CN109897011B - IDO inhibitor and application thereof - Google Patents

IDO inhibitor and application thereof Download PDF

Info

Publication number
CN109897011B
CN109897011B CN201811493232.XA CN201811493232A CN109897011B CN 109897011 B CN109897011 B CN 109897011B CN 201811493232 A CN201811493232 A CN 201811493232A CN 109897011 B CN109897011 B CN 109897011B
Authority
CN
China
Prior art keywords
mmol
compound
amino
fluorophenyl
added
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.)
Active
Application number
CN201811493232.XA
Other languages
Chinese (zh)
Other versions
CN109897011A (en
Inventor
许忻
张瑱
李云飞
王贯
朱伟波
李东升
秦成刚
刘传夺
刘磊
武琦梅
杨旭芹
贾婕
王影
陈宇浩
王艺瑾
葛建
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.)
Zhejiang Huahai Pharmaceutical Co Ltd
Shanghai Synergy Pharmaceutical Sciences Co Ltd
Original Assignee
Zhejiang Huahai Pharmaceutical Co Ltd
Shanghai Synergy Pharmaceutical Sciences Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zhejiang Huahai Pharmaceutical Co Ltd, Shanghai Synergy Pharmaceutical Sciences Co Ltd filed Critical Zhejiang Huahai Pharmaceutical Co Ltd
Publication of CN109897011A publication Critical patent/CN109897011A/en
Application granted granted Critical
Publication of CN109897011B publication Critical patent/CN109897011B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Embodiments of the present invention provide compounds of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein the substituents R 1 、R 2 、R 0 The definition of (2) is as described in the specification; the synthesized small-molecule IDO inhibitor has remarkable drug effect and high safety, and can be hopefully used as a novel antitumor drug.

