-isopropylcyclopropyl ] pyridazine (1.10g,3.29mmol), dichlorobis (triphenylphosphine) palladium (II) (230mg, 328. mu. mol) and tributyl (prop-1-ynyl) stannane (1.30g,3.94mmol) were dissolved in N, N-dimethylformamide (10mL) and then reacted at 110 ℃ for 2 hours under nitrogen. After completion of the reaction, the reaction mixture was poured into water (20mL), followed by extraction with ethyl acetate (50mL × 2), and the organic phase was washed with brine (50mL), dried over sodium sulfate, and concentrated and purified by silica gel column separation (petroleum ether: ethyl acetate (V/V) ═ 10:1-5:1) to obtain 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S,2R) -2-isopropylcyclopropyl) -3- (prop-1-yn-1-yl) pyridazine (11H) as a yellow oily compound (1.00g, yield 89.1%).
LC-MS,M/Z(ESI):339.1[M+H]+
Eighth step: synthesis of 5- (5- ((1S,2R) -2-isopropylcyclopropyl) -6- (prop-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (11)
Figure BDA0003339874850000323
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S,2R) -2-isopropylcyclopropyl) -3- (prop-1-yn-1-yl) pyridazine (1.00g,2.67mmol) was dissolved in hydrochloric acid (1M,5.00mL) and reacted at 50 ℃ for 12 hours. After the completion of the reaction, the reaction solution was concentrated and then separated by reverse-phase high performance liquid chromatography (column: Phenomenex luna C18150 × 40mm × 15 μm; mobile phase: a ═ water + 0.225% by volume of formic acid (99%), B ═ acetonitrile; gradient: 28% -58%, 10 minutes) to give 5- (5- ((1S,2R) -2-isopropylcyclopropyl) -6- (prop-1-yn-1-yl) pyridazin-3-yl) pyrimidine-2, 4(1H,3H) -dione (11) (295mg, yield 36.7%) as a yellow solid.
1H NMR(400MHz,CD3OD):δ8.40(s,1H),7.84(s,1H),2.22(s,3H),2.14-2.17(m,1H),1.19-1.21(m,1H),1.13-1.17(m,1H),1.09-1.11(m,1H),1.08(s,3H),1.06(s,3H),1.04-1.05(m,1H).
LC-MS,M/Z(ESI):311.1[M+H]+
Test example 1: in vitro inhibitory activity of compound on recombinant human CD73 enzyme
The assay was performed in the presence of 25mM Tris (Biosharp; 77-86-1), 25mM MgCl2Tris-MgCl of (Nanjing chemical reagents Ltd.; 7791-18-6)2In buffer. With Tris-MgCl2buffer A3 Xstock solution of Human-CD73 (Novoprotein; C446) was prepared and added to a 96-well white plate at 20. mu.L/well to a final concentration of 0.1. mu.g/mL; with Tris-MgCl2Diluting the final concentration of the compound to be 3 multiplied mother liquor with a proper concentration gradient by buffer, adding the compound into the 96-well test white board according to 20 mu L/well, uniformly mixing, incubating at normal temperature for 30min, and setting a positive control group (without adding the compound) and a negative control group (without adding CD 73); with Tris-MgCl2buffer prepared AMP (Sigma; A1752-5G) as 3 Xmother liquor, added to the above 96-well white plate at 20. mu.L/well to a final concentration of 100. mu.M, mixed well, and incubated at 37 ℃ for 60 min; Tris-MgCl for ATP2buffer prepared ATP (Sigma; A7699-1G) as 7 Xmother liquor, added to the above 96-well white plate at a final concentration of 100. mu.M per well, mixed well, incubated for 5min, and detected with ATP-GLO kit (Promega; G7573).
