WO2017167150A1 - 一种3-氟吡啶杂环化合物及其应用 - Google Patents
一种3-氟吡啶杂环化合物及其应用 Download PDFInfo
- Publication number
- WO2017167150A1 WO2017167150A1 PCT/CN2017/078305 CN2017078305W WO2017167150A1 WO 2017167150 A1 WO2017167150 A1 WO 2017167150A1 CN 2017078305 W CN2017078305 W CN 2017078305W WO 2017167150 A1 WO2017167150 A1 WO 2017167150A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- group
- substituted
- alkyl
- hydrogen atom
- unsubstituted
- 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.)
- Ceased
Links
- 0 C*(*=C(C)C=C1)C1=O Chemical compound C*(*=C(C)C=C1)C1=O 0.000 description 3
- ULZCOWMSBOJCLT-UHFFFAOYSA-N CN(CC1)CCS1(=O)=O Chemical compound CN(CC1)CCS1(=O)=O ULZCOWMSBOJCLT-UHFFFAOYSA-N 0.000 description 1
- MKWDLLNKRKUEHO-UHFFFAOYSA-N CN(CC1)CCS1=O Chemical compound CN(CC1)CCS1=O MKWDLLNKRKUEHO-UHFFFAOYSA-N 0.000 description 1
- KDTVWEHAAISPNW-UHFFFAOYSA-N CN1CCSCC1 Chemical compound CN1CCSCC1 KDTVWEHAAISPNW-UHFFFAOYSA-N 0.000 description 1
- DIQYDDOLHDIWQK-UHFFFAOYSA-N CS(CCC[NH-])(=O)=O Chemical compound CS(CCC[NH-])(=O)=O DIQYDDOLHDIWQK-UHFFFAOYSA-N 0.000 description 1
- WCQFINPOEGRJKR-UHFFFAOYSA-N CS(CC[NH-])(=O)=O Chemical compound CS(CC[NH-])(=O)=O WCQFINPOEGRJKR-UHFFFAOYSA-N 0.000 description 1
- JYYNAJVZFGKDEQ-UHFFFAOYSA-N Cc1cc(C)ncc1 Chemical compound Cc1cc(C)ncc1 JYYNAJVZFGKDEQ-UHFFFAOYSA-N 0.000 description 1
- ITQTTZVARXURQS-UHFFFAOYSA-N Cc1cccnc1 Chemical compound Cc1cccnc1 ITQTTZVARXURQS-UHFFFAOYSA-N 0.000 description 1
- LSZQMSSIUQNTDX-UHFFFAOYSA-N Cc1ccn[n]1C Chemical compound Cc1ccn[n]1C LSZQMSSIUQNTDX-UHFFFAOYSA-N 0.000 description 1
- FKNQCJSGGFJEIZ-UHFFFAOYSA-N Cc1ccncc1 Chemical compound Cc1ccncc1 FKNQCJSGGFJEIZ-UHFFFAOYSA-N 0.000 description 1
- BSKHPKMHTQYZBB-UHFFFAOYSA-N Cc1ncccc1 Chemical compound Cc1ncccc1 BSKHPKMHTQYZBB-UHFFFAOYSA-N 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
- A61K31/444—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/47—Quinolines; Isoquinolines
- A61K31/4709—Non-condensed quinolines and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/50—Pyridazines; Hydrogenated pyridazines
- A61K31/502—Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- Y—GENERAL 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
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- the invention relates to a 3-fluoropyridine heterocyclic compound and an application thereof, and belongs to the technical field of medicine.
- the Wnt gene was discovered in 1982 and was originally identified as a site for preferential integration of mouse mammary tumor virus. This gene is capable of transmitting proliferation and differentiation information between cells. It is an oncogene and was named the Int gene at that time. Mouse Int-1 and Int-3). It was subsequently found to belong to the same source gene (orthologous gene) as the wingless gene of Drosophila, thereby naming the two as the Wnt gene.
- the Wnt pathway is a very conserved signaling pathway.
- Members of the lower biological flies to higher mammals have a high degree of homology.
- the Wnt signaling pathway is involved in the regulation of a variety of biological processes, including embryo growth and morphogenesis, tissue stability, energy metabolism balance, and stem cell maintenance (Logan et al., Annu. Rev. Cell. Dev. Biol. , 2004, 20, 781-810).
- Wnt signaling pathway plays an important role in the regulation of epidermal stem cells, intestinal stem cells, hematopoietic stem cells, neural stem cells, embryonic stem cells, and cancer stem cells (Reya et al., Nature, 2005, 434, 843-850).
- the classical Wnt signaling pathway is driven by the extracellular ligand Wnts protein simultaneously binding to the 7-cell transmembrane Frizzled receptor and the helper receptor LRP5/6 to initiate Wnt/ ⁇ -catenin signaling pathway and activate cytoplasmic Dsh protein
- activated Dsh protein can inhibit the activity of GSK-3 ⁇ , a key component in the degradation complex formed by APC protein, GSK-3 ⁇ , Axin, ⁇ -catenin, etc., so that ⁇ -catenin is not phosphorylated by GSK-3 ⁇ .
- the recognition and degradation of the ubiquitin proteasome is avoided, which in turn accumulates in the cytoplasm (Boutros et al, Mech.
- ⁇ -catenin When ⁇ -catenin accumulates to a certain concentration in the cytoplasm, it begins to transfer to the nucleus, and binds to the transcription factor TCF/LEFs in the nucleus, causing the promoter of the downstream target gene of ⁇ -catenin to be exposed and activated. Activation of c-myc, cyclin-D1, survivin, gasstrin, VEGF, ASEF, etc. leads to abnormal cell proliferation.
- cytoplasmic ⁇ -catenin binds to the membrane adhesion proteins E-cadherin and ⁇ -catenin to form a complex that participates in the regulation of the cytoskeleton, maintains the adhesion of the same type of cells, and prevents cell migration.
- a small amount of free ⁇ -catenin is recognized and degraded by the ubiquitin proteasome after being phosphorylated by the degradation complex in the cytoplasm, keeping the intracellular The ⁇ -catenin is in a low level state, thereby turning the Wnt signaling pathway off.
- Wnt gene itself or any other member of the pathway changes to cause abnormal activation, it may cause tumors.
- regulators of the Wnt pathway including mutations in genes such as APC, ⁇ -catenin, Axin, and TCF, are widely present in colon cancer patients, resulting in overexpression of growth-related genes (Klaus et al., Nat. Rev. Cancer, 2008, 8, 387-398).
- Lozzo et al. found that overexpression of Wnt-5a mRNA, especially in breast cancer, was found in breast cancer, lung cancer, prostate cancer, and melanoma by studying more than 100 normal and tumor tissues and 10 human tumor cell lines. Abnormally apparent (Lozzo et al., Cancer Research, 1995, 55, 3495).
- Wnt signaling pathway has a better anti-tumor targeting effect.
- Activation of the Wnt signaling pathway by ⁇ -catenin increases the circulation and expansion of neural progenitor cells, whereas deletions result in the loss of progenitor cell spacing (Chenn et al., Science, 2002, 297, 365-369).
- Abnormal activation of the Wnt signal is tumorigenic in the nervous system (Dahmen et al, Cancer Res., 2001, 61, 7039-7043).
- the Wnt signaling pathway promotes the renewal and maintenance of pluripotent hematopoietic stem cells, and abnormal Wnt signaling is responsible for various diseases and other blood-related cancers caused by pluripotent hematopoietic stem cells (Reya et al, Nature, 2005, 434, 843- 850; Willert et al, Nature, 2003, 423, 448-452).
- Deregulation of Wnt signaling also leads to the development of diabetic retinopathy by inducing retinal inflammation, vascular leakage, and neovascularization.
- tumor immunotherapy has become a research hotspot.
- the main strategy of tumor immunotherapy is to block the activation of immunosuppressive receptors on T cells using immunological checkpoint inhibitors such as CTLA-4, PD-1 and PD-L1 monoclonal antibodies.
- This strategy is effective in patients with a variety of tumors, such as melanoma and lung cancer (especially T cell infiltrating tumors), whereas in tumor tissue without T cell infiltration, it is often anti-tumor immunity.
- the object of the present invention is to propose a 3-fluoropyridine heterocyclic compound and its use, which can effectively antagonize the Wnt signaling pathway and can be used for treating or preventing disorders caused by Wnt signaling pathway disorders. .
- A is a 5-14 membered heteroaryl ring having 1 to 4 hetero atoms, a 6-12 membered aromatic ring, a 3-6 membered cycloalkyl group or 3-substituted or substituted with 1 to 6 R 6 groups.
- a 6-membered heterocycloalkyl group, the hetero atom of the heteroaryl ring and the heterocycloalkyl group comprising one or more of N, O and S; or A being unsubstituted or 1-2 R 6 groups replaced Wherein X 1 is N or CR 6 ;
- B is selected from a hydrogen atom, a cyano group, a halogeno group, a hydroxyl group, a C 3-8 cycloalkyl group, a C 2-8 alkenyl group or a C 2-8 alkynyl group, a C 1-8 sulfonyl group, a C 1-8 sulfonamide.
- B is selected from a benzene ring which is unsubstituted or substituted with 1-3 R 9 groups, unsubstituted or substituted with 1-3 R 10 groups 5-6 membered heteroaromatic ring containing 1-3 heteroatoms, unsubstituted or substituted with 1-3 R 11 groups containing 5-7 membered 1-3 a saturated heterocyclic ring of a hetero atom, the hetero atom of the heteroaromatic ring and the saturated heterocyclic ring including one or more of N, O and S;
- U 6 is unsubstituted or substituted with R 13 6-12 membered aromatic ring, unsubstituted or substituted with 1-6 R 13 groups substituted 5-14 membered heteroaryl ring, unsubstituted or 1-6 R 13 groups, and substituted with 5-7 membered heterocyclic ring, unsubstituted or substituted with 1-6 R 13 substituted 5-7 membered aromatic heterocycle and 5-6 membered heteroaryl a ring, -OR 14 , -NR 14 R 15 , wherein the heterocyclic or heteroaromatic ring is a heterocyclic or heteroaromatic ring containing from 1 to 4 heteroatoms, and the hetero atom of the heterocyclic or heteroaromatic ring includes N Or one or more of O and S; or U is a C 2-8 alkenyl or C 2-8 alkynyl group substituted by a 5-6 membered aromatic or heteroaryl ring, said heterocyclic ring, heteroaromatic ring a heterocyclic
- L is the price key or Wherein Y is a valence bond, -O-, -S-, -N(R 18 )- or -C(R 18 )(R 19 )-;
- R 1, R 2, R 3 are each independently selected from hydrogen atom, C 1-6 alkyl, 1 to 3 groups independently selected from halo, hydroxy, cyano, C 1-3 alkyl, C 3- a C 1-6 alkyl group substituted with a 5- cycloalkyl group and a C 1-3 alkoxy group;
- R 4 and R 5 are each independently selected from the group consisting of a hydrogen atom, a cyano group, an amino group, a halogen group, a C 1-8 alkyl group, a C 3-8 cycloalkyl group, a C 1-8 alkoxy group, and -NH-C 1-3. alkyl;
- R 6 is selected from a hydrogen atom, a halogeno group, a cyano group, a hydroxyl group, an amino group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkoxy group; or R 6 is selected from the group consisting of 1-3 An amino group substituted with a substituent selected from a halogeno group, a C 1-3 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group or a C 1-6 alkoxy group base;
- R 7 and R 8 are each independently selected from a hydrogen atom which is unsubstituted or substituted with 1-3 R 12 , a C 1-8 alkyl group, a C 3-8 cycloalkyl group; or an atom to which R 7 and R 8 are bonded together Loop into
- R 9 and R 10 are each independently selected from a hydrogen atom, a halogeno group, a cyano group, a hydroxyl group, an amino group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkoxy group; or R 9 , R 10 each selected from an amino group substituted with 1 to 3 substituents selected from a halogeno group, a C 1-3 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group Or a C 1-6 alkoxy group;
- R 11 is selected from a hydrogen atom, a halogen group, a cyano group, a hydroxyl group, an amino group, a C 1-6 alkoxy group, a C 3-6 cycloalkyl group, an oxo group, a C 1 which is unsubstituted or substituted by a halogen group. -6 alkyl;
- R 12 is selected from the group consisting of a hydrogen atom, a cyano group, a hydroxyl group, an amino group, a C 1-6 alkoxy group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 2-6 ureido group, and a C 2-6 oxo group.
- Urea -S(O) 2 -C 1-3 alkyl, -N(R 16 )S(O) 2 -C 1-3 alkyl, -N(R 16 )C(O)-C 1- 3- alkyl;
- R 13 is selected from the group consisting of a hydrogen atom, a cyano group, a halogeno group, a hydroxyl group, an amino group, a C 1-6 alkoxy group, a C 3-6 cycloalkyl group, a -S(O) 2 -C 1-3 alkyl group, -C (O)-C 1-3 alkyl, -C(O)OC 1-3 alkyl, oxo, C 1-6 alkyl unsubstituted or substituted by halo;
- R 14 and R 15 are each independently selected from the group consisting of a hydrogen atom, a C 3-6 cycloalkyl group, a C 1-6 alkyl group, a 5-6 membered aromatic ring, a 5-6 membered heteroaryl ring having 1-3 hetero atoms, and 5 6-membered heterocyclic ring having 1-3 heteroatoms, 20 is substituted with 1-3 R C 3-6 cycloalkyl, 20 substituted with 1-3 R C 1-6 alkyl, 1- 3 R 20 substituted 5-6 membered aromatic ring substituted with 1 to 3 R 20 is 5-6 membered heteroaromatic ring containing 1-3 heteroatoms, and is substituted with 1-3 R & lt 20 5-6 a heterocyclic ring containing 1-3 heteroatoms; the hetero atom comprising one or more of N, O and S; or R 14 and R 15 together with the N atom to which they are attached;
- R 16 , R 17 , R 18 , and R 19 are each independently selected from a hydrogen atom, a C 1-6 alkyl group which is unsubstituted or substituted with a halogen group;
- R 20 is selected from the group consisting of a hydrogen atom, a cyano group, a hydroxyl group, an amino group, a C 1-6 alkoxy group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a 5-6 membered aromatic ring, and a 5-6 member containing 1 a heteroatom ring of 3 heteroatoms comprising one or more of N, O and S;
- R 21 is selected from a hydrogen atom or a C 1-6 alkyl group.
- B is selected from the group consisting of a hydrogen atom, a cyano group, a halogeno group, a hydroxyl group, a C 1-8 sulfonyl group, a C 1-8 sulfonylamino group, a C 1-8 alkoxy group, a C 1-8 acyl group, and a C 1-8 amide group.
- B is selected from benzene which is unsubstituted or substituted with 1-3 R 9 groups ring, unsubstituted or substituted with 1 to 3 R 10 groups substituted with a 5-6 membered heteroaromatic ring containing 1-3 heteroatoms, unsubstituted or substituted with 1-3 R 11 groups 5- a 7-membered saturated heterocyclic ring having 1 to 3 hetero atoms, the hetero atom of the heteroaromatic ring and the saturated heterocyclic ring including one or more of N, O and S;
- R 1 is a hydrogen atom
- R 2 and R 3 are each independently selected from a hydrogen atom and a C 1-6 alkyl group
- R 4 and R 5 are each independently selected from a hydrogen atom, a cyano group, a halogeno group, and a C 1-8 alkyl group;
- R 6 is selected from a hydrogen atom, a halogeno group, a cyano group, a hydroxyl group, an amino group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkoxy group; or R 6 is selected from the group consisting of 1-3 An amino group substituted with a substituent selected from a halogeno group, a C 1-3 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group or a C 1-6 alkoxy group base;
- R 7 and R 8 are each independently selected from the group consisting of a hydrogen atom, a C 3-8 cycloalkyl group, a C 1-8 alkyl group unsubstituted or substituted with 1-3 R 12 groups; or R 7 and R 8 together with an atom to which they are attached Loop into
- R 9, R 10 are independently selected from hydrogen, halo, cyano, C 1-6 alkyl, C 1-6 alkoxy; or R 9, R 10 are independently selected from 1-3 halo groups Substituted C 1-6 alkyl or C 1-6 alkoxy;
- R 11 is selected from a hydrogen atom, an oxo group, a C 1-6 alkyl group which is unsubstituted or substituted by a halogen group;
- R 12 is selected from a hydrogen atom, a cyano group, C 1-6 alkyl, C 1-6 sulfonamido group;
- R 13 is selected from the group consisting of a hydrogen atom, a cyano group, a halogeno group, an oxo group, a C 1-6 alkoxy group, a C 3-6 cycloalkyl group, a C 1-6 acyl group, which is unsubstituted or substituted by a halogen group.
- R 14 and R 15 are each independently selected from a hydrogen atom, a C 3-6 cycloalkyl group, a C 1-6 alkyl group which is unsubstituted or substituted with 1-3 R 20 groups; or R 14 and R 15 together with the N to which they are attached Atom into a ring;
- R 16 , R 17 , R 18 , R 19 are each independently selected from a hydrogen atom, a C 1-6 alkyl group which is unsubstituted or substituted with a halogen group;
- R 20 is selected from the group consisting of a hydrogen atom, a cyano group, a hydroxyl group, a C 1-6 alkoxy group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a 5-6 membered aromatic ring, and a 5-6 member having 1-3.
- a heteroatom heteroaromatic ring comprising one or more of N, O and S;
- R 21 is selected from a hydrogen atom or a C 1-6 alkyl group.
- A is one of the following groups which are unsubstituted or substituted by 1-6 R 6 groups:
- R 6 is as defined above.
- A is one of the following groups which are unsubstituted or substituted by one R 6 group:
- R 6 is as defined above.
- A is one of the following groups which are unsubstituted or substituted by one R 6 group:
- R 6 is as defined above.
- A is one of the following groups that are unsubstituted or substituted with one R 6 group:
- R 6 is a hydrogen atom, a halogeno group (preferably F), a cyano group or a C 1-6 alkoxy group (preferably a methoxy group).
- B is one of the following groups:
- n 0, 1, or 2
- R 10 is as defined above.
- n 0, 1, or 2
- R 10 is as defined above.
- Scheme 8 Among the 3-fluoropyridine heterocyclic compounds of Scheme 6, most preferably, B is Wherein n is 1, and R 10 is a C 1-6 alkyl group (preferably a methyl group).
- L is a valence bond.
