CN108191857B - 6-substituted pyrido [2,3-D ] pyrimidines as protein kinase inhibitors - Google Patents

6-substituted pyrido [2,3-D ] pyrimidines as protein kinase inhibitors Download PDF

Info

Publication number
CN108191857B
CN108191857B CN201810053736.3A CN201810053736A CN108191857B CN 108191857 B CN108191857 B CN 108191857B CN 201810053736 A CN201810053736 A CN 201810053736A CN 108191857 B CN108191857 B CN 108191857B
Authority
CN
China
Prior art keywords
compound
alkyl
group
cyclopentyl
hrms
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201810053736.3A
Other languages
Chinese (zh)
Other versions
CN108191857A (en
Inventor
徐亮
蒋昌盛
何皓
程航
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shengke Pharmaceutical (Jiangsu) Co., Ltd.
Original Assignee
Shengke Pharmaceutical Jiangsu Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shengke Pharmaceutical Jiangsu Co ltd filed Critical Shengke Pharmaceutical Jiangsu Co ltd
Publication of CN108191857A publication Critical patent/CN108191857A/en
Application granted granted Critical
Publication of CN108191857B publication Critical patent/CN108191857B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides 6-substituted pyrido [2,3-d ] of the general formula (I)]Pyrimidines which are useful in the treatment of cell proliferative disorders. The novel compounds of the present invention are potent inhibitors of cyclin-dependent kinases 4 and 6(CDK 4/6).