Description

IDO inhibitor and application thereof
Technical Field
The invention belongs to the field of medicinal chemistry, and relates to novel compounds of IDO inhibitors, a preparation method thereof and application of the novel compounds in the aspect of preventing and treating IDO related diseases.
Background
Tumor immunotherapy has recently been paid attention to, is a focus of the field of tumor therapy, is evaluated as the most important scientific breakthrough in the year by the journal of science in 2013 due to its excellent therapeutic effect and innovation, has now demonstrated strong antitumor activity in the treatment of some tumor types such as melanoma, non-small cell lung cancer and the like, and has been approved by the FDA in the united states for clinical use as a tumor immunotherapeutic drug (macromolecular antibody). Tumor immunotherapy is expected to become a innovation in the field of tumor treatment after surgery, chemotherapy and radiotherapy.
There are many immunosuppressive molecules in tumor microenvironment, and immunotherapeutic strategies that improve tumor immune microenvironment by modulating the functions of these inhibitory molecules are important strategies for current research of small molecule drugs for tumor immunotherapy. L-tryptophan (L-Trp) is an essential amino acid required for biosynthesis of protein, nicotinic acid and neurotransmitter 5-hydroxytryptamine (serotonin), indoleamine-2,3-dioxygenase 1 (also known as IDO 1), indoleamine-2,3-dioxygenase 2 (indo 2,3-dioxygenase 2, IDO 2) and tryptophan-2,3-dioxygenase (TDO) are key rate limiting enzymes in the kynurenine (kynurenine) pathway, the predominant metabolic pathway of L-tryptophan, and catalysis occurs in the process of L-tryptophan to N-formyl-L-kynurenine. IDO1 is widely distributed and is commonly expressed in placenta, large and small intestine, colon, spleen, kidney, lung, liver, stomach and brain (King, n.j.c.; thomas, s.r.molecular in focus: indoleamine 2,3-dioxygenase, int.j.biochem.cell biol.,2007,39,2167-2172), and IDO1 is also induced to express (Yuasa, h.j.; takubo, m.; takahashi, a.; hasegawa, t.; noma, h.; zuki, t. of vertebrate Indoleamine, 3-dioxygenase, 35. 714-714) by a number of cytokines (e.g., interferon α, β, γ, interleukin IL-1, IL-2, tumor necrosis factor TNF, etc.) in addition to many cells such as myeloid cells (dendritic cells, monocytes, macrophages, etc.) and tumor cells; IDO2 is mainly expressed in the kidney and tissues such as epididymis, testis, liver, ovary, uterus, placenta, etc., although IDO1 and IDO2 are widely distributed, TDO is mainly expressed in the liver in different cell types, responsible for regulating tryptophan content of the peripheral system, and TDO is also distributed in the brain (Kanai, m.; funakoshi, h.; takahashi, h.; hayakawa, t.; mizuno, s.; matsumoto, k.; nakamura, t.trytophan 2,3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice.mol. Brain,2009,2,1-16). Although these enzymes catalyze the same biochemical reaction and all contain heme, their structural similarity is not high, IDO1 is monomeric, TDO exists in tetrameric form, IDO1, which is more closely related in structure, is 43% identical to the amino acid sequence of IDO2, TDO is only 10% identical to IDO1, IDO1 and TDO have different substrate specificities, TDO has greater specificity for L-tryptophan and L-tryptophan derivatives substituted at the 5-and 6-positions of the indole group, and IDO1 has a broader substrate range, a wider range of substrates such as L-and D-tryptophan, tryptamine, 5-hydroxytryptamine and 1-methyl-L-tryptophan (Forouhar, f.et al molecular insights into substrate recognition and catalysis by tryptophan, 3-dioxygenase. Proc. Natl. Acad. Sci. U.S.A.,2007,104,473-478).
In recent years, the regulation of the kynurenine pathway in immune response has been studied more fully, and as the first most widely distributed rate-limiting enzyme in this pathway, the study on IDO1 has been most widespread. The immunosuppressive effect of IDO1 is mainly due to two aspects, i.e. IDO1 reduces local tryptophan concentration, which affects proliferation of many types of cells, e.g. T-lymphocytes are particularly sensitive to tryptophan deficiency, there is a very sensitive regulatory point in the cell cycle of T-cells, local tryptophan deficiency causes T-cells to stagnate in the middle of G1 phase of the cell cycle, which inhibits proliferation of T-cells, and IDO 1-mediated downstream products of the kynurenine pathway, such as L-kynurenine, 3-hydroxy-anthranilic acid, etc., inhibit proliferation of helper T-cells 1 (Th 1) and natural killer cells (NK cells), but also induce apoptosis, in addition tryptophan deficiency in the kynurenine pathway and downstream metabolites and CD8 + T-cell interactions, while reducing CD8 + Immunoreactivity of T-cell cells (Dounay, a.b., tube, j.b., verhoest, p.r. changes and opportunities in the discovery of new therapeutics targeting the kynurenine pathway j. Med. Chem.,2015,58,8762-8782). At present, a great deal of research shows that IDO is highly expressed in leukemia cells, so that local T-cell proliferation is inhibited, T-cell mediated immune response is inhibited, T-cell activation signal transduction is blocked, and thus attack of tumor cells escaping from an immune system is mediated.
Compared to IDO1, little research on TDO was done, platten et al found that TDO is highly expressed in cells of human glioma, and that inhibition or knockout of TDO reduced kynurenine production, but knockout of IDO1 and IDO2 did not affect kynurenine concentration, suggesting that TDO is a major enzyme of tryptophan metabolism in human glioma. Van den Eynde et al demonstrated that inhibition of TDO could inhibit tumor growth by the P815 mast cell tumor (mastocytoma) model, and they also studied the expression of TDO2 and IDO1 in 104 human tumor cell lines, and found 20 expressed TDO2, 17 expressed IDO1, and 16 expressed both (Van den Eynde, B.J.et al, proc.Natl.Acad.Sci.U.S.A.,2012,109,2497-2502).
Searching for highly potent inhibitors based on IDO and/or TDO as targets has become a research hotspot for drug development in recent years (Austin, c.j.d., rendina, l.m. targeting key dioxygenases in tryptophan-kynurenine metabolism for immunomodulation and cancer chemotherapy, drug Discovery Today,2015,20,609-617; gostner, j.m., becker, k.,f. Fuchs, D.the potential of targeting indoleamine, 3-dioxygenase for cancer treatment, expert Opin. Ther. Targets,2015,19,605-615), related patents currently published for IDO and/or TDO as targets, such as WO2010005958, WO2012142237, WO2014159248, WO2015067782, WO2016071283, WO2016147144, WO2016165613, WO2017107979, and the like.
At present, no IDO and/or TDO as target agents are marketed, and some candidate agents are currently subjected to clinical tests in I-III stage, so that in order to achieve the aim of better tumor treatment effect, the new generation of high-efficiency low-toxicity high-efficiency IDO and/or TDO inhibitor agents are expected to be developed for treating tumors, as well as diseases such as virus infection, depression, neurodegenerative diseases, cataract, organ transplant rejection and autoimmune.
Disclosure of Invention
The invention relates to a novel compound of a small molecular IDO inhibitor, and pharmaceutically acceptable salts, stereoisomers, tautomeric forms, polymorphs, solvates, prodrugs, metabolites and isotopic derivatives thereof, wherein the structural general formula of the compound is shown in a formula I. The invention also relates to pharmaceutical compositions comprising said compounds, and pharmaceutically acceptable salts, stereoisomers, tautomeric forms, polymorphs, solvates, prodrugs, metabolites, isotopic derivatives thereof. The research shows that the synthesized small molecular IDO inhibitor has obvious drug effect and high safety, and is hopeful to become a novel antitumor drug.
One aspect of the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof:
wherein R is 0 Is that
R 1 And R is 2 Respectively and independently halogen, cyano and C 1-3 Alkyl, halogen substituted C 1-6 Alkyl or C 1-6 An alkoxy group; preferably halogen, cyano, C 1-3 Alkyl, halogen substituted C 1-3 Alkyl or C 1-3 An alkoxy group;
r' is
X is-NH-, -O-, and-S-or-NR "-;
y is a single bond, -NH-or-O-;
when Y is a single bond, R is
When Y is-NH-, R is
When Y is-O-, R is
R 3 is-NH 2 、C 1-10 Alkyl, C 3-10 Cycloalkyl, C 3-10 Heterocyclyl, C 6-20 Aryl or C 3-14 Heteroaryl; wherein said C 1-10 Alkyl, C 3-10 Cycloalkyl, C 3-10 Heterocyclyl, C 6-20 Aryl or C 3-14 Heteroaryl groups may be substituted with one or more of the following groups: halogen, C 1-8 Alkyl, C 1-8 Alkoxy, C 1-8 Carboxyl, hydroxyl, amino, nitro, cyano, aldehyde, alkanoyl, trifluoromethyl, p-nitrophenyl, sulfonamide, alkylsulfonyl, benzenesulfonyl, arylsulfonyl or C 1-8 Nitric acid esters;
preferably, R 3 is-NH 2 3-6 membered cycloalkyl, 4-6 membered heterocyclyl or C 1-4 Alkyl, wherein said 3-6 membered cycloalkyl, 4-6 membered heterocyclyl and C 1-4 Alkyl groups may be mono-or di-substituted with: -OH, C 1-4 Alkyl, -O-C 1-4 Alkyl, -NH 2 Halogen, phenyl, cyano;
R 4 、R 5 、R 6 、R 7 、R 8 、R 9 respectively and independently-H or C 1-3 An alkyl group;
R 10 is-H, C 1-10 Alkyl, C 3-10 Cycloalkyl, C 3-10 Heterocyclyl, C 6-20 Aryl, C 3-14 Heteroaryl orWherein said C 1-10 Alkyl, C 3-10 Cycloalkyl, C 3-10 Heterocyclyl, C 6-20 Aryl, C 3-14 Heteroaryl groups may be substituted with one or more of the following groups: halogen, C 1-8 Alkyl, C 1-8 Alkoxy, C 1-8 Carboxyl, hydroxyl, amino, nitro, cyano, aldehyde, alkanoyl, trifluoromethyl, p-nitrophenyl, sulfonamide, alkylsulfonyl, benzenesulfonyl, arylsulfonyl or C 1-8 Nitric acid esters;
preferably, R 10 is-H,A 4-6 membered heterocyclyl wherein the heteroatom in the heterocyclyl is O or N;
or R is 9 And R is 10 Together with the attached nitrogen atom, form a 4-6 membered ring or form a morpholinyl group;
R 11 and R is 12 Respectively and independently C 1-3 Alkyl, -O-C 1-4 Alkyl, or R 11 And R is 12 Together with the phosphorus atom to which it is attached, form a 4-6 membered ring;
r' and R 13 Respectively and independently C 1-3 An alkyl group;
R 14 is-H, -CN,3-6 membered cycloalkyl or 4-6 membered heterocyclyl;
R 20 、R 21 respectively and independently-H or C 1-3 Alkyl, or R 20 And R is 21 Together forming a 3-6 membered cycloalkyl;
R 22 、R 23 respectively and independently-H or C 1-3 An alkyl group;
R 24 is-CN, C 1-10 Alkyl, C 3-10 Cycloalkyl, C 3-10 Heterocyclyl, C 6-20 Aryl, C 3-14 Heteroaryl; wherein said C 1-10 Alkyl, C 3-10 Cycloalkyl, C 3-10 Heterocyclyl, C 6-20 Aryl, C 3-14 Heteroaryl groups may be substituted with one or more of the following groups: halogen, C 1-8 Alkyl, C 1-8 Alkoxy, C 1-8 Carboxyl, hydroxyl, amino, nitro, cyano, aldehyde, alkanoyl, trifluoromethyl, p-nitrophenyl, sulfonamide, alkylsulfonyl, benzenesulfonyl, arylsulfonyl or C 1-8 Nitric acid esters;
preferably, R 24 Is C 1-3 Alkyl, -CN, 3-6 membered cycloalkyl or 4-6 membered heterocyclyl;
n is 0, 1 or 2;
m is 1, 2 or 3.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is that
Some embodiments of the present invention relate to compounds of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof, wherein X is-NH-or-NR "-; y is-NH-; r isn is 1.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 1 And R is 2 Respectively and independently of one another are-C 1-3 Alkyl, -C 1-3 Alkoxy, -F, -Br, -Cl, -CF 3 or-CHF 2 ;R 10 is-H.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatX is-S-or-O-; y is a single bond; n is 1.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite thereofAn object or isotope derivative, wherein R 1 And R is 2 Respectively and independently C 1-3 Alkyl, C 1-3 Alkoxy, -F, -Br, -Cl, -CF 3 or-CHF 2 ;R 13 Is methyl.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatX is-NR "-; y is-NH-; n is 1.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 1 And R is 2 Respectively and independently C 1-3 Alkyl, C 1-3 Alkoxy, -F, -Br, -Cl, -CF 3 or-CHF 2
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatR 2 is-CHF 2
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 1 is-F; n is 1.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatY is a single bond or-NH-;
when Y is a single bond, R is
When Y is-NH-, R is
R 10 Is thatOr a 4-6 membered heterocyclic group, wherein the heteroatom in the heterocyclic group is O or N;
or R is 9 And R is 10 Together with the nitrogen atom to which they are attached form a 4-6 membered ring or form a morpholinyl group.
Some embodiments of the present invention relate to compounds of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof, wherein X is-NH-or-NR "-; the 4-6 membered heterocyclic group is n is 1.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 1 And R is 2 Respectively and independently of one another are-C 1-3 Alkyl, -F, -Br, -Cl, -CF 3 or-CHF 2
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or equivalent thereofDerivatives of the potential element, wherein R 0 Is thatY is-O-;
r isR 10 Is->Or a 4-6 membered heterocyclic group, wherein the heteroatom in the heterocyclic group is O or N.
Some embodiments of the present invention relate to compounds of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof, wherein X is-NH-or-NR "-; the 4-6 membered heterocyclic group is n is 1.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 1 And R is 2 Respectively and independently of one another are-C 1-3 Alkyl, -F, -Br, -Cl, -CF 3 or-CHF 2
Some embodiments of the present invention relate to compounds of the aforementioned general formula (I) or pharmaceutically acceptable salts, stereoisomers, tautomeric forms, polymorphs, solvates, prodrugs, metabolites or isotopic derivatives thereof, wherein Y is-NH-; r is
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or equivalent thereofDerivatives of a hormone, wherein R 1 And R is 2 Respectively and independently C 1-3 Alkyl, C 1-3 Alkoxy, -F, -Br, -Cl, -CF 3 、-CHF 2
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatR' is->
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 1 And R is 2 Respectively and independently of one another are-C 1-3 Alkyl, -C 1-3 Alkoxy, -F, -Br, -Cl, -CF 3 、-CHF 2 ;R 10 is-H.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatR' is->
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 2 is-CHF 2
Some embodiments of the present invention relate to the aforementioned compounds of formula (I) or a pharmaceutically acceptable salt, stereoisomer thereofA isomer, tautomeric form, polymorph, solvate, prodrug, metabolite or isotopic derivative, wherein R 1 is-F.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatY is a single bond; r is R 3 is-C 1-3 Alkyl, -CH 2 CH 2 OH、-CH 2 CH 2 -O-CH 3 3-6 membered cycloalkyl, 4-6 membered heterocyclyl, wherein the heteroatom in the heterocyclyl is O or N, any carbon atom on the 3-6 membered cycloalkyl being mono-or di-substituted by halogen.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, wherein R 0 Is thatY is-NH-, R is-> R 10 Is->Or a 4-6 membered heterocyclic group, wherein the heteroatom in the heterocyclic group is O or N; or R is 10 And R is 9 Together with the attached nitrogen atom, form a 4-6 membered ring or form a morpholinyl group; r is R 11 And R is 12 At the same time be-C 1-3 Alkyl or-O-C 1-3 An alkyl group.
Some embodiments of the present invention relate to a compound of the aforementioned general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, precursor thereofA drug, metabolite or isotopic derivative, wherein R 1 And R is 2 Respectively and independently of one another are-C 1-3 Alkyl, -F, -Br, -Cl, -CF 3 or-CHF 2
The synthesis flow of the compound provided by the invention is as follows:
(one) when R in the formula (I) 0 Is thatWherein X is-NH-or-NR "-, when Y is a single bond, R is
When Y is-NH-or-NR' -R isWhen Y is-O-, R is->Wherein R is 10 Is->
The synthetic route of the above structure is as follows:
compounds Ia are in a base (e.g. NaHCO 3 ,Na 2 CO 3 NaOH, etc.), with R 1 And R is 2 Substituted anilines are reacted to give the compounds Ib.
Compound Ib reacts with CDI under heating to give the ring-closing product Ic.
Compound Ic is in acidic conditions (e.g. concentrated H 2 SO 4 TFA, etc.) with an oxidizing agent (e.g., H) 2 O 2 Urea peroxide, percarbonateSodium, etc.) to give nitro compound Id.
Compounds Id are reacted with amines Ie-1 (or under basic conditions (e.g. NaHCO) 3 Triethylamine, etc.) and salts thereof to give the compound If.
Compounds If under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) to give the target compound I-1.
(II) when R in the formula (I) 0 Is thatWherein X is-NH-or-NR', Y is-NH-, and R isR 10 Is->Wherein R is 23 is-H, R may be
The synthetic route of the above structure is as follows:
compounds Id are reacted with amines Ie-2 (or under basic conditions (e.g. NaHCO) 3 Triethylamine, etc.) and salts thereof) to give compound Ig.
Compound Ig in TBAF, NH 4 F, removing TBDPS protecting groups under the action of HF and the like to obtain a compound Ih.
Compound Ih under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-2.
(III) when R in the formula (I) 0 Is thatWherein X is-NH-or-NR "-, Y is-NH-or-O-, R is->R 10 Is 4-6 membered heteroA cyclic group in which the hetero atom in the heterocyclic group may be-O-or-NH-, and the 4-6 membered heterocyclic group may be specifically +.>
The synthetic route of the above structure is as follows:
Compound Id is reacted with amine Ie-3 (or under basic conditions (e.g. NaHCO 3 Triethylamine, etc.) and salts thereof to give compound Ii.
The Boc protecting group of the compound Ii is removed under acidic conditions (such as HCl, HBr, TFA and the like) to obtain the compound Ij.
Compounds Ij under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-3.
(IV) when R in the formula (I) 0 Is thatWherein X is-NH-or-NR "-, Y is-NH-, R isR 10 Is->Wherein R is 23 Is C 1-3 An alkyl group; r is R 9 And R is 10 May form a 4-6 membered ring together with the nitrogen atom to which it is attached or form a morpholinyl group; r can be specifically
The synthetic route of the above structure is as follows:
the compound Id is reacted with a mono-Boc protected diamine Ie-4 to give the compound Ik.
Removing Boc protecting group from compound Ik under acidic condition (such as HCl, HBr, TFA, etc.), to obtain compound IL or its salt.
Compound Il under basic conditions (e.g. triethylamine, diisopropylethylamine, naHCO) 3 ,Na 2 CO 3 ,K 2 CO 3 Etc.) with sulfonyl chloride or phosphoryl chloride to give compound Im.
Compound Im is prepared under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-4.
(V) when R in the formula (I) 0 Is thatWherein X is-NR "-, Y is-NH-, R is +.>Wherein R is 9 And R is 10 When the two groups are all-H, the two groups are respectively,
the synthetic route of the above structure is as follows:
The reaction product of chlorosulfonyl isocyanate with t-butanol is reacted with compound Il under basic conditions (e.g. triethylamine, diisopropylethylamine, naHCO 3 ,Na 2 CO 3 ,K 2 CO 3 Etc.) to give compound In.
And removing the Boc protecting group of the compound In under acidic conditions (such as HCl, HBr, TFA and the like) to obtain the compound Io.
Compounds Io under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-5.
(VI) when R in the formula (I) 0 Is thatWherein Y is a single bond and R is->R 14 is-H orIn the time-course of which the first and second contact surfaces,
the synthetic route of the above structure is as follows:
compound Id is reacted with an amine or alcohol Ie-5 under basic conditions (e.g., triethylamine, naOH, tBuOK, naH, etc.) to give compound Ip.
The compound Ip is oxidized in an oxidizing agent (e.g. m-CPBA, naIO 4 Etc.) to obtain sulfoxide product Iq.
Compound Iq in PhI (OAc) 2 Under the action of ammonium carbamate or concentrated sulfuric acid, naN 3 The compound Ir is produced.
Ir compounds under alkaline conditions (e.g. NaOH, K) 2 CO 3 Etc.) ring opening to give compound I-6.
Ir compound under basic conditions (e.g. triethylamine, diisopropylethylamine, naHCO) 3 ,Na 2 CO 3 ,K 2 CO 3 Etc.) with a cycloalkylsulfonyl chloride compound to give compound Is.
Compounds Is under alkaline conditions (e.g. NaOH, K) 2 CO 3 Etc.) ring opening to give compound I-7.
(seventh) when R in the formula (I) 0 Is thatWherein Y is a single bond and R is->R 14 In the case of the-CN,
the synthetic route of the above structure is as follows:
compound Ip in PhI (OAc) 2 Under the action of the compound, the compound It is obtained by the action of cyanamide.
Compound It is oxidized in an oxidizing agent (e.g. m-CPBA, naIO 4 Etc.), compound Iu is obtained.
Compounds Iu are under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-8.
(eight) when R in the formula (I) 0 Is thatWherein Y is-NH-, R isR 10 In the case of the compound of formula-H,
the synthetic route of the above structure is as follows:
wherein ring A is
The compound Iaa being over Pd catalyst (e.g. Pd (OAc) 2 ,Pd 2 (dba) 3 ,Pd(CH 3 CN) 2 Cl 2 Etc.), phosphine ligands (e.g., BINAP, xantPhos, P (t-Bu) 3 BrettPhos et al), under alkaline conditions (e.g., cs 2 CO 3 ,t-BuONa,K 3 PO 4 Etc.) with a mono-Boc protected diamine to give the compound Iab.
The compound Iab is hydrolyzed under alkaline conditions (such as LiOH, naOH, etc.), and the compound Iac is obtained.
Compound Iac is reacted with substituted aniline under the action of condensing agent (e.g., HATU, HBTU, HOAt, HOBt, EDC, DCC, etc.) to give compound Iad.
Compound Iad is subjected to removal of Boc protecting groups under acidic conditions (e.g., HCl, HBr, TFA, etc.) to yield compound Iae.
Compound Iae under basic conditions (e.g. NaHCO 3 ,Na 2 CO 3 Triethylamine, diisopropylethylamine, etc.) with FmocCl to give compound Iaf.
Compound Iaf in PCl 5 Under the action of the compound Iag is obtained.
Compound Iag under basic conditions (e.g. NaHCO 3 ,Na 2 CO 3 NaOAc, etc.), with hydroxylamine hydrochloride (or hydroxylamine sulfate) to give compound Iah.
The compound Iah reacts with CDI under heating to obtain a ring-closing product Iai.
Removing Fmoc protecting group from the compound Iai under the action of piperidine, pyrrolidine, TBAF or the like to obtain the compound Iaj.
The reaction product of chlorosulfonyl isocyanate with t-butanol is reacted with compound Iaj under basic conditions (e.g., triethylamine, diisopropylethylamine, naHCO) 3 ,Na 2 CO 3 ,K 2 CO 3 Etc.) to give compound Iak.
Compound Iak is subjected to removal of Boc protecting groups under acidic conditions (e.g., HCl, HBr, TFA, etc.) to yield compound Ial.
Compound Ial under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-9.
(nine) when R in the formula (I) 0 Is thatWherein Y is-NH-, R is->R 10 In the case of the compound of formula-H,
the synthetic route of the above structure is as follows:
wherein ring A is
The compound Iba is hydrolyzed under alkaline conditions (e.g., liOH, naOH, etc.), to obtain the compound Ibb.
The compound Ibb reacts with substituted aniline under the action of condensing agent (such as HATU, HBTU, HOAt, HOBt, EDC, DCC and the like) to obtain the compound Ibc.
Compound Ibc at PCl 5 Under the action of this, the compound Ibd is obtained.
Compound Ibd is prepared under basic conditions (e.g. NaHCO 3 ,Na 2 CO 3 NaOAc, etc.), with hydroxylamine hydrochloride (or hydroxylamine sulfate) to give compound Ibe.
The compound Ibe reacts with CDI under the heating condition to obtain a ring-closing product Ibf.
The reaction product of chlorosulfonyl isocyanate with t-butanol is reacted with mono-Boc protected diamine under basic conditions (e.g., triethylamine, diisopropylethylamine, naHCO 3 ,Na 2 CO 3 ,K 2 CO 3 Etc.) to give the compound Ibh.
And removing the Boc protecting group of the compound Ibh under acidic conditions (such as HCl, HBr, TFA and the like) to obtain the compound Ibi.
The compound Ibf reacts with the compound Ibi under the condition of room temperature or heating to obtain the compound Ibg.
Compound Ibg is prepared under alkaline conditions (e.g. NaOH, K) 2 CO 3 Etc.) ring opening to give compound I-10.
(ten) when R in the formula (I) 0 Is thatWherein Y is-NH-, R is->R 10 In the case of the compound of formula-H,
the synthetic route of the above structure is as follows:
wherein ring A is
The compound Ica reacts with substituted aniline under the action of condensing agent (such as HATU, HBTU, HOAt, HOBt, EDC, DCC, etc.) to obtain compound Icb.
Compound Icb in PCl 5 Under the action of this, compound Icc is obtained.
Compound Icc under basic conditions (e.g. NaHCO 3 ,Na 2 CO 3 NaOAc, etc.), with hydroxylamine hydrochloride (or hydroxylamine sulfate) to give compound Icd.
The compound Icd reacts with CDI under heating to obtain a ring-closing product Ice.
The compound Ice is reacted with an aldehyde Icf under heating, and the resulting imine product is further reacted with a reducing agent (e.g., naBH 4 ,NaBH 3 (CN),NaBH(OAc) 3 Etc.) to give compound Icg. Or the compound Ice and the aldehyde Icf are directly reacted with a reducing agent (such as NaBH 3 (CN),NaBH(OAc) 3 Etc.), the compound Icg is obtained.
The Boc protecting group of the compound Icg is removed under acidic conditions (such as HCl, HBr, TFA and the like) to obtain the compound ICh.
The reaction product of chlorosulfonyl isocyanate with t-butanol is reacted with compound ICh under basic conditions (e.g., triethylamine, diisopropylethylamine, naHCO 3 ,Na 2 CO 3 ,K 2 CO 3 Etc.) to give the compound Ici.
The Boc protecting group of the compound Ici is removed under acidic conditions (such as HCl, HBr, TFA and the like) to obtain a compound Icj.
Compound Icj is prepared under alkaline conditions (e.g. NaOH, K 2 CO 3 Etc.) ring opening to give compound I-11.
Representative compounds of formula (I) of the present invention are as follows:
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3-aminosulfonyl propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (methylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (2-methoxyethyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (N- (2-hydroxypropyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (2-hydroxyethyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (1-hydroxymethyl) cyclopropyl) sulfamoyl) amino) ethyl) amino-1, 2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((N- (1-hydroxy-2-methylpropan-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((N- (1-hydroxypropane-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N-methyl-N- (pyrrolidin-3-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((2- (2, 2-dioxido-1, 2, 3-oxathiazolidin-3-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((2- (1, 1-isothiazolidin-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazol-3-carboxamidine;
N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonylimidazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-pyrrolidin-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-cimetidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-methyl-2-oxo-1, 3, 2-diazaphospholan-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2-oxo-1, 3, 2-dioxaphosphorin-2-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((2- ((dimethylphosphoryl) amino) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((2- ((diethylphosphoryl) amino) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((1-oxo-2, 5-dihydropyrazol-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((1-oxo-2, 5-phosphol-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonylpyrrolidin-2-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonylpyrrolidin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonyloxazolidin-5-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonyloxazolidin-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (3-methyl-2-carbonyloxazolidin-5-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (3-methyl-2-carbonyloxazolidin-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (1-methyl-2-carbonylimidazolidin-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (3-methyl-2-carbonylimidazolidin-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazolidin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonyl-imidazolidin-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1H-tetrazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-oxo-1, 6-dihydropyridin-2-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-oxo-1, 6-dihydropyridin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-carbonyl-1, 6-dihydropyridazin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) amino) -1,2, 5-oxadiazol-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (morpholine-4-sulfonylamino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (S-methylsulfonylimine) ethyl) mercapto) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- (2- (S-methylsulfonylimino) ethoxy) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
4- ((2- (N-cyclopropylsulfonyl) -S-methylsulfonylimino) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
4- ((2- (S-methyl sulfoxide cyanoimine) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
methyl (pyrrolidin-3-yl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester;
(2-methoxyethyl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester;
(1-azetidin-3-yl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- (methyl (2- (aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxy-formamidino) -N- (2- ((sulfamoyl) amino) ethyl) -1,2, 5-oxadiazole-3-carboxamide;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-5- ((2- ((aminosulfonyl) amino) ethyl) amino) -1,2, 3-thiadiazole-4-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((aminosulfonyl) amino) ethyl) amino) -1,2, 5-thiadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((aminosulfonyl) amino) ethyl) amino) furan-3-carboxamidine;
N- (3-bromo-4-fluorophenyl) -N' -hydroxy-3- ((2- ((aminosulfonyl) amino) ethyl) amino) thiophene-2-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-3- ((2- ((aminosulfonyl) amino) ethyl) amino) -1H-pyrazole-4-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((aminosulfonyl) amino) ethyl) amino) -1H-pyrazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (ethylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((1-methylethyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (cyclopropylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((2-hydroxyethyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (((tetrahydrofuran-3-yl) methyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (((tetrahydrofuran-2-yl) methyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((3- (((3, 3-difluorocyclobutyl) methyl) sulfonyl) propyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((2-methoxyethyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((tetrahydro-2H-pyran-4-yl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -4- ((3- (cyclohexylsulfonyl) propyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonylimidazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (2-pyrrolidin-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazol-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2-oxo-1, 3, 2-dioxaphosphorin-2-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (1-hydroxy-2-methylpropan-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (S-methylsulfoxide imino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
(2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) phosphoramidate;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((1- (2-oxo-1, 3, 2-dioxaphosphorin-2-yl) piperidin-4-yl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (aminosulfonyl (1-aminosulfonyl azetidin-3-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine;
n- (3-chloro-4-fluorophenyl) -N' -hydroxy-4- ((3- (methylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine.
The term "pharmaceutically acceptable salt" as used herein includes acid addition salts and base addition salts thereof.
Suitable acid addition salts are formed from acids that form non-toxic salts. Examples include, but are not limited to, hydrochloride, sulfate/bisulfate, nitrate, phosphate/hydrogen phosphate/dihydrogen phosphate, hydrobromide, hydroiodide, acetate, lactate, methanesulfonate, citrate, malate, maleate, fumarate, tartrate, salicylate, stearate, and the like.
Suitable base addition salts are formed from bases that form non-toxic salts. Examples include, but are not limited to, sodium, potassium, calcium, magnesium salts, or salts with ammonia or organic amines such as ethylamine, ethanolamine, triethanolamine, or amino acids, for example, as alkali metal salts, alkaline earth metal salts, or as ammonium salts.
For a review of suitable salts, see Stahl, P.H., and Wermuth, C.G., et al, "Handbook of Pharmaceutical Salts: properties, selection, and Use,2 nd Revised Edition”(Wiley-VCH,2011)。
If the compounds of the general formula (I) contain both acidic and basic groups in the molecule, the invention also includes internal salts or betaines (zwitterionic) other than the salt forms mentioned.
Methods for preparing pharmaceutically acceptable salts of the compounds of the invention are known to those skilled in the art, for example by contacting the compounds with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts.
The compounds of the general formula (I) may be present in crystalline or amorphous form. Furthermore, certain crystalline forms of the compounds of formula (I) may exist as polymorphs, which are included in the scope of the present invention. The polymorphs of the compound of formula (I) can be distinguished using many conventional analytical techniques including, but not limited to, single crystal X-ray powder diffraction (XRPD) patterns, infrared (IR) spectra, raman spectra, differential Scanning Calorimetry (DSC), thermogravimetric analysis (TGA), and solid state nuclear magnetic resonance (ssNMR) characterization.
The compounds of the invention also include tautomeric forms. Tautomeric forms result from the exchange of single bonds with adjacent double bonds and concomitant proton migration. Tautomeric forms include proton transfer tautomers, which are isomerically protonated states of the same empirical formula and total charge. Examples of proton transfer tautomers include keto-enol pairs, amide-imidic acid pairs, lactam-lactam pairs, enamine-imine pairs, and cyclic forms in which protons can occupy two or more positions of a heterocyclic ring system, for example, 1H-and 3H-imidazoles, 1H-, 2H-and 4H-1,2,4 triazoles, 1H-and 2H-isoindoles, and 1H-and 2H-pyrazoles. Tautomeric forms may be in equilibrium or spatially fixed to one form by appropriate substitution.
Another aspect of the invention relates to a pharmaceutical composition comprising a therapeutically effective dose of a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof.
In some embodiments of the invention, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers, diluents or excipients.
The present invention also relates to a process for preparing the above composition, which comprises mixing a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof with a pharmaceutically acceptable carrier, diluent or excipient.
The invention further relates to the use of a compound of general formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the prophylaxis and/or treatment of a disease having the pathological features of IDO-mediated tryptophan metabolic pathways. IDO inhibitors may be used for inhibition of cardiac disorders and for treatment of other diseases characterized by the pathology of IDO mediated tryptophan metabolic pathways, including infections with viruses such as AIDS, cellular infections such as lyme disease and streptococcal infections, neurodegenerative disorders (e.g. alzheimer's disease, huntington's disease and parkinson's disease), autoimmune diseases, depression, anxiety, cataracts, psychological disorders, cancers (including T-cell leukemia and colon cancer), eye disease states (e.g. cataracts and age-related yellowing), and autoimmune diseases, wherein the cancers may be selected from breast cancer, cervical cancer, colon cancer, lung cancer, gastric cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumors, ovarian tumors, peritoneal tumors, stage IV melanoma, solid tumors, glioma, glioblastoma, hepatoma, mastocytosis, renal tumor, head and neck tumors, leukemia, lymphoma, myeloma, and non-small cell lung cancer.
The invention also relates to compounds of the general formula (I) or pharmaceutically acceptable salts, stereoisomers, tautomeric forms, polymorphs, solvates, prodrugs, metabolites or isotopic derivatives thereof, or pharmaceutical compositions comprising the same, for use in the prevention and/or treatment of diseases that are prevented from being characterized by the pathology of the IDO mediated tryptophan metabolic pathway, these diseases include infections by viruses such as AIDS, cellular infections such as Lyme disease and streptococcal infection, neurodegenerative disorders (e.g. Alzheimer's disease, huntington's disease and Parkinson's disease), autoimmune diseases, depression, anxiety, cataracts, psychological disorders, cancers (including T-cell leukemia and colon cancer), eye disease states (e.g. cataracts and age-related yellowing) and autoimmune diseases, wherein the cancer may be selected from breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumors, ovarian tumors, peritoneal tumors, stage IV melanoma, solid tumors, glioma, glioblastoma, hepatocellular carcinoma, mastoid kidney tumor, head and neck tumors, leukemia, lymphoma, myeloma, and non-small cell lung cancer.
The present invention also relates to a method of treatment and/or prophylaxis of a disease characterized by the pathology of the IDO-mediated tryptophan metabolic pathway, comprising administering to a patient a therapeutically effective dose of a compound of formula (I), or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof, or a pharmaceutical composition comprising the same. These diseases include infections with viruses such as AIDS, cellular infections such as lyme disease and streptococcal infection, neurodegenerative disorders (e.g. alzheimer's disease, huntington's disease and parkinson's disease), autoimmune diseases, depression, anxiety, cataracts, psychological disorders, cancers (including T-cell leukemias and colon cancers), eye disease states (e.g. cataracts and age-related yellowing), and autoimmune diseases, wherein the cancers may be selected from breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumors, ovarian tumors, peritoneal tumors, stage IV melanoma, solid tumors, gliomas, glioblastomas, hepatocellular carcinoma, mastocytosis, head and neck tumors, leukemias, lymphomas, myelomas and non-small cell lung cancer.
Another aspect of the invention relates to a method of treating cancer comprising administering to a patient a therapeutically effective dose of a compound of formula (I) of the invention or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, or a pharmaceutical composition comprising the same. The method shows outstanding efficacy and fewer side effects, wherein the cancer may be selected from breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, stage IV melanoma, solid tumor, glioma, glioblastoma, hepatocellular carcinoma, mastocytosis renal tumor, head and neck tumor, leukemia, lymphoma, myeloma, and non-small cell lung cancer.
The pharmaceutical compositions of the present invention may be formulated in conventional manner using one or more pharmaceutically acceptable carriers. Thus, the active compounds of the present invention may be formulated for oral, buccal, intranasal, parenteral (e.g., intravenous, intramuscular, or subcutaneous) or rectal administration. The compounds of the invention may also be formulated in sustained release dosage forms.
Depending on the purpose of the treatment, the pharmaceutical compositions may be formulated into various types of dosage unit forms, such as tablets, pills, powders, liquids, suspensions, emulsions, granules, capsules, suppositories, and injections (solutions and suspensions).
For shaping pharmaceutical compositions in tablet form, any excipient known and widely used in the art may be used. For example, carriers such as lactose, white sugar, sodium chloride, dextrose, urea, starch, calcium carbonate, kaolin, crystalline cellulose, silicic acid, and the like; binders such as water, ethanol, propanol, ordinary syrup, dextrose solution, starch solution, gelatin solution, carboxymethyl cellulose, shellac, methyl cellulose and potassium phosphate, polyvinyl pyrrolidone, and the like; disintegrants such as dry starch, sodium alginate, agar powder and kelp powder, sodium bicarbonate, calcium carbonate, fatty acid esters of polyethylene sorbitan, sodium lauryl sulfate, monoglyceride of stearic acid, starch, lactose, etc.; disintegration inhibitors such as white sugar, glycerol tristearate, coconut oil and hydrogenated oil; adsorption promoters such as quaternary ammonium bases and sodium dodecyl sulfate; wetting agents such as glycerin, starch, and the like; adsorbents such as starch, lactose, kaolin, bentonite, colloidal silicic acid, etc.; and lubricants such as pure talc, stearate, boric acid powder, polyethylene glycol, and the like. The composition can be made into sugar coated tablet, tu Ming film coated tablet, enteric coated tablet, film coated tablet, double-layer film tablet and multilayer tablet by selecting common coating materials as required.
For shaping pharmaceutical compositions in the form of pellets, any excipient known and widely used in the art may be used, for example, carriers such as lactose, starch, coconut oil, hardened vegetable oils, kaolin and talc, etc.; binders such as gum arabic powder, tragacanth powder, gelatin, ethanol and the like; disintegrating agents such as agar and kelp powder.
For shaping pharmaceutical compositions in the form of suppositories, any excipient known and widely used in the art may be used, for example polyethylene glycol, coconut oil, higher alcohols, esters of higher alcohols, gelatin and semisynthetic glycerides, and the like.
For preparing pharmaceutical compositions in the form of injections, the solutions or suspensions may be sterilized (preferably by adding appropriate amounts of sodium chloride, glucose or glycerol, etc.), and then prepared into injections isotonic with blood. In preparing the injection, any carrier commonly used in the art may be used. For example, water, ethanol, propylene glycol, ethoxylated isostearyl alcohol, polyoxylated isostearyl alcohol, fatty acid esters of polyethylene sorbitan, and the like. In addition, usual dissolving agents, buffers, analgesics, etc. may be added.
The invention further relates to the combination of a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomeric form, polymorph, solvate, prodrug, metabolite, or isotopic derivative thereof, or a pharmaceutical composition comprising the same, with an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an antiviral agent, a chemotherapeutic agent, an immunosuppressant, radiation, an anti-tumor vaccine, an antiviral vaccine, a cytokine therapy, or a tyrosine kinase inhibitor.
As is well known to those skilled in the art, the amount of drug administered depends on a variety of factors, including, but not limited to, the following: the activity of the particular compound employed, the age of the patient, the weight of the patient, the health of the patient, the diet of the patient, the time of administration, the mode of administration, the rate of excretion, the combination of the drugs, and the like. In addition, the optimal mode of treatment, such as the mode of treatment, the daily amount of compound (I) of formula (I) or the type of pharmaceutically acceptable salt, can be verified according to conventional treatment protocols.
Detailed description of the invention
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
The term "alkyl" includes saturated aliphatic groups including straight chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, and the like), branched chain alkyl groups (isopropyl, tert-butyl, isobutyl, and the like), cycloalkyl groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl), alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
In certain embodiments, the linear or branched alkyl group has 6 or fewer carbon atoms in the backbone (e.g., linear C 1-6 Branched chain is C 3-6 ) And more preferably 4 or fewer carbon atoms. Likewise, preferred cycloalkyl groups have 3 to 8 carbon atoms in their ring structure, more preferably 5 or 6 carbons in their ring structure.
The term "C 1-10 Alkyl "includes alkyl groups containing 1 to 10 carbon atoms.
In addition, hydrogen on one or more carbons in the hydrocarbon backbone of the alkyl group may be replaced by a substituent. The substituents may include: alkenyl, alkynyl, halogen, hydroxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthio carbonyl, alkoxy, phosphate, phosphonate, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, sulfate, alkylsulfinyl, sulfonic acid, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl or aromatic or heteroaromatic groups.
The term "halogen" includes fluorine, chlorine, bromine, iodine, and the like.
The term "heterocyclyl" or "heterocycle" includes saturated or unsaturated non-aromatic rings or ring systems, for example, which are 4-, 5-, 6-or 7-membered monocyclic, 7-, 8-, 9-, 10-, 11-or 12-membered bicyclic or 10-, 11-, 12-, 13-, 14-or 15-membered tricyclic ring systems and contain at least one heteroatom selected from O, S and N. Wherein the N and S can also optionally be oxidized to various oxidation states. May be attached to a heteroatom or carbon atom of a heterocyclic group. Heterocyclyl groups may include fused or bridged rings and spiro rings. Examples of heterocycles include Tetrahydrofuran (THF), dihydrofuran, 1, 4-dioxane, morpholine, 1, 4-dithiane, piperazine, piperidine, 1, 3-dioxolane, imidazolidine, imidazoline, pyrroline, pyrrolidine, tetrahydropyran, dihydropyran, oxathiolane (oxathiolane), dithiolane, 1, 3-dioxane, 1, 3-dithiane, oxathiane, thiomorpholine and the like.
The heterocyclic group may be substituted with 1, 2 or 3 substituents such as: alkyl, hydroxy (or protected hydroxy), halogen, oxo (e.g., =o), amino, alkylamino or dialkylamino, alkoxy, cycloalkyl, carboxyl, heterocycloxy (where hetero-epoxy refers to a heterocycle attached via an oxygen bridge), alkyl-O-C (O) -, mercapto, nitro, cyano, sulfamoyl or sulfonamide, aryl, alkyl-C (O) -O-, aryl-S-, aryloxy, alkyl-S-, formyl (e.g., HC (O) -), carbamoyl, aryl-alkyl-, and aryl substituted with alkyl, cycloalkyl, alkoxy, hydroxy, amino, alkyl-C (O) -NH-, alkylamino, dialkylamino, or halogen.
The term "aryl" includes 6-to 8-membered monocyclic aromatic groups, such as phenyl, cyclooctenyl; "aryl" also includes 8-to 14-membered polycyclic (bicyclic, tricyclic) aryl groups such as naphthalene, phenanthrene, and the like.
The term "heteroaryl" refers to an aromatic cyclic group having at least one ring carbon atom replaced with a heteroatom selected from O, S, N, which may contain 1-4 heteroatoms; heteroaryl groups may be monocyclic or polycyclic heteroaryl groups and may be 5-14 membered heteroaryl, 5-12 membered heteroaryl, 5-10 membered heteroaryl, 5-8 membered heteroaryl, 5-6 membered heteroaryl, such as pyrrole, furan, thiophene, thiazole, isothiazole, imidazole, triazole, tetrazole, pyrazole, oxazole, isoxazole, pyridine, pyrazine, pyridazine, pyrimidine, and the like; furthermore, the term "aryl" also includes polycyclic aryl groups such as tricyclic, bicyclic, e.g. naphthalene, benzoxazole, benzodiazole, benzothiazole, benzimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, naphthyridine, indole, benzofuran, purine, benzofuran, deazapurine, or indolizine. These heteroaryl groups are also referred to as "arylheterocycle", "heterocycle", "heteroaryl" or "heteroaromatic groups".
Typical heteroaryl groups include 2-or 3-thienyl; 2-or 3-furyl; 2-or 3-pyrrolyl; 2-, 4-, or 5-imidazolyl; 3-, 4-, or 5-pyrazolyl; 2-, 4-, or 5-thiazolyl; 3-, 4-, or 5-isothiazolyl; 2-, 4-, or 5-oxazolyl; 3-, 4-, or 5-isoxazolyl; 3-or 5-1,2, 4-triazolyl; 4-or 5-1,2, 3-triazolyl; tetrazolyl; 2-, 3-, or 4-pyridyl; 3-or 4-pyridazinyl; 3-, 4-, or 5-pyrazinyl; 2-pyrazinyl; 2-, 4-or 5-pyrimidinyl.
The term "heteroaryl" also includes groups in which the heteroaromatic ring is fused to one or more aryl, cycloaliphatic or heterocyclic rings in which the linking group or point of attachment is on the heteroaromatic ring. Examples thereof include, but are not limited to, 1-, 2-, 3-, 5-, 6-, 7-or 8-indolizinyl; 1-, 3-, 4-, 5-, 6-or 7-isoindolyl; 2-, 3-, 4-, 5-, 6-, or 7-indolyl; 2-, 3-, 4-, 5-, 6-, or 7-indazolyl; 2-, 4-, 5-, 6-, 7-or 8-purinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-or 9-quinolizinyl; 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolinyl; 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolinyl; 1-, 4-, 5-, 6-, 7-or 8-phthalazinyl; 2-, 3-, 4-, 5-or 6-naphthyridinyl; 2-, 3-, 5-, 6-, 7-or 8-quinazolinyl; 3-, 4-, 5-, 6-, 7-or 8-cinnolinyl; 2-, 4-, 6-or 7-pteridinyl; 1-, 2-, 3-, 4-, 5-, 6-, 7-or 8-4aH carbazolyl; 1-, 2-, 3-, 4-, 5-, 6-, 7-or 8-carbazolyl; 1-, 3-, 4-, 5-, 6-, 7-, 8-or 9-carboline group; 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-or 10-phenanthridinyl; 1-, 2-, 3-, 4-, 5-, 6-, 7-, 8-or 9-acridinyl; 1-, 2-, 4-, 5-, 6-, 7-, 8-or 9-pyridyl; 2-, 3-, 4-, 5-, 6-, 8-, 9-or 10-phenanthroline groups; 1-, 2-, 3-, 4-, 6-, 7-, 8-or 9-phenazinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-or 10-phenothiazinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-or 10-phenazinyl; 2-, 3-, 4-, 5-, 6-, or 1-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, or 10-benzisoquinolinyl; 2-, 3-, 4-or thieno [2,3-b ] furyl; 2-, 3-, 5-, 6-, 7-, 8-, 9-, 10-, or 11-7H-pyrazino [2,3-c ] carbazolyl; 2-, 3-, 5-, 6-, or 7-2H-furo [3,2-b ] -pyranyl; 2-, 3-, 4-, 5-, 7-or 8-5H-pyrido [2,3-d ] -o-oxazinyl; 1-, 3-or 5-1H-pyrazolo [4,3-d ] -azolyl; 2-, 4-, or 5-4H-imidazo [4,5-d ] thiazolyl; 3-, 5-or 8-pyrazino [2,3-d ] pyridazinyl; 2-, 3-, 5-, or 6-imidazo [2,1-b ] thiazolyl; 1-, 3-, 6-, 7-, 8-or 9-furo [3,4-c ] cinnolinyl; 1-, 2-, 3-, 4-, 5-, 6-, 8-, 9-, 10-, or 11-4H-pyrido [2,3-c ] carbazolyl; 2-, 3-, 6-, or 7-imidazo [1,2-b ] [1,2,4] triazinyl; 7-benzo [ b ] thienyl; 2-, 4-, 5-, 6-, or 7-benzoxazolyl; 2-, 4-, 5-, 6-, or 7-benzimidazolyl; 2-, 3-, 4-, 5-, 6-, or 7-benzothiazolyl; 1-, 2-, 4-, 5-, 6-, 7-, 8-or 9-benzoxepin; 2-, 4-, 5-, 6-, 7-or 8-benzoxazinyl; 1-, 2-, 3-, 5-, 6-, 7-, 8-, 9-, 10-or 11-1H-pyrrolo [1,2-b ] [2] benzazepine. Typical fused heteroaryl groups include 2-, 3-, 4-, 5-, 6-, 7-or 8-quinolinyl; 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolinyl; 2-, 3-, 4-, 5-, 6-, or 7-indolyl; 2-, 3-, 4-, 5-, 6-, or 7-benzo [ b ] thienyl; 2-, 4-, 5-, 6-, or 7-benzoxazolyl; 2-, 4-, 5-, 6-, or 7-benzimidazolyl; 2-, 4-, 5-, 6-or 7-benzothiazolyl.