- the 3-fluoropyridine heterocyclic compound in a 3-fluoropyridine heterocyclic compound according to any one of the above aspects 1 to 9, preferably, the 3-fluoropyridine heterocyclic compound, and a pharmaceutically acceptable salt thereof, have the formula II structure:
- X 3 is N or CR 22 ;
- Y 1 , Y 2 , Y 3 , Y 4 are each independently selected from N or CR 6 ;
- U 6 is unsubstituted or substituted with R 13 6-12 membered aromatic ring, unsubstituted or substituted with 1-6 R 13 groups substituted 5-14 membered heteroaryl ring, unsubstituted or 1-6 R 13 groups, and substituted with 5-7 membered heterocyclic ring, unsubstituted or substituted with 1-6 R 13 substituted 5-7 membered aromatic heterocycle and 5-6 membered heteroaryl a ring, -OR 14 or -NR 14 R 15 , wherein the heterocyclic or heteroaromatic ring is a heterocyclic or heteroaromatic ring containing from 1 to 4 heteroatoms, and the hetero atom of the heterocyclic or heteroaromatic ring includes N Or one or more of O and S; or U is a C 2-8 alkenyl or C 2-8 alkynyl group substituted by a 5-6 membered aromatic or heteroaryl ring, said heterocyclic ring, heteroaromatic ring a heterocyclic or
- L is a valence bond
- R 1 and R 4 are a hydrogen atom
- R 2 and R 3 are each independently a hydrogen atom or a C 1-3 alkyl group
- R 5 is selected from the group consisting of a hydrogen atom, a halogeno group, a cyano group, and a C 1-3 alkyl group;
- R 6 is selected from the group consisting of a hydrogen atom, a halogeno group, a cyano group, a hydroxyl group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkoxy group; or R 6 is selected from the group consisting of 1-3 from halo, C 1-3 alkyl, C 3-6 cycloalkyl substituted with a substituent C 1-6 alkyl, C 3-6 cycloalkyl or C 1-6 alkoxy;
- R 10 is selected from a hydrogen atom, a halogeno group, a C 1-6 alkyl group which is unsubstituted or substituted by a halogen group;
- R 13 is selected from the group consisting of a hydrogen atom, a cyano group, a halogeno group, an oxo group, a C 1-6 alkoxy group, a C 3-6 cycloalkyl group, a C 1-6 acyl group, which is unsubstituted or substituted by a halogen group.
- R 14 and R 15 are each independently selected from a hydrogen atom, a C 3-6 cycloalkyl group, a C 1-6 alkyl group which is unsubstituted or substituted with 1-3 R 20 groups; or R 14 and R 15 together with the N to which they are attached Atom into a ring;
- R 20 is selected from the group consisting of a hydrogen atom, a cyano group, a hydroxyl group, a C 1-6 alkoxy group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a 5-6 membered aromatic ring, and a 5-6 member having 1-3.
- a heteroatom heteroaromatic ring comprising one or more of N, O and S;
- R 21 and R 22 are each independently selected from a hydrogen atom or a C 1-6 alkyl group.
- Scheme 11 In the 3-fluoropyridine heterocyclic compound of Scheme 10, preferably, for Wherein the group is unsubstituted or substituted by 1 R 6 wherein R 6 is a hydrogen atom, a halo group (preferably F), a cyano group or a C 1-6 alkoxy group (preferably a methoxy group). .
- R 6 is a hydrogen atom, a halo group (preferably F), a cyano group or a C 1-6 alkoxy group (preferably a methoxy group).
- 3-fluoropyridine heterocyclic compound according to any one of the above aspects 1 to 11, preferably, the 3-fluoropyridine heterocyclic compound, and a pharmaceutically acceptable salt thereof, have the formula III Structure:
- Y 5 , Y 6 , Y 7 , Y 8 , Y 9 , Y 10 are each independently selected from N or CR 6 ;
- B is a cyano group
- U 6 is unsubstituted or substituted with R 13 6-12 membered aromatic ring, unsubstituted or substituted with 1-6 R 13 groups substituted 5-14 membered heteroaryl ring, unsubstituted or 1-6 R 13 groups, and substituted with 5-7 membered heterocyclic ring, unsubstituted or substituted with 1-6 R 13 substituted 5-7 membered aromatic heterocycle and 5-6 membered heteroaryl a ring, -OR 14 or -NR 14 R 15 , wherein the heterocyclic or heteroaromatic ring is a heterocyclic or heteroaromatic ring containing from 1 to 4 heteroatoms, and the hetero atom of the heterocyclic or heteroaromatic ring includes N Or one or more of O and S; or U is a C 2-8 alkenyl or C 2-8 alkynyl group substituted by a 5-6 membered aromatic or heteroaryl ring, said heterocyclic ring, heteroaromatic ring a heterocyclic or
- L is a valence bond
- R 1 and R 4 are a hydrogen atom
- R 2 and R 3 are each selected from a hydrogen atom and a methyl group
- R 5 is selected from the group consisting of a hydrogen atom, a halogeno group, a cyano group, and a C 1-3 alkyl group;
- R 6 is selected from the group consisting of a hydrogen atom, a halogeno group, a cyano group, a hydroxyl group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkoxy group; or R 6 is selected from the group consisting of 1-3 from halo, C 1-3 alkyl, C 3-6 cycloalkyl substituted with a substituent C 1-6 alkyl, C 3-6 cycloalkyl or C 1-6 alkoxy;
- R 13 is selected from the group consisting of a hydrogen atom, a cyano group, a halogeno group, an oxo group, a C 1-6 alkoxy group, a C 3-6 cycloalkyl group, a C 1-6 acyl group, which is unsubstituted or substituted by a halogen group.
- R 14 and R 15 are each independently selected from a hydrogen atom, a C 3-6 cycloalkyl group, a C 1-6 alkyl group which is unsubstituted or substituted with 1-3 R 20 groups; or R 14 and R 15 together with the N to which they are attached Atom into a ring;
- R 20 is selected from the group consisting of a hydrogen atom, a cyano group, a hydroxyl group, a C 1-6 alkoxy group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a 5-6 membered aromatic ring, and a 5-6 member having 1-3.
- a heteroatom heteroaromatic ring comprising one or more of N, O and S;
- R 21 is a hydrogen atom or a C 1-6 alkyl group.
- U is one of the following groups which are unsubstituted or substituted by 1 to 6 R 13 groups. :
- R 13 is as defined above.
- R 13 is as defined above.
- R 13 is halogen (preferably F or Cl), C 1-6 alkyl (preferably methyl), C 1-6 alkoxy (preferably methoxy) or cyano.
- the 3-fluoropyridine heterocyclic compound in a 3-fluoropyridine heterocyclic compound according to any one of the above aspects 1 to 13, preferably, the 3-fluoropyridine heterocyclic compound, and a pharmaceutically acceptable salt thereof, have Formula IV or Formula V The structure shown:
- X 4 , X 5 , X 6 , X 7 , X 8 , X 9 are each independently selected from N or CR 13 ;
- L is a valence bond
- R 1 and R 4 are a hydrogen atom
- R 2 and R 3 are each independently a hydrogen atom or a C 1-3 alkyl group
- R 5 is selected from the group consisting of a hydrogen atom, a halogeno group, a cyano group, and a C 1-3 alkyl group;
- R 6 is selected from the group consisting of a hydrogen atom, a halogeno group, a cyano group, a hydroxyl group, a C 1-6 alkyl group, a C 3-6 cycloalkyl group, a C 1-6 alkoxy group; or R 6 is selected from the group consisting of 1-3 from halo, C 1-3 alkyl, C 3-6 cycloalkyl substituted with a substituent C 1-6 alkyl, C 3-6 cycloalkyl or C 1-6 alkoxy;
- R 10 is selected from a hydrogen atom, a halogeno group, a C 1-6 alkyl group which is unsubstituted or substituted by a halogen group;
- R 13 is selected from the group consisting of a hydrogen atom, a cyano group, a halogeno group, a C 1-6 alkoxy group, a C 3-6 cycloalkyl group, a C 1-6 alkyl group which is unsubstituted or substituted with a halogen group;
- R 23 and R 24 are each independently selected from a hydrogen atom, a C 1-3 alkyl group; or R 23 and R 24 are combined into an oxo group;
- R 25 , R 26 , R 27 and R 28 are each independently selected from a hydrogen atom or a C 1-3 alkyl group.
- the 3-fluoropyridine heterocyclic compound includes one or more of the following compounds:
- the present invention also provides a pharmaceutical composition
- a pharmaceutical composition comprising a therapeutically effective amount of the above 3-fluoropyridine heterocyclic compound and a pharmaceutically acceptable carrier therefor.
- the present invention also provides a pharmaceutical combination product of a 3-fluoropyridine heterocyclic compound, which comprises the 3-fluoropyridine heterocyclic compound of the present invention or a pharmaceutically acceptable salt thereof, and an antitumor drug, an antibacterial drug, and an antiviral agent.
- a combination of one or a combination of a drug, an anti-parasitic drug, a central nervous system drug, an anti-osteogenesis drug, and a diabetes drug is used in combination.
- the present invention also provides the use of the above 3-fluoropyridine heterocyclic compound for the preparation of a medicament for treating or preventing a disease associated with abnormal Wnt signaling pathway;
- the diseases associated with abnormal Wnt signaling pathway include breast cancer, lung cancer, Bladder cancer, pancreatic cancer, liver cancer, head and neck squamous cell carcinoma, thyroid cancer, sarcoma, osteosarcoma, dural fibroma, melanoma, prostate cancer, colorectal cancer, ovarian cancer, cervical cancer, esophageal cancer, gastric cancer, myeloma, Lymphoma, mantle cell lymphoma, cutaneous T-cell lymphoma, chronic and non-progressive anemia, spontaneous or essential thrombocytosis, idiopathic bone Myeloid fibrosis, pulmonary fibrosis, renal fibrosis, liver fibrosis, cirrhosis, diabetic retinopathy, macroglobulinemia, leukemia, acute
- the above pharmaceutical composition means a composition comprising the above 3-fluoropyridine heterocyclic compound and a pharmaceutically acceptable salt or carrier thereof as a component, or the above 3-fluoropyridine heterocyclic compound and pharmaceutically acceptable thereof a combination of the accepted salt and a combination of one or more of an anti-tumor drug, an antibacterial drug, an antiviral drug, an anti-parasitic drug, a central nervous system drug, an anti-osteogenesis drug, and a diabetes drug Things.
- the 3-fluoropyridine heterocyclic compound of the present invention is capable of effectively antagonizing the Wnt signaling pathway and can be used for treating or preventing a disorder caused by a malfunction of the Wnt signaling pathway or a disease associated with abnormal Wnt signaling pathway.
- Fig. 2 is a graph showing the results of cytostatic test of 3-fluoropyridine heterocyclic compound B7 in Example 25.
- Alkyl means a straight or branched saturated aliphatic hydrocarbon group.
- the alkyl group includes an alkyl group containing 1 to 18, such as 1 to 12, further such as 1 to 10, still more preferably 1 to 6 carbon atoms, examples of which are selected from methyl, ethyl, 1-propyl or N-propyl ("n-Pr"), 2-propyl or isopropyl ("i-Pr"), 1-butyl or n-butyl (“n-Bu”), 2-methyl-1- Propyl or isobutyl (“i-Bu”), 1-methylpropyl or sec-butyl (“s-Bu”) and 1,1-dimethylethyl or tert-butyl ("t-Bu” ").
- alkyl groups may be selected from the group consisting of 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-1-Butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl (-CH, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4- Methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3-dimethyl-2-butyl and 3,3-dimethyl-2 - butyl.
- Alkynyl means a straight or branched hydrocarbon radical comprising at least one C ⁇ C triple bond and 2-18 such as 2-8, 2-6 or 2-4 carbon atoms.
- alkynyl groups include ethynyl, 1-propynyl, propargyl, 1-butynyl, 2-butynyl and 3-butynyl.
- Cycloalkyl means a saturated or partially unsaturated, cyclic aliphatic hydrocarbon group including monocyclic and polycyclic (eg, bicyclic and tricyclic) groups.
- a cycloalkyl group can contain 3-12, such as 3-8, further such as 3-6, 3-5 or 3-4 carbon atoms.
- examples of cycloalkyl groups are selected from monocyclic rings containing from 3 to 12, such as from 3 to 8, from 3 to 6 carbon atoms.
- Examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, Cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclodecyl , cyclodecyl, cyclodecyl and cyclododeyl.
- bicyclic cycloalkyl group examples include a bicyclic cycloalkyl group having 7 to 12 carbon atoms, which are arranged to be selected from [4, 4], [4, 5], [5, 5], [5, 6] And a bicyclic ring of the [6,6] ring system, or a bridged bicyclic ring selected from the group consisting of bicyclo [2.2.1] heptane, bicyclo [2.2.2] octane, and bicyclo [3.2.2] decane .
- Other examples of bicyclic cycloalkyl groups include those arranged in a bicyclic ring selected from the group consisting of the [5,6] and [6,6] ring systems.
- the ring may be saturated or have at least one double bond (ie, partially unsaturated), but not fully conjugated.
- Aryl or “aryl ring” means a monocyclic or bicyclic aromatic hydrocarbon group containing from 6 to 10 carbon atoms.
- Examples of the aryl or aromatic ring include a phenyl or benzene ring, a naphthyl group or a naphthalene ring (including 1-naphthyl or 2-naphthyl).
- a divalent group formed from a substituted benzene derivative and having a free valence at a ring atom is designated as a substituted phenylene group.
- the divalent group derived from a monovalent polycyclic hydrocarbon group designated by a "- group” is removed by removing a hydrogen atom from a carbon atom having a free valence as follows: a "-subunit" is added to the corresponding monovalent group, for example A naphthyl group having two linked sites is referred to as a naphthylene group.
- the aryl group does not encompass or overlap the heteroaryl group in any way, as defined separately below.
- Halogen or "halo” or “halo” means F, Cl, Br or I.
- Heteroaryl or “heteroaryl” refers to a group selected from the group consisting of:
- a -5-7 membered aromatic monocyclic ring comprising at least one hetero atom selected from N, O and S, for example from 1 to 4, or in some embodiments from 1 to 3 heteroatoms, the remaining ring atoms being carbon ;
- -8-12 membered bicyclic ring comprising at least one heteroatom selected from N, O and S, for example from 1 to 4, or in some embodiments from 1 to 3, or in other embodiments 1 Or 2 heteroatoms, the remaining ring atoms being carbon, and wherein at least one of the rings is aromatic and at least one heteroatom is present in the aromatic ring;
- a -11-14 membered tricyclic ring comprising at least one heteroatom selected from N, O and S, for example from 1 to 4, or in some embodiments from 1 to 3, or in other embodiments 1 Or 2 heteroatoms, the remaining ring atoms are carbon, and at least one of the rings is aromatic and at least one heteroatom is present in the aromatic ring.
- the hetero atoms in the heteroaryl group exceeds 1, the hetero atoms are not adjacent to each other. In some embodiments, the total number of S and O atoms in the heteroaryl group is at most 2. In some embodiments, the total number of S and O atoms in the aromatic heterocycle is at most one.
- heteroaryl groups include, but are not limited to, (numbered starting from the position of the linkage designated as priority 1), pyridyl (such as 2-pyridyl, 3-pyridyl or 4-pyridyl), porphyrinyl, pyridyl Azinyl, 2,4-pyrimidinyl, 3,5-pyrimidinyl, 2,4-imidazolyl, imidazopyridyl, isoxazolyl, oxazolyl, thiazolyl, isothiazolyl, thiadiazolyl, Tetrazolyl, thienyl, triazinyl, benzothienyl, furyl, benzofuranyl, benzimidazolyl, indolyl, isodecyl, indanyl, pyridazinyl, pyrazine , pyridazinyl, pyrrolyl, triazolyl, quinolyl, isoquinolyl, pyri
- Heterocyclic or “heterocyclic” or “heterocyclyl” or “heterocycloalkyl” are mutually replaceable and refer to a 4-12 membered monocyclic, bicyclic, and tricyclic saturated or partially unsaturated non.
- An aromatic ring comprising at least one carbon atom in addition to at least one (such as 1-4, again such as 1-3 or further such as 1 or 2) heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen .
- the ring can be saturated or have at least one double bond (ie, partially unsaturated).
- the heterocyclic ring is optionally substituted for oxo.
- the site of attachment in the heterocycle can be carbon or a hetero atom.
- the heterocyclic ring is not a heteroaryl group as defined herein.
- the heterocyclic ring may also form a fused ring with an aromatic ring (e.g., a benzene ring) or a heteroaryl ring, such as a 5-7 membered heterocyclic benzophenone ring or a 5-7 membered heterocyclic ring and a 5-6 membered heteroaryl ring.
- heterocycles include, but are not limited to, (numbered starting from the position specified as priority 1), 1-pyrrolidinyl, 2-pyrrolidinyl, 2,4-imidazolidinyl, 2,3-pyrazole Alkyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2,5-piperazinyl, pyranyl, 2-morpholinyl, 3-morpholinyl , oxacyclopropyl, azacyclopropyl, thioheteropropyl, azetidinyl, oxetanyl, thioheterobutyl, 1,2-dithiot-butyl, 1, 3-dithiot-butyl, dihydropyridyl, tetrahydropyridyl, thiomorpholinyl, oxathiaridyl, piperazinyl, homopiperazinyl, homopiperidinyl, piperaz
- the substituted heterocyclic ring also includes a ring system substituted with one or more oxo moieties such as piperidinyl N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl and 1,1-dioxo-1-thiomorpholinyl.
- oxo moieties such as piperidinyl N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl and 1,1-dioxo-1-thiomorpholinyl.
- fused ring refers to a polycyclic ring system, for example, a bicyclic or tricyclic ring system in which two rings share only two ring atoms and one common bond.
- the fused ring may include a fused bicyclic cycloalkyl ring such as a bicyclic cycloalkyl ring having 7 to 12 ring atoms, which is arranged to be selected from the above [4, 4], [4, 5] a bicyclic ring of the [5,5], [5,6] and [6,6] ring systems; a fused bicyclic aryl ring such as the above 7-12 membered bicyclic aryl ring system; fused three a cycloaryl ring such as the above 10-15 membered tricyclic aryl ring system; a fused bicyclic heteroaryl ring such as the above 8-12-membered bicyclic heteroaryl ring; a fused tricyclic heteroaryl group; A ring such as the above 11
- Alkanoyl means a group formed by the combination of an alkyl group and a carbonyl group.
- a C 1-8 acyl group means a C 1-8 alkyl acyl group.
- Alkoxy means a group formed by the combination of an alkyl group and an oxygen atom.
- amide or “amido” refers to an amide group attached through a nitrogen atom.
- a C 1-8 amide group refers to a C 1-8 alkyl amide group.
- Sulfonyl means RS(O) 2 -, wherein R includes alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, meaning alkyl, cycloalkyl, heterocycloalkyl, aryl
- the base or heteroaryl is attached to the parent structure via -S(O) 2 -.
- C 1-8sulfonyl represents a C 1-8 alkylsulfonyl group.
- Sulfonylamino refers to a sulfonamide substituent attached to a nitrogen atom.
- C 1-8 sulfonylamino means a C 1-8 alkylsulfonylamino group.
- “Ureido” refers to a substituted or unsubstituted urea group.
- Ester group means a group containing a carbonyloxy group.
- a C 1-8 ester group means a C 1-8 alkylcarbonyloxy group.
- the compounds described herein may contain asymmetric centers and may thus exist as enantiomers. When the compounds described herein have two or more asymmetric centers, they may also exist as diastereomers. Both enantiomers and diastereomers fall into a wider variety of stereoisomers.
- the present application is intended to include all possible stereoisomers, such as substantially pure analytical enantiomers, their racemic mixtures, and mixtures of diastereomers.
- the present application is intended to include all stereoisomers of the compounds described herein and/or pharmaceutically acceptable salts thereof. Unless otherwise specified, reference to one isomer applies to any possible isomer. Wherever the composition of the isomer is not specified, all possible isomers are included.