Description

6-substituted pyrido [2,3-D ] pyrimidines as protein kinase inhibitors
Technical Field
The invention provides a 6-substituted pyrido [2,3-d ] pyrimidine compound as a cyclin dependent kinase inhibitor, in particular cyclin dependent kinases 4 and 6. The compounds of the invention are useful for the treatment of cancer, inflammation, and also for the treatment of neurodegenerative diseases such as parkinson's syndrome.
Background
Cancer is the second leading disease of death worldwide, and global cancer report 2014 issued by the world health organization states that global cancer patients and death cases are all increasing disturbingly, and nearly half of newly added cancer cases appear in asia, most of which are in china. It is also reported that global cancer cases are predicted to present a rapidly growing situation, increasing from 1400 million people in 2012 to 1900 million people in 2025 year by year to 2400 million people in 2035 year, where china will account for 21.9% of the global population. Organic structural data show that the cancer mortality rate of China is increased by 80 percent in the last 30 years, the number of cancer diseases is about 260 thousands every year, and the number of deaths is about 180 thousands.
The cell cycle is an important part of the vital activity of a cell. Research shows that the occurrence and development of various malignant tumors are closely related to disorder of cell cycle regulatory mechanism, so that the tumors are also considered as a cell cycle disease. Three scientists in the united states and the united kingdom discovered, after the 70's of the 20 th century, the important roles of cyclin-dependent kinases (CDKs) and cyclins in cell regulation. Thus, the 2001 nobel physiological/medical prize was won. With the progress of cell cycle regulation mechanism research, especially the core role of CDK in cell cycle regulation, cyclin dependent kinase has become the hot spot for current anticancer drugs in recent years.
Cyclin-dependent kinases are a class of serine (Ser)/threonine (Thr) kinases. As important signal transduction molecules within the cell, and the CDK-cyclin complex formed by cyclins, are involved in the growth, reproduction, dormancy or apoptosis of cells (Morgan d.o., annu. rev. cell. dev.biol.1997; 13261-. Unlike other kinases, CDKs must form a corresponding dimeric complex with cyclins in order to function. Cyclins are continuously expressed and degraded periodically and are respectively combined with CDKs which are activated by the cyclins instantaneously, and the phosphorylation of different substrates is catalyzed through the activity of the CDKs, so that the promotion and conversion effects on different phases of the cell cycle are realized. In addition, CDKs may bind to CDK inhibitors, blocking cell division. In humans, 13 CDK members (CDK 1-13) have been identified, corresponding to which 12 cyclins (A-L) are present. It is currently believed that CDK1, 2,4, 6 and 7 are primarily responsible for regulation in the cell cycle.
It has been shown that increased activity or transient abnormalities in activation of cyclin-dependent kinases lead to the formation of human tumors (Sherr C.J., Science 1996; 274: 1672-. Indeed, the formation of human tumors is commonly associated with alterations in the CDK protein itself or its regulators (Cordon-Cardo C., am. J. Pathol. 1995; 147, 545. 560; Karp J. E. and Broder S., nat. Med. 1995; 1: 309. 320; Hall. M. et al., adv. cancer Res. 1996; 68: 67-108). Protein inhibitors of naturally occurring CDKs such as p16 and p27 result in growth inhibition of lung cancer cell lines in vitro (Kamb A., Curr. Top. Microbiol. Immunol. 1998; 227: 139-148). Small molecule CDK inhibitors may also be useful in the treatment of cardiovascular disorders such as restenosis and atherosclerosis and other vascular disorders caused by abnormal cell proliferation. Vascular smooth muscle proliferation and intimal hyperplasia secondary to balloon angioplasty are inhibited by overexpression of the cyclin-dependent kinase inhibitor p21 protein (Chang M.W.et al, J.Clin.Invest., 1995; 96: 2260; Yang Z-Y.et al, Proc.Natl.Acad.Sci.1996; 93: 9905). Furthermore, CVT-313(Ki ═ 95nM), a purine CDK2 inhibitor, resulted in greater than 80% inhibition of neointimal formation in rats (Brooks e.e.et al, j.biol.chem.1997: 29207-. CDK inhibitors can be used to treat diseases caused by a variety of infectious agents, including fungi, protozoan parasites (e.g., plasmodium falciparum), and DNA and RNA viruses. For example, cyclin-dependent kinases are essential for viral replication secondary to Herpes Simplex Virus (HSV) infection (Schang L.M.et al, J.Virol.1998; 72:5626), a homologue of CDKs known to play a key role in yeast. Selective CDK inhibitors can be used to ameliorate the consequences of various autoimmune disorders. Chronic inflammatory disease rheumatoid arthritis is characterized by synovial tissue hyperplasia; inhibition of synovial tissue proliferation minimizes inflammation and prevents joint destruction. Expression of the CDK inhibitor p16 protein in synovial fibroblasts causes growth inhibition (Taniguchi K.et al, nat. Med.1999; 5: 760-767). Similarly, joint swelling was substantially inhibited by treatment with p 16-expressing adenovirus in a rat model of arthritis. CDK inhibitors may be effective against other cell proliferative disorders including psoriasis (characterized by keratinocyte hyperproliferation), glomerulonephritis and lupus. Certain CDK inhibitors may be used as chemoprotectants by their ability to inhibit cell cycle progression in normal untransformed cells (Chen et al, j. nat. cancer institute, 2000; 92: 1999-2008). Pretreatment of cancer patients with CDK inhibitors prior to the use of cytotoxic agents can reduce the side effects commonly associated with chemotherapy. Normally proliferating tissues are protected from cytotoxic effects by the action of selective CDK inhibitors. Review articles on small molecule inhibitors of cyclin-dependent kinases have noted the difficulty of identifying compounds that inhibit specific CDK proteins without inhibiting other enzymes.
A selective targeted therapeutic against CDK4 and/or CDK6 may play a role in (1) CDK4 gene amplification or high expression of proteins, such changes often occurring in well-differentiated and de-differentiated liposarcomas, and also in some other solid and hematologic malignancies; (2) the expansion or high expression of cyclin Dl, which is observed in mantle cell lymphomas, as well as in a variety of solid tumors; (3) loss of the p16INK4A gene (CDKN2A), also a common event in many cancers. In cells, the p16INK4A protein is a natural inhibitor of CDK4, and cancers with p16 loss are generally sensitive to inhibition by CDK 4. In ovarian cancer, for example, these cancer cell lines have low p16 protein levels and high Rb expression and would be sensitive to inhibitors of CDK 4. Because of the high similarity of the proteins of CDK6 and CDK4, current CDK4 inhibitors also have similar inhibitory effects on CDK6 protein kinases. Three CDK4/CDK6 inhibitors, Palbociclib (Palbociclib, PD0332991), LEE011 and LY2835219, which fail CDK4/6 to form a complex with cyclin D, effectively block the progression of the cell cycle from the G1 phase to the S phase, thereby achieving the purpose of inhibiting tumor cell proliferation, and exhibit antitumor activity in breast cancer, neuroblastoma, malignant rhabdomyoma, lymphoma, sarcoma and other tumors, either as a single agent or in combination with other targeted therapeutic drugs, have been clinically tested.
Disclosure of Invention
The present invention provides 6-substituted pyrido [2,3-d ] pyrimidines as inhibitors of cyclin dependent kinases, particularly cyclin dependent kinases 4 and 6(CDK 4/6). The compounds of the present invention are useful in the treatment of cell proliferative disorders such as restenosis, cancer and inflammation. The compounds of the invention further improve the pharmacokinetic properties, including metabolic stability and clearance, over existing drugs. Meanwhile, the compound has stronger activity in more specifically inhibiting cyclin-dependent kinase 4 or 6 and better selectivity for other kinases. In addition, the compounds of the present invention are readily synthesized and can be administered to a patient by a variety of methods.
In one aspect, the present invention provides a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug, or isotopic variation thereof, and mixtures thereof:
Figure BDA0001553119070000041
wherein:
A1selected from the group consisting of CR3Or N;
A2selected from the group consisting of CR3Or N;
A3selected from the group consisting of CR3Or N;
b is selected from C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl, wherein said C3-7Cycloalkyl or 3-7 membered heterocyclyl is optionally substituted by oxo or thioxo;
z is selected from O or S;
R1selected from H, halogen, -CN, -NO2、-OR5、-S-R5、-NR6R7、-C(O)OR5、-C(O)NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl;
R2selected from H, -C (O) OR5、-C(O)NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl, wherein said C is1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl is optionally substituted with 1,2 or 3RaSubstituted by groups;
R3selected from H, halogen, -CN, -NO2、-L-OR5、-L-S-R5、-L-NR6R7、-L-C(O)OR5、-L-C(O)NR6R7、-L-S(O)mR5、-L-S(O)mOR5、-L-S(O)mNR6R7、-O-C1-6alkylene-R4、C1-6Alkyl radical, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-11 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl, said groups optionally substituted with 1,2 or 3R4Substituted by groups;
R4selected from H, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Haloalkyl, -C1-6alkylene-OH, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl;
R5independently selected from H, C1-6Alkyl radical, C2-8Alkenyl radical, C2-8Alkynyl, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl;
R6and R7Independently selected from H, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl; or, R6、R7Together with the N atom, form a 3-7 membered heterocyclyl;
R8independently selected from H, C1-6Alkyl radical, C2-8Alkenyl radical, C2-8Alkynyl, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl;
Raindependently selected from H, halogen, -CN, -NO2、-OR5、-S-R5、-NR6R7、-C(O)OR5、-C(O)NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl;
R3and R5-R8Optionally substituted by 1,2 or 3RbIs substituted by radicals in which RbIndependently selected from H, halogen, -NO2Carbonyl, -L-CN, -L-OR5、-L-S-R5、-L-NR6R7、-L-C(O)R5、-L-C(S)R5、-L-C(O)OR5、-L-C(S)OR5、-L-C(O)-NR6R7、-L-C(S)-NR6R7、-L-O-C(O)R5、-L-O-C(S)R5、-L-N(R6)-C(O)-R5、-L-N(R6)-C(S)-R5、-L-S(O)mR5、-L-S(O)mOR5、-L-S(O)mNR6R7、-L-N(R6)-S(O)m-R5、-L-N(R6)-S(O)m-NR6R7、-L-N(R6)-C(O)OR5、-L-N(R6)-C(S)OR5、-L-O-C1-6alkylene-OR5、-L-C(O)-C1-6alkylene-NR6R7、-L-N(R6)-C(O)-NR6R7、-L-N(R6)-C(S)-NR6R7、-L-O-C(O)-NR6R7、-L-O-C(S)-NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl; wherein said C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl optionally further substituted with one or more of the following groups:
-L-CN、-NO2carbonyl, -L-OR5、-L-S-R5、-L-NR6R7、-L-C(O)R5、-L-C(S)R5、-L-C(O)OR5、-L-C(S)OR5、-L-C(O)-NR6R7、-L-C(S)-NR6R7、-L-O-C(O)R5、-L-O-C(S)R5、-L-N(R6)-C(O)-R5、-L-N(R6)-C(S)-R5、-L-S(O)mR5、-L-S(O)mOR5、-L-S(O)mNR6R7、-L-N(R6)-S(O)m-R5、-L-N(R6)-S(O)m-R6R7、-L-N(R6)-C(O)OR5、-L-N(R6)-C(S)OR5、-L-O-C1-6alkylene-OR5、-L-C(O)-C1-6alkylene-NR6R7、-L-N(R6)-C(O)-NR6R7、-L-N(R6)-C(S)-NR6R7、-L-O-C(O)-NR6R7、-L-O-C(S)-NR6R7
L is selected from the group consisting of a bond, -C1-6Alkylene-, -C2-6alkenylene-or-C2-6Alkynylene-;
m represents 0, 1 or 2;
n represents 0, 1,2,3,4, 5 or 6.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention, and optionally a pharmaceutically acceptable excipient.
In another aspect, the invention provides pharmaceutical compositions comprising a compound of the invention and a pharmaceutically acceptable excipient, which also contain an additional therapeutic agent.
In another aspect, the invention provides kits comprising a compound of the invention, and other therapeutic agents, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
In another aspect, the invention provides the use of a compound of the invention in the manufacture of a medicament for the treatment and/or prevention of a CDK (particularly CDK4 and/or CDK6) mediated disorder.
In another aspect, the invention provides a method of treating and/or preventing a CDK (particularly CDK4 and/or CDK6) mediated disorder in a subject, comprising administering to said subject a compound of the invention or a composition of the invention.
In another aspect, the present invention provides a compound of the invention or a composition of the invention for use in the treatment and/or prevention of a CDK (particularly CDK4 and/or CDK6) mediated disorder.
In particular embodiments, the disease includes a cell proliferative disorder, including but not limited to cancer, cardiovascular disorders, infectious diseases, chronic inflammatory diseases, autoimmune disorders, and other cell proliferative disorders. More specifically, the cancers include, but are not limited to, solid tumors and hematologic malignancies, such as breast cancer, neuroblastoma, malignant rhabdomyoma, well-differentiated and de-differentiated liposarcoma, glioma, lung cancer, colorectal cancer, gastric cancer, gastrointestinal stromal tumor (GIST), hepatocellular carcinoma, prostate tumor, sarcoma, ovarian cancer, cervical cancer, pancreatic cancer, melanoma, thyroid cancer, cholangiocarcinoma, endometrial cancer, kidney cancer, mesothelioma, lymphoma, leukemia, non-hodgkin's lymphoma, mantle cell lymphoma, anaplastic large cell lymphoma, Acute Myeloid Leukemia (AML), multiple myeloma. Such as restenosis, atherosclerosis, vascular smooth muscle proliferation and intimal hyperplasia following balloon angioplasty, and other vascular disorders caused by abnormal cell proliferation. Such infectious diseases include fungal, protozoan parasites (e.g., plasmodium falciparum), and DNA and RNA viral infections, such as Herpes Simplex Virus (HSV) infections. Such as rheumatoid arthritis. Such other cell proliferative disorders include psoriasis (characterized by keratinocyte hyperproliferation), glomerulonephritis, lupus, and the like.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
Definition of
Chemical definition
The definitions of specific functional groups and chemical terms are described in more detail below.
When a range of values is recited, it is intended to include each value and every subrange within the range. E.g. "C1-6Alkyl "includes C1、C2、C3、C4、C5、C6、C1-6、C1-5、C1-4、C1-3、C1-2、C2-6、C2-5、C2-4、C2-3、C3-6、C3-5、C3-4、C4-6、C4-5And C5-6An alkyl group.
It is to be understood that any of the moieties defined below may be substituted with a number of substituents when described herein, and that the corresponding definitions are within their scope as set forth below, including such substituted moieties. Unless otherwise specified, the term "substituted" is as defined below.
“C1-12Alkyl "means a straight or branched chain saturated monovalent alk (en) yl group containing 1 to 12 carbon atoms. In some embodiments, C1-6Alkyl groups are preferred. In some embodiments, C1-4Alkyl groups are particularly preferred. Typical C1-12Alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, dimethylpentyl, octyl, 2, 4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl and the like. The alkyl group may be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
“C2-8Alkenyl "represents a straight or branched chain hydrocarbon group having 2 to 8 carbon atoms and at least one carbon-carbon double bond, including but not limited to vinyl, 3-buten-1-yl, 2-vinylbutyl, 3-hexen-1-yl, and the like. In some embodiments, C2-6Alkenyl groups are preferred. In some embodiments, C2-4Alkenyl groups are particularly preferred. The term "C2-8Alkenyl "includes cycloalkenyl and heteroalkenyl, wherein from 1 to 3 nitrogen atoms selected from O, S, N or substituted may be substituted for a carbon atom. The alkenyl group may be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
“C2-8Alkynyl "refers to a straight or branched chain hydrocarbon group having 2 to 8 carbon atoms with one or more unsaturated carbon-carbon double bonds, with at least one carbon-carbon triple bond. In some embodiments, C2-6Alkynyl groups are preferred. In some embodiments, C2-4Alkynyl groups are particularly preferred. Typical alkynyl groups include ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl and hexenyl. The alkynyl group may be substituted at any available point of attachment,for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
“-C1-6Alkylene-, -C2-6alkenylene-or-C2-6Alkynylene- "means" C "as defined above1-6Alkyl radical, C2-6Alkenyl or C2-6Alkynyl "is a divalent radical.
“C1-6Alkylene "means removal of C1-6One hydrogen of the alkyl group forms a divalent alkylene group and may be a substituted or unsubstituted alkylene group. In some embodiments, C1-4Alkylene groups are particularly preferred. Unsubstituted said alkylene groups include, but are not limited to: methylene (-CH)2-) ethylene (-CH2CH2-) propylene (-CH)2CH2CH2-) butylene (-CH)2CH2CH2CH2-) pentylene (-CH)2CH2CH2CH2CH2-) and hexylene (-CH2CH2CH2CH2CH2CH2-) and the like. Exemplary substituted said alkylene groups, for example, said alkylene groups substituted with one or more alkyl (methyl) groups, include, but are not limited to: substituted methylene (-CH (CH)3)-、-C(CH3)2-) substituted ethylene (-CH (CH)3)CH2-、-CH2CH(CH3)-、-C(CH3)2CH2-、-CH2C(CH3)2-) Substituted propylene (-CH (CH)3)CH2CH2-、-CH2CH(CH3)CH2-、-CH2CH2CH(CH3)-、-C(CH3)2CH2CH2-、-CH2C(CH3)2CH2-、-CH2CH2C(CH3)2-) and the like.
“C2-6Alkenylene "means the removal of C2-6One hydrogen of an alkenyl group forms a divalent alkenylene group, and may be a substituted or unsubstituted alkenylene group. In some embodiments, C2-4Alkenylene is particularly preferred. Exemplary embodiments of the inventionThe unsubstituted alkenylene group of (a) includes, but is not limited to: vinylidene (-CH-) and propenylene (e.g., -CH-) CHCH2-、-CH2-CH ═ CH —). Exemplary substituted alkenylene groups, for example, alkenylene substituted with one or more alkyl (methyl) groups, include, but are not limited to: substituted ethylene (-C (CH)3)=CH-、-CH=C(CH3) -) substituted propenylene (-C (CH)3)=CHCH2-、-CH=C(CH3)CH2-、-CH=CHCH(CH3)-、-CH=CHC(CH3)2-、-CH(CH3)-CH=CH-、-C(CH3)2-CH=CH-、-CH2-C(CH3)=CH-、-CH2-CH=C(CH3) -) and the like.
“C2-6Alkynylene "means removal of C2-6One hydrogen of the alkynyl group forms a divalent alkynylene group, and may be a substituted or unsubstituted alkynylene group. In some embodiments, C2-4Alkynylene groups are particularly preferred. Exemplary such alkynylene groups include, but are not limited to: ethynylene (-C [ identical to ] C-), substituted or unsubstituted propynyl (-C [ identical to ] CCH)2-) and the like.
“C1-6Alkoxy "refers to the group-OR, where R is substituted OR unsubstituted C1-6An alkyl group. In some embodiments, C1-4Alkoxy groups are particularly preferred. Specific said alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, n-pentoxy, isopentoxy, n-hexoxy, cyclohexoxy, n-heptoxy, n-octoxy and 2-ethylhexoxy. The alkoxy group may be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
"halo" or "halogen" refers to fluorine, chlorine, bromine or iodine.
“C1-12Haloalkyl "represents the above-mentioned" C1-12Alkyl "substituted with one or more halo groups. Examples include monohalogen substituted, dihalogen substituted and polyhaloalkyl including perhalo. A single halogenThe substituents may have an iodine, bromine, chlorine or fluorine atom in the radical; two halogen substituents and multiple halogen substituents may have two or more of the same halogen atom or a combination of different halogens. The "lower haloalkyl" group contains 1 to 6 carbon atoms, i.e. C1-6A haloalkyl group. Examples of preferred haloalkyl groups include monofluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl. The haloalkyl group can be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
“C3-7Cycloalkyl "refers to a non-aromatic cyclic hydrocarbon group having 3 to 7 ring carbon atoms and zero heteroatoms. In some embodiments, C3-6Cycloalkyl is particularly preferred, more preferably C5-6A cycloalkyl group. Cycloalkyl also includes ring systems in which the aforementioned cycloalkyl ring is fused to one or more aryl or heteroaryl groups, where the point of attachment is on the cycloalkyl ring, and in such cases the number of carbons continues to represent the number of carbons in the cycloalkyl system. Exemplary such cycloalkyl groups include, but are not limited to: cyclopropyl (C)3) Cyclopropenyl group (C)3) Cyclobutyl (C)4) Cyclobutenyl radical (C)4) Cyclopentyl (C)5) Cyclopentenyl group (C)5) Cyclohexyl (C)6) Cyclohexenyl (C)6) Cyclohexyldienyl (C)6) Cycloheptyl (C)7) Cycloheptenyl (C)7) Cycloheptadienyl (C)7) Cycloheptatrienyl (C)7) And so on.
"3-11 membered heterocyclyl" refers to a radical of a 3 to 11 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon. In heterocyclic groups containing one or more nitrogen atoms, the point of attachment may be carbon or a nitrogen atom, as valency permits. In some embodiments, a 3-9 membered heterocyclic group is preferred which is a 3 to 9 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, the composition has advantagesA 3-7 membered heterocyclic group which is a 3-to 7-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms; preferably a 3-6 membered heterocyclic group which is a 3 to 6 membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; preferably a 4-6 membered heterocyclic group which is a 4 to 6 membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; more preferred are 5-6 membered heterocyclic groups which are 5 to 6 membered non-aromatic ring systems having ring carbon atoms and 1 to 3 ring heteroatoms. Heterocyclyl also includes ring systems wherein the aforementioned heterocyclyl ring is fused to one or more cycloalkyl groups, wherein the point of attachment is on the cycloalkyl ring, or ring systems wherein the aforementioned heterocyclyl ring is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring; and in such cases the number of ring members continues to represent the number of ring members in the heterocyclyl ring system. Exemplary 3-membered heterocyclic groups containing one heteroatom include, but are not limited to: aziridinyl, oxacyclopropaneyl, thienylyl. Exemplary 4-membered heterocyclic groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclic groups containing one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2, 5-dione. Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to: dioxolanyl, oxathiolanyl (oxathiolanyl), dithiolanyl (disulphuryl), and oxazolidin-2-one. Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl and thiadiazolinyl. Exemplary 6-membered heterocyclic groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridinyl and thiacyclohexyl (thianyl). Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithiinyl, dioxanyl. Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to: hexahydrotriazinyl (triazinanyl). Exemplary 7-membered heterocyclic groups containing one heteroatom include, but are not limited to: azepane, oxepanyl and thiepane.Exemplary with C6Aryl ring fused 5-membered heterocyclyl (also referred to herein as 5, 6-bicyclic heterocyclyl) includes, but is not limited to: indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolonyl, and the like. Exemplary with C6Aryl ring fused 6-membered heterocyclyl (also referred to herein as 6, 6-bicyclic heterocyclyl) includes, but is not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
3-11 membered heterocyclyl also includes spiroheterocyclyl, i.e., groups in which two rings (e.g., heterocycle and carbone) share a carbon atom, at least one of which is heterocyclyl as defined above. More specifically, the spiroheterocyclyl is a spirocyclic ring formed of two 4-membered rings, two 5-membered rings, two 6-membered rings, one 4-membered ring and one 5-membered ring, one 4-membered ring and one 6-membered ring, or one 5-membered ring and one 6-membered ring, wherein at least one ring is a 4-6-membered heterocyclyl group as defined above, preferably a 4-6-membered heterocyclyl group containing 1,2 or 3O, N or S heteroatoms, more preferably a 4-6-membered heterocyclyl group containing 1N heteroatom.
Specific examples of preferred heterocyclic groups include: pyrrolinyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, dihydropyranyl, dihydrofuranyl, thiazolidinyl, dihydrothiazolyl, 2, 3-dihydro-benzo [ l,4] dioxanyl, indolinyl, isoindolinyl, dihydrobenzothiophene, dihydrobenzofuranyl, isochromanyl, chromanyl, 1, 2-dihydroisoquinoline, 1,2,3, 4-tetrahydroisoquinoline, 1,2,3, 4-tetrahydroquinoline, 2,3,4,4a,9,9 a-hexahydro-lH-3-azafluorene, 5,6, 7-trihydro-1, 2, 4-triazolo [3,4-a ] isoquinolinyl, 3, 4-dihydro-2H-benzo [ l,4] oxazinyl, benzo [ l,4] dioxanyl, 2, 3-dihydro-lH-lk' -benzo [ d ] isothiazol-6-yl, 2, 3-di-benzo [ l,4] dioxinyl, dihydrobenzofuran, 2-oxoaziridin-1-yl, 2-oxoazetidin-1-yl, 2-oxopyrrolidin-1-yl, 2-oxoazacyclohex-1-yl, 2-oxoazepin-1-yl, 2-oxoazocin-1-yl, 2-oxoazepin-1-yl, 2-oxoazacyclodec-1-yl, aziridine, azetidine, pyrrolidinyl, azacyclohexane, azepane, azooctane, azocane, Azacyclononane, azacyclodecane, piperidinyl, piperazinyl, morpholinyl, diazaspiro [3.3] heptane, diazaspiro [3.4] octane, diazaspiro [3.5] nonane, diazaspiro [4.4] nonane, diazaspiro [4.5] decane, and diazaspiro [5.5] undecane.
“C6-10Aryl "refers to a group having a monocyclic or polycyclic (e.g., bicyclic) 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) of 6 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, an aryl group has six ring carbon atoms ("C)6Aryl "; for example, phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C)10Aryl "; e.g., naphthyl, e.g., 1-naphthyl and 2-naphthyl). Aryl also includes ring systems in which the aforementioned aryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the aryl ring system.
"5-10 membered heteroaryl" refers to a group having a 5-10 membered monocyclic or bicyclic 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) with ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur. In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems may include one or more heteroatoms in one or both rings. Heteroaryl also includes ring systems in which the aforementioned heteroaryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the heteroaryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the heteroaryl ring system. In some embodiments, 5-6 membered heteroaryl groups are particularly preferred, which are 5-6 membered monocyclic or bicyclic 4n +2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms. Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furanyl and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: a tetrazolyl group. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: a pyridyl group. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to: triazinyl and tetrazinyl. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepinyl, oxacycloheptyl, and thiacycloheptyl trienyl groups. Exemplary 5, 6-bicyclic heteroaryls include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl, benzisothiafuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, indezinyl, and purinyl. Exemplary 6, 6-bicyclic heteroaryls include, but are not limited to: naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl and quinazolinyl.
Specific examples of preferred heteroaryl groups include: pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl (4H-l,2, 4-triazolyl, 1H-1,2, 3-triazolyl, 2H-l,2, 3-triazolyl, pyranyl, 2-furyl, 3-furan, etc., 2-thienyl, 3-thienyl, oxazolyl, isoxazolyl, oxazolyl (1,2, 4-oxazolyl, 1,3, 4-oxazolyl, 1,2, 5-oxazolyl, thiazolyl, thiadiazolyl (1,2, 4-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl).
"carbonyl", whether used alone or in combination with other terms (e.g., aminocarbonyl), is designated as-C (O) -.
"oxo" represents ═ O.
Alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, and the like, as defined herein, are optionally substituted groups. Generally, the term "substituted," whether preceded by the term "optionally" or not, refers to the substitution of at least one hydrogen present on a group (e.g., a carbon or nitrogen atom) with an allowable substituent, e.g., a substituent that, upon substitution, results in a stable compound, e.g., a compound that does not spontaneously undergo transformation (e.g., by rearrangement, cyclization, elimination, or other reaction). Unless otherwise specified, a "substituted" group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, the substituent at each position is the same or different. The term "substituted" includes substitution with all permissible substituents of organic compounds (any of the substituents described herein that result in the formation of stable compounds). For the purposes of the present invention, a heteroatom such as nitrogen may have a hydrogen substituent and/or any suitable substituent described herein that satisfies the valence of the heteroatom and results in the formation of a stable moiety.
Exemplary substituents on carbon atoms include, but are not limited to: halogen, -CN, -NO2、-N3、-SO2H、-SO3H、-OH、-ORaa、-ON(Rbb)2、-N(Rbb)2、-N(Rbb)3 +X-、-N(ORcc)Rbb、-SH、-SRaa、-SSRcc、-C(=O)Raa、-CO2H、-CHO、-C(ORcc)2、-CO2Raa、-OC(=O)Raa、-OCO2Raa、-C(=O)N(Rbb)2、-OC(=O)N(Rbb)2、-NRbbC(=O)Raa、-NRbbCO2Raa、-NRbbC(=O)N(Rbb)2、-C(=NRbb)Raa、-C(=NRbb)ORaa、-OC(=NRbb)Raa、-OC(=NRbb)ORaa、-C(=NRbb)N(Rbb)2、-OC(=NRbb)N(Rbb)2、-NRbbC(=NRbb)N(Rbb)2、-C(=O)NRbbSO2Raa、-NRbbSO2Raa、-SO2N(Rbb)2、-SO2Raa、-SO2ORaa、-OSO2Raa、-S(=O)Raa、-OS(=O)Raa、-Si(Raa)3、-OSi(Raa)3、-C(=S)N(Rbb)2、-C(=O)SRaa、-C(=S)SRaa、-SC(=S)SRaa、-SC(=O)SRaa、-OC(=O)SRaa、-SC(=O)ORaa、-SC(=O)Raa、-P(=O)2Raa、-OP(=O)2Raa、-P(=O)(Raa)2、-OP(=O)(Raa)2、-OP(=O)(ORcc)2、-P(=O)2N(Rbb)2、-OP(=O)2N(Rbb)2、-P(=O)(NRbb)2、-OP(=O)(NRbb)2、-NRbbP(=O)(ORcc)2、-NRbbP(=O)(NRbb)2、-P(Rcc)2、-P(Rcc)3、-OP(Rcc)2、-OP(Rcc)3、-B(Raa)2、-B(ORcc)2、-BRaa(ORcc) Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RddSubstituted by groups;
or two geminal hydrogens on a carbon atom are replaced by groups ═ O, ═ S, ═ NN (R)bb)2、=NNRbbC(=O)Raa、=NNRbbC(=O)ORaa、=NNRbbS(=O)2Raa、=NRbbOr as NORccSubstitution;
Raaeach is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two RaaThe radicals combining to form a heterocyclic or heteroaromatic radicalRing wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RddSubstituted by groups;
Rbbeach independently selected from: hydrogen, -OH, -ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)2Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two RbbThe groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RddSubstituted by groups;
Rcceach is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two RccThe groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RddSubstituted by groups;
Rddeach independently selected from: halogen, -CN, -NO2、-N3、-SO2H、-SO3H、-OH、-ORee、-ON(Rff)2、-N(Rff)2,、-N(Rff)3 +X-、-N(ORee)Rff、-SH、-SRee、-SSRee、-C(=O)Ree、-CO2H、-CO2Ree、-OC(=O)Ree、-OCO2Ree、-C(=O)N(Rff)2、-OC(=O)N(Rff)2、-NRffC(=O)Ree、-NRffCO2Ree、-NRffC(=O)N(Rff)2、-C(=NRff)ORee、-OC(=NRff)Ree、-OC(=NRff)ORee、-C(=NRff)N(Rff)2、-OC(=NRff)N(Rff)2、-NRffC(=NRff)N(Rff)2、-NRffSO2Ree、-SO2N(Rff)2、-SO2Ree、-SO2ORee、-OSO2Ree、-S(=O)Ree、-Si(Ree)3、-OSi(Ree)3、-C(=S)N(Rff)2、-C(=O)SRee、-C(=S)SRee、-SC(=S)SRee、-P(=O)2Ree、-P(=O)(Ree)2、-OP(=O)(Ree)2、-OP(=O)(ORee)2Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RggSubstituted by radicals, or two geminal RddSubstituents may combine to form ═ O or ═ S;
Reeis independently selected from the group consisting of alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RggSubstituted by groups;
Rffeach is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two RffThe groups combine to form a heterocyclyl or heteroaryl ring, wherein,each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RggSubstituted by groups;
Rggis independently from each other: halogen, -CN, -NO2、-N3、-SO2H、-SO3H、-OH、-OC1-6Alkyl, -ON (C)1-6Alkyl radical)2、-N(C1-6Alkyl radical)2、-N(C1-6Alkyl radical)3 +X-、-NH(C1-6Alkyl radical)2 +X-、-NH2(C1-6Alkyl radical)+X-、-NH3 +X-、-N(OC1-6Alkyl) (C1-6Alkyl), -N (OH) (C)1-6Alkyl), -NH (OH), -SH, -SC1-6Alkyl, -SS (C)1-6Alkyl), -C (═ O) (C)1-6Alkyl), -CO2H、-CO2(C1-6Alkyl), -OC (═ O) (C)1-6Alkyl), -OCO2(C1-6Alkyl), -C (═ O) NH2、-C(=O)N(C1-6Alkyl radical)2、-OC(=O)NH(C1-6Alkyl), -NHC (═ O) (C)1-6Alkyl), -N (C)1-6Alkyl) C (═ O) (C)1-6Alkyl), -NHCO2(C1-6Alkyl), -NHC (═ O) N (C)1-6Alkyl radical)2、-NHC(=O)NH(C1-6Alkyl), -NHC (═ O) NH2、-C(=NH)O(C1-6Alkyl), -OC (═ NH) (C)1-6Alkyl), -OC (═ NH) OC1-6Alkyl, -C (═ NH) N (C)1-6Alkyl radical)2、-C(=NH)NH(C1-6Alkyl), -C (═ NH) NH2、-OC(=NH)N(C1-6Alkyl radical)2、-OC(NH)NH(C1-6Alkyl), -OC (NH) NH2、-NHC(NH)N(C1-6Alkyl radical)2、-NHC(=NH)NH2、-NHSO2(C1-6Alkyl), -SO2N(C1-6Alkyl radical)2、-SO2NH(C1-6Alkyl), -SO2NH2、-SO2C1-6Alkyl, -SO2OC1-6Alkyl, -OSO2C1-6Alkyl, -SOC1-6Alkyl, -Si (C)1-6Alkyl radical)3、-OSi(C1-6Alkyl radical)3、-C(=S)N(C1-6Alkyl radical)2、C(=S)NH(C1-6Alkyl), C (═ S) NH2、-C(=O)S(C1-6Alkyl), -C (═ S) SC1-6Alkyl, -SC (═ S) SC1-6Alkyl, -P (═ O)2(C1-6Alkyl), -P (═ O) (C)1-6Alkyl radical)2、-OP(=O)(C1-6Alkyl radical)2、-OP(=O)(OC1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Haloalkyl, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C7Carbocyclyl, C6-C10Aryl radical, C3-C7Heterocyclic group, C5-C10A heteroaryl group; or two geminal RggSubstituents may combine to form ═ O or ═ S; wherein, X-Are counter ions.
Exemplary substituents on the nitrogen atom include, but are not limited to: hydrogen, -OH, -ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRbb)Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)2Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R's attached to a nitrogen atomccThe groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3,4 or 5RddIs substituted by radicals, and wherein Raa、Rbb、RccAnd RddAs described above.
Other definitions
The term "cancer" includes, but is not limited to, the following cancers: breast, ovary, cervix, prostate, testis, esophagus, stomach, skin, lung, bone, colon, pancreas, thyroid, biliary tract, buccal and pharyngeal (oral), lip, tongue, oral cavity, pharynx, small intestine, colorectal, large intestine, rectum, brain and central nervous system, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, adenocarcinoma, adenoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma, kidney carcinoma, myeloid disorders, lymphoid disorders, hodgkin's disease, hairy cell carcinoma, and leukemia.
The term "treating" as used herein relates to reversing, alleviating, inhibiting the progression of, or preventing the disorder or condition to which the term applies, or one or more symptoms of such disorder or condition. The term "treatment" as used herein relates to the act of verb treatment, the latter being as just defined.
The term "pharmaceutically acceptable salts, esters, amides and prodrugs" as used herein means those carboxylic acid salts, amino acid addition salts, esters, amides and prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without undue toxicity, irritation, allergic response and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, including, where possible, the zwitterionic forms of the compounds of the present invention.
The term "salts" denotes the relatively non-toxic inorganic and organic acid addition salts of the compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds, or separately by reacting the purified free base form of the compound with a suitable organic or inorganic acid and isolating the resulting salt. As long as the compounds of the present invention are basic compounds, they are capable of forming a wide variety of different salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animals, in practice it is often necessary to first isolate a pharmaceutically unacceptable salt of the base compound from the reaction mixture and then simply convert it to the free base compound by treatment with a basic agent, after which the free base is converted to a pharmaceutically acceptable acid addition salt. The acid addition salts of the basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to form the salt in a conventional manner. The free base may be regenerated by contacting the salt form with a base and isolating the free base in a conventional manner. The free base forms differ somewhat from their respective salt forms in certain physical properties, such as solubility in polar solvents, but for the purposes of the present invention, the salts are also equivalent to their respective free bases.
Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali metal and alkaline earth metal hydroxides or organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines are N, N' -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine and procaine.
Base addition salts of acidic compounds can be prepared by contacting the free acid form with a sufficient amount of the desired base to form the salt in a conventional manner. The free acid may be regenerated by contacting the salt form with an acid and separating the free acid in a conventional manner. The free acid forms differ somewhat from their respective salt forms in certain physical properties, such as solubility in polar solvents, but the salts are also equivalent to their respective free acids for the purposes of the present invention.
The salts can be sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogen phosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides prepared from inorganic acids such as hydrochloric acid, nitric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, phosphoric acid, and the like. Representative salts include: hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthoate, mesylate, glucoheptonate, lactobionate, lauryl sulfonate, isethionate and the like. Salts may also be prepared from organic acids such as aliphatic mono-and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like. Representative salts include acetate, propionate, octanoate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoic acid basin, methylbenzoate, dinitrobenzoate, naphthoate, benzenesulfonate, tosylate, phenylacetate, citrate, lactate, maleate, tartrate, mesylate, and the like. Pharmaceutically acceptable salts can include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations, including but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Also contemplated are salts of amino acids such as arginate, gluconate, galacturonate, and the like (see, e.g., "pharmaceutical salts," J.pharm.Sci., 1977; 66:1-19, incorporated herein by reference).
Examples of pharmaceutically acceptable non-toxic amides of the compounds of the present invention include those derived from Cl-C6Alkyl esters, wherein the alkyl group is linear or branched. Acceptable esters also include C5-C7Cycloalkyl esters and arylalkyl esters, such as but not limited to benzyl esters. C1-C4Alkyl esters are preferred. Esters of the compounds of the invention may be prepared according to conventional methods, for example: march's advanced organic Chemistry,5Edition "m.b. smith&J.March,John Wiley&Sons,2001。
Examples of pharmaceutically acceptable non-toxic amides of the compounds of the present invention include those derived from ammonia, primary C1-C6Alkylamine and secondary C1-C6Dialkylamine derived amides in which the alkyl group is linear or branched. In the case of secondary amines, the amines may also beIn the form of a 5-or 6-membered heterocyclic ring containing one nitrogen atom. From ammonia, C1-C3Alkyl primary amines and C1-C2Secondary dialkylamine derived amides are preferred. Amides of the compounds of the invention may be prepared according to conventional methods, for example: march's Advanced organic chemistry,5Edition ", m.b. smith&J.March,John Wiley&Sons,2001。
The term "prodrug" means a compound that is rapidly converted in vivo to the parent compound of the above formula, for example, by hydrolysis in blood. For a thorough discussion see T.Higuchi and V.Ste11a, "Pro-drugs as Novel delivery systems," Vol.14of the A.C.S.Sympossium Series and Bioversiblecommunications in drug delivery, ed.Edward B.Roche, American Pharmaceutical Association and PergammonPress, 1987, both of which are incorporated by reference.
The "subject" to which the drug is administered includes, but is not limited to: a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an older adult)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. The terms "human", "patient" and "subject" are used interchangeably herein.
"disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise specified, the term "treatment" includes the effect that occurs when a subject has a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), and also includes the effect that occurs before the subject begins to have the particular disease, disorder or condition ("prophylactic treatment").
Generally, an "effective amount" of a compound is an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of a compound of the present invention may vary depending on the following factors: for example, biological goals, pharmacokinetics of the compound, the disease being treated, mode of administration, and the age, health, and condition of the subject. An effective amount includes a therapeutically effective amount and a prophylactically effective amount.
As used herein, unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with a disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" can include an amount that improves the overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
As used herein, unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or one or more symptoms associated with a disease, disorder, or condition, or to prevent the recurrence of a disease, disorder, or condition. A prophylactically effective amount of a compound refers to an amount of a therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in the prevention of a disease, disorder, or condition. The term "prophylactically effective amount" can include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic effect of other prophylactic agents.
"combination" and related terms refer to the simultaneous or sequential administration of a compound of the invention and another therapeutic agent. For example, the compounds of the present invention may be administered simultaneously or sequentially with the other therapeutic agent in separate unit dosage forms, or simultaneously with the other therapeutic agent in a single unit dosage form.
Detailed description of the preferred embodiments
Herein, "compound of the present invention" refers to the following compounds of formula (I) -formula (VIII), pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variations thereof, and mixtures thereof.
Compounds are generally described herein using standard nomenclature. Compounds having asymmetric centers, it is understood (unless otherwise indicated) that all optical isomers and mixtures thereof are included. Furthermore, all isomeric compounds and carbon-carbon double bonds encompassed by the present invention may occur as Z and E unless otherwise specified. Compounds that exist in different tautomeric forms, one such compound is not limited to any particular tautomer, but is intended to encompass all tautomeric forms. The general formula used for some compounds includes description, variables. Unless otherwise specified, each variable in such a formula is defined as a plurality of variables that are independent of any other variable and that independently define any one variable in a formula at each occurrence.
In one embodiment, the present invention relates to a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug, or isotopic variation thereof, and mixtures thereof:
Figure BDA0001553119070000181
wherein:
A1selected from the group consisting of CR3Or N;
A2selected from the group consisting of CR3Or N;
A3selected from the group consisting of CR3Or N;
b is selected from C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl, wherein said C3-7Cycloalkyl or 3-7 membered heterocyclyl is optionally substituted by oxo or thioxo;
z is selected from O or S;
R1selected from H, halogen, -CN, -NO2、-OR5、-S-R5、-NR6R7、-C(O)OR5、-C(O)NR6R7、C1-6Alkyl radical、C1-6Haloalkyl, C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl;
R2selected from H, -C (O) OR5、-C(O)NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl, wherein said C is1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl is optionally substituted with 1,2 or 3RaSubstituted by groups;
R3selected from H, halogen, -CN, -NO2、-L-OR5、-L-S-R5、-L-NR6R7、-L-C(O)OR5、-L-C(O)NR6R7、-L-S(O)mR5、-L-S(O)mOR5、-L-S(O)mNR6R7、-O-C1-6alkylene-R4、C1-6Alkyl radical, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-11 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl, said groups optionally substituted with 1,2 or 3R4Substituted by groups;
R4selected from H, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Haloalkyl, -C1-6alkylene-OH, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl;
R5independently selected from H, C1-6Alkyl radical, C2-8Alkenyl radical, C2-8Alkynyl, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl;
R6and R7Independently selected from H, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl; or, R6、R7Together with the N atomTo form a 3-7 membered heterocyclic group;
R8independently selected from H, C1-6Alkyl radical, C2-8Alkenyl radical, C2-8Alkynyl, C1-6Haloalkyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl;
Raindependently selected from H, halogen, -CN, -NO2、-OR5、-S-R5、-NR6R7、-C(O)OR5、-C(O)NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl;
R3and R5-R8Optionally substituted by 1,2 or 3RbIs substituted by radicals in which RbIndependently selected from H, halogen, -NO2Carbonyl, -L-CN, -L-OR5、-L-S-R5、-L-NR6R7、-L-C(O)R5、-L-C(S)R5、-L-C(O)OR5、-L-C(S)OR5、-L-C(O)-NR6R7、-L-C(S)-NR6R7、-L-O-C(O)R5、-L-O-C(S)R5、-L-N(R6)-C(O)-R5、-L-N(R6)-C(S)-R5、-L-S(O)mR5、-L-S(O)mOR5、-L-S(O)mNR6R7、-L-N(R6)-S(O)m-R5、-L-N(R6)-S(O)m-NR6R7、-L-N(R6)-C(O)OR5、-L-N(R6)-C(S)OR5、-L-O-C1-6alkylene-OR5、-L-C(O)-C1-6alkylene-NR6R7、-L-N(R6)-C(O)-NR6R7、-L-N(R6)-C(S)-NR6R7、-L-O-C(O)-NR6R7、-L-O-C(S)-NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl; wherein said C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, -L-C3-7Cycloalkyl, -L-3-7 membered heterocyclyl, -L-C6-10Aryl or-L-5-10 membered heteroaryl optionally further substituted with one or more of the following groups:
-L-CN、-NO2carbonyl, -L-OR5、-L-S-R5、-L-NR6R7、-L-C(O)R5、-L-C(S)R5、-L-C(O)OR5、-L-C(S)OR5、-L-C(O)-NR6R7、-L-C(S)-NR6R7、-L-O-C(O)R5、-L-O-C(S)R5、-L-N(R6)-C(O)-R5、-L-N(R6)-C(S)-R5、-L-S(O)mR5、-L-S(O)mOR5、-L-S(O)mNR6R7、-L-N(R6)-S(O)m-R5、-L-N(R6)-S(O)m-R6R7、-L-N(R6)-C(O)OR5、-L-N(R6)-C(S)OR5、-L-O-C1-6alkylene-OR5、-L-C(O)-C1-6alkylene-NR6R7、-L-N(R6)-C(O)-NR6R7、-L-N(R6)-C(S)-NR6R7、-L-O-C(O)-NR6R7、-L-O-C(S)-NR6R7
L is selected from the group consisting of a bond, -C1-6Alkylene-, -C2-6alkenylene-or-C2-6Alkynylene-;
m represents 0, 1 or 2;
n represents 0, 1,2,3,4, 5 or 6.
In another embodiment, the present invention relates to the above compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variations thereof, and mixtures thereof, having the general formula (II):
Figure BDA0001553119070000201
wherein the content of the first and second substances,
A1selected from the group consisting of CR3Or N;
A2selected from the group consisting of CR3Or N;
A3selected from the group consisting of CR3Or N;
b is selected from 5-6 membered heteroaryl and 3-7 membered heterocyclyl optionally substituted with oxo or thioxo;
R1selected from H, halogen, -CN, -NO2、-OR5、-S-R5、-NR6R7、-C(O)OR5、-C(O)NR6R7、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl;
R3is selected from C1-6Alkyl radical, C1-6Haloalkyl, -O-C1-6alkylene-R4Or 3-11 membered heterocyclyl, said group optionally substituted with 1,2 or 3R4Substituted by groups;
R4selected from H, C1-6Alkyl radical, C1-6Haloalkyl, -NH2、-NHC1-6Alkyl or-N (C)1-6Alkyl radical)2
R5、R6、R7And R8Independently selected from H, C1-6Alkyl or C1-6A haloalkyl group;
n is 0, 1 or 2.
In another embodiment, the present invention relates to the above compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variations thereof, and mixtures thereof, having the general formula (III):
Figure BDA0001553119070000211
wherein the content of the first and second substances,
b is selected from:
Figure BDA0001553119070000212
is the point of attachment to the parent nucleus;
R1selected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl, 3-7 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl;
R3is selected from C1-6Alkyl radical, C1-6Haloalkyl, -O-C1-6alkylene-R4Or the following groups:
Figure BDA0001553119070000213
Figure BDA0001553119070000221
R4selected from H, C1-6Alkyl radical, C1-6Haloalkyl, -NH2、-NHC1-6Alkyl or-N (C)1-6Alkyl radical)2
R5、R6、R7、R8Independently selected from H, C1-6Alkyl or C1-6A haloalkyl group;
n is 0, 1 or 2.
In another embodiment, the present invention relates to the above compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variations thereof, and mixtures thereof, having the general formula (IV):
Figure BDA0001553119070000222
wherein:
b is selected from:
Figure BDA0001553119070000223
is the point of attachment to the parent nucleus;
R1selected from H or Me;
R4selected from H or Me;
R8selected from H or Me;
n is 0 or 1.
In another embodiment, the present invention relates to the above compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variations thereof, and mixtures thereof, having the general formulae (V) - (VIII):
Figure BDA0001553119070000224
wherein A is1-A3、R1-R3、R8And n is as defined in any one of claims 1 to 4.
In another embodiment, the present invention relates to a compound of formula (V), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug, or isotopic variation thereof, and mixtures thereof:
Figure BDA0001553119070000231
wherein the content of the first and second substances,
A1selected from N, CH, C (halogen), C (C)1-6Alkyl group), C (C)1-6Haloalkyl) Or C (OC)1-6Alkyl groups); preferably, A1Selected from N, CH Or C (OC)1-6Alkyl groups); preferably, A1Selected from N, CH Or C (OCH)3);
A2Selected from N, CH, C (halogen), C (C)1-6Alkyl group), C (C)1-6Haloalkyl) Or C (OC)1-6Alkyl groups); preferably, A2Selected from N, CH, C (halogen), C (C)1-6Alkyl) Or C (OC)1-6Alkyl groups); preferably,A2Selected from N, CH, CF, CCH3Or C (OCH)3);
A3Selected from N, CH, C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A3Selected from N, CH or C (C)1-6Alkyl groups); preferably, A3Selected from N, CH or CCH3
R1Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R1Is selected from H or C1-6An alkyl group; preferably, R1Selected from H or Me;
R2is selected from C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R2Is C3-7A cycloalkyl group; preferably, R2Is cyclopentyl;
R3is selected from C1-6Alkyl radical, C1-6Haloalkyl, -O-C1-6alkylene-R4Or optionally substituted with 1,2 or 3R4The following groups substituted with groups:
Figure BDA0001553119070000232
preferably, R3Is selected from C1-6Alkyl radical, C1-6Haloalkyl, -O-C1-6alkylene-R4Or the following groups:
Figure BDA0001553119070000241
R4selected from H, C1-6Alkyl radical, C1-6Haloalkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2or-C1-6alkylene-OH; preferably, R4Selected from H, C1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2or-C1-6alkylene-OH; preferably, R4Selected from H, Me, Et, iPr, NH2、NMe2、CH2CH2OH or CH2CH2CH2OH;
R8Selected from H, C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R8Selected from H, C1-6Alkyl radical, C1-6Haloalkyl or C3-7A cycloalkyl group; preferably, R8Selected from H, Me, CF3Et, cyclopropyl or cyclobutyl.
In another embodiment, the present invention relates to a compound of formula (V), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug, or isotopic variation thereof, and mixtures thereof:
Figure BDA0001553119070000242
wherein the content of the first and second substances,
A1selected from N, CH, C (halogen), C (C)1-6Alkyl group), C (C)1-6Haloalkyl) Or C (OC)1-6Alkyl groups); preferably, A1Selected from N, CH Or C (OC)1-6Alkyl groups); preferably, A1Selected from N, CH Or C (OCH)3);
A2Selected from N, CH, C (halogen), C (C)1-6Alkyl group), C (C)1-6Haloalkyl) Or C (OC)1-6Alkyl groups); preferably, A2Selected from N, CH, C (halogen), C (C)1-6Alkyl) Or C (OC)1-6Alkyl groups); preferably, A2Selected from N, CH, CF, CCH3Or C (OCH)3);
A3Selected from CH and C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A3Selected from CH or C (C)1-6Alkyl groups); preferably, A3Selected from CH or CCH3
R1Is selected from C1-6Alkyl or C1-6A haloalkyl group; preferably, R1Is C1-6An alkyl group; preferably, R1Is Me;
R2is selected from C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R2Is C3-7A cycloalkyl group; preferably, R2Is cyclopentyl;
R3is selected from
Figure BDA0001553119070000251
Figure BDA0001553119070000252
R4Selected from H, C1-6Alkyl radical, C1-6Haloalkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2or-C1-6alkylene-OH; preferably, R4Selected from H, C1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2or-C1-6alkylene-OH; preferably, R4Selected from H, Me, Et, iPr, NH2、NMe2、CH2CH2OH or CH2CH2CH2OH;
R8Selected from H, C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R8Selected from H, C1-6Alkyl radical, C1-6Haloalkyl or C3-7A cycloalkyl group; preferably, R8Selected from H, Me, CF3Et, cyclopropyl or cyclobutyl.
In another embodiment, the present invention relates to a compound of formula (V), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug, or isotopic variation thereof, and mixtures thereof:
Figure BDA0001553119070000253
wherein the content of the first and second substances,
A1is N;
A2selected from CH and C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A2Selected from CH or C (C)1-6Alkyl groups); preferably, A2Selected from CH or CCH3
A3Selected from CH and C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A3Selected from CH or C (C)1-6Alkyl groups); preferably, A3Selected from CH or CCH3
R1Is selected from C1-6Alkyl or C1-6A haloalkyl group; preferably, R1Is C1-6An alkyl group; preferably, R1Is Me;
R2is selected from C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R2Is C3-7A cycloalkyl group; preferably, R2Is cyclopentyl;
R3is piperazinyl, optionally substituted with 1,2 or 3R4Substituted by groups; preferably, R3Is composed of
Figure BDA0001553119070000261
R4Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R4Is selected from H or C1-6An alkyl group; preferably, R4Selected from H, Me or Et;
R8selected from H, C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R8Selected from H, C1-6Alkyl radical, C1-6Haloalkyl or C3-7A cycloalkyl group; preferably, R8Selected from H, Me, CF3Et or cyclopropyl.
In another embodiment, the present invention relates to a compound of formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug, or isotopic variation thereof, and mixtures thereof:
Figure BDA0001553119070000262
wherein the content of the first and second substances,
A1is N;
A2selected from CH or C (C)1-6Alkyl groups); preferably, A2Is CH;
A3selected from CH or C (C)1-6Alkyl groups); preferably, A3Is CH;
R1is selected from C1-6Alkyl or C1-6A haloalkyl group; preferably, R1Is C1-6An alkyl group; preferably, R1Is Me;
R2is selected from C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R2Is C3-7A cycloalkyl group; preferably, R2Is cyclopentyl;
R3is a 3-7 membered heterocyclyl optionally substituted with 1,2 or 3R4Substituted by groups; preferably, R3Is composed of
Figure BDA0001553119070000263
R4Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R4Is selected from H or C1-6An alkyl group; preferably, R4Selected from H or Me;
R8is selected from C1-6Alkyl or C1-6A haloalkyl group; preferably, R8Is C1-6An alkyl group; preferably, R8Is Me.
In another embodiment, the present invention relates to compounds of general formula (VII) or (VIII), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variations thereof, and mixtures thereof:
Figure BDA0001553119070000271
wherein the content of the first and second substances,
A1is selected from N;
A2selected from CH and C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A2Is CH;
A3selected from CH and C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A3Selected from CH and C (C)1-6Alkyl) or C (C)1-6Haloalkyl); preferably, A3Selected from CH or C (C)1-6Alkyl groups); preferably, A3Selected from CH and C (CH)3);
R1Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R1Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R1Is selected from H or C1-6An alkyl group; preferably, R1Selected from H or Me;
R2is selected from C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R2Is selected from C3-7Cycloalkyl or 3-7 membered heterocyclyl; preferably, R2Is C3-7A cycloalkyl group; preferably, R2Is cyclopentyl;
R3is a 3-7 membered heterocyclyl optionally substituted with 1,2 or 3R4Substituted by groups; preferably, R3Is composed of
Figure BDA0001553119070000272
R4Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R4Selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R4Is selected from H or C1-6An alkyl group; preferably, R4Is Me;
R8selected from H, C1-6Alkyl or C1-6A haloalkyl group; preferably, R8Is selected from H or C1-6An alkyl group; preferably, R8Is H;
n is 0, 1 or 2; preferably, n is 0.
A1、A2And A3
In a particular embodiment, A1Is CR3(ii) a In another specific embodiment, A1Is CH; in another specific embodiment, A1Is N.
In a particular embodiment, A2Is CR3(ii) a In another specific embodiment, A2Is CH; in another specific embodiment, A2Is N.
In a particular embodiment, A3Is CR3(ii) a In another specific embodiment, A3Is CH; in another specific embodiment, A3Is N.
B
In one embodiment, B is C3-7A cycloalkyl group; in a particular embodiment, B is C substituted by oxo3-7A cycloalkyl group; in one embodiment, B is C substituted with thio3-7A cycloalkyl group; in another embodiment, B is C3-7A heterocycloalkyl group; in another embodiment, B is C substituted with oxo3-7A heterocycloalkyl group; in another embodiment, B is C substituted with thio3-7A heterocycloalkyl group; in another embodiment, B is C6-10An aryl group; in another embodiment, B is a 5-10 membered heteroaryl.
In a more specific embodiment, B is selected from
Figure BDA0001553119070000281
Figure BDA0001553119070000282
In another more particular embodiment, B is
Figure BDA0001553119070000283
In another more particular embodiment, B is
Figure BDA0001553119070000284
In another more particular embodiment, B is
Figure BDA0001553119070000285
At another placeIn a more particular embodiment, B is
Figure BDA0001553119070000286
In another more particular embodiment, B is
Figure BDA0001553119070000287
In another more particular embodiment, B is
Figure BDA0001553119070000288
In another more particular embodiment, B is
Figure BDA0001553119070000289
In another more particular embodiment, B is
Figure BDA00015531190700002810
In another more particular embodiment, B is
Figure BDA00015531190700002811
In another more particular embodiment, B is
Figure BDA00015531190700002812
In another more particular embodiment, B is
Figure BDA00015531190700002813
In another more particular embodiment, B is
Figure BDA00015531190700002814
In another more particular embodiment, B is
Figure BDA0001553119070000291
In another more particular embodiment, B is
Figure BDA0001553119070000292
In another more particular embodiment, B is
Figure BDA0001553119070000293
In another more particular embodiment, B is
Figure BDA0001553119070000294
In another more particular embodiment, B is
Figure BDA0001553119070000295
In another more particular embodiment, B is
Figure BDA0001553119070000296
In another more particular embodiment, B is
Figure BDA0001553119070000297
In another more particular embodiment, B is
Figure BDA0001553119070000298
In another more particular embodiment, B is
Figure BDA0001553119070000299
In another more particular embodiment, B is
Figure BDA00015531190700002910
In another more particular embodiment, B is
Figure BDA00015531190700002911
Wherein x is the point of attachment to the parent nucleus.
Z
In a specific embodiment, Z is O; in another specific embodiment, Z is S.
R1
In one embodiment, R1Is H; in another embodiment, R1Is halogen; in another embodiment, R1is-CN; in another embodiment, R1is-NO2(ii) a In another embodiment, R1is-OR5(ii) a In addition toIn one embodiment, R1is-S-R5(ii) a In another embodiment, R1is-NR6R7(ii) a In another embodiment, R1is-C (O) OR5(ii) a In another embodiment, R1is-C (O) NR6R7(ii) a In another embodiment, R1Is C1-6An alkyl group; in another embodiment, R1Is C1-6A haloalkyl group; in another embodiment, R1Is C3-7A cycloalkyl group; in another embodiment, R1Is a 3-7 membered heterocyclic group; in another embodiment, R1Is C6-10An aryl group; in another embodiment, R1Is a 5-10 membered heteroaryl.
R2
In one embodiment, R2Is H; in another embodiment, R2is-C (O) OR5(ii) a In another embodiment, R2is-C (O) NR6R7(ii) a In another embodiment, R2Is C1-6An alkyl group; in another embodiment, R2Is C1-6A haloalkyl group; in another embodiment, R2Is C3-7A cycloalkyl group; in another embodiment, R2Is a 3-7 membered heterocyclic group.
At R2In the above specific embodiment, the C1-6Alkyl radical, C1-6Haloalkyl, C3-7Cycloalkyl or 3-7 membered heterocyclyl is optionally substituted with 1,2 or 3RaAnd (4) substituting the group. In one embodiment, the group is substituted with 1RaSubstituted by groups; in another embodiment, the group is substituted with 2RaSubstituted by groups; in another embodiment, the group is substituted with 3RaAnd (4) substituting the group.
R3
In one embodiment, R3Is H; in another embodiment, R3Is halogenA peptide; in another embodiment, R3is-CN; in another embodiment, R3is-NO2(ii) a In another embodiment, R3is-L-OR5(ii) a In another embodiment, R3is-L-S-R5(ii) a In another embodiment, R3is-L-NR6R7(ii) a In another embodiment, R3is-L-C (O) OR5(ii) a In another embodiment, R3is-L-C (O) NR6R7(ii) a In another embodiment, R3is-L-S (O)mR5(ii) a In another embodiment, R3is-L-S (O)mOR5(ii) a In another embodiment, R3is-L-S (O)mNR6R7(ii) a In another embodiment, R3is-O-C1-6alkylene-R4(ii) a In another embodiment, R3Is C1-6An alkyl group; in another embodiment, R3Is C1-6A haloalkyl group; in another embodiment, R3is-L-C3-7A cycloalkyl group; in another embodiment, R3is-L-3-11 membered heterocyclyl; in another embodiment, R3is-L-3-9 membered heterocyclyl; in another embodiment, R3is-L-C6-10An aryl group; in another embodiment, R3is-L-5-10 membered heteroaryl.
At R3In the above specific embodiments, the group is optionally substituted with 1,2 or 3R4And (4) substituting the group. In one embodiment, the group is substituted with 1R4Substituted by groups; in another embodiment, the group is substituted with 2R4Substituted by groups; in another embodiment, the group is substituted with 3R4And (4) substituting the group.
In more specific embodiments, R3Is composed of
Figure BDA0001553119070000301
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000302
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000303
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000311
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000312
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000313
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000314
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000315
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000316
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000317
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000318
In another more particular embodiment, R3Is composed of
Figure BDA0001553119070000319
In another more particular embodiment, R3Is composed of
Figure BDA00015531190700003110
R4
In one embodiment, R4Is H; in another embodiment, R4is-NH2(ii) a In another embodiment, R4is-NHC1-6An alkyl group; in another embodiment, R4is-N (C)1-6Alkyl radical)2(ii) a In another embodiment, R4Is C1-6An alkyl group; in another embodiment, R4Is C1-6A haloalkyl group; in another embodiment, R4is-C1-6alkylene-OH; in another embodiment, R4is-L-C3-7A cycloalkyl group; in another embodiment, R4is-L-3-7 membered heterocyclyl; in another embodiment, R4is-L-C6-10An aryl group; in another embodiment, R4is-L-5-10 membered heteroaryl.
R5
In one embodiment, R5Is H; in another embodiment, R5Is C1-6An alkyl group; in another embodiment, R5Is C2-8An alkenyl group; in another embodiment, R5Is C2-8An alkynyl group; in another embodiment, R5Is C1-6A haloalkyl group; in another embodiment, R5is-L-C3-7A cycloalkyl group; in another embodiment, R5is-L-3-7 membered heterocyclyl; in another embodiment, R5is-L-C6-10An aryl group; in another embodiment, R5is-L-5-10 membered heteroaryl.
R6And R7
In one embodiment, R6And R7Independently is H; in another embodiment, R6And R7Independently is C1-6An alkyl group; in another embodiment, R6And R7Independently is C2-6An alkenyl group; in another embodiment, R6And R7Independently is C2-6An alkynyl group; in another embodiment, R6And R7Independently is C1-6A haloalkyl group; in another embodiment, R6And R7Independently is-L-C3-7A cycloalkyl group; in another embodiment, R6And R7independently-L-3-7 membered heterocyclyl; in another embodiment, R6And R7Independently is-L-C6-10An aryl group; in another embodiment, R6And R7independently-L-5-10 membered heteroaryl; or, R6、R7Together with the N atom, form a 3-7 membered heterocyclic group.
R8
In one embodiment, R8Is H; in another embodiment, R8Is C1-6An alkyl group; in another embodiment, R8Is C2-8An alkenyl group; in another embodiment, R8Is C2-8An alkynyl group; in another embodiment, R8Is C1-6A haloalkyl group; in another embodiment, R8is-L-C3-7A cycloalkyl group; in another embodiment, R8is-L-3-7 membered heterocyclyl; in another embodiment, R8is-L-C6-10An aryl group; in another embodiment, R8is-L-5-10 membered heteroaryl.
L
In a specific embodiment, L is a bond; in another embodiment, L is-C1-6Alkylene-; in another embodiment, L is-C2-6Alkenylene-; in another embodiment, L is-C2-6Alkynylene-.
m
In a specific embodiment, m is 0; in another embodiment, m is 1; in another embodiment, m is 2.
n
In a specific embodiment, n is 0; in another embodiment, n is 1; in another embodiment, n is 2; in another embodiment, n is 3; in another embodiment, n is 4; in another embodiment, n is 5; in another embodiment, n is 6.
Any of the above embodiments, or any combination thereof, may be combined with any of the other embodiments, or any combination thereof. For example, A1Any one or any combination of the above technical means, can be combined with A2、A3、B、Z、R1-R8L, m and n, or any combination thereof. The present invention is intended to include all combinations of these solutions, limited to space, not listed one by one.
The compounds of the invention may include one or more asymmetric centers and may therefore exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. Isomers may be separated from mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
One skilled in the art will appreciate that the organic compound may form a complex with a solvent in which it reacts or from which it precipitates or crystallizes. These complexes are referred to as "solvates". When the solvent is water, the complex is referred to as a "hydrate". The present invention encompasses all solvates of the compounds of the present invention.
The term "solvate" refers to a form of a compound or salt thereof that is combined with a solvent, typically formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like. The compounds described herein can be prepared, for example, in crystalline form, and can be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. "solvate" includes solvates in solution and isolatable solvates. Representative solvates include hydrates, ethanolates, and methanolates.
The term "hydrate" refers to a compound that is associated with an aqueous phase. In general, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, hydrates of the compounds can be used, for example, with the formula R.xH2O represents, wherein R is the compound, and x is a number greater than 0. A given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrates (x is a number greater than 0 and less than 1), e.g., hemihydrate (R0.5H)2O)) and polyhydrates (x is a number greater than 1, e.g. dihydrate (R.2H)2O) and hexahydrate (R.6H)2O))。
The compounds of the invention may be in amorphous or crystalline form (polymorphs). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "polymorph" refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof) in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may cause a crystalline form to dominate. Various polymorphs of a compound may be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those recited in formulas I-VIII, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and36and (4) Cl. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the invention, e.g. by incorporation of radioactive isotopes (e.g. by introducing3H and14C) can be used in drug and/or substrate tissue distribution assays. Tritium, i.e.3H and carbon-14, i.e.14The C isotopes are particularly preferred because of their ease of preparation and detection. Further, by heavier isotopes, e.g. deuterium, i.e.2H, may be preferred in some cases because of the higher metabolic stability that may provide therapeutic benefits, such as increased in vivo half-life or reduced dosage requirements. Isotopically labeled compounds of formulae I-VIII of the present invention and prodrugs thereof can generally be prepared by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent in carrying out the procedures disclosed in the schemes and/or in the examples and preparations below.
In addition, prodrugs are also included within the context of the present invention. The term "prodrug" as used herein refers to a compound that is converted in vivo by hydrolysis, for example in the blood, to its active form with a medicinal effect. Pharmaceutically acceptable Prodrugs are described in t.higuchi and v.stella, Prodrugs as Novel Delivery Systems, vol.14of a.c.s.symposium series, Edward b.roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press,1987, and d.fleisher, s.ramon and h.bara "Improved oral Drug Delivery: the solubility limits over company the use of drivers, Advanced Drug Delivery Reviews (1996)19(2)115-130, each of which is incorporated herein by reference.
A prodrug is any covalently bonded compound of the present invention that releases the parent compound in vivo when such prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a manner such that the modification is effected by routine manipulation or in vivo cleavage to produce the parent compound. Prodrugs include, for example, compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, cleaves to form a hydroxy, amino, or sulfhydryl group. Thus, representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of hydroxy, mercapto, and amino functional groups of the compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like may be used. The ester itself may be active and/or may hydrolyze under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which readily break down in the human body to release the parent acid or salt thereof.
The invention also provides a pharmaceutical formulation comprising a therapeutically effective amount of a compound of formulae I-VIII, or a therapeutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient thereof. All of these forms are within the scope of the present invention.
Pharmaceutical compositions, formulations and kits
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of a compound of the invention. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a compound of the invention. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of a compound of the present invention.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds formulated therewith. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The invention also includes kits (e.g., pharmaceutical packages). The provided kits can include a compound of the invention, an additional therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispensable packages or other suitable containers) containing the compound of the invention, the additional therapeutic agent. In some embodiments, provided kits may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent. In some embodiments, the compound of the present invention and the additional therapeutic agent provided in the first container and the second container are combined to form one unit dosage form.
Administration of drugs
The pharmaceutical compositions provided by the present invention may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implant or other modes of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of compound actually administered can be determined by a physician, as the case may be, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a condition according to the invention, a subject at risk of developing the condition is administered a compound provided herein, typically based on physician's advice and under the supervision of a physician, at a dosage level as described above. Subjects at risk of developing a particular disorder, typically include subjects with a family history of the disorder, or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