The aromatic ring of an "aryl" or "heteroaryl" group may be substituted at one or more ring positions with substituents as described above, such as halogen, hydroxy, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, alkylaminocarbonyl, arylalkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, arylalkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthio carbonyl, phosphate, phosphonate, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylaryl amino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, sulfate, alkylsulfinyl, sulfonate, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic group, where the aryl group may also be fused to a non-aromatic ring or to form a tetrahydroheterocyclic ring (e.g., a tetrahydronaphthalene ring).
The term "alkoxy" includes alkyl groups covalently attached to an oxygen atom. Examples of alkoxy groups include methoxy, ethoxy, isopropoxy, propoxy, butoxy and pentoxy. The hydrogen in the alkoxy group may be substituted with a substituent, and examples of the substituted alkoxy group include haloalkoxy groups. Alkoxy groups may also be substituted with: alkenyl, alkynyl, halogen, hydroxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthio carbonyl, phosphate, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylaryl amino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, alkylsulfinyl, sulfonic acid, sulfamoyl, sulfonylamino, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl or aromatic groups.
The term "alkanoyl" includes the use of acyl (CH 3 A compound or group of CO-) or carbonyl groups. Which includes a substituted acyl group. One or more hydrogen atoms in the alkanoyl group may be replaced by: for example, alkyl, alkynyl, halogen, hydroxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthio carbonyl, alkoxy, phosphate, -P (O) (O-) 2 (phosphonato), -PH (O) (O-) (phosphonato), cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, sulfate (sulfo), alkylsulfinyl, -SO 2 O- (sulfonato), sulfamoyl, sulfonylamino, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic group.
Herein, each group, including but not limited to R 0 、R’、R、R 10 R is R 14 For exampleIn>Indicating the position of attachment of the group to the parent.
Herein, general formula (I)And +.sub.f. in each of the specific compounds below it>Represents a bond between a hydroxyl group and N.
The term "stereoisomer" refers to an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., one, two, three, or four) asymmetric centers, it may result in racemates, racemic mixtures, meso, single enantiomers, diastereomeric mixtures, and individual diastereomers. Specific individual molecules may also exist as geometric isomers (cis/trans).
The term "solvate" refers to a form of a compound that is physically bound to a solvent, typically by a solvolysis reaction. This physical bonding includes hydrogen bonding. Conventional solvents include water, ethanol, methanol, acetic acid, and the like. The compounds of formulas (I-III) may be prepared in crystalline form and may be in solvate form (e.g., hydrated form). Suitable solvates include pharmaceutically acceptable solvates (e.g., hydrates), and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, for example when one or more solvent molecules are incorporated into the lattice of a crystalline solid, the solvate will be able to dissociate. "solvate" encompasses solution phases and dissociable solvates. Representative solvates include hydrates, ethanolates, methanolates, and the like.
The term "prodrug" refers to a derivative which is converted into a compound of the present invention by reaction with an enzyme, gastric acid or the like under physiological conditions in vivo, for example, by oxidation, reduction, hydrolysis or the like each carried out under the catalysis of an enzyme.
The term "metabolite" refers to all molecules derived from any of the compounds of the invention in a cell or organism, preferably a human.
The term "isotopic derivative" refers to a compound comprising an isotope at one or more atoms constituting the compound, in a non-natural proportion. For example deuterium 2 H or D) tritium 3 H or T, C-13% 13 C) Nitrogen-15% 15 N) and oxygen-18% 18 O), and the like.
The term "pharmaceutical composition" means a composition comprising one or more compounds of the general formula (I) of the present invention or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, prodrug, metabolite or isotopic derivative thereof; the pharmaceutical composition may also comprise other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to organisms, facilitate the absorption of active ingredients and thus exert biological activity.
The carriers include all pharmaceutical formulations useful in the pharmaceutical arts for making injectable and non-injectable routes of administration, such as diluents, wetting agents, fillers, binders, wetting agents, disintegrants, absorption enhancers, surfactants, retarders, adsorbents, suspending agents, flocculants, deflocculants, emulsifiers, conventional matrices, solubilizers, co-solvents, cosolvents, preservatives, flavoring agents, colorants, antioxidants, buffers, bacteriostats, isotonicity adjusting agents, pH adjusting agents, metal ion complexing agents, hardeners, thickening agents, and the like.
Detailed Description
The invention will be further described with reference to the following examples, which are not intended to limit the scope of the invention. The experimental methods of the present invention, in which specific conditions are not specified, are generally performed according to conventional conditions or according to conditions suggested by the manufacturer of the raw materials or goods. The reagents of specific origin are not noted and are commercially available conventional reagents.
Abbreviations in the present invention have the following meanings:
/>
/>
the structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR measurements were performed using Varian or Bruker 400MHz nuclear magnetic instruments. NMR chemical shift (delta) of 10 -6 Units of (ppm) are given. The measuring solvent is deuterated chloroform (CDCl) 3 ) Deuterated dimethylBase sulfoxide (DMSO-d) 6 ) Deuterated methanol (CD) 3 OD), internal standard is Tetramethylsilane (TMS).
An Agilent 1260 high performance liquid chromatograph (Agilent ZORBAX C18X4.6mm, 3.5uM column) and an Agilent 6120 mass spectrometer were used for LC-MS measurement.
HPLC determination was performed using a Waters 2695-2996 high pressure liquid chromatograph (Gimini C18.times.4.6 mm column).
The reaction was monitored by Thin Layer Chromatography (TLC) or LC-MS.
The thin layer chromatography silica gel plate uses a smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification of the silica gel plate used by the Thin Layer Chromatography (TLC) is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
Column chromatography generally uses tobacco stage yellow sea silica gel 200-300 mesh or 300-400 mesh silica gel.
The starting materials in the examples of the present invention are known and commercially available or may be synthesized using or according to methods known in the art.
The hydrogenation reaction is usually vacuumized, filled with hydrogen and repeatedly operated for 3 times.
The developing agent system used was: petroleum ether and ethyl acetate systems, n-hexane and ethyl acetate systems, methylene chloride and methanol systems. The volume ratio of the solvent is adjusted according to the polarity of the compound.
The eluent system for column chromatography and the developing agent system for thin layer chromatography used for purifying the compound include: petroleum ether and ethyl acetate systems, n-hexane and ethyl acetate systems, methylene chloride and methanol systems. The volume ratio of the solvent is adjusted according to the polarity of the compound, and can be adjusted by adding a small amount of alkaline or acidic reagents such as triethylamine, acetic acid and the like.
In the following examples, the reaction temperature was room temperature (20℃to 30 ℃) unless otherwise specified.
In the following experimental steps, each starting material, if it is an intermediate prepared in the foregoing steps, may not necessarily be prepared in the batch mentioned for the foregoing intermediate.
Synthesis of intermediates
Synthesis of intermediate Int-1 4- (3-bromo-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one
First step Synthesis of 4-amino-N- (3-bromo-4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-formamidine Int-1b
The compound 4-amino-N-hydroxy-1, 2, 5-oxadiazole-3-carboxamidine chloro Int-1a (20 g,123.4mmol, prepared as disclosed in patent application WO 2010005958) was dissolved in ethyl acetate (100 mL) and water (100 mL), 3-bromo-4-fluoroaniline (23.2 g,123.4 mmol), sodium bicarbonate (15.5 g,185.1 mmol) was added, heated to 60 ℃, reacted for 3 hours, cooled, separated, the aqueous phase extracted with ethyl acetate, the organic phases combined and washed with water, saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated to give the title compound Int-1b (38 g,120.9 mmol), yield 98%.
MS m/z(ESI):316[M+H] +
Second step of synthesizing 3- (4-amino-1, 2, 5-oxadiazol-3-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one Int-1c
Compound Int-1b (20 g,63.4 mmol) was dissolved in ethyl acetate (200 mL), CDI (15.4 g,95.2 mmol) was added, heated to 60℃and reacted for 1 hour, cooled, the reaction mixture was washed with 1M HCl (2X 200 mL), the organic phase was further washed with water, saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound Int-1c (21 g,61.5 mmol) in 97% yield.
MS m/z(ESI):342[M+H] +
Third step of synthesizing 4- (3-bromo-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -ketone Int-1
Compound Int-1c (5 g,14.6 mmol) was dissolved in concentrated sulfuric acid (40 mL), 30% hydrogen peroxide (13.8 mL,117.3 mmol) was slowly added dropwise under ice bath, then stirred under ice bath for 1 hour, warmed to room temperature and stirred for 6 hours, and the reaction was completedAll of them. Poured into ice water, extracted with ethyl acetate (2×200 mL), the organic phases were combined, and dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography (PE: ea=4:1) to give the title compound Int-1 (2.9 g,7.8 mmol) in 53% yield. 1 H NMR(400MHz,CDCl 3 )δ7.61–7.58(m,1H),7.27–7.26(m,1H),7.25(d,J=1.4Hz,1H)。
Synthesis of intermediate Int-2 3- (4- ((2-aminoethyl) amino) -1,2, 5-oxadiazol-3-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride
Synthesis of tert-butyl (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-oxa-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethylcarbamate Int-2a in a first step
Compound Int-1 (1.0 g,2.7 mmol) was dissolved in THF (30 mL), N-t-butoxycarbonyl-1, 2-ethylenediamine (1.3 g,8.1 mmol) was added and stirred at room temperature for 10 min, and TLC monitored for complete consumption of starting material. The reaction was concentrated and the crude product purified by column chromatography (PE: ea=1:1) to give the title compound Int-2a (1.2 g,2.5 mmol) in 92% yield.
1 H NMR(400MHz,MeOD-d 4 )δ7.86(dd,J=6.0,2.5Hz,1H),7.53(ddd,J=8.8,4.2,2.6Hz,1H),7.39(t,J=8.6Hz,1H),3.41(dd,J=6.5,5.0Hz,2H),3.32-3.31(m,2H),1.40(s,9H)。
MS m/z(ESI):507[M+Na] +
Second step Synthesis of 3- (4- ((2-aminoethyl) amino) -1,2, 5-oxadiazol-3-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride Int-2
Compound Int-2a (0.24 g,0.5 mmol) was dissolved in 2M HCl in ethyl acetate (5 mL) and stirred at room temperature for 2h. TLC detection reaction was complete. The solvent was evaporated to dryness under reduced pressure to give the title compound Int-2 (0.21 g,0.5mmol, 100%).
MS m/z(ESI):385[M+H] +
Synthetic intermediate Int-3 3-difluoromethyl-4-fluoroaniline
First step of synthesizing 2-difluoromethyl-1-fluoro-4-nitrobenzene Int-3b
2-fluoro-5-nitrobenzaldehyde Int-3a (3.7 g,22.0 mmol) was dissolved in anhydrous DCM (35 mL), and DAST (17.7 g,110 mmol) in DCM (15 mL) was slowly added dropwise under ice-water bath cooling, and the reaction was carried out for 1h in ice-water bath and overnight at room temperature. The reaction was slowly added dropwise to ice water (100 mL), DCM (100 mL) was added and the organic layer was extracted with saturated NaHCO 3 After washing (2X 50 mL), dried over anhydrous sodium sulfate, filtered, concentrated to dryness, and purified by column chromatography to give the title compound Int-3b (4.1 g) in 98% yield.
Second step of synthesizing 3-difluoromethyl-4-fluoroaniline Int-3
Compound Int-3b (4.1 g,21.5 mmol) was dissolved in MeOH (90 mL), concentrated hydrochloric acid (22 mL) was slowly added dropwise under cooling in an ice-water bath, and iron powder (4.8 g,86.0 mmol) was added in portions to react for 1.5h at room temperature. Drop the reaction solution into saturated NaHCO 3 The solution (150 mL) was extracted with ethyl acetate (2X 100 mL), and the organic layers were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated to dryness to give the title compound Int-3 (3.3 g) in 94% yield.
MS m/z(ESI):162[M+H] +
Synthesis of intermediate Int-4 4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one
Prepared by the same method as Int-1. The first step is to react with Int-1a by taking Int-3 as a raw material.
1 H NMR(400MHz,CDCl 3 )δ7.59–7.56(m,1H),7.51–7.45(m,1H),7.32(tt,J=9.0,1.2Hz,1H),6.88(t,J=54.4Hz,1H)。
Synthesis of intermediate Int-5 4- (3-chloro-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one
3-chloro-4-fluoroaniline is taken as a raw material to react with Int-1 a. The subsequent step was prepared in the same manner as intermediate Int-1.
EXAMPLE 1 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3-sulfamoylpropyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 1
First step of Synthesis of 3- (1, 3-dicarbonyl isoindolin-2-yl) propane-1-sulfonic acid potassium salt 1b
Compound 1, 3-propane sultone 1a (1.0 g,8.2 mmol) and potassium phthalimide (1.5 g,8.2 mmol) were dissolved in ethanol (50 mL) and refluxed for 1 hour. Cooled to room temperature, the white insoluble was filtered and dried to give the title compound 1b (1.5 g,5.5 mmol) as a white solid in 68% yield.
1 H NMR(400MHz,DMSO-d 6 )δ7.86–7.75(m,4H),3.58(t,J=7.2Hz,2H),2.41–2.34(m,2H),1.88–1.79(m,2H)。
MS m/z(ESI):270[M+H] + (sulfonic acid).
Second step of synthesizing 3- (1, 3-dicarbonyl isoindolin-2-yl) propane-1-sulfonamide 1c
Compound 1c (1.0 g,3.3 mmol) was dissolved in dichloromethane (50 mL), and thionyl chloride (4.4 g,37 mmol) was added to the reaction system. The reaction mixture was stirred under reflux for 2 hours. The reaction solution is concentrated to obtain a crude sulfonyl chloride product. The crude product was dissolved in tetrahydrofuran (5 mL), added slowly dropwise to aqueous ammonia (5 mL), stirred at room temperature for 1 hour, and the reaction concentrated to give the crude product which was purified by column chromatography (DCM: meoh=10:1) to give the title compound 1c (460 mg,1.7 mmol) in 53% yield.
MS m/z(ESI):269[M+H] +
Synthesis of 3-aminopropane-1-sulfonylamine hydrochloride 1d in the third step
Compound 1c (1 g,3.7 mmol) was dissolved in ethanol (20 mL) and 98% aqueous hydrazine hydrate (1 mL) was added. Reflux for 2 hours, cool to room temperature, filter the precipitated white solid, and concentrate the filtrate. The solid remaining after concentration was dissolved in water, adjusted to pH 4 by the addition of 3N hydrochloric acid, the white solid was precipitated and filtered, and the filtrate was concentrated to give crude title compound 1d (300 mg,1.6 mmol) in 43% yield.
Fourth step Synthesis of 3- ((4- (4- (3-bromo-4-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) propane-1-sulfonamide 1e
Compound Int-1 (150 mg,0.4 mmol) and compound 1d (83 mg,0.6 mmol) were dissolved in tetrahydrofuran (10 mL), triethylamine (121 mg,1.2 mmol) was added, and stirring was carried out at room temperature for 2 hours, and the disappearance of starting material was monitored by spotting. The reaction was concentrated to give crude product, which was purified by plate chromatography (DCM: meoh=20:1) to give the title compound 1e (80 mg,0.17 mmol) in 43% yield.
MS m/z(ESI):463[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3-sulfamoyl propyl) amino) -1,2, 5-oxadiazole-3-formamidine 1
Compound 1e (80 mg,0.17 mmol) was dissolved in tetrahydrofuran (10 mL), 2M aqueous sodium hydroxide solution (3 mL) was added, stirred at room temperature for 1 hour, quenched with 3M aqueous hydrochloric acid, pH adjusted to 8, the mixture extracted with ethyl acetate (3X 10 mL), and the organic phases combined. Washing with saturated common salt water, drying with anhydrous sodium sulfate, filtering, and concentrating to obtain crude product. The crude product was purified by plate chromatography (DCM: meoh=10:1) to give the title compound 1 (40 mg,0.09 mmol) in 54% yield.
1 H NMR(400MHz,DMSO-d 6 )δ8.87(s,1H),7.19–7.12(m,1H),7.08(dd,J=6.1,2.7Hz,1H),6.78(s,2H),6.73(ddd,J=8.9,4.1,2.8Hz,1H),6.27(t,J=6.0Hz,1H),3.32(d,J=6.6Hz,1H),3.04–2.95(m,2H),1.97(p,J=7.0Hz,2H).MS m/z(ESI):437[M+H] +
EXAMPLE 2 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (methylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 2
First step of Synthesis of tert-butyl (3- (methylthio) propyl) carbamate 2b
The compound (3-bromopropyl) carbamic acid tert-butyl ester 2a (1.0 g,4.48 mmol) was dissolved in DMSO (5 mL), sodium methyl mercaptide (345 mg,4.93 mmol) was added and stirred overnight at room temperature. Water was added and stirred for 0.5h, then ethyl acetate was added to extract, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 2b (82mg, 4.0 mmol) in 89% yield.
Second step of synthesizing (3- (methylsulfonyl) propyl) carbamic acid tert-butyl ester 2c
Compound 2b (82mg, 4.0 mmol) was dissolved in methanol (20 mL), and an aqueous solution (20 mL) of Oxone (3.97 g,6.5 mmol) was added dropwise and stirred at room temperature for 1h. Water and methylene chloride were added to extract, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 2c (1.0 g,4.2 mmol) in 98% yield.
1 H NMR(400MHz,CDCl 3 )δ4.72(s,1H),3.30(t,J=6.7Hz,2H),3.16–3.01(m,2H),2.92(s,3H),2.15–1.92(m,2H),1.44(s,9H)。
Third step of synthesizing 3- (methylsulfonyl) propyl-1-amine 2d
Compound 2c (170 mg,0.72 mmol) was dissolved in dichloromethane (20 mL), TFA (2 mL) was added dropwise, and the mixture was stirred at room temperature for 1h. After the completion of the reaction, the reaction mixture was basified with aqueous ammonia, extracted three times with methylene chloride, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 2d (60 mg,0.44 mmol) in 61% yield.
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((3- (methylsulfonyl) propyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 2e
Compound 2d (60 mg,0.44 mmol) and triethylamine (44 mg,0.44 mmol) were dissolved in tetrahydrofuran (10 mL), and Compound Int-1 (162 mg,0.44 mmol) was added and stirred at room temperature for 1h. Ethyl acetate and water were added, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 2e (180 mg,0.39 mmol) as a product in 88% yield.
1 H NMR(400MHz,CDCl 3 )δ7.64(dd,J=5.8,2.5Hz,1H),7.38–7.33(m,1H),7.32–7.27(m,1H),5.55(t,J=6.6Hz,1H),3.63(q,J=6.4Hz,2H),3.13(t,J=7.4Hz,2H),2.95(s,3H),2.29(p,J=6.8Hz,2H)。
MS m/z(ESI):462[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (methylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-formamidine 2
Compound 2e (180 mg,0.39 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (376 mg,2.73 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, and after concentration, the purification was performed by plate analysis to give the title compound 2 (120 mg,0.28 mmol) in 71% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,1H),8.86(s,1H),7.15(t,J=8.8Hz,1H),7.08(dd,J=6.1,2.7Hz,1H),6.73(ddd,J=8.9,4.2,2.8Hz,1H),6.29(t,J=6.0Hz,1H),3.31(t,J=6.4Hz,2H),3.14–3.07(m,2H),1.99–1.91(m,2H)。
MS m/z(ESI):436[M+H] +
EXAMPLE 3 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (2-methoxyethyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 3
First step of synthesizing (2-methoxyethyl) sulfamoyl chloride 3b
A mixture of 2-methoxyethyl-1-amine 3a (200 mg,2.6 mmol) and triethylamine (0.56 mL,4 mmol) was slowly added dropwise to a 1M solution of sulfonyl chloride in dichloromethane (4 mL) under an ice bath. The reaction mixture was stirred at 0℃for 2 hours, then poured into ice water and extracted with dichloromethane (3X 10 mL). The organic phases were combined, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the crude title compound 3b as a colorless oil. The crude product was used directly in the next reaction.
Second step Synthesis of N- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((N- (2-methoxyethyl) sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 3c
Compound Int-2 (175 mg,0.41 mmol) and triethylamine (83 mg,0.82 mmol) were dissolved in dichloromethane (4 mL), and a dichloromethane solution of compound 3b (84 mg,0.49 mmol) was added dropwise to the system at room temperature. After the completion of the dropwise addition, the reaction mixture was stirred at room temperature for 1 hour, and the reaction solution was concentrated to obtain a crude product. The crude product was purified by plate chromatography (DCM: meoh=10:1) to give the title compound 3c (210 mg,0.4 mmol) in 96% yield.
MS m/z(ESI):522[M+H] +
Third step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (2-methoxyethyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 3
Compound 3c (210 mg,0.4 mmol) was dissolved in tetrahydrofuran (5 mL), and 2M aqueous sodium hydroxide solution (2 mL) was added to the system at room temperature. The reaction mixture was stirred at room temperature for 1 hour, quenched with 3M aqueous hydrochloric acid in an ice bath, pH adjusted to 8, extracted with ethyl acetate (3×10 mL), the organic phases were combined, washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give crude product. The crude product was purified by plate chromatography (DCM: meoh=10:1) to give the title compound 3 (90 mg,0.18 mmol) as a white solid in 45% yield.
1 H NMR(400MHz,MeOD-d 4 )δ7.11(dd,J=6.0,2.7Hz,1H),7.02(t,J=8.6Hz,1H),6.83(ddd,J=8.8,4.1,2.7Hz,1H),3.44(dt,J=7.9,5.9Hz,4H),3.31(s,3H),3.20(dd,J=6.6,5.4Hz,2H),3.11(t,J=5.6Hz,2H)。
MS m/z(ESI):496[M+H] +
EXAMPLE 4 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (N- (2-hydroxypropyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 4
First step Synthesis of 2- ((tert-butyldiphenylsilyl) oxy) propyl-1-amine 4b
Compound 1-amino-2-propanol 4a (500 mg,6.66 mmol), triethylamine (178 mg,7.99 mmol) were dissolved in dichloromethane (20 mL), and tert-butyldiphenylchlorosilane (2.12 g,7.32 mmol) was added thereto and stirred at room temperature for 4h. Saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 4b (1.35 g,4.31 mmol) by column chromatography in 65% yield.
Second step Synthesis of tert-butyl (2- ((2-oxooxazolidine) -3-sulfamoyl) ethyl) carbamate 4e
The compound chlorosulfonyl isocyanate 4c (5 g,35.3 mmol) was dissolved in dichloromethane (10 mL), and a solution of bromoethanol (4.41 g,35.3 mmol) in dichloromethane (10 mL) was added dropwise under ice bath, and reacted under ice bath for 2h.
The compound N-t-butoxycarbonyl-1, 2-ethylenediamine (6.23 g,38.9 mmol) and triethylamine (7.15 g,70.7 mmol) were dissolved in methylene chloride (20 mL), and the reaction solution was added dropwise under an ice bath. After the dripping is finished, naturally heating to room temperature overnight. Water was added thereto, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration and concentration gave the title compound 4e (10 g,32.4 mmol) in 91% yield. The crude product was used directly in the next step.
Third step Synthesis of tert-butyl (2- ((N- (2- ((tert-butyldiphenylsilyl) oxy) propyl) aminosulfonyl) amino) ethyl) carbamate 4f
Compound 4b (1 g,3.23 mmol), 4e (1 g,3.23 mmol) and triethylamine (234 mg,6.47 mmol) were dissolved in acetonitrile (30 mL) and heated to 80℃overnight. Water was added thereto, extraction was performed three times with ethyl acetate, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, and column chromatography after concentration gave the title compound 4f (1.1 g,2.06 mmol) in 64% yield.
1 H NMR(400MHz,CDCl 3 )δ7.70–7.61(m,4H),7.48–7.35(m,6H),4.98–4.34(m,3H),4.06–3.96(m,1H),3.23(brs,2H),3.08–2.90(m,4H),1.43(s,9H),1.11(d,J=6.2Hz,3H),1.06(s,9H)。
MS m/z(ESI):436[M+H-100] +
Fourth step Synthesis of 2- ((N- (2- ((tert-butyldiphenylsilyl) oxy) propyl) aminosulfonyl) amino) ethylamine 4g
Compound 4f (300 mg,0.56 mmol) was dissolved in dichloromethane (20 mL), TFA (2 mL) was added dropwise, and the mixture was stirred at room temperature for 1h. After completion of the reaction, the reaction mixture was basified with aqueous ammonia, extracted three times with methylene chloride, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give 4g (150 mg,0.34 mmol) of the title compound in 62% yield.
Fifth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (N- (2- ((tert-butyldiphenylsilyl) oxy) propyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one for 4H
Compound 4g (210 mg,0.48 mmol) was dissolved in tetrahydrofuran (10 mL), and compound Int-1 (178 mg,0.48 mmol) was added and stirred at room temperature for 1h. Ethyl acetate and water were added, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound (250 mg,0.33 mmol) in 69% yield as purified by plate chromatography.
1 H NMR(400MHz,CDCl 3 )δ7.70–7.61(m,5H),7.48–7.37(m,6H),7.37–7.31(m,1H),7.29(d,J=7.7Hz,1H),5.63–5.56(m,1H),4.48(brs,1H),4.06–3.96(m,1H),3.52(q,J=5.8Hz,2H),3.22(t,J=5.6Hz,2H),3.08–2.89(m,2H),1.12(d,J=6.2Hz,3H),1.06(s,9H)。
MS m/z(ESI):682[M+H-76] +
Sixth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (N- (2-hydroxypropyl) sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 4i
Compound 4h (250 mg,0.33 mmol) was dissolved in tetrahydrofuran (10 mL), TBAF (860 mg,3.3 mmol) was added and stirred at room temperature for 1h. After the completion of the reaction, ethyl acetate was added, and the mixture was washed three times with a saturated ammonium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 4i (100 mg,0.19 mmol) in 58% yield.
MS m/z(ESI):522[M+H] +
Seventh step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (N- (2-hydroxypropyl) sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 4
Compound 4i (100 mg,0.19 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (185 mg,1.34 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration and then purification by plate chromatography gave the title compound 4 (60 mg,0.12 mmol) in 64% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.49(s,1H),8.89(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.0,2.7Hz,1H),6.97(t,J=5.9Hz,1H),6.80–6.73(m,2H),6.23(t,J=6.0Hz,1H),4.67(d,J=4.7Hz,1H),3.73–3.61(m,1H),3.35(q,J=6.1Hz,2H),3.05(q,J=6.2Hz,2H),2.82–2.64(m,2H),1.03(d,J=6.2Hz,3H)。MS m/z(ESI):496[M+H] +
EXAMPLE 5 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (2-hydroxyethyl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 5
Prepared in the same manner as in example 4, starting with ethanolamine instead of 4a.
1 H NMR(400MHz,MeOD-d 4 )δ7.14(dd,J=6.0,2.7Hz,1H),7.05(t,J=8.7Hz,1H),6.85(ddd,J=8.8,4.1,2.7Hz,1H),3.64(t,J=5.9Hz,2H),3.46(dd,J=6.6,5.4Hz,2H),3.24(dd,J=6.6,5.4Hz,2H),3.09(t,J=5.9Hz,2H).
MS m/z(ESI):482[M+H] +
EXAMPLE 6 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (1-hydroxymethyl) cyclopropyl) aminosulfonyl) amino) ethyl) amino-1, 2, 5-oxadiazole-3-carboxamidine 6
Prepared in the same manner as in example 4, starting with (1-aminocyclopropyl) methanol instead of 4a.
1 H NMR(400MHz,DMSO-d 6 )δ11.50(s,1H),8.89(s,1H),7.29(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.89(t,J=5.9Hz,1H),6.82–6.72(m,1H),6.24(t,J=6.0Hz,1H),4.65(t,J=5.8Hz,1H),3.47(d,J=5.8Hz,2H),3.37–3.33(m,2H),3.02(q,J=6.2Hz,2H),0.77–0.71(m,2H),0.63–0.57(m,2H)。MS m/z(ESI):508[M+H] +
EXAMPLE 7 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((N- (1-hydroxy-2-methylpropan-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 7
Prepared in the same manner as in example 4 using 2-amino-2-methyl-1-propanol as a starting material instead of 4a.
1 H NMR(400MHz,Methanol-d 4 )δ.16–6.66(m,5H),3.44(d,J=5.5Hz,4H),3.23(dt,J=6.4,4.9Hz,2H),1.25(s,5H).
MS m/z(ESI):510[M+H] +
EXAMPLE 8 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((N- (1-hydroxypropan-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 8
By the same procedure as in example 4The preparation is carried out by taking 2-amino-1-propanol as initial raw material to replace 4a. 1 H NMR(400MHz,Methanol-d 4 )δ7.11(dd,J=6.0,2.7Hz,1H),7.02(t,J=8.7Hz,1H),6.83(ddd,J=8.8,4.1,2.7Hz,1H),3.55–3.31(m,5H),3.22(dd,J=6.5,5.4Hz,2H),1.16(d,J=6.6Hz,3H).
MS m/z(ESI):496[M+H] +
EXAMPLE 9 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N-methyl-N- (pyrrolidin-3-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 9
First step Synthesis of tert-butyl 3- ((chlorosulfonyl) (meth) amino) pyrrolidine-1-carboxylate 9b
Sulfonyl chloride SO under the protection of nitrogen 2 Cl 2 (0.135 g,1.0 mmol) in dry dichloromethane (5 mL), cooled to-10deg.C, and then a solution of 3- (methylamino) pyrrolidine-1-carboxylic acid tert-butyl ester 9a (1.0 g,1.0 mmol) and triethylamine (0.1 g,1.0 mmol) in dichloromethane (10 mL) was slowly added dropwise. The reaction was allowed to warm to room temperature for 2 hours and was used directly in the next step.
Second step Synthesis of 3- ((N- (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) sulfamoyl) (methyl) amino) pyrrolidine-1-carboxylic acid tert-butyl ester 9c
Under nitrogen protection, the compound Int-2 (0.42 g,1.0 mmol), triethylamine (0.20 g,2.0 mmol) was dissolved in anhydrous dichloromethane (5 mL), cooled to 0deg.C by ice-water bath, and then the one-step reaction solution of compound 9b was added dropwise and stirred at room temperature overnight. Saturated ammonium chloride solution was added, shaking, the organic phase dried over anhydrous sodium sulfate, filtered, and the crude product obtained after concentration was purified by plate chromatography (DCM: meoh=20:1) to give the title compound 9c (200 mg,0.31 mmol) in 31% yield.
MS m/z(ESI):669[M+Na] +
Third step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (N-methyl-N' - (pyrrolidin-3-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one trifluoroacetate 9d
Compound 9c (200 mg,0.31 mmol) was dissolved in DCM (5 mL) and TFA (0.5 mL) was added and reacted at room temperature for 2 hours. The reaction solution was concentrated to dryness under reduced pressure to give the crude title compound 9d (190 mg,0.3 mmol) in 97% yield.
MS m/z(ESI):547[M+H] +
Fourth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((N-methyl-N- (pyrrolidin-3-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 9
Compound 9d (190 mg,0.3 mmol) was dissolved in THF (5 mL) and aqueous NaOH (1.0 mL, 2M) was added. Stirring is carried out for 1 hour. Ammonium chloride solution and ethyl acetate were added to extract, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained after concentration was purified by plate chromatography (DCM: meoh=10:1) to give the title compound 9 (50 mg,0.1 mmol) in 33% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.48(s,1H),9.24(s,1H),9.07(s,1H),8.90(s,1H),7.52(d,J=6.0Hz,1H),7.19(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.78(dd,J=7.7,4.3Hz,1H),6.27(d,J=6.5Hz,1H),4.45(q,J=8.3Hz,1H),3.32(dd,J=14.2,7.7Hz,4H),3.04(d,J=6.1Hz,4H),2.68(s,3H),2.10–1.94(m,2H).
MS m/z(ESI):521[M+H] +
EXAMPLE 10 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- (2, 2-dioxo-1, 2, 3-oxathiazolidin-3-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamide 10
First step of Synthesis of tert-butyl 2-oxo-1, 2, 3-oxathiazolidine-3-carboxylate 10b
Imidazole (2.9 g,43.4 mmol) was dissolved in dichloromethane (50 mL), a solution of thionyl chloride (0.94 mL,13 mmol) in dichloromethane (4 mL) was slowly added dropwise to the system at 0deg.C, and the reaction system was warmed to room temperature and stirred for 1 hour. The reaction was again cooled to-78 ℃, a dichloromethane solution (4 mL) of t-butyl (2-hydroxyethyl) carbamate (1 g,6.2 mmol) of compound 10a was slowly added dropwise to the reaction mixture, then slowly warmed to room temperature and stirred continuously overnight. The solids in the reaction were filtered off, the filter cake was washed twice with dichloromethane, the filtrates were combined and washed twice with water, the organic phase was dried over anhydrous sodium sulfate, filtered and the concentrated crude title compound 10b (1.2 g,5.7 mmol) was 92% yield as a pale brown oil. The crude product was used directly in the next reaction.
Second step of synthesizing 2, 2-dioxido-1, 2, 3-oxathiazolidine-3-tert-butyl formate 10c
Compound 10b (1.2 g,5.7 mmol) was dissolved in dichloromethane (40 mL), an aqueous solution (5 mL) of sodium periodate (3.0 g,14.3 mmol) was added, and after the addition was complete, the reaction mixture was cooled to 0℃and ruthenium trichloride (0.15 g,2.45 mmol) was added to the reaction mixture. The reaction mixture was stirred at 0 ℃ for 2 hours, then slowly warmed to room temperature and stirred overnight. The reaction solution was extracted three times with methylene chloride, and the organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 10c (0.5 g,2.2 mmol) in the form of a milky white solid in 38% yield.
1 H NMR(400MHz,CDCl 3 )δ4.61(t,J=6.4Hz,2H),4.04(t,J=6.4Hz,2H),1.54(s,9H)。
Third step of synthesizing 2, 2-dioxido-1, 2, 3-oxathiazolidine 10d
Compound 10c (0.5 g,2.2 mmol) was dissolved in dichloromethane (10 mL) and trifluoroacetic acid (4 mL) was added slowly. The reaction mixture was stirred at room temperature for 1 hour, and the reaction mixture was concentrated to give the crude title compound 10d, which was used directly in the next reaction.
Fourth step of synthesizing tert-butyl (2- (2, 2-Dioxo-1, 2, 3-oxathiazolidin-3-yl) ethyl) carbamate 10e
Compound 10d (270 mg,2.2 mmol) was dissolved in DMF (5 mL), tert-butyl (2-bromoethyl) carbamate (560 mg,2.6 mmol) and anhydrous potassium carbonate (910 mg,6.6 mmol) were added at room temperature and reacted overnight at room temperature. Saturated ammonium chloride solution was added, extraction was performed three times with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to give the title compound 10e (230 mg,0.86 mmol) in 39% yield.
MS m/z(ESI):289[M+Na] +
Fifth step of synthesizing 2, 2-dioxido-3- (2-amino ethyl) -1,2, 3-oxathiazolidine trifluoroacetate 10f
Compound 10e (230 mg,0.86 mmol) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (1 mL) was added slowly. The reaction mixture was stirred at room temperature for 1 hour, and the reaction mixture was concentrated to give the crude title compound 10f, which was used directly in the next reaction.
Sixth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (2, 2-dioxathiazolidine-3-yl) -2, 3-oxa-thiazolidin-3-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 10g
Compound 10f (200 mg,0.76 mmol) was dissolved in tetrahydrofuran (5 mL) and compound Int-1 (284 mg,0.76 mmol) and triethylamine (154 mg,1.52 mmol) were added. The reaction mixture was stirred at room temperature for 1 hour, and the reaction was concentrated to give crude product, which was purified by plate chromatography (DCM: meoh=10:1) to give the title compound 10g (155 mg,0.32 mmol), yield 42%
MS m/z(ESI):491[M+H] +
Seventh step Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- (2, 2-dioxo-1, 2, 3-oxathiazolidin-3-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamide 10
10g (150 mg,0.31 mmol) of the compound was dissolved in tetrahydrofuran (5 mL), then 2N aqueous sodium hydroxide solution (2 mL) was added, stirred at room temperature for 1 hour, quenched with 3N aqueous hydrochloric acid, pH was adjusted to 8, extracted three times with ethyl acetate, the organic phases were combined, the organic phases were washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give crude product, which was purified by plate chromatography (DCM: meOH=8:1) to give the title compound 10 (45 mg,0.097 mmol) in 32% yield.
MS m/z(ESI):465[M+H] +
EXAMPLE 11 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- (1, 1-isothiazolidin-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 11
/>
First step Synthesis of tert-butyl (2- ((3-chloropropyl) sulfonamide) ethyl) carbamate 11b
N-Boc-1, 2-ethylenediamine (1.6 g,10.0 mmol) and DIPEA (1.50 g,11.6 mmol) were dissolved in THF (20 mL), and a solution of 3-chloropropane-1-sulfonyl chloride 11a (1.77 g,10.0 mmol) in THF (5 mL) was added dropwise with cooling in an ice-water bath. Gradually warmed to room temperature overnight after the completion of the dropping. Spin-drying, adding ethyl acetate and ammonium chloride solution to separate, washing the organic phase with saline solution, drying over anhydrous sodium sulfate, filtering, concentrating to obtain the title compound 11b (3.0 g,10.0mmol, 100%).
1 H NMR(400MHz,CDCl 3 )δ5.03(brs,1H),4.90(brs,1H),3.68(t,J=6.2Hz,2H),3.30–3.26(m,4H),3.23–3.15(m,2H),2.35–2.22(m,2H),1.45(s,9H)。
MS m/z(ESI):301[M+H] +
Second step of synthesizing (2- (1, 1-Dioxisothiazolidin-2-yl) ethyl) carbamic acid tert-butyl ester 11c
Compound 11b (0.6 g,2.0 mmol) was dissolved in anhydrous DMF (10 mL) under nitrogen, naH (120 mg,3.0mmol,60% wt.) was added under ice-bath cooling, and the mixture was gradually warmed to room temperature after addition and stirred overnight. The reaction mixture was quenched by adding an ammonium chloride solution, extracted with ethyl acetate 3 times, the organic phases were combined, washed with saturated brine 2 times, and the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 11c (0.27 g,1.0 mmol) in 51% yield.
1 H NMR(400MHz,CDCl 3 )δ4.92(brs,1H),3.33–3.27(m,4H),3.20–3.11(m,4H),2.41–2.30(m,2H),1.43(s,9H)。
MS m/z(ESI):287[M+Na] +
Third step of synthesizing 2- (1, 1-dioxid isothiazolin-2-yl) ethylamine hydrochloride salt 11d
Compound 11c (0.3 g,1.13 mmol) was dissolved in 2M HCl in ethyl acetate (5 mL) and stirred at room temperature for 4 hours. The title compound 11d (0.23 g,1.13 mmol) was obtained in 100% yield by spin drying. Directly used in the next step.
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (1, 1-isothiazolidin-2-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 11e
Compound 11d (226 mg,1.13 mmol) and triethylamine (230 mg,2.27 mmol) were dissolved in DCM (10 mL), and compound Int-1 (430 mg,1.16 mmol) was added and stirred at room temperature for 30min. The reaction solution was dried by spinning, ethyl acetate and a small amount of ammonium chloride solution were added to wash, and the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product, which was purified by plate chromatography to give the title compound 11e (150 mg,0.3 mmol) in 27% yield.
MS m/z(ESI):489[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- (1, 1-isothiazolidin-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-formamidine 11
Compound 11e (0.10 g,0.2 mmol) was dissolved in THF (10 mL) and aqueous NaOH (1 mL, 2M) was added. Stirring is carried out for 30min at room temperature. Saturated ammonium chloride solution and ethyl acetate were added, the organic phase was separated, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by spin-drying was purified by plate analysis (DCM: meoh=10:1) to give the title compound 11 (42 mg,0.09 mmol) in 44% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.90(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.9,4.2,2.7Hz,1H),6.28(t,J=6.1Hz,1H),3.41(q,J=6.3Hz,2H),3.24(t,J=6.7Hz,2H),3.20–3.15(m,2H),3.13(t,J=6.2Hz,2H),2.27–2.14(m,2H)。
MS m/z(ESI):463[M+H] +
EXAMPLE 12 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazol-3-carboxamide 12
First step Synthesis of 2-benzyl-1, 2, 5-thiadiazole 1, 1-dioxide 12b
Compound N 1 Benzyl-1, 2-ethylenediamine 12a (1.0 g,6.66 mmol) was dissolved in 1, 4-dioxane (30 mL), sulfonamide (640 mg,6.66 mmol) and triethylamine (1.35 g,1.92 mmol) were added, and the mixture was heated under reflux with stirring for 16h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with ethyl acetate, and the organic phase was combined, dried over saturated brine, filtered, concentrated, and the crude product was purified by column chromatography to give the title compound 12b (700 mg,3.3 mmol) in 50% yield.
1 H NMR(400MHz,CDCl 3 )δ7.41–7.27(m,5H),4.56(t,J=6.7Hz,1H),4.18(s,2H),3.47(q,J=6.6Hz,2H),3.27(t,J=6.6Hz,2H)。
MS m/z(ESI):213[M+H] +
Second step of Synthesis of tert-butyl (2- (5-benzyl-1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) carbamate 12c
Compound 12b (300 mg,1.41 mmol) was dissolved in DMF (10 mL), and tert-butyl (2-bromoethyl) carbamate (348 mg,1.55 mmol) and anhydrous potassium carbonate (390 mg,2.82 mmol) were added and reacted overnight at room temperature. A saturated ammonium chloride solution was added, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated, followed by plate analysis to give the title compound 12c (400 mg,1.13 mmol) in 80% yield.
MS m/z(ESI):378[M+Na] +
Third step of synthesizing (2- (1, 1-Dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) carbamic acid tert-butyl ester 12d
Compound 12c (300 mg,0.84 mmol) was dissolved in methanol (10 mL), palladium on carbon (10% Pd,300 mg) was added, ammonium formate (800 mg,12.6 mmol) was added, and the mixture was heated under reflux for 5h. Filtration and concentration gave the crude title compound 12d (200 mg,0.75 mmol) in 89% yield.
1 H NMR(400MHz,CDCl 3 )δ4.91(brs,1H),4.32(brs,1H),3.57–3.44(m,4H),3.39(q,J=5.7Hz,2H),3.17(t,J=6.0Hz,2H),1.44(s,9H)。
MS m/z(ESI):288[M+Na] +
Synthesis of 2- (2-aminoethyl) -1,2, 5-thiadiazole 1, 1-dioxide 12e in the fourth step
Compound 12d (80 mg,0.3 mmol) was dissolved in dichloromethane (5 mL) and TFA (1 mL) was added dropwise at room temperature. Stir at room temperature for 1h. After the completion of the reaction, the solution was alkalified with water and aqueous ammonia, extracted three times with methylene chloride, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 12e (50 mg,0.3 mmol) in 100% yield.
MS m/z(ESI):166[M+H] +
Fifth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((2- (1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 12f
Compound 12e (50 mg,0.3 mmol) and Int-1 (113 mg,0.3 mmol) were dissolved in tetrahydrofuran (3 mL) and stirred at room temperature for 1h. Ethyl acetate and water were added, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 12f (80 mg,0.16 mmol) in 54% yield.
1 H NMR(400MHz,CDCl 3 )δ7.64(dd,J=5.8,2.6Hz,1H),7.35(ddd,J=8.8,4.2,2.6Hz,1H),7.29(d,J=7.7Hz,1H),5.65(t,J=6.2Hz,1H),4.32(brs,1H),3.74–3.64(m,2H),3.60–3.44(m,4H),3.42–3.28(m,2H)。
MS m/z(ESI):490[M+H] +
Sixth step Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazol-3-formamidine 12
Compound 12f (80 mg,0.16 mmol) was dissolved in methanol (5 mL), followed by the addition of potassium carbonate (160 mg,1.16 mmol) and stirring at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, and after concentration, the purification was performed by plate analysis to obtain the title compound 12 (25 mg,0.05 mmol) in 33% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.91(s,1H),7.21–7.15(m,2H),7.12(dd,J=6.1,2.7Hz,1H),6.75(ddd,J=8.9,4.2,2.7Hz,1H),6.31(t,J=6.0Hz,1H),3.44(q,J=6.3Hz,2H),3.38–3.33(m,2H),3.31–3.24(m,2H),3.10(t,J=6.3Hz,2H)。
MS m/z(ESI):464[M+H] +
EXAMPLE 13 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonylimidazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 13
First step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (2-carbonylimidazol-1-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 13b
Compound Int-1 (150 mg,0.4 mmol) was dissolved in acetonitrile (2 mL), and compound 1- (2-aminoethyl) imidazolidin-2-one 13a (78 mg,0.6 mmol) and triethylamine (121 mg,1.2 mmol) were added successively under stirring at room temperature, and the reaction mixture was stirred at room temperature for 10 minutes to precipitate insoluble materials. Filtration gave the title compound 13b (70 mg,0.15 mmol) as a white powder of 96% purity. The filtrate was concentrated to give the title compound 13b (120 mg,0.26 mmol) in 63% purity, 78% overall yield.
MS m/z(ESI):454[M+H] +
Second step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonylimidazolidin-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 13
Compound 13b (120 mg,0.26 mmol) having a purity of 63% was dissolved in tetrahydrofuran (5 mL), and a 2N aqueous solution of sodium hydroxide (2 mL) was added at room temperature. After stirring at room temperature for 1 hour, the mixture was quenched with 3N aqueous hydrochloric acid in an ice bath, and the pH was adjusted to 8. Then extracted with ethyl acetate (3X 30 mL) and the organic phases combined. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the crude product. The crude product was purified twice by plate chromatography to give the title compound 13 (15 mg,0.03 mmol) in 21% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.89(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.74(ddd,J=9.0,4.2,2.7Hz,1H),6.33(s,1H),6.24(t,J=5.8Hz,1H),3.39–3.30(m,4H),3.28–3.18(m,4H)。
MS m/z(ESI):428[M+H] +
EXAMPLE 14 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-pyrrolidin-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-cimetidine 14
First step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (2-pyrrolidin-1-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 14b
Compound Int-1 (370 mg,1.0 mmol) was dissolved in THF (10 mL) and compound 1- (2-aminoethyl) pyrrolidin-2-one hydrochloride 14a (160 mg,1.0 mmol) was added followed by saturated NaHCO 3 The aqueous solution (1 mL) was stirred at room temperature for 3 hours. Saturated ammonium chloride solution and a small amount of water were added, and extraction was performed twice with ethyl acetate. The organic phase was washed once with saturated brine, dried over anhydrous sodium sulfate, filtered and dried by spinning. The crude product was purified by plate chromatography (PE: ea=1:2) to give the title compound 14b (135 mg,0.3 mmol) in 30% yield.
MS m/z(ESI):453[M+H] +
Second step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-pyrrolidone-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-methylimidine 14
Compound 14b (135 mg,0.3 mmol) was dissolved in THF (5 mL), 2M aqueous NaOH (1 mL) was added, and the reaction was complete after stirring at room temperature for 10 min. The reaction was quenched by addition of saturated ammonium chloride solution and extracted twice with ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, filtered and spun-dried. The crude pale yellow solid was slurried with a small amount of ethyl acetate and petroleum ether and filtered to give the title compound 14 (50 mg,0.12 mmol) in 39% yield as a white solid.
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.89(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.9,4.2,2.7Hz,1H),6.25(t,J=5.7Hz,1H),3.43–3.32(m,6H),2.17(t,J=8.1Hz,2H),1.95–1.83(m,2H)。
MS m/z(ESI):427[M+H] +
EXAMPLE 15 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-methyl-2-oxo-1, 3, 2-diazaphospholan-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 15
First step Synthesis of 1-benzyl-2-methyl-2-oxo-1, 3, 2-diazaphospholane 15a
Compound N 1 Benzyl-1, 2-ethylenediamine 12a (500 mg,3.3 mmol) and triethylamine (840 mg,8.3 mmol) were dissolved in dichloromethane (20 mL), and a dichloromethane solution (5 mL) of dichlorophosphine (420 mg,3.2 mmol) was slowly added dropwise at 0deg.C and gradually warmed to room temperature overnight. Adding saturated ammonium chloride solution for quenching, washing the organic phase with saturated common salt water once, drying with anhydrous sodium sulfate, filtering, and concentrating to obtain crude product. The crude product was purified by column chromatography to give the title compound 15a (220 mg,1.0 mmol) in 31% yield. MS m/z (ESI) 211[ M+H ] ] +
The subsequent steps were carried out in a similar manner to example 12, so as to obtain compound 15.
MS m/z(ESI):462[M+H] +
EXAMPLE 16 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2-oxo-1, 3, 2-dioxaphosphorin-2-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 16
First step Synthesis of 2-chloro-2-oxo-1, 3, 2-dioxaphosphorinane 16b
Compound 1, 3-propanediol 16a (270 mg,3.54 mmol) and triethylamine (719 mg,7.08 mmol) were dissolved in dichloromethane (10 mL), cooled to 0℃and phosphorus oxychloride (543 mg,3.54 mmol) was added dropwise to the solution. The solution was stirred at room temperature overnight and used directly in the next step.
Second step Synthesis of (2- ((2-oxo-1, 3, 2-dioxaphosphorin-2-yl) amino) ethyl) carbamic acid (9H-fluoren-9-yl) methyl ester 16c
Under nitrogen, N-fluorenylmethoxycarbonyl ethylenediamine hydrochloride (300 mg,0.9 mmol) and triethylamine (190 mg,1.8 mmol) were dissolved in dichloromethane (10 mL), cooled to 0℃and a dichloromethane solution of the compound (147 mg,3.54 mmol) in the next step was added dropwise. Stirring was carried out at room temperature for 1h. The reaction solution was washed with water, saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 16c (240 mg,0.6 mmol) in 63% yield.
MS m/z(ESI):403[M+H] +
Third step Synthesis of 2- ((2-aminoethyl) amino) -2-oxo-1, 3, 2-dioxaphosphorinane 16d
Compound 16c (100 mg,0.25 mmol) was dissolved in tetrahydrofuran (10 mL), TBAF (650 mg,2.5 mmol) was added, and the mixture was stirred at room temperature for 1h. After the reaction is completed, the reaction mixture is directly used for the next step.
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((2-oxo-1, 3, 2-dioxaphosphorin-2-yl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 16e