- the term “substantially pure” as used herein means that the target stereoisomer contains up to 35% by weight, such as at most 30% by weight, such as up to 25% by weight, and further such as up to 20% by weight of any one or more other stereoisomers. In some embodiments, the term “substantially pure” means that the target stereoisomer contains up to 10% by weight, such as up to 5% by weight, such as up to 1% by weight, of any one or more other stereoisomers.
- Certain of the compounds described herein may exist as attachment sites for different hydrogens, referred to as tautomers.
- the application is intended to cover both the keto form and the enol form, alone and in mixtures thereof.
- the reaction products can advantageously be separated from each other and/or from the starting materials.
- the desired product of each step or series of steps is separated and/or purified (hereinafter referred to as separation) to the desired degree of homogenization by conventional techniques in the art.
- separation involves multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation or chromatography.
- Chromatography can involve a number of methods including, for example, reverse phase and normal phase chromatography; size exclusion chromatography; ion exchange chromatography; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical chromatography; simulated moving bed (“SMB”) Chromatography and preparative thin or thick layer chromatography and small-scale thin layer and flash chromatography techniques.
- SMB simulated moving bed
- Diastereomeric mixtures can be separated into their individual diastereomers by methods well known to those skilled in the art, such as chromatography and/or fractional crystallization, based on physicochemical differences in diastereomers. .
- the enantiomers can be separated by converting the enantiomeric mixture to a diastereomeric reaction by reaction with a suitably optically active compound (for example, a chiral auxiliary such as a chiral alcohol or Mosher's acid chloride).
- a suitably optically active compound for example, a chiral auxiliary such as a chiral alcohol or Mosher's acid chloride.
- the isomeric mixture is separated, the diastereomers are separated, and the individual diastereomers are then converted (e.g., hydrolyzed) to the corresponding pure enantiomers.
- the enantiomers can also be separated using a chiral HPLC column.
- a single stereoisomer for example, a substantially pure enantiomer
- Mixture (Eliel, E. and Wilen, S. Stereochemistry of Organic Compounds. New York: John Wiley & Sons, Inc., 1994; Lochmuller, CH, et al. "Chromatographic resolution of enantiomers: Selective review.” J. Chromatogr., 113 (3) (1975): pp. 283-302).
- the racemic mixture of the chiral compounds of the present invention can be separated and isolated by any suitable method, including: (1) formation of an ionic diastereomeric salt with a chiral compound, followed by fractional crystallization or other Method separation, (2) formation of diastereomeric compounds with a chiral derivatization reagent, separation of the diastereomers, then conversion to pure stereoisomers, (3) direct under chiral conditions The substantially pure or enriched stereoisomer is isolated. See: Wainer, Irving W., Ed. Drug Stereochemistry: Analytical Methods and Pharmacology. New York: Marcel Dekker, Inc., 1993.
- “Pharmaceutically acceptable salts” include, but are not limited to, salts formed with inorganic acids, and salts with organic acids.
- the free base can be obtained by alkalizing a solution of the acid salt.
- an addition salt such as a pharmaceutically acceptable addition salt
- an addition salt can be prepared by dissolving the free base in a suitable organic solvent and then following the preparation of the acid addition salt from the base compound. The solution is treated with an acid in a conventional operation.
- Treatment refers to administering to a subject deemed to have such a need (with, for example, a cancer) at least one compound described herein or a pharmaceutically acceptable thereof Salt.
- an effective amount refers to an amount of at least one compound described herein, or a pharmaceutically acceptable salt thereof, that is effective to "treat” (as defined above) a disease or condition in a subject.
- the solvent and the drug used were analytically pure or chemically pure; the solvent was re-distilled before use; and the anhydrous solvent was treated according to standard methods or literature methods.
- Column chromatography silica gel (100-200 mesh) and thin layer chromatography silica gel (GF254) are products of Qingdao Ocean Chemical Plant and Yantai Chemical Plant; unless otherwise specified, petroleum ether (60-90 ° C) / ethyl acetate ( v / v) as eluant; color-developing agent with iodine or phosphomolybdic acid ethanol solution; all unexplained extraction solvent are dried over anhydrous Na 2 SO 4.
- LC-MS High Performance Liquid Chromatography-Ion Trap Mass Spectrometer
- DAD diode array detector
- detection wavelengths 214 nm and 254 nm ion trap mass spectrometry (ESI source).
- the HPLC column was AgelaDurashell C18 (4.6 x 50 mm, 3.5 ⁇ m); the mobile phase was 0.1% aqueous NH4HCO3: acetonitrile (from 5:95 to 95:5 in 5 min); the flow rate was 1.8 mL/min.
- This embodiment provides a 3-fluoropyridine heterocyclic compound and a synthesis method thereof.
- a heterocyclic compound B1 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B2 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B3 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B4 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B5 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B6 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B7 which is synthesized by the following method:
- B7-1 (167 mg, 0.6 mmol) and 2,3-difluoropyridine-4-boronic acid (105 mg, 0.66 mmol) were dissolved in dioxane (4 mL) and water (1 mL), and Pd(dppf)Cl was added. 2 (39 mg, 0.048 mmol), dppf (26 mg, 0.048 mmol) and potassium phosphate (254 mg, 1.2 mmol). Under nitrogen protection, it was replaced with nitrogen five times, heated to 100 ° C, and reacted for 12 hours. After cooling to room temperature, the residue was evaporated.
- This example provides a 3-fluoropyridine heterocyclic compound B8 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B9 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B10 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B11 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B12 which is synthesized by the following method:
- B12-1 164 mg, 1.0 mmol
- 2,3-difluoropyridine-4-boronic acid (175 mg, 1.1 mmol) were dissolved in dioxane (4 mL) and water (1 mL), and Pd(dppf)Cl was added.
- 2 65 mg, 0.08 mmol
- dppf 44 mg, 0.08 mmol
- potassium phosphate 424 mg, 2.0 mmol
- This example provides a 3-fluoropyridine heterocyclic compound B13 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B14 which is synthesized by the following method:
- B14-1 (209 mg, 1.0 mmol) and 2,3-difluoropyridine-4-boronic acid (175 mg, 1.1 mmol) were dissolved in dioxane (4 mL) and water (1 mL), and Pd(dppf)Cl was added. 2 (65 mg, 0.08 mmol), dppf (44 mg, 0.08 mmol) and potassium phosphate (424 mg, 2.0 mmol). Under nitrogen protection, it was replaced with nitrogen five times, heated to 100 ° C, and reacted for 12 hours. After cooling to room temperature, it was concentrated under reduced vacuo.
- This example provides a 3-fluoropyridine heterocyclic compound B15 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B16 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B17 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B18 which is synthesized by the following method:
- B18-1 200 mg, 1.0 mmol
- B1-2 (238 mg, 1.5 mmol) were dissolved in dioxane (4 mL) and water (1 mL), and Pd(dppf)Cl 2 (65 mg, 0.08 mmol), Dppf (44 mg, 0.08 mmol) and potassium phosphate (424 mg, 2.0 mmol). Under nitrogen protection, it was replaced with nitrogen five times, heated to 100 ° C, and reacted for 12 hours. After cooling to room temperature, the residue was evaporated.
- This example provides a 3-fluoropyridine heterocyclic compound B19 which is synthesized by the following method:
- B19-1 (264 mg, 1.5 mmol) and B1-2 (358 mg, 2.25 mmol) were dissolved in dioxane (8 mL) and water (2 mL), and Pd(dppf)Cl 2 (55 mg, 0.075 mmol), Dppf (42 mg, 0.075 mmol) and potassium phosphate (798 mg, 3.0 mmol). Under nitrogen protection, it was replaced with nitrogen five times, heated to 100 ° C, and reacted for 12 hours. After cooling to room temperature, it was concentrated under reduced pressure and purified by column chromatography.
- This example provides a 3-fluoropyridine heterocyclic compound B20 which is synthesized by the following method:
- B20-1 (288 mg, 1.5 mmol) and B1-2 (358 mg, 2.25 mmol) were dissolved in dioxane (8 mL) and water (2 mL), and Pd(dppf)Cl 2 (55 mg, 0.075 mmol), Dppf (42 mg, 0.075 mmol) and potassium phosphate (798 mg, 3.0 mmol). Under nitrogen protection, it was replaced with nitrogen five times, heated to 100 ° C, and reacted for 12 hours. After cooling to room temperature, it was concentrated under reduced vacuo.
- This example provides a 3-fluoropyridine heterocyclic compound B21 which is synthesized by the following method:
- B21-2 (199 mg, 1.0 mmol) and B1-2 (191 mg, 1.2 mmol) were dissolved in dioxane (8 mL) and water (2 mL), and Pd(dppf)Cl 2 (37 mg, 0.05 mmol), Dppf (28 mg, 0.05 mmol) and potassium phosphate (532 mg, 2.0 mmol). Under nitrogen protection, it was replaced with nitrogen five times, heated to 100 ° C, and reacted for 12 hours. After cooling to room temperature, it was concentrated under reduced vacuo.
- This example provides a 3-fluoropyridine heterocyclic compound B22 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B23 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B24 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B25 which is synthesized by the following method:
- This example provides a 3-fluoropyridine heterocyclic compound B38 which is synthesized by the following method:
- Table 1 lists the analytical structures and spectral data of the compounds B1-B38 obtained in Examples 1-26 and the like.
- This example measures the inhibitory ability of the 3-fluoropyridine heterocyclic compounds B1-B25 and B38 prepared in Examples 1-26, and the compounds B26-B37 obtained according to the similar procedures of Examples 1-26, to the Wnt signaling pathway. .
- LWnt3A cells (CRL-2647, ATCC) were cultured in DMEM medium (Gibico) containing 10% fetal bovine serum (Hyclone).
- HEK293STF stable clonal cells (HEK293 cells transfected with "Super-TopFlash” TCF fluorescence reporter plasmid) were cultured in complete medium (containing 4mML-glutamine, 1.5g/L sodium bicarbonate, 4.5g/L glucose, 6 ⁇ g/ mL of blasticidin and 10% fetal bovine serum in DMEM medium).
- LWnt3A cells and HEK293STF stable cloned cells were cultured to a confluence of 90%, they were harvested separately and mixed at a ratio of 1:1.
- test compound 100 ⁇ L/well of the mixed cell culture medium was added to a 96-well plate to give a final cell concentration of 12,000 cells/well, followed by further culture for 24 hours.
- the test compound was diluted stepwise with DMSO and then diluted to the desired concentration with DMEM medium. 20 ⁇ L of the compound solution was added to the aforementioned 96-well plate containing the cell culture medium, followed by incubation at 37 ° C for 48 hours. Finally, 50 ⁇ L of luciferase solution (Brigh-Glo, Promega) was added to each well, and shaken at room temperature for 5 minutes.
- the luminescent signal was measured with a microplate reader (PHERAstar FS, BMG), and then the IC 50 value (potency) of the compound was calculated based on the inhibition of the luminescent signal by the compound at different concentrations.
- a microplate reader PHERAstar FS, BMG
- the IC 50 value potency
- a graph of inhibition results is obtained; the test patterns of the remaining compounds are basically the same.
- the overall results are shown in Table 2 (Results of the experiment for determining the ability of the heterocyclic compound B1-B38 to inhibit the Wnt signaling pathway).
- Compound IC 50 (nM) Compound IC 50 (nM) Compound IC 50 (nM) Compound IC 50 (nM) B1 0.18 B2 0.06 B3 0.11 B4 0.27 B5 0.31 B6 0.54 B7 0.12 B8 0.11 B9 5.2 B10 0.24 B11 16 B12 1 B13 10 B14 1 B15 10 B16 10 B17 16 B18 8 B19 7 B20 8 B21 2 B22 318 B23 10 B24 12 B25 0.06 B26 0.13 B27 0.12 B28 0.12 B29 0.06 B30 0.04 B31 0.34 B32 1.8 B33 0.4 B34 0.11 B35 1.0 B36 0.25 B37 0.2 B38 0.5
- the heterocyclic compound obtained in the examples of the present invention can be used as an effective antagonist of the Wnt signaling pathway, and can effectively block the Wnt signaling pathway, thereby being capable of treating or preventing diseases caused by abnormal Wnt signaling pathway. disease.
- the cytochrome P450 enzyme inhibition rate of the substrate to be tested was carried out in human liver microsomes.
- the five CYP isozymes and their respective probe substrates and their concentrations are as follows: CYP-1A2 (Phenacetin, 30 ⁇ M), CYP-2C9 (Tolutamide, 100 ⁇ M), CYP-2C19 (S-mephenytoin, 40 ⁇ M), CYP-2D6 (Dextromethorphan, 5 ⁇ M) and CYP-3A4 (Midazolam, 1 ⁇ M). All probe substrates were used at concentrations close to or below their Kms.
- HLM 0.2 mg/mL
- phosphate buffer 100 mM, pH 7.4
- NADPH 1 ⁇ M
- test compound 10 ⁇ M
- substrate of each CYP enzyme was incubated in a constant temperature water bath at 37 ° C. .
- the compound was incubated for 10 min before the start of the reaction with NADPH to allow the enzyme-inhibitor to function adequately. It was then reacted within each CYP enzyme specific reaction time period (CYP1A2, CYP2D6 and CYP3A4 for 10 min; CYP2C9 and CYP2C19 for 30 min).
- the reaction was quenched by the addition of an appropriate amount of an internal standard of ice-cold acetonitrile (100 ⁇ M). After high-speed centrifugation (13,000 rpm) for 10 min, the supernatant sample LC-MS/MS was used to quantitatively analyze the concentration of the unique metabolite formed by the probe substrate and the CYP enzyme to determine the CYP inhibition rate. Each test substrate was tested at least three times independently.
- LC-MS/MS analysis conditions All samples were analyzed by LC-MS/MS system using API4000QTRAP mass spectrometer.
- the instrument is equipped with two LC-20AD pumps and a CBM-20A controller, a DGU-20A solvent auto-degassing pump and a SIL-20A autosampler (Shimadzu, Columbia, MD, USA).
- Bona Aigel's Venusil XBP C18 column (50 x 2.1 mm, 5 ⁇ M) was used as an HPLC separation column with a column temperature of 40 ° C, a flow rate of 0.3 mL/min, and a total run time of 6 min.
- MS/MS Quantification of MS/MS: The API4000QTRAP mass spectrometer was operated in electrospray (ESI) positive ion mode with multiple reaction monitors (MRM). All compounds and internal standards were tested over a 100 ms residence time period. The remaining MS/MS parameters were set as follows: ion source curtain pressure, 30 psi; atomizing gas pressure, 55 psi; ion source spray voltage 4500V; ion source temperature 500 °C. For the detection of specific selected ions, the de-clustered voltage (DP) and collision energy (CE) need to be optimized according to the different detectors. The final data processing was performed using AB SCIEX Analysist 1.5.2 data collection and integration software.