The pharmaceutical compositions provided herein may also be administered chronically ("chronic administration"). By long-term administration is meant administration of the compound or pharmaceutical composition thereof over a long period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue for an indefinite period of time, e.g., for the remainder of the subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, e.g., within the therapeutic window.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the targeted systemic level of the active ingredient through the body, e.g., intramuscular or subcutaneous bolus doses result in slow release of the active ingredient, while a bolus delivered directly to the vein (e.g., by IV intravenous drip) can be delivered more rapidly, resulting in a rapid rise in the concentration of the active ingredient in the blood to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV intravenous drip, to provide a steady state concentration of the active ingredient in the body of the subject. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More generally, however, the compositions are provided in unit dosage form for convenient administration of the precise dosage. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable for the purpose of producing the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of liquid compositions, or in the case of solid compositions, pills, tablets, capsules and the like. In such compositions, the compound is typically a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being various carriers or excipients and processing aids useful in forming the desired form of administration.
For oral dosages, a representative regimen is one to five oral dosages, particularly two to four oral dosages, typically three oral dosages per day. Using these dosing modes, each dose provides about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing about 0.1 to about 10mg/kg, especially about 1 to about 5 mg/kg.
In order to provide a blood level similar to, or lower than, the use of the injected dose, a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
From about 1 to about 120 hours, especially 24 to 96 hours, the injection dosage level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour. To obtain sufficient steady state levels, a preload bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered. For human patients of 40 to 80kg, the maximum total dose cannot exceed about 2 g/day.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, as well as buffers, suspending and dispersing agents, coloring and flavoring agents, and the like. Solid forms may include, for example, any of the following components, or compounds with similar properties: a binder, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose, disintegrants, for example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injection, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with a cream base, for example of the oil-in-water type. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and compositions are included within the scope of the present invention.
The compounds of the present invention may also be administered by transdermal means. Thus, transdermal administration can be achieved using a reservoir (reservoir) or porous membrane type, or a patch of various solid matrices.
The above components of the compositions for oral, injectable or topical administration are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences,17th edition,1985, Mack Publishing Company, Easton, Pennsylvania, section 8, which is incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6,7 and 8 α -1, 4-linked glucose units, respectively, which optionally include one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated and sulfoalkyl ether substitution. In some embodiments, the cyclodextrin is sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, e.g., U.S.5,376,645. In some embodiments, the formulation includes hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Treatment of
The present invention provides methods of treating a mammal (including a human) for a disorder or condition selected from the group consisting of: cell proliferation disorders such as cancer, vascular smooth muscle proliferation associated with atherosclerosis, post-operative vascular stenosis, restenosis and endometriosis; infections, including viral infections, e.g., DNA viruses, such as herpes, and RNA viruses, such as HIV, and fungal infections; autoimmune diseases, such as psoriasis, inflammation, such as rheumatoid arthritis, lupus, type I diabetes, diabetic nephropathy, multiple sclerosis and glomerulonephritis; organ transplant rejection, including host versus graft disease, comprising administering to said mammal a therapeutically effective amount of a compound of the present invention or a composition thereof.
The invention further provides compounds of the invention useful for treating abnormal cell proliferation, such as cancer. The invention further provides a method of treating abnormal cell proliferation, such as a cancer selected from: breast, ovary, cervix, prostate, testis, esophagus, stomach, skin, lung, bone, colon, pancreas, thyroid, biliary tract, buccal and pharyngeal (oral), lip, tongue, oral cavity, pharynx, small intestine, colorectal, large intestine, rectum, brain and central nervous system, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, adenocarcinoma, adenoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma, kidney carcinoma, myeloid disorders, lymphoid disorders, hodgkin's disease, hairy cell carcinoma, and leukemia comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of the present invention or a composition thereof.
Further, the present invention relates to a method of treating a subject suffering from a disease caused by vascular smooth muscle cell proliferation. The compounds of the present invention effectively inhibit the proliferation and migration of vascular smooth muscle cells. The method comprises administering to a subject in need thereof an amount of a compound of the invention or composition thereof sufficient to inhibit vascular smooth muscle proliferation and/or migration.
The present invention further provides a method of treating a subject suffering from gout comprising administering to said subject in need of treatment a compound of the present invention or a composition thereof in an amount sufficient to treat the condition.
The invention further provides a method of treating a subject having a renal disease, e.g., polycystic kidney disease, comprising administering to said subject in need thereof a compound of the invention or composition thereof in an amount sufficient to treat the condition.
Due to their inhibitory activity against CDKs and other kinases, the compounds of the present invention are also useful research tools for studying the mechanism of action of these kinases in vitro and in vivo.
The above treatment method is preferably carried out as follows: administering to a subject in need thereof a therapeutically effective amount of a compound of the invention (as described below). The compounds of the invention are potent inhibitors of cyclin dependent kinase 4(CDK 4). These compounds are readily synthesized, can be administered by a variety of routes, including oral and parenteral, with little or no toxicity.
The compounds of the present invention are selective inhibitors of the cyclin dependent kinase CDK4, that is, they inhibit CDK4 more efficiently than they inhibit tyrosine kinases and other serine-threonine kinases, including other cyclin dependent kinases, such as CDK 2. Although they are selective for CDK4 inhibition, the compounds of the invention may also inhibit other kinases, although at concentrations above those at which they inhibit CDK 4. However, the compounds of the invention may also inhibit CDK6 at concentrations similar to those necessary to inhibit CDK4, since CDK6 is structurally similar to CDK4 and performs similar functions.
The compounds of the invention are useful in the treatment of cancer (e.g., leukemia and cancers of the lung, breast, prostate and skin, such as melanoma) and other proliferative diseases, including but not limited to psoriasis, HSV, HIV, restenosis and atherosclerosis. To treat cancer with a compound of the present invention, a therapeutically effective amount of a pharmaceutically acceptable composition comprising at least one compound of the present invention is administered to a patient in need of such treatment, e.g., a patient suffering from cancer or another proliferative disease.
An effective amount of a compound of the invention is generally in a mean daily dose of from 0.01mg to 50mg of compound per kilogram of body weight of the patient, preferably from 0.1mg to 25mg of compound per kilogram of body weight of the patient, in single or multiple administrations. Generally, the compounds of the present invention may be administered to such patients in need of such treatment in a daily dosage range of from about 1mg to about 3500mg, preferably from 10mg to 1000mg per patient. For example, the daily dose per patient may be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000 mg. Administration may be once or more daily, weekly (or at intervals of several days), or on an intermittent schedule. For example, the compound may be administered one or more times per day on a weekly basis (e.g., monday), indefinitely or for several weeks, e.g., 4-10 weeks. Alternatively, the administration may be continued daily for several days (e.g., 2-10 days), followed by several days (e.g., 1-30 days) without administration of the compound, with the cycle repeated indefinitely or for a given number of times, e.g., 4-10 cycles. For example, the compounds of the invention may be administered daily for 5 days, followed by 9 days, followed by 5 days, followed by 9 days, and so on, with the cycle repeated indefinitely or 4-10 times in total.
Combination therapy
The compounds or compositions of the present invention may be administered concurrently with, before, or after one or more additional agents, for use as a combination therapy. The medicament includes a therapeutically active agent. The medicament also includes a prophylactically active agent. Agents include small organic molecules, such as pharmaceutical compounds (e.g., human or veterinary compounds approved by the U.S. food and drug administration as provided in the federal regulations Compilation (CFR)), peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucins, lipoproteins, synthetic polypeptides or proteins, small molecules linked proteins, glycoproteins, steroids, nucleic acids, DNA, RNA, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins and cells. In some embodiments, the additional agent is an agent for treating and/or preventing a disease described herein. Each additional agent may be administered at a dose and/or on a schedule determined by the agent. The additional agents may also be administered together with each other and/or with the compounds or compositions described herein, in a single dose or separately in different doses. The particular combination employed in this regimen will take into account the compatibility of the compounds of the present invention with additional agents and/or the desired therapeutic and/or prophylactic effect that will be achieved. Typically, the additional agents are used in combination at a level that is no greater than the level at which they are used individually. In some embodiments, the levels used in combination will be lower than they are used individually.
Such additional agents include, but are not limited to, antiproliferative agents, anticancer agents, antiangiogenic agents, anti-inflammatory agents, immunosuppressive agents, antibacterial agents, antiviral agents, cardiovascular agents, lipid-lowering agents, antidiabetic agents, antiallergic agents, contraceptives, and analgesics. In particular embodiments, the additional agent is an antiproliferative agent. In a specific embodiment, the additional agent is an anti-cancer agent. In particular embodiments, the additional agent is an anti-leukemic agent. In particular embodiments, the additional agent is ABITREXATE (methotrexate), ADE, Adriamycin RDF (doxorubicin HCl), Ambochlorin (chlorambucil), ARRANON (nelarabine), ARZERRA (Oxaliumab), BOSULIF (bosutinib), BUSULFEX (busulfan), CAMPATH (alemtuzumab), CERUBIDINE (daunorubicin HCl), CLAFEN (cyclophosphamide), CLOFAREX (clofarabine), CLOFARAR (clofarabine), CVP, CYTOSAR-U (cytarabine), CYTOXAN (cyclophosphamide), ERWINE (Alopecuroidea), FLUDARA (fludarabine phosphate), FOLEX (methotrexate), FOLEX PFS (methotrexate), GAZYVA (methotrexate), GAZUTYVA (Involumab), GLEEVEC (imatinib mesylate), Hydrab-SIG (CVB), vincristine hydrochloride (Nitrogen mustard), Neosincalix (Nitrogen mustard), Neobrevicine hydrochloride (Nitrogen mustard), Neobrevicine (IV), Neobrevudine hydrochloride (N B), Neobrevicine (N-L), Neobrevicine (N-B), Neoprene (N-L), Neoprene, Neobrene, Neobrevudine hydrochloride (N-L, Neoprene, METHOTREXATE LPF (METHOTREXATE), MEXATE-AQ (METHOTREXATE), mitoxantrone hydrochloride, mustagen (nitrogen mustard hydrochloride), myreran (busulfan), NEOSAR (cyclophosphamide), ONCASPAR (peimepirnase), PURINETHOL (mercaptopurine), PURIXAN (mercaptopurine), Rubidomycin (daunorubicin hydrochloride), SPRYCEL (dasatinib), triribo (cephalotaxine), TARABINE PFS (cytarabine), TASIGNA (nilotinib), trenda (bendamustine hydrochloride), senox (arsenic trioxide), VINCASAR PFS (vincristine sulfate), zydelig (idelalisib), or combinations thereof. In a specific embodiment, the additional agent is an anti-lymphoma drug. In particular embodiments, the additional agent is ABITREXATE (methotrexate), ABVD, ABVE-PC, ADCETRIS (brentuximab vedotin), ADRIAMYCIN PFS (doxorubicin hydrochloride), ADRIAMYCIN RDF (doxorubicin hydrochloride), AMBOCHLORIN (chlorambucil), AMBOCLORIN (chlorambucil), ARRANON (nelarabine), BEACOPP, BECENUM (carmustine), BELEODAQ (belinostat), BEXXAR (tositumomab and I131 tositumomab), BICNU (carmustine), BLENOXANE (bleomycin), CARMUBRIS (carmustine), CHOP, CLAFEN (cyclophosphamide), COPP-ABV, CVP, CYXAN (cyclophosphamide), DECYPOT (cytarabine), DTIC-DOME (EPOCAPIN), FOPHOSPHOLIFA (FOURACH), FOUREA-2, FOURACEAX (FOURACEAX), FORCONITAX-2, FORCONITAX (FOURACAT), FORCONITON (FORCONITON), FORCONITAX-2, FORCONITON (FORCONITX), FORCONITON (FORCAT), BENCA, BENC, Isotaxx (romidepsin), LEUKERAN (chlorambucil), LINFOLIZIN (chlorambucil), lomustine, MATULANE (procarbazine hydrochloride), METHOTREXATE LPF (METHOTREXATE), MEXATE-AQ (METHOTREXATE), MOPP, MOZOBIL (plerixafof), MUSTARGEN (mechlorethamine hydrochloride), NEOSAR (cyclophosphamide), OEPA, ONTAK (dinil 2), OPPA, R-CHOP, revalid (lenalidomide), rituximab (rituximab), STANFORD V, TREANDA (bendamustine hydrochloride), VAMP, VELBAN (vinblastine sulfate), VELCADE (bortezomib), velsa (vinblastine sulfate), VINCASAR PFS (vincristine sulfate), zetimumab), zovovaciz (vinelalin), linidizein (zuelidlyigin), or combinations thereof. In specific embodiments, the additional agent is revalimid (lenalidomide), DACOGEN (decitabine), vidaka (azacitidine), cytar-U (cytarabine), IDAMYCIN (idarubicin), CERUBIDINE (daunorubicin), LEUKERAN (chlorambucil), NEOSAR (cyclophosphamide), FLUDARA (fludarabine), LEUSTATIN (cladribine), or a combination thereof. In particular embodiments, the additional agent is ABITREXATE (methotrexate), ABRAXANE (paclitaxel albumin-stabilized nanoparticulate formulation), AC-T, ADE, ADRIAMYCIN PFS (doxorubicin hydrochloride), ADRUCIL (fluorouracil), AFINITOR (everolimus), AFINITOR DISPERZ (everolimus), ALDARA (imiquimod), ALIMTA (disodium pemetrexed), AREDIA (disodium pamidronate), ARANIDEX (anastrozole), AROMASIN (exemestane), AVASTIN (bevacizumab), BECENUM (carmustine), BECEP, BICNU (carmustine), BLENOXANOX (bleomycin), CAF, CAMPAR (irinotecan hydrochloride), CAPRELA (vandetanib), BOPLATIN-TAXOL, CARBRMUMUIS (carmustine), CASOX (DEMOX), BILUX (CLARITUIX (HPV), HOMORPE-HCL (CINOX), HOMORPE (CINOX), HALCOME (CIT-C), HALCOME (CIE), HALON (CIT-C (CIE), HALOX (CALCOME), HALOX (CALCO-C), HALOX), HALON (CALCO-C (CALCO, COSMEGEN (Actinomycin D), CYFOS (ifosfamide), CYRAMZA (ramucirumab), CYTOSAR-U (cytarabine), CYTOXAN (cyclophosphamide), DACOGEN (decitabine), DEGARELIX, DOXIL (doxorubicin hydrochloride liposome), doxorubicin hydrochloride, DOX-SL (doxorubicin hydrochloride liposome), DTIC-DOME (dacarbazine), EFUDEX (fluorouracil), ELLENCE (epirubicin hydrochloride), ELOXATIN (oxaliplatin), ERBITIX (CETUXIMAB), ERIVEGEL (VIVAGID), ERIVEGE (etoposide phosphate), EVACET (doxorubicin hydrochloride liposome), FARESTON (toremifene), FASLODEX (fulvestrant), FEC, FEMARA (letrozole), FLUOROPELEX (fluorouracil), FOLEX (methotrexate), FOLELFPFS (FOLFIRI), FOBEI-BEFU, FOXIFU, FOIREFI, FOXIDOXIFU-like vaccine (FOIREFI), FOLIX (FOIRIFLOX, FOIRIF, GEMCITABINE-CISPLATIN, GEMCITABINE-OXALLATIN, GEMZAR (GEMCITABINE hydrochloride), GILOTRIF (Afatinib dimaleate), GLEEVEC (imatinib mesylate), GLIADEL (carmustine implant), GLIADEL WAFER (carmustine implant), HERCEPTIN (trastuzumab), HYCAMTIN (topotecan hydrochloride), IFEX (ifosfamide), IFOSFAMIDUM (ifosfamide), INLYTA (acitinib), INTRON A (recombinant interferon alfa-2b), IRESSA (gefitinib), IXEMPRA (ixabepilone), JAFI (ruxolitinib phosphate), JEVTANA (cabazitaxel), KADCYLA (ado-trastuzumab emsine), KEYTRUDA (KYrolizumab), PROLIS (Carbamizumab), EPOLIPY (Liposome), Lipolax (Lipolax), Lipolamin (Dellulupulin hydrochloride), Leuprolin (Pro-4-acetate), Leuprolin (Pro-4-L, LUPRON DEPOT-PED (leuprolide acetate), MEGACE (megestrol), MEKINIST (trimetatinib), METHAZOLASTONE (temozolomide), METHOTREE LPF (METHOTREXATE), MEXATE-AQ (METHOTREXATE), mitoxantrone hydrochloride, MITOTZYTREX (mitomycin c), MOZOBIL (plerixafor), MUSTARGEN (mechlorethamine hydrochloride), MUTAMYCIN (mitomycin c), MYLOSAR (azacitidine), ELBINAVIREBIN (vinorelbine tartrate), NEOSAR (cyclophosphamide), NEXAVAR (sorafenib tosylate), NOLVADEX (tamoxifen citrate), NOVALDEX (tamoxifen citrate), NOVALAPSE (tamoxifen citrate), PAD, PARLATAPIN (carboplatin), cisplatin (carboplatin), PEG-interferon-2 b), pemetrexen (polyethylene glycol), dipotassium citrate), prednisone (prednisone), cisplatin (prednisone), prednisone (prednisone), cisplatin), prednisone (prednisone, cisplatin), cisplatin, prednisone (prednisone, cisplatin, prednisone, and cisplatin, and, PROLIA (dinolizumab), PROVENGE (sipuleucel-t), revalimid (lenalidomide), RUBIDOMYCIN (daunorubicin hydrochloride), SPRYCEL (dasatinib), STIVARGA (regorafenib), SUTENT (sunitinib malate), sylaron (peginterferon alfa-2b), sylvatan (cetuximab), SYNOVIR (thalidomide), TAC, TAFINLAR (dalafenib), TARABINE PFS (cytarabine), TARCEVA (erlotinib hydrochloride), tasiua (nilotinib), TAXOL (paclitaxel), TAXOTERE (docetaxel), TEMODAR (temozolomide), THALOMID (thalidomide), topveveveveveveovar (etoposide), ristol (sirolimus), tresx (trisafen), tykerabenzene (tyxol), vinpocetine (perimidin), vinpocetine (peril), perimidin (peril), vinpocetine), perimidin (vinpocetine), perimidin (valbutrin), perimidin (valacil), perimidin (valbutrin), vinpocetine), tremulin (valacil), tremulin (valbutrin), tremulin (valacil), tremulin (val, Vidazane (azacitidine), VINCASAR PFS (vincristine sulfate), VOTRIENT (pazopanib hydrochloride), WELLCOVORIN (calcium folinate), XALKORI (crizotinib), XELODA (capecitabine), XELOX, XGEVA (dinomab), xolago (radium dichloride 223), xtanid (enzalutamide), yeloy (ipilimumab), ZALTRAP (aflibercept), zelborafenib (william), ZOLADEX (goserelin acetate), ZOMETA (zoledronic acid), ZYKADIA (ceritinib), ZYTIGA (abiraterone acetate), or combinations thereof.
In a specific embodiment, the additional agent is an antiviral agent. In particular embodiments, the additional agent is selected from the group consisting of epigenetic or transcriptional modulators (e.g., DNA methyltransferase inhibitors, histone deacetylase inhibitors (HDAC inhibitors), lysine methyltransferase inhibitors), antimitotic drugs (e.g., taxanes and vinca alkaloids), hormone receptor modulators (e.g., estrogen receptor modulators and androgen receptor modulators), cell signaling pathway inhibitors (e.g., tyrosine kinase inhibitors), modulators of protein stability (e.g., proteasome inhibitors), Hsp90 inhibitors, glucocorticoids, all-trans retinoic acid, and other differentiation promoting drugs. In particular embodiments, the compounds or pharmaceutical compositions of the present invention may be administered in combination with anti-cancer therapies, including, but not limited to, surgery, radiation therapy, transplantation (e.g., stem cell transplantation, bone marrow transplantation), immunotherapy, and chemotherapy.
Examples
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the methods and compounds claimed herein are carried out, prepared, and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what is claimed herein.
The preparation schemes of the compounds of the present invention are shown in, for example, schemes 1 to 4.
Reaction scheme 1
Figure BDA0001553119070000451
The compounds of formula V may be prepared according to the general reaction scheme above. First, 6-bromo-2-methylthio-8H-pyrido [2,3-d ] is prepared according to the reported literature (J.Med.chem.2005,48, 2371-2387)]Pyrimidin-7-one (1). Then converting the bromide (1) into 2-methylthio-7-oxo-7, 8-dihydropyrido [2,3-d ] under the action of cuprous cyanide]Intermediate 2 of pyrimidine-6-carbonitrile. When it is desired to synthesize a compound having no substituent at the 5-position (R)1H), intermediate 2 can also be obtained by direct ring closure using (4-amino-2- (methylthio) pyrimidin-5-yl) carbaldehyde and cyanoacetic acid using the method of reference (j.med.chem.2014,57, 578-599). The cyano group at the 6-position of the intermediate 2 is then further modified with hydroxylamine to give an amidoxime compound 3 which can be cyclized with triethyl orthoformate or the corresponding anhydride to give a compound 4, thus forming a five-membered heterocycle of 1,2, 4-oxadiazole at the 6-position of the mother ring. After the compound 4 has oxidized the methylthio group at the 2-position to methylsulfinyl group with m-chloroperoxybenzoic acid or oxaziridine, this group is substituted under heating with a different amino compound (6) to give the corresponding compound having the structure of formula V.
Reaction formula 2
Figure BDA0001553119070000461
The compounds of formula VI may be prepared according to the general reaction scheme above. Hydrolyzing an intermediate 2 prepared by the same reaction formula as the compound shown in the general formula V by using a sodium hydroxide solution, converting 6-cyano into carboxyl to obtain a compound 7, condensing the carboxyl and different hydrazides to obtain a compound 8 from the compound 7, and then obtaining a compound 9 of which the 6-position forms a 1,3, 4-oxadiazole five-membered heterocyclic ring under the action of phosphorus oxychloride. Oxidizing the compound 9 with m-chloroperoxybenzoic acid to obtain a methanesulfinyl compound 10, and finally substituting with different amino compounds (6) under heating to obtain the corresponding compound with the structure of the general formula VI.
Reaction formula 3
Figure BDA0001553119070000462
The compounds of formula VII may be prepared according to the general reaction scheme above. 6-bromo-2-methylthio-8H-pyrido [2,3-d ] prepared by the literature method (J.Med.chem.2005,48, 2371-2387)]Pyrimidine-7-one (1) with various substitutions (R) under the catalytic condition of cuprous iodide8) Or coupling unsubstituted 2-azetidinone to obtain a compound 11, oxidizing by m-chloroperoxybenzoic acid to obtain a methanesulfinyl compound 12, and finally substituting by different amino compounds (6) under heating to obtain the corresponding compound with the structure of a general formula VII.
Reaction formula 4
Figure BDA0001553119070000471
The compound of formula VIII can be prepared according to the same reaction scheme as the compound of formula VII except that the coupling with bromide 1 is catalyzed by copper powder for various substitutions (R)8) Or the reaction of non-substituted 2-pyrrolidone can obtain a coupling product 13 with the catalytic yield equivalent to that of cuprous iodide, then the coupling product is oxidized by m-chloroperoxybenzoic acid to obtain a methanesulfinyl compound 14, and finally the coupling product is substituted by different amino compounds (6) under the heating condition to obtain the corresponding compound with the structure of the general formula VIII.
Example 1
Figure BDA0001553119070000472
(8-cyclopentyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -N' -hydroxyformamidine (3a)
2a (5g,17.46mmol), hydroxylamine hydrochloride (2.67g,38.41mmol) and sodium bicarbonate (3.23g,38.41mmol) were placed in a reaction flask, t-butanol (60mL) was added, and the reaction was heated to 70 ℃ with stirring overnight. After bringing to room temperature, the reaction was diluted with methanol (80mL) and filtered, the filtrate was concentrated and the resulting crude product was filtered again and washed with solvent (methyl tert-butyl ether/petroleum ether ═ 3:1, ca. 15mL) to give 3a (5.2g, 93.4%) as a yellow solid.
1H NMR(400MHz,DMSO-d6,ppm):9.70(s,1H),8.99(s,1H),8.19(s,1H),5.94(br,2H),5.94–5.82(m,1H),2.59(s,3H),2.31–2.17(m,2H),2.07–1.93(m,2H),1.90–1.77(m,2H),1.71–1.58(m,2H).
8-cyclopentyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (4a)
3a (3.9g,12.21mmol) and acetic anhydride (6.23g,61.05mmol) were dissolved in pyridine (30mL), heated to 120 ℃ with stirring for 3 hours, and then allowed to cool to room temperature naturally and stirred overnight. Most of pyridine was removed by rotary evaporation, and the reaction mixture was diluted with ethyl acetate (about 40mL), washed with saturated aqueous copper sulfate solution 2 times, washed with saturated brine 1 time, and the organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (5: 1 to 1:1 petroleum ether/ethyl acetate) to give 4a (2.1g, 45.8%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):8.71(s,1H),8.44(s,1H),6.12–6.03(m,1H),2.66(s,3H),2.64(s,3H),2.42–2.33(m,2H),2.18–2.09(m,2H),1.95–1.87(m,2H),1.73–1.66(m,2H).
8-cyclopentyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (5a)
4a (2.1g,6.11mmol) was dissolved in dichloromethane (20mL) and then m-chloroperoxybenzoic acid (85%, 0.993g,4.89mmol) was added and stirred at room temperature for 12 hours. Saturated aqueous sodium sulfite (20mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (1: 1 to 0:1 petroleum ether/ethyl acetate) to give 5a (1.49g, 67.6%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):9.05(s,1H),8.59(s,1H),6.16–6.07(m,1H),3.02(s,3H),2.70(s,3H),2.37–2.28(m,2H),2.22–2.14(m,2H),2.02–1.93(m,2H),1.76–1.67(m,2H).
8-cyclopentyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one (V-1)
5a (100mg,0.278mmol) and 6a (107mg,0.556mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon 3 times, toluene (3mL) was added, and the mixture was heated to 100 ℃ under stirring with an argon balloon overnight for reaction. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered and washed with ethyl acetate to give V-1(40mg, 29.5%) as a yellow solid.
1H NMR(400MHz,CDCl3,ppm):9.15(br,1H),8.78(s,1H),8.41(s,1H),8.19(d,J=9.2Hz,1H),8.13(d,J=2.8Hz,1H),7.34(dd,J=9.0,3.0Hz,1H),6.05–5.96(m,1H),3.24(t,J=5.0Hz,4H),2.66(s,3H),2.61(t,J=4.8Hz,4H),2.49–2.34(m,2H),2.37(s,3H),2.20–2.10(m,2H),1.95–1.87(m,2H),1.74–1.64(m,2H).
HRMS (ESI) for C25H29N9O2[M+H]+488.2517 for the calculated value m/z; measured value m/z is 488.2515.
Table 1 examples 2-7
Figure BDA0001553119070000491
Figure BDA0001553119070000501
Example 2
Figure BDA0001553119070000502
4- (6- (8-cyclopentyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-2a)
5a (200mg,0.556mmol) and 6b (170.4mg,0.612mmol) were placed in a dry 15mL reaction tube, sealed with a rubber stopper and replaced with argon for 3 times, toluene (5mL) was added, and the mixture was heated to 100 ℃ under stirring with an argon balloon for 6 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered, and washed with ethyl acetate to give a yellow solid, i.e., V-2a (102mg, 32%).
1H NMR(400MHz,CDCl3,ppm):9.15(br,1H),8.79(s,1H),8.41(s,1H),8.21(d,J=9.2Hz,1H),8.13(d,J=2.8Hz,1H),7.34(dd,J=9.2,2.8Hz,1H),6.05–5.96(m,1H),3.62(t,J=5.0Hz,4H),3.15(t,J=5.0Hz,4H),2.66(s,3H),2.47–2.39(m,2H),2.20–2.11(m,2H),1.95–1.87(m,2H),1.74–1.65(m,2H),1.49(s,9H).
8-cyclopentyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (V-2)
Placing V-2a (90mg,0.157mmol) in a reaction bottle, adding 1, 4-dioxane solution of hydrogen chloride (4N,4mL) and methanol (4mL), stirring at room temperature for 4 hours, removing the solvent by rotary evaporation under reduced pressure, and washing the obtained crude product with ethyl acetate and ether sequentially to obtain yellow powder, namely V-2(76mg, 95%).
1H NMR(400MHz,DMSO-d6,ppm):11.41(br,1H),9.57(br,2H),9.13(s,1H),8.71(s,1H),8.07(s,1H),8.01(d,J=9.2Hz,1H),7.84(d,J=9.2Hz,1H),5.95–5.87(m,1H),3.51–3.43(m,4H),3.29–3.20(m,4H),2.66(s,3H),2.30–2.21(m,2H),2.05–1.95(m,2H),1.87–1.79(m,2H),1.66–1.57(m,2H).
HRMS (ESI) for C24H27N9O2[M+H]+474.2360 for the calculated value m/z; measured value m/z is 474.2359.
Example 3
Referring to the method for the synthesis of V-2a in example 2, compound V-3a (36.9mg, 27%) was obtained by reacting compound 5a (80mg,0.223mmol) with 6c (74.4mg,0.234 mmol).
HRMS (ESI) for C32H39N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3206.
With reference to the method for synthesizing V-2 in example 2, the title compound V-3(28mg, 96%) was prepared from compound V-3a (32.5mg,0.053 mmol).
HRMS (ESI) for C27H31N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2679.
Example 4
Referring to the method for the synthesis of V-2a in example 2, compound 5a (63.4mg,0.176mmol) and 6d (59mg,0.185mmol) were reacted to give compound V-4a (26mg, 24%).
HRMS (ESI) for C32H39N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3203.
With reference to the method for synthesizing V-2 in example 2, the title compound V-4(19mg, 97%) was prepared from compound V-4a (21.9mg,0.036 mmol).
HRMS (ESI) for C27H31N9O2[M+H]+514.2674 for the calculated value m/z; measured value m/z is 514.2677.
Example 5
Referring to the method for synthesizing V-1 in example 1, compound 5a (90mg,0.25mmol) and 6e (58.2mg,0.263mmol) were reacted to give the title compound V-5(29.2mg, 22.6%).
HRMS (ESI) for C27H34N9O2[M+H]+516.2830 for the calculated value m/z; measured value m/z is 516.2837.
Example 6
Referring to the method for synthesizing V-1 in example 1, compound 5a (90mg,0.25mmol) and 6f (55.3mg,0.263mmol) were reacted to give the title compound V-6(36mg, 28.5%).
HRMS (ESI) for C26H31N7O4[M+H]+506.2510 for the calculated value m/z; measured value m/z is 506.2514.
Example 7
Referring to the method for synthesizing V-2a in example 2, compound V-7a (45.2mg, 30.7%) was obtained by reacting compound 5a (90mg,0.25mmol) with 6g (76.9mg,0.263 mmol).
HRMS (ESI) for C30H37N9O4[M+H]+588.3041 for the calculated value m/z; measured value m/z is 588.3047.
With reference to the method for synthesizing V-2 in example 2, the title compound V-7(35mg, 98%) was prepared from compound V-7a (40mg,0.068 mmol).
HRMS (ESI) for C25H29N9O2[M+H]+488.2517 for the calculated value m/z; measured value m/z is 488.2523.
Example 8
Figure BDA0001553119070000531
8-cyclopentyl-2-methylsulfanyl-6- (1,2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (4b)
3a (2g,6.26mmol) was placed in a reaction flask, triethyl orthoformate (30mL) was added, and the mixture was heated to 130 ℃ with stirring for 12 hours. The excess triethyl orthoformate was removed by rotary evaporation under reduced pressure and the crude product obtained was purified by column chromatography (3: 1 to 1:1 petroleum ether/ethyl acetate) to give 4b (1g, 44.2%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):8.79(s,1H),8.74(s,1H),8.53(s,1H),6.14–6.05(m,1H),2.65(s,3H),2.43–2.34(m,2H),2.20–2.10(m,2H),1.97–1.88(m,2H),1.75–1.65(m,2H).
8-cyclopentyl-2-methanesulfinyl-6- (1,2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (5b)
4b (1g,3.04mmol) was dissolved in dichloromethane (20mL) and then m-chloroperoxybenzoic acid (85%, 0.506g,2.49mmol) was added and stirred at room temperature for 3 hours. Saturated aqueous sodium sulfite (20mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (petroleum ether/ethyl acetate 2:1 to 0:1) to give 5b (0.73g, 69.7%) as a pale yellow solid.
1H NMR(400MHz,CDCl3,ppm):9.09(s,1H),8.84(s,1H),8.67(s,1H),6.18–6.09(m,1H),3.03(s,3H),2.37–2.29(m,2H),2.24–2.14(m,2H),2.04–1.94(m,2H),1.77–1.67(m,2H).
8-cyclopentyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (1,2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (V-8)
With reference to the synthesis of V-1 in example 1, the title compound V-8(88mg, 32%) was prepared from 5b (200mg,0.579mmol) and 6a (122.5mg,0.637 mmol).
1H NMR(400MHz,CDCl3,ppm):8.78(s,1H),8.76(s,1H),8.49(s,1H),8.19(d,J=9.2Hz,1H),8.11(d,J=2.8Hz,1H),7.35(dd,J=9.2,2.8Hz,1H),6.06–5.97(m,1H),3.24(t,J=5.0Hz,4H),2.62(t,J=5.0Hz,4H),2.47–2.37(m,2H),2.38(s,3H),2.21–2.11(m,2H),1.97–1.88(m,2H),1.75–1.69(m,2H).
HRMS (ESI) for C24H27N9O2[M+H]+474.2360 for the calculated value m/z; measured value m/z is 474.2368.
TABLE 2 examples 9-14
Figure BDA0001553119070000541
Example 9
Referring to the procedure for the synthesis of V-2a in example 2, compound V-9a (55.7mg, 17.2%) was obtained by reacting compound 5b (200mg,0.579mmol) and 6c (177.3mg,0.637 mmol).
1H NMR(400MHz,CDCl3,ppm):8.77(s,1H),8.76(s,1H),8.61(br,1H),8.50(s,1H),8.21(d,J=8.8Hz,1H),8.09(d,J=2.8Hz,1H),7.35(dd,J=9.2,3.2Hz,1H),6.06–5.97(m,1H),3.62(t,J=5.0Hz,4H),3.15(t,J=5.0Hz,4H),2.48–2.38(m,2H),2.22–2.12(m,2H),1.97–1.89(m,2H),1.75–1.67(m,2H),1.49(s,9H).
Referring to the method for synthesizing V-2 in example 2, the title compound V-9(49mg, 99%)
1H NMR(400MHz,DMSO-d6,ppm):11.24(br,1H),9.72(s,1H),9.48(br,2H),9.13(s,1H),8.74(s,1H),8.08(s,1H),7.94(d,J=8.4Hz,1H),7.85(d,J=9.2Hz,1H),5.96–5.87(m,1H),3.48–3.45(m,4H),3.29–3.21(m,4H),2.33–2.22(m,2H),2.03–1.94(m,2H),1.87–1.80(m,2H),1.67–1.58(m,2H).
HRMS (ESI) for C23H25N9O2[M+H]+460.2204 for the calculated value m/z; measured value m/z is 460.2207.
Example 10
Referring to the method for the synthesis of V-2a in example 2, compound 5b (100mg,0.29mmol) and 6c (96.8mg,0.3mmol) were reacted to give compound V-10a (50.4mg, 29%).
HRMS (ESI) for C31H37N9O4[M+H]+600.3041 for the calculated value m/z; measured value m/z is 600.3048.
Referring to the synthesis of V-2 in example 2, the title compound V-10(39.8mg, 99%) was prepared from compound V-10a (45mg,0.075 mmol).
HRMS (ESI) for C26H29N9O2[M+H]+500.2517 for the calculated value m/z; measured value m/z is 500.2510.
Example 11
Referring to the method for the synthesis of V-2a in example 2, compound 5b (100mg,0.29mmol) and 6d (96.8mg,0.3mmol) were reacted to give compound V-11a (48.7mg, 28%).
HRMS (ESI) for C31H37N9O4[M+H]+600.3041 for the calculated value m/z; measured value m/z is 600.3037.
Referring to the synthesis of V-2 in example 2, the title compound V-11(38mg, 99%) was prepared from compound V-11a (43mg,0.072 mmol).
HRMS (ESI) for C26H29N9O2[M+H]+500.2517 for the calculated value m/z; measured value m/z is 500.2505.
Example 12
Referring to the method for synthesizing V-1 in example 1, compound 5b (100mg,0.29mmol) and 6e (67.3mg,0.3mmol) were reacted to give the title compound V-12(33.5mg, 23%).
HRMS (ESI) for C26H32N9O2[M+H]+502.2673 for the calculated value m/z; measured value m/z is 502.2684.
Example 13
Referring to the method for synthesizing V-1 in example 1, compound 5b (100mg,0.29mmol) and 6f (63.9mg,0.3mmol) were reacted to give the title compound V-13(41.8mg, 29.3%).
HRMS (ESI) for C25H29N7O4[M+H]+492.2354 for the calculated value m/z; measured value m/z is 492.2356.
Example 14
Referring to the method for synthesizing V-2a in example 2, compound V-14a (47.7mg, 28.7%) was obtained by reacting compound 5b (100mg,0.29mmol) with 6g (88.9mg,0.3 mmol).
HRMS (ESI) for C29H35N9O4[M+H]+574.2885 for the calculated value m/z; measured value m/z is 574.2879.
Referring to the method for synthesizing V-2 in example 2, the title compound V-14(37mg, 99%) was prepared from compound V-14a (42mg,0.073 mmol).
HRMS (ESI) for C24H27N9O2[M+H]+474.2360 for the calculated value m/z; measured value m/z is 474.2366.
Example 15
Figure BDA0001553119070000571
8-cyclopentyl-N' - ((cyclopropanecarbonyl) oxy) -2- (methylthio) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxamidine (3ac)