To the reaction solution of the compound 16d in the previous step was added the compound Int-1 (93 mg,0.25 mmol), and the mixture was stirred at room temperature for 1h. Ethyl acetate (20 mL) was added to dilute, followed by saturated ammonium chloride solution and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 16e (50 mg,0.1 mmol) in 40% yield.
MS m/z(ESI):505[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2-oxo-1, 3, 2-dioxaphosphorin-2-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 16
Compound 16e (50 mg,0.1 mmol) was dissolved in methanol (20 mL), and potassium carbonate (97 mg,0.7 mmol) was added thereto and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration and then purification by plate analysis gave the title compound 16 (40 mg,0.08 mmol) in 84% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,1H),8.89(s,1H),7.22-7.15(m,1H),7.13-7.07(m,1H),6.81-6.72(m,1H),6.25-6.16(m,1H),5.36-5.25(m,1H),4.38-4.13(m,4H),3.30-3.20(m,2H),3.01-2.87(m,2H),1.93-1.75(m,2H)。
MS m/z(ESI):479[M+H] +
EXAMPLE 17 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- ((dimethylphosphoryl) amino) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 17
First step Synthesis of N- (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) -P, P-dimethylphosphinamide 17b
Compound Int-2 (0.125 g,0.30 mmol) was dissolved in acetonitrile (10 mL), triethylamine (0.10 g,1.0 mmol) was added, and a solution of dimethylphosphoryl chloride 17a (0.12 g,1.1 mmol) in acetonitrile (3 mL) was added dropwise with cooling in an ice-water bath. Gradually warmed to room temperature and stirred overnight. The crude product was purified by plate chromatography (DCM: meoh=20:1) by spin-drying to give the title compound 17b (65 mg,0.14 mmol) in 47% yield.
MS m/z(ESI):461[M+H] +
Second step Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- ((dimethylphosphoryl) amino) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 17
Compound 17b (110 mg,0.24 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (230 mg,1.67 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration and then purification by plate analysis gave the title compound 17 (45 mg,0.10 mmol) in 43% yield.
MS m/z(ESI):435[M+H] +
EXAMPLE 18 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((2- ((diethylphosphoryl) amino) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 18
Prepared in the same manner as in example 17, except for using diethylphosphoryl chloride as a starting material in place of 17a.
MS m/z(ESI):463[M+H] +
EXAMPLE 19 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((1-oxo-2, 5-dihydro-phosphol-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 19
First step of synthesizing 1-hydroxy-1-oxo-2, 5-dihydro-phosphole 19b
The compound ammonium hypophosphite (3.0 g,36.13 mmol) was slowly added to the compound N, O-bis (trimethylsilyl) acetamide (22.05 g,108.4 mmol) under an ice-water bath followed by the compound 1, 4-dichloro-2-butene 19a (4.97 g,39.75 mmol) under nitrogen protection for 7 days at 110 ℃. After cooling, 1% dilute hydrochloric acid was added, extracted with ethyl acetate, the aqueous phase was concentrated, dichloromethane was added, and sonication was performed for 1h. Drying over anhydrous sodium sulfate, filtration, and concentration gave the crude title compound 19b (500 mg,4.2 mmol) in 12% yield.
MS m/z(ESI):119[M+H] +
Second step of synthesizing 1-chloro-1-oxo-2, 5-dihydro-phosphole 19c
Compound 19b (500 mg,4.2 mmol) was dissolved in dichloromethane (20 mL), oxalyl chloride (1.07 g,8.47 mmol) was added dropwise under ice-water bath, and stirred overnight at room temperature. Direct concentration, toluene addition, continued concentration, and repeated three times gave the title compound 19c (500 mg,3.7 mmol) in 88% yield.
Third step Synthesis of (2- ((1-oxo-2, 5-dihydro-phosphol-1-yl) amino) ethyl) formate (9H-fluoren-9-yl) methyl ester 19d
Compound N-fluorenylmethoxycarbonyl ethylenediamine hydrochloride (300 mg,0.9 mmol) and triethylamine (190 mg,1.8 mmol) were dissolved in methylene chloride (10 mL), the resulting solution was cooled to 0℃and a methylene chloride solution (3 mL) of compound 19c (122 mg,0.9 mmol) was added dropwise thereto under nitrogen atmosphere, followed by stirring at room temperature for 1h. The reaction solution was washed with water, and the organic phase was dried over saturated brine, filtered and concentrated, followed by column chromatography purification to give the title compound 19d (229 mg,0.6 mmol) in 67% yield.
MS m/z(ESI):383[M+H] +
Fourth step Synthesis of 1-oxo-1- ((2-aminoethyl) amino) -2, 5-dihydro-phosphole 19e
Compound 19d (229 mg,0.6 mmol) was dissolved in tetrahydrofuran (10 mL), TBAF (1.3 g,5.1 mmol) was added and stirred at room temperature for 1h. After the completion of the reaction, ethyl acetate was added, and the mixture was washed three times with a saturated ammonium chloride solution, and the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 19e (86 mg,0.54 mmol) in 90% yield.
MS m/z(ESI):161[M+H] +
Fifth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((1-oxo-2, 5-dihydro-phosphol-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 19f
Compound 19e (40 mg,0.25 mmol) and compound Int-1 (93 mg,0.25 mmol) were dissolved in tetrahydrofuran (10 mL) and stirred at room temperature for 1h. Ethyl acetate and water were added, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated then purified by plate chromatography to give the title compound 19f (77 mg,0.16 mmol) in 64% yield.
MS m/z(ESI):485[M+H] +
Sixth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((1-oxo-2, 5-dihydro-phosphol-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 19
Compound 19f (77 mg,0.16 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (155 mg,1.12 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 19 (41 mg,0.09 mmol) in 56% yield.
MS m/z(ESI):459[M+H] +
EXAMPLE 20 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((1-oxo-2, 5-phosphacycle-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 20
First step Synthesis of 1-oxo-1- ((2-aminoethyl) amino) -2, 5-phospholane 20a
Compound 19e (40 mg,0.25 mmol) was dissolved in methanol (10 mL), palladium on carbon (10% Pd,10 mg) was added and stirred overnight at room temperature under hydrogen atmosphere. Filtration and concentration gave the title compound 20a (30 mg,0.19 mmol) in 74% yield.
MS m/z(ESI):163[M+H] +
Second step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((1-oxo-2, 5-phospholanyl-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 20b
Compound 20a (30 mg,0.19 mmol) and compound Int-1 (70 mg,0.19 mmol) were dissolved in tetrahydrofuran (10 mL) and stirred at room temperature for 1h. Ethyl acetate and water were added, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated then purified by plate chromatography to give the title compound 20b (68 mg,0.14 mmol) in 74% yield.
MS m/z(ESI):487[M+H] +
Third step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((1-oxo-2, 5-phosphacycle-1-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 20
Compound 20b (68 mg,0.14 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (155 mg,1.12 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 20 (37 mg,0.08 mmol) in 57% yield.
MS m/z(ESI):461[M+H] +
EXAMPLE 21 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonylpyrrolidin-2-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 21
First step of synthesizing 5-carbonyl pyrrolidine-2-formaldehyde 21b
Under nitrogen, compound 5- (hydroxymethyl) pyrrolidin-2-one 21a (1.15 g,10.0 mmol) was dissolved in dichloromethane (50 mL) and DMP (5.1 g,12.0 mmol) was added in portions under an ice-water bath and the reaction was allowed to warm to room temperature for 1 hour. The filtrate was filtered, washed with a saturated sodium sulfite solution, dried over anhydrous sodium sulfate, filtered, concentrated and purified by column chromatography to give the title compound 21b (0.8 g,7.1 mmol) in 71% yield.
MS m/z(ESI):114[M+H] +
Second step of synthesizing 2- (5-carbonyl pyrrolidin-2-yl) acetaldehyde 21c
Under nitrogen, methoxymethyl triphenylphosphine chloride (0.7 g,2.0 mmol) was dissolved in anhydrous tetrahydrofuran (30 mL), cooled to-78deg.C, and n-BuLi (2.5M, 1.7mL,4.2 mmol) was added dropwise. After the addition was completed, the mixture was gradually warmed to room temperature and stirred for 1 hour, cooled to-78℃and an anhydrous tetrahydrofuran solution (5 mL) of compound 21b (0.22 g,2.0 mmol) was slowly added. After the addition was completed, the temperature was naturally raised and stirred overnight. After quenching the saturated ammonium chloride solution, 1mL of concentrated hydrochloric acid was added and refluxed at 70℃for 1 hour. Ethyl acetate was added for extraction, and the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the crude title compound 21c (0.20 g,1.57 mmol) in 78% yield.
MS m/z(ESI):128[M+H] +
Third step 5- (2-aminoethyl) pyrrolidin-2-one 21d
Compound 21c (0.20 g,1.57 mmol) was dissolved in methanol and ammonium acetate (0.22 g,3.0 mmol) was added. After stirring for 1 hour, sodium cyanoborohydride (0.24 g,4.0 mmol) was added and stirred overnight at room temperature. Adding saturated NaHCO 3 The solution was quenched, concentrated, ethyl acetate and water were added, the extract was dried over anhydrous sodium sulfate, filtered, and the crude product obtained after concentration was purified by column chromatography to give the title compound 21d (0.13 g,1.0 mmol) in 60% yield.
MS m/z(ESI):129[M+H] +
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (5-carbonylpyrrolidin-2-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 21e
Compound 21d (0.13 g,1.0 mmol) and Int-1 (0.372 g,1.0 mmol) were dissolved in tetrahydrofuran (5 mL) and reacted at room temperature for 1h. After concentration, the product was purified by plate chromatography to give the title compound 21e (350 mg,0.77 mmol) in 77% yield
MS m/z(ESI):453[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonylpyrrolidin-2-yl) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 21
Compound 21e (135 mg,0.3 mmol) was dissolved in methanol (5 mL), anhydrous potassium carbonate (330 mg,2.4 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 21 (60 mg,0.14 mmol) in 47% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,1H),8.09(s,1H),7.76(s,1H),7.10–7.20(m,2H),6.73–6.77(m,1H),6.19–6.22(m,1H),3.45–3.51(m,1H),3.17–3.32(m,2H),2.07–2.16(m,3H),1.57–1.74(m,3H)。
MS m/z(ESI):427[M+H] +
EXAMPLE 22 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonylpyrrolidin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 22
Prepared in the same manner as in example 21 except for using 4- (hydroxymethyl) pyrrolidin-2-one 22a as a raw material instead of 21a.
MS m/z(ESI):427[M+H] +
EXAMPLE 23 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonyloxazolidin-5-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 23
Prepared in the same manner as in example 21 except for using 5- (hydroxymethyl) oxazolidin-2-one 23a as a starting material instead of 21a.
MS m/z(ESI):429[M+H] +
EXAMPLE 24 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonyloxazolidine-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 24
Prepared in the same manner as in example 21 except for using 4- (hydroxymethyl) oxazolidin-2-one 24a as a starting material instead of 21a.
MS m/z(ESI):429[M+H] +
EXAMPLE 25 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (3-methyl-2-carbonyloxazolid in-5-yl) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 25
First step Synthesis of 5- (((tert-butyldimethylsilyl) oxo) methyl) oxazolidin-2-one 25a
Compound 5- (hydroxymethyl) oxazolidin-2-one 23a (2.34 g,20.0 mmol), imidazole (1.7 g,25 mmol) was dissolved in acetonitrile (30 mL), ice-cooled to 0deg.C, and t-butyldimethylchlorosilane (3.3 g,22.0 mmol) was then added. The reaction was stirred at 0 ℃ for 0.5 hours, then allowed to warm to room temperature and stirred overnight. Saturated ammonium chloride solution and ethyl acetate were added, extraction was performed, the organic phase was washed three times with saturated ammonium chloride solution, dried over anhydrous sodium sulfate, filtered, and the crude product obtained after concentration was purified by column chromatography to obtain the title compound 25a (3.7 g,18 mmol) in 80% yield.
Second step Synthesis of 5- (((tert-butyldimethylsilyl) oxo) methyl) -3-methyl oxazolidin-2-one 25b
Compound 25a (3.7 g,18 mmol) was dissolved in DMF (40 mL), cooled to 0deg.C, and NaH (1.08 g,27mmol,60% wt) was added. The reaction was stirred at 0 ℃ for 0.5 hours, then warmed to room temperature and stirred for half an hour. Methyl iodide (3.8 g,27 mmol) was added and stirred overnight at room temperature. Saturated aqueous ammonium chloride and ethyl acetate were added, extraction was performed, the organic phase was washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 25b (3.0 g,12.2 mmol) in 68% yield.
MS m/z(ESI):246[M+H] +
Third step of synthesizing 5- (hydroxymethyl) -3-methyl oxazolidin-2-one 25c
Compound 25b (3.0 g,12.2 mmol) was dissolved in THF (20 mL), tetra-n-butyl ammonium fluoride (30 g,114 mmol) was added, after stirring for 2 hours, water and ethyl acetate were added, the organic phase was extracted, washed with saturated ammonium chloride and brine, dried over anhydrous sodium sulfate, filtered, and the resulting crude product was concentrated to give the title compound 25c (0.8 g,6.1 mmol) in 50% yield.
The subsequent steps were conducted in the same manner as in example 21 to obtain compound 25.
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.93(s,1H),7.16–7.21(t,J=8.8Hz,1H),7.11–7.13(dd,J=6.0,2.8,1H),6.72–6.77(m,1H),6.33–6.35(t,J=6.0Hz,1H),5.76(s,1H),4.42–4.48(m,1H),3.58–3.62(t,J=8.4Hz,1H),3.26–3.32(m,1H),3.17–3.21(m,1H),2.73(s,3H),1.88–1.93(m,2H).
MS m/z(ESI):443[M+H] +
EXAMPLE 26 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (3-methyl-2-carbonyloxazolid in-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 26
Prepared in the same manner as in example 25 using 4- (hydroxymethyl) oxazolidin-2-one 24a as a starting material in place of 23a.
MS m/z(ESI):429[M+H] +
EXAMPLE 27 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (1-methyl-2-carbonylimidazol-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 27
First step Synthesis of 5-formyl-3-methyl-2-carbonyl imidazolidine-1-carboxylic acid benzyl ester 27b
Under nitrogen, compound 5-hydroxymethyl-3-methyl-2-carbonylimidazolidine-1-carboxylate 27a (2.0 g,7.6mmol, prepared by the method disclosed in patent application "WO 2015150995") was dissolved in dichloromethane (20 mL), DMP (4.8 g,11.4 mmol) was added in portions under an ice-water bath, and the reaction was allowed to warm to room temperature for 1 hour. The filtrate was filtered, washed with a saturated sodium sulfite solution, dried over anhydrous sodium sulfate, filtered, concentrated and purified by column chromatography to give the title compound 27b (1.2 g,4.6 mmol) in 60% yield.
MS m/z(ESI):265[M+H] +
Second step Synthesis of benzyl 3-methyl-2-carbonyl-5- (2-carboxyethyl) imidazolidine-1-carboxylate 27c
Under nitrogen, methoxymethyl triphenylphosphine chloride (1.44 g,4.2 mmol) was dissolved in anhydrous tetrahydrofuran (20 mL), cooled to-78deg.C, and n-BuLi (2.5M, 1.8mL,4.6 mmol) was added dropwise. After the addition was completed, the mixture was gradually warmed to room temperature and stirred for 1 hour, cooled to-78℃and a solution of compound 27b (1.0 g,3.8 mmol) in anhydrous tetrahydrofuran (5 mL) was slowly added. After the addition was completed, the temperature was naturally raised and stirred overnight. After quenching the saturated ammonium chloride solution, 2mL of concentrated hydrochloric acid was added and refluxed at 70 ℃ for 1 hour. Ethyl acetate was added for extraction, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give a crude product which was purified by column chromatography to give the title compound 27c (0.65 g,2.4 mmol) in 62% yield.
MS m/z(ESI):277[M+H] +
Third step of synthesizing 5- (2-aminoethyl) -3-methyl-2-carbonyl imidazolidine-1-carboxylic acid benzyl ester 27d
Compound 27c (600 mg,2.17 mmol) was dissolved in methanol and ammonium acetate (335 mg,4.34 mmol) was added. After stirring for 1 hour, sodium triacetoxyborohydride (1.15 g,5.43 mmol) was added and stirred overnight at room temperature. Adding saturated NaHCO 3 The solution was quenched, concentrated, ethyl acetate and water were added, the extract was dried over anhydrous sodium sulfate, filtered, and the crude product obtained after concentration was purified by column chromatography to give the title compound 27d (355 mg,1.28 mmol) in 59% yield.
MS m/z(ESI):278[M+H] +
Fourth step of synthesizing 4- (2-amino ethyl) -1-methylimidazolidin-2-one 27e
Compound 27d (350 mg,1.26 mmol) was dissolved in methanol (10 mL), palladium on carbon (10% Pd,40 mg) was added, and the mixture was stirred overnight at room temperature under hydrogen atmosphere. Filtration and concentration gave the title compound 27e (170 mg,1.19 mmol) in 94% yield.
MS m/z(ESI):144[M+H] +
Fifth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (1-methyl-2-carbonylimidazol-4-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 27f
Compound 27e (150 mg,1.05 mmol) and Int-1 (400 mg,1.08 mmol) were dissolved in tetrahydrofuran (5 mL) and reacted at room temperature for 1h. After concentration, the title compound 27f (350 mg,0.75 mmol) was obtained in 71% yield by analytical purification
MS m/z(ESI):468[M+H] +
Sixth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (1-methyl-2-carbonylimidazol-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 27
Compound 27f (150 mg,0.32 mmol) was dissolved in methanol (5 mL), anhydrous potassium carbonate (350 mg,2.53 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 27 (68 mg,0.15 mmol) in 48% yield.
MS m/z(ESI):442[M+H] +
EXAMPLE 28 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (3-methyl-2-carbonylimidazol-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 28
/>
First step Synthesis of 4-hydroxymethyl-3-methyl-2-carbonylimidazolidine-1-carboxylic acid benzyl ester 28b
The compound methyl 1-benzyloxycarbonyl-3-methyl-2-carbonylimidazolidine-4-carboxylate 28a (2.9 g,10.0mmol, prepared by the method disclosed in patent application "WO 2008119825") was dissolved in methanol (30 mL) and sodium borohydride (0.46 g,12.0 mmol) was added in portions under an ice-water bath. Stirring was continued for 2 hours after the addition was completed. Quench with water and stir at room temperature for 1 hour. Concentrated to smaller volume under reduced pressure, then extracted with water and ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 28b (2.5 g,9.5 mmol) in 95% yield.
MS m/z(ESI):265[M+H] +
The subsequent steps were conducted in the same manner as in example 27 to obtain a compound 28.
MS m/z(ESI):442[M+H] +
EXAMPLE 29 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazolidin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamide 29
First step of synthesizing 1, 1-Dioxo-1, 2, 5-thiadiazolidine-3-carboxylic acid methyl ester 29b
Compound 2, 3-Di-amino methyl propionate dihydrochloride 29a (1.0 g,5.23 mmol) and triethylamine (1.85 g,18.3 mmol) were dissolved in 1, 4-dioxane (20 mL), and sulfonamide (503 mg,5.23 mmol) was added, and the mixture was heated under reflux and stirred for 16h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with ethyl acetate, and the organic phase was combined, dried over saturated brine, filtered, concentrated, and the crude product was purified by column chromatography to give the title compound 29b (450 mg,2.5 mmol) in 48% yield.
MS m/z(ESI):181[M+H] +
The subsequent steps were conducted in the same manner as in example 28 to obtain compound 29.
MS m/z(ESI):464[M+H] +
EXAMPLE 30 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonyl-imidazolidin-4-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 30
First step of synthesizing 2-carbonyl imidazolidine-4-methyl formate 30a
Compound 2, 3-Di-amino methyl propionate dihydrochloride 29a (1.0 g,5.23 mmol) and triethylamine (1.17 g,11.5 mmol) were dissolved in THF (20 mL) and CDI (1.02 g,6.3 mmol) was added. Stirring overnight at room temperature, quenching with ammonium chloride, extracting the aqueous phase 3 times with EtOAc, washing the organic phase with dilute hydrochloric acid, drying over anhydrous sodium sulfate, filtering, concentrating to give the title compound 30a (520 mg,3.6 mmol) in 69% yield.
MS m/z(ESI):145[M+H] +
Subsequent to the procedure of example 28, the preparation of compound 30 was performed in the same manner.
MS m/z(ESI):428[M+H] +
EXAMPLE 31 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 31
First step Synthesis of N- (2, 4-dimethoxybenzyl) -2- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-3-yl) acetamide 31b
Compound 2- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-3-yl) acetic acid 31a (0.43 g,3.0 mmol) and 2, 4-dimethoxybenzylamine (0.55 g,3.3 mmol) were dissolved in DMF (5 mL), EDC (0.86 g,6.0 mmol) was added and reacted overnight at room temperature. The crude product was concentrated and purified by column chromatography to give the title compound 31b (490 mg,1.7 mmol) in 56% yield.
MS m/z(ESI):293[M+H] +
Second step Synthesis of 5- (2- ((2, 4-dimethoxybenzyl) amino) ethyl) -2, 4-dihydro-3H-1, 2, 4-triazol-3-one 31c
Compound 31b (460 mg,1.6 mmol) was dissolved in anhydrous THF (5 mL), and a 2M solution of borane in dimethyl sulfide (1.6 mL,3.2 mmol) was added dropwise with cooling in an ice-water bath and heated at reflux for 4h. Cooled to room temperature, quenched with a small amount of methanol, concentrated, and the crude product was purified by column chromatography to give the title compound 31c (360 mg,1.3 mmol) in 82% yield.
MS m/z(ESI):279[M+H] +
Third step of synthesizing 5- (2-amino ethyl) -2, 4-dihydro-3H-1, 2, 4-triazole-3-ketone trifluoroacetate 31d
Compound 31c (350 mg,1.3 mmol) was dissolved in DCM (5 mL), TFA (1 mL) was added dropwise and stirred at room temperature for 1h. Spin-drying afforded the crude title compound 31d, which was used directly in the next step.
MS m/z(ESI):129[M+H] +
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-3-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 31e
The crude compound 31d, compound Int-1 (284 mg,1.3 mmol) was dissolved in THF (5 mL), triethylamine (330 mg,3.3 mmol) was added and reacted at room temperature for 1h. The crude product was concentrated and purified by plate chromatography to give the title compound 31e (405 mg,0.9 mmol) in 69% yield in two steps.
MS m/z(ESI):453[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 31
Compound 31e (150 mg,0.33 mmol) was dissolved in methanol (5 mL), anhydrous potassium carbonate (366 mg,2.65 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 31 (85 mg,0.2 mmol) in 60% yield.
MS m/z(ESI):427[M+H] +
EXAMPLE 32 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1H-tetrazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 32
First step Synthesis of 1- (2-chloroethyl) -1, 4-dihydro-5H-tetrazol-5-one 32b
Compound 1-chloro-2-isocyanatoethane 32a (10 g,95.2 mmol) and a catalytic amount of boron trifluoride etherate were mixed and stirred at reflux overnight. The reaction solution was concentrated, quenched with methanol, and concentrated again to give a crude product, which was purified by column chromatography (PE: ea=1:1) to give the title compound 32b (10.5 g,70.9 mmol) in 74% yield.
MS m/z(ESI):149[M+H] +
Second step of Synthesis of 2- (2- (5-carbonyl-4, 5-dihydro-1H-tetrazol-1-yl) ethyl) isoindoline-1, 3-dione 32c
Compound 32b (2.0 g,13.5 mmol), potassium phthalimide (2.5 g,13.5 mmol), potassium carbonate (5.6 g,40.5 mmol) and a catalytic amount of potassium iodide were dissolved in acetonitrile (150 mL), stirred for 4 hours under reflux, cooled to room temperature, filtered, and the filtrate was concentrated to give a crude product, which was directly used for the next reaction.
MS m/z(ESI):260[M+H] +
Third step of synthesizing 1- (2-amino ethyl) -1, 4-dihydro-5H-tetrazol-5-one 32d
Compound 32c (3.0 g,11.5 mmol) was dissolved in ethanol (100 mL) and 98% hydrazine hydrate (5 mL) was added, the reaction mixture was stirred at reflux for 5 hours, cooled to room temperature, the white insoluble material was filtered off, the filtrate was concentrated to give crude product, and the crude product was purified by column chromatography to give the title compound 32d (1.22 g,9.5 mmol) in 82% yield
MS m/z(ESI):130[M+H] +
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (5-carbonyl-4, 5-dihydro-1H-tetrazol-1-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 32e
Compound 32d (0.5 g,3.8 mmol) and compound Int-1 (1.43 g,3.8 mmol) were dissolved in tetrahydrofuran (50 mL) and the reaction mixture was stirred at room temperature for 1 hour. The reaction solution was concentrated to give a crude product, which was purified by column chromatography to give the title compound 32e (0.95 g,2.1 mmol) in 55% yield. MS m/z (ESI) 454[ M+H ]] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1H-tetrazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 32
Compound 32e (150 mg,0.33 mmol) was dissolved in tetrahydrofuran (10 mL) and then 2N aqueous sodium hydroxide solution (4 mL) was added. The reaction mixture was stirred at room temperature for 1 hour, quenched with 3N aqueous hydrochloric acid, pH was adjusted to 8, extracted three times with ethyl acetate, the organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated to give crude product, which was purified by plate chromatography to give the title compound 32 (45 mg,0.11 mmol) in 31% yield.
MS m/z(ESI):428[M+H] +
EXAMPLE 33 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-oxo-1, 6-dihydropyridin-2-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 33
First step Synthesis of 2- (6-methoxypyridin-2-yl) acetonitrile 33b
N-butyllithium (2.5M, 8.0mL,20.0 mmol) was added dropwise to a solution of acetonitrile (1.1 mL,21.2mmol, molecular sieve dried) in anhydrous tetrahydrofuran (20 mL) under nitrogen at-78deg.C, and reacted at that temperature for 30min. A solution of the compound 2-bromo-6-methoxypyridine 33a (1 g,5.32 mmol) in anhydrous tetrahydrofuran (5 mL) was then slowly added dropwise. After the dripping is finished, the temperature is gradually raised to the room temperature, and the mixture is stirred for 4 hours. Pouring into ice water, extracting with ethyl acetate three times, washing the organic phase with saturated saline, drying over anhydrous sodium sulfate, filtering, concentrating, and purifying by column chromatography to give the title compound 33b (470 mg,3.2 mmol) in 60% yield. MS m/z (ESI): 149[ M+H ]] +
Second step Synthesis of 2- (6-methoxypyridin-2-yl) ethyl-1-amine 33c
Compound 33b (474 mg,3.2 mmol) was dissolved in methanol (10 mL), and sodium borohydride (122 mg,3.2 mmol) was added in portions while cobalt chloride hexahydrate (71 mg,0.3 mmol) was added in an ice-water bath. Gradually heating to room temperature and stirring for 4h. The mixture was quenched with saturated ammonium chloride solution, extracted three times with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated, followed by column chromatography purification to give the title compound 33c (426 mg,2.8 mmol) in 88% yield.
MS m/z(ESI):153[M+H] +
Third step of synthesizing 6- (2-aminoethyl) pyridin-2 (1H) one 33d
Compound 33c (426 mg,2.8 mmol) was dissolved in hydrogen bromide in methanol (. About.10% HBr,25 mL) and heated at reflux for 2h. Spin-drying, adding ethyl acetate and saturated sodium bicarbonate solution, extracting three times, combining the organic phases, washing with saturated saline, drying over anhydrous sodium sulfate, filtering, concentrating to give the title compound 33d (110 mg,0.8 mmol), yield 29%.
MS m/z(ESI):139[M+H] +
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- (6-oxo-1, 6-dihydropyridin-2-yl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 33e
Compound 33d (110 mg,0.8 mmol) was dissolved in tetrahydrofuran (10 mL), and compound Int-1 (294 mg,0.8 mmol) was added and stirred at room temperature for 1h. After concentration, the crude product was purified by plate chromatography to give the title compound 33e (250 mg,0.54 mmol) in 67% yield.
MS m/z(ESI):463[M+H] +
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-oxo-1, 6-dihydropyridin-2-yl) ethyl) amino) -1,2, 5-oxadiazole-3-formamidine 33
Compound 33e (250 mg,0.54 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (522 mg,3.78 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration, and then purification by plate analysis gave the title compound 33 (122 mg,0.28 mmol) in 49% yield.
MS m/z(ESI):437[M+H] +
EXAMPLE 34 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-oxo-1, 6-dihydropyridin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 34
Prepared by a method similar to that of example 33 using 5-bromo-2-methoxypyridine as a starting material, replacing 33a. 1 H NMR(400MHz,Methanol-d 4 )δ7.54(dd,J=9.4,2.6Hz,1H),7.24(d,J=2.5Hz,1H),7.14–6.98(m,2H),6.85–6.74(m,1H),6.51(d,J=9.3Hz,1H),3.46(t,J=6.9Hz,2H),2.73(t,J=6.8Hz,2H).
MS m/z(ESI):437[M+H] +
EXAMPLE 35 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (6-carbonyl-1, 6-dihydropyridazin-3-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 35
First step Synthesis of 6- (hydroxymethyl) pyridazin-3 (2H) -one 35b
Compound 35a (1.0 g,6.5 mmol) was dissolved in anhydrous tetrahydrofuran (20 mL), DIBAL-H (1.0M, 13mL,13.0 mmol) was added dropwise under ice-water bath, and the mixture was gradually warmed to room temperature and stirred for 1H. Quenching with saturated ammonium chloride, extracting with ethyl acetate three times, washing with saturated saline, drying with anhydrous sodium sulfate, filtering, concentrating, and purifying by column chromatography to give the title compound 35b (510 mg,4.0 mmol), yield 62%. MS m/z (ESI): 127[ M+H)] +
The subsequent steps were conducted in the same manner as in example 21 to obtain compound 35.
MS m/z(ESI):438[M+H] +
EXAMPLE 36 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) amino) -1,2, 5-oxadiazol-3-carboxamidine 36
First step of Synthesis of tert-butyl (2-cyanoethyl) carbamate 36b
Compound 3-aminopropionitrile 36a (1.0 g,14.3 mmol) and triethylamine (1.59 g,15.7 mmol) were dissolved in DCM (15 mL), and di-tert-butyl dicarbonate (3.43 g,15.7 mmol) was added to the solution in ice-water bath and stirred overnight at room temperature. The reaction mixture was washed with 0.5M hydrochloric acid (2X 20 mL), saturated sodium hydrogencarbonate solution (20 mL) and saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 36b (2.4 g,14.3 mmol) in 100% yield.
MS m/z(ESI):193[M+Na] +
Second step of Synthesis of (3- (hydroxyamino) -3-carbamimidopropyl) carbamic acid tert-butyl ester 36c
Compound 36b (500 mg,2.9 mmol) was dissolved in ethanol (20 mL), hydroxylamine hydrochloride (409 mg,5.9 mmol) was added, sodium ethoxide (600 mg,8.8 mmol) was added in portions, and heated at 60℃overnight. After cooling, the mixture was concentrated, water and ethyl acetate were added, the aqueous phase was extracted 3 times with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude title compound 36c (480 mg,2.4 mmol) in 80% yield.
1 H NMR(400MHz,DMSO-d 6 )δ8.81(s,1H),6.64(t,J=4.8Hz,1H),5.35(s,2H),3.08(q,J=6.9Hz,2H),2.09(t,J=7.5Hz,2H),1.37(s,9H)。
MS m/z(ESI):204[M+H] +
Third step of synthesizing (2- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) carbamic acid tert-butyl ester 36d
Compound 36c (450 mg,2.2 mmol) was dissolved in ethanol (8 mL), CDI (710 mg,4.4 mmol) and sodium ethoxide (204 mg,4.4 mmol) were added and heated at 60℃overnight. After cooling, concentrating, adding water and ethyl acetate, extracting the aqueous phase with ethyl acetate for 3 times, combining the organic phases, drying over anhydrous sodium sulfate, filtering, concentrating to obtain crude product, and purifying by column chromatography to obtain the title compound 36d (270 mg,1.2 mmol), yield 53%.
MS m/z(ESI):230[M+H] +
Fourth step of synthesizing 3- (2-amino ethyl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride 36e
Compound 36d (270 mg,1.2 mmol) was dissolved in 2M HCl in ethyl acetate (10 mL) and stirred at room temperature overnight. The title compound 36e was concentrated and used directly in the next step.
Fifth step of synthesizing N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4-nitro-1, 2, 5-oxadiazole-3-formamidine 36f
Compound Int-1 (186 mg,0.5 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (553 mg,4.0 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration and concentration gave the crude title compound 36f (150 mg,0.4 mmol) in 87% yield.
Sixth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) amino) -1,2, 5-oxadiazol-3-formamidine 36
Compounds 36e (66 mg,0.4 mmol) and 36f (150 mg,0.4 mmol) were dissolved in THF (5 mL), triethylamine (80 mg,0.8 mmol) was added and stirred at room temperature for 1h. After concentration, the crude product was purified by chromatography to give the title compound 36 (103 mg,0.24 mmol) in 60% yield.
MS m/z(ESI):428[M+H] +
EXAMPLE 37 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (morpholine-4-sulfonylamino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 37
Prepared in the same manner as in example 17 using morpholine-4-sulfonyl chloride as a starting material instead of 17a. 1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.90(s,1H),7.49(t,J=5.8Hz,1H),7.18(t,J=8.8Hz,1H),7.10(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.9,4.2,2.7Hz,1H),6.26(t,J=6.1Hz,1H),3.64–3.57(m,4H),3.37–3.29(m,2H),3.12(q,J=6.2Hz,2H),3.04–2.96(m,4H)。
MS m/z(ESI):508[M+H] +
EXAMPLE 38 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (S-methylsulfonylimine) ethyl) mercapto) -1,2, 5-oxadiazole-3-carboxamidine 38
/>
First step Synthesis of (2- (benzyloxy) ethyl) (methyl) thioether 38b
The compound ((2-bromoethoxy) methyl) benzene 38a (2.15 g,1.0 mmol) was dissolved in DMF (30 mL) and sodium methyl mercaptide (0.11 g,1.5 mmol) was added. The mixture was stirred at room temperature for 1 hour, the reaction mixture was concentrated under reduced pressure, ethyl acetate and water were added, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 38b (1.40 g,0.8 mmol) in 80% yield.
MS m/z(ESI):183[M+H] +
Second step of Synthesis of ((2- (methylsulfonyl) ethoxy) methyl) benzene 38c
Compound 38b (0.18 g,1.0 mmol) was dissolved in DCM (20 mL), cooled to-40℃and a solution of mCPBA (0.21 g,1.2 mmol) in DCM (5 mL) was added dropwise. After the addition was completed, the mixture was slowly warmed to room temperature and stirred at room temperature for 1 hour. The reaction was quenched by the addition of sodium sulfite solution, and the organic phase was washed three times with saturated sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 38c (118 mg,0.6 mmol) in 60% yield.
MS m/z(ESI):199[M+H] +
Third step ((2- ((S-methyl) (2, 2-trifluoroacetyl) sulfoxide imino) ethoxy) methyl) benzene 38d
Compound 38c (198 mg,1.0 mmol) was dissolved in DCM (20 mL), trifluoroacetamide (226 mg,2.0 mmol), magnesium oxide (201 mg,5.0 mmol), rhodium acetate (22 mg,0.05 mmol) and iodobenzene acetate (640 mg,2.0 mmol) were added. Stirring overnight at room temperature, filtering, concentrating the mother liquor to dryness, adding water and ethyl acetate to separate the organic phases, combining, drying with anhydrous sodium sulfate, filtering, purifying the concentrated crude product by column chromatography to obtain the title compound 38d (150 mg,0.48 mmol) in 48% yield.
MS m/z(ESI):310[M+H] +
Fourth step Synthesis of 2- ((S-methyl) (2, 2-trifluoroacetyl) sulfoxide imino) ethanol 38e
Compound 38d (0.3 g,1.0 mmol) was dissolved in MeOH (10 mL), pd/C (10% Pd,50 mg) was added and stirred overnight under 6MPa of hydrogen. Filtration and concentration gave the crude title compound 38e (0.2 g,1.0 mmol) in 100% yield.
MS m/z(ESI):220[M+H] +
Fifth step of synthesizing 2- (S-methyl-N- (2, 2-trifluoro acetyl) sulfoxide imino) ethyl methane sulfonate 38f
Compound 38e (0.15 g,0.7 mmol), triethylamine (0.15 g,0.7 mmol) was dissolved in dichloromethane (10 mL) and a solution of methanesulfonyl chloride (125 mg,1.1 mmol) in dichloromethane (2 mL) was slowly added dropwise under an ice-water bath. After the addition, the ice bath was removed and stirred at room temperature for 2 hours. The reaction solution was washed twice with saturated ammonium chloride solution, saturated sodium bicarbonate solution and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude title compound 38f, which was used directly in the next reaction.
Sixth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((S-methyl) (2, 2-trifluoroacetyl) sulfoxide imino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 38g
Compound Int-1 (0.19 g,0.5 mmol) was dissolved in DMF (8 mL) and sodium hydrosulfide (67 mg,0.6 mmol) was added. Stirred at room temperature for 1 hour, then compound 38f (0.15 g,0.5 mmol) and sodium carbonate (0.11 g,1.0 mmol) were added and the reaction was stirred at room temperature overnight. Diluting with water, extracting with ethyl acetate for 3 times, saturated brine, drying over anhydrous sodium sulfate, filtering, concentrating, and purifying by column chromatography to obtain 38g (0.12 g,0.2mmol, 40%) of the title compound.
MS m/z(ESI):560[M+H] +
Seventh step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (S-methyl sulfoxide imine) ethyl) mercapto) -1,2, 5-oxadiazole-3-formamidine 38
38g (0.25 g,0.45 mmol) of the compound was dissolved in THF (5 mL) and then a 2M NaOH solution (1.5 mL) was added. Stirring at room temperature for 1 hour. Saturated ammonium chloride solution was added, extraction was performed three times with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give crude product, which was purified by plate chromatography to give the title compound 38 (50 mg,0.11 mmol) in 26% yield.
MS m/z(ESI):438[M+H] +
EXAMPLE 39 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- (2- (S-methylsulfonylimino) ethoxy) -1,2, 5-oxadiazole-3-carboxamidine 39
First step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- (2- (methylthio) ethoxy) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 39a
Compound Int-1 (0.372 g,1.0 mmol) and 2- (methylthio) ethanol (0.27 g,3.0 mmol) were dissolved in THF (20 mL) and Cs was added 2 CO 3 (0.33 g,1.0 mmol) was stirred at 60℃for 4 hours. Saturated ammonium chloride solution was added, extraction was performed 3 times with ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give a crude product, which was purified by column chromatography to give the title compound 39a (0.21 g,0.5 mmol) in 50% yield.
Second step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- (2- (methylsulfonyl) ethoxy) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 39b
Compound 39a (0.21 g,0.5 mmol) was dissolved in DCM (10 mL), cooled to-40℃and a solution of mCPBA (0.21 g,1.2 mmol) in DCM (5 mL) was added dropwise. After the addition was completed, the mixture was slowly warmed to room temperature and stirred at room temperature for 1 hour. The reaction was quenched by addition of sodium sulfite solution, the organic phase was washed three times with saturated sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by plate chromatography to give the title compound 39b (85 mg,0.2 mmol) in 40% yield.
MS m/z(ESI):433[M+H] +
Third step of synthesizing 4- (3-bromo-4-fluorophenyl) -3- (4- (2- (S-methyl sulfoxide imino) ethoxy) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 39c
Compound 39b (85 mg,0.2 mmol) was dissolved in MeOH (2 mL) and ammonium carbamate (63 mg,0.8 mmol) and PhI (OAc) were added 2 (190 mg,0.6 mmol) was stirred at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure, and the title compound 39c (45 mg,0.1 mmol) was obtained by plate chromatography and purification in 51% yield. MS m/z (ESI): 448[ M+H ]] +
Fourth step of synthesizing N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- (2- (S-methyl sulfoxide imino) ethoxy) -1,2, 5-oxadiazole-3-formamidine 39
Compound 39c (45 mg,0.1 mmol) was dissolved in THF (5 mL), 2M NaOH solution (0.5 mL) was added and stirred at room temperature for 30min. Saturated ammonium chloride solution and ethyl acetate were added, the solution was separated, the aqueous phase was extracted twice with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the concentrated crude product was purified by plate chromatography to give the title compound 39 (24 mg,0.057 mmol) in 57% yield.
MS m/z(ESI):422[M+H] +
EXAMPLE 40 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 40
First step Synthesis of (2- ((4- (4- (3- (difluoromethyl) -4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) carbamic acid tert-butyl ester 40a
Compound Int-4 (300 mg,0.87 mmol) and N-t-butoxycarbonyl-1, 2-ethylenediamine (420 mg,2.6 mmol) were dissolved in THF (10 mL) and reacted at room temperature for 30min. To the residue was added ethyl acetate (15 mL), which was washed successively with 0.5M HCl (10 mL), water (10 mL) and saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 40a (400 mg,0.87 mmol), yield 100%.
MS m/z(ESI):457[M+H] +
Second step Synthesis of 3- (4- ((2-aminoethyl) amino) -1,2, 5-oxadiazol-3-yl) -4- (3- (difluoromethyl) -4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one 40b
Compound 40a (400 mg,0.87 mmol) was dissolved in 2M HCl ethyl acetate (6 mL) and reacted overnight at room temperature. The reaction solution was added to a saturated sodium hydrogencarbonate solution (50 mL), extracted with ethyl acetate (3X 20 mL), and the organic layers were combined, washed with water (15 mL) and saturated brine (15 mL) in this order, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 40b (250 mg,0.7 mmol) in 80% yield. MS m/z (ESI): 357[ M+H ]] +
Third step Synthesis of (N- (2- ((4- (3- (difluoromethyl) -4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) sulfamoyl) carbamic acid tert-butyl ester 40c
The compound chlorosulfonic acid isocyanate (0.24 g,1.7 mmol) was dissolved in anhydrous DCM (2 mL), and t-BuOH (0.16 mL,1.7 mmol) was added dropwise under ice-water bath cooling and reacted at room temperature for 1h for use (reaction solution A).
Compound 40b (250 mg,0.7 mmol) was suspended in anhydrous DCM (2 mL), TEA (0.47 mL,3.4 mmol) was added dropwise under ice-water cooling, and reaction solution A was slowly added dropwise, and gradually warmed to room temperature to react for 2h. Ethyl acetate (20 mL) was added for dilution, and the mixture was washed successively with 0.1M HCl (2X 10 mL) and saturated brine (2X 10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product, which was purified by column chromatography to give the title compound 40c (100 mg,0.19 mmol) in 27% yield.
MS m/z(ESI):536[M+H] +
Fourth step Synthesis of 4- (3- (difluoromethyl) -4-fluorophenyl) -3- (4- ((2- (sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 40d
Compound 40c (100 mg,0.19 mmol) was suspended in DCM (7 mL) and TFA (3.5 mL) was added and the reaction was allowed to settle and react at room temperature for 2h. Concentrated to dryness and purified by column chromatography to give the title compound 40d (60 mg,0.14 mmol) in 74% yield.
MS m/z(ESI):436[M+H] +
Fifth step Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 40
Compound 40d (60 mg,0.14 mmol) was dissolved in methanol (3 mL), and 2M aqueous NaOH solution (0.4 mL,0.8 mmol) was added dropwise and reacted at room temperature for 1.5h. 3M HCl was added dropwise to adjust pH to 7, methanol was distilled off under reduced pressure, water (5 mL), ethyl acetate extraction (2X 5 mL) was added, the organic layers were combined, washed with saturated brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 40 (55 mg,0.13 mmol) in 97% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.49(s,1H),8.95(s,1H),7.18(t,J=9.5Hz,1H),7.12(t,J=54.4Hz,1H),7.06–7.01(m,1H),6.96–6.90(m,1H),6.71(t,J=6.0Hz,1H),6.60(s,2H),6.25(t,J=6.1Hz,1H),3.39–3.35(m,2H),3.10(q,J=6.2Hz,2H)。
MS m/z(ESI):410[M+H] +
EXAMPLE 41 Synthesis of 4- ((2- (N-cyclopropylsulfonyl) -S-methylsulfonylimino) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 41
First step Synthesis of 4- (3- (difluoromethyl) -4-fluorophenyl) -3- (4- ((2- (methylsulfanyl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 41a
Compound Int-4 (6.0 g,17.5 mmol) and 2-methylmercaptoethylamine hydrochloride (1.9 g,21.0 mmol) were dissolved in acetonitrile (50 mL), triethylamine (2.65 g,26.2 mmol) was added and reacted at room temperature for 20min. Acetonitrile was distilled off under reduced pressure, ethyl acetate (50 mL) was added to the residue, which was dried over 0.5M HCl (25 mL) and saturated brine (30 mL) in this order, anhydrous sodium sulfate, filtered, and the resulting crude product was concentrated and purified by column chromatography (PE: ea=5:1) to give the title compound 41a (4.9 g,12.6 mmol), yield 72%.
MS m/z(ESI):388[M+H] +
Second step Synthesis of 4- (3- (difluoromethyl) -4-fluorophenyl) -3- (4- ((2-methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 41b
Compound 41a (1.0 g,2.6 mmol) was dissolved in DCM/MeOH (10:1, 15 mL) and MMPP (0.71 g,1.43 mmol) was added and reacted overnight at room temperature. Ethyl acetate (30 mL) was added to dilute the mixture, and the organic layer was washed with water (10 mL) and then with saturated brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 41b (0.73 g,1.8 mmol) in 70% yield.
MS m/z(ESI):404[M+H] +
Third step Synthesis of 4- (3- (difluoromethyl) -4-fluorophenyl) -3- (4- ((2- (S-methylsulfoxide imino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 41c
Compound 41b (0.7 g,1.7 mmol) was dissolved in CHCl 3 (20 mL) NaN was added 3 (0.22 g,3.4 mmol) and concentrated sulfuric acid (0.4 mL) were added dropwise under ice-water cooling. After the addition, the mixture was heated to 40℃overnight. Saturated sodium bicarbonate solution (30 mL) was added, extracted with ethyl acetate (3X 30 mL), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 41c (0.5 g,1.2 mmol) in 70% yield. MS m/z (ESI) 419[ M+H ]] +
Fourth step Synthesis of 4- (3- (difluoromethyl) -4-fluorophenyl) -3- (4- ((2- (N-cyclopropylsulfonyl) -S-methylsulfonyliminoethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 41d
Compound 41c (0.5 g,1.2 mmol) was dissolved in pyridine (10 mL), cyclopropylsulfonyl chloride (0.51 g,3.6 mmol) and DMAP (0.15 g,1.2 mmol) were added and reacted overnight at room temperature. Saturated sodium bicarbonate (15 mL) was added, extraction was performed with ethyl acetate (2X 30 mL), the organic phases were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to give the title compound 41d (0.13 g,0.25 mmol) in 21% yield.
MS m/z(ESI):523[M+H] +
Fifth step Synthesis of 4- ((2- (N-cyclopropylsulfonyl) -S-methylsulfonylimino) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 41
Compound 41d (0.1 g,0.19 mmol) was dissolved in THF/MeOH (1:1, 10 mL) and 2M aqueous NaOH (1 mL) was added and reacted at room temperature for 2h. Saturated ammonium chloride solution (20 mL) was added, extraction was performed with ethyl acetate (2X 25 mL), the organic layers were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by plate chromatography to give title compound 41 (75 mg,0.15 mmol) in 79% yield.
1 H NMR(400MHz,DMSO-d6)δ11.41(s,1H),8.96(s,1H),7.18(t,J=9.6Hz,1H),7.12(t,J=54.4Hz,1H),7.05–7.03(m,1H),6.95–6.92(m,1H),6.57(t,J=5.9Hz,1H),3.95-3.75(m,,4H),3.46(s,3H),2.60–2.67(m,1H),0.90-0.96(m,4H).
MS m/z(ESI):497[M+H] +
EXAMPLE 42 Synthesis of 4- ((2- (S-methyl sulfoxide cyanoimine) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 42
First step Synthesis of 4- (3-difluoromethyl) -4-fluorophenyl) -3- (4- ((2- (methylthiocyanatomine) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 42a
Compound 41a (250 mg,0.65 mmol) and cyanamide (52 mg,1.3 mmol) were dissolved in acetonitrile (5 mL), iodobenzene acetate (0.23 g,0.72 mmol) was added under ice-water bath cooling, and the reaction was continued for 3h. Concentration and column chromatography purification gave the title compound 42a (0.22 g,0.51 mmol) in 80% yield.
MS m/z(ESI):428[M+H] +
Second step Synthesis of 4- (3-difluoromethyl) -4-fluorophenyl) -3- (4- ((2- (methylsulfoxide-thiocyanatomine) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 42b
Compound 42a (0.22 g,0.51 mmol) was dissolved in absolute ethanol (5 mL) and K was added 2 CO 3 (0.21 g,1.5 mmol) and m-chloroperoxybenzoic acid (0.13 g,0.77 mmol) were reacted at room temperature for 2h. Ethyl acetate (20 mL) was added to dilute the mixture, and the organic phase was washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated, and the obtained crude product was purified by column chromatography to give the title compound 42b (0.12 g,0.27 mmol) in 55% yield.
MS m/z(ESI):444[M+H] +
Third step Synthesis of 4- ((2- (S-methyl sulfoxide cyanoimine) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-formamidine 42
Compound 42b (120 mg,0.27 mmol) was dissolved in THF (5 mL) and 2M aqueous NaOH (0.5 mL) was added and reacted at room temperature for 1h. The mixture was concentrated to pH 6-7 with 3M HCl solution, ethyl acetate (10 mL) was added to the mixture, the organic layer was washed with saturated brine (5 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 42 (102 mg,0.24 mmol) in 90% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.97(s,1H),7.18(t,J=9.6Hz,1H),7.12(t,J=54.4Hz,1H),7.08–7.02(m,1H),6.97–6.89(m,1H),6.61(t,J=5.9Hz,1H),3.94(t,J=6.3Hz,2H),3.79(q,J=6.0Hz,2H),3.54(s,3H)。
MS m/z(ESI):418[M+H] +
EXAMPLE 43 Synthesis of methyl (pyrrolidin-3-yl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester 43
First step Synthesis of tert-butyl 3- (((2- (((benzyloxy) carbonyl) amino) ethoxy) sulfonyl) (methyl) amino) pyrrolidine-1-carboxylate 43a
Benzyl (2-hydroxyethyl) carbamate (1.5 g,5.0 mmol) and pyridine (0.40 g,5.0 mmol) were dissolved in anhydrous DCM (10 mL), and compound 9b was slowly added dropwise with cooling in an ice-water bath and reacted overnight at room temperature. The reaction mixture was washed with 0.1M HCl (2X 10 mL) and then with saturated brine (15 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to give the title compound 43a (0.57 g,1.2 mmol) in 25% yield.
MS m/z(ESI):458[M+H] +