- ESI electrospray
- MRM multiple reaction monitors
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
本发明提供了一种3-氟吡啶杂环化合物,该3-氟吡啶杂环化合物及其药学上可接受的盐,具有式I所示的结构。本发明还提供一种药物组合物,其包含治疗有效量的上述的3-氟吡啶杂环化合物及其药学上可接受的载体。本发明还提供上述的3-氟吡啶杂环化合物在制备治疗或预防Wnt信号通路异常的药物中的应用。本发明的3-氟吡啶杂环化合物能够有效拮抗Wnt信号通路,从而能够用于治疗或预防因Wnt信号通路失常引起的病症。
Description
本发明涉及一种3-氟吡啶杂环化合物及其应用,属于医药技术领域。
最早的Wnt信号通路的发现来自于对致癌病毒和果蝇发育机制的研究。Wnt基因于1982年发现,最初是作为小鼠乳腺肿瘤病毒优先整合的位点而被鉴定的,该基因能在细胞间传递增殖和分化信息,是一种癌基因,当时被命名为Int基因(小鼠Int-1和Int-3)。随后发现它与果蝇的无翅基因(wingless)属于相同源基因(orthologousgene),从而将二者结合命名为Wnt基因。
至今已发现并克隆出19种Wnt基因家族成员,人们将Wnt基因所介导的信号转导通路称为Wnt信号通路。Wnt通路是一条十分保守的信号传导通路。从低等生物果蝇直至高等哺乳动物,其成员都具有高度的同源性。Wnt信号转导通路参与了多种生物学过程的调控,包括胚胎的生长和形态发育、组织的稳定、能量代谢的平衡以及干细胞的维持(Logan等人,Annu.Rev.Cell.Dev.Biol.,2004,20,781-810)。近年来人们在对干细胞的研究中发现,Wnt信号通路对于表皮干细胞、肠干细胞、造血干细胞、神经干细胞、胚胎干细胞以及肿瘤干细胞的维持都起着重要的调控作用(Reya等人,Nature,2005,434,843-850)。
经典Wnt信号通路是由胞膜外配体Wnts蛋白同时与7次细胞跨膜的Frizzled受体和辅助性受体LRP5/6结合后开启Wnt/β-catenin信号通路的传导,并活化胞质内Dsh蛋白,活化的Dsh蛋白能抑制由APC蛋白、GSK-3β、Axin、β-catenin等形成的降解复合体中的关键成分GSK-3β的活性,使β-catenin不被GSK-3β磷酸化从而避免了泛素蛋白酶体对其识别和降解,进而在胞质中逐渐积聚(Boutros等人,Mech.Dev.,1999,83,27-37;Perrimon,Cell,1994,76,781-784)。当β-catenin在胞浆中积聚到一定浓度时就开始向细胞核转移,并与胞核内的转录因子TCF/LEFs结合导致β-cateninn的下游靶基因的启动因子暴露出来而被激活表达,如激活c-myc、cyclin-D1、survivin、gastrin、VEGF、ASEF等而导致细胞异常增殖。而在正常体细胞中,胞质内的β-catenin大部分与胞膜粘附蛋白E-cadherin及α-catenin结合形成复合体参与细胞骨架的调节,维持同型细胞的粘附,防止细胞转移,少部分游离的β-catenin在胞浆内被降解复合体磷酸化后由泛素蛋白酶体识别并降解,保持胞内
β-catenin低水平状态,从而使Wnt信号通路处于关闭状态。
研究发现当Wnt基因本身或通路其他任一成员因素发生变化使其不正常活化时,均有可能引起肿瘤的发生。例如,在结肠癌患者中广泛存在Wnt通路的调节因子包括APC、β-catenin、Axin、TCF等基因的突变,从而造成与生长相关的基因的过量表达(Klaus等人,Nat.Rev.Cancer,2008,8,387-398)。Lozzo等通过对100多份正常和肿瘤组织及10个人类肿瘤细胞系的研究发现在乳腺癌、肺癌、前列腺癌及黑色素瘤中均存在Wnt-5amRNA的过度表达,尤其是在乳腺癌中的表达异常明显(Lozzo等人,CancerResearch,1995,55,3495)。
Wnt途径的异常活化在细胞癌变、肿瘤发生及肿瘤侵袭性过程中具有重要作用,而阻断异常的Wnt信号通路可以抑制肿瘤细胞增殖,诱导肿瘤细胞凋亡。因此,Wnt信号通路具有较好的抗肿瘤靶向作用。
研究表明Wnt信号通路中LRP5过表达引起的异常Wnt信号和一些癌症相联系(Hoang等人,Int.J.Cancer,2004,109,106-111)。如前列腺癌、结肠直肠癌、卵巢癌、食管癌和胃癌。
β-catenin激活Wnt信号通路能增加神经祖细胞的循环和扩增,而缺失会导致祖细胞间隔的缺失(Chenn等人,Science,2002,297,365-369)。Wnt信号的异常活化在神经系统中是致瘤的(Dahmen等人,Cancer Res.,2001,61,7039-7043)。
Wnt信号通路可以促进全能造血干细胞的更新和维持,并且异常的Wnt信号对于由全能造血干细胞导致的各种疾病和其它血液相关的癌症负有责任(Reya等人,Nature,2005,434,843-850;Willert等人,Nature,2003,423,448-452)。
Wnt信号传导的失调也会通过诱导视网膜炎症、血管渗漏、和新血管形成,导致糖尿病性视网膜病的发展。
近年来,肿瘤免疫治疗成为研究热点。肿瘤免疫治疗的主要策略是利用免疫检查点抑制剂,如CTLA-4,PD-1和PD-L1的单抗等,阻断T细胞上免疫抑制受体的活化。这种策略在多种肿瘤(如黑色素瘤和肺癌)的病人中效果显著(特别是T细胞浸润的肿瘤),然而在没有T细胞浸润的肿瘤组织中,常常抗肿瘤免疫。研究表明(Stefani Spranger等人,Nature,2015,523,231-235),在T细胞浸润和无T细胞浸润的黑色素瘤的病人样本,49%无T细胞浸润的黑色素瘤存在高水平的活性β-catenin,而T细胞浸润的病人仅有4%。活化的Wnt/β-catenin信号通路在肿瘤中,使CD103+树突细胞(DC细胞,T细胞免疫的哨卫)募集受阻,因而T细胞活化受阻不能浸润肿瘤。因此,抑制Wnt信号通路将有助于增强免疫检查点抑制剂的抗肿瘤效果。Wnt信号通路抑制剂与免疫检查点抑制剂的联合应用具有加成作用,优于二者的单独应用。
由上可见,要改善因上述Wnt信号通路失常引起的的病症,就需要提出有效的Wnt信号通路调节剂。
发明内容
鉴于上述现有技术存在的缺陷,本发明的目的是提出一种3-氟吡啶杂环化合物及其应用,能够有效拮抗Wnt信号通路,从而能够用于治疗或预防因Wnt信号通路失常引起的病症。
本发明的目的通过以下的化合物技术方案1-20、药物组合物、药物联用产品和化合物的用途得以实现:
方案1:一种3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式I所示的结构:
其中:
A为未被取代或被1-6个R6基团取代的5-14元含有1-4个杂原子的杂芳环、6-12元芳环、3-6元环烷基或3-6元杂环烷基,所述杂芳环和杂环烷基的杂原子包括N、O和S中的一种或多种;或者A为未被取代或被1-2个R6基团取代的其中X1为N或CR6;
B选自氢原子、氰基、卤代基、羟基、C3-8环烷基、C2-8烯基或C2-8炔基、C1-8磺酰基、C1-8磺酰胺基、C1-8烷氧基、C1-8酯基、C1-8酰基、C1-8酰胺基、C1-8氧代脲基、C1-8脲基、未被取代或被1-3个R12取代基取代的C1-8烷基、-NR7R8;或者B选自未被取代或被1-3个R9基团取代的苯环、未被取代或被1-3个R10基团取代的5-6元含有1-3个杂原子的杂芳环、未被取代或被1-3个R11基团取代的5-7元含有1-3个杂原子的饱和杂环,所述杂芳环和饱和杂环的杂原子包括N、O和S中的一种或多种;
U为未被取代或被1-6个R13基团取代的6-12元芳环、未被取代或被1-6个R13基团取代的5-14元杂芳环、未被取代或被1-6个R13基团取代的5-7元杂环并苯环、未被取代或被1-6个R13基团取代的5-7元杂环并5-6元杂芳环、-OR14、-NR14R15,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为被5-6元芳环或杂芳环取代的C2-8烯基或C2-8炔基,所述杂环、
杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为未被取代或被1-2个R13基团取代的其中X2为N或CR13;
R1、R2、R3各自独立地选自氢原子、C1-6烷基、被1-3个分别选自卤代基、羟基、氰基、C1-3烷基、C3-5环烷基和C1-3烷氧基的基团取代的C1-6烷基;
R4、R5分别选自氢原子、氰基、氨基、卤代基、C1-8烷基、C3-8环烷基、C1-8烷氧基、-NH-C1-3烷基;
R6选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的氨基、C1-6烷基、C3-6环烷基或C1-6烷氧基;
R7、R8分别选自未被取代或被1-3个R12取代的氢原子、C1-8烷基、C3-8环烷基;或者R7和R8连同它们连接的原子成环;
R9、R10分别选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R9、R10分别选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基取代的氨基、C1-6烷基、C3-6环烷基或C1-6烷氧基;
R11选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷氧基、C3-6环烷基、氧代基、未被取代或被卤代基取代的C1-6烷基;
R12选自氢原子、氰基、羟基、氨基、C1-6烷氧基、C1-6烷基、C3-6环烷基、C2-6脲基、C2-6氧代脲基、-S(O)2-C1-3烷基、-N(R16)S(O)2-C1-3烷基、-N(R16)C(O)-C1-3烷基;
R13选自氢原子、氰基、卤代基、羟基、氨基、C1-6烷氧基、C3-6环烷基、-S(O)2-C1-3烷基、-C(O)-C1-3烷基、-C(O)O-C1-3烷基、氧代基、未被取代或被卤代基取代的C1-6烷基;
R14、R15分别选自氢原子、C3-6环烷基、C1-6烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环、5-6元含有1-3个杂原子的杂环、被1-3个R20取代的C3-6环烷基、被1-3个R20取代的C1-6烷基、被1-3个R20取代的5-6元芳环、被1-3个R20取代的5-6元含有1-3个杂原子的杂芳环、被1-3个R20取代的5-6元含有1-3个杂原子的杂环;所述杂原子包括N、O和S中的一种或多种;或者R14和R15连同它们连接的N原子成环;
R16、R17、R18、R19分别选自氢原子、未被取代或被卤代基取代的C1-6烷基;
R20选自氢原子、氰基、羟基、氨基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、
5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;
R21选自氢原子或C1-6烷基。
上述的3-氟吡啶杂环化合物中,优选的,
B选自氢原子、氰基、卤代基、羟基、C1-8磺酰基、C1-8磺酰胺基、C1-8烷氧基、C1-8酰基、C1-8酰胺基、C1-8氧代脲基、C1-8脲基、C1-8烷基、-NR7R8;或者B选自未被取代或被1-3个R9基团取代的苯环、未被取代或被1-3个R10基团取代的5-6元含有1-3个杂原子的杂芳环、未被取代或被1-3个R11基团取代的5-7元含有1-3个杂原子的饱和杂环,所述杂芳环和饱和杂环的杂原子包括N、O和S中的一种或多种;
R1为氢原子;
R2、R3分别选自氢原子、C1-6烷基;
R4、R5分别选自氢原子、氰基、卤代基、C1-8烷基;
R6选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的氨基、C1-6烷基、C3-6环烷基或C1-6烷氧基;
R7、R8分别选自氢原子、C3-8环烷基、未被取代或被1-3个R12取代的C1-8烷基;或者R7和R8连同它们连接的原子成环;
R9、R10分别选自氢原子、卤代基、氰基、C1-6烷基、C1-6烷氧基;或者R9、R10分别选自被1-3个卤代基取代的C1-6烷基或C1-6烷氧基;
R11选自氢原子、氧代基、未被取代或被卤代基取代的C1-6烷基;
R12选自氢原子、氰基、C1-6烷基、C1-6磺酰胺基;
R13选自氢原子、氰基、卤代基、氧代基、C1-6烷氧基、C3-6环烷基、C1-6酰基、未被取代或被卤代基取代的C1-6烷基;
R14、R15分别选自氢原子、C3-6环烷基、未被取代或被1-3个R20取代的C1-6烷基;或者R14和R15连同它们连接的N原子成环;
R16、R17、R18、R19各自独立地选自氢原子、未被取代或被卤代基取代的C1-6烷基;
R20选自氢原子、氰基、羟基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;
R21选自氢原子、或C1-6烷基。
方案2:在方案1的3-氟吡啶杂环化合物中,优选的,A为未被取代或被1-6个R6基团取代的下列基团中的一种:
其中R6为如上所定义。
方案3:在方案2的3-氟吡啶杂环化合物中,更优选的,A为未被取代或被1个R6基团取代的下列基团中的一种:
其中R6为如上所定义。
方案4:在方案2的3-氟吡啶杂环化合物中,最优选的,A为未被取代或被1个R6基团取代的下列基团中的一种:
其中R6为如上所定义。
方案5:在方案2的3-氟吡啶杂环化合物中,甚至最优选的,A为未被取代或被1个R6基团取代的下列基团中的一种:
其中R6为氢原子、卤代基(优选为F)、氰基或C1-6烷氧基(优选为甲氧基)。
方案6:在以上方案1至5中任一方案的3-氟吡啶杂环化合物中,优选的,B为下列基团中的一种:
其中,n为0、1或2,R10为如上所定义。
方案7:在方案6的3-氟吡啶杂环化合物中,更优选的,B为下列基团中的一种:
其中,n为0、1或2,R10为如上所定义。
方案9:在以上方案1至8中任一方案的3-氟吡啶杂环化合物中,优选的,L为价键。
方案10:在以上方案1至9中任一方案的3-氟吡啶杂环化合物中,优选的,该3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式II所示的结构:
其中,X3为N或CR22;
Y1、Y2、Y3、Y4各自独立地选自N或CR6;
U为未被取代或被1-6个R13基团取代的6-12元芳环、未被取代或被1-6个R13基团取代的5-14元杂芳环、未被取代或被1-6个R13基团取代的5-7元杂环并苯环、未被取代或被1-6个R13基团取代的5-7元杂环并5-6元杂芳环、-OR14或-NR14R15,所述杂环、杂芳
环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为被5-6元芳环或杂芳环取代的C2-8烯基或C2-8炔基,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为未被取代或被1-2个R13基团取代的其中X2为N或CR13;
L是价键;
R1和R4为氢原子;
R2和R3各自独立地为氢原子或C1-3烷基;
R5选自氢原子、卤代基、氰基、C1-3烷基;
R6选自氢原子、卤代基、氰基、羟基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的C1-6烷基、C3-6环烷基或C1-6烷氧基;
R10选自氢原子、卤代基、未被取代或被卤代基取代的C1-6烷基;
R13选自氢原子、氰基、卤代基、氧代基、C1-6烷氧基、C3-6环烷基、C1-6酰基、未被取代或被卤代基取代的C1-6烷基;
R14、R15分别选自氢原子、C3-6环烷基、未被取代或被1-3个R20取代的C1-6烷基;或者R14和R15连同它们连接的N原子成环;
R20选自氢原子、氰基、羟基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;
R21、R22分别独立选自氢原子或C1-6烷基。
方案12:在以上方案1至11中任一方案的3-氟吡啶杂环化合物中,优选的,该3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式III所示的结构:
其中:
Y5、Y6、Y7、Y8、Y9、Y10各自独立地选自N或CR6;
B为氰基;
U为未被取代或被1-6个R13基团取代的6-12元芳环、未被取代或被1-6个R13基团取代的5-14元杂芳环、未被取代或被1-6个R13基团取代的5-7元杂环并苯环、未被取代或被1-6个R13基团取代的5-7元杂环并5-6元杂芳环、-OR14或-NR14R15,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为被5-6元芳环或杂芳环取代的C2-8烯基或C2-8炔基,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为未被取代或被1-2个R13基团取代的其中X2为N或CR13;
L是价键;
R1和R4是氢原子;
R2和R3分别选自氢原子、甲基;
R5选自氢原子、卤代基、氰基、C1-3烷基;
R6选自氢原子、卤代基、氰基、羟基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的C1-6烷基、C3-6环烷基或C1-6烷氧基;
R13选自氢原子、氰基、卤代基、氧代基、C1-6烷氧基、C3-6环烷基、C1-6酰基、未被取代或被卤代基取代的C1-6烷基;
R14、R15分别选自氢原子、C3-6环烷基、未被取代或被1-3个R20取代的C1-6烷基;或者R14和R15连同它们连接的N原子成环;
R20选自氢原子、氰基、羟基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;
R21为氢原子或C1-6烷基。
方案14:在以上方案1至13中任一方案的3-氟吡啶杂环化合物中,优选的,U为未被取代或被1-6个R13基团取代的下列基团中的一种:
其中R13如上所定义。
方案15:在方案14中的3-氟吡啶杂环化合物中,更优选的,U为未被取代或被1个R13基团取代的下列基团中的一种:
其中R13如上所定义。
方案16:在方案14中的3-氟吡啶杂环化合物中,最优选的,U为未被取代或被1个R13基团取代的下列基团中的一种:
其中R13为卤素(优选为F或Cl)、C1-6烷基(优选为甲基)、C1-6烷氧基(优选为甲氧基)或氰基。
方案18:在以上方案1至13中任一方案的3-氟吡啶杂环化合物中,优选的,该3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式IV或式V所示的结构:
其中,X4、X5、X6、X7、X8、X9各自独立选自N或CR13;
L是价键;
R1和R4为氢原子;
R2和R3各自独立地为氢原子或C1-3烷基;
R5选自氢原子、卤代基、氰基、C1-3烷基;
R6选自氢原子、卤代基、氰基、羟基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的C1-6烷基、C3-6环烷基或C1-6烷氧基;
R10选自氢原子、卤代基、未被取代或被卤代基取代的C1-6烷基;
R13选自氢原子、氰基、卤代基、C1-6烷氧基、C3-6环烷基、未被取代或被卤代基取代的C1-6烷基;
R23、R24分别选自氢原子、C1-3烷基;或者R23和R24合并为氧代基;
R25、R26、R27、R28各自独立选自氢原子或C1-3烷基。
方案20:上述的3-氟吡啶杂环化合物中,优选的,该3-氟吡啶杂环化合物包括下列化合物中的一种或多种:
本发明还提供一种药物组合物,其包含治疗有效量的上述的3-氟吡啶杂环化合物及其药学上可接受的载体。
本发明还提供一种3-氟吡啶杂环化合物的药物联合产品,其包括本发明所述的3-氟吡啶杂环化合物或其药学上可接受的盐与抗肿瘤药物、抗菌药物、抗病毒药物、抗寄生虫病药物、中枢神经系统系统药物、抗骨质增生药物、糖尿病药物中的一种或几种的组合进行联合应用得到的组合物。