Cyclopropanecarboxylic acid (568mg,6.6mmol) was dissolved in dichloromethane (25mL) under argon and 1-hydroxybenzotriazole (892mg,6.6mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (1.27g,6.6mmol) were added. The resulting solution was stirred at room temperature for 20 minutes, then crude 3c (1.89g,6mmol) was added and stirring continued for 2 hours. The reaction solution was quenched with saturated aqueous sodium bicarbonate solution and then extracted with dichloromethane. After the organic phases are combined, the organic phases are washed by sodium hydroxide aqueous solution (1N) and saturated saline solution in sequence, dried by anhydrous sodium sulfate, filtered and dried in a spinning way to obtain yellow foamy solid, namely a crude product of 3ac, which can be directly used for the next reaction.
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (4c)
Under the protection of argon, crude 3ac (6mmol) was dissolved in a mixed solvent of ethanol and water (4:1,30mL), sodium acetate trihydrate (2.04g,15mmol) was added, and the resulting reaction solution was sealed and heated to 90 ℃ for reaction for 14 hours. After the reaction solution was cooled, it was diluted with water and extracted with dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and subjected to column chromatography (petroleum ether: dichloromethane: acetone: 7:1:1) to give 4c as a yellow foamy solid (1.01g, 45.3% overall yield in three steps).
HRMS (ESI) for C18H20N5O2S[M+H]+370.1332 for the calculated value m/z; measured value m/z is 370.1323.
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (5c)
After 4c (922mg,2.5mmol) was dissolved in chloroform (30mL), 2-benzenesulfonyl-3-phenylazepino propane (718mg,2.75mmol) was added, and the mixture was stirred at room temperature for 15 hours. After evaporation of the solvent to dryness, the crude product was purified by column chromatography (dichloromethane: methanol 50:1) to yield 5c (876mg, 91%) as a yellow solid.
HRMS (ESI) for C18H20N5O3S[M+H]+386.1281 for the calculated value m/z; measured value m/z is 386.1293.
(6- ((8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-15a)
5c (193mg,0.5mmol) and 6b (139mg,0.5mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon for 3 times, and then benzotrifluoride (1mL) was added and heated to 80 ℃ under stirring with an argon balloon for 16 hours. After cooling to room temperature, the solvent was evaporated under reduced pressure, and the obtained reaction residue was subjected to column chromatography (petroleum ether: dichloromethane: acetone ═ 5:2:1) to obtain a crude product, which was then washed with a mixed solvent of methyl tert-butyl ether and ethyl acetate (4:3) to obtain a yellow solid, i.e., V-15a (72.2mg, 24.1%).
HRMS (ESI) for C31H38N9O4[M+H]+600.3041 for the calculated value m/z; measured value m/z is 600.3023.
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one hydrochloride (V-15)
With reference to the synthesis of V-2 in example 2, the title compound V-15(26mg, 97%) was prepared from V-15a (30mg, 50. mu. mol).
HRMS (ESI) for C26H30N9O2[M+H]+500.2517 for the calculated value m/z; measured value m/z is 500.2539.
Table 3 examples 16 to 19
Figure BDA0001553119070000591
Example 16
Referring to the method for synthesizing V-1 in example 1, compound 5c (115.5mg,0.3mmol) and 6h (53.7mg,0.3mmol) were reacted to give the title compound V-16(40.5mg, 27%).
HRMS (ESI) for C26H29N8O3[M+H]+501.2357 for the calculated value m/z; measured value m/z is 501.2359.
Example 17
Referring to the method for synthesizing V-1 in example 1, compound 5c (115.5mg,0.3mmol) and 6a (57.6mg,0.3mmol) were reacted to give the title compound V-17(47.7mg, 31%).
HRMS (ESI) for C27H32N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2689.
Example 18
Referring to the method for synthesizing V-2a in example 2, compound V-18a (31.3mg, 21%) was obtained by reacting compound 5c (115.5mg,0.3mmol) with 6i (88mg,0.3 mmol).
HRMS (ESI) for C32H40N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3203.
The title compound V-18(31.3mg, 99%) was prepared from compound V-18a (35.0mg, 57. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C27H32N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2688.
Example 19
Referring to the method for synthesizing V-2a in example 2, compound V-19a (60.7mg, 33%) was obtained by reacting compound 5c (115.5mg,0.3mmol) with 6g (88mg,0.3 mmol).
HRMS (ESI) for C32H40N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3183.
The title compound V-19(35.7mg, 99%) was prepared from compound V-19a (40mg, 65. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C27H32N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2679.
Example 20
Figure BDA0001553119070000611
8-cyclopentyl-5-methyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carbonitrile (2b)
After sealing 10mL under argon atmosphere, 1b (100mg,0.28mmol), cuprous cyanide (112mg,1.25mmol) and 1-methyl-2-pyrrolidone (1mL) were added, and the mixture was heated and stirred at 150 ℃ for 3 hours. After cooling to room temperature, the tube was opened, the reaction solution was diluted with ethyl acetate and washed with saturated brine, and the organic phase was dried over anhydrous sodium sulfate and then filtered. The filtrate was concentrated and column chromatographed to give 2b (29mg, 34.2%) as an off-white solid.
1H NMR(400MHz,CDCl3,ppm):8.81(s,1H),5.98–5.89(m,1H),2.72(s,3H),2.63(s,3H),2.31–2.23(m,2H),2.12–2.05(m,2H),1.92–1.84(m,2H),1.73–1.66(m,2H).
(8-cyclopentyl-5-methyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -N' -hydroxyformamidine (3b)
2b (750mg,2.5mmol), hydroxylamine hydrochloride (433mg,6.24mmol) and sodium bicarbonate (524mg,6.24mmol) were placed in a reaction flask, tert-butanol (30mL) was added and the reaction was heated to 70 ℃ with stirring overnight. After bringing to room temperature, the reaction was diluted with methanol (10mL) and filtered, the filtrate was concentrated and the resulting crude product was filtered again and washed with solvent (methyl tert-butyl ether/petroleum ether ═ 3:1, ca. 5mL) to give 3b (378mg, 45.4%) as a yellow solid.
1H NMR(400MHz,DMSO-d6,ppm):8.79(s,1H),6.00–5.91(m,1H),4.99(br,2H),2.62(s,3H),2.52(s,3H),2.35–2.25(m,2H),2.09–2.01(m,2H),1.90–1.82(m,2H),1.71–1.61(m,2H).
8-cyclopentyl-5-methyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (4d)
3b (378mg,1.13mmol) and acetic anhydride (289mg,2.83mmol) were dissolved in pyridine (3mL) and heated to 120 ℃ with stirring for 2 hours. Most of pyridine was removed by rotary evaporation, and the reaction mixture was diluted with ethyl acetate (about 30mL), washed with saturated aqueous copper sulfate solution 2 times, washed with saturated brine 1 time, and the organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (petroleum ether/ethyl acetate 10:1) to give 4d (243mg, 60%) as a yellow oil.
1H NMR(400MHz,CDCl3,ppm):8.82(s,1H),6.05–5.96(m,1H),2.70(s,3H),2.64(s,3H),2.40(s,3H),2.37–2.28(m,2H),2.11–2.01(m,2H),1.92–1.84(m,2H),1.71–1.62(m,2H).
8-cyclopentyl-5-methyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (5d)
After 4d (247mg,0.69mmol) was dissolved in chloroform (2mL), 2-benzenesulfonyl-3-phenyloxaziridine (239.7mg,0.91mmol) was added, and the mixture was stirred at room temperature for 3 hours. After evaporation of the solvent to dryness, the crude product was purified by column chromatography (petroleum ether/ethyl acetate 1:1 to 0:1) to give 5d (170mg, 65.9%) as a yellow oil.
1H NMR(400MHz,CDCl3,ppm):9.14(s,1H),6.06–5.97(m,1H),3.00(s,3H),2.71(s,3H),2.49(s,3H),2.29–2.21(m,2H),2.13–2.05(m,2H),1.98–1.88(m,2H),1.71–1.63(m,2H).
4- (6- (8-cyclopentyl-5-methyl-6- (5-methyl-1, 2, 4-oxadiazol-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-20a)
Referring to the synthesis of V-15a in example 15, V-20a (23.9mg, 14.7%) was prepared from 5d (103mg,0.276mmol) and 6b (80.6mg,0.29 mmol).
HRMS (ESI) for C30H37N9O4[M+H]+588.3041 for the calculated value m/z; measured value m/z is 588.3037.
With reference to the synthesis of V-15 in example 15, the title compound V-20(21mg, 99%) was prepared from V-20a (23.9mg,0.04 mmol).
1H NMR(400MHz,D2O,ppm):9.07(s,1H),8.06(d,J=7.6Hz,1H),7.86(s,1H),7.51(d,J=9.6Hz,1H),5.88–5.79(m,1H),3.54–3.52(m,4H),3.46–3.44(m,4H),2.72(s,3H),2.41(s,3H),2.14–2.07(m,2H),2.02–1.89(m,4H),1.71–1.65(m,2H).
TABLE 4 examples 21 to 25
Figure BDA0001553119070000631
Example 21
Referring to the method for synthesizing V-15a in example 15, compound V-21a (31.4mg, 18.7%) was obtained by reacting compound 5d (100mg,0.268mmol) and 6c (89.5mg,0.281 mmol).
HRMS (ESI) for C33H41N9O4[M+H]+628.3354 for the calculated value m/z; measured value m/z is 628.3361.
Referring to the method for synthesizing V-2 in example 2, the title compound V-21(23.6mg, 99%) was prepared from compound V-21a (26.5mg,0.042 mmol).
HRMS (ESI) for C28H33N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2833.
Example 22
Referring to the method for synthesizing V-15a in example 15, compound V-22a (28.9mg, 17.2%) was obtained by reacting compound 5d (100mg,0.268mmol) and 6d (89.5mg,0.281 mmol).
HRMS (ESI) for C33H41N9O4[M+H]+628.3354 for the calculated value m/z; measured value m/z is 628.3357.
Referring to the method for synthesizing V-2 in example 2, the title compound V-22(22mg, 98%) was prepared from compound V-22a (25mg,0.04 mmol).
HRMS (ESI) for C28H33N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2826.
Example 23
Referring to the method for synthesizing V-1 in example 1, compound 5d (100mg,0.268mmol) and 6e (62.2mg,0.281mmol) were reacted to give the title compound V-23(22.5mg, 15.8%).
HRMS (ESI) for C28H36N9O2[M+H]+530.2986 for the calculated value m/z; measured value m/z is 530.3001.
Example 24
Referring to the method for synthesizing V-1 in example 1, compound 5d (100mg,0.268mmol) and 6f (59.1mg,0.281mmol) were reacted to give the title compound V-24(19.3mg, 13.9%).
HRMS (ESI) for C27H33N7O4[M+H]+520.2667 for the calculated value m/z; measured value m/z is 520.2678.
Example 25
Referring to the method for synthesizing V-15a in example 15, compound V-25a (21.9mg, 13.6%) was obtained by reacting compound 5d (100mg,0.268mmol) with 6g (82.2mg,0.281 mmol).
Referring to the method for synthesizing V-2 in example 2, the title compound V-25(15.8mg, 98%) was prepared from compound V-25a (18mg,0.03 mmol).
HRMS (ESI) for C26H31N9O2[M+H]+502.2673 for the calculated value m/z; measured value m/z is 502.2670.
Example 26
Figure BDA0001553119070000651
8-cyclopentyl-5-methyl-2-methylsulfanyl-6- (1,2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (4e)
3b (300mg,0.9mmol) was placed in a reaction flask, triethyl orthoformate (3mL) was added, and the mixture was heated to 130 ℃ with stirring for reaction for 3 hours. The excess triethyl orthoformate was removed by rotary evaporation under reduced pressure and the crude product was purified by column chromatography to give 4e (110mg, 35.6%) as a yellow oil.
1H NMR(400MHz,CDCl3,ppm):8.91(s,1H),8.84(s,1H),6.06–5.97(m,1H),2.65(s,3H),2.40(s,3H),2.36–2.28(m,2H),2.11–2.01(m,2H),1.93–1.85(m,2H),1.71–1.64(m,2H).
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (1,2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (5e)
4e (110mg,0.32mmol) was dissolved in dichloromethane (2mL), cooled to 0 ℃ in an ice water bath, and then m-chloroperoxybenzoic acid (44.2mg,0.256mmol) was added in portions and stirred at room temperature for 3 hours. Saturated aqueous sodium sulfite (2mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (1: 1 to 0:1 petroleum ether/ethyl acetate) to give 5e (80mg, 69.5%) as a yellow solid.
1H NMR(400MHz,CDCl3,ppm):9.16(s,1H),8.95(s,1H),6.08–6.00(m,1H),3.02(s,3H),2.50(s,3H),2.30–2.21(m,2H),2.15–2.06(m,2H),2.00–1.90(m,2H),1.75–1.65(m,2H).
4- (6- (8-cyclopentyl-5-methyl-6- (1,2, 4-oxadiazol-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-26a)
Referring to the procedure for the synthesis of V-15a in example 15, compound V-26a (17.7mg, 12.3%) was reacted from compound 5e (90mg,0.25mmol) and 6b (73.2mg,0.263 mmol).
1H NMR(400MHz,CDCl3,ppm):8.90(s,1H),8.87(s,1H),8.46(br,1H),8.21(d,J=9.2Hz,1H),8.06(d,J=3.2Hz,1H),7.35(dd,J=9.2,2.8Hz,1H),6.00–5.91(m,1H),3.62(t,J=5.0Hz,4H),3.14(t,J=5.0Hz,4H),2.39(s,3H),2.39–2.31(m,2H),2.10–2.02(m,2H),1.93–1.85(m,2H),1.70–1.63(m,2H),1.49(s,9H).
8-cyclopentyl-5-methyl-6- (1,2, 4-oxadiazol-3-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (V-26)
Referring to the method for synthesizing V-2 in example 2, the title compound V-26(15mg, 98%) was prepared from compound V-26a (17.2mg,0.03 mmol).
1H NMR(400MHz,D2O,ppm):9.44(s,1H),9.05(s,1H),8.02(d,J=4.0Hz,1H),7.85(s,1H),7.49(d,J=6.4Hz,1H),5.83–5.79(m,1H),3.53–3.52(m,4H),3.46–3.45(m,4H),2.38(s,3H),2.11–2.07(m,2H),1.99–1.91(m,4H),1.70–1.66(m,2H).
TABLE 5 examples 27 to 31
Figure BDA0001553119070000661
Figure BDA0001553119070000671
Example 27
Referring to the method for synthesizing V-15a in example 15, compound V-27a (18.1mg, 11.8%) was obtained by reacting compound 5e (90mg,0.25mmol) with 6c (83.7mg,0.263 mmol).
HRMS (ESI) for C32H39N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3203.
Referring to the synthesis of V-2 in example 2, the title compound V-27(13.2mg, 98%) was prepared from compound V-27a (15mg,0.024 mmol).
HRMS (ESI) for C27H31N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2667.
Example 28
Referring to the procedure for the synthesis of V-15a in example 15, compound 5e (90mg,0.25mmol) and 6d (83.7mg,0.263mmol) were reacted to give compound V-28a (19.6mg, 12.8%).
HRMS (ESI) for C32H39N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3194.
Referring to the synthesis of V-2 in example 2, the title compound V-28(13mg, 97%) was prepared from compound V-28a (15mg,0.024 mmol).
HRMS (ESI) for C27H31N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2677.
Example 29
Referring to the method for synthesizing V-1 in example 1, compound 5e (90mg,0.25mmol) and 6e (58.2mg,0.263mmol) were reacted to give the title compound V-29(17.2mg, 13.3%).
HRMS (ESI) for C27H34N9O2[M+H]+516.2830 for the calculated value m/z; measured value m/z is 516.2844.
Example 30
Referring to the method for synthesizing V-1 in example 1, compound 5e (90mg,0.25mmol) and 6f (55.3mg,0.263mmol) were reacted to give the title compound V-30(22.4mg, 17.7%).
HRMS (ESI) for C26H31N7O4[M+H]+506.2510 for the calculated value m/z; measured value m/z is 506.2512.
Example 31
Referring to the method for synthesizing V-15a in example 15, compound V-31a (15.6mg, 10.6%) was obtained by reacting compound 5e (90mg,0.25mmol) with 6g (76.9mg,0.263 mmol).
HRMS (ESI) for C30H37N9O4[M+H]+588.3041 for the calculated value m/z; measured value m/z is 588.3038.
With reference to the method for synthesizing V-2 in example 2, the title compound V-31(11.9mg, 99%) was prepared from compound V-31a (13.5mg,0.023 mmol).
HRMS (ESI) for C25H29N9O2[M+H]+488.2517 for the calculated value m/z; measured value m/z is 488.2520.
Example 32
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one hydrochloride (V-32)
Figure BDA0001553119070000691
8-cyclopentyl-N' - ((cyclopropanecarbonyl) oxy) -5-methyl-2- (methylthio) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxamidine (3bf)
Cyclopropanecarboxylic acid (1.33g,15.5mmol) was dissolved in dichloromethane (150mL) under argon and 1-hydroxybenzotriazole (2.09g,15.5mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (2.97g,15.5mmol) were added. The resulting solution was stirred at room temperature for 20 minutes, then crude 3b (14.1mmol) was added and stirring continued for 2 hours. The reaction solution was quenched with saturated aqueous sodium bicarbonate solution and then extracted with dichloromethane. The organic phases are combined and washed by sodium hydroxide aqueous solution (1N) and saturated saline solution in sequence, dried by anhydrous sodium sulfate, filtered and dried in a spinning way to obtain yellow foamy solid, namely a crude product of 3bf, which can be directly used for the next reaction.
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (4f)
Under the protection of argon, crude 3bf (15mmol) is dissolved in a mixed solvent of ethanol and water (4:1,75mL), sodium acetate trihydrate (4.80g,35.3mmol) is added, and the obtained reaction solution is heated to 90 ℃ for reaction for 14 hours after being sealed. After the reaction solution was cooled, it was diluted with water and extracted with dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and subjected to column chromatography (petroleum ether: dichloromethane: acetone: 7:1:1) to give 4f (2.47g, 45.7% overall yield in three steps) as a yellow foamy solid.
1H NMR(400MHz,CDCl3,ppm):8.81(s,1H),6.03–5.94(m,1H),2.64(s,3H),2.37(s,3H),2.35–2.27(m,3H),2.09–2.01(m,2H),1.91–1.83(m,2H),1.71–1.64(m,2H),1.34–1.32(m,2H),1.30–1.27(m,2H).
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (5f)
After 4f (2.43g,6.33mmol) was dissolved in chloroform (110mL), 2-benzenesulfonyl-3-phenyloxaziridine (776.9mg,2.973mmol) was added and the mixture was stirred at room temperature for 15 hours. The solvent was evaporated to dryness and the crude product was purified by column chromatography (dichloromethane: methanol ═ 50:1) to yield 5f (2.5g, 99%) as a yellow solid.
1H NMR(400MHz,CDCl3,ppm):9.12(s,1H),6.05–5.96(m,1H),3.01(s,3H),2.46(s,3H),2.38–2.23(m,3H),2.13–2.07(m,2H),1.98–1.88(m,2H),1.71–1.65(m,2H),1.37–1.27(m,4H).
(6- ((8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-7-oxo-7, 8-dihydropyrido [2,3-3-d ] pyrimidin-2-yl) amino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-32a)
Referring to the synthesis of V-2a in example 2, compound V-32a (18.4mg, 20.8%) was prepared from 5f (57.5mg,0.144mmol) and 6b (42mg,0.151 mmol).
1H NMR(400MHz,CDCl3,ppm):8.84(s,1H),8.38(br,1H),8.20(d,J=9.2Hz,1H),8.06(d,J=2.4Hz,1H),7.34(dd,J=9.2,2.8Hz,1H),5.97–5.88(m,1H),3.62(t,J=4.8Hz,4H),3.13(t,J=4.4Hz,4H),2.36(s,3H),2.33–2.27(m,3H),2.07–2.02(m,2H),1.90–1.85(m,2H),1.72–1.66(m,2H),1.49(s,9H),1.34–1.31(m,2H),1.29–1.27(m,2H).
8-cyclopentyl-6- (5-cyclopropyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one hydrochloride (V-32)
With reference to the synthesis of V-2 in example 2, the title compound V-32(16mg, 97%) was prepared from V-32a (18.4mg,0.03 mmol).
1H NMR(400MHz,D2O,ppm):9.07(s,1H),8.06(d,J=9.2Hz,1H),7.87(s,1H),7.52(d,J=9.2Hz,1H),5.87–5.78(m,1H),3.55–3.47(m,8H),2.45–2.39(m,1H),2.41(s,3H),2.15–2.08(m,2H),2.01–1.90(m,4H),1.73–1.66(m,2H),1.43–1.38(m,2H),1.31–1.27(m,2H).
TABLE 6 examples 34 to 36
Figure BDA0001553119070000711
Figure BDA0001553119070000721
Figure BDA0001553119070000731
Figure BDA0001553119070000741
Example 33
Referring to the method for synthesizing V-1 in example 1, the title compound V-33(20.5mg, 26%) was obtained by reacting compound 5f (60mg,0.15mmol) and 6a (29mg,0.15 mmol).
HRMS (ESI) for C28H34N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2833.
Example 34
Referring to the method for the synthesis of V-2a in example 2, compound V-34a (26.5mg, 27%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6c (47.7mg,0.15 mmol).
HRMS (ESI) for C35H44N9O4[M+H]+654.3511 for the calculated value m/z; measured value m/z is 654.3519.
The title compound V-34(18mg, 99%) was prepared from compound V-34a (20mg, 30.6. mu. mol) by following the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C30H36N9O2[M+H]+554.2986 for the calculated value m/z; measured value m/z is 554.2991.
Example 35
Referring to the method for the synthesis of V-2a in example 2, compound V-35a (20.5mg, 21%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6d (47.7mg,0.15 mmol).
HRMS (ESI) for C35H44N9O4[M+H]+654.3511 for the calculated value m/z; measured value m/z is 654.3519.
The title compound V-35(13.5mg, 99%) was prepared from compound V-35a (15mg,23. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C30H36N9O2[M+H]+554.2986 for the calculated value m/z; measured value m/z is 554.2977.
Example 36
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6e (33mg,0.15mmol) were reacted to give the title compound V-36(15mg, 18%).
HRMS (ESI) for C30H38N9O2[M+H]+556.3143 for the calculated value m/z; measured value m/z is 556.3153.
Example 37
Referring to the method for synthesizing V-1 in example 1, the title compound V-37(9mg, 11%) was obtained by reacting compound 5f (60mg,0.15mmol) and 6f (31.5mg,0.15 mmol).
HRMS (ESI) for C29H36N7O4[M+H]+546.2823 for the calculated value m/z; measured value m/z is 546.2836.
Example 38
Referring to the method for the synthesis of V-2a in example 2, compound V-38a (27.3mg, 29%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6i (44mg,0.15 mmol).
HRMS (ESI), forIn C33H41N9O4[M]+627.3276 for the calculated value m/z; measured value m/z is 627.3271.
The title compound V-38(21.3mg, 95%) was prepared from compound V-38a (25mg, 40.0. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C28H34N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2839.
Example 39
Referring to the method for synthesizing V-2a in example 2, compound V-39a (21.6mg, 23%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6g (44mg,0.15 mmol).
HRMS (ESI) for C33H41N9O4[M]+627.3276 for the calculated value m/z; measured value m/z is 627.3276.
The title compound V-39(16.1mg, 93%) was prepared from compound V-39a (18mg, 28.7. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C28H34N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2841.
Example 40
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6h (26.9mg,0.15mmol) were reacted to give the title compound V-40(23.9mg, 31%).
HRMS (ESI) for C27H31N8O3[M+H]+515.2514 for the calculated value m/z; measured value m/z is 515.2519.
EXAMPLE 41
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6j (30.9mg,0.15mmol) were reacted to give the title compound V-41(28.4mg, 35%).
HRMS (ESI) for C29H36N9O2[M+H]+542.2986 for the calculated value m/z; measured value m/z is 542.3001.
Example 42
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6k (33.2mg,0.15mmol) were reacted to give the title compound V-42(19.2mg, 23%).
HRMS (ESI) for C30H36N8O3[M]+557.2905 for the calculated value m/z; measured value m/z is 556.2906.
Example 43
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6l (35.3mg,0.15mmol) were reacted to give the title compound V-43(7.7mg, 9%).
HRMS (ESI) for C31H38N7O4[M+H]+572.2980 for the calculated value m/z; measured value m/z is 572.2983.
Example 44
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6m (39.5mg,0.15mmol) were reacted to give the title compound V-44(11.7mg, 13%).
HRMS (ESI) for C32H39N8O4[M+H]+599.3089 for the calculated value m/z; measured value m/z is 599.3106.
Example 45
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6n (35.1mg,0.15mmol) were reacted to give the title compound V-45(16.2mg, 19%).
HRMS (ESI) for C32H40N7O3[M+H]+570.3187 for the calculated value m/z; measured value m/z is 570.3187.
Example 46
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6o (26.6mg,0.15mmol) were reacted to give the title compound V-46(21.5mg, 28%).
HRMS (ESI) for C28H33N8O2[M+H]+513.2721 for the calculated value m/z; measured value m/z is 513.2739.
Example 47
Referring to the method for the synthesis of V-2a in example 2, compound V-47a (25.8mg, 28%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6p (41.7mg,0.15 mmol).
HRMS (ESI) for C32H40N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3200.
The title compound V-47(17.4mg, 97%) was prepared from compound V-47a (20mg, 32.6. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C27H32N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2686.
Example 48
Referring to the method for the synthesis of V-2a in example 2, compound V-48a (29.2mg, 31%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6q (43.8mg,0.15 mmol).
HRMS (ESI) for C33H42N9O4[M+H]+628.3354 for the calculated value m/z; measured value m/z is 628.3355.
The title compound V-48(22.4mg, 99%) was prepared from compound V-48a (25mg, 40.0. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C28H34N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2833.
Example 49
Referring to the method for the synthesis of V-2a in example 2, compound V-49a (26.0mg, 27%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6r (45.9mg,0.15 mmol).
HRMS (ESI) for C34H44N9O4[M+H]+642.3511 for the calculated value m/z; measured value m/z is 642.3511.
The title compound V-49(18.3mg, 96%) was prepared from compound V-49a (21mg, 32.7. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C26H36N9O2[M+H]+542.2986 for the calculated value m/z; measured value m/z is 542.2991.
Example 50
Referring to the method for synthesizing V-1 in example 1, the reaction of compound 5f (60mg,0.15mmol) and 6s (33.3mg,0.15mmol) gave the title compound V-50(23.4mg, 28%).
HRMS (ESI) for C29H36N9O3[M+H]+558.2936 for the calculated value m/z; measured value m/z is 558.2942.
Example 51
Referring to the method for synthesizing V-2a in example 2, compound V-51a (23.1mg, 24%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6t (45.9mg,0.15 mmol).
HRMS (ESI) for C34H44N9O4[M+H]+642.3511 for the calculated value m/z; measured value m/z is 642.3523.
With reference to the method for synthesizing V-2 in example 2, the title compound V-51(17.8mg, 99%) was prepared from compound V-51a (20mg, 31.2. mu. mol).
HRMS (ESI) for C29H36N9O2[M+H]+542.2986 for the calculated value m/z; measured value m/z is 542.2989.
Example 52
Referring to the method for synthesizing V-1 in example 1, compound 5f (60mg,0.15mmol) and 6u (35.3mg,0.15mmol) were reacted to give the title compound V-52(18.0mg, 21%).
HRMS (ESI) for C31H39N8O3[M+H]+571.3140 for the calculated value m/z; measured value m/z is 571.3149.
Example 53
Referring to the method for the synthesis of V-2a in example 2, compound V-53a (17.8mg, 19%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6V (43.5mg,0.15 mmol).
HRMS (ESI) for C33H40N9O4[M+H]+626.3198 for the calculated value m/z; measured value m/z is 626.3203.
The title compound V-53(12.5mg, 93%) was prepared from compound V-53a (15mg, 24.0. mu. mol) by following the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C28H32N9O2[M+H]+526.2673 for the calculated value m/z; measured value m/z is 526.2685.
Example 54
Referring to the method for the synthesis of V-2a in example 2, compound V-54a (17.6mg, 18%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6w (47.7mg,0.15 mmol).
HRMS (ESI) for C35H44N9O4[M+H]+654.3511 for the calculated value m/z; measured value m/z is 654.3519.
The title compound V-54(10.7mg, 91%) was prepared from compound V-54a (13mg, 19.9. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C30H36N9O2[M+H]+554.2986 for the calculated value m/z; measured value m/z is 554.2993.
Example 55
Referring to the method for the synthesis of V-2a in example 2, compound V-55a (12.7mg, 13%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6X (47.7mg,0.15 mmol).
HRMS (ESI) for C35H44N9O4[M+H]+654.3511 for the calculated value m/z; measured value m/z is 654.3513.
The title compound V-55(9.0mg, 99%) was prepared from compound V-55a (10mg, 15.3. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C30H36N9O2[M+H]+554.2986 for the calculated value m/z; measured value m/z is 554.2993.
Example 56
Referring to the method for the synthesis of V-2a in example 2, compound V-56a (22mg, 22%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6y (49.8mg,0.15 mmol).
HRMS (ESI) for C36H46N9O4[M+H]+668.3667 for the calculated value m/z; measured value m/z is 668.3671.
The title compound V-56(12.7mg, 94%) was prepared from compound V-56a (15mg, 22.5. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C31H38N9O2[M+H]+568.3143 for the calculated value m/z; measured value m/z is 568.3161.
Example 57
Referring to the method for the synthesis of V-2a in example 2, compound V-57a (18.2mg, 19%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6z (45.6mg,0.15 mmol).
HRMS (ESI) for C34H42N9O4[M+H]+640.3354 for the calculated value m/z; measured value m/z is 640.3355.
The title compound V-57(13.4mg, 99%) was prepared from compound V-57a (15mg, 23.5. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C29H34N9O2[M+H]+540.2830 for the calculated value m/z; measured value m/z is 540.2839.
Example 58
Referring to the method for the synthesis of V-1 in example 1, compound 5f (60mg,0.15mmol) and 6za (39.0mg,0.15mmol) were reacted to give the title compound V-58(24.1mg, 27%).
HRMS (ESI) for C32H38N9O3[M+H]+596.3092 for the calculated value m/z; measured value m/z is 596.3091.
Example 59
Referring to the method for the synthesis of V-1 in example 1, compound 5f (60mg,0.15mmol) and 6zb (36.9mg,0.15mmol) were reacted to give the title compound V-59(22.7mg, 26%).
HRMS (ESI) for C31H36N9O3[M+H]+582.2936 for the calculated value m/z; measured value m/z is 582.2941.
Example 60
Referring to the method for the synthesis of V-1 in example 1, compound 5f (60mg,0.15mmol) and 6zc (41.4mg,0.15mmol) were reacted to give the title compound V-60(21.1mg, 23%).
HRMS (ESI) for C32H38N9O4[M+H]+612.3041 for the calculated value m/z; measured value m/z is 612.3058.
Example 61
Referring to the method for the synthesis of V-2a in example 2, compound V-61a (23.5mg, 25%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6zd (43.8mg,0.15 mmol).
HRMS (ESI) for C33H42N9O4[M+H]+628.3354 for the calculated value m/z; measured value m/z is 628.3355.
The title compound V-61(13.3mg, 99%) was prepared from compound V-61a (15mg, 23.9. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C28H34N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2821.
Example 62
Referring to the procedure for the synthesis of V-2a in example 2, reaction of compound 5f (60mg,0.15mmol) and 6ze (41.9mg,0.15mmol) gave compound V-62a (12.0mg, 13%).
HRMS (ESI) for C31H39N10O4[M+H]+615.3150 for the calculated value m/z; measured value m/z is 615.3157.
The title compound V-62(8.9mg, 99%) was prepared from compound V-62a (10mg, 16.3. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C26H31N10O2[M+H]+515.2626 for the calculated value m/z; measured value m/z is 515.2633.
Example 63
Referring to the method for the synthesis of V-1 in example 1, compound 5f (60mg,0.15mmol) and 6zf (33mg,0.15mmol) were reacted to give the title compound V-63(25.8mg, 31%).
HRMS (ESI) for C30H38N9O2[M+H]+556.3143 for the calculated value m/z; measured value m/z ═556.3133.
Example 64
Referring to the method for the synthesis of V-1 in example 1, compound 5f (60mg,0.15mmol) and 6zg (35.7mg,0.15mmol) were reacted to give the title compound V-64(22.4mg, 26%).
HRMS (ESI) for C30H37FN9O2[M+H]+574.3049 for the calculated value m/z; measured value m/z is 574.3044.
Example 65
Referring to the method for the synthesis of V-1 in example 1, compound 5f (60mg,0.15mmol) and 6zh (37.5mg,0.15mmol) were reacted to give the title compound V-65(19.3mg, 22%).
HRMS (ESI) for C31H40N9O3[M+H]+586.3249 for the calculated value m/z; measured value m/z is 586.3253.
Example 66
Referring to the method for synthesizing V-2a in example 2, compound V-66a (21.2mg, 23%) was obtained by reacting compound 5f (60mg,0.15mmol) with 6zi (41.7mg,0.15 mmol).
HRMS (ESI) for C32H40N9O4[M+H]+614.3198 for the calculated value m/z; measured value m/z is 614.3200.
The title compound V-66(13.1mg, 97%) was prepared from compound V-66a (15mg, 24.5. mu. mol) by reference to the procedure for the synthesis of V-2a in example 2.
HRMS (ESI) for C27H32N9O2[M+H]+514.2673 for the calculated value m/z; measured value m/z is 514.2673.
Example 67
8-cyclopentyl-6- (5-ethyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one hydrochloride (V-67)
Figure BDA0001553119070000831
8-cyclopentyl-5-methyl-2- (methylthio) -7-oxo-N' - (propionyloxy) -7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxamidine (3bg)
Propionic acid (408mg,5.5mmol) was dissolved in dichloromethane (20mL) under argon and 1-hydroxybenzotriazole (743mg,5.5mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (1.05g,5.5mmol) were added. The resulting solution was stirred at room temperature for 20 minutes, then crude 3b (1.67,5mmol) was added and stirring continued for 2 hours. The reaction solution was quenched with saturated aqueous sodium bicarbonate solution and then extracted with dichloromethane. The organic phases are combined and washed by sodium hydroxide aqueous solution (1N) and saturated saline solution in sequence, dried by anhydrous sodium sulfate, filtered and dried in a spinning way to obtain yellow foamy solid which is a crude product of 3bg and can be directly used for the next reaction.
8-cyclopentyl-6- (5-ethyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2- (methylthio) pyrido [2,3-d ] pyrimidin-7 (8H) -one (4g)
Under the protection of argon, 3bg (5mmol) of the crude product is dissolved in a mixed solvent of ethanol and water (4:1,30mL), sodium acetate trihydrate (1.7g,12.5mmol) is added, and the obtained reaction solution is heated to 90 ℃ for reaction for 14 hours after being sealed. After the reaction solution was cooled, it was diluted with water and extracted with dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and subjected to column chromatography (petroleum ether: dichloromethane: acetone: 7:1:1) to give 4g (835mg, 45% overall yield in three steps) of a yellow foamy solid.
HRMS (ESI) for C18H22N5O2[M+H]+372.1489 for the calculated value m/z; measured value m/z is 372.1490.
8-cyclopentyl-6- (5-ethyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2- (methylsulfinyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (5g)
After 4g (800mg,2.16mmol) was dissolved in chloroform (12mL), 2-benzenesulfonyl-3-phenylazepin (679mg,2.6mmol) was added, and the mixture was stirred at room temperature for 15 hours. The solvent was evaporated to dryness and the crude product was purified by column chromatography (dichloromethane: methanol 50:1) to give 5g (936mg, 93%) of a yellow solid.
HRMS (ESI) for C18H22N5O3S[M+H]+388.1438 for the calculated value m/z; measured value m/z is 388.1441.
(6- ((8-cyclopentyl-6- (5-ethyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-7-oxo-7, 8-dihydropyrido [2,3-3-d ] pyrimidin-2-yl) amino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-67a)
5g (116mg,0.3mmol) and 6b (83.5mg,0.3mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon for 3 times, and then benzotrifluoride (0.6mL) was added and heated to 80 ℃ under stirring with an argon balloon for 16 hours. After cooling to room temperature, the solvent was evaporated under reduced pressure, and the obtained reaction residue was subjected to column chromatography (petroleum ether: dichloromethane: acetone ═ 5:2:1) to obtain a crude product, which was then washed with a mixed solvent of methyl tert-butyl ether and ethyl acetate (4:3) to obtain a yellow solid, i.e., V-67a (48.7mg, 27%).
HRMS (ESI) for C31H40N9O4[M+H]+602.3198 for the calculated value m/z; measured value m/z is 602.3205.
8-cyclopentyl-6- (5-ethyl-1, 2, 4-oxadiazol-3-yl) -5-methyl-2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) pyrido [2,3-d ] pyrimidine-7 (8H) -hydrochloride (V-67)
V67a (30mg, 50. mu. mol) was dissolved in dichloromethane (2mL), the resulting solution was cooled to 0 ℃ with an ice water bath, and then a 1, 4-dioxane solution of hydrogen chloride (4N,0.25mL) was added dropwise, followed by warming to room temperature and stirring for 16 hours. The resulting suspension was filtered and washed with dichloromethane to give the title compound V-67(26.5mg, 99%) as a yellow powder.
HRMS (ESI) for C26H32N9O2[M+H]+502.2673 for the calculated value m/z; measured value m/z is 502.2681.
TABLE 7 examples 68-71
Figure BDA0001553119070000851
Example 68
Referring to the method for synthesizing V-1 in example 1, the reaction of compound 5g (116mg,0.3mmol) and 6h (53.7mg,0.3mmol) gave the title compound V-68(31.6mg, 21%).
HRMS (ESI) for C26H31N8O3[M+H]+503.2514 for the calculated value m/z; measured value m/z is 503.2515.
Example 69
Referring to the method for synthesizing V-1 in example 1, the reaction of compound 5g (116mg,0.3mmol) and 6a (57.6mg,0.3mmol) gave the title compound V-69(27.8mg, 18%).
HRMS (ESI) for C27H34N9O2[M+H]+516.2830 for the calculated value m/z; measured value m/z is 516.2839.
Example 70
Referring to the method for synthesizing V-2 in example 2, compound V-70a (42.5mg, 23%) was obtained by reacting 5g (116mg,0.3mmol) of compound with 6i (87.7mg,0.3 mmol).
HRMS (ESI) for C32H42N9O4[M+H]+616.3354 for the calculated value m/z; measured value m/z is 616.3359.
The title compound V-70(25.6mg, 95%) was prepared from compound V-70a (30mg, 48.8. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C27H34N9O2[M+H]+516.2830 for the calculated value m/z; measured value m/z is 516.2822.
Example 71
Referring to the method for synthesizing V-2a in example 2, 5g (116mg,0.3mmol) and 6g (87.7mg,0.3mmol) of the compound were reacted to obtain compound V-71a (38.8mg, 21%).