Second step Synthesis of tert-butyl 3- (((2-aminoethoxy) sulfonyl) (methyl) amino) pyrrolidine-1-carboxylate 43b
Compound 43a (0.57 g,1.2 mmol) was dissolved in MeOH (10 mL) and Pd/C (10% Pd,0.1 g) was added and reacted overnight at room temperature under a hydrogen atmosphere. Filtration and concentration gave the title compound 43b (0.35 g,1.1 mmol) in 90% yield.
MS m/z(ESI):324[M+H] +
Third step Synthesis of tert-butyl 3- (((2- ((4- (4- (3-bromo-4-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethoxy) sulfonyl) (methyl) amino) pyrrolidine-1-carboxylate 43c
Compound 43b (0.35 g,1.1 mmol) and compound Int-1 (0.32 g,0.85 mmol) were dissolved in acetonitrile (3 mL), reacted at room temperature for 30min, diluted with ethyl acetate (20 mL), washed with saturated brine (5 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography to give the title compound 43c (0.34 g) in 62% yield.
MS m/z(ESI):648[M+H] +
Fourth step Synthesis of 2- ((4- (4- (3-bromo-4-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl methyl (pyrrolidin-3-yl) sulfamate 43d
Compound 43c (0.34 g,0.52 mmol) was dissolved in DCM (10 mL) and TFA (5 mL) was added and reacted at room temperature for 2h. The residue was concentrated, dissolved with ethyl acetate and water, saturated sodium bicarbonate solution was added, the aqueous phase was extracted 2 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 43d (0.24 g,0.44 mmol) in 85% yield.
MS m/z(ESI):548[M+H] +
Fifth step Synthesis of methyl (pyrrolidin-3-yl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester 43
Compound 43d (0.24 mg,0.44 mmol) was dissolved in methanol (5 mL), and 2M aqueous NaOH (0.5 mL,1 mmol) was added dropwise and reacted at room temperature for 1.5h. 3M HCl was added dropwise to adjust pH to 6-7, meOH was distilled off under reduced pressure, water was added, extraction was performed with ethyl acetate (2X 10 mL), the organic layers were combined, washed with saturated brine (5 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product obtained was purified by column chromatography to give the title compound 43 (0.18 g,0.34 mmol) in 78% yield.
MS m/z(ESI):522[M+H] +
EXAMPLE 44 Synthesis of (2-methoxyethyl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester 44
/>
Prepared in the same manner as in example 43 except for using (2-methoxyethyl) sulfamoyl chloride 3b as a raw material instead of 9b.
MS m/z(ESI):497[M+H] +
EXAMPLE 45 Synthesis of (1-azetidin-3-yl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester 45
First step Synthesis of 2- ((((9H-fluoren-9-yl) methoxy) carbonyl) amino) ethyl) chlorosulfonate 45b
The compound (2-hydroxyethyl) carbamic acid ((9H-fluoren-9-yl) methyl) 45a (1.0 g,3.53 mmol) and pyridine (279 mg,3.53 mmol) were dissolved in toluene (20 mL), and a toluene solution (5 mL) of sulfonyl chloride (476 mg,3.53 mmol) was added dropwise at-50℃to-60℃and reacted at this temperature for 2 hours, followed by natural warming to room temperature and stirring continued for 2 hours. The filtrate was filtered, washed with water, 1M hydrochloric acid, saturated sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 45b (1.2 g,3.14 mmol) in 89% yield.
Second step Synthesis of tert-butyl 3- (((2- ((((9H-fluoren-9-yl) methoxy) carbonyl) amino) ethoxy) sulfonyl) amino) azetidine-1-carboxylate 45c
Compound 45b (1.2 g,3.14 mmol), triethylamine (636 mg,6.29 mmol) were dissolved in tetrahydrofuran (20 mL), followed by slow addition of tert-butyl 3-aminoazetidine-1-carboxylate (433 mg,2.51 mmol) and reaction at room temperature for 2h. Water was added thereto, extraction was performed three times with ethyl acetate, drying over anhydrous sodium sulfate, filtration and column chromatography after concentration gave the title compound 45c (650 mg,1.26 mmol) in 50% yield.
MS m/z(ESI):518[M+H] +
Third step of Synthesis of tert-butyl 3- (((2-aminoethoxy) sulfonyl) amino) azetidine-1-carboxylate 45d
Compound 45c (650 mg,1.26 mmol) was dissolved in tetrahydrofuran (10 mL), TBAF (650 mg,2.5 mmol) was added and reacted at room temperature for 1h. Ethyl acetate was added for dilution, and the mixture was washed three times with saturated ammonium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 45d (330 mg,1.12 mmol) in 89% yield.
Fourth step Synthesis of tert-butyl 3- (((2- ((4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethoxy) sulfonyl) amino) azetidine-1-carboxylate 45e
Compound 45d (330 mg,1.12 mmol) and compound Int-1 (418 mg,1.12 mmol) were dissolved in tetrahydrofuran (10 mL) and reacted at room temperature for 1h. Concentration and purification by plate chromatography gave the title compound 45e (425 mg,0.69 mmol) in 61% yield.
MS m/z(ESI):620[M+H] +
Fifth step Synthesis of (1-azetidin-3-yl) sulfamic acid (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester 45f
Compound 45e (425 mg,0.69 mmol) was dissolved in dichloromethane (20 mL), TFA (2 mL) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1h. The solution was basified with water and saturated sodium bicarbonate solution, extracted three times with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give compound 45f (320 mg,0.62 mmol) in 90% yield.
Sixth step Synthesis of (1-azetidin-3-yl) sulfamic acid (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) ester 45
Compound 45f (320 mg,0.62 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (600 mg,4.34 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 45 (92 mg,0.19 mmol) in 30% yield.
1 H NMR(400MHz,MeOD-d 4 )δ7.15(dd,J=6.0,2.7Hz,1H),7.05(t,J=8.7Hz,1H),6.89–6.80(m,1H),4.40(p,J=7.3Hz,1H),3.93(d,J=7.4Hz,4H),3.58(t,J=6.4Hz,2H),3.41(t,J=6.4Hz,2H)。
MS m/z(ESI):494[M+H] +
EXAMPLE 46 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- (methyl (2- (sulfamoyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 46
/>
Prepared in the same manner as in example 40, starting from tert-butyl (2- (methylamino) ethyl) carbamate and substituting Int-1 for Int-4.
1 H NMR(400MHz,DMSO-d 6 )δ11.14(s,1H),10.32(s,1H),9.22(s,1H),9.10(s,1H),8.05(dd,J=6.3,2.7Hz,1H),7.35(ddd,J=9.0,4.3,2.7Hz,1H),7.27(t,J=8.8Hz,1H),7.14(t,J=8.8Hz,1H),7.09(dd,J=6.0,2.7Hz,1H),6.69(ddd,J=8.9,4.1,2.8Hz,1H),6.60–6.54(m,2H),6.53(s,4H),3.40(t,J=6.8Hz,2H),3.29(t,J=6.8Hz,1H),3.05(q,J=6.5Hz,2H),2.99–2.92(m,5H),2.89(s,3H).MS m/z(ESI):452[M+H] +
EXAMPLE 47 Synthesis of 4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -N- (2- ((sulfamoyl) amino) ethyl) -1,2, 5-oxadiazole-3-carboxamide 47
/>
First step of Synthesis of (2- (sulfamido) ethyl) carbamic acid (9H-fluoren-9-yl) methyl ester 47n
Under the protection of nitrogen, the compound N-fluorenylmethoxycarbonyl ethylenediamine hydrochloride 47m (300 mg,0.9 mmol) and triethylamine (190 mg,1.8 mmol) were dissolved in methylene chloride (10 mL), and sulfamoyl chloride (207 mg,1.8 mmol) was added dropwise to the solution under cooling in an ice-water bath and stirred at room temperature for 1h. Water and saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated and purified by column chromatography to give the title compound 47n (255 mg,0.7 mmol) in 78% yield.
MS m/z(ESI):362[M+H] +
Second step Synthesis of 2- ((aminosulfonyl) amino) ethyl-1-amine 47o
Compound 47n (255 mg,0.7 mmol) and TBAF (1.3 g,5.1 mmol) were dissolved in tetrahydrofuran (10 mL) and stirred at room temperature for 1h. Ethyl acetate was added for dilution, and the mixture was washed three times with saturated ammonium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 47o (75 mg,0.54 mmol) in 77% yield.
Third step of synthesizing 3, 4-di (oximino) dihydrofuran-2 (3H) -one 47b
The compound 4-hydroxy-2 (5H) -furanone 47a (10 g,0.1 mol) was dissolved in water (60 mL), and sodium nitrite (6.9 g,0.1 mol) was added in portions and stirred at room temperature for 10min. Then, the reaction mixture was added dropwise to an aqueous solution (20 mL) of hydroxylamine sulfate (16.4 g,0.1 mol), stirred at room temperature for 2 hours, filtered, and the cake was washed with water and dried to give the title compound 47b (3.0 g,23.8 mmol) in 24% yield.
Fourth step of synthesizing 4H, 6H-furo [3,4-c ] [1,2,5] oxadiazol-4-one 47c
Compound 47b (3.0 g,23.8 mmol) was dissolved in 1, 4-dioxane (60 mL), and thionyl chloride (3.93 g,33.3 mmol) was slowly added dropwise and stirred at room temperature overnight. Concentration and recrystallization of the resulting solid from ethanol gave the title compound 47c (2.1 g,16.4 mmol) in 69% yield.
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -4- (hydroxymethyl) -1,2, 5-oxadiazole-3-carboxamide 47d
Compound 47c (2.1 g,16.4 mmol) was added to ethanol (80 mL) and DMF (10 mL), and a solution of 3-bromo-4-fluoroaniline (3.1 g,16.4 mmol) in ethanol (5 mL) was slowly added dropwise under ice-bath and stirred overnight at 80 ℃. Concentrated, water was added, extracted 3 times with ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by column chromatography to give the title compound 47d (3.1 g,9.8 mmol) in 60% yield. MS m/z (ESI) 316[ M+H ] ] +
Sixth step Synthesis of N- (3-bromo-4-fluorophenyl) -4- (((triisopropylsilyl) oxy) methyl) -1,2, 5-oxadiazole-3-carboxamide 47e
Compound 47d (4.5 g,14.3 mmol) and triethylamine (1.44 g,14.3 mmol) were dissolved in dichloromethane (20 mL), triisopropylchlorosilane (2.75 g,14.3 mmol) was added and stirred overnight at room temperature. Saturated ammonium chloride solution was added, extracted three times with methylene chloride, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated and purified by column chromatography to give the title compound 47e (5.13 g,10.9 mmol) in 76% yield.
Seventh step of synthesizing N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- (((triisopropylsilyl) oxy) methyl) -1,2, 5-oxadiazole-3-formamidine 47f
Compound 47e (3.53 g,7.5 mmol) was dissolved in acetonitrile (20 mL), phosphorus pentachloride (2.25 g,10.9 mmol) was added and stirred at 80℃for 5h. After cooling, concentrate, the residue was dissolved in ethanol (20 mL), sodium acetate (0.71 g,8.6 mmol), and hydroxylamine sulfate (0.71 g,4.3 mmol) were added and stirred at 70℃for 4h. After concentration, purification by column chromatography gave the title compound 47f (1.65 g,3.4 mmol) in 45% yield.
Eighth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3-4- (((triisopropylsilyl) oxy) methyl) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 47g
Compound 47f (3.16 g,6.5 mmol) was dissolved in THF (20 mL), CDI (1.1 g,6.5 mmol) was added and heated under reflux for 4h. After cooling, ethyl acetate was added, followed by washing with 1M HCl, saturated brine, drying over anhydrous sodium sulfate, filtration, and column chromatography purification after concentration gave 47g (2.6 g,5.1 mmol) of the title compound in 79% yield.
MS m/z(ESI):513[M+H] +
Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- (hydroxymethyl) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 47H
47g (2.6 g,5.1 mmol) of the compound and TBAF (2.6 g,10.2 mmol) were dissolved in tetrahydrofuran (10 mL) and stirred at room temperature for 2h. Ethyl acetate was added for dilution, and the mixture was washed three times with saturated ammonium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 47h (1.50 g,4.2 mmol) in 82% yield.
Tenth step Synthesis of 4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-carbaldehyde 47i
Under nitrogen, compound 47h (1.50 g,4.2 mmol) was dissolved in dichloromethane (20 mL) and DMP (2.67 g,6.3 mmol) was added in portions under an ice-water bath and the reaction was allowed to warm to room temperature for 2 hours. The filtrate was filtered, washed with a saturated sodium sulfite solution, dried over anhydrous sodium sulfate, filtered, concentrated and purified by column chromatography to give the title compound 47i (0.95 g,2.7 mmol) in 64% yield.
MS m/z(ESI):355[M+H] +
Eleventh step synthesis of 4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-carboxylic acid 47j
Compound 47i (0.95 g,2.7 mmol) was dissolved in t-butanol (20 mL) and water (8 mL), a solution of 2-methyl-2-butene in THF (2M, 10.7mL,21.4 mmol), sodium dihydrogen phosphate (0.64 g,5.4 mmol) and sodium chlorite (3.0 g,80%,26.7 mmol) was added and stirred overnight at room temperature. Diluting with water, adjusting pH to 4 with 1M HCl, extracting with ethyl acetate three times, mixing the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, filtering, concentrating, and purifying by column chromatography to give the title compound 47j (460 mg,1.2 mmol) in 46% yield.
MS m/z(ESI):371[M+H] +
Synthesis of 4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -N- (2- ((sulfamoyl) amino) ethyl) -1,2, 5-oxadiazol-3-carboxamide 47k in the twelfth step
Compound 47j (200 mg,0.54 mmol) was dissolved in dichloromethane (10 mL), EDCI (104 mg,0.54 mmol) and HOBt (73 mg,0.54 mmol) were added, stirred at room temperature for 30min, compound 47o (75 mg,0.54 mmol) was added, and stirred at room temperature overnight. Water was added thereto, extracted with methylene chloride, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain the title compound 47k (216 mg,0.44 mmol) in 61% yield.
MS m/z(ESI):492[M+H] +
Thirteenth step Synthesis of 4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -N- (2- ((sulfamoyl) amino) ethyl) -1,2, 5-oxadiazole-3-carboxamide 47
Compound 47k (216 mg,0.44 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (425 mg,3.08 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration and then purification by plate analysis gave the title compound 47 (107 mg,0.23 mmol) in 52% yield.
MS m/z(ESI):466[M+H] +
EXAMPLE 48 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-5- ((2- ((sulfamoyl) amino) ethyl) amino) -1,2, 3-thiadiazole-4-carboxamidine 48
First step Synthesis of 5-chloro-1, 2, 3-thiadiazole-4-carboxylic acid 48b
The compound 5-chloro-1, 2, 3-thiadiazole-4-carboxylic acid ethyl ester 48a (1.0 g,5.2 mmol) was dissolved in 1, 4-dioxane (20 mL), concentrated hydrochloric acid (10 mL) was added, and the mixture was heated under reflux for 3h. After cooling, most of the solvent was removed by concentration, extracted three times with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 48b (787 mg,4.8 mmol) in 92% yield.
Second step Synthesis of N- (3-bromo-4-fluorophenyl) -5-chloro-1, 2, 3-thiadiazole-4-carboxamide 48c
Compound 48b (787 mg,4.8 mmol) was dissolved in dichloromethane (10 mL), EDCI (920 mg,4.8 mmol) and HOBt (640 mg,4.8 mmol) were added, stirred at room temperature for 30min, and then 3-bromo-4-fluoroaniline (227 mg,4.8 mmol) was added and stirred at room temperature overnight. Water was added and extracted twice with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography to give the title compound 48c (1.4 g,4.2 mmol) in 88% yield.
MS m/z(ESI):336[M+H] +
Third step of synthesizing N- (3-bromo-4-fluorophenyl) -5-chloro-N' -hydroxy-1, 2, 3-thiadiazole-4-formamidine 48d
Compound 48c (1.4 g,4.2 mmol) was dissolved in acetonitrile (20 mL), phosphorus pentachloride (1.13 g,4.2 mmol) was added and stirred at 80℃for 5h. After cooling, concentrate, the residue was dissolved in ethanol (20 mL), sodium acetate (0.42 g,5.0 mmol), and hydroxylamine sulfate (0.83 g,5.0 mmol) were added and stirred at 70℃for 4h. After concentration, column chromatography purification gave the title compound 48d (0.7 g,2.0 mmol) in 48% yield.
MS m/z(ESI):351[M+H] +
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (5-chloro-1, 2, 3-thiadiazol-4-yl) -1,2, 4-oxadiazol-5 (4H) -one 48e
Compound 48d (910 mg,2.6 mmol) was dissolved in THF (10 mL) and CDI (440 mg,2.6 mmol) was added and heated at reflux for 4h. After cooling, ethyl acetate was added, followed by washing with 1M HCl, saturated brine, drying over anhydrous sodium sulfate, filtration, and column chromatography purification after concentration gave the title compound 48e (865 mg,2.3 mmol) in 88% yield.
MS m/z(ESI):377[M+H] +
Fifth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (5- ((2- ((sulfamoyl) amino) ethyl) amino) -1,2, 3-thiadiazol-4-yl) -1,2, 4-oxadiazol-5 (4H) -one 48f
Compound 48e (79mg, 2.1 mmol) and compound 47o (29 mg,2.1 mmol) were dissolved in ethanol (15 mL) and stirred at room temperature for 2h. After concentration, purification by column chromatography gave the title compound 48f (294 mg,0.62 mmol) in 30% yield.
MS m/z(ESI):480[M+H] +
Sixth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-5- ((2- ((sulfamoyl) amino) ethyl) amino) -1,2, 3-thiadiazole-4-carboxamidine 48
Compound 48f (294 mg,0.62 mmol) was dissolved in methanol (10 mL), anhydrous potassium carbonate (600 mg,4.34 mmol) was added, and the mixture was stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration and then purification by plate analysis gave the title compound 48 (217 mg,0.48 mmol) in 77% yield.
1 H NMR(400MHz,DMSO-d 6 )δ9.98(s,1H),7.78(d,J=7.3Hz,1H),7.53(d,J=7.4Hz,1H),7.47(d,J=7.4Hz,1H),7.39(p,J=7.3Hz,2H),7.21(t,J=7.6Hz,1H),6.95(d,J=7.8Hz,2H),5.23(s,1H),4.02(t,J=4.8Hz,2H),3.76(s,3H),3.00(s,2H),2.83(s,1H),2.69(s,2H),2.51(s,3H).
MS m/z(ESI):454[M+H] +
EXAMPLE 49 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((sulfamoyl) amino) ethyl) amino) -1,2, 5-thiadiazole-3-carboxamidine 49
Prepared in the same manner as in example 48 using ethyl 4-chloro-1, 2, 5-thiadiazole-3-carboxylate as a starting material in place of 48a.
1 H NMR(400MHz,DMSO-d 6 )δ8.88(s,1H),7.18(t,J=8.8Hz,1H),7.05(dd,J=6.1,2.7Hz,1H),6.91(s,2H),6.70(m,1H),6.63(m,3H),4.19(s,2H),3.22(q,J=5.9Hz,2H).
MS m/z(ESI):454[M+H] +
EXAMPLE 50 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((sulfamoyl) amino) ethyl) amino) furan-3-carboxamidine 50
First step Synthesis of 4-bromofuran-3-carboxylic acid ethyl ester 50b
Compound 3, 4-dibromofuran 50a (5 g,22.3 mmol) was dissolved in tetrahydrofuran (100 mL), and 1M isopropyl magnesium bromide (33.4 mL,33.4 mmol) was added dropwise to the system at zero temperature, and the reaction was carried out at zero temperature for 1 hour. Ethyl chloroformate (2.4 g,22.3 mmol) was then slowly added dropwise to the reaction system while maintaining zero degrees. After the completion of the dropwise addition, the temperature was slowly raised and stirring was maintained for 2 hours. Quench with saturated ammonium chloride solution, extract three times with ethyl acetate, combine the organic phases, wash with water and saturated brine, dry over anhydrous sodium sulfate, filter, concentrate, and purify the crude by column chromatography to give the title compound 50b (3.4 g,15.6 mmol) in 70% yield. MS m/z (ESI) 219[ M+H ] ] +
Second step Synthesis of ethyl 4- ((2- ((tert-butoxycarbonyl) amino) ethyl) amino) furan-3-carboxylate 50c
Compound 50b (2.5 g,11.4 mmol), (2-aminoethyl) carbamic acid tert-butyl ester (2.75 g,17.4 mmol), palladium acetate (0.25 g,1.14 mmol), xantPhos (0.66 g,1.14 mmol) and potassium carbonate (3.1 g,22.8 mmol) were dissolved in anhydrous dioxane (100 mL) and stirred under reflux under nitrogen overnight. The reaction solution was filtered, the filtrate was concentrated, and the crude product was purified by column chromatography to give the title compound 50c (0.74 g,2.5 mmol) in 22% yield.
MS m/z(ESI):299[M+H] +
Third step Synthesis of 4- ((2- ((tert-Butoxycarbonyl) amino) ethyl) amino) furan-3-carboxylic acid 50d
Compound 50c (0.74 g,2.5 mmol) was dissolved in tetrahydrofuran (10 mL) and 2M aqueous sodium hydroxide (3 mL) was added. Stirring at room temperature for 2 hours, quenching with 3M hydrochloric acid, adjusting pH to 4, extracting three times with ethyl acetate, combining the organic phases, washing with water and saturated brine, drying over anhydrous sodium sulfate, filtering, concentrating the filtrate to give the title compound 50d (0.6 g,2.3 mmol) in 92% yield.
MS m/z(ESI):271[M+H] +
Fourth step Synthesis of tert-butyl (2- ((4- ((3-bromo-4-fluorophenyl) carbamoyl) furan-3-yl) amino) ethyl) carbamate 50e
Compound 50d (0.6 g,2.3 mmol), 3-bromo-4-fluoroaniline (0.47 g,2.5 mmol), HATU (0.95 g,2.5 mmol) and triethylamine (0.35 g,3.5 mmol) were dissolved in N, N-dimethylformamide (15 mL) and stirred at room temperature for 3 hours. Ethyl acetate was added and the organic phase was washed four times with water, twice with saturated brine, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated and the crude product was purified by column chromatography to give the title compound 50e (0.88 g,2.0 mmol) in 87% yield.
MS m/z(ESI):442[M+H] +
Fifth step Synthesis of 4- ((2-aminoethyl) amino) -N- (3-bromo-4-fluorophenyl) furan-3-carboxamide trifluoroacetate salt 50f
Compound 50e (0.88 g,2.0 mmol) was dissolved in dichloromethane (15 mL) and trifluoroacetic acid (8 mL) was added. The reaction mixture was stirred at room temperature for 2 hours. The reaction was concentrated to give crude title compound 50f (0.75 g,1.7 mmol) which was used directly in the next reaction.
MS m/z(ESI):342[M+H] +
Sixth step Synthesis of (9H-fluoren-9-yl) methyl (2- ((4- ((3-bromo-4-fluorophenyl) carbamoyl) furan-3-yl) amino) ethyl) carbamate 50g
Compound 50f (0.75 g,1.7 mmol) and triethylamine (0.51 g,5.1 mmol) were dissolved in dichloromethane (10 mL), 9-fluorenylmethylchloroformate (0.54 g,2.1 mmol) was added at room temperature, and stirred overnight at room temperature. The reaction was quenched by the addition of saturated sodium bicarbonate solution, extracted three times with dichloromethane, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give the crude compound 50g (0.78 g,1.4 mmol) as a 82% yield by column chromatography.
MS m/z(ESI):564[M+H] +
Seventh step of synthesizing (9H-fluoren-9-yl) methyl (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) furan-3-yl) amino) ethyl) carbamate for 50H
50g (0.7 g,1.2 mmol) of the compound was dissolved in toluene (10 mL), and phosphorus pentachloride (0.3 g,1.5 mmol) was added. The reaction mixture was stirred under reflux for 4 hours under nitrogen, the reaction solution was concentrated, and the residue was dissolved in ethanol (10 mL) and added dropwise to a 50% aqueous hydroxylamine solution at room temperature. The reaction mixture was stirred at room temperature for 2 hours. The excess ethanol was evaporated, extracted three times with dichloromethane and the organic phases were combined, dried over saturated brine, filtered, the filtrate concentrated and the crude product purified by column chromatography to give the title compound 50h (0.46 g,0.8 mmol) in 66% yield.
MS m/z(ESI):579[M+H] +
Eighth step Synthesis of (9H-fluoren-9-yl) methyl (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) furan-3-yl) amino) ethyl) carbamate 50i
The compound 50h (0.46 g,0.8 mmol) and CDI (0.19 g,1.2 mmol) were dissolved in tetrahydrofuran (20 mL) and stirred at reflux for 2 h. The reaction was concentrated and the crude product was purified by column chromatography to give the title compound 50i (0.3 g,0.5 mmol) in 62% yield.
MS m/z(ESI):605[M+H] +
Ninth step Synthesis of 3- (4- ((2-aminoethyl) amino) furan-3-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one 50j
Compound 50i (0.3 g,0.5 mmol) was dissolved in tetrahydrofuran (10 mL), tetrabutylammonium fluoride (0.65 g,2.5 mmol) was added to the reaction mixture, and the reaction mixture was stirred at room temperature for 2 hours. The reaction solution was quenched with saturated ammonium chloride, extracted three times with dichloromethane, the organic phases were combined, washed three times with saturated ammonium chloride, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product. The crude product was purified by column chromatography to give the title compound 50j (0.15 g,0.4 mmol) in 80% yield.
MS m/z(ESI):383[M+H] +
Tenth step Synthesis of (N- (2- ((4- (4- (3-bromo-fluorophenyl) -5-carbonyl-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) furan-3-yl) amino) ethyl) sulfamoyl) carbamic acid tert-butyl ester 50k
Chlorosulfonyl isocyanate (70 mg,0.5 mmol) is dissolved in dichloromethane (5 mL) and a solution of t-butanol (59 mg,0.8 mmol) in dichloromethane (2 mL) is added dropwise under ice-bath. After the completion of the addition, the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was directly added dropwise to a solution of compound 50j (0.15 g,0.4 mmol) in dichloromethane (5 mL), the reaction mixture was stirred at room temperature for 1 hour, concentrated to give a crude product, which was purified by column chromatography to give the title compound 50k (168 mg,0.3 mmol) in 75% yield.
MS m/z(ESI):562[M+H] +
Eleventh step synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((aminosulfonyl) amino) ethyl) amino) -furan-3-yl) -1,2, 4-oxadiazol-5 (4H) -one trifluoroacetate salt 50l
Compound 50k (168 mg,0.3 mmol) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (2 mL) was added. The reaction mixture was stirred at room temperature for 1 hour. The reaction was concentrated to give 50l of the crude title compound. The crude product was used directly in the next reaction.
MS m/z(ESI):462[M+H] +
Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((sulfamoyl) amino) ethyl) amino) furan-3-carboxamidine 50
Compound 50l was dissolved in tetrahydrofuran (10 mL) and then 2M aqueous sodium hydroxide (4 mL) was added. The reaction mixture was stirred at room temperature for 1 hour, quenched with 3M aqueous hydrochloric acid, pH was adjusted to 8, extracted three times with ethyl acetate, the organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated to give crude product, which was purified by plate chromatography to give the title compound 50 (35 mg,0.08 mmol) in 26% yield in two steps.
MS m/z(ESI):436[M+H] +
EXAMPLE 51 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-3- ((2- ((sulfamoyl) amino) ethyl) amino) thiophene-2-carboxamidine 51
Prepared in the same manner as in example 50 using methyl 3-bromothiophene-2-carboxylate as a starting material in place of 50b.
MS m/z(ESI):452[M+H] +
EXAMPLE 52 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-3- ((2- ((sulfamoyl) amino) ethyl) amino) -1H-pyrazole-4-carboxamidine 52
First step Synthesis of 3-amino-N- (3-bromo-4-fluorophenyl) -1H-pyrazole-4-carboxamide 52b
The compound 3-amino-1H-pyrazole-4-carboxylic acid 52a (5.0 g,39.3 mmol), EDCI (11.3 g,59.0 mmol) and HOBt (8.0 g,59.0 mmol) were dissolved in DMF (50 mL), reacted at room temperature for 1H, 3-bromo-4-fluoroaniline (8.2 g,43.2 mmol) was added, and reacted at room temperature overnight. Concentrated to dryness under reduced pressure, and purified by column chromatography to give the title compound 52b (7.7 g,23.4 mmol) in 65% yield.
MS m/z(ESI):299[M+H] +
Second step Synthesis of 3-amino-N- (3-bromo-4-fluorophenyl) -N' -hydroxy-1H-pyrazole-4-carboxamidine 52c
Compound 52b (7.0 g,23.4 mmol) was dissolved in toluene (50 mL) and phosphorus pentachloride (6.3 g,30.4 mmol) was added. The reaction mixture was stirred under reflux for 4 hours under nitrogen, the reaction solution was concentrated, and the residue was dissolved in ethanol (30 mL) and added dropwise to a 50% aqueous hydroxylamine solution at room temperature. The reaction mixture was stirred at room temperature for 2 hours. The excess ethanol was distilled off, extracted three times with methylene chloride, the organic phases were combined, dried with saturated brine, filtered, and the filtrate was concentrated, and the crude product was purified by column chromatography to give the title compound 52c (3.9 g,12.4 mmol) in 53% yield.
MS m/z(ESI):314[M+H] +
Third step of synthesizing 3- (3-amino-1H-pyrazol-4-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one 52d
Compound 52c (3.9 g,12.4 mmol) and CDI (2.2 g,13.6 mmol) were dissolved in THF (20 mL) and reacted under reflux for 2h. Cooled to room temperature, concentrated, ethyl acetate (50 mL) was added, washed successively with 0.5M HCl (2X 20 mL) and saturated brine (25 mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 52d (3.8 g,11.2 mmol) in 90% yield.
MS m/z(ESI):340[M+H] +
Fourth step Synthesis of tert-butyl (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1H-pyrazol-3-yl) amino) ethyl) carbamate 52e
Compound 52d (3.8 g,11.2 mmol) and tert-butyl (3-oxopropyl) carbamate (2.1 g,12.1 mmol) were dissolved in dichloromethane (50 mL), small amounts of HOAc were added, and NaBH (OAc) was added in portions 3 (7.1 g,33.5 mmol) was reacted overnight at room temperature. Adding saturated NaHCO 3 The solution was separated, the aqueous layer was extracted with DCM (50 mL), the combined organic layers dried over anhydrous sodium sulfate, filtered, concentrated and purified by column chromatography to give the title compound 52e (2.85 g,5.9 mmol) in 53% yield.
MS m/z(ESI):483[M+H] +
Fifth step Synthesis of 3- (3- ((2-aminoethyl) amino) -1H-pyrazol-4-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one trifluoroformate salt 52f
Compound 52e (2.0 g,4.1 mmol) was dissolved in DCM (10 mL), TFA (5 mL) was added and reacted at room temperature for 2h. Concentration to dryness afforded crude title compound 52f, which was used directly in the next step.
MS m/z(ESI):383[M+H] +
Sixth step Synthesis of (N- (2- ((4- (4- (3-bromo-4-fluorophenyl-5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1H-pyrazol-3-yl) amino) ethyl) sulfamoyl) carbamic acid tert-butyl ester 52g
Chlorosulfonic acid isocyanate (0.37 g,2.6 mmol) was dissolved in anhydrous DCM (2 mL), and t-BuOH (0.19 g,2.6 mmol) was added dropwise thereto under ice-water bath cooling and reacted at room temperature for 1h for use (reaction solution A).
Compound 52f (0.5 g,1.3 mmol) was dissolved in anhydrous DCM (2 mL), TEA (0.53 g,5.2 mmol) was added dropwise under ice-water cooling, and reaction solution A was slowly added dropwise, and gradually warmed to room temperature to react for 2h. Ethyl acetate (20 mL) was added to dilute, and the organic layer was washed successively with 0.1M HCl (2X 10 mL) and saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and the crude product was concentrated and purified by column chromatography to give 52g (0.40 g,0.71 mmol) of the title compound in 55% yield.
MS m/z(ESI):562[M+H] +
Seventh step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((sulfamoyl) amino) ethyl) amino) -1H-pyrazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 52H
52g (0.40 g,0.71 mmol) of compound was suspended in DCM (7 mL), TFA (3.5 mL) was added and reacted at room temperature for 2h. Concentrated to dryness, saturated sodium bicarbonate solution was added, extracted 3 times with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and the resulting crude product was concentrated and purified by column chromatography to give the title compound 52h (0.27 g,0.58 mmol) in 81% yield.
MS m/z(ESI):462[M+H] +
Eighth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-3- ((2- ((sulfamoyl) amino) ethyl) amino) -1H-pyrazole-4-carboxamidine 52
Compound 52h (0.27 g,0.58 mmol) was dissolved in methanol (8 mL) and 2M aqueous NaOH solution (0.6 mL,1.2 mmol) was added dropwise and reacted at room temperature for 1.5h. 3M HCl is added dropwise to adjust the pH to 6-7, methanol is distilled off under reduced pressure, water is added, extraction is carried out twice with ethyl acetate, the organic layers are combined, saturated brine is washed, dried over anhydrous sodium sulfate, filtered, the crude product obtained is concentrated, and the title compound 52 (0.23 g,0.53 mmol) is obtained by plate chromatography in 90% yield.
MS m/z(ESI):436[M+H] +
EXAMPLE 53 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- ((sulfamoyl) amino) ethyl) amino) -1H-pyrazole-3-carboxamidine 53
Prepared in the same manner as in example 52 using 4-amino-1H-pyrazole-3-carboxylic acid as a starting material in place of 52a.
MS m/z(ESI):436[M+H] +
EXAMPLE 54 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (ethylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 54
Prepared in the same manner as in example 2, starting with sodium ethanethiolate instead of sodium methanethiol. 1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,1H),8.94(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.9,4.2,2.7Hz,1H),6.35(t,J=5.9Hz,1H),3.33(q,J=6.6Hz,2H),3.21–2.99(m,4H),2.06–1.91(m,2H),1.21(t,J=7.4Hz,3H)。
MS m/z(ESI):450[M+H] +
Example 55 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((1-methylethyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 55
First step of Synthesis of tert-butyl (3- (1' -methylethylsulfanyl) propyl) carbamate 55b
The compound (3-bromopropyl) carbamic acid tert-butyl ester 2a (1.0 g,4.48 mmol) was dissolved in DMSO (5 mL), isopropyl mercaptan (374 mg,4.93 mmol) and sodium hydroxide (197mg, 4.93 mmol) were added, and stirred overnight at room temperature. Water was added and stirred for 0.5h, then ethyl acetate was added to extract, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 55b (854 mg,3.20 mmol) in 71% yield.
The subsequent steps were conducted in the same manner as in example 2 to obtain compound 55.
1H NMR(400MHz,DMSO-d6)δ11.45(s,1H),8.92(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.80–6.73(m,1H),6.34(t,J=5.9Hz,1H),3.37–3.32(m,2H),3.29–3.21(m,1H),3.15–3.07(m,2H),2.03–1.93(m,2H),1.23(s,6H).
MS m/z(ESI):464[M+H]+。
EXAMPLE 56 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (cyclopropylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 56
First step of Synthesis of cyclopropyl mercaptan 56b
The compound cyclopropyl magnesium bromide solution 56a (10 mL,5.0 mmol) was dissolved in tetrahydrofuran (20 mL), and under ice bath, sulfur powder (128 mg,4.0 mmol) was added in portions, after the addition was completed, the mixture was reacted at 50℃for 3 hours, then cooled to 0℃and LiAlH was slowly added 4 (95 mg,2.5 mmol) and, after addition, refluxed for half an hour. After cooling on ice, water (3 mL), 5% dilute sulfuric acid was carefully added, the resulting mixture was extracted with ethyl acetate (3X 25 mL), and the organic phases were combined. Washing with 5% dilute sulfuric acid, 5% sodium carbonate solution, saturated saline water, drying with anhydrous sodium sulfate, filtering, and concentrating to obtain crude product.
The subsequent step was conducted in the same manner as in example 55 to obtain compound 56.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.92(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.80–6.73(m,1H),6.35(t,J=5.9Hz,1H),3.37–3.33(m,2H),3.23–3.14(m,2H),2.74–2.65(m,1H),2.06–1.95(m,2H),1.05–0.91(m,4H).
MS m/z(ESI):462[M+H] +
Example 57 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((2-hydroxyethyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 57
First step Synthesis of 2- ((tert-butyldiphenylsilyl) oxo) ethane-1-thiol 57b
Compound 1-mercapto-ethanol 57a (1.0 g,12.82 mmol), triethylamine (1.94 g,19.23 mmol) were dissolved in dichloromethane (30 mL), and tert-butyldiphenylchlorosilane (3.71 g,12.82 mmol) was added and stirred at room temperature for 4h. Saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 57b (2.43 g,7.69 mmol) by column chromatography in 60% yield.
Second step Synthesis of tert-butyl (3- ((2- ((tert-butyldiphenylsilyl) oxo) ethyl) thio) propyl) carbamate 57c
Compound (3-bromopropyl) carbamic acid tert-butyl ester 2a (1.0 g,4.48 mmol) was dissolved in DMSO (5 mL), and compound 57b (1.56 g,4.93 mmol) and sodium hydroxide (197mg, 4.93 mmol) were added and stirred overnight at room temperature. Water was added and stirred for 0.5h, then ethyl acetate was added to extract, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 57c (1.44 g,3.05 mmol) in 68% yield.
Third step Synthesis of tert-butyl (3- ((2- ((tert-butyldiphenylsilyl) oxo) ethyl) sulfonyl) propyl) carbamate 57d
The compound (3- ((2- ((tert-butyldiphenylsilyl) oxo) ethyl) thio) propyl) carbamic acid tert-butyl ester 57c (1.44 g,3.05 mmol) was dissolved in methanol (20 mL) and Oxone (2.79 g,4.58 mmol) in water (20 mL) was added dropwise and stirred at room temperature for 1h. Water and methylene chloride were added to extract, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 57d (1.29 g,2.56 mmol) in 84% yield.
Fourth step Synthesis of tert-butyl (3- ((2-hydroxyethyl) sulfonyl) propyl) carbamate 57e
Compound 57d (1.29 g,2.56 mmol) was dissolved in tetrahydrofuran (30 mL), TBAF (6.67 g,25.6 mmol) was added, and the mixture was stirred at room temperature for 1h. After the reaction is completed, the reaction mixture is directly used for the next step.
The subsequent steps were conducted in the same manner as in example 2 to obtain compound 57.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.90(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.80–6.71(m,1H),6.32(t,J=6.0Hz,1H),5.12(t,J=5.1Hz,1H),3.78(q,J=5.5Hz,2H),3.37–3.29(m,2H),3.22(t,J=5.8Hz,2H),3.20–3.13(m,2H),2.05–1.94(m,2H)。
MS m/z(ESI):466[M+H] +
EXAMPLE 58 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (((tetrahydrofuran-3-yl) methyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 58
First step Synthesis of S- (3- (((benzyloxy) carbonyl) amino) propyl) thioacetate 58b
The compound benzyl (3-bromopropyl) carbamate 58a (10 g,36.9 mmol) was dissolved in DMF (50 mL) and potassium thioacetate (5 g,44.2 mmol) was added to the system. Stir at room temperature overnight. Quenched with water, extracted three times with ethyl acetate, the organic phases were combined, dried over saturated brine, filtered, and the filtrate concentrated to give the crude product as a colorless oil, 58b (9 g,33.7 mmol) in 91% yield as a crude product by column chromatography.
MS m/z(ESI):290[M+Na] +
Second step of synthesizing benzyl (3-mercaptopropyl) carbamate 58c
Compound 58b (9 g,33.7 mmol) was dissolved in methanol (100 mL) and 2N sodium hydroxide solution (150 mL) was added. The reaction mixture was stirred at room temperature for 4 hours. The pH was adjusted to 7 by addition of 3N hydrochloric acid, extracted three times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound 58c (7 g,31.1 mmol) as an orange oil in 91% yield.
1 H NMR(400MHz,DMSO-d 6 )δ7.30(dt,J=9.6,7.4Hz,5H),3.05(q,J=6.4Hz,2H),2.45–2.38(m,2H),2.30(dd,J=8.5,7.3Hz,1H),1.62(p,J=6.9Hz,2H)。
MS m/z(ESI):248[M+Na] +
Third step of Synthesis of benzyl (3- (((tetrahydrofuran-3-yl) methyl) thio) propyl) carbamate 58d
Compound 58c (200 mg,0.88 mmol), 3- (bromomethyl) tetrahydrofuran (175 mg,1.06 mmol) and potassium tert-butoxide (197mg, 1.76 mmol) were dispersed in ethanol (5 mL), reacted for 10 minutes under reflux, cooled to room temperature, quenched with water, extracted three times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate=2:1) to afford the title compound 58d (100 mg,0.32 mmol) as a colorless oil in 36% yield.
MS m/z(ESI):310[M+H] +
Fourth step Synthesis of benzyl (3- (((tetrahydrofuran-3-yl) methyl) sulfonyl) propyl) carbamate 58e
Compound 58d (100 mg,0.32 mmol) was dissolved in methanol (2 mL), and an aqueous solution of potassium hydrogen peroxymonosulfate complex (107 mg,0.64 mmol) was added. The reaction mixture was stirred at room temperature for 2 hours, saturated NaHCO was added 3 The aqueous solution was quenched, extracted three times with ethyl acetate and the organic phases combined. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound 58e (102 mg,0.30 mmol) as a brown oil in 93% yield.
MS m/z(ESI):342[M+H] +
Fifth step Synthesis of 3- (((tetrahydrofuran-3-yl) methyl) sulfonyl) propane-1-amine 58f
Compound 58e (102 mg,0.30 mmol) was dissolved in methanol (3 mL), 10% Pd/C (3 mg) was added and stirred overnight under hydrogen at ambient temperature. The reaction solution was filtered, and the filtrate was concentrated to give the title compound 58f (50 mg,0.24 mmol) as a pale brown oil in 80% yield.
Sixth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((3- (((tetrahydrofuran-3-yl) methyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 58g
Compound 58f (50 mg,0.24 mmol), compound Int-1 (90 mg,0.24 mmol) and triethylamine (72 mg,0.72 mmol) were dissolved in acetonitrile (5 mL) and stirred at room temperature for 1 hour. The reaction was concentrated and the crude product purified by column chromatography (DCM: meoh=10:1) to give 58g (90 mg,0.17 mmol) of the title compound as a colourless oil in 70% yield.
MS m/z(ESI):532[M+H] +
Seventh step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (((tetrahydrofuran-3-yl) methyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 58
58g (90 mg,0.17 mmol) of the compound was dissolved in tetrahydrofuran (5 mL), and a 2N aqueous solution (4 mL) of sodium hydroxide was added thereto, followed by stirring at room temperature for 1 hour. Quench with 3N aqueous hydrochloric acid, adjust pH to 8, extract three times with ethyl acetate and combine the organic phases. The organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was purified by plate chromatography (DCM: meoh=8:1) to give the title compound 58 (12 mg,0.023 mmol) as a white solid in 13% yield.
1 H NMR(400MHz,Methanol-d4)δ7.11(dd,J=6.0,2.7Hz,1H),7.03(t,J=8.6Hz,1H),6.82(ddd,J=8.8,4.1,2.7Hz,1H),3.97(dd,J=8.7,7.1Hz,1H),3.84(td,J=8.3,4.8Hz,1H),3.74(dt,J=8.3,7.5Hz,1H),3.51(dd,J=8.7,7.1Hz,1H),3.44(t,J=6.7Hz,2H),3.26–3.07(m,5H),2.76(dq,J=14.5,7.3Hz,1H),2.27–2.06(m,3H),1.75(dq,J=12.5,7.9Hz,1H).
MS m/z(ESI):506[M+H] +
EXAMPLE 59 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- (((tetrahydrofuran-2-yl) methyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 59
2- (bromomethyl) tetrahydrofuran was used as a starting material and reacted with 58 c. Subsequent steps were prepared in the same manner as in example 58.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.91(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.9,4.2,2.7Hz,1H),6.32(t,J=6.0Hz,1H),4.22–4.14(m,1H),3.78–3.70(m,1H),3.68–3.59(m,1H),3.45–3.26(m,3H),3.25–3.07(m,3H),2.10–1.92(m,3H),1.90–1.73(m,2H),1.66–1.54(m,1H)。
MS m/z(ESI):506[M+H] +
EXAMPLE 60 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((3- (((3, 3-difluorocyclobutyl) methyl) sulfonyl) propyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 60
3- (bromomethyl) -1, 1-difluorocyclobutane is taken as a raw material to react with 58 c. Subsequent steps were prepared in the same manner as in example 58.
1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,1H),8.92(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.77(ddd,J=8.9,4.2,2.7Hz,1H),6.34(t,J=5.9Hz,1H),3.40(d,J=6.9Hz,2H),3.36–3.30(m,2H),3.17–3.09(m,2H),2.84–2.70(m,2H),2.63–2.42(m,3H),2.05–1.93(m,2H)。
MS m/z(ESI):526[M+H] +
Example 61 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((2-methoxyethyl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 61
1-bromo-2-methoxyethane is taken as a raw material to react with 58 c. Subsequent steps were prepared in the same manner as in example 58.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.91(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.9,4.2,2.7Hz,1H),6.32(t,J=6.0Hz,1H),3.68(t,J=5.7Hz,2H),3.36(t,J=5.7Hz,2H),3.35–3.30(m,2H),3.24(s,3H),3.18–3.09(m,2H),1.99(p,J=7.0Hz,2H)。
MS m/z(ESI):480[M+H] +
EXAMPLE 62 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((3- ((tetrahydro-2H-pyran-4-yl) sulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 62
Starting from 4-bromo-tetrahydropyran, it is reacted with 58 c. Subsequent steps were prepared in the same manner as in example 58.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.91(s,1H),7.18(t,J=8.8Hz,1H),7.12(dd,J=6.1,2.7Hz,1H),6.76(ddd,J=8.8,4.1,2.7Hz,1H),6.33(t,J=6.0Hz,1H),3.99–3.92(m,2H),3.45–3.29(m,5H),3.15–3.08(m,2H),2.04–1.94(m,2H),1.92–1.84(m,2H),1.68–1.55(m,2H)。
MS m/z(ESI):506[M+H] +
EXAMPLE 63 Synthesis of N- (3-bromo-4-fluorophenyl) -4- ((3- (cyclohexylsulfonyl) propyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 63
Starting from cyclohexyl mercaptan, reacting with 2 a. Subsequent steps were prepared in the same manner as in example 56.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.92(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.79–6.73(m,1H),6.33(t,J=5.9Hz,1H),3.36–3.29(m,2H),3.12–2.99(m,3H),2.06–1.92(m,4H),1.84–1.76(m,2H),1.67–1.59(m,1H),1.41–1.10(m,5H).
MS m/z(ESI):504[M+H] +
EXAMPLE 64 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 64
The (2-bromoethyl) carbamic acid tert-butyl ester 64a is taken as a raw material and reacts with sodium methyl mercaptan. The subsequent steps were prepared in the same manner as in example 2.
1 H NMR(400MHz,DMSO-d 6 )δ11.50(s,1H),8.92(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.2,2.7Hz,1H),6.79–6.72(m,1H),6.49(t,J=6.0Hz,1H),3.67(q,J=6.5Hz,2H),3.44(t,J=6.5Hz,2H),3.04(s,3H)。
MS m/z(ESI):422[M+H] +
EXAMPLE 65 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 65
4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-4 is taken as a raw material to replace Int-1 to react with 64 d. Subsequent steps were prepared in the same manner as in example 64.
1 H NMR(400MHz,DMSO-d 6 )δ11.48(s,1H),8.96(s,1H),7.18(t,J=9.5Hz,1H),7.13(t,J=54.2Hz,1H),7.03(dd,J=6.1,2.8Hz,1H),6.96–6.90(m,1H),6.51(t,J=6.0Hz,1H),3.67(q,J=6.4Hz,2H),3.44(t,J=6.5Hz,2H),3.04(s,3H)。
MS m/z(ESI):394[M+H] +
EXAMPLE 66 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (methylsulfonyl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 66
4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-4 is taken as a raw material to replace Int-1 to react with 2 d. The subsequent steps were prepared in the same manner as in example 2.
1 H NMR(400MHz,DMSO-d 6 )δ11.44(s,1H),8.96(s,1H),7.19(t,J=9.5Hz,1H),7.12(t,J=54.9Hz,1H),7.06–7.01(m,1H),6.97–6.91(m,1H),6.35(t,J=5.9Hz,1H),3.36–3.29(m,2H),3.17–3.10(m,2H),2.97(s,3H),1.98(p,J=7.0Hz,2H)。
MS m/z(ESI):408[M+H] +
EXAMPLE 67 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (2-carbonylimidazol-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 67
4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-4 is taken as a raw material to replace Int-1 to react with 13 a. The subsequent steps were prepared in the same manner as in example 13.
1 H NMR(400MHz,MeOD-d 4 )δ7.10–7.04(m,2H),7.03–6.97(m,1H),6.91(t,J=55.8Hz,1H),3.57–3.51(m,2H),3.46–3.42(m,4H),3.42–3.36(m,2H)。
MS m/z(ESI):400[M+H] +
EXAMPLE 68 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (2-pyrrolidin-1-yl) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 68
4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-4 is taken as a raw material to replace Int-1 to react with 14 a. The subsequent steps were prepared in the same manner as in example 14.
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,1H),8.94(s,1H),7.18(t,J=9.4Hz,1H),7.13(t,J=54.6Hz,1H),7.02(dd,J=6.3,2.7Hz,1H),6.97–6.91(m,1H),6.27(t,J=5.7Hz,1H),3.42–3.31(m,6H),2.17(t,J=8.1Hz,2H),1.89(p,J=7.5Hz,2H)。
MS m/z(ESI):399[M+H] +
EXAMPLE 69 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -4- ((2- (1, 1-dioxo-1, 2, 5-thiadiazol-2-yl) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazol-3-carboxamide 69
4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-4 is taken as a raw material to replace Int-1 to react with 12 e. The subsequent steps were prepared in the same manner as in example 12.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.96(s,1H),7.22–7.15(m,2H),7.12(t,J=54.7Hz,1H),7.05–7.00(m,1H),6.96–6.90(m,1H),6.32(t,J=5.9Hz,1H),3.43(q,J=6.1Hz,2H),3.38–3.31(m,2H),3.28(t,J=6.3Hz,2H),3.10(t,J=6.3Hz,2H)。
MS m/z(ESI):436[M+H] +
EXAMPLE 70 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2-oxo-1, 3, 2-dioxaphosphori-l-2-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 70
4- (3-difluoromethyl-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-4 is taken as a raw material to replace Int-1 to react with 16 d. The subsequent steps were prepared in the same manner as in example 16.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.93(s,1H),7.18(t,J=9.5Hz,1H),7.12(t,J=54.3Hz,1H),7.04(dd,J=6.1,2.7Hz,1H),6.97–6.91(m,1H),6.22(t,J=6.1Hz,1H),5.30(dt,J=13.3,6.8Hz,1H),4.34–4.12(m,4H),3.27(q,J=6.3Hz,2H),2.97(dq,J=13.0,6.5Hz,2H),1.93–1.75(m,2H)。
MS m/z(ESI):451[M+H] +
EXAMPLE 71 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (1-hydroxy-2-methylpropan-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 71
First step Synthesis of tert-butyl (2- ((N- (1-hydroxy-2-methylpropyl-2-yl) aminosulfonyl) amino) ethyl) carbamate 71b
Compound 2-amino-2-methylpropyl-1-ol 71a (200 mg,2.24 mmol), compound 4e (800 mg,2.59 mmol) and triethylamine (450 mg,4.45 mmol) were dissolved in acetonitrile (15 mL) and heated at 100℃for 6h. After cooling, concentrated, and the crude product was purified by column chromatography (DCM: meoh=5:1) to give the title compound 71b (600 mg,1.93 mmol) in 86% yield.
Second step Synthesis of 2- ((N- (1-hydroxy-2-methylpropyl-2-yl) aminosulfonyl) amino) ethylamine trifluoroacetate salt 71c
Compound 71b (300 mg,0.96 mmol) was dissolved in dichloromethane (10 mL) and TFA (2 mL) was added and reacted at room temperature for 1h. Concentration gave the crude title compound 71c, which was used directly in the next step.
Third step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((2- ((N- (1-hydroxy-2-methylpropyl-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 71d
Compound 71c, int-1 (325 mg,0.94 mmol) and triethylamine (190 mg,1.88 mmol) were dissolved in acetonitrile (5 mL) and stirred at room temperature for 1 hour. The reaction was concentrated, and the crude product was purified by column chromatography to give the title compound 71d (100 mg,0.19 mmol) in 20% yield.
Fourth step Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- ((N- (1-hydroxy-2-methylpropan-2-yl) aminosulfonyl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 71
Compound 71d (100 mg,0.19 mmol) was dissolved in tetrahydrofuran (5 mL), and a 2N aqueous solution of sodium hydroxide (1 mL) was added thereto and stirred at room temperature for 1 hour. The pH was adjusted to 8 with 1N aqueous hydrochloric acid, extracted three times with ethyl acetate and the organic phases combined. The organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated, and the crude product was purified by plate chromatography to give the title compound 71 (25 mg,0.05 mmol) in 28% yield.
1 H NMR(400MHz,MeOD-d 4 )δ7.12–7.04(m,2H),7.04–6.99(m,1H),6.91(t,J=54.9Hz,1H),3.49–3.43(m,4H),3.28–3.23(m,2H),1.28(s,6H)。
MS m/z(ESI):482[M+H] +
EXAMPLE 72 Synthesis of N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-4- ((2- (S-methylsulfonylimino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 72