本发明还提供上述的3-氟吡啶杂环化合物在制备用于治疗或预防与Wnt信号通路异常相关的疾病的药物中的应用;所述与Wnt信号通路异常相关的疾病包括乳腺癌、肺癌、膀胱癌、胰腺癌、肝癌、头颈鳞状上皮癌、甲状腺癌、肉瘤、骨肉瘤、硬纤维瘤、黑色素瘤、前列腺癌、结肠直肠癌、卵巢癌、宫颈癌、食管癌、胃癌、骨髓瘤、淋巴瘤、套细胞淋巴瘤、皮肤T细胞淋巴瘤、慢性和非进行性贫血、自发性或原发性血小板增多症、特发性骨
髓纤维化、肺纤维化、肾脏纤维化、肝纤维化、肝硬化、糖尿病性视网膜病、巨球蛋白血症、白血病、急性白血病、慢性白血病、淋巴性白血病、髓性白血病、骨髓增生异常综合症、骨髓增殖性病症、脑瘤、星形细胞瘤、髓母细胞瘤、许旺细胞瘤、原发性神经外胚瘤、垂体瘤、以及寄生虫病、血吸虫病和疟疾中的一种病症或几种病症的组合的药物及药物组合物。
上述的药物组合物是指以上述的3-氟吡啶杂环化合物及其药学上可接受的盐或载体作为组分的组合物,或者是上述的3-氟吡啶杂环化合物及其药学上可接受的盐与抗肿瘤药物、抗菌药物、抗病毒药物、抗寄生虫病药物、中枢神经系统系统药物、抗骨质增生药物、糖尿病药物中的一种或几种的组合进行联合应用得到的组合物。
本发明的突出效果为:
本发明的3-氟吡啶杂环化合物能够有效拮抗Wnt信号通路,从而能够用于治疗或预防因Wnt信号通路失常引起的病症或与Wnt信号通路异常相关的疾病。
图1是实施例25中3-氟吡啶杂环化合物B4的细胞抑制测试结果曲线图;
图2是实施例25中3-氟吡啶杂环化合物B7的细胞抑制测试结果曲线图。
定义
除非另有具体说明,本申请使用的词语、短语和符号具有其通常含义。
“烷基”是指直链或支化的饱和脂族烃基。烷基包括包含1-18个、诸如1-12个、进一步诸如1-10个、更进一步诸如1-6个碳原子的烷基,其实例选自甲基、乙基、1-丙基或正丙基(″n-Pr″)、2-丙基或异丙基(″i-Pr″)、1-丁基或正丁基(″n-Bu″)、2-甲基-1-丙基或异丁基(″i-Bu″)、1-甲基丙基或仲丁基(″s-Bu″)以及1,1-二甲基乙基或叔丁基(″t-Bu″)。烷基的其它实例可选自1-戊基、2-戊基、3-戊基、2-甲基-2-丁基、3-甲基-2-丁基、3-甲基-1-丁基、2-甲基-1-丁基、1-己基、2-己基、3-己基(-CH、2-甲基-2-戊基、3-甲基-2-戊基、4-甲基-2-戊基、3-甲基-3-戊基、2-甲基-3-戊基、2,3-二甲基-2-丁基和3,3-二甲基-2-丁基。
“烯基”是指包含至少一个C=C双键和2-18个诸如2-8、2-6或2-4个碳原子的直链或支化的烃基。烯基的实例选自乙烯基、丙-1-烯基、丙-2-烯基(-CH2CH=CH2)、2-甲基丙-1-烯基、丁-1-烯基、丁-2-烯基、丁-3-烯基、丁-1,3-二烯基、2-甲基丁-1,3-二烯基、己-1-烯基、己-2-烯基、己-3-烯基、己-4-烯基和己-1,3-二烯基。
“炔基”是指包含至少一个C≡C三键和2-18个诸如2-8、2-6或2-4个碳原子的直链或支化的烃基。炔基的实例包括乙炔基、1-丙炔基、炔丙基、1-丁炔基、2-丁炔基和3-丁炔基。
“环烷基”是指饱和或部分不饱和的环状脂族烃基,所述环状脂族烃基包括单环和多环(例如,二环和三环)基团。例如环烷基可包含3-12个、诸如3-8个、进一步诸如3-6个、3-5个或3-4个碳原子。进一步例如,环烷基的实例选自包含3-12个,诸如3-8个、3-6个碳原子的单环。单环环烷基的实例包括环丙基、环丁基、环戊基、1-环戊-1-烯基、1-环戊-2-烯基、1-环戊-3-烯基、环己基、1-环己-1-烯基、1-环己-2-烯基、1-环己-3-烯基、环己二烯基、环庚基、环辛基、环壬基、环癸基、环十一基和环十二基。二环环烷基的实例包括具有7-12个碳原子的二环环烷基,其排列成选自[4,4]、[4,5]、[5,5]、[5,6]和[6,6]环系统的二环,或排列成选自二环[2.2.1]庚烷、二环[2.2.2]辛烷和二环[3.2.2]壬烷的桥接二环。二环环烷基的其它实例包括排列成选自[5,6]和[6,6]环系统的二环的那些。所述环可为饱和的或具有至少一个双键(即部分不饱和的),但不完全共轭。
“芳基”或“芳环”是指含有6-10个碳原子的单环或二环芳族烃基。所述芳基或芳环的实例包括苯基或苯环、萘基或萘环(包括1-萘基或2-萘基)。由经取代的苯衍生物形成的且在环原子处具有自由价的二价基团被命名为经取代的亚苯基。通过从具有自由价的碳原子除去一个氢原子由命名以“-基”结尾的一价多环烃基衍生的二价基团如下命名:向相应的一价基团添加“-亚基”,例如,具有两个连接的位点的萘基被称为亚萘基。然而,芳基不以任何方式涵盖杂芳基或与杂芳基重叠,单独在下文定义。
“卤素”或“卤代”或“卤代基”是指F、Cl、Br或I。
“杂芳基”或“杂芳环”是指选自以下的基团:
-5-7元芳族单环,其包含至少一个选自N、O和S的杂原子,例如1-4个,或者在一些实施方案中为1-3个杂原子,剩余环原子为碳;
-8-12元二环,其包含至少一个选自N、O和S的杂原子,例如1-4个,或者在一些实施方案中为1-3个,或者在其它实施方案中为1个或2个杂原子,剩余环原子为碳,且其中至少一个环为芳族的且芳族环中存在至少一个杂原子;以及
-11-14元三环,其包含至少一个选自N、O和S的杂原子,例如1-4个,或者在一些实施方案中为1-3个,或者在其它实施方案中为1个或2个杂原子,剩余环原子为碳,且其中至少一个环为芳族的且芳族环中存在至少一个杂原子。
当杂芳基中S和O原子的总数超过1时,所述杂原子彼此不相邻。在一些实施方案中,杂芳基中S和O原子的总数至多为2。在一些实施方案中,芳族杂环中S和O原子的总数至多为1。
杂芳基的实例包括,但不限于,(从指定为优先次序1的连接位置开始编号),吡啶基(诸如2-吡啶基、3-吡啶基或4-吡啶基)、噌啉基、吡嗪基、2,4-嘧啶基、3,5-嘧啶基、2,4-咪唑基、咪唑并吡啶基、异噁唑基、噁唑基、噻唑基、异噻唑基、噻二唑基、四唑基、噻吩基、三嗪基、苯并噻吩基、呋喃基、苯并呋喃基、苯并咪唑基、吲哚基、异吲哚基、二氢吲哚基、酞嗪基、吡嗪基、哒嗪基、吡咯基、三唑基、喹啉基、异喹啉基、吡唑基、吡咯并吡啶基(诸如1H-吡咯并[2,3-b]吡啶-5-基)、吡唑并吡啶基(诸如1H-吡唑并[3,4-b]吡啶-5-基)、苯并噁唑基(诸如苯并[d]噁唑-6-基)、蝶啶基、嘌呤基、1-氧杂-2,3-二唑基、1-氧杂-2,4-二唑基、1-氧杂-2,5-二唑基、1-氧杂-3,4-二唑基、1-硫杂-2,3-二唑基、1-硫杂-2,4-二唑基、1-硫杂-2,5-二唑基、1-硫杂-3,4-二唑基、呋咱基、苯并呋咱基、苯并噻吩基、苯并噻唑基、苯并噁唑基、喹唑啉基、喹喔啉基、二氮杂萘基、呋喃并吡啶基、苯并噻唑基(诸如苯并[d]噻唑-6-基)、吲唑基(诸如1H-吲唑-5-基)和5,6,7,8-四氢异喹啉基。
“杂环的”或“杂环”或“杂环基”或“杂环烷基”是可相互替换的,是指4-12元单环、二环和三环饱和或部分不饱和的非芳族环,所述环除了至少一个(诸如1-4个、再诸如1-3个或者再诸如1个或2个)选自氧、硫和氮的杂原子之外还包含至少一个碳原子。所述环可为饱和的或具有至少一个双键(即部分不饱和的)。所述杂环任选地取代有氧代。杂环中连接的位点可为碳或杂原子。所述杂环不是本申请所定义的杂芳基。所述杂环也可与芳环(例如苯环)或杂芳环形成稠合环,例如5-7元杂环并苯环或5-7元杂环并5-6元杂芳环。
杂环的实例包括,但不限于,(从指定为优先次序1的连接位置开始编号),1-吡咯烷基、2-吡咯烷基、2,4-咪唑烷基、2,3-吡唑烷基、1-哌啶基、2-哌啶基、3-哌啶基、4-哌啶基、2,5-哌嗪基、吡喃基、2-吗啉基、3-吗啉基、氧杂环丙基、氮杂环丙基、硫杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、1,2-二硫杂环丁基、1,3-二硫杂环丁基、二氢吡啶基、四氢吡啶基、硫吗啉基、氧硫杂环己基、哌嗪基、高哌嗪基、高哌啶基、氮杂环庚基、氧杂环庚基、硫杂环庚基、1,4-氧硫杂环己基、1,4-二氧杂环庚基、1,4-氧硫杂环庚基、1,4-氧氮杂环庚基、1,4-二硫杂环庚基、1,4-硫氮杂环庚基和1,4-二氮杂环庚基、1,4-二噻烷基、1,4-氮硫杂环己基、氧氮杂基、二氮杂基、硫氮杂基、二氢噻吩基、二氢吡喃基、二氢呋喃基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、四氢萘啶基(例如四氢-1,6-萘啶基)、1-吡咯啉基、2-吡咯啉基、3-吡咯啉基、二氢吲哚基、2H-吡喃基、4H-吡喃基、
1,4-二噁烷基、1,3-二氧杂环戊基、吡唑啉基、吡唑烷基、二噻烷基、二硫杂环戊基、吡唑烷基、咪唑啉基、嘧啶酮基、1,1-二氧代-硫吗啉基、3-氮杂二环[3.1.0]己基、3-氮杂二环[4.1.0]环庚基和氮杂二环[2.2.2]环己基。经取代的杂环还包括取代有一个或多个氧代部分的环系统,诸如哌啶基N-氧化物、吗啉基-N-氧化物、1-氧代-1-硫吗啉基和1,1-二氧代-1-硫吗啉基。
“稠合环”是指多环系统,例如,双环或三环系统,其中两个环仅共享两个环原子和一个共同的键。稠合环的实例可包括稠合的二环环烷基环,诸如具有7-12个环原子的二环环烷基环,其排列成选自上述[4,4]、[4,5]、[5,5]、[5,6]和[6,6]环系统的二环;稠合的二环芳基环,诸如上述7-12元二环芳基环系统;稠合的三环芳基环,诸如上述10-15元三环芳基环系统;稠合的二环杂芳基环,诸如上述8-12-元二环杂芳基环;稠合的三环杂芳基环,诸如上述11-14元三环杂芳基环;以及上述稠合的二环或三环杂环基环。
“烷酰基”是指由烷基与羰基结合所形成的基团。例如C1-8酰基表示C1-8烷基酰基。
“烷氧基”是指由烷基与氧原子结合所形成的基团。
“酰氨基”或“酰胺基”是指经氮原子连接的酰胺基。例如C1-8酰胺基是指C1-8烷基酰胺基。
“磺酰基”是指R-S(O)2-,其中R包括烷基、环烷基、杂环烷基、芳基和杂芳基,是指烷基、环烷基、杂环烷基、芳基或杂芳基通过-S(O)2-与母体结构连接。例如,C1-8磺酰基表示C1-8烷基磺酰基。
“磺酰氨基”是指通氮原子连接的磺酰胺取代基。例如,C1-8磺酰氨基是指C1-8烷基磺酰基氨基。
“脲基”是指取代或非取代的脲基团。
“酯基”是指含有羰基氧基的基团。例如,C1-8酯基是指C1-8烷基羰基氧基。
本申请所述化合物可含有不对称中心,并且由此可以对映异构体存在。当本申请所述化合物具有两个或更多个不对称中心时,它们还可以非对映异构体存在。对映异构体和非对映异构体均落入更宽种类的立体异构体中。本申请意在包括所有可能的立体异构体,诸如基本上纯的解析的对映异构体、它们的外消旋混合物以及非对映异构体的混合物。本申请意在包括所有本申请所述化合物的立体异构体和/或其药学上可接受的盐。除非另有具体说明,否则提及一种异构体适用于任意可能的异构体。但凡未指明异构体组成时,均包括所有可能的异构体。
本申请使用的术语“基本上纯的”意指目标立体异构体含有至多35重量%,诸如至多
30重量%,再诸如至多25重量%,再诸如至多20重量%的任意一种或多种其它立体异构体。在一些实施方案中,术语“基本上纯的”意指目标立体异构体含有至多10重量%,例如至多5重量%,诸如至多1重量%的任意一种或多种其它立体异构体。
当本申请所述化合物含有烯烃双键,除非另有指明,否则这类双键意欲包括E和Z两种几何异构体。
某些本申请所述化合物可以具有不同的氢的连接位点存在,称为互变异构体。例如,包括羰基-CH2C(O)-的化合物(酮形式)可经受互变异构,形成羟基-CH=C(OH)-(烯醇形式)。适用时,本申请意欲,单独地以及它们的混合物,包括酮形式和烯醇形式两者。
可有利地从彼此和/或从起始物质中分离出反应产物。通过本领域中的常规技术将每一步或一系列步骤的期望产物分离和/或纯化(以下称为分离)至期望的均质程度。通常上述分离涉及多相萃取、从溶剂或溶剂混合物中结晶、蒸馏、升华或色谱。色谱可涉及许多方法,其包括例如:反相和正相色谱;尺寸排阻色谱;离子交换色谱;高、中和低压液相色谱方法和仪器;小规模分析色谱;模拟移动床(″SMB″)色谱和制备型薄层或厚层色谱以及小规模薄层和快速色谱的技术。本领域技术人员应当使用最可能实现期望分离的技术。
通过本领域技术人员熟知的方法,诸如色谱和/或分级结晶,基于非对映异构体的物理化学差异,可将非对映异构的混合物分离成它们的单独的非对映异构体。对映异构体可通过以下方式进行分离:通过与适当光学活性的化合物(例如,手性助剂诸如手性醇或Mosher′s酰氯)进行反应将对映异构的混合物转化成非对映异构的混合物,分离非对映异构体,然后将单独的非对映异构体转化(例如,水解)成对应的纯的对映异构体。也可使用手性HPLC柱分离对映异构体。
单一的立体异构体,例如,基本上纯的对映异构体,可通过以下方式获得:使用诸如形成非对映异构体的方法,用光学活性的拆分剂来拆分外消旋混合物(Eliel,E.和Wilen,S.Stereochemistry of Organic Compounds.New York:John Wiley&Sons,Inc.,1994;Lochmuller,C.H.,等″Chromatographic resolution of enantiomers:Selective review.″J.Chromatogr.,113(3)(1975):pp.283-302)。本发明的手性化合物的外消旋混合物可通过任何合适的方法分开和离析,所述方法包括:(1)与手性化合物形成离子性非对映异构的盐,然后通过分级结晶或其它方法分离,(2)与手性衍生化试剂形成非对映异构的化合物,分离所述非对映异构体,然后转化为纯的立体异构体,(3)在手性条件下直接分离基本上纯的或富含的立体异构体。参见:Wainer,Irving W.,Ed.Drug Stereochemistry:Analytical Methods and Pharmacology.New York:Marcel Dekker,Inc.,1993。
“药学上可接受的盐”包括,但不限于与无机酸形成的盐,以及与有机酸形成的盐。
此外,如果本申请所述化合物以酸加成盐的形式来获得,则可通过碱化酸式盐的溶液获得游离碱。相反,如果产物为游离碱,则可通过以下方式制备加成盐(诸如药用加成盐)∶将游离碱溶于合适的有机溶剂中,然后按照用于由碱化合物制备酸加成盐的常规操作用酸处理该溶液。本领域技术人员无需创造性努力就应当知道可用于制备无毒药用加成盐的多种合成方法。
“治疗”、“处理”或“处置”或“减轻”是指对被认为有此需要的(患有例如癌症)受试者给药本申请所述的至少一种化合物或其药学上可接受的盐。
术语“有效量”是指在受试者中有效“治疗”(如上文所定义)疾病或病症的本申请所述的至少一种化合物或其药学上可接受的盐的量。
为了对本发明的技术特征、目的和有益效果有更加清楚的理解,现对本发明的技术方案进行以下详细说明,但不能理解为对本发明的可实施范围的限定。下述实施例中所述实验方法,如无特殊说明,均为常规方法;所述试剂和材料,如无特殊说明,均可从商业途径获得。
下述的实施例中,所用溶剂和药品均为分析纯或化学纯;溶剂在使用前均经过重新蒸馏;无水溶剂均按照标准方法或文献方法进行处理。柱层析硅胶(100-200目)和薄层层析硅胶(GF254)为青岛海洋化工厂和烟台化工厂产品;如未特别说明,均采用石油醚(60-90℃)/乙酸乙酯(v/v)作为洗脱剂;显色剂用碘或磷钼酸的乙醇溶液;所有萃取溶剂未经说明均用无水Na2SO4干燥。1HNMR用varian-400型核磁共振仪记录,TMS为内标。LC-MS用美国Agilent公司1100型高效液相色谱-离子阱质谱联用仪(LC-MSDTrap)记录,二极管阵列检测器(DAD),检测波长214nm和254nm,离子阱质谱(ESI源)。HPLC柱为AgelaDurashellC18(4.6×50mm,3.5μm);流动相为0.1%NH4HCO3水溶液∶乙腈(5分钟内从5∶95到95∶5);流速为1.8mL/min。
实施例1
本实施例提供一种3-氟吡啶杂环化合物及其合成方法。
(1)杂环化合物B1,其是由如下方法合成的:
1)中间体B1-2的合成:
50mL的两口烧瓶,加入无水四氢呋喃20mL,氮气保护,冷却至-78℃,加入2.5N的LDA四氢呋喃溶液(4.4mL,11mmol),慢慢加入B1-1(1.15g,10mmol)。-78℃下搅拌1h,然后慢慢加入硼酸三异丙酯(2.82g,15mmol)。加完再在-78℃搅拌1.5h,慢慢升至室温。2N氢氧化钠水溶液5mL加入,搅拌10分钟。分层,水相调PH为5,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗一次,无水硫酸钠干燥,减压蒸除溶剂,得白色固体(700mg,44%)。1HNMR(400MHz,DMSO-d6)δ8.86(s,2H),7.99(d,J=4.4Hz,1H),7.43(m,1H)。
2)中间体B1-3的合成:
将B1-2(690mg,4.34mmol)和6-溴喹啉(749mg,3.61mmol)溶于35mL二氧六环和7mL水,加入碳酸钾(1.25g,9.03mmol)和四三苯基磷钯(161mg,0.14mmol)。置换氮气,在100℃下搅拌反应8h。冷却至室温,加入10mL水,乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗。有机相经无水硫酸钠干燥,浓缩。剩余物经柱层析纯化(石油醚∶乙酸乙酯=10∶1-5∶1),得到白色固体(95mg,9%)。1HNMR(400MHz,CDCl3)δ9.02(d,J=4.0Hz,1H),8.26(m,J=4.4Hz,2H),8.12(s,1H),8.08(d,J=5.2Hz,1H),7.93(m,1H),7.51(m,J=4.0Hz,1H),7.41(m,1H)。
3)中间体B1-4的合成:
将B1-3(70mg,0.29mmol)和对溴苄胺(107mg,0.58mmol)溶于2mL二甲基亚砜,加入碳酸钾(80mg,0.58mmol),加热到120℃反应8h。待冷却至室温,水2mL加入,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗。有机相经无水硫酸钠干燥,浓缩除去溶剂。剩余物经柱层析纯化(石油醚∶乙酸乙酯=10∶1-5∶1),得到棕色固体(70mg,59%)。1HNMR(400MHz,CDCl3)δ8.97(d,J=2.8Hz,1H),8.21(m,2H),8.05(s,1H),7.99(d,J=5.2Hz,1H),7.92(d,J=8.8Hz,1H),7.47(m,3H),7.30(d,J=8.2Hz,2H),6.78(m,1H),5.08(s,1H),4.72(d,J=5.7Hz,2H)。
3)产物B1的合成:
将中间体B1-4(50mg,0.12mmol)溶于3mL二氧六环和0.6mL水,加入2-甲基吡啶-4-
频哪醇硼酸酯(54mg,0.18mmol),1,1′-二(二苯膦基)二茂铁二氯化钯(II)(10mg,0.012mmol),碳酸钠(33mg,0.31mmol)。氮气置换,100℃下反应8h。冷却至室温,加入10mL水,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗涤。有机相经无水硫酸钠干燥,浓缩除去溶剂。剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1)得到白色固体(18mg,35%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B1。
实施例2
本实施例提供一种3-氟吡啶杂环化合物B2,其是由如下方法合成的:
1)中间体B2-2的合成:
将化合物B2-1(5g,34.3mmol)溶于乙腈(100mL),加入NBS(18.3g,103mmol)和过氧化苯甲酰(829mg,3.43mmol),回流过夜,冷却到常温,加入水(200mL),乙酸乙酯(200mL×3)萃取,有机相用饱和食盐水(300mL×3)洗,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=100∶1),得到黄色油状物(7.45g,97%)。1HNMR(400MHz,CDCl3)δ8.23(d,J=1.2Hz,1H),7.56-7.53(m,1H),4.45(s,2H)。
2)中间体B2-3的合成:
将中间体B2-2(7.45g,33.1mmol)溶于DMF(50mL),在冰浴下加入NaN3(8.61g,132mmol),50℃搅拌过夜,冷却到常温,加入水(100mL),乙酸乙酯(100mL×3)萃取,有机相用饱和食盐水(200mL×3)洗,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=50∶1),得到无色油状物(5.6g,90%)。1HNMR(400MHz,CDCl3)δ8.19(s,1H),7.50(d,J=8.0Hz,1H),4.45(s,2H)。
3)中间体B2-4的合成:
将中间体B2-3(5.6g,29.9mmol)溶于四氢呋喃(50mL),冰浴搅拌下分批加入PPh3(8.6g,32.9mmol),加热回流,缓慢滴加水(10mL),回流搅拌8小时,冷却到常温,乙酸乙酯(100mL)稀释,用0.2M的盐酸(100mL×2)萃取,水相用乙酸乙酯(100mL)洗一次,然后水相用2M的NaOH水溶液把PH调节到9,乙酸乙酯(200mL×3)萃取,得到的有机相用无水硫酸钠干
燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(二氯甲烷∶甲醇=50∶1),得到棕色油状物(2.57g,53%)。1HNMR(400MHz,DMSO)δ8.23(s,1H),7.89(d,J=9.6Hz,1H),3.75(s,2H)。
4)中间体B2-5的合成:
将中间体B2-4(2.36g,14.6mmol)溶于二氧六环/水(50mL/10mL),加入2-甲基吡啶-4-频哪醇硼酸酯(1.7g,16.1mmol),碳酸钾(8.09g,58.6mmol),四三苯基磷钯(847mg,0.73mmol),氮气置换,100℃搅拌过夜,冷却到常温,加入水(100mL),二氯甲烷(100mL×6)萃取,有机相用无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(二氯甲烷∶甲醇=50∶1-20∶1加氨水),得到棕色固体(2.5g,79%)。1HNMR(400MHz,CDCl3)δ8.61(d,J=4.8Hz,1H),8.50(s,1H),7.77(s,1H),7.69(d,J=4.0Hz,1H),7.58(d,J=12.0Hz,1H),4.01(s,2H),2.65(s,3H)。
5)产物B2的合成:
将B1-3(55mg,0.23mmol)和B2-5(59mg,0.27mmol)溶于2mL二甲基亚砜,加入碳酸钾(63mg,0.45mmol),加热到120℃反应8h。待冷却至室温,加入2mL水,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1-1∶1),得棕色固体(30mg,30%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B2。
实施例3
本实施例提供一种3-氟吡啶杂环化合物B3,其是由如下方法合成的:
1)中间体B3-2的合成:
将化合物B3-1(5g,25.4mmol)溶于乙腈(100mL),加入NBS(6.8g,38mmol)和过氧化苯甲酰(100mg,0.41mmol),回流过夜,然后补加过氧化苯甲酰(50mg,0.21mmol),再回流12h。冷却到常温,浓缩蒸除溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=100∶1-50∶1),得到固体(4.3g,61%)。
2)中间体B3-3的合成:
将中间体B3-2(2.3g,8.2mmol)溶于DMSO(40mL),在冰浴下加入NaN3(637mg,9.8mmol),常温下搅拌30分钟,加入水(100mL),乙酸乙酯(100mL×3)萃取,有机相用饱和食盐水(50mL×3)洗,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,得到白色固体(940mg,48%),直接用于下一步反应。
3)中间体B3-4的合成:
将中间体B3-3(940mg,3.95mmol)溶于四氢呋喃(15mL),冰浴搅拌下分批加入PPh3(1.14g,4.34mmol),然后缓慢滴加水(3mL),加热回流,搅拌过夜。待冷却至室温,乙酸乙酯(100mL)稀释,用0.2M的盐酸(20mL×2)萃取。水相合并,用乙酸乙酯(20mL)洗一次,然后水相用饱和碳酸钠水溶液把PH调节到9,二氯甲烷(40mL×5)萃取,得到的有机相用饱和氯化钠水溶液洗,经无水硫酸钠干燥,减压蒸除溶剂,得到固体(570mg,68%),直接用于下一步反应。
4)中间体B3-5的合成:
将中间体B3-4(570mg,2.69mmol)溶于二氧六环/水(15mL/3mL),加入2-甲基吡啶-4-频哪醇硼酸酯(45%,1.43g,2.96mmol),碳酸钾(1.12g,8.07mmol)和四(三苯基磷)钯(310mg,0.27mmol),氮气置换,100℃反应8h。待冷却到常温,反应液经硅藻土过滤,水(10mL)加入,乙酸乙酯(50mL×3)萃取。有机相合并,用无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1-1∶1),得到灰色固体(570mg,95%)。
5)产物B3的合成:
将B1-3(50mg,0.21mmol)和B3-5(56mg,0.25mmol)溶于1mL二甲基亚砜,加入碳酸钾(57mg,0.41mmol),加热到135℃反应12h。待冷却至室温,加入2mL水,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1-1∶1),得白色固体(20mg,23%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B3。
实施例4
本实施例提供一种3-氟吡啶杂环化合物B4,其是由如下方法合成的:
1)中间体B4-2的合成:
将化合物B4-1(3g,23.4mmol)溶于乙腈(60mL),加入NBS(5g,28mmol)和过氧化苯甲酰(57mg,0.23mmol),回流18h,然后补加过氧化苯甲酰(28mg,0.12mmol),再回流12h。冷却到常温,减压浓缩,剩余物经柱层析纯化(石油醚∶乙酸乙酯=100∶1-50∶1),得到固体(2.45g,51%)。
2)中间体B4-3的合成:
将中间体B4-2(2.9g,14mmol)溶于DMSO(58mL),在冰浴下加入NaN3(1g,15mmol),常温下搅拌30分钟,加入水(100mL),乙酸乙酯(100mL×3)萃取,有机相用饱和食盐水(100mL×3)洗,无水硫酸钠干燥,滤除硫酸钠,减压蒸除溶剂,得到白色固体(2.2g,90%),直接用于下一步反应。
3)中间体B4-4的合成:
将中间体B4-3(2.2g,13mmol)溶于四氢呋喃(50mL),冰浴搅拌下分批加入三苯基磷(3.8g,14mmol),然后缓慢滴加水(10mL),氮气气氛下加热回流,搅拌过夜。待冷却至室温,乙酸乙酯(100mL)稀释,用0.2M的盐酸(20mL×2)萃取。水相合并,用乙酸乙酯(20mL)洗一次,然后水相用饱和碳酸钠水溶液把PH调节到9,二氯甲烷(80mL×5)萃取,得到的有机相用饱和氯化钠水溶液洗,经无水硫酸钠干燥,减压蒸除溶剂,得到固体(1.4g,75%),直接用于下一步反应。
4)中间体B4-5的合成:
将中间体B4-4(1g,7mmol)溶于二氧六环/水(30mL/6mL),加入2-甲基吡啶-4-频哪醇硼酸酯(45%,3.7g,7.7mmol),碳酸钾(2.9g,21mmol)和四(三苯基磷)钯(809mg,0.7mmol),氮气置换,100℃反应8h。待冷却到常温,反应液经硅藻土过滤,水(10mL)加入,乙酸乙酯(50mL×3)萃取。有机相合并,用饱和食盐水洗,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1-1∶1),得到灰色固体(1.3g,93%)。
5)产物B4的合成:
将B1-3(50mg,0.21mmol)和B4-5(50mg,0.25mmol)溶于1mL二甲基亚砜,加入碳酸钾(57mg,0.41mmol),加热到135℃反应12h。待冷却至室温,加入2mL水,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1-1∶1),得白色固体(18mg,20%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B4。
实施例5
本实施例提供一种3-氟吡啶杂环化合物B5,其是由如下方法合成的:
将B1-3(80mg,0.33mmol)和B5-1(80mg,0.4mmol)溶于2mL二甲基亚砜,加入碳酸钾(91mg,0.66mmol),加热到135℃反应12h。待冷却至室温,加入20mL水,用乙酸乙酯萃取三次。合并有机相,用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=2∶1-1∶1),得白色固体(50mg,30%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B5。
实施例6
本实施例提供一种3-氟吡啶杂环化合物B6,其是由如下方法合成的:
将B1-2(84mg,0.35mmol)和B5-1(90mg,0.42mmol)溶于2mL二甲基亚砜,加入碳酸钾(98mg,0.7mmol),加热到135℃反应18h。待冷却至室温,加入30mL水,用乙酸乙酯(30mL*3)萃取。合并有机相,用饱和食盐水(30mL*3)洗涤,无水硫酸钠干燥,减压浓缩后经柱层析纯化(二氯甲烷∶甲醇=30∶1),得浅黄色固体(21mg,14%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B6。
实施例7
本实施例提供一种3-氟吡啶杂环化合物B7,其是由如下方法合成的:
1)中间体B7-2的合成:
将B7-1(167mg,0.6mmol)和2,3-二氟吡啶-4-硼酸(105mg,0.66mmol)溶于二氧六环(4mL)和水(1mL)中,加入Pd(dppf)Cl2(39mg,0.048mmol)、dppf(26mg,0.048mmol)和磷酸钾(254mg,1.2mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减压浓缩后经柱层析得白色固体(80mg,47%)。
2)产物B7的合成:
将B7-2(78mg,0.32mmol)和K2CO3(88mg,0.64mmol)溶于DMSO(2mL)中,加入B2-5(104mg,0.48mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得黄色固体(15mg,11%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B7。
实施例8
本实施例提供一种3-氟吡啶杂环化合物B8,其是由如下方法合成的:
将B1-3(121mg,0.5mmol)和K2CO3(138mg,1.0mmol)溶于DMSO(2mL)中,加入B8-1(120mg,0.65mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得黄色固体(15mg,7%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B8。
实施例9
本实施例提供一种3-氟吡啶杂环化合物B9,其是由如下方法合成的:
将B1-3(121mg,0.5mmol)和K2CO3(138mg,1.0mmol)溶于DMSO(2mL)中,加入B9-1(112mg,0.65mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(20mg,10%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B9。
实施例10
本实施例提供一种3-氟吡啶杂环化合物B10,其是由如下方法合成的:
1)中间体B10-2的合成:
将B10-1(2.5g,16.3mmol)溶于四氢呋喃/水(50mL/50mL),加入碳酸钠(5.2g,49mmol),碘(11g,41mmol),常温搅拌4小时,加入水(50mL),乙酸乙酯萃取(100mL*3),有机相用硫代硫酸钠溶液洗至颜色明显退去,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,得到灰白色固体(4.1g,90%)。图谱数据如下:1HNMR(400MHz,DMSO)δ11.41(s,1H),8.33(d,J=1.6Hz,1H),7.53(d,J=2.0Hz,1H),3.86(s,3H)。
2)中间体B10-3的合成:
将B10-2(2.7g,9.6mmol)溶于DMF(50mL),加入碳酸钾(2.0g,14.5mmol),碘甲烷(1.65g,11.6mmol),常温搅拌过夜。倒入水中(100mL),乙酸乙酯(100mL*3)萃取,有机相用饱和食盐水洗(150mL*6),无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=20∶1)得到白色固体(1.88g,66%)。图谱数据如下:1HNMR(400MHz,
CDCl3)δ8.58(s,1H),7.53(s,1H),3.97(s,3H),3.96(s,3H)。
3)中间体B10-4的合成:
将硼氢化钠(1.65g,43.3mmol)溶于乙醇(100mL),将B10-3(2.54g,8.67mmol)溶于乙醇(30mL)在冰浴搅拌下滴加到上述溶液中,滴毕常温搅拌20小时。减压蒸馏除去大部分乙醇,加入水(100mL),二氯甲烷萃取(100mL*3),有机相用无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,得到无色油状物(2.0g,87%)。图谱数据如下:1HNMR(400MHz,CDCl3)δ8.18(d,J=2.0Hz,1H),8.13(s,1H),7.94(s,1H),7.09(s,1H),4.71(s,2H),3.92(s,3H)。
4)中间体B10-5的合成:
将B10-4(2.0g,7.55mmol)溶于二氯甲烷(40mL),加入三乙胺(3.05g,30.2mmol),常温搅拌下缓慢滴加甲基磺酰氯(1.72g,15.1mmol),常温搅拌过夜。饱和食盐水洗(50mL*2),有机相用无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,得到黄色油状物(1.9g,89%)。
5)中间体B10-6的合成:
将B10-5(1.9g,6.71mmol)溶于DMSO(40mL),加入叠氮化钠(873mg,13.4mmol),常温搅拌8小时,加入水(50mL),乙酸乙酯(50mL*3)萃取,有机相用饱和食盐水洗(100mL*3),无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物过柱子(石油醚∶乙酸乙酯=10∶1)得浅黄色油状物(1.35g,69%)。图谱数据如下:1HNMR(400MHz,CDCl3)δ7.96(d,J=1.6Hz,1H),6.96(d,J=1.6Hz,1H),4.38(s,2H),3.94(s,3H)。
6)中间体B10-7的合成:
将B10-6(1.35g,4.66mmol)溶于THF(20mL),搅拌下缓慢加入三苯基膦(1.34g,5.12mmol),加完加热回流,加入水(2mL),回流搅拌8小时,冷却到常温,加入乙酸乙酯(40mL)稀释,用0.2M盐酸(50mL)萃取,水相用乙酸乙酯(50mL)洗一次,氢氧化钠固体调PH到10左右,二氯甲烷(50mL*6)萃取,合并有机相,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,得到浅黄色油状物(1.18g,96%)。图谱数据如下:1HNMR(400MHz,CDCl3)δ7.95(s,1H),7.07(s,1H),3.92(s,3H),3.89(s,2H)。
7)中间体B10-8的合成:
将B10-7(1.12g,4.24mmol),中间体11(1.51g,50%,5.52mmol)溶于二氧六环/水(20mL/4mL),加入碳酸钾(2.34g,17.0mmol)、四三苯基膦钯(245mg,0.212mmol)、氮气置换,回流搅拌过夜。冷却到常温,用硅藻土抽滤,滤液用乙酸乙酯(100mL)稀释后弃去水层,有机相用无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物过柱子(二氯甲烷∶甲醇=100∶1到50∶1)得到棕色油状物(730mg,75%)。图谱数据如下:1HNMR(400MHz,CDCl3)δ8.55(d,
J=5.2Hz,1H),8.27(s,1H),7.71(s,1H),7.67(d,J=5.2Hz,1H),7.38(s,1H),3.99(s,2H),3.93(s,3H),2.62(s,3H)。
8)产物B10的合成:
将B1-3(104mg,0.43mmol)和K2CO3(119mg,0.86mmol)溶于DMSO(2mL)中,加入B10-8(100mg,0.43mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得浅黄色固体(45mg,23%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B10。
实施例11
本实施例提供一种3-氟吡啶杂环化合物B11,其是由如下方法合成的:
将B1-3(121mg,0.5mmol)和K2CO3(138mg,1.0mmol)溶于DMSO(2mL)中,加入B11-1(122mg,0.65mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(30mg,14%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B11。
实施例12
本实施例提供一种3-氟吡啶杂环化合物B12,其是由如下方法合成的:
1)中间体B12-2的合成:
将B12-1(164mg,1.0mmol)和2,3-二氟吡啶-4-硼酸(175mg,1.1mmol)溶于二氧六环(4mL)和水(1mL)中,加入Pd(dppf)Cl2(65mg,0.08mmol)、dppf(44mg,0.08mmol)和磷酸钾(424mg,2.0mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减压浓缩后经柱层析得白色固体(130mg,53%)。
2)产物B12的合成:
将B12-2(10mg,0.04mmol)和DIPEA(41mg,0.32mmol)溶于NMP(2mL)中,加入B2-5(17mg,0.08mmol)。氮气保护下,加热至130℃,反应24小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=20∶1)得浅黄色固体(5mg,28%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B12。
实施例13
本实施例提供一种3-氟吡啶杂环化合物B13,其是由如下方法合成的:
1)中间体B13-2的合成:
将B13-1(3.5g,15.7mmol)溶于DMF(15mL),加入氰化锌(3.7g,36.4mmol)和Pd(PPh3)4(1.8g,0.157mmol),置换氮气,120℃搅拌过夜,冷却至室温,加硅藻土过滤,滤液加200mL乙酸乙酯稀释,饱和食盐水洗,乙酸乙酯相用无水硫酸钠干燥,减压浓缩,经柱层析纯化(石油醚∶乙酸乙酯=5∶1-2∶1),得白色固体(2.3g,86%)。
2)中间体B13-3的合成:
将B13-2(2.3g,13.6mmol)溶于乙腈(50mL),加入NBS(2.92g,16.4mmol)和过氧苯甲酰(331mg,0.14mmol),搅拌回流过夜。待冷却到常温,加入100mL乙酸乙酯和100mL水分层,水相用乙酸乙酯萃取三次,有机相合并,用无水硫酸钠干燥,减压浓缩,经柱层析纯化(石油醚∶乙酸乙酯=10∶1-5∶1),得到白色固体(2.5g,74.2%)。