HRMS (ESI) for C32H42N9O4[M+H]+616.3354 for the calculated value m/z; measured value m/z is 616.3361.
The title compound V-71(26.9mg, 99%) was prepared from compound V-71a (30mg, 48.8. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C27H34N9O2[M+H]+516.2830 for the calculated value m/z; measured value m/z is 516.2826.
Example 72
Figure BDA0001553119070000871
N' - ((cyclobutanecarbonyl) oxy) -8-cyclopentyl-5-methyl-2- (methylthio) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxamidine (3bh)
Cyclobutylcarboxylic acid (550mg,5.5mmol) was dissolved in dichloromethane (20mL) under argon and 1-hydroxybenzotriazole (743mg,5.5mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (1.05g,5.5mmol) were added. The resulting solution was stirred at room temperature for 20 minutes, then crude 3b (5mmol) was added and stirring continued for 2 hours. The reaction solution was quenched with saturated aqueous sodium bicarbonate solution and then extracted with dichloromethane. After the organic phases are combined, the organic phases are washed by sodium hydroxide aqueous solution (1N) and saturated saline solution in sequence, dried by anhydrous sodium sulfate, filtered and dried in a spinning way to obtain yellow foamy solid, namely a crude product of 3bh, which can be directly used for the next reaction.
6- (5-cyclobutyl-1, 2, 4-oxadiazol-3-yl) -8-cyclopentyl-5-methyl-2- (methylthio) pyrido [2,3-d ] pyrimidin-7 (8H) -one (4H)
Under the protection of argon, the crude product 3bh (5mmol) was dissolved in a mixed solvent of ethanol and water (4:1,30mL), sodium acetate trihydrate (1.7g,12.5mmol) was added, and the resulting reaction solution was sealed and heated to 90 ℃ for reaction for 14 hours. After the reaction solution was cooled, it was diluted with water and extracted with dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated and column chromatographed (petroleum ether: dichloromethane: acetone: 7:1:1) to give a yellow foamy solid of 4h (774.5mg, 39% overall yield over three steps).
HRMS (ESI) for C20H24N5O2S[M+H]+398.1645 for the calculated value m/z; measured value m/z is 398.1641.
6- (5-cyclobutyl-1, 2, 4-oxadiazol-3-yl) -8-cyclopentyl-5-methyl-2- (methylsulfinyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (5H)
After 4h (700mg,1.76mmol) was dissolved in chloroform (12mL), 2-benzenesulfonyl-3-phenylazepine (506mg,1.94mmol) was added, and the mixture was stirred at room temperature for 15 hours. After evaporation of the solvent to dryness, the crude product was purified by column chromatography (dichloromethane: methanol 50:1) to give a yellow solid, i.e. 5h (690mg, 95%).
HRMS (ESI) for C20H24N5O3S[M+H]+414.1594 for the calculated value m/z; measured value m/z is 414.1590.
(6- ((6- (5-cyclobutyl-1, 2, 4-oxadiazol-3-yl) -8-cyclopentyl-5-methyl-7-oxo-7, 8-dihydropyrido [2,3-3-d ] pyrimidin-2-yl) amino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-72a)
5h (124mg,0.3mmol) and 6b (83.4mg,0.3mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon for 3 times, benzotrifluoride (0.6mL) was added, and the mixture was heated to 80 ℃ under stirring with an argon balloon for 16 hours. After cooling to room temperature, the solvent was evaporated under reduced pressure, and the obtained reaction residue was subjected to column chromatography (petroleum ether: dichloromethane: acetone ═ 5:2:1) to obtain a crude product, which was then washed with a mixed solvent of methyl tert-butyl ether and ethyl acetate (4:3) to obtain a yellow solid, i.e., V-72a (35.8mg, 19%).
HRMS (ESI) for C33H42N9O4[M+H]+628.3354 for the calculated value m/z; measured value m/z is 628.3361.
6- (5-cyclobutyl-1, 2, 4-oxadiazol-3-yl) -8-cyclopentyl-5-methyl-2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one hydrochloride (V-72)
V-72a (25mg, 39.9. mu. mol) was dissolved in methylene chloride (2mL), and the resulting solution was cooled to 0 ℃ with an ice water bath, then a 1, 4-dioxane solution of hydrogen chloride (4N,0.2mL) was added dropwise, and then warmed to room temperature and stirred for 16 hours. The resulting suspension was filtered and washed with dichloromethane to give a yellow powder as the title compound V-72(21.0mg, 93%).
HRMS (ESI) for C28H34N9O2[M+H]+528.2830 for the calculated value m/z; measured value m/z is 528.2841.
TABLE 8 examples 73-76
Figure BDA0001553119070000891
Example 73
Referring to the method for synthesizing V-1 in example 1, the title compound V-73(36.4mg, 23%) was obtained by reacting compound 5h (124mg,0.3mmol) and 6h (53.7mg,0.3 mmol).
HRMS (ESI) for C28H33N8O3[M+H]+529.2670 for the calculated value m/z; measured value m/z is 529.2659.
Example 74
Referring to the method for synthesizing V-1 in example 1, the title compound V-74(34.1mg, 21%) was obtained by reacting compound 5h (124mg,0.3mmol) and 6a (57.7mg,0.3 mmol).
HRMS (ESI) for C29H36N9O2[M+H]+542.2986 for the calculated value m/z; measured value m/z is 542.2971.
Example 75
Referring to the method for the synthesis of V-2a in example 2, compound V-75a (42.3mg, 22%) was obtained by reacting compound 5h (124mg,0.3mmol) with 6i (87.7mg,0.3 mmol).
HRMS (ESI) for C34H44N9O4[M+H]+642.3511 for the calculated value m/z; measured value m/z is 642.3522.
The title compound V-75(17.5mg, 97%) was prepared from compound V-75a (20mg, 31.2. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C29H36N9O2[M+H]+542.2986 for the calculated value m/z; measured value m/z is 542.2993.
Example 76
Referring to the method for synthesizing V-2a in example 2, compound V-76a (46.2mg, 24%) was obtained by reacting compound xx (124mg,0.3mmol) with 6g (87.7mg,0.3 mmol).
HRMS (ESI) for C34H44N9O4[M+H]+642.3511 for the calculated value m/z; measured value m/z is 642.3519.
The title compound V-76(16.4mg, 91%) was prepared from compound V-76a (20mg, 31.2. mu. mol) by reference to the procedure for the synthesis of V-2 in example 2.
HRMS (ESI) for C29H36N9O2[M+H]+542.2986 for the calculated value m/z; measured value m/z is 542.2991.
Example 77
Figure BDA0001553119070000901
8-cyclopentyl-5-methyl-2-methylsulfanyl-6- (5-trifluoromethyl-1, 2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (4i)
Crude 3b (0.5mmol) was dissolved in trifluoroacetic anhydride (1mL) and heated to 110 ℃ with stirring for 2 h. The reaction solution was cooled, then washed with saturated aqueous sodium bicarbonate solution in an ice-water bath to pH 8, followed by extraction with dichloromethane, and the organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatography (petroleum ether: ethyl acetate: 10:1) gave 4i (131mg, 63.7%) as a yellow viscous solid.
1H NMR(400MHz,CDCl3,ppm):8.86(s,1H),6.04–5.95(m,1H),2.65(s,3H),2.43(s,3H),2.37–2.28(m,2H),2.11–2.01(m,2H),1.94–1.86(m,2H),1.73–1.65(m,2H).
19F NMR(376MHz,CDCl3,ppm):–65.01(s,3F).
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (5-trifluoromethyl-1, 2, 4-oxadiazol-3-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (5i)
After 4i (114.5mg,0.278mmol) was dissolved in chloroform (2.8mL), 2-benzenesulfonyl-3-phenyloxaziridine (123.6mg,0.473mmol) was added and the mixture was stirred at room temperature for 40 hours. After evaporation of the solvent to dryness, the crude product was purified by column chromatography (dichloromethane: methanol 50:1) to yield 5i (77mg, 64.7%) as a white powder.
1H NMR(400MHz,CDCl3,ppm):9.18(s,1H),6.07–5.98(m,1H),3.02(s,3H),2.53(s,3H),2.30–2.22(m,2H),2.13–2.07(m,2H),2.01–1.91(m,2H),1.72–1.67(m,2H).
19F NMR(376MHz,CDCl3,ppm):–64.97(s,3F).
(6- ((8-cyclopentyl-5-methyl-7-oxo-6- (5- (trifluoromethyl) -1,2, 4-oxadiazol-3-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (V-77a)
5i (72mg,0.168mmol) and tert-butyl 4- (6-aminopyridin-3-yl) piperazine-1-carboxylate (6b,49.2mg,0.177mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon 3 times, benzotrifluoride (0.7mL) was added, and the mixture was heated to 80 ℃ under stirring with an argon balloon for 16 hours. After cooling to room temperature, the solvent was evaporated to dryness under reduced pressure, and the obtained reaction residue was subjected to column chromatography (petroleum ether: dichloromethane: acetone ═ 5:1:1) to obtain a crude product, which was then washed with methyl tert-butyl ether to obtain a yellow powder, i.e., V-77a (17mg, 15.7%).
1H NMR(400MHz,CDCl3,ppm):8.89(s,1H),8.43(br,1H),8.20(d,J=8.8Hz,1H),8.07(s,1H),7.35(dd,J=9.0,2.2Hz,1H),5.98–5.89(m,1H),3.62(t,J=5.0Hz,4H),3.14(t,J=4.8Hz,4H),2.42(s,3H),2.38–2.31(m,2H),2.11–2.01(m,2H),1.94–1.86(m,2H),1.73–1.67(m,2H),1.49(s,9H).
19F NMR(376MHz,CDCl3,ppm):–65.06(s,3F).
8-cyclopentyl-5-methyl-2- ((5- (piperazin-1-yl) pyridin-2-yl) amino) -6- (5- (trifluoromethyl) -1,2, 4-oxadiazol-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one hydrochloride (V-77)
V-77a (16.5mg,0.0257mmol) was dissolved in dichloromethane (1mL), the resulting solution was cooled to 0 ℃ with an ice water bath, and then a solution of hydrogen chloride in 1, 4-dioxane (4N,0.48mL) was added dropwise, followed by warming to room temperature and stirring for 21 hours. The resulting suspension was filtered and washed with dichloromethane to give a yellow powder as the title compound V-77(13mg, 87.5%).
1H NMR(400MHz,D2O,ppm):9.10(s,1H),8.08(s,1H),7.87(s,1H),7.51(s,1H),5.86–5.79(m,1H),3.59–3.42(m,8H),2.45(s,3H),2.17–2.07(m,2H),2.02–1.90(m,4H),1.73–1.64(m,2H).
Example 78
Figure BDA0001553119070000921
8-cyclopentyl-5-methyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxylic acid (7a)
8-cyclopentyl-5-methyl-2-methylthio-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carbonitrile (2b,60mg,0.2mmol) was suspended in a mixed solvent of 5% aqueous sodium hydroxide (1mL) and ethanol (0.5mL), and the mixture was heated to 60 ℃ with stirring for 18 hours to react, whereby the solution gradually became clear. After cooling to room temperature, hydrochloric acid (6N) was added dropwise until the solution became weakly acidic (pH 4-5), and the precipitated solid was filtered and washed with ethyl acetate to remove a small amount of organic impurities from the solid. Then the filtrate was removed, the remaining crude solid was washed with ethanol, and the resulting filtrate was evaporated to dryness to give a pale solid, 8-cyclopentyl-5-methyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxylic acid (7a,31.4mg, 49.2%).
1H NMR(400MHz,CDCl3,ppm):9.35(s,1H),5.27–5.19(m,1H),2.68(s,3H),2.64(s,3H),2.45–2.37(m,2H),2.12–1.98(m,4H),1.73–1.64(m,2H).
N' -acetyl-8-cyclopentyl-5-methyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxylic acid hydrazide (8a)
8-cyclopentyl-5-methyl-2-methylsulfanyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carboxylic acid (7a,280mg,0.88mmol) and O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (433.4mg,1.14mmol) were placed in a reaction flask, N, N-dimethylformamide (5mL) and N, N-diisopropylethylamine (339.9mg,2.63mmol) were added, and after stirring at room temperature for 5 minutes, acetyl hydrazine (71.7mg,0.97mmol) was added. Stirring is carried out for 15 hours at room temperature, ethyl acetate is added, the mixture is washed by saturated salt water for 3 times, an organic phase is dried by anhydrous sodium sulfate, the mixture is filtered, a filtrate is decompressed and evaporated to dryness, and a crude product is subjected to column chromatography to obtain a light brown solid, namely N' -acetyl-8-cyclopentyl-5-methyl-2-methylthio-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-formylhydrazine (8a,203mg, 61.7%).
1H NMR(300MHz,CDCl3,ppm):10.34(br,1H),8.92(s,1H),8.33(br,1H),6.04–5.92(m,1H),2.75(s,3H),2.64(s,3H),2.34–2.21(m,2H),2.13(s,3H),2.13–2.02(m,2H),1.89–1.82(m,2H),1.75–1.67(m,2H).
8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (9a)
N' -acetyl-8-cyclopentyl-5-methyl-2-methylthio-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidine-6-carbohydrazide (8a,200mg,0.53mmol) was placed in a dry reaction flask under argon protection, dried toluene (2mL) and phosphorus oxychloride (89.4mg,0.58mmol) were added, and the mixture was stirred and heated at reflux for 4 hours. The reaction solution was cooled to room temperature, poured into ice water, adjusted to neutral (pH 7) with aqueous sodium hydroxide (1N), extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was evaporated to dryness under reduced pressure and subjected to column chromatography to obtain a yellow solid, i.e., 8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (9a,42mg, 22%).
1H NMR(300MHz,CDCl3,ppm):8.89(s,1H),6.07–5.95(m,1H),2.69(s,3H),2.67(s,3H),2.56(s,3H),2.40–2.29(m,2H),2.09–1.98(m,2H),1.94–1.87(m,2H),1.78–1.70(m,2H).
8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (10a)
8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (9a,112mg,0.31mmol) was dissolved in dichloromethane (2mL), cooled to 0 ℃ in a ice water bath, and then m-chloroperoxybenzoic acid (85%, 50.9mg,0.25mmol) was added in portions and stirred at room temperature for 3 hours. Saturated aqueous sodium sulfite (2mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and subjected to column chromatography to give a yellow solid, 8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (10a,86mg, 73.5%).
HRMS (ESI) for C17H19N5O3S[M+H]+374.1281 for the calculated value m/z; measured value m/z is 374.1276.
8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one (VI-1)
8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2-methanesulfinyl-8H-pyrido [2,3-d ] pyrimidin-7-one (10a,82mg,0.22mmol) and 2-amino-5- (4-methylpiperazin-1-yl) pyridine (6a,44.3mg,0.23mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon 3 times, benzotrifluoride (1mL) was added, and the mixture was heated to 80 ℃ under argon balloon protection with stirring for 16 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered, and washed with ethyl acetate to give a yellow solid, i.e., 8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one (VI-1,20.8mg, 18.9%).
HRMS (ESI) for C26H31N9O2[M+H]+502.2674 for the calculated value m/z; measured value m/z is 502.2680.
Example 79
Figure BDA0001553119070000951
4- (6- (8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) -4-methylpyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (VI-2a)
Referring to the procedure for the synthesis of V-15a in example 15, compound VI-2a (18.7mg, 14.5%) was reacted from compound 10a (80mg,0.214mmol) and 6g (65.8mg,0.225 mmol).
HRMS (ESI) for C31H39N9O4[M+H]+602.3198 for the calculated value m/z; measured value m/z is 602.3193.
8-cyclopentyl-5-methyl-6- (5-methyl-1, 3, 4-oxadiazol-2-yl) -2- (4-methyl-5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VI-2)
Referring to the synthesis of V-15 in example 15, the title compound VI-2(14mg, 99%) was prepared from compound VI-2a (16mg,0.027 mmol).
HRMS (ESI) for C26H31N9O2[M+H]+502.2673 for the calculated value m/z; measured value m/z is 502.2682.
Example 80
Figure BDA0001553119070000961
8-cyclopentyl-2-methylsulfanyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (11a)
Under argon, cuprous iodide (56mg,0.294mmol), 6-bromo-8-cyclopentyl-2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (1a,1g,2.94mmol), 2-azetidinone (418mg,5.88mmol) and potassium carbonate (609mg,4.41mmol) were added to a 25mL stopcock. The reaction tube was sealed with a rubber stopper, evacuated and returned to argon 3 times, and then N, N' -dimethylethylenediamine (52mg,0.588mmol) and toluene (4mL) were injected. After replacing the rubber stopper with a teflon stopper, the reaction mixture was stirred and heated to 100 ℃ for overnight reaction. After the reaction is cooled to room temperature, the cover is opened, the reaction solution is filtered, the filtrate is evaporated to dryness under reduced pressure, and a white solid obtained by column chromatography (petroleum ether/ethyl acetate ═ 2:1 to 0:1) is 8-cyclopentyl-2-methylthio-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (11a,890mg, 91.6%).
1H NMR(400MHz,CDCl3,ppm):8.59(s,1H),8.18(s,1H),5.98–5.89(m,1H),4.20(t,J=4.8Hz,2H),3.15(t,J=4.6Hz,2H),2.60(s,3H),2.36–2.27(m,2H),2.12–2.02(m,2H),1.92–1.84(m,2H),1.74–1.66(m,2H).
8-cyclopentyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12a)
8-cyclopentyl-2-methylsulfanyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (11a,820mg,2.48mmol) was dissolved in chloroform (10mL), and m-chloroperoxybenzoic acid (85%, 453mg,2.23mmol) was added and stirred at room temperature for 3 hours. Saturated aqueous sodium sulfite (10mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (petrol ether/ethyl acetate 2:1 to 0:1) to give 8-cyclopentyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12a,700mg, 81.4%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):8.88(s,1H),8.30(s,1H),6.02–5.93(m,1H),4.26(t,J=4.8Hz,2H),3.19(t,J=4.8Hz,2H),2.96(s,3H),2.29–2.19(m,2H),2.15–2.08(m,2H),1.98–1.89(m,2H),1.73–1.63(m,2H).
8-cyclopentyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VII-1)
8-cyclopentyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12a,120mg,0.346mmol) and 2-amino-5- (4-methylpiperazin-1-yl) pyridine (6a,69.9mg,0.364mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and then replaced with argon 3 times, benzotrifluoride (1.7mL) was added, and the mixture was heated to 95 ℃ under argon balloon protection with stirring for 24 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered and washed with ethyl acetate to give a yellow solid, 8-cyclopentyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VII-1,26mg, 15.8%).
1H NMR(400MHz,CDCl3,ppm):8.59(s,1H),8.22(br,1H),8.16–8.14(m,2H),8.05(d,J=2.0Hz,1H),7.32(dd,J=9.2,2.4Hz,1H),5.94–5.85(m,1H),4.17(t,J=4.2Hz,2H),3.22(t,J=4.4Hz,4H),3.13(t,J=4.2Hz,2H),2.63(t,J=4.2Hz,4H),2.38(s,3H),2.38–2.30(m,2H),2.11–2.02(m,2H),1.92–1.87(m,2H),1.74–1.66(m,2H).
HRMS (ESI) for C25H30N8O2[M+H]+475.2564 for the calculated value m/z; measured value m/z is 475.2561.
Example 81
Figure BDA0001553119070000981
4- (6- (8-cyclopentyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (VII-2a)
8-cyclopentyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12a,173.2mg,0.5mmol) and tert-butyl 4- (6-aminopyridin-3-yl) piperazine-1-carboxylate (6b,146mg,0.525mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and then replaced with argon 3 times, benzotrifluoride (2mL) was added, and the mixture was heated to 90 ℃ under stirring with an argon balloon for 48 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered and washed with ethyl acetate to give a yellow solid, i.e., tert-butyl 4- (6- (8-cyclopentyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylate (VII-2a,62mg, 22%).
1H NMR(400MHz,CDCl3,ppm):8.61(s,1H),8.35(br,1H),8.19–8.16(m,2H),8.06(d,J=2.4Hz,1H),7.33(dd,J=9.0,2.6Hz,1H),5.94–5.85(m,1H),4.17(t,J=4.6Hz,2H),3.61(t,J=4.8Hz,4H),3.15–3.10(m,6H),2.39–2.31(m,2H),2.11–2.02(m,2H),1.92–1.85(m,2H),1.75–1.69(m,2H),1.49(s,9H).
8-cyclopentyl-6- (2-oxoazetidin-1-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VII-2)
Tert-butyl 4- (6- (8-cyclopentyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylate (VII-2a,45mg,0.08mmol) was placed in a reaction flask, and a solution of hydrogen chloride in 1, 4-dioxane (4N,2mL) and methanol (2mL) were added. After stirring at room temperature for 2 hours, the solvent was removed by rotary evaporation under reduced pressure, and the resulting crude product was stirred in ethyl acetate for half an hour and then filtered to give a yellow powder, 8-cyclopentyl-6- (2-oxoazetidin-1-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VII-2,39mg, 98%).
1H NMR(400MHz,D2O,ppm):8.42(s,1H),7.98(dd,J=9.6,2.4Hz,1H),7.59(d,J=2.8Hz,1H),7.30(d,J=9.6Hz,1H),6.33(s,1H),5.82–5.74(m,1H),3.48(s,8H),3.34(t,J=6.6Hz,2H),2.71(t,J=6.6Hz,2H),2.21–2.13(m,2H),2.09–1.93(m,4H),1.78–1.71(m,2H).
Example 82
Figure BDA0001553119070000991
8-cyclopentyl-5-methyl-2-methylsulfanyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (11b)
Under argon, cuprous iodide (53mg,0.282mmol), 6-bromo-8-cyclopentyl-5-methyl-2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (1b,1g,2.82mmol), 2-azetidinone (0.4g,5.64mmol) and potassium carbonate (584.6mg,4.23mmol) were added to a 25mL block. The reaction tube was sealed with a rubber stopper, evacuated and returned to argon 3 times, and then N, N' -dimethylethylenediamine (50mg,0.564mmol) and toluene (5mL) were injected. After replacing the rubber stopper with a teflon stopper, the reaction mixture was stirred and heated to 100 ℃ for overnight reaction. After the reaction is cooled to room temperature, the reaction solution is uncapped, the reaction solution is filtered, the filtrate is evaporated to dryness under reduced pressure, and a white solid obtained by column chromatography (petroleum ether/ethyl acetate ═ 2:1 to 0:1) is 8-cyclopentyl-5-methyl-2-methylthio-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (11b,500mg, 51.5%).
1H NMR(400MHz,CDCl3,ppm):8.77(s,1H),5.97–5.88(m,1H),3.95(t,J=4.6Hz,2H),3.19(t,J=4.6Hz,2H),2.61(s,3H),2.42(s,3H),2.34–2.25(m,2H),2.11–2.01(m,2H),1.91–1.83(m,2H),1.73–1.67(m,2H).
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12b)
8-cyclopentyl-5-methyl-2-methylsulfanyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (11b,550mg,1.6mmol) was dissolved in chloroform (8mL), and m-chloroperoxybenzoic acid (85%, 292mg,1.44mmol) was added and stirred at room temperature for 3 hours. Saturated aqueous sodium sulfite (10mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (petrol ether/ethyl acetate 2:1 to 0:1) to give 8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12b,435mg, 75.6%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):9.08(s,1H),6.01–5.92(m,1H),4.09–4.02(m,2H),3.23(t,J=4.6Hz,2H),2.97(s,3H),2.50(s,3H),2.27–2.19(m,2H),2.14–2.08(m,2H),1.98–1.89(m,2H),1.74–1.67(m,2H).
8-cyclopentyl-5-methyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VII-3)
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12b,120mg,0.333mmol) and 2-amino-5- (4-methylpiperazin-1-yl) pyridine (6a,67.2mg,0.35mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and then replaced with argon 3 times, benzotrifluoride (1.7mL) was added, and the mixture was heated to 90 ℃ under stirring with an argon balloon for 18 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered and washed with ethyl acetate to give a yellow solid, 8-cyclopentyl-5-methyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VII-3,46mg, 28%).
1H NMR(400MHz,CDCl3,ppm):8.79(s,1H),8.25(br,1H),8.15(d,J=9.2Hz,1H),8.05(d,J=2.4Hz,1H),7.33(dd,J=9.2,2.8Hz,1H),5.93–5.84(m,1H),3.91(t,J=4.2Hz,2H),3.23–3.17(m,6H),2.61(t,J=4.8Hz,4H),2.41(s,3H),2.37(s,3H),2.37–2.29(m,2H),2.10–2.01(m,2H),1.91–1.83(m,2H),1.71–1.63(m,2H).
HRMS (ESI) for C26H32N8O2[M+H]+489.2721 for the calculated value m/z; measured value m/z is 489.2715.
Example 83
Figure BDA0001553119070001011
4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (VII-4a)
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (12b,120mg,0.333mmol) and tert-butyl 4- (6-aminopyridin-3-yl) piperazine-1-carboxylate (6b,94.4mg,0.34mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon 3 times, benzotrifluoride (1.3mL) was added, and the mixture was heated to 85 ℃ under argon balloon protection for 17 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 3 hours. The precipitated solid was filtered and washed with ethyl acetate to give a bright yellow solid, i.e., tert-butyl 4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylate (VII-4a,53.3mg, 27.9%).
1H NMR(400MHz,CDCl3,ppm):8.82(s,1H),8.44(br,1H),8.18(d,J=9.2Hz,1H),8.06(d,J=2.8Hz,1H),7.33(dd,J=9.0,3.0Hz,1H),5.93–5.84(m,1H),3.92(t,J=4.4Hz,2H),3.61(t,J=5.0Hz,4H),3.18(t,J=4.4Hz,2H),3.12(t,J=4.8Hz,4H),2.42(s,3H),2.37–2.29(m,2H),2.11–2.01(m,2H),1.91–1.83(m,2H),1.74–1.64(m,2H),1.49(s,9H).
8-cyclopentyl-5-methyl-6- (2-oxoazetidin-1-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VII-4)
Tert-butyl 4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylate (VII-4a,43.4mg,0.0755mmol) was placed in a reaction flask and a solution of hydrogen chloride in 1, 4-dioxane (4N,2mL) and methanol (2mL) were added. After stirring at room temperature for 2 hours, the solvent was removed by rotary evaporation under reduced pressure, and the resulting crude product was stirred in ethyl acetate for half an hour and then filtered to give a yellow powder, 8-cyclopentyl-5-methyl-6- (2-oxoazetidin-1-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VII-4,35mg, 90.7%).
1H NMR(400MHz,D2O,ppm):9.05(s,1H),8.14(dd,J=9.6,2.8Hz,1H),7.87(d,J=2.8Hz,1H),7.53(d,J=9.6Hz,1H),5.91–5.82(m,1H),3.61(t,J=6.0Hz,2H),3.57–3.54(m,4H),3.49–3.47(m,4H),2.81(t,J=6.0Hz,2H),2.60(s,3H),2.23–2.15(m,2H),2.07–1.91(m,4H),1.76–1.66(m,2H).
Example 84
Figure BDA0001553119070001021
4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxoazetidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) -4-methylpyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (VII-5a)
Referring to the procedure for the synthesis of VII-4a in example 32, compound 12b (100mg,0.277mmol) and 6g (84.8mg,0.29mmol) were reacted to give compound VII-5a (43.3mg, 26.5%).
HRMS (ESI) for C31H40N8O4[M+H]+589.3245 for the calculated value m/z; measured value m/z is 589.3253.
8-cyclopentyl-5-methyl-2- (4-methyl-5- (piperazin-1-yl) pyridin-2-ylamino) -6- (2-oxoazetidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VII-5)
The title compound VII-5(34.3mg, 96%) was prepared from compound VII-5a (40mg,0.068mmol) by referring to the procedure for the synthesis of VII-4 in example 32.
HRMS (ESI) for C26H32N8O2[M+H]+489.2721 for the calculated value m/z; measured value m/z is 489.2724.
Example 85
Figure BDA0001553119070001031
8-cyclopentyl-2-methylsulfanyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (13a)
Copper powder (13mg,0.206mmol), 6-bromo-8-cyclopentyl-2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (1a,700mg,2.06mmol), 2-pyrrolidone (350mg,4.11mmol) and potassium carbonate (426mg,3.09mmol) were added to a 25mL sealed tube under argon. The reaction tube was sealed with a rubber stopper, evacuated and returned to argon 3 times, and then N, N' -dimethylethylenediamine (36mg,0.411mmol) and toluene (4mL) were injected. After replacing the rubber stopper with a teflon stopper, the reaction mixture was stirred and heated to 135 ℃ for overnight reaction. After the reaction was cooled to room temperature, the reaction solution was uncapped, the reaction solution was filtered, the filtrate was evaporated to dryness under reduced pressure, and a white solid obtained by column chromatography (petroleum ether/ethyl acetate ═ 2:1 to 0:1) was 8-cyclopentyl-2-methylthio-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (13a,490mg, 69%).
1H NMR(400MHz,CDCl3,ppm):8.61(s,1H),7.77(s,1H),6.01–5.92(m,1H),3.96(t,J=7.0Hz,2H),2.62(s,3H),2.57(t,J=8.0Hz,2H),2.37–2.28(m,2H),2.22–2.14(m,2H),2.12–2.03(m,2H),1.93–1.85(m,2H),1.74–1.64(m,2H).
8-cyclopentyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14a)
8-cyclopentyl-2-methylsulfanyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (13a,490mg,1.42mmol) was dissolved in dichloromethane (10mL), m-chloroperoxybenzoic acid (85%, 260mg,1.28mmol) was added, and the mixture was stirred at room temperature for 3 hours. Saturated aqueous sodium sulfite (10mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (petrol ether/ethyl acetate 2:1 to 0:1) to give 8-cyclopentyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14a,320mg, 62.4%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):8.93(s,1H),7.97(s,1H),6.04–5.95(m,1H),4.05–4.01(m,2H),2.97(s,3H),2.58(t,J=8.0Hz,2H),2.29–2.07(m,6H),1.99–1.89(m,2H),1.74–1.65(m,2H).
8-cyclopentyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VIII-1)
8-cyclopentyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14a,120mg,0.333mmol) and 2-amino-5- (4-methylpiperazin-1-yl) pyridine (6a,67.2mg,0.35mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and replaced with argon 3 times, benzotrifluoride (1.7mL) was added, and the mixture was heated to 95 ℃ under stirring with an argon balloon for 24 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered and washed with ethyl acetate to give a yellow solid, i.e., 8-cyclopentyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VIII-1,46mg, 28.3%).
1H NMR(400MHz,CDCl3,ppm):8.62(s,1H),8.44(br,1H),8.16(d,J=9.2Hz,1H),8.07(d,J=2.4Hz,1H),7.70(s,1H),7.33(dd,J=9.0,3.0Hz,1H),5.96–5.87(m,1H),3.93(t,J=7.0Hz,2H),3.22(t,J=4.8Hz,4H),2.61(t,J=4.6Hz,4H),2.56(t,J=8.0Hz,2H),2.39–2.30(m,2H),2.37(s,3H),2.22–2.14(m,2H),2.10–2.02(m,2H),1.93–1.85(m,2H),1.71–1.64(m,2H).
HRMS (ESI) for C26H32N8O2[M+H]+489.2721 for the calculated value m/z; measured value m/z is 489.2718.
Example 86
Figure BDA0001553119070001051
8-cyclopentyl-5-methyl-2-methylsulfanyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (13b)
Under argon, cuprous iodide (268mg,1.41mmol), 6-bromo-8-cyclopentyl-5-methyl-2-methylsulfanyl-8H-pyrido [2,3-d ] pyrimidin-7-one (1b,5g,14.1mmol), 2-pyrrolidone (2.4g,28.2mmol) and potassium carbonate (2.92g,21.15mmol) were added to a 100mL block. The reaction tube was sealed with a rubber stopper, evacuated and returned to argon 3 times, and then N, N' -dimethylethylenediamine (248mg,2.82mmol) and toluene (25mL) were injected. After replacing the rubber stopper with a teflon stopper, the reaction mixture was stirred and heated to 100 ℃ for overnight reaction. After the reaction is cooled to room temperature, the cover is opened, the reaction solution is filtered, the filtrate is evaporated to dryness under reduced pressure, and a white solid obtained by column chromatography (petroleum ether/ethyl acetate is 2:1 to 0:1) is 8-cyclopentyl-5-methyl-2-methylthio-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (13b,420mg, 8.3%).
1H NMR(400MHz,CDCl3,ppm):8.76(s,1H),5.98–5.89(m,1H),4.14–4.08(m,1H),3.42–3.36(m,1H),2.67–2.50(m,2H),2.61(s,3H),2.34(s,3H),2.34–2.15(m,4H),2.09–2.01(m,2H),1.90–1.82(m,2H),1.70–1.65(m,2H).
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14b)
8-cyclopentyl-5-methyl-2-methylsulfanyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (13b,420mg,1.17mmol) was dissolved in chloroform (6mL), and m-chloroperoxybenzoic acid (85%, 214mg,1.05mmol) was added and the mixture was stirred at room temperature for 40 minutes. Saturated aqueous sodium sulfite (8mL) was added to extract the reaction out, followed by three extractions with dichloromethane. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and column chromatographed (petrol ether/ethyl acetate 2:1 to 0:1) to give 8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14b,120mg, 27.3%) as a white solid.
1H NMR(400MHz,CDCl3,ppm):9.07(d,J=8.0Hz,1H),6.01–5.89(m,1H),4.18–4.12(m,1H),3.44–3.38(m,1H),2.95(s,3H),2.67–2.51(m,2H),2.41(s,3H),2.35–2.18(m,4H),2.12–2.04(m,2H),1.94–1.88(m,2H),1.71–1.64(m,2H).
8-cyclopentyl-5-methyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VIII-2)
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14b,92mg,0.246mmol) and 2-amino-5- (4-methylpiperazin-1-yl) pyridine (6a,49.6mg,0.258mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and then replaced with argon 3 times, benzotrifluoride (1.2mL) was added, and the mixture was heated to 90 ℃ under stirring with an argon balloon for 18 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 2 hours. The precipitated solid was filtered and subjected to column chromatography to obtain a yellow solid, i.e., 8-cyclopentyl-5-methyl-2- (5- (4-methylpiperazin-1-yl) pyridin-2-ylamino) -6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (VIII-2,18mg, 14.6%).
1H NMR(400MHz,CDCl3,ppm):8.78(s,1H),8.34(br,1H),8.16(d,J=9.2Hz,1H),8.05(d,J=2.0Hz,1H),7.33(dd,J=8.8,2.4Hz,1H),5.93–5.84(m,1H),4.13–4.07(m,1H),3.40–3.37(m,1H),3.21(t,J=4.4Hz,4H),2.67–2.50(m,6H),2.37(s,3H),2.33(s,3H),2.29–2.16(m,2H),2.06–1.82(m,6H),1.70–1.63(m,2H).
HRMS (ESI) for C27H34N8O2[M+H]+503.2877 for the calculated value m/z; measured value m/z is 503.2878.
Example 87
Figure BDA0001553119070001071
4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxopyrrolidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (VIII-3a)
8-cyclopentyl-5-methyl-2-methanesulfinyl-6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one (14b,120mg,0.32mmol) and tert-butyl 4- (6-aminopyridin-3-yl) piperazine-1-carboxylate (98mg,0.35mmol) were placed in a dry 10mL reaction tube, sealed with a rubber stopper and then replaced with argon 3 times, benzotrifluoride (1.3mL) was added, and the mixture was heated to 80 ℃ under argon balloon protection for 24 hours. After cooling to room temperature, a small amount of ethyl acetate was added and stirring was continued for about 6 hours. The precipitated solid was filtered and washed with ethyl acetate to give a yellow solid, i.e., tert-butyl 4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxopyrrolidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylate (VIII-3a,33.6mg, 17.8%).
1H NMR(400MHz,CDCl3,ppm):8.79(s,1H),8.28(br,1H),8.19(d,J=9.2Hz,1H),8.05(d,J=2.8Hz,1H),7.33(dd,J=8.8,2.8Hz,1H),5.93–5.84(m,1H),4.13–4.07(m,1H),3.61(t,J=5.0Hz,4H),3.42–3.36(m,1H),3.12(t,J=4.6Hz,4H),2.68–2.50(m,2H),2.38–2.18(m,4H),2.33(s,3H),2.09–2.01(m,2H),1.90–1.82(m,2H),1.67–1.62(m,2H),1.49(s,9H).
8-cyclopentyl-5-methyl-6- (2-oxopyrrolidin-1-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VIII-3)
Tert-butyl 4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxopyrrolidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) pyridin-3-yl) piperazine-1-carboxylate (VIII-3a,25mg,0.0425mmol) was placed in a reaction flask and a solution of hydrogen chloride in 1, 4-dioxane (4N,1.1mL) and methanol (1.1mL) were added. After stirring at room temperature for 2 hours, the solvent was removed by rotary evaporation under reduced pressure, and the resulting crude product was stirred in ethyl acetate for half an hour and then filtered to give a yellow powder, 8-cyclopentyl-5-methyl-6- (2-oxopyrrolidin-1-yl) -2- (5- (piperazin-1-yl) pyridin-2-ylamino) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VIII-3,22mg, 99%).
1H NMR(400MHz,D2O,ppm):9.08(s,1H),8.11(dd,J=9.6,2.4Hz,1H),7.90(d,J=2.8Hz,1H),7.54(d,J=9.6Hz,1H),5.92–5.83(m,1H),3.83–3.77(m,1H),3.65–3.63(m,1H),3.56–3.54(m,4H),3.49–3.46(m,4H),2.69–2.65(m,2H),2.43(s,3H),2.36–2.29(m,2H),2.16–2.07(m,2H),2.00–1.88(m,4H),1.72–1.66(m,2H).
Example 88
Figure BDA0001553119070001081
4- (6- (8-cyclopentyl-5-methyl-7-oxo-6- (2-oxopyrrolidin-1-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-ylamino) -4-methylpyridin-3-yl) piperazine-1-carboxylic acid tert-butyl ester (VIII-4a)
Referring to the procedure for the synthesis of VIII-3a in example 36, compound 14b (100mg,0.267mmol) and 6g (82mg,0.28mmol) were reacted to give compound VIII-4a (24.6mg, 15.3%).
HRMS (ESI) for C32H42N8O4[M+H]+603.3402 for the calculated value m/z; measured value m/z is 603.3996.
8-cyclopentyl-5-methyl-2- (4-methyl-5- (piperazin-1-yl) pyridin-2-ylamino) -6- (2-oxopyrrolidin-1-yl) -8H-pyrido [2,3-d ] pyrimidin-7-one hydrochloride (VIII-4)
The title compound VIII-4(19mg, 97%) was prepared from compound VIII-4a (22mg,0.037mmol) by referring to the procedure for the synthesis of VIII-3 in example 36.
HRMS (ESI) for C27H34N8O2[M+H]+503.2877 for the calculated value m/z; measured value m/z is 503.2883.
Example 89
And (3) measuring the potency of the biochemical kinase inhibition test. Kinase activity assay and IC50 assay. First, 10ng of recombinant CDK 4/cyclin D1 or recombinant CDK 6/cyclin D3(Life Technologies PV4204) was diluted in kinase buffer (20mM Tris pH7.5,10mM MgCl. RTM20.01% NP-40,2mM DTT), at the indicated concentration of inhibitor for 30 minutes at room temperature. The kinase reaction was initiated by the addition of 1. mu.g (1.5. mu.M) of recombinant retinoblastoma (Rb) protein, 5. mu.M ATP and 10. mu. Ci of γ -32P-ATP. The reaction was incubated at 30 ℃ for 20 minutes, quenched by the addition of 2 × Laemmli sample buffer, heated at 95 ℃ for three minutes, solubilized by 12% acrylamide SDS-PAGE, and autoradiographed. The autoradiogram was scanned and the corresponding phosphorylated substrate protein bands were quantified using a densitometer (Bio-Rad). The resulting optical density values were plotted as a function of log drug concentration using Prism 4 Graphpad software, and IC50 values were determined by plotting a sigmoidal nonlinear regression curve with variable slope.
The results of the enzyme inhibitory activity of the compounds of the present invention are shown in the following table. Compounds were tested in a 10-fold amount of IC50 in a 10 μ M initial concentration three-fold serial dilution mode. Control compound: staurosporine, tested in a 10-fold-amount IC50, 20 μ M starting concentration four-fold serial dilution mode. The reaction was run under 10. mu.M ATP.
Figure BDA0001553119070001101
Figure BDA0001553119070001111
Figure BDA0001553119070001121
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (25)