Compound 41c (130 mg,0.31 mmol) was dissolved in THF/MeOH (1:1, 10 mL) and 2M aqueous NaOH (0.6 mL) was added and reacted at room temperature for 1h. The pH was adjusted to 7 with 1M HCl, diluted with ethyl acetate (25 mL), washed successively with water (10 mL) and saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated to give the title compound 72 (115 mg,0.29 mmol) in 94% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,1H),8.94(s,1H),7.18(t,J=9.5Hz,1H),7.13(t,J=54.4Hz,1H),7.02(dd,J=6.1,2.8Hz,1H),6.96–6.89(m,1H),6.59(t,J=6.0Hz,1H),3.87(s,1H),3.65(q,J=6.4Hz,2H),3.42–3.27(m,2H),2.95(s,3H)。
MS m/z(ESI):393[M+H] +
EXAMPLE 73 Synthesis of dimethyl (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) phosphoramidate 73
/>
First step Synthesis of tert-butyl (2- ((dimethoxyphosphono) amino) ethyl) carbamate 73b
The compound N-t-butoxycarbonyl-1, 2-ethylenediamine hydrochloride 73a (500 mg,2.5 mmol) and triethylamine (514 mg,5.0 mmol) were dissolved in dichloromethane (8 mL), and dimethoxyphosphoryl chloride (404 mg,2.8 mmol) was added under an ice-water bath to react overnight at room temperature. After dilution with water, extraction was performed 3 times with dichloromethane, the organic phases were combined, washed once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound 73b (600 mg,0.22 mmol) in 88% yield.
MS m/z(ESI):291[M+Na] +
Second step of synthesizing dimethyl (2-aminoethyl) phosphoramidate 73c
Compound 73b (200 mg,0.75 mmol) was dissolved in dichloromethane (10 mL), TFA (1 mL) was added and reacted at room temperature for 0.5h. Concentration gave the crude title compound 73c, which was used directly in the next step.
Third step Synthesis of dimethyl (2- ((4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) phosphoramidate 73d
Compound 73c was dissolved in tetrahydrofuran (10 mL) and water (1 mL), and potassium carbonate (207 mg,1.5 mmol) and compound Int-1 (100 mg,0.27 mmol) were added and reacted at room temperature for 2h. Water was added thereto, extraction was performed with ethyl acetate, drying over anhydrous sodium sulfate, filtration and concentration were performed, and the crude product was purified by plate analysis to give the title compound 73d (10 mg,0.02 mmol) in a yield of 7.5%.
MS m/z(ESI):493[M+H] +
Fourth step Synthesis of dimethyl (2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxycarbamimidoyl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) phosphoramidate 73
Compound 73d (10 mg,0.02 mmol) was dissolved in methanol (10 mL), potassium carbonate (14 mg,0.1 mmol) was added, and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extracted three times with methylene chloride, dried over anhydrous sodium sulfate, filtered, and concentrated, and then purified by plate chromatography to give the title compound 73 (9 mg,0.019 mmol) in 95% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.89(s,1H),7.19(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.80–6.74(m,1H),6.22(t,J=6.2Hz,1H),5.15–5.07(m,1H),3.56(s,3H),3.53(s,3H),3.26(q,J=6.2Hz,2H),3.00–2.91(m,2H)。
MS m/z(ESI):467[M+H] +
EXAMPLE 74 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((1- (2-oxo-1, 3, 2-dioxaphosphorin-2-yl) piperidin-4-yl) amino) -1,2, 5-oxadiazole-3-carboxamidine 74
First step Synthesis of 2-chloro-2-oxo-1, 3, 2-dioxaphosphorinane 16b
Compound 1, 3-propanediol 16a (497 mg,6.54 mmol) and triethylamine (1.32 g,13.08 mmol) were dissolved in dichloromethane (20 mL), cooled to 0℃under nitrogen, and phosphorus oxychloride (1.0 g,6.54 mmol) was added dropwise to the solution. The solution was stirred at room temperature overnight and used directly in the next step.
Second step Synthesis of tert-butyl (1- (2-oxo-1, 3, 2-dioxaphosphorinan-2-yl) piperidin-4-yl) carbamate 74a
Under nitrogen, the compound 4- (tert-butoxycarbonylamino) piperidine (1.0 g,5.0 mmol) and triethylamine (505 mg,5.0 mmol) were dissolved in dichloromethane (10 mL), cooled to 0℃and a one-step dichloromethane solution of compound 16b was added dropwise to the solution. Stirring was carried out at room temperature for 2h. The reaction solution was washed with water, saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated and recrystallized to give the title compound 74a (1.0 g,3.1 mmol) in 63% yield.
MS m/z(ESI):343[M+Na] +
Third step Synthesis of 2-oxo-2- (4-aminopiperidin-1-yl) -1,3, 2-dioxaphosphorinane trifluoroacetate 74b
Compound 74a (200 mg,0.62 mmol) was dissolved in dichloromethane (10 mL), TFA (2 mL) was added and reacted at room temperature for 1h. Concentration gave the crude title compound 74b, which was used directly in the next step.
Fourth step Synthesis of 4- (3-bromo-4-fluorophenyl) -3- (4- ((1- (2-oxo-1, 3, 2-dioxaphosphorinan-2-yl) piperidin-4-yl) amino) -1,2, 5-oxadiazol-3-yl) -1,2, 4-oxadiazol-5 (4H) -one 74c
The above compound 74b and potassium carbonate (75 mg,0.54 mmol) were added to a tetrahydrofuran (10 mL) and water (1 mL) system, and the compound Int-1 (100 mg,0.27 mmol) was added and stirred at room temperature for 1h. Diluting with water, extracting with ethyl acetate, drying over anhydrous sodium sulfate, filtering, concentrating, and purifying the crude product by plate chromatography to give the title compound 74c (60 mg,0.11 mmol) in 41% yield.
Fifth step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((1- (2-oxo-1, 3, 2-dioxaphosphorin-2-yl) piperidin-4-yl) amino) -1,2, 5-oxadiazole-3-carboxamidine 74
Compound 74c (60 mg,0.11 mmol) was dissolved in methanol (10 mL), and potassium carbonate (107 mg,0.77 mmol) was added thereto and stirred at 50℃for 1h. After cooling, a saturated ammonium chloride solution was added, extraction was performed three times with methylene chloride, washing with saturated brine, drying over anhydrous sodium sulfate, filtration, concentration, and then purification by plate analysis gave the title compound 74 (50 mg,0.096 mmol) in 88% yield.
1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,1H),8.90(s,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.1,2.7Hz,1H),6.78(ddd,J=8.9,4.2,2.7Hz,1H),6.18(d,J=7.4Hz,1H),4.39–4.15(m,4H),3.52–3.42(m,1H),3.42–3.34(m,2H),2.88–2.75(m,2H),2.07–1.72(m,4H),1.44–1.30(m,2H)。
MS m/z(ESI):519[M+H] +
EXAMPLE 75 Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (aminosulfonyl (1-aminosulfonyl azetidin-3-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 75
First step Synthesis of tert-butyl 3- ((2- ((fluorenylmethoxycarbonyl) amino) ethyl) amino) azetidine-1-carbamate 75b
Compound N-fluorenylmethoxycarbonyl ethylenediamine hydrochloride 75a (284 mg,2.92 mmol), 1-tert-butoxycarbonyl-3-azetidinone (500 mg,2.92 mmol) was dissolved in dichloromethane (15 mL), acetic acid (0.5 mL) was added, and NaBH (OAc) was added in portions 3 (928 mg,4.38 mmol) was stirred at room temperature overnight. Saturated sodium bicarbonate solution and dichloromethane were added, extracted 2 times, and dried over anhydrous sodium sulfate. Filtration, concentration and purification of the crude product by column chromatography gave the title compound 75b (500 mg,1.14 mmol) in 44% yield.
Second step Synthesis of tert-butyl 3- ((2-aminoethyl) amino) azetidine-1-carbamate 75c
Compound 75b (540 mg,1.23 mmol) was dissolved in tetrahydrofuran (5 mL), TBAF (470 mg,1.8 mmol) was added and reacted at room temperature for 2h. The resulting solution of crude title compound 75c was used directly in the next step.
Third step Synthesis of 3- ((2- ((4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) amino) azetidine-1-carbamic acid tert-butyl ester 75d
To the tetrahydrofuran solution of the above-mentioned compound 75c, compound Int-1 (270 mg,0.72 mmol) was added and reacted at room temperature for 0.5h. After dilution with water, extraction with ethyl acetate, aqueous saturated saline, drying over anhydrous sodium sulfate, filtration, concentration, and purification of the crude product by column chromatography gave the title compound 75d (170 mg,0.31 mmol) in 43% yield.
Fourth step Synthesis of 3- (4- ((2- ((azetidin-3-yl) amino) ethyl) amino) -1,2, 5-oxadiazol-3-yl) -4- (3-bromo-4-fluorophenyl) -1,2, 4-oxadiazol-5 (4H) -one trifluoroacetate 75e
Compound 75d (220 mg,0.41 mmol) was dissolved in dichloromethane (5 mL), TFA (2 mL) was added, and the mixture was stirred at room temperature for 1h. Concentration gave the crude title compound 75e, which was used directly in the next step.
Fifth step Synthesis of tert-butyl ((3- ((2- ((4- (4- (3-bromo-4-fluorophenyl) -5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) (N- (tert-butoxycarbonyl) aminosulfonyl) amino) azetidin-1-yl) aminosulfonyl) carbamate 75f
The compound chlorosulfonic acid isocyanate (23 mg,0.16 mmol) was dissolved in anhydrous dichloromethane (1 mL), and t-BuOH (13 mg,0.18 mmol) was added thereto under ice-water bath cooling to react at room temperature for 1h, followed by preparation (reaction solution A).
Compound 75e (80 mg,0.15 mmol) was dissolved in anhydrous dichloromethane (1 mL), triethylamine (61 mg,0.6 mmol) was added under ice-water bath cooling, and the reaction solution A was slowly dropped and gradually warmed to room temperature to react for 2 hours. Dichloromethane (10 mL) was added for dilution, washed with water and saturated brine in this order, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product obtained was purified by plate analysis to give the title compound 75f (40 mg,0.05 mmol) in 35% yield.
Sixth step Synthesis of tert-butyl ((3- ((2- ((4- (N- (3-bromo-4-fluorophenyl) -N' -hydroxyformamidino) -1,2, 5-oxadiazol-3-yl) amino) ethyl) (N- (tert-butoxycarbonyl) aminosulfonyl) amino) azetidin-1-yl) aminosulfonyl) carbamate 75g
Compound 75f (40 mg,0.05 mmol) was dissolved in tetrahydrofuran (1 mL), and 2N aqueous NaOH (0.5 mL) was added thereto and stirred at room temperature for 1h. The pH was adjusted to 7 with 2N HCl and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by plate chromatography to give 75g (20 mg,0.026 mmol) of the title compound in 52% yield.
MS m/z(ESI):772[M+H] +
Seventh step Synthesis of N- (3-bromo-4-fluorophenyl) -N' -hydroxy-4- ((2- (sulfamoyl (1-sulfamoyl azetidin-3-yl) amino) ethyl) amino) -1,2, 5-oxadiazole-3-carboxamide 75
75g (20 mg,0.026 mmol) of the compound was dissolved in methylene chloride (1 mL), TFA (0.5 mL) was added, and the mixture was reacted at room temperature for 1h. Concentrated, and the crude product was purified by reverse phase column chromatography to give the title compound 75 (5.6 mg,0.01 mmol) in 38% yield.
1 H NMR(400MHz,MeOD-d 4 )δ7.15(dd,J=6.0,2.7Hz,1H),7.05(t,J=8.7Hz,1H),6.89–6.80(m,1H),4.40(p,J=7.3Hz,1H),3.93(d,J=7.4Hz,4H),3.58(t,J=6.4Hz,2H),3.41(t,J=6.4Hz,2H)。
MS m/z(ESI):572[M+H] +
EXAMPLE 76 Synthesis of N- (3-chloro-4-fluorophenyl) -N' -hydroxy-4- ((3- (methylsulfonyl) propyl) amino) -1,2, 5-oxadiazole-3-carboxamidine 76
4- (3-chloro-4-fluorophenyl) -3- (4-nitro-1, 2, 5-oxadiazole-3-yl) -1,2, 4-oxadiazole-5 (4H) -ketone Int-5 is taken as a raw material to replace the reaction of Int-1 and 2 d. The subsequent steps were prepared in the same manner as in example 2. 1 H NMR(400MHz,MeOD-d 4 )δ11.47(s,1H),8.93(s,1H),7.22(t,J=9.1Hz,1H),6.99(dd,J=6.6,2.8Hz,1H),6.80–6.66(m,1H),6.33(t,J=6.0Hz,1H),3.38-3.28(m,2H),3.14(t,J=7.9Hz,2H),2.97(s,3H),2.09–1.88(m,2H).
MS m/z(ESI):392[M+H] +
Example 77- ((2- (N-methoxyacyl) -S-methylsulfonylimino) ethyl) amino) -N- (3- (difluoromethyl) -4-fluorophenyl) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine 77
Prepared in the same manner as in compound 41, substituting methyl chloroformate for cyclopropylsulfonyl chloride.
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.97(s,1H),7.19(t,J=9.5Hz,1H),7.13(t,J=54.4Hz,1H)7.04(dd,J=6.2,2.8Hz,1H),6.97–6.89(m,1H),6.56(t,J=5.7Hz,1H),3.89–3.65(m,4H),3.53(s,3H),3.37(s,3H).
MS m/z(ESI):451[M+H] +
Biological experimental examples:
EXAMPLE 1 determination of the in vitro enzyme inhibitory Activity of the Compounds of the invention against human indoleamine-2, 3 dioxygenase 1 (hIDO 1)
1. Purpose of experiment and method
The purpose of this experiment was to test the in vitro enzyme inhibition activity of the compounds of the present invention against human indoleamine-2, 3 dioxygenase (hIDO 1) and to evaluate the in vitro activity of the compounds according to IC 50.
1.1 configuration of the reaction System
The total reaction system was 100uL, and final concentration was 400uM L-tryptophan (Sigma, cat#T8941), 20mM ascorbic acid (Sigma, cat#A4034), 20uM methylene blue (Sigma, cat#M9140), 1000U/ml catalase (Sigma, cat#9322), 100mM phosphate buffer (Corning, cat#21-040-CVR) PH6.5 were mixed, and the mixture was preheated in a 37℃water bath (Shanghai precision macro laboratory equipment) for 5 minutes.
1.2 configuration of Compounds
The compound was diluted with 100% DMSO (Sigma, cat#D2650) starting at a concentration of 10000nM and diluted 1:4 to a final test concentration of 9.765nM-10000nM. 10uL of each dilution group compound was taken into the above reaction system.
1.3 detection
To the reaction system containing the compound was added hIDO1 (self-contained) at a final concentration of 20nM, reacted at 37 ℃ for 30 minutes, after enzymatic reaction, 30% w/v trichloroacetic acid 50uL (Sigma, cat#t 0699) was added to terminate the reaction, the reaction solution was heated at 52 ℃ for 30 minutes to complete the conversion from N-formylkynurenine to kynurenine, and then centrifuged at 12000rpm for 10 minutes at normal temperature, 100uL of the supernatant was mixed with an equal volume of Ehrlich's reagent (Sigma, cat# 156477), and absorbance (OD) was measured at 480nM using a microplate reader (BIOTEK).
1.4 data processing and result analysis
Using the general inhibitor formula lgIC50 = Xm-I (P- (3-Pm-Pn)/4), xm: lg maximum dose, I: lg (maximum dose/adjacent dose), P: sum of positive response rates, pm: maximum positive response rate, pn: minimum positive response rate. Concentration-dependent statistical analysis was performed to calculate the median inhibitor concentration IC50 of the compound.
The positive compounds used in the experiments were: INCB024360 (and EW. Cat# 1204669-58-8).
2. Test results
Specific IC50 values for in vitro enzyme activity assays of the compounds of the invention on human indoleamine-2, 3 dioxygenase (hIDO 1) are shown in Table 1 below. As can be seen from Table 1 below, the compounds of the present invention have superior in vitro enzyme inhibition effect on human indoleamine-2, 3 dioxygenase (hIDO 1) than INCB024360.
Table 1: half maximal inhibitory concentration (IC 50) of the compounds of the invention for the inhibitory activity of hIDO1 enzyme
/>
Conclusion: the compound has stronger enzyme inhibition activity on human indoleamine-2, 3 dioxygenase (hIDO 1).
EXAMPLE 2 determination of the intracellular enzyme inhibitory Activity of the Compounds of the invention on human Indolylamine-2, 3 dioxygenase 1 (hIDO 1)
1. Purpose of experiment and method
The purpose of this experiment was to test the in vitro activity of the compounds according to the invention on the intracellular enzyme inhibition activity of human indoleamine-2, 3 dioxygenase (hIDO 1) by evaluating the compounds according to IC 50.
1.1. Cell culture and preparation
HEK293 cells (from Proc. Natl. Acad. Sci.) were used to control the cell count of human embryonic kidney cells at 2.5X10 5 Inoculating/ml into 96-well plate (Corning, cat: 3599), 100ul per well, placing at 37deg.C, humidity 95%, 5% CO 2 Incubator (Thermo) overnight. After 24 hours, recombinant plasmid PcDNA3-hIDO1 was transfected using liposome Lipofectamin2000 (Invitrogen, cat# 11668-027). The information on the medium used for cell culture is as follows: DMEM (Corning, cat#11960), FBS (Gibco, cat#35-076-CV), green, streptomycin diabody (Corning, cat: 30-002-CI).
1.2 configuration of Compounds
The compound was diluted with 100% DMSO (Sigma, cat#D2650) starting at a concentration of 100000nM and 1:4 and the final assay concentration was 9.765nM-10000nM.
1.3 Experimental procedure
After the recombinant plasmid PcDNA3-hIDO1 is transfected into 293 cells for 24 hours, 10uL of diluted compounds with different concentrations are added, and the compound of the control group is INCB024360 (and the chemical engineering, cat# 1204669-58-8).
1.4 detection
After further incubation for 24 hours, 100uL of the cell supernatant was separately taken into another 96 well plate, 50uL of 30% w/v trichloroacetic acid (Sigma, cat#t0699) was added to terminate the reaction, heated at 52 ℃ for 30 minutes, then centrifuged at 12000rpm for 10 minutes at room temperature, 100uL of the supernatant was mixed with an equal volume of Ehrlich's (Sigma, cat# 156477) reagent for color development, and finally absorbance (OD) values were detected at 480nm using a microplate reader (BIOTEK).
1.5 data processing and result analysis
The half inhibitor concentration IC50 of the compound was calculated using the general inhibitor formula (lgIC 50 = Xm-I (P- (3-Pm-Pn)/4), xm: lg maximum dose, I: lg (maximum dose/adjacent dose), P: sum of positive response rates, pm: maximum positive response rate, pn: minimum positive response rate, by concentration-dependent statistical analysis.
2. Test results
Specific IC50 values for the measurement of the cytostatic activity of the compounds of the invention against human indoleamine-2, 3 dioxygenase (hIDO 1) are shown in Table 2 below. As can be seen from Table 2 below, the inhibition effect of the compounds of the present invention on human indoleamine-2, 3 dioxygenase (hIDO 1) is significantly better than INCB024360.
Table 2: half maximal inhibitory concentration (IC 50) of the compounds of the invention on the cytostatic activity of hIDO1
/>
Conclusion: the compound has stronger enzyme inhibition activity on human indoleamine-2, 3 dioxygenase (hIDO 1).
The compounds of the present invention were evaluated for their in vitro activity on IC50 in human endometrial cancer Hela intracellular indoleamine-2, 3 dioxygenase (IDO 1) enzyme inhibition assay.
1.1. Cell culture and preparation
Human endometrial cancer HeLa cells (from the department of Chinese sciences) were isolated at 1X 10 5 Inoculating/ml into 96-well plate (Corning, cat#3599), 100uL per well, placing at 37deg.C, humidity 95%, 5% CO 2 Incubator (Thermo) overnight. The information on the medium used for cell culture is as follows: DMEM (Corning, cat#11960), FBS (Gibco, cat#35-076-CV), green, streptomycin double antibody (Corning, cat#30-002-CI).
1.2 configuration of Compounds
The compound was diluted with 100% DMSO (Sigma, cat#D2650) starting at a concentration of 100000nM and 1:4 and the final assay concentration was 9.765nM-10000nM.
1.3 Experimental procedure
Cells were cultured for 24 hours, the supernatant was removed, and 100uL of DMEM high-sugar medium (Corning, cat#11960) containing INF-gamma (R & D, cat#285-IF-100) and 10uL of diluted compound was added to each well and cultured overnight to express indoleamine-2, 3 dioxygenase (IDO 1), and the control compound was INCB024360 (vs. Oncomelani, cat# 1204669-58-8).
1.4 detection
After 24 hours incubation, 100uL of the cell supernatant was separately taken into another 96-well plate, 50uL of 30% w/v trichloroacetic acid (Sigma, cat#T0699) was added to terminate the reaction, the mixture was heated at 52℃for 30 minutes, centrifuged at 12000rpm for 10 minutes at room temperature, 100uL of the supernatant was mixed with an equal volume of Ehrlich's reagent (Sigma, cat# 156477) to develop a color, and finally absorbance (OD) was measured at 480nm using a microplate reader (BIOTEK).
1.5 data processing and result analysis
The concentration-dependent statistical analysis was performed using the general inhibitor formula lgic50=xm-I (P- (3-Pm-Pn)/4), xm: lg maximum dose, I: lg (maximum dose/adjacent dose), P: sum of positive response rates, pm: maximum positive response rate, pn: minimum positive response rate, to calculate the half inhibitor concentration (IC 50) of the compound.
2. Test results
Specific IC50 values for the measurement of the human endometrial cancer Hela cell enzyme inhibitory activity of the compounds of the invention are shown in table 3 below. As can be seen from Table 3 below, the enzyme inhibition effect of the compounds of the present invention on indoleamine-2, 3 dioxygenase (IDO 1) in human endometrial cancer Hela cells is significantly better than INCB024360.
Table 3: half inhibitory concentration (IC 50) of the inventive Compounds against the inhibitory Activity of IDO1 enzyme in human endometrial cancer Hela cells
Conclusion: the compound has stronger inhibitory activity on indoleamine-2, 3 dioxygenase (IDO 1) in human endometrial cancer Hela cells.
Example 4 in vitro cytotoxicity test of the Compounds of the invention against murine melanoma cells B16F10
1. Purpose of experiment and method
The purpose of this experiment was to test the cytotoxicity test of the compounds of the invention against murine melanoma cells B16F 10.
1.1. Cell culture and preparation
The melanoma cells B16F10 (derived from cell line of the national academy of sciences) of the mice were cultured at 1X 10 4 Inoculating/ml into 96-well plate (Corning, cat: 3599), 100uL per well, placing at 37deg.C, humidity 95%, 5% CO 2 Incubator (Thermo) overnight. The information on the medium used for cell culture is as follows: DMEM (Corning, cat#11960), FBS (Gibco, cat#35-076-CV), green, streptomycin double antibody (Corning, cat#30-002-CI).
1.2 configuration of Compounds
The compound was diluted with 100% DMSO (Sigma, cat # 2650) starting at a concentration of 100000nM and performed at a 1:4 dilution and a final assay concentration of 9.765nM-10000nM.
1.3 Experimental procedure
Adding 10uL of diluted compound into cells for further culturing for 48 hours, and setting 3 compound holes (experimental group) for each sample; a blank (medium), background zeroing (containing only cells and medium), negative control wells (cells, medium, same concentration of drug dissolution medium, thiazole blue (MTT), DMSO) and positive control wells (cells, same concentration of INCB024360 (vs. freon, cat# 1204669-58-8), medium, thiazole blue (MTT), DMSO) were also set.
1.4 detection
20uL of thiazole blue (MTT) solution (5 mg/ml, sigma, cat#M2003) was added to each well, the culture was continued for 4 hours, the culture solution in the well was discarded, 150uL of DMSO (Sigma, cat#D2650) was added to each well, and the mixture was placed on a shaking table and shaken at a low speed for 10 minutes to dissolve the crystals sufficiently. The absorbance (OD) values of each well were measured at 490nm using a microplate reader (BIOTEK).
1.5 data processing and result analysis
Calculation of cell proliferation inhibition ratio=100% × (1- (average OD value of experimental group-average OD value of negative control group)/average OD value of negative control group) using the formula)
2. Test results
Specific data for the measurement of the proliferation inhibition rate of the compounds of the present invention on mouse melanoma cells B16F10 are shown in table 4 below. From Table 4 below, it can be seen that the compounds of the present invention are not cytotoxic to murine melanoma cells B16F10 in vitro.
Table 4: proliferation inhibition rate of the compound of the invention on mouse melanoma cells B16F10 in vitro
/>
Conclusion: the compound of the invention has no cytotoxicity to the melanoma cells B16F10 of mice.
EXAMPLE 5 inhibition experiments of the Compounds of the invention on mature dendritic cell kynurenine levels
1. Purpose of experiment and method
The purpose of this experiment was to test the inhibition assay of the kynurenine level of the mature dendritic cells by the compounds of the present invention.
1.1. Cell culture and preparation
Human lymphocyte isolates (Sigma, cat# 10771) were density gradient centrifuged at 2500rpm for 30 minutes to obtain human Peripheral Blood Mononuclear Cells (PBMC) according to reference (Liu X, shin N, koblish H K, et al, select inhibition of IDO, 1, effectively regulates mediators of antitumor, immunity, blood,2010,115 (17): 3520), and human peripheral blood mononuclear cells were seeded in 24 well plates (Corning, cat# 3473). The information on the medium for cell culture is as follows: DMEM (Corning, cat#11960), FBS (Gibco, cat#35-076-CV), green, streptomycin double antibody (Corning, cat#30-002-CI).
1.2 configuration of Compounds
The compound was diluted with 100% DMSO (Sigma, cat#D2650) starting at a concentration of 100000nM and 1:4 and the final assay concentration was 9.765nM-10000nM.
1.3 Experimental procedure
The medium was induced to culture with serum-free cell culture medium AIMV (Invitrogen, cat# 12055091) containing 100ng/mL granulocyte-macrophage colony stimulating factor GM-CSF (R & D, cat#215-GM-010) and 200ng/mL IL-4 (R & D, cat#204-IL-010) for 5 days, and further with lipopolysaccharide LPS (Invitrogen, cat#00-4976-93) and IFN-gamma (R & D, cat#285-IF-100) for 48 hours to induce indoleamine-2, 3 dioxygenase (IDO) expression, example compounds at different concentrations after dilution and control samples INCB024360 (vs. the chemical, cat# 1204669-58-8) were added, and the vehicle group (DMSO group) and negative control group (blank) were set and incubation was continued for 48 hours.
1.4 detection
After 48 hours of incubation, 100uL of the supernatant was taken and the concentration of kynurenine in the supernatant was determined by high performance liquid chromatography-fluorescence detection.
1.5 data processing and result analysis
The concentration-dependent statistical analysis was performed using the general inhibitor formula lgic50=xm-I (P- (3-Pm-Pn)/4), xm: lg maximum dose, I: lg (maximum dose/adjacent dose), P: sum of positive response rates, pm: maximum positive response rate, pn: minimum positive response rate, to calculate the half inhibitor concentration (IC 50) of the compound to the mature dendritic cell kynurenine level.
2. Test results
The compounds of the examples of the present invention can inhibit the kynurenine production level of mature dendritic cells, and the specific data are shown in table 5 below, and the inhibition effect of the compounds of the present invention on the kynurenine level of mature dendritic cells is more obvious than that of the control group INCB024360 as shown in table 5 below.
Table 5: half maximal inhibitory concentration of the inventive Compounds on kynurenine level of mature dendritic cells
Conclusion: the compound has a strong inhibition effect on the kynurenine level of mature dendritic cells.
Example 6 in vivo efficacy test of the Compound of the invention in mice
1. Purpose of experiment
The effect of example compound 18, example compound 24, example compound 37, example compound 48, example compound 67 on the tumor-inhibiting effect of ct26.wt in a colon cancer model of mice after oral administration was measured using mice as experimental animals. The tumor inhibiting effect of the compound of the present invention on ct26.wt tumor-bearing mice was studied.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 18, example compound 24, example compound 37, example compound 48, example compound 67
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% of N, N-dimethylacetamide (Guozu, cat# 30066818), 30% of PEG400 (Taitan, cat#G 41713B) and 65% of 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasonic, and making into suspension.
2.3 operation
2.3.1 Experimental procedure
Under aseptic condition, the colon cancer cells CT26.wt (derived from cell site of Chinese academy) of mice in logarithmic growth phase are digested into single cells, and transplanted under the right side of the back of Balb/c mice (purchased from Shanghai Sipule-BiKai laboratory animal Co., ltd.) each inoculated with 1×10 5 Cells, 100uL in volume, 9 days after inoculation, until the tumor of the mice grows to an average value of 100mm 3 When the mice were left and right, the mice were randomly divided into 7 groups, 6 in vivo efficacy experiments were performed in each group, the positive control group was INCB024360 (and Kat #1204669-58-8, chemical engineering), and the negative control group was given an equal amount of vehicle. The specific design is shown in Table 6.
Table 6: in vivo efficacy experiment of Compounds
2.3.2 determination of tumor inhibition Rate
Mouse tumor volume = length x width/2 (mm) calculated according to the formula 3 ) Formula of tumor inhibition rate = 1- (tumor size of dosing group/tumor size of negative control group) ×100%
2.4 experimental results
The compound of this example can inhibit the tumor growth rate of CT26 tumor-bearing mice, and the specific data are shown in table 7 below, and it can be seen from table 7 below that the inhibition effect of the compound of the present invention on the tumor growth rate of CT26 tumor-bearing mice is more obvious than that of the control group INCB 024360.
Table 7: tumor growth inhibition rate of the compound of the invention on CT26 tumor-bearing mice
Conclusion: the compound has better inhibition effect on the tumor growth of CT26 tumor-bearing mice.
EXAMPLE 7 in vivo efficacy test of the Compound of the invention in mice
1.1. Purpose of experiment
The effect of each of example compound 20, example compound 56, and example compound 62 on the antitumor effect of PAN02 in the pancreatic cancer model of mice was measured by using mice as experimental animals. The tumor inhibiting effect of the compound of the invention combined with axitinib on PAN02 tumor-bearing mice is discussed.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 20, example compound 56, example compound 62
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% NN dimethylacetamide (Guozu, cat# 30066818), 30% PEG400 (Taitan, cat#G 41713B) and 65% 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasound, and making into suspension.
2.3 operation
2.3.1 Experimental procedure
Under the aseptic condition, the PAN02 (from Nanjing Bai biotechnology Co., ltd.) of the pancreatic cancer cells in the logarithmic growth phase is digested into single cells, and then the single cells are mixed with Matrigel (Corning, cat# 354234) and transplanted under the skin on the back right side of a C57BL/6 mouse, each mouse is inoculated with 5 x 10≡6 cells, the volume of 100uL, after the inoculation, when the tumor of the mouse grows to about 100mm3 on the average value, the mice are randomly divided into 10 groups, each group is subjected to 6 drug effect experiments, the positive control group is INCB024360 (and the ond chemical industry, cat# 1204669-58-8), and the negative control group is given an equal amount of solvent. The specific design is shown in Table 8.
Table 8: in vivo efficacy experiment of Compounds
2.3.2 determination of tumor inhibition Rate
The formula of tumor volume = length x width/2 (mm 3) of mice was calculated according to the formula = 1- (tumor size of dosing group/tumor size of negative control group) x100% of tumor inhibition rate
2.4 experimental results
The compound of the example can inhibit the tumor growth rate of PAN02 tumor-bearing mice by combining with the axitinib, the specific data are shown in the following table 9, and the inhibition effect of the compound of the invention on the tumor growth rate of the PAN02 tumor-bearing mice by combining with the axitinib is more obvious than that of a control group INCB 024360.
Table 9: tumor growth inhibition rate of PAN02 tumor-bearing mice by combining compound and axitinib
Conclusion: the compound of the invention has better inhibition effect on the tumor growth of PAN02 tumor-bearing mice in combination with the axitinib.
Example 8 in vivo efficacy test of the Compound of the invention in mice
1. Purpose of experiment
Taking a C57BL/6 mouse as an experimental animal, subcutaneously inoculating a mouse melanoma cell B16F10, and when the average volume of tumors grows to 100mm3, respectively giving the compound 17, the compound 23, the compound 47 and the compound 58 of the example in combination with cyclophosphamide, and determining the tumor inhibition effect of the compound in combination with cyclophosphamide on the B16F10 tumor-bearing mice.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 17, example compound 23, example compound 47, example compound 58
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% of N, N-dimethylacetamide (Guozu, cat# 30066818), 30% of PEG400 (Taitan, cat#G 41713B) and 65% of 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasonic, and making into suspension.
2.3 operation
2.3.1 Experimental procedure
Under aseptic condition, the mouse melanoma cells B16F10 (from cell site of Chinese academy) in logarithmic growth phase are digested into single cells, mixed with Matrigel (Corning, cat# 354234), transplanted under the right side of the back of C57BL/6 mouse, and each mouse is inoculated with 1×10 5 Cells, 100uL in volume, after inoculation, until the mice tumors grow to an average of 100mm 3 When left and right, mice were randomly divided into 12 groups, 6 drug efficacy experiments were performed in each group, the positive control group was INCB024360 (and Kat #1204669-58-8, chemical engineering), and the negative control group was given an equal amount of vehicle. The specific design is shown in Table 10.
Table 10: in vivo efficacy experiment of Compounds
2.3.2 determination of tumor inhibition Rate
Mouse tumor volume = length x width/2 (mm) calculated according to the formula 3 ) Formula of tumor inhibition rate = 1- (tumor size of dosing group/tumor size of negative control group) ×100%
2.4 experimental results
The compound of this example can inhibit the tumor growth rate of B16F10 tumor-bearing mice in combination with cyclophosphamide, and the specific data are shown in Table 11 below, and it can be seen from Table 11 below that the compound of the present invention in combination with cyclophosphamide has a more effective tumor growth inhibition effect on B16F10 tumor-bearing mice than the combination of INCB024360 and cyclophosphamide in the control group.
Table 11: tumor growth inhibition rate of B16F10 tumor-bearing mice by combining compound and cyclophosphamide
Conclusion: the compound of the invention has better inhibition effect on the tumor growth of B16F10 tumor-bearing mice in combination with cyclophosphamide
Example 9 in vivo efficacy test of the Compound of the invention in mice
1. Purpose of experiment
Taking Balb/c mice as experimental animals, subcutaneously inoculating 4T1 mice tumor cells (derived from cell line of the department of Chinese sciences), and growing until the average volume of tumor reaches 100mm 3 In this case, the compound 14, the compound 33 and the compound 45 were administered in combination with doxorubicin, respectively, and the tumor-inhibiting effect of the compound of the present invention in combination with doxorubicin on 4T1 tumor-bearing mice was measured.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 14, example compound 33, example compound 45
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% of N, N-dimethylacetamide (Guozu, cat# 30066818), 30% of PEG400 (Taitan, cat#G 41713B) and 65% of 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasonic, and making into suspension.
2.3 operation
2.3.1 Experimental procedure
Under the aseptic condition, 4T1 digestion of breast cancer cells of mice in logarithmic growth phase into single cells are transplanted under the skin of the right side of the back of Balb/c mice, and each mouse is inoculated with 1 multiplied by 10 5 Cells, 100uL in volume, mice tumor grown to an average of 100mm after inoculation 3 When left and right, the mice are randomly divided into 10 groups, 6 drug effect experiments are carried out on each group, the positive control group is INCB024360 (and the chemical industry, cat# 1204669-58-8), and the negative control group is given with the same amount of solvent. The specific design is shown in Table 12.
Table 12: in vivo efficacy experiment of Compounds
2.3.2 determination of tumor inhibition Rate
Calculation of mouse tumor volume according to formulaProduct = length x width/2 (mm) 3 ) Formula of tumor inhibition rate = 1- (tumor size of dosing group/tumor size of negative control group) ×100%
2.4 experimental results
The compound of the present example can inhibit the tumor growth rate of 4T1 tumor-bearing mice in combination with doxorubicin, and the specific data are shown in Table 13 below, and it can be seen from Table 13 below that the compound of the present invention in combination with doxorubicin has a more effective tumor growth inhibition effect on 4T1 tumor-bearing mice model than the combination of control INCB024360 with doxorubicin.
Table 13: tumor growth inhibition rate of 4T1 tumor-bearing mice by combining compound of the invention and doxorubicin
Conclusion: the compound of the invention has better inhibition effect on the tumor growth of the 4T1 tumor-bearing mice in combination with doxorubicin
Example 10 in vivo efficacy test in mice of the Compound of the invention
1. Purpose of experiment
Taking Severe Combined Immunodeficiency (SCID) mice as experimental animals, subcutaneously inoculating B16F10 mice tumor cells, and growing until the average volume of tumor reaches 100mm 3 In this case, the tumor inhibitory effect of the compound of the present invention on B16F10 tumor-bearing mice was measured by administering IFN-gamma in combination with each of example compound 11, example compound 54 and example compound 67.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 11, example compound 54, example compound 67
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% of N, N-dimethylacetamide (Guozu, cat# 30066818), 30% of PEG400 (Taitan, cat#G 41713B) and 65% of 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasonic, and making into suspension.
2.3 operation
2.3.1 Experimental procedure
Under aseptic condition, the mouse melanocyte B16F10 (derived from cell site of Chinese academy of sciences) in logarithmic growth phase was digested into single cell, mixed with Matrigel (Corning, cat # 354234) and transplanted under the back right side of Severe Combined Immunodeficiency (SCID) mouse (purchased from Beijing Vitolihua laboratory animal technologies Co., ltd.), each mouse was inoculated with 1×10 5 Cells, 100uL in volume, were vaccinated for 3 days, and mice were randomly divided into 5 groups of 6 drug efficacy experiments each based on body weight, the positive control group was INCB024360 (and the chemical industry, cat# 1204669-58-8), and the negative control group was given an equal amount of vehicle. The specific design is shown in Table 14.
Table 14: in vivo efficacy experiment of Compounds
2.3.2 determination of tumor inhibition Rate
Mouse tumor volume = length x width/2 (mm) calculated according to the formula 3 ) Formula of tumor inhibition rate = 1- (tumor size of dosing group/tumor size of negative control group) ×100%
2.4 experimental results
The compound of this example can inhibit the tumor growth rate of B16F10 tumor-bearing mice by combining with IFN-gamma, the specific data are shown in the following table 15, and the following table 15 shows that the compound of the invention has more effective tumor growth inhibition effect on B16F10 tumor-bearing mice model than the combination of the control group INCB024360 and IFN-gamma.
Table 15: tumor growth inhibition rate of B16F10 tumor-bearing mice by combining the compound of the invention with IFN-gamma
Conclusion: the compound of the invention has better inhibition effect on the tumor growth of B16F10 tumor-bearing mice in combination with IFN-gamma
Example 11 in vivo efficacy test of the Compounds of the invention in mice Using Anti-PD-1 antibodies
1. Purpose of experiment
Taking a C57BL/6 mouse (purchased from Shanghai Laike laboratory animal Co., ltd.) as a laboratory animal, subcutaneously inoculating MC38 mouse tumor cells (from Nanjac Bai biotechnology Co., ltd.) and when the average volume of the tumor grows to 100mm3, respectively combining the example compound 15, the example compound 18 and the example compound 63 with an Anti-PD-1 antibody, and determining the tumor inhibition effect of the compound of the invention combined with the Anti-PD-1 antibody on the MC38 tumor-bearing mouse.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 15, example compound 18, example compound 63
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% NN dimethylacetamide (Guozu, cat# 30066818), 30% PEG400 (Taitan, cat#G 41713B) and 65% 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasound, and making into suspension.
2.3 operation
2.3.1 Experimental procedure
Under aseptic condition, the colon cancer cells MC38 of the mice in logarithmic growth phase are digested into single cells and mixed with Matrigel (Corning, cat# 354234) to be transplanted under the right back of C57BL/6 mice, each of which is inoculated with 1×10 6 Cells, 100uL in volume, after inoculation, when tumor volume reaches 100mm 3 In the range, tumor-bearing mice were randomly divided into 8 groups, each group was subjected to 6 drug effect experiments, and the specific design is shown in Table 16.
Table 16: in vivo efficacy experiment of Compounds
2.3.2 determination of tumor inhibition Rate
Mouse tumor volume = length x width/2 (mm) calculated according to the formula 3 ) Formula of tumor inhibition rate = 1- (tumor size of dosing group/tumor size of negative control group) ×100%
2.4 experimental results
The compound of this example, in combination with Anti-PD-1 antibody, was able to inhibit the tumor growth rate of MC38 tumor-bearing mice, and the specific data are shown in Table 17 below, as can be seen from Table 17 below, the combination of the compound of the invention with Anti-PD-1 antibody has a more potent tumor growth inhibition effect on MC38 tumor-bearing mice model than the combination of control INCB024360 with Anti-PD-1 antibody.
Table 17: tumor growth inhibition ratio of MC38 tumor-bearing mice by combining compound of the invention and Anti-PD-1 antibody
Conclusion: the compound of the invention combined with the Anti-PD-1 antibody has better inhibition effect on the tumor growth of MC38 tumor-bearing mice
EXAMPLE 12 rat pharmacokinetic experiments of Compounds of the invention
1. Purpose of experiment
The drug concentrations in plasma after oral administration of example 6, example 9, example 11, example 13, example 15, example 17, example 18, example 20, example 24, example 33, example 37, example 45, example 47, example 48, example 51, example 54, example 56, example 58, example 62, example 65, example 67 were determined by LC/MS method using 200-250g,8 week old male SD rats (purchased from Shanghai cetrap-bikava laboratory animals limited) as laboratory animals. The pharmacokinetic behavior of the compound of the invention in rats is studied and its pharmacokinetic profile is evaluated.
2. Experimental protocol
2.1 Compounds for experiments
Determination of example compound 6, example compound 9, example compound 11, example compound 13, example compound 15, example compound 17, example compound 18, example compound 20, example compound 24, example compound 33, example compound 37, example compound 45, example compound 47, example compound 48, example compound 51, example compound 54, example compound 56, example compound 58, example compound 62, example compound 65, example compound 67
2.2 formulation of Compounds
Configuration of the compound: weighing a certain amount of the compound, dissolving in 5% of N, N-dimethylacetamide (Guozu, cat# 30066818), 30% of PEG400 (Taitan, cat#G 41713B) and 65% of 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasonic, and making into suspension.
2.3 operation
Rats were orally administered the above compounds, respectively, 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 12.0, 24.0 hours before and after administration, 0.2ml was collected via the orbit, placed in an anticoagulation tube, centrifuged at 6000 rpm for 10 minutes at 4℃to separate plasma, and stored at-80 ℃.
Specific dosing was performed according to the protocol, details of which are shown in table 18.
Table 18: rat pharmacokinetics test protocol
50. Mu.L of the plasma at the different time points was taken, 150. Mu.L of acetonitrile (Allatin, cat#A998-4) solution containing an internal standard was added, mixed, shaken for 5 minutes, centrifuged at 12000rpm for 5 minutes, 100. Mu.L of the supernatant was taken out, and mixed with 200. Mu.L of water for sample injection analysis.
2.4 experimental results
The content of the compound in the plasma of the mice was determined by liquid chromatography-mass spectrometry (LC/MS/MS). Pharmacokinetic parameters of the post-administration compounds were calculated using the non-compartmental model of the winnonlin5.3 software. The pharmacokinetic profile of the compounds of the invention is shown in table 19.
Table 19: pharmacokinetic parameters of the compounds of the invention
/>
Conclusion: the compound of the invention has better absorption in drug substitution and better pharmacodynamic characteristics.
EXAMPLE 13 acute toxicity test of the Compounds of the invention
1. Purpose of experiment
Example compound 3, example compound 7, example compound 11, example compound 15, example compound 17, example compound 25, example compound 27, example compound 30, example compound 34, example compound 39, example compound 42, example compound 47, example compound 61, example compound 67, example compound 76 were administered orally 1 time to each half of the day of administration, and continuous observation for 14 days including clinical observation, weight and pathology examination was performed using 20-22g, 8-week-old ICR mice as experimental animals.
2. Experimental protocol
2.1 Compounds for experiments
Assay example compound 3, example compound 7, example compound 11, example compound 15, example compound 17, example compound 25, example compound 27, example compound 30, example compound 34, example compound 39, example compound 42, example compound 47, example compound 61, example compound 67, example compound 76
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% of N, N-dimethylacetamide (Guozu, cat# 30066818), 30% of PEG400 (Taitan, cat#G 41713B) and 65% of 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasonic, and making into suspension.
2.3 operation
Mice were observed for acute toxicity following single oral administration of example compound 3, example compound 7, example compound 11, example compound 15, example compound 17, example compound 25, example compound 27, example compound 30, example compound 34, example compound 39, example compound 42, example compound 47, example compound 61, example compound 67, example compound 76, respectively, in each dose group of 10 mice, each half of the male and female animals. The compound of the example was formulated for administration at 200mg/mL, with a volume of 10mL/kg, for 1 administration.
2.4 experimental results
The observation was continued for 14 days after dosing, and no other abnormal manifestations were seen in all animals. The animals given to each group had a slight decrease in body weight on day 2, but showed no significant differences from the vehicle control group (5% N, N dimethylacetamide (Guozu, cat # 30066818), 30% PEG400 (Taitan, cat # G41713B), 65% 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat # H1088B), and no macroscopic lesions were seen in the chest, abdomen, pelvis, and cranial cavity after euthanasia of all animals at the end of the 14 day observation period.
Under the experimental conditions, the LD50 value of the compound of the invention after oral administration is more than 2000mg/kg, and the safety is better.
EXAMPLE 14 rat long-toxicity Pre-test of the Compounds of the invention
1. Purpose of experiment
100-140g SD rats are used as experimental animals, the male and female animals are divided into 4 groups randomly, each group of rats is 10, the example compound 11, the example compound 15, the example compound 47 and the example compound 67 are orally administered, 600mg/kg, 300mg/kg and 100mg/kg of each compound are respectively administered, and the continuous administration is carried out for 28 days, so that the death condition, the poisoning reaction, the weight change, the diet, the appearance, the behavior and the like of the animals are recorded. The animals were continuously observed for appearance, behavioral activity, body weight, faeces and death 32 days after discontinuation of dosing.
2. Experimental protocol
2.1 Compounds for experiments
Determination of example Compound 11, example Compound 15, example Compound 47, example Compound 67
2.2 formulation of Compounds
Weighing a certain amount of the compound, dissolving in 5% NN dimethylacetamide (Guozu, cat# 30066818), 30% PEG400 (Taitan, cat#G 41713B) and 65% 10% (2-hydroxypropyl) beta-cyclodextrin (Allatin, cat#H21088B), mixing by ultrasound, and making into suspension.
2.3 operation
Each group of rats was orally administered with corresponding concentrations of example compound 11, example compound 15, example compound 47, example compound 67 for 28 consecutive days, 10 mice per dose group, each half of the male and female, with a dosing volume of 10mL/kg, and the condition of each group of animals, including the appearance characteristics of the animals, behavioural activities, body weight, faeces and animal death, was observed; after stopping the drug for 32 days, animals were euthanized and the organs of each group of animals were dissected for observation.
2.4 experimental results
The administration was continued for 28 days, and all animals had substantially no abnormalities in behavior, gait, hair, feces, etc. After stopping the medicine for 32 days, dissecting each group of animals for examination, wherein obvious abnormality is not seen on the body surface, and lesions are not seen on each viscera.
The obvious adverse reaction dosage (NOEAL) of the compound is more than 600mg/kg, and the safety is good. Compared with the mice in the control group, the mice in the administration group have no body weight and abnormal behavior within 60 days from the administration day, and the compound of the invention does not show obvious toxicity.
The foregoing description is only of the preferred embodiments of the present invention and is not intended to limit the scope of the present invention. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention are included in the protection scope of the present invention.