The inhibition rate of the compound on Human-CD73 is calculated according to the following formula, and then the IC of the compound on the inhibition of Human-CD73 is calculated by Prism software by taking the concentration of the compound as an X axis and the inhibition rate as a Y axis50The value:
Figure BDA0003339874850000331
TABLE 1 test Compounds for in vitro inhibition Activity on Human-CD73 enzyme
Test compounds IC50(nM)
Control Compound 2 27.03
1 21.61
2 9.118
3 4.638
4 83.94
5 196.2
6 34.80
7 33.74
8 60.03
9 247.6
10 11.19
11 14.71
In vitro enzyme test results show that the compound of the invention has good inhibition effect on CD73 enzyme, and compared with a control compound, part of the compounds of the invention show more excellent inhibition effect on CD73 enzyme.
Test example 2: pharmacokinetic testing
Mouse pharmacokinetic experiments using male ICR mice, 20-25g, fasted overnight. 3 mice were taken and orally administered by gavage (10 mg/kg). Blood was collected before dosing, and at 15, 30 minutes and 1,2, 4, 8, 24 hours post-dosing; 3 mice were also administered by intravenous injection (3mg/kg), and blood was collected before administration, 15 and 30 minutes and 1,2, 4, 8 and 24 hours after administration. Blood samples, 6800g, were centrifuged at 2-8 ℃ for 6 minutes, plasma was collected and stored at-80 ℃. And (3) adding 3-5 times of acetonitrile solution containing an internal standard into the plasma at each time point, mixing, carrying out vortex mixing for 1 minute, centrifuging at 4 ℃ for 10 minutes at 13000 rpm, taking supernatant, adding 3 times of water, mixing, and taking a proper amount of mixed solution to carry out LC-MS/MS analysis. The major pharmacokinetic parameters were analyzed using the WinNonlin 7.0 software non-compartmental model.
Rat pharmacokinetic experiments were performed using male SD rats, 250-280g, fasted overnight. 3 rats were orally administered by gavage (10 mg/kg). Blood was collected before dosing, and at 15, 30 minutes and 1,2, 4, 8, 24 hours post-dosing; 3 rats were also taken and administered intravenously (3mg/kg), and blood was collected before administration, 15 and 30 minutes and 1,2, 4, 8 and 24 hours after administration. Blood samples, 6800g, were centrifuged at 2-8 ℃ for 6 minutes, plasma was collected and stored at-80 ℃. And (3) adding 3-5 times of acetonitrile solution containing an internal standard into the plasma at each time point, mixing, carrying out vortex mixing for 1 minute, centrifuging at 4 ℃ for 10 minutes at 13000 rpm, taking supernatant, adding 3 times of water, mixing, and taking a proper amount of mixed solution to carry out LC-MS/MS analysis. The major pharmacokinetic parameters were analyzed using the WinNonlin 7.0 software non-compartmental model.
TABLE 2 mouse pharmacokinetic test results
Figure BDA0003339874850000341
Note: -indicating not tested
The result of a mouse pharmacokinetic test shows that the compound shows excellent pharmacokinetic property, and compared with a control compound, the compound has larger exposure and good pharmacy.
TABLE 3 rat pharmacokinetic test results
Figure BDA0003339874850000342
Note: -indicating not tested
The result of rat pharmacokinetic experiment shows that the compound of the invention shows excellent pharmacokinetic property, and compared with a control compound, the compound of the invention has larger exposure and good drug forming property.