1. A compound of formula (V), or a pharmaceutically acceptable salt thereof:
Figure FDA0002632462510000011
wherein the content of the first and second substances,
A1is selected from N;
A2selected from CH, C (halogen), C (C)1-6Alkyl) Or C (OC)1-6Alkyl groups);
A3selected from CH or C (C)1-6Alkyl groups);
R1is selected from H or C1-6An alkyl group;
R2is C3-7A cycloalkyl group;
R3is selected from C1-6Alkyl radical, C1-6Haloalkyl, -O-C1-6alkylene-R4Or optionally substituted with 1,2 or 3R4The following groups substituted with groups:
Figure FDA0002632462510000012
R4selected from H, C1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2or-C1-6alkylene-OH;
R8selected from H, C1-6Alkyl radical, C1-6Haloalkyl or C3-7A cycloalkyl group.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,
A1is selected from N;
A2selected from CH, CF, CCH3Or C (OCH)3);
A3Selected from CH or CCH3
R1Selected from H or Me;
R2is cyclopentyl;
R3is selected from C1-6Alkyl radical, C1-6Haloalkyl, -O-C1-6alkylene-R4Or the following groups:
Figure FDA0002632462510000021
R4selected from H, Me, Et, iPr, NH2、NMe2、CH2CH2OH or CH2CH2CH2OH;
R8Selected from H, Me, CF3Et, cyclopropyl or cyclobutyl.
3. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,
A1is selected from N;
A2selected from CH, C (halogen), C (C)1-6Alkyl) Or C (OC)1-6Alkyl groups);
A3selected from CH or C (C)1-6Alkyl groups);
R1is C1-6An alkyl group;
R2is C3-7A cycloalkyl group;
R3is selected from
Figure FDA0002632462510000022
Figure FDA0002632462510000023
R4Selected from H, C1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2or-C1-6alkylene-OH;
R8selected from H, C1-6Alkyl radical, C1-6Haloalkyl or C3-7A cycloalkyl group.
4. A compound according to claim 3, or a pharmaceutically acceptable salt thereof,
A1is selected from N;
A2selected from CH, CF, CCH3Or C (OCH)3);
A3Selected from CH or CCH3
R1Is Me;
R2is cyclopentyl;
R3is selected from
Figure FDA0002632462510000031
Figure FDA0002632462510000032
R4Selected from H, Me, Et, iPr, NH2、NMe2、CH2CH2OH or CH2CH2CH2OH;
R8Selected from H, Me, CF3Et, cyclopropyl or cyclobutyl.
5. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,
A1is N;
A2selected from CH or C (C)1-6Alkyl groups);
A3selected from CH or C (C)1-6Alkyl groups);
R1is C1-6An alkyl group;
R2is C3-7A cycloalkyl group;
R3is composed of
Figure FDA0002632462510000033
R4Is selected from H or C1-6An alkyl group;
R8selected from H, C1-6Alkyl radical, C1-6Haloalkyl or C3-7A cycloalkyl group.
6. The compound according to claim 5, or a pharmaceutically acceptable salt thereof,
A1is N;
A2selected from CH or CCH3
A3Selected from CH or CCH3
R1Is Me;
R2is cyclopentyl;
R3is composed of
Figure FDA0002632462510000034
R4Selected from H, Me or Et;
R8selected from H, Me, CF3Et or cyclopropyl.
7. A compound of formula (VI), or a pharmaceutically acceptable salt thereof:
Figure FDA0002632462510000041
wherein the content of the first and second substances,
A1is N;
A2is CH;
A3is CH;
R1is C1-6An alkyl group;
R2is C3-7A cycloalkyl group;
R3is composed of
Figure FDA0002632462510000042
R4Is selected from H or C1-6An alkyl group;
R8is C1-6An alkyl group.
8. The compound according to claim 7, or a pharmaceutically acceptable salt thereof,
A1is N;
A2is CH;
A3is CH;
R1is Me;
R2is cyclopentyl;
R3is composed of
Figure FDA0002632462510000043
R4Selected from H or Me;
R8is Me.
9. A compound, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the following:
Figure FDA0002632462510000044
Figure FDA0002632462510000051
Figure FDA0002632462510000061
Figure FDA0002632462510000071
10. a pharmaceutical composition comprising a compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
11. The pharmaceutical composition of claim 10, further comprising an additional therapeutic agent.
12. Use of a compound according to any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment and/or prevention of a CDK mediated disorder.
13. Use according to claim 12 wherein the CDK mediated disorder is mediated by CDK4 and/or CDK 6.
14. Use according to claim 12 or 13, wherein the disease is selected from the group consisting of cell proliferative diseases, cardiovascular disorders, infectious diseases, chronic inflammatory diseases, autoimmune disorders and other cell proliferative disorders.
15. The use of claim 14, wherein the cell proliferative disorder is cancer.
16. The use of claim 15, wherein the cancer is selected from the group consisting of a solid tumor and a hematologic malignancy.
17. The use of claim 16, wherein the solid and hematological malignancies are selected from the group consisting of breast cancer, neuroblastoma, malignant rhabdomyoma, glioma, lung cancer, colorectal cancer, gastric cancer, gastrointestinal stromal tumor, hepatocellular carcinoma, prostate cancer, sarcoma, ovarian cancer, cervical cancer, pancreatic cancer, melanoma, thyroid cancer, cholangiocarcinoma, endometrial cancer, renal cancer, mesothelioma, lymphoma, leukemia and multiple myeloma.
18. The use of claim 16, wherein the solid and hematological malignancies are well-differentiated and de-differentiated liposarcomas, non-hodgkin's lymphomas, mantle cell lymphomas, anaplastic large cell lymphomas, or acute myeloid leukemia.
19. The use of claim 14, wherein the cardiovascular disorder is selected from the group consisting of restenosis, atherosclerosis, vascular smooth muscle proliferation and intimal hyperplasia following balloon angioplasty, and other vascular disorders caused by abnormal cell proliferation.
20. The use of claim 14, wherein the infectious disease is selected from the group consisting of fungal, protozoal parasite and DNA and RNA virus infections.
21. The use of claim 20, wherein the infectious disease is an infection by plasmodium falciparum.
22. The use of claim 20, wherein the infectious disease is selected from herpes simplex virus infection.
23. The use of claim 14, wherein the chronic inflammatory disease is rheumatoid arthritis.
24. The use of claim 14, wherein the other cell proliferative disorder is selected from psoriasis, glomerulonephritis and lupus.
25. The use of claim 24, wherein said psoriasis is characterized by keratinocyte hyperproliferation.
CN201810053736.3A 2017-01-24 2018-01-19 6-substituted pyrido [2,3-D ] pyrimidines as protein kinase inhibitors Active CN108191857B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2017100596526 2017-01-24
CN201710059652 2017-01-24

Publications (2)

Publication Number Publication Date
CN108191857A CN108191857A (en) 2018-06-22
CN108191857B true CN108191857B (en) 2020-10-23

Family

ID=62590224

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810053736.3A Active CN108191857B (en) 2017-01-24 2018-01-19 6-substituted pyrido [2,3-D ] pyrimidines as protein kinase inhibitors

Country Status (2)

Country Link
CN (1) CN108191857B (en)
HK (1) HK1257799A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109956940A (en) * 2019-05-14 2019-07-02 上海贤鼎生物科技有限公司 A kind of method that the rich former times cloth intermediate cyano reaction of pa prepares heteroaryl cyanide
TWI823420B (en) * 2021-06-09 2023-11-21 大陸商鄭州同源康醫藥有限公司 Compounds useful as CDK kinase inhibitors and uses thereof
WO2023177356A2 (en) * 2022-03-18 2023-09-21 Engine Biosciences Pte. Ltd. Compounds and method for pkmyt1 inhibition
CN117430597A (en) * 2022-07-14 2024-01-23 浙江同源康医药股份有限公司 Compounds useful as CDK4 kinase inhibitors and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4343534B2 (en) * 2001-03-02 2009-10-14 ゲーペーツェー バイオテック アクチェンゲゼルシャフト Three hybrid assay system
CN101001857B (en) * 2002-01-22 2011-06-22 沃尼尔·朗伯有限责任公司 2-(pyridin-2-ylamino)-pyrido [2,3-d]pyrimidin-7-ones
JP2007523151A (en) * 2004-02-18 2007-08-16 ワーナー−ランバート カンパニー リミテッド ライアビリティー カンパニー 2- (Pyridin-3-ylamino) -pyrido [2,3-d] pyrimidin-7-one
JP2013507395A (en) * 2009-10-09 2013-03-04 アフラクシス・インコーポレイテッド 8-Ethyl-6- (aryl) pyrido [2,3-D] pyrimidin-7 (8H) -one for the treatment of CNS disorders

Also Published As

Publication number Publication date
HK1257799A1 (en) 2019-11-01
CN108191857A (en) 2018-06-22

Similar Documents

Publication Publication Date Title
CN109803972B (en) 1H-imidazole [4,5-H ] quinazoline compound as protein kinase inhibitor
CN108602776B (en) Heteroaryl compounds as IRAK inhibitors and uses thereof
CN107074859B (en) Cyclohexylethyl substituted diaza-and triaza-tricyclics as indoleamine-2, 3-dioxygenase (IDO) antagonists for the treatment of cancer
CN108191857B (en) 6-substituted pyrido [2,3-D ] pyrimidines as protein kinase inhibitors
CN114502158A (en) IRAK degradation agent and application thereof
TW201838966A (en) Substituted dihydroindene-4-carboxamides and analogs thereof, and methods using same
JP6835727B2 (en) Macrocycles and their use as IRAK1 / 4 inhibitors
JP2018531225A6 (en) Heteroaryl compounds as IRAK inhibitors and uses thereof
CN112047938B (en) 2,4, 6-trisubstituted pyrimidine compounds as ATR kinase inhibitors
KR102464677B1 (en) 1H-pyrazolo[4,3-H]quinazoline compound serving as protein kinase inhibitor
CN108473478A (en) TBK/IKK epsilon inhibitor compounds and application thereof
JP2019142874A (en) Piperidine cxcr7 receptor modulators
JP2020537671A (en) Pyrimidine TBK / IKKε inhibitor compounds and their use
CN109923115B (en) Imidazo [1',2':1,6] pyrido [2,3-D ] pyrimidines as protein kinase inhibitors
JP7316288B2 (en) Quinoline compounds and their use as IRAK inhibitors
CN112574208B (en) Substituted fused tricyclic derivatives, compositions and uses thereof
US11993604B2 (en) Substituted pyrazolo[4,3-H]quinazolines as protein kinase inhibitors
TWI802605B (en) PYRIMIDINE TBK/IKKε INHIBITOR COMPOUNDS AND USES THEREOF
TW202409011A (en) Inhibitors of human respiratory syncytial virus and metapneumovirus
CN116096721A (en) 1H-imidazo [4,5-H ] quinazoline compounds as novel selective FLT3 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20190124

Address after: 221000 office building 7, Oriental Pearl District, Yunlong District, Xuzhou, Jiangsu.

Applicant after: Shengke Pharmaceutical (Jiangsu) Co., Ltd.

Address before: 610041 13-2 Building No. 66, Fourth Street, Tianfu, Chengdu High-tech Zone, Sichuan Province

Applicant before: Chengdu excellent Sally medical science and Technology Co., Ltd.

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1257799

Country of ref document: HK

GR01 Patent grant
GR01 Patent grant
CP02 Change in the address of a patent holder
CP02 Change in the address of a patent holder

Address after: 5 / F, building 6, 88 tonganzhen North Road, high tech Zone, Suzhou City, Jiangsu Province

Patentee after: Shengke Pharmaceutical (Jiangsu) Co.,Ltd.

Address before: 221000 office building 7, Oriental Pearl District, Yunlong District, Xuzhou, Jiangsu.

Patentee before: Shengke Pharmaceutical (Jiangsu) Co.,Ltd.