Claims (5)

1. A compound or a pharmaceutically acceptable salt, stereoisomer or tautomeric form thereof:
2. a pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, tautomer thereof, and optionally one or more pharmaceutically acceptable carriers and/or diluents.
3. Use of a compound according to claim 1 or a pharmaceutically acceptable salt, stereoisomer, tautomer form thereof or a pharmaceutical composition according to claim 2 for the manufacture of a medicament for the prevention and/or treatment of a disease having the pathological characteristics of IDO-mediated tryptophan metabolic pathways.
4. The use according to claim 3, characterized in that said diseases are selected from the group consisting of infections with the AIDS virus, lyme disease and streptococcal infections, autoimmune diseases, depression, anxiety, cataracts, psychological disorders, cancers.
5. The use according to claim 4, wherein the cancer is selected from breast cancer, cervical cancer, colon cancer, lung cancer, gastric cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, stage IV melanoma, glioma, glioblastoma, hepatocellular carcinoma, mastoid kidney tumor, head and neck tumor, leukemia, lymphoma, myeloma.
CN201811493232.XA 2017-12-08 2018-12-07 IDO inhibitor and application thereof Active CN109897011B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201711294943X 2017-12-08
CN201711294943 2017-12-08
CN201810213011 2018-03-15
CN2018102130116 2018-03-15