3)中间体B13-4的合成:
将B13-3(2.5g,10mmol)溶于DMSO(50mL),加入NaN3(986mg,15mmol),50℃搅拌反应1h。冷却到常温,加入100mL乙酸乙酯和100mL水分层,水相用乙酸乙酯萃取三次,有机相合并,用无水硫酸钠干燥,减压浓缩,经柱层析纯化(石油醚∶乙酸乙酯=10∶1-5∶1),得到淡黄色固体(2g,94%)。
4)中间体B13-5的合成:
将B13-4(2g,9.5mmol)溶于四氢呋喃(90mL),加入三苯基膦(2.77g,10.5mmol),回流搅拌5min,再加入9mL水,继续回流搅拌1h。加入100mL乙酸乙酯和100mL1NHCl分层。水
相加入氨水调碱性,用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩,得到紫色粗品(890mg,51%)。
5)产物B13的合成:
将B13-5(90mg,0.49mmol)溶于NMP(1.5mL),加入B1-3(100mg,0.41mmol),DIPEA(420mg,3.3mmol),120℃搅拌过夜。冷却到常温,加入50mL乙酸乙酯和50mL水分层,水相用乙酸乙酯萃取三次。有机相合并,用无水硫酸钠干燥,减压浓缩,经柱层析纯化(二氯甲烷∶甲醇=100∶1-20∶1),得到淡黄色固体(18mg,9%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B13。
实施例14
本实施例提供一种3-氟吡啶杂环化合物B14,其是由如下方法合成的:
1)中间体B14-2的合成:
将B14-1(209mg,1.0mmol)和2,3-二氟吡啶-4-硼酸(175mg,1.1mmol)溶于二氧六环(4mL)和水(1mL)中,加入Pd(dppf)Cl2(65mg,0.08mmol)、dppf(44mg,0.08mmol)和磷酸钾(424mg,2.0mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减压浓缩后经柱层析得白色固体(100mg,41%)。
2)产物B14的合成:
将B14-2(40mg,0.17mmol)和DIPEA(170mg,1.32mmol)溶于NMP(2mL)中,加入B2-5(54mg,0.25mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得红色固体(20mg,28%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B14。
实施例15
本实施例提供一种3-氟吡啶杂环化合物B15,其是由如下方法合成的:
1)中间体B15-2的合成:
将B15-1(1.0g,7.2mmol)溶于DMF(20mL),加入硫代吗啉(890mg,8.63mmol),二异丙基乙基胺(1.9g,14mmol),40℃反应过夜,冷却到常温,加入水(100mL),有固体析出,抽滤并干燥得到灰白色固体(1.36g,92%)。1HNMR(400MHz,CDCl3)δ8.41(s,1H),7.62(d,J=8.8Hz,1H),6.59(d,J=8.8Hz,1H),4.05(s,4H),2.67(s,4H)。
2)中间体B15-3的合成:
将中间体B15-2(1.1g,5.4mmol)溶于二氯甲烷(50mL),冰浴搅拌下缓慢分批加入mCPBA(2.18g,85%,10.7mmol),0℃继续反应2小时。饱和碳酸氢钠溶液(20mL)加入,再搅拌30分钟,分出有机相。水相再用二氯甲烷萃取(20mL)。合并有机相,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂得到白色固体(1.28g,100%)。图谱数据如下:1HNMR(400MHz,CDCl3)δ8.48(s,1H),7.75(dd,J=8.8,1.6Hz,1H),6.77(d,J=8.8Hz,1H),4.25(s,4H),3.08(t,J=4.6Hz,4H)。
3)中间体B15-4的合成:
将中间体B15-3(600mg,2.53mmol)溶于THF/MeOH(15mL/9mL),加入雷尼镍(300mg),氨水(3mL),35℃反应4小时,用硅藻土抽滤,旋干溶剂,得到浅绿色固体(630mg,100%)。
4)产物B15的合成:
将B1-3(97mg,0.4mmol)和K2CO3(110mg,0.8mmol)溶于DMSO(2mL)中,加入B15-4(96mg,0.4mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(35mg,19%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B15。
实施例16
本实施例提供一种3-氟吡啶杂环化合物B16,其是由如下方法合成的:
1)中间体B16-2的合成:
将6-氯-3-氰基吡啶(1.5g,10.8mmol)溶于DMF(50mL),加入B16-1(2.59g,16.2mmol),DIPEA(2.79g,21.6mmol),100℃反应过夜。冷却到常温,水(100mL)加入,乙酸乙酯萃取(100mL*3),有机相用饱和食盐水洗(200mL*6),无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=5∶1-1∶2),得到灰白色固体(700mg,29%)。图谱数据如下:1HNMR(400MHz,CDCl3)δ8.39(s,1H),7.58(dd,J=8.8,1.6Hz,1H),6.48(d,J=8.8Hz,1H),5.58(s,1H),4.05-4.01(m,2H),3.37(t,J=5.8Hz,2H),2.98(s,3H)。
2)中间体B16-3的合成:
将B16-2(660mg,2.93mmol)溶于THF/MeOH(15mL/9mL),加入雷尼镍(300mg),氨水(3mL),35℃反应4小时,用硅藻土抽滤,旋干溶剂,得到浅绿色油状物(680mg,100%)。
3)产物B16的合成:
将B1-3(97mg,0.4mmol)和K2CO3(110mg,0.8mmol)溶于DMSO(2mL)中,加入B16-3(91mg,0.4mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(53mg,29%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B16。
实施例17
本实施例提供一种3-氟吡啶杂环化合物B17,其是由如下方法合成的:
将B1-3(97mg,0.4mmol)和K2CO3(110mg,0.8mmol)溶于DMSO(2mL)中,加入B17-1(82mg,0.4mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入
水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(20mg,12%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B17
实施例18
本实施例提供一种3-氟吡啶杂环化合物B18,其是由如下方法合成的:
1)中间体B18-2的合成:
将B18-1(200mg,1.0mmol)和B1-2(238mg,1.5mmol)溶于二氧六环(4mL)和水(1mL)中,加入Pd(dppf)Cl2(65mg,0.08mmol)、dppf(44mg,0.08mmol)和磷酸钾(424mg,2.0mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减压浓缩后经柱层析得白色固体(135mg,58%)。
2)产物B18的合成:
将B18-2(50mg,0.21mmol)和DIPEA(217mg,1.68mmol)溶于NMP(2mL)中,加入B2-5(69mg,0.32mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得黄色固体(40mg,44%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B18。
实施例19
本实施例提供一种3-氟吡啶杂环化合物B19,其是由如下方法合成的:
1)中间体B19-2的合成:
将B19-1(264mg,1.5mmol)和B1-2(358mg,2.25mmol)溶于二氧六环(8mL)和水(2mL)中,加入Pd(dppf)Cl2(55mg,0.075mmol)、dppf(42mg,0.075mmol)和磷酸钾(798mg,3.0mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减
压浓缩后经柱层析得白色固体(72mg,23%)。
2)产物B19的合成:
将B19-2(44mg,0.21mmol)和DIPEA(217mg,1.68mmol)溶于NMP(2mL)中,加入B2-5(69mg,0.32mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(50mg,59%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B19。
实施例20
本实施例提供一种3-氟吡啶杂环化合物B20,其是由如下方法合成的:
1)中间体B20-2的合成:
将B20-1(288mg,1.5mmol)和B1-2(358mg,2.25mmol)溶于二氧六环(8mL)和水(2mL)中,加入Pd(dppf)Cl2(55mg,0.075mmol)、dppf(42mg,0.075mmol)和磷酸钾(798mg,3.0mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减压浓缩后经柱层析得白色固体(90mg,26%)。
2)产物B20的合成:
将B20-2(47mg,0.21mmol)和DIPEA(217mg,1.68mmol)溶于NMP(2mL)中,加入B2-5(69mg,0.32mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(55mg,62%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B20。
实施例21
本实施例提供一种3-氟吡啶杂环化合物B21,其是由如下方法合成的:
1)中间体B21-2的合成:
将NaH(2.4g,60%,60mmol)置于100mL烧瓶中,加入干燥THF(40mL),冰浴下加入
B21-1(2.6g,15mmol),反应1小时,加入碘甲烷,升至常温反应3小时,加入饱和氯化铵溶液(40mL),用二氯甲烷萃取(50mL*3),无水硫酸钠干燥,减压浓缩后经柱层析(石油醚∶乙酸乙酯=1∶2)得黄色固体(2.5g,89%)。
2)中间体B21-3的合成:
将B21-2(199mg,1.0mmol)和B1-2(191mg,1.2mmol)溶于二氧六环(8mL)和水(2mL)中,加入Pd(dppf)Cl2(37mg,0.05mmol)、dppf(28mg,0.05mmol)和磷酸钾(532mg,2.0mmol)。氮气保护下,用氮气置换五次,加热至100℃,反应12小时。冷却至室温后,减压浓缩后经柱层析得白色固体(30mg,14%)。
3)产物B21的合成:
将B21-3(30mg,0.135mmol)和DIPEA(139mg,1.08mmol)溶于NMP(1mL)中,加入B2-5(44mg,0.202mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得白色固体(15mg,27%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B21。
实施例22
本实施例提供一种3-氟吡啶杂环化合物B22,其是由如下方法合成的:
1)中间体B22-2的合成:
向500mL的圆底烧瓶中加入DMF(6.1g,83.5mmol),冰浴下滴加POCl3(36g,235mmol),搅拌5分钟后加入化合物B22-1,回流搅拌1h。冷却到常温,反应液倒入冰水中,搅拌五分钟后滤出固体。固体用甲苯除水,剩余物经柱层析纯化(石油醚∶二氯甲烷=1∶1)得白色固体(5.1g,74%)。
2)中间体B22-3的合成:
将中间体B22-2(5.1g,25mmol)溶于四氢呋喃(150mL),加入碘单质(7.6g,30mmol)NH4OH(125mL),常温搅拌过夜。加入硫代硫酸钠固体搅拌30min,旋掉部分四氢呋喃,用乙酸乙酯(200mL*3)萃取。合并乙酸乙酯相,经无水硫酸钠干燥,减压浓缩,得黄色固
体(4.25g,84%)。
3)中间体B22-4的合成:
将中间体B22-3(2g,9.8mmol)溶于DMF(30mL),加入Pd(PPh3)4(1.14g,0.98mmol),三乙胺(16g,158mmol),甲酸(2.45g,53.3mmol),110℃搅拌2h。冷却到常温,加硅藻土过滤。滤液加100mL乙酸乙酯稀释,用100mL饱和食盐水洗。乙酸乙酯相干燥旋干,剩余物经柱层析纯化(石油醚∶乙酸乙酯=10∶1)得到白色固体(1.5g,91%)。
4)中间体B22-5的合成:
将中间体B22-4(1.5g,8.9mmol)溶于乙腈(50mL),加入NBS(2.06g,11.57mmol),过氧苯甲酰(216mg,0.89mmol),搅拌回流过夜。冷却到常温,加100mL乙酸乙酯和100mL水萃取。乙酸乙酯相干燥旋干,剩余物经柱层析纯化(石油醚∶二氯甲烷=2∶3)得到白色固体(743mg,34%)。
5)中间体B22-6的合成:
将中间体B22-5(743mg,3mmol)溶于DMSO(15mL),加入NaN3(293mg,4.5mmol),50℃搅拌1h。冷却到常温,加75mL乙酸乙酯和75mL水萃取。乙酸乙酯相干燥旋干,剩余物经柱层析纯化(石油醚∶乙酸乙酯=10∶1-5∶1)得到白色固体(392mg,62%)。
6)中间体B22-7的合成:
将中间体B22-6(392mg,1.87mmol)溶于THF(10mL),加入PPh3(541mg,2.1mmol),回流搅拌5min,加入1mL水,继续回流搅拌1h,加乙酸乙酯和1N HCl分层。水相加入氨水调成碱性,加入100mL乙酸乙酯萃取,合并乙酸乙酯相,减压浓缩得到粗品(340mg,99%),为淡黄色固体。
7)产物B22的合成:
将中间体B22-7(62mg,est.0.34mmol)溶于DMSO(1.5mL),加入B1-3(51mg,0.21mmol),K2CO3(58mg,0.42mmol),135℃搅拌过夜,冷却到常温,加入50mL乙酸乙酯和50mL水萃取,乙酸乙酯相干燥旋干,固体柱层析(二氯甲烷∶甲醇=100∶1到20∶1),得到红色固体(4mg,5%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B22。
实施例23
本实施例提供一种3-氟吡啶杂环化合物B23,其是由如下方法合成的:
1)中间体B23-2的合成:
将B23-1(2g,9.1mmol)溶于DMF(15mL),加入氰化锌(2.1g,18.0mmol)和Pd(PPh3)4(1.1g,0.09mmol),置换氮气,120℃搅拌过夜,冷却至室温,加硅藻土过滤,滤液加200mL乙酸乙酯稀释,饱和食盐水洗,乙酸乙酯相用无水硫酸钠干燥,减压浓缩,经柱层析纯化(石油醚∶乙酸乙酯=4∶1),得白色固体(1.5g,98%)。
2)中间体B23-3的合成:
将B23-2(1.5g,8.9mmol)溶于乙腈(80mL),加入NBS(1.9g,17.8mmol)和过氧苯甲酰(216mg,0.9mmol),搅拌回流过夜。待冷却到常温,加入100mL乙酸乙酯和100mL水分层,水相用乙酸乙酯萃取三次,有机相合并,用无水硫酸钠干燥,减压浓缩,经柱层析纯化(二氯甲烷∶石油醚=8∶1),得到白淡黄色固体(1g,45%)。
3)中间体B23-4的合成:
将B23-4(1g,4.1mmol)溶于DMSO(20mL),加入NaN3(395mg,6.2mmol),50℃搅拌反应1h。冷却到常温,加入100mL乙酸乙酯和100mL水分层,水相用乙酸乙酯萃取三次,有机相合并,用无水硫酸钠干燥,减压浓缩,经柱层析纯化(石油醚∶乙酸乙酯=10∶1-5∶1),得到棕黄色固体(800mg,93%)。
4)中间体B23-5的合成:
将B23-4(800mg,3.8mmol)溶于四氢呋喃(20mL),加入三苯基膦(1.1g,4.2mmol),回流搅拌5min,再加入2mL水,继续回流搅拌1h。加入100mL乙酸乙酯和100mL 1N HCl分层。水相加入氨水调碱性,用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩,得到淡黄色粗品(400mg,56%)。
5)产物B23的合成:
将B23-5(56mg,0.31mmol)溶于NMP(1.5mL),加入B1-3(50mg,0.21mmol),DIPEA(212mg,1.6mmol),120℃搅拌过夜。冷却到常温,加入50mL乙酸乙酯和50mL水分层,水相用乙酸乙酯萃取三次。有机相合并,用无水硫酸钠干燥,减压浓缩,经柱层析纯化(二
氯甲烷∶甲醇=100∶1-20∶1),得到淡绿色固体(4mg,3.2%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B23。
实施例24
本实施例提供一种3-氟吡啶杂环化合物B24,其是由如下方法合成的:
1)中间体B24-2的合成:
将乙二胺(14.5mL,216mmol)加入到B24-1(1g,7.2mmol)中,常温搅拌2小时。碳酸钾(4g,28.8mmol)加入,继续搅拌1小时。抽滤滤除固体,滤液减压浓缩。然后加入DMF,再减压浓缩,重复两次,至完全除去剩余的乙二胺,得褐色油状液体(1.2g,粗品),直接用于下一步。
2)产物B24的合成:
将中间体B24-2(40mg,0.24mmol)溶于NMP(1.5mL),加入B1-2(50mg,0.21mmol),DIPEA(215mg,1.64mmol),135℃搅拌过夜。冷却到常温,加入50mL乙酸乙酯和50mL水分层。水相用乙酸乙酯萃取三次。合并乙酸乙酯相,经无水硫酸钠干燥,减压浓缩。剩余物经柱层析纯化(二氯甲烷∶甲醇=200∶1-100∶1),得到白色固体(28mg,30%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B24。
实施例25
本实施例提供一种3-氟吡啶杂环化合物B25,其是由如下方法合成的:
将B3-5(104mg,0.4mmol)和DIPEA(206mg,1.6mmol)溶于NMP(1mL)中,加入B7-2(49mg,0.2mmol)。氮气保护下,加热至135℃,反应18小时。冷却至室温后,加入水(30mL),用乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(30mL*3)洗,无水硫酸钠干燥,减压浓缩后经柱层析(二氯甲烷∶甲醇=30∶1)得黄色固体(42mg,48%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B25。
实施例26
本实施例提供一种3-氟吡啶杂环化合物B38,其是由如下方法合成的:
1)中间体B38-2的合成:
将B38-1(1.7g,10mmol),2,3-二氟-4-碘吡啶(4.8g,20mmol),碳酸色(16.3g,50mmol),三(二亚苄基丙酮)二钯(460mg,0.5mmol)溶于甲苯中,在氮气保护下,加热回流12小时,冷却至室温,经硅藻土过滤旋干柱层析纯化(石油醚∶乙酸乙酯=4∶1-1∶1)得黄色固体(2.3g,88%).1H NMR(400MHz,CDCl3)δ8.48(d,J=4.2Hz,1H),7.72(d,J=5.6Hz,1H),7.45(d,J=7.6Hz,1H),7.17(m,1H),6.69(m,1H),4.57(s,2H),3.81(m,2H),3.18(m,2H).
2)中间体B38-2的合成:
将B38-2(50mg,0.21mmol)和B2-5(66mg,0.3mmol)溶于1mL二甲基亚砜,加入碳酸钾(131mg,1.0mmol),加热到160℃反应12h。待冷却至室温,加入5mL水,用乙酸乙酯萃取(10mL×3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,滤除硫酸钠,旋干溶剂,剩余物经柱层析纯化(石油醚∶乙酸乙酯=1∶1-0∶1),得黄色固体(4mg,5%)。经过核磁图谱解析(图谱数据见表1),所得到的固体为化合物B38。
表1列出了实施例1-26及其类似步骤获得的化合物B1-B38的解析结构和波谱数据。
表1
实施例27
本实施例对实施例1-26制备得到的3-氟吡啶杂环化合物B1-B25和B38,以及按照实施例1-26的类似步骤获得的化合物B26-B37对Wnt信号通路的抑制能力进行测定。
LWnt3A细胞(CRL-2647,ATCC)培养在含有10%胎牛血清(Hyclone)的DMEM培养基(Gibico)中。HEK293STF稳定克隆细胞(转染有“Super-TopFlash”TCF荧光报告质粒的HEK293细胞)培养在完全培养基(含有4mML-谷氨酰胺、1.5g/L碳酸氢钠、4.5g/L葡萄糖、6μg/mL杀稻瘟菌素和10%胎牛血清的DMEM培养基)中。当LWnt3A细胞和HEK293STF稳定克隆细胞培养至汇合度为90%时分别收获,并以1∶1的比例混合。100μL/孔的混合细胞培养液加入到96孔板中,使最终的细胞浓度为12000个细胞/孔,然后再培养24h。被测化合物用DMSO逐级稀释,然后再用DMEM培养基稀释到所要浓度。取20μL化合物溶液加入到前述的装有细胞培养液的96孔板中,然后于37℃孵化48h。最后每孔中加入50μL荧光素酶溶液(Brigh-Glo,Promega),常温下振摇5分钟。发光信号用酶标仪(PHERAstarFS,BMG)测定,然后根据不同浓度化合物对发光信号的抑制结果计算出化合物的IC50值(效价)。以B4和B7为例,如图1和图2所示,得到抑制结果图;其余化合物的测试图基本相同。整体结果如表2(杂环化合物B1-B38对Wnt信号通路的抑制能力测定实验的结果)所示。
表2
| 化合物 | IC50(nM) | 化合物 | IC50(nM) | 化合物 | IC50(nM) |
| B1 | 0.18 | B2 | 0.06 | B3 | 0.11 |
| B4 | 0.27 | B5 | 0.31 | B6 | 0.54 |
| B7 | 0.12 | B8 | 0.11 | B9 | 5.2 |
| B10 | 0.24 | B11 | 16 | B12 | 1 |
| B13 | 10 | B14 | 1 | B15 | 10 |
| B16 | 10 | B17 | 16 | B18 | 8 |
| B19 | 7 | B20 | 8 | B21 | 2 |
| B22 | 318 | B23 | 10 | B24 | 12 |
| B25 | 0.06 | B26 | 0.13 | B27 | 0.12 |
| B28 | 0.12 | B29 | 0.06 | B30 | 0.04 |
| B31 | 0.34 | B32 | 1.8 | B33 | 0.4 |
| B34 | 0.11 | B35 | 1.0 | B36 | 0.25 |
| B37 | 0.2 | B38 | 0.5 |
由上表2可见,本发明实施例得到的杂环化合物,能够作为Wnt信号通路的有效拮抗剂,能够有效阻断Wnt信号通路,从而能够用于治疗或预防因Wnt信号通路失常引起的病
症。
实施例28-CYP抑制率测试
待测底物的细胞色素P450酶抑制率是在人类肝微粒体中进行的。五个CYP同工酶和各自的探针底物及其浓度如下:CYP-1A2(Phenacetin,30μM),CYP-2C9(Tolutamide,100μM),CYP-2C19(S-mephenytoin,40μM),CYP-2D6(Dextromethorphan,5μM)和CYP-3A4(Midazolam,1μM)。所有的探针底物都是在接近或者低于其Kms的浓度下使用。将混合有HLM(0.2mg/mL),磷酸盐缓冲液(100mM,pH 7.4),NADPH(1μM),待测化合物(10μM)和各自CYP酶的底物在37℃的恒温水浴锅内进行孵育。在与NADPH的反应混合开始之前,需先将化合物孵育10min,使酶-抑制剂充分作用。之后在每种CYP酶特定的反应时间段(CYP1A2,CYP2D6和CYP3A4为10min;CYP2C9和CYP2C19为30min)内反应。反应结束后加入含适量内标的冰冷乙腈(100μM)淬灭反应。高速离心(13000转/分)10min后,将上清液待测样品LC-MS/MS,定量分析探针底物与CYP酶形成的独有代谢产物的浓度来测定CYP抑制率。每个待测底物至少独立地完成三次测试。
LC-MS/MS分析条件:所有样品都使用API4000QTRAP质谱仪的LC-MS/MS系统进行数据分析。该仪器配有两台LC-20AD泵和CBM-20A控制器,DGU-20A溶剂自动脱气泵和SIL-20A自动进样器(Shimadzu,Columbia,MD,USA)。博纳艾杰尔的Venusil XBP C18柱(50×2.1mm,5μM)作为HPLC分离柱,柱温为40℃,流速为0.3mL/min,总运行时间为6min。
MS/MS的定量:API4000QTRAP质谱仪在配有多个反应监测器(MRM)的电喷雾(ESI)正离子模式下运行。所有化合物和内标都在100ms的停留时间段内进行检测。其余MS/MS参数设置如下:离子源气帘压力,30psi;雾化气流压力,55psi;离子源喷雾电压4500V;离子源温度500℃。对于特定的选定离子的检测,去簇电压(DP)和碰撞能(CE)都需要根据检测物的不同进行最优化。最后的数据处理利用AB SCIEX Analysist 1.5.2数据收集和集成软件进行。
所得CYP抑制活性结果数据,见表3。
表3
Claims (13)
- 一种3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式I所示的结构:其中:A为未被取代或被1-6个R6基团取代的5-14元含有1-4个杂原子的杂芳环、6-12元芳环、3-6元环烷基或3-6元杂环烷基,所述杂芳环和杂环烷基的杂原子包括N、O和S中的一种或多种;或者A为未被取代或被1-2个R6基团取代的其中X1为N或CR6;B选自氢原子、氰基、卤代基、羟基、C3-8环烷基、C2-8烯基或C2-8炔基、C1-8磺酰基、C1-8磺酰胺基、C1-8烷氧基、C1-8酯基、C1-8酰基、C1-8酰胺基、C1-8氧代脲基、C1-8脲基、未被取代或被1-3个R12取代基取代的C1-8烷基、未被取代或被1-3个R12取代基取代的-NR7R8;或者B选自未被取代或被1-3个R9基团取代的苯环、未被取代或被1-3个R10基团取代的5-6元含有1-3个杂原子的杂芳环、未被取代或被1-3个R11基团取代的5-7元含有1-3个杂原子的饱和杂环,所述杂芳环和饱和杂环的杂原子包括N、O和S中的一种或多种;U为未被取代或被1-6个R13基团取代的6-12元芳环、未被取代或被1-6个R13基团取代的5-14元杂芳环、未被取代或被1-6个R13基团取代的5-7元杂环并苯环、未被取代或被1-6个R13基团取代的5-7元杂环并5-6元杂芳环、-OR14或-NR14R15,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为被5-6元芳环或杂芳环取代的C2-8烯基或C2-8炔基,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为未被取代或被1-2个R13基团取代的其中X2为N或CR13;R1、R2、R3各自独立地选自氢原子、C1-6烷基、被1-3个分别选自卤代基、羟基、氰 基、C1-3烷基、C3-5环烷基和C1-3烷氧基的基团取代的C1-6烷基;R4、R5分别选自氢原子、氰基、氨基、卤代基、C1-8烷基、C3-8环烷基、C1-8烷氧基、-NH-C1-3烷基;R6选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的氨基、C1-6烷基、C3-6环烷基或C1-6烷氧基;R7、R8分别选自未被取代或被1-3个R12取代的氢原子、C1-8烷基、C3-8环烷基;或者R7和R8连同它们连接的原子成环;R9、R10分别选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R9、R10分别选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基取代的氨基、C1-6烷基、C3-6环烷基或C1-6烷氧基;R11选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷氧基、C3-6环烷基、氧代、未被取代或被卤代基取代的C1-6烷基;R12选自氢原子、氰基、羟基、氨基、C1-6烷氧基、C1-6烷基、C3-6环烷基、C2-6脲基、C2-6氧代脲基、-S(O)2-C1-3烷基、-N(R16)S(O)2-C1-3烷基、-N(R16)C(O)-C1-3烷基;R13选自氢原子、氰基、卤代基、羟基、氨基、C1-6烷氧基、C3-6环烷基、-S(O)2-C1-3烷基、-C(O)-C1-3烷基、-C(O)O-C1-3烷基、氧代基、未被取代或被卤代基取代的C1-6烷基;R14、R15分别选自氢原子、C3-6环烷基、C1-6烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环、5-6元含有1-3个杂原子的杂环、被1-3个R20取代的C3-6环烷基、被1-3个R20取代的C1-6烷基、被1-3个R20取代的5-6元芳环、被1-3个R20取代的5-6元含有1-3个杂原子的杂芳环、被1-3个R20取代的5-6元含有1-3个杂原子的杂环;所述杂原子包括N、O和S中的一种或多种;或者R14和R15连同它们连接的N原子成环;R16、R17、R18、R19分别选自氢原子、未被取代或被卤代基取代的C1-6烷基;R20选自氢原子、氰基、羟基、氨基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;R21为氢原子或C1-6烷基。
- 根据权利要求1所述的3-氟吡啶杂环化合物,其特征在于:B选自氢原子、氰基、卤代基、羟基、C1-8磺酰基、C1-8磺酰胺基、C1-8烷氧基、C1-8酰基、C1-8酰胺基、C1-8氧代脲基、C1-8脲基、C1-8烷基、-NR7R8;或者B选自未被取代或被1-3个R9基团取代的苯环、未被取代或被1-3个R10基团取代的5-6元含有1-3个杂原 子的杂芳环、未被取代或被1-3个R11基团取代的5-7元含有1-3个杂原子的饱和杂环,所述杂芳环和饱和杂环的杂原子包括N、O和S中的一种或多种;R1为氢原子;R2、R3分别选自氢原子、C1-6烷基;R4、R5分别选自氢原子、氰基、卤代基、C1-8烷基;R6选自氢原子、卤代基、氰基、羟基、氨基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的氨基、C1-6烷基、C3-6环烷基或C1-6烷氧基;R7、R8分别选自氢原子、C3-8环烷基、未被取代或被1-3个R12取代的C1-8烷基;或者R7和R8连同它们连接的原子成环;R9、R10分别选自氢原子、卤代基、氰基、C1-6烷基、C1-6烷氧基;或者R9、R10分别选自被1-3个卤代基取代的C1-6烷基、被1-3个卤代基取代的C1-6烷氧基;R11选自氢原子、氧代基、未被取代或被卤代基取代的C1-6烷基;R12选自氢原子、氰基、C1-6烷基、C1-6磺酰胺基;R13选自氢原子、氰基、卤代基、氧代基、C1-6烷氧基、C3-6环烷基、C1-6酰基、未被取代或被卤代基取代的C1-6烷基;R14、R15分别选自氢原子、C3-6环烷基、未被取代或被1-3个R20取代的C1-6烷基;或者R14和R15连同它们连接的N原子成环;R16、R17、R18、R19各自独立地选自氢原子、未被取代或被卤代基取代的C1-6烷基;R20选自氢原子、氰基、羟基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;R21选自氢原子或C1-6烷基。
- 根据权利要求1-4任一项所述的3-氟吡啶杂环化合物,其特征在于:L为价键。
- 根据权利要求1-5任一项所述的3-氟吡啶杂环化合物,其特征在于:该3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式II所示的结构:其中,X3为N、或CR22;Y1、Y2、Y3、Y4各自独立地选自N或CR6;U为未被取代或被1-6个R13基团取代的6-12元芳环、未被取代或被1-6个R13基团取代的5-14元杂芳环、未被取代或被1-6个R13基团取代的5-7元杂环并苯环、未被取代或被1-6个R13基团取代的5-7元杂环并5-6元杂芳环、-OR14或-NR14R15,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为被5-6元芳环或杂芳环取代的C2-8烯基或C2-8炔基,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为未被取代或被1-2个R13基团取代的其中X2为N或CR13;L是价键;R1和R4为氢原子;R2和R3各自独立地为氢原子或C1-3烷基;R5选自氢原子、卤代基、氰基、C1-3烷基;R6选自氢原子、卤代基、氰基、羟基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的C1-6烷基、C3-6环烷基或C1-6烷氧基;R10选自氢原子、卤代基、未被取代或被卤代基取代的C1-6烷基;R13选自氢原子、氰基、卤代基、氧代基、C1-6烷氧基、C3-6环烷基、C1-6酰基、未被取代或被卤代基取代的C1-6烷基;R14、R15分别选自氢原子、C3-6环烷基、未被取代或被1-3个R20取代的C1-6烷基;或者R14和R15连同它们连接的N原子成环;R20选自氢原子、氰基、羟基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;R21、R22分别选自氢原子或C1-6烷基。
- 根据权利要求1-5任一项所述的3-氟吡啶杂环化合物,其特征在于:该3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式III所示的结构:其中:Y5、Y6、Y7、Y8、Y9、Y10各自独立地选自N或CR6;B为氰基;U为未被取代或被1-6个R13基团取代的6-12元芳环、未被取代或被1-6个R13基团取代的5-14元杂芳环、未被取代或被1-6个R13基团取代的5-7元杂环并苯环、未被取代或被1-6个R13基团取代的5-7元杂环并5-6元杂芳环、-OR14或-NR14R15,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为被5-6元芳环或杂芳环取代的C2-8烯基或C2-8炔基,所述杂环、杂芳环分别为含有1-4个杂原子的杂环或杂芳环,所述杂环或杂芳环的杂原子包括N、O和S中的一种或多种;或者U为未被取代或被1-2个R13基团取代的其中X2为N或CR13;L是价键;R1和R4是氢原子;R2和R3分别选自氢原子、甲基;R5选自氢原子、卤代基、氰基、C1-3烷基;R6选自氢原子、卤代基、氰基、羟基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的C1-6烷基、C3-6环烷基 或C1-6烷氧基;R13选自氢原子、氰基、卤代基、氧代基、C1-6烷氧基、C3-6环烷基、C1-6酰基、未被取代或被卤代基取代的C1-6烷基;R14、R15分别选自氢原子、C3-6环烷基、未被取代或被1-3个R20取代的C1-6烷基;或者R14和R15连同它们连接的N原子成环;R20选自氢原子、氰基、羟基、C1-6烷氧基、C1-6烷基、C3-6环烷基、5-6元芳环、5-6元含有1-3个杂原子的杂芳环,所述杂原子包括N、O和S中的一种或多种;R21为氢原子或C1-6烷基。
- 根据权利要求1-6任一项所述的3-氟吡啶杂环化合物,其特征在于该3-氟吡啶杂环化合物,及其药学上可接受的盐,具有式IV或式V所示的结构:其中,X4、X5、X6、X7、X8、X9各自独立选自N或CR13;L是价键;R1和R4为氢原子;R2和R3各自独立地为氢原子或C1-3烷基;R5选自氢原子、卤代基、氰基、C1-3烷基;R6选自氢原子、卤代基、氰基、羟基、C1-6烷基、C3-6环烷基、C1-6烷氧基;或者R6选自被1-3个选自卤代基、C1-3烷基、C3-6环烷基的取代基所取代的C1-6烷基、C3-6环烷基或C1-6烷氧基;R10选自氢原子、卤代基、未被取代或被卤代基取代的C1-6烷基;R13选自氢原子、氰基、卤代基、C1-6烷氧基、C3-6环烷基、未被取代或被卤代基取代的C1-6烷基;R23、R24分别选自氢原子、C1-3烷基;或者R23和R24合并为氧代基;R25、R26、R27、R28各自独立选自氢原子或C1-3烷基。
- 一种药物组合物,其包含治疗有效量的权利要求1-10任一项所述的3-氟吡啶杂环化合物及其药学上可接受的载体。
- 一种药物联用产品,其包括权利要求1-11任一项所述的3-氟吡啶杂环化合物或其药学上可接受的盐与抗肿瘤药物、抗菌药物、抗病毒药物、抗寄生虫病药物、中枢神经系统系统药物、抗骨质增生药物、糖尿病药物中的一种或几种。
- 权利要求1-11任一项所述的3-氟吡啶杂环化合物或者权利要求12所述的药物联合产品在制备用于治疗或预防与Wnt信号通路异常相关的病症的药物中的应用;所述与Wnt信号通路异常相关的病症包括乳腺癌、肺癌、膀胱癌、胰腺癌、肝癌、头颈鳞状上皮 癌、甲状腺癌、肉瘤、骨肉瘤、硬纤维瘤、黑色素瘤、前列腺癌、结肠直肠癌、卵巢癌、宫颈癌、食管癌、胃癌、骨髓瘤、淋巴瘤、套细胞淋巴瘤、皮肤T细胞淋巴瘤、慢性和非进行性贫血、自发性或原发性血小板增多症、特发性骨髓纤维化、肺纤维化、肾脏纤维化、肝纤维化、肝硬化、糖尿病性视网膜病、巨球蛋白血症、白血病、急性白血病、慢性白血病、淋巴性白血病、髓性白血病、骨髓增生异常综合症、骨髓增殖性病症、脑瘤、星形细胞瘤、髓母细胞瘤、许旺细胞瘤、原发性神经外胚瘤、垂体瘤、以及寄生虫病、血吸虫病和疟疾。
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN201610195733.4 | 2016-03-31 | ||
| CN201610195733 | 2016-03-31 | ||
| CN201610850358.2A CN107286136B (zh) | 2016-03-31 | 2016-09-26 | 一种3-氟吡啶杂环化合物及其应用 |
| CN201610850358.2 | 2016-09-26 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2017167150A1 true WO2017167150A1 (zh) | 2017-10-05 |
Family
ID=59962617
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2017/078305 Ceased WO2017167150A1 (zh) | 2016-03-31 | 2017-03-27 | 一种3-氟吡啶杂环化合物及其应用 |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2017167150A1 (zh) |
Cited By (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2020007273A1 (zh) * | 2018-07-02 | 2020-01-09 | 四川大学 | 新型雄激素受体抑制剂及其合成方法和应用 |
| WO2022086892A1 (en) * | 2020-10-21 | 2022-04-28 | Calithera Biosciences, Inc. | Interleukin 4 (il4)-induced gene 1 inhibitors and methods of use thereof |
Citations (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2013185353A1 (en) * | 2012-06-15 | 2013-12-19 | Curegenix Inc. | Compound as wnt signaling inhibitor, composition, and use thereof |
| WO2014141038A2 (en) * | 2013-03-11 | 2014-09-18 | Irm Llc | Markers associated with wnt inhibitors |
| CN104876912A (zh) * | 2015-04-08 | 2015-09-02 | 苏州云轩医药科技有限公司 | Wnt信号通路抑制剂及其应用 |
| WO2015140195A1 (en) * | 2014-03-20 | 2015-09-24 | Bayer Pharma Aktiengesellschaft | Novel compounds |
| WO2015140196A1 (en) * | 2014-03-20 | 2015-09-24 | Bayer Pharma Aktiengesellschaft | Inhibitors of the wnt signalling pathways |
| CN105142641A (zh) * | 2013-03-12 | 2015-12-09 | 广州源生医药科技有限公司 | 用于治疗癌症的化合物 |
| CN105254613A (zh) * | 2015-10-08 | 2016-01-20 | 苏州云轩医药科技有限公司 | 具有Wnt信号通路抑制活性的杂环化合物及其应用 |
-
2017
- 2017-03-27 WO PCT/CN2017/078305 patent/WO2017167150A1/zh not_active Ceased
Patent Citations (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2013185353A1 (en) * | 2012-06-15 | 2013-12-19 | Curegenix Inc. | Compound as wnt signaling inhibitor, composition, and use thereof |
| WO2014141038A2 (en) * | 2013-03-11 | 2014-09-18 | Irm Llc | Markers associated with wnt inhibitors |
| CN105142641A (zh) * | 2013-03-12 | 2015-12-09 | 广州源生医药科技有限公司 | 用于治疗癌症的化合物 |
| WO2015140195A1 (en) * | 2014-03-20 | 2015-09-24 | Bayer Pharma Aktiengesellschaft | Novel compounds |
| WO2015140196A1 (en) * | 2014-03-20 | 2015-09-24 | Bayer Pharma Aktiengesellschaft | Inhibitors of the wnt signalling pathways |
| CN104876912A (zh) * | 2015-04-08 | 2015-09-02 | 苏州云轩医药科技有限公司 | Wnt信号通路抑制剂及其应用 |
| CN105254613A (zh) * | 2015-10-08 | 2016-01-20 | 苏州云轩医药科技有限公司 | 具有Wnt信号通路抑制活性的杂环化合物及其应用 |
Cited By (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2020007273A1 (zh) * | 2018-07-02 | 2020-01-09 | 四川大学 | 新型雄激素受体抑制剂及其合成方法和应用 |
| CN110669049A (zh) * | 2018-07-02 | 2020-01-10 | 四川大学 | 新型雄激素受体抑制剂及其合成方法和应用 |
| CN110669049B (zh) * | 2018-07-02 | 2022-03-04 | 四川大学 | 新型雄激素受体抑制剂及其合成方法和应用 |
| WO2022086892A1 (en) * | 2020-10-21 | 2022-04-28 | Calithera Biosciences, Inc. | Interleukin 4 (il4)-induced gene 1 inhibitors and methods of use thereof |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20240425477A1 (en) | Ire1 small molecule inhibitors | |
| AU2018278311B2 (en) | IRE1 small molecule inhibitors | |
| KR101457966B1 (ko) | Pgds 억제제로서 피리미딘 아미드 화합물 | |
| WO2021143701A1 (zh) | 嘧啶-4(3h)-酮类杂环化合物、其制备方法及其在医药学上的应用 | |
| TWI865480B (zh) | 抑制pge2/ep4信號傳導的化合物、其製備方法及其在醫藥上的應用 | |
| KR20250067967A (ko) | 면역조절제로서 복소환형 화합물 | |
| WO2022253326A1 (zh) | Nlrp3炎症小体抑制剂及其应用 | |
| CN109867675B (zh) | 一种吡咯并嘧啶衍生的化合物、药物组合物以及其用途 | |
| TWI810172B (zh) | 嘧啶化合物及包括其的藥學組成物 | |
| CN101528744A (zh) | 用于调节c-kit和pdgfr受体的组合物和方法 | |
| WO2018010514A1 (zh) | 作为fgfr抑制剂的杂环化合物 | |
| WO2022171123A1 (en) | Egfr degraders and methods of use | |
| CN109415341B (zh) | 作为TGF-βR1抑制剂的苯并三氮唑衍生的α,β不饱和酰胺类化合物 | |
| WO2022012623A1 (en) | Degradation of (egfr) by conjugation of egfr inhibitors with e3 ligase ligand and methods of use | |
| WO2022206939A1 (zh) | 作为fgfr抑制剂的杂环化合物及其应用 | |
| WO2021078022A1 (zh) | 一种小分子化合物 | |
| BR112014004741B1 (pt) | Entidade química, seu uso e composição farmacêutica compreendendo a mesma | |
| IL305046A (en) | Pyridopyrimidinone derivative, the method of preparation thereof and its use | |
| EP4615850A1 (en) | Compounds for the degradation of egfr kinase | |
| WO2019057112A1 (zh) | 2-取代吡唑氨基-4-取代氨基-5-嘧啶甲酰胺类化合物、组合物及其应用 | |
| KR20090074179A (ko) | Ppar 조절제로서 유용한 1h인돌2카르복실산 유도체 | |
| WO2017167150A1 (zh) | 一种3-氟吡啶杂环化合物及其应用 | |
| EP3891130B1 (en) | Ire1 small molecule inhibitors | |
| JP2016531947A (ja) | 配座固定されたPI3K及びmTOR阻害剤 | |
| JP2018535949A (ja) | Wntシグナル伝達経路阻害剤およびその治療用途 |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 17773180 Country of ref document: EP Kind code of ref document: A1 |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 17773180 Country of ref document: EP Kind code of ref document: A1 |