Claims (16)

1. A compound of formula I, tautomers, stereoisomers, hydrates, solvates, pharmaceutically acceptable salts, or prodrugs thereof:
Figure FDA0003339874840000011
wherein the content of the first and second substances,
m is 0,1, 2,3 or 4;
Figure FDA0003339874840000012
in R1Independently selected from hydrogen, halogen, hydroxy, cyano, amino, unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-; the quilt RaSubstituted C1-C6Alkyl, or said RaSubstituted C1-C6In alkyl-O-, said is substituted by RaThe substitution can be one or more, and R isaEach independently is the following substituent: halogen, hydroxy, cyano, amino、C1-C6Alkyl radical, C1-C6alkyl-O-, -COOH, -C (═ O) NH2(ii) a When the number of the substituents is plural, the substituents may be the same or different; when m is not 0 or 1, R1Independently are the same or different;
n is 0,1, 2 or 3;
R2selected from hydrogen, unsubstituted or substituted by RbSubstituted C1-C6Alkyl, unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl, unsubstituted or substituted by RbSubstituted 5-8 membered aryl, unsubstituted or substituted by RbSubstituted 5-8 membered heteroaryl, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, or, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkenyl; the quilt RbSubstituted C1-C6Alkyl, said by RbSubstituted C3-C6Cycloalkyl radicals, said being RbSubstituted 5-8 membered aryl, said substituted RbSubstituted 5-8 membered heteroaryl, said substituted by RbSubstituted 4-8 membered heterocycloalkyl, or said substituted RbIn the substituted 4-8 membered heterocycloalkenyl group, the group represented by RbThe substitution can be one or more, and R isbEach independently is the following substituent: halogen, hydroxy, cyano, amino, carboxy, C3-C6Cycloalkyl radical, C1-C6Alkyl, C substituted by 1-5 identical or different halogens1-C6Alkyl, or, C1-C6alkyl-O-; when the number of the substituents is plural, the substituents may be the same or different;
said unsubstituted or substituted by RbIn the substituted 5-8 membered heteroaryl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocyclic alkyl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocycloalkenyl, the heteroatoms are selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3.
2. A compound of formula I, tautomers, stereoisomers, hydrates, solvates, pharmaceutically acceptable salts, or prodrugs thereof:
Figure FDA0003339874840000021
wherein the content of the first and second substances,
m is 0,1, 2,3 or 4;
Figure FDA0003339874840000022
in R1Independently selected from hydrogen, halogen, hydroxy, cyano, amino, unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-; the quilt RaSubstituted C1-C6Alkyl, or said RaSubstituted C1-C6In alkyl-O-, the substituents each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C1-C6Alkyl radical, C1-C6alkyl-O-, -COOH, -C (═ O) NH2(ii) a When the number of the substituents is plural, the substituents may be the same or different; when m is not 0 or 1, R1Independently are the same or different;
n is 0,1, 2 or 3;
R2selected from hydrogen, unsubstituted or substituted by RbSubstituted C1-C6Alkyl, unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl, unsubstituted or substituted by RbSubstituted 5-8 membered aryl, unsubstituted or substituted by RbSubstituted 5-8 membered heteroaryl, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, or unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkenyl; the quilt RbSubstituted C1-C6Alkyl, said by RbSubstituted C3-C6Cycloalkyl radicals, said being RbSubstitutionThe 5-to 8-membered aryl of (A), the said group consisting ofbSubstituted 5-8 membered heteroaryl, said substituted by RbSubstituted 4-8 membered heterocycloalkyl, or said substituted RbIn substituted 4-8 membered heterocycloalkenyl, the substitutions each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C3-C6Cycloalkyl radical, C1-C6Alkyl, C substituted by 1-5 identical or different halogens1-C6Alkyl, or, C1-C6alkyl-O-; when the number of the substituents is plural, the substituents may be the same or different;
said unsubstituted or substituted by RbIn the substituted 5-8 membered heteroaryl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocyclic alkyl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocycloalkenyl, the heteroatoms are selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3.
3. The compound of formula I, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt, or prodrug of claim 1 or 2, characterized in that,
when R is2Is unsubstituted or substituted by RbSubstituted C1-C6When alkyl, said C1-C6Alkyl is C1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl or isobutyl;
and/or when R2Is unsubstituted or substituted by RbSubstituted C1-C6When it is alkyl, the substituent RbThe number of (2) is 1-3, preferably 1;
and/or when R2Is unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl radical, said C3-C6Cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, preferably cyclopropyl orA cyclobutyl group;
and/or when R2Is unsubstituted or substituted by RbSubstituted 5-8 membered aryl, said 5-8 membered aryl is independently phenyl or naphthyl, preferably phenyl;
and/or when R2Is unsubstituted or substituted by RbWhen substituted 5-8 membered heteroaryl, said 5-8 membered heteroaryl is independently pyrrole, pyrazole, triazole, furan, oxazole, thiophene, thiazole, pyridine, pyrazine or pyrimidine, preferably pyrazole, furan, thiophene, pyridine;
and/or when R2Is unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, said 4-8 membered heterocycloalkyl is independently azetidine, oxetane, tetrahydropyrrolyl, tetrahydrofuranyl, hexahydropyran or tetrahydro-2H-thiopyran 1, 1-dioxide, preferably azetidine or oxetane;
and/or when R2Is unsubstituted or substituted by RbWhen substituted 4-8 membered heterocycloalkenyl, said 4-8 membered heterocycloalkenyl is independently dihydropyridinyl, tetrahydropyridinyl, tetrahydropyrimidinyl, pyrrolinyl, imidazolinyl, pyrazolinyl, dihydroimidazolyl, dihydropyrazolyl, dihydrooxazolyl, dihydrooxadiazolyl, dihydrothiazolyl, dihydroisothiazolyl, dihydrothienyl, dihydropyrrolyl, 3, 4-dihydro-2H-pyranyl, dihydrofuranyl, dihydropyrazinyl, dihydropyrimidyl, or fluorodihydrofuranyl, preferably 1,2,3, 4-tetrahydropyridinyl, 1, 2-dihydropyridinyl, 1, 4-dihydropyridinyl, 1,2,3, 6-tetrahydropyridinyl, 3, 4-dihydro-2H-pyranyl, or dihydrofuranyl;
and/or, RbIs a hydroxyl group;
and/or when RbIs C1-C6When alkyl, said C1-C6Alkyl is C1-C4Alkyl, preferably methyl, ethyl, n-propyl or isopropyl;
and/or when RbWhen the halogen is F, Cl, Br, I, preferably F or Cl.
4. The compound of formula I, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug according to claim 1 or 2,
the above-mentioned
Figure FDA0003339874840000031
Is composed of
Figure FDA0003339874840000032
Or
Figure FDA0003339874840000033
Preferably is
Figure FDA0003339874840000034
And/or when R1When halogen is used, the halogen is F, Cl, Br or I, preferably F or Cl;
and/or when R1When it is halogen, m is 0,1 or 2;
and/or when R1Is unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-said C1-C6Alkyl is independently C1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl or isobutyl;
and/or when R1Is unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6When alkyl-O-, m is 1 or 2, preferably m is 1;
and/or when R1Is as a quilt RaSubstituted C1-C6Alkyl, or, by RaSubstituted C1-C6alkyl-O-, the number of said substitutions is independently 1 to 3, preferably 2;
and/or when R1Is as a quilt RaSubstituted C1-C6Alkyl radicalOr, by RaSubstituted C1-C6When alkyl-O-, the substituents are each independently C1-C6Alkyl, or C1-C6alkyl-O-, C as described for said substitution1-C6Alkyl is independently C1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl or isobutyl;
and/or when RaWhen halogen is used, the halogen is F, Cl, Br or I, preferably F or Cl.
5. The compound as shown in the formula I, the tautomer, the stereoisomer, the hydrate, the solvate, the pharmaceutically acceptable salt or the prodrug thereof according to the claim 1 or 2,
when in use
Figure FDA0003339874840000041
Is composed of
Figure FDA0003339874840000042
When is in use, the
Figure FDA0003339874840000043
Is composed of
Figure FDA0003339874840000044
Figure FDA0003339874840000045
And/or when
Figure FDA0003339874840000046
Is composed of
Figure FDA0003339874840000047
When is in use, the
Figure FDA0003339874840000048
Is composed of
Figure FDA0003339874840000049
And/or when
Figure FDA00033398748400000410
Is composed of
Figure FDA00033398748400000411
When is in use, the
Figure FDA00033398748400000412
Is composed of
Figure FDA00033398748400000413
And/or when
Figure FDA00033398748400000414
Is composed of
Figure FDA00033398748400000415
When is in use, the
Figure FDA00033398748400000416
Is composed of
Figure FDA00033398748400000417
6. The compound as shown in the formula I, the tautomer, the stereoisomer, the hydrate, the solvate, the pharmaceutically acceptable salt or the prodrug thereof according to the claim 1 or 2,
Figure FDA00033398748400000418
is composed of
Figure FDA00033398748400000419
Or
Figure FDA00033398748400000420
And/or, R2Is composed of
Figure FDA0003339874840000051
Or
Figure FDA0003339874840000052
7. The compound as shown in the formula I, the tautomer, the stereoisomer, the hydrate, the solvate, the pharmaceutically acceptable salt or the prodrug thereof according to the claim 1 or 2,
Figure FDA0003339874840000053
is composed of
Figure FDA0003339874840000054
And/or, R2Is composed of
Figure FDA0003339874840000055
Or
Figure FDA0003339874840000056
8. The compound of formula I, its tautomers, its stereoisomers, its hydrates, its solvates, its pharmaceutically acceptable salts or its prodrugs according to claim 1 or 2, characterized in that it is
Figure FDA0003339874840000057
Wherein the content of the first and second substances,
R1independently selected from hydrogen, halogen, hydroxy, cyano, amino, unsubstituted or substituted by RaSubstituted C1-C6Alkyl, or, unsubstituted or substituted by RaSubstituted C1-C6alkyl-O-; the quilt RaSubstituted C1-C6Alkyl, or said is RaSubstituted C1-C6In alkyl-O-, the substituents each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C1-C6Alkyl radical, C1-C6alkyl-O-, -COOH, -C (═ O) NH2(ii) a When the number of the substituents is plural, the substituents may be the same or different;
R2selected from hydrogen, unsubstituted or substituted by RbSubstituted C1-C6Alkyl, unsubstituted or substituted by RbSubstituted C3-C6Cycloalkyl, unsubstituted or substituted by RbSubstituted 5-8 membered aryl, unsubstituted or substituted by RbSubstituted 5-8 membered heteroaryl, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkyl, or, unsubstituted or substituted by RbSubstituted 4-8 membered heterocycloalkenyl; the quilt RbSubstituted C1-C6Alkyl, said by RbSubstituted C3-C6Cycloalkyl radicals, said being RbSubstituted 5-8 membered aryl, said substituted RbSubstituted 5-8 membered heteroaryl, said substituted by RbSubstituted 4-8 membered heterocycloalkyl, or said substituted RbIn substituted 4-8 membered heterocycloalkenyl, the substitutions each independently refer to one or more of the following substituents: halogen, hydroxy, cyano, amino, C3-C6Cycloalkyl, C1-C6Alkyl, C substituted by 1-5 identical or different halogens1-C6Alkyl, or, C1-C6alkyl-O-; when the number of the substituents is plural, the substituents may be the same or different;
said unsubstituted or substituted by RbIn the substituted 5-8 membered heteroaryl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3(ii) a Said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocyclic alkyl, the heteroatom is selected from one or more of N, S, O and P, and the number of the heteroatom is 1-3; said unsubstituted or substituted by RbIn the substituted 4-8 membered heterocycloalkenyl, the heteroatoms are selected from one or more of N, S, O and P, and the number of the heteroatoms is 1-3.
9. The compound of formula I, its hydrate, solvate, pharmaceutically acceptable salt or prodrug according to claim 1 or 2, characterized in that it is
Figure FDA0003339874840000061
Wherein R is1And R2Having the definition as set forth in claim 1.
10. The compound of formula I, a hydrate, a solvate, a pharmaceutically acceptable salt or a prodrug thereof according to claim 9, wherein R is1Selected from difluoromethyl, trifluoromethyl, dichloromethyl, trichloromethyl or isopropyl;
R2selected from methyl, ethyl or cyclopropyl.
11. The compound, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug according to claim 1 or 2, wherein the compound represented by formula I is selected from any one of the following compounds:
Figure FDA0003339874840000062
12. a pharmaceutical composition, comprising a compound of formula I, a tautomer, a stereoisomer, a hydrate, a solvate, a pharmaceutically acceptable salt or a prodrug thereof according to any one of claims 1 to 11, and a pharmaceutically acceptable excipient.
13. Use of a compound according to any one of claims 1 to 11, a tautomer, a stereoisomer, a hydrate, a solvate, a pharmaceutically acceptable salt or a prodrug thereof, a pharmaceutical composition according to claim 12 in combination with a PD-1 antibody, a PD-L1 antibody, a CTLA-4 antibody or a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor for the manufacture of a medicament for the treatment of a CD 73-associated disease.
14. Use of a compound according to any one of claims 1-11, a tautomer, a stereoisomer, a hydrate, a solvate, a pharmaceutically acceptable salt, or a prodrug thereof, or a pharmaceutical composition of claim 12, for the manufacture of a medicament for the treatment of a CD 73-related disease.
15. The use according to claim 13 or 14, wherein the CD 73-related disease is cancer.
16. The use according to claim 15, wherein the cancer is bladder cancer, breast cancer, bile duct cancer, rectal cancer, colon cancer, stomach cancer, gallbladder cancer, glioblastoma, head and neck cancer, liver cancer, lung cancer, lymphoma, medulloblastoma, melanoma, ovarian cancer, pancreatic cancer, prostate cancer or renal cancer.
CN202111305068.7A 2020-11-05 2021-11-05 Pyridazine alkyne compound and application thereof Pending CN114437038A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011224592 2020-11-05
CN2020112245927 2020-11-05

Publications (1)

Publication Number Publication Date
CN114437038A true CN114437038A (en) 2022-05-06

Family

ID=81362818

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111305068.7A Pending CN114437038A (en) 2020-11-05 2021-11-05 Pyridazine alkyne compound and application thereof

Country Status (2)

Country Link
CN (1) CN114437038A (en)
WO (1) WO2022095953A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2528298A (en) * 2014-07-16 2016-01-20 Redx Pharma Plc Compounds
TW202227095A (en) * 2016-01-08 2022-07-16 美商阿克思生物科學有限公司 Modulators of 5'-nucleotidase, ecto and the use thereof
TWI702954B (en) * 2018-03-01 2020-09-01 美商美國禮來大藥廠 Cd73 inhibitors

Also Published As

Publication number Publication date
WO2022095953A1 (en) 2022-05-12

Similar Documents

Publication Publication Date Title
CN111484477B (en) Benzopyridone heterocyclic compound and application thereof
CN111647000B (en) Pyrazine derivative and application thereof in inhibition of SHP2
CN112105610A (en) Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
EP3848367A1 (en) Tricyclic compounds acting on crbn proteins
KR102590310B1 (en) KEAP1-Nrf2 protein interaction inhibitor
CN114315839A (en) Pyrimidinedione compounds and use thereof
WO2022095975A1 (en) Cd73 inhibitor and use thereof
CN114456159A (en) Nitrogen-substituted heterocyclic thiophene compound and application thereof
KR20200142508A (en) Aminopyrazine diol compounds as PI3K-γ inhibitors
CN114456157A (en) Nitrogen-substituted amino carbonate thiophene compound and application thereof
CN114456147A (en) Oxygen substituted amino carbonate thiophene compounds
AU2021285467A1 (en) N-phenylaminocarbonyl pyridino-, pyrimidino and benzo-tropanes as modulators of GPR65
US20230142274A1 (en) Substituted aryl compound
WO2021078274A1 (en) Pyridazinone or pyridazine compound and derivative and pharmaceutical composition thereof
JP2023522725A (en) 3-Azabicycloalkyl derivative and pharmaceutical composition containing the same
JP7420403B2 (en) Compounds used as kinase inhibitors and their applications
WO2022037630A1 (en) Tetracyclic derivative, method for preparing same and use thereof in medicine
CN114437038A (en) Pyridazine alkyne compound and application thereof
WO2023143147A1 (en) Pyridazopyridone compounds, pharmaceutical composition thereof and use thereof
CN113173924A (en) Pyridine acetamide derivative as CDK inhibitor, and preparation method and application thereof
TWI823420B (en) Compounds useful as CDK kinase inhibitors and uses thereof
CN112209933B (en) BTK inhibitors containing 4-azacycloheptane
CN111718351B (en) Oxygen-containing substituted pyrazolopyrimidine compound, pharmaceutical composition and application thereof
WO2023116763A1 (en) Pyridazine compound, and pharmaceutical composition and use thereof
KR20230123004A (en) Pharmaceutical composition for treating cancer comprising novel compounds for inhibiting prostaglandin e2 receptor and anticancer drug

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