Publications (2)

Publication Number Publication Date
CN109897011A CN109897011A (en) 2019-06-18
CN109897011B true CN109897011B (en) 2023-10-31

Family

ID=66943382

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811493232.XA Active CN109897011B (en) 2017-12-08 2018-12-07 IDO inhibitor and application thereof

Country Status (1)

Country Link
CN (1) CN109897011B (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110143955B (en) * 2018-02-11 2023-01-20 中国科学院上海药物研究所 Oxadiazole derivative containing heterocyclic side chain, synthetic method and application thereof
CN111689925A (en) * 2019-03-14 2020-09-22 复旦大学 Indoleamine-2, 3-dioxygenase inhibitor containing sulfonamide structure and preparation method and application thereof
CN112300012A (en) * 2019-07-26 2021-02-02 上海睿瓦科技有限公司 Method for preparing 2-methoxy ethylamine hydrochloride
CN111138425A (en) * 2020-01-06 2020-05-12 中国药科大学 Triazole derivative and preparation method and application thereof
CN112679393A (en) * 2020-12-31 2021-04-20 江苏科本药业有限公司 Preparation method of sofosbuvir impurity

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102164902A (en) * 2008-07-08 2011-08-24 因塞特公司 1,2, 5-oxadiazole as an inhibitor of indoleamine 2, 3-dioxygenase
CN105646389A (en) * 2016-01-28 2016-06-08 中国科学院上海有机化学研究所 Sulfamate serving as indoleamine-2, 3-dioxygenase inhibitor and preparation method and application thereof
CN106456753A (en) * 2014-02-04 2017-02-22 因塞特公司 Combination of a PD-1 antagonist and an ido1 inhibitor for treating cancer
CN106565696A (en) * 2015-10-09 2017-04-19 江苏恒瑞医药股份有限公司 Oxadiazole derivative, preparing method of oxadiazole derivative and application of oxadiazole derivative to medicines
WO2017106062A1 (en) * 2015-12-15 2017-06-22 Merck Sharp & Dohme Corp. Novel compounds as indoleamine 2,3-dioxygenase inhibitors
CN106967005A (en) * 2017-04-07 2017-07-21 钟燕 A kind of compound that can suppress IDO, Its Preparation Method And Use
CN108863976A (en) * 2017-09-11 2018-11-23 郑州泰基鸿诺医药股份有限公司 Compound and its application as IDO regulator
CN109111438A (en) * 2017-06-26 2019-01-01 正大天晴药业集团股份有限公司 amidine compound for IDO inhibitor
CN109574950A (en) * 2017-09-28 2019-04-05 上海翔锦生物科技有限公司 1,2,5- furodiazole derivative and application thereof
CN110128367A (en) * 2018-02-02 2019-08-16 成都海创药业有限公司 A kind of indoles amine -2,3- dioxygenase inhibitor and its preparation method and application
CN110143955A (en) * 2018-02-11 2019-08-20 中国科学院上海药物研究所 Oxadiazole derivatives, synthetic method and its application containing heterocyclic side chain
CN110407765A (en) * 2018-04-28 2019-11-05 上海挚盟医药科技有限公司 1,2,5- furodiazole derivative, preparation method and its application in medicine
CN111116572A (en) * 2018-10-31 2020-05-08 石药集团中奇制药技术(石家庄)有限公司 Oxadiazole derivatives, their preparation and use

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102164902A (en) * 2008-07-08 2011-08-24 因塞特公司 1,2, 5-oxadiazole as an inhibitor of indoleamine 2, 3-dioxygenase
CN106456753A (en) * 2014-02-04 2017-02-22 因塞特公司 Combination of a PD-1 antagonist and an ido1 inhibitor for treating cancer
CN106565696A (en) * 2015-10-09 2017-04-19 江苏恒瑞医药股份有限公司 Oxadiazole derivative, preparing method of oxadiazole derivative and application of oxadiazole derivative to medicines
WO2017106062A1 (en) * 2015-12-15 2017-06-22 Merck Sharp & Dohme Corp. Novel compounds as indoleamine 2,3-dioxygenase inhibitors
CN105646389A (en) * 2016-01-28 2016-06-08 中国科学院上海有机化学研究所 Sulfamate serving as indoleamine-2, 3-dioxygenase inhibitor and preparation method and application thereof
CN106967005A (en) * 2017-04-07 2017-07-21 钟燕 A kind of compound that can suppress IDO, Its Preparation Method And Use
CN109111438A (en) * 2017-06-26 2019-01-01 正大天晴药业集团股份有限公司 amidine compound for IDO inhibitor
CN108863976A (en) * 2017-09-11 2018-11-23 郑州泰基鸿诺医药股份有限公司 Compound and its application as IDO regulator
CN109574950A (en) * 2017-09-28 2019-04-05 上海翔锦生物科技有限公司 1,2,5- furodiazole derivative and application thereof
CN110128367A (en) * 2018-02-02 2019-08-16 成都海创药业有限公司 A kind of indoles amine -2,3- dioxygenase inhibitor and its preparation method and application
CN110143955A (en) * 2018-02-11 2019-08-20 中国科学院上海药物研究所 Oxadiazole derivatives, synthetic method and its application containing heterocyclic side chain
CN110407765A (en) * 2018-04-28 2019-11-05 上海挚盟医药科技有限公司 1,2,5- furodiazole derivative, preparation method and its application in medicine
CN111116572A (en) * 2018-10-31 2020-05-08 石药集团中奇制药技术(石家庄)有限公司 Oxadiazole derivatives, their preparation and use

Also Published As

Publication number Publication date
CN109897011A (en) 2019-06-18

Similar Documents

Publication Publication Date Title
CN109897011B (en) IDO inhibitor and application thereof
JP6916795B2 (en) LSD1 inhibitor
KR102644798B1 (en) Heterocyclic inhibitors of erk1 and erk2 and their use in the treatment of cancer
WO2021143693A1 (en) Aryl or heteroaryl pyridone or pyrimidine derivative, preparation method and use thereof
TWI386405B (en) An imidazole derivative
CA2993929A1 (en) 1,3,4-oxadiazole sulfonamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
JP2020530446A (en) Compounds, compositions, and methods
CA3051645A1 (en) Aryl hydrocarbon receptor (ahr) modulator compounds
AU2016292450B2 (en) Aniline pyrimidine derivatives and uses thereof
AU2006237920A1 (en) Nitrogen-containing heterocyclic compound
KR20230043943A (en) Compounds as CDK7 kinase inhibitors and uses thereof
WO2021249563A1 (en) Aryl or heteroaryl pyridone or pyrimidone derivative, preparation method therefor and application thereof
JP2014521595A (en) Substituted sulfonamides useful as anti-apoptotic Bcl inhibitors
CA2872000A1 (en) Complement pathway modulators and uses thereof
CN107001271B (en) Hydroxyamidines analog derivative, preparation method and its application in medicine
CA3054826A1 (en) Mk2 inhibitors, synthesis thereof, and intermediates thereto
CA2901770A1 (en) Acyclic cyanoethylpyrazolo pyridones as janus kinase inhibitors
CA3054953A1 (en) Haloallylamine pyrazole derivative inhibitors of lysyl oxidases and uses thereof
JP2017512796A (en) Substituted nitrogen-containing heterocyclic derivatives, pharmaceutical compositions containing them, and their antitumor applications
JP2022540580A (en) Tetrahydroisoquinoline compound
JP2023527242A (en) Pyrimidine compounds that are AXL inhibitors
CN115991706A (en) PIM kinase inhibitors
JP6795525B2 (en) Arylsulfonamide compounds as carbonic anhydrase inhibitors and their therapeutic use
JP6454413B2 (en) Aminosulfonyl compounds, methods for their production, and uses
CA2815055A1 (en) Viral polymerase